51
|
González-Hernández S, Mukouyama YS. Lymphatic vasculature in the central nervous system. Front Cell Dev Biol 2023; 11:1150775. [PMID: 37091974 PMCID: PMC10119411 DOI: 10.3389/fcell.2023.1150775] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 03/28/2023] [Indexed: 04/25/2023] Open
Abstract
The central nervous system (CNS) is considered as an immune privilege organ, based on experiments in the mid 20th century showing that the brain fails to mount an efficient immune response against an allogeneic graft. This suggests that in addition to the presence of the blood-brain barrier (BBB), the apparent absence of classical lymphatic vasculature in the CNS parenchyma limits the capacity for an immune response. Although this view is partially overturned by the recent discovery of the lymphatic-like hybrid vessels in the Schlemm's canal in the eye and the lymphatic vasculature in the outmost layer of the meninges, the existence of lymphatic vessels in the CNS parenchyma has not been reported. Two potential mechanisms by which lymphatic vasculature may arise in the organs are: 1) sprouting and invasion of lymphatic vessels from the surrounding tissues into the parenchyma and 2) differentiation of blood endothelial cells into lymphatic endothelial cells in the parenchyma. Considering these mechanisms, we here discuss what causes the dearth of lymphatic vessels specifically in the CNS parenchyma.
Collapse
|
52
|
Moses J, Sinclair B, Law M, O'Brien TJ, Vivash L. Automated Methods for Detecting and Quantitation of Enlarged Perivascular spaces on MRI. J Magn Reson Imaging 2023; 57:11-24. [PMID: 35866259 PMCID: PMC10083963 DOI: 10.1002/jmri.28369] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/06/2022] [Accepted: 07/08/2022] [Indexed: 02/03/2023] Open
Abstract
The brain's glymphatic system is a network of intracerebral vessels that function to remove "waste products" such as degraded proteins from the brain. It comprises of the vasculature, perivascular spaces (PVS), and astrocytes. Poor glymphatic function has been implicated in numerous diseases; however, its contribution is still unknown. Efforts have been made to image the glymphatic system to further assess its role in the pathogenesis of different diseases. Numerous imaging modalities have been utilized including two-photon microscopy and contrast-enhanced magnetic resonance imaging (MRI). However, these are associated with limitations for clinical use. PVS form a part of the glymphatic system and can be visualized on standard MRI sequences when enlarged. It is thought that PVS become enlarged secondary to poor glymphatic drainage of metabolites. Thus, quantitating PVS could be a good surrogate marker for glymphatic function. Numerous manual rating scales have been developed to measure the PVS number and size on MRI scans; however, these are associated with many limitations. Instead, automated methods have been created to measure PVS more accurately in different diseases. In this review, we discuss the imaging techniques currently available to visualize the glymphatic system as well as the automated methods currently available to measure PVS, and the strengths and limitations associated with each technique. EVIDENCE LEVEL: 1 TECHNICAL EFFICACY: Stage 1.
Collapse
Affiliation(s)
- Jasmine Moses
- Department of Neurosciences, Central Clinical School, Monash University, Melbourne, Australia
| | - Ben Sinclair
- Department of Neurosciences, Central Clinical School, Monash University, Melbourne, Australia.,Department of Neurology, Alfred Hospital, Melbourne, Australia.,Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Melbourne, Australia
| | - Meng Law
- Department of Neurosciences, Central Clinical School, Monash University, Melbourne, Australia.,Department of Radiology, Alfred Health, Melbourne, Victoria, Australia.,Department of Electrical and Computer Systems Engineering, Monash University, Melbourne, Victoria, Australia
| | - Terence J O'Brien
- Department of Neurosciences, Central Clinical School, Monash University, Melbourne, Australia.,Department of Neurology, Alfred Hospital, Melbourne, Australia.,Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Melbourne, Australia.,Department of Neurology, Royal Melbourne Hospital, University of Melbourne, Victoria, Australia
| | - Lucy Vivash
- Department of Neurosciences, Central Clinical School, Monash University, Melbourne, Australia.,Department of Neurology, Alfred Hospital, Melbourne, Australia.,Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Melbourne, Australia.,Department of Neurology, Royal Melbourne Hospital, University of Melbourne, Victoria, Australia
| |
Collapse
|
53
|
Chachaj A, Gąsiorowski K, Szuba A, Sieradzki A, Leszek J. The Lymphatic System In The Brain Clearance Mechanisms - New Therapeutic Perspectives For Alzheimer's Disease. Curr Neuropharmacol 2023; 21:380-391. [PMID: 35410605 PMCID: PMC10190136 DOI: 10.2174/1570159x20666220411091332] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 03/05/2022] [Accepted: 04/06/2022] [Indexed: 11/22/2022] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia worldwide. Pathological deposits of neurotoxic proteins within the brain, such as amyloid-ß and hyperphosphorylated tau tangles, are the prominent features in AD. According to recent studies, the newly discovered brain lymphatic system was demonstrated to be crucial in the clearance of metabolic macromolecules from the brain. Meningeal lymphatic vessels located in the dura mater drain the fluid, macromolecules, and immune cells from cerebrospinal fluid (CSF) and transport them, as lymph, to the deep cervical lymph nodes. The lymphatic system provides the perivascular exchange of CSF with interstitial fluid (ISF) and ensures the homeostasis of neuronal interstitial space. In this review, we aim to summarize recent findings on the role of the lymphatic system in AD pathophysiology and discuss possible therapeutic perspectives, targeting the lymphatic clearance mechanisms within the brain.
Collapse
Affiliation(s)
- Angelika Chachaj
- Department of Angiology, Hypertension and Diabetology, Wroclaw Medical University, Wroclaw, Poland
| | | | - Andrzej Szuba
- Department of Angiology, Hypertension and Diabetology, Wroclaw Medical University, Wroclaw, Poland
| | - Adrian Sieradzki
- Department of Nervous System Diseases, Wroclaw Medical University, Wroclaw, Poland
| | - Jerzy Leszek
- Department of Psychiatry, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
54
|
Guo X, Zhang G, Peng Q, Huang L, Zhang Z, Zhang Z. Emerging Roles of Meningeal Lymphatic Vessels in Alzheimer's Disease. J Alzheimers Dis 2023; 94:S355-S366. [PMID: 36683509 PMCID: PMC10473149 DOI: 10.3233/jad-221016] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/17/2022] [Indexed: 01/22/2023]
Abstract
Meningeal lymphatic vessels (mLVs), the functional lymphatic system present in the meninges, are the key drainage route responsible for the clearance of molecules, immune cells, and cellular debris from the cerebrospinal fluid and interstitial fluid into deep cervical lymph nodes. Aging and ApoE4, the two most important risk factors for Alzheimer's disease (AD), induce mLV dysfunction, decrease cerebrospinal fluid influx and outflux, and exacerbate amyloid pathology and cognitive dysfunction. Dysfunction of mLVs results in the deposition of metabolic products, accelerates neuroinflammation, and promotes the release of pro-inflammatory cytokines in the brain. Thus, mLVs represent a novel therapeutic target for treating neurodegenerative and neuroinflammatory diseases. This review aims to summarize the structure and function of mLVs and to discuss the potential effect of aging and ApoE4 on mLV dysfunction, as well as their roles in the pathogenesis of AD.
Collapse
Affiliation(s)
- Xiaodi Guo
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Guoxin Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qinyu Peng
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Liqin Huang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhaohui Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
55
|
Jiang H, Wei H, Zhou Y, Xiao X, Zhou C, Ji X. Overview of the meningeal lymphatic vessels in aging and central nervous system disorders. Cell Biosci 2022; 12:202. [PMID: 36528776 PMCID: PMC9759913 DOI: 10.1186/s13578-022-00942-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
In the aging process and central nervous system (CNS) diseases, the functions of the meningeal lymphatic vessels (MLVs) are impaired. Alterations in MLVs have been observed in aging-related neurodegenerative diseases, brain tumors, and even cerebrovascular disease. These findings reveal a new perspective on aging and CNS disorders and provide a promising therapeutic target. Additionally, recent neuropathological studies have shown that MLVs exchange soluble components between the cerebrospinal fluid (CSF) and interstitial fluid (ISF) and drain metabolites, cellular debris, misfolded proteins, and immune cells from the CSF into the deep cervical lymph nodes (dCLNs), directly connecting the brain with the peripheral circulation. Impairment and dysfunction of meningeal lymphatics can lead to the accumulation of toxic proteins in the brain, exacerbating the progression of neurological disorders. However, for many CNS diseases, the causal relationship between MLVs and neuropathological changes is not fully clear. Here, after a brief historical retrospection, we review recent discoveries about the hallmarks of MLVs and their roles in the aging and CNS diseases, as well as potential therapeutic targets for the treatment of neurologic diseases.
Collapse
Affiliation(s)
- Huimin Jiang
- grid.24696.3f0000 0004 0369 153XBeijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing, 100069 China
| | - Huimin Wei
- grid.64939.310000 0000 9999 1211Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191 China
| | - Yifan Zhou
- grid.24696.3f0000 0004 0369 153XBeijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing, 100069 China
| | - Xuechun Xiao
- grid.64939.310000 0000 9999 1211Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191 China
| | - Chen Zhou
- grid.24696.3f0000 0004 0369 153XBeijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing, 100069 China
| | - Xunming Ji
- grid.24696.3f0000 0004 0369 153XBeijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing, 100069 China ,grid.24696.3f0000 0004 0369 153XDepartment of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053 China
| |
Collapse
|
56
|
Brain borders at the central stage of neuroimmunology. Nature 2022; 612:417-429. [PMID: 36517712 DOI: 10.1038/s41586-022-05474-7] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 10/24/2022] [Indexed: 12/16/2022]
Abstract
The concept of immune privilege suggests that the central nervous system is isolated from the immune system. However, recent studies have highlighted the borders of the central nervous system as central sites of neuro-immune interactions. Although the nervous and immune systems both function to maintain homeostasis, under rare circumstances, they can develop pathological interactions that lead to neurological or psychiatric diseases. Here we discuss recent findings that dissect the key anatomical, cellular and molecular mechanisms that enable neuro-immune responses at the borders of the brain and spinal cord and the implications of these interactions for diseases of the central nervous system.
Collapse
|
57
|
Xiang T, Feng D, Zhang X, Chen Y, Wang H, Liu X, Gong Z, Yuan J, Liu M, Sha Z, Lv C, Jiang W, Nie M, Fan Y, Wu D, Dong S, Feng J, Ponomarev ED, Zhang J, Jiang R. Effects of increased intracranial pressure on cerebrospinal fluid influx, cerebral vascular hemodynamic indexes, and cerebrospinal fluid lymphatic efflux. J Cereb Blood Flow Metab 2022; 42:2287-2302. [PMID: 35962479 PMCID: PMC9670008 DOI: 10.1177/0271678x221119855] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 06/22/2022] [Accepted: 06/23/2022] [Indexed: 12/14/2022]
Abstract
The glymphatic-lymphatic fluid transport system (GLFTS) consists of glymphatic pathway and cerebrospinal fluid (CSF) lymphatic outflow routes, allowing biological liquids from the brain parenchyma to access the CSF along with perivascular space and to be cleaned out of the skull through lymphatic vessels. It is known that increased local pressure due to physical compression of tissue improves lymphatic transport in peripheral organs, but little is known about the exact relationship between increased intracranial pressure (IICP) and GLFTS. In this study, we verify our hypothesis that IICP significantly impacts GLFTS, and this effect depends on severity of the IICP. Using a previously developed inflating balloon model to induce IICP and inject fluorescent tracers into the cisterna magna, we found significant impairment of the glymphatic circulation after IICP. We further found that cerebrovascular occlusion occurred, and cerebrovascular pulsation decreased after IICP. IICP also interrupted the drainage of deep cervical lymph nodes and dorsal meningeal lymphatic function, enhancing spinal lymphatic outflow to the sacral lymph nodes. Notably, these effects were associated with the severity of IICP. Thus, our findings proved that the intensity of IICP significantly impacts GLFTS. This may have translational applications for preventing and treating related neurological disorders.
Collapse
Affiliation(s)
- Tangtang Xiang
- 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 City, Tianjin, China
| | - Dongyi 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 City, Tianjin, China
| | - Xinjie Zhang
- 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 City, Tianjin, 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 City, Tianjin, China
| | - Hanhua Wang
- 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 City, Tianjin, China
| | - Xuanhui 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 City, Tianjin, China
| | - Zhitao Gong
- 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 City, 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 City, 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 City, 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 City, Tianjin, China
| | - Chuanxiang Lv
- Department of Neurosurgery, The First Clinical Hospital, Jilin
University, Changchun, 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 City, Tianjin, 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 City, Tianjin, China
| | - Yibing Fan
- 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 City, Tianjin, China
| | - Di Wu
- 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 City, Tianjin, China
| | - 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 City, 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 City, Tianjin, China
| | - Eugene D Ponomarev
- School of Biomedical Sciences, Faculty of Medicine, The Chinese
University of Hong Kong, Shatin, NT, Hong Kong, China
| | - Jianning Zhang
- 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 City, 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 City, Tianjin, China
| |
Collapse
|
58
|
Hier DB, Azizi S, Thimgan MS, Wunsch DC. Tau kinetics in Alzheimer's disease. Front Aging Neurosci 2022; 14:1055170. [PMID: 36437992 PMCID: PMC9682289 DOI: 10.3389/fnagi.2022.1055170] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 10/27/2022] [Indexed: 07/20/2023] Open
Abstract
The cytoskeletal protein tau is implicated in the pathogenesis of Alzheimer's disease which is characterized by intra-neuronal neurofibrillary tangles containing abnormally phosphorylated insoluble tau. Levels of soluble tau are elevated in the brain, the CSF, and the plasma of patients with Alzheimer's disease. To better understand the causes of these elevated levels of tau, we propose a three-compartment kinetic model (brain, CSF, and plasma). The model assumes that the synthesis of tau follows zero-order kinetics (uncorrelated with compartmental tau levels) and that the release, absorption, and clearance of tau is governed by first-order kinetics (linearly related to compartmental tau levels). Tau that is synthesized in the brain compartment can be released into the interstitial fluid, catabolized, or retained in neurofibrillary tangles. Tau released into the interstitial fluid can mix with the CSF and eventually drain to the plasma compartment. However, losses of tau in the drainage pathways may be significant. The kinetic model estimates half-life of tau in each compartment (552 h in the brain, 9.9 h in the CSF, and 10 h in the plasma). The kinetic model predicts that an increase in the neuronal tau synthesis rate or a decrease in tau catabolism rate best accounts for observed increases in tau levels in the brain, CSF, and plasma found in Alzheimer's disease. Furthermore, the model predicts that increases in brain half-life of tau in Alzheimer's disease should be attributed to decreased tau catabolism and not to increased tau synthesis. Most clearance of tau in the neuron occurs through catabolism rather than release to the CSF compartment. Additional experimental data would make ascertainment of the model parameters more precise.
