1
|
Patel PU, Regmi A, Dass AI, Rojas OL. Immune conversations at the border: meningeal immunity in health and disease. Front Immunol 2025; 16:1531068. [PMID: 39944687 PMCID: PMC11813769 DOI: 10.3389/fimmu.2025.1531068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 01/10/2025] [Indexed: 05/09/2025] Open
Abstract
The brain and spinal cord, collectively known as the central nervous system, are encapsulated by an overlapping series of membranes known as the meninges. Once considered primarily a physical barrier for central nervous system protection, the bordering meninges are now recognized as highly immunologically active. The meninges host diverse resident immune cells and serve as a critical interface with peripheral immunity, playing multifaceted roles in maintaining central nervous system homeostasis, responding to pathogenic threats, and neurological disorders. This review summarizes recent advancements in our understanding of meningeal immunity including its structural composition, physiological functions, and role in health and disease.
Collapse
Affiliation(s)
- Preya U. Patel
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Aryan Regmi
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Angelina I. Dass
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Olga L. Rojas
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
| |
Collapse
|
2
|
Glück C, Zhou Q, Droux J, Chen Z, Glandorf L, Wegener S, Razansky D, Weber B, El Amki M. Pia-FLOW: Deciphering hemodynamic maps of the pial vascular connectome and its response to arterial occlusion. Proc Natl Acad Sci U S A 2024; 121:e2402624121. [PMID: 38954543 PMCID: PMC11252916 DOI: 10.1073/pnas.2402624121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 06/04/2024] [Indexed: 07/04/2024] Open
Abstract
The pial vasculature is the sole source of blood supply to the neocortex. The brain is contained within the skull, a vascularized bone marrow with a unique anatomical connection to the brain meninges. Recent developments in tissue clearing have enabled detailed mapping of the entire pial and calvarial vasculature. However, what are the absolute flow rate values of those vascular networks? This information cannot accurately be retrieved with the commonly used bioimaging methods. Here, we introduce Pia-FLOW, a unique approach based on large-scale transcranial fluorescence localization microscopy, to attain hemodynamic imaging of the whole murine pial and calvarial vasculature at frame rates up to 1,000 Hz and spatial resolution reaching 5.4 µm. Using Pia-FLOW, we provide detailed maps of flow velocity, direction, and vascular diameters which can serve as ground-truth data for further studies, advancing our understanding of brain fluid dynamics. Furthermore, Pia-FLOW revealed that the pial vascular network functions as one unit for robust allocation of blood after stroke.
Collapse
Affiliation(s)
- Chaim Glück
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich8057, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich8057, Switzerland
| | - Quanyu Zhou
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich8057, Switzerland
- Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich8092, Switzerland
| | - Jeanne Droux
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich8057, Switzerland
- Department of Neurology, University Hospital and University of Zurich, Zurich8091, Switzerland
| | - Zhenyue Chen
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich8057, Switzerland
- Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich8092, Switzerland
| | - Lukas Glandorf
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich8057, Switzerland
- Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich8092, Switzerland
| | - Susanne Wegener
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich8057, Switzerland
- Department of Neurology, University Hospital and University of Zurich, Zurich8091, Switzerland
| | - Daniel Razansky
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich8057, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich8057, Switzerland
- Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich8092, Switzerland
| | - Bruno Weber
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich8057, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich8057, Switzerland
| | - Mohamad El Amki
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich8057, Switzerland
- Department of Neurology, University Hospital and University of Zurich, Zurich8091, Switzerland
| |
Collapse
|
3
|
Xiao J, Li Y, Rowley T, Huang J, Yolken RH, Viscidi RP. Immunotherapy targeting the PD-1 pathway alleviates neuroinflammation caused by chronic Toxoplasma infection. Sci Rep 2023; 13:1288. [PMID: 36690687 PMCID: PMC9870997 DOI: 10.1038/s41598-023-28322-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 01/17/2023] [Indexed: 01/25/2023] Open
Abstract
Toxoplasma gondii can infect the host brain and trigger neuroinflammation. Such neuroinflammation might persist for years if the infection is not resolved, resulting in harmful outcomes for the brain. We have previously demonstrated the efficacy of immunotherapy targeting the programmed cell death protein 1 (PD-1) pathway on clearance of Toxoplasma tissue cysts. We aimed to test whether parasite clearance would lead to the resolution of neuroinflammation in infected brains. We established chronic Toxoplasma infection in BALB/c mice using the cyst-forming Prugniaud strain. Mice then received αPD-L1 or isotype control antibodies. After completion of the therapy, mice were euthanized six weeks later. The number of brain tissue cysts, Toxoplasma-specific CD8 + T cell proliferation and IFN-γ secretion, serum cytokine and chemokine levels, and CNS inflammation were measured. In αPD-L1-treated mice, we observed reduced brain tissue cysts, increased spleen weight, elevated IFN-γ production by antigen-specific CD8 + T cells, and a general increase in multiple serum cytokines and chemokines. Importantly, αPD-L1-treated mice displayed attenuation of meningeal lymphocytes, reactive astrocytes, and C1q expression. The reduction in inflammation-related proteins is correlated with reduced parasite burden. These results suggest that promoting systemic immunity results in parasite clearance, which in turn alleviates neuroinflammation. Our study may have implications for some brain infections where neuroinflammation is a critical component.
Collapse
Affiliation(s)
- Jianchun Xiao
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD, 21287, USA.
| | - Ye Li
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD, 21287, USA
| | - Treva Rowley
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD, 21287, USA
| | - Jing Huang
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD, 21287, USA
| | - Robert H Yolken
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD, 21287, USA
| | - Raphael P Viscidi
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD, 21287, USA
| |
Collapse
|
4
|
Audshasai T, Coles JA, Panagiotou S, Khandaker S, Scales HE, Kjos M, Baltazar M, Vignau J, Brewer JM, Kadioglu A, Yang M. Streptococcus pneumoniae Rapidly Translocate from the Nasopharynx through the Cribriform Plate to Invade the Outer Meninges. mBio 2022; 13:e0102422. [PMID: 35924840 PMCID: PMC9426477 DOI: 10.1128/mbio.01024-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 07/05/2022] [Indexed: 11/20/2022] Open
Abstract
The entry routes and translocation mechanisms of microorganisms or particulate materials into the central nervous system remain obscure We report here that Streptococcus pneumoniae (pneumococcus), or polystyrene microspheres of similar size, appear in the meninges of the dorsal cortex of mice within minutes of inhaled delivery. Recovery of viable bacteria from dissected tissue and fluorescence microscopy show that up to at least 72 h, pneumococci and microspheres were predominantly found in the outer of the two meninges: the pachymeninx. No pneumococci were found in blood or cerebrospinal fluid. Intravital imaging through the skull, aligned with flow cytometry showed recruitment and activation of LysM+ cells in the dorsal pachymeninx at 5 and 10 hours following intranasal infection. Imaging of the cribriform plate suggested that both pneumococci and microspheres entered through the foramina via an inward flow of fluid connecting the nose to the pachymeninx. Our findings bring new insight into the varied mechanisms of pneumococcal invasion of the central nervous system, but they are also pertinent to the delivery of drugs to the brain and the entry of airborne particulate matter into the cranium. IMPORTANCE Using two-photon imaging, we show that pneumococci translocate from the nasopharynx to the dorsal meninges of a mouse in the absence of any bacteria found in blood or cerebrospinal fluid. Strikingly, this takes place within minutes of inhaled delivery of pneumococci, suggesting the existence of an inward flow of fluid connecting the nasopharynx to the meninges, rather than a receptor-mediated mechanism. We also show that this process is size dependent, as microspheres of the same size as pneumococci can translocate along the same pathway, while larger size microspheres cannot. Furthermore, we describe the host response to invasion of the outer meninges. Our study provides a completely new insight into the key initial events that occur during the translocation of pneumococci directly from the nasal cavity to the meninges, with relevance to the development of intranasal drug delivery systems and the investigations of brain damage caused by inhaled air pollutants.
