1
|
Liu Z, Cheng T, Dong H, Sun D, Wang Y, Li J, Yu Z, Cao L. Roles of central nervous system resident and recruited macrophages in the brain barrier system. Neural Regen Res 2026; 21:855-868. [PMID: 39885670 DOI: 10.4103/nrr.nrr-d-24-00986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 01/06/2025] [Indexed: 02/01/2025] Open
Abstract
Macrophages in the brain barrier system include microglia in the brain parenchyma, border-associated macrophages at the brain's borders, and recruited macrophages. They are responsible for neural development, maintenance of homeostasis, and orchestrating immune responses. With the rapid exploitation and development of new technologies, there is a deeper understanding of macrophages in the brain barrier system. Here we review the origin, development, important molecules, and functions of macrophages, mainly focusing on microglia and border-associated macrophages. We also highlight some advances in single-cell sequencing and significant cell markers. We anticipate that more advanced methods will emerge to study resident and recruited macrophages in the future, opening new horizons for neuroimmunology and related peripheral immune fields.
Collapse
Affiliation(s)
- Ze Liu
- Institute of Neuroscience Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science SMMU, Shanghai, China
| | - Teng Cheng
- Institute of Neuroscience Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science SMMU, Shanghai, China
| | - Hongtian Dong
- Institute of Neuroscience Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science SMMU, Shanghai, China
| | - Dingya Sun
- Institute of Neuroscience Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science SMMU, Shanghai, China
| | - Yan Wang
- Department of Pharmacy, Shanghai Baoshan District Wusong Central Hospital (Zhongshan Hospital Wusong Branch, Fudan University), Shanghai, China
| | - Jiayan Li
- Neurovascular Center, Changhai Hospital SMMU, Shanghai, China
| | - Zhongwang Yu
- Institute of Neuroscience Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science SMMU, Shanghai, China
| | - Li Cao
- Institute of Neuroscience Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science SMMU, Shanghai, China
| |
Collapse
|
2
|
Machado RS, Mathias K, Joaquim L, da Costa MDA, Tiscoski A, Gonçalves CL, Rezin GT, Petronilho F. Emerging Roles of Meningeal Lymphatic Vessels in Ischemic Stroke. Mol Neurobiol 2025:10.1007/s12035-025-04983-6. [PMID: 40289061 DOI: 10.1007/s12035-025-04983-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
This review highlights the emerging relevance of meningeal lymphatic vessels (MLVs) in the context of ischemic stroke, challenging the conventional view of a privileged immunological central nervous system. MLVs facilitate immunological surveillance by modulating the entry of peripheral immune cells into the meningeal compartment, a process not impeded by the blood-brain barrier. In ischemic stroke, these vessels play a crucial role in the neuroinflammatory cascade, contributing to immune responses by draining antigens and signals to cervical lymph nodes. Their involvement extends to potential contributions to resolving ischemia-induced cerebral edema, impacting fluid homeostasis. The dynamic interaction among MLVs, neuroinflammation, and fluid dynamics suggests promising therapeutic approaches. Targeting these vessels for immunomodulation, fluid drainage, and preserving blood-brain barrier integrity emerges as an innovative approach to improve ischemic stroke outcomes. However, successful clinical translation awaits further exploration of the therapeutic potential of these vessels. The multifaceted contributions of MLVs provide a compelling rationale for ongoing research, aiming to fully harness their therapeutic impact in ischemic stroke management.
Collapse
Affiliation(s)
- Richard Simon Machado
- Laboratory of Experimental Neurology, Health Sciences Unit, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, Santa Catarina, Brazil
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Health Sciences Unit, Graduate Program in Health Sciences, University of South Santa Catarina, Tubarão, Santa Catarina, Brazil
| | - Khiany Mathias
- Laboratory of Experimental Neurology, Health Sciences Unit, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, Santa Catarina, Brazil
| | - Larissa Joaquim
- Laboratory of Experimental Neurology, Health Sciences Unit, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, Santa Catarina, Brazil
| | - Maiara de Aguiar da Costa
- Laboratory of Experimental Neurology, Health Sciences Unit, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, Santa Catarina, Brazil
| | - Anita Tiscoski
- Laboratory of Experimental Neurology, Health Sciences Unit, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, Santa Catarina, Brazil
| | - Cinara Ludvig Gonçalves
- Laboratory of Experimental Neurology, Health Sciences Unit, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, Santa Catarina, Brazil
| | - Gislaine Tezza Rezin
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Health Sciences Unit, Graduate Program in Health Sciences, University of South Santa Catarina, Tubarão, Santa Catarina, Brazil
| | - Fabricia Petronilho
- Laboratory of Experimental Neurology, Health Sciences Unit, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, Santa Catarina, Brazil.
| |
Collapse
|
3
|
Manavi Z, Melchor GS, Bullard MR, Gross PS, Ray S, Gaur P, Baydyuk M, Huang JK. Senescent cell reduction does not improve recovery in mice under experimental autoimmune encephalomyelitis (EAE) induced demyelination. J Neuroinflammation 2025; 22:101. [PMID: 40197319 PMCID: PMC11974124 DOI: 10.1186/s12974-025-03425-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 03/21/2025] [Indexed: 04/10/2025] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) characterized by immune cell-driven demyelination and progressive neurodegeneration. Senescent cells (SCs) have recently been observed in chronic MS lesions indicating their possible involvement in disease progression. However, the role of SCs and the potential therapeutic benefit of their reduction through senolytic therapy remains to be determined in experimental autoimmune encephalomyelitis (EAE), a widely used preclinical model of MS. Here, we show that senescent-like myeloid cells accumulate in the spinal cord parenchyma and meninges in mice after myelin oligodendrocyte glycoprotein (MOG33-55) EAE induction. Treatment with the senolytic cocktail, Dasatinib and Quercetin (DQ), effectively reduces the senescent-like myeloid cells, but this does not translate into improved clinical outcomes in EAE mice. Increasing DQ dosage or using INK-ATTAC transgenic mice also failed to ameliorate EAE severity. Additionally, histopathological analysis shows no significant differences in demyelination or axonal degeneration between treated and control groups. Our findings indicate that senescent-like myeloid cells are present in an immune-mediated demyelinating model of MS and can be reduced through senolytic therapy with Dasatinib and Quercetin. However, their reduction through DQ does not significantly impact inflammation or recovery, suggesting that the therapeutic potential of senolytics as disease-modifying drugs in MS may be limited.
Collapse
Affiliation(s)
- Zeeba Manavi
- Department of Biology, Georgetown University, Washington, DC, USA
| | - George S Melchor
- Department of Biology, Georgetown University, Washington, DC, USA
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC, USA
| | - Meghan R Bullard
- Department of Biology, Georgetown University, Washington, DC, USA
| | - Phillip S Gross
- Department of Biology, Georgetown University, Washington, DC, USA
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC, USA
| | - Shinjini Ray
- Department of Biology, Georgetown University, Washington, DC, USA
| | - Pankaj Gaur
- Department of Oncology, Georgetown University, Washington, DC, USA
| | - Maryna Baydyuk
- Department of Biology, Georgetown University, Washington, DC, USA
| | - Jeffrey K Huang
- Department of Biology, Georgetown University, Washington, DC, USA.
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC, USA.
| |
Collapse
|
4
|
Hiraki C, Tsuruta F. The Meninges as CNS Interfaces and the Roles of Meningeal Macrophages. Biomolecules 2025; 15:497. [PMID: 40305192 PMCID: PMC12024811 DOI: 10.3390/biom15040497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/19/2025] [Accepted: 03/26/2025] [Indexed: 05/02/2025] Open
Abstract
The brain, the most important component of the central nervous system (CNS), is protected by multiple intricate barriers that strictly regulate the entry of proteins and cells. Thus, the brain is often described as an organ with immune privilege. Within the brain parenchyma, microglia are thought to be the primary resident immune cells, with no other immune-related cells present under normal conditions. On the other hand, recent studies in the meningeal border regions have revealed the presence of meningeal-specific lymphatic vessels and channels that connect to the skull bone marrow. Importantly, resident macrophage populations specific to these boundary regions, known as CNS-associated macrophages (CAMs) or border-associated macrophages (BAMs), have been identified. In contrast to the brain parenchyma, the meninges contain many immune-related structures and cells, making them an important immune interface at the CNS border. CAMs serve a dual function, triggering immune responses under pathological conditions and supporting the maintenance of brain homeostasis. This review focuses on the immune architecture of the meninges and the roles of CAMs in humans and mice, summarizing and discussing recent advances in this field.
Collapse
Affiliation(s)
- Chihiro Hiraki
- Master’s and Doctoral Program in Biology, Degree Programs in Life and Earth Sciences, Graduate School of Science and Technology, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8577, Ibaraki, Japan;
| | - Fuminori Tsuruta
- Master’s and Doctoral Program in Biology, Institute of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8577, Ibaraki, Japan
- Doctoral Program in Human Biology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8577, Ibaraki, Japan
- Doctoral Program in Humanics, School of Integrative and Global Majors, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8577, Ibaraki, Japan
- Master’s and Doctoral Program in Neuroscience, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8577, Ibaraki, Japan
- Center for Quantum and Information Life Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8577, Ibaraki, Japan
| |
Collapse
|
5
|
Zhang Y, Yang Z, Jiang N, Tan X, Jiang P, Cao G, Yang Q. MAIT cell deficiency exacerbates neuroinflammation in P301S human tau transgenic mice. J Neuroinflammation 2025; 22:90. [PMID: 40114233 PMCID: PMC11927249 DOI: 10.1186/s12974-025-03413-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Accepted: 03/08/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND The role of immune cells in neurodegeneration remains incompletely understood. Accumulation of misfolded tau proteins is a hallmark of neurodegenerative diseases. Our recent study revealed the presence of mucosal-associated invariant T (MAIT) cells in the meninges, where they express antioxidant molecules to maintain meningeal barrier integrity. However, the role of MAIT cells in tau-related neuroinflammation and neurodegeneration remains unknown. METHODS Flow cytometry analysis was performed to examine MAIT cells in human Tau P301S transgenic mice. Tau pathology, hippocampus atrophy, meningeal integrity, and microglial gene expression were examined in Mr1-/- P301S mice that lacked MAIT cells and control P301S transgenic mice, as well as Mr1-/- P301S mice with adoptive transfer of MAIT cells. RESULTS The meninges of P301S mutant human tau transgenic mice had increased numbers of MAIT cells, which retained their expression of antioxidant molecules. Mr1-/-P301S mice that lacked MAIT cells exhibited increased tau pathology and hippocampus atrophy compared to control Mr1+/+P301S mice. Adoptive transfer of MAIT cells reduced tau pathology and hippocampus atrophy in Mr1-/- P301S mice. Meningeal barrier integrity was compromised in Mr1-/-P301S mice, but not in control Mr1+/+P301S mice. A distinctive microglia subset with a proinflammatory gene expression profile (M-inflammatory) was enriched in the hippocampus of Mr1-/-P301S mice. The transcriptomes of the remaining microglia in these mice also shifted towards a proinflammatory state, with increased expression of inflammatory cytokines, chemokines, and genes related to ribosome biogenesis and immune responses to toxic substances. The transfer of MAIT cells restored meningeal barrier integrity and suppressed microglial inflammation in the Mr1-/- P301S mice. CONCLUSIONS Our data indicate an important role for MAIT cells in regulating tau-pathology-related neuroinflammation and neurodegeneration.
Collapse
Affiliation(s)
- Yuanyue Zhang
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, 89 French St, New Brunswick, NJ, 08901, USA
| | - Zhi Yang
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, 89 French St, New Brunswick, NJ, 08901, USA
| | - Na Jiang
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, 89 French St, New Brunswick, NJ, 08901, USA
| | - Xiaosheng Tan
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, 89 French St, New Brunswick, NJ, 08901, USA
| | - Peng Jiang
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, 08854, USA
| | - Gaoyuan Cao
- Rutgers Institute for Translational Medicine and Science, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA
| | - Qi Yang
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, 89 French St, New Brunswick, NJ, 08901, USA.
- Rutgers Institute for Translational Medicine and Science, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA.
- Department of Pediatrics, Johnson Medical School, Rutgers Robert Wood, New Brunswick, NJ, 08901, USA.
| |
Collapse
|
6
|
Londoño AC, Mora CA. High efficacy therapy to prevent the formation of meningeal tertiary lymphoid organs after CXCL13 index screening in early multiple sclerosis. Front Neurosci 2025; 19:1558810. [PMID: 40165834 PMCID: PMC11955623 DOI: 10.3389/fnins.2025.1558810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Accepted: 02/24/2025] [Indexed: 04/02/2025] Open
Abstract
Postmortem studies have shown the presence of subpial inflammation with tertiary lymphoid organs (TLO) in the meninges of patients with progressive multiple sclerosis, playing an important role in the pathophysiology of the disease. The chemokine (C-X-C motif) ligand 13 (CXCL13) induces the formation of these lymphoid organs, thus promoting activity of disease. The progression to disability in multiple sclerosis has been reduced, thanks to the effect of disease modifying therapy. However, despite advances in the treatment of disease with immunomodulatory agents, we still lack specific laboratory biomarkers that could indicate the state of activity of disease, either at time of diagnosis or when escalation therapy seems to be mandatory. In patients with multiple sclerosis, MRI studies have not demonstrated the presence of TLO in the CNS, so far. The determination of the CXCL13 index (ICXCL 13), in clinical specimens, could become a reliable biomarker for the verification of the presence and activity of the TLO, thus contributing to improving therapy outcome, with high efficacy therapy, in the clinical setting.
Collapse
Affiliation(s)
- Ana C. Londoño
- Instituto Neurologico de Colombia (INDEC), Medellin, Colombia
- Retired (2022), Medellin, Colombia
| | - Carlos A. Mora
- Department of Medicine, Neurology Unit, Virginia Tech Carilion School of Medicine, Roanoke, VA, United States
| |
Collapse
|
7
|
Hammarlund-Udenaes M, Loryan I. Assessing central nervous system drug delivery. Expert Opin Drug Deliv 2025; 22:421-439. [PMID: 39895003 DOI: 10.1080/17425247.2025.2462767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/15/2025] [Accepted: 01/31/2025] [Indexed: 02/04/2025]
Abstract
INTRODUCTION Delivering drugs to the central nervous system (CNS) remains a major challenge due to the blood-brain barrier, restricting the entry of drugs into the brain. This limitation contributes to the ongoing lack of effective treatments for CNS diseases. To improve the process of drug discovery and development, it is crucial to streamline methods that measure clinically relevant parameters, allowing for good selection of drug candidates. AREA COVERED In this paper, we discuss the essential prerequisites for successful CNS drug delivery and review relevant methods. We emphasize the need for closer collaboration between in vitro and in vivo scientists to improve the relevance of these methods and increase the success rate of developing effective CNS therapies. While our focus is on small molecule drugs, we also touch on some aspects of larger molecules. EXPERT OPINION Significant progress has been made in recent years in method development and their application. However, there is still work to be done before the use of in silico models, in vitro cell systems, and AI can consistently offer meaningful correlations and relationships to clinical data. This gap is partly due to limited patient data, but a lot can be achieved through in vivo research in animal models.
