1
|
Cui Z, He J, Li A, Wang J, Yang Y, Wang K, Liu Z, Ouyang Q, Su Z, Hu P, Xiao G. Novel insights into non-coding RNAs and their role in hydrocephalus. Neural Regen Res 2026; 21:636-647. [PMID: 39688559 DOI: 10.4103/nrr.nrr-d-24-00963] [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: 08/20/2024] [Accepted: 11/16/2024] [Indexed: 12/18/2024] Open
Abstract
A large body of evidence has highlighted the role of non-coding RNAs in neurodevelopment and neuroinflammation. This evidence has led to increasing speculation that non-coding RNAs may be involved in the pathophysiological mechanisms underlying hydrocephalus, one of the most common neurological conditions worldwide. In this review, we first outline the basic concepts and incidence of hydrocephalus along with the limitations of existing treatments for this condition. Then, we outline the definition, classification, and biological role of non-coding RNAs. Subsequently, we analyze the roles of non-coding RNAs in the formation of hydrocephalus in detail. Specifically, we have focused on the potential significance of non-coding RNAs in the pathophysiology of hydrocephalus, including glymphatic pathways, neuroinflammatory processes, and neurological dysplasia, on the basis of the existing evidence. Lastly, we review the potential of non-coding RNAs as biomarkers of hydrocephalus and for the creation of innovative treatments.
Collapse
Affiliation(s)
- Zhiyue Cui
- Department of Diagnostic Radiology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan Province, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Jian He
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - An Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Junqiang Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Yijian Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Kaiyue Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Zhikun Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Qian Ouyang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Department of Neurosurgery, Zhuzhou Hospital, Central South University Xiangya School of Medicine, Zhuzhou, Hunan Province, China
| | - Zhangjie Su
- Department of Neurosurgery, Addenbrooke 's Hospital, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge, UK
| | - Pingsheng Hu
- Department of Diagnostic Radiology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan Province, China
| | - Gelei Xiao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| |
Collapse
|
2
|
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
|
3
|
Fang Y, Peng J, Chu T, Gao F, Xiong F, Tu Y. Glymphatic system dysfunction in adult ADHD: Relationship to cognitive performance. J Affect Disord 2025; 379:150-158. [PMID: 40081578 DOI: 10.1016/j.jad.2025.03.059] [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: 10/29/2024] [Revised: 03/09/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
OBJECTIVES While attention-deficit/hyperactivity disorder (ADHD) persists into adulthood, the relationship between glymphatic system function and cognitive performance in adult ADHD remains unclear. This study investigated the association between glymphatic system markers and cognitive outcomes in adults with ADHD. METHODS This case-control study includes 41 adults with ADHD and 108 age-matched healthy controls (HCs). Glymphatic function was evaluated using choroid plexus volume (CPV), diffusion tensor imaging along the perivascular space (DTI-ALPS) index and coupling between blood‑oxygen-level-dependent signals and cerebrospinal fluid signals (BOLD-CSF coupling). Cognitive performance was measured using standardized neuropsychological tests. RESULTS Compared with HCs, adults with ADHD exhibited significantly lower bilateral and whole-brain ALPS indices (P < 0.05). Although CPV was increased in the ADHD group, this difference did not reach statistical significance, and no significant differences were observed in BOLD-CSF coupling between the two groups. Furthermore, whole-brain ALPS indices were positively associated with visual memory performance (r = 0.422, P = 0.005), an effect that was more pronounced in the right hemisphere (r = 0.458, P = 0.002). LIMITATIONS The cross-sectional design limits causal inferences, and the effects of medication were not fully accounted for. CONCLUSIONS These findings identify an association between glymphatic dysfunction and cognitive impairment in adults with ADHD. The observed correlation suggests that alterations in glymphatic function may underlie ADHD-related cognitive deficits. Targeting these pathways could provide novel therapeutic opportunities in the management of adult ADHD.
Collapse
Affiliation(s)
- Yan Fang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Juan Peng
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Tiantian Chu
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Feng Gao
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Fei Xiong
- Department of Radiology, General Hospital of Central Theater Command, Wuhan 430070, China
| | - Ye Tu
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
4
|
McInvale JJ, Kuper LC, Li E, Bonanno J, Lorman D, Gumenick R, Vincenti SL, Newman LA. Estradiol effects on astrocytic aquaporin 4 and glutamate transporter 1 expression contribute to shifts in brain dynamics supporting spatial working memory. Behav Brain Res 2025; 487:115578. [PMID: 40199402 DOI: 10.1016/j.bbr.2025.115578] [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: 12/31/2024] [Revised: 03/04/2025] [Accepted: 04/04/2025] [Indexed: 04/10/2025]
Abstract
Estrogenic effects on astrocytes improve glutamate recycling and water homeostasis for neuroprotection in pathology. Estrogens also enhance spatial learning and memory. The current study looked at the effect of 17β-estradiol (E2) on astrocytic glutamate transporter 1 (GLT-1) and aquaporin 4 (AQP4) in the prelimbic cortex (PrL) and dorsal hippocampus (dHC), areas active in spatial (allocentric) working memory in comparison to dorsolateral striatum (dlStr) which is involved in response or egocentric learning. Ovariectomized, female, Long Evans rats received 0, 4.5 µg/kg, or 45 µg/kg of E2 in a sesame oil vehicle 24 and 48 h prior to a delayed spontaneous alternation task (dSA). In line with previous research dSA performance significantly improved with administration of E2 as compared to sesame oil vehicle. AQP4 and GLT-1 levels were brain area specific and E2 enhanced AQP4 and GLT-1 in brain areas associated with spatial working memory (PrL and dHC) as compared to dlStr. Additionally, AQP4 was found to have the highest density in the unmyelinated axon rich hilus while GLT-1 showed the highest density in the synaptically dense molecular layer. However, AQP4 density in the stratum radiatum was similar to the hilus after dSA, potentially supporting dynamic changes in AQP4 response to natural cognitive activity. Hilar and prelimbic AQP4 area stained was also negatively correlated with performance on the dSA, which supports the theory of increased polarity of AQP4 with healthy cognitive function. These data suggest astrocytic water and glutamate homeostasis shift with high levels of estrogens to support spatial strategies.
Collapse
Affiliation(s)
- Julie J McInvale
- Department of Psychological Science, Neuroscience and Behavior Program, Vassar College, Poughkeepsie, NY, USA
| | - Louisa C Kuper
- Department of Psychological Science, Neuroscience and Behavior Program, Vassar College, Poughkeepsie, NY, USA
| | - Evelyn Li
- Department of Psychological Science, Neuroscience and Behavior Program, Vassar College, Poughkeepsie, NY, USA
| | - James Bonanno
- Department of Psychological Science, Neuroscience and Behavior Program, Vassar College, Poughkeepsie, NY, USA
| | - Daniella Lorman
- Department of Psychological Science, Neuroscience and Behavior Program, Vassar College, Poughkeepsie, NY, USA
| | - Ruby Gumenick
- Department of Psychological Science, Neuroscience and Behavior Program, Vassar College, Poughkeepsie, NY, USA
| | - Sydney L Vincenti
- Department of Psychological Science, Neuroscience and Behavior Program, Vassar College, Poughkeepsie, NY, USA
| | - Lori A Newman
- Department of Psychological Science, Neuroscience and Behavior Program, Vassar College, Poughkeepsie, NY, USA.
| |
Collapse
|
5
|
Wilson AC, Pountney DL, Khoo TK. Therapeutic Mechanisms of Exercise in Parkinson's Disease. Int J Mol Sci 2025; 26:4860. [PMID: 40429998 DOI: 10.3390/ijms26104860] [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: 04/17/2025] [Revised: 05/15/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025] Open
Abstract
Despite being the second-most common neurodegenerative disease, the etiology of Parkinson's disease (PD) remains uncertain with current knowledge suggestive of multiple risk factors. Furthermore, curative treatment does not yet exist, and treatment is primarily symptomatic in nature. For this reason, supportive therapies such as exercise are a crucial tool in PD management. It is useful to better understand how exercise affects the brain and body in the context of PD to guide clinical decision-making and determine the optimal exercise intensity and modality for PD patients. This review outlines the various mechanisms by which exercise can be beneficial as a therapeutic option in PD.
Collapse
Affiliation(s)
- Alice C Wilson
- School of Medicine and Dentistry, Griffith University, Gold Coast, QLD 4222, Australia
| | - Dean L Pountney
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, QLD 4222, Australia
| | - Tien K Khoo
- School of Medicine and Dentistry, Griffith University, Gold Coast, QLD 4222, Australia
- Graduate School of Medicine, University of Wollongong, Wollongong, NSW 2500, Australia
- Northern New South Wales Local Health District, NSW Health, Lismore, NSW 2480, Australia
| |
Collapse
|
6
|
Yang Z, Gong S, Zhang J, Zhang Y, Liu H, Luo Y, Zhong L, Ou Z, Yan Z, Zhang W, Xu J, Peng K, Zhi L, Liu G. Sleep disturbances are related to glymphatic dysfunction in blepharospasm. Neuroscience 2025; 573:228-236. [PMID: 40127757 DOI: 10.1016/j.neuroscience.2025.03.044] [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: 12/11/2024] [Revised: 03/13/2025] [Accepted: 03/19/2025] [Indexed: 03/26/2025]
Abstract
Research has shown a close relationship between sleep and glymphatic function, with impaired glymphatic function potentially contributing to sleep problems in a bidirectional way. However, its role in sleep disturbances associated with dystonia remains unknown. This cross-sectional study aimed to investigate whether glymphatic function is impaired in dystonia and to explore its relationship with sleep disturbances. We conducted structural magnetic resonance imaging and diffusion tensor imaging (DTI) on two large cohorts: patients with blepharospasm (BSP, n = 45) and cervical dystonia (CD, n = 43), alongside age- and sex-matched healthy controls (HCs). Anxiety, depression, and sleep quality were evaluated using the Hamilton Anxiety Scale (HAMA), Hamilton Depression Scale (HAMD), and Pittsburgh Sleep Quality Index (PSQI), respectively. Analysis along the perivascular space (DTI-ALPS) index and choroid plexus volume (CPV) was used to assess glymphatic function in these participants. Patients with BSP and CD had higher HAMA, HAMD, and PSQI scores than those of HCs. Patients with BSP exhibited a lower DTI-ALPS index and larger CPV than those of HCs, while no significant differences were found between CD and HCs. In BSP and CD, PSQI scores positively correlated with HAMA and HAMD scores and negatively with the DTI-ALPS index in BSP. Multivariate analysis identified the DTI-ALPS index as a dependent predictive factor of the PSQI in patients with BSP. Our findings suggest that glymphatic function varies across types of focal dystonia, with glymphatic dysfunction potentially playing an important role in the pathogenesis of sleep disturbances in BSP.
Collapse
Affiliation(s)
- Zhengkun Yang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, China
| | - Shiyuan Gong
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, China
| | - Jiana Zhang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, China
| | - Yue Zhang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, China
| | - Huiming Liu
- Department of Medical Imaging, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yuhan Luo
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, China
| | - Linchang Zhong
- Department of Medical Imaging, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Zilin Ou
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, China
| | - Zhicong Yan
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, China
| | - Weixi Zhang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, China
| | - Jinping Xu
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Kangqiang Peng
- Department of Medical Imaging, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Lanlan Zhi
- Department of Medical Imaging, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| | - Gang Liu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, China.
| |
Collapse
|
7
|
Rieff M, Holzberger F, Lapina O, Ringstad G, Magnus Valnes L, Warsza B, Kristian Eide P, Mardal K, Wohlmuth B. U-Net-Based Prediction of Cerebrospinal Fluid Distribution and Ventricular Reflux Grading. NMR IN BIOMEDICINE 2025; 38:e70029. [PMID: 40229147 PMCID: PMC11996590 DOI: 10.1002/nbm.70029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 03/04/2025] [Accepted: 03/05/2025] [Indexed: 04/16/2025]
Abstract
Previous work indicates evidence that cerebrospinal fluid (CSF) plays a crucial role in brain waste clearance processes and that altered flow patterns are associated with various diseases of the central nervous system. In this study, we investigate the potential of deep learning to predict the distribution in human brain of a gadolinium-based CSF contrast agent (tracer) administered intrathecal. For this, T1-weighted magnetic resonance imaging (MRI) scans taken at multiple time points before and after injection were utilized. We propose a U-net-based supervised learning model to predict pixel-wise signal increase at its peak after 24 h. Performance is evaluated based on different tracer distribution stages provided during training, including predictions from baseline scans taken before injection. Our findings show that training with imaging data from only the first 2-h postinjection yields tracer flow predictions comparable to models trained with additional later-stage scans. Validation against ventricular reflux gradings from neuroradiologists confirmed alignment with expert evaluations. These results demonstrate that deep learning-based methods for CSF flow prediction deserve more attention, as minimizing MR imaging without compromising clinical analysis could enhance efficiency, improve patient well-being and lower healthcare costs.
Collapse
Affiliation(s)
- Melanie Rieff
- Department of Mathematics, School of Computation, Information, and TechnologyTechnical University of MunichGarchingGermany
- Department of Computer ScienceETH ZurichZurichSwitzerland
| | - Fabian Holzberger
- Department of Mathematics, School of Computation, Information, and TechnologyTechnical University of MunichGarchingGermany
| | - Oksana Lapina
- Department of RadiologyOslo University Hospital RikshospitaletOsloNorway
| | - Geir Ringstad
- Department of RadiologyOslo University Hospital RikshospitaletOsloNorway
- Department of Geriatrics and Internal MedicineSorlandet HospitalArendalNorway
- KG Jebsen Centre for Brain Fluid Research, Institute of Clinical Medicine, Faculty of MedicineUniversity of OsloOsloNorway
- Institute of Clinical MedicineUniversity of OsloOsloNorway
| | - Lars Magnus Valnes
- Department of NeurosurgeryOslo University Hospital RikshospitaletOsloNorway
| | - Bogna Warsza
- Department of RadiologyOslo University Hospital RikshospitaletOsloNorway
| | - Per Kristian Eide
- KG Jebsen Centre for Brain Fluid Research, Institute of Clinical Medicine, Faculty of MedicineUniversity of OsloOsloNorway
- Institute of Clinical MedicineUniversity of OsloOsloNorway
- Department of NeurosurgeryOslo University Hospital RikshospitaletOsloNorway
| | - Kent‐André Mardal
- KG Jebsen Centre for Brain Fluid Research, Institute of Clinical Medicine, Faculty of MedicineUniversity of OsloOsloNorway
- Department of MathematicsUniversity of OsloOsloNorway
- Department of Numerical Analysis and Scientific ComputingSimula Research LaboratoryOsloNorway
| | - Barbara Wohlmuth
- Department of Mathematics, School of Computation, Information, and TechnologyTechnical University of MunichGarchingGermany
| |
Collapse
|
8
|
Massoud AT, Noltensmeyer DA, Juranek J, Cox CS, Velasquez FC, Zhu B, Sevick-Muraca EM, Shah MN. Insights into the Role of the Glymphatic System in the Pathogenesis of Post-hemorrhagic Hydrocephalus. Mol Neurobiol 2025; 62:6537-6543. [PMID: 39821726 PMCID: PMC11952971 DOI: 10.1007/s12035-025-04692-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 01/06/2025] [Indexed: 01/19/2025]
Abstract
Recently, it has been well-established that the glymphatic or glial-lymphatic system plays a vital role in the pathophysiology of various neurological compromise, especially hydrocephalus (HCP). Till now, the complete pathway is not yet fully understood, and little evidence is available from the literature that links hydrocephalus to disorders of the glymphatic system. Most published molecular studies and animal research have shown that, in models with hydrocephalus, the drainage of cerebrospinal fluid (CSF) via the glymphatic system is disrupted. This is strongly observed in normal pressure and post-hemorrhagic hydrocephalus cases. A thorough search of the literature to date yields scarce evidence on studies conducted on humans. Despite major similarities between non-human and human glymphatic pathways, the need for studies conducted on humans is becoming more urgent as the glymphatic pathway has been shown to be a good candidate for therapeutic intervention. In this review, we collect and report the most updated evidence addressing the glymphatic drainage pathways and their associations with the development of various types of hydrocephalus. In addition, we reveal the current scientific gap in human studies and our recommendations for the conduction of future clinical studies.