Collapse
Affiliation(s)
- Teerawit Audshasai
- Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
- Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
| | - Jonathan A. Coles
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Stavros Panagiotou
- Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Shadia Khandaker
- Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Hannah E. Scales
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Morten Kjos
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | - Murielle Baltazar
- Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Julie Vignau
- Centre de Recherche en Cancérologie et Immunologie Nantes Angers, Université de Nantes, Nantes, France
| | - James M. Brewer
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Aras Kadioglu
- Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Marie Yang
- Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
5
|
Santos-Lima B, Pietronigro EC, Terrabuio E, Zenaro E, Constantin G. The role of neutrophils in the dysfunction of central nervous system barriers. Front Aging Neurosci 2022; 14:965169. [PMID: 36034148 PMCID: PMC9404376 DOI: 10.3389/fnagi.2022.965169] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 07/21/2022] [Indexed: 12/04/2022] Open
Abstract
Leukocyte migration into the central nervous system (CNS) represents a central process in the development of neurological diseases with a detrimental inflammatory component. Infiltrating neutrophils have been detected inside the brain of patients with several neuroinflammatory disorders, including stroke, multiple sclerosis and Alzheimer’s disease. During inflammatory responses, these highly reactive innate immune cells can rapidly extravasate and release a plethora of pro-inflammatory and cytotoxic factors, potentially inducing significant collateral tissue damage. Indeed, several studies have shown that neutrophils promote blood-brain barrier damage and increased vascular permeability during neuroinflammatory diseases. Recent studies have shown that neutrophils migrate into the meninges and choroid plexus, suggesting these cells can also damage the blood-cerebrospinal fluid barrier (BCSFB). In this review, we discuss the emerging role of neutrophils in the dysfunction of brain barriers across different neuroinflammatory conditions and describe the molecular basis and cellular interplays involved in neutrophil-mediated injury of the CNS borders.
Collapse
|
6
|
Mansilla MJ, Presas-Rodríguez S, Teniente-Serra A, González-Larreategui I, Quirant-Sánchez B, Fondelli F, Djedovic N, Iwaszkiewicz-Grześ D, Chwojnicki K, Miljković Đ, Trzonkowski P, Ramo-Tello C, Martínez-Cáceres EM. Paving the way towards an effective treatment for multiple sclerosis: advances in cell therapy. Cell Mol Immunol 2021; 18:1353-1374. [PMID: 33958746 PMCID: PMC8167140 DOI: 10.1038/s41423-020-00618-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 12/03/2020] [Indexed: 12/13/2022] Open
Abstract
Multiple sclerosis (MS) is a leading cause of chronic neurological disability in young to middle-aged adults, affecting ~2.5 million people worldwide. Currently, most therapeutics for MS are systemic immunosuppressive or immunomodulatory drugs, but these drugs are unable to halt or reverse the disease and have the potential to cause serious adverse events. Hence, there is an urgent need for the development of next-generation treatments that, alone or in combination, stop the undesired autoimmune response and contribute to the restoration of homeostasis. This review analyzes current MS treatments as well as different cell-based therapies that have been proposed to restore homeostasis in MS patients (tolerogenic dendritic cells, regulatory T cells, mesenchymal stem cells, and vaccination with T cells). Data collected from preclinical studies performed in the experimental autoimmune encephalomyelitis (EAE) model of MS in animals, in vitro cultures of cells from MS patients and the initial results of phase I/II clinical trials are analyzed to better understand which parameters are relevant for obtaining an efficient cell-based therapy for MS.
Collapse
Affiliation(s)
- M J Mansilla
- Division of Immunology, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Barcelona, Spain. .,Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain.
| | - S Presas-Rodríguez
- Multiple Sclerosis Unit, Department of Neurosciences, Germans Trias i Pujol University Hospital, Barcelona, Spain.,Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - A Teniente-Serra
- Division of Immunology, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Barcelona, Spain.,Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - I González-Larreategui
- Division of Immunology, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Barcelona, Spain.,Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - B Quirant-Sánchez
- Division of Immunology, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Barcelona, Spain.,Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - F Fondelli
- Division of Immunology, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Barcelona, Spain.,Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - N Djedovic
- Department of Immunology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - D Iwaszkiewicz-Grześ
- Department of Medical Immunology, Medical University of Gdańsk, Gdańsk, Poland.,Poltreg S.A., Gdańsk, Poland
| | - K Chwojnicki
- Department of Anaesthesiology & Intensive Care, Medical University of Gdańsk, Gdańsk, Poland
| | - Đ Miljković
- Department of Immunology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - P Trzonkowski
- Department of Medical Immunology, Medical University of Gdańsk, Gdańsk, Poland.,Poltreg S.A., Gdańsk, Poland
| | - C Ramo-Tello
- Multiple Sclerosis Unit, Department of Neurosciences, Germans Trias i Pujol University Hospital, Barcelona, Spain.,Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - E M Martínez-Cáceres
- Division of Immunology, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Barcelona, Spain. .,Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain.
| |
Collapse
|
7
|
Cardiac glycosides target barrier inflammation of the vasculature, meninges and choroid plexus. Commun Biol 2021; 4:260. [PMID: 33637884 PMCID: PMC7910294 DOI: 10.1038/s42003-021-01787-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 02/03/2021] [Indexed: 01/31/2023] Open
Abstract
Neuroinflammation is a key component of virtually all neurodegenerative diseases, preceding neuronal loss and associating directly with cognitive impairment. Neuroinflammatory signals can originate and be amplified at barrier tissues such as brain vasculature, surrounding meninges and the choroid plexus. We designed a high content screening system to target inflammation in human brain-derived cells of the blood-brain barrier (pericytes and endothelial cells) to identify inflammatory modifiers. Screening an FDA-approved drug library we identify digoxin and lanatoside C, members of the cardiac glycoside family, as inflammatory-modulating drugs that work in blood-brain barrier cells. An ex vivo assay of leptomeningeal and choroid plexus explants confirm that these drugs maintain their function in 3D cultures of brain border tissues. These results suggest that cardiac glycosides may be useful in targeting inflammation at border regions of the brain and offer new options for drug discovery approaches for neuroinflammatory driven degeneration.