Collapse
Affiliation(s)
| | - Irena Loryan
- Translational PKPD Group, Department of Pharmacy, Uppsala University, Uppsala, Sweden
| |
Collapse
|
8
|
Shirvani A, Shirvani P, Jonah U, Moore BE, Holick MF. Suspected Mitochondrial Dysfunction and Complex Pathophysiology in Fatal Hypermobile Ehlers-Danlos Syndrome: Insights from a Case Report and Post-Mortem Findings. Biomedicines 2025; 13:469. [PMID: 40002882 PMCID: PMC11852713 DOI: 10.3390/biomedicines13020469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Hypermobile Ehlers-Danlos Syndrome (hEDS) is a complex connective tissue disorder with multi-systemic manifestations that significantly impact quality of life. This case report investigates the clinical course and molecular mechanisms of advanced hEDS through an in-depth case study and post-mortem findings. Methods: The clinical history of a 24-year-old patient with advanced hEDS was analyzed, focusing on progressive complications across multiple systems. Post-mortem examination and genetic analysis were performed to elucidate the underlying pathophysiology. Results: The patient's clinical course was marked by gastrointestinal, neurological, and immune complications requiring numerous surgical interventions. Post-mortem findings revealed severe gastrointestinal dysmotility and Alzheimer's Type II astrocytes. Genetic analysis identified variants in mtDNA genes ATP6, CYB, and ND, suggesting a potential role of impaired mitochondrial function in hEDS pathogenesis but requiring further validation through functional studies. Conclusions: This case report provides valuable insights into the potential role of mitochondrial dysfunction in advanced hEDS and highlights the need for further research in this area. Future studies should include comprehensive functional assays, longitudinal tissue sampling, family genetic analyses, and muscle biopsies to better understand the complex interplay between genetic factors, mitochondrial function, and clinical manifestations in hEDS. Establishing genetic bases and developing targeted therapies addressing both structural and metabolic aspects are crucial. The patient's legacy offers invaluable information that could significantly contribute to enhancing diagnostic accuracy and developing personalized treatment strategies for this challenging disorder, potentially leading to better care for individuals living with hEDS.
Collapse
Affiliation(s)
- Arash Shirvani
- Ehlers-Danlos Syndrome Clinical Research Program, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA; (A.S.); (P.S.)
| | - Purusha Shirvani
- Ehlers-Danlos Syndrome Clinical Research Program, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA; (A.S.); (P.S.)
| | - Ugochukwu Jonah
- The Department of Pathology & Laboratory Medicine at Boston Medical Center and Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA; (U.J.); (B.E.M.)
| | - Brian E. Moore
- The Department of Pathology & Laboratory Medicine at Boston Medical Center and Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA; (U.J.); (B.E.M.)
| | - Michael F. Holick
- Ehlers-Danlos Syndrome Clinical Research Program, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA; (A.S.); (P.S.)
| |
Collapse
|
9
|
Garcia FJ, Heiman M. Molecular and cellular characteristics of cerebrovascular cell types and their contribution to neurodegenerative diseases. Mol Neurodegener 2025; 20:13. [PMID: 39881338 PMCID: PMC11780804 DOI: 10.1186/s13024-025-00799-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 01/07/2025] [Indexed: 01/31/2025] Open
Abstract
Many diseases and disorders of the nervous system suffer from a lack of adequate therapeutics to halt or slow disease progression, and to this day, no cure exists for any of the fatal neurodegenerative diseases. In part this is due to the incredible diversity of cell types that comprise the brain, knowledge gaps in understanding basic mechanisms of disease, as well as a lack of reliable strategies for delivering new therapeutic modalities to affected areas. With the advent of single cell genomics, it is now possible to interrogate the molecular characteristics of diverse cell populations and their alterations in diseased states. More recently, much attention has been devoted to cell populations that have historically been difficult to profile with bulk single cell technologies. In particular, cell types that comprise the cerebrovasculature have become increasingly better characterized in normal and neurodegenerative disease contexts. In this review, we describe the current understanding of cerebrovasculature structure, function, and cell type diversity and its role in the mechanisms underlying various neurodegenerative diseases. We focus on human and mouse cerebrovasculature studies and discuss both origins and consequences of cerebrovascular dysfunction, emphasizing known cell type-specific vulnerabilities in neuronal and cerebrovascular cell populations. Lastly, we highlight how novel insights into cerebrovascular biology have impacted the development of modern therapeutic approaches and discuss outstanding questions in the field.
Collapse
Affiliation(s)
- Francisco J Garcia
- The Picower Institute for Learning and Memory, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| | - Myriam Heiman
- The Picower Institute for Learning and Memory, Cambridge, MA, USA.
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA.
| |
Collapse
|
10
|
Zhan T, Tian S, Chen S. Border-Associated Macrophages: From Embryogenesis to Immune Regulation. CNS Neurosci Ther 2024; 30:e70105. [PMID: 39496482 PMCID: PMC11534460 DOI: 10.1111/cns.70105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/17/2024] [Accepted: 10/16/2024] [Indexed: 11/06/2024] Open
Abstract
Border-associated macrophages (BAMs) play a pivotal role in maintaining brain homeostasis and responding to pathological conditions. Understanding their origins, characteristics, and roles in both healthy and diseased brains is crucial for advancing our knowledge of neuroinflammatory and neurodegenerative diseases. This review addresses the ontogeny, replenishment, microenvironmental regulation, and transcriptomic heterogeneity of BAMs, highlighting recent advancements in lineage tracing and fate-mapping studies. Furthermore, we examine the roles of BAMs in maintaining brain homeostasis, immune surveillance, and responses to injury and neurodegenerative diseases. Further research is crucial to clarify the dynamic interplay between BAMs and the brain's microenvironment in health and disease. This effort will not only resolve existing controversies but also reveal new therapeutic targets for neuroinflammatory and neurodegenerative disorders, pushing the boundaries of neuroscience.
Collapse
Affiliation(s)
- Tiantong Zhan
- Department of Neurosurgery, School of Medicine, The Second Affiliated HospitalZhejiang UniversityHangzhouChina
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological DiseasesHangzhouZhejiangChina
| | - Sixuan Tian
- Department of Neurosurgery, School of Medicine, The Second Affiliated HospitalZhejiang UniversityHangzhouChina
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological DiseasesHangzhouZhejiangChina
| | - Sheng Chen
- Department of Neurosurgery, School of Medicine, The Second Affiliated HospitalZhejiang UniversityHangzhouChina
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological DiseasesHangzhouZhejiangChina
| |
Collapse
|
11
|
Betsholtz C, Engelhardt B, Koh GY, McDonald DM, Proulx ST, Siegenthaler J. Advances and controversies in meningeal biology. Nat Neurosci 2024; 27:2056-2072. [PMID: 39333784 PMCID: PMC11862877 DOI: 10.1038/s41593-024-01701-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 06/12/2024] [Indexed: 09/30/2024]
Abstract
The dura, arachnoid and pia mater, as the constituent layers of the meninges, along with cerebrospinal fluid in the subarachnoid space and ventricles, are essential protectors of the brain and spinal cord. Complemented by immune cells, blood vessels, lymphatic vessels and nerves, these connective tissue layers have held many secrets that have only recently begun to be revealed. Each meningeal layer is now known to have molecularly distinct types of fibroblasts. Cerebrospinal fluid clearance through peripheral lymphatics and lymph nodes is well documented, but its routes and flow dynamics are debated. Advances made in meningeal immune functions are also debated. This Review considers the cellular and molecular structure and function of the dura, arachnoid and pia mater in the context of conventional views, recent progress, and what is uncertain or unknown. The hallmarks of meningeal pathophysiology are identified toward developing a more complete understanding of the meninges in health and disease.
Collapse
Affiliation(s)
- Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden and Department of Medicine-Huddinge, Karolinska Institutet, Huddinge, Sweden
| | | | - Gou Young Koh
- Center for Vascular Research, Institute for Basic Science and Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Donald M McDonald
- Department of Anatomy, Cardiovascular Research Institute, and UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Steven T Proulx
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Julie Siegenthaler
- Department of Pediatrics, Section of Developmental Biology, University of Colorado, Anschutz Medical Campus Aurora, Colorado, CO, USA.
| |
Collapse
|
12
|
Severance S, Daylor V, Petrucci T, Gensemer C, Patel S, Norris RA. Hypermobile Ehlers-Danlos syndrome and spontaneous CSF leaks: the connective tissue conundrum. Front Neurol 2024; 15:1452409. [PMID: 39087003 PMCID: PMC11289524 DOI: 10.3389/fneur.2024.1452409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 07/03/2024] [Indexed: 08/02/2024] Open
Abstract
Collagen, the most abundant protein in the body, is a key component of the extracellular matrix (ECM), which plays a crucial role in the structure and support of connective tissues. Abnormalities in collagen associated with connective tissue disorders (CTD) can lead to neuroinflammation and weaken the integrity of the blood-brain barrier (BBB), a semi-permeable membrane that separates the brain's extracellular fluid from the bloodstream. This compromise in the BBB can result from disruptions in ECM components, leading to neuroinflammatory responses, neuronal damage, and increased risks of neurological disorders. These changes impact central nervous system homeostasis and may exacerbate neurological conditions linked to CTD, manifesting as cognitive impairment, sensory disturbances, headaches, sleep issues, and psychiatric symptoms. The Ehlers-Danlos syndromes (EDS) are a group of heritable CTDs that result from varying defects in collagen and the ECM. The most prevalent subtype, hypermobile EDS (hEDS), involves clinical manifestations that include joint hypermobility, skin hyperextensibility, autonomic dysfunction, mast cell activation, chronic pain, as well as neurological manifestations like chronic headaches and cerebrospinal fluid (CSF) leaks. Understanding the connections between collagen, CSF, inflammation, and the BBB could provide insights into neurological diseases associated with connective tissue abnormalities and guide future research.
Collapse
Affiliation(s)
- Sydney Severance
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States
| | - Victoria Daylor
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States
| | - Taylor Petrucci
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC, United States
| | - Cortney Gensemer
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC, United States
| | - Sunil Patel
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC, United States
| | - Russell A. Norris
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
13
|
Karan AA, Gerasimov KA, Spivak YS, Suleymanova EM, Vinogradova LV. Inflammatory response of leptomeninges to a single cortical spreading depolarization. J Headache Pain 2024; 25:113. [PMID: 39009958 PMCID: PMC11251126 DOI: 10.1186/s10194-024-01823-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 07/07/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND Neurogenic meningeal inflammation is regarded as a key driver of migraine headache. Multiple evidence show importance of inflammatory processes in the dura mater for pain generation but contribution of the leptomeninges is less clear. We assessed effects of cortical spreading depolarization (CSD), the pathophysiological mechanism of migraine aura, on expression of inflammatory mediators in the leptomeninges. METHODS A single CSD event was produced by a focal unilateral microdamage of the cortex in freely behaving rats. Three hours later intact cortical leptomeninges and parenchyma of ipsi-lesional (invaded by CSD) and sham-treated contra-lesional (unaffected by CSD) hemispheres were collected and mRNA levels of genes associated with inflammation (Il1b, Tnf, Ccl2; Cx3cl1, Zc3h12a) and endocannabinoid CB2 receptors (Cnr2) were measured using qPCR. RESULTS Three hours after a single unilateral CSD, most inflammatory factors changed their expression levels in the leptomeninges, mainly on the side of CSD. The meninges overlying affected cortex increased mRNA expression of all proinflammatory cytokines (Il1b, Tnf, Ccl2) and anti-inflammatory factors Zc3h12a and Cx3cl1. Upregulation of proinflammatory cytokines was found in both meninges and parenchyma while anti-inflammatory markers increased only meningeal expression. CONCLUSION A single CSD is sufficient to produce pronounced leptomeningeal inflammation that lasts for at least three hours and involves mostly meninges overlying the cortex affected by CSD. The prolonged post-CSD inflammation of the leptomeninges can contribute to mechanisms of headache generation following aura phase of migraine attack.
Collapse
Affiliation(s)
- Anna A Karan
- Department of Molecular Neurobiology, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Butlerova Street 5A, 117485, Moscow, Russia
| | - Konstantin A Gerasimov
- Department of Molecular Neurobiology, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Butlerova Street 5A, 117485, Moscow, Russia
- Pirogov Russian National Research Medical University, Ostrovityanova Street 1, 117997, Moscow, Russia
| | - Yulia S Spivak
- Department of Molecular Neurobiology, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Butlerova Street 5A, 117485, Moscow, Russia
| | - Elena M Suleymanova
- Department of Molecular Neurobiology, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Butlerova Street 5A, 117485, Moscow, Russia
| | - Lyudmila V Vinogradova
- Department of Molecular Neurobiology, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Butlerova Street 5A, 117485, Moscow, Russia.
| |
Collapse
|
14
|
Martin CG, Bent JS, Hill T, Topalidou I, Singhvi A. Epithelial UNC-23 limits mechanical stress to maintain glia-neuron architecture in C. elegans. Dev Cell 2024; 59:1668-1688.e7. [PMID: 38670103 PMCID: PMC11233253 DOI: 10.1016/j.devcel.2024.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/23/2023] [Accepted: 04/03/2024] [Indexed: 04/28/2024]
Abstract
For an organ to maintain correct architecture and function, its diverse cellular components must coordinate their size and shape. Although cell-intrinsic mechanisms driving homotypic cell-cell coordination are known, it is unclear how cell shape is regulated across heterotypic cells. We find that epithelial cells maintain the shape of neighboring sense-organ glia-neuron units in adult Caenorhabditis elegans (C. elegans). Hsp co-chaperone UNC-23/BAG2 prevents epithelial cell shape from deforming, and its loss causes head epithelia to stretch aberrantly during animal movement. In the sense-organ glia, amphid sheath (AMsh), this causes progressive fibroblast growth factor receptor (FGFR)-dependent disruption of the glial apical cytoskeleton. Resultant glial cell shape alteration causes concomitant shape change in glia-associated neuron endings. Epithelial UNC-23 maintenance of glia-neuron shape is specific both spatially, within a defined anatomical zone, and temporally, in a developmentally critical period. As all molecular components uncovered are broadly conserved across central and peripheral nervous systems, we posit that epithelia may similarly regulate glia-neuron architecture cross-species.
