1
|
Stasiak K, Stevens AD, Bolte AC, Curley CT, Perusina Lanfranca M, Lindsay RS, Eyo UB, Lukens JR, Price RJ, Bullock TNJ, Engelhard VH. Differential T cell accumulation within intracranial and subcutaneous melanomas is associated with differences in intratumoral myeloid cells. Cancer Immunol Immunother 2024; 74:10. [PMID: 39487854 DOI: 10.1007/s00262-024-03832-0] [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: 07/29/2024] [Accepted: 09/06/2024] [Indexed: 11/04/2024]
Abstract
Patients with metastatic brain melanomas (MBM) experience shorter-lasting survival than patients with extracranial metastases, and this is associated with a higher fraction of dysfunctional CD8 T cells. The goal of this study was to understand the underlying cause of T cell dysfunction in MBM. To accomplish this, we compared murine B16 melanomas implanted intracranially (IC) or subcutaneously (SC). CD8 T cell activation was not altered, but representation in IC tumors was lower. Transferred activated or naïve CD8 T cells accumulated in similar numbers in both tumors, suggesting that the vasculature does not differentially impair T cell presence. Surprisingly, we found no evidence for T cell activation in draining lymph nodes of SC or IC tumor-bearing mice, consistent with the fact that dendritic cells (DC) that had acquired tumor antigen showed an immature phenotype. Instead, T cell activation occurred within both tumors, where the majority of tumor antigen+ myeloid cells were found. While, the numbers of intratumoral DC were comparable, those in IC tumors acquired less tumor antigen, and were alternatively matured based on upregulation of MHCII without upregulation of CD86. Additionally, in IC tumors, the largest population of tumor antigen+ myeloid cells were microglia. However, their presence did not influence either antigen acquisition or the phenotype of other myeloid cell populations. Overall, our data suggest that diminished representation of CD8 T cells in IC tumors is a consequence of alternatively matured DC and/or microglia that induce distinctly activated T cells, which ultimately fail to continue to accumulate inside the tumor.
Collapse
Affiliation(s)
- Katarzyna Stasiak
- Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, USA
| | - Aaron D Stevens
- Department of Pathology, University of Virginia, Charlottesville, VA, USA
| | - Ashley C Bolte
- Department of Neuroscience, University of Virginia, Charlottesville, USA
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, USA
| | - Colleen T Curley
- Department of Biomedical Engineering, University of Virgnia, Charlottesville, VA, USA
| | - Mirna Perusina Lanfranca
- Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, USA
| | - Robin S Lindsay
- Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, USA
| | - Ukpong B Eyo
- Department of Neuroscience, University of Virginia, Charlottesville, USA
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, USA
| | - John R Lukens
- Department of Neuroscience, University of Virginia, Charlottesville, USA
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, USA
| | - Richard J Price
- Department of Biomedical Engineering, University of Virgnia, Charlottesville, VA, USA
- Department of Radiology & Medical Imaging, University of Virginia, Charlottesville, USA
| | | | - Victor H Engelhard
- Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA.
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, USA.