Collapse
|
8
|
Borst K, Prinz M. Deciphering the heterogeneity of myeloid cells during neuroinflammation in the single-cell era. Brain Pathol 2020; 30:1192-1207. [PMID: 33058309 PMCID: PMC8018048 DOI: 10.1111/bpa.12910] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 08/23/2020] [Accepted: 10/12/2020] [Indexed: 12/13/2022] Open
Abstract
Multiple sclerosis (MS) is a disabling neuroinflammatory disease, which is little understood and lacks a sufficient therapeutic regimen. Myeloid cells have repeatedly shown to play a pivotal role in the disease progression. During homeostasis, only the CNS‐resident microglia and CNS‐associated macrophages are present in the CNS. Neuroinflammation causes peripheral immune cells to infiltrate the CNS contributing to disease progression and neurological sequelae. The differential involvement of the diverse peripheral and resident myeloid cell subsets to the disease pathogenesis and outcome are highly debated and difficult to assess. However, novel technological advances (new mouse models, single‐cell RNA‐Sequencing, and CYTOF) have improved the depth of immune profiling, which allows the characterization of distinct myeloid subsets. This review provides an overview of current knowledge on the phenotypes and roles of these different myeloid subsets in neuroinflammatory disease and their therapeutic relevance.
Collapse
Affiliation(s)
- Katharina Borst
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, Medical Faculty, 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
|
9
|
Pinke KH, Zorzella-Pezavento SFG, Lara VS, Sartori A. Should mast cells be considered therapeutic targets in multiple sclerosis? Neural Regen Res 2020; 15:1995-2007. [PMID: 32394947 PMCID: PMC7716037 DOI: 10.4103/1673-5374.282238] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 12/19/2019] [Accepted: 01/16/2020] [Indexed: 12/23/2022] Open
Abstract
Mast cells are immune cells of the myeloid lineage that are found throughout the body, including the central nervous system. They perform many functions associated with innate and specific immunity, angiogenesis, and vascular homeostasis. Moreover, they have been implicated in a series of pathologies (e.g., hypersensitivity reactions, tumors, and inflammatory disorders). In this review, we propose that this cell could be a relevant therapeutic target in multiple sclerosis, which is a central nervous system degenerative disease. To support this proposition, we describe the general biological properties of mast cells, their contribution to innate and specific immunity, and the participation of mast cells in the various stages of multiple sclerosis and experimental autoimmune encephalomyelitis development. The final part of this review is dedicated to an overview of the available mast cells immunomodulatory drugs and their activity on multiple sclerosis and experimental autoimmune encephalomyelitis, including our own experience related to the effect of ketotifen fumarate on experimental autoimmune encephalomyelitis evolution.
Collapse
Affiliation(s)
- Karen Henriette Pinke
- Institute of Biosciences, Department of Microbiology and Immunology, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | | | - Vanessa Soares Lara
- Bauru School of Dentistry, Department of Surgery, Stomatology, Pathology and Radiology, University of São Paulo, Bauru, São Paulo, Brazil
| | - Alexandrina Sartori
- Institute of Biosciences, Department of Microbiology and Immunology, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| |
Collapse
|
10
|
Decimo I, Dolci S, Panuccio G, Riva M, Fumagalli G, Bifari F. Meninges: A Widespread Niche of Neural Progenitors for the Brain. Neuroscientist 2020; 27:506-528. [PMID: 32935634 PMCID: PMC8442137 DOI: 10.1177/1073858420954826] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Emerging evidence highlights the several roles that meninges play in
relevant brain functions as they are a protective membrane for the
brain, produce and release several trophic factors important for
neural cell migration and survival, control cerebrospinal fluid
dynamics, and embrace numerous immune interactions affecting neural
parenchymal functions. Furthermore, different groups have identified
subsets of neural progenitors residing in the meninges during
development and in the adulthood in different mammalian species,
including humans. Interestingly, these immature neural cells are able
to migrate from the meninges to the neural parenchyma and
differentiate into functional cortical neurons or oligodendrocytes.
Immature neural cells residing in the meninges promptly react to brain
disease. Injury-induced expansion and migration of meningeal neural
progenitors have been observed following experimental demyelination,
traumatic spinal cord and brain injury, amygdala lesion, stroke, and
progressive ataxia. In this review, we summarize data on the function
of meninges as stem cell niche and on the presence of immature neural
cells in the meninges, and discuss their roles in brain health and
disease. Furthermore, we consider the potential exploitation of
meningeal neural progenitors for the regenerative medicine to treat
neurological disorders.
Collapse
Affiliation(s)
- Ilaria Decimo
- Laboratory of Pharmacology, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Sissi Dolci
- Laboratory of Pharmacology, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Gabriella Panuccio
- Enhanced Regenerative Medicine, Istituto Italiano di Tecnologia, Genova, Italy
| | - Marco Riva
- Unit of Neurosurgery, Fondazione IRCCS Ca'Granda Ospedale Maggiore Policlinico, Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Guido Fumagalli
- Laboratory of Pharmacology, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Francesco Bifari
- Laboratory of Cell Metabolism and Regenerative Medicine, Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| |
Collapse
|
11
|
Michel L, Grasmuck C, Charabati M, Lécuyer MA, Zandee S, Dhaeze T, Alvarez JI, Li R, Larouche S, Bourbonnière L, Moumdjian R, Bouthillier A, Lahav B, Duquette P, Bar-Or A, Gommerman JL, Peelen E, Prat A. Activated leukocyte cell adhesion molecule regulates B lymphocyte migration across central nervous system barriers. Sci Transl Med 2020; 11:11/518/eaaw0475. [PMID: 31723036 DOI: 10.1126/scitranslmed.aaw0475] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 07/10/2019] [Accepted: 10/21/2019] [Indexed: 12/19/2022]
Abstract
The presence of B lymphocyte-associated oligoclonal immunoglobulins in the cerebrospinal fluid is a classic hallmark of multiple sclerosis (MS). The clinical efficacy of anti-CD20 therapies supports a major role for B lymphocytes in MS development. Although activated oligoclonal populations of pathogenic B lymphocytes are able to traffic between the peripheral circulation and the central nervous system (CNS) in patients with MS, molecular players involved in this migration have not yet been elucidated. In this study, we demonstrated that activated leukocyte cell adhesion molecule (ALCAM/CD166) identifies subsets of proinflammatory B lymphocytes and drives their transmigration across different CNS barriers in mouse and human. We also showcased that blocking ALCAM alleviated disease severity in animals affected by a B cell-dependent form of experimental autoimmune encephalomyelitis. Last, we determined that the proportion of ALCAM+ B lymphocytes was increased in the peripheral blood and within brain lesions of patients with MS. Our findings indicate that restricting access to the CNS by targeting ALCAM on pathogenic B lymphocytes might represent a promising strategy for the development of next-generation B lymphocyte-targeting therapies for the treatment of MS.
