1
|
Singh V, Choudhury A, Ahn HJ. Fibrinogen's potential role in connecting cerebrovascular abnormalities with glymphatic dysfunction in Alzheimer's disease. Neural Regen Res 2025; 20:203-204. [PMID: 39657091 DOI: 10.4103/nrr.nrr-d-23-02093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 03/13/2024] [Indexed: 12/17/2024] Open
Affiliation(s)
- Vishal Singh
- Department of Pharmacology, Physiology and Neurosciences, Rutgers-New Jersey Medical School, Newark, NJ, USA (Singh V, Choudhury A, Ahn HJ)
| | - Arnab Choudhury
- Department of Pharmacology, Physiology and Neurosciences, Rutgers-New Jersey Medical School, Newark, NJ, USA (Singh V, Choudhury A, Ahn HJ)
| | - Hyung Jin Ahn
- Department of Pharmacology, Physiology and Neurosciences, Rutgers-New Jersey Medical School, Newark, NJ, USA (Singh V, Choudhury A, Ahn HJ)
- Brain Health Institute, Rutgers University, Piscataway, NJ, USA (Ahn HJ)
| |
Collapse
|
2
|
Hasegawa S, Yoshimaru D, Hayashi N, Shibukawa S, Takagi M, Murai H. Analyzing the relationship between specific brain structural changes and the diffusion tensor image analysis along the perivascular space index in idiopathic normal pressure hydrocephalus. J Neurol 2024; 272:56. [PMID: 39666072 DOI: 10.1007/s00415-024-12850-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 11/30/2024] [Accepted: 12/02/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND Diffusion Tensor Image Analysis Along the Perivascular Space (DTI-ALPS) evaluates the glymphatic system in patients with idiopathic normal-pressure hydrocephalus (iNPH). However, white matter compression due to ventricular enlargement may affect the ALPS index. This study aimed to investigate the relationship among the ALPS index, white matter changes, and clinical symptoms in patients with iNPH. METHODS We calculated the ALPS index in 30 patients with iNPH, aged 70 and above, using DTI data and correlated it with various clinical and imaging indices, including the Evans index, callosal angle, cognitive tests, gait assessment (timed up-and-go [TUG] test), cerebrospinal fluid (CSF) medullary pressure, and various DTI indices (axial diffusivity [AD], radial diffusivity [RD], mean diffusivity [MD], fractional anisotropy [FA]). RESULTS Significant negative correlations were observed between the ALPS index and the rate of change in step count in the TUG test after the tap test (r = -0.5014, p = 0.0048), as well as CSF medullary pressure (r = -0.4651, p = 0.0096). Positive correlations were identified between the ALPS index and both AD (r = 0.4984, p = 0.0051) and MD (r = 0.3631, p = 0.0486). CONCLUSION A lower ALPS index was associated with gait improvement following the tap test as well as higher CSF medullary pressure. The ALPS index may detect subtle periventricular compression-induced changes in iNPH. Consequently, it could potentially serve as a predictor for tap test effectiveness in patients with iNPH, offering a new perspective on its application in iNPH diagnosis and treatment.
Collapse
Affiliation(s)
- Shinya Hasegawa
- Department of Radiology, Chiba Saiseikai Narashino Hospital, 1-1-1 Izumicho, Narashino City, Chiba, 275-8580, Japan.
- Graduate School of Radiological Technology, Gunma Prefectural College of Health Sciences, 23-1 Kamiochicho, Maebashi City, Gunma, 371-0052, Japan.
| | - Daisuke Yoshimaru
- Division of Regenerative Medicine, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan.
- National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan.
- Department of Radiology, Tokyo Medical University, Tokyo, Japan.
| | - Norio Hayashi
- Graduate School of Radiological Technology, Gunma Prefectural College of Health Sciences, 23-1 Kamiochicho, Maebashi City, Gunma, 371-0052, Japan
| | - Shuhei Shibukawa
- Department of Radiology, Tokyo Medical University, Tokyo, Japan
- Faculty of Health Science, Department of Radiological Technology, Juntendo University, Tokyo, Japan
- Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan
| | - Mika Takagi
- Department of Radiology, Chiba Saiseikai Narashino Hospital, 1-1-1 Izumicho, Narashino City, Chiba, 275-8580, Japan
| | - Hisayuki Murai
- Department of Neurosurgery, Chiba Saiseikai Narashino Hospital, Chiba, Japan
| |
Collapse
|
3
|
Wu CH, Liao WH, Chu YC, Hsiao MY, Kung Y, Wang JL, Chen WS. Very Low-Intensity Ultrasound Facilitates Glymphatic Influx and Clearance via Modulation of the TRPV4-AQP4 Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401039. [PMID: 39494466 DOI: 10.1002/advs.202401039] [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: 01/28/2024] [Revised: 10/18/2024] [Indexed: 11/05/2024]
Abstract
Recently, the glymphatic system has been proposed as a mechanism for waste clearance from the brain parenchyma. Glymphatic dysfunction has previously been shown to be associated with several neurological diseases, including Alzheimer's disease, traumatic brain injury, and stroke. As such, it may serve as an important target for therapeutic interventions. In the present study, very low-intensity ultrasound (VLIUS) (center frequency, 1 MHz; pulse repetition frequency, 1 kHz; duty factor, 1%; spatial peak temporal average intensity [Ispta] = 3.68 mW cm2; and duration, 5 min) is found to significantly enhance the influx of cerebrospinal fluid tracers into the paravascular spaces of the brain, and further facilitate interstitial substance clearance from the brain parenchyma, including exogenous β-amyloid. Notably, no evidence of brain damage is observed following VLIUS stimulation. VLIUS may enhance glymphatic influx via the transient receptor potential vanilloid-4-aquaporin-4 pathway in astrocytes. This mechanism may provide insights into VLIUS-regulated glymphatic function that modifies the natural course of central nervous system disorders related to waste clearance dysfunction.
Collapse
Affiliation(s)
- Chueh-Hung Wu
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, 100, Taiwan
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu, 300, Taiwan
| | - Wei-Hao Liao
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, 100, Taiwan
| | - Ya-Cherng Chu
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, 106, Taiwan
| | - Ming-Yen Hsiao
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, 100, Taiwan
| | - Yi Kung
- Department of Biomechatronic Engineering, National Chiayi University, Chiayi, 600, Taiwan
| | - Jaw-Lin Wang
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, 106, Taiwan
| | - Wen-Shiang Chen
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, 100, Taiwan
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli, 350, Taiwan
| |
Collapse
|
4
|
Wang K, Tang Z, Yang Y, Guo Y, Liu Z, Su Z, Li X, Xiao G. Zebrafish as a Model Organism for Congenital Hydrocephalus: Characteristics and Insights. Zebrafish 2024; 21:361-384. [PMID: 39510565 DOI: 10.1089/zeb.2024.0148] [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: 11/15/2024] Open
Abstract
Hydrocephalus is a cerebrospinal fluid-related disease that usually manifests as abnormal dilation of the ventricles, with a triad of clinical findings including walking difficulty, reduced attention span, and urinary frequency or incontinence. The onset of congenital hydrocephalus is closely related to mutations in genes that regulate brain development. Currently, our understanding of the mechanisms of congenital hydrocephalus remains limited, and the prognosis of existing treatments is unsatisfactory. Additionally, there are no suitable or dedicated model organisms for congenital hydrocephalus. Therefore, it is significant to determine the mechanism and develop special animal models of congenital hydrocephalus. Recently, zebrafish have emerged as a popular model organism in many fields, including developmental biology, genetics, and toxicology. Its genome shares high similarity with that of humans, and it has fast and low-cost reproduction. These advantages make it suitable for studying the pathogenesis and therapeutic approaches for various diseases, specifically congenital diseases. This study explored the possibility of using zebrafish as a model organism for congenital hydrocephalus. This review describes the characteristics of zebrafish and discusses specific congenital hydrocephalus models. The advantages and limitations of using zebrafish for hydrocephalus research are highlighted, and insights for further model development are provided.
Collapse
Affiliation(s)
- Kaiyue Wang
- Department of Neurosurgery, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, PR China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, PR China
| | - Zhi Tang
- Department of Neurosurgery, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, PR China
| | - Yijian Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, PR China
| | - Yating Guo
- Department of Neurosurgery, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, PR China
| | - Zhikun Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, PR China
| | - Zhangjie Su
- Department of Neurosurgery, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge, United Kingdom
| | - Xuejun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, PR China
| | - Gelei Xiao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, PR China
| |
Collapse
|
5
|
Li H, Yao Q, Huang X, Yang X, Yu C. The role and mechanism of Aβ clearance dysfunction in the glymphatic system in Alzheimer's disease comorbidity. Front Neurol 2024; 15:1474439. [PMID: 39655162 PMCID: PMC11626247 DOI: 10.3389/fneur.2024.1474439] [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: 08/01/2024] [Accepted: 11/12/2024] [Indexed: 12/12/2024] Open
Abstract
Alzheimer's disease (AD) is the leading type of dementia globally, characterized by a complex pathogenesis that involves various comorbidities. An imbalance in the production and clearance of amyloid β-protein (Aβ) peptides in the brain is a key pathological mechanism of AD, with the glymphatic system playing a crucial role in Aβ clearance. Comorbidities associated with AD, such as diabetes, depression, and hypertension, not only affect Aβ production but also impair the brain's lymphatic system. Abnormalities in the structure and function of this system further weaken Aβ clearance capabilities, and the presence of comorbidities may exacerbate this process. This paper aims to review the role and specific mechanisms of impaired Aβ clearance via the glymphatic system in the context of AD comorbidities, providing new insights for the prevention and treatment of AD. Overall, the damage to the glymphatic system primarily focuses on aquaporin-4 (AQP4) and perivascular spaces (PVS), suggesting that maintaining the health of the glymphatic system may help slow the progression of AD and its comorbidities. Additionally, given the ongoing controversies regarding the structure of the glymphatic system, this paper revisits this structure and discusses the principles and characteristics of current detection methods for the glymphatic system.
Collapse
Affiliation(s)
| | | | | | - Xiaoyan Yang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Changyin Yu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
6
|
Singh V, Rochakim N, Ferraresso F, Choudhury A, Kastrup CJ, Ahn HJ. Caveolin-1 and Aquaporin-4 as Mediators of Fibrinogen-Driven Cerebrovascular Pathology in Hereditary Cerebral Amyloid Angiopathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.11.623066. [PMID: 39605467 PMCID: PMC11601418 DOI: 10.1101/2024.11.11.623066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Hereditary Cerebral Amyloid Angiopathy (HCAA) is a rare inherited form of CAA, characterized by increased vascular deposits of amyloid peptides. HCAA provides a unique opportunity to study the pathogenic mechanisms linked to CAA, as it is associated with severe cerebrovascular pathology. Some of HCAA-associated amyloid-β (Aβ) mutations significantly enhance the interaction between fibrinogen and Aβ, resulting in altered fibrin structure and co-deposition with Aβ in the perivascular space. However, the mechanisms underlying perivascular fibrinogen deposition and the associated cerebrovascular pathology in HCAA remain unclear. To investigate this, we analyzed TgSwDI transgenic mice carrying HCAA-associated mutations and observed a significant age-dependent increase in fibrin(ogen) extravasation and fibrin(ogen)-Aβ colocalization in the perivascular space. Moreover, Caveolin-1, a protein involved in non-specific transcytosis across the endothelium, significantly increased with age in TgSwDI mice and correlated with fibrin(ogen) extravasation. Additionally, we noted significant aquaporin-4 (AQP4) depolarization in the CAA-laden blood vessels of TgSwDI mice, which also correlated with fibrin(ogen) extravasation and fibrin(ogen)-Aβ colocalization. Given that AQP4 plays a crucial role in Aβ clearance via the glymphatic pathway, its depolarization may disrupt this critical clearance mechanism, thereby exacerbating CAA pathology. To further explore the relationship between fibrin(ogen) and these factors, we depleted fibrinogen in TgSwDI mice using siRNA against fibrinogen. This intervention resulted in decreased CAA, reduced caveolin-1 levels, attenuated microglial activation, restored polarized expression of AQP4, and improved spatial memory in fibrinogen-depleted TgSwDI mice. These findings suggest that targeting fibrinogen could be a promising strategy for mitigating CAA pathology and its associated cerebrovascular pathology. Significance Statement Our study reveals the mechanism by which fibrin(ogen)-Aβ colocalization could exacerbates CAA pathology. Our findings highlight that the age-dependent increase of endothelial caveolin-1 could facilitate fibrin(ogen) extravasation, which binds with Aβ in the perivascular space inducing microglial neuroinflammation and AQP4 depolarization, thus exacerbating CAA pathology. Furthermore, fibrinogen depletion could mitigate CAA severity, reduce microglial activation, restore AQP4 polarization and memory impairment. These results suggest that targeting fibrinogen and caveolin-1-mediated transcytosis may offer new strategies to address CAA-associated cerebrovascular pathology.
Collapse
|
7
|
MohanaSundaram A, Mofatteh M, Ashraf GM, Praticò D. Glymphotherapeutics for Alzheimer's disease: Time to move the needle. Ageing Res Rev 2024; 101:102478. [PMID: 39222666 DOI: 10.1016/j.arr.2024.102478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/15/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Alzheimer's disease (AD), the most predominant neurodegenerative disease and a quintessential entity within the dementia umbrella, is a global public health crisis. While the lack of disease modifying therapies has been a weak point in AD treatment, the success of recently approved monoclonal antibody-based therapeutics (aducanumab and lecanemab) targeted at the removal of amyloid-beta (Aβ) peptides in the brain is still under debate. There are multiple safety concerns about these approved neurotherapeutics including amyloid-related imaging abnormalities, stroke, meningitis, encephalitis, and even death. Novel paradigms focused on aquaporin-4-mediated neuro-perivascular Aβ and Tau protein clearance pathway are garnering attention. In this paper, we argue that orchestrating the drug discovery focused on glymphatic clearance-facilitating drugs ("glymphotherapeutics") might be a potentially novel and viable strategy to mitigate the progression and improve the clinical outcomes of AD.
Collapse
Affiliation(s)
- ArunSundar MohanaSundaram
- School of Pharmacy, Sathyabama Institute of Science and Technology, Chennai, Tamilnadu 600119, India
| | - Mohammad Mofatteh
- School of Medicine, Dentistry, and Biomedical Sciences, Queen's University Belfast, Belfast BT9 7BL, UK
| | - Ghulam Md Ashraf
- Department of Biological Sciences, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou 215123, Jiangsu, China
| | - Domenico Praticò
- Department of Neurosciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
8
|
Burke MR, Sotiropoulos I, Waites CL. The multiple roles of chronic stress and glucocorticoids in Alzheimer's disease pathogenesis. Trends Neurosci 2024; 47:933-948. [PMID: 39307629 PMCID: PMC11563862 DOI: 10.1016/j.tins.2024.08.015] [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: 05/02/2024] [Revised: 08/22/2024] [Accepted: 08/29/2024] [Indexed: 11/15/2024]
Abstract
Chronic stress and the accompanying long-term elevation of glucocorticoids (GCs), the stress hormones of the body, increase the risk and accelerate the progression of Alzheimer's disease (AD). Signatures of AD include intracellular tau (MAPT) tangles, extracellular amyloid β (Aβ) plaques, and neuroinflammation. A growing body of work indicates that stress and GCs initiate cellular processes underlying these pathologies through dysregulation of protein homeostasis and trafficking, mitochondrial bioenergetics, and response to damage-associated stimuli. In this review, we integrate findings from mechanistic studies in rodent and cellular models, wherein defined chronic stress protocols or GC administration have been shown to elicit AD-related pathology. We specifically discuss the effects of chronic stress and GCs on tau pathogenesis, including hyperphosphorylation, aggregation, and spreading, amyloid precursor protein (APP) processing and trafficking culminating in Aβ production, immune priming by proinflammatory cytokines and disease-associated molecular patterns, and alterations to glial cell and blood-brain barrier (BBB) function.