| |
Collapse
|
2
|
Wang M, Caryotakis SE, Smith GG, Nguyen AV, Pleasure DE, Soulika AM. CSF1R antagonism results in increased supraspinal infiltration in EAE. J Neuroinflammation 2024; 21:103. [PMID: 38643194 PMCID: PMC11031888 DOI: 10.1186/s12974-024-03063-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 03/11/2024] [Indexed: 04/22/2024] Open
Abstract
BACKGROUND Colony stimulating factor 1 receptor (CSF1R) signaling is crucial for the maintenance and function of various myeloid subsets. CSF1R antagonism was previously shown to mitigate clinical severity in experimental autoimmune encephalomyelitis (EAE). The associated mechanisms are still not well delineated. METHODS To assess the effect of CSF1R signaling, we employed the CSF1R antagonist PLX5622 formulated in chow (PLX5622 diet, PD) and its control chow (control diet, CD). We examined the effect of PD in steady state and EAE by analyzing cells isolated from peripheral immune organs and from the CNS via flow cytometry. We determined CNS infiltration sites and assessed the extent of demyelination using immunohistochemistry of cerebella and spinal cords. Transcripts of genes associated with neuroinflammation were also analyzed in these tissues. RESULTS In addition to microglial depletion, PD treatment reduced dendritic cells and macrophages in peripheral immune organs, both during steady state and during EAE. Furthermore, CSF1R antagonism modulated numbers and relative frequencies of T effector cells both in the periphery and in the CNS during the early stages of the disease. Classical neurological symptoms were milder in PD compared to CD mice. Interestingly, a subset of PD mice developed atypical EAE symptoms. Unlike previous studies, we observed that the CNS of PD mice was infiltrated by increased numbers of peripheral immune cells compared to that of CD mice. Immunohistochemical analysis showed that CNS infiltrates in PD mice were mainly localized in the cerebellum while in CD mice infiltrates were primarily localized in the spinal cords during the onset of neurological deficits. Accordingly, during the same timepoint, cerebella of PD but not of CD mice had extensive demyelinating lesions, while spinal cords of CD but not of PD mice were heavily demyelinated. CONCLUSIONS Our findings suggest that CSF1R activity modulates the cellular composition of immune cells both in the periphery and within the CNS, and affects lesion localization during the early EAE stages.
Collapse
Affiliation(s)
- Marilyn Wang
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Sofia E Caryotakis
- Shriners Hospitals for Children, Northern California, Sacramento, CA, USA
- University of California, San Francisco, San Francisco, CA, USA
| | - Glendalyn G Smith
- Shriners Hospitals for Children, Northern California, Sacramento, CA, USA
| | - Alan V Nguyen
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA, USA
- Sutro Biosciences, South San Francisco, CA, USA
| | - David E Pleasure
- Shriners Hospitals for Children, Northern California, Sacramento, CA, USA
- Department of Neurology, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Athena M Soulika
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA, USA.
- Shriners Hospitals for Children, Northern California, Sacramento, CA, USA.
| |
Collapse
|
3
|
Nguyen LTM, Hassan S, Pan H, Wu S, Wen Z. Interplay of Zeb2a, Id2a and Batf3 regulates microglia and dendritic cell development in the zebrafish brain. Development 2024; 151:dev201829. [PMID: 38240311 DOI: 10.1242/dev.201829] [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: 04/01/2023] [Accepted: 01/08/2024] [Indexed: 02/06/2024]
Abstract
In vertebrates, the central nervous system (CNS) harbours various immune cells, including parenchymal microglia, perivascular macrophages and dendritic cells, which act in coordination to establish an immune network to regulate neurogenesis and neural function, and to maintain the homeostasis of the CNS. Recent single cell transcriptomic profiling has revealed that the adult zebrafish CNS contains microglia, plasmacytoid dendritic cells (pDCs) and two conventional dendritic cells (cDCs), ccl35+ cDCs and cnn3a+cDCs. However, how these distinct myeloid cells are established in the adult zebrafish CNS remains incompletely defined. Here, we show that the Inhibitor of DNA binding 2a (Id2a) is essential for the development of pDCs and cDCs but is dispensable for the formation of microglia, whereas the Basic leucine zipper transcription factor ATF-like 3 (Batf3) acts downstream of id2a and is required exclusively for the formation of the cnn3a+ cDC subset. In contrast, the Zinc finger E-box-binding homeobox 2a (Zeb2a) promotes the expansion of microglia and inhibits the DC specification, possibly through repressing id2a expression. Our study unravels the genetic networks that govern the development of microglia and brain-associated DCs in the zebrafish CNS.