Collapse
Affiliation(s)
- Ahmed T Massoud
- Department of Pediatric Surgery, Division of Neurosurgery, McGovern Medical School at UTHealth, Houston, TX, USA.
- Department of Neurosurgery, McGovern Medical School, UTHealth Houston, Houston, TX, USA.
| | - Dillon A Noltensmeyer
- Department of Neurosurgery, McGovern Medical School, UTHealth Houston, Houston, TX, USA
| | - Jenifer Juranek
- Department of Pediatric Surgery, Division of Neurosurgery, McGovern Medical School at UTHealth, Houston, TX, USA
| | - Charles S Cox
- Department of Pediatric Surgery, Division of Neurosurgery, McGovern Medical School at UTHealth, Houston, TX, USA
| | - Fred Christian Velasquez
- Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Banghe Zhu
- Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Eva M Sevick-Muraca
- Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Manish N Shah
- Department of Pediatric Surgery, Division of Neurosurgery, McGovern Medical School at UTHealth, Houston, TX, USA
- Department of Neurosurgery, McGovern Medical School, UTHealth Houston, Houston, TX, USA
| |
Collapse
|
9
|
Valdes-Hernandez PA, Montesino-Goicolea S, Laffitte Nodarse C, Johnson AJ, Fillingim RB, Cruz-Almeida Y. Widespread and prolonged pain may reduce brain clearance capacity only via sleep impairment: Evidence from participants with knee pain. THE JOURNAL OF PAIN 2025; 30:105356. [PMID: 40032053 DOI: 10.1016/j.jpain.2025.105356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/14/2025] [Accepted: 02/25/2025] [Indexed: 03/05/2025]
Abstract
The brain is key in the pain-sleep relationship, and sleep is needed for glymphatic clearance. However, no studies have examined how pain relates to the glymphatic system (GS). Characterizing the GS alongside sleep quality in well-characterized pain samples is essential for advancing this understanding. Non-invasive MRI techniques, such as Diffusion Tensor Imaging-Analysis aLong the Perivascular Space (DTI-ALPS), are particularly valuable as they are approved for humans. Although the relationship between the DTI-ALPS index and the GS is primarily deductive, the index may be a proxy for waste clearance capacity in deep white matter. Its sensitivity to interstitial space changes-known to be modulated by norepinephrine-offers a unique opportunity to investigate how sleep impairment and chronic pain regulation affect specific components of brain waste clearance. We thus fitted two longitudinal models linking pain, sleep quality and the DTI-ALPS index. We hypothesized that variations in pain characteristics would predict DTI-ALPS index changes, either directly or mediated by sleep quality changes. Alternatively, we hypothesized that variations in sleep quality would predict changes in pain characteristics via DTI-ALPS index modifications. Knee pain participants (n=87) completed an MRI and self-reported measures of pain and sleep impairment at baseline and two years later. We only found evidence supporting that more widespread and longer pain at baseline significantly influenced decreases in the DTI-ALPS index in the left hemisphere through increased sleep impairment two years later (p=0.039, corrected). PERSPECTIVE: Findings highlight the need for research on the relationship between pain and sleep quality and its implications for brain health.
Collapse
Affiliation(s)
- Pedro A Valdes-Hernandez
- Department of Community Dentistry and Behavioral Science, University of Florida, USA; Pain Research and Intervention Center of Excellence, University of Florida, USA; Center for Cognitive Aging and Memory, McKnight Brain Institute, University of Florida, USA
| | - Soamy Montesino-Goicolea
- Department of Community Dentistry and Behavioral Science, University of Florida, USA; Pain Research and Intervention Center of Excellence, University of Florida, USA; Center for Cognitive Aging and Memory, McKnight Brain Institute, University of Florida, USA
| | - Chavier Laffitte Nodarse
- Department of Community Dentistry and Behavioral Science, University of Florida, USA; Pain Research and Intervention Center of Excellence, University of Florida, USA; Center for Cognitive Aging and Memory, McKnight Brain Institute, University of Florida, USA
| | - Alisa J Johnson
- Department of Community Dentistry and Behavioral Science, University of Florida, USA; Pain Research and Intervention Center of Excellence, University of Florida, USA
| | - Roger B Fillingim
- Department of Community Dentistry and Behavioral Science, University of Florida, USA; Pain Research and Intervention Center of Excellence, University of Florida, USA
| | - Yenisel Cruz-Almeida
- Department of Community Dentistry and Behavioral Science, University of Florida, USA; Pain Research and Intervention Center of Excellence, University of Florida, USA; Center for Cognitive Aging and Memory, McKnight Brain Institute, University of Florida, USA; Department of Neuroscience, College of Medicine, University of Florida, USA.
| |
Collapse
|
10
|
Kawiková I, Špička V, Lai JCK, Askenase PW, Wen L, Kejík Z, Jakubek M, Valeš K, Španiel F. Extracellular vesicles as precision therapeutics for psychiatric conditions: targeting interactions among neuronal, glial, and immune networks. Front Immunol 2025; 16:1454306. [PMID: 40264776 PMCID: PMC12011847 DOI: 10.3389/fimmu.2025.1454306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 02/14/2025] [Indexed: 04/24/2025] Open
Abstract
The critical role of the immune system in brain function and dysfunction is well recognized, yet development of immune therapies for psychiatric diseases has been slow due to concerns about iatrogenic immune deficiencies. These concerns are emphasized by the lack of objective diagnostic tools in psychiatry. A promise to resolve this conundrum lies in the exploitation of extracellular vesicles (EVs) that are physiologically produced or can be synthetized. EVs regulate recipient cell functions and offer potential for EVs-based therapies. Intranasal EVs administration enables the targeting of specific brain regions and functions, thereby facilitating the design of precise treatments for psychiatric diseases. The development of such therapies requires navigating four dynamically interacting networks: neuronal, glial, immune, and EVs. These networks are profoundly influenced by brain fluid distribution. They are crucial for homeostasis, cellular functions, and intercellular communication. Fluid abnormalities, like edema or altered cerebrospinal fluid (CSF) dynamics, disrupt these networks, thereby negatively impacting brain health. A deeper understanding of the above-mentioned four dynamically interacting networks is vital for creating diagnostic biomarker panels to identify distinct patient subsets with similar neuro-behavioral symptoms. Testing the functional pathways of these biomarkers could lead to new therapeutic tools. Regulatory approval will depend on robust preclinical data reflecting progress in these interdisciplinary areas, which could pave the way for the design of innovative and precise treatments. Highly collaborative interdisciplinary teams will be needed to achieve these ambitious goals.
Collapse
Affiliation(s)
- Ivana Kawiková
- National Institute of Mental Health, Klecany, Czechia
- Department of Medicine, Yale School of Medicine, New Haven, CT, United States
- Department of Biology, Hartford University, West Hartford, CT, United States
| | - Václav Špička
- Institute of Physics of the Czech Academy of Sciences, Prague, Czechia
| | - James C. K. Lai
- Department of Biomedical and Pharmaceutical Sciences, Idaho State University College of Pharmacy, Pocatello, ID, United States
- Department of Diagnostic Radiology and Biomedical Imaging, Magnetic Resonance Research Center, Yale School of Medicine, New Haven, CT, United States
| | - Philip W. Askenase
- Department of Medicine, Yale School of Medicine, New Haven, CT, United States
| | - Li Wen
- Department of Medicine, Yale School of Medicine, New Haven, CT, United States
| | - Zdeněk Kejík
- Biotechnology and Biomedical Center in Vestec (BIOCEV) , First Faculty of Medicine, Charles University, Vestec, Czechia
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - Milan Jakubek
- Biotechnology and Biomedical Center in Vestec (BIOCEV) , First Faculty of Medicine, Charles University, Vestec, Czechia
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - Karel Valeš
- National Institute of Mental Health, Klecany, Czechia
- 3rd Medical Faculty, Charles University, Prague, Czechia
| | - Filip Španiel
- National Institute of Mental Health, Klecany, Czechia
- 3rd Medical Faculty, Charles University, Prague, Czechia
| |
Collapse
|
11
|
Liang W, Sun W, Li C, Zhou J, Long C, Li H, Xu D, Xu H. Glymphatic system dysfunction and cerebrospinal fluid retention in gliomas: evidence from perivascular space diffusion and volumetric analysis. Cancer Imaging 2025; 25:51. [PMID: 40197529 PMCID: PMC11974089 DOI: 10.1186/s40644-025-00868-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 03/25/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Gliomas may impair glymphatic function and alter cerebrospinal fluid (CSF) dynamics through structural brain changes, potentially affecting peritumoral brain edema (PTBE) and fluid clearance. This study investigated the impact of gliomas on glymphatic system function and CSF volume via diffusion tensor imaging analysis along the perivascular space (DTI-ALPS) and volumetric magnetic resonance imaging (MRI), which clarified the relationships between tumor characteristics and glymphatic system disruption. METHODS In this prospective study, 112 glioma patients and 56 healthy controls underwent MRI to calculate DTI-ALPS indices and perform volumetric analyses of CSF, tumor, and PTBE. Statistical analyses were used to assess the relationships between the DTI-ALPS index, tumor volume, PTBE volume, and clinical characteristics. RESULTS Glioma patients had significantly lower DTI-ALPS indices (1.266 ± 0.258 vs. 1.395 ± 0.174, p < 0.001) and greater CSF volumes (174.53 ± 34.89 cm³ vs. 154.25 ± 20.89 cm³, p < 0.001) than controls did. The DTI-ALPS index was inversely correlated with tumor volume (r = -0.353, p < 0.001) and PTBE volume (r = -0.266, p = 0.015). High-grade gliomas were associated with lower DTI-ALPS indices and larger PTBE volumes (all p < 0.001). Tumor grade emerged as an independent predictor of the DTI-ALPS index in multivariate analysis (β = -0.244, p = 0.011). CONCLUSION Gliomas are associated with significant glymphatic dysfunction, as evidenced by reduced DTI-ALPS indices and increased CSF and PTBE volumes. The DTI-ALPS index serves as a potential biomarker of glymphatic disruption in glioma patients, offering insights into tumor-related fluid changes and the pathophysiology of brain-tumor interactions.
Collapse
Affiliation(s)
- Weiqiang Liang
- Department of Radiology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China
- Hubei Provincial Engineering Research Center of Multimodal Medical Imaging Technology and Clinical Application, Wuhan, China
| | - Wenbo Sun
- Department of Radiology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China
- Hubei Provincial Engineering Research Center of Multimodal Medical Imaging Technology and Clinical Application, Wuhan, China
| | - Chunyan Li
- Department of Radiology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China
- Hubei Provincial Engineering Research Center of Multimodal Medical Imaging Technology and Clinical Application, Wuhan, China
| | - Jie Zhou
- Department of Radiology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China
- Hubei Provincial Engineering Research Center of Multimodal Medical Imaging Technology and Clinical Application, Wuhan, China
| | - Changyou Long
- Department of Radiology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China
- Hubei Provincial Engineering Research Center of Multimodal Medical Imaging Technology and Clinical Application, Wuhan, China
| | - Huan Li
- Department of Radiology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China
- Hubei Provincial Engineering Research Center of Multimodal Medical Imaging Technology and Clinical Application, Wuhan, China
| | - Dan Xu
- Department of Nuclear Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, 169 Donghu Road, Wuhan, 430071, China.
- Hubei Provincial Engineering Research Center of Multimodal Medical Imaging Technology and Clinical Application, Wuhan, China.
| | - Haibo Xu
- Department of Radiology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China.
- Hubei Provincial Engineering Research Center of Multimodal Medical Imaging Technology and Clinical Application, Wuhan, China.
| |
Collapse
|
12
|
Zimmermann J, Boudriot C, Eipert C, Hoffmann G, Nuttall R, Neumaier V, Bonhoeffer M, Schneider S, Schmitzer L, Kufer J, Kaczmarz S, Hedderich DM, Ranft A, Golkowski D, Priller J, Zimmer C, Ilg R, Schneider G, Preibisch C, Sorg C, Zott B. Total cerebral blood volume changes drive macroscopic cerebrospinal fluid flux in humans. PLoS Biol 2025; 23:e3003138. [PMID: 40273212 PMCID: PMC12061420 DOI: 10.1371/journal.pbio.3003138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 05/08/2025] [Accepted: 04/01/2025] [Indexed: 04/26/2025] Open
Abstract
In the mammalian brain, the directed motion of cerebrospinal fluid (CSF-flux) is instrumental in the distribution and removal of solutes. Changes in total cerebral blood volume (CBV) have been hypothesized to drive CSF-flux. We tested this hypothesis in two multimodal brain imaging experiments in healthy humans, in which we drove large changes in total CBV by neuronal burst-suppression under anesthesia or by transient global vasodilation in a hypercapnic challenge. We indirectly monitored CBV changes with a high temporal resolution based on associated changes in total brain volume by functional MRI (fMRI) and measured cerebral blood flow by arterial spin-labeling. Relating CBV-sensitive signals to fMRI-derived measures of macroscopic CSF flow across the basal cisternae, we demonstrate that increasing total CBV extrudes CSF from the skull and decreasing CBV allows its influx. Moreover, CSF largely stagnates when CBV is stable. Together, our results establish the direct coupling between total CBV changes and CSF-flux.