Collapse
Affiliation(s)
- Daniel B. Hier
- Applied Computational Intelligence Laboratory, Department of Electrical & Computer Engineering, Missouri University of Science & Technology, Rolla, MO, United States
- Department of Neurology and Rehabilitation, University of Illinois at Chicago, Chicago, IL, United States
| | - Sima Azizi
- Applied Computational Intelligence Laboratory, Department of Electrical & Computer Engineering, Missouri University of Science & Technology, Rolla, MO, United States
| | - Matthew S. Thimgan
- Department of Biological Sciences, Missouri University of Science & Technology, Rolla, MO, United States
| | - Donald C. Wunsch
- Applied Computational Intelligence Laboratory, Department of Electrical & Computer Engineering, Missouri University of Science & Technology, Rolla, MO, United States
- ECCS Division, National Science Foundation, Alexandria, VA, United States
| |
Collapse
|
59
|
Zhao L, Tannenbaum A, Bakker ENTP, Benveniste H. Physiology of Glymphatic Solute Transport and Waste Clearance from the Brain. Physiology (Bethesda) 2022; 37:0. [PMID: 35881783 PMCID: PMC9550574 DOI: 10.1152/physiol.00015.2022] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/12/2022] [Accepted: 07/20/2022] [Indexed: 12/25/2022] Open
Abstract
This review focuses on the physiology of glymphatic solute transport and waste clearance, using evidence from experimental animal models as well as from human studies. Specific topics addressed include the biophysical characteristics of fluid and solute transport in the central nervous system, glymphatic-lymphatic coupling, as well as the role of cerebrospinal fluid movement for brain waste clearance. We also discuss the current understanding of mechanisms underlying increased waste clearance during sleep.
Collapse
Affiliation(s)
- Lucy Zhao
- Department of Anesthesiology, Yale School of Medicine, New Haven, Connecticut
| | - Allen Tannenbaum
- Departments of Computer Science and Applied Mathematics and Statistics, Stony Brook University, Stony Brook, New York
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York City, New York
| | - Erik N T P Bakker
- Department of Biomedical Engineering and Physics, Amsterdam UMC, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Helene Benveniste
- Department of Anesthesiology, Yale School of Medicine, New Haven, Connecticut
- Department of Biomedical Engineering, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
60
|
Lampejo AO, Jo M, Murfee WL, Breslin JW. The Microvascular-Lymphatic Interface and Tissue Homeostasis: Critical Questions That Challenge Current Understanding. J Vasc Res 2022; 59:327-342. [PMID: 36315992 PMCID: PMC9780194 DOI: 10.1159/000525787] [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: 03/22/2022] [Accepted: 06/20/2022] [Indexed: 11/07/2022] Open
Abstract
Lymphatic and blood microvascular networks play critical roles in the clearance of excess fluid from local tissue spaces. Given the importance of these dynamics in inflammation, tumor metastasis, and lymphedema, understanding the coordinated function and remodeling between lymphatic and blood vessels in adult tissues is necessary. Knowledge gaps exist because the functions of these two systems are typically considered separately. The objective of this review was to highlight the coordinated functional relationships between blood and lymphatic vessels in adult microvascular networks. Structural, functional, temporal, and spatial relationships will be framed in the context of maintaining tissue homeostasis, vessel permeability, and system remodeling. The integration across systems will emphasize the influence of the local environment on cellular and molecular dynamics involved in fluid flow from blood capillaries to initial lymphatic vessels in microvascular networks.
Collapse
Affiliation(s)
- Arinola O. Lampejo
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, 32611, USA
| | - Michiko Jo
- Division of Presymptomatic Disease, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Walter L. Murfee
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, 32611, USA
| | - Jerome W. Breslin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA
| |
Collapse
|
61
|
Moses J, Sinclair B, Schwartz DL, Silbert LC, O’Brien TJ, Law M, Vivash L. Perivascular spaces as a marker of disease severity and neurodegeneration in patients with behavioral variant frontotemporal dementia. Front Neurosci 2022; 16:1003522. [PMID: 36340772 PMCID: PMC9633276 DOI: 10.3389/fnins.2022.1003522] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 10/06/2022] [Indexed: 11/19/2022] Open
Abstract
Background Behavioural Variant Frontotemporal Dementia (bvFTD) is a rapidly progressing neurodegenerative proteinopathy. Perivascular spaces (PVS) form a part of the brain’s glymphatic clearance system. When enlarged due to poor glymphatic clearance of toxic proteins, PVS become larger and more conspicuous on MRI. Therefore, enlarged PVS may be a useful biomarker of disease severity and progression in neurodegenerative proteinopathies such as bvFTD. This study aimed to determine the utility of PVS as a biomarker of disease progression in patients with bvFTD. Materials and methods Serial baseline and week 52 MRIs acquired from ten patients with bvFTD prospectively recruited and followed in a Phase 1b open label trial of sodium selenate for bvFTD were used in this study. An automated algorithm quantified PVS on MRI, which was visually inspected and validated by a member of the study team. The number and volume of PVS were extracted and mixed models used to assess the relationship between PVS burden and other measures of disease (cognition, carer burden scale, protein biomarkers). Additional exploratory analysis investigated PVS burden in patients who appeared to not progress over the 12 months of selenate treatment (i.e., “non-progressors”). Results Overall, PVS cluster number (ß = −3.27, CI [−7.80 – 1.27], p = 0.267) and PVS volume (ß = −36.8, CI [−84.9 – 11.3], p = 0.171) did not change over the paired MRI scans 12 months apart. There was association between cognition total composite scores and the PVS burden (PVS cluster ß = −0.802e–3, CI [9.45e–3 – −6.60e–3, p ≤ 0.001; PVS volume ß = −1.30e–3, CI [−1.55e–3 – −1.05e–3], p ≤ 0.001), as well as between the change in the cognition total composite score and the change in PVS volume (ß = 4.36e–3, CI [1.33e–3 – 7.40e–3], p = 0.046) over the trial period. There was a significant association between CSF t-tau and the number of PVS clusters (ß = 2.845, CI [0.630 – 5.06], p = 0.036). Additionally, there was a significant relationship between the change in CSF t-tau and the change in the number of PVS (ß = 1.54, CI [0.918 – 2.16], p < 0.001) and PVS volume (ß = 13.8, CI [6.37 – 21.1], p = 0.003) over the trial period. An association was found between the change in NfL and the change in PVS volume (ß = 1.40, CI [0.272 – 2.52], p = 0.045) over time. Within the “non-progressor” group (n = 7), there was a significant relationship between the change in the CSF total-tau (t-tau) levels and the change in the PVS burden (PVS cluster (ß = 1.46, CI [0.577 – 2.34], p = 0.014; PVS volume ß = 14.6, CI [3.86 – 25.4], p = 0.032) over the trial period. Additionally, there was evidence of a significant relationship between the change in NfL levels and the change in the PVS burden over time (PVS cluster ß = 0.296, CI [0.229 – 0.361], p ≤ 0.001; PVS volume ß = 3.67, CI [2.42 – 4.92], p = 0.002). Conclusion Analysis of serial MRI scans 12 months apart in patients with bvFTD demonstrated a relationship between PVS burden and disease severity as measured by the total cognitive composite score and CSF t-tau. Further studies are needed to confirm PVS as a robust marker of neurodegeneration in proteinopathies.
Collapse
Affiliation(s)
- Jasmine Moses
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Benjamin Sinclair
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Neurology, Alfred Hospital, Melbourne, VIC, Australia
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Melbourne, VIC, Australia
| | - Daniel L. Schwartz
- NIA-Layton Oregon Aging and Alzheimer’s Disease Research Center, Oregon Health & Science University, Portland, OR, United States
- Advanced Imaging Research Center, Oregon Health and Science University, Portland, OR, United States
| | - Lisa C. Silbert
- NIA-Layton Oregon Aging and Alzheimer’s Disease Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Portland Veterans Affairs Health Care System, Portland, OR, United States
| | - Terence J. O’Brien
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Neurology, Alfred Hospital, Melbourne, VIC, Australia
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Melbourne, VIC, Australia
- Department of Neurology, Royal Melbourne Hospital, University of Melbourne, Melbourne, VIC, Australia
| | - Meng Law
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Radiology, Alfred Health, Melbourne, VIC, Australia
- Department of Electrical and Computer Systems Engineering, Monash University, Melbourne, VIC, Australia
| | - Lucy Vivash
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Neurology, Alfred Hospital, Melbourne, VIC, Australia
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Melbourne, VIC, Australia
- Department of Neurology, Royal Melbourne Hospital, University of Melbourne, Melbourne, VIC, Australia
- *Correspondence: Lucy Vivash,
| |
Collapse
|
62
|
Xu JQ, Liu QQ, Huang SY, Duan CY, Lu HB, Cao Y, Hu JZ. The lymphatic system: a therapeutic target for central nervous system disorders. Neural Regen Res 2022; 18:1249-1256. [PMID: 36453401 PMCID: PMC9838139 DOI: 10.4103/1673-5374.355741] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
The lymphatic vasculature forms an organized network that covers the whole body and is involved in fluid homeostasis, metabolite clearance, and immune surveillance. The recent identification of functional lymphatic vessels in the meninges of the brain and the spinal cord has provided novel insights into neurophysiology. They emerge as major pathways for fluid exchange. The abundance of immune cells in lymphatic vessels and meninges also suggests that lymphatic vessels are actively involved in neuroimmunity. The lymphatic system, through its role in the clearance of neurotoxic proteins, autoimmune cell infiltration, and the transmission of pro-inflammatory signals, participates in the pathogenesis of a variety of neurological disorders, including neurodegenerative and neuroinflammatory diseases and traumatic injury. Vascular endothelial growth factor C is the master regulator of lymphangiogenesis, a process that is critical for the maintenance of central nervous system homeostasis. In this review, we summarize current knowledge and recent advances relating to the anatomical features and immunological functions of the lymphatic system of the central nervous system and highlight its potential as a therapeutic target for neurological disorders and central nervous system repair.
Collapse
Affiliation(s)
- Jia-Qi Xu
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan Province, China,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, Hunan Province, China,Mobile Health Ministry of Education - China Mobile Joint Laboratory, Changsha, Hunan Province, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Qian-Qi Liu
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan Province, China,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, Hunan Province, China,Mobile Health Ministry of Education - China Mobile Joint Laboratory, Changsha, Hunan Province, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Sheng-Yuan Huang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Chun-Yue Duan
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan Province, China,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, Hunan Province, China,Mobile Health Ministry of Education - China Mobile Joint Laboratory, Changsha, Hunan Province, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Hong-Bin Lu
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, Hunan Province, China,Mobile Health Ministry of Education - China Mobile Joint Laboratory, Changsha, Hunan Province, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China,Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, Hunan Province, China,Correspondence to: Yong Cao, or ; Hong-Bin Lu, ; Jian-Zhong Hu, .
| | - Yong Cao
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan Province, China,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, Hunan Province, China,Mobile Health Ministry of Education - China Mobile Joint Laboratory, Changsha, Hunan Province, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China,Correspondence to: Yong Cao, or ; Hong-Bin Lu, ; Jian-Zhong Hu, .
| | - Jian-Zhong Hu
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan Province, China,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, Hunan Province, China,Mobile Health Ministry of Education - China Mobile Joint Laboratory, Changsha, Hunan Province, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China,Correspondence to: Yong Cao, or ; Hong-Bin Lu, ; Jian-Zhong Hu, .
| |
Collapse
|
63
|
Bohr T, Hjorth PG, Holst SC, Hrabětová S, Kiviniemi V, Lilius T, Lundgaard I, Mardal KA, Martens EA, Mori Y, Nägerl UV, Nicholson C, Tannenbaum A, Thomas JH, Tithof J, Benveniste H, Iliff JJ, Kelley DH, Nedergaard M. The glymphatic system: Current understanding and modeling. iScience 2022; 25:104987. [PMID: 36093063 PMCID: PMC9460186 DOI: 10.1016/j.isci.2022.104987] [Citation(s) in RCA: 94] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
We review theoretical and numerical models of the glymphatic system, which circulates cerebrospinal fluid and interstitial fluid around the brain, facilitating solute transport. Models enable hypothesis development and predictions of transport, with clinical applications including drug delivery, stroke, cardiac arrest, and neurodegenerative disorders like Alzheimer's disease. We sort existing models into broad categories by anatomical function: Perivascular flow, transport in brain parenchyma, interfaces to perivascular spaces, efflux routes, and links to neuronal activity. Needs and opportunities for future work are highlighted wherever possible; new models, expanded models, and novel experiments to inform models could all have tremendous value for advancing the field.