Collapse
Affiliation(s)
- Laure Michel
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, Montréal, QC Canada.,Neuroimmunology Unit, Centre de recherche du CHUM (CRCHUM), Montréal, QC H2X 0A9, Canada
| | - Camille Grasmuck
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, Montréal, QC Canada.,Neuroimmunology Unit, Centre de recherche du CHUM (CRCHUM), Montréal, QC H2X 0A9, Canada
| | - Marc Charabati
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, Montréal, QC Canada.,Neuroimmunology Unit, Centre de recherche du CHUM (CRCHUM), Montréal, QC H2X 0A9, Canada
| | - Marc-André Lécuyer
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, Montréal, QC Canada.,Neuroimmunology Unit, Centre de recherche du CHUM (CRCHUM), Montréal, QC H2X 0A9, Canada
| | - Stephanie Zandee
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, Montréal, QC Canada.,Neuroimmunology Unit, Centre de recherche du CHUM (CRCHUM), Montréal, QC H2X 0A9, Canada
| | - Tessa Dhaeze
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, Montréal, QC Canada.,Neuroimmunology Unit, Centre de recherche du CHUM (CRCHUM), Montréal, QC H2X 0A9, Canada
| | - Jorge I Alvarez
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, Montréal, QC Canada.,Neuroimmunology Unit, Centre de recherche du CHUM (CRCHUM), Montréal, QC H2X 0A9, Canada
| | - Rui Li
- Center for Neuroinflammation and Experimental Therapeutics and Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sandra Larouche
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, Montréal, QC Canada.,Neuroimmunology Unit, Centre de recherche du CHUM (CRCHUM), Montréal, QC H2X 0A9, Canada
| | - Lyne Bourbonnière
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, Montréal, QC Canada.,Neuroimmunology Unit, Centre de recherche du CHUM (CRCHUM), Montréal, QC H2X 0A9, Canada
| | | | | | - Boaz Lahav
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, Montréal, QC Canada.,Neuroimmunology Unit, Centre de recherche du CHUM (CRCHUM), Montréal, QC H2X 0A9, Canada
| | - Pierre Duquette
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, Montréal, QC Canada.,Neuroimmunology Unit, Centre de recherche du CHUM (CRCHUM), Montréal, QC H2X 0A9, Canada
| | - Amit Bar-Or
- Center for Neuroinflammation and Experimental Therapeutics and Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Evelyn Peelen
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, Montréal, QC Canada.,Neuroimmunology Unit, Centre de recherche du CHUM (CRCHUM), Montréal, QC H2X 0A9, Canada
| | - Alexandre Prat
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, Montréal, QC Canada. .,Neuroimmunology Unit, Centre de recherche du CHUM (CRCHUM), Montréal, QC H2X 0A9, Canada
| |
Collapse
|
12
|
Karpus WJ. Cytokines and Chemokines in the Pathogenesis of Experimental Autoimmune Encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2020; 204:316-326. [PMID: 31907274 DOI: 10.4049/jimmunol.1900914] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 11/13/2019] [Indexed: 12/25/2022]
Abstract
Experimental autoimmune encephalomyelitis is a CD4+ T cell-mediated demyelinating disease of the CNS that serves as a model for multiple sclerosis. Cytokines and chemokines shape Th1 and Th17 effector responses as well as regulate migration of leukocytes to the CNS during disease. The CNS cellular infiltrate consists of Ag-specific and nonspecific CD4+ and CD8+ T cells, neutrophils, B cells, monocytes, macrophages, and dendritic cells. The mechanism of immune-mediated inflammation in experimental autoimmune encephalomyelitis has been extensively studied in an effort to develop therapeutic modalities for multiple sclerosis and, indeed, has provided insight in modern drug discovery. The present Brief Review highlights critical pathogenic aspects of cytokines and chemokines involved in generation of effector T cell responses and migration of inflammatory cells to the CNS. Select cytokines and chemokines are certainly important in the regulatory response, which involves T regulatory, B regulatory, and myeloid-derived suppressor cells. However, that discussion is beyond the scope of this brief review.
Collapse
Affiliation(s)
- William J Karpus
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53706
| |
Collapse
|
13
|
de Oliveira LRC, Mimura LAN, Fraga-Silva TFDC, Ishikawa LLW, Fernandes AAH, Zorzella-Pezavento SFG, Sartori A. Calcitriol Prevents Neuroinflammation and Reduces Blood-Brain Barrier Disruption and Local Macrophage/Microglia Activation. Front Pharmacol 2020; 11:161. [PMID: 32226379 PMCID: PMC7080989 DOI: 10.3389/fphar.2020.00161] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Accepted: 02/07/2020] [Indexed: 12/14/2022] Open
Abstract
Multiple sclerosis (MS) is a progressive disease of the central nervous system (CNS) that involves damage to the myelin sheath surrounding axons. MS therapy is based on immunomodulatory drugs that reduce disease recurrence and severity. Vitamin D is a hormone whose immunomodulatory ability has been widely demonstrated, including in experimental autoimmune encephalomyelitis (EAE), which is an animal model of CNS inflammation. In this study, we evaluated the potential of very early intervention with the active form of vitamin D (1,25-dihydroxyvitamin D3) to control neuroinflammation during EAE development. EAE was induced in C57BL/6J mice and 1,25-dihydroxyvitamin D3 administration began 1 day after disease induction. This procedure decreased prevalence, clinical score, inflammation, and demyelination. It also reduced MHCII expression in macrophages and microglia as well as the level of oxidative stress and messenger RNA (mRNA) expression for NLRP3, caspase-1, interleukin (IL)-1β, CX3CR1, CCL17, RORc and Tbx21 at the CNS. Otherwise, mRNA expression for ZO-1 increased at the lumbar spinal cord. These effects were accompanied by the stabilization of blood-spinal cord barrier permeability. The results of this study indicate that early intervention with 1,25-dihydroxyvitamin D3 can control the neuroinflammatory process that is the hallmark of EAE and MS immunopathogenesis and should thus be explored as an adjunct therapy for MS patients.
Collapse
Affiliation(s)
| | - Luiza Ayumi Nishiyama Mimura
- Department of Microbiology and Immunology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, Brazil
| | | | - Larissa Lumi Watanabe Ishikawa
- Department of Microbiology and Immunology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, Brazil
| | | | | | - Alexandrina Sartori
- Department of Microbiology and Immunology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, Brazil
| |
Collapse
|
14
|
Kierdorf K, Masuda T, Jordão MJC, Prinz M. Macrophages at CNS interfaces: ontogeny and function in health and disease. Nat Rev Neurosci 2019; 20:547-562. [PMID: 31358892 DOI: 10.1038/s41583-019-0201-x] [Citation(s) in RCA: 260] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2019] [Indexed: 12/16/2022]
Abstract
The segregation and limited regenerative capacity of the CNS necessitate a specialized and tightly regulated resident immune system that continuously guards the CNS against invading pathogens and injury. Immunity in the CNS has generally been attributed to neuron-associated microglia in the parenchyma, whose origin and functions have recently been elucidated. However, there are several other specialized macrophage populations at the CNS borders, including dural, leptomeningeal, perivascular and choroid plexus macrophages (collectively known as CNS-associated macrophages (CAMs)), whose origins and roles in health and disease have remained largely uncharted. CAMs are thought to be involved in regulating the fine balance between the proper segregation of the CNS, on the one hand, and the essential exchange between the CNS parenchyma and the periphery, on the other. Recent studies that have been empowered by major technological advances have shed new light on these cells and suggest central roles for CAMs in CNS physiology and in the pathogenesis of diseases.