Collapse
Affiliation(s)
- Juliana Zimmermann
- Department of Neuroradiology, School of Medicine and Health, Technical University of Munich, Munich, Germany
- Department of Anesthesiology and Intensive Care, School of Medicine and Health, Technical University of Munich, Munich, Germany
- TUM-Neuroimaging Center, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Clara Boudriot
- Department of Neuroradiology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Christiane Eipert
- Department of Neuroradiology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Gabriel Hoffmann
- Department of Neuroradiology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Rachel Nuttall
- Department of Neuroradiology, School of Medicine and Health, Technical University of Munich, Munich, Germany
- Department of Anesthesiology and Intensive Care, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Viktor Neumaier
- Department of Neuroradiology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Moritz Bonhoeffer
- Department of Neuroradiology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Sebastian Schneider
- Department of Neuroradiology, School of Medicine and Health, Technical University of Munich, Munich, Germany
- TUM-Neuroimaging Center, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Lena Schmitzer
- Department of Neuroradiology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Jan Kufer
- Department of Neuroradiology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Stephan Kaczmarz
- Department of Neuroradiology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Dennis M Hedderich
- Department of Neuroradiology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Andreas Ranft
- Department of Anesthesiology and Intensive Care, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Daniel Golkowski
- Department of Neurology, School of Medicine, Technical University of Munich, Munich, Germany
- Department of Neurology, Heidelberg University Hospital, Heidelberg, Germany
| | - Josef Priller
- Department of Psychiatry and Psychotherapy, School of Medicine and Health, Technical University of Munich, Munich, Germany
- Charité - Universitätsmedizin Berlin and DZNE, Neuropsychiatry, Berlin, Germany
- University of Edinburgh and UKI DRI, Edinburgh, United Kingdom
| | - Claus Zimmer
- Department of Neuroradiology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Rüdiger Ilg
- Department of Neurology, School of Medicine, Technical University of Munich, Munich, Germany
- Department of Neurology, Asklepios Stadtklinik Bad Tölz, Bad Tölz, Germany
| | - Gerhard Schneider
- Department of Anesthesiology and Intensive Care, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Christine Preibisch
- Department of Neuroradiology, School of Medicine and Health, Technical University of Munich, Munich, Germany
- TUM-Neuroimaging Center, School of Medicine and Health, Technical University of Munich, Munich, Germany
- Department of Neurology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Christian Sorg
- Department of Neuroradiology, School of Medicine and Health, Technical University of Munich, Munich, Germany
- TUM-Neuroimaging Center, School of Medicine and Health, Technical University of Munich, Munich, Germany
- Department of Psychiatry and Psychotherapy, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Benedikt Zott
- Department of Neuroradiology, School of Medicine and Health, Technical University of Munich, Munich, Germany
- TUM-Neuroimaging Center, School of Medicine and Health, Technical University of Munich, Munich, Germany
- Institute for Neuroscience, Technical University of Munich, Germany
- TUM Institute for Advanced Study, Garching, Germany
| |
Collapse
|
13
|
Huang S, Lin L, Gao X, Li Y, Qiu H, Deng C, Qian L, Chen Y, Tang W, Liang Y, Su S, Yang Z. Glymphatic system dysfunction in pediatric tourette syndrome Neuroimaging evidence from MRI metrics. J Psychiatr Res 2025; 184:1-7. [PMID: 40031125 DOI: 10.1016/j.jpsychires.2025.02.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND AND OBJECTIVES The glymphatic system, vital for brain waste clearance, is implicated in neurodevelopmental diseases, but its role in pediatric Tourette syndrome (TS) is not well understood. This study investigates structural and functional alterations in the glymphatic system in pediatric TS using non-invasive MRI techniques. METHODS This case-control study included 37 children with Tourette syndrome (TS) and 37 age- and gender-matched typically developing (TD) controls. We assessed brain volumetric differences and glymphatic function using two MRI metrics: perivascular space (PVS) burden for glymphatic influx and the DTI-ALPS index for waste clearance, with PVS quantified via semi-automated analysis of axial T2-weighted images. Correlations between MRI metrics and clinical symptoms in TS children were analyzed using partial correlations. RESULTS Children with Tourette syndrome (TS) exhibited significant reductions in brain parenchymal and white matter volume compared to typically developing (TD) children (all PFDR < 0.001), along with a higher perivascular space (PVS) volume (6.29 ± 3.62 mL vs. 4.76 ± 2.13 mL; PFDR = 0.046), indicating impaired glymphatic influx. The DTI-ALPS index was lower in TS (1.21 ± 0.18 vs. 1.46 ± 0.12; PFDR < 0.001), reflecting reduced waste clearance, and Dzassoc and Dzproj metrics were positively correlated with motor tic severity in TS (all P ≤ 0.02). CONCLUSIONS Our findings suggest significant glymphatic dysfunction in pediatric TS, indicating its role in the disorder's pathogenesis. Increased PVS burden and decreased DTI-ALPS index may serve as non-invasive biomarkers for diagnosing and understanding TS mechanisms.
Collapse
Affiliation(s)
- Shuzhen Huang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Liping Lin
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiang Gao
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yufen Li
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Huaqiong Qiu
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Chengfen Deng
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Long Qian
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, 100817, China
| | - Yingqian Chen
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Wen Tang
- Department of Pediatric Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yujian Liang
- Department of Pediatric Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Shu Su
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zhiyun Yang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
14
|
Huang Y, Zhen Z, Deng L, Ou P, Shi L, Shi F, Hua R, Wu J, Chen W, Wen R, Wang J, Liu C. Beyond the cerebellum: perivascular space burden in spinocerebellar ataxia type 3 extends to multiple brain regions. Brain Commun 2025; 7:fcaf118. [PMID: 40190350 PMCID: PMC11969673 DOI: 10.1093/braincomms/fcaf118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 02/05/2025] [Accepted: 03/26/2025] [Indexed: 04/09/2025] Open
Abstract
Spinocerebellar ataxia type 3 (SCA3) is an uncommon inherited (autosomal dominant) neurodegenerative disorder caused by abnormal accumulation of ataxin-3 protein. The perivascular space (PVS) burden reflects protein clearance and may worsen in SCA3 disease. This study aimed to quantify the PVS burden and investigate the relationship between the PVS burden and clinical characteristics in individuals with SCA3. This study enrolled 43 SCA3 patients and 43 age- and sex-matched healthy controls (HCs). The cross-sectional study assessed the severity of ataxia in SCA3 patients using the Scale for the Assessment and Rating of Ataxia (SARA) and the International Cooperative Ataxia Rating Scale (ICARS). Various cognitive functions were evaluated in all subjects using the Montreal Cognitive Assessment (MoCA), Rapid Verbal Retrieval (RAR) and Digital Span Test (DST) scales. MRI was used to automatically segment the PVS in all subjects and quantify the PVS burden in 15 brain regions. Compared with the HCs, the SCA3 patients showed a significantly higher PVS burden in the basal ganglia, temporal lobe, right parietal lobe and right cerebellum. There was a positive correlation in motor dysfunction between the PVS volume in the left parietal lobe, right cerebellum and PVS number in the right cerebellum with the SARA and ICARS scores. This study showed that SCA3 patients have an increased PVS burden in many brain regions, leading to motor impairment. The PVS burden could be a new imaging biomarker for disease monitoring and a therapeutic target for SCA3.
Collapse
Affiliation(s)
- Yonghua Huang
- 7T Magnetic Resonance Imaging Translational Medical Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
- Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
- Department of Radiology, The 940th Hospital of the Joint Logistics Support Force of the Chinese People's Liberation Army, Lanzhou 730050, China
| | - Zhiming Zhen
- 7T Magnetic Resonance Imaging Translational Medical Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
- Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Lihua Deng
- 7T Magnetic Resonance Imaging Translational Medical Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
- Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Peiling Ou
- 7T Magnetic Resonance Imaging Translational Medical Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
- Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Linfeng Shi
- 7T Magnetic Resonance Imaging Translational Medical Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
- Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Feng Shi
- Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd., Shanghai 200232, China
| | - Rui Hua
- Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd., Shanghai 200232, China
| | - Jiaojiao Wu
- Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd., Shanghai 200232, China
| | - Wei Chen
- MR Research Collaboration Teams, Siemens Healthineers Ltd., Guangzhou 510630, China
| | - Ru Wen
- 7T Magnetic Resonance Imaging Translational Medical Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
- Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Jian Wang
- 7T Magnetic Resonance Imaging Translational Medical Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
- Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Chen Liu
- 7T Magnetic Resonance Imaging Translational Medical Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
- Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| |
Collapse
|
15
|
Wu J, Li R, Wang J, Zhu H, Ma Y, You C, Shu K. Reactive Astrocytes in Glioma: Emerging Opportunities and Challenges. Int J Mol Sci 2025; 26:2907. [PMID: 40243478 PMCID: PMC11989224 DOI: 10.3390/ijms26072907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/16/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Gliomas are the most prevalent malignant tumors in the adult central nervous system (CNS). Glioblastoma (GBM) accounts for approximately 60-70% of primary gliomas. It is a great challenge to human health because of its high degree of malignancy, rapid progression, short survival time, and susceptibility to recurrence. Owing to the specificity of the CNS, the glioma microenvironment often contains numerous glial cells. Astrocytes are most widely distributed in the human brain and form reactive astrocyte proliferation regions around glioma tissue. In addition, astrocytes are activated under pathological conditions and regulate tumor and microenvironmental cells through cell-to-cell contact or the secretion of active substances. Therefore, astrocytes have attracted attention as important components of the glioma microenvironment. Here, we focus on the mechanisms of reactive astrocyte activation under glioma conditions, their contribution to the mechanisms of glioma genesis and progression, and their potential value as targets for clinical intervention in gliomas.
Collapse
Affiliation(s)
| | | | | | | | | | - Chao You
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095, Jie Fang Avenue, Qiao Kou District, Wuhan 430030, China; (J.W.); (J.W.); (H.Z.); (Y.M.)
| | - Kai Shu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095, Jie Fang Avenue, Qiao Kou District, Wuhan 430030, China; (J.W.); (J.W.); (H.Z.); (Y.M.)
| |
Collapse
|
16
|
Wang C, Fan X, Shi Y, Tang F. Radiation-Induced Brain Injury with Special Reference to Astrocytes as a Therapeutic Target. J Integr Neurosci 2025; 24:25907. [PMID: 40152565 DOI: 10.31083/jin25907] [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: 07/30/2024] [Revised: 10/22/2024] [Accepted: 11/06/2024] [Indexed: 03/29/2025] Open
Abstract
Radiotherapy is one of the primary modalities for oncologic treatment and has been utilized at least once in over half of newly diagnosed cancer patients. Cranial radiotherapy has significantly enhanced the long-term survival rates of patients with brain tumors. However, radiation-induced brain injury, particularly hippocampal neuronal damage along with impairment of neurogenesis, inflammation, and gliosis, adversely affects the quality of life for these patients. Astrocytes, a type of glial cell that are abundant in the brain, play essential roles in maintaining brain homeostasis and function. Despite their importance, the pathophysiological changes in astrocytes induced by radiation have not been thoroughly investigated, and no systematic or comprehensive review addressing the effects of radiation on astrocytes and related diseases has been conducted. In this paper, we review current studies on the neurophysiological roles of astrocytes following radiation exposure. We describe the pathophysiological changes in astrocytes, including astrogliosis, astrosenescence, and the associated cellular and molecular mechanisms. Additionally, we summarize the roles of astrocytes in radiation-induced impairments of neurogenesis and the blood-brain barrier (BBB). Based on current research, we propose that brain astrocytes may serve as potential therapeutic targets for treating radiation-induced brain injury (RIBI) and subsequent neurological and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Caiping Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 226001 Nantong, Jiangsu, China
- Radiation Physiology Laboratory, Singapore Nuclear Research and Safety Initiative, National University of Singapore, 138602 Singapore, Singapore
| | - Xingjuan Fan
- Department of Neurology, Affiliated Hospital of Nantong University, 226001 Nantong, Jiangsu, China
| | - Yunwei Shi
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 226001 Nantong, Jiangsu, China
- Radiation Physiology Laboratory, Singapore Nuclear Research and Safety Initiative, National University of Singapore, 138602 Singapore, Singapore
| | - Fengru Tang
- Radiation Physiology Laboratory, Singapore Nuclear Research and Safety Initiative, National University of Singapore, 138602 Singapore, Singapore
| |
Collapse
|
17
|
Lin B, Leong YY, Mohamad M. Glymphatic system dysfunction in cerebral infarction: advances and perspectives based on DTI-derived ALPS measures. Am J Transl Res 2025; 17:1630-1642. [PMID: 40226042 PMCID: PMC11982858 DOI: 10.62347/oqre2088] [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: 09/23/2024] [Accepted: 01/25/2025] [Indexed: 04/15/2025]
Abstract
The glymphatic pathway plays a crucial role in the clearance of metabolic byproducts and solutes from cerebral tissue. Dysfunction of the glymphatic pathway has been associated with various neurological disorders, including ischemic stroke. Diffusion tensor imaging (DTI) and the derived Analysis aLong the Perivascular Space (ALPS) have emerged as promising tools for evaluating glymphatic pathway function. This review aims to summarize the current evidence on the use of DTI-derived ALPS measures in assessing glymphatic dysfunction in ischemic stroke patients, and to explore their potential implications for diagnosis, prognostication, and treatment monitoring in this patient population.
Collapse
Affiliation(s)
- Bomiao Lin
- Center for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan MalaysiaJalan Raja Muda Abdul Aziz, Wilayah Persekutuan Kuala Lumpur, 50300, Malaysia
- Department of Radiology, Zhujiang Hospital of Southern Medical UniversityGuangzhou, Guangdong, China
| | - Yuh Yang Leong
- Department of Radiology, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif Bandar Tun Razak Cheras56000 Kuala Lumpur, Malaysia
| | - Mazlyfarina Mohamad
- Center for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan MalaysiaJalan Raja Muda Abdul Aziz, Wilayah Persekutuan Kuala Lumpur, 50300, Malaysia
| |
Collapse
|
18
|
Jackson ON, Keenan TF, Nelson-Maney NP, Rommel SA, McLellan WA, Pabst DA, Costidis AM, Caron KM, Kernagis DN, Rotstein DS, Thayer VG, Harms CA, Piscitelli-Doshkov MA, Doshkov P, Schweikert LE, Yopak KE, Braun M, Tift MS. Meningeal Lymphatic and Glymphatic Structures in a Pelagic Delphinid ( Delphinus delphis). Animals (Basel) 2025; 15:729. [PMID: 40076012 PMCID: PMC11899484 DOI: 10.3390/ani15050729] [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: 01/31/2025] [Revised: 02/23/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
The glymphatic system, an analog of the peripheral lymphatic system in the brain, and the meningeal lymphatic system are critical to central nervous system health. The glymphatic system functions to distribute cerebrospinal fluid and important compounds throughout the brain and to remove metabolic waste. The flow of cerebrospinal fluid through this system is affected by changes in cerebral blood flow, intracranial pressure, and vascular tone. Cetaceans experience profound cardiorespiratory alterations while diving that can directly affect cerebrospinal fluid and blood flow and, thus, glymphatic function. Our goal was to investigate glymphatic and lymphatic system structures, including perivascular spaces, aquaporin-4 water channels, meningeal lymphatic, and dural venous sinus vessels in the common dolphin (Delphinus delphis), using immunofluorescent labeling, histochemical staining, and postmortem computed tomography (CT) angiography. We highlight perivascular spaces and aquaporin-4 water channels surrounding blood vessels in the parenchyma and demonstrate evidence of meningeal lymphatic vessels and associated dural venous sinuses. These results demonstrate that common dolphins possess the key anatomical structures required for functional glymphatic and meningeal lymphatic systems. Future studies can build upon these anatomical discoveries to study the function and role of these systems in brain health in this species.
Collapse
Affiliation(s)
- Olivia N. Jackson
- Department of Biology and Marine Biology, College of Science and Engineering, University of North Carolina Wilmington, Wilmington, NC 28403, USA; (T.F.K.); (S.A.R.); (W.A.M.); (D.A.P.); (L.E.S.); (K.E.Y.); (M.S.T.)
| | - Tiffany F. Keenan
- Department of Biology and Marine Biology, College of Science and Engineering, University of North Carolina Wilmington, Wilmington, NC 28403, USA; (T.F.K.); (S.A.R.); (W.A.M.); (D.A.P.); (L.E.S.); (K.E.Y.); (M.S.T.)
| | - Nathan P. Nelson-Maney
- Department of Cell Biology and Physiology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (N.P.N.-M.); (K.M.C.)
| | - Sentiel A. Rommel
- Department of Biology and Marine Biology, College of Science and Engineering, University of North Carolina Wilmington, Wilmington, NC 28403, USA; (T.F.K.); (S.A.R.); (W.A.M.); (D.A.P.); (L.E.S.); (K.E.Y.); (M.S.T.)
| | - William A. McLellan
- Department of Biology and Marine Biology, College of Science and Engineering, University of North Carolina Wilmington, Wilmington, NC 28403, USA; (T.F.K.); (S.A.R.); (W.A.M.); (D.A.P.); (L.E.S.); (K.E.Y.); (M.S.T.)
| | - D. Ann Pabst
- Department of Biology and Marine Biology, College of Science and Engineering, University of North Carolina Wilmington, Wilmington, NC 28403, USA; (T.F.K.); (S.A.R.); (W.A.M.); (D.A.P.); (L.E.S.); (K.E.Y.); (M.S.T.)
| | - Alexander M. Costidis
- Department of Biology and Marine Biology, College of Science and Engineering, University of North Carolina Wilmington, Wilmington, NC 28403, USA; (T.F.K.); (S.A.R.); (W.A.M.); (D.A.P.); (L.E.S.); (K.E.Y.); (M.S.T.)
- Marine Mammal Solutions LLC, Norfolk, VA 23502, USA
| | - Kathleen M. Caron
- Department of Cell Biology and Physiology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (N.P.N.-M.); (K.M.C.)
| | - Dawn N. Kernagis
- DEEP, Bristol BS11 8AR, UK;
- Department of Neurosurgery, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | - Victoria G. Thayer
- Center for Marine Science and Technologies, North Carolina State University, Morehead City, NC 28557, USA; (V.G.T.); (C.A.H.)