Collapse
Affiliation(s)
- Mia R Burke
- Department of Pathology and Cell Biology, Taub Institute for Research on Alzheimer's Disease and Aging Brain, Columbia University Irving Medical Center, New York, NY, USA; Pathobiology and Mechanisms of Disease Graduate Program, Columbia University Irving Medical Center, New York, NY, USA
| | - Ioannis Sotiropoulos
- Institute of Biosciences and Applications, National Centre for Scientific Research (NCSR) Demokritos, Agia Paraskevi, Greece
| | - Clarissa L Waites
- Department of Pathology and Cell Biology, Taub Institute for Research on Alzheimer's Disease and Aging Brain, Columbia University Irving Medical Center, New York, NY, USA; Department of Neuroscience, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
9
|
Panagopoulos D, Stranjalis G, Gavra M, Boviatsis E, Korfias S, Karydakis P. Current Trends in the Treatment of Pediatric Hydrocephalus: A Narrative Review Centered on the Indications, Safety, Efficacy, and Long-Term Outcomes of Available Treatment Modalities. CHILDREN (BASEL, SWITZERLAND) 2024; 11:1334. [PMID: 39594909 PMCID: PMC11593021 DOI: 10.3390/children11111334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 10/17/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024]
Abstract
The pathophysiologic substrate of pediatric hydrocephalus has not been thoroughly elucidated. Valve-based shunt systems have constituted the main therapeutic option since the late 1950s. The initially used systems were concerning the ventricular system and the atrium. In the 1970s, VA shunts were not the main stay of treatment as the preferred option for the terminal end of the drainage system was the peritoneum. Our review analyzes these valve types and attempts a comparison, based on their functional characteristics. Nowadays, the only available surgical alternative for the treatment of hydrocephalus is ETV. This technique is associated with lower infection rates as well as, on average, a lower re-operation rate. Another term that deserves special mention is related to the outcome of ETV in children who had a medical history of previously incorporated shunts and who were subsequently suffering from shunt malfunction. Well-recognized predictive factors associated with secondary ETV failure include age, early onset of hydrocephalus, and prematurity. Although several attempts have been made in order to establish the optimum surgical treatment management in the different subgroups of patientswho are suffering from shunt dysfunction, there is no universal agreement. Therefore, this review attempts to identify the specific subpopulations of patients in whom the insertion of a drainage system as the preferred treatment modality is associated with an optimum long-term prognosis, compared to ETV, and vice versa. The objective of our study is to analyze the safety, efficacy, and outcomes of drainage devices and ETV in pediatric hydrocephalus patients.
Collapse
Affiliation(s)
- Dimitrios Panagopoulos
- Neurosurgical Department, Pediatric Hospital of Athens, ‘Agia Sophia’, 45701 Athens, Greece
| | - Georgios Stranjalis
- 1st University Neurosurgical Department, Medical School, ‘Evangelismos’ Hospital, University of Athens, 10676 Athens, Greece; (G.S.); (S.K.)
| | - Maro Gavra
- Radiology Department, Pediatric Hospital of Athens, ‘Agia Sophia’, 10676 Athens, Greece;
| | - Efstathios Boviatsis
- 2nd University Neurosurgical Department, Medical School, ‘Attikon’ Hospital, University of Athens, 10676 Athens, Greece;
| | - Stefanos Korfias
- 1st University Neurosurgical Department, Medical School, ‘Evangelismos’ Hospital, University of Athens, 10676 Athens, Greece; (G.S.); (S.K.)
| | - Ploutarchos Karydakis
- Neurosurgical Department, General Hospital of Athens ‘Gennimatas’, 10676 Athens, Greece;
| |
Collapse
|
10
|
Grosu S, Nikolova T, Lorbeer R, Stoecklein VM, Rospleszcz S, Fink N, Schlett CL, Storz C, Beller E, Keeser D, Heier M, Kiefer LS, Maurer E, Walter SS, Ertl-Wagner BB, Ricke J, Bamberg F, Peters A, Stoecklein S. The spine-brain axis: is spinal anatomy associated with brain volume? Brain Commun 2024; 6:fcae365. [PMID: 39464212 PMCID: PMC11503949 DOI: 10.1093/braincomms/fcae365] [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: 04/29/2024] [Revised: 08/20/2024] [Accepted: 10/18/2024] [Indexed: 10/29/2024] Open
Abstract
First small sample studies indicate that disturbances of spinal morphology may impair craniospinal flow of cerebrospinal fluid and result in neurodegeneration. The aim of this study was to evaluate the association of cervical spinal canal width and scoliosis with grey matter, white matter, ventricular and white matter hyperintensity volumes of the brain in a large study sample. Four hundred participants underwent whole-body 3 T magnetic resonance imaging. Grey matter, white matter and ventricular volumes were quantified using a warp-based automated brain volumetric approach. Spinal canal diameters were measured manually at the cervical vertebrae 2/3 level. Scoliosis was evaluated using manual measurements of the Cobb angle. Linear binomial regression analyses of measures of brain volumes and spine anatomy were performed while adjusting for age, sex, hypertension, cholesterol levels, body mass index, smoking and alcohol consumption. Three hundred eighty-three participants were included [57% male; age: 56.3 (±9.2) years]. After adjustment, smaller spinal canal width at the cervical vertebrae 2/3 level was associated with lower grey matter (P = 0.034), lower white matter (P = 0.012) and higher ventricular (P = 0.006, inverse association) volume. Participants with scoliosis had lower grey matter (P = 0.005), lower white matter (P = 0.011) and larger brain ventricular (P = 0.003) volumes than participants without scoliosis. However, these associations were attenuated after adjustment. Spinal canal width at the cervical vertebrae 2/3 level and scoliosis were not associated with white matter hyperintensity volume before and after adjustment (P > 0.864). In our study, cohort smaller spinal canal width at the cervical vertebrae 2/3 level and scoliosis were associated with lower grey and white matter volumes and larger ventricle size. These characteristics of the spine might constitute independent risk factors for neurodegeneration.
Collapse
Affiliation(s)
- Sergio Grosu
- Department of Radiology, LMU University Hospital, LMU Munich, 81377 Munich, Germany
| | - Trayana Nikolova
- Department of Radiology, LMU University Hospital, LMU Munich, 81377 Munich, Germany
| | - Roberto Lorbeer
- Department of Radiology, LMU University Hospital, LMU Munich, 81377 Munich, Germany
| | - Veit M Stoecklein
- Department of Neurosurgery, LMU University Hospital, LMU Munich, 81377 Munich, Germany
| | - Susanne Rospleszcz
- German Research Center for Environmental Health, Institute of Epidemiology, Helmholtz Center Munich, 85764 Neuherberg, Germany
- Department of Epidemiology, Biometry and Epidemiology, Institute for Medical Information Processing, LMU Munich, 81377 Munich, Germany
| | - Nicola Fink
- Department of Radiology, LMU University Hospital, LMU Munich, 81377 Munich, Germany
| | - Christopher L Schlett
- Department of Diagnostic and Interventional Radiology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Corinna Storz
- Department of Neuroradiology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Ebba Beller
- Paediatric Radiology and Neuroradiology, Institute of Diagnostic and Interventional Radiology, University Medical Centre Rostock, 18057 Rostock, Germany
| | - Daniel Keeser
- Department of Radiology, LMU University Hospital, LMU Munich, 81377 Munich, Germany
- Department of Psychiatry, LMU University Hospital, LMU Munich, 80336 Munich, Germany
| | - Margit Heier
- German Research Center for Environmental Health, Institute of Epidemiology, Helmholtz Center Munich, 85764 Neuherberg, Germany
- KORA Study Centre, University Hospital of Augsburg, 86153 Augsburg, Germany
| | - Lena S Kiefer
- Department of Diagnostic and Interventional Radiology, University of Tuebingen, 72076 Tuebingen, Germany
- Department of Nuclear Medicine and Clinical Molecular Imaging, University of Tuebingen, 72076 Tuebingen, Germany
| | - Elke Maurer
- Department for Trauma and Reconstructive Surgery, BG Unfallklinik Tuebingen, University of Tuebingen, 72076 Tuebingen, Germany
| | - Sven S Walter
- KORA Study Centre, University Hospital of Augsburg, 86153 Augsburg, Germany
- Department of Radiology, Division of Musculoskeletal Radiology, New York University, Grossman School of Medicine, New York, NY 10016, USA
| | - Birgit B Ertl-Wagner
- Department of Radiology, LMU University Hospital, LMU Munich, 81377 Munich, Germany
- Department of Diagnostic Imaging, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada, M5G 1E8
- Department of Medical Imaging, University of Toronto, Toronto, ON, Canada, M5T 1W7
| | - Jens Ricke
- Department of Radiology, LMU University Hospital, LMU Munich, 81377 Munich, Germany
| | - Fabian Bamberg
- Department of Diagnostic and Interventional Radiology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Annette Peters
- German Research Center for Environmental Health, Institute of Epidemiology, Helmholtz Center Munich, 85764 Neuherberg, Germany
- Department of Epidemiology, Biometry and Epidemiology, Institute for Medical Information Processing, LMU Munich, 81377 Munich, Germany
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Sophia Stoecklein
- Department of Radiology, LMU University Hospital, LMU Munich, 81377 Munich, Germany
| |
Collapse
|
11
|
Ma J, Chen M, Liu GH, Gao M, Chen NH, Toh CH, Hsu JL, Wu KY, Huang CM, Lin CM, Fang JT, Lee SH, Lee TMC. Effects of sleep on the glymphatic functioning and multimodal human brain network affecting memory in older adults. Mol Psychiatry 2024:10.1038/s41380-024-02778-0. [PMID: 39397082 DOI: 10.1038/s41380-024-02778-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 10/15/2024]
Abstract
Understanding how sleep affects the glymphatic system and human brain networks is crucial for elucidating the neurophysiological mechanism underpinning aging-related memory declines. We analyzed a multimodal dataset collected through magnetic resonance imaging (MRI) and polysomnographic recording from 72 older adults. A proxy of the glymphatic functioning was obtained from the Diffusion Tensor Image Analysis along the Perivascular Space (DTI-ALPS) index. Structural and functional brain networks were constructed based on MRI data, and coupling between the two networks (SC-FC coupling) was also calculated. Correlation analyses revealed that DTI-ALPS was negatively correlated with sleep quality measures [e.g., Pittsburgh Sleep Quality Index (PSQI) and apnea-hypopnea index]. Regarding human brain networks, DTI-ALPS was associated with the strength of both functional connectivity (FC) and structural connectivity (SC) involving regions such as the middle temporal gyrus and parahippocampal gyrus, as well as with the SC-FC coupling of rich-club connections. Furthermore, we found that DTI-ALPS positively mediated the association between sleep quality and rich-club SC-FC coupling. The rich-club SC-FC coupling further mediated the association between DTI-ALPS and memory function in good sleepers but not in poor sleepers. The results suggest a disrupted glymphatic-brain relationship in poor sleepers, which underlies memory decline. Our findings add important evidence that sleep quality affects cognitive health through the underlying neural relationships and the interplay between the glymphatic system and multimodal brain networks.
Collapse
Affiliation(s)
- Junji Ma
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong SAR, China
- Laboratory of Neuropsychology & Human Neuroscience, The University of Hong Kong, Hong Kong SAR, China
| | - Menglu Chen
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong SAR, China
- Laboratory of Neuropsychology & Human Neuroscience, The University of Hong Kong, Hong Kong SAR, China
| | - Geng-Hao Liu
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Division of Acupuncture and Moxibustion, Center for Traditional Chinese Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- Sleep Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Mengxia Gao
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong SAR, China
- Laboratory of Neuropsychology & Human Neuroscience, The University of Hong Kong, Hong Kong SAR, China
| | - Ning-Hung Chen
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Sleep Center, Respiratory Therapy, Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Cheng Hong Toh
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan County, Taiwan
| | - Jung-Lung Hsu
- Department of Neurology, New Taipei Municipal TuCheng Hospital, New Taipei City, Taiwan
- Department of Neurology, at Linkou, Chang Gung Memorial Hospital and College of Medicine, Neuroscience Research Center, Chang-Gung University, Taoyuan, Taiwan
- Graduate Institute of Mind, Brain, & Consciousness, Taipei Medical University, Taipei, Taiwan
| | - Kuan-Yi Wu
- College of Medicine, Chang Gung University, Taoyuan County, Taiwan
- Department of Psychiatry, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Chih-Mao Huang
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Chih-Ming Lin
- College of Medicine, Chang Gung University, Taoyuan County, Taiwan
- Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Ji-Tseng Fang
- College of Medicine, Chang Gung University, Taoyuan County, Taiwan.
- Department of Nephrology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.
| | - Shwu-Hua Lee
- College of Medicine, Chang Gung University, Taoyuan County, Taiwan.
- Department of Psychiatry, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.
| | - Tatia M C Lee
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong SAR, China.
- Laboratory of Neuropsychology & Human Neuroscience, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
12
|
Ferreira Machado M, Muela HCS, Costa-Hong VA, Cristina Moraes N, Maia Memória C, Sanches Yassuda M, Bor-Seng-Shu E, Nitrini R, Aparecido Bortolotto L, de Carvalho Nogueira R. Angiotensin-converting enzyme inhibitors: a therapeutic option for controlling blood pressure associated with delayed cognitive processing speed. J Hum Hypertens 2024:10.1038/s41371-024-00965-8. [PMID: 39367178 DOI: 10.1038/s41371-024-00965-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 09/26/2024] [Indexed: 10/06/2024]
Abstract
Antihypertensive treatment (AT) is essential for preventing hypertension-related cognitive decline. The goals of this observational study were to compare cognitive performance (CP) between non-hypertensive (NH) volunteers and hypertensive patients and to evaluate the correlation between CP and antihypertensive drugs (AHD). Three groups were constituted: NH (n = 30) [group 1], hypertensive with systolic blood pressure (SBP) < 140 mmHg and diastolic blood pressure (DBP) < 90 mmHg (n = 54) [group 2] and hypertensive with SBP ≥ 140 or DBP ≥ 90 (n = 31) [group 3]. To analyze the cognitive domains, a neuropsychological battery was applied and the raw performance values in these tests were transformed into z-scores. The domain was considered impaired if it presented a z-score below -1.5 SD. Compared to group 1, both groups of hypertensive were older (51 [ ± 12] years) and showed a worse CP in episodic memory (p = 0.014), language (p = 0.003) and processing speed (PS) [p = 0.05]. Angiotensin-converting enzyme inhibitors (ACEi) and angiotensin receptor blockers (ARB) were the most used AHD (46.3%, p = 0.01 [group 2] and 64.5%, p = 0.005 [group 3]) and showed correlations with PS. Linear regression models revealed a negative association of PS with the use of ACEi (β = -0.230, p = 0.004), but not with the use of ARB (β = 0.208, p = 0.008). The effect of AT on cognition appears to go beyond the search for lower blood pressure targets and also includes the mechanism of action of AHD on the brain, so that additional benefits may possibly be achieved with simple adaptations in the treatment regimen, particularly in patients without clinically manifest cognitive impairment.