Collapse
Affiliation(s)
- Katrin Kierdorf
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Centre for Integrative Biological Signalling Studies (CIBSS), University of Freiburg, Freiburg, Germany.,Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Takahiro Masuda
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany. .,Centre for Integrative Biological Signalling Studies (CIBSS), University of Freiburg, Freiburg, Germany. .,Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany. .,Centre for Biological Signalling Studies (BIOSS), University of Freiburg, Freiburg, Germany.
| |
Collapse
|
15
|
Microglia in Central Nervous System Inflammation and Multiple Sclerosis Pathology. Trends Mol Med 2018; 25:112-123. [PMID: 30578090 DOI: 10.1016/j.molmed.2018.11.005] [Citation(s) in RCA: 340] [Impact Index Per Article: 48.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 11/22/2018] [Accepted: 11/26/2018] [Indexed: 02/06/2023]
Abstract
Microglia are the resident macrophages of the central nervous system (CNS). They have important physiological functions in maintaining tissue homeostasis but also contribute to CNS pathology. Microglia respond to changes in the microenvironment, and the resulting reactive phenotype can be very diverse, with both neuroinflammatory and neuroprotective properties, illustrating the plasticity of these cells. Recent progress in understanding the autoimmune neuroinflammatory disease multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis suggests major roles for microglia in the disease, which have drastically changed our view on the function of microglia in MS.
Collapse
|
16
|
Russi AE, Walker-Caulfield ME, Brown MA. Mast cell inflammasome activity in the meninges regulates EAE disease severity. Clin Immunol 2018; 189:14-22. [DOI: 10.1016/j.clim.2016.04.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 04/18/2016] [Accepted: 04/19/2016] [Indexed: 12/16/2022]
|
17
|
Brown MA, Weinberg RB. Mast Cells and Innate Lymphoid Cells: Underappreciated Players in CNS Autoimmune Demyelinating Disease. Front Immunol 2018; 9:514. [PMID: 29619025 PMCID: PMC5871669 DOI: 10.3389/fimmu.2018.00514] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 02/27/2018] [Indexed: 12/29/2022] Open
Abstract
Multiple sclerosis (MS) and its mouse model, experimental autoimmune encephalomyelitis, are autoimmune CNS inflammatory diseases. As a result of a breakdown in the relatively impermeable blood–brain barrier (BBB) in affected individuals, myelin-specific CD4+ and CD8+ T cells gain entry into the immune privileged CNS and initiate myelin, oligodendrocyte, and nerve axon destruction. However, despite the absolute requirement for T cells, there is increasing evidence that innate immune cells also play critical amplifying roles in disease pathogenesis. By modulating the character and magnitude of the myelin-reactive T cell response and regulating BBB integrity, innate cells affect both disease initiation and progression. Two classes of innate cells, mast cells and innate lymphoid cells (ILCs), have been best studied in models of allergic and gastrointestinal inflammatory diseases. Yet, there is emerging evidence that these cell types also exert a profound influence in CNS inflammatory disease. Both cell types are residents within the meninges and can be activated early in disease to express a wide variety of disease-modifying cytokines and chemokines. In this review, we discuss how mast cells and ILCs can have either disease-promoting or -protecting effects on MS and other CNS inflammatory diseases and how sex hormones may influence this outcome. These observations suggest that targeting these cells and their unique mediators can be exploited therapeutically.
Collapse
Affiliation(s)
- Melissa A Brown
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Rebecca B Weinberg
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
18
|
Brown MA. Studies of Mast Cells: Adventures in Serendipity. Front Immunol 2018; 9:520. [PMID: 29593744 PMCID: PMC5859373 DOI: 10.3389/fimmu.2018.00520] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 02/28/2018] [Indexed: 01/14/2023] Open
Abstract
Like many of us who had the great fortune to work with Bill Paul, my science life was immeasurably altered by my interactions with him. Although intimidating at first because of his stature in the immunology world, it was soon clear that he not only truly cared about the specific research we were doing together, but he wished to convey to his trainees an approach to science that was open, always questioning, and infinitely fun. His enthusiasm was infectious and after my training with him, despite stresses due to funding and publishing hurdles, I never regretted the path I took. My research took a sharp turn from the studies of adaptive immunity I had planned on pursuing after my fellowship with Bill to a life long quest to understand the wonders of the mast cell, a relatively rare innate immune cell. This came about because Bill’s curiosity and expectation of the unexpected allowed him to view, in retrospect, a rather mundane observation we made together involving a non-physiological transformed mast cell line as something that might be really interesting. I have never forgotten that lesson: Look at the data with an eye on the big picture. Sometimes the unexpected is more interesting than predicted results. His example in this regard was incredibly important when as an independent investigator a mistake in mouse sex determination led to unexpected and very confusing data. Yet, these data ultimately revealed a role for mast cells in male-specific protection in experimental autoimmune encephalomyelitis, the mouse model of multiple sclerosis. Bill’s influence in immunology is far-reaching and will continue to be felt as those of us who train our own students and post-doctoral fellows pass on his wisdom and approach to scientific research.
Collapse
Affiliation(s)
- Melissa A Brown
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
19
|
Male-specific IL-33 expression regulates sex-dimorphic EAE susceptibility. Proc Natl Acad Sci U S A 2018; 115:E1520-E1529. [PMID: 29378942 DOI: 10.1073/pnas.1710401115] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The cellular and molecular basis of sex-dimorphic autoimmune diseases, such as the CNS demyelinating disease multiple sclerosis (MS), remains unclear. Our studies in the SJL mouse model of MS, experimental autoimmune encephalomyelitis (EAE), reveal that sex-determined differences in Il33 expression by innate immune cells in response to myelin peptide immunization regulate EAE susceptibility. IL-33 is selectively induced in PLP139-151-immunized males and activates type 2 innate lymphoid cells (ILC2s), cells that promote and sustain a nonpathogenic Th2 myelin-specific response. Without this attenuating IL-33 response, females generate an encephalitogenic Th17-dominant response, which can be reversed by IL-33 treatment. Mast cells are one source of IL-33 and we provide evidence that testosterone directly induces Il33 gene expression and also exerts effects on the potential for Il33 gene expression during mast cell development. Thus, in contrast to their pathogenic role in allergy, we propose a sex-specific role for both mast cells and ILC2s as attenuators of the pathogenic Th response in CNS inflammatory disease.