Collapse
Affiliation(s)
- Tomas Bohr
- Department of Physics, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Poul G. Hjorth
- Department of Applied Mathematics and Computer Science, Technical University of Denmark, Richard Petersens Plads, 2800 Kgs. Lyngby, Denmark
| | - Sebastian C. Holst
- Neuroscience and Rare Diseases Discovery and Translational Area, Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Sabina Hrabětová
- Department of Cell Biology and The Robert Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Medical Center, Brooklyn, NY, USA
| | - Vesa Kiviniemi
- Oulu Functional NeuroImaging, Department of Diagnostic Radiology, MRC, Oulu University Hospital, Oulu, Finland
- Medical Imaging, Physics and Technology, the Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Tuomas Lilius
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Emergency Medicine and Services, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Iben Lundgaard
- Department of Experimental Medical Science, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Kent-Andre Mardal
- Department of Mathematics, University of Oslo, Oslo, Norway
- Simula Research Laboratory, Department of Numerical Analysis and Scientific Computing, Oslo, Norway
| | | | - Yuki Mori
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - U. Valentin Nägerl
- Instítut Interdisciplinaire de Neurosciences, Université de Bordeaux / CNRS UMR 5297, Centre Broca Nouvelle-Aquitaine, 146 rue Léo Saignat, CS 61292 Case 130, 33076 Bordeaux Cedex France
| | - Charles Nicholson
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA
- Department of Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Allen Tannenbaum
- Departments of Computer Science/ Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY, USA
| | - John H. Thomas
- Department of Mechanical Engineering, University of Rochester, Rochester, 14627 NY, USA
| | - Jeffrey Tithof
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, USA
| | - Helene Benveniste
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA
- Department of Biomedical Engineering, Yale School of Medicine, New Haven, CT, USA
| | - Jeffrey J. Iliff
- VISN 20 Mental Illness Research, Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA, USA
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA
| | - Douglas H. Kelley
- Department of Mechanical Engineering, University of Rochester, Rochester, 14627 NY, USA
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, 14642 NY, USA
| |
Collapse
|
64
|
Ocular Lymphatic and Glymphatic Systems: Implications for Retinal Health and Disease. Int J Mol Sci 2022; 23:ijms231710139. [PMID: 36077535 PMCID: PMC9456449 DOI: 10.3390/ijms231710139] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 11/17/2022] Open
Abstract
Clearance of ocular fluid and metabolic waste is a critical function of the eye in health and disease. The eye has distinct fluid outflow pathways in both the anterior and posterior segments. Although the anterior outflow pathway is well characterized, little is known about posterior outflow routes. Recent studies suggest that lymphatic and glymphatic systems play an important role in the clearance of fluid and waste products from the posterior segment of the eye. The lymphatic system is a vascular network that runs parallel to the blood circulatory system. It plays an essential role in maintenance of fluid homeostasis and immune surveillance in the body. Recent studies have reported lymphatics in the cornea (under pathological conditions), ciliary body, choroid, and optic nerve meninges. The evidence of lymphatics in optic nerve meninges is, however, limited. An alternative lymphatic system termed the glymphatic system was recently discovered in the rodent eye and brain. This system is a glial cell-based perivascular network responsible for the clearance of interstitial fluid and metabolic waste. In this review, we will discuss our current knowledge of ocular lymphatic and glymphatic systems and their role in retinal degenerative diseases.
Collapse
|
65
|
Odfalk KF, Bieniek KF, Hopp SC. Microglia: Friend and foe in tauopathy. Prog Neurobiol 2022; 216:102306. [PMID: 35714860 PMCID: PMC9378545 DOI: 10.1016/j.pneurobio.2022.102306] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 04/24/2022] [Accepted: 06/10/2022] [Indexed: 12/16/2022]
Abstract
Aggregation of misfolded microtubule associated protein tau into abnormal intracellular inclusions defines a class of neurodegenerative diseases known as tauopathies. The consistent spatiotemporal progression of tau pathology in Alzheimer's disease (AD) led to the hypothesis that tau aggregates spread in the brain via bioactive tau "seeds" underlying advancing disease course. Recent studies implicate microglia, the resident immune cells of the central nervous system, in both negative and positive regulation of tau pathology. Polymorphisms in genes that alter microglial function are associated with the development of AD and other tauopathies. Experimental manipulation of microglia function can alter tau pathology and microglia-mediated neuroinflammatory cascades can exacerbate tau pathology. Microglia also exert protective functions by mitigating tau spread: microglia internalize tau seeds and have the capacity to degrade them. However, when microglia fail to degrade these tau seeds there are deleterious consequences, including secretion of exosomes containing tau that can spread to neurons. This review explores the intersection of microglia and tau from the perspective of neuropathology, neuroimaging, genetics, transcriptomics, and molecular biology. As tau-targeted therapies such as anti-tau antibodies advance through clinical trials, it is critical to understand the interaction between tau and microglia.
Collapse
Affiliation(s)
- Kristian F Odfalk
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA; Department of Pharmacology, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Kevin F Bieniek
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA; Department of Pathology and Laboratory Medicine, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Sarah C Hopp
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA; Department of Pharmacology, University of Texas Health Science Center San Antonio, San Antonio, TX, USA.
| |
Collapse
|
66
|
Abstract
The central nervous system (CNS) has been viewed as an immunologically privileged site, but emerging works are uncovering a large array of neuroimmune interactions primarily occurring at its borders. CNS barriers sites host diverse population of both innate and adaptive immune cells capable of, directly and indirectly, influence the function of the residing cells of the brain parenchyma. These structures are only starting to reveal their role in controlling brain function under normal and pathological conditions and represent an underexplored therapeutic target for the treatment of brain disorders. This review will highlight the development of the CNS barriers to host neuro-immune interactions and emphasize their newly described roles in neurodevelopmental, neurological, and neurodegenerative disorders, particularly for the meninges.
Collapse
Affiliation(s)
- Natalie M Frederick
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Gabriel A Tavares
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Antoine Louveau
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Molecular Medicine, Cleveland Clinic College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA.,Kent University, Neurosciences, School of Biomedical Sciences, Cleveland, Ohio, USA
| |
Collapse
|
67
|
Dai W, Yang M, Xia P, Xiao C, Huang S, Zhang Z, Cheng X, Li W, Jin J, Zhang J, Wu B, Zhang Y, Wu PH, Lin Y, Wu W, Zhao H, Zhang Y, Lin WJ, Ye X. A functional role of meningeal lymphatics in sex difference of stress susceptibility in mice. Nat Commun 2022; 13:4825. [PMID: 35974004 PMCID: PMC9381547 DOI: 10.1038/s41467-022-32556-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 08/03/2022] [Indexed: 11/09/2022] Open
Abstract
Major depressive disorder is one of the most common mental health conditions. Meningeal lymphatics are essential for drainage of molecules in the cerebrospinal fluid to the peripheral immune system. Their potential role in depression-like behaviour has not been investigated. Here, we show in mice, sub-chronic variable stress as a model of depression-like behaviour impairs meningeal lymphatics in females but not in males. Manipulations of meningeal lymphatics regulate the sex difference in the susceptibility to stress-induced depression- and anxiety-like behaviors in mice, as well as alterations of the medial prefrontal cortex and the ventral tegmental area, brain regions critical for emotional regulation. Together, our findings suggest meningeal lymphatic impairment contributes to susceptibility to stress in mice, and that restoration of the meningeal lymphatics might have potential for modulation of depression-like behaviour.
Collapse
Affiliation(s)
- Weiping Dai
- Brain Research Center, Sun Yat-sen Memorial Hospital and Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Mengqian Yang
- Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Pei Xia
- Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Chuan Xiao
- Brain Research Center, Sun Yat-sen Memorial Hospital and Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shuying Huang
- Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhan Zhang
- Brain Research Center, Sun Yat-sen Memorial Hospital and Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xin Cheng
- Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Wenchang Li
- Department of Joint Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jian Jin
- Department of Rehabilitation, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jingyun Zhang
- Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Binghuo Wu
- Key Laboratory of Stem Cells and Tissue Engineering, Zhongshan School of Medicine, Sun Yat-sen University, Ministry of Education, Guangzhou, China
| | - Yingying Zhang
- Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Pei-Hui Wu
- Department of Joint Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yangyang Lin
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Department of Rehabilitation Medicine, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wen Wu
- Department of Rehabilitation, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Hu Zhao
- Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yan Zhang
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Wei-Jye Lin
- Brain Research Center, Sun Yat-sen Memorial Hospital and Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China. .,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China. .,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Xiaojing Ye
- Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China. .,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
68
|
Virenque A, Koivisto H, Antila S, Zub E, Rooney EJ, Miszczuk D, Müller A, Stoka E, Marchi N, Alitalo K, Tanila H, Noe FM. Significance of developmental meningeal lymphatic dysfunction in experimental post-traumatic injury. Brain Behav Immun Health 2022; 23:100466. [PMID: 35694175 PMCID: PMC9184565 DOI: 10.1016/j.bbih.2022.100466] [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: 01/13/2022] [Revised: 04/24/2022] [Accepted: 04/26/2022] [Indexed: 12/01/2022] Open
Abstract
Understanding the pathological mechanisms unfolding after chronic traumatic brain injury (TBI) could reveal new therapeutic entry points. During the post-TBI sequel, the involvement of cerebrospinal fluid drainage through the meningeal lymphatic vessels was proposed. Here, we used K14-VEGFR3-Ig transgenic mice to analyze whether a developmental dysfunction of meningeal lymphatic vessels modifies post-TBI pathology. To this end, a moderate TBI was delivered by controlled cortical injury over the temporal lobe in male transgenic mice or their littermate controls. We performed MRI and a battery of behavioral tests over time to define the post-TBI trajectories. In vivo analyses were integrated by ex-vivo quantitative and morphometric examinations of the cortical lesion and glial cells. In post-TBI K14-VEGFR3-Ig mice, the recovery from motor deficits was protracted compared to littermates. This outcome is coherent with the observed slower hematoma clearance in transgenic mice during the first two weeks post-TBI. No other genotype-related behavioral differences were observed, and the volume of cortical lesions imaged by MRI in vivo, and confirmed by histology ex-vivo, were comparable in both groups. However, at the cellular level, post-TBI K14-VEGFR3-Ig mice exhibited an increased percentage of activated Iba1 microglia in the hippocampus and auditory cortex, areas that are proximal to the lesion. Although not impacting or modifying the structural brain damage and post-TBI behavior, a pre-existing dysfunction of meningeal lymphatic vessels is associated with morphological microglial activation over time, possibly representing a sub-clinical pathological imprint or a vulnerability factor. Our findings suggest that pre-existing mLV deficits could represent a possible risk factor for the overall outcome of TBI pathology. Developmental deficit in the meningeal lymphatic vessels contributes to sustain the chronic neuroinflammation and represent a susceptibility factor in TBI, despite the lack of a functional phenotype. Development and progression of TBI-related cortical lesion is not exacerbated by developmental deficit in meningeal lymphatics. Meningeal lymphatic developmental deficits result in increased neuroinflammation, suggesting a sub-clinical pathological imprint or a vulnerability factor. Congenital mLV deficit affects the interstitial fluid dynamics and the post-TBI hematoma resolution.
Collapse
Affiliation(s)
- Anaïs Virenque
- Neuroscience Center, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, 00290, Helsinki, Finland
| | - Hennariikka Koivisto
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland
| | - Salli Antila
- Wihuri Research Institute and Translational Cancer Medicine Program, Biomedicum Helsinki, University of Helsinki, 00290, Helsinki, Finland
| | - Emma Zub
- Cerebrovascular and Glia Research, Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Erin Jane Rooney
- Neuroscience Center, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, 00290, Helsinki, Finland
| | - Diana Miszczuk
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland
| | - Adrian Müller
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland
| | - Enija Stoka
- Neuroscience Center, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, 00290, Helsinki, Finland
| | - Nicola Marchi
- Cerebrovascular and Glia Research, Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Kari Alitalo
- Wihuri Research Institute and Translational Cancer Medicine Program, Biomedicum Helsinki, University of Helsinki, 00290, Helsinki, Finland
| | - Heikki Tanila
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland
| | - Francesco Mattia Noe
- Neuroscience Center, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, 00290, Helsinki, Finland
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland
- Corresponding author. HiLIFE, Neuroscience Center, Helsinki University, Helsinki, Finland.
| |
Collapse
|
69
|
Hovd MH, Mariussen E, Uggerud H, Lashkarivand A, Christensen H, Ringstad G, Eide PK. Population pharmacokinetic modeling of CSF to blood clearance: prospective tracer study of 161 patients under work-up for CSF disorders. Fluids Barriers CNS 2022; 19:55. [PMID: 35778719 PMCID: PMC9250213 DOI: 10.1186/s12987-022-00352-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 06/22/2022] [Indexed: 11/30/2022] Open
Abstract
Background Quantitative measurements of cerebrospinal fluid to blood clearance has previously not been established for neurological diseases. Possibly, variability in cerebrospinal fluid clearance may affect the underlying disease process and may possibly be a source of under- or over-dosage of intrathecally administered drugs. The aim of this study was to characterize the cerebrospinal fluid to blood clearance of the intrathecally administered magnetic resonance imaging contrast agent gadobutrol (Gadovist, Bayer Pharma AG, GE). For this, we established a population pharmacokinetic model, hypothesizing that cerebrospinal fluid to blood clearance differs between cerebrospinal fluid diseases. Methods Gadobutrol served as a surrogate tracer for extra-vascular pathways taken by several brain metabolites and drugs in cerebrospinal fluid. We estimated cerebrospinal fluid to blood clearance in patients with different cerebrospinal fluid disorders, i.e. symptomatic pineal and arachnoid cysts, as well as tentative spontaneous intracranial hypotension due to cerebrospinal fluid leakage, idiopathic intracranial hypertension, or different types of hydrocephalus (idiopathic normal pressure hydrocephalus, communicating- and non-communicating hydrocephalus). Individuals with no verified cerebrospinal fluid disturbance at clinical work-up were denoted references. Results Population pharmacokinetic modelling based on 1,140 blood samples from 161 individuals revealed marked inter-individual variability in pharmacokinetic profiles, including differences in absorption half-life (time to 50% of tracer absorbed from cerebrospinal fluid to blood), time to maximum concentration in blood and the maximum concentration in blood as well as the area under the plasma concentration time curve from zero to infinity. In addition, the different disease categories of cerebrospinal fluid diseases demonstrated different profiles. Conclusions The present observations of considerable variation in cerebrospinal fluid to blood clearance between individuals in general and across neurological diseases, may suggest that defining cerebrospinal fluid to blood clearance can become a useful diagnostic adjunct for work-up of cerebrospinal fluid disorders. We also suggest that it may become useful for assessing clearance capacity of endogenous brain metabolites from cerebrospinal fluid, as well as measuring individual cerebrospinal fluid to blood clearance of intrathecal drugs.