Collapse
Affiliation(s)
- Michel Ferreira Machado
- Department of Neurology, Hospital das Clínicas, University of São Paulo Medical School, São Paulo, Brazil.
| | | | | | - Natalia Cristina Moraes
- Department of Neurology, Hospital das Clínicas, University of São Paulo Medical School, São Paulo, Brazil
| | - Claudia Maia Memória
- Department of Neurology, Hospital das Clínicas, University of São Paulo Medical School, São Paulo, Brazil
| | - Monica Sanches Yassuda
- Gerontology, School of Arts, Sciences and Humanities, University of São Paulo Medical School, São Paulo, Brazil
| | - Edson Bor-Seng-Shu
- Department of Neurology, Hospital das Clínicas, University of São Paulo Medical School, São Paulo, Brazil
| | - Ricardo Nitrini
- Department of Neurology, Hospital das Clínicas, University of São Paulo Medical School, São Paulo, Brazil
| | - Luiz Aparecido Bortolotto
- Hypertension Unit, Instituto do Coração (INCOR), University of São Paulo Medical School, São Paulo, Brazil
| | | |
Collapse
|
13
|
Duy PQ, Mehta NH, Kahle KT. Biomechanical instability of the brain-CSF interface in hydrocephalus. Brain 2024; 147:3274-3285. [PMID: 38798141 PMCID: PMC11449143 DOI: 10.1093/brain/awae155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 02/15/2024] [Accepted: 05/05/2024] [Indexed: 05/29/2024] Open
Abstract
Hydrocephalus, characterized by progressive expansion of the CSF-filled ventricles (ventriculomegaly), is the most common reason for brain surgery. 'Communicating' (i.e. non-obstructive) hydrocephalus is classically attributed to a primary derangement in CSF homeostasis, such as choroid plexus-dependent CSF hypersecretion, impaired cilia-mediated CSF flow currents, or decreased CSF reabsorption via the arachnoid granulations or other pathways. Emerging data suggest that abnormal biomechanical properties of the brain parenchyma are an under-appreciated driver of ventriculomegaly in multiple forms of communicating hydrocephalus across the lifespan. We discuss recent evidence from human and animal studies that suggests impaired neurodevelopment in congenital hydrocephalus, neurodegeneration in elderly normal pressure hydrocephalus and, in all age groups, inflammation-related neural injury in post-infectious and post-haemorrhagic hydrocephalus, can result in loss of stiffness and viscoelasticity of the brain parenchyma. Abnormal brain biomechanics create barrier alterations at the brain-CSF interface that pathologically facilitates secondary enlargement of the ventricles, even at normal or low intracranial pressures. This 'brain-centric' paradigm has implications for the diagnosis, treatment and study of hydrocephalus from womb to tomb.
Collapse
Affiliation(s)
- Phan Q Duy
- Department of Neurosurgery, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Neel H Mehta
- Department of Neurosurgery, Harvard Medical School and Massachusetts General Hospital, Boston, MA 02115, USA
| | - Kristopher T Kahle
- Department of Neurosurgery, Harvard Medical School and Massachusetts General Hospital, Boston, MA 02115, USA
- Department of Neurosurgery, Boston Children’s Hospital, Boston, MA 02115, USA
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA 02114, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Developmental Brain and CSF Disorders Program, Massachusetts General Hospital, Boston, MA 02114, USA
- Program in Neuroscience, Harvard University, Cambridge, MA 02142, USA
| |
Collapse
|
14
|
Shlobin NA, Staple BL, Sclafani M, Harter DH. The Glymphatic System and Subarachnoid Lymphatic-Like Membrane: Recent Developments in Cerebrospinal Fluid Research. World Neurosurg 2024; 190:147-156. [PMID: 39002777 DOI: 10.1016/j.wneu.2024.07.062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 07/05/2024] [Accepted: 07/06/2024] [Indexed: 07/15/2024]
Abstract
BACKGROUND Cerebrospinal fluid (CSF) circulates throughout the ventricles, cranial and spinal subarachnoid spaces, and central spinal cord canal. CSF protects the central nervous system through mechanical cushioning, regulation of intracranial pressure, regulation of metabolic homeostasis, and provision of nutrients. Recently, investigators have characterized the glial-lymphatic (glymphatic) system, the analog of the lymphatic system in the central nervous system, and described a fourth meningeal layer; the subarachnoid lymphatic-like membrane (SLYM)relevant to the CSF. METHODS A narrative review was conducted. RESULTS In this review, we summarize these advances. We describe the development of the original model, controversies, a revised model, and a new conceptual framework. We characterize the biological functions, influence of sleep-wake cycles, and effect of aging with relevance to the glymphatic system. We highlight the role of the glymphatic system in Alzheimer's disease, idiopathic normal pressure hydrocephalus, ischemic stroke, subarachnoid hemorrhage, and traumatic brain injury. Next, we characterize the structure and role of the SLYM. Finally, we explore the relevance of the glymphatic system and SLYM to neurosurgery. CONCLUSIONS This manuscript will inform clinicians and scientists regarding preclinical and translational advances in the understanding of the structure, dynamics, and function of the CSF.
Collapse
Affiliation(s)
- Nathan A Shlobin
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
| | - Brandon L Staple
- College of Medicine, University of Nebraska, Omaha, Nebraska, USA
| | | | - David H Harter
- Department of Neurosurgery, NYU Langone, New York, New York, USA
| |
Collapse
|
15
|
Sun M, Zhang M, Di F, Bai W, Sun J, Zhang M, Sun J, Li M, Liang X. Polystyrene nanoplastics induced learning and memory impairments in mice by damaging the glymphatic system. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 284:116874. [PMID: 39153278 DOI: 10.1016/j.ecoenv.2024.116874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 08/02/2024] [Accepted: 08/09/2024] [Indexed: 08/19/2024]
Abstract
The excessive usage of nanoplastics (NPs) has posed a serious threat to the ecological environment and human health, which can enter the brain and then result in neurotoxicity. However, research on the neurotoxic effects of NPs based on different exposure routes and modifications of functional groups is lacking. In this study, the neurotoxicity induced by NPs was studied using polystyrene nanoplastics (PS-NPs) of different modifications (PS, PS-COOH, and PS-NH2). It was found that PS-NH2 through intranasal administration (INA) exposure route exhibited the greatest accumulation in the mice brain after exposure for 7 days. After the mice were exposed to PS-NH2 by INA means for 28 days, the exploratory ability and spatial learning ability were obviously damaged in a dose-dependent manner. Further analysis indicated that these damages induced by PS-NH2 were closely related to the decreased ability of glymphatic system to clear β-amyloid (Aβ) and phosphorylated Tau (P-Tau) proteins, which was ascribed to the loss of aquaporin-4 (AQP4) polarization in the astrocytic endfeet. Moreover, the loss of AQP4 polarization might be regulated by the NF-κB pathway. Our current study establishes the connection between the neurotoxicity induced by PS-NPs and the glymphatic system dysfunction for the first time, which will contribute to future research on the neurotoxicity of NPs.
Collapse
Affiliation(s)
- Meng Sun
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250014, China; School of Public Health, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Min Zhang
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Fanglin Di
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Weijie Bai
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250014, China
| | - Jikui Sun
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250014, China
| | - Mingkun Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250014, China
| | - Jinlong Sun
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250014, China
| | - Meng Li
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250014, China.
| | - Xue Liang
- School of Public Health, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China; Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China.
| |
Collapse
|
16
|
Yuan Y, Peng W, Lei J, Zhao Y, Zhao B, Li Y, Wang J, Qu Q. AQP4 Endocytosis-Lysosome Degradation Mediated by MMP-9/β-DG Involved in Diabetes Cognitive Impairment. Mol Neurobiol 2024; 61:8438-8453. [PMID: 38512439 DOI: 10.1007/s12035-024-04085-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 02/29/2024] [Indexed: 03/23/2024]
Abstract
Cognitive impairment is considered to be one of the important comorbidities of diabetes, but the underlying mechanisms are widely unknown. Aquaporin-4 (AQP4) is the most abundant water channel in the central nervous system, which plays a neuroprotective role in various neurological diseases by maintaining the function of glymphatic system and synaptic plasticity. However, whether AQP4 is involved in diabetes-related cognitive impairment remains unknown. β-dystroglycan (β-DG), a key molecule for anchoring AQP4 on the plasma membrane of astrocytes and avoiding its targeting to lysosomes for degradation, can be cleaved by matrix metalloproteinase-9 (MMP-9). β-DG deficiency can cause a decline in AQP4 via regulating its endocytosis. However, whether cleavage of β-DG can affect the expression of AQP4 remains unreported. In this study, we observed that diabetes mice displayed cognitive disorder accompanied by reduction of AQP4 in prefrontal cortex. And we found that bafilomycin A1, a widely used lysosome inhibitor, could reverse the downregulation of AQP4 in diabetes, further demonstrating that the reduction of AQP4 in diabetes is a result of more endocytosis-lysosome degradation. In further experiments, we found diabetes caused the excessive activation of MMP-9/β-DG which leaded to the loss of connection between AQP4 and β-DG, further inducing the endocytosis of AQP4. Moreover, inhibition of MMP-9/β-DG restored the endocytosis-lysosome degradation of AQP4 and partially alleviated cognitive dysfunction in diabetes. Our study sheds new light on the role of AQP4 in diabetes-associated cognitive disorder. And we provide a promising therapeutic target to reverse the endocytosis-lysosome degradation of AQP4 in diabetes, such as MMP-9/β-DG.
Collapse
Affiliation(s)
- Ye Yuan
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Rd, Xi'an, 710061, China
| | - Wei Peng
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Rd, Xi'an, 710061, China
| | - Jingna Lei
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Rd, Xi'an, 710061, China
| | - Yi Zhao
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Rd, Xi'an, 710061, China
| | - Beiyu Zhao
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Rd, Xi'an, 710061, China
| | - Yan Li
- Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jin Wang
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Rd, Xi'an, 710061, China.
| | - Qiumin Qu
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Rd, Xi'an, 710061, China.
- Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
17
|
Hu Y, Cao C, Li M, He H, Luo L, Guo Y. Association between idiopathic normal pressure hydrocephalus and Alzheimer's disease: a bidirectional Mendelian randomization study. Sci Rep 2024; 14:22744. [PMID: 39349954 PMCID: PMC11443089 DOI: 10.1038/s41598-024-72559-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 09/09/2024] [Indexed: 10/04/2024] Open
Abstract
Observational studies have suggested a bidirectional relationship between idiopathic normal pressure hydrocephalus (iNPH) and Alzheimer's disease (AD). However, the causal association between these two neurodegenerative disorders remains unclear. This study aimed to explore the causal relationship between iNPH and AD using a two-sample bidirectional Mendelian randomization (MR) method. Large-scale genome-wide association studies of iNPH (Ncase = 767, Ncontrol = 375,610) and AD (Ncase/proxy = 111,326, Ncontrol = 677,663) in European individuals were used to screen genetic instruments for MR analysis. Inverse variance-weighted (IVW) method was used as the main analysis, other MR methods and a series of sensitivity analyses were performed to ensure the reliability. In the forward MR analysis, genetic predisposition to iNPH had no effects on the risk of AD development. Likewise, in the reverse MR analysis, AD did not demonstrate a significant causal effect on iNPH. Sensitivity analyses bolstered the reliability of the MR results. Our MR study indicated no genetic evidence supporting a suggestive association between AD and iNPH in either direction, and provided evidence on the dichotomy between true iNPH and neurodegenerative NPH.
Collapse
Affiliation(s)
- Yuanjun Hu
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, China
| | - Chengan Cao
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Manting Li
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, China
| | - Haiyong He
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, China
| | - Lun Luo
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, China.
| | - Ying Guo
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, China.
| |
Collapse
|
18
|
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
|
19
|
Santiago J, Pocevičiūtė D, Wennström M. Perivascular phosphorylated TDP-43 inclusions are associated with Alzheimer's disease pathology and loss of CD146 and Aquaporin-4. Brain Pathol 2024:e13304. [PMID: 39251230 DOI: 10.1111/bpa.13304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 08/16/2024] [Indexed: 09/11/2024] Open
Abstract
The majority of patients with Alzheimer's disease (AD) exhibit aggregates of Trans-active response DNA binding protein 43 (TDP-43) in their hippocampus, which is associated with a more aggressive disease progression. The TDP-43 inclusions are commonly found in neurons, but also in astrocytes. The impact of the inclusions in astrocytes is less known. In the current study, we investigate the presence of phosphorylated TDP-43 (pTDP-43) inclusions in astrocytic endfeet and their potential association with blood-brain barrier (BBB) damage, glymphatic system dysfunction, and AD pathology. By staining postmortem hippocampal sections from AD patients and non-demented controls against TDP-43 and pTDP-43 together with the astrocytic markers glial fibrillary acidic protein (GFAP), astrocytic endfeet marker Aquaporin-4 (AQP4), and markers for BBB alterations (CD146) and leakiness (Immunoglobulin A), we demonstrate a close association between perivascular pTDP-43 or TDP-43 inclusions and GFAP or AQP4. These perivascular inclusions were more prominent in AD and correlated with the disease severity and loss of CD146 and AQP4. The findings indicate a relationship between pTDP-43 accumulation in astrocytic endfeet and BBB and glymphatic system dysfunction, which may contribute to the downstream pathological events seen in AD patients and the aggressive disease progression.
Collapse
Affiliation(s)
- Jessica Santiago
- Cognitive Disorder Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Dovilė Pocevičiūtė
- Cognitive Disorder Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Malin Wennström
- Cognitive Disorder Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| |
Collapse
|
20
|
Watkins L, Mukherjee S, Tithof J. Dynamics of waste proteins in brain tissue: Numerical insights into Alzheimer's risk factors. Phys Rev E 2024; 110:034401. [PMID: 39425375 DOI: 10.1103/physreve.110.034401] [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: 10/06/2023] [Accepted: 08/14/2024] [Indexed: 10/21/2024]
Abstract
Over the past few decades, research has indicated that the buildup of waste proteins, like amyloid-β (Aβ), in the brain's interstitial spaces is linked to neurodegenerative diseases like Alzheimer's, but the details of how such proteins are removed from the brain are not well understood. We have developed a numerical model to investigate the aggregation and clearance mechanisms of Aβ in the interstitial spaces of the brain. The model describes the volume-averaged transport of Aβ in a segment of the brain interstitium modeled as a porous medium, oriented between the perivascular space (fluid-filled channel surrounding a blood vessel) of a penetrating arteriole and that of a venule. Our numerical approach solves N coupled advection-diffusion-aggregation equations that model the production, aggregation, fragmentation, and clearance of N species of Aβ. We simulate N=50 species to investigate the oligomer-size dependence of clearance and aggregation. We introduce a timescale plot that helps predict Aβ buildup for different neurological conditions. We show that a sudden increase in monomer concentration, as occurs in conditions like traumatic brain injury, leads to significant plaque formation, which can qualitatively be predicted using the timescale plot. Our results also indicate that impaired protein clearance (as occurs with aging) and fragmentation are both mechanisms that sustain large intermediate oligomer concentrations. Our results provide novel insight into several known risk factors for Alzheimer's disease and cognitive decline, and we introduce a unique framing of Aβ dynamics as a competition between different timescales associated with production rates, aggregation rates, and clearance conditions.
Collapse
|
21
|
Choi S, Kum J, Hyun SY, Park TY, Kim H, Kim SK, Kim J. Transcranial focused ultrasound stimulation enhances cerebrospinal fluid movement: Real-time in vivo two-photon and widefield imaging evidence. Brain Stimul 2024; 17:1119-1130. [PMID: 39277129 DOI: 10.1016/j.brs.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 09/10/2024] [Accepted: 09/10/2024] [Indexed: 09/17/2024] Open
Abstract
BACKGROUND Cerebrospinal fluid (CSF) flow is crucial for brain homeostasis and its dysfunction is highly associated with neurodegenerative diseases. Restoring CSF circulation is proposed as a key strategy for the treatment of the diseases. Among the methods to improve CSF circulation, focused ultrasound (FUS) stimulation has emerged as a promising non-invasive brain stimulation technique, with effectiveness evidenced by ex vivo studies. However, due to technical disturbances in in vivo imaging combined with FUS, direct evidence of real-time in vivo CSF flow enhancement by FUS remains elusive. OBJECTIVE To investigate whether FUS administered through the skull base can enhance CSF influx in living animals with various real-time imaging techniques. METHODS We demonstrate a novel method of applying FUS through the skull base, facilitating cortical CSF influx, evidenced by diverse in vivo imaging techniques. Acoustic simulation confirmed effective sonication of our approach through the skull base. After injecting fluorescent CSF tracers into cisterna magna, FUS was administered at the midline of the jaw through the skull base for 30 min, during which imaging was performed concurrently. RESULTS Enhanced CSF influx was observed in macroscopic imaging, demonstrated by the influx area and intensity of the fluorescent dyes after FUS. In two-photon imaging, increased fluorescence was observed in the perivascular space (PVS) after stimulation. Moreover, particle tracking of microspheres showed more microspheres entering the imaging field, with increased mean speed after FUS. CONCLUSION Our findings provide direct real-time in vivo imaging evidence that FUS promotes CSF influx and flow in the PVS.
