201
|
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: 15] [Impact Index Per Article: 7.5] [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
|
202
|
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: 4.3] [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
|
203
|
Semyachkina-Glushkovskaya O, Karavaev A, Prokhorov M, Runnova A, Borovkova E, Yu.M. I, Hramkov A, Kulminskiy D, Semenova N, Sergeev K, Slepnev A, Yu. SE, Zhuravlev M, Fedosov I, Shirokov A, Blokhina I, Dubrovski A, Terskov A, Khorovodov A, Ageev V, Elovenko D, Evsukova A, Adushkina V, Telnova V, Postnov D, Penzel T, Kurths J. EEG biomarkers of activation of the lymphatic drainage system of the brain during sleep and opening of the blood-brain barrier. Comput Struct Biotechnol J 2022; 21:758-768. [PMID: 36698965 PMCID: PMC9841170 DOI: 10.1016/j.csbj.2022.12.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 12/12/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
The lymphatic drainage system of the brain (LDSB) is the removal of metabolites and wastes from its tissues. A dysfunction of LDSB is an important sign of aging, brain oncology, the Alzheimer's and Parkinson's diseases. The development of new strategies for diagnosis of LDSB injuries can improve prevention of age-related cerebral amyloid angiopathy, neurodegenerative and cerebrovascular diseases. There are two conditions, such as deep sleep and opening of the blood-brain-barrier (OBBB) associated with the LDSB activation. A promising candidate for measurement of LDSB could be electroencephalography (EEG). In this pilot study on rats, we tested the hypothesis, whether deep sleep and OBBB can be an informative platform for an effective extracting of information about the LDSB functions. Using the nonlinear analysis of EEG dynamics and machine learning technology, we discovered that the LDSB activation during OBBB and sleep is associated with similar changes in the EEG θ-activity. The OBBB causes the higher LDSB activation vs. sleep that is accompanied by specific changes in the low frequency EEG activity extracted by the power spectra analysis of the EEG dynamics combined with the coherence function. Thus, our findings demonstrate a link between neural activity associated with the LDSB activation during sleep and OBBB that is an important informative platform for extraction of the EEG-biomarkers of the LDSB activity. These results open new perspectives for the development of technology for the LDSB diagnostics that would open a novel era in the prognosis of brain diseases caused by the LDSB disorders, including OBBB.
Collapse
Affiliation(s)
- O.V. Semyachkina-Glushkovskaya
- Physics Department, Humboldt University, Newtonstrasse 15, 12489 Berlin, Germany,Saratov State University, Astrakhanskaya str., 83, Saratov, 410012, Russia,Corresponding author at: Physics Department, Humboldt University, Newtonstrasse 15, 12489 Berlin, Germany.
| | - A.S. Karavaev
- Charité – Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany,Saratov Branchof the Institute of Radio Engineering and Electronics of Russian Academy of Sciences, Zelyonaya, 38, Saratov, 410019, Russia,Saratov State Medical University, B.Kazachaya str., 112, Saratov, 410012, Russia,Institute of Higher Nervous Activity and Neurophysiology of Russian Academy of Sciences, (IHNA&NPh RAS), 5AButlerova St., Moscow 117485, Russia
| | - M.D. Prokhorov
- Saratov State University, Astrakhanskaya str., 83, Saratov, 410012, Russia,Charité – Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany,Saratov Branchof the Institute of Radio Engineering and Electronics of Russian Academy of Sciences, Zelyonaya, 38, Saratov, 410019, Russia
| | - A.E. Runnova
- Saratov State University, Astrakhanskaya str., 83, Saratov, 410012, Russia,Saratov State Medical University, B.Kazachaya str., 112, Saratov, 410012, Russia
| | - E.I. Borovkova
- Saratov State University, Astrakhanskaya str., 83, Saratov, 410012, Russia,Charité – Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany,Saratov State Medical University, B.Kazachaya str., 112, Saratov, 410012, Russia
| | - Ishbulatov Yu.M.
- Saratov State University, Astrakhanskaya str., 83, Saratov, 410012, Russia,Charité – Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany,Saratov Branchof the Institute of Radio Engineering and Electronics of Russian Academy of Sciences, Zelyonaya, 38, Saratov, 410019, Russia,Saratov State Medical University, B.Kazachaya str., 112, Saratov, 410012, Russia
| | - A.N. Hramkov
- Saratov State University, Astrakhanskaya str., 83, Saratov, 410012, Russia
| | - D.D. Kulminskiy
- Saratov State University, Astrakhanskaya str., 83, Saratov, 410012, Russia
| | - N.I. Semenova
- Saratov State University, Astrakhanskaya str., 83, Saratov, 410012, Russia
| | - K.S. Sergeev
- Saratov State University, Astrakhanskaya str., 83, Saratov, 410012, Russia
| | - A.V. Slepnev
- Saratov State University, Astrakhanskaya str., 83, Saratov, 410012, Russia
| | - Sitnikova E. Yu.
- Saratov State University, Astrakhanskaya str., 83, Saratov, 410012, Russia,Institute of Higher Nervous Activity and Neurophysiology of Russian Academy of Sciences, (IHNA&NPh RAS), 5AButlerova St., Moscow 117485, Russia
| | - M.O. Zhuravlev
- Saratov State University, Astrakhanskaya str., 83, Saratov, 410012, Russia,Saratov State Medical University, B.Kazachaya str., 112, Saratov, 410012, Russia
| | - I.V. Fedosov
- Saratov State University, Astrakhanskaya str., 83, Saratov, 410012, Russia
| | - A.A. Shirokov
- Saratov State University, Astrakhanskaya str., 83, Saratov, 410012, Russia,Institute of Biochemistry and Physiology of Plants and Microorganisms, Russian Academy of Sciences, ProspektEntuziastov13, Saratov 410049, Russia
| | - I.A. Blokhina
- Saratov State University, Astrakhanskaya str., 83, Saratov, 410012, Russia
| | - A.I. Dubrovski
- Saratov State University, Astrakhanskaya str., 83, Saratov, 410012, Russia
| | - A.V. Terskov
- Saratov State University, Astrakhanskaya str., 83, Saratov, 410012, Russia
| | - A.P. Khorovodov
- Saratov State University, Astrakhanskaya str., 83, Saratov, 410012, Russia
| | - V.B. Ageev
- Saratov State University, Astrakhanskaya str., 83, Saratov, 410012, Russia
| | - D.A. Elovenko
- Saratov State University, Astrakhanskaya str., 83, Saratov, 410012, Russia
| | - A.S. Evsukova
- Saratov State University, Astrakhanskaya str., 83, Saratov, 410012, Russia
| | - V.V. Adushkina
- Saratov State University, Astrakhanskaya str., 83, Saratov, 410012, Russia
| | - V.V. Telnova
- Saratov State University, Astrakhanskaya str., 83, Saratov, 410012, Russia
| | - D.E. Postnov
- Saratov State University, Astrakhanskaya str., 83, Saratov, 410012, Russia
| | - T.U. Penzel
- Saratov State University, Astrakhanskaya str., 83, Saratov, 410012, Russia,Charité – Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - J.G. Kurths
- Physics Department, Humboldt University, Newtonstrasse 15, 12489 Berlin, Germany,Saratov State University, Astrakhanskaya str., 83, Saratov, 410012, Russia,Potsdam Institute for Climate Impact Research, Telegrafenberg A31, 14473 Potsdam, Germany
| |
Collapse
|
204
|
Chen X, Holtzman DM. Emerging roles of innate and adaptive immunity in Alzheimer's disease. Immunity 2022; 55:2236-2254. [PMID: 36351425 PMCID: PMC9772134 DOI: 10.1016/j.immuni.2022.10.016] [Citation(s) in RCA: 93] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 08/15/2022] [Accepted: 10/18/2022] [Indexed: 11/09/2022]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease, with characteristic extracellular amyloid-β (Aβ) deposition and intracellular accumulation of hyperphosphorylated, aggregated tau. Several key regulators of innate immune pathways are genetic risk factors for AD. While these genetic risk factors as well as in vivo data point to key roles for microglia, emerging evidence also points to a role of the adaptive immune response in disease pathogenesis. We review the roles of innate and adaptive immunity, their niches, their communication, and their contributions to AD development and progression. We also summarize the cellular compositions and physiological functions of immune cells in the parenchyma, together with those in the brain border structures that form a dynamic disease-related immune niche. We propose that both innate and adaptive immune responses in brain parenchyma and border structures could serve as important therapeutic targets for treating both the pre-symptomatic and the symptomatic stages of AD.
Collapse
Affiliation(s)
- Xiaoying Chen
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
205
|
Suresh S, Larson J, Jenrow KA. Chronic neuroinflammation impairs waste clearance in the rat brain. Front Neuroanat 2022; 16:1013808. [PMID: 36569282 PMCID: PMC9768431 DOI: 10.3389/fnana.2022.1013808] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 11/21/2022] [Indexed: 12/13/2022] Open
Abstract
Background Previous reports have established an association between impaired clearance of macromolecular waste from the brain parenchyma and a variety of brain insults for which chronic neuroinflammation is a common pathological feature. Here we investigate whether chronic neuroinflammation is sufficient to impair macromolecular waste clearance from the rat brain. Methods Using a rodent model of chronic neuroinflammation induced by a single high-dose injection of lipopolysaccharide, the clearance kinetics of two fluorophore-conjugated dextran tracers were assayed at 8-weeks post-induction. The expression and distribution of amyloid β and aquaporin-4 proteins within selected brain regions were assayed at 36-weeks post-induction, following open-field, novel object recognition, and contextual fear conditioning assays. Results Chronic neuroinflammation significantly impaired the clearance kinetics of both dextran tracers and resulted in significantly elevated levels of amyloid β within the hippocampus. Aquaporin-4 density on astrocytic endfeet processes was also reduced within multiple brain regions. These pathologies were associated with significantly enhanced contextual fear memory. Conclusion Our results suggest that chronic neuroinflammation is sufficient to compromise the clearance of macromolecular waste from the brain parenchyma and may be the root cause of impaired waste clearance associated with a variety of brain pathologies.