- North Carolina Marine Fisheries, Department of Environmental Quality, Morehead City, NC 28557, USA
| | - Craig A. Harms
- Center for Marine Science and Technologies, North Carolina State University, Morehead City, NC 28557, USA; (V.G.T.); (C.A.H.)
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27695, USA
| | | | - Paul Doshkov
- Cape Hatteras National Seashore, Manteo, NC 27954, USA;
| | - Lorian E. Schweikert
- Department of Biology and Marine Biology, College of Science and Engineering, University of North Carolina Wilmington, Wilmington, NC 28403, USA; (T.F.K.); (S.A.R.); (W.A.M.); (D.A.P.); (L.E.S.); (K.E.Y.); (M.S.T.)
| | - Kara E. Yopak
- Department of Biology and Marine Biology, College of Science and Engineering, University of North Carolina Wilmington, Wilmington, NC 28403, USA; (T.F.K.); (S.A.R.); (W.A.M.); (D.A.P.); (L.E.S.); (K.E.Y.); (M.S.T.)
| | - Molly Braun
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
| | - Michael S. Tift
- Department of Biology and Marine Biology, College of Science and Engineering, University of North Carolina Wilmington, Wilmington, NC 28403, USA; (T.F.K.); (S.A.R.); (W.A.M.); (D.A.P.); (L.E.S.); (K.E.Y.); (M.S.T.)
| |
Collapse
|
19
|
Ajijola OA, Aksu T, Arora R, Biaggioni I, Chen PS, De Ferrari G, Dusi V, Fudim M, Goldberger JJ, Green AL, Herring N, Khalsa SS, Kumar R, Lakatta E, Mehra R, Meyer C, Po S, Stavrakis S, Somers VK, Tan AY, Valderrabano M, Shivkumar K. Clinical neurocardiology: defining the value of neuroscience-based cardiovascular therapeutics - 2024 update. J Physiol 2025; 603:1781-1839. [PMID: 40056025 DOI: 10.1113/jp284741] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 01/28/2025] [Indexed: 04/01/2025] Open
Abstract
The intricate role of the autonomic nervous system (ANS) in regulating cardiac physiology has long been recognized. Aberrant function of the ANS is central to the pathophysiology of cardiovascular diseases. It stands to reason, therefore, that neuroscience-based cardiovascular therapeutics hold great promise in the treatment of cardiovascular diseases in humans. A decade after the inaugural edition, this White Paper reviews the current state of understanding of human cardiac neuroanatomy, neurophysiology and pathophysiology in specific disease conditions, autonomic testing, risk stratification, and neuromodulatory strategies to mitigate the progression of cardiovascular diseases.
Collapse
Affiliation(s)
- Olujimi A Ajijola
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Center of Excellence, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Tolga Aksu
- Division of Cardiology, Yeditepe University Hospital, Istanbul, Türkiye
| | - Rishi Arora
- Division of Cardiology, Northwestern Feinberg School of Medicine, Chicago, IL, USA
| | - Italo Biaggioni
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Peng-Sheng Chen
- Department of Cardiology, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Gaetano De Ferrari
- Department of Medical Sciences, University of Turin, Italy and Division of Cardiology, Cardiovascular and Thoracic Department, 'Città della Salute e della Scienza' Hospital, Torino, Italy
| | - Veronica Dusi
- Department of Medical Sciences, University of Turin, Italy and Division of Cardiology, Cardiovascular and Thoracic Department, 'Città della Salute e della Scienza' Hospital, Torino, Italy
| | - Marat Fudim
- Division of Cardiology, Duke University Medical Center, Durham, NC, USA
| | - Jeffrey J Goldberger
- Division of Cardiology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Alexander L Green
- Department of Clinical Neurosciences, John Radcliffe Hospital, and Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Neil Herring
- Department for Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Sahib S Khalsa
- Department of Psychiatry, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Rajesh Kumar
- Department of Neurobiology and the Brain Research Institute, University of California, Los Angeles, CA, USA
| | - Edward Lakatta
- National Institute of Aging, National Institutes of Health, Bethesda, MD, USA
| | - Reena Mehra
- Division of Pulmonary Medicine, University of Washington, Seattle, WA, USA
| | - Christian Meyer
- Klinik für Kardiologie, Angiologie, Intensivmedizin, cNEP Research Consortium EVK, Düsseldorf, Germany
- Heart Rhythm Institute, Overland Park, KS, USA
| | - Sunny Po
- University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Stavros Stavrakis
- University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Virend K Somers
- Division of Cardiovascular Diseases, Mayo Clinic and Mayo Foundation, Rochester, MN, USA
| | - Alex Y Tan
- Division of Cardiology, Richmond Veterans Affairs Hospital, Richmond, VA, USA
| | - Miguel Valderrabano
- Methodist DeBakey Heart and Vascular Center and Methodist Hospital Research Institute, Houston Methodist Hospital, Houston, TX, USA
| | - Kalyanam Shivkumar
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Center of Excellence, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
20
|
Ji KH, Yun CH. Emerging Technologies to Track and Improve Sleep Health. Sleep Med Clin 2025; 20:47-55. [PMID: 39894598 DOI: 10.1016/j.jsmc.2024.10.011] [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
This review explores cutting-edge advancements, including wearable sleep trackers, brain age assessments, transcranial electrical stimulation (TES), acoustic stimulation, and glymphatic system modulation. Sleep trackers provide continuous monitoring of sleep patterns, while brain age estimation offers insights into brain health and early detection of accelerated aging. TES shows promise in improving mood, memory, and sleep. Acoustic stimulation during slow-wave sleep has been demonstrated to enhance memory consolidation. Additionally, optimizing the glymphatic system may facilitate brain waste clearance, crucial in preventing neurodegenerative diseases like Alzheimer's. However, significant challenges remain, including the need for rigorous longitudinal studies to validate these technologies' efficacy and safety.
Collapse
Affiliation(s)
- Ki-Hwan Ji
- Department of Neurology, Inje University Busan Paik Hospital, College of Medicine, Inje University, 75 Bokjiro, 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
|
21
|
Kalaria R, Englund E. Neuropathological features of cerebrovascular diseases. Pathology 2025; 57:207-219. [PMID: 39718486 DOI: 10.1016/j.pathol.2024.10.003] [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: 09/17/2024] [Revised: 10/14/2024] [Accepted: 10/22/2024] [Indexed: 12/25/2024]
Abstract
Optimal blood flow through a patent cerebral circulation is critical for supply of oxygen and nutrients for brain function. The integrity of vascular elements within arterial vessels of any calibre can be compromised by various disease processes. Pathological changes in the walls of veins and the venous system may also alter the dynamics of cerebral perfusion. The consequences of both systemic vascular and cerebrovascular diseases range from acute focal changes to irreversible chronic restructuring of the brain parenchyma. Cerebral infarcts of different sizes may instigate a cascade of programmed cell death mechanisms including autophagy and mitophagy and processes that range from necroptosis to ferroptosis. Recent advances also emphasise the role of the vascular inflammasome in the pathology of cerebral infarction. Here, we summarise current knowledge on frequencies, epidemiological features and the neuropathology of common cerebrovascular disorders among which cerebral small vessel diseases have become of particular interest. We also highlight the current spectrum of monogenic and polygenic genetic disorders affecting the intracranial vasculature. With the advent of DNA screening technologies, it is now realised that several cerebrovascular disorders exhibit strong genetic traits. Whilst several gene defects and their aberrant products are identified, the precise role or mechanisms of how they influence angiogenesis, vasculogenesis, vessel integrity or the extracellular matrix remain largely unclear. Despite such genetic advances, histopathological examination remains the gold standard for diagnosis and characterisation of most cerebrovascular disorders.
Collapse
Affiliation(s)
- Raj Kalaria
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, United Kingdom.
| | - Elisabet Englund
- Institutionen för kliniska vetenskaper, Lunds Universitet, Klinisk Patologi & Medicinsk Service, Region Skåne, Lund, Sweden
| |
Collapse
|
22
|
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
|
23
|
Chen Y, Xu J, Kong Y, Kang Y, Gong Z, Wang H, Huang Y, Zhan S, Yu Y, Lv X, Tan W. Cortical Morphology Alterations Mediate the Relationship Between Glymphatic System Function and the Severity of Asthenopia. Int J Biomed Imaging 2025; 2025:4464776. [PMID: 40041480 PMCID: PMC11879604 DOI: 10.1155/ijbi/4464776] [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: 03/15/2024] [Revised: 09/27/2024] [Accepted: 01/30/2025] [Indexed: 03/06/2025] Open
Abstract
Objectives: This study is aimed at assessing glymphatic function by diffusion tensor image analysis along the perivascular space (DTI-ALPS) and its associations with cortical morphological changes and severity of accommodative asthenopia (AA). Methods: We prospectively enrolled 50 patients with AA and 47 healthy controls (HCs). All participants underwent diffusion tensor imaging (DTI) and T1-weighted imaging and completed the asthenopia survey scale (ASS). Differences in brain morphometry and the analysis along the perivascular space (ALPS) index between the two groups were compared. The correlation and mediation analyses were conducted to explore the relationships between them. Results: Compared to HCs, patients with AA exhibited significantly increased sulcal depth in the left superior occipital gyrus (SOG.L) and increased cortical thickness in the left superior temporal gyrus (STG.L), left middle occipital gyrus (MOG.L), left postcentral gyrus (PoCG.L), and left precuneus (PCUN.L). Additionally, patients with AA had a significantly lower ALPS index than HCs. The sulcal depth of the SOG.L was significantly positively correlated with the ASS score in patients with AA, and a positive correlation was found between the cortical thickness of the MOG.L and ASS score. The ALPS index was negatively associated with the sulcal depth of the SOG.L and cortical thickness of the MOG.L. Mediation analysis revealed that the sulcal depth of SOG.L and cortical thickness of MOG.L partially mediated the impact of the DTI-ALPS index on the ASS score. Conclusion: Our findings suggested that patients with AA exhibit impaired glymphatic function, which may contribute to the severity of asthenopia through its influence on cortical morphological changes. The ALPS index is anticipated to become a potential imaging biomarker for patients with AA. Trial Registration: Chinese Registry of Clinical Trials: ChiCTR1900028306.
Collapse
Affiliation(s)
- Yilei Chen
- Department of Radiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jun Xu
- Department of Pharmacy, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yingnan Kong
- Department of Radiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yingjie Kang
- Department of Radiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhigang Gong
- Department of Radiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hui Wang
- Department of Radiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yanwen Huang
- Department of Radiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Songhua Zhan
- Department of Radiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ying Yu
- Department of Ophthalmology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaoli Lv
- Department of Ophthalmology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Wenli Tan
- Department of Radiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
24
|
Galgani A, Scotto M, Faraguna U, Giorgi FS. Fading Blue: Exploring the Causes of Locus Coeruleus Damage Across the Lifespan. Antioxidants (Basel) 2025; 14:255. [PMID: 40227216 PMCID: PMC11939699 DOI: 10.3390/antiox14030255] [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: 01/31/2025] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 04/15/2025] Open
Abstract
Locus Coeruleus (LC) is a brain nucleus that is involved in a variety of key functions (ranging from attention modulation to sleep-wake cycle regulation, to memory encoding); its proper function is necessary both during brain development and for brain integrity maintenance, and both at the microscale and macroscale level. Due to their specific intrinsic and extrinsic features, LC cells are considered particularly susceptible to damage concerning a variety of insults. This explains LC involvement in degenerative diseases not only in adults (in the context of neurodegenerative disease, mainly), but also in children (in relation to early hypoxic damage and Down's Syndrome, among others). In this narrative review, we dissect the potential mechanisms through which LC is affected in different diseases, with a special emphasis on the high rate of activity it is subjected to and the oxidative stress associated with it. Further research aimed at deepening our understanding of these mechanisms is needed to enable the development of potential strategies in the future that could slow down LC degeneration in subjects predisposed to specific brain disorders.
Collapse
Affiliation(s)
- Alessandro Galgani
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56125 Pisa, Italy
| | - Marco Scotto
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56125 Pisa, Italy
- Istituto Italiano di Tecnologia, 16163 Genova, Italy
| | - Ugo Faraguna
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56125 Pisa, Italy
- I.R.C.C.S. Stella Maris, Calambrone, 56128 Pisa, Italy
| | - Filippo S. Giorgi
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56125 Pisa, Italy
- I.R.C.C.S. Stella Maris, Calambrone, 56128 Pisa, Italy
| |
Collapse
|
25
|
Sarkkinen J, Yohannes DA, Kreivi N, Dürnsteiner P, Elsakova A, Huuhtanen J, Nowlan K, Kurdo G, Linden R, Saarela M, Tienari PJ, Kekäläinen E, Perdomo M, Laakso SM. Altered immune landscape of cervical lymph nodes reveals Epstein-Barr virus signature in multiple sclerosis. Sci Immunol 2025; 10:eadl3604. [PMID: 39982975 DOI: 10.1126/sciimmunol.adl3604] [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: 10/17/2023] [Revised: 07/17/2024] [Accepted: 01/29/2025] [Indexed: 02/23/2025]
Abstract
Multiple sclerosis (MS) is an autoimmune disease of the central nervous system, and Epstein-Barr virus (EBV) infection is a prerequisite for developing the disease. However, the pathogenic mechanisms that lead to MS remain to be determined. Here, we characterized the immune landscape of deep cervical lymph nodes (dcLNs) in newly diagnosed untreated patients with MS (pwMS) using fine-needle aspirations. By combining single-cell RNA sequencing and cellular indexing of transcriptomes and epitopes by sequencing, we observed increased memory B cells and reduced germinal center B cells with decreased clonality in pwMS. Double-negative memory B cells were increased in pwMS that transcriptionally resembled B cells with a lytic EBV infection. Moreover, EBV-targeting memory CD8 T cells were detected in a subset of pwMS. We also detected increased EBV DNA in dcLNs and elevated viral loads in patient saliva. These findings suggest that EBV-driven B cell dysregulation is a critical mechanism in MS pathogenesis.
Collapse
Affiliation(s)
- Joona Sarkkinen
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Dawit A Yohannes
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Nea Kreivi
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Pia Dürnsteiner
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Alexandra Elsakova
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Jani Huuhtanen
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- Hematology Research Unit Helsinki, Department of Hematology, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Department of Computer Science, Aalto University School of Science, Espoo, Finland
| | - Kirsten Nowlan
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Goran Kurdo
- Department of Radiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Riikka Linden
- Department of Radiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Mika Saarela
- Department of Neurology, Brain Center, Helsinki University Hospital, Helsinki, Finland
| | - Pentti J Tienari
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- Department of Neurology, Brain Center, Helsinki University Hospital, Helsinki, Finland
| | - Eliisa Kekäläinen
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Maria Perdomo
- Department of Virology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Sini M Laakso
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- Department of Neurology, Brain Center, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
26
|
Choi Y, Jung HJ, Jung HK, Jeong E, Kim S, Kim JY, Lee EJ, Lim YM, Kim H. In vivo imaging markers of glymphatic dysfunction in amyotrophic lateral sclerosis: Analysis of ALPS index and choroid plexus volume. J Neurol Sci 2025; 469:123393. [PMID: 39818026 DOI: 10.1016/j.jns.2025.123393] [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: 11/22/2024] [Revised: 12/24/2024] [Accepted: 01/08/2025] [Indexed: 01/18/2025]
Abstract
BACKGROUND The glymphatic system, essential for brain waste clearance, has been implicated in neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). Emerging imaging markers, such as the analysis along the perivascular space (ALPS) index and choroid plexus volume (CPV), may provide insights into glymphatic function, but their relevance to ALS remains unclear. OBJECTIVE To assess glymphatic dysfunction in ALS patients using the ALPS index and CPV. METHODS In this prospective single-center study, we analyzed 51 ALS patients and 51 age- and sex-matched healthy controls (HC). The ALPS index was calculated using diffusion tensor imaging, and 3D T1-weighted MRI was used for automated estimation of CPV and its fraction (CPV/total intracranial volume). Diagnostic performance was assessed using area under the receiver operating curve (AUC). Correlations between imaging markers and clinical parameters were also examined. RESULTS ALS patients had a significantly lower ALPS index (ALS: 1.45 ± 0.15; HC: 1.55 ± 0.16; p = 0.002) and higher CPV fraction (ALS: 0.12 ± 0.04 %; HC: 0.10 ± 0.02 %; p < 0.001). The ALPS index and CPV fraction had AUCs of 0.70 and 0.72, respectively. A significant inverse correlation was observed between the ALPS index and CPV fraction (r = -0.31, p = 0.002). Both markers correlated with aging but not with clinical disability or progression rate. CONCLUSION This study identifies glymphatic dysfunction in ALS, as evidenced by changes in the ALPS index and CPV. Larger studies are warranted to validate these findings and assess their potential as biomarkers for ALS.