Collapse
Affiliation(s)
- Seunghwan Choi
- Department of East-West Medicine, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Jeungeun Kum
- Bionics Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Seon Young Hyun
- KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Tae Young Park
- Bionics Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Hyungmin Kim
- Bionics Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea; KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul, 02447, Republic of Korea.
| | - Sun Kwang Kim
- Department of East-West Medicine, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea; KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul, 02447, Republic of Korea; Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea.
| | - Jaeho Kim
- Department of Neurology, Dongtan Sacred Heart Hospital, Hallym University College of Medicine, Hwaseong-si, Gyeonggi-do, 18450, Republic of Korea.
| |
Collapse
|
22
|
Zhong W, Li W, Li Z, Wang Q, Zhang W. [Causal relationship between sleep phenotype and idiopathic normal pressure hydrocephalus: a two-sample bidirectional Mendelian randomization study]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:1612-1619. [PMID: 39276058 PMCID: PMC11378049 DOI: 10.12122/j.issn.1673-4254.2024.08.21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 09/16/2024]
Abstract
OBJECTIVE To explore the causal relationship between sleep phenotype and idiopathic normal pressure hydrocephalus (iNPH) using two-sample bidirectional Mendelian randomization. METHODS The exposure data including 8 sleep phenotypes used in this study were obtained from GWAS catalog, FinnGenR10 and MRCIEU GWAS. The outcome data for idiopathic normal-pressure hydrocephalus were obtained from FinnGen R10. We used the inverse-variance weighted (IVW) method to perform the principal analyses. Cochrane Q-statistics test was used to assess the heterogeneity and MR Egger‑intercept test performed to evaluate the pleiotropy for sensitivity analyses. RESULTS IVW result showed that frequent daytime nap was associated with higher odds of iNPH (OR=3.3393, 95 CI% : 1.0646-10.4742, P=0.0270). Cochrane Q-statistics test and MR Egger‑intercept test showed that the MR analysis had no pleiotropy or heterogeneity (P > 0.05). The external validation reproduced this result (OR=2.5660, 95 CI% : 1.1680-5.6373, P=0.0189; OR=4.0424, 95 CI% : 1.5709-10.4024, P=0.0038). Reverse Mendelian randomization suggested that iNPH did not have significant impact on sleep phenotype. CONCLUSION The frequency of daytime naps is causally associated with iNPH, and reducing the frequency of weekly daytime naps can reduce the risk of iNPH in the elderly population.
Collapse
Affiliation(s)
- W Zhong
- Department of Pediatric Neurosurgery, Center of Neurosurgery, Zhujiang Hospital, Southern Medical University//Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration//The National Key Clinical Specialty//The Engineering Technology Research Center of Education Ministry of China, Guangzhou 510282, China
| | - W Li
- Department of Pediatric Neurosurgery, Center of Neurosurgery, Zhujiang Hospital, Southern Medical University//Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration//The National Key Clinical Specialty//The Engineering Technology Research Center of Education Ministry of China, Guangzhou 510282, China
| | - Z Li
- Department of Pediatric Neurosurgery, Center of Neurosurgery, Zhujiang Hospital, Southern Medical University//Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration//The National Key Clinical Specialty//The Engineering Technology Research Center of Education Ministry of China, Guangzhou 510282, China
| | - Q Wang
- Department of Pediatric Neurosurgery, Center of Neurosurgery, Zhujiang Hospital, Southern Medical University//Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration//The National Key Clinical Specialty//The Engineering Technology Research Center of Education Ministry of China, Guangzhou 510282, China
| | - W Zhang
- Department of Pediatric Neurosurgery, Center of Neurosurgery, Zhujiang Hospital, Southern Medical University//Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration//The National Key Clinical Specialty//The Engineering Technology Research Center of Education Ministry of China, Guangzhou 510282, China
| |
Collapse
|
23
|
Kamalian A, Shirzadeh Barough S, Ho SG, Albert M, Luciano MG, Yasar S, Moghekar A. Molecular signatures of normal pressure hydrocephalus: a large-scale proteomic analysis of cerebrospinal fluid. Fluids Barriers CNS 2024; 21:64. [PMID: 39118132 PMCID: PMC11312837 DOI: 10.1186/s12987-024-00561-5] [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: 03/25/2024] [Accepted: 07/24/2024] [Indexed: 08/10/2024] Open
Abstract
Given the persistent challenge of differentiating idiopathic Normal Pressure Hydrocephalus (iNPH) from similar clinical entities, we conducted an in-depth proteomic study of cerebrospinal fluid (CSF) in 28 shunt-responsive iNPH patients, 38 Mild Cognitive Impairment (MCI) due to Alzheimer's disease, and 49 healthy controls. Utilizing the Olink Explore 3072 panel, we identified distinct proteomic profiles in iNPH that highlight significant downregulation of synaptic markers and cell-cell adhesion proteins. Alongside vimentin and inflammatory markers upregulation, these results suggest ependymal layer and transependymal flow dysfunction. Moreover, downregulation of multiple proteins associated with congenital hydrocephalus (e.g., L1CAM, PCDH9, ISLR2, ADAMTSL2, and B4GAT1) points to a possible shared molecular foundation between congenital hydrocephalus and iNPH. Through orthogonal partial least squares discriminant analysis (OPLS-DA), a panel comprising 13 proteins has been identified as potential diagnostic biomarkers of iNPH, pending external validation. These findings offer novel insights into the pathophysiology of iNPH, with implications for improved diagnosis.
Collapse
Affiliation(s)
- Aida Kamalian
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
| | | | - Sara G Ho
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
| | - Marilyn Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
| | - Mark G Luciano
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
| | - Sevil Yasar
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
| | - Abhay Moghekar
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA.
| |
Collapse
|
24
|
Seidler RD, Mao XW, Tays GD, Wang T, Zu Eulenburg P. Effects of spaceflight on the brain. Lancet Neurol 2024; 23:826-835. [PMID: 38945144 DOI: 10.1016/s1474-4422(24)00224-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 04/06/2024] [Accepted: 05/14/2024] [Indexed: 07/02/2024]
Abstract
The number of long duration human spaceflights has increased substantially over the past 15 years, leading to the discovery of numerous effects on the CNS. Microgravity results in headward fluid shifts, ventricular expansion, an upward shift of the brain within the skull, and remodelling of grey and white matter. The fluid changes are correlated with changes to perivascular space and spaceflight associated neuro-ocular syndrome. Microgravity alters the vestibular processing of head tilt and results in reduced tactile and proprioceptive inputs during spaceflight. Sensory adaptation is reflected in postflight effects, evident as transient sensorimotor impairment. Another major concern is that galactic cosmic radiation, which spacefarers will be exposed to when going beyond the magnetosphere around Earth, might have a negative effect on CNS function. Research with rodents points to the potential disruptive effects of space radiation on blood-brain barrier integrity and brain structures. More work is needed to understand and mitigate these effects on the CNS before humans travel to Mars, as the flight durations will be longer than anyone has previously experienced.
Collapse
Affiliation(s)
- Rachael D Seidler
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA; Department of Neurology, University of Florida, Gainesville, FL, USA.
| | - Xiao Wen Mao
- Department of Basic Sciences, Division of Biomedical Engineering Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Grant D Tays
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
| | - Tianyi Wang
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
| | - Peter Zu Eulenburg
- Institute for Neuroradiology, Ludwig-Maximilians University Munich, Munich, Germany
| |
Collapse
|
25
|
Dong S, Zhao H, Nie M, Sha Z, Feng J, Liu M, Lv C, Chen Y, Jiang W, Yuan J, Qian Y, Wan H, Gao C, Jiang R. Cannabidiol Alleviates Neurological Deficits After Traumatic Brain Injury by Improving Intracranial Lymphatic Drainage. J Neurotrauma 2024; 41:e2009-e2025. [PMID: 38553903 DOI: 10.1089/neu.2023.0539] [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: 04/30/2024] Open
Abstract
Traumatic brain injury (TBI) persists as a substantial clinical dilemma, largely because of the absence of effective treatments. This challenge is exacerbated by the hindered clearance of intracranial metabolic byproducts and the continual accrual of deleterious proteins. The glymphatic system (GS) and meningeal lymphatic vessels (MLVs), key elements of the intracranial lymphatic network, play critical roles in the clearance of harmful substances. Cannabidiol (CBD) has shown promise in reducing metabolite overload and bolstering cognitive performance in various neurodegenerative diseases. The precise mechanisms attributing to its beneficial effects in TBI scenarios, however, are yet to be distinctly understood. Utilizing a fluid percussion injury paradigm, our research adopted a multifaceted approach, encompassing behavioral testing, immunofluorescence and immunohistochemical analyses, laser speckle imaging, western blot techniques, and bilateral cervical efferent lymphatic ligation. This methodology aimed to discern the influence of CBD on both neurological outcomes and intracranial lymphatic clearance in a murine TBI model. We observed that CBD administration notably ameliorated motor, memory, and cognitive functions, concurrently with a significant reduction in the concentration of phosphorylated tau protein and amyloid-β. In addition, CBD expedited the turnover and elimination of intracranial tracers, increased cerebral blood flow, and enhanced the efficacy of fluorescent tracer migration from MLVs to deep cervical lymph nodes (dCLNs). Remarkably, CBD treatment also induced a reversion in aquaporin-4 (AQP-4) polarization and curtailed neuroinflammatory indices. A pivotal discovery was that the surgical interruption of efferent lymphatic conduits in the neck nullified CBD's positive contributions to intracranial waste disposal and cognitive improvement, yet the anti-neuroinflammatory actions remained unaffected. These insights suggest that CBD may enhance intracranial metabolite clearance, potentially via the regulation of the intracranial lymphatic system, thereby offering neurofunctional prognostic improvement in TBI models. Our findings underscore the potential therapeutic applicability of CBD in TBI interventions, necessitating further comprehensive investigations and clinical validations to substantiate these initial conclusions.
Collapse
Affiliation(s)
- Shiying Dong
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Post Neuro-injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin, Tianjin, China
| | - Hongwei Zhao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Post Neuro-injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin, Tianjin, China
- Department of Neurosurgery, Binzhou Medical University Hospital, Binzhou, China
| | - Meng Nie
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Post Neuro-injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin, Tianjin, China
| | - Zhuang Sha
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Post Neuro-injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin, Tianjin, China
| | - Jiancheng Feng
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Post Neuro-injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin, Tianjin, China
| | - Mingqi Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Post Neuro-injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin, Tianjin, China
| | - Chuanxiang Lv
- Department of Neurosurgery, The First Clinical Hospital, Jilin University, Changchun, China
| | - Yupeng Chen
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Post Neuro-injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin, Tianjin, China
| | - Weiwei Jiang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Post Neuro-injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin, Tianjin, China
| | - Jiangyuan Yuan
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Post Neuro-injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin, Tianjin, China
| | - Yu Qian
- Tianjin Neurological Institute, Key Laboratory of Post Neuro-injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin, Tianjin, China
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Honggang Wan
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Post Neuro-injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin, Tianjin, China
| | - Chuang Gao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Post Neuro-injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin, Tianjin, China
| | - Rongcai Jiang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Post Neuro-injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin, Tianjin, China
- State Key Laboratory of Experimental Hematology, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
26
|
Pramotton FM, Spitz S, Kamm RD. Challenges and Future Perspectives in Modeling Neurodegenerative Diseases Using Organ-on-a-Chip Technology. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403892. [PMID: 38922799 PMCID: PMC11348103 DOI: 10.1002/advs.202403892] [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: 04/12/2024] [Revised: 06/01/2024] [Indexed: 06/28/2024]
Abstract
Neurodegenerative diseases (NDDs) affect more than 50 million people worldwide, posing a significant global health challenge as well as a high socioeconomic burden. With aging constituting one of the main risk factors for some NDDs such as Alzheimer's disease (AD) and Parkinson's disease (PD), this societal toll is expected to rise considering the predicted increase in the aging population as well as the limited progress in the development of effective therapeutics. To address the high failure rates in clinical trials, legislative changes permitting the use of alternatives to traditional pre-clinical in vivo models are implemented. In this regard, microphysiological systems (MPS) such as organ-on-a-chip (OoC) platforms constitute a promising tool, due to their ability to mimic complex and human-specific tissue niches in vitro. This review summarizes the current progress in modeling NDDs using OoC technology and discusses five critical aspects still insufficiently addressed in OoC models to date. Taking these aspects into consideration in the future MPS will advance the modeling of NDDs in vitro and increase their translational value in the clinical setting.
Collapse
Affiliation(s)
- Francesca Michela Pramotton
- Department of Mechanical Engineering and Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Sarah Spitz
- Department of Mechanical Engineering and Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Roger D. Kamm
- Department of Mechanical Engineering and Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| |
Collapse
|
27
|
Liu X, Barisano G, Shao X, Jann K, Ringman JM, Lu H, Arfanakis K, Caprihan A, DeCarli C, Gold BT, Maillard P, Satizabal CL, Fadaee E, Habes M, Stables L, Singh H, Fischl B, van der Kouwe A, Schwab K, Helmer KG, Greenberg SM, Wang DJ. Cross-Vendor Test-Retest Validation of Diffusion Tensor Image Analysis along the Perivascular Space (DTI-ALPS) for Evaluating Glymphatic System Function. Aging Dis 2024; 15:1885-1898. [PMID: 37307817 PMCID: PMC11272201 DOI: 10.14336/ad.2023.0321-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 03/21/2023] [Indexed: 06/14/2023] Open
Abstract
The diffusion tensor image analysis along the perivascular space (DTI-ALPS) method was proposed to evaluate glymphatic system (GS) function. However, few studies have validated its reliability and reproducibility. Fifty participants' DTI data from the MarkVCID consortium were included in this study. Two pipelines by using DSI studio and FSL software were developed for data processing and ALPS index calculation. The ALPS index was obtained by the average of bilateral ALPS index and was used for testing the cross-vendor, inter-rater and test-retest reliability by using R studio software. The ALPS index demonstrated favorable inter-scanner reproducibility (ICC=0.77 to 0.95, P< 0.001), inter-rater reliability (ICC=0.96 to 1, P< 0.001) and test-retest repeatability (ICC=0.89 to 0.95, P< 0.001), offering a potential biomarker for in vivo evaluation of GS function.
Collapse
Affiliation(s)
- Xiaodan Liu
- Laboratory of FMRI Technology (LOFT), Mark & Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA.
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, USA.
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA.
| | | | - Xingfeng Shao
- Laboratory of FMRI Technology (LOFT), Mark & Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA.
| | - Kay Jann
- Laboratory of FMRI Technology (LOFT), Mark & Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA.
| | - John M Ringman
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Hanzhang Lu
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Konstantinos Arfanakis
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, USA.
- Department of Diagnostic Radiology and Nuclear Medicine, Rush University, Chicago, IL, USA.
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA.
| | | | - Charles DeCarli
- Department of Neurology, University of California, Davis, Davis, CA, USA.
| | - Brian T Gold
- Department of Neuroscience, University of Kentucky, Lexington, KY, USA.
| | - Pauline Maillard
- Department of Neurology, University of California, Davis, Davis, CA, USA.
| | - Claudia L Satizabal
- Population Health Sciences and Glenn Biggs Institute for Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| | - Elyas Fadaee
- Neuroimage Analytics Laboratory and Biggs Institute Neuroimaging Core, Glenn Biggs Institute for Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| | - Mohamad Habes
- Neuroimage Analytics Laboratory and Biggs Institute Neuroimaging Core, Glenn Biggs Institute for Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| | - Lara Stables
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA.
| | - Herpreet Singh
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
| | - Bruce Fischl
- Department of Radiology, Harvard Medical School, Boston, MA, USA.
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA.
- Division of Health Sciences and Technology, Massachusetts Institute of Technology, Computer Science and AI Lab, Cambridge, Massachusetts, USA.
| | - Andre van der Kouwe
- Department of Radiology, Harvard Medical School, Boston, MA, USA.