Collapse
Affiliation(s)
- Swathi Suresh
- Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, United States
| | - Jacob Larson
- Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, United States,Department of Physics, Central Michigan University, Mount Pleasant, MI, United States
| | - Kenneth Allen Jenrow
- Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, United States,Department of Psychology, Central Michigan University, Mount Pleasant, MI, United States,*Correspondence: Kenneth Allen Jenrow,
| |
Collapse
|
206
|
Fonken LK, Gaudet AD. Neuroimmunology of healthy brain aging. Curr Opin Neurobiol 2022; 77:102649. [PMID: 36368270 PMCID: PMC9826730 DOI: 10.1016/j.conb.2022.102649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 10/05/2022] [Accepted: 10/06/2022] [Indexed: 11/10/2022]
Abstract
Aging involves progressive deterioration away from homeostasis. Whereas the healthy adult brain maintains neuroimmune cells in a surveillant and homeostatic state, aged glial cells have a hyperreactive phenotype. These age-related pro-inflammatory biases are driven in part by cell-intrinsic factors, including increased cell priming and pro-inflammatory cell states. In addition, the aged inflammatory milieu is shaped by an altered environment, such as amplified danger signals and cytokines and dysregulated glymphatic function. These cell-instrinsic and environmental factors conspire to heighten the age-related risk for neuroimmune activation and associated pathology. In this review, we discuss cellular and molecular neuroimmune shifts with "healthy" aging; how these age-related changes affect physiology and behavior; and how recent research has revealed neuroimmune pathways and targets for improving health span.
Collapse
Affiliation(s)
- Laura K Fonken
- Division of Pharmacology and Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX, USA.
| | - Andrew D Gaudet
- Department of Psychology, College of Liberal Arts, University of Texas at Austin, Austin, TX, USA; Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, USA. https://twitter.com/Gaudet_91
| |
Collapse
|
207
|
Rustenhoven J, Kipnis J. Brain borders at the central stage of neuroimmunology. Nature 2022; 612:417-429. [PMID: 36517712 PMCID: PMC10205171 DOI: 10.1038/s41586-022-05474-7] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [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
Affiliation(s)
- Justin Rustenhoven
- Center for Brain immunology and Glia (BIG), Washington University in St Louis, School of Medicine, St Louis, MO, USA.
- Department of Pathology and Immunology, Washington University in St Louis, School of Medicine, St Louis, MO, USA.
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Auckland, New Zealand.
- Center for Brain Research, The University of Auckland, Auckland, New Zealand.
| | - Jonathan Kipnis
- Center for Brain immunology and Glia (BIG), Washington University in St Louis, School of Medicine, St Louis, MO, USA.
- Department of Pathology and Immunology, Washington University in St Louis, School of Medicine, St Louis, MO, USA.
| |
Collapse
|
208
|
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: 5.7] [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
|
209
|
Deciphering the heterogeneity of the Lyve1 + perivascular macrophages in the mouse brain. Nat Commun 2022; 13:7366. [PMID: 36450771 PMCID: PMC9712536 DOI: 10.1038/s41467-022-35166-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 11/21/2022] [Indexed: 12/05/2022] Open
Abstract
Perivascular macrophages (pvMs) are associated with cerebral vasculature and mediate brain drainage and immune regulation. Here, using reporter mouse models, whole brain and section immunofluorescence, flow cytometry, and single cell RNA sequencing, besides the Lyve1+F4/80+CD206+CX3CR1+ pvMs, we identify a CX3CR1- pvM population that shares phagocytic functions and location. Furthermore, the brain parenchyma vasculature mostly hosts Lyve1+MHCII- pvMs with low to intermediate CD45 expression. Using the double Cx3cr1GFP x Cx3cr1-Cre;RosatdT reporter mice for finer mapping of the lineages, we establish that CD45lowCX3CR1- pvMs are derived from CX3CR1+ precursors and require PU.1 during their ontogeny. In parallel, results from the Cxcr4-CreErt2;Rosa26tdT lineage tracing model support a bone marrow-independent replenishment of all Lyve1+ pvMs in the adult mouse brain. Lastly, flow cytometry and 3D immunofluorescence analysis uncover increased percentage of pvMs following photothrombotic induced stroke. Our results thus show that the parenchymal pvM population is more heterogenous than previously described, and includes a CD45low and CX3CR1- pvM population.
Collapse
|
210
|
Zhou Y, Ran W, Luo Z, Wang J, Fang M, Wei K, Sun J, Lou M. Impaired peri-olfactory cerebrospinal fluid clearance is associated with ageing, cognitive decline and dyssomnia. EBioMedicine 2022; 86:104381. [PMID: 36442319 PMCID: PMC9706530 DOI: 10.1016/j.ebiom.2022.104381] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/09/2022] [Accepted: 11/09/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Animal experiments have demonstrated the dependency of cerebrospinal fluid clearance function on age and sleep, which partially underlay the cognitive decline in the elderly. However, human evidence is lacking, which could be mainly attributed to the limited methods of cerebrospinal fluid clearance function assessment. METHOD Serial T1-weighted and T2-fluid attenuated inversion recovery imaging were performed in 92 patients before and at multiple time points including 4.5 h, 15 h and 39 h after intrathecal injection of contrast agent to visualize the putative meningeal lymphatic pathway, peri-olfactory nerve pathway, and peri-optic nerve pathway. We defined the clearance function as the percentage change in signal unit ratio of critical locations in these pathways from baseline to 39 h after intrathecal injection, and further analysed their relationships with age, sleep, and cognitive function. FINDINGS Cerebrospinal fluid clearance through the putative meningeal lymphatic and perineural pathways were clearly visualized. The clearance function of putative meningeal lymphatic and perineural pathways were impaired with ageing (all P < 0.05). The clearance function through peri-olfactory nerve pathway in inferior turbinate was positively correlated with sleep quality and cognitive function (both P < 0.05), and mediated the association of sleep quality with cognitive function (percent change in β [bootstrap 95% CI]: 33% [-0.220, -0.007]). INTERPRETATION The impaired clearance through putative peri-olfactory nerve pathway may explain the cognitive decline in patients with sleep disturbance. The study shows a promising method to assess cerebrospinal fluid clearance function of putative peri-neural pathways via dynamic magnetic resonance imaging with intrathecal injection of contrast agent. FUNDING This work was supported by the National Natural Science Foundation of China (81971101, 82171276 and 82101365).
Collapse
Affiliation(s)
- Ying Zhou
- Department of Neurology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Wang Ran
- Department of Neurology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Zhongyu Luo
- Department of Neurology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Jianan Wang
- Department of Neurology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Mengmeng Fang
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Kai Wei
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Jianzhong Sun
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Min Lou
- Department of Neurology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China,Corresponding author. Department of Neurology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, #88 Jiefang Road, Hangzhou, China.
| |
Collapse
|
211
|
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: 19] [Impact Index Per Article: 6.3] [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
|
212
|
Drieu A, Du S, Storck SE, Rustenhoven J, Papadopoulos Z, Dykstra T, Zhong F, Kim K, Blackburn S, Mamuladze T, Harari O, Karch CM, Bateman RJ, Perrin R, Farlow M, Chhatwal J, Hu S, Randolph GJ, Smirnov I, Kipnis J. Parenchymal border macrophages regulate the flow dynamics of the cerebrospinal fluid. Nature 2022; 611:585-593. [PMID: 36352225 PMCID: PMC9899827 DOI: 10.1038/s41586-022-05397-3] [Citation(s) in RCA: 117] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 09/29/2022] [Indexed: 11/11/2022]
Abstract
Macrophages are important players in the maintenance of tissue homeostasis1. Perivascular and leptomeningeal macrophages reside near the central nervous system (CNS) parenchyma2, and their role in CNS physiology has not been sufficiently well studied. Given their continuous interaction with the cerebrospinal fluid (CSF) and strategic positioning, we refer to these cells collectively as parenchymal border macrophages (PBMs). Here we demonstrate that PBMs regulate CSF flow dynamics. We identify a subpopulation of PBMs that express high levels of CD163 and LYVE1 (scavenger receptor proteins), closely associated with the brain arterial tree, and show that LYVE1+ PBMs regulate arterial motion that drives CSF flow. Pharmacological or genetic depletion of PBMs led to accumulation of extracellular matrix proteins, obstructing CSF access to perivascular spaces and impairing CNS perfusion and clearance. Ageing-associated alterations in PBMs and impairment of CSF dynamics were restored after intracisternal injection of macrophage colony-stimulating factor. Single-nucleus RNA sequencing data obtained from patients with Alzheimer's disease (AD) and from non-AD individuals point to changes in phagocytosis, endocytosis and interferon-γ signalling on PBMs, pathways that are corroborated in a mouse model of AD. Collectively, our results identify PBMs as new cellular regulators of CSF flow dynamics, which could be targeted pharmacologically to alleviate brain clearance deficits associated with ageing and AD.
Collapse
Affiliation(s)
- Antoine Drieu
- Center for Brain Immunology and Glia (BIG), Washington University in St Louis, St Louis, MO, USA.
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA.
| | - Siling Du
- Center for Brain Immunology and Glia (BIG), Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
- Immunology Graduate Program, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Steffen E Storck
- Center for Brain Immunology and Glia (BIG), Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Justin Rustenhoven
- Center for Brain Immunology and Glia (BIG), Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Zachary Papadopoulos
- Center for Brain Immunology and Glia (BIG), Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
- Immunology Graduate Program, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Taitea Dykstra
- Center for Brain Immunology and Glia (BIG), Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Fenghe Zhong
- Department of Biomedical Engineering, Danforth Campus, Washington University in St Louis, St Louis, MO, USA
| | - Kyungdeok Kim
- Center for Brain Immunology and Glia (BIG), Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Susan Blackburn
- Center for Brain Immunology and Glia (BIG), Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Tornike Mamuladze
- Center for Brain Immunology and Glia (BIG), Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
- Immunology Graduate Program, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Oscar Harari
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
| | - Celeste M Karch
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Randall J Bateman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Richard Perrin
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | | | - Jasmeer Chhatwal
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Song Hu
- Department of Biomedical Engineering, Danforth Campus, Washington University in St Louis, St Louis, MO, USA
| | - Gwendalyn J Randolph
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Igor Smirnov
- Center for Brain Immunology and Glia (BIG), Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Jonathan Kipnis
- Center for Brain Immunology and Glia (BIG), Washington University in St Louis, St Louis, MO, USA.