Collapse
Affiliation(s)
- Cecilia G Martin
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - James S Bent
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Tyler Hill
- Department of Biology, Brandeis University, Waltham, MA 02454, USA
| | - Irini Topalidou
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Aakanksha Singhvi
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; Department of Biological Structure, University of Washington School of Medicine, Seattle, WA 98195, USA.
| |
Collapse
|
15
|
Whiteley AE, Ma D, Wang L, Yu SY, Yin C, Price TT, Simon BG, Xu KR, Marsh KA, Brockman ML, Prioleau TM, Zhou KI, Cui X, Fecci PE, Jeck WR, McCall CM, Neff JL, Sipkins DA. Breast cancer exploits neural signaling pathways for bone-to-meninges metastasis. Science 2024; 384:eadh5548. [PMID: 38900896 PMCID: PMC12167639 DOI: 10.1126/science.adh5548] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 04/23/2024] [Indexed: 06/22/2024]
Abstract
The molecular mechanisms that regulate breast cancer cell (BCC) metastasis and proliferation within the leptomeninges (LM) are poorly understood, which limits the development of effective therapies. In this work, we show that BCCs in mice can invade the LM by abluminal migration along blood vessels that connect vertebral or calvarial bone marrow and meninges, bypassing the blood-brain barrier. This process is dependent on BCC engagement with vascular basement membrane laminin through expression of the neuronal pathfinding molecule integrin α6. Once in the LM, BCCs colocalize with perivascular meningeal macrophages and induce their expression of the prosurvival neurotrophin glial-derived neurotrophic factor (GDNF). Intrathecal GDNF blockade, macrophage-specific GDNF ablation, or deletion of the GDNF receptor neural cell adhesion molecule (NCAM) from BCCs inhibits breast cancer growth within the LM. These data suggest integrin α6 and the GDNF signaling axis as new therapeutic targets against breast cancer LM metastasis.
Collapse
Affiliation(s)
- Andrew E. Whiteley
- Department of Medicine, Division of Hematologic Malignancies and Cellular Therapy, Duke University, Durham, NC, 27710, USA
- Presently at: Vividion Therapeutics, San Diego, CA 92121
| | - Danhui Ma
- Department of Medicine, Division of Hematologic Malignancies and Cellular Therapy, Duke University, Durham, NC, 27710, USA
| | - Lihua Wang
- Department of Medicine, Division of Hematologic Malignancies and Cellular Therapy, Duke University, Durham, NC, 27710, USA
| | - Seok-Yeong Yu
- Department of Medicine, Division of Hematologic Malignancies and Cellular Therapy, Duke University, Durham, NC, 27710, USA
| | - Claire Yin
- Department of Medicine, Division of Hematologic Malignancies and Cellular Therapy, Duke University, Durham, NC, 27710, USA
| | - Trevor T. Price
- Department of Medicine, Division of Hematologic Malignancies and Cellular Therapy, Duke University, Durham, NC, 27710, USA
- Presently at: Aktis Oncology; Durham, NC 27710, USA
| | - Brennan G. Simon
- Department of Medicine, Division of Hematologic Malignancies and Cellular Therapy, Duke University, Durham, NC, 27710, USA
- Presently at: Stanford University School of Medicine; Stanford, CA,94305, USA
| | - Katie R. Xu
- Department of Medicine, Division of Hematologic Malignancies and Cellular Therapy, Duke University, Durham, NC, 27710, USA
- Presently at: Case Western Reserve University School of Medicine; Cleveland, OH 44106, USA
| | - Kathleen A. Marsh
- Department of Medicine, Division of Hematologic Malignancies and Cellular Therapy, Duke University, Durham, NC, 27710, USA
- Presently at: Wake Forest University School of Medicine; Winston-Salem, NC 27101, USA
| | - Maegan L. Brockman
- Department of Medicine, Division of Hematologic Malignancies and Cellular Therapy, Duke University, Durham, NC, 27710, USA
- Presently at: Laney Graduate School, Emory University; Atlanta, GA 30307, USA
| | - Tatiana M. Prioleau
- Department of Medicine, Division of Hematologic Malignancies and Cellular Therapy, Duke University, Durham, NC, 27710, USA
| | - Katherine I. Zhou
- Department of Medicine, Division of Hematologic Malignancies and Cellular Therapy, Duke University, Durham, NC, 27710, USA
| | - Xiuyu Cui
- Department of Neurosurgery, Duke University; Durham, NC 27710, USA
| | - Peter E. Fecci
- Department of Neurosurgery, Duke University; Durham, NC 27710, USA
| | - William R. Jeck
- Department of Pathology, Duke University; Durham, NC 27710, USA
| | - Chad M. McCall
- Presently at: Carolinas Pathology Group, Charlotte, NC 28217, USA
| | - Jadee L. Neff
- Department of Pathology, Duke University; Durham, NC 27710, USA
| | - Dorothy A. Sipkins
- Department of Medicine, Division of Hematologic Malignancies and Cellular Therapy, Duke University, Durham, NC, 27710, USA
| |
Collapse
|
16
|
Muzio L, Perego J. CNS Resident Innate Immune Cells: Guardians of CNS Homeostasis. Int J Mol Sci 2024; 25:4865. [PMID: 38732082 PMCID: PMC11084235 DOI: 10.3390/ijms25094865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/22/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Although the CNS has been considered for a long time an immune-privileged organ, it is now well known that both the parenchyma and non-parenchymal tissue (meninges, perivascular space, and choroid plexus) are richly populated in resident immune cells. The advent of more powerful tools for multiplex immunophenotyping, such as single-cell RNA sequencing technique and upscale multiparametric flow and mass spectrometry, helped in discriminating between resident and infiltrating cells and, above all, the different spectrum of phenotypes distinguishing border-associated macrophages. Here, we focus our attention on resident innate immune players and their primary role in both CNS homeostasis and pathological neuroinflammation and neurodegeneration, two key interconnected aspects of the immunopathology of multiple sclerosis.
Collapse
Affiliation(s)
- Luca Muzio
- Neuroimmunology Lab, IRCCS San Raffaele Scientific Institute, Institute of Experimental Neurology, 20133 Milan, Italy;
| | | |
Collapse
|
17
|
Koller BH, Nguyen M, Snouwaert JN, Gabel CA, Ting JPY. Species-specific NLRP3 regulation and its role in CNS autoinflammatory diseases. Cell Rep 2024; 43:113852. [PMID: 38427558 PMCID: PMC12054400 DOI: 10.1016/j.celrep.2024.113852] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/16/2024] [Accepted: 02/07/2024] [Indexed: 03/03/2024] Open
Abstract
The NLRP3 inflammasome is essential for caspase-1 activation and the release of interleukin (IL)-1β, IL-18, and gasdermin-D in myeloid cells. However, research on species-specific NLRP3's physiological impact is limited. We engineer mice with the human NLRP3 gene, driven by either the human or mouse promoter, via syntenic replacement at the mouse Nlrp3 locus. Both promoters facilitate hNLRP3 expression in myeloid cells, but the mouse promoter responds more robustly to LPS. Investigating the disease impact of differential NLRP3 regulation, we introduce the D305N gain-of-function mutation into both humanized lines. Chronic inflammation is evident with both promoters; however, CNS outcomes vary significantly. Despite poor response to LPS, the human promoter results in D305N-associated aseptic meningitis, mirroring human pathology. The mouse promoter, although leading to increased CNS expression post-LPS, does not induce meningitis in D305N mutants. Therefore, human-like NLRP3 expression may be crucial for accurate modeling of its role in disease pathogenesis.
Collapse
Affiliation(s)
- Beverly H Koller
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - MyTrang Nguyen
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - John N Snouwaert
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | - Jenny P-Y Ting
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Center for Translational Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
18
|
Sun R, Jiang H. Border-associated macrophages in the central nervous system. J Neuroinflammation 2024; 21:67. [PMID: 38481312 PMCID: PMC10938757 DOI: 10.1186/s12974-024-03059-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 03/05/2024] [Indexed: 03/17/2024] Open
Abstract
Tissue-resident macrophages play an important role in the local maintenance of homeostasis and immune surveillance. In the central nervous system (CNS), brain macrophages are anatomically divided into parenchymal microglia and non-parenchymal border-associated macrophages (BAMs). Among these immune cell populations, microglia have been well-studied for their roles during development as well as in health and disease. BAMs, mostly located in the choroid plexus, meningeal and perivascular spaces, are now gaining increased attention due to advancements in multi-omics technologies and genetic methodologies. Research on BAMs over the past decade has focused on their ontogeny, immunophenotypes, involvement in various CNS diseases, and potential as therapeutic targets. Unlike microglia, BAMs display mixed origins and distinct self-renewal capacity. BAMs are believed to regulate neuroimmune responses associated with brain barriers and contribute to immune-mediated neuropathology. Notably, BAMs have been observed to function in diverse cerebral pathologies, including Alzheimer's disease, Parkinson's disease, multiple sclerosis, ischemic stroke, and gliomas. The elucidation of the heterogeneity and diverse functions of BAMs during homeostasis and neuroinflammation is mesmerizing, since it may shed light on the precision medicine that emphasizes deep insights into programming cues in the unique brain immune microenvironment. In this review, we delve into the latest findings on BAMs, covering aspects like their origins, self-renewal capacity, adaptability, and implications in different brain disorders.
Collapse
Affiliation(s)
- Rui Sun
- Department of Neurological Surgery, Washington University School of Medicine in St. Louis, 660 S. Euclid Ave., Box 8057, St. Louis, MO, 63110, USA.
| | - Haowu Jiang
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine in St. Louis, 660 S. Euclid Ave., CB 8054, St. Louis, MO, 63110, USA.
| |
Collapse
|
19
|
Lummis NC, Gastfriend BD, Daneman R. Dural mural cells paint an anti-inflammatory picture. J Exp Med 2024; 221:e20232263. [PMID: 38270593 PMCID: PMC10818063 DOI: 10.1084/jem.20232263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2024] Open
Abstract
Mural cells directly contact macrophages in the dural layer of the meninges to suppress pro-inflammatory phenotypes, including antigen presentation and lymphocyte differentiation. These mechanisms represent new targets for modulating CNS immune surveillance and pathological inflammation (Min et al. 2024. J. Exp. Med.https://doi.org/10.1084/jem.20230326).
Collapse
Affiliation(s)
- Nicole C. Lummis
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Benjamin D. Gastfriend
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Richard Daneman
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
20
|
Rebejac J, Eme-Scolan E, Rua R. Role of meningeal immunity in brain function and protection against pathogens. J Inflamm (Lond) 2024; 21:3. [PMID: 38291415 PMCID: PMC10829400 DOI: 10.1186/s12950-023-00374-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 12/22/2023] [Indexed: 02/01/2024] Open
Abstract
The brain and spinal cord collectively referred to as the Central Nervous System (CNS) are protected by the blood-brain barrier that limits molecular, microbial and immunological trafficking. However, in the last decade, many studies have emphasized the protective role of 'border regions' at the surface of the CNS which are highly immunologically active, in contrast with the CNS parenchyma. In the steady-state, lymphoid and myeloid cells residing in the cranial meninges can affect brain function and behavior. Upon infection, they provide a first layer of protection against microbial neuroinvasion. The maturation of border sites over time enables more effective brain protection in adults as compared to neonates. Here, we provide a comprehensive update on the meningeal immune system and its role in physiological brain function and protection against infectious agents.
Collapse
Affiliation(s)
- Julie Rebejac
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, Inserm, CNRS, Marseille, France
| | - Elisa Eme-Scolan
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, Inserm, CNRS, Marseille, France
| | - Rejane Rua
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, Inserm, CNRS, Marseille, France.
| |
Collapse
|
21
|
Aspden JW, Murphy MA, Kashlan RD, Xiong Y, Poznansky MC, Sîrbulescu RF. Intruders or protectors - the multifaceted role of B cells in CNS disorders. Front Cell Neurosci 2024; 17:1329823. [PMID: 38269112 PMCID: PMC10806081 DOI: 10.3389/fncel.2023.1329823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 12/20/2023] [Indexed: 01/26/2024] Open
Abstract
B lymphocytes are immune cells studied predominantly in the context of peripheral humoral immune responses against pathogens. Evidence has been accumulating in recent years on the diversity of immunomodulatory functions that B cells undertake, with particular relevance for pathologies of the central nervous system (CNS). This review summarizes current knowledge on B cell populations, localization, infiltration mechanisms, and function in the CNS and associated tissues. Acute and chronic neurodegenerative pathologies are examined in order to explore the complex, and sometimes conflicting, effects that B cells can have in each context, with implications for disease progression and treatment outcomes. Additional factors such as aging modulate the proportions and function of B cell subpopulations over time and are also discussed in the context of neuroinflammatory response and disease susceptibility. A better understanding of the multifactorial role of B cell populations in the CNS may ultimately lead to innovative therapeutic strategies for a variety of neurological conditions.
Collapse
Affiliation(s)
- James W. Aspden
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Matthew A. Murphy
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Rommi D. Kashlan
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Yueyue Xiong
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Mark C. Poznansky
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Ruxandra F. Sîrbulescu
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
22
|
Barnacle JR, Davis AG, Wilkinson RJ. Recent advances in understanding the human host immune response in tuberculous meningitis. Front Immunol 2024; 14:1326651. [PMID: 38264653 PMCID: PMC10803428 DOI: 10.3389/fimmu.2023.1326651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 12/18/2023] [Indexed: 01/25/2024] Open
Abstract
Tuberculous meningitis (TBM), the most severe form of tuberculosis, causes death in approximately 25% cases despite antibiotic therapy, and half of survivors are left with neurological disability. Mortality and morbidity are contributed to by a dysregulated immune response, and adjunctive host-directed therapies are required to modulate this response and improve outcomes. Developing such therapies relies on improved understanding of the host immune response to TBM. The historical challenges in TBM research of limited in vivo and in vitro models have been partially overcome by recent developments in proteomics, transcriptomics, and metabolomics, and the use of these technologies in nested substudies of large clinical trials. We review the current understanding of the human immune response in TBM. We begin with M. tuberculosis entry into the central nervous system (CNS), microglial infection and blood-brain and other CNS barrier dysfunction. We then outline the innate response, including the early cytokine response, role of canonical and non-canonical inflammasomes, eicosanoids and specialised pro-resolving mediators. Next, we review the adaptive response including T cells, microRNAs and B cells, followed by the role of the glutamate-GABA neurotransmitter cycle and the tryptophan pathway. We discuss host genetic immune factors, differences between adults and children, paradoxical reaction, and the impact of HIV-1 co-infection including immune reconstitution inflammatory syndrome. Promising immunomodulatory therapies, research gaps, ongoing challenges and future paths are discussed.