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA.
| | - Kristin Schwab
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
| | - Karl G Helmer
- Department of Radiology, Harvard Medical School, Boston, MA, USA.
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA.
| | - Steven M Greenberg
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
| | - Danny J.J Wang
- Laboratory of FMRI Technology (LOFT), Mark & Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA.
| | | |
Collapse
|
28
|
Boyd ED, Kaur J, Ding G, Chopp M, Jiang Q. Clinical magnetic resonance imaging evaluation of glymphatic function. NMR IN BIOMEDICINE 2024; 37:e5132. [PMID: 38465514 DOI: 10.1002/nbm.5132] [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: 06/07/2023] [Revised: 01/31/2024] [Accepted: 02/06/2024] [Indexed: 03/12/2024]
Abstract
The glymphatic system is a system of specialized perivascular spaces in the brain that facilitates removal of toxic waste solutes from the brain. Evaluation of glymphatic system function by means of magnetic resonance imaging (MRI) has thus far been largely focused on rodents because of the limitations of intrathecal delivery of gadolinium-based contrast agents to humans. This review discusses MRI methods that can be employed clinically for glymphatic-related measurements intended for early diagnosis, prevention, and the treatment of various neurological conditions. Although glymphatic system-based MRI research is in its early stages, recent studies have identified promising noninvasive MRI markers associated with glymphatic system alterations in neurological diseases. However, further optimization in data acquisition, validation, and modeling are needed to investigate the glymphatic system within the clinical setting.
Collapse
Affiliation(s)
- Edward D Boyd
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, USA
- Department of Radiology, Michigan State University, East Lansing, Michigan, USA
| | - Jasleen Kaur
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, USA
- Department of Physics, Oakland University, Rochester, Michigan, USA
| | - Guangliang Ding
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, USA
- Department of Radiology, Michigan State University, East Lansing, Michigan, USA
| | - Michael Chopp
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, USA
- Department of Physics, Oakland University, Rochester, Michigan, USA
| | - Quan Jiang
- Department of Neurology, Henry Ford Health System, Detroit, Michigan, USA
- Department of Radiology, Michigan State University, East Lansing, Michigan, USA
- Department of Physics, Oakland University, Rochester, Michigan, USA
| |
Collapse
|
29
|
Chang T, Huang X, Zhang X, Li J, Bai W, Wang J. A bibliometric analysis and visualization of normal pressure hydrocephalus. Front Neurol 2024; 15:1442493. [PMID: 39144708 PMCID: PMC11322097 DOI: 10.3389/fneur.2024.1442493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 07/18/2024] [Indexed: 08/16/2024] Open
Abstract
Background Normal pressure hydrocephalus (NPH) has drawn an increasing amount of attention over the last 20 years. At present, there is a shortage of intuitive analysis on the trends in development, key contributors, and research hotspots topics in the NPH field. This study aims to analyze the evolution of NPH research, evaluate publications both qualitatively and quantitatively, and summarize the current research hotspots. Methods A bibliometric analysis was conducted on data retrieved from the Web of Science Core Collection (WoSCC) database between 2003 and 2023. Quantitative assessments were conducted using bibliometric analysis tools such as VOSviewer and CiteSpace software. Results A total of 2,248 articles published between 2003 and 2023 were retrieved. During this period, the number of publications steadily increased. The United States was the largest contributor. The University of Gothenburg led among institutions conducting relevant research. Eide P. K. was the most prolific author. The Journal of Neurosurgery is the leading journal on NPH. According to the analysis of the co-occurrence of keywords and co-cited references, the primary research directions identified were pathophysiology, precise diagnosis, and individualized treatment. Recent research hotspots have mainly focused on epidemiology, the glymphatic system, and CSF biomarkers. Conclusion The comprehensive bibliometric analysis of NPH offers insights into the main research directions, highlights key countries, contributors, and journals, and identifies significant research hotspots. This information serves as a valuable reference for scholars to further study NPH.
Collapse
Affiliation(s)
- Tengwu Chang
- Department of Neurosurgery, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumchi, China
| | - Xiaoyuan Huang
- Department of Neurosurgery, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumchi, China
| | - Xu Zhang
- Xinjiang Second Medical College, Karamay, China
| | - JinYong Li
- Department of Neurosurgery, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumchi, China
| | - Wenju Bai
- Department of Neurosurgery, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumchi, China
| | - Jichao Wang
- Department of Neurosurgery, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumchi, China
| |
Collapse
|
30
|
Ge GR, Song W, Giannetto MJ, Rolland JP, Nedergaard M, Parker KJ. Mouse brain elastography changes with sleep/wake cycles, aging, and Alzheimer's disease. Neuroimage 2024; 295:120662. [PMID: 38823503 PMCID: PMC11409907 DOI: 10.1016/j.neuroimage.2024.120662] [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: 02/27/2024] [Revised: 05/05/2024] [Accepted: 05/30/2024] [Indexed: 06/03/2024] Open
Abstract
Understanding the physiological processes in aging and how neurodegenerative disorders affect cognitive function is a high priority for advancing human health. One specific area of recently enabled research is the in vivo biomechanical state of the brain. This study utilized reverberant optical coherence elastography, a high-resolution elasticity imaging method, to investigate stiffness changes during the sleep/wake cycle, aging, and Alzheimer's disease in murine models. Four-dimensional scans of 44 wildtype mice, 13 mice with deletion of aquaporin-4 water channel, and 12 mice with Alzheimer-related pathology (APP/PS1) demonstrated that (1) cortical tissue became softer (on the order of a 10% decrease in shear wave speed) when young wildtype mice transitioned from wake to anesthetized, yet this effect was lost in aging and with mice overexpressing amyloid-β or lacking the water channel AQP4. (2) Cortical stiffness increased with age in all mice lines, but wildtype mice exhibited the most prominent changes as a function of aging. The study provides novel insight into the brain's biomechanics, the constraints of fluid flow, and how the state of brain activity affects basic properties of cortical tissues.
Collapse
Affiliation(s)
- Gary R Ge
- The Institute of Optics, University of Rochester, 480 Intercampus Drive, Rochester, NY 14627, USA
| | - Wei Song
- Center for Translational Neuromedicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Michael J Giannetto
- Center for Translational Neuromedicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Jannick P Rolland
- The Institute of Optics, University of Rochester, 480 Intercampus Drive, Rochester, NY 14627, USA; Department of Biomedical Engineering, University of Rochester, 204 Robert B. Goergen Hall, Rochester, NY 14627, USA; Center for Visual Science, University of Rochester, 361 Meliora Hall, Rochester, NY 14627, USA
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA; Center for Translational Neuromedicine, University of Copenhagen, Blegdamsvej 3B, 2200-N, Denmark.
| | - Kevin J Parker
- Department of Biomedical Engineering, University of Rochester, 204 Robert B. Goergen Hall, Rochester, NY 14627, USA; Department of Electrical and Computer Engineering, University of Rochester, 500 Computer Studies Building, Rochester, NY 14627, USA; Department of Imaging Sciences (Radiology), University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA.
| |
Collapse
|
31
|
Taranov A, Bedolla A, Iwasawa E, Brown FN, Baumgartner S, Fugate EM, Levoy J, Crone SA, Goto J, Luo Y. The choroid plexus maintains adult brain ventricles and subventricular zone neuroblast pool, which facilitates poststroke neurogenesis. Proc Natl Acad Sci U S A 2024; 121:e2400213121. [PMID: 38954546 PMCID: PMC11252789 DOI: 10.1073/pnas.2400213121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 06/07/2024] [Indexed: 07/04/2024] Open
Abstract
The brain's neuroreparative capacity after injuries such as ischemic stroke is partly contained in the brain's neurogenic niches, primarily the subventricular zone (SVZ), which lies in close contact with the cerebrospinal fluid (CSF) produced by the choroid plexus (ChP). Despite the wide range of their proposed functions, the ChP/CSF remain among the most understudied compartments of the central nervous system (CNS). Here, we report a mouse genetic tool (the ROSA26iDTR mouse line) for noninvasive, specific, and temporally controllable ablation of CSF-producing ChP epithelial cells to assess the roles of the ChP and CSF in brain homeostasis and injury. Using this model, we demonstrate that ChP ablation causes rapid and permanent CSF volume loss in both aged and young adult brains, accompanied by disruption of ependymal cilia bundles. Surprisingly, ChP ablation did not result in overt neurological deficits at 1 mo postablation. However, we observed a pronounced decrease in the pool of SVZ neuroblasts (NBs) following ChP ablation, which occurs due to their enhanced migration into the olfactory bulb. In the middle cerebral artery occlusion model of ischemic stroke, NB migration into the lesion site was also reduced in the CSF-depleted mice. Thus, our study establishes an important role of ChP/CSF in regulating the regenerative capacity of the adult brain under normal conditions and after ischemic stroke.
Collapse
Affiliation(s)
- Aleksandr Taranov
- Department of Molecular and Cellular Biosciences, College of Medicine, University of Cincinnati, Cincinnati, OH45229
| | - Alicia Bedolla
- Department of Molecular and Cellular Biosciences, College of Medicine, University of Cincinnati, Cincinnati, OH45229
| | - Eri Iwasawa
- Division of Pediatric Neurosurgery, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
| | - Farrah N. Brown
- Division of Pediatric Neurosurgery, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
| | - Sarah Baumgartner
- Division of Pediatric Neurosurgery, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
| | - Elizabeth M. Fugate
- Imaging Research Center, Department of Radiology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH45229
| | - Joel Levoy
- Imaging Research Center, Department of Radiology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH45229
| | - Steven A. Crone
- Division of Pediatric Neurosurgery, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
- Department of Neurosurgery, College of Medicine, University of Cincinnati, Cincinnati, OH45267
| | - June Goto
- Division of Pediatric Neurosurgery, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
- Department of Neurosurgery, College of Medicine, University of Cincinnati, Cincinnati, OH45267
| | - Yu Luo
- Department of Molecular and Cellular Biosciences, College of Medicine, University of Cincinnati, Cincinnati, OH45229
| |
Collapse
|
32
|
Mayo F, González-Vinceiro L, Hiraldo-González L, Rodríguez-Gómez FD, Calle-Castillejo C, Mayo M, Netti V, Ramírez-Lorca R, Echevarría M. Impact of aquaporin-4 and CD11c + microglia in the development of ependymal cells in the aqueduct: inferences to hydrocephalus. Fluids Barriers CNS 2024; 21:53. [PMID: 38956598 PMCID: PMC11221146 DOI: 10.1186/s12987-024-00548-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 05/07/2024] [Indexed: 07/04/2024] Open
Abstract
AQP4 is expressed in the endfeet membranes of subpial and perivascular astrocytes and in the ependymal cells that line the ventricular system. The sporadic appearance of obstructive congenital hydrocephalus (OCHC) has been observed in the offspring of AQP4-/- mice (KO) due to stenosis of Silvio's aqueduct. Here, we explore whether the lack of AQP4 expression leads to abnormal development of ependymal cells in the aqueduct of mice. We compared periaqueductal samples from wild-type and KO mice. The microarray-based transcriptome analysis reflected a large number of genes with differential expression (809). Gene sets (GS) associated with ependymal development, ciliary function and the immune system were specially modified qPCR confirmed reduced expression in the KO mice genes: (i) coding for transcription factors for ependymal differentiation (Rfx4 and FoxJ1), (ii) involved in the constitution of the central apparatus of the axoneme (Spag16 and Hydin), (iii) associated with ciliary assembly (Cfap43, Cfap69 and Ccdc170), and (iv) involved in intercellular junction complexes of the ependyma (Cdhr4). By contrast, genes such as Spp1, Gpnmb, Itgax, and Cd68, associated with a Cd11c-positive microglial population, were overexpressed in the KO mice. Electron microscopy and Immunofluorescence of vimentin and γ-tubulin revealed a disorganized ependyma in the KO mice, with changes in the intercellular complex union, unevenly orientated cilia, and variations in the planar cell polarity of the apical membrane. These structural alterations translate into reduced cilia beat frequency, which might alter cerebrospinal fluid movement. The presence of CD11c + microglia cells in the periaqueductal zone of mice during the first postnatal week is a novel finding. In AQP4-/- mice, these cells remain present around the aqueduct for an extended period, showing peak expression at P11. We propose that these cells play an important role in the normal development of the ependyma and that their overexpression in KO mice is crucial to reduce ependyma abnormalities that could otherwise contribute to the development of obstructive hydrocephalus.
Collapse
Affiliation(s)
- Francisco Mayo
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Av. Manuel Siurot s/n, 41013, Seville, Spain
- Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, 41009, Seville, Spain
| | - Lourdes González-Vinceiro
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Av. Manuel Siurot s/n, 41013, Seville, Spain
- Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, 41009, Seville, Spain
| | - Laura Hiraldo-González
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Av. Manuel Siurot s/n, 41013, Seville, Spain
- Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, 41009, Seville, Spain
| | - Francisco D Rodríguez-Gómez
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Av. Manuel Siurot s/n, 41013, Seville, Spain
| | - Claudia Calle-Castillejo
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Av. Manuel Siurot s/n, 41013, Seville, Spain
| | - Manuel Mayo
- Física Teórica, Universidad de Sevilla, Apartado de Correos 1065, 41080, Seville, Spain
| | - Vanina Netti
- Facultad de Medicina, Departamento de Ciencias Fisiológicas, Laboratorio de Biomembranas, Universidad de Buenos Aires- CONICET, Instituto de Fisiología y Biofísica ''Bernardo Houssay'' (IFIBIO-HOUSSAY), Buenos Aires, Argentina
| | - Reposo Ramírez-Lorca
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Av. Manuel Siurot s/n, 41013, Seville, Spain
- Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, 41009, Seville, Spain
| | - Miriam Echevarría
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Av. Manuel Siurot s/n, 41013, Seville, Spain.
- Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, 41009, Seville, Spain.
| |
Collapse
|
33
|
Salminen A. Aryl hydrocarbon receptor impairs circadian regulation in Alzheimer's disease: Potential impact on glymphatic system dysfunction. Eur J Neurosci 2024; 60:3901-3920. [PMID: 38924210 DOI: 10.1111/ejn.16450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 05/23/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024]
Abstract
Circadian clocks maintain diurnal rhythms of sleep-wake cycle of 24 h that regulate not only the metabolism of an organism but also many other periodical processes. There is substantial evidence that circadian regulation is impaired in Alzheimer's disease. Circadian clocks regulate many properties known to be disturbed in Alzheimer's patients, such as the integrity of the blood-brain barrier (BBB) as well as the diurnal glymphatic flow that controls waste clearance from the brain. Interestingly, an evolutionarily conserved transcription factor, that is, aryl hydrocarbon receptor (AhR), impairs the function of the core clock proteins and thus could disturb diurnal rhythmicity in the BBB. There is abundant evidence that the activation of AhR signalling inhibits the expression of the major core clock proteins, such as the brain and muscle arnt-like 1 (BMAL1), clock circadian regulator (CLOCK) and period circadian regulator 1 (PER1) in different experimental models. The expression of AhR is robustly increased in the brains of Alzheimer's patients, and protein level is enriched in astrocytes of the BBB. It seems that AhR signalling inhibits glymphatic flow since it is known that (i) activation of AhR impairs the function of the BBB, which is cooperatively interconnected with the glymphatic system in the brain, and (ii) neuroinflammation and dysbiosis of gut microbiota generate potent activators of AhR, which are able to impair glymphatic flow. I will examine current evidence indicating that activation of AhR signalling could disturb circadian functions of the BBB and impair glymphatic flow and thus be involved in the development of Alzheimer's pathology.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
34
|
Xiong Z, Bai M, Wang Z, Wang R, Tian C, Wang L, Nie L, Zeng X. Resting-state fMRI network efficiency as a mediator in the relationship between the glymphatic system and cognitive function in obstructive sleep apnea hypopnea syndrome: Insights from a DTI-ALPS investigation. Sleep Med 2024; 119:250-257. [PMID: 38704873 DOI: 10.1016/j.sleep.2024.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/15/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
INTRODUCTION Obstructive sleep apnea hypopnea syndrome (OSAHS) is associated with cognitive impairment and physiological complications, necessitating further understanding of its mechanisms. This study investigates the relationship between glymphatic system function, brain network efficiency, and cognitive impairment in OSAHS patients using diffusion tensor image analysis along the perivascular space (DTI-ALPS) and resting-state fMRI. MATERIALS AND METHODS This study included 31 OSAHS patients and 34 age- and gender-matched healthy controls (HC). All participants underwent GE 3.0T magnetic resonance imaging (MRI) with diffusion tensor image (DTI) and resting-state fMRI scans. The DTI-ALPS index and brain functional networks were assessed. Differences between groups and correlations with clinical characteristics were analyzed. Additionally, the mediating role of brain network efficiency was explored. Finally, receiver operating characteristics (ROC) analysis assessed diagnostic performance. RESULTS OSAHS patients had significantly lower ALPS-index (1.268 vs. 1.431, p < 0.0001) and moderate negative correlation with Apnea Hypopnea Index (AHI) (r = -0.389, p = 0.031), as well as moderate positive correlation with Montreal Cognitive Assessment (MoCA) (r = 0.525, p = 0.002). Moreover, global efficiency (Eg) of the brain network was positively correlated with the ALPS-index and MoCA scores in OSAHS patients (r = 0.405, p = 0.024; r = 0.56, p = 0.001, respectively). Furthermore, mediation analysis showed that global efficiency partially mediated the impact of glymphatic system dysfunction on cognitive impairment in OSAHS patients (indirect effect = 4.58, mediation effect = 26.9 %). The AUROC for identifying OSAHS and HC was 0.80 (95 % CI 0.69 to 0.91) using an ALPS-index cut-off of 1.35. CONCLUSIONS OSAHS patients exhibit decreased ALPS-index, indicating impaired glymphatic system function. Dysfunction of the glymphatic system can affect cognitive function in OSAHS by disrupting brain functional network, suggesting a potential underlying pathological mechanism. Additionally, preliminary findings suggest that the ALPS-index may offer promise as a potential indicator for OSAHS.