Collapse
Affiliation(s)
- Yangsean Choi
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Hee-Jae Jung
- Department of Neurology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Ha-Kyung Jung
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Eunseon Jeong
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Shina Kim
- Department of Neurology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Ji-Yon Kim
- Department of Neurology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Eun-Jae Lee
- Department of Neurology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Young-Min Lim
- Department of Neurology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Hyunjin Kim
- Department of Neurology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea.
| |
Collapse
|
27
|
Kawalec-Rutkowska AM, Czaja J, Skuła M, Simka M. Blood Flow in the Internal Jugular Veins in the Lateral Decubitus Body Position in the Healthy People. J Clin Med 2025; 14:1211. [PMID: 40004742 PMCID: PMC11856538 DOI: 10.3390/jcm14041211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/04/2025] [Accepted: 02/09/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Some studies have suggested that the lateral decubitus position during sleep may protect the brain from neurodegenerative processes. Although the mechanisms of such possible protection are not known, an optimal venous outflow may be responsible. Venous outflow from the cranial cavity is dependent on the body's position. However, to date, flow in the internal jugular veins (IJVs) in the lateral position has not been studied quantitatively. Methods: Using ultrasonography, we measured the cross-sectional areas and flow volumes in the IJVs in a group of 25 healthy individuals aged 20-52 ± 12.1 years. These measurements were performed in the supine, upright, and lateral decubitus positions. Results: In the lateral decubitus position, we revealed a collapse of the IJV located higher, dilatation of the opposite vein, and a shift in flow from one vein to the opposite. In the right lateral position, the mean cross-sectional area and flow in the right IJV were 88.6 ± 71.1 mm2 and 74.3 ± 97.5 mL/min, in the left IJV: 37.2 ± 33.4 mm2 and 48.8 ± 82.8 mL/min. In the left lateral position, the right IJV was 38.4 ± 30.7 mm2 and 56.7 ± 56.1 mL/min, and the left IJV was 75.9 ± 51.9 mm2 and 99.7 ± 123.9 mL/min. However, there was also a high heterogeneity of the cross-sectional area changes, and in many participants, this pattern was not observed. Regarding flow volumes in the lateral body positions, in comparison with the supine position, the total outflow through both internal jugular veins was not significantly different. Conclusions: In terms of venous outflow, the lateral decubitus position did not differ significantly from the supine position. The working hypothesis of a potentially protective effect of this body position during sleep against neurodegeneration through improved venous outflow has not been proven, at least in healthy individuals.
Collapse
Affiliation(s)
| | | | | | - Marian Simka
- Institute of Medical Sciences, University of Opole, 45-060 Opole, Poland; (A.M.K.-R.); (J.C.); (M.S.)
| |
Collapse
|
28
|
Wostyn P, Goddaer P. Immersive gamma music as a tool for enhancing glymphatic clearance in astronauts while improving their mental well-being. LIFE SCIENCES IN SPACE RESEARCH 2025; 44:86-89. [PMID: 39864916 DOI: 10.1016/j.lssr.2024.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 10/24/2024] [Accepted: 10/27/2024] [Indexed: 01/28/2025]
Abstract
Spaceflight occurs under extreme environmental conditions that pose significant risks to the physical and mental health and well-being of astronauts. Certain factors, such as prolonged isolation, monotony, disrupted circadian rhythms, heavy workload, and weightlessness in space, can trigger psychological distress and may contribute to a variety of mental health problems, including mood and anxiety disturbances. Recent findings regarding spaceflight-associated alterations in cerebrospinal fluid spaces, demonstrating enlargement of the brain's perivascular spaces from preflight to postflight, at least suggest reduced glymphatic clearance in microgravity, and have raised concerns about long-term cognitive health in astronauts. Therefore, it is critical for future long-duration human exploration missions to identify, develop and validate all potentially effective long-term countermeasures capable of reducing the risk of perivascular space enlargement and impaired glymphatic transport in space mission crews. Furthermore, it is crucial to implement effective strategies that would allow crew members to maintain optimal psychological well-being during future long-duration space exploration. In the present paper, we propose "immersive gamma music" as an add-on countermeasure that in combination with existing countermeasures can optimize glymphatic clearance in astronauts while improving their mental well-being. If confirmed, this approach could enrich the practice of space medicine, and might become increasingly important, given the plans for future human missions, including a return to the Moon and manned missions to Mars.
Collapse
Affiliation(s)
- Peter Wostyn
- Department of Psychiatry, PC Sint-Amandus, Reigerlostraat 10, Beernem, Belgium.
| | - Piet Goddaer
- Studio Ozark Henry, Conterdijk 23, Wulpen, Belgium.
| |
Collapse
|
29
|
Chen W, Liang C, Peng S, Bao S, Xue F, Lian X, Liu Y, Wang G. Aquaporin-4 activation facilitates glymphatic system function and hematoma clearance post-intracerebral hemorrhage. Glia 2025; 73:368-380. [PMID: 39530196 DOI: 10.1002/glia.24639] [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: 08/06/2024] [Revised: 10/14/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
Efficient clearance of hematomas is crucial for improving clinical outcomes in patients with intracerebral hemorrhage (ICH). The glymphatic system, facilitated by aquaporin-4 (AQP4), plays a crucial role in cerebrospinal fluid (CSF) entry and metabolic waste clearance. This study examined the role of the glymphatic system in ICH pathology, with a focus on AQP4. Collagenase-induced ICH models were established, with AQP4 expression regulated through mifepristone as an agonist, TGN-020 as an inhibitor, and Aqp4 gene knockout. Fluorescence tracing and multimodal magnetic resonance imaging (MRI) were employed to observe glymphatic system functionality, hematoma, and edema volumes. Neurological deficit scoring was performed using the modified Garcia Scale. AQP4 expression was quantified using RT-qPCR and Western blotting, and cellular localization was explored using immunofluorescence. The brain tissue sections were examined for neuronal morphology, degenerative changes, and iron deposition. Three days post-ICH, the AQP4 agonist group showed increased AQP4 protein expression and perivascular polarization, decreased hemoglobin levels, and reduced iron deposition. Conversely, the inhibition group exhibited contrasting trends. AQP4 activation improved glymphatic system function, leading to a wider distribution, improved neurological function, and reduced hematoma. Pharmacological inhibition and genetic knockout of AQP4 have opposing effects. The glymphatic system, facilitated by AQP4, plays a crucial role in hematoma clearance following cerebral hemorrhage. Upregulation of AQP4 improves glymphatic system function, facilitates hematoma clearance, and promotes brain tissue recovery.
Collapse
Affiliation(s)
- Wenchao Chen
- Department of Neurology, Second Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Chuntian Liang
- Department of Neurology, Second Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Neurology, Sanya Central Hospital (Hainan Third People's Hospital), Hainan Medical University, Sanya, Hainan, China
| | - Shasha Peng
- Department of Pharmacology, School of Basical Medical Sciences, Shanxi Medical University, Taiyuan, China
- Department of Pharmacy, Sanya Central Hospital (Hainan Third People's Hospital), Hainan Medical University, Sanya, China
| | - Shuangjin Bao
- Department of Pathology and Pathophysiology, Basic Medical College, Shanxi Medical University, Taiyuan, China
- Department of Pathology, West China Fourth Hospital, Chengdu, China
| | - Fang Xue
- Department of Neurology, Second Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xia Lian
- Department of Neurology, Second Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yinghong Liu
- Department of Neurology, Second Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Gaiqing Wang
- Department of Neurology, Second Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Neurology, Sanya Central Hospital (Hainan Third People's Hospital), Hainan Medical University, Sanya, Hainan, China
| |
Collapse
|
30
|
Cheng Q, Liu Y, Yang Z, Zhang M, Liu T, Niu Y, Liu W, Huang L, Feng Y, Zhang X, Luo X, Ning Q, Chen T. Evaluation of Plasma Neurodegenerative Biomarkers for Diagnosing Minimal Hepatic Encephalopathy and Predicting Overt Hepatic Encephalopathy in Chinese Patients with Hepatic Cirrhosis. J Clin Transl Hepatol 2025; 13:35-46. [PMID: 39801785 PMCID: PMC11712093 DOI: 10.14218/jcth.2024.00413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 01/16/2025] Open
Abstract
Background and Aims The performance of neurodegenerative biomarkers-neurofilament light chain (NfL), glial fibrillary acidic protein (GFAP), tau, and ubiquitin carboxy-terminal hydrolase L1 (UCHL1)-in diagnosing minimal hepatic encephalopathy (MHE) has not been systematically evaluated, simultaneously, nor have their associations with the development of overt hepatic encephalopathy (OHE). This study aimed to evaluate the performance of plasma NfL, GFAP, tau, and UCHL1 in diagnosing MHE and predicting the development of OHE in Chinese patients with hepatic cirrhosis. Methods In this prospective study, 124 patients with hepatic cirrhosis were recruited. The Psychometric Hepatic Encephalopathy Score was used to diagnose MHE, and OHE development was observed during a 30-day follow-up period. Plasma levels of NfL, GFAP, tau, and UCHL1 were measured using the highly sensitive single-molecule array when MHE was diagnosed. Additionally, serum interleukin-6 (IL-6) levels and the model for end-stage liver disease (MELD) and MELD-Na scores were also measured. Results MHE was diagnosed in 57 (46.0%) patients. Patients with MHE had significantly higher plasma levels of NfL and GFAP (34.2 vs. 22.4 pg/mL and 173 vs. 97.6 pg/mL, respectively; both p < 0.001) and lower tau levels (8.4 vs. 11.6 pg/mL, p = 0.048) compared to those without MHE. Plasma NfL (odds ratios = 1.027, 95% confidence interval [CI]: 1.006-1.048; p = 0.013) and serum ammonia levels (odds ratios = 1.021, 95% CI: 1.006-1.036; p = 0.007) were independently associated with MHE occurrence. A combination of NfL, GFAP, tau, and UCHL1 was effective in diagnosing MHE in all cirrhotic patients (area under the receiver operating characteristic curve [hereinafter referred to as AUROC]: 0.748, 95% CI: 0.662-0.821), with an accuracy, sensitivity, and specificity of 71.0%, 71.9%, and 71.6%, respectively. In patients without previous OHE, the combination had an AUROC of 0.764 (95% CI: 0.673-0.840), with an accuracy, sensitivity, and specificity of 72.5%, 71.7%, and 73.0%, respectively. Furthermore, GFAP (hazard ratio (HR) = 1.003, 95% CI: 1.000-1.005; p = 0.044), IL-6 (HR = 1.003, 95% CI: 1.001-1.004; p < 0.001), and MELD score (HR = 1.139, 95% CI: 1.072-1.210; p < 0.001)-but not NfL, tau, and UCHL1-were identified as risk factors for 30-day OHE development. Conclusions The combination of plasma levels of NfL, GFAP, tau, and UCHL1 performs well in diagnosing MHE. Additionally, MELD score, IL-6, and GFAP appear to be significant predictors of OHE development in patients with hepatic cirrhosis.
Collapse
Affiliation(s)
- Qiuyu Cheng
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yunhui Liu
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhongyuan Yang
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Meng Zhang
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tingting Liu
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuxin Niu
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wei Liu
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lanyue Huang
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuzhao Feng
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaoyun Zhang
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaoping Luo
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qin Ning
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tao Chen
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
31
|
Wang L, Yang W, Ran Y, Song H, Yan X, Guo J. An Improved Method for Extracting Rat Cerebrospinal Fluid with Repeatable Large-Scale Collection. Vet Sci 2025; 12:58. [PMID: 39852933 PMCID: PMC11769370 DOI: 10.3390/vetsci12010058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/10/2025] [Accepted: 01/13/2025] [Indexed: 01/26/2025] Open
Abstract
The aim of this study was to explore an improved method for extracting rat cerebrospinal fluid (CSF), observing the impact on animal health under conditions of large-scale CSF collection and evaluating the feasibility of repeated collections. A total of 20 rats were anesthetized and fixed in a stereotactic frame. A 26G scalp needle, combined with a 1 mL syringe, was used to puncture the atlanto-occipital membrane and collect approximately 170 μL of CSF. CSF was collected twice within 14 days. During the study, animals were monitored daily for food intake, body weight, and hematological parameters, and at the end of the study, histopathological examination was performed. The health of the animals remained good, and repeated CSF collections were feasible. The success rate of the procedure was 100%, with blood contamination in the CSF decreasing from 70% in the first collection to 35% in the second. This technique is convenient, accurate, and suitable for widespread applications.
Collapse
Affiliation(s)
- Limei Wang
- Guangzhou Bay Area Institute of Biomedicine, Guangdong Lewwin Pharmaceutical Research Institute Co., Ltd., Guangdong Provincial Key Laboratory of Drug Non-Clinical Evaluation and Research, TCM Non-Clinic Evaluation Branch of National Engineering Research Center for Modernization of Traditional Chinese Medicine, Guangdong Engineering Research Center for Innovative Drug Evaluation and Research, Guangzhou 510990, China
| | - Wei Yang
- Guangzhou Bay Area Institute of Biomedicine, Guangdong Lewwin Pharmaceutical Research Institute Co., Ltd., Guangdong Provincial Key Laboratory of Drug Non-Clinical Evaluation and Research, TCM Non-Clinic Evaluation Branch of National Engineering Research Center for Modernization of Traditional Chinese Medicine, Guangdong Engineering Research Center for Innovative Drug Evaluation and Research, Guangzhou 510990, China
- Division of Life Science and State Key Lab of Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong 999077, China
| | - Yanhong Ran
- Guangzhou Bay Area Institute of Biomedicine, Guangdong Lewwin Pharmaceutical Research Institute Co., Ltd., Guangdong Provincial Key Laboratory of Drug Non-Clinical Evaluation and Research, TCM Non-Clinic Evaluation Branch of National Engineering Research Center for Modernization of Traditional Chinese Medicine, Guangdong Engineering Research Center for Innovative Drug Evaluation and Research, Guangzhou 510990, China
| | - Hui Song
- Guangzhou Bay Area Institute of Biomedicine, Guangdong Lewwin Pharmaceutical Research Institute Co., Ltd., Guangdong Provincial Key Laboratory of Drug Non-Clinical Evaluation and Research, TCM Non-Clinic Evaluation Branch of National Engineering Research Center for Modernization of Traditional Chinese Medicine, Guangdong Engineering Research Center for Innovative Drug Evaluation and Research, Guangzhou 510990, China
| | - Xinxin Yan
- Guangzhou Bay Area Institute of Biomedicine, Guangdong Lewwin Pharmaceutical Research Institute Co., Ltd., Guangdong Provincial Key Laboratory of Drug Non-Clinical Evaluation and Research, TCM Non-Clinic Evaluation Branch of National Engineering Research Center for Modernization of Traditional Chinese Medicine, Guangdong Engineering Research Center for Innovative Drug Evaluation and Research, Guangzhou 510990, China
| | - Jianmin Guo
- Guangzhou Bay Area Institute of Biomedicine, Guangdong Lewwin Pharmaceutical Research Institute Co., Ltd., Guangdong Provincial Key Laboratory of Drug Non-Clinical Evaluation and Research, TCM Non-Clinic Evaluation Branch of National Engineering Research Center for Modernization of Traditional Chinese Medicine, Guangdong Engineering Research Center for Innovative Drug Evaluation and Research, Guangzhou 510990, China
- Division of Life Science and State Key Lab of Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong 999077, China
| |
Collapse
|
32
|
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
|
33
|
Kumar A, Kumar R, Narayan RK, Nath B, Datusalia AK, Rastogi AK, Jha RK, Kumar P, Pareek V, Prasoon P, Faiq MA, Agrawal P, Prasad SN, Kumari C, Asghar A. Anatomical correlates for the newly discovered meningeal layer in the existing literature: A systematic review. Anat Rec (Hoboken) 2025; 308:191-210. [PMID: 38924700 DOI: 10.1002/ar.25524] [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: 02/23/2024] [Revised: 05/07/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024]
Abstract
The existence of a previously unrecognized subarachnoid lymphatic-like membrane (SLYM) was reported in a recent study. SLYM is described as an intermediate leptomeningeal layer between the arachnoid and pia mater in mouse and human brains, which divides the subarachnoid space (SAS) into two functional compartments. Being a macroscopic structure, having missed detection in previous studies is surprising. We systematically reviewed the published reports in animals and humans to explore whether prior descriptions of this meningeal layer were reported in some way. A comprehensive search was conducted in PubMed/Medline, EMBASE, Google Scholar, Science Direct, and Web of Science databases using combinations of MeSH terms and keywords with Boolean operators from inception until 31 December 2023. We found at least eight studies that provided structural evidence of an intermediate leptomeningeal layer in the brain or spinal cord. However, unequivocal descriptions for this layer all along the central nervous system were scarce. Obscure names like the epipial, intermediate meningeal, outer pial layers, or intermediate lamella were used to describe it. Its microscopic/ultrastructural details closely resemble the recently reported SLYM. We further examined the counterarguments in current literature that are skeptical of the existence of this layer. The potential physiological and clinical implications of this new meningeal layer are significant, underscoring the urgent need for further exploration of its structural and functional details.