Collapse
Affiliation(s)
- James R. Barnacle
- The Francis Crick Institute, London, United Kingdom
- Department of Infectious Disease, Imperial College, London, United Kingdom
- Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
| | - Angharad G. Davis
- The Francis Crick Institute, London, United Kingdom
- Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
| | - Robert J. Wilkinson
- The Francis Crick Institute, London, United Kingdom
- Department of Infectious Disease, Imperial College, London, United Kingdom
- Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
| |
Collapse
|
23
|
Pan S, Koleske JP, Koller GM, Halupnik GL, Alli AHO, Koneru S, DeFreitas D, Ramagiri S, Strahle JM. Postnatal meningeal CSF transport is primarily mediated by the arachnoid and pia maters and is not altered after intraventricular hemorrhage-posthemorrhagic hydrocephalus. Fluids Barriers CNS 2024; 21:4. [PMID: 38191402 PMCID: PMC10773070 DOI: 10.1186/s12987-023-00503-7] [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: 08/10/2023] [Accepted: 12/12/2023] [Indexed: 01/10/2024] Open
Abstract
BACKGROUND CSF has long been accepted to circulate throughout the subarachnoid space, which lies between the arachnoid and pia maters of the meninges. How the CSF interacts with the cellular components of the developing postnatal meninges including the dura, arachnoid, and pia of both the meninges at the surface of the brain and the intracranial meninges, prior to its eventual efflux from the cranium and spine, is less understood. Here, we characterize small and large CSF solute distribution patterns along the intracranial and surface meninges in neonatal rodents and compare our findings to meningeal CSF solute distribution in a rodent model of intraventricular hemorrhage-posthemorrhagic hydrocephalus. We also examine CSF solute interactions with the tela choroidea and its pial invaginations into the choroid plexuses of the lateral, third, and fourth ventricles. METHODS 1.9-nm gold nanoparticles, 15-nm gold nanoparticles, or 3 kDa Red Dextran Tetramethylrhodamine constituted in aCSF were infused into the right lateral ventricle of P7 rats to track CSF circulation. 10 min post-1.9-nm gold nanoparticle and Red Dextran Tetramethylrhodamine injection and 4 h post-15-nm gold nanoparticle injection, animals were sacrificed and brains harvested for histologic analysis to identify CSF tracer localization in the cranial and spine meninges and choroid plexus. Spinal dura and leptomeninges (arachnoid and pia) wholemounts were also evaluated. RESULTS There was significantly less CSF tracer distribution in the dura compared to the arachnoid and pia maters in neonatal rodents. Both small and large CSF tracers were transported intracranially to the arachnoid and pia mater of the perimesencephalic cisterns and tela choroidea, but not the falx cerebri. CSF tracers followed a similar distribution pattern in the spinal meninges. In the choroid plexus, there was large CSF tracer distribution in the apical surface of epithelial cells, and small CSF tracer along the basolateral surface. There were no significant differences in tracer intensity in the intracranial meninges of control vs. intraventricular hemorrhage-posthemorrhagic hydrocephalus (PHH) rodents, indicating preserved meningeal transport in the setting of PHH. CONCLUSIONS Differential CSF tracer handling by the meninges suggests that there are distinct roles for CSF handling between the arachnoid-pia and dura maters in the developing brain. Similarly, differences in apical vs. luminal choroid plexus CSF handling may provide insight into particle-size dependent CSF transport at the CSF-choroid plexus border.
Collapse
Affiliation(s)
- Shelei Pan
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Joshua P Koleske
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Gretchen M Koller
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Grace L Halupnik
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Abdul-Haq O Alli
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Shriya Koneru
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Dakota DeFreitas
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Sruthi Ramagiri
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Jennifer M Strahle
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA.
| |
Collapse
|
24
|
Yao Z, van Velthoven CTJ, Kunst M, Zhang M, McMillen D, Lee C, Jung W, Goldy J, Abdelhak A, Aitken M, Baker K, Baker P, Barkan E, Bertagnolli D, Bhandiwad A, Bielstein C, Bishwakarma P, Campos J, Carey D, Casper T, Chakka AB, Chakrabarty R, Chavan S, Chen M, Clark M, Close J, Crichton K, Daniel S, DiValentin P, Dolbeare T, Ellingwood L, Fiabane E, Fliss T, Gee J, Gerstenberger J, Glandon A, Gloe J, Gould J, Gray J, Guilford N, Guzman J, Hirschstein D, Ho W, Hooper M, Huang M, Hupp M, Jin K, Kroll M, Lathia K, Leon A, Li S, Long B, Madigan Z, Malloy J, Malone J, Maltzer Z, Martin N, McCue R, McGinty R, Mei N, Melchor J, Meyerdierks E, Mollenkopf T, Moonsman S, Nguyen TN, Otto S, Pham T, Rimorin C, Ruiz A, Sanchez R, Sawyer L, Shapovalova N, Shepard N, Slaughterbeck C, Sulc J, Tieu M, Torkelson A, Tung H, Valera Cuevas N, Vance S, Wadhwani K, Ward K, Levi B, Farrell C, Young R, Staats B, Wang MQM, Thompson CL, Mufti S, Pagan CM, Kruse L, Dee N, Sunkin SM, Esposito L, Hawrylycz MJ, Waters J, Ng L, Smith K, Tasic B, Zhuang X, et alYao Z, van Velthoven CTJ, Kunst M, Zhang M, McMillen D, Lee C, Jung W, Goldy J, Abdelhak A, Aitken M, Baker K, Baker P, Barkan E, Bertagnolli D, Bhandiwad A, Bielstein C, Bishwakarma P, Campos J, Carey D, Casper T, Chakka AB, Chakrabarty R, Chavan S, Chen M, Clark M, Close J, Crichton K, Daniel S, DiValentin P, Dolbeare T, Ellingwood L, Fiabane E, Fliss T, Gee J, Gerstenberger J, Glandon A, Gloe J, Gould J, Gray J, Guilford N, Guzman J, Hirschstein D, Ho W, Hooper M, Huang M, Hupp M, Jin K, Kroll M, Lathia K, Leon A, Li S, Long B, Madigan Z, Malloy J, Malone J, Maltzer Z, Martin N, McCue R, McGinty R, Mei N, Melchor J, Meyerdierks E, Mollenkopf T, Moonsman S, Nguyen TN, Otto S, Pham T, Rimorin C, Ruiz A, Sanchez R, Sawyer L, Shapovalova N, Shepard N, Slaughterbeck C, Sulc J, Tieu M, Torkelson A, Tung H, Valera Cuevas N, Vance S, Wadhwani K, Ward K, Levi B, Farrell C, Young R, Staats B, Wang MQM, Thompson CL, Mufti S, Pagan CM, Kruse L, Dee N, Sunkin SM, Esposito L, Hawrylycz MJ, Waters J, Ng L, Smith K, Tasic B, Zhuang X, Zeng H. A high-resolution transcriptomic and spatial atlas of cell types in the whole mouse brain. Nature 2023; 624:317-332. [PMID: 38092916 PMCID: PMC10719114 DOI: 10.1038/s41586-023-06812-z] [Show More Authors] [Citation(s) in RCA: 312] [Impact Index Per Article: 156.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 10/31/2023] [Indexed: 12/17/2023]
Abstract
The mammalian brain consists of millions to billions of cells that are organized into many cell types with specific spatial distribution patterns and structural and functional properties1-3. Here we report a comprehensive and high-resolution transcriptomic and spatial cell-type atlas for the whole adult mouse brain. The cell-type atlas was created by combining a single-cell RNA-sequencing (scRNA-seq) dataset of around 7 million cells profiled (approximately 4.0 million cells passing quality control), and a spatial transcriptomic dataset of approximately 4.3 million cells using multiplexed error-robust fluorescence in situ hybridization (MERFISH). The atlas is hierarchically organized into 4 nested levels of classification: 34 classes, 338 subclasses, 1,201 supertypes and 5,322 clusters. We present an online platform, Allen Brain Cell Atlas, to visualize the mouse whole-brain cell-type atlas along with the single-cell RNA-sequencing and MERFISH datasets. We systematically analysed the neuronal and non-neuronal cell types across the brain and identified a high degree of correspondence between transcriptomic identity and spatial specificity for each cell type. The results reveal unique features of cell-type organization in different brain regions-in particular, a dichotomy between the dorsal and ventral parts of the brain. The dorsal part contains relatively fewer yet highly divergent neuronal types, whereas the ventral part contains more numerous neuronal types that are more closely related to each other. Our study also uncovered extraordinary diversity and heterogeneity in neurotransmitter and neuropeptide expression and co-expression patterns in different cell types. Finally, we found that transcription factors are major determinants of cell-type classification and identified a combinatorial transcription factor code that defines cell types across all parts of the brain. The whole mouse brain transcriptomic and spatial cell-type atlas establishes a benchmark reference atlas and a foundational resource for integrative investigations of cellular and circuit function, development and evolution of the mammalian brain.
Collapse
Affiliation(s)
- Zizhen Yao
- Allen Institute for Brain Science, Seattle, WA, USA.
| | | | | | - Meng Zhang
- Howard Hughes Medical Institute, Department of Chemistry and Chemical Biology, Department of Physics, Harvard University, Cambridge, MA, USA
| | | | - Changkyu Lee
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Won Jung
- Howard Hughes Medical Institute, Department of Chemistry and Chemical Biology, Department of Physics, Harvard University, Cambridge, MA, USA
| | - Jeff Goldy
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | - Pamela Baker
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Eliza Barkan
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | | | - Daniel Carey
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | - Min Chen
- University of Pennsylvania, Philadelphia, PA, USA
| | | | - Jennie Close
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Scott Daniel
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Tim Dolbeare
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | - James Gee
- University of Pennsylvania, Philadelphia, PA, USA
| | | | | | - Jessica Gloe
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - James Gray
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | - Windy Ho
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Mike Huang
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Madie Hupp
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Kelly Jin
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Kanan Lathia
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Arielle Leon
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Su Li
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Brian Long
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Zach Madigan
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Zoe Maltzer
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Naomi Martin
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Rachel McCue
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Ryan McGinty
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Nicholas Mei
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Jose Melchor
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | - Sven Otto
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | - Lane Sawyer
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Noah Shepard
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Josef Sulc
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Michael Tieu
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Herman Tung
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Shane Vance
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Katelyn Ward
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Boaz Levi
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Rob Young
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Brian Staats
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Shoaib Mufti
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Lauren Kruse
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Nick Dee
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | - Jack Waters
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Lydia Ng
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Xiaowei Zhuang
- Howard Hughes Medical Institute, Department of Chemistry and Chemical Biology, Department of Physics, Harvard University, Cambridge, MA, USA
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA, USA.
| |
Collapse
|
25
|
Kearns NA, Iatrou A, Flood DJ, De Tissera S, Mullaney ZM, Xu J, Gaiteri C, Bennett DA, Wang Y. Dissecting the human leptomeninges at single-cell resolution. Nat Commun 2023; 14:7036. [PMID: 37923721 PMCID: PMC10624900 DOI: 10.1038/s41467-023-42825-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 10/23/2023] [Indexed: 11/06/2023] Open
Abstract
Emerging evidence shows that the meninges conduct essential immune surveillance and immune defense at the brain border, and the dysfunction of meningeal immunity contributes to aging and neurodegeneration. However, no study exists on the molecular properties of cell types within human leptomeninges. Here, we provide single nuclei profiling of dissected postmortem leptomeninges from aged individuals. We detect diverse cell types, including unique meningeal endothelial, mural, and fibroblast subtypes. For immune cells, we show that most T cells express CD8 and bear characteristics of tissue-resident memory T cells. We also identify distinct subtypes of border-associated macrophages (BAMs) that display differential gene expressions from microglia and express risk genes for Alzheimer's Disease (AD), as nominated by genome-wide association studies (GWAS). We discover cell-type-specific differentially expressed genes in individuals with Alzheimer's dementia, particularly in fibroblasts and BAMs. Indeed, when cultured, leptomeningeal cells display the signature of ex vivo AD fibroblasts upon amyloid-β treatment. We further explore ligand-receptor interactions within the leptomeningeal niche and computationally infer intercellular communications in AD. Thus, our study establishes a molecular map of human leptomeningeal cell types, providing significant insight into the border immune and fibrotic responses in AD.
Collapse
Affiliation(s)
- Nicola A Kearns
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Artemis Iatrou
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
| | - Daniel J Flood
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Sashini De Tissera
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Zachary M Mullaney
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Jishu Xu
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Chris Gaiteri
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
- Department of Psychiatry, Upstate Medical University, Syracuse, NY, 13210, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Yanling Wang
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA.
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, 60612, USA.
- Rush Graduate College, Rush University, Chicago IL, 60612, USA.
| |
Collapse
|
26
|
Ma L, Chang Q, Pei F, Liu M, Zhang W, Hong YK, Chai Y, Chen JF. Skull progenitor cell-driven meningeal lymphatic restoration improves neurocognitive functions in craniosynostosis. Cell Stem Cell 2023; 30:1472-1485.e7. [PMID: 37863055 PMCID: PMC10842404 DOI: 10.1016/j.stem.2023.09.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 08/21/2023] [Accepted: 09/27/2023] [Indexed: 10/22/2023]
Abstract
The meninges lie in the interface between the skull and brain, harboring lymphatic vasculature and skull progenitor cells (SPCs). How the skull and brain communicate remains largely unknown. We found that impaired meningeal lymphatics and brain perfusion drive neurocognitive defects in Twist1+/- mice, an animal model of craniosynostosis recapitulating human Saethre-Chotzen syndrome. Loss of SPCs leads to skull deformities and elevated intracranial pressure (ICP), whereas transplanting SPCs back into mutant mice mitigates lymphatic and brain defects through two mechanisms: (1) decreasing elevated ICP by skull correction and (2) promoting the growth and migration of lymphatic endothelial cells (LECs) via SPC-secreted vascular endothelial growth factor-C (VEGF-C). Treating Twist1+/- mice with VEGF-C promotes meningeal lymphatic growth and rescues defects in ICP, brain perfusion, and neurocognitive functions. Thus, the skull functionally integrates with the brain via meningeal lymphatics, which is impaired in craniosynostosis and can be restored by SPC-driven lymphatic activation via VEGF-C.