Collapse
Affiliation(s)
- Zhenliang Xiong
- Engineering Research Center of Text Computing & Cognitive Intelligence, Ministry of Education, Key Laboratory of Intelligent Medical Image Analysis and Precise Diagnosis of Guizhou Province, State Key Laboratory of Public Big Data, College of Computer Science and Technology, Guizhou University, Guiyang, China; Department of Radiology, Guizhou Provincial People's Hospital, Key Laboratory of Intelligent Medical Imaging Analysis and Accurate Diagnosis of Guizhou Province, International Exemplary Cooperation Base of Precision Imaging for Diagnosis and Treatment, Guiyang, China
| | - Mingxian Bai
- Department of Radiology, Guizhou Provincial People's Hospital, Key Laboratory of Intelligent Medical Imaging Analysis and Accurate Diagnosis of Guizhou Province, International Exemplary Cooperation Base of Precision Imaging for Diagnosis and Treatment, Guiyang, China
| | - Zhongxin Wang
- Department of Pulmonary and Critical Care Medicine, Guizhou Provincial People's Hospital, Guiyang, China
| | - Rongpin Wang
- Department of Radiology, Guizhou Provincial People's Hospital, Key Laboratory of Intelligent Medical Imaging Analysis and Accurate Diagnosis of Guizhou Province, International Exemplary Cooperation Base of Precision Imaging for Diagnosis and Treatment, Guiyang, China
| | - Chong Tian
- Department of Radiology, Guizhou Provincial People's Hospital, Key Laboratory of Intelligent Medical Imaging Analysis and Accurate Diagnosis of Guizhou Province, International Exemplary Cooperation Base of Precision Imaging for Diagnosis and Treatment, Guiyang, China
| | - Lihui Wang
- Engineering Research Center of Text Computing & Cognitive Intelligence, Ministry of Education, Key Laboratory of Intelligent Medical Image Analysis and Precise Diagnosis of Guizhou Province, State Key Laboratory of Public Big Data, College of Computer Science and Technology, Guizhou University, Guiyang, China
| | - Lisha Nie
- GE Healthcare, MR Research China, Beijing, China.
| | - Xianchun Zeng
- Department of Radiology, Guizhou Provincial People's Hospital, Key Laboratory of Intelligent Medical Imaging Analysis and Accurate Diagnosis of Guizhou Province, International Exemplary Cooperation Base of Precision Imaging for Diagnosis and Treatment, Guiyang, China.
| |
Collapse
|
35
|
Drenthen GS, Elschot EP, van der Knaap N, Uher D, Voorter PHM, Backes WH, Jansen JFA, van der Thiel MM. Imaging Interstitial Fluid With MRI: A Narrative Review on the Associations of Altered Interstitial Fluid With Vascular and Neurodegenerative Abnormalities. J Magn Reson Imaging 2024; 60:40-53. [PMID: 37823526 DOI: 10.1002/jmri.29056] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/27/2023] [Accepted: 09/27/2023] [Indexed: 10/13/2023] Open
Abstract
Interstitial fluid (ISF) refers to the fluid between the parenchymal cells and along the perivascular spaces (PVS). ISF plays a crucial role in delivering nutrients and clearing waste products from the brain. This narrative review focuses on the use of MRI techniques to measure various ISF characteristics in humans. The complementary value of contrast-enhanced and noncontrast-enhanced techniques is highlighted. While contrast-enhanced MRI methods allow measurement of ISF transport and flow, they lack quantitative assessment of ISF properties. Noninvasive MRI techniques, including multi-b-value diffusion imaging, free-water-imaging, T2-decay imaging, and DTI along the PVS, offer promising alternatives to derive ISF measures, such as ISF volume and diffusivity. The emerging role of these MRI techniques in investigating ISF alterations in neurodegenerative diseases (eg, Alzheimer's disease and Parkinson's disease) and cerebrovascular diseases (eg, cerebral small vessel disease and stroke) is discussed. This review also emphasizes current challenges of ISF imaging, such as the microscopic scale at which ISF has to be measured, and discusses potential focus points for future research to overcome these challenges, for example, the use of high-resolution imaging techniques. Noninvasive MRI methods for measuring ISF characteristics hold significant potential and may have a high clinical impact in understanding the pathophysiology of neurodegenerative and cerebrovascular disorders, as well as in evaluating the efficacy of ISF-targeted therapies in clinical trials. LEVEL OF EVIDENCE: 3 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Gerhard S Drenthen
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
- School for Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Elles P Elschot
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
- School for Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Noa van der Knaap
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
- School for Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Daniel Uher
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
- School for Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, the Netherlands
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Paulien H M Voorter
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
- School for Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Walter H Backes
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
- School for Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Jacobus F A Jansen
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
- School for Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, the Netherlands
- Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Merel M van der Thiel
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
- School for Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, the Netherlands
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
36
|
Wang Z, Zuo M, Li W, Chen S, Yuan Y, He Y, Yang Y, Mao Q, Liu Y. The impact of telomere length on the risk of idiopathic normal pressure hydrocephalus: a bidirectional Mendelian randomization study. Sci Rep 2024; 14:14713. [PMID: 38926610 PMCID: PMC11208170 DOI: 10.1038/s41598-024-65725-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 06/24/2024] [Indexed: 06/28/2024] Open
Abstract
Idiopathic normal pressure hydrocephalus (iNPH) affects mainly aged populations. The gradual shortening of telomere length (TL) is one of the hallmarks of aging. Whereas the genetic contribution of TL to the iNPH is incompletely understood. We aimed to investigate the causal relationship between TL and iNPH through the Mendelian randomization (MR) analysis. We respectively obtained 186 qualified single nucleotide polymorphisms (SNPs) of TL and 20 eligible SNPs of iNPH for MR analysis. The result of MR analysis showed that genetically predicted longer TL was significantly associated with a reduced odd of iNPH (odds ratio [OR] = 0.634 95% Confidence interval [CI] 0.447-0.899, p = 0.011). The causal association remained consistent in multivariable MR (OR = 0.530 95% CI 0.327-0.860, p = 0.010). However, there was no evidence that the iNPH was causally associated with the TL (OR = 1.000 95% CI 0.996-1.004, p = 0.955). Our study reveals a potential genetic contribution of TL to the etiology of iNPH, that is a genetically predicted increased TL might be associated with a reduced risk of iNPH.
Collapse
Affiliation(s)
- Zhihao Wang
- Department of Neurosurgery, West China Hospital, Sichuan University, No 37 Guoxue Avenue, Chengdu, 610041, Sichuan, China
| | - Mingrong Zuo
- Department of Pediatric Neurosurgery, West China Women's and Children's Hospital: Sichuan University West China Second University Hospital, Chengdu, 610041, China
| | - Wenhao Li
- Department of Neurosurgery, West China Hospital, Sichuan University, No 37 Guoxue Avenue, Chengdu, 610041, Sichuan, China
| | - Siliang Chen
- Department of Neurosurgery, West China Hospital, Sichuan University, No 37 Guoxue Avenue, Chengdu, 610041, Sichuan, China
| | - Yunbo Yuan
- Department of Neurosurgery, West China Hospital, Sichuan University, No 37 Guoxue Avenue, Chengdu, 610041, Sichuan, China
| | - Yuze He
- Department of Neurosurgery, West China Hospital, Sichuan University, No 37 Guoxue Avenue, Chengdu, 610041, Sichuan, China
| | - Yuan Yang
- Department of Neurosurgery, West China Hospital, Sichuan University, No 37 Guoxue Avenue, Chengdu, 610041, Sichuan, China
| | - Qing Mao
- Department of Neurosurgery, West China Hospital, Sichuan University, No 37 Guoxue Avenue, Chengdu, 610041, Sichuan, China
| | - Yanhui Liu
- Department of Neurosurgery, West China Hospital, Sichuan University, No 37 Guoxue Avenue, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
37
|
Lu J, Wang X, Xu F, Rao C, Guo Y, Su Z, Chen S, Li Q. Exploring causal correlations of inflammatory biomarkers in idiopathic normal-pressure hydrocephalus: insights from bidirectional Mendelian randomization analysis. Front Aging Neurosci 2024; 16:1412434. [PMID: 38974901 PMCID: PMC11224557 DOI: 10.3389/fnagi.2024.1412434] [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: 04/04/2024] [Accepted: 06/11/2024] [Indexed: 07/09/2024] Open
Abstract
Background and objective Neuroinflammatory processes have been identified as playing a crucial role in the pathophysiology of various neurodegenerative diseases, including idiopathic normal-pressure hydrocephalus (iNPH). iNPH, defined as a common disease of cognitive impairment in older adults, poses major challenges for therapeutic interventions owing to the stringent methodological requirements of relevant studies, clinical heterogeneity, unclear etiology, and uncertain diagnostic criteria. This study aims to assess the relationship between circulating inflammatory biomarkers and iNPH risk using bidirectional two-sample Mendelian randomization (MR) combined with meta-analysis. Methods In our bidirectional MR study, genetic data from a genome-wide association study (GWAS) involving 1,456 iNPH cases and 409,726 controls of European ancestry were employed. Single-nucleotide polymorphisms (SNPs) associated with exposures served as instrumental variables for estimating the causal relationships between iNPH and 132 types of circulating inflammatory biomarkers from corresponding GWAS data. Causal associations were primarily examined using the inverse variance-weighted method, supplemented by MR-Egger, weighted median, simple mode, and weighted mode analyses. In the results, heterogeneity was assessed using the Cochran Q test. Horizontal pleiotropy was evaluated through the MR-Egger intercept test and the MR pleiotropy residual sum and outliers test. Sensitivity analysis was conducted through leave-one-out analysis. Reverse MR analyses were performed to mitigate bias from reverse causality. Meta-analyses of identical inflammatory biomarkers from both data sources strengthened the findings. Results Results indicated a genetically predicted association between Interleukin-16 (IL-16) [OR: 1.228, 95% CI: 1.049-1.439, p = 0.011], TNF-related apoptosis ligand (TRAIL) [OR: 1.111, 95% CI: 1.019-1.210, p = 0.017] and Urokinase-type plasminogen activator (uPA) [OR: 1.303, 95% CI: 1.025-1.658, p = 0.031] and the risk of iNPH. Additionally, changes in human Glial cell line-derived neurotrophic factor (hGDNF) [OR: 1.044, 95% CI: 1.006-1.084, p = 0.023], Matrix metalloproteinase-1 (MMP-1) [OR: 1.058, 95% CI: 1.020, 1.098, p = 0.003] and Interleukin-12p70 (IL-12p70) [OR: 0.897, 95% CI: 0.946-0.997, p = 0.037] levels were identified as possible consequences of iNPH. Conclusion Our MR study of inflammatory biomarkers and iNPH, indicated that IL-16, TRAIL, and uPA contribute to iNPH pathogenesis. Furthermore, iNPH may influence the expression of hGDNF, MMP-1, and IL-12p70. Therefore, targeting specific inflammatory biomarkers could be promising strategy for future iNPH treatment and prevention.
Collapse
Affiliation(s)
- Jianglong Lu
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xianpeng Wang
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Fanjie Xu
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Changjun Rao
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yuhang Guo
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhipeng Su
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Siyan Chen
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qun Li
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
38
|
Elias-Mas A, Wang JY, Rodríguez-Revenga L, Kim K, Tassone F, Hessl D, Rivera SM, Hagerman R. Enlarged perivascular spaces and their association with motor, cognition, MRI markers and cerebrovascular risk factors in male fragile X premutation carriers. J Neurol Sci 2024; 461:123056. [PMID: 38772058 DOI: 10.1016/j.jns.2024.123056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 05/15/2024] [Accepted: 05/16/2024] [Indexed: 05/23/2024]
Abstract
FMR1 premutation carriers (55-200 CGG repeats) are at risk of developing fragile X-associated tremor/ataxia syndrome (FXTAS), a neurodegenerative disorder associated with motor and cognitive impairment. Bilateral hyperintensities of the middle cerebellar peduncles (MCP sign) are the major radiological hallmarks of FXTAS. In the general population, enlarged perivascular spaces (PVS) are biomarkers of small vessel disease and glymphatic dysfunction and are associated with cognitive decline. Our aim was to determine if premutation carriers show higher ratings of PVS than controls and whether enlarged PVS are associated with motor and cognitive impairment, MRI features of neurodegeneration, cerebrovascular risk factors and CGG repeat length. We evaluated 655 MRIs (1-10 visits/participant) from 229 carriers (164 with FXTAS and 65 without FXTAS) and 133 controls. PVS in the basal ganglia (BG-EPVS), centrum semiovale, and midbrain were evaluated with a semiquantitative scale. Mixed-effects models were used for statistical analysis adjusting for age. In carriers with FXTAS, we revealed that (1) BG-PVS ratings were higher than those of controls and carriers without FXTAS; (2) BG-PVS severity was associated with brain atrophy, white matter hyperintensities, enlarged ventricles, FXTAS stage and abnormal gait; (3) age-related increase in BG-PVS was associated with cognitive dysfunction; and (4) PVS ratings of all three regions showed robust associations with CGG repeat length and were higher in carriers with the MCP sign than carriers without the sign. This study demonstrates clinical relevance of PVS in FXTAS especially in the basal ganglia region and suggests microangiopathy and dysfunctional cerebrospinal fluid circulation in FXTAS physiopathology.