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA.
- Immunology Graduate Program, School of Medicine, Washington University in St Louis, St Louis, MO, USA.
| |
Collapse
|
213
|
Hidaka Y, Hashimoto M, Suehiro T, Fukuhara R, Ishikawa T, Tsunoda N, Koyama A, Honda K, Miyagawa Y, Yoshiura K, Boku S, Ishii K, Ikeda M, Takebayashi M. Impact of age on the cerebrospinal fluid spaces: high-convexity and medial subarachnoid spaces decrease with age. Fluids Barriers CNS 2022; 19:82. [PMID: 36307853 PMCID: PMC9615391 DOI: 10.1186/s12987-022-00381-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 10/13/2022] [Indexed: 11/26/2022] Open
Abstract
Background Impaired cerebrospinal fluid (CSF) dynamics may contribute to the pathophysiology of neurodegenerative diseases, and play a crucial role in brain health in older people; nonetheless, such age-related changes have not been well elucidated. Disproportionately enlarged subarachnoid-space hydrocephalus (DESH) is a neuroimaging phenotype of idiopathic normal-pressure hydrocephalus, originating from impaired CSF dynamics, and closely associated with aging. This study aimed to investigate the pathophysiology of DESH and determine age-related changes in CSF dynamics. Methods Using magnetic resonance imaging, we investigated the pathophysiology of DESH by quantitatively evaluating the volumes of DESH-related regions (ventricles [VS], Sylvian fissure [SF], and subarachnoid spaces at high convexity and midline [SHM]) and brain parenchyma in community-dwelling individuals aged ≥ 65 years. DESH-related regions were assessed using a visual rating scale, and volumes measured using voxel-based morphometry. Brain parenchyma volumes were measured using FreeSurfer software. Results Data from 1,356 individuals were analyzed, and 25 (1.8%) individuals had DESH. Regarding the relationships between the volume of each CSF space and age, VS and SF volumes increased with age, whereas SHM volume did not increase. VS and SF volumes increased as the whole brain volume decreased, whereas SHM volume did not increase even if the whole brain volume decreased; that is, SHM did not expand even if brain atrophy progressed. Moreover, lower Mini-Mental State Examination scores were significantly associated with lower SHM volume and higher VS volume. These associations remained significant even when individuals with DESH were excluded. Conclusions This study showed that the volume of high-convexity and medial subarachnoid spaces did not expand and tended to decrease with age; the human brain continuously progresses toward a “DESH-like” morphology with aging in community-dwelling older persons (i.e., DESH might be an “accelerated aging stage” rather than an “age-related disorder”). Our results indicated that brain atrophy may be associated with the development of “DESH-like” morphology. In addition, this morphological change, as well as brain atrophy, is an important condition associated with cognitive decline in older adults. Our findings highlight the importance of investigating the aging process of CSF dynamics in the human brain to preserve brain health in older people. Supplementary Information The online version contains supplementary material available at 10.1186/s12987-022-00381-5.
Collapse
|
214
|
Boccalatte F, Mina R, Aroldi A, Leone S, Suryadevara CM, Placantonakis DG, Bruno B. Advances and Hurdles in CAR T Cell Immune Therapy for Solid Tumors. Cancers (Basel) 2022; 14:5108. [PMID: 36291891 PMCID: PMC9600451 DOI: 10.3390/cancers14205108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/03/2022] [Accepted: 10/10/2022] [Indexed: 11/28/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cells in solid tumors have so far yielded limited results, in terms of therapeutic effects, as compared to the dramatic results observed for hematological malignancies. Many factors involve both the tumor cells and the microenvironment. The lack of specific target antigens and severe, potentially fatal, toxicities caused by on-target off-tumor toxicities constitute major hurdles. Furthermore, the tumor microenvironment is usually characterized by chronic inflammation, the presence of immunosuppressive molecules, and immune cells that can reduce CAR T cell efficacy and facilitate antigen escape. Nonetheless, solid tumors are under investigation as possible targets despite their complexity, which represents a significant challenge. In preclinical mouse models, CAR T cells are able to efficiently recognize and kill several tumor xenografts. Overall, in the next few years, there will be intensive research into optimizing novel cell therapies to improve their effector functions and keep untoward effects in check. In this review, we provide an update on the state-of-the-art CAR T cell therapies in solid tumors, focusing on the preclinical studies and preliminary clinical findings aimed at developing optimal strategies to reduce toxicity and improve efficacy.
Collapse
Affiliation(s)
- Francesco Boccalatte
- Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016, USA
- Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA
| | - Roberto Mina
- Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, University of Torino, 10126 Torino, TO, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, TO, Italy
| | - Andrea Aroldi
- Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, MB, Italy
| | - Sarah Leone
- Department of Population Health, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Carter M. Suryadevara
- Department of Neurosurgery, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Dimitris G. Placantonakis
- Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA
- Department of Neurosurgery, NYU Grossman School of Medicine, New York, NY 10016, USA
- Brain and Spine Tumor Center/Neuroscience Institute, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Benedetto Bruno
- Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, University of Torino, 10126 Torino, TO, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, TO, Italy
| |
Collapse
|
215
|
Semyachkina-Glushkovskaya O, Diduk S, Anna E, Elina D, Artem K, Khorovodov A, Shirokov A, Fedosov I, Dubrovsky A, Blokhina I, Terskov A, Navolokin N, Evsukova A, Elovenko D, Adushkina V, Kurths J. Music improves the therapeutic effects of bevacizumab in rats with glioblastoma: Modulation of drug distribution to the brain. Front Oncol 2022; 12:1010188. [PMID: 36313687 PMCID: PMC9606698 DOI: 10.3389/fonc.2022.1010188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Background The development of new methods for modulation of drug distribution across to the brain is a crucial step in the effective therapies for glioblastoma (GBM). In our previous work, we discovered the phenomenon of music-induced opening of the blood-brain barrier (OBBB) in healthy rodents. In this pilot study on rats, we clearly demonstrate that music-induced BBB opening improves the therapeutic effects of bevacizumab (BZM) in rats with GBM via increasing BZM distribution to the brain along the cerebral vessels. Methods The experiments were performed on Wistar male rats (200–250 g, n=161) using transfected C6-TagRFP cell line and the loud rock music for OBBB. The OBBB was assessed by spectrofluorometric assay of Evans Blue (EB) extravasation and confocal imaging of fluorescent BZM (fBZM) delivery into the brain. Additionally, distribution of fBZM and Omniscan in the brain was studied using fluorescent and magnetic resonance imaging (MRI), respectively. To analyze the therapeutic effects of BZM on the GBM growth in rats without and with OBBB, the GBM volume (MRI scans), as well as immunohistochemistry assay of proliferation (Ki67 marker) and apoptosis (Bax marker) in the GBM cells were studied. The Mann–Whitney–Wilcoxon test was used for all analysis, the significance level was p < 0.05, n=7 in each group. Results Our finding clearly demonstrates that music-induced OBBB increases the delivery of EB into the brain tissues and the extravasation of BZM into the brain around the cerebral vessels of rats with GBM. Music significantly increases distribution of tracers (fBZM and Omniscan) in the rat brain through the pathways of brain drainage system (perivascular and lymphatic), which are an important route of drug delivery into the brain. The music-induced OBBB improves the suppressive effects of BZM on the GBM volume and the cellular mechanisms of tumor progression that was accompanied by higher survival among rats in the GBM+BZM+Music group vs. other groups. Conclusion We hypothesized that music improves the therapeutic effects of BZM via OBBB in the normal cerebral vessels and lymphatic drainage of the brain tissues. This contributes better distribution of BZM in the brain fluids and among the normal cerebral vessels, which are used by GBM for invasion and co-opt existing vessels as a satellite tumor form. These results open the new perspectives for an improvement of therapeutic effects of BZM via the music-induced OBBB for BZM in the normal cerebral vessels, which are used by GBM for migration and progression.