Collapse
Affiliation(s)
- Ashutosh Kumar
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), Patna, India
| | - Rajesh Kumar
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), Patna, India
| | - Ravi K Narayan
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), Patna, India
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), Bhubaneswar, India
| | - Banshi Nath
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), Patna, India
| | - Ashok K Datusalia
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, India
| | - Ashok K Rastogi
- Department of Forensic Medicine and Toxicology, All India Institute of Medical Sciences (AIIMS), Patna, India
| | - Rakesh K Jha
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), Patna, India
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), Bhubaneswar, India
| | - Pankaj Kumar
- Regional Institute of Ophthalmology, Indira Gandhi Institute of Medical Sciences, Patna, India
| | - Vikas Pareek
- Haskins Laboratories, Yale Child Study Centre, Yale School of Medicine, University of Connecticut, New Haven, Connecticut, USA
| | - Pranav Prasoon
- Department of Anatomy and Cell Biology, George Washington University, Washington, DC, USA
| | - Muneeb A Faiq
- New York University (NYU) Langone Health Center, NYU Robert I Grossman School of Medicine, New York, New York, USA
| | - Prabhat Agrawal
- Spine Surgery Clinic, Department of Orthopedics, All India Institute of Medical Sciences (AIIMS), Patna, India
| | - Surya Nandan Prasad
- Department of Radiodiagnosis, All India Institute of Medical Sciences (AIIMS), Patna, India
| | - Chiman Kumari
- Department of Anatomy, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Adil Asghar
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), Patna, India
| |
Collapse
|
34
|
Chen Y, Zhang C, Feng Y. Medicinal plants for the management of post-COVID-19 fatigue: A literature review on the role and mechanisms. J Tradit Complement Med 2025; 15:15-23. [PMID: 39807273 PMCID: PMC11725095 DOI: 10.1016/j.jtcme.2024.05.006] [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: 01/22/2024] [Revised: 05/19/2024] [Accepted: 05/23/2024] [Indexed: 01/16/2025] Open
Abstract
Background COVID-19 infection has a lasting impact on human health, which is known as post-COVID-19 conditions. Fatigue is one of the most commonly reported post-COVID-19 conditions. Management of fatigue in the post-COVID-19 era is necessary and emerging. The use of medicinal plants may provide a strategy for the management of post-COVID-19 fatigue. Methods A literature search has been conducted by using PubMed, Embase and Cochrane library databases is performed for studies published up to March 2024. Keywords, such as "post-COVID-19 conditions, persistent COVID-19 symptoms, chronic COVID-19, long-term sequelae, fatigue, post-COVID-19 fatigue, herbal plants, medicinal herbs, traditional Chinese medicine, pharmacological mechanisms, pharmacological actions" are thoroughly searched in Englsih and Chinese. This study reviews the pathophysiology of post-COVID-19 fatigue and potential herbal plants for managing post-COVID-19 fatigue. Results and conclusion Representative medicinal plants that have been extensively investigated by previous studies are presented in the study. Three common mechanisms among the most extensively studied for post-COVID-19 fatigue, with each mechanism having medicinal plants as an example. The latest clinical studies concerning the management of post-COVID-19 fatigue using medicinal plants have also been summarized. The study shows the potential for improving post-COVID-19 fatigue by consuming medicinal plants.
Collapse
Affiliation(s)
- Yuanyuan Chen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, PR China
| | - Cheng Zhang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, PR China
| | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, PR China
| |
Collapse
|
35
|
Hong SJ, De Souza BJ, Penberthy KK, Hwang L, Procaccini DE, Kheir JN, Bembea MM. Plasma brain-related biomarkers and potential therapeutic targets in pediatric ECMO. Neurotherapeutics 2025; 22:e00521. [PMID: 39765416 PMCID: PMC11840354 DOI: 10.1016/j.neurot.2024.e00521] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 02/04/2025] Open
Abstract
Extracorporeal membrane oxygenation (ECMO) is a technique used to support severe cardiopulmonary failure. Its potential life-saving benefits are tempered by the significant risk for acute brain injury (ABI), from both primary pathophysiologic factors and ECMO-related complications through central nervous system cellular injury, blood-brain barrier dysfunction (BBB), systemic inflammation and neuroinflammation, and coagulopathy. Plasma biomarkers are an emerging tool used to stratify risk for and diagnose ABI, and prognosticate neurofunctional outcomes. Components of the neurovascular unit have been rational targets for this inquiry in ECMO. Central nervous system (CNS) neuronal and astroglial cellular-derived neuron-specific enolase (NSE), tau, glial fibrillary acidic protein (GFAP) and S100β elevations have been detected in ABI and are associated with poorer outcomes. Evidence of BBB breakdown through peripheral blood detection of CNS cellular components NSE, GFAP, and S100β, as well as evidence of elevated BBB components vWF and PDGFRβ are associated with higher mortality and worse neurofunctional outcomes. Higher concentrations of pro-inflammatory cytokines (IL-1β, IL-6, IFN-γ, TNF-α) are associated with abnormal neuroimaging, and proteomic expression panels reveal different coagulation and inflammatory responses. Abnormal coagulation profiles are common in ECMO with ongoing studies attempting to describe specific abnormalities either being causal or associated with neurologic outcomes; vWF has shown some promise. Understanding these mechanisms of injury through biomarker analysis supports potential neuroprotective strategies such as individualized blood pressure targets, judicious hypercarbia and hypoxemia correction, and immunomodulation (inhaled hydrogen and N-acetylcysteine). Further research continues to elucidate the role of biomarkers as predictors, prognosticators, and therapeutic targets.
Collapse
Affiliation(s)
- Sue J Hong
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bradley J De Souza
- Department of Critical Care Medicine, Children's Hospital Los Angeles and Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Kristen K Penberthy
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lisa Hwang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - John N Kheir
- Department of Cardiology, Boston Children's Hospital, and the Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Melania M Bembea
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
36
|
Zhu Y, Ma J, Li Y, Gu M, Feng X, Shao Y, Tan L, Lou HF, Sun L, Liu Y, Zeng LH, Qiu Z, Li XM, Duan S, Yu YQ. Adenosine-Dependent Arousal Induced by Astrocytes in a Brainstem Circuit. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407706. [PMID: 39494592 DOI: 10.1002/advs.202407706] [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] [Received: 07/09/2024] [Revised: 09/20/2024] [Indexed: 11/05/2024]
Abstract
Astrocytes play a crucial role in regulating sleep-wake behavior. However, how astrocytes govern a specific sleep-arousal circuit remains unknown. Here, the authors show that parafacial zone (PZ) astrocytes responded to sleep-wake cycles with state-differential Ca2+ activity, peaking during transitions from sleep to wakefulness. Using chemogenetic and optogenetic approaches, they find that activating PZ astrocytes elicited and sustained wakefulness by prolonging arousal episodes while impeding transitions from wakefulness to non-rapid eye movement (NREM) sleep. Activation of PZ astrocytes specially induced the elevation of extracellular adenosine through the ATP hydrolysis pathway but not equilibrative nucleoside transporter (ENT) mediated transportation. Strikingly, the rise in adenosine levels induced arousal by activating A1 receptors, suggesting a distinct role for adenosine in the PZ beyond its conventional sleep homeostasis modulation observed in the basal forebrain (BF) and cortex. Moreover, at the circuit level, PZ astrocyte activation induced arousal by suppressing the GABA release from the PZGABA neurons, which promote NREM sleep and project to the parabrachial nucleus (PB). Thus, their study unveils a distinctive arousal-promoting effect of astrocytes within the PZ through extracellular adenosine and elucidates the underlying mechanism at the neural circuit level.
Collapse
Affiliation(s)
- Yuwei Zhu
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, 311121, China
| | - Jiale Ma
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Nanhu Brain-computer Interface Institute, Hangzhou, 311100, China
| | - Yulan Li
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Mengyang Gu
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Xiang Feng
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, 311121, China
| | - Yujin Shao
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Lei Tan
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Hui-Fang Lou
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Li Sun
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, 311121, China
| | - Yijun Liu
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, 311121, China
| | - Ling-Hui Zeng
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, China
| | - Zilong Qiu
- Department of Neurology, Songjiang Hospital, Songjiang Research Institute, MOE-Shanghai Key Laboratory for Children's Environmental Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiao-Ming Li
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, 311121, China
- Nanhu Brain-computer Interface Institute, Hangzhou, 311100, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, 310058, China
| | - Shumin Duan
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, 311121, China
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, China
- Department of Neurology, Songjiang Hospital, Songjiang Research Institute, MOE-Shanghai Key Laboratory for Children's Environmental Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, 310058, China
| | - Yan-Qin Yu
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, 311121, China
- Nanhu Brain-computer Interface Institute, Hangzhou, 311100, China
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, 310058, China
| |
Collapse
|
37
|
Siri S, Burchett A, Datta M. Simulating the impact of tumor mechanical forces on glymphatic networks in the brain parenchyma. Biomech Model Mechanobiol 2024; 23:2229-2241. [PMID: 39298038 PMCID: PMC11554883 DOI: 10.1007/s10237-024-01890-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: 07/23/2024] [Accepted: 09/12/2024] [Indexed: 09/25/2024]
Abstract
The brain glymphatic system is currently being explored in the context of many neurological disorders and diseases, including traumatic brain injury, Alzheimer's disease, and ischemic stroke. However, little is known about the impact of brain tumors on glymphatic function. Mechanical forces generated during tumor development and growth may be responsible for compromised glymphatic transport pathways, reducing waste clearance and cerebrospinal fluid (CSF) transport in the brain parenchyma. One such force is solid stress, i.e., growth-induced forces from cell hyperproliferation and excess matrix deposition. Because there are no prior studies assessing the impact of tumor-derived solid stress on glymphatic system structure and performance in the brain parenchyma, this study serves to fill an important gap in the field. We adapted a previously developed Electrical Analog Model using MATLAB Simulink for glymphatic transport coupled with Finite Element Analysis for tumor mechanical stresses and strains in COMSOL. This allowed simulation of the impact of tumor mechanical force generation on fluid transport within brain parenchymal glymphatic units-which include perivascular spaces, astrocytic networks, interstitial spaces, and capillary basement membranes. We conducted a parametric analysis to compare the contributions of tumor size, tumor proximity, and ratio of glymphatic subunits to the stress and strain experienced by the glymphatic unit and corresponding reduction in flow rate of CSF. Mechanical stresses intensify with proximity to the tumor and increasing tumor size, highlighting the vulnerability of nearby glymphatic units to tumor-derived forces. Our stress and strain profiles reveal compressive deformation of these surrounding glymphatics and demonstrate that varying the relative contributions of astrocytes vs. interstitial spaces impact the resulting glymphatic structure significantly under tumor mechanical forces. Increased tumor size and proximity caused increased stress and strain across all glymphatic subunits, as does decreased astrocyte composition. Indeed, our model reveals an inverse correlation between extent of astrocyte contribution to the composition of the glymphatic unit and the resulting mechanical stress. This increased mechanical strain across the glymphatic unit decreases the venous efflux rate of CSF, dependent on the degree of strain and the specific glymphatic subunit of interest. For example, a 20% mechanical strain on capillary basement membranes does not significantly decrease venous efflux (2% decrease in flow rates), while the same magnitude of strain on astrocyte networks and interstitial spaces decreases efflux flow rates by 7% and 22%, respectively. Our simulations reveal that solid stress from growing brain tumors directly reduces glymphatic fluid transport, independently from biochemical effects from cancer cells. Understanding these pathophysiological implications is crucial for developing targeted interventions aimed at restoring effective waste clearance mechanisms in the brain. This study opens potential avenues for future experimental research in brain tumor-related glymphatic dysfunction.
Collapse
Affiliation(s)
- Saeed Siri
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Alice Burchett
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Meenal Datta
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA.
| |
Collapse
|
38
|
Sibilia F, Sheikh-Bahaei N, Mack WJ, Barisano G, Choupan J. Neuroinflammation modifies the relationship between stress and perivascular spaces in an elderly population with different levels of cognitive impairment. Front Cell Neurosci 2024; 18:1480405. [PMID: 39610697 PMCID: PMC11603360 DOI: 10.3389/fncel.2024.1480405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 10/30/2024] [Indexed: 11/30/2024] Open
Abstract
Background Perivascular spaces (PVS) are fluid-filled spaces surrounding the brain parenchymal vasculature. Literature suggests that PVS may play a significant role in aging and neurological disorders, including Alzheimer's disease (AD). The aim of this study is to investigate whether the relationship between MRI-visible PVS and stress is influenced by neuroinflammation in an elderly population with different levels of cognitive impairment. Methods Using brain MRI scans acquired at 1.5 T, PVS were quantified in a cohort of 461 individuals, consisting of cognitively healthy controls (n = 48), people with mild cognitive impairment (MCI, n = 322) and Alzheimer's disease (AD, n = 91). PVS volume fraction was calculated in the basal ganglia and centrum semiovale using a semi-automated segmentation approach. Stress was quantified with levels of salivary cortisol. Inflammatory biomarkers measured from plasma included cytokines, matrix metalloproteinases and C-reactive protein. General linear models were used to test the relationship between PVS and cortisol, when interacting with inflammatory markers. This was done on the whole cohort and within each clinical cognitive group. Results In the centrum semiovale, higher inflammation levels reduced the relationship of cortisol with PVS. In basal ganglia, higher levels of C-reactive protein reduced the negative relationship of cortisol with PVS. All analyses were accounted for age, sex, body mass index (BMI) and total hippocampal volume. There was a significant interaction effect between cortisol and C-reactive protein on PVS volume fraction in the MCI group. Discussion These findings suggest an influence of neuroinflammation on the PVS structure in Alzheimer's disease spectrum, and offer insight for better understanding physiological processes of cognitive impairment onset.