Collapse
Affiliation(s)
- Andrea Elias-Mas
- Radiology Department, Hospital Universitari Mútua de Terrassa, Terrassa, Barcelona, Spain; Institute for Research and Innovation Parc Taulí (I3PT), Sabadell, Spain; Genetics Doctorate Program, Universitat de Barcelona (UB), Barcelona, Spain.
| | - Jun Yi Wang
- Center for Mind and Brain, University of California Davis, CA, United States.
| | - Laia Rodríguez-Revenga
- Biochemistry and Molecular Genetics Department, Hospital Clinic of Barcelona, Barcelona, Spain; CIBER of Rare Diseases (CIBERER), Instituto de Salud Carlos III, Barcelona, Spain; Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| | - Kyoungmi Kim
- Department of Public Health Sciences, University of California Davis School of Medicine, Sacramento, CA, United States.
| | - Flora Tassone
- MIND Institute, University of California Davis, Sacramento, CA, United States; Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, Sacramento, CA, United States.
| | - David Hessl
- MIND Institute, University of California Davis, Sacramento, CA, United States; Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, United States.
| | - Susan M Rivera
- Center for Mind and Brain, University of California Davis, CA, United States; MIND Institute, University of California Davis, Sacramento, CA, United States; Department of Psychology, University of Maryland, College Park, MD, United States.
| | - Randi Hagerman
- MIND Institute, University of California Davis, Sacramento, CA, United States; Department of Pediatrics, University of California Davis Medical Center, Sacramento, CA, United States.
| |
Collapse
|
39
|
Margoni M, Pagani E, Meani A, Preziosa P, Mistri D, Gueye M, Moiola L, Filippi M, Rocca MA. Cognitive Impairment Is Related to Glymphatic System Dysfunction in Pediatric Multiple Sclerosis. Ann Neurol 2024; 95:1080-1092. [PMID: 38481063 DOI: 10.1002/ana.26911] [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: 10/30/2023] [Revised: 03/04/2024] [Accepted: 03/04/2024] [Indexed: 05/18/2024]
Abstract
OBJECTIVE The aim of this study was to investigate whether, compared to pediatric healthy controls (HCs), the glymphatic system is impaired in pediatric multiple sclerosis (MS) patients according to their cognitive status, and to assess its association with clinical disability and MRI measures of brain structural damage. METHODS Sixty-five pediatric MS patients (females = 62%; median age = 15.5 [interquartile range, IQR = 14.5;17.0] years) and 23 age- and sex-matched HCs (females = 44%; median age = 14.1 [IQR = 11.8;16.2] years) underwent neurological, neuropsychological and 3.0 Tesla MRI assessment, including conventional and diffusion tensor imaging (DTI). We calculated the diffusion along the perivascular space (DTI-ALPS) index, a proxy of glymphatic function. Cognitive impairment (Co-I) was defined as impairment in at least 2 cognitive domains. RESULTS No significant differences in DTI-ALPS index were found between HCs and cognitively preserved (Co-P) pediatric MS patients (estimated mean difference [EMD] = -0.002 [95% confidence interval = -0.069; 0.065], FDR-p = 0.956). Compared to HCs and Co-P patients, Co-I pediatric MS patients (n = 20) showed significantly lower DTI-ALPS index (EMD = -0.136 [95% confidence interval = -0.214; -0.058], FDR-p ≤ 0.004). In HCs, no associations were observed between DTI-ALPS index and normalized brain, cortical and thalamic volumes, and normal-appearing white matter (NAWM) fractional anisotropy (FA) and mean diffusivity (MD) (FDR-p ≥ 0.348). In pediatric MS patients, higher brain WM lesion volume (LV), higher NAWM MD, lower normalized thalamic volume, and lower NAWM FA were associated with lower DTI-ALPS index (FDR-p ≤ 0.016). Random Forest selected lower DTI-ALPS index (relative importance [RI] = 100%), higher brain WM LV (RI = 59.5%) NAWM MD (RI = 57.1%) and intelligence quotient (RI = 51.3%) as informative predictors of cognitive impairment (out-of-bag area under the curve = 0.762). INTERPRETATION Glymphatic system dysfunction occurs in pediatric MS, is associated with brain focal lesions, irreversible tissue loss accumulation and cognitive impairment. ANN NEUROL 2024;95:1080-1092.
Collapse
Affiliation(s)
- Monica Margoni
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elisabetta Pagani
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alessandro Meani
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Paolo Preziosa
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Damiano Mistri
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Mor Gueye
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lucia Moiola
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Massimo Filippi
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
- Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maria Assunta Rocca
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
40
|
Xiong Y, Yu Q, Zhi H, Peng H, Xie M, Li R, Li K, Ma Y, Sun P. Advances in the study of the glymphatic system and aging. CNS Neurosci Ther 2024; 30:e14803. [PMID: 38887168 PMCID: PMC11183173 DOI: 10.1111/cns.14803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 04/26/2024] [Accepted: 05/29/2024] [Indexed: 06/20/2024] Open
Abstract
The glymphatic system is cerebrospinal fluid-brain tissue fluid exchange flow mediated by aquaporin-4 (AQP4) on the end feet of astrocytes for a system, which is capable of rapidly removing brain metabolites and thus maintaining brain homeostasis, and is known as the central immune system. Dysfunction of the glymphatic system causes accumulation of misfolded and highly phosphorylated proteins (amyloid-β and Tau proteins), which destabilizes the proteins, and the body's neuroinflammatory factors are altered causing aging of the immune system and leading to neurodegenerative diseases. Damage to the glymphatic system and aging share common manifestations, as well as unstudied biological mechanisms that are also linked, such as mitochondria, oxidative stress, chronic inflammation, and sleep. In this paper, we first summarize the structure, function, and research methods of the glymphatic system and the relationship between the glymphatic system and the peripheral immune system, and second, sort out and summarize the factors of the glymphatic system in removing metabolites and resolving aging-related diseases and factors affecting aging, to explore its related biological mechanisms, and moreover, to provide a new way of thinking for treating or intervening aging-related diseases.
Collapse
Affiliation(s)
- Ying Xiong
- School of Traditional Chinese MedicineShandong University of Traditional Chinese MedicineJinanChina
| | - Qingying Yu
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical SciencesGuangzhou University of Chinese MedicineGuangzhouChina
| | - Haimei Zhi
- Qilu Hospital of Shandong UniversityJinanChina
| | - Huiyuan Peng
- Department of RehabilitationZhongshan Hospital of Traditional Chinese MedicineZhongshanChina
| | - Mingjun Xie
- School of Traditional Chinese MedicineShandong University of Traditional Chinese MedicineJinanChina
| | - Renjun Li
- Department of PsychiatryJinan Mental Health CenterJinanChina
| | - Kejian Li
- Innovative Institute of Chinese Medicine and PharmacyShandong University of Traditional Chinese MedicineJinanChina
| | - Yuexiang Ma
- School of Traditional Chinese MedicineShandong University of Traditional Chinese MedicineJinanChina
| | - Peng Sun
- Innovative Institute of Chinese Medicine and PharmacyShandong University of Traditional Chinese MedicineJinanChina
| |
Collapse
|
41
|
Yu S, Jiang H, Yu L, Liu T, Yang C, Cao J, Song Q, Miao Y, Wang W. DTI-ALPS index decreased in patients with Type 2 Diabetes Mellitus. Front Neurosci 2024; 18:1383780. [PMID: 38841097 PMCID: PMC11150564 DOI: 10.3389/fnins.2024.1383780] [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: 03/07/2024] [Accepted: 05/03/2024] [Indexed: 06/07/2024] Open
Abstract
Backgrounds Type 2 Diabetes Mellitus (T2DM) has become a significant global public health issue, characterized by a rising prevalence and associated deficits across multiple organ systems. Our study aims to utilize the DTI-ALPS technique to assess the change of ALPS index in T2DM patients, and to explore whether such changes are correlated with cognition level and diffusion parameters. Methods The study involved 41 patients with T2DM (mean age, 60.49 ± 8.88 years) and 27 healthy controls (mean age, 58.00 ± 7.63 years). All subjects underwent MRI examination, cognitive assessment, and laboratory tests. Tract-based spatial statistics (TBSS) was used to evaluate white matter changes. GLM was performed to check the DTI-ALPS index difference between T2DM and HC groups. Spearman correlation analysis and partial correlation analysis were used to analyze the correlation between the DTI-ALPS index and diffusion properties & cognitive scores. Results The results show that the ALPS index was lower in T2DM patients. MoCA score was significantly correlated with the ALPS index. Patients with T2DM had a significant increase in both mean diffusivity (MD) and radial diffusivity (RD) and decrease in fractional anisotropy (FA) compared to the HC group. Conclusion The results suggest that the ALPS index is decreased in T2DM patients and associates with cognitive level.
Collapse
Affiliation(s)
- Shuncheng Yu
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Hongjun Jiang
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Langxuan Yu
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Tieli Liu
- College of Medical Imaging, Dalian Medical University, Dalian, Liaoning, China
| | - Chun Yang
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Jiajun Cao
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Qingwei Song
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yanwei Miao
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Weiwei Wang
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
42
|
Buscemi F, Torregrossa F, Grasso G. Potential Role of Gut Microbiota in Development of Idiopathic Normal Pressure Hydrocephalus: A Matter or Coincidence? World Neurosurg 2024; 185:307-309. [PMID: 38467373 DOI: 10.1016/j.wneu.2024.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 03/04/2024] [Indexed: 03/13/2024]
Affiliation(s)
- Felice Buscemi
- Section of Neurosurgery, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Fabio Torregrossa
- Section of Neurosurgery, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Giovanni Grasso
- Section of Neurosurgery, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy.
| |
Collapse
|
43
|
He Y, Wang Z, Zuo M, Zhang S, Li W, Chen S, Yuan Y, Yang Y, Liu Y. The impact of neurocognitive and psychiatric disorders on the risk of idiopathic normal pressure hydrocephalus: A bidirectional Mendelian randomization study. Brain Behav 2024; 14:e3532. [PMID: 38779749 PMCID: PMC11112403 DOI: 10.1002/brb3.3532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Neurocognitive and psychiatric disorders have been proved that they can comorbid more often with idiopathic normal pressure hydrocephalus (iNPH) than general population. However, the potential causal association between these disorders and iNPH has not been assessed. Thus, our study aims to investigate the causal relationship between them based on a bidirectional Mendelian randomization (MR) analysis. METHODS Random effects of the inverse variance weighted (IVW) method were conducted to obtain the causal association among the neurocognitive disorders, psychiatric disorders, and iNPH. Genome-wide association studies (GWAS) of 12 neurocognitive and psychiatric disorders were downloaded via the OpenGWAS database, GWAS Catalog, and Psychiatric Genomics Consortium, whereas GWAS data of iNPH were obtained from the FinnGen consortium round 9 release, with 767 cases and 375,610 controls of European ancestry. We also conducted the sensitivity analysis in these significant causal inferences using weighted median model, Cochrane's Q test, MR-Egger regression, MR Pleiotropy Residual Sum and Outlier detect and the leave-one-out analysis. RESULTS For most of the neurocognitive and psychiatric disorders, no causal association was established between them and iNPH. We have found that iNPH (odds ratio [OR] = 1.030, 95% confidence interval [CI]: 1.011-1.048, p = .001) is associated with increased risk for schizophrenia, which failed in validation of sensitivity analysis. Notably, genetically predicted Parkinson's disease (PD) is associated with increased risk of iNPH (OR = 1.256, 95% CI: 1.045-1.511, p = .015). CONCLUSION Our study has revealed the potential causal effect in which PD associated with an increased risk of iNPH. Further study is warranted to investigate the association between PD and iNPH and the potential underlying mechanism.
Collapse
Affiliation(s)
- Yuze He
- Department of NeurosurgeryWest China HospitalSichuan UniversityChengduChina
| | - Zhihao Wang
- Department of NeurosurgeryWest China HospitalSichuan UniversityChengduChina
| | - Mingrong Zuo
- Department of Pediatric NeurosurgeryWest China Women's and Children's Hospital, Sichuan University West China Second University HospitalChengduChina
| | - Shuxin Zhang
- Department of Critical Care MedicineWest China HospitalSichuan UniversityChengduChina
| | - Wenhao Li
- Department of NeurosurgeryWest China HospitalSichuan UniversityChengduChina
| | - Siliang Chen
- Department of NeurosurgeryWest China HospitalSichuan UniversityChengduChina
| | - Yunbo Yuan
- Department of NeurosurgeryWest China HospitalSichuan UniversityChengduChina
| | - Yuan Yang
- Department of NeurosurgeryWest China HospitalSichuan UniversityChengduChina
| | - Yanhui Liu
- Department of NeurosurgeryWest China HospitalSichuan UniversityChengduChina
| |
Collapse
|
44
|
Prasuhn J, Xu J, Hua J, van Zijl P, Knutsson L. Exploring neurodegenerative disorders using advanced magnetic resonance imaging of the glymphatic system. Front Psychiatry 2024; 15:1368489. [PMID: 38651012 PMCID: PMC11033437 DOI: 10.3389/fpsyt.2024.1368489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 03/22/2024] [Indexed: 04/25/2024] Open
Abstract
The glymphatic system, a macroscopic waste clearance system in the brain, is crucial for maintaining neural health. It facilitates the exchange of cerebrospinal and interstitial fluid, aiding the clearance of soluble proteins and metabolites and distributing essential nutrients and signaling molecules. Emerging evidence suggests a link between glymphatic dysfunction and the pathogenesis of neurodegenerative disorders, including Alzheimer's, Parkinson's, and Huntington's disease. These disorders are characterized by the accumulation and propagation of misfolded or mutant proteins, a process in which the glymphatic system is likely involved. Impaired glymphatic clearance could lead to the buildup of these toxic proteins, contributing to neurodegeneration. Understanding the glymphatic system's role in these disorders could provide insights into their pathophysiology and pave the way for new therapeutic strategies. Pharmacological enhancement of glymphatic clearance could reduce the burden of toxic proteins and slow disease progression. Neuroimaging techniques, particularly MRI-based methods, have emerged as promising tools for studying the glymphatic system in vivo. These techniques allow for the visualization of glymphatic flow, providing insights into its function under healthy and pathological conditions. This narrative review highlights current MRI-based methodologies, such as motion-sensitizing pulsed field gradient (PFG) based methods, as well as dynamic gadolinium-based and glucose-enhanced methodologies currently used in the study of neurodegenerative disorders.
Collapse
Affiliation(s)
- Jannik Prasuhn
- Division of Magnetic Resonance (MR) Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- F. M. Kirby Research Center for Functional Brain Imaging, Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD, United States
- Department of Neurology, University Medical Center Schleswig-Holstein, Lübeck, Germany
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
- Center for Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany
| | - Jiadi Xu
- Division of Magnetic Resonance (MR) Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- F. M. Kirby Research Center for Functional Brain Imaging, Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD, United States
| | - Jun Hua
- Division of Magnetic Resonance (MR) Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- F. M. Kirby Research Center for Functional Brain Imaging, Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD, United States
| | - Peter van Zijl
- Division of Magnetic Resonance (MR) Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- F. M. Kirby Research Center for Functional Brain Imaging, Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD, United States
| | - Linda Knutsson
- F. M. Kirby Research Center for Functional Brain Imaging, Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD, United States
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Medical Radiation Physics, Lund University, Lund, Sweden
| |
Collapse
|
45
|
Lopes DM, Wells JA, Ma D, Wallis L, Park D, Llewellyn SK, Ahmed Z, Lythgoe MF, Harrison IF. Glymphatic inhibition exacerbates tau propagation in an Alzheimer's disease model. Alzheimers Res Ther 2024; 16:71. [PMID: 38576025 PMCID: PMC10996277 DOI: 10.1186/s13195-024-01439-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 03/26/2024] [Indexed: 04/06/2024]
Abstract
BACKGROUND The aggregation and spread of misfolded amyloid structured proteins, such as tau and α-synuclein, are key pathological features associated with neurodegenerative disorders, including Alzheimer's and Parkinson's disease. These proteins possess a prion-like property, enabling their transmission from cell to cell leading to propagation throughout the central and peripheral nervous systems. While the mechanisms underlying their intracellular spread are still being elucidated, targeting the extracellular space has emerged as a potential therapeutic approach. The glymphatic system, a brain-wide pathway responsible for clearing extracellular metabolic waste from the central nervous system, has gained attention as a promising target for removing these toxic proteins. METHODS In this study, we investigated the impact of long-term modulation of glymphatic function on tau aggregation and spread by chronically treating a mouse model of tau propagation with a pharmacological inhibitor of AQP4, TGN-020. Thy1-hTau.P301S mice were intracerebrally inoculated with tau into the hippocampus and overlying cortex, and subsequently treated with TGN-020 (3 doses/week, 50 mg/kg TGN-020, i.p.) for 10-weeks. During this time, animal memory was studied using cognitive behavioural tasks, and structural MR images were acquired of the brain in vivo prior to brain extraction for immunohistochemical characterisation. RESULTS Our findings demonstrate increased tau aggregation in the brain and transhemispheric propagation in the hippocampus following the inhibition of glymphatic clearance. Moreover, disruption of the glymphatic system aggravated recognition memory in tau inoculated mice and exacerbated regional changes in brain volume detected in the model. When initiation of drug treatment was delayed for several weeks post-inoculation, the alterations were attenuated. CONCLUSIONS These results indicate that by modulating AQP4 function and, consequently, glymphatic clearance, it is possible to modify the propagation and pathological impact of tau in the brain, particularly during the initial stages of the disease. These findings highlight the critical role of the glymphatic system in preserving healthy brain homeostasis and offer valuable insights into the therapeutic implications of targeting this system for managing neurodegenerative diseases characterized by protein aggregation and spread.