Collapse
Affiliation(s)
- Markus Herberg Hovd
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Espen Mariussen
- Norwegian Institute for Air Research, Kjeller, Norway.,Department of Air Quality and Noise, Norwegian Institute of Public Health, Oslo, Norway
| | - Hilde Uggerud
- Norwegian Institute for Air Research, Kjeller, Norway
| | - Aslan Lashkarivand
- Department of Neurosurgery, Oslo University Hospital-Rikshospitalet, Pb 4950 Nydalen, 0424, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Hege Christensen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Geir Ringstad
- Division of Radiology and Nuclear Medicine, Department of Radiology, Oslo University Hospital-Rikshospitalet, Oslo, Norway.,Department of Geriatrics and Internal Medicine, Sorlandet Hospital, Arendal, Norway
| | - Per Kristian Eide
- Department of Neurosurgery, Oslo University Hospital-Rikshospitalet, Pb 4950 Nydalen, 0424, Oslo, Norway. .,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
70
|
Li Q, Chen Y, Feng W, Cai J, Gao J, Ge F, Zhou T, Wang Z, Ding F, Marshall C, Sheng C, Zhang Y, Sun M, Shi J, Xiao M. Drainage of senescent astrocytes from brain via meningeal lymphatic routes. Brain Behav Immun 2022; 103:85-96. [PMID: 35427759 DOI: 10.1016/j.bbi.2022.04.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 03/16/2022] [Accepted: 04/10/2022] [Indexed: 12/30/2022] Open
Abstract
Recent progress on the central lymphatic system has greatly increased our understanding of how the brain maintains its own waste homeostasis. Here, we showed that perivascular spaces and meningeal lymphatic vessels form a functional route for clearance of senescent astrocytes from the aging brain. Blocking meningeal lymphatic drainage by ligation of the deep cervical lymph nodes impaired clearance of senescent astrocytes from brain parenchyma, subsequently increasing neuroinflammation in aged mice. By contrast, enhancing meningeal lymphatic vessel diameter by a recombinant adeno-associated virus encoding mouse vascular endothelial growth factor-C (VEGF-C) improved clearance of senescent astrocytes and mitigated neuroinflammation. Mechanistically, VEGF-C was highly expressed in senescent astrocytes, contributing themselves to migrate across lymphatic vessels along C-C motif chemokine ligand 21 (CCL21) gradient by interacting with VEGF receptor 3. Moreover, intra-cisternal injection of antibody against CCL21 hampered senescent astrocytes into the lymphatic vessels and exacerbated short memory defects of aged mice. Together, these findings reveal a new perspective for the meningeal lymphatics in the removal of senescent astrocytes, thus offering a valuable target for therapeutic intervention.
Collapse
Affiliation(s)
- Qian Li
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, 211166, China; Department of Neurology, the Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, 210029, China; Center for Global Health, Nanjing Medical University, Nanjing, 211166, China
| | - Yan Chen
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, 211166, China; Department of Neurology, the Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, 210029, China; Center for Global Health, Nanjing Medical University, Nanjing, 211166, China
| | - Weixi Feng
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, 211166, China; Department of Neurology, the Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, 210029, China; Center for Global Health, Nanjing Medical University, Nanjing, 211166, China
| | - Jiachen Cai
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, 211166, China
| | - Junying Gao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, 211166, China; Department of Anatomy, Nanjing Medical University, Nanjing, 211166, China
| | - Feifei Ge
- Department of Neurology, the Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Tiantian Zhou
- Department of Anesthesia, Nanjing Integrated Traditional Chinese and Western Medicine Hospital, Nanjing, 210028, China
| | - Ze Wang
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, 211166, China
| | - Fengfei Ding
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China; Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, Department of Neurosurgery, University of Rochester Medical Center, Rochester, NY, 14642, United States
| | - Charles Marshall
- Department of Physical Therapy, University of Kentucky Center of Excellence in Rural Health, Hazard, KY, 41701, USA
| | - Chengyu Sheng
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, 211166, China
| | - Yongjie Zhang
- Department of Anatomy, Nanjing Medical University, Nanjing, 211166, China
| | - Mingkuan Sun
- Department of Toxicology, Nanjing Medical University, Nanjing, 211166, China
| | - Jingping Shi
- Department of Neurology, the Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Ming Xiao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, 211166, China; Department of Neurology, the Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, 210029, China; Center for Global Health, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
71
|
Chen Z, Liu P, Xia X, Wang L, Li X. Living on the border of the CNS: Dural immune cells in health and disease. Cell Immunol 2022; 377:104545. [DOI: 10.1016/j.cellimm.2022.104545] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/26/2022] [Accepted: 05/09/2022] [Indexed: 12/31/2022]
|
72
|
Singla B, Aithabathula RV, Kiran S, Kapil S, Kumar S, Singh UP. Reactive Oxygen Species in Regulating Lymphangiogenesis and Lymphatic Function. Cells 2022; 11:1750. [PMID: 35681445 PMCID: PMC9179518 DOI: 10.3390/cells11111750] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/18/2022] [Accepted: 05/24/2022] [Indexed: 11/17/2022] Open
Abstract
The lymphatic system is pivotal for immunosurveillance and the maintenance of tissue homeostasis. Lymphangiogenesis, the formation of new lymphatic vessels from pre-existing vessels, has both physiological and pathological roles. Recent advances in the molecular mechanisms regulating lymphangiogenesis have opened a new area of research on reparative lymphangiogenesis for the treatment of various pathological disorders comprising neurological disorders, cardiac repair, autoimmune disease, obesity, atherosclerosis, etc. Reactive oxygen species (ROS) produced by the various cell types serve as signaling molecules in several cellular mechanisms and regulate various aspects of growth-factor-mediated responses, including lymphangiogenesis. The ROS, including superoxide anion, hydrogen peroxide, and nitric oxide, play both beneficial and detrimental roles depending upon their levels and cellular microenvironment. Low ROS levels are essential for lymphangiogenesis. On the contrary, oxidative stress due to enhanced ROS generation and/or reduced levels of antioxidants suppresses lymphangiogenesis via promoting lymphatic endothelial cell apoptosis and death. In this review article, we provide an overview of types and sources of ROS, discuss the role of ROS in governing lymphangiogenesis and lymphatic function, and summarize the role of lymphatics in various diseases.
Collapse
Affiliation(s)
- Bhupesh Singla
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, Memphis, TN 38017, USA; (R.V.A.); (S.K.); (S.K.); (U.P.S.)
| | - Ravi Varma Aithabathula
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, Memphis, TN 38017, USA; (R.V.A.); (S.K.); (S.K.); (U.P.S.)
| | - Sonia Kiran
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, Memphis, TN 38017, USA; (R.V.A.); (S.K.); (S.K.); (U.P.S.)
| | - Shweta Kapil
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children′s Hospital Medical Center, Cincinnati, OH 45229, USA;
| | - Santosh Kumar
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, Memphis, TN 38017, USA; (R.V.A.); (S.K.); (S.K.); (U.P.S.)
| | - Udai P. Singh
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, Memphis, TN 38017, USA; (R.V.A.); (S.K.); (S.K.); (U.P.S.)
| |
Collapse
|
73
|
The role of the meningeal lymphatic system in local meningeal inflammation and trigeminal nociception. Sci Rep 2022; 12:8804. [PMID: 35614095 PMCID: PMC9133044 DOI: 10.1038/s41598-022-12540-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 04/20/2022] [Indexed: 12/23/2022] Open
Abstract
A system of lymphatic vessels has been recently characterized in the meninges, with a postulated role in ‘cleaning’ the brain via cerebral fluid drainage. As meninges are the origin site of migraine pain, we hypothesized that malfunctioning of the lymphatic system should affect the local trigeminal nociception. To test this hypothesis, we studied nociceptive and inflammatory mechanisms in the hemiskull preparations (containing the meninges) of K14-VEGFR3-Ig (K14) mice lacking the meningeal lymphatic system. We recorded the spiking activity of meningeal afferents and estimated the local mast cells population, calcitonin gene-related peptide (CGRP) and cytokine levels as well as the dural trigeminal innervation in freshly-isolated hemiskull preparations from K14-VEGFR3-Ig (K14) or wild type C57BL/6 mice (WT). Spiking activity data have been confirmed in an acquired model of meningeal lymphatic dysfunction (AAV-mVEGFR3(1–4)Ig induced lymphatic ablation). We found that levels of the pro-inflammatory cytokine IL12-p70 and CGRP, implicated in migraine, were reduced in the meninges of K14 mice, while the levels of the mast cell activator MCP-1 were increased. The other migraine-related pro-inflammatory cytokines (basal and stimulated), did not differ between the two genotypes. The patterns of trigeminal innervation in meninges remained unchanged and we did not observe alterations in basal or ATP-induced nociceptive firing in the meningeal afferents associated with meningeal lymphatic dysfunction. In summary, the lack of meningeal lymphatic system is associated with a new balance between pro- and anti-migraine mediators but does not directly trigger meningeal nociceptive state.
Collapse
|
74
|
Zhang D, Li X, Li B. Glymphatic System Dysfunction in Central Nervous System Diseases and Mood Disorders. Front Aging Neurosci 2022; 14:873697. [PMID: 35547631 PMCID: PMC9082304 DOI: 10.3389/fnagi.2022.873697] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/31/2022] [Indexed: 12/13/2022] Open
Abstract
The glymphatic system, a recently discovered macroscopic waste removal system in the brain, has many unknown aspects, especially its driving forces and relationship with sleep, and thus further explorations of the relationship between the glymphatic system and a variety of possible related diseases are urgently needed. Here, we focus on the progress in current research on the role of the glymphatic system in several common central nervous system diseases and mood disorders, discuss the structural and functional abnormalities of the glymphatic system which may occur before or during the pathophysiological progress and the possible underlying mechanisms. We emphasize the relationship between sleep and the glymphatic system under pathological conditions and summarize the common imaging techniques for the glymphatic system currently available. The perfection of the glymphatic system hypothesis and the exploration of the effects of aging and endocrine factors on the central and peripheral regulatory pathways through the glymphatic system still require exploration in the future.
Collapse
Affiliation(s)
- Dianjun Zhang
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, School of Forensic Medicine, China Medical University, Shenyang, China
- China Medical University Center of Forensic Investigation, School of Forensic Medicine, China Medical University, Shenyang, China
| | - Xinyu Li
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, School of Forensic Medicine, China Medical University, Shenyang, China
- China Medical University Center of Forensic Investigation, School of Forensic Medicine, China Medical University, Shenyang, China
| | - Baoman Li
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, School of Forensic Medicine, China Medical University, Shenyang, China
- China Medical University Center of Forensic Investigation, School of Forensic Medicine, China Medical University, Shenyang, China
| |
Collapse
|
75
|
Olate-Briones A, Escalona E, Salazar C, Herrada MJ, Liu C, Herrada AA, Escobedo N. The meningeal lymphatic vasculature in neuroinflammation. FASEB J 2022; 36:e22276. [PMID: 35344212 DOI: 10.1096/fj.202101574rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 03/08/2022] [Accepted: 03/14/2022] [Indexed: 12/13/2022]
Abstract
The lymphatic vasculature is a unidirectional network of lymphatic endothelial cells, whose main role is to maintain fluid homeostasis along with the absorption of dietary fat in the gastrointestinal organs and management and coordination of immune cell trafficking into lymph nodes during homeostasis and under inflammatory conditions. In homeostatic conditions, immune cells, such as dendritic cells, macrophages, or T cells can enter into the lymphatic vasculature and move easily through the lymph reaching secondary lymph nodes where immune cell activation or peripheral tolerance can be modulated. However, under inflammatory conditions such as pathogen infection, increased permeabilization of lymphatic vessels allows faster immune cell migration into inflamed tissues following a chemokine gradient, facilitating pathogen clearance and the resolution of inflammation. Interestingly, since the re-discovery of lymphatic vasculature in the central nervous system, known as the meningeal lymphatic vasculature, the role of these lymphatics as a key player in several neurological disorders has been described, with emphasis on the neurodegenerative process. Alternatively, less has been discussed about meningeal lymphatics and its role in neuroinflammation. In this review, we discuss current knowledge about the anatomy and function of the meningeal lymphatic vasculature and specifically analyze its contribution to different neuroinflammatory processes, highlighting the potential therapeutic target of meningeal lymphatic vasculature in these pathological conditions.
Collapse
Affiliation(s)
- Alexandra Olate-Briones
- Lymphatic Vasculature and Inflammation Research Laboratory, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Talca, Chile
| | - Emilia Escalona
- Lymphatic Vasculature and Inflammation Research Laboratory, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Talca, Chile
| | - Celia Salazar
- Lymphatic Vasculature and Inflammation Research Laboratory, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Talca, Chile
| | | | - Chaohong Liu
- Department of Microbiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Andrés A Herrada
- Lymphatic Vasculature and Inflammation Research Laboratory, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Talca, Chile
| | - Noelia Escobedo
- Lymphatic Vasculature and Inflammation Research Laboratory, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Talca, Chile
| |
Collapse
|
76
|
Onder H, Kocer B, Comoglu S. Idiopathic normal pressure hydrocephalus-like MRI features in patients with progressive supranuclear palsy: a comparative case-control study. Neurol Res 2022; 44:807-813. [PMID: 35297741 DOI: 10.1080/01616412.2022.2052622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
BACKGROUND The occurrence of neurodegenerative disease in patients with normal pressure hydrocephalus (NPH) is emphasized in recent reports. Based on this common co-occurrence, some of the hydrocephalic disorders appearing in late adulthood have been hypothesized to result from initially unapparent parenchymal abnormalities of neurodegenerative origin. Among these diseases, progressive supranuclear palsy (PSP) has been specifically remarked on. We aimed to comparatively investigate the neuroimaging clues of iNPH in our PSP subjects. METHODS Eighteen patients with a clinical diagnosis of PSP, 44 with Parkinson's disease (PD), and 44 healthy control (HC) individuals were enrolled. The disproportionately enlarged subarachnoid space hydrocephalus (DESH) score, the Evans' index (EI), and the callosal angle (CA) were measured on the conventional magnetic resonance imaging (MRI). The comparative analyses were performed using IBM SPSS Statistics 26. RESULTS We found that dilated Sylvian fissures score (p = 0.016) and focal sulcal dilatation score (p = 0.037) were higher in the PSP group in comparison to HC whereas the CA score was higher in PSP subjects in comparison to both PD patients and HC (p = 0.000). Remarkably, the DESH score was also found to be higher in the PSP group in comparison to the age-matched HC group (p = 0.024). CONCLUSIONS We found that the NPH-like MRI features were more common in PSP subjects in comparison to PD subjects and age-matched HC. These results may provide critical contributions to the literature regarding the overlap between PSP and NPH.