Collapse
Affiliation(s)
- Oxana Semyachkina-Glushkovskaya
- Humboldt University, Institute of Physics, Berlin, Germany
- Deparment of Biology, Saratov State University, Saratov, Russia
- *Correspondence: Oxana Semyachkina-Glushkovskaya,
| | - Sergey Diduk
- Laboratory of Pharmaceutical Biotechnology, Pushchino State Institute of Natural Science, Pushchino, Russia
- Department of Biotechnology, Leeners LLС, Moscow, Russia
| | - Eroshova Anna
- Laboratory of Pharmaceutical Biotechnology, Pushchino State Institute of Natural Science, Pushchino, Russia
- Department of Biotechnology, Leeners LLС, Moscow, Russia
| | - Dosadina Elina
- Laboratory of Pharmaceutical Biotechnology, Pushchino State Institute of Natural Science, Pushchino, Russia
- Department of Biotechnology, Leeners LLС, Moscow, Russia
| | - Kruglov Artem
- Laboratory of Pharmaceutical Biotechnology, Pushchino State Institute of Natural Science, Pushchino, Russia
- Department of Biotechnology, Leeners LLС, Moscow, Russia
| | | | - Alexander Shirokov
- Deparment of Biology, Saratov State University, Saratov, Russia
- Institute of Biochemistry and Physiology of Plants and Microorganisms, Saratov Scientific Centre of the Russian Academy of Sciences (IBPPM RAS), Saratov, Russia
| | - Ivan Fedosov
- Deparment of Biology, Saratov State University, Saratov, Russia
| | | | - Inna Blokhina
- Deparment of Biology, Saratov State University, Saratov, Russia
| | - Andrey Terskov
- Deparment of Biology, Saratov State University, Saratov, Russia
| | - Nikita Navolokin
- Deparment of Biology, Saratov State University, Saratov, Russia
- Department of Pathological Anatomy, Saratov Medical State University, Saratov, Russia
| | - Arina Evsukova
- Deparment of Biology, Saratov State University, Saratov, Russia
| | - Daria Elovenko
- Deparment of Biology, Saratov State University, Saratov, Russia
| | | | - Jürgen Kurths
- Humboldt University, Institute of Physics, Berlin, Germany
- Deparment of Biology, Saratov State University, Saratov, Russia
- Potsdam Institute for Climate Impact Research, Department of Complexity Science, Potsdam, Germany
| |
Collapse
|
216
|
van Bree NFHN, Wilhelm M. The Tumor Microenvironment of Medulloblastoma: An Intricate Multicellular Network with Therapeutic Potential. Cancers (Basel) 2022; 14:5009. [PMID: 36291792 PMCID: PMC9599673 DOI: 10.3390/cancers14205009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/08/2022] [Accepted: 10/10/2022] [Indexed: 11/25/2022] Open
Abstract
Medulloblastoma (MB) is a heterogeneous disease in which survival is highly affected by the underlying subgroup-specific characteristics. Although the current treatment modalities have increased the overall survival rates of MB up to 70-80%, MB remains a major cause of cancer-related mortality among children. This indicates that novel therapeutic approaches against MB are needed. New promising treatment options comprise the targeting of cells and components of the tumor microenvironment (TME). The TME of MB consists of an intricate multicellular network of tumor cells, progenitor cells, astrocytes, neurons, supporting stromal cells, microglia, immune cells, extracellular matrix components, and vasculature systems. In this review, we will discuss all the different components of the MB TME and their role in MB initiation, progression, metastasis, and relapse. Additionally, we briefly introduce the effect that age plays on the TME of brain malignancies and discuss the MB subgroup-specific differences in TME components and how all of these variations could affect the progression of MB. Finally, we highlight the TME-directed treatments, in which we will focus on therapies that are being evaluated in clinical trials.
Collapse
Affiliation(s)
| | - Margareta Wilhelm
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institute, 17165 Stockholm, Sweden
| |
Collapse
|
217
|
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: 1.7] [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
|
218
|
Mills WA, Coburn MA, Eyo UB. The emergence of the calvarial hematopoietic niche in health and disease. Immunol Rev 2022; 311:26-38. [PMID: 35880587 PMCID: PMC9489662 DOI: 10.1111/imr.13120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The diploë region of skull has recently been discovered to act as a myeloid cell reservoir to the underlying meninges. The presence of ossified vascular channels traversing the inner skull of cortex provides a passageway for the cells to traffic from the niche, and CNS-derived antigens traveling through cerebrospinal fluid in a perivascular manner reaches the niche to signal myeloid cell egress. This review will highlight the recent findings establishing this burgeoning field along with the known role this niche plays in CNS aging and disease. It will further highlight the anatomical routes and physiological properties of the vascular structures these cells use for trafficking, spanning from skull to brain parenchyma.
Collapse
Affiliation(s)
- William A. Mills
- Brain, Immunology, and Glia CenterUniversity of VirginiaCharlottesvilleVirginiaUSA,Department of NeuroscienceUniversity of VirginiaCharlottesvilleVirginiaUSA,Robert M. Berne Cardiovascular Research CenterUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Morgan A Coburn
- Brain, Immunology, and Glia CenterUniversity of VirginiaCharlottesvilleVirginiaUSA,Department of NeuroscienceUniversity of VirginiaCharlottesvilleVirginiaUSA,Robert M. Berne Cardiovascular Research CenterUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Ukpong B. Eyo
- Brain, Immunology, and Glia CenterUniversity of VirginiaCharlottesvilleVirginiaUSA,Department of NeuroscienceUniversity of VirginiaCharlottesvilleVirginiaUSA,Robert M. Berne Cardiovascular Research CenterUniversity of VirginiaCharlottesvilleVirginiaUSA
| |
Collapse
|
219
|
Mason HD, McGavern DB. How the immune system shapes neurodegenerative diseases. Trends Neurosci 2022; 45:733-748. [PMID: 36075783 PMCID: PMC9746609 DOI: 10.1016/j.tins.2022.08.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/10/2022] [Accepted: 08/01/2022] [Indexed: 12/15/2022]
Abstract
Neurodegenerative diseases are a major cause of death and disability worldwide and are influenced by many factors including age, genetics, and injuries. While these diseases are often thought to result from the accumulation and spread of aberrant proteins, recent studies have demonstrated that they can be shaped by the innate and adaptive immune system. Resident myeloid cells typically mount a sustained response to the degenerating CNS, but peripheral leukocytes such as T and B cells can also alter disease trajectories. Here, we review the sometimes-dichotomous roles played by immune cells during neurodegenerative diseases and explore how brain trauma can serve as a disease initiator or accelerant. We also offer insights into how failure to properly resolve a CNS injury might promote the development of a neurodegenerative disease.
Collapse
Affiliation(s)
- Hannah D Mason
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dorian B McGavern
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
220
|
Abstract
Circulation of cerebrospinal fluid and interstitial fluid around the central nervous system and through the brain transports not only those water-like fluids but also any solutes they carry, including nutrients, drugs, and metabolic wastes. Passing through brain tissue primarily during sleep, this circulation has implications for neurodegenerative disorders including Alzheimer's disease, for tissue damage during stroke and cardiac arrest, and for flow-related disorders such as hydrocephalus and syringomyelia. Recent experimental results reveal several features of this flow, but other aspects are not fully understood, including its driving mechanisms. We review the experimental evidence and theoretical modeling of cerebrospinal fluid flow, including the roles of advection and diffusion in transporting solutes. We discuss both local, detailed fluid-dynamic models of specific components of the system and global hydraulic models of the overall network of flow paths.
Collapse
Affiliation(s)
- Douglas H Kelley
- Department of Mechanical Engineering, University of Rochester, Rochester, New York, USA
| | - John H Thomas
- Department of Mechanical Engineering and Department of Physics and Astronomy, University of Rochester, Rochester, New York, USA
| |
Collapse
|
221
|
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: 107] [Impact Index Per Article: 35.7] [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
|
222
|
Tu T, Peng Z, Song Z, Ma Y, Zhang H. New insight into DAVF pathology—Clues from meningeal immunity. Front Immunol 2022; 13:858924. [PMID: 36189220 PMCID: PMC9520480 DOI: 10.3389/fimmu.2022.858924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 08/26/2022] [Indexed: 11/13/2022] Open
Abstract
In recent years, with the current access in techniques, studies have significantly advanced the knowledge on meningeal immunity, revealing that the central nervous system (CNS) border acts as an immune landscape. The latest concept of meningeal immune system is a tertiary structure, which is a comprehensive overview of the meningeal immune system from macro to micro. We comprehensively reviewed recent advances in meningeal immunity, particularly the new understanding of the dural sinus and meningeal lymphatics. Moreover, based on the clues from the meningeal immunity, new insights were proposed into the dural arteriovenous fistula (DAVF) pathology, aiming to provide novel ideas for DAVF understanding.
Collapse
Affiliation(s)
- Tianqi Tu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- International Neuroscience Institute (China-INI), Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zhenghong Peng
- Department of Health Management Center, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Zihao Song
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- International Neuroscience Institute (China-INI), Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yongjie Ma
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- International Neuroscience Institute (China-INI), Xuanwu Hospital, Capital Medical University, Beijing, China
- *Correspondence: Yongjie Ma, ; Hongqi Zhang,
| | - Hongqi Zhang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- International Neuroscience Institute (China-INI), Xuanwu Hospital, Capital Medical University, Beijing, China
- *Correspondence: Yongjie Ma, ; Hongqi Zhang,
| |
Collapse
|
223
|
The Role of Glymphatic System in Alzheimer’s and Parkinson’s Disease Pathogenesis. Biomedicines 2022; 10:biomedicines10092261. [PMID: 36140362 PMCID: PMC9496080 DOI: 10.3390/biomedicines10092261] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 11/30/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common cause of neurodegenerative dementia, whilst Parkinson’s disease (PD) is a neurodegenerative movement disorder. These two neurodegenerative disorders share the accumulation of toxic proteins as a pathological hallmark. The lack of definitive disease-modifying treatments for these neurogenerative diseases has led to the hypothesis of new pathogenic mechanisms to target and design new potential therapeutic approaches. The recent observation that the glymphatic system is supposed to be responsible for the movement of cerebrospinal fluid into the brain and clearance of metabolic waste has led to study its involvement in the pathogenesis of these classic proteinopathies. Aquaporin-4 (AQP4), a water channel located in the endfeet of astrocyte membrane, is considered a primary driver of the glymphatic clearance system, and defective AQP4-mediated glymphatic drainage has been linked to proteinopathies. The objective of the present review is to present the recent body of knowledge that links the glymphatic system to the pathogenesis of AD and PD disease and other lifestyle factors such as sleep deprivation and exercise that may influence glymphatic system function. We will also focus on the potential neuroimaging approaches that could identify a neuroimaging marker to detect glymphatic system changes.
Collapse
|
224
|
Thompson D, Brissette CA, Watt JA. The choroid plexus and its role in the pathogenesis of neurological infections. Fluids Barriers CNS 2022; 19:75. [PMID: 36088417 PMCID: PMC9463972 DOI: 10.1186/s12987-022-00372-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 08/27/2022] [Indexed: 11/10/2022] Open
Abstract
The choroid plexus is situated at an anatomically and functionally important interface within the ventricles of the brain, forming the blood-cerebrospinal fluid barrier that separates the periphery from the central nervous system. In contrast to the blood-brain barrier, the choroid plexus and its epithelial barrier have received considerably less attention. As the main producer of cerebrospinal fluid, the secretory functions of the epithelial cells aid in the maintenance of CNS homeostasis and are capable of relaying inflammatory signals to the brain. The choroid plexus acts as an immunological niche where several types of peripheral immune cells can be found within the stroma including dendritic cells, macrophages, and T cells. Including the epithelia cells, these cells perform immunosurveillance, detecting pathogens and changes in the cytokine milieu. As such, their activation leads to the release of homing molecules to induce chemotaxis of circulating immune cells, driving an immune response at the choroid plexus. Research into the barrier properties have shown how inflammation can alter the structural junctions and promote increased bidirectional transmigration of cells and pathogens. The goal of this review is to highlight our foundational knowledge of the choroid plexus and discuss how recent research has shifted our understanding towards viewing the choroid plexus as a highly dynamic and important contributor to the pathogenesis of neurological infections. With the emergence of several high-profile diseases, including ZIKA and SARS-CoV-2, this review provides a pertinent update on the cellular response of the choroid plexus to these diseases. Historically, pharmacological interventions of CNS disorders have proven difficult to develop, however, a greater focus on the role of the choroid plexus in driving these disorders would provide for novel targets and routes for therapeutics.