Collapse
Affiliation(s)
- Li Ma
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033, USA
| | - Qing Chang
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033, USA
| | - Fei Pei
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033, USA
| | - Mengmeng Liu
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033, USA
| | - Wei Zhang
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033, USA
| | - Young-Kwon Hong
- Department of Surgery, Norris Comprehensive Cancer Center, Keck School of Medicine of USC, Los Angeles, CA 90033, USA
| | - Yang Chai
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033, USA.
| | - Jian-Fu Chen
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
27
|
Kwon W, Yoo C, Kim JH, Kim T, Kim A, Hwang M, Choi H. Role of human dural fibroblasts in the angiogenic responses of human endothelial cells: An in vitro dural model and the application of lab-on-a-chip for EDAS. Bioeng Transl Med 2023; 8:e10589. [PMID: 38023706 PMCID: PMC10658529 DOI: 10.1002/btm2.10589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/18/2023] [Accepted: 08/01/2023] [Indexed: 12/01/2023] Open
Abstract
Encephaloduroarteriosynangiosis (EDAS), an indirect anastomosis procedure, is widely accepted as a primary treatment for moyamoya disease (MMD) to improve collateral blood flow. During surgical intervention, dural fibroblasts (DuF) are thought to produce various proteins that create an angiogenic microenvironment. However, the biophysiological evidence supporting the angiogenic properties of this surgical technique has not been thoroughly elucidated. The purpose of these studies was to determine whether DuF releases pro-angiogenic factors and chemokines and promotes angiogenic properties in human endothelial cells (ECs) under IL-1β-mediated wound conditions, which are expected to occur during the process of neo-vascularization within the dura mater. Furthermore, a microfluidic chemotaxis platform was implemented to investigate the angiogenic activity of ECs in response to a reconstituted dura model. Transcriptome sequencing revealed that IL-1β stimulation on DuF induced a significant upregulation of various pro-angiogenic genes, including IL-6, IL-8, CCL-2, CCL-5, SMOC-1, and SCG-2 (p < 0.05). Moreover, compared to ECs cultured in naïve media or naïve DuF media, those exposed to IL-1β-DuF conditioned media expressed higher mRNA and protein levels of these pro-angiogenic factors (p < 0.001). ECs co-cultured with IL-1β-DuF also exhibited considerable migration on the microfluidic chemotaxis platform. Furthermore, the chemotactic effects on the ECs were reduced upon neutralization of IL-8 or inhibition of NF-κB signaling. Our findings demonstrate that IL-1β-DuFs release factors that activate and enhance the angiogenic properties of ECs. These results suggest a potential interaction between DuF and ECs following EDAS for MMD, and these components could be targeted for the development of therapeutic biomarkers.
Collapse
Affiliation(s)
- Woo‐Keun Kwon
- Department of Neurosurgery, Korea University Guro HospitalKorea University College of MedicineSeoulSouth Korea
| | - Chang‐Min Yoo
- Department of Medical Sciences, Graduate School of MedicineKorea UniversitySeoulSouth Korea
| | - Jang Hun Kim
- Department of Neurosurgery, Korea University Anam HospitalKorea University College of MedicineSeoulSouth Korea
| | - Tae‐Won Kim
- Department of Medical Sciences, Graduate School of MedicineKorea UniversitySeoulSouth Korea
| | - An‐Gi Kim
- Department of Medical Sciences, Graduate School of MedicineKorea UniversitySeoulSouth Korea
| | - Min‐Ho Hwang
- Department of Medical Sciences, Graduate School of MedicineKorea UniversitySeoulSouth Korea
| | - Hyuk Choi
- Department of Medical Sciences, Graduate School of MedicineKorea UniversitySeoulSouth Korea
| |
Collapse
|
28
|
Santorella E, Balsbaugh JL, Ge S, Saboori P, Baker D, Pachter JS. Proteomic interrogation of the meninges reveals the molecular identities of structural components and regional distinctions along the CNS axis. Fluids Barriers CNS 2023; 20:74. [PMID: 37858244 PMCID: PMC10588166 DOI: 10.1186/s12987-023-00473-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 10/04/2023] [Indexed: 10/21/2023] Open
Abstract
The meninges surround the brain and spinal cord, affording physical protection while also serving as a niche of neuroimmune activity. Though possessing stromal qualities, its complex cellular and extracellular makeup has yet to be elaborated, and it remains unclear whether the meninges vary along the neuroaxis. Hence, studies were carried-out to elucidate the protein composition and structural organization of brain and spinal cord meninges in normal, adult Biozzi ABH mice. First, shotgun, bottom-up proteomics was carried-out. Prominent proteins at both brain and spinal levels included Type II collagen and Type II keratins, representing extracellular matrix (ECM) and cytoskeletal categories, respectively. While the vast majority of total proteins detected was shared between both meningeal locales, more were uniquely detected in brain than in spine. This pattern was also seen when total proteins were subdivided by cellular compartment, except in the case of the ECM category where brain and spinal meninges each had near equal number of unique proteins, and Type V and type III collagen registered exclusively in the spine. Quantitative analysis revealed differential expression of several collagens and cytoskeletal proteins between brain and spinal meninges. High-resolution immunofluorescence and immunogold-scanning electronmicroscopy on sections from whole brain and spinal cord - still encased within bone -identified major proteins detected by proteomics, and highlighted their association with cellular and extracellular elements of variously shaped arachnoid trabeculae. Western blotting aligned with the proteomic and immunohistological analyses, reinforcing differential appearance of proteins in brain vs spinal meninges. Results could reflect regional distinctions in meninges that govern protective and/or neuroimmune functions.
Collapse
Affiliation(s)
- Elise Santorella
- Department of Immunology, UConn Health, 263 Farmington Ave, Farmington, CT, 06030, USA
| | - Jeremy L Balsbaugh
- Proteomics and Metabolomics Facility, Center for Open Research Resources & Equipment, University of Connecticut, Storrs, CT, 06269, USA
| | - Shujun Ge
- Department of Immunology, UConn Health, 263 Farmington Ave, Farmington, CT, 06030, USA
| | - Parisa Saboori
- Department of Mechanical Engineering, Manhattan College, Bronx, NY, 10071, USA
| | - David Baker
- Blizard Institute, Queen Mary University of London, London, England
| | - Joel S Pachter
- Department of Immunology, UConn Health, 263 Farmington Ave, Farmington, CT, 06030, USA.
| |
Collapse
|
29
|
Kim YC, Ahn JH, Jin H, Yang MJ, Hong SP, Yoon JH, Kim SH, Gebre TN, Lee HJ, Kim YM, Koh GY. Immaturity of immune cells around the dural venous sinuses contributes to viral meningoencephalitis in neonates. Sci Immunol 2023; 8:eadg6155. [PMID: 37801517 DOI: 10.1126/sciimmunol.adg6155] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 07/24/2023] [Indexed: 10/08/2023]
Abstract
High neonatal susceptibility to meningitis has been attributed to the anatomical barriers that act to protect the central nervous system (CNS) from infection being immature and not fully developed. However, the mechanisms by which pathogens breach CNS barriers are poorly understood. Using the Armstrong strain of lymphocytic choriomeningitis virus (LCMV) to study virus propagation into the CNS during systemic infection, we demonstrate that mortality in neonatal, but not adult, mice is high after infection. Virus propagated extensively from the perivenous sinus region of the dura mater to the leptomeninges, choroid plexus, and cerebral cortex. Although the structural barrier of CNS border tissues is comparable between neonates and adults, immunofluorescence staining and single-cell RNA sequencing analyses revealed that the neonatal dural immune cells are immature and predominantly composed of CD206hi macrophages, with major histocompatibility complex class II (MHCII)hi macrophages being rare. In adults, however, perivenous sinus immune cells were enriched in MHCIIhi macrophages that are specialized for producing antiviral molecules and chemokines compared with CD206hi macrophages and protected the CNS against systemic virus invasion. Our findings clarify how systemic pathogens enter the CNS through its border tissues and how the immune barrier at the perivenous sinus region of the dura blocks pathogen access to the CNS.
Collapse
Affiliation(s)
- Young-Chan Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
- Center for Vascular Research, Institute for Basic Science (IBS), Daejeon 34141, Republic of Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Ji Hoon Ahn
- Center for Vascular Research, Institute for Basic Science (IBS), Daejeon 34141, Republic of Korea
| | - Hokyung Jin
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
- Center for Vascular Research, Institute for Basic Science (IBS), Daejeon 34141, Republic of Korea
| | - Myung Jin Yang
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
- Center for Vascular Research, Institute for Basic Science (IBS), Daejeon 34141, Republic of Korea
| | - Seon Pyo Hong
- Center for Vascular Research, Institute for Basic Science (IBS), Daejeon 34141, Republic of Korea
| | - Jin-Hui Yoon
- Center for Vascular Research, Institute for Basic Science (IBS), Daejeon 34141, Republic of Korea
| | - Sang-Hoon Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Tirhas Niguse Gebre
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Hyuek Jong Lee
- Center for Vascular Research, Institute for Basic Science (IBS), Daejeon 34141, Republic of Korea
| | - You-Me Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Gou Young Koh
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
- Center for Vascular Research, Institute for Basic Science (IBS), Daejeon 34141, Republic of Korea
| |
Collapse
|
30
|
Orrico-Sanchez A, Guiard BP, Manta S, Callebert J, Launay JM, Louis F, Paccard A, Gruszczynski C, Betancur C, Vialou V, Gautron S. Organic cation transporter 2 contributes to SSRI antidepressant efficacy by controlling tryptophan availability in the brain. Transl Psychiatry 2023; 13:302. [PMID: 37775532 PMCID: PMC10542329 DOI: 10.1038/s41398-023-02596-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 09/15/2023] [Accepted: 09/15/2023] [Indexed: 10/01/2023] Open
Abstract
Selective serotonin reuptake inhibitors (SSRI) are common first-line treatments for major depression. However, a significant number of depressed patients do not respond adequately to these pharmacological treatments. In the present preclinical study, we demonstrate that organic cation transporter 2 (OCT2), an atypical monoamine transporter, contributes to the effects of SSRI by regulating the routing of the essential amino acid tryptophan to the brain. Contrarily to wild-type mice, OCT2-invalidated mice failed to respond to prolonged fluoxetine treatment in a chronic depression model induced by corticosterone exposure recapitulating core symptoms of depression, i.e., anhedonia, social withdrawal, anxiety, and memory impairment. After corticosterone and fluoxetine treatment, the levels of tryptophan and its metabolites serotonin and kynurenine were decreased in the brain of OCT2 mutant mice compared to wild-type mice and reciprocally tryptophan and kynurenine levels were increased in mutants' plasma. OCT2 was detected by immunofluorescence in several structures at the blood-cerebrospinal fluid (CSF) or brain-CSF interface. Tryptophan supplementation during fluoxetine treatment increased brain concentrations of tryptophan and, more discreetly, of 5-HT in wild-type and OCT2 mutant mice. Importantly, tryptophan supplementation improved the sensitivity to fluoxetine treatment of OCT2 mutant mice, impacting chiefly anhedonia and short-term memory. Western blot analysis showed that glycogen synthase kinase-3β (GSK3β) and mammalian/mechanistic target of rapamycin (mTOR) intracellular signaling was impaired in OCT2 mutant mice brain after corticosterone and fluoxetine treatment and, conversely, tryptophan supplementation recruited selectively the mTOR protein complex 2. This study provides the first evidence of the physiological relevance of OCT2-mediated tryptophan transport, and its biological consequences on serotonin homeostasis in the brain and SSRI efficacy.
Collapse
Affiliation(s)
| | - Bruno P Guiard
- Université Paul Sabatier, CNRS, Research Center on Animal Cognition, Toulouse, France
| | - Stella Manta
- Université Paul Sabatier, CNRS, Research Center on Animal Cognition, Toulouse, France
| | - Jacques Callebert
- Sorbonne Paris Cité, Hôpital Lariboisière, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Jean-Marie Launay
- Sorbonne Paris Cité, Hôpital Lariboisière, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Franck Louis
- Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine, Paris, France
| | - Antoine Paccard
- Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine, Paris, France
| | | | - Catalina Betancur
- Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine, Paris, France
| | - Vincent Vialou
- Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine, Paris, France.
| | - Sophie Gautron
- Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine, Paris, France.
| |
Collapse
|
31
|
Allen CA, Goderie SK, Liu M, Kiehl TR, Farjood F, Wang Y, Boles NC, Temple S. Adult Mouse Leptomeninges Exhibit Regional and Age-related Cellular Heterogeneity Implicating Mental Disorders. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.10.557097. [PMID: 37745502 PMCID: PMC10515796 DOI: 10.1101/2023.09.10.557097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
The leptomeninges envelop the central nervous system (CNS) and contribute to cerebrospinal fluid (CSF) production and homeostasis. We analyzed the meninges overlying the anterior or posterior forebrain in the adult mouse by single nuclear RNA-sequencing (snucRNA-seq). This revealed regional differences in fibroblast and endothelial cell composition and gene expression. Surprisingly, these non-neuronal cells co-expressed genes implicated in neural functions. The regional differences changed with aging, from 3 to 18 months. Cytokine analysis revealed specific soluble factor production from anterior vs posterior meninges that also altered with age. Secreted factors from the leptomeninges from different regions and ages differentially impacted the survival of anterior or posterior cortical neuronal subsets, neuron morphology, and glia proliferation. These findings suggest that meningeal dysfunction in different brain regions could contribute to specific neural pathologies. The disease-associations of meningeal cell genes differentially expressed with region and age were significantly enriched for mental and substance abuse disorders.
Collapse
Affiliation(s)
| | | | - Mo Liu
- Neural Stem Cell Institute, Rensselaer, NY 12144, USA
| | | | | | - Yue Wang
- Neural Stem Cell Institute, Rensselaer, NY 12144, USA
| | | | - Sally Temple
- Neural Stem Cell Institute, Rensselaer, NY 12144, USA
| |
Collapse
|
32
|
Rego S, Sanchez G, Da Mesquita S. Current views on meningeal lymphatics and immunity in aging and Alzheimer's disease. Mol Neurodegener 2023; 18:55. [PMID: 37580702 PMCID: PMC10424377 DOI: 10.1186/s13024-023-00645-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 08/02/2023] [Indexed: 08/16/2023] Open
Abstract
Alzheimer's disease (AD) is an aging-related form of dementia associated with the accumulation of pathological aggregates of amyloid beta and neurofibrillary tangles in the brain. These phenomena are accompanied by exacerbated inflammation and marked neuronal loss, which altogether contribute to accelerated cognitive decline. The multifactorial nature of AD, allied to our still limited knowledge of its etiology and pathophysiology, have lessened our capacity to develop effective treatments for AD patients. Over the last few decades, genome wide association studies and biomarker development, alongside mechanistic experiments involving animal models, have identified different immune components that play key roles in the modulation of brain pathology in AD, affecting its progression and severity. As we will relay in this review, much of the recent efforts have been directed to better understanding the role of brain innate immunity, and particularly of microglia. However, and despite the lack of diversity within brain resident immune cells, the brain border tissues, especially the meninges, harbour a considerable number of different types and subtypes of adaptive and innate immune cells. Alongside microglia, which have taken the centre stage as important players in AD research, there is new and exciting evidence pointing to adaptive immune cells, namely T and B cells found in the brain and its meninges, as important modulators of neuroinflammation and neuronal (dys)function in AD. Importantly, a genuine and functional lymphatic vascular network is present around the brain in the outermost meningeal layer, the dura. The meningeal lymphatics are directly connected to the peripheral lymphatic system in different mammalian species, including humans, and play a crucial role in preserving a "healthy" immune surveillance of the CNS, by shaping immune responses, not only locally at the meninges, but also at the level of the brain tissue. In this review, we will provide a comprehensive view on our current knowledge about the meningeal lymphatic vasculature, emphasizing its described roles in modulating CNS fluid and macromolecule drainage, meningeal and brain immunity, as well as glial and neuronal function in aging and in AD.