Collapse
Affiliation(s)
- Halil Onder
- Neurology Clinic, Diskapi Yildirim Beyazit Training and Research Hospital, Ankara, Turkey
| | - Bilge Kocer
- Neurology Clinic, Diskapi Yildirim Beyazit Training and Research Hospital, Ankara, Turkey
| | - Selcuk Comoglu
- Neurology Clinic, Diskapi Yildirim Beyazit Training and Research Hospital, Ankara, Turkey
| |
Collapse
|
77
|
Ishida K, Yamada K, Nishiyama R, Hashimoto T, Nishida I, Abe Y, Yasui M, Iwatsubo T. Glymphatic system clears extracellular tau and protects from tau aggregation and neurodegeneration. J Exp Med 2022; 219:213037. [PMID: 35212707 PMCID: PMC8932543 DOI: 10.1084/jem.20211275] [Citation(s) in RCA: 86] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 12/21/2021] [Accepted: 01/24/2022] [Indexed: 12/16/2022] Open
Abstract
Accumulation of tau has been implicated in various neurodegenerative diseases termed tauopathies. Tau is a microtubule-associated protein but is also actively released into the extracellular fluids including brain interstitial fluid and cerebrospinal fluid (CSF). However, it remains elusive whether clearance of extracellular tau impacts tau-associated neurodegeneration. Here, we show that aquaporin-4 (AQP4), a major driver of the glymphatic clearance system, facilitates the elimination of extracellular tau from the brain to CSF and subsequently to deep cervical lymph nodes. Strikingly, deletion of AQP4 not only elevated tau in CSF but also markedly exacerbated phosphorylated tau deposition and the associated neurodegeneration in the brains of transgenic mice expressing P301S mutant tau. The current study identified the clearance pathway of extracellular tau in the central nervous system, suggesting that glymphatic clearance of extracellular tau is a novel regulatory mechanism whose impairment contributes to tau aggregation and neurodegeneration.
Collapse
Affiliation(s)
- Kazuhisa Ishida
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kaoru Yamada
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Risa Nishiyama
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tadafumi Hashimoto
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Innovative Dementia Prevention, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Itaru Nishida
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoichiro Abe
- Department of Pharmacology, Keio University School of Medicine, Tokyo, Japan.,Keio University Global Research Institute, Center for Water Biology and Medicine, Tokyo, Japan
| | - Masato Yasui
- Department of Pharmacology, Keio University School of Medicine, Tokyo, Japan.,Keio University Global Research Institute, Center for Water Biology and Medicine, Tokyo, Japan
| | - Takeshi Iwatsubo
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| |
Collapse
|
78
|
Connexin 43 gap junction-mediated astrocytic network reconstruction attenuates isoflurane-induced cognitive dysfunction in mice. J Neuroinflammation 2022; 19:64. [PMID: 35255943 PMCID: PMC8903726 DOI: 10.1186/s12974-022-02424-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 02/23/2022] [Indexed: 01/18/2023] Open
Abstract
Background Postoperative cognitive dysfunction (POCD) is a common complication following anesthesia and surgery. General anesthetic isoflurane has potential neurotoxicity and induces cognitive impairments, but the exact mechanism remains unclear. Astrocytes form interconnected networks in the adult brain through gap junctions (GJs), which primarily comprise connexin 43 (Cx43), and play important roles in brain homeostasis and functions such as memory. However, the role of the GJ-Cx43-mediated astrocytic network in isoflurane-induced cognitive dysfunction has not been defined. Methods 4-month-old male C57BL/6 mice were exposure to long-term isoflurane to induce cognitive impairment. To simulate an in vitro isoflurane-induced cognitive dysfunction‐like condition, primary mouse astrocytes were subjected to long-term isoflurane exposure. Cognitive function was assessed by Y-maze and fear conditioning tests. Western blot was used to determine the expression levels of different functional configurations of Cx43. The morphology of the GJs-Cx43 was evaluated by immunofluorescence staining. Levels of IL-1β and IL-6 were examined by ELISA. The ability of GJs-Cx43-mediated intercellular communication was examined by lucifer yellow dye transfer assay. Ethidium bromide uptake assays were used to measure the activity of Cx43 hemichannels. The ultrastructural morphology of astrocyte gap junctions and tripartite synapse were observed by transmission electron microscopy. Results After long-term isoflurane anesthesia, the GJs formed by Cx43 in the mouse hippocampus and primary mouse astrocytes were significantly reduced, GJs function was impaired, hemichannel activity was enhanced, the levels of IL-1β and IL-6 were increased, and mice showed significant cognitive impairment. After treatment with the novel GJ-Cx43 enhancer ZP1609, GJ-Cx43-mediated astrocytic network function was enhanced, neuroinflammation was alleviated, and ameliorated cognition dysfunction induced by long-term isoflurane exposure. However, ZP1609 enhances the astrocytic network by promoting Cx43 to form GJs without affecting hemichannel activity. Additionally, our data showed that long-term isoflurane exposure does not alter the structure of tripartite synapse. Conclusion Our results reveal a novel mechanism of the GJ-Cx43-mediated astrocytic network involved in isoflurane-induced neuroinflammation and cognitive impairments, which provides new mechanistic insight into the pathogenesis of POCD and identifies potential targets for its treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02424-y.
Collapse
|
79
|
Bush N, O’Reilly L, Louveau A. Meningeal Lymphatic vasculature in health and disease. Curr Opin Hematol 2022; 29:151-155. [DOI: 10.1097/moh.0000000000000711] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
80
|
Wardlaw JM, Benveniste H, Williams A. Cerebral Vascular Dysfunctions Detected in Human Small Vessel Disease and Implications for Preclinical Studies. Annu Rev Physiol 2022; 84:409-434. [PMID: 34699267 DOI: 10.1146/annurev-physiol-060821-014521] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cerebral small vessel disease (SVD) is highly prevalent and a common cause of ischemic and hemorrhagic stroke and dementia, yet the pathophysiology is poorly understood. Its clinical expression is highly varied, and prognostic implications are frequently overlooked in clinics; thus, treatment is currently confined to vascular risk factor management. Traditionally, SVD is considered the small vessel equivalent of large artery stroke (occlusion, rupture), but data emerging from human neuroimaging and genetic studies refute this, instead showing microvessel endothelial dysfunction impacting on cell-cell interactions and leading to brain damage. These dysfunctions reflect defects that appear to be inherited and secondary to environmental exposures, including vascular risk factors. Interrogation in preclinical models shows consistent and converging molecular and cellular interactions across the endothelial-glial-neural unit that increasingly explain the human macroscopic observations and identify common patterns of pathology despite different triggers. Importantly, these insights may offer new targets for therapeutic intervention focused on restoring endothelial-glial physiology.
Collapse
Affiliation(s)
- Joanna M Wardlaw
- Division of Neuroimaging Sciences, Centre for Clinical Brain Sciences; UK Dementia Research Institute; and Edinburgh Imaging, University of Edinburgh, Edinburgh, United Kingdom;
| | - Helene Benveniste
- Department of Anesthesiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Anna Williams
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
81
|
Nguyen B, Bix G, Yao Y. Basal lamina changes in neurodegenerative disorders. Mol Neurodegener 2021; 16:81. [PMID: 34876200 PMCID: PMC8650282 DOI: 10.1186/s13024-021-00502-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 11/17/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Neurodegenerative disorders are a group of age-associated diseases characterized by progressive degeneration of the structure and function of the CNS. Two key pathological features of these disorders are blood-brain barrier (BBB) breakdown and protein aggregation. MAIN BODY The BBB is composed of various cell types and a non-cellular component---the basal lamina (BL). Although how different cells affect the BBB is well studied, the roles of the BL in BBB maintenance and function remain largely unknown. In addition, located in the perivascular space, the BL is also speculated to regulate protein clearance via the meningeal lymphatic/glymphatic system. Recent studies from our laboratory and others have shown that the BL actively regulates BBB integrity and meningeal lymphatic/glymphatic function in both physiological and pathological conditions, suggesting that it may play an important role in the pathogenesis and/or progression of neurodegenerative disorders. In this review, we focus on changes of the BL and its major components during aging and in neurodegenerative disorders, including Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). First, we introduce the vascular and lymphatic systems in the CNS. Next, we discuss the BL and its major components under homeostatic conditions, and summarize their changes during aging and in AD, PD, and ALS in both rodents and humans. The functional significance of these alterations and potential therapeutic targets are also reviewed. Finally, key challenges in the field and future directions are discussed. CONCLUSIONS Understanding BL changes and the functional significance of these changes in neurodegenerative disorders will fill the gap of knowledge in the field. Our goal is to provide a clear and concise review of the complex relationship between the BL and neurodegenerative disorders to stimulate new hypotheses and further research in this field.
Collapse
Affiliation(s)
- Benjamin Nguyen
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA, USA
| | - Gregory Bix
- Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Departments of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Yao Yao
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA, USA.
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, MDC 8, Tampa, Florida, 33612, USA.
| |
Collapse
|
82
|
Eide PK, Ringstad G. Cerebrospinal fluid egress to human parasagittal dura and the impact of sleep deprivation. Brain Res 2021; 1772:147669. [PMID: 34587499 DOI: 10.1016/j.brainres.2021.147669] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/26/2021] [Accepted: 09/22/2021] [Indexed: 10/20/2022]
Abstract
Emerging evidence suggests that the glymphatic system and meningeal lymphatic vessels are instrumental for clearance of toxic metabolites from the brain. Animal and human studies suggest that glymphatic circulation is up-regulated during sleep. Meningeal lymphatic clearance may be more efficient in the wake state, as shown in rodents. We have previously shown clearance of cerebrospinal fluid directly from the subarachnoid space to the parasagittal dura, which harbors meningeal lymphatic vessels. Hence, assessing molecular clearance from parasagittal dura provides an opportunity to decipher the role of sleep/sleep deprivation in human lymphatic clearance function. In this study, we applied magnetic resonance imaging to explore whether sleep deprivation modifies molecular clearance from human parasagittal dura, utilizing an intrathecal magnetic resonance imaging contrast agent as tracer. We hypothesized that tracer enhancement in parasagittal dura would differ after sleep deprivation. One group of individuals (n = 7) underwent one night's total sleep deprivation while a control group (n = 9) was allowed unrestricted sleep. There were no sleep restrictions after the 24-hour time point. After one night of sleep deprivation (at 24 h), we found neither evidence for altered tracer enrichment in the parasagittal dura, nor after a day of unrestricted sleep (at 48 h). The hypothesis of altered molecular egress to parasagittal dura after sleep deprivation was not supported by our data. Further studies are required to determine the role of sleep for molecular clearance from cerebrospinal fluid to meningeal lymphatic vessels in humans.
Collapse
Affiliation(s)
- Per Kristian Eide
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Neurosurgery, Oslo University Hospital - Rikshospitalet, Oslo, Norway.
| | - Geir Ringstad
- Department of Radiology, Oslo University Hospital - Rikshospitalet, Oslo, Norway.
| |
Collapse
|
83
|
Meningeal Lymphatics: An Immune Gateway for the Central Nervous System. Cells 2021; 10:cells10123385. [PMID: 34943894 PMCID: PMC8699870 DOI: 10.3390/cells10123385] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/26/2021] [Accepted: 11/28/2021] [Indexed: 01/30/2023] Open
Abstract
The recent (re)discovery of the meningeal lymphatic system has opened new theories as to how immune cells traffic and interact with the central nervous system (CNS). While evidence is accumulating on the contribution of the meningeal lymphatic system in both homeostatic and disease conditions, a lot remains unknown about the mechanisms that allow for interaction between the meningeal lymphatic system and immune cells. In this review, we synthesize the knowledge about the lymphatic immune interaction in the CNS and highlight the important questions that remain to be answered.
Collapse
|
84
|
Hier DB, Obafemi-Ajayi T, Thimgan MS, Olbricht GR, Azizi S, Allen B, Hadi BA, Wunsch DC. Blood biomarkers for mild traumatic brain injury: a selective review of unresolved issues. Biomark Res 2021; 9:70. [PMID: 34530937 PMCID: PMC8447604 DOI: 10.1186/s40364-021-00325-5] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 08/26/2021] [Indexed: 01/03/2023] Open
Abstract
Background The use of blood biomarkers after mild traumatic brain injury (mTBI) has been widely studied. We have identified eight unresolved issues related to the use of five commonly investigated blood biomarkers: neurofilament light chain, ubiquitin carboxy-terminal hydrolase-L1, tau, S100B, and glial acidic fibrillary protein. We conducted a focused literature review of unresolved issues in three areas: mode of entry into and exit from the blood, kinetics of blood biomarkers in the blood, and predictive capacity of the blood biomarkers after mTBI. Findings Although a disruption of the blood brain barrier has been demonstrated in mild and severe traumatic brain injury, biomarkers can enter the blood through pathways that do not require a breach in this barrier. A definitive accounting for the pathways that biomarkers follow from the brain to the blood after mTBI has not been performed. Although preliminary investigations of blood biomarkers kinetics after TBI are available, our current knowledge is incomplete and definitive studies are needed. Optimal sampling times for biomarkers after mTBI have not been established. Kinetic models of blood biomarkers can be informative, but more precise estimates of kinetic parameters are needed. Confounding factors for blood biomarker levels have been identified, but corrections for these factors are not routinely made. Little evidence has emerged to date to suggest that blood biomarker levels correlate with clinical measures of mTBI severity. The significance of elevated biomarker levels thirty or more days following mTBI is uncertain. Blood biomarkers have shown a modest but not definitive ability to distinguish concussed from non-concussed subjects, to detect sub-concussive hits to the head, and to predict recovery from mTBI. Blood biomarkers have performed best at distinguishing CT scan positive from CT scan negative subjects after mTBI.