Collapse
Affiliation(s)
- Derick Thompson
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, USA
| | - Catherine A Brissette
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, USA
| | - John A Watt
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, USA.
| |
Collapse
|
225
|
Gimple RC, Yang K, Halbert ME, Agnihotri S, Rich JN. Brain cancer stem cells: resilience through adaptive plasticity and hierarchical heterogeneity. Nat Rev Cancer 2022; 22:497-514. [PMID: 35710946 DOI: 10.1038/s41568-022-00486-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/03/2022] [Indexed: 02/07/2023]
Abstract
Malignant brain tumours are complex ecosystems containing neoplastic and stromal components that generate adaptive and evolutionarily driven aberrant tissues in the central nervous system. Brain cancers are cultivated by a dynamic population of stem-like cells that enforce intratumoural heterogeneity and respond to intrinsic microenvironment or therapeutically guided insults through proliferation, plasticity and restructuring of neoplastic and stromal components. Far from a rigid hierarchy, heterogeneous neoplastic populations transition between cellular states with differential self-renewal capacities, endowing them with powerful resilience. Here we review the biological machinery used by brain tumour stem cells to commandeer tissues in the intracranial space, evade immune responses and resist chemoradiotherapy. Through recent advances in single-cell sequencing, improved models to investigate the role of the tumour microenvironment and a deeper understanding of the fundamental role of the immune system in cancer biology, we are now better equipped to explore mechanisms by which these processes can be exploited for therapeutic benefit.
Collapse
Affiliation(s)
- Ryan C Gimple
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Kailin Yang
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA
| | - Matthew E Halbert
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sameer Agnihotri
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jeremy N Rich
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA, USA.
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
226
|
Cao M, Ong MTY, Yung PSH, Tuan RS, Jiang Y. Role of synovial lymphatic function in osteoarthritis. Osteoarthritis Cartilage 2022; 30:1186-1197. [PMID: 35487439 DOI: 10.1016/j.joca.2022.04.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 04/01/2022] [Accepted: 04/20/2022] [Indexed: 02/02/2023]
Abstract
BACKGROUND Osteoarthritis (OA) affects the entire joint, initially with a low degree of inflammation. Synovitis is correlated with the severity of OA clinical symptoms and cartilage degradation. The synovial lymphatic system (SLS) plays a prominent role in clearing macromolecules within the joint, including the pro-inflammatory cytokines in arthritic status. Scattered evidence shows that impaired SLS drainage function leads to the accumulation of inflammatory factors in the joint and aggravates the progression of OA, and the role of SLS function in OA is less studied. DESIGN This review summarizes the current understanding of synovial lymphatic function in OA progression and potential regulatory pathways and aims to provide a framework of knowledge for the development of OA treatments targeting lymphatic structure and functions. RESULTS SLS locates in the subintima layer of the synovium and consists of lymphatic capillaries and lymphatic collecting vessels. Vascular endothelial growth factor C (VEGF-C) is the most critical regulating factor of lymphatic endothelial cells (LECs) and SLS. Nitric oxide production-induced impairment of lymphatic muscle cells (LMCs) and contractile function may attribute to drainage dysfunction. Preclinical evidence suggests that promoting lymphatic drainage may help restore intra-articular homeostasis to attenuate the progression of OA. CONCLUSION SLS is actively involved in the homeostatic maintenance of the joint. Understanding the drainage function of the SLS at different stages of OA development is essential for further design of therapies targeting the function of these vessels.
Collapse
Affiliation(s)
- M Cao
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - M T Y Ong
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - P S H Yung
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Institute for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - R S Tuan
- Institute for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Y Jiang
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Institute for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.
| |
Collapse
|
227
|
Aryal M, Azadian MM, Hart AR, Macedo N, Zhou Q, Rosenthal EL, Airan RD. Noninvasive ultrasonic induction of cerebrospinal fluid flow enhances intrathecal drug delivery. J Control Release 2022; 349:434-442. [PMID: 35798095 DOI: 10.1016/j.jconrel.2022.06.067] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 06/15/2022] [Accepted: 06/30/2022] [Indexed: 10/17/2022]
Abstract
Intrathecal drug delivery is routinely used in the treatment and prophylaxis of varied central nervous system conditions, as doing so allows drugs to directly bypass the blood-brain barrier. However, the utility of this route of administration is limited by poor brain and spinal cord parenchymal drug uptake from the cerebrospinal fluid. We demonstrate that a simple noninvasive transcranial ultrasound protocol can significantly increase influx of cerebrospinal fluid into the perivascular spaces of the brain, to enhance the uptake of intrathecally administered drugs. Specifically, we administered small (~1 kDa) and large (~155 kDa) molecule agents into the cisterna magna of rats and then applied low, diagnostic-intensity focused ultrasound in a scanning protocol throughout the brain. Using real-time magnetic resonance imaging and ex vivo histologic analyses, we observed significantly increased uptake of small molecule agents into the brain parenchyma, and of both small and large molecule agents into the perivascular space from the cerebrospinal fluid. Notably, there was no evidence of brain parenchymal damage following this intervention. The low intensity and noninvasive approach of transcranial ultrasound in this protocol underscores the ready path to clinical translation of this technique. In this manner, this protocol can be used to directly bypass the blood-brain barrier for whole-brain delivery of a variety of agents. Additionally, this technique can potentially be used as a means to probe the causal role of the glymphatic system in the variety of disease and physiologic processes to which it has been correlated.
Collapse
Affiliation(s)
- Muna Aryal
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, United States; Departments of Engineering and Radiation Oncology, Loyola University Chicago, Chicago, IL, United States
| | - Matine M Azadian
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, United States
| | - Alex R Hart
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, United States
| | - Nicholas Macedo
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, United States
| | - Quan Zhou
- Department of Otolaryngology, Stanford University School of Medicine, Stanford, CA, United States; Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Eben L Rosenthal
- Department of Otolaryngology, Stanford University School of Medicine, Stanford, CA, United States; Stanford Cancer Center, Stanford Medical Center, Stanford, CA, United States; Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Raag D Airan
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, United States; Department of Materials Science and Engineering, Stanford University School of Medicine, Stanford, CA, United States; Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, United States.
| |
Collapse
|
228
|
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
|
229
|
Heming M, Börsch AL, Wiendl H, Meyer Zu Hörste G. High-dimensional investigation of the cerebrospinal fluid to explore and monitor CNS immune responses. Genome Med 2022; 14:94. [PMID: 35978442 PMCID: PMC9385102 DOI: 10.1186/s13073-022-01097-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 07/28/2022] [Indexed: 01/15/2023] Open
Abstract
The cerebrospinal fluid (CSF) features a unique immune cell composition and is in constant contact with the brain borders, thus permitting insights into the brain to diagnose and monitor diseases. Recently, the meninges, which are filled with CSF, were identified as a neuroimmunological interface, highlighting the potential of exploring central nervous system (CNS) immunity by studying CNS border compartments. Here, we summarize how single-cell transcriptomics of such border compartments advance our understanding of neurological diseases, the challenges that remain, and what opportunities novel multi-omic methods offer. Single-cell transcriptomics studies have detected cytotoxic CD4+ T cells and clonally expanded T and B cells in the CSF in the autoimmune disease multiple sclerosis; clonally expanded pathogenic CD8+ T cells were found in the CSF and in the brain adjacent to β-amyloid plaques of dementia patients; in patients with brain metastases, CD8+ T cell clonotypes were shared between the brain parenchyma and the CSF and persisted after therapy. We also outline how novel multi-omic approaches permit the simultaneous measurements of gene expression, chromatin accessibility, and protein in the same cells, which remain to be explored in the CSF. This calls for multicenter initiatives to create single-cell atlases, posing challenges in integrating patients and modalities across centers. While high-dimensional analyses of CSF cells are challenging, they hold potential for personalized medicine by better resolving heterogeneous diseases and stratifying patients.
Collapse
Affiliation(s)
- Michael Heming
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Anna-Lena Börsch
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Heinz Wiendl
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Gerd Meyer Zu Hörste
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany.
| |
Collapse
|
230
|
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: 18] [Impact Index Per Article: 6.0] [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
|
231
|
Da Mesquita S. Charting the meningeal lymphatic network. J Exp Med 2022; 219:e20220891. [PMID: 35789368 PMCID: PMC9257528 DOI: 10.1084/jem.20220891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
A genuine network of lymphatic vessels can be found in the dural layer of the meninges that ensheathe the brain and spinal cord of mammalians. In this issue, Jacob et al. (2022. J. Exp. Med.https://doi.org/10.1084/jem.20220035) employ light sheet fluorescence imaging of intact mouse heads to provide a more comprehensive chart of the meningeal lymphatic vasculature and draw a parallel between lymphatic drainage of cerebrospinal fluid in mice and humans.