Collapse
|
20
|
Shrestha B, Jiang X, Ge S, Paul D, Chianchiano P, Pachter JS. Spatiotemporal resolution of spinal meningeal and parenchymal inflammation during experimental autoimmune encephalomyelitis. Neurobiol Dis 2017; 108:159-172. [PMID: 28844788 DOI: 10.1016/j.nbd.2017.08.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 08/10/2017] [Accepted: 08/18/2017] [Indexed: 01/14/2023] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) induced by active immunization of C57BL/6 mice with peptide from myelin oligodendrocyte protein (MOG35-55), is a neuroinflammatory, demyelinating disease widely recognized as an animal model of multiple sclerosis (MS). Typically, EAE presents with an ascending course of paralysis, and inflammation that is predominantly localized to the spinal cord. Recent studies have further indicated that inflammation - in both MS and EAE - might initiate within the meninges and propagate from there to the underlying parenchyma. However, the patterns of inflammation within the respective meningeal and parenchymal compartments along the length of the spinal cord, and the progression with which these patterns develop during EAE, have yet to be detailed. Such analysis could hold key to identifying factors critical for spreading, as well as constraining, inflammation along the neuraxis. To address this issue, high-resolution 3-dimensional (3D) confocal microscopy was performed to visualize, in detail, the sequence of leukocyte infiltration at distinct regions of the spinal cord. High quality virtual slide scanning for imaging the entire spinal cord using epifluorescence was further conducted to highlight the directionality and relative degree of inflammation. Meningeal inflammation was found to precede parenchymal inflammation at all levels of the spinal cord, but did not develop equally or simultaneously throughout the subarachnoid space (SAS) of the meninges. Instead, meningeal inflammation was initially most obvious in the caudal SAS, from which it progressed to the immediate underlying parenchyma, paralleling the first signs of clinical disease in the tail and hind limbs. Meningeal inflammation could then be seen to extend in the caudal-to-rostral direction, followed by a similar, but delayed, trajectory of parenchymal inflammation. To additionally determine whether the course of ascending paralysis and leukocyte infiltration during EAE is reflected in differences in inflammatory gene expression by meningeal and parenchymal microvessels along the spinal cord, laser capture microdissection (LCM) coupled with gene expression profiling was performed. Expression profiles varied between these respective vessel populations at both the cervical and caudal levels of the spinal cord during disease progression, and within each vessel population at different levels of the cord at a given time during disease. These results reinforce a significant role for the meninges in the development and propagation of central nervous system inflammation associated with MS and EAE.
Collapse
Affiliation(s)
- Bandana Shrestha
- Blood-Brain Barrier Laboratory, Dept. of Cell Biology, UConn Health, 263 Farmington Ave, Farmington, CT 06030, United States.
| | - Xi Jiang
- Blood-Brain Barrier Laboratory, Dept. of Cell Biology, UConn Health, 263 Farmington Ave, Farmington, CT 06030, United States.
| | - Shujun Ge
- Blood-Brain Barrier Laboratory, Dept. of Cell Biology, UConn Health, 263 Farmington Ave, Farmington, CT 06030, United States.
| | - Debayon Paul
- Blood-Brain Barrier Laboratory, Dept. of Cell Biology, UConn Health, 263 Farmington Ave, Farmington, CT 06030, United States.
| | - Peter Chianchiano
- Blood-Brain Barrier Laboratory, Dept. of Cell Biology, UConn Health, 263 Farmington Ave, Farmington, CT 06030, United States.
| | - Joel S Pachter
- Blood-Brain Barrier Laboratory, Dept. of Cell Biology, UConn Health, 263 Farmington Ave, Farmington, CT 06030, United States.
| |
Collapse
|
21
|
Tesfagiorgis Y, Zhu SL, Jain R, Kerfoot SM. Activated B Cells Participating in the Anti-Myelin Response Are Excluded from the Inflamed Central Nervous System in a Model of Autoimmunity that Allows for B Cell Recognition of Autoantigen. THE JOURNAL OF IMMUNOLOGY 2017; 199:449-457. [DOI: 10.4049/jimmunol.1602042] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 05/09/2017] [Indexed: 11/19/2022]
|
22
|
Coles JA, Myburgh E, Brewer JM, McMenamin PG. Where are we? The anatomy of the murine cortical meninges revisited for intravital imaging, immunology, and clearance of waste from the brain. Prog Neurobiol 2017; 156:107-148. [PMID: 28552391 DOI: 10.1016/j.pneurobio.2017.05.002] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 04/25/2017] [Accepted: 05/08/2017] [Indexed: 12/15/2022]
Abstract
Rapid progress is being made in understanding the roles of the cerebral meninges in the maintenance of normal brain function, in immune surveillance, and as a site of disease. Most basic research on the meninges and the neural brain is now done on mice, major attractions being the availability of reporter mice with fluorescent cells, and of a huge range of antibodies useful for immunocytochemistry and the characterization of isolated cells. In addition, two-photon microscopy through the unperforated calvaria allows intravital imaging of the undisturbed meninges with sub-micron resolution. The anatomy of the dorsal meninges of the mouse (and, indeed, of all mammals) differs considerably from that shown in many published diagrams: over cortical convexities, the outer layer, the dura, is usually thicker than the inner layer, the leptomeninx, and both layers are richly vascularized and innervated, and communicate with the lymphatic system. A membrane barrier separates them and, in disease, inflammation can be localized to one layer or the other, so experimentalists must be able to identify the compartment they are studying. Here, we present current knowledge of the functional anatomy of the meninges, particularly as it appears in intravital imaging, and review their role as a gateway between the brain, blood, and lymphatics, drawing on information that is scattered among works on different pathologies.
Collapse
Affiliation(s)
- Jonathan A Coles
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, Sir Graeme Davis Building, University of Glasgow, Glasgow, G12 8TA, United Kingdom.
| | - Elmarie Myburgh
- Centre for Immunology and Infection Department of Biology, University of York, Wentworth Way, Heslington, York YO10 5DD, United Kingdom
| | - James M Brewer
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, Sir Graeme Davis Building, University of Glasgow, Glasgow, G12 8TA, United Kingdom
| | - Paul G McMenamin
- Department of Anatomy & Developmental Biology, School of Biomedical and Psychological Sciences and Monash Biomedical Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, 10 Chancellor's Walk, Clayton, Victoria, 3800, Australia
| |
Collapse
|
23
|
The Involvement of Pial Microvessels in Leukocyte Invasion after Mild Traumatic Brain Injury. PLoS One 2016; 11:e0167677. [PMID: 28030563 PMCID: PMC5193324 DOI: 10.1371/journal.pone.0167677] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 11/20/2016] [Indexed: 12/23/2022] Open
Abstract
The pathophysiological mechanisms underlying mild traumatic brain injury (mTBI) are not well understood, but likely involve neuroinflammation. Here the controlled cortical impact model of mTBI in rats was used to test this hypothesis. Mild TBI caused a rapid (within 6 h post-mTBI) upregulation of synthesis of TNF-α and IL-1β in the cerebral cortex and hippocampus, followed by an increase in production of neutrophil (CXCL1-3) and monocyte (CCL2) chemoattractants. While astrocytes were not a significant source of CXC chemokines, they highly expressed CCL2. An increase in production of CXC chemokines coincided with the influx of neutrophils into the injured brain. At 6 h post-mTBI, we observed a robust influx of CCL2-expressing neutrophils across pial microvessels into the subarachnoid space (SAS) near the injury site. Mild TBI was not accompanied by any significant influx of neutrophils into the brain parenchyma until 24 h after injury. This was associated with an early induction of expression of intercellular adhesion molecule 1 on the endothelium of the ipsilateral pial, but not intraparenchymal, microvessels. At 6 h post-mTBI, we also observed a robust influx of neutrophils into the ipsilateral cistern of velum interpositum (CVI), a slit-shaped cerebrospinal fluid space located above the 3rd ventricle with highly vascularized pia mater. From SAS and CVI, neutrophils appeared to move along the perivascular spaces to enter the brain parenchyma. The monocyte influx was not observed until 24 h post-mTBI, and these inflammatory cells predominantly entered the ipsilateral SAS and CVI, with a limited invasion of brain parenchyma. These observations indicate that the endothelium of pial microvessels responds to injury differently than that of intraparenchymal microvessels, which may be associated with the lack of astrocytic ensheathment of cerebrovascular endothelium in pial microvessels. These findings also suggest that neuroinflammation represents the potential therapeutic target in mTBI.