Collapse
Affiliation(s)
- Daniel B Hier
- Department of Electrical and Computer Engineering, Missouri University of Science and Technology, Rolla, MO 65401, USA.
| | - Tayo Obafemi-Ajayi
- Cooperative Engineering Program, Missouri State University, Springfield, MO 65897, United States
| | - Matthew S Thimgan
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO 65409, United States
| | - Gayla R Olbricht
- Department of Mathematics and Statistics, Missouri University of Science and Technology, Rolla, MO 65409, United States
| | - Sima Azizi
- Department of Electrical and Computer Engineering, Missouri University of Science and Technology, Rolla, MO 65401, USA
| | - Blaine Allen
- Department of Electrical and Computer Engineering, Missouri University of Science and Technology, Rolla, MO 65401, USA
| | - Bassam A Hadi
- Department of Surgery, Mercy Hospital, St. Louis MO, Missouri, MO 63141, United States
| | - Donald C Wunsch
- Department of Electrical and Computer Engineering, Missouri University of Science and Technology, Rolla, MO 65401, USA.,National Science Foundation, ECCS Division, Virginia, 22314, USA
| |
Collapse
|
85
|
Rockson SG. Comorbidity and Lymphatic Disease: The Lymphatic Continuum Re-Examined. Lymphat Res Biol 2021; 19:17-19. [PMID: 33625889 DOI: 10.1089/lrb.2021.0001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
It has now been ∼20 years since the original Lymphatic Continuum conference was convened, and this continuum has transitioned from a compelling concept to a reality. The explosive growth in our comprehension of lymphatic genetics, development, and function has expanded and modified our traditional views regarding what is, and is not, lymphatic disease. Groundbreaking investigations over the past decade have now defined a large and growing list of pathological conditions in which morphological or function lymphatic alterations can be identified. This list includes atherosclerosis and dyslipidemia, hypertension and other cardiovascular diseases, inflammation and inflammatory bowel disease, obesity, narrow angle glaucoma, and, most recently and compellingly, neurodegenerative disease. The sometimes overlapping but largely disparate nature of these various aforementioned disease categories suggests that the presence, or absence, of structural or functional lymphatic derangements may represent a previously unrecognized unifying influence in the maintenance of health and the promotion of disease. Future investigation of lymphatic mechanisms in disease will likely continue to elucidate the influences of lymphatic dysfunction, perhaps subtle, that can invest other, seemingly unrelated, diseases. In future, such discoveries will provide mechanistic insights and may potentiate the development of a new lymphatic-based approach to human disease diagnosis and therapeutics.
Collapse
Affiliation(s)
- Stanley G Rockson
- Department of Cardiovascular Medicine, Stanford Center for Lymphatic and Venous Disorders, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
86
|
Kylkilahti TM, Berends E, Ramos M, Shanbhag NC, Töger J, Markenroth Bloch K, Lundgaard I. Achieving brain clearance and preventing neurodegenerative diseases-A glymphatic perspective. J Cereb Blood Flow Metab 2021; 41:2137-2149. [PMID: 33461408 PMCID: PMC8392766 DOI: 10.1177/0271678x20982388] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/28/2020] [Accepted: 11/20/2020] [Indexed: 12/12/2022]
Abstract
Age-related neurodegenerative diseases are a growing burden to society, and many are sporadic, meaning that the environment, diet and lifestyle play significant roles. Cerebrospinal fluid (CSF)-mediated clearing of brain waste products via perivascular pathways, named the glymphatic system, is receiving increasing interest, as it offers unexplored perspectives on understanding neurodegenerative diseases. The glymphatic system is involved in clearance of metabolic by-products such as amyloid-β from the brain, and its function is believed to lower the risk of developing some of the most common neurodegenerative diseases. Here, we present magnetic resonance imaging (MRI) data on the heart cycle's control of CSF flow in humans which corroborates findings from animal studies. We also review the importance of sleep, diet, vascular health for glymphatic clearance and find that these factors are also known players in brain longevity.
Collapse
Affiliation(s)
- Tekla Maria Kylkilahti
- Department of Experimental Medical Science, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Eline Berends
- Department of Experimental Medical Science, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Marta Ramos
- Department of Experimental Medical Science, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Nagesh C Shanbhag
- Department of Experimental Medical Science, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Johannes Töger
- Diagnostic Radiology, Department of Clinical Sciences, Lund University and Skane University Hospital Lund, Lund, Sweden
| | | | - Iben Lundgaard
- Department of Experimental Medical Science, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| |
Collapse
|
87
|
Choi C, Park J, Kim H, Chang KT, Park J, Min KT. DSCR1 upregulation enhances dural meningeal lymphatic drainage to attenuate amyloid pathology of Alzheimer's disease. J Pathol 2021; 255:296-310. [PMID: 34312845 DOI: 10.1002/path.5767] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 06/29/2021] [Accepted: 07/19/2021] [Indexed: 01/11/2023]
Abstract
Highly developed meningeal lymphatics remove waste products from the brain. Disruption of meningeal lymphatic vessels in a mouse model of amyloid pathology (5XFAD) accelerates the accumulation of amyloid plaques in the meninges and brain, and causes learning and memory deficits, suggesting that clearance of toxic wastes by lymphatic vessels plays a key role in neurodegenerative diseases. Here, we discovered that DSCR1 (Down syndrome critical region 1, known also as RCAN1, regulator of calcineurin 1) facilitates the drainage of waste products by increasing the coverage of dorsal meningeal lymphatic vessels. Furthermore, upregulation of DSCR1 in 5XFAD mice diminishes Aβ pathology in the brain and improves memory defects. Surgical ligation of cervical lymphatic vessels afferent to dcLN blocks the beneficial effects of DSCR1 on Aβ accumulation and cognitive function. Interestingly, intracerebroventricular delivery of AAV1-DSCR1 to 5XFAD mice is sufficient to rebuild the meningeal lymphatic system and re-establish cognitive performance. Collectively, our data indicate that DSCR1 facilitates the growth of dorsal meningeal lymphatics to improve drainage efficiency and protect against Alzheimer's disease (AD) pathologies, further highlighting that improving meningeal lymphatic function is a feasible treatment strategy for AD. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Chiyeol Choi
- Department of Biological Sciences, College of Information and Biotechnology, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Jiwon Park
- Department of Biological Sciences, College of Information and Biotechnology, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Hyerin Kim
- Department of Biological Sciences, College of Information and Biotechnology, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Karen T Chang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jiyoung Park
- Department of Biological Sciences, College of Information and Biotechnology, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Kyung-Tai Min
- Department of Biological Sciences, College of Information and Biotechnology, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| |
Collapse
|
88
|
Rustenhoven J, Tanumihardja C, Kipnis J. Cerebrovascular Anomalies: Perspectives From Immunology and Cerebrospinal Fluid Flow. Circ Res 2021; 129:174-194. [PMID: 34166075 DOI: 10.1161/circresaha.121.318173] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Appropriate vascular function is essential for the maintenance of central nervous system homeostasis and is achieved through virtue of the blood-brain barrier; a specialized structure consisting of endothelial, mural, and astrocytic interactions. While appropriate blood-brain barrier function is typically achieved, the central nervous system vasculature is not infallible and cerebrovascular anomalies, a collective terminology for diverse vascular lesions, are present in meningeal and cerebral vasculature supplying and draining the brain. These conditions, including aneurysmal formation and rupture, arteriovenous malformations, dural arteriovenous fistulas, and cerebral cavernous malformations, and their associated neurological sequelae, are typically managed with neurosurgical or pharmacological approaches. However, increasing evidence implicates interacting roles for inflammatory responses and disrupted central nervous system fluid flow with respect to vascular perturbations. Here, we discuss cerebrovascular anomalies from an immunologic angle and fluid flow perspective. We describe immune contributions, both common and distinct, to the formation and progression of diverse cerebrovascular anomalies. Next, we summarize how cerebrovascular anomalies precipitate diverse neurological sequelae, including seizures, hydrocephalus, and cognitive effects and possible contributions through the recently identified lymphatic and glymphatic systems. Finally, we speculate on and provide testable hypotheses for novel nonsurgical therapeutic approaches for alleviating neurological impairments arising from cerebrovascular anomalies, with a particular emphasis on the normalization of fluid flow and alleviation of inflammation through manipulations of the lymphatic and glymphatic central nervous system clearance pathways.
Collapse
Affiliation(s)
- Justin Rustenhoven
- Center for Brain Immunology and Glia (J.R., J.K.), Washington University in St. Louis, St Louis, MO.,Department of Pathology and Immunology, School of Medicine (J.R., J.K.), Washington University in St. Louis, St Louis, MO
| | | | - Jonathan Kipnis
- Center for Brain Immunology and Glia (J.R., J.K.), Washington University in St. Louis, St Louis, MO.,Department of Pathology and Immunology, School of Medicine (J.R., J.K.), Washington University in St. Louis, St Louis, MO
| |
Collapse
|
89
|
Gao Y, Han H, Du J, He Q, Jia Y, Yan J, Dai H, Cui B, Yang J, Wei X, Yang L, Wang R, Long R, Ren Q, Yang X, Lu J. Early changes to the extracellular space in the hippocampus under simulated microgravity conditions. SCIENCE CHINA-LIFE SCIENCES 2021; 65:604-617. [PMID: 34185240 DOI: 10.1007/s11427-021-1932-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 05/26/2021] [Indexed: 01/11/2023]
Abstract
The smooth transportation of substances through the brain extracellular space (ECS) is crucial to maintaining brain function; however, the way this occurs under simulated microgravity remains unclear. In this study, tracer-based magnetic resonance imaging (MRI) and DECS-mapping techniques were used to image the drainage of brain interstitial fluid (ISF) from the ECS of the hippocampus in a tail-suspended hindlimb-unloading rat model at day 3 (HU-3) and 7 (HU-7). The results indicated that drainage of the ISF was accelerated in the HU-3 group but slowed markedly in the HU-7 group. The tortuosity of the ECS decreased in the HU-3 group but increased in the HU-7 group, while the volume fraction of the ECS increased in both groups. The diffusion rate within the ECS increased in the HU-3 group and decreased in the HU-7 group. The alterations to ISF drainage and diffusion in the ECS were recoverable in the HU-3 group, but neither parameter was restored in the HU-7 group. Our findings suggest that early changes to the hippocampal ECS and ISF drainage under simulated microgravity can be detected by tracer-based MRI, providing a new perspective for studying microgravity-induced nano-scale structure abnormities and developing neuroprotective approaches involving the brain ECS.
Collapse
Affiliation(s)
- Yajuan Gao
- Department of Radiology, Peking University Third Hospital, Beijing, 100191, China.,Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China.,Beijing Key Laboratory of Magnetic Resonance Imaging Technology, Beijing, 100191, China
| | - Hongbin Han
- Department of Radiology, Peking University Third Hospital, Beijing, 100191, China. .,Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China. .,Beijing Key Laboratory of Magnetic Resonance Imaging Technology, Beijing, 100191, China.
| | - Jichen Du
- Beijing Key Laboratory of Magnetic Resonance Imaging Technology, Beijing, 100191, China.,Department of Neurology, Aerospace Center Hospital, Peking University Aerospace Clinical College, Beijing, 100039, China
| | - Qingyuan He
- Department of Radiology, Peking University Third Hospital, Beijing, 100191, China.,Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China.,Beijing Key Laboratory of Magnetic Resonance Imaging Technology, Beijing, 100191, China
| | - Yanxing Jia
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Junhao Yan
- Department of Anatomy and Histology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Hui Dai
- NHC Key Laboratory of Medical Immunology, Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Bin Cui
- Department of Radiology, Aerospace Center Hospital, Peking University Aerospace Clinical College, Beijing, 100039, China
| | - Jing Yang
- Department of Neurology, Aerospace Center Hospital, Peking University Aerospace Clinical College, Beijing, 100039, China
| | - Xunbin Wei
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China
| | - Liu Yang
- Department of Radiology, Peking University Third Hospital, Beijing, 100191, China.,Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China.,Beijing Key Laboratory of Magnetic Resonance Imaging Technology, Beijing, 100191, China
| | - Rui Wang
- Department of Radiology, Peking University Third Hospital, Beijing, 100191, China.,Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China.,Beijing Key Laboratory of Magnetic Resonance Imaging Technology, Beijing, 100191, China
| | - Ren Long
- Department of Radiology, Peking University Third Hospital, Beijing, 100191, China.,Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China.,Beijing Key Laboratory of Magnetic Resonance Imaging Technology, Beijing, 100191, China
| | - Qiushi Ren
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China
| | - Xing Yang
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China
| | - Jiabin Lu
- Department of Radiology, Peking University Third Hospital, Beijing, 100191, China.,Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China.,Beijing Key Laboratory of Magnetic Resonance Imaging Technology, Beijing, 100191, China
| |
Collapse
|
90
|
Lee S, Yoo RE, Choi SH, Oh SH, Ji S, Lee J, Huh KY, Lee JY, Hwang I, Kang KM, Yun TJ, Kim JH, Sohn CH. Contrast-enhanced MRI T1 Mapping for Quantitative Evaluation of Putative Dynamic Glymphatic Activity in the Human Brain in Sleep-Wake States. Radiology 2021; 300:661-668. [PMID: 34156299 DOI: 10.1148/radiol.2021203784] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background Evaluation of the glymphatic system with intrathecal contrast material injection has limited clinical use. Purpose To investigate the feasibility of using serial intravenous contrast-enhanced T1 mapping in the quantitative evaluation of putative dynamic glymphatic activity in various brain regions and to demonstrate the effect of sleep on glymphatic activity in humans. Materials and Methods In this prospective study from May 2019 to February 2020, 25 healthy participants (mean age, 25 years ± 2 [standard deviation]; 15 men) underwent two cycles of MRI (day and night cycles). For each cycle, T1 maps were acquired at baseline and 0.5, 1, 1.5, 2, and 12 hours after intravenous contrast material injection. For the night cycle, participants had a normal night of sleep between 2 and 12 hours. The time (tmin) to reach the minimum T1 value (T1min), the absolute difference between baseline T1 and T1min (peak ΔT1), and the slope between two measurements at 2 and 12 hours (slope[2h-12h]) were determined from T1 value-time curves in cerebral gray matter (GM), cerebral white matter (WM), cerebellar GM, cerebellar WM, and putamen. Mixed-model analysis of variance (ANOVA), Friedman test, and repeated-measures ANOVA were used to assess the effect of sleep on slope(2h-12h) and to compare tmin and peak ΔT1 among different regions. Results The slope(2h-12h) increased from the day to night cycles in cerebral GM, cerebellar GM, and putamen (geometric mean ratio [night/day] = 1.4 [95% CI: 1.2, 1.7], 1.3 [95% CI: 1.1, 1.4], and 2.4 [95% CI: 1.6, 3.6], respectively; P = .001, P < .001, and P < .001, respectively). Median tmin values were 0.5 hour in cerebral and cerebellar GM and putamen for both cycles. Cerebellar GM had the highest mean peak ΔT1, followed by cerebral GM and putamen in both day (159 msec ± 6, 99 msec ± 4, and 62 msec ± 5, respectively) and night (152 msec ± 6, 104 msec ± 6, and 58 msec ± 4, respectively) cycles. Conclusion Clearance of a gadolinium-based contrast agent was greater after sleep compared with daytime wakefulness. These results suggest that sleep was associated with greater glymphatic clearance compared with wakefulness. © RSNA, 2021 Online supplemental material is available for this article. See also the editorial by Anzai and Minoshima in this issue.