Collapse
|
232
|
Li G, Cao Y, Tang X, Huang J, Cai L, Zhou L. The meningeal lymphatic vessels and the glymphatic system: Potential therapeutic targets in neurological disorders. J Cereb Blood Flow Metab 2022; 42:1364-1382. [PMID: 35484910 PMCID: PMC9274866 DOI: 10.1177/0271678x221098145] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 04/03/2022] [Accepted: 04/14/2022] [Indexed: 02/05/2023]
Abstract
The recent discovery of the meningeal lymphatic vessels (mLVs) and glymphatic pathways has challenged the long-lasting dogma that the central nervous system (CNS) lacks a lymphatic system and therefore does not interact with peripheral immunity. This discovery has reshaped our understanding of mechanisms underlying CNS drainage. Under normal conditions, a close connection between mLVs and the glymphatic system enables metabolic waste removal, immune cell trafficking, and CNS immune surveillance. Dysfunction of the glymphatic-mLV system can lead to toxic protein accumulation in the brain, and it contributes to the development of a series of neurodegenerative disorders, such as Alzheimer's and Parkinson's diseases. The identification of precise cerebral transport routes is based mainly on indirect, invasive imaging of animals, and the results cannot always be applied to humans. Here we review the functions of the glymphatic-mLV system and evidence for its involvement in some CNS diseases. We focus on emerging noninvasive imaging techniques to evaluate the human glymphatic-mLV system and their potential for preclinical diagnosis and prevention of neurodegenerative diseases. Potential strategies that target the glymphatic-mLV system in order to treat and prevent neurological disorders are also discussed.
Collapse
Affiliation(s)
- Gaowei Li
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Cao
- Department of Neurosurgery, Chengdu Second People's hospital, Chengdu, China
| | - Xin Tang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Jianhan Huang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Linjun Cai
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Liangxue Zhou
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
233
|
Preventive effects of a standardized flavonoid extract of safflower in rotenone-induced Parkinson's disease rat model. Neuropharmacology 2022; 217:109209. [DOI: 10.1016/j.neuropharm.2022.109209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/22/2022] [Accepted: 07/29/2022] [Indexed: 11/24/2022]
|
234
|
Jacob L, de Brito Neto J, Lenck S, Corcy C, Benbelkacem F, Geraldo LH, Xu Y, Thomas JM, El Kamouh MR, Spajer M, Potier MC, Haik S, Kalamarides M, Stankoff B, Lehericy S, Eichmann A, Thomas JL. Conserved meningeal lymphatic drainage circuits in mice and humans. J Exp Med 2022; 219:e20220035. [PMID: 35776089 PMCID: PMC9253621 DOI: 10.1084/jem.20220035] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 04/07/2022] [Accepted: 06/08/2022] [Indexed: 12/19/2022] Open
Abstract
Meningeal lymphatic vessels (MLVs) were identified in the dorsal and caudobasal regions of the dura mater, where they ensure waste product elimination and immune surveillance of brain tissues. Whether MLVs exist in the anterior part of the murine and human skull and how they connect with the glymphatic system and extracranial lymphatics remained unclear. Here, we used light-sheet fluorescence microscopy (LSFM) imaging of mouse whole-head preparations after OVA-A555 tracer injection into the cerebrospinal fluid (CSF) and performed real-time vessel-wall (VW) magnetic resonance imaging (VW-MRI) after systemic injection of gadobutrol in patients with neurological pathologies. We observed a conserved three-dimensional anatomy of MLVs in mice and humans that aligned with dural venous sinuses but not with nasal CSF outflow, and we discovered an extended anterior MLV network around the cavernous sinus, with exit routes through the foramina of emissary veins. VW-MRI may provide a diagnostic tool for patients with CSF drainage defects and neurological diseases.
Collapse
Affiliation(s)
- Laurent Jacob
- Institut du Cerveau, Pitié-Salpêtrière Hospital, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Paris, France
- Paris Cardiovascular Research Center, Institut National de la Santé et de la Recherche Médicale, Université de Paris, Paris, France
| | - Jose de Brito Neto
- Institut du Cerveau, Pitié-Salpêtrière Hospital, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Paris, France
- Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Stephanie Lenck
- Institut du Cerveau, Pitié-Salpêtrière Hospital, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Paris, France
- Department of Neuroradiology, Pitie-Salpêtrière Hospital, Sorbonne University, Paris, France
| | - Celine Corcy
- Department of Neuroradiology, Pitie-Salpêtrière Hospital, Sorbonne University, Paris, France
| | | | - Luiz Henrique Geraldo
- Paris Cardiovascular Research Center, Institut National de la Santé et de la Recherche Médicale, Université de Paris, Paris, France
- Department of Internal Medicine, Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT
| | - Yunling Xu
- Paris Cardiovascular Research Center, Institut National de la Santé et de la Recherche Médicale, Université de Paris, Paris, France
| | - Jean-Mickael Thomas
- Institut du Cerveau, Pitié-Salpêtrière Hospital, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Paris, France
| | - Marie-Renee El Kamouh
- Institut du Cerveau, Pitié-Salpêtrière Hospital, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Paris, France
| | - Myriam Spajer
- Institut du Cerveau, Pitié-Salpêtrière Hospital, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Paris, France
| | - Marie-Claude Potier
- Institut du Cerveau, Pitié-Salpêtrière Hospital, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Paris, France
| | - Stephane Haik
- Institut du Cerveau, Pitié-Salpêtrière Hospital, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Paris, France
| | - Michel Kalamarides
- Institut du Cerveau, Pitié-Salpêtrière Hospital, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Paris, France
- Department of Neurosurgery, Pitie-Salpêtrière Hospital, Sorbonne University, Paris, France
| | - Bruno Stankoff
- Institut du Cerveau, Pitié-Salpêtrière Hospital, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Paris, France
- Department of Neurology, St Antoine Hospital, Assistance Publique Hôpitaux de Paris – Sorbonne, Paris, France
| | - Stephane Lehericy
- Institut du Cerveau, Pitié-Salpêtrière Hospital, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Paris, France
- Department of Neuroradiology, Pitie-Salpêtrière Hospital, Sorbonne University, Paris, France
- Centre for NeuroImaging Research, Institut du Cerveau et de la Moelle épinière, Paris, France
| | - Anne Eichmann
- Paris Cardiovascular Research Center, Institut National de la Santé et de la Recherche Médicale, Université de Paris, Paris, France
- Department of Internal Medicine, Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT
| | - Jean-Leon Thomas
- Institut du Cerveau, Pitié-Salpêtrière Hospital, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Paris, France
- Department of Neurology, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
235
|
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
|
236
|
Choi D, Park E, Yu RP, Cooper MN, Cho IT, Choi J, Yu J, Zhao L, Yum JEI, Yu JS, Nakashima B, Lee S, Seong YJ, Jiao W, Koh CJ, Baluk P, McDonald DM, Saraswathy S, Lee JY, Jeon NL, Zhang Z, Huang AS, Zhou B, Wong AK, Hong YK. Piezo1-Regulated Mechanotransduction Controls Flow-Activated Lymphatic Expansion. Circ Res 2022; 131:e2-e21. [PMID: 35701867 PMCID: PMC9308715 DOI: 10.1161/circresaha.121.320565] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
BACKGROUND Mutations in PIEZO1 (Piezo type mechanosensitive ion channel component 1) cause human lymphatic malformations. We have previously uncovered an ORAI1 (ORAI calcium release-activated calcium modulator 1)-mediated mechanotransduction pathway that triggers lymphatic sprouting through Notch downregulation in response to fluid flow. However, the identity of its upstream mechanosensor remains unknown. This study aimed to identify and characterize the molecular sensor that translates the flow-mediated external signal to the Orai1-regulated lymphatic expansion. METHODS Various mutant mouse models, cellular, biochemical, and molecular biology tools, and a mouse tail lymphedema model were employed to elucidate the role of Piezo1 in flow-induced lymphatic growth and regeneration. RESULTS Piezo1 was found to be abundantly expressed in lymphatic endothelial cells. Piezo1 knockdown in cultured lymphatic endothelial cells inhibited the laminar flow-induced calcium influx and abrogated the flow-mediated regulation of the Orai1 downstream genes, such as KLF2 (Krüppel-like factor 2), DTX1 (Deltex E3 ubiquitin ligase 1), DTX3L (Deltex E3 ubiquitin ligase 3L,) and NOTCH1 (Notch receptor 1), which are involved in lymphatic sprouting. Conversely, stimulation of Piezo1 activated the Orai1-regulated mechanotransduction in the absence of fluid flow. Piezo1-mediated mechanotransduction was significantly blocked by Orai1 inhibition, establishing the epistatic relationship between Piezo1 and Orai1. Lymphatic-specific conditional Piezo1 knockout largely phenocopied sprouting defects shown in Orai1- or Klf2- knockout lymphatics during embryo development. Postnatal deletion of Piezo1 induced lymphatic regression in adults. Ectopic Dtx3L expression rescued the lymphatic defects caused by Piezo1 knockout, affirming that the Piezo1 promotes lymphatic sprouting through Notch downregulation. Consistently, transgenic Piezo1 expression or pharmacological Piezo1 activation enhanced lymphatic sprouting. Finally, we assessed a potential therapeutic value of Piezo1 activation in lymphatic regeneration and found that a Piezo1 agonist, Yoda1, effectively suppressed postsurgical lymphedema development. CONCLUSIONS Piezo1 is an upstream mechanosensor for the lymphatic mechanotransduction pathway and regulates lymphatic growth in response to external physical stimuli. Piezo1 activation presents a novel therapeutic opportunity for preventing postsurgical lymphedema. The Piezo1-regulated lymphangiogenesis mechanism offers a molecular basis for Piezo1-associated lymphatic malformation in humans.