Collapse
|
24
|
Louveau A, Da Mesquita S, Kipnis J. Lymphatics in Neurological Disorders: A Neuro-Lympho-Vascular Component of Multiple Sclerosis and Alzheimer's Disease? Neuron 2016; 91:957-973. [PMID: 27608759 PMCID: PMC5019121 DOI: 10.1016/j.neuron.2016.08.027] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Lymphatic vasculature drains interstitial fluids, which contain the tissue's waste products, and ensures immune surveillance of the tissues, allowing immune cell recirculation. Until recently, the CNS was considered to be devoid of a conventional lymphatic vasculature. The recent discovery in the meninges of a lymphatic network that drains the CNS calls into question classic models for the drainage of macromolecules and immune cells from the CNS. In the context of neurological disorders, the presence of a lymphatic system draining the CNS potentially offers a new player and a new avenue for therapy. In this review, we will attempt to integrate the known primary functions of the tissue lymphatic vasculature that exists in peripheral organs with the proposed function of meningeal lymphatic vessels in neurological disorders, specifically multiple sclerosis and Alzheimer's disease. We propose that these (and potentially other) neurological afflictions can be viewed as diseases with a neuro-lympho-vascular component and should be therapeutically targeted as such.
Collapse
Affiliation(s)
- Antoine Louveau
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Sandro Da Mesquita
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Jonathan Kipnis
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
25
|
Mast cell activation disease and the modern epidemic of chronic inflammatory disease. Transl Res 2016; 174:33-59. [PMID: 26850903 DOI: 10.1016/j.trsl.2016.01.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Revised: 01/07/2016] [Accepted: 01/08/2016] [Indexed: 12/18/2022]
Abstract
A large and growing portion of the human population, especially in developed countries, suffers 1 or more chronic, often quite burdensome ailments which either are overtly inflammatory in nature or are suspected to be of inflammatory origin, but for which investigations to date have failed to identify specific causes, let alone unifying mechanisms underlying the multiple such ailments that often afflict such patients. Relatively recently described as a non-neoplastic cousin of the rare hematologic disease mastocytosis, mast cell (MC) activation syndrome-suspected to be of greatly heterogeneous, complex acquired clonality in many cases-is a potential underlying/unifying explanation for a diverse assortment of inflammatory ailments. A brief review of MC biology and how aberrant primary MC activation might lead to such a vast range of illness is presented.
Collapse
|
26
|
van den Berg R, Laman JD, van Meurs M, Hintzen RQ, Hoogenraad CC. Rotarod motor performance and advanced spinal cord lesion image analysis refine assessment of neurodegeneration in experimental autoimmune encephalomyelitis. J Neurosci Methods 2016; 262:66-76. [PMID: 26784021 DOI: 10.1016/j.jneumeth.2016.01.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Revised: 01/07/2016] [Accepted: 01/07/2016] [Indexed: 12/28/2022]
Abstract
BACKGROUND Experimental autoimmune encephalomyelitis (EAE) is a commonly used experimental model for multiple sclerosis (MS). Experience with this model mainly comes from the field of immunology, while data on its use in studying the neurodegenerative aspects of MS is scarce. NEW METHOD The aim of this study is to improve and refine methods to assess neurodegeneration and function in EAE. Using the rotarod, a tool used in neuroscience to monitor motor performance, we evaluated the correlation between motor performance, disease severity as measured using a clinical scale and area covered by inflammatory lesions. RESULTS The included parameters are highly correlated in a non-linear manner, with motor performance rapidly decreasing in the intermediate values of the clinical scale. The relation between motor performance and histopathological damage is exclusively determined by lesions in the ventral and lateral columns, based on a new method of analysis of the entire spinal cord. Using a set of definitions for distinct disease milestones, we quantified disease duration as well as severity. COMPARISON WITH EXISTING METHODS The rotarod measures motor performance in a more objective and quantitative manner compared to using a clinical score. The outcome shows a strong correlation to the surface area of inflammatory lesions in the motor systems of the spinal cord. CONCLUSIONS These results provide an improved workflow for interpreting the outcome of EAE from a neurological point of view, with the eventual goal of dissecting neurodegeneration and evaluating neuroprotective drugs in EAE for application in MS.
Collapse
Affiliation(s)
- Robert van den Berg
- Cell Biology, Utrecht University, Utrecht, The Netherlands; Department of Neurology, Erasmus MC, Rotterdam, The Netherlands
| | - Jon D Laman
- Department of Neuroscience, University Groningen, University Medical Center Groningen, The Netherlands
| | - Marjan van Meurs
- Department of Immunology, Erasmus MC, Rotterdam, The Netherlands
| | | | | |
Collapse
|
27
|
Pikor NB, Prat A, Bar-Or A, Gommerman JL. Meningeal Tertiary Lymphoid Tissues and Multiple Sclerosis: A Gathering Place for Diverse Types of Immune Cells during CNS Autoimmunity. Front Immunol 2016; 6:657. [PMID: 26793195 PMCID: PMC4710700 DOI: 10.3389/fimmu.2015.00657] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 12/22/2015] [Indexed: 11/13/2022] Open
Abstract
Collections of leukocytes in the meningeal space have been documented in Multiple Sclerosis (MS). These meningeal aggregates, which in the context of other autoimmune diseases have often been termed tertiary lymphoid tissues (TLT), have been associated with sub-pial cortical damage and disease progression. However, the key molecular and cellular signals required for their formation and maintenance remain unclear. Herein, we review TLT structures in other disease states in order to provide a framework for understanding these structures in the MS meninges. We then assess the evidence that the meningeal compartment serves as an important nexus for immune cells as well as a location for drainage of antigen into cervical lymph nodes. Extrapolating what is known about the molecular and cellular cues that initiate the formation of leukocyte aggregates in non-lymphoid tissues, we speculate on what signals lead to the formation and maintenance of meningeal TLT structures. Referring to the animal model of MS [experimental autoimmune encephalomyelitis (EAE)], we also explore what is known about these structures in supporting B cell and T cell responses during neuroinflammation. Last, we examine the evidence that connects these structures to ongoing neuropathology. Collectively, our review points to the meningeal compartment as an important player in neuroinflammatory processes. Moreover, we hypothesize that in order to gain insights into pro- and anti-inflammatory properties of lymphocytes in MS, one must understand the cellular scaffolds that support lymphocyte retention within the meninges, thus highlighting the importance of non-immune cells (stromal cells) in the neuroinflammatory process.