Collapse
Affiliation(s)
- Linh Thi My Nguyen
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, the Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Shaoli Hassan
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, the Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Hongru Pan
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, the Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Shuting Wu
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Zilong Wen
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, the Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen 518055, China
- Department of Immunology and Microbiology, School of Life Science, the Southern University of Science and Technology, Shenzhen 518000, China
| |
Collapse
|
4
|
Butic AB, Spencer SA, Shaheen SK, Lukacher AE. Polyomavirus Wakes Up and Chooses Neurovirulence. Viruses 2023; 15:2112. [PMID: 37896889 PMCID: PMC10612099 DOI: 10.3390/v15102112] [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: 09/29/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
JC polyomavirus (JCPyV) is a human-specific polyomavirus that establishes a silent lifelong infection in multiple peripheral organs, predominantly those of the urinary tract, of immunocompetent individuals. In immunocompromised settings, however, JCPyV can infiltrate the central nervous system (CNS), where it causes several encephalopathies of high morbidity and mortality. JCPyV-induced progressive multifocal leukoencephalopathy (PML), a devastating demyelinating brain disease, was an AIDS-defining illness before antiretroviral therapy that has "reemerged" as a complication of immunomodulating and chemotherapeutic agents. No effective anti-polyomavirus therapeutics are currently available. How depressed immune status sets the stage for JCPyV resurgence in the urinary tract, how the virus evades pre-existing antiviral antibodies to become viremic, and where/how it enters the CNS are incompletely understood. Addressing these questions requires a tractable animal model of JCPyV CNS infection. Although no animal model can replicate all aspects of any human disease, mouse polyomavirus (MuPyV) in mice and JCPyV in humans share key features of peripheral and CNS infection and antiviral immunity. In this review, we discuss the evidence suggesting how JCPyV migrates from the periphery to the CNS, innate and adaptive immune responses to polyomavirus infection, and how the MuPyV-mouse model provides insights into the pathogenesis of JCPyV CNS disease.
Collapse
Affiliation(s)
| | | | | | - Aron E. Lukacher
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, PA 17033, USA; (A.B.B.); (S.A.S.); (S.K.S.)
| |
Collapse
|
5
|
Zhou Q, Zhao C, Yang Z, Qu R, Li Y, Fan Y, Tang J, Xie T, Wen Z. Cross-organ single-cell transcriptome profiling reveals macrophage and dendritic cell heterogeneity in zebrafish. Cell Rep 2023; 42:112793. [PMID: 37453064 DOI: 10.1016/j.celrep.2023.112793] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 06/02/2023] [Accepted: 06/27/2023] [Indexed: 07/18/2023] Open
Abstract
Tissue-resident macrophages (TRMs) and dendritic cells (DCs) are highly heterogeneous and essential for immunity, tissue regeneration, and homeostasis maintenance. Here, we comprehensively profile the heterogeneity of TRMs and DCs across adult zebrafish organs via single-cell RNA sequencing. We identify two macrophage subsets: pro-inflammatory macrophages with potent phagocytosis signatures and pro-remodeling macrophages with tissue regeneration signatures in barrier tissues, liver, and heart. In parallel, one conventional dendritic cell (cDC) population with prominent antigen presentation capacity and plasmacytoid dendritic cells (pDCs) featured by anti-virus properties are also observed in these organs. Remarkably, in addition to a single macrophage/microglia population with potent phagocytosis capacity, a pDC population and two distinct cDC populations are identified in the brain. Finally, we generate specific reporter lines for in vivo tracking of macrophage and DC subsets. Our study depicts the landscape of TRMs and DCs and creates valuable tools for in-depth study of these cells in zebrafish.