Collapse
Affiliation(s)
- Douglas M Lopes
- Centre for Advanced Biomedical Imaging, Department of Imaging, Division of Medicine, University College London, Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| | - Jack A Wells
- Centre for Advanced Biomedical Imaging, Department of Imaging, Division of Medicine, University College London, Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| | - Da Ma
- Department of Internal Medicine, Section of Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Lauren Wallis
- Centre for Advanced Biomedical Imaging, Department of Imaging, Division of Medicine, University College London, Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| | - Daniel Park
- Centre for Advanced Biomedical Imaging, Department of Imaging, Division of Medicine, University College London, Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| | - Sophie K Llewellyn
- Centre for Advanced Biomedical Imaging, Department of Imaging, Division of Medicine, University College London, Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| | - Zeshan Ahmed
- Neuroscience Next Generation Therapeutics (NGTx), Eli Lilly and Company, Cambridge, MA, USA
| | - Mark F Lythgoe
- Centre for Advanced Biomedical Imaging, Department of Imaging, Division of Medicine, University College London, Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| | - Ian F Harrison
- Centre for Advanced Biomedical Imaging, Department of Imaging, Division of Medicine, University College London, Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK.
| |
Collapse
|
46
|
Edison P. Astroglial activation: Current concepts and future directions. Alzheimers Dement 2024; 20:3034-3053. [PMID: 38305570 PMCID: PMC11032537 DOI: 10.1002/alz.13678] [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: 08/24/2023] [Revised: 11/27/2023] [Accepted: 12/11/2023] [Indexed: 02/03/2024]
Abstract
Astrocytes are abundantly and ubiquitously expressed cell types with diverse functions throughout the central nervous system. Astrocytes show remarkable plasticity and exhibit morphological, molecular, and functional remodeling in response to injury, disease, or infection of the central nervous system, as evident in neurodegenerative diseases. Astroglial mediated inflammation plays a prominent role in the pathogenesis of neurodegenerative diseases. This review focus on the role of astrocytes as essential players in neuroinflammation and discuss their morphological and functional heterogeneity in the normal central nervous system and explore the spatial and temporal variations in astroglial phenotypes observed under different disease conditions. This review discusses the intimate relationship of astrocytes to pathological hallmarks of neurodegenerative diseases. Finally, this review considers the putative therapeutic strategies that can be deployed to modulate the astroglial functions in neurodegenerative diseases. HIGHLIGHTS: Astroglia mediated neuroinflammation plays a key role in the pathogenesis of neurodegenerative diseases. Activated astrocytes exhibit diverse phenotypes in a region-specific manner in brain and interact with β-amyloid, tau, and α-synuclein species as well as with microglia and neuronal circuits. Activated astrocytes are likely to influence the trajectory of disease progression of neurodegenerative diseases, as determined by the stage of disease, individual susceptibility, and state of astroglial priming. Modulation of astroglial activation may be a therapeutic strategy at various stages in the trajectory of neurodegenerative diseases to modify the disease course.
Collapse
Affiliation(s)
- Paul Edison
- Division of NeurologyDepartment of Brain SciencesFaculty of Medicine, Imperial College LondonLondonUK
- Division of Psychological medicine and clinical neurosciencesSchool of Medicine, Cardiff UniversityWalesUK
| |
Collapse
|
47
|
Wen Q, Wang H, Haacke EM, Jiang Q, Hu J. Contribution of Direct Cerebral Vascular Transport in Brain Substance Clearance. Aging Dis 2024; 15:584-600. [PMID: 37611901 PMCID: PMC10917538 DOI: 10.14336/ad.2023.0426] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 04/26/2023] [Indexed: 08/25/2023] Open
Abstract
The accumulation of harmful substances has long been recognized as a likely cause of many neurodegenerative diseases. The two classic brain clearance pathways are cerebrospinal fluid (CSF) and vascular circulation systems. Since the discovery of the glymphatic system, research on the CSF pathway has gained momentum, and impaired CSF clearance has been implicated in virtually all neurodegenerative animal models. However, the contribution of the direct participation of vascular transport across the blood-brain barrier in clearing substances is often ignored in glymphatic papers. Supportive evidence for the direct involvement of parenchymal vasculature in substance clearance is accumulated. First, multiple mechanisms have been proposed for the vascular drainage of exogenous and endogenous substances across the blood-brain barriers. Second, the "traditional" role of arachnoid villi and granulations as the main site for CSF draining into the vasculature system has been questioned. Third, MRI studies using different CSF tracers indicate that parenchymal vasculature directly participates in tracer efflux, consistent with immunohistochemical findings. Here we will review evidence in the literature that supports the direct participation of the parenchymal vascular system in substance clearance, in addition to the CSF clearance pathways.
Collapse
Affiliation(s)
- Qiuting Wen
- Department of Radiology and Imaging Sciences, Indiana University, Indianapolis, IN, USA.
| | - Haoyu Wang
- Beijing Institute of Radiation Medicine, Beijing, China.
| | - E. Mark Haacke
- Department of Radiology, Wayne State University, Detroit, MI 48201 USA.
| | - Quan Jiang
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202 USA.
| | - Jiani Hu
- Department of Radiology, Wayne State University, Detroit, MI 48201 USA.
| |
Collapse
|
48
|
Kaur J, Boyd ED, Ding G, Zhang L, Luo H, Li Q, Li L, Wei M, Landschoot-Ward J, Chopp M, Zhang Z, Jiang Q. The Association between Glymphatic System and Perivascular Macrophages in Brain Waste Clearance. Diagnostics (Basel) 2024; 14:731. [PMID: 38611644 PMCID: PMC11011895 DOI: 10.3390/diagnostics14070731] [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: 01/17/2024] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 04/14/2024] Open
Abstract
The glymphatic system suggests the convective bulk flow of cerebrospinal fluid (CSF) through perivascular spaces and the interstitial spaces of the brain parenchyma for the rapid removal of toxic waste solutes from the brain. However, the presence of convective bulk flow within the brain interstitial spaces is still under debate. We first addressed this argument to determine the involvement of the glymphatic system in brain waste clearance utilizing contrast-enhanced 3D T1-weighted imaging (T1WI), diffusion tensor imaging (DTI), and confocal microscopy imaging. Furthermore, perivascular macrophages (PVMs), which are immune cells located within perivascular spaces, have not been thoroughly explored for their association with the glymphatic system. Therefore, we investigated tracer uptake by PVMs in the perivascular spaces of both the arteries/arterioles and veins/venules and the potential association of PVMs in assisting the glymphatic system for interstitial waste clearance. Our findings demonstrated that both convective bulk flow and diffusion are responsible for the clearance of interstitial waste solutes from the brain parenchyma. Furthermore, our results suggested that PVMs may play an important function in glymphatic system-mediated interstitial waste clearance. The glymphatic system and PVMs could be targeted to enhance interstitial waste clearance in patients with waste-associated neurological conditions and aging.
Collapse
Affiliation(s)
- Jasleen Kaur
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (J.K.); (E.D.B.); (G.D.); (L.Z.); (H.L.); (Q.L.); (L.L.); (M.W.); (J.L.-W.); (M.C.); (Z.Z.)
- Department of Physics, Oakland University, Rochester, MI 48309, USA
| | - Edward D. Boyd
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (J.K.); (E.D.B.); (G.D.); (L.Z.); (H.L.); (Q.L.); (L.L.); (M.W.); (J.L.-W.); (M.C.); (Z.Z.)
- Department of Radiology, Michigan State University, East Lansing, MI 48824, USA
| | - Guangliang Ding
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (J.K.); (E.D.B.); (G.D.); (L.Z.); (H.L.); (Q.L.); (L.L.); (M.W.); (J.L.-W.); (M.C.); (Z.Z.)
- Department of Radiology, Michigan State University, East Lansing, MI 48824, USA
| | - Li Zhang
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (J.K.); (E.D.B.); (G.D.); (L.Z.); (H.L.); (Q.L.); (L.L.); (M.W.); (J.L.-W.); (M.C.); (Z.Z.)
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | - Hao Luo
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (J.K.); (E.D.B.); (G.D.); (L.Z.); (H.L.); (Q.L.); (L.L.); (M.W.); (J.L.-W.); (M.C.); (Z.Z.)
| | - Qingjiang Li
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (J.K.); (E.D.B.); (G.D.); (L.Z.); (H.L.); (Q.L.); (L.L.); (M.W.); (J.L.-W.); (M.C.); (Z.Z.)
| | - Lian Li
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (J.K.); (E.D.B.); (G.D.); (L.Z.); (H.L.); (Q.L.); (L.L.); (M.W.); (J.L.-W.); (M.C.); (Z.Z.)
| | - Min Wei
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (J.K.); (E.D.B.); (G.D.); (L.Z.); (H.L.); (Q.L.); (L.L.); (M.W.); (J.L.-W.); (M.C.); (Z.Z.)
| | - Julie Landschoot-Ward
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (J.K.); (E.D.B.); (G.D.); (L.Z.); (H.L.); (Q.L.); (L.L.); (M.W.); (J.L.-W.); (M.C.); (Z.Z.)
| | - Michael Chopp
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (J.K.); (E.D.B.); (G.D.); (L.Z.); (H.L.); (Q.L.); (L.L.); (M.W.); (J.L.-W.); (M.C.); (Z.Z.)
- Department of Physics, Oakland University, Rochester, MI 48309, USA
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
- Department of Neurology, Wayne State University, Detroit, MI 48202, USA
| | - Zhenggang Zhang
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (J.K.); (E.D.B.); (G.D.); (L.Z.); (H.L.); (Q.L.); (L.L.); (M.W.); (J.L.-W.); (M.C.); (Z.Z.)
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
- Department of Neurology, Wayne State University, Detroit, MI 48202, USA
| | - Quan Jiang
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (J.K.); (E.D.B.); (G.D.); (L.Z.); (H.L.); (Q.L.); (L.L.); (M.W.); (J.L.-W.); (M.C.); (Z.Z.)
- Department of Physics, Oakland University, Rochester, MI 48309, USA
- Department of Radiology, Michigan State University, East Lansing, MI 48824, USA
- Department of Neurology, Wayne State University, Detroit, MI 48202, USA
| |
Collapse
|
49
|
Colvee-Martin H, Parra JR, Gonzalez GA, Barker W, Duara R. Neuropathology, Neuroimaging, and Fluid Biomarkers in Alzheimer's Disease. Diagnostics (Basel) 2024; 14:704. [PMID: 38611617 PMCID: PMC11012058 DOI: 10.3390/diagnostics14070704] [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: 01/11/2024] [Revised: 02/05/2024] [Accepted: 02/17/2024] [Indexed: 04/14/2024] Open
Abstract
An improved understanding of the pathobiology of Alzheimer's disease (AD) should lead ultimately to an earlier and more accurate diagnosis of AD, providing the opportunity to intervene earlier in the disease process and to improve outcomes. The known hallmarks of Alzheimer's disease include amyloid-β plaques and neurofibrillary tau tangles. It is now clear that an imbalance between production and clearance of the amyloid beta protein and related Aβ peptides, especially Aβ42, is a very early, initiating factor in Alzheimer's disease (AD) pathogenesis, leading to aggregates of hyperphosphorylation and misfolded tau protein, inflammation, and neurodegeneration. In this article, we review how the AD diagnostic process has been transformed in recent decades by our ability to measure these various elements of the pathological cascade through the use of imaging and fluid biomarkers. The more recently developed plasma biomarkers, especially phosphorylated-tau217 (p-tau217), have utility for screening and diagnosis of the earliest stages of AD. These biomarkers can also be used to measure target engagement by disease-modifying therapies and the response to treatment.
Collapse
Affiliation(s)
- Helena Colvee-Martin
- Wien Center for Alzheimer’s Disease & Memory Disorders, Mount Sinai Medical Center, Miami Beach, FL 33140, USA; (H.C.-M.); (W.B.)
| | - Juan Rayo Parra
- Human & Molecular Genetics, Florida International University, Miami, FL 33199, USA; (J.R.P.); (G.A.G.)
| | - Gabriel Antonio Gonzalez
- Human & Molecular Genetics, Florida International University, Miami, FL 33199, USA; (J.R.P.); (G.A.G.)
| | - Warren Barker
- Wien Center for Alzheimer’s Disease & Memory Disorders, Mount Sinai Medical Center, Miami Beach, FL 33140, USA; (H.C.-M.); (W.B.)
| | - Ranjan Duara
- Wien Center for Alzheimer’s Disease & Memory Disorders, Mount Sinai Medical Center, Miami Beach, FL 33140, USA; (H.C.-M.); (W.B.)
| |
Collapse
|
50
|
Castaneyra-Ruiz L, Ledbetter J, Lee S, Rangel A, Torres E, Romero B, Muhonen M. Intraventricular dimethyl sulfoxide (DMSO) induces hydrocephalus in a dose-dependent pattern. Heliyon 2024; 10:e27295. [PMID: 38486744 PMCID: PMC10937698 DOI: 10.1016/j.heliyon.2024.e27295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/17/2024] Open
Abstract
Introduction Dimethyl sulfoxide (DMSO), a widely utilized solvent in the medical industry, has been associated with various adverse effects, even at low concentrations, including damage to mitochondrial integrity, altered membrane potentials, caspase activation, and apoptosis. Notably, therapeutic molecules for central nervous system treatments, such as embolic agents or some chemotherapy drugs that are dissolved in DMSO, have been associated with hydrocephalus as a secondary complication. Our study investigated the potential adverse effects of DMSO on the brain, specifically focusing on the development of hydrocephalus and the effect on astrocytes. Methods Varied concentrations of DMSO were intraventricularly injected into 3-day-old mice, and astrocyte cultures were exposed to similar concentrations of DMSO. After 14 days of injection, magnetic resonance imaging (MRI) was employed to quantify the brain ventricular volumes in mice. Immunofluorescence analysis was conducted to delineate DMSO-dependent effects in the brain. Additionally, astrocyte cultures were utilized to assess astrocyte viability and the effects of cellular apoptosis. Results Our findings revealed a dose-dependent induction of ventriculomegaly in mice with 2%, 10%, and 100% DMSO injections (p < 0.001). The ciliated cells of the ventricles were also proportionally affected by DMSO concentration (p < 0.0001). Furthermore, cultured astrocytes exhibited increased apoptosis after DMSO exposure (p < 0.001). Conclusion Our study establishes that intraventricular administration of DMSO induces hydrocephalus in a dose-dependent manner. This observation sheds light on a potential explanation for the occurrence of hydrocephalus as a secondary complication in intracranial treatments utilizing DMSO as a solvent.
Collapse
Affiliation(s)
| | | | - Seunghyun Lee
- CHOC Children's Research Institute, Orange, CA, 92868, USA
| | - Anthony Rangel
- CHOC Children's Research Institute, Orange, CA, 92868, USA
| | - Evelyn Torres
- CHOC Children's Research Institute, Orange, CA, 92868, USA
| | - Bianca Romero
- Neurosurgery Department at CHOC Children's Hospital, Orange, CA, 92868, USA
| | - Michael Muhonen
- Neurosurgery Department at CHOC Children's Hospital, Orange, CA, 92868, USA
| |
Collapse
|