Collapse
Affiliation(s)
- Natalia B Pikor
- Department of Immunology, University of Toronto , Toronto, ON , Canada
| | - Alexandre Prat
- Neuroimmunology Unit, Department of Neuroscience, Centre de Recherche de CHUM, Université de Montréal , Montreal, QC , Canada
| | - Amit Bar-Or
- Neuroimmunology Unit, Montreal Neurological Institute and Hospital, McGill University , Montreal, QC , Canada
| | | |
Collapse
|
28
|
NFκB signaling drives pro-granulocytic astroglial responses to neuromyelitis optica patient IgG. J Neuroinflammation 2015; 12:185. [PMID: 26423139 PMCID: PMC4590277 DOI: 10.1186/s12974-015-0403-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 09/22/2015] [Indexed: 01/21/2023] Open
Abstract
Background Astrocytes expressing the aquaporin-4 water channel are a primary target of pathogenic, disease-specific immunoglobulins (IgG) found in patients with neuromyelitis optica (NMO). Immunopathological analyses of active NMO lesions highlight a unique inflammatory phenotype marked by infiltration of granulocytes. Previous studies characterized this granulocytic infiltrate as a response to vasculocentric complement activation and localized tissue destruction. In contrast, we observe that granulocytic infiltration in NMO lesions occurs independently of complement-mediated tissue destruction or active demyelination. These immunopathological findings led to the hypothesis that NMO IgG stimulates astrocyte signaling that is responsible for granulocytic recruitment in NMO. Methods Histopathology was performed on archival formalin-fixed paraffin-embedded autopsy-derived CNS tissue from 23 patients clinically and pathologically diagnosed with NMO or NMO spectrum disorder. Primary murine astroglial cultures were stimulated with IgG isolated from NMO patients or control IgG from healthy donors. Transcriptional responses were assessed by microarray, and translational responses were measured by ELISA. Signaling through the NFκB pathway was measured by western blotting and immunostaining. Results Stimulation of primary murine astroglial cultures with NMO IgG elicited a reactive and inflammatory transcriptional response that involved signaling through the canonical NFκB pathway. This signaling resulted in the release of pro-granulocytic chemokines and was inhibited by the clinically relevant proteasome inhibitors bortezomib and PR-957. Conclusions We propose that the astrocytic NFκB-dependent inflammatory response to stimulation by NMO IgG represents one of the earliest events in NMO pathogenesis, providing a target for therapeutic intervention upstream of irreversible cell death and tissue damage. Electronic supplementary material The online version of this article (doi:10.1186/s12974-015-0403-8) contains supplementary material, which is available to authorized users.
Collapse
|
29
|
Do not judge a cell by its cover--diversity of CNS resident, adjoining and infiltrating myeloid cells in inflammation. Semin Immunopathol 2015; 37:591-605. [PMID: 26251238 DOI: 10.1007/s00281-015-0520-6] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 07/27/2015] [Indexed: 12/24/2022]
Abstract
Specialized populations of tissue-resident myeloid cells inhabit every organ of the body. While many of these populations appear similar morphologically and phenotypically, they exhibit great functional diversity. The central nervous system (CNS), as an immune privileged organ, possesses a unique tissue-resident macrophage population, the microglia, as well as numerous myeloid cell subsets at its boarders and barriers in CNS-adjoining tissues, namely the meninges, the perivascular space, and the choroid plexus. Recent research has added much to our knowledge about microglia, whereas the populations of CNS-surrounding phagocytes are just starting to be appreciated. As guardians of CNS homeostasis, these myeloid cells perform immune surveillance and immune modulatory tasks in health and disease. As such, microglia and CNS-surrounding antigen-presenting cells have been shown to be crucially involved not only in the initiation and progression but also resolution of multiple sclerosis (MS). MS and its rodent model, experimental autoimmune encephalomyelitis, are autoimmune inflammatory demyelinating CNS pathologies. While some crucial aspects of the disease pathogenesis have been solved, much of the complex involvement and interplay of the innate immune compartment remains yet to be clarified. Here, we will discuss the current understanding of the scope of phenotypes and functions of myeloid cells involved in CNS neuroinflammation.
Collapse
|
30
|
Group 3 innate lymphoid cells accumulate and exhibit disease-induced activation in the meninges in EAE. Cell Immunol 2015; 297:69-79. [PMID: 26163773 DOI: 10.1016/j.cellimm.2015.06.006] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 05/31/2015] [Accepted: 06/18/2015] [Indexed: 12/11/2022]
Abstract
Innate lymphoid cells are immune cells that reside in tissues that interface with the external environment and contribute to the first line defense against pathogens. However, they also have roles in promoting chronic inflammation. Here we demonstrate that group 3 ILCs, (ILC3s - CD45+Lin-IL-7Rα+RORγt+), are normal residents of the meninges and exhibit disease-induced accumulation and activation in EAE. In addition to production of the pro-inflammatory cytokines IL-17 and GM-CSF, ILC3s constitutively express CD30L and OX40L, molecules required for memory T cell survival. We show that disease-induced trafficking of transferred wild type T cells to the meninges is impaired in ILC3-deficient Rorc-/- mice. Furthermore, lymphoid tissue inducer cells, a c-kit+ ILC3 subset that promotes ectopic lymphoid follicle development, a hallmark of many autoimmune diseases, are reduced in the meninges of EAE-resistant c-kit mutant Kit(W/Wv) mice. We propose that ILC3s sustain neuroinflammation by supporting T cell survival and reactivation in the meninges.
Collapse
|
31
|
Russi AE, Walker-Caulfield ME, Ebel ME, Brown MA. Cutting edge: c-Kit signaling differentially regulates type 2 innate lymphoid cell accumulation and susceptibility to central nervous system demyelination in male and female SJL mice. THE JOURNAL OF IMMUNOLOGY 2015; 194:5609-13. [PMID: 25972476 DOI: 10.4049/jimmunol.1500068] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 04/10/2015] [Indexed: 12/20/2022]
Abstract
Multiple sclerosis preferentially affects women, and this sexual dimorphism is recapitulated in the SJL mouse model of multiple sclerosis, experimental autoimmune encephalomyelitis (EAE). In this study, we demonstrate that signaling through c-Kit exerts distinct effects on EAE susceptibility in male and female SJL mice. Previous studies in females show that Kit mutant (W/W(v)) mice are less susceptible to EAE than are wild-type mice. However, male W/W(v) mice exhibit exacerbated disease, a phenotype independent of mast cells and corresponding to a shift from a Th2- to a Th17-dominated T cell response. We demonstrate a previously undescribed deficit in c-Kit(+) type 2 innate lymphoid cells (ILC2s) in W/W(v) mice. ILC2s are also significantly reduced in EAE-susceptible wild-type females, indicating that both c-Kit signals and undefined male-specific factors are required for ILC2 function. We propose that deficiencies in Th2-promoting ILC2s remove an attenuating influence on the encephalitogenic T cell response and therefore increases disease susceptibility.
Collapse
Affiliation(s)
- Abigail E Russi
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611; and
| | | | - Mark E Ebel
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611; and
| | - Melissa A Brown
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611; and
| |
Collapse
|