Collapse
Affiliation(s)
- Qiuxia Zhou
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, the Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Changlong Zhao
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, the Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Zhiyong Yang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, the Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Rui Qu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, the Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Yunbo Li
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, the Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Yining Fan
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, the Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Jinlin Tang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, the Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Ting Xie
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, the Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Zilong Wen
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, the Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China; Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen 518055, China; Department of Immunology and Microbiology, School of Life Science, Southern University of Science and Technology, Shenzhen 518055, China.
| |
Collapse
|
6
|
Alakhras NS, Kaplan MH. Dendritic Cells as a Nexus for the Development of Multiple Sclerosis and Models of Disease. Adv Biol (Weinh) 2023:e2300073. [PMID: 37133870 DOI: 10.1002/adbi.202300073] [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: 02/12/2023] [Revised: 04/13/2023] [Indexed: 05/04/2023]
Abstract
Multiple sclerosis (MS) results from an autoimmune attack on the central nervous system (CNS). Dysregulated immune cells invade the CNS, causing demyelination, neuronal and axonal damage, and subsequent neurological disorders. Although antigen-specific T cells mediate the immunopathology of MS, innate myeloid cells have essential contributions to CNS tissue damage. Dendritic cells (DCs) are professional antigen-presenting cells (APCs) that promote inflammation and modulate adaptive immune responses. This review focuses on DCs as critical components of CNS inflammation. Here, evidence from studies is summarized with animal models of MS and MS patients that support the critical role of DCs in orchestrating CNS inflammation.
Collapse
Affiliation(s)
- Nada S Alakhras
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, 635 Barnhill Dr, Indianapolis, IN, 46202, USA
| | - Mark H Kaplan
- Department of Microbiology and Immunology, Indiana University School of Medicine, 635 Barnhill Dr, MS420, Indianapolis, IN, 46202, USA
| |
Collapse
|
7
|
Chen HY, Zhao Y, Xie YZ. Immunosenescence of brain accelerates Alzheimer's disease progression. Rev Neurosci 2023; 34:85-101. [PMID: 35791032 DOI: 10.1515/revneuro-2022-0021] [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: 02/25/2022] [Accepted: 06/04/2022] [Indexed: 01/07/2023]
Abstract
Most of Alzheimer's disease (AD) cases are sporadic and occur after age 65. With prolonged life expectancy and general population aging, AD is becoming a significant public health concern. The immune system supports brain development, plasticity, and homeostasis, yet it is particularly vulnerable to aging-related changes. Aging of the immune system, called immunosenescence, is the multifaceted remodeling of the immune system during aging. Immunosenescence is a contributing factor to various age-related diseases, including AD. Age-related changes in brain immune cell phenotype and function, crosstalk between immune cells and neural cells, and neuroinflammation work together to promote neurodegeneration and age-related cognitive impairment. Although numerous studies have confirmed the correlation between systemic immune changes and AD, few studies focus on the immune state of brain microenvironment in aging and AD. This review mainly addresses the changes of brain immune microenvironment in aging and AD. Specifically, we delineate how various aspects of the brain immune microenvironment, including immune gateways, immune cells, and molecules, and the interplay between immune cells and neural cells, accelerate AD pathogenesis during aging. We also propose a theoretical framework of therapeutic strategies selectively targeting the different mechanisms to restore brain immune homeostasis.
Collapse
Affiliation(s)
- Hou-Yu Chen
- Department of Abdominal Surgery, Affiliated Cancer Hospital, Institute of Guangzhou Medical University, Guangdong 510095, China
| | - Yan Zhao
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.,Institute of Aging and Age-related Disease Research, Central South University, Changsha, Hunan 410011, China
| | - Yong-Zhi Xie
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| |
Collapse
|
8
|
Saunders NR, Dziegielewska KM, Fame RM, Lehtinen MK, Liddelow SA. The choroid plexus: a missing link in our understanding of brain development and function. Physiol Rev 2023; 103:919-956. [PMID: 36173801 PMCID: PMC9678431 DOI: 10.1152/physrev.00060.2021] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 09/01/2022] [Accepted: 09/17/2022] [Indexed: 11/22/2022] Open
Abstract
Studies of the choroid plexus lag behind those of the more widely known blood-brain barrier, despite a