Collapse
Affiliation(s)
- Dongwon Choi
- Department of Surgery, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA,Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Eunkyung Park
- Department of Surgery, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA,Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Roy P. Yu
- Department of Surgery, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Michael N. Cooper
- Department of Surgery, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Il-Taeg Cho
- Department of Surgery, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA,Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Joshua Choi
- Department of Surgery, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - James Yu
- Department of Surgery, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Luping Zhao
- Department of Surgery, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA,Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Ji-Eun Irene Yum
- Department of Surgery, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA,Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Jin Suh Yu
- Department of Surgery, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA,Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Brandon Nakashima
- Department of Surgery, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Sunju Lee
- Department of Surgery, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA,Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Young Jin Seong
- Department of Surgery, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA,Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Wan Jiao
- Department of Surgery, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Chester J. Koh
- Division of Pediatric Urology, Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - Peter Baluk
- Cardiovascular Research Institute, UCSF Helen Diller Family Comprehensive Cancer Center, and Department of Anatomy, University of California, San Francisco, San Francisco, California, USA
| | - Donald M. McDonald
- Cardiovascular Research Institute, UCSF Helen Diller Family Comprehensive Cancer Center, and Department of Anatomy, University of California, San Francisco, San Francisco, California, USA
| | - Sindhu Saraswathy
- Doheny Eye Institute and Department of Ophthalmology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Jong Y. Lee
- Doheny Eye Institute and Department of Ophthalmology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Noo Li Jeon
- Department of Mechanical and Aerospace Engineering, Seoul National University, Seoul, Republic of Korea
| | - Zhenqian Zhang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Alex S. Huang
- Doheny Eye Institute and Department of Ophthalmology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Bin Zhou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Alex K. Wong
- Division of Plastic Surgery, City of Hope National Medical Center, Duarte, California, USA
| | - Young-Kwon Hong
- Department of Surgery, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA,Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
237
|
Evaluation of the Glymphatic System Using the DTI-ALPS Index in Patients with Spontaneous Intracerebral Haemorrhage. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2694316. [PMID: 35847591 PMCID: PMC9277160 DOI: 10.1155/2022/2694316] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/02/2022] [Indexed: 12/26/2022]
Abstract
Objective To investigate the function of the human glymphatic system (GS) in patients with spontaneous intracerebral haemorrhage (sICH) using diffusion tensor imaging analysis along with the perivascular space (DTI-ALPS). Methods Twenty patients with sICH and 31 healthy controls (HCs) were recruited for DTI and susceptibility-weighted imaging scanning. The diffusivity along the perivascular spaces, as well as the projection fibres and association fibres, was evaluated separately. The DTI-ALPS index of each subject was also calculated. Two-sample t-tests and paired t-tests were performed to analyse the difference in ALPS scores between patients and HCs, as well as that between the lesion side and contralateral side. Pearson correlation analysis was used to observe the relationship between disease duration and GS function. Results The DTI-ALPS index on the lesion side was significantly lower than that of the contralateral side in patients with sICH (p < 0.01, t = −5.77), and it was also significantly lower than that of the ipsilateral side of HCs (p < 0.01, t = −9.50). No significant differences were found in the DTI-ALPS index on the nonlesion side between patients and HCs (p = 0.96, t = 0.05) or between the left and right cerebral hemispheres of HCs (p = 0.41, t = −0.83). The DTI-ALPS index of the lesion side in patients with sICH was significantly correlated with disease duration (p = 0.018, r = 0.537). Conclusions The present study confirmed that GS dysfunction on the ipsilateral side of the lesion is impaired in patients with haemorrhagic stroke, indicating that the GS may be a separate system in the left and right cerebral hemispheres. The DTI-ALPS index can reflect disease duration. These findings have significant implications for understanding sICH from a new perspective.
Collapse
|
238
|
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: 19] [Impact Index Per Article: 6.3] [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
|
239
|
Proulx ST, Engelhardt B. Central nervous system zoning: How brain barriers establish subdivisions for CNS immune privilege and immune surveillance. J Intern Med 2022; 292:47-67. [PMID: 35184353 PMCID: PMC9314672 DOI: 10.1111/joim.13469] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The central nervous system (CNS) coordinates all our body functions. Neurons in the CNS parenchyma achieve this computational task by high speed communication via electrical and chemical signals and thus rely on a strictly regulated homeostatic environment, which does not tolerate uncontrolled entry of blood components including immune cells. The CNS thus has a unique relationship with the immune system known as CNS immune privilege. Previously ascribed to the presence of blood-brain barriers and the lack of lymphatic vessels in the CNS parenchyma prohibiting, respectively, efferent and afferent connections with the peripheral immune system, it is now appreciated that CNS immune surveillance is ensured by cellular and acellular brain barriers that limit immune cell and mediator accessibility to specific compartments at the borders of the CNS. CNS immune privilege is established by a brain barriers anatomy resembling the architecture of a medieval castle surrounded by two walls bordering a castle moat. Built for protection and defense this two-walled rampart at the outer perimeter of the CNS parenchyma allows for accommodation of different immune cell subsets and efficient monitoring of potential danger signals derived from inside or outside of the CNS parenchyma. It enables effective mounting of immune responses within the subarachnoid or perivascular spaces, while leaving the CNS parenchyma relatively undisturbed. In this study, we propose that CNS immune privilege rests on the proper function of the brain barriers, which allow for CNS immune surveillance but prohibit activation of immune responses from the CNS parenchyma unless it is directly injured.
Collapse
Affiliation(s)
- Steven T Proulx
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | | |
Collapse
|
240
|
Khang M, Bindra RS, Mark Saltzman W. Intrathecal delivery and its applications in leptomeningeal disease. Adv Drug Deliv Rev 2022; 186:114338. [PMID: 35561835 DOI: 10.1016/j.addr.2022.114338] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 04/26/2022] [Accepted: 05/06/2022] [Indexed: 12/22/2022]
Abstract
Intrathecal delivery (IT) of opiates into the cerebrospinal fluid (CSF) for anesthesia and pain relief has been used clinically for decades, but this relatively straightforward approach of bypassing the blood-brain barrier has been underutilized for other indications because of its lack of utility in delivering small lipid-soluble drugs. However, emerging evidence suggests that IT drug delivery be an efficacious strategy for the treatment of cancers in which there is leptomeningeal spread of disease. In this review, we discuss CSF flow dynamics and CSF clearance pathways in the context of intrathecal delivery. We discuss human and animal studies of several new classes of therapeutic agents-cellular, protein, nucleic acid, and nanoparticle-based small molecules-that may benefit from IT delivery. The complexity of the CSF compartment presents several key challenges in predicting biodistribution of IT-delivered drugs. New approaches and strategies are needed that can overcome the high rates of turnover in the CSF to reach specific tissues or cellular targets.
Collapse
|
241
|
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: 0.3] [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]
|
242
|
Distinct roles of the meningeal layers in CNS autoimmunity. Nat Neurosci 2022; 25:887-899. [PMID: 35773544 DOI: 10.1038/s41593-022-01108-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 05/23/2022] [Indexed: 12/27/2022]
Abstract
The meninges, comprising the leptomeninges (pia and arachnoid layers) and the pachymeninx (dura layer), participate in central nervous system (CNS) autoimmunity, but their relative contributions remain unclear. Here we report on findings in animal models of CNS autoimmunity and in patients with multiple sclerosis, where, in acute and chronic disease, the leptomeninges were highly inflamed and showed structural changes, while the dura mater was only marginally affected. Although dural vessels were leakier than leptomeningeal vessels, effector T cells adhered more weakly to the dural endothelium. Furthermore, local antigen-presenting cells presented myelin and neuronal autoantigens less efficiently, and the activation of autoreactive T cells was lower in dural than leptomeningeal layers, preventing local inflammatory processes. Direct antigen application was required to evoke a local inflammatory response in the dura. Together, our data demonstrate an uneven involvement of the meningeal layers in CNS autoimmunity, in which effector T cell trafficking and activation are functionally confined to the leptomeninges, while the dura remains largely excluded from CNS autoimmune processes.
Collapse
|
243
|
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: 2.7] [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
|
244
|
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: 4.0] [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
|
245
|
The Underlying Role of the Glymphatic System and Meningeal Lymphatic Vessels in Cerebral Small Vessel Disease. Biomolecules 2022; 12:biom12060748. [PMID: 35740873 PMCID: PMC9221030 DOI: 10.3390/biom12060748] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/21/2022] [Accepted: 05/24/2022] [Indexed: 02/01/2023] Open
Abstract
There is a growing prevalence of vascular cognitive impairment (VCI) worldwide, and most research has suggested that cerebral small vessel disease (CSVD) is the main contributor to VCI. Several potential physiopathologic mechanisms have been proven to be involved in the process of CSVD, such as blood-brain barrier damage, small vessels stiffening, venous collagenosis, cerebral blood flow reduction, white matter rarefaction, chronic ischaemia, neuroinflammation, myelin damage, and subsequent neurodegeneration. However, there still is a limited overall understanding of the sequence and the relative importance of these mechanisms. The glymphatic system (GS) and meningeal lymphatic vessels (mLVs) are the analogs of the lymphatic system in the central nervous system (CNS). As such, these systems play critical roles in regulating cerebrospinal fluid (CSF) and interstitial fluid (ISF) transport, waste clearance, and, potentially, neuroinflammation. Accumulating evidence has suggested that the glymphatic and meningeal lymphatic vessels played vital roles in animal models of CSVD and patients with CSVD. Given the complexity of CSVD, it was significant to understand the underlying interaction between glymphatic and meningeal lymphatic transport with CSVD. Here, we provide a novel framework based on new advances in main four aspects, including vascular risk factors, potential mechanisms, clinical subtypes, and cognition, which aims to explain how the glymphatic system and meningeal lymphatic vessels contribute to the progression of CSVD and proposes a comprehensive insight into the novel therapeutic strategy of CSVD.
Collapse
|
246
|
Ampie L, McGavern DB. Immunological defense of CNS barriers against infections. Immunity 2022; 55:781-799. [PMID: 35545028 PMCID: PMC9087878 DOI: 10.1016/j.immuni.2022.04.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/12/2022] [Accepted: 04/15/2022] [Indexed: 12/24/2022]
Abstract
Neuroanatomical barriers with physical, chemical, and immunological properties play an essential role in preventing the spread of peripheral infections into the CNS. A failure to contain pathogens within these barriers can result in very serious CNS diseases. CNS barriers are inhabited by an elaborate conglomerate of innate and adaptive immune cells that are highly responsive to environmental challenges. The CNS and its barriers can also be protected by memory T and B cells elicited by prior infection or vaccination. Here, we discuss the different CNS barriers from a developmental, anatomical, and immunological standpoint and summarize our current understanding of how memory cells protect the CNS compartment. We then discuss a contemporary challenge to CNS-barrier system (SARS-CoV-2 infection) and highlight approaches to promote immunological protection of the CNS via vaccination.