Collapse
Affiliation(s)
- Shanon Rego
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
- Post-baccalaureate Research Education Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Guadalupe Sanchez
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
- Neuroscience Ph.D. Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Sandro Da Mesquita
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA.
- Post-baccalaureate Research Education Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL, 32224, USA.
- Neuroscience Ph.D. Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL, 32224, USA.
| |
Collapse
|
33
|
Semple BD, Panagiotopoulou O. Cranial Bone Changes Induced by Mild Traumatic Brain Injuries: A Neglected Player in Concussion Outcomes? Neurotrauma Rep 2023; 4:396-403. [PMID: 37350792 PMCID: PMC10282977 DOI: 10.1089/neur.2023.0025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/24/2023] Open
Abstract
Mild traumatic brain injuries (TBIs), particularly when repetitive in nature, are increasingly recognized to have a range of significant negative implications for brain health. Much of the ongoing research in the field is focused on the neurological consequences of these injuries and the relationship between TBIs and long-term neurodegenerative conditions such as chronic traumatic encephalopathy and Alzheimer's disease. However, our understanding of the complex relationship between applied mechanical force at impact, brain pathophysiology, and neurological function remains incomplete. Past research has shown that mild TBIs, even below the threshold that results in cranial fracture, induce changes in cranial bone structure and morphology. These structural and physiological changes likely have implications for the transmission of mechanical force into the underlying brain parenchyma. Here, we review this evidence in the context of the current understanding of bone mechanosensitivity and the consequences of TBIs or concussions. We postulate that heterogeneity of the calvarium, including differing bone thickness attributable to past impacts, age, or individual variability, may be a modulator of outcomes after subsequent TBIs. We advocate for greater consideration of cranial responses to TBI in both experimental and computer modeling of impact biomechanics, and raise the hypothesis that calvarial bone thickness represents a novel biomarker of brain injury vulnerability post-TBI.
Collapse
Affiliation(s)
- Bridgette D. Semple
- Department of Neuroscience, Monash University, Prahran, Victoria, Australia
- Department of Neurology, Alfred Health, Prahran, Victoria, Australia
- Department of Medicine (Royal Melbourne Hospital), University of Melbourne, Parkville, Victoria, Australia
| | - Olga Panagiotopoulou
- Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
34
|
Shrewsbury SB. The Upper Nasal Space: Option for Systemic Drug Delivery, Mucosal Vaccines and "Nose-to-Brain". Pharmaceutics 2023; 15:1720. [PMID: 37376168 PMCID: PMC10303426 DOI: 10.3390/pharmaceutics15061720] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/06/2023] [Accepted: 06/10/2023] [Indexed: 06/29/2023] Open
Abstract
Sino-nasal disease is appropriately treated with topical treatment, where the nasal mucosa acts as a barrier to systemic absorption. Non-invasive nasal delivery of drugs has produced some small molecule products with good bioavailability. With the recent COVID pandemic and the need for nasal mucosal immunity becoming more appreciated, more interest has become focused on the nasal cavity for vaccine delivery. In parallel, it has been recognized that drug delivery to different parts of the nose can have different results and for "nose-to-brain" delivery, deposition on the olfactory epithelium of the upper nasal space is desirable. Here the non-motile cilia and reduced mucociliary clearance lead to longer residence time that permits enhanced absorption, either into the systemic circulation or directly into the CNS. Many of the developments in nasal delivery have been to add bioadhesives and absorption/permeation enhancers, creating more complicated formulations and development pathways, but other projects have shown that the delivery device itself may allow more differential targeting of the upper nasal space without these additions and that could allow faster and more efficient programs to bring a wider range of drugs-and vaccines-to market.
Collapse
|
35
|
Gómez-Domínguez EG, Toriz CG, González-Pozos S, González-Del-Pliego M, Aguirre-Benítez EL, Pérez-Torres A, Flores-Martinez YM, Solano-Agama C, Rodríguez-Mata V, García-Godínez A, Martínez-Fong D, Mendoza-Garrido ME. Characterization of the rat pituitary capsule: Evidence that the cerebrospinal fluid filled the pituitary cleft and the inner side of the capsule. PLoS One 2023; 18:e0286399. [PMID: 37235567 DOI: 10.1371/journal.pone.0286399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
In humans, the pituitary gland is covered by a fibrous capsule and is considered a continuation of the meningeal sheath. However, in rodents some studies concluded that only the pars tuberalis (PT) and pars nervosa (PN) are enwrapped by the pia mater, while others showed that the whole gland is covered by this sheath. At PT the median eminence subarachnoid drains cerebrospinal fluid (CSF) to its cisternal system representing a pathway to the hypothalamus. In the present study we examined the rat pituitary capsule to elucidate its configuration, its physical interaction with the pituitary border and its relationship with the CSF. Furthermore, we also revisited the histology of the pituitary cleft and looked whether CSF drained in it. To answer such questions, we used scanning and transmission electron microscopy, intracerebroventricular infusion of Evan´s blue, fluorescent beads, and sodium fluorescein. The latter was measured in the pars distalis (PD) and various intracranial tissues. We found a pituitary capsule resembling leptomeninges, thick at the dorsal side of the pars intermedia (PI) and PD, thicker at the level of PI in contiguity with the PN and thinner at the rostro-ventral side as a thin membrane of fibroblast-like cells embedded in a fibrous layer. The capsule has abundant capillaries on all sides. Our results showed that the CSFs bathe between the capsule and the surface of the whole gland, and ciliate cells are present in the pituitary border. Our data suggest that the pituitary gland intercommunicates with the central nervous system (CNS) through the CSF.
Collapse
Affiliation(s)
- Edgar Giovanhi Gómez-Domínguez
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados, Instituto Politécnico Nacional, Ciudad de México, México
| | - César Gabriel Toriz
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados, Instituto Politécnico Nacional, Ciudad de México, México
| | - Sirenia González-Pozos
- Coordinación General de Servicios Experimentales, Microscopía Electrónica, Centro de Investigación y de Estudios Avanzados, Instituto Politécnico Nacional, Ciudad de México, México
| | - Margarita González-Del-Pliego
- Departamento de Embriología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Elsa Liliana Aguirre-Benítez
- Departamento de Embriología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Armando Pérez-Torres
- Departamento de Biología Celular y Tisular, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Yazmin Monserrat Flores-Martinez
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados, Instituto Politécnico Nacional, Ciudad de México, México
| | - Carmen Solano-Agama
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados, Instituto Politécnico Nacional, Ciudad de México, México
| | - Verónica Rodríguez-Mata
- Departamento de Biología Celular y Tisular, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Alejandro García-Godínez
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados, Instituto Politécnico Nacional, Ciudad de México, México
| | - Daniel Martínez-Fong
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados, Instituto Politécnico Nacional, Ciudad de México, México
| | - María Eugenia Mendoza-Garrido
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados, Instituto Politécnico Nacional, Ciudad de México, México
| |
Collapse
|
36
|
Derk J, Como CN, Jones HE, Joyce LR, Kim S, Spencer BL, Bonney S, O'Rourke R, Pawlikowski B, Doran KS, Siegenthaler JA. Formation and function of the meningeal arachnoid barrier around the developing mouse brain. Dev Cell 2023; 58:635-644.e4. [PMID: 36996816 PMCID: PMC10231667 DOI: 10.1016/j.devcel.2023.03.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 12/13/2022] [Accepted: 03/08/2023] [Indexed: 03/31/2023]
Abstract
The arachnoid barrier, a component of the blood-cerebrospinal fluid barrier (B-CSFB) in the meninges, is composed of epithelial-like, tight-junction-expressing cells. Unlike other central nervous system (CNS) barriers, its' developmental mechanisms and timing are largely unknown. Here, we show that mouse arachnoid barrier cell specification requires the repression of Wnt-β-catenin signaling and that constitutively active β-catenin can prevent its formation. We also show that the arachnoid barrier is functional prenatally and, in its absence, a small molecular weight tracer and the bacterium group B Streptococcus can cross into the CNS following peripheral injection. Acquisition of barrier properties prenatally coincides with the junctional localization of Claudin 11, and increased E-cadherin and maturation continues after birth, where postnatal expansion is marked by proliferation and re-organization of junctional domains. This work identifies fundamental mechanisms that drive arachnoid barrier formation, highlights arachnoid barrier fetal functions, and provides novel tools for future studies on CNS barrier development.
Collapse
Affiliation(s)
- Julia Derk
- University of Colorado Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO 80045, USA
| | - Christina N Como
- University of Colorado Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO 80045, USA; University of Colorado Anschutz Medical Campus, Neuroscience Graduate Program, Aurora, CO 80045, USA
| | - Hannah E Jones
- University of Colorado Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO 80045, USA; University of Colorado Anschutz Medical Campus, Cell Biology Stem Cells and Development Graduate Program, Aurora, CO 80045, USA
| | - Luke R Joyce
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Sol Kim
- University of Colorado Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO 80045, USA; University of Colorado Anschutz Medical Campus, Cell Biology Stem Cells and Development Graduate Program, Aurora, CO 80045, USA
| | - Brady L Spencer
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Stephanie Bonney
- University of Colorado Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO 80045, USA
| | - Rebecca O'Rourke
- University of Colorado Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO 80045, USA
| | - Brad Pawlikowski
- University of Colorado Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO 80045, USA
| | - Kelly S Doran
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Julie A Siegenthaler
- University of Colorado Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO 80045, USA; University of Colorado Anschutz Medical Campus, Neuroscience Graduate Program, Aurora, CO 80045, USA; University of Colorado Anschutz Medical Campus, Cell Biology Stem Cells and Development Graduate Program, Aurora, CO 80045, USA.
| |
Collapse
|
37
|
Yao Z, van Velthoven CTJ, Kunst M, Zhang M, McMillen D, Lee C, Jung W, Goldy J, Abdelhak A, Baker P, Barkan E, Bertagnolli D, Campos J, Carey D, Casper T, Chakka AB, Chakrabarty R, Chavan S, Chen M, Clark M, Close J, Crichton K, Daniel S, Dolbeare T, Ellingwood L, Gee J, Glandon A, Gloe J, Gould J, Gray J, Guilford N, Guzman J, Hirschstein D, Ho W, Jin K, Kroll M, Lathia K, Leon A, Long B, Maltzer Z, Martin N, McCue R, Meyerdierks E, Nguyen TN, Pham T, Rimorin C, Ruiz A, Shapovalova N, Slaughterbeck C, Sulc J, Tieu M, Torkelson A, Tung H, Cuevas NV, Wadhwani K, Ward K, Levi B, Farrell C, Thompson CL, Mufti S, Pagan CM, Kruse L, Dee N, Sunkin SM, Esposito L, Hawrylycz MJ, Waters J, Ng L, Smith KA, Tasic B, Zhuang X, Zeng H. A high-resolution transcriptomic and spatial atlas of cell types in the whole mouse brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.06.531121. [PMID: 37034735 PMCID: PMC10081189 DOI: 10.1101/2023.03.06.531121] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
The mammalian brain is composed of millions to billions of cells that are organized into numerous cell types with specific spatial distribution patterns and structural and functional properties. An essential step towards understanding brain function is to obtain a parts list, i.e., a catalog of cell types, of the brain. Here, we report a comprehensive and high-resolution transcriptomic and spatial cell type atlas for the whole adult mouse brain. The cell type atlas was created based on the combination of two single-cell-level, whole-brain-scale datasets: a single-cell RNA-sequencing (scRNA-seq) dataset of ~7 million cells profiled, and a spatially resolved transcriptomic dataset of ~4.3 million cells using MERFISH. The atlas is hierarchically organized into five nested levels of classification: 7 divisions, 32 classes, 306 subclasses, 1,045 supertypes and 5,200 clusters. We systematically analyzed the neuronal, non-neuronal, and immature neuronal cell types across the brain and identified a high degree of correspondence between transcriptomic identity and spatial specificity for each cell type. The results reveal unique features of cell type organization in different brain regions, in particular, a dichotomy between the dorsal and ventral parts of the brain: the dorsal part contains relatively fewer yet highly divergent neuronal types, whereas the ventral part contains more numerous neuronal types that are more closely related to each other. We also systematically characterized cell-type specific expression of neurotransmitters, neuropeptides, and transcription factors. The study uncovered extraordinary diversity and heterogeneity in neurotransmitter and neuropeptide expression and co-expression patterns in different cell types across the brain, suggesting they mediate a myriad of modes of intercellular communications. Finally, we found that transcription factors are major determinants of cell type classification in the adult mouse brain and identified a combinatorial transcription factor code that defines cell types across all parts of the brain. The whole-mouse-brain transcriptomic and spatial cell type atlas establishes a benchmark reference atlas and a foundational resource for deep and integrative investigations of cell type and circuit function, development, and evolution of the mammalian brain.