Collapse
Affiliation(s)
- Sanghyup Lee
- From the Departments of Radiology (S.L., R.E.Y., S.H.C., J.Y.L., I.H., K.M.K., T.J.Y., J.H.K., C.H.S.) and Clinical Pharmacology and Therapeutics (K.Y.H.), Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehangno, Jongno-gu, Seoul 03080, Republic of Korea; Center for Nanoparticle Research, Institute for Basic Science, Seoul, Republic of Korea (S.H.C.); School of Chemical and Biological Engineering (S.H.C.) and Department of Electrical and Computer Engineering (S.J., J.L.), Seoul National University, Seoul, Republic of Korea; and Department of Biomedical Engineering, Hankuk University of Foreign Studies, Yongin-si, Republic of Korea (S.H.O.)
| | - Roh-Eul Yoo
- From the Departments of Radiology (S.L., R.E.Y., S.H.C., J.Y.L., I.H., K.M.K., T.J.Y., J.H.K., C.H.S.) and Clinical Pharmacology and Therapeutics (K.Y.H.), Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehangno, Jongno-gu, Seoul 03080, Republic of Korea; Center for Nanoparticle Research, Institute for Basic Science, Seoul, Republic of Korea (S.H.C.); School of Chemical and Biological Engineering (S.H.C.) and Department of Electrical and Computer Engineering (S.J., J.L.), Seoul National University, Seoul, Republic of Korea; and Department of Biomedical Engineering, Hankuk University of Foreign Studies, Yongin-si, Republic of Korea (S.H.O.)
| | - Seung Hong Choi
- From the Departments of Radiology (S.L., R.E.Y., S.H.C., J.Y.L., I.H., K.M.K., T.J.Y., J.H.K., C.H.S.) and Clinical Pharmacology and Therapeutics (K.Y.H.), Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehangno, Jongno-gu, Seoul 03080, Republic of Korea; Center for Nanoparticle Research, Institute for Basic Science, Seoul, Republic of Korea (S.H.C.); School of Chemical and Biological Engineering (S.H.C.) and Department of Electrical and Computer Engineering (S.J., J.L.), Seoul National University, Seoul, Republic of Korea; and Department of Biomedical Engineering, Hankuk University of Foreign Studies, Yongin-si, Republic of Korea (S.H.O.)
| | - Se-Hong Oh
- From the Departments of Radiology (S.L., R.E.Y., S.H.C., J.Y.L., I.H., K.M.K., T.J.Y., J.H.K., C.H.S.) and Clinical Pharmacology and Therapeutics (K.Y.H.), Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehangno, Jongno-gu, Seoul 03080, Republic of Korea; Center for Nanoparticle Research, Institute for Basic Science, Seoul, Republic of Korea (S.H.C.); School of Chemical and Biological Engineering (S.H.C.) and Department of Electrical and Computer Engineering (S.J., J.L.), Seoul National University, Seoul, Republic of Korea; and Department of Biomedical Engineering, Hankuk University of Foreign Studies, Yongin-si, Republic of Korea (S.H.O.)
| | - Sooyeon Ji
- From the Departments of Radiology (S.L., R.E.Y., S.H.C., J.Y.L., I.H., K.M.K., T.J.Y., J.H.K., C.H.S.) and Clinical Pharmacology and Therapeutics (K.Y.H.), Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehangno, Jongno-gu, Seoul 03080, Republic of Korea; Center for Nanoparticle Research, Institute for Basic Science, Seoul, Republic of Korea (S.H.C.); School of Chemical and Biological Engineering (S.H.C.) and Department of Electrical and Computer Engineering (S.J., J.L.), Seoul National University, Seoul, Republic of Korea; and Department of Biomedical Engineering, Hankuk University of Foreign Studies, Yongin-si, Republic of Korea (S.H.O.)
| | - Jongho Lee
- From the Departments of Radiology (S.L., R.E.Y., S.H.C., J.Y.L., I.H., K.M.K., T.J.Y., J.H.K., C.H.S.) and Clinical Pharmacology and Therapeutics (K.Y.H.), Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehangno, Jongno-gu, Seoul 03080, Republic of Korea; Center for Nanoparticle Research, Institute for Basic Science, Seoul, Republic of Korea (S.H.C.); School of Chemical and Biological Engineering (S.H.C.) and Department of Electrical and Computer Engineering (S.J., J.L.), Seoul National University, Seoul, Republic of Korea; and Department of Biomedical Engineering, Hankuk University of Foreign Studies, Yongin-si, Republic of Korea (S.H.O.)
| | - Ki Young Huh
- From the Departments of Radiology (S.L., R.E.Y., S.H.C., J.Y.L., I.H., K.M.K., T.J.Y., J.H.K., C.H.S.) and Clinical Pharmacology and Therapeutics (K.Y.H.), Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehangno, Jongno-gu, Seoul 03080, Republic of Korea; Center for Nanoparticle Research, Institute for Basic Science, Seoul, Republic of Korea (S.H.C.); School of Chemical and Biological Engineering (S.H.C.) and Department of Electrical and Computer Engineering (S.J., J.L.), Seoul National University, Seoul, Republic of Korea; and Department of Biomedical Engineering, Hankuk University of Foreign Studies, Yongin-si, Republic of Korea (S.H.O.)
| | - Ji Ye Lee
- From the Departments of Radiology (S.L., R.E.Y., S.H.C., J.Y.L., I.H., K.M.K., T.J.Y., J.H.K., C.H.S.) and Clinical Pharmacology and Therapeutics (K.Y.H.), Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehangno, Jongno-gu, Seoul 03080, Republic of Korea; Center for Nanoparticle Research, Institute for Basic Science, Seoul, Republic of Korea (S.H.C.); School of Chemical and Biological Engineering (S.H.C.) and Department of Electrical and Computer Engineering (S.J., J.L.), Seoul National University, Seoul, Republic of Korea; and Department of Biomedical Engineering, Hankuk University of Foreign Studies, Yongin-si, Republic of Korea (S.H.O.)
| | - Inpyeong Hwang
- From the Departments of Radiology (S.L., R.E.Y., S.H.C., J.Y.L., I.H., K.M.K., T.J.Y., J.H.K., C.H.S.) and Clinical Pharmacology and Therapeutics (K.Y.H.), Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehangno, Jongno-gu, Seoul 03080, Republic of Korea; Center for Nanoparticle Research, Institute for Basic Science, Seoul, Republic of Korea (S.H.C.); School of Chemical and Biological Engineering (S.H.C.) and Department of Electrical and Computer Engineering (S.J., J.L.), Seoul National University, Seoul, Republic of Korea; and Department of Biomedical Engineering, Hankuk University of Foreign Studies, Yongin-si, Republic of Korea (S.H.O.)
| | - Koung Mi Kang
- From the Departments of Radiology (S.L., R.E.Y., S.H.C., J.Y.L., I.H., K.M.K., T.J.Y., J.H.K., C.H.S.) and Clinical Pharmacology and Therapeutics (K.Y.H.), Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehangno, Jongno-gu, Seoul 03080, Republic of Korea; Center for Nanoparticle Research, Institute for Basic Science, Seoul, Republic of Korea (S.H.C.); School of Chemical and Biological Engineering (S.H.C.) and Department of Electrical and Computer Engineering (S.J., J.L.), Seoul National University, Seoul, Republic of Korea; and Department of Biomedical Engineering, Hankuk University of Foreign Studies, Yongin-si, Republic of Korea (S.H.O.)
| | - Tae Jin Yun
- From the Departments of Radiology (S.L., R.E.Y., S.H.C., J.Y.L., I.H., K.M.K., T.J.Y., J.H.K., C.H.S.) and Clinical Pharmacology and Therapeutics (K.Y.H.), Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehangno, Jongno-gu, Seoul 03080, Republic of Korea; Center for Nanoparticle Research, Institute for Basic Science, Seoul, Republic of Korea (S.H.C.); School of Chemical and Biological Engineering (S.H.C.) and Department of Electrical and Computer Engineering (S.J., J.L.), Seoul National University, Seoul, Republic of Korea; and Department of Biomedical Engineering, Hankuk University of Foreign Studies, Yongin-si, Republic of Korea (S.H.O.)
| | - Ji-Hoon Kim
- From the Departments of Radiology (S.L., R.E.Y., S.H.C., J.Y.L., I.H., K.M.K., T.J.Y., J.H.K., C.H.S.) and Clinical Pharmacology and Therapeutics (K.Y.H.), Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehangno, Jongno-gu, Seoul 03080, Republic of Korea; Center for Nanoparticle Research, Institute for Basic Science, Seoul, Republic of Korea (S.H.C.); School of Chemical and Biological Engineering (S.H.C.) and Department of Electrical and Computer Engineering (S.J., J.L.), Seoul National University, Seoul, Republic of Korea; and Department of Biomedical Engineering, Hankuk University of Foreign Studies, Yongin-si, Republic of Korea (S.H.O.)
| | - Chul-Ho Sohn
- From the Departments of Radiology (S.L., R.E.Y., S.H.C., J.Y.L., I.H., K.M.K., T.J.Y., J.H.K., C.H.S.) and Clinical Pharmacology and Therapeutics (K.Y.H.), Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehangno, Jongno-gu, Seoul 03080, Republic of Korea; Center for Nanoparticle Research, Institute for Basic Science, Seoul, Republic of Korea (S.H.C.); School of Chemical and Biological Engineering (S.H.C.) and Department of Electrical and Computer Engineering (S.J., J.L.), Seoul National University, Seoul, Republic of Korea; and Department of Biomedical Engineering, Hankuk University of Foreign Studies, Yongin-si, Republic of Korea (S.H.O.)
| |
Collapse
|
91
|
Abstract
Lymphedema is a common, complex, and inexplicably underappreciated human disease. Despite a history of relative neglect by health care providers and by governmental health care agencies, the last decade has seen an explosive growth of insights into, and approaches to, the problem of human lymphedema. The current review highlights the significant advances that have occurred in the investigative and clinical approaches to lymphedema, particularly over the last decade. This review summarizes the progress that has been attained in the realms of genetics, lymphatic imaging, and lymphatic surgery. Newer molecular insights are explored, along with their relationship to future molecular therapeutics. Growing insights into the relationships among lymphedema, obesity, and other comorbidities are important to consider in current and future responses to patients with lymphedema.
Collapse
Affiliation(s)
- Stanley G Rockson
- Allan and Tina Neill Professor of Lymphatic Research and Medicine, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
92
|
Eide PK, Mariussen E, Uggerud H, Pripp AH, Lashkarivand A, Hassel B, Christensen H, Hovd MH, Ringstad G. Clinical application of intrathecal gadobutrol for assessment of cerebrospinal fluid tracer clearance to blood. JCI Insight 2021; 6:147063. [PMID: 33822769 PMCID: PMC8262318 DOI: 10.1172/jci.insight.147063] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 03/31/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUNDMethodology for estimation of cerebrospinal fluid (CSF) tracer clearance could have wide clinical application in predicting excretion of intrathecal drugs and metabolic solutes from brain metabolism and for diagnostic workup of CSF disturbances.METHODSThe MRI contrast agent gadobutrol (Gadovist) was used as a CSF tracer and injected into the lumbar CSF. Gadobutrol is contained outside blood vessels of the CNS and is eliminated along extravascular pathways, analogous to many CNS metabolites and intrathecal drugs. Tracer enrichment was verified and assessed in CSF by MRI at the level of the cisterna magna in parallel with obtaining blood samples through 48 hours.RESULTSIn a reference patient cohort (n = 29), both enrichment within CSF and blood coincided in time. Blood concentration profiles of gadobutrol through 48 hours varied between patients diagnosed with CSF leakage (n = 4), idiopathic normal pressure hydrocephalus dementia (n = 7), pineal cysts (n = 8), and idiopathic intracranial hypertension (n = 4).CONCLUSIONAssessment of CSF tracer clearance is clinically feasible and may provide a way to predict extravascular clearance of intrathecal drugs and endogenous metabolites from the CNS. The peak concentration in blood (at about 10 hours) was preceded by far peak tracer enhancement at MRI in extracranial lymphatic structures (at about 24 hours), as shown in previous studies, indicating a major role of the spinal canal in CSF clearance capacity.FUNDINGThe work was supported by the Department of Neurosurgery, Oslo University Hospital; the Norwegian Institute for Air Research; and the University of Oslo.
Collapse
Affiliation(s)
- Per K Eide
- Department of Neurosurgery, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | | | - Hilde Uggerud
- Norwegian Institute for Air Research, Kjeller, Norway
| | - Are H Pripp
- Oslo Centre of Biostatistics and Epidemiology, Research Support Services
| | - Aslan Lashkarivand
- Department of Neurosurgery, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Bjørnar Hassel
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Neurohabilitation, and
| | - Hege Christensen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Markus Herberg Hovd
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Geir Ringstad
- Division of Radiology and Nuclear Medicine, Department of Radiology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| |
Collapse
|
93
|
Neuroimmune cleanup crews in brain injury. Trends Immunol 2021; 42:480-494. [PMID: 33941486 DOI: 10.1016/j.it.2021.04.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/05/2021] [Accepted: 04/06/2021] [Indexed: 12/21/2022]
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability. Mounting evidence indicates that the immune system is critically involved in TBI pathogenesis, where it is deployed to dispose of neurotoxic material generated from head trauma and to instruct the wound healing process. However, the immune response to brain damage must be carefully held in check as aberrant regulation of immune signaling can lead to deleterious neuroinflammation, brain pathology, and neurological dysfunction. Efficient clearance of neurotoxic material by microglia (the brain's resident phagocytes) and the glymphatic-meningeal lymphatic drainage system are paramount to keeping the immune system in balance following head trauma. In this review, we highlight emerging evidence that defines pivotal roles for microglia and the recently discovered glymphatic-meningeal lymphatic system in TBI pathogenesis.