Collapse
Affiliation(s)
- Francesca Sibilia
- Laboratory of Neuro Imaging, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Nasim Sheikh-Bahaei
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Wendy J. Mack
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Giuseppe Barisano
- Department of Neurosurgery, Stanford University, Stanford, CA, United States
| | - Jeiran Choupan
- Laboratory of Neuro Imaging, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- NeuroScope Inc,Scarsdale, NY, United States
| |
Collapse
|
39
|
Gao M, Liu Z, Zang H, Wu X, Yan Y, Lin H, Yuan J, Liu T, Zhou Y, Liu J. A Histopathologic Correlation Study Evaluating Glymphatic Function in Brain Tumors by Multiparametric MRI. Clin Cancer Res 2024; 30:4876-4886. [PMID: 38848042 PMCID: PMC11528195 DOI: 10.1158/1078-0432.ccr-24-0150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 03/15/2024] [Accepted: 06/05/2024] [Indexed: 11/02/2024]
Abstract
PURPOSE This study aimed to elucidate the impact of brain tumors on cerebral edema and glymphatic drainage by leveraging advanced MRI techniques to explore the relationships among tumor characteristics, glymphatic function, and aquaporin-4 (AQP4) expression levels. EXPERIMENTAL DESIGN In a prospective cohort from March 2022 to April 2023, patients with glioblastoma, brain metastases, and aggressive meningiomas, alongside age- and sex-matched healthy controls, underwent 3.0T MRI, including diffusion tensor imaging analysis along the perivascular space (DTI-ALPS) index and multiparametric MRI for quantitative brain mapping. Tumor and peritumor tissues were analyzed for AQP4 expression levels via immunofluorescence. Correlations among MRI parameters, glymphatic function (DTI-ALPS index), and AQP4 expression levels were statistically assessed. RESULTS Among 84 patients (mean age: 55 ± 12 years; 38 males) and 59 controls (mean age: 54 ± 8 years; 23 males), patients with brain tumor exhibited significantly reduced glymphatic function (DTI-ALPS index: 2.315 vs. 2.879; P = 0.001) and increased cerebrospinal fluid volume (201.376 cm³ vs. 115.957 cm³; P = 0.001). A negative correlation was observed between tumor volume and the DTI-ALPS index (r: -0.715, P < 0.001), whereas AQP4 expression levels correlated positively with peritumoral brain edema volume (r: 0.989, P < 0.001) and negatively with proton density in peritumoral brain edema areas (ρ: -0.506, P < 0.001). CONCLUSIONS Our findings highlight the interplay among tumor-induced compression, glymphatic dysfunction, and altered fluid dynamics, demonstrating the utility of DTI-ALPS and multiparametric MRI in understanding the pathophysiology of tumor-related cerebral edema. These insights provide a radiological foundation for further neuro-oncological investigations into the glymphatic system. See related commentary by Surov and Borggrefe, p. 4813.
Collapse
Affiliation(s)
- Min Gao
- Department of Radiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zhengliang Liu
- School of Computing, The University of Georgia, Athens, Georgia
| | - Hongjing Zang
- Department of Pathology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xiong Wu
- Department of Radiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yizhong Yan
- National Engineering Research Center of Human Stem Cell, Changsha, China
| | - Hai Lin
- Central Research Institute, United Imaging Healthcare, Shanghai, China
| | - Jianmin Yuan
- Central Research Institute, United Imaging Healthcare, Shanghai, China
| | - Tianming Liu
- School of Computing, The University of Georgia, Athens, Georgia
| | - Yu Zhou
- Department of Neurosurgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jun Liu
- Department of Radiology, The Second Xiangya Hospital of Central South University, Changsha, China
- Department of Radiology Quality Control Center, Hunan, China
| |
Collapse
|
40
|
Lin S, Guo M, Liang Q, Lin X, Chen S, Li Y, Chen P, Qiu Y. Evaluation of Glymphatic System Development in Neonatal Brain via Diffusion Analysis along the Perivascular Space Index. Ann Neurol 2024; 96:970-980. [PMID: 39096048 DOI: 10.1002/ana.27047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 06/22/2024] [Accepted: 07/15/2024] [Indexed: 08/04/2024]
Abstract
OBJECTIVE Glymphatic system is a recently discovered macroscopic waste clearance system associated with numerous neurological diseases. However, little is known about glymphatic system development in neonates. We sought to evaluate diffusion along the perivascular space (ALPS) index, a proxy for glymphatic system function, in neonates and investigate its potential associations with maturation, sex, and preterm birth. METHODS Diffusion magnetic resonance imaging (MRI) data in 418 neonates, including 92 preterm neonates (57 males) and 326 term neonates (175 males), from the Developing Human Connectome Project were used for evaluating ALPS index. Linear regression modeling was performed to assess group differences in the ALPS index according to preterm birth and sex. Pearson's and partial correlation analysis were performed to assess the association between the ALPS index and gestational age (GA) as well as postmenstrual age (PMA) at MRI. Moderation analysis was performed to assess the moderation effect of preterm birth on the relationship between the ALPS index and PMA. RESULTS Compared to term neonates, preterm neonates exhibited lower ALPS indices (p < 0.001). The ALPS index positively correlated with PMA (p = 0.004) and GA (p < 0.001). Preterm birth (p = 0.013) had a significant moderation effect on the relationship between the ALPS index and PMA. Sex had no significant direct effect (p = 0.639) or moderation effect (p = 0.333) on ALPS index. INTERPRETATION Glymphatic system development is a dynamic process in neonates, which can be moderated by preterm birth, the ALPS index could serve as a sensitive biomarker for monitoring this process. ANN NEUROL 2024;96:970-980.
Collapse
Affiliation(s)
- Shiwei Lin
- Department of Radiology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Meifen Guo
- Department of Radiology, the Affiliated TCM Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qunjun Liang
- Department of Radiology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Xiaoshan Lin
- Department of Radiology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Shengli Chen
- Department of Radiology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Ying Li
- Department of Radiology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Peiqi Chen
- Department of Radiology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Yingwei Qiu
- Department of Radiology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
41
|
Zhang J, Zhang X, Zhao J, Wu J. The Effects of Vitamin D on Movement and Cognitive Function in Senile Mice After Sevoflurane Anaesthesia. Exp Aging Res 2024; 50:817-831. [PMID: 37990880 DOI: 10.1080/0361073x.2023.2282350] [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: 05/17/2023] [Accepted: 11/08/2023] [Indexed: 11/23/2023]
Abstract
BACKGROUND Vitamin D (VD) is a neuroactive steroid involved in many brain functions, such as neurotrophic, neuroimmune control and neurotransmission, which affects the growth and function of the brain. The purpose of this study is to explore the effect of VD on motor and cognitive function of aged mice after sevoflurane anesthesia. METHOD We established sevoflurane anesthesia model and VD(-) and VD(+) mice model. The VD concentration of mice in each group was determined by enzyme-linked immunosorbent assay (ELISA). An open-field test was used to evaluate the mice's capacity for movement and exploration. A Y-maze test was used to gauge the mice's short-term memory. The primary purpose of the water-maze experiment was to examine mice's long-term spatial memory. RESULTS The ELISA results showed that the model was successfully constructed. In the open-field test, VD increased the exercise distance of mice (P < .05). In the Y-maze experiment, VD improved short-term memory impairment in mice (P < .05). In the water-maze test, VD increased the activity time and platform crossing number of mice in the target quadrant. (P < .05). CONCLUSION Sevoflurane anesthesia caused cognitive dysfunction in aged mice, including reduced learning ability, memory loss, lower motor and exploratory abilities and depression, and VD deficiency aggravated these impairments. By supplementing with VD, learning ability and long-term memory were enhanced, motor and exploratory abilities were improved, and depression levels were reduced. Anxiety was also improved.
Collapse
Affiliation(s)
- Jialei Zhang
- Department of Anesthesiology, Changzhi People's Hospital Affiliated with Shanxi Medical University, Changzhi, China
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaoling Zhang
- Department of Oncology, Changzhi People's Hospital Affiliated with Shanxi Medical University, Changzhi, China
| | - Jun Zhao
- Department of Oncology, Changzhi People's Hospital Affiliated with Shanxi Medical University, Changzhi, China
| | - Jie Wu
- Department of pain treatment, Changzhi People's Hospital Affiliated with Shanxi Medical University, Changzhi, China
| |
Collapse
|
42
|
Tastan B, Heneka MT. The impact of neuroinflammation on neuronal integrity. Immunol Rev 2024; 327:8-32. [PMID: 39470038 DOI: 10.1111/imr.13419] [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: 10/30/2024]
Abstract
Neuroinflammation, characterized by a complex interplay among innate and adaptive immune responses within the central nervous system (CNS), is crucial in responding to infections, injuries, and disease pathologies. However, the dysregulation of the neuroinflammatory response could significantly affect neurons in terms of function and structure, leading to profound health implications. Although tremendous progress has been made in understanding the relationship between neuroinflammatory processes and alterations in neuronal integrity, the specific implications concerning both structure and function have not been extensively covered, with the exception of perspectives on glial activation and neurodegeneration. Thus, this review aims to provide a comprehensive overview of the multifaceted interactions among neurons and key inflammatory players, exploring mechanisms through which inflammation influences neuronal functionality and structural integrity in the CNS. Further, it will discuss how these inflammatory mechanisms lead to impairment in neuronal functions and architecture and highlight the consequences caused by dysregulated neuronal functions, such as cognitive dysfunction and mood disorders. By integrating insights from recent research findings, this review will enhance our understanding of the neuroinflammatory landscape and set the stage for future interventions that could transform current approaches to preserve neuronal integrity and function in CNS-related inflammatory conditions.
Collapse
Affiliation(s)
- Bora Tastan
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Michael T Heneka
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, North Worcester, Massachusetts, USA
| |
Collapse
|
43
|
Liu H, Meng L, Wang J, Qin C, Feng R, Chen Y, Chen P, Zhu Q, Ma M, Teng J, Ding X. Enlarged perivascular spaces in alcohol-related brain damage induced by dyslipidemia. J Cereb Blood Flow Metab 2024; 44:1867-1880. [PMID: 38700501 PMCID: PMC11494831 DOI: 10.1177/0271678x241251570] [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: 10/24/2023] [Revised: 03/28/2024] [Accepted: 04/02/2024] [Indexed: 05/05/2024]
Abstract
Perivascular spaces (PVSs) as the anatomical basis of the glymphatic system, are increasingly recognized as potential imaging biomarkers of neurological conditions. However, it is not clear whether enlarged PVSs are associated with alcohol-related brain damage (ARBD). We aimed to investigate the effect of long-term alcohol exposure on dyslipidemia and the glymphatic system in ARBD. We found that patients with ARBD exhibited significantly enlargement of PVSs in the frontal cortex and basal ganglia, as well as a notable increased levels of total cholesterol (TC) and triglycerides (TG). The anatomical changes of the glymphatic drainage system mentioned above were positively associated with TC and TG. To further explore whether enlarged PVSs affects the function of the glymphatic system in ARBD, we constructed long alcohol exposure and high fat diet mice models. The mouse model of long alcohol exposure exhibited increased levels of TC and TG, enlarged PVSs, the loss of aquaporin-4 polarity caused by reactive astrocytes and impaired glymphatic drainage function which ultimately caused cognitive deficits, in a similar way as high fat diet leading to impairment in glymphatic drainage. Our study highlights the contribution of dyslipidemia due to long-term alcohol abuse in the impairment of the glymphatic drainage system.
Collapse
Affiliation(s)
- Han Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
- Henan Key Laboratory of Chronic Disease Prevention and Therapy & Intelligent Health Management, Henan 450052, China
| | - Lin Meng
- Department of Neurology, Zhengzhou Central Hospital, Zhengzhou, Henan 450000, China
| | - Jiuqi Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
- Henan Key Laboratory of Chronic Disease Prevention and Therapy & Intelligent Health Management, Henan 450052, China
| | - Chi Qin
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
- Henan Key Laboratory of Chronic Disease Prevention and Therapy & Intelligent Health Management, Henan 450052, China
| | - Renyi Feng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
- Henan Key Laboratory of Chronic Disease Prevention and Therapy & Intelligent Health Management, Henan 450052, China
| | - Yongkang Chen
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
- Henan Key Laboratory of Chronic Disease Prevention and Therapy & Intelligent Health Management, Henan 450052, China
| | - Pei Chen
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
- Henan Key Laboratory of Chronic Disease Prevention and Therapy & Intelligent Health Management, Henan 450052, China
| | - Qingyong Zhu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
- Henan Key Laboratory of Chronic Disease Prevention and Therapy & Intelligent Health Management, Henan 450052, China
| | - Mingming Ma
- Department of Neurology, Henan Provincial People’s Hospital, Zhengzhou, Henan 450000, China
| | - Junfang Teng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
- Henan Key Laboratory of Chronic Disease Prevention and Therapy & Intelligent Health Management, Henan 450052, China
| | - Xuebing Ding
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
- Henan Key Laboratory of Chronic Disease Prevention and Therapy & Intelligent Health Management, Henan 450052, China
| |
Collapse
|
44
|
Mineiro R, Rodrigues Cardoso M, Catarina Duarte A, Santos C, Cipolla-Neto J, Gaspar do Amaral F, Costa D, Quintela T. Melatonin and brain barriers: The protection conferred by melatonin to the blood-brain barrier and blood-cerebrospinal fluid barrier. Front Neuroendocrinol 2024; 75:101158. [PMID: 39395545 DOI: 10.1016/j.yfrne.2024.101158] [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: 05/02/2024] [Revised: 07/29/2024] [Accepted: 10/05/2024] [Indexed: 10/14/2024]
Abstract
The blood-brain barrier and the blood-cerebrospinal fluid barrier separate the blood from brain tissue and cerebrospinal fluid. These brain barriers are important to maintain homeostasis and complex functions by protecting the brain from xenobiotics and harmful endogenous compounds. The disruption of brain barriers is a characteristic of neurologic diseases. Melatonin is a lipophilic hormone that is mainly produced by the pineal gland. The blood-brain barrier and the blood-cerebrospinal fluid barriers are melatonin-binding sites. Among the several melatonin actions, the most characteristic one is the regulation of sleep-wake cycles, melatonin has anti-inflammatory and antioxidant properties. Since brain barriers disruption can arise from inflammation and oxidative stress, knowing the influence of melatonin on the integrity of brain barriers is extremely important. Therefore, the objective of this review is to gather and discuss the available literature about the regulation of brain barriers by melatonin.
Collapse
Affiliation(s)
- Rafael Mineiro
- CICS-UBI-Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Maria Rodrigues Cardoso
- CICS-UBI-Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Ana Catarina Duarte
- CICS-UBI-Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Cecília Santos
- CICS-UBI-Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Jose Cipolla-Neto
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Diana Costa
- CICS-UBI-Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Telma Quintela
- CICS-UBI-Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal; Instituto Politécnico da Guarda, 6300-559 Guarda, Portugal.
| |
Collapse
|
45
|
Zhao Y, Huang Y, Cao Y, Yang J. Astrocyte-Mediated Neuroinflammation in Neurological Conditions. Biomolecules 2024; 14:1204. [PMID: 39456137 PMCID: PMC11505625 DOI: 10.3390/biom14101204] [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: 08/09/2024] [Revised: 09/15/2024] [Accepted: 09/18/2024] [Indexed: 10/28/2024] Open
Abstract
Astrocytes are one of the key glial types of the central nervous system (CNS), accounting for over 20% of total glial cells in the brain. Extensive evidence has established their indispensable functions in the maintenance of CNS homeostasis, as well as their broad involvement in neurological conditions. In particular, astrocytes can participate in various neuroinflammatory processes, e.g., releasing a repertoire of cytokines and chemokines or specific neurotrophic factors, which result in both beneficial and detrimental effects. It has become increasingly clear that such astrocyte-mediated neuroinflammation, together with its complex crosstalk with other glial cells or immune cells, designates neuronal survival and the functional integrity of neurocircuits, thus critically contributing to disease onset and progression. In this review, we focus on the current knowledge of the neuroinflammatory responses of astrocytes, summarizing their common features in neurological conditions. Moreover, we highlight several vital questions for future research that promise novel insights into diagnostic or therapeutic strategies against those debilitating CNS diseases.