Collapse
Affiliation(s)
- Zizhen Yao
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Meng Zhang
- Howard Hughes Medical Institute, Department of Chemistry and Chemical Biology, Department of Physics, Harvard University, Cambridge, MA, USA
| | | | - Changkyu Lee
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Won Jung
- Howard Hughes Medical Institute, Department of Chemistry and Chemical Biology, Department of Physics, Harvard University, Cambridge, MA, USA
| | - Jeff Goldy
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Pamela Baker
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Eliza Barkan
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Daniel Carey
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | - Min Chen
- University of Pennsylvania, Philadelphia, PA, USA
| | | | - Jennie Close
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Scott Daniel
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Tim Dolbeare
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - James Gee
- University of Pennsylvania, Philadelphia, PA, USA
| | | | - Jessica Gloe
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - James Gray
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | - Windy Ho
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Kelly Jin
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Kanan Lathia
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Arielle Leon
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Brian Long
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Zoe Maltzer
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Naomi Martin
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Rachel McCue
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | | | | | | | - Josef Sulc
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Michael Tieu
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Herman Tung
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Katelyn Ward
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Boaz Levi
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Shoaib Mufti
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Lauren Kruse
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Nick Dee
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | - Jack Waters
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Lydia Ng
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Xiaowei Zhuang
- Howard Hughes Medical Institute, Department of Chemistry and Chemical Biology, Department of Physics, Harvard University, Cambridge, MA, USA
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA, USA
| |
Collapse
|
38
|
Amann L, Masuda T, Prinz M. Mechanisms of myeloid cell entry to the healthy and diseased central nervous system. Nat Immunol 2023; 24:393-407. [PMID: 36759712 DOI: 10.1038/s41590-022-01415-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 12/15/2022] [Indexed: 02/11/2023]
Abstract
Myeloid cells in the central nervous system (CNS), such as microglia, CNS-associated macrophages (CAMs), dendritic cells and monocytes, are vital for steady-state immune homeostasis as well as the resolution of tissue damage during brain development or disease-related pathology. The complementary usage of multimodal high-throughput and high-dimensional single-cell technologies along with recent advances in cell-fate mapping has revealed remarkable myeloid cell heterogeneity in the CNS. Despite the establishment of extensive expression profiles revealing myeloid cell multiplicity, the local anatomical conditions for the temporal- and spatial-dependent cellular engraftment are poorly understood. Here we highlight recent discoveries of the context-dependent mechanisms of myeloid cell migration and settlement into distinct subtissular structures in the CNS. These insights offer better understanding of the factors needed for compartment-specific myeloid cell recruitment, integration and residence during development and perturbation, which may lead to better treatment of CNS diseases.
Collapse
Affiliation(s)
- Lukas Amann
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Takahiro Masuda
- Division of Molecular Neuroimmunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan.
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany. .,Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany. .,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
39
|
Neutzner M, Kohler C, Frank S, Killer HE, Neutzner A. Impact of aging on meningeal gene expression. Fluids Barriers CNS 2023; 20:12. [PMID: 36747230 PMCID: PMC9903605 DOI: 10.1186/s12987-023-00412-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 01/25/2023] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The three-layered meninges cover and protect the central nervous system and form the interface between cerebrospinal fluid and the brain. They are host to a lymphatic system essential for maintaining fluid dynamics inside the cerebrospinal fluid-filled subarachnoid space and across the brain parenchyma via their connection to glymphatic structures. Meningeal fibroblasts lining and traversing the subarachnoid space have direct impact on the composition of the cerebrospinal fluid through endocytotic uptake as well as extensive protein secretion. In addition, the meninges are an active site for immunological processes and act as gatekeeper for immune cells entering the brain. During aging in mice, lymphatic drainage from the brain is less efficient contributing to neurodegenerative processes. Aging also affects the immunological status of the meninges, with increasing numbers of T cells, changing B cell make-up, and altered macrophage complement. METHODS We employed RNASeq to measure gene expression and to identify differentially expressed genes in meninges isolated from young and aged mice. Using Ingenuity pathway, GO term, and MeSH analyses, we identified regulatory pathways and cellular functions in meninges affected by aging. RESULTS Aging had profound impact on meningeal gene expression. Pathways related to innate as well as adaptive immunity were affected. We found evidence for increasing numbers of T and B lymphocytes and altered activity profiles for macrophages and other myeloid cells. Furthermore, expression of pro-inflammatory cytokine and chemokine genes increased with aging. Similarly, the complement system seemed to be more active in meninges of aged mice. Altered expression of solute carrier genes pointed to age-dependent changes in cerebrospinal fluid composition. In addition, gene expression for secreted proteins showed age-dependent changes, in particular, genes related to extracellular matrix composition and organization were affected. CONCLUSIONS Aging has profound effects on meningeal gene expression; thereby affecting the multifaceted functions meninges perform to maintain the homeostasis of the central nervous system. Thus, age-dependent neurodegenerative processes and cognitive decline are potentially in part driven by altered meningeal function.
Collapse
Affiliation(s)
- Melanie Neutzner
- grid.6612.30000 0004 1937 0642Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Corina Kohler
- grid.6612.30000 0004 1937 0642Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Stephan Frank
- grid.6612.30000 0004 1937 0642Department of Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Hanspeter E. Killer
- grid.6612.30000 0004 1937 0642Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Albert Neutzner
- Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031, Basel, Switzerland.
| |
Collapse
|
40
|
Multiple Genetic Loci Associated with Pug Dog Thoracolumbar Myelopathy. Genes (Basel) 2023; 14:genes14020385. [PMID: 36833311 PMCID: PMC9957375 DOI: 10.3390/genes14020385] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/24/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023] Open
Abstract
Pug dogs with thoracolumbar myelopathy (PDM) present with a specific clinical phenotype that includes progressive pelvic limb ataxia and paresis, commonly accompanied by incontinence. Vertebral column malformations and lesions, excessive scar tissue of the meninges, and central nervous system inflammation have been described. PDM has a late onset and affects more male than female dogs. The breed-specific presentation of the disorder suggests that genetic risk factors are involved in the disease development. To perform a genome-wide search for PDM-associated loci, we applied a Bayesian model adapted for mapping complex traits (BayesR) and a cross-population extended haplotype homozygosity test (XP-EHH) in 51 affected and 38 control pugs. Nineteen associated loci (harboring 67 genes in total, including 34 potential candidate genes) and three candidate regions under selection (with four genes within or next to the signal) were identified. The multiple candidate genes identified have implicated functions in bone homeostasis, fibrotic scar tissue, inflammatory responses, or the formation, regulation, and differentiation of cartilage, suggesting the potential relevance of these processes to the pathogenesis of PDM.
Collapse
|
41
|
Miao YB, Zhao W, Renchi G, Gong Y, Shi Y. Customizing delivery nano-vehicles for precise brain tumor therapy. J Nanobiotechnology 2023; 21:32. [PMID: 36707835 PMCID: PMC9883977 DOI: 10.1186/s12951-023-01775-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 01/09/2023] [Indexed: 01/29/2023] Open
Abstract
Although some tumor has become a curable disease for many patients, involvement of the central nervous system (CNS) is still a major concern. The blood-brain barrier (BBB), a special structure in the CNS, protects the brain from bloodborne pathogens via its excellent barrier properties and hinders new drug development for brain tumor. Recent breakthroughs in nanotechnology have resulted in various nanovehicless (NPs) as drug carriers to cross the BBB by different strategys. Here, the complex compositions and special characteristics of causes of brain tumor formation and BBB are elucidated exhaustively. Additionally, versatile drug nanovehicles with their recent applications and their pathways on different drug delivery strategies to overcome the BBB obstacle for anti-brain tumor are briefly discussed. Customizing nanoparticles for brain tumor treatments is proposed to improve the efficacy of brain tumor treatments via drug delivery from the gut to the brain. This review provides a broad perspective on customizing delivery nano-vehicles characteristics facilitate drug distribution across the brain and pave the way for the creation of innovative nanotechnology-based nanomaterials for brain tumor treatments.
Collapse
Affiliation(s)
- Yang-Bao Miao
- grid.410646.10000 0004 1808 0950Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000 China ,Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, 610072 Sichuan China
| | - Wang Zhao
- grid.410646.10000 0004 1808 0950Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000 China ,Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, 610072 Sichuan China
| | - Gao Renchi
- grid.410646.10000 0004 1808 0950Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000 China ,Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, 610072 Sichuan China
| | - Ying Gong
- grid.263901.f0000 0004 1791 7667School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031 People’s Republic of China
| | - Yi Shi
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, 610072 Sichuan China ,grid.9227.e0000000119573309Natural Products Research Center, Institute of Chengdu Biology, Sichuan Translational Medicine Hospital, Chinese Academy of Sciences, Chengdu, 610072 Sichuan China ,grid.410646.10000 0004 1808 0950Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, Chengdu, 610072 Sichuan China
| |
Collapse
|
42
|
Lin MS. Subdural Lesions Linking Additional Intracranial Spaces and Chronic Subdural Hematomas: A Narrative Review with Mutual Correlation and Possible Mechanisms behind High Recurrence. Diagnostics (Basel) 2023; 13:diagnostics13020235. [PMID: 36673045 PMCID: PMC9857428 DOI: 10.3390/diagnostics13020235] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/11/2022] [Accepted: 01/05/2023] [Indexed: 01/10/2023] Open
Abstract
The purpose of this study was two-fold. The first was to investigate the pathologic mechanisms underlying the formation of subdural fluid collection, an umbrella term referring to a condition commonly seen in the clinical setting. Accumulation of the cerebrospinal fluid (CSF) in the subdural space can be referred to in this disease category, disregarding the underlying source of the subdural fluid. However, in these two clinical situations, especially after trauma or brain surgery, fluid collection from the subarachnoid space (subdural hygroma) or from the ventricle to the subarachnoid space and infusion into the subdural space (external hydrocephalus), surgical management of critical patients may adopt the strategies of burr-hole, subduroperitoneal shunt, or ventriculoperitoneal shunt, which present distinctly different thoughts. Crucially, the former can be further transformed into chronic subdural hematoma (CSDH). The second significant theme was the pathogenesis of CSDH. Once the potential dural border cell (DBC) layer is separated such as if a wound is formed, the physiological mechanisms that seem to promote wound healing will resume in the subdural space as follows: coagulation, inflammation, fibroblast proliferation, neovascularization, and fibrinolysis. These aptly correspond to several key characteristics of CSDH formation such as the presence of both coagulation and fibrinolysis signals within the clot, neomembrane formation, angiogenesis, and recurrent bleeding, which contribute to CSDH failing to coagulate and absorb easily. Such a complexity of genesis and the possibility of arising from multiple pathological patterns provide a reasonable explanation for the high recurrence rate, even after surgery. Among the various complex and clinically challenging subdural lesions, namely, CSDH (confined to the subdural space alone), subdural hygroma (linked in two spaces), and external hydrocephalus (linked in three spaces), the ability to fully understand the different pathological mechanisms of each, differentiate them clinically, and devote more interventional strategies (including anti-inflammatory, anti-angiogenic, and anti-fibrinolysis) will be important themes in the future.
Collapse
Affiliation(s)
- Muh-Shi Lin
- Division of Neurosurgery, Department of Surgery, Kuang Tien General Hospital, Taichung 43303, Taiwan; ; Tel.: +886-4-2665-1900
- Department of Biotechnology and Animal Science, College of Bioresources, National Ilan University, Yilan 26047, Taiwan
- Department of Biotechnology, College of Medical and Health Care, Hung Kuang University, Taichung 43302, Taiwan
- Department of Health Business Administration, College of Medical and Health Care, Hung Kuang University, Taichung 43302, Taiwan
| |
Collapse
|
43
|
Møllgård K, Beinlich FRM, Kusk P, Miyakoshi LM, Delle C, Plá V, Hauglund NL, Esmail T, Rasmussen MK, Gomolka RS, Mori Y, Nedergaard M. A mesothelium divides the subarachnoid space into functional compartments. Science 2023; 379:84-88. [PMID: 36603070 DOI: 10.1126/science.adc8810] [Citation(s) in RCA: 127] [Impact Index Per Article: 63.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The central nervous system is lined by meninges, classically known as dura, arachnoid, and pia mater. We show the existence of a fourth meningeal layer that compartmentalizes the subarachnoid space in the mouse and human brain, designated the subarachnoid lymphatic-like membrane (SLYM). SLYM is morpho- and immunophenotypically similar to the mesothelial membrane lining of peripheral organs and body cavities, and it encases blood vessels and harbors immune cells. Functionally, the close apposition of SLYM with the endothelial lining of the meningeal venous sinus permits direct exchange of small solutes between cerebrospinal fluid and venous blood, thus representing the mouse equivalent of the arachnoid granulations. The functional characterization of SLYM provides fundamental insights into brain immune barriers and fluid transport.
Collapse
Affiliation(s)
- Kjeld Møllgård
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Felix R M Beinlich
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Peter Kusk
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Leo M Miyakoshi
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Christine Delle
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Virginia Plá
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Natalie L Hauglund
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Tina Esmail
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Martin K Rasmussen
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Ryszard S Gomolka
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Yuki Mori
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Maiken Nedergaard
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
44
|
Riew TR, Hwang JW, Jin X, Kim HL, Lee MY. Infiltration of meningeal macrophages into the Virchow-Robin space after ischemic stroke in rats: Correlation with activated PDGFR-β-positive adventitial fibroblasts. Front Mol Neurosci 2022; 15:1033271. [PMID: 36644619 PMCID: PMC9837109 DOI: 10.3389/fnmol.2022.1033271] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 12/09/2022] [Indexed: 12/31/2022] Open
Abstract
Macrophages play a crucial role in wound healing and fibrosis progression after brain injury. However, a detailed analysis of their initial infiltration and interaction with fibroblasts is yet to be conducted. This study aimed to investigate the possible route for migration of meningeal macrophages into the ischemic brain and whether these macrophages closely interact with neighboring platelet-derived growth factor beta receptor (PDGFR-β)-positive adventitial fibroblasts during this process. A rat model of ischemic stroke induced by middle cerebral artery occlusion (MCAO) was developed. In sham-operated rats, CD206-positive meningeal macrophages were confined to the leptomeninges and the perivascular spaces, and they were not found in the cortical parenchyma. In MCAO rats, the number of CD206-positive meningeal macrophages increased both at the leptomeninges and along the vessels penetrating the cortex 1 day after reperfusion and increased progressively in the extravascular area of the cortical parenchyma by 3 days. Immunoelectron microscopy and correlative light and electron microscopy showed that in the ischemic brain, macrophages were frequently located in the Virchow-Robin space around the penetrating arterioles and ascending venules at the pial surface. This was identified by cells expressing PDGFR-β, a novel biomarker of leptomeningeal cells. Macrophages within penetrating vessels were localized in the perivascular space between smooth muscle cells and PDGFR-β-positive adventitial fibroblasts. In addition, these PDGFR-β-positive fibroblasts showed morphological and molecular characteristics similar to those of leptomeningeal cells: they had large euchromatic nuclei with prominent nucleoli and well-developed rough endoplasmic reticulum; expressed nestin, vimentin, and type I collagen; and were frequently surrounded by collagen fibrils, indicating active collagen synthesis. In conclusion, the perivascular Virchow-Robin space surrounding the penetrating vessels could be an entry route of meningeal macrophages from the subarachnoid space into the ischemic cortical parenchyma, implying that activated PDGFR-β-positive adventitial fibroblasts could be involved in this process.