Collapse
|
94
|
das Neves SP, Delivanoglou N, Da Mesquita S. CNS-Draining Meningeal Lymphatic Vasculature: Roles, Conundrums and Future Challenges. Front Pharmacol 2021; 12:655052. [PMID: 33995074 PMCID: PMC8113819 DOI: 10.3389/fphar.2021.655052] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 04/13/2021] [Indexed: 12/11/2022] Open
Abstract
A genuine and functional lymphatic vascular system is found in the meninges that sheath the central nervous system (CNS). This unexpected (re)discovery led to a reevaluation of CNS fluid and solute drainage mechanisms, neuroimmune interactions and the involvement of meningeal lymphatics in the initiation and progression of neurological disorders. In this manuscript, we provide an overview of the development, morphology and unique functional features of meningeal lymphatics. An outline of the different factors that affect meningeal lymphatic function, such as growth factor signaling and aging, and their impact on the continuous drainage of brain-derived molecules and meningeal immune cells into the cervical lymph nodes is also provided. We also highlight the most recent discoveries about the roles of the CNS-draining lymphatic vasculature in different pathologies that have a strong neuroinflammatory component, including brain trauma, tumors, and aging-associated neurodegenerative diseases like Alzheimer's and Parkinson's. Lastly, we provide a critical appraisal of the conundrums, challenges and exciting questions involving the meningeal lymphatic system that ought to be investigated in years to come.
Collapse
Affiliation(s)
| | | | - Sandro Da Mesquita
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States
| |
Collapse
|
95
|
Chen J, Wang L, Xu H, Wang Y, Liang Q. The lymphatic drainage system of the CNS plays a role in lymphatic drainage, immunity, and neuroinflammation in stroke. J Leukoc Biol 2021; 110:283-291. [PMID: 33884651 DOI: 10.1002/jlb.5mr0321-632r] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 03/19/2021] [Accepted: 03/26/2021] [Indexed: 12/31/2022] Open
Abstract
The lymphatic drainage system of the central nervous system (CNS) plays an important role in maintaining interstitial fluid balance and regulating immune responses and immune surveillance. The impaired lymphatic drainage system of the CNS might be involved in the onset and progression of various neurodegenerative diseases, neuroinflammation, and cerebrovascular diseases. A significant immune response and brain edema are observed after stroke, resulting from disrupted homeostasis in the brain. Thus, understanding the lymphatic drainage system of the CNS in stroke may lead to the development of new approaches for therapeutic interventions in the future. Here, we review recent evidence implicating the lymphatic drainage system of the CNS in stroke.
Collapse
Affiliation(s)
- Jinman Chen
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Key Laboratory of theory and therapy of muscles and bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), Shanghai, China
| | - Linmei Wang
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hao Xu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Key Laboratory of theory and therapy of muscles and bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), Shanghai, China
| | - Yongjun Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Key Laboratory of theory and therapy of muscles and bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), Shanghai, China
| | - Qianqian Liang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Key Laboratory of theory and therapy of muscles and bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), Shanghai, China
| |
Collapse
|
96
|
Mentis AFA, Dardiotis E, Chrousos GP. Apolipoprotein E4 and meningeal lymphatics in Alzheimer disease: a conceptual framework. Mol Psychiatry 2021; 26:1075-1097. [PMID: 32355332 PMCID: PMC7985019 DOI: 10.1038/s41380-020-0731-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 04/01/2020] [Accepted: 04/09/2020] [Indexed: 12/11/2022]
Abstract
The potential existence and roles of the meningeal lymphatic system in normal and pathological brain function have been a long-standing enigma. Recent evidence suggests that meningeal lymphatic vessels are present in both the mouse and human brain; in mice, they seem to play a role in clearing toxic amyloid-beta peptides, which have been connected with Alzheimer disease (AD). Here, we review the evidence linking the meningeal lymphatic system with human AD. Novel findings suggest that the recently described meningeal lymphatic vessels could be linked to, and possibly drain, the efferent paravascular glial lymphatic (glymphatic) system carrying cerebrospinal fluid, after solute and immune cell exchange with brain interstitial fluid. In so doing, the glymphatic system could contribute to the export of toxic solutes and immune cells from the brain (an exported fluid we wish to describe as glymph, similarly to lymph) to the meningeal lymphatic system; the latter, by being connected with downstream anatomic regions, carries the glymph to the conventional cervical lymphatic vessels and nodes. Thus, abnormal function in the meningeal lymphatic system could, in theory, lead to the accumulation, in the brain, of amyloid-beta, cellular debris, and inflammatory mediators, as well as immune cells, resulting in damage of the brain parenchyma and, in turn, cognitive and other neurologic dysfunctions. In addition, we provide novel insights into APOE4-the leading genetic risk factor for AD-and its relation to the meningeal lymphatic system. In this regard, we have reanalyzed previously published RNA-Seq data to show that induced pluripotent stem cells (iPSCs) carrying the APOE4 allele (either as APOE4 knock-in or stemming from APOE4 patients) express lower levels of (a) genes associated with lymphatic markers, and (b) genes for which well-characterized missense mutations have been linked to peripheral lymphedema. Taking into account this evidence, we propose a new conceptual framework, according to which APOE4 could play a novel role in the premature shrinkage of meningeal lymphatic vessels (meningeal lymphosclerosis), leading to abnormal meningeal lymphatic functions (meningeal lymphedema), and, in turn, reduction in the clearance of amyloid-beta and other macromolecules and inflammatory mediators, as well as immune cells, from the brain, exacerbation of AD manifestations, and progression of the disease. Altogether, these findings and their potential interpretations may herald novel diagnostic tools and therapeutic approaches in patients with AD.
Collapse
Affiliation(s)
- Alexios-Fotios A Mentis
- Public Health Laboratories, Hellenic Pasteur Institute, Vas. Sofias Avenue 127, 115 21, Athens, Greece.
- Department of Microbiology, University of Thessaly, Panepistimiou 3, Viopolis, 41 500, Larissa, Greece.
| | - Efthimios Dardiotis
- Department of Neurology, University of Thessaly, Panepistimiou 3, Viopolis, 41 500, Larissa, Greece
| | - George P Chrousos
- University Research Institute of Maternal and Child Health and Precision Medicine, National and Kapodistrian University of Athens, Medical School, Aghia Sophia Children's Hospital, Livadias 8, 115 27, Athens, Greece
- UNESCO Chair on Adolescent Health Care, Athens, Greece
| |
Collapse
|
97
|
Ma T, Wang F, Xu S, Huang JH. Meningeal immunity: Structure, function and a potential therapeutic target of neurodegenerative diseases. Brain Behav Immun 2021; 93:264-276. [PMID: 33548498 DOI: 10.1016/j.bbi.2021.01.028] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 01/14/2021] [Accepted: 01/23/2021] [Indexed: 12/25/2022] Open
Abstract
Meningeal immunity refers to immune surveillance and immune defense in the meningeal immune compartment, which depends on the unique position, structural composition of the meninges and functional characteristics of the meningeal immune cells. Recent research advances in meningeal immunity have demonstrated many new ways in which a sophisticated immune landscape affects central nervous system (CNS) function under physiological or pathological conditions. The proper function of the meningeal compartment might protect the CNS from pathogens or contribute to neurological disorders. Since the concept of meningeal immunity, especially the meningeal lymphatic system and the glymphatic system, is relatively new, we will provide a general review of the meninges' basic structural elements, organization, regulation, and functions with regards to meningeal immunity. At the same time, we will emphasize recent evidence for the role of meningeal immunity in neurodegenerative diseases. More importantly, we will speculate about the feasibility of the meningeal immune region as a drug target to provide some insights for future research of meningeal immunity.
Collapse
Affiliation(s)
- Tengyun Ma
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, PR China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, PR China
| | - Fushun Wang
- Institute of Brain and Psychological Sciences, Sichuan Normal University, Chengdu 610060, PR China.
| | - Shijun Xu
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, PR China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, PR China.
| | - Jason H Huang
- Department of Neurosurgery, Baylor Scott & White Health Center, Temple, TX 76502, United States; Department of Surgery, Texas A&M University College of Medicine, Temple, TX 76502, United States
| |
Collapse
|
98
|
Ding XB, Wang XX, Xia DH, Liu H, Tian HY, Fu Y, Chen YK, Qin C, Wang JQ, Xiang Z, Zhang ZX, Cao QC, Wang W, Li JY, Wu E, Tang BS, Ma MM, Teng JF, Wang XJ. Impaired meningeal lymphatic drainage in patients with idiopathic Parkinson's disease. Nat Med 2021; 27:411-418. [PMID: 33462448 DOI: 10.1038/s41591-020-01198-1] [Citation(s) in RCA: 168] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 12/01/2020] [Indexed: 01/29/2023]
Abstract
Animal studies implicate meningeal lymphatic dysfunction in the pathogenesis of neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease (PD). However, there is no direct evidence in humans to support this role1-5. In this study, we used dynamic contrast-enhanced magnetic resonance imaging to assess meningeal lymphatic flow in cognitively normal controls and patients with idiopathic PD (iPD) or atypical Parkinsonian (AP) disorders. We found that patients with iPD exhibited significantly reduced flow through the meningeal lymphatic vessels (mLVs) along the superior sagittal sinus and sigmoid sinus, as well as a notable delay in deep cervical lymph node perfusion, compared to patients with AP. There was no significant difference in the size (cross-sectional area) of mLVs in patients with iPD or AP versus controls. In mice injected with α-synuclein (α-syn) preformed fibrils, we showed that the emergence of α-syn pathology was followed by delayed meningeal lymphatic drainage, loss of tight junctions among meningeal lymphatic endothelial cells and increased inflammation of the meninges. Finally, blocking flow through the mLVs in mice treated with α-syn preformed fibrils increased α-syn pathology and exacerbated motor and memory deficits. These results suggest that meningeal lymphatic drainage dysfunction aggravates α-syn pathology and contributes to the progression of PD.
Collapse
Affiliation(s)
- Xue-Bing Ding
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, China
| | - Xin-Xin Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, China
| | - Dan-Hao Xia
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, China
| | - Han Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, China
| | - Hai-Yan Tian
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, China
| | - Yu Fu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, China
| | - Yong-Kang Chen
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, China
| | - Chi Qin
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, China
| | - Jiu-Qi Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, China
| | - Zhi Xiang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, China
| | - Zhong-Xian Zhang
- National Centre for International Research in Cell and Gene Therapy, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Qin-Chen Cao
- Department of Radiation Therapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wei Wang
- Henan Medical Association, Zhengzhou, China
| | - Jia-Yi Li
- Neural Plasticity and Repair Unit, Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, Lund, Sweden.,Institute of Health Sciences, China Medical University, Shenyang, China
| | - Erxi Wu
- Neuroscience Institute and Department of Neurosurgery, Baylor Scott & White Health, Temple, TX, USA.,Texas A & M University Colleges of Medicine and Pharmacy, College Station, TX, USA.,Livestrong Cancer Institutes and Department of Oncology, Dell Medical School, the University of Texas at Austin, Austin, TX, USA
| | - Bei-Sha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Ming-Ming Ma
- Department of Neurology, Affiliated People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, China.
| | - Jun-Fang Teng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China. .,Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, China.
| | - Xue-Jing Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China. .,Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
99
|
Yankova G, Bogomyakova O, Tulupov A. The glymphatic system and meningeal lymphatics of the brain: new understanding of brain clearance. Rev Neurosci 2021; 32:693-705. [PMID: 33618444 DOI: 10.1515/revneuro-2020-0106] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 01/31/2021] [Indexed: 12/25/2022]
Abstract
The glymphatic system and meningeal lymphatics have recently been characterized. Glymphatic system is a glia-dependent system of perivascular channels, and it plays an important role in the removal of interstitial metabolic waste products. The meningeal lymphatics may be a key drainage route for cerebrospinal fluid into the peripheral blood, may contribute to inflammatory reaction and central nervous system (CNS) immune surveillance. Breakdowns and dysfunction of the glymphatic system and meningeal lymphatics play a crucial role in age-related brain changes, the pathogenesis of neurovascular and neurodegenerative diseases, as well as in brain injuries and tumors. This review discusses the relationship recently characterized meningeal lymphatic vessels with the glymphatic system, which provides perfusion of the CNS with cerebrospinal and interstitial fluids. The review also presents the results of human studies concerning both the presence of meningeal lymphatics and the glymphatic system. A new understanding of how aging, medications, sleep and wake cycles, genetic predisposition, and even body posture affect the brain drainage system has not only changed the idea of brain fluid circulation but has also contributed to an understanding of the pathology and mechanisms of neurodegenerative diseases.
Collapse
Affiliation(s)
- Galina Yankova
- Lavrentyev Institute of Hydrodynamics, Siberian Branch, Russian Academy of Sciences, Novosibirsk630090, Russia.,Novosibirsk State University, Novosibirsk630090,Russia
| | - Olga Bogomyakova
- International Tomography Center, Siberian Branch, Russian Academy of Sciences, Novosibirsk630090, Russia
| | - Andrey Tulupov
- Novosibirsk State University, Novosibirsk630090,Russia.,International Tomography Center, Siberian Branch, Russian Academy of Sciences, Novosibirsk630090, Russia.,Meshalkin National Medical Research Center, Ministry of Health of Russian Federation, Novosibirsk 630055, Russia
| |
Collapse
|
100
|
Onder H. Co-occurrence of idiopathic normal-pressure hydrocephalus-like magnetic resonance imaging features in progressive supranuclear palsy. Eur J Neurol 2021; 28:e48-e49. [PMID: 33459447 DOI: 10.1111/ene.14738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 12/30/2020] [Indexed: 11/28/2022]
Affiliation(s)
- Halil Onder
- Department of Neurology, Yozgat City Hospital, Yozgat, Turkey
| |
Collapse
|