Collapse
Affiliation(s)
- Yanxiang Zhao
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
- The Affiliated High School, Peking University, Beijing 100080, China
| | - Yingying Huang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
| | - Ying Cao
- Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Jing Yang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
- Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
- Peking University Third Hospital Cancer Center, Beijing 100191, China
| |
Collapse
|
46
|
Garcia KJ, Brolly G, Ng D, Bederson M, Martinez P, Whiting MD. Lifetime history of head injury is associated with reduced perivascular space number in acute mild traumatic brain injury. Brain Commun 2024; 6:fcae314. [PMID: 39329080 PMCID: PMC11426355 DOI: 10.1093/braincomms/fcae314] [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: 05/14/2024] [Revised: 07/08/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024] Open
Abstract
Traumatic brain injury impairs function of the glymphatic system, a perivascular network involved in waste clearance. Enlarged perivascular spaces visible on MRI are an emerging biomarker of glymphatic function. This study characterized enlarged perivascular spaces in acute head injury with 7 T MRI. Healthy controls (n = 8) and patients (n = 11) with mild traumatic brain injury underwent MRI within 7 days of injury and were evaluated for lifetime history of head injury, neurobehavioral symptoms and sleep disturbances. MRI-visible perivascular spaces were quantified and assessed according to published criteria. The number of enlarged perivascular spaces was significantly higher in traumatic brain injury patients than controls (P = 0.015). Among healthy controls, 6/8 scored 'none' or 'mild' on the perivascular space rating scale, while 10/11 patients scored 'moderate', 'frequent' or 'severe'. There was an inverse relationship between perivascular space number and number of lifetime head injuries. Patients with more prior head injuries exhibited fewer enlarged perivascular spaces (P = 0.014). These results indicate that mild head injury results in acute alterations in perivascular space number, and this effect is mediated by previous head injury history. Enlarged perivascular spaces may reflect a glymphatic response that is diminished after multiple head injuries, although this will require further study.
Collapse
Affiliation(s)
- Kiersten J Garcia
- Stephens Family Clinical Research Institute, Carle Health, Urbana, IL 61801, USA
- Carle Illinois Advanced Imaging Center, Carle Health, Urbana, IL 61801, USA
| | - Grace Brolly
- Carle Illinois College of Medicine, Urbana, IL 61801, USA
| | - Daniel Ng
- Carle Illinois College of Medicine, Urbana, IL 61801, USA
| | - Maria Bederson
- Carle Illinois College of Medicine, Urbana, IL 61801, USA
| | - Pedro Martinez
- Department of Neuroscience, Wartburg College, Waverly, IA 50677, USA
| | - Mark D Whiting
- Stephens Family Clinical Research Institute, Carle Health, Urbana, IL 61801, USA
- Carle Illinois Advanced Imaging Center, Carle Health, Urbana, IL 61801, USA
- Carle Illinois College of Medicine, Urbana, IL 61801, USA
| |
Collapse
|
47
|
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
|
48
|
Guo Y, Wu L, Liu J, Liu J, Sun Z. Correlation between glymphatic dysfunction and cranial defect in severe traumatic brain injury: a retrospective case-control study based on a diffusion tensor image analysis along the perivascular space (DTI-ALPS) investigation. Quant Imaging Med Surg 2024; 14:6756-6766. [PMID: 39281142 PMCID: PMC11400707 DOI: 10.21037/qims-24-348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 07/17/2024] [Indexed: 09/18/2024]
Abstract
Background To date, limited research has been conducted on the functionality of the glymphatic system during the recovery phase of severe traumatic brain injury (sTBI). This study aimed to use a diffusion tensor image analysis along the perivascular space (DTI-ALPS) to evaluate glymphatic system function in patients recovering from sTBI who underwent unilateral decompressive craniectomy, and to examine the correlation between the ALPS index and the size of the cranial defect. We hypothesized that assessments would reveal ongoing impairments in glymphatic system function among sTBI patients during the recovery phase. Methods A total of 23 patients with a history of sTBI who had previously undergone unilateral decompressive craniectomy at Xiangya Hospital of Central South University from January 2020 to December 2020 were enrolled in the study, along with 33 healthy control (HC) subjects. All the subjects underwent magnetic resonance imaging (MRI) with DTI scans, and the ALPS index was subsequently calculated to assess glymphatic system functionality. Additionally, the circumference and sectional area of the cranial defect were measured for each patient. An analysis of variance (ANOVA) was used to compare the ALPS index values between the sTBI patients and HC subjects, while a Pearson correlation analysis was used to examine the correlation between the ALPS index and cranial defect characteristics. Results The ALPS index values of both the craniectomy side (t=-9.08, P<0.001) and non-craniectomy side (t=-5.06, P<0.001) of the sTBI group were significantly lower than those of the HC group. However, no statistically significant differences were observed between the ALPS index values of the craniectomy and non-craniectomy sides. Additionally, no significant differences were observed in the ALPS index values of both the craniectomy and non-craniectomy sides among the early, intermediate, and late recovery phases. In the sTBI patients, a moderately strong negative correlation was found between the circumference of the cranial defect and the ALPS index of the craniectomy side (r=-0.62, P=0.002), and a moderately negative correlation was found between the sectional area of the cranial defect and the ALPS index of the craniectomy side (r=-0.56, P=0.005). Conclusions The non-invasive DTI-ALPS technique revealed significantly reduced ALPS index values during the recovery phase of sTBI, indicating persistent impairment in glymphatic system function. A significant negative correlation was found between the ALPS index value of the craniectomy side and the size of the cranial defect. These findings suggest that the ALPS index may serve as a valuable prognostic factor in the recovery phase of sTBI.
Collapse
Affiliation(s)
- Yong Guo
- Department of Neurosurgery, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, China
| | - Lin Wu
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jiacheng Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Jinfang Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Zhongyi Sun
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
49
|
Deike K, Decker A, Scheyhing P, Harten J, Zimmermann N, Paech D, Peters O, Freiesleben SD, Schneider LS, Preis L, Priller J, Spruth E, Altenstein S, Lohse A, Fliessbach K, Kimmich O, Wiltfang J, Bartels C, Hansen N, Jessen F, Rostamzadeh A, Düzel E, Glanz W, Incesoy EI, Butryn M, Buerger K, Janowitz D, Ewers M, Perneczky R, Rauchmann BS, Teipel S, Kilimann I, Goerss D, Laske C, Munk MH, Spottke A, Roy N, Wagner M, Roeske S, Heneka MT, Brosseron F, Ramirez A, Dobisch L, Wolfsgruber S, Kleineidam L, Yakupov R, Stark M, Schmid MC, Berger M, Hetzer S, Dechent P, Scheffler K, Petzold GC, Schneider A, Effland A, Radbruch A. Machine Learning-Based Perivascular Space Volumetry in Alzheimer Disease. Invest Radiol 2024; 59:667-676. [PMID: 38652067 DOI: 10.1097/rli.0000000000001077] [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: 04/25/2024]
Abstract
OBJECTIVES Impaired perivascular clearance has been suggested as a contributing factor to the pathogenesis of Alzheimer disease (AD). However, it remains unresolved when the anatomy of the perivascular space (PVS) is altered during AD progression. Therefore, this study investigates the association between PVS volume and AD progression in cognitively unimpaired (CU) individuals, both with and without subjective cognitive decline (SCD), and in those clinically diagnosed with mild cognitive impairment (MCI) or mild AD. MATERIALS AND METHODS A convolutional neural network was trained using manually corrected, filter-based segmentations (n = 1000) to automatically segment the PVS in the centrum semiovale from interpolated, coronal T2-weighted magnetic resonance imaging scans (n = 894). These scans were sourced from the national German Center for Neurodegenerative Diseases Longitudinal Cognitive Impairment and Dementia Study. Convolutional neural network-based segmentations and those performed by a human rater were compared in terms of segmentation volume, identified PVS clusters, as well as Dice score. The comparison revealed good segmentation quality (Pearson correlation coefficient r = 0.70 with P < 0.0001 for PVS volume, detection rate in cluster analysis = 84.3%, and Dice score = 59.0%). Subsequent multivariate linear regression analysis, adjusted for participants' age, was performed to correlate PVS volume with clinical diagnoses, disease progression, cerebrospinal fluid biomarkers, lifestyle factors, and cognitive function. Cognitive function was assessed using the Mini-Mental State Examination, the Comprehensive Neuropsychological Test Battery, and the Cognitive Subscale of the 13-Item Alzheimer's Disease Assessment Scale. RESULTS Multivariate analysis, adjusted for age, revealed that participants with AD and MCI, but not those with SCD, had significantly higher PVS volumes compared with CU participants without SCD ( P = 0.001 for each group). Furthermore, CU participants who developed incident MCI within 4.5 years after the baseline assessment showed significantly higher PVS volumes at baseline compared with those who did not progress to MCI ( P = 0.03). Cognitive function was negatively correlated with PVS volume across all participant groups ( P ≤ 0.005 for each). No significant correlation was found between PVS volume and any of the following parameters: cerebrospinal fluid biomarkers, sleep quality, body mass index, nicotine consumption, or alcohol abuse. CONCLUSIONS The very early changes of PVS volume may suggest that alterations in PVS function are involved in the pathophysiology of AD. Overall, the volumetric assessment of centrum semiovale PVS represents a very early imaging biomarker for AD.
Collapse
Affiliation(s)
- Katerina Deike
- From the German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany (K.D., A.D., K.F., O.K., F.J., Annika Spottke, N.R., M.W., S.R., M.T.H., F.B., Alfredo Ramirez, S.W., L.K., M.S., M.C.S., G.C.P., Anja Schneider, Alexander Radbruch); Department of Neuroradiology, University Hospital, Bonn, Germany (K.D., P.S., D.P., Alexander Radbruch); Department of Neurodegenerative Disease and Geriatric Psychiatry/Psychiatry, University Hospital Bonn, Bonn, Germany (J.H., N.Z., K.F., M.W., Alfredo Ramirez, S.W., L.K., Anja Schneider); Department of Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany (D.P.); German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany (O.P., S.D.F., J.P., E.S., S.A.); Institute of Psychiatry and Psychotherapy, Charité-Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany (O.P., S.D.F., L.-S.S., L.P.); Department of Psychiatry and Psychotherapy, Charité, Berlin, Germany (J.P., E.S., S.A., A.L.); Department of Psychiatry and Psychotherapy, School of Medicine, Munich, Germany (J.P.); University of Edinburgh and UK DRI, Edinburgh, United Kingdom (J.P.); German Center for Neurodegenerative Diseases (DZNE), Goettingen, Germany (J.W.); Department of Psychiatry and Psychotherapy, University Medical Center, Goettingen, Germany (J.W., C.B., N.H.); Neurosciences and Signaling Group, Institute of Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal (J.W.); Department of Psychiatry, University of Cologne, Cologne, Germany (F.J., Ayda Rostamzadeh); Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany (F.J., Alfredo Ramirez); German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany (E.D., W.G., E.I.I., Michaela Butryn, L.D., R.Y.); Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke University, Magdeburg, Germany (E.D., W.G., E.I.I., Michaela Butryn); Department for Psychiatry and Psychotherapy, University Clinic Magdeburg, Magdeburg, Germany (E.I.I.); German Center for Neurodegenerative Diseases (DZNE), Munich, Germany (K.B., M.E., R.P.); Institute for Stroke and Dementia Research, LMU Munich, Germany (K.B., D.J., M.E.); Department of Psychiatry and Psychotherapy, LMU Munich, Germany (R.P., B.-S.R.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (R.P.); Ageing Epidemiology Research Unit, School of Public Health, Imperial College London, London, United Kingdom (R.P.); Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, United Kingdom (R.P., B.-S.R.); Department of Neuroradiology, University Hospital Munich, Munich, Germany (B.-S.R.); German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany (S.T., I.K., D.G.); Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany (S.T., I.K., D.G.); German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany (C.L., M.H.M.); Section for Dementia Research, Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy, Tübingen, Germany (C.L.); Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen Germany (M.H.M.); Department of Neurology, University of Bonn, Bonn, Germany (Annika Spottke); Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Cologne, Germany (Alfredo Ramirez); Department of Psychiatry and Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, San Antonio, TX (Alfredo Ramirez); Institute for Medical Biometry, Informatics, and Epidemiology, University Hospital Bonn, Bonn, Germany (M.C.S., Moritz Berger); Berlin Center for Advanced Neuroimaging, Charité-Universitätsmedizin, Berlin, Germany (S.H.); MR-Research in Neurosciences, Department of Cognitive Neurology, Göttingen, Germany (P.D.); Department for Biomedical Magnetic Resonance, University of Tübingen, Tübingen, Germany (K.S.); Division of Vascular Neurology, Department of Neurology, University Hospital Bonn, Bonn, Germany (G.C.P.); and Institute for Applied Mathematics, University of Bonn, Bonn, Germany (A.E.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Colpaert M, Singh PK, Donohue KJ, Pires NT, Fuller DD, Corti M, Byrne BJ, Sun RC, Vander Kooi CW, Gentry MS. Neurological glycogen storage diseases and emerging therapeutics. Neurotherapeutics 2024; 21:e00446. [PMID: 39277505 PMCID: PMC11581880 DOI: 10.1016/j.neurot.2024.e00446] [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: 05/09/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/17/2024] Open
Abstract
Glycogen storage diseases (GSDs) comprise a group of inherited metabolic disorders characterized by defects in glycogen metabolism, leading to abnormal glycogen accumulation in multiple tissues, most notably affecting the liver, skeletal muscle, and heart. Recent findings have uncovered the importance of glycogen metabolism in the brain, sustaining a myriad of physiological functions and linking its perturbation to central nervous system (CNS) pathology. This link resulted in classification of neurological-GSDs (n-GSDs), a group of diseases with shared deficits in neurological glycogen metabolism. The n-GSD patients exhibit a spectrum of clinical presentations with common etiology while requiring tailored therapeutic approaches from the traditional GSDs. Recent research has elucidated the genetic and biochemical mechanisms and pathophysiological basis underlying different n-GSDs. Further, the last decade has witnessed some promising developments in novel therapeutic approaches, including enzyme replacement therapy (ERT), substrate reduction therapy (SRT), small molecule drugs, and gene therapy targeting key aspects of glycogen metabolism in specific n-GSDs. This preclinical progress has generated noticeable success in potentially modifying disease course and improving clinical outcomes in patients. Herein, we provide an overview of current perspectives on n-GSDs, emphasizing recent advances in understanding their molecular basis, therapeutic developments, underscore key challenges and the need to deepen our understanding of n-GSDs pathogenesis to develop better therapeutic strategies that could offer improved treatment and sustainable benefits to the patients.
Collapse
Affiliation(s)
- Matthieu Colpaert
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Pankaj K Singh
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
| | | | | | - David D Fuller
- Department of Physical Therapy and Breathing Research and Therapeutics Center, University of Florida, Gainesville, FL, USA
| | - Manuela Corti
- Department of Pediatrics, Powell Gene Therapy Center, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Barry J Byrne
- Department of Pediatrics, Powell Gene Therapy Center, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Ramon C Sun
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA; Center for Advanced Spatial Biomolecule Research (CASBR), University of Florida, Gainesville, FL, USA
| | - Craig W Vander Kooi
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA; Center for Advanced Spatial Biomolecule Research (CASBR), University of Florida, Gainesville, FL, USA
| | - Matthew S Gentry
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA; Center for Advanced Spatial Biomolecule Research (CASBR), University of Florida, Gainesville, FL, USA.
| |
Collapse
|