Collapse
Affiliation(s)
- Tae-Ryong Riew
- Department of Anatomy, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Ji-Won Hwang
- Department of Anatomy, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Xuyan Jin
- Department of Anatomy, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul, South Korea,Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Hong Lim Kim
- Integrative Research Support Center, Laboratory of Electron Microscope, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Mun-Yong Lee
- Department of Anatomy, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul, South Korea,Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, South Korea,*Correspondence: Mun-Yong Lee, ✉
| |
Collapse
|
45
|
Zhang Y, Bailey JT, Xu E, Singh K, Lavaert M, Link VM, D'Souza S, Hafiz A, Cao J, Cao G, Sant'Angelo DB, Sun W, Belkaid Y, Bhandoola A, McGavern DB, Yang Q. Mucosal-associated invariant T cells restrict reactive oxidative damage and preserve meningeal barrier integrity and cognitive function. Nat Immunol 2022; 23:1714-1725. [PMID: 36411380 PMCID: PMC10202031 DOI: 10.1038/s41590-022-01349-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 10/03/2022] [Indexed: 11/22/2022]
Abstract
Increasing evidence indicates close interaction between immune cells and the brain, revising the traditional view of the immune privilege of the brain. However, the specific mechanisms by which immune cells promote normal neural function are not entirely understood. Mucosal-associated invariant T cells (MAIT cells) are a unique type of innate-like T cell with molecular and functional properties that remain to be better characterized. In the present study, we report that MAIT cells are present in the meninges and express high levels of antioxidant molecules. MAIT cell deficiency in mice results in the accumulation of reactive oxidative species in the meninges, leading to reduced expression of junctional protein and meningeal barrier leakage. The presence of MAIT cells restricts neuroinflammation in the brain and preserves learning and memory. Together, our work reveals a new functional role for MAIT cells in the meninges and suggests that meningeal immune cells can help maintain normal neural function by preserving meningeal barrier homeostasis and integrity.
Collapse
Affiliation(s)
- Yuanyue Zhang
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Jacob T Bailey
- Department of Immunology & Microbial Disease, Albany Medical College, Albany, NY, USA
| | - En Xu
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Kunal Singh
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Marieke Lavaert
- Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Verena M Link
- Metaorganism Immunity Section, Laboratory of Immune System Biology and Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Shanti D'Souza
- Department of Immunology & Microbial Disease, Albany Medical College, Albany, NY, USA
| | - Alex Hafiz
- Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Jian Cao
- Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Gaoyuan Cao
- Rutgers Institute for Translational Medicine and Science, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Derek B Sant'Angelo
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
- Department of Pediatrics, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Wei Sun
- Department of Immunology & Microbial Disease, Albany Medical College, Albany, NY, USA
| | - Yasmine Belkaid
- Metaorganism Immunity Section, Laboratory of Immune System Biology and Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Avinash Bhandoola
- Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, 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
| | - Qi Yang
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA.
- Rutgers Institute for Translational Medicine and Science, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA.
- Department of Pediatrics, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA.
| |
Collapse
|
46
|
Yang R, Pan J, Wang Y, Xia P, Tai M, Jiang Z, Chen G. Application and prospects of somatic cell reprogramming technology for spinal cord injury treatment. Front Cell Neurosci 2022; 16:1005399. [PMID: 36467604 PMCID: PMC9712200 DOI: 10.3389/fncel.2022.1005399] [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: 07/28/2022] [Accepted: 11/02/2022] [Indexed: 08/10/2023] Open
Abstract
Spinal cord injury (SCI) is a serious neurological trauma that is challenging to treat. After SCI, many neurons in the injured area die due to necrosis or apoptosis, and astrocytes, oligodendrocytes, microglia and other non-neuronal cells become dysfunctional, hindering the repair of the injured spinal cord. Corrective surgery and biological, physical and pharmacological therapies are commonly used treatment modalities for SCI; however, no current therapeutic strategies can achieve complete recovery. Somatic cell reprogramming is a promising technology that has gradually become a feasible therapeutic approach for repairing the injured spinal cord. This revolutionary technology can reprogram fibroblasts, astrocytes, NG2 cells and neural progenitor cells into neurons or oligodendrocytes for spinal cord repair. In this review, we provide an overview of the transcription factors, genes, microRNAs (miRNAs), small molecules and combinations of these factors that can mediate somatic cell reprogramming to repair the injured spinal cord. Although many challenges and questions related to this technique remain, we believe that the beneficial effect of somatic cell reprogramming provides new ideas for achieving functional recovery after SCI and a direction for the development of treatments for SCI.
Collapse
Affiliation(s)
- Riyun Yang
- Department of Histology and Embryology, Medical School of Nantong University, Nantong, China
| | - Jingying Pan
- Department of Histology and Embryology, Medical School of Nantong University, Nantong, China
| | - Yankai Wang
- Center for Basic Medical Research, Medical School of Nantong University, Nantong, China
| | - Panhui Xia
- Center for Basic Medical Research, Medical School of Nantong University, Nantong, China
| | - Mingliang Tai
- Center for Basic Medical Research, Medical School of Nantong University, Nantong, China
| | - Zhihao Jiang
- Center for Basic Medical Research, Medical School of Nantong University, Nantong, China
| | - Gang Chen
- Center for Basic Medical Research, Medical School of Nantong University, Nantong, China
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
47
|
Endres LM, Jungblut M, Divyapicigil M, Sauer M, Stigloher C, Christodoulides M, Kim BJ, Schubert-Unkmeir A. Development of a multicellular in vitro model of the meningeal blood-CSF barrier to study Neisseria meningitidis infection. Fluids Barriers CNS 2022; 19:81. [PMID: 36289516 PMCID: PMC9597984 DOI: 10.1186/s12987-022-00379-z] [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: 06/08/2022] [Accepted: 10/06/2022] [Indexed: 12/01/2022] Open
Abstract
Background Bacterial meningitis is a life-threatening disease that occurs when pathogens such as Neisseria meningitidis cross the meningeal blood cerebrospinal fluid barrier (mBCSFB) and infect the meninges. Due to the human-specific nature of N. meningitidis, previous research investigating this complex host–pathogen interaction has mostly been done in vitro using immortalized brain endothelial cells (BECs) alone, which often do not retain relevant barrier properties in culture. Here, we developed physiologically relevant mBCSFB models using BECs in co-culture with leptomeningeal cells (LMCs) to examine N. meningitidis interaction. Methods We used BEC-like cells derived from induced pluripotent stem cells (iBECs) or hCMEC/D3 cells in co-culture with LMCs derived from tumor biopsies. We employed TEM and structured illumination microscopy to characterize the models as well as bacterial interaction. We measured TEER and sodium fluorescein (NaF) permeability to determine barrier tightness and integrity. We then analyzed bacterial adherence and penetration of the cell barrier and examined changes in host gene expression of tight junctions as well as chemokines and cytokines in response to infection. Results Both cell types remained distinct in co-culture and iBECs showed characteristic expression of BEC markers including tight junction proteins and endothelial markers. iBEC barrier function as determined by TEER and NaF permeability was improved by LMC co-culture and remained stable for seven days. BEC response to N. meningitidis infection was not affected by LMC co-culture. We detected considerable amounts of BEC-adherent meningococci and a relatively small number of intracellular bacteria. Interestingly, we discovered bacteria traversing the BEC-LMC barrier within the first 24 h post-infection, when barrier integrity was still high, suggesting a transcellular route for N. meningitidis into the CNS. Finally, we observed deterioration of barrier properties including loss of TEER and reduced expression of cell-junction components at late time points of infection. Conclusions Here, we report, for the first time, on co-culture of human iPSC derived BECs or hCMEC/D3 with meningioma derived LMCs and find that LMC co-culture improves barrier properties of iBECs. These novel models allow for a better understanding of N. meningitidis interaction at the mBCSFB in a physiologically relevant setting. Supplementary Information The online version contains supplementary material available at 10.1186/s12987-022-00379-z.
Collapse
Affiliation(s)
- Leo M. Endres
- grid.8379.50000 0001 1958 8658Institute for Hygiene and Microbiology, University of Würzburg, Josef-Schneider-Strasse 2, 97080 Würzburg, Germany
| | - Marvin Jungblut
- grid.8379.50000 0001 1958 8658Department of Biotechnology and Biophysics, Biocenter, University of Würzburg, Würzburg, Germany
| | - Mustafa Divyapicigil
- grid.411015.00000 0001 0727 7545Department of Biological Sciences, University of Alabama, Tuscaloosa, AL USA ,grid.265892.20000000106344187Department of Microbiology Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL USA ,grid.411015.00000 0001 0727 7545Center for Convergent Biosciences & Medicine, University of Alabama, Tuscaloosa, AL USA ,grid.411015.00000 0001 0727 7545Alabama Life Research Institute, University of Alabama, Tuscaloosa, AL USA
| | - Markus Sauer
- grid.8379.50000 0001 1958 8658Department of Biotechnology and Biophysics, Biocenter, University of Würzburg, Würzburg, Germany
| | - Christian Stigloher
- grid.8379.50000 0001 1958 8658Imaging Core Facility, Biocenter, University of Würzburg, Würzburg, Germany
| | - Myron Christodoulides
- grid.5491.90000 0004 1936 9297Molecular Microbiology, School of Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, UK
| | - Brandon J. Kim
- grid.411015.00000 0001 0727 7545Department of Biological Sciences, University of Alabama, Tuscaloosa, AL USA ,grid.265892.20000000106344187Department of Microbiology Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL USA ,grid.411015.00000 0001 0727 7545Center for Convergent Biosciences & Medicine, University of Alabama, Tuscaloosa, AL USA ,grid.411015.00000 0001 0727 7545Alabama Life Research Institute, University of Alabama, Tuscaloosa, AL USA
| | - Alexandra Schubert-Unkmeir
- grid.8379.50000 0001 1958 8658Institute for Hygiene and Microbiology, University of Würzburg, Josef-Schneider-Strasse 2, 97080 Würzburg, Germany
| |
Collapse
|
48
|
Kee R, Naughton M, McDonnell GV, Howell OW, Fitzgerald DC. A Review of Compartmentalised Inflammation and Tertiary Lymphoid Structures in the Pathophysiology of Multiple Sclerosis. Biomedicines 2022; 10:biomedicines10102604. [PMID: 36289863 PMCID: PMC9599335 DOI: 10.3390/biomedicines10102604] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 10/10/2022] [Indexed: 11/24/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic, immune-mediated, demyelinating disease of the central nervous system (CNS). The most common form of MS is a relapsing–remitting disease characterised by acute episodes of demyelination associated with the breakdown of the blood–brain barrier (BBB). In the relapsing–remitting phase there is often relative recovery (remission) from relapses characterised clinically by complete or partial resolution of neurological symptoms. In the later and progressive stages of the disease process, accrual of neurological disability occurs in a pathological process independent of acute episodes of demyelination and is accompanied by a trapped or compartmentalised inflammatory response, most notable in the connective tissue spaces of the vasculature and leptomeninges occurring behind an intact BBB. This review focuses on compartmentalised inflammation in MS and in particular, what we know about meningeal tertiary lymphoid structures (TLS; also called B cell follicles) which are organised clusters of immune cells, associated with more severe and progressive forms of MS. Meningeal inflammation and TLS could represent an important fluid or imaging marker of disease activity, whose therapeutic abrogation might be necessary to stop the most severe outcomes of disease.
Collapse
Affiliation(s)
- Rachael Kee
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7BL, UK
- Department of Neurology, Royal Victoria Hospital, Belfast BT12 6BA, UK
- Correspondence:
| | - Michelle Naughton
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7BL, UK
| | | | - Owain W. Howell
- Institute of Life Sciences, Swansea University, Wales SA2 8QA, UK
| | - Denise C. Fitzgerald
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7BL, UK
| |
Collapse
|
49
|
Palackdkharry CS, Wottrich S, Dienes E, Bydon M, Steinmetz MP, Traynelis VC. The leptomeninges as a critical organ for normal CNS development and function: First patient and public involved systematic review of arachnoiditis (chronic meningitis). PLoS One 2022; 17:e0274634. [PMID: 36178925 PMCID: PMC9524710 DOI: 10.1371/journal.pone.0274634] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 08/31/2022] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND & IMPORTANCE This patient and public-involved systematic review originally focused on arachnoiditis, a supposedly rare "iatrogenic chronic meningitis" causing permanent neurologic damage and intractable pain. We sought to prove disease existence, causation, symptoms, and inform future directions. After 63 terms for the same pathology were found, the study was renamed Diseases of the Leptomeninges (DLMs). We present results that nullify traditional clinical thinking about DLMs, answer study questions, and create a unified path forward. METHODS The prospective PRISMA protocol is published at Arcsology.org. We used four platforms, 10 sources, extraction software, and critical review with ≥2 researchers at each phase. All human sources to 12/6/2020 were eligible for qualitative synthesis utilizing R. Weekly updates since cutoff strengthen conclusions. RESULTS Included were 887/14286 sources containing 12721 DLMs patients. Pathology involves the subarachnoid space (SAS) and pia. DLMs occurred in all countries as a contributor to the top 10 causes of disability-adjusted life years lost, with communicable diseases (CDs) predominating. In the USA, the ratio of CDs to iatrogenic causes is 2.4:1, contradicting arachnoiditis literature. Spinal fusion surgery comprised 54.7% of the iatrogenic category, with rhBMP-2 resulting in 2.4x more DLMs than no use (p<0.0001). Spinal injections and neuraxial anesthesia procedures cause 1.1%, and 0.2% permanent DLMs, respectively. Syringomyelia, hydrocephalus, and arachnoid cysts are complications caused by blocked CSF flow. CNS neuron death occurs due to insufficient arterial supply from compromised vasculature and nerves traversing the SAS. Contrast MRI is currently the diagnostic test of choice. Lack of radiologist recognition is problematic. DISCUSSION & CONCLUSION DLMs are common. The LM clinically functions as an organ with critical CNS-sustaining roles involving the SAS-pia structure, enclosed cells, lymphatics, and biologic pathways. Cases involve all specialties. Causes are numerous, symptoms predictable, and outcomes dependent on time to treatment and extent of residual SAS damage. An international disease classification and possible treatment trials are proposed.
Collapse
Affiliation(s)
| | - Stephanie Wottrich
- Case Western Reserve School of Medicine, Cleveland, Ohio, United States of America
| | - Erin Dienes
- Arcsology®, Mead, Colorado, United States of America
| | - Mohamad Bydon
- Department of Neurologic Surgery, Orthopedic Surgery, and Health Services Research, Mayo Clinic School of Medicine, Rochester, Minnesota, United States of America
| | - Michael P. Steinmetz
- Department of Neurological Surgery, Cleveland Clinic Lerner College of Medicine Neurologic Institute, Cleveland, Ohio, United States of America
| | - Vincent C. Traynelis
- Department of Neurosurgery, Rush University School of Medicine, Chicago, Illinois, United States of America
| |
Collapse
|
50
|
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
|