Collapse
Affiliation(s)
- Leonel Ampie
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA; Department of Surgical Neurology, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Dorian B McGavern
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
247
|
Measurement of CSF pulsation from EPI-based human fMRI. Neuroimage 2022; 257:119293. [PMID: 35551990 DOI: 10.1016/j.neuroimage.2022.119293] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 05/01/2022] [Accepted: 05/08/2022] [Indexed: 11/22/2022] Open
Abstract
It is recently discovered that the glymphatic system and meningeal lymphatic system are the primary routes for the clearance of brain waste products. The CSF flow is part of these systems, facilitating the clearance procedure. Nonetheless, the relationship between CSF flow and brain functional activity has been underexplored. To investigate CSF dynamics and functional brain activity simultaneously, recent studies have proposed a CSF inflow index measured on edge slices (CSFedge) of echo-planar imaging (EPI) based functional magnetic resonance imaging (fMRI), however, it lacks the quantitative aspect of the CSF pulsation. We proposed a new method for quantifying CSF pulsation (CSFpulse) based on an interslice CSF pulsation model in the 4th ventricle of EPI-based fMRI. The proposed CSFpulse successfully detected the higher CSF flow during the resting state than the typical task states (visual and motor) (p<.05), which is consistent with previous studies based on phase contrast (PC) MRI and CSF volume MRI, while it was not detected in CSFedge based indices or baseline CSF signals in various regions of interest (ROIs). Moreover, CSFpulse demonstrated dynamic functional changes in CSF pulsation: it decreased during the activation-on blocks while it increased during the activation-off blocks. CSFpulse significantly correlated with stroke volume measured using PC MRI, a standard method for CSF pulsation quantification, under the same functional state, while CSFedge based indices or CSF ROIs showed no correlation with the PC MRI stroke volume. Lastly, the correlation of CSFpulse with global BOLD was weaker than that of CSFedge, suggesting that CSFpulse may reflect distinct CSF physiological information that is less affected by global BOLD effects. Based on these results, the proposed CSFpulse provides CSF pulsatility information more accurately in a quantitative manner than CSFedge based indices from the recent CSF studies or the conventional ROI-based analysis. In addition to the high correlation with PC MRI, CSFpulse is much faster than PC MRI and provides information of functional brain activations simultaneously, advantageous over PC MRI or CSF volume MRI. Accordingly, the suggested CSFpulse can be used for investigating intra-subject functional changes in BOLD and CSF pulsation simultaneously and inter-subject CSF pulsation variations based on conventional EPI-based fMRI, which warrants further investigation.
Collapse
|
248
|
Choudhury A, Magill ST, Eaton CD, Prager BC, Chen WC, Cady MA, Seo K, Lucas CHG, Casey-Clyde TJ, Vasudevan HN, Liu SJ, Villanueva-Meyer JE, Lam TC, Pu JKS, Li LF, Leung GKK, Swaney DL, Zhang MY, Chan JW, Qiu Z, Martin MV, Susko MS, Braunstein SE, Bush NAO, Schulte JD, Butowski N, Sneed PK, Berger MS, Krogan NJ, Perry A, Phillips JJ, Solomon DA, Costello JF, McDermott MW, Rich JN, Raleigh DR. Meningioma DNA methylation groups identify biological drivers and therapeutic vulnerabilities. Nat Genet 2022; 54:649-659. [PMID: 35534562 PMCID: PMC9374001 DOI: 10.1038/s41588-022-01061-8] [Citation(s) in RCA: 122] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 03/22/2022] [Indexed: 02/06/2023]
Abstract
Meningiomas are the most common primary intracranial tumors. There are no effective medical therapies for meningioma patients, and new treatments have been encumbered by limited understanding of meningioma biology. Here, we use DNA methylation profiling on 565 meningiomas integrated with genetic, transcriptomic, biochemical, proteomic and single-cell approaches to show meningiomas are composed of three DNA methylation groups with distinct clinical outcomes, biological drivers and therapeutic vulnerabilities. Merlin-intact meningiomas (34%) have the best outcomes and are distinguished by NF2/Merlin regulation of susceptibility to cytotoxic therapy. Immune-enriched meningiomas (38%) have intermediate outcomes and are distinguished by immune infiltration, HLA expression and lymphatic vessels. Hypermitotic meningiomas (28%) have the worst outcomes and are distinguished by convergent genetic and epigenetic mechanisms driving the cell cycle and resistance to cytotoxic therapy. To translate these findings into clinical practice, we show cytostatic cell cycle inhibitors attenuate meningioma growth in cell culture, organoids, xenografts and patients.
Collapse
Affiliation(s)
- Abrar Choudhury
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
- Medical Scientist Training Program, University of California, San Francisco, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Stephen T Magill
- Department of Neurological Surgery, Northwestern University, Chicago, IL, USA.
| | - Charlotte D Eaton
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Briana C Prager
- Department of Medicine, University of California, San Diego, San Diego, CA, USA
| | - William C Chen
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Martha A Cady
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Kyounghee Seo
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Calixto-Hope G Lucas
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Tim J Casey-Clyde
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Harish N Vasudevan
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - S John Liu
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Javier E Villanueva-Meyer
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Tai-Chung Lam
- Department of Clinical Oncology, The University of Hong Kong, Pokfulam, Hong Kong
| | - Jenny Kan-Suen Pu
- Division of Neurosurgery, Department of Surgery, The University of Hong Kong, Pokfulam, Hong Kong
| | - Lai-Fung Li
- Division of Neurosurgery, Department of Surgery, The University of Hong Kong, Pokfulam, Hong Kong
| | - Gilberto Ka-Kit Leung
- Division of Neurosurgery, Department of Surgery, The University of Hong Kong, Pokfulam, Hong Kong
| | - Danielle L Swaney
- J. David Gladstone Institutes, California Institute for Quantitative Biosciences, San Francisco, CA, USA
- California Institute for Quantitative Biosciences, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| | - Michael Y Zhang
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Jason W Chan
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, USA
| | - Zhixin Qiu
- Department of Medicine, University of California, San Diego, San Diego, CA, USA
| | - Michael V Martin
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Matthew S Susko
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, USA
| | - Steve E Braunstein
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, USA
| | - Nancy Ann Oberheim Bush
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Jessica D Schulte
- Department of Neurosciences, University of California, San Diego, San Diego, CA, USA
| | - Nicholas Butowski
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Penny K Sneed
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, USA
| | - Mitchel S Berger
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Nevan J Krogan
- J. David Gladstone Institutes, California Institute for Quantitative Biosciences, San Francisco, CA, USA
- California Institute for Quantitative Biosciences, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| | - Arie Perry
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Joanna J Phillips
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - David A Solomon
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Joseph F Costello
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Michael W McDermott
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
- Miami Neuroscience Institute, Baptist Health, Miami, FL, USA
| | - Jeremy N Rich
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - David R Raleigh
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, USA.
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
249
|
Melatonin in ventricular and subarachnoid cerebrospinal fluid: Its function in the neural glymphatic network and biological significance for neurocognitive health. Biochem Biophys Res Commun 2022; 605:70-81. [DOI: 10.1016/j.bbrc.2022.03.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/02/2022] [Accepted: 03/04/2022] [Indexed: 12/13/2022]
|
250
|
Li W, Liu YH, Estrada H, Rebling J, Reiss M, Galli S, Nombela-Arrieta C, Razansky D. Tracking Strain-Specific Morphogenesis and Angiogenesis of Murine Calvaria with Large-Scale Optoacoustic and Ultrasound Microscopy. J Bone Miner Res 2022; 37:1032-1043. [PMID: 35220594 PMCID: PMC9311448 DOI: 10.1002/jbmr.4533] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 01/30/2022] [Accepted: 02/20/2022] [Indexed: 11/25/2022]
Abstract
Skull bone development is a dynamic and well-coordinated process playing a key role in maturation and maintenance of the bone marrow (BM), fracture healing, and progression of diseases such as osteoarthritis or osteoporosis. At present, dynamic transformation of the growing bone (osteogenesis) as well as its vascularization (angiogenesis) remain largely unexplored due to the lack of suitable in vivo imaging techniques capable of noninvasive visualization of the whole developing calvaria at capillary-level resolution. We present a longitudinal study on skull bone development using ultrasound-aided large-scale optoacoustic microscopy (U-LSOM). Skull bone morphogenesis and microvascular growth patterns were monitored in three common mouse strains (C57BL/6J, CD-1, and Athymic Nude-Foxn1nu) at the whole-calvaria scale over a 3-month period. Strain-specific differences in skull development were revealed by quantitative analysis of bone and vessel parameters, indicating the coupling between angiogenesis and osteogenesis during skull bone growth in a minimally invasive and label-free manner. The method further enabled identifying BM-specific sinusoidal vessels, and superficial skull vessels penetrating into BM compartments. Our approach furnishes a new high-throughput longitudinal in vivo imaging platform to study morphological and vascular skull alterations in health and disease, shedding light on the critical links between blood vessel formation, skull growth, and regeneration. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Weiye Li
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, Zurich, Switzerland.,Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, Switzerland
| | - Yu-Hang Liu
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, Zurich, Switzerland.,Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, Switzerland
| | - Héctor Estrada
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, Zurich, Switzerland.,Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, Switzerland
| | - Johannes Rebling
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, Zurich, Switzerland.,Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, Switzerland
| | - Michael Reiss
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, Zurich, Switzerland.,Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, Switzerland
| | - Serena Galli
- Department of Medical Oncology and Hematology, University and University Hospital Zurich, Zurich, Switzerland
| | - César Nombela-Arrieta
- Department of Medical Oncology and Hematology, University and University Hospital Zurich, Zurich, Switzerland
| | - Daniel Razansky
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, Zurich, Switzerland.,Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, Switzerland
| |
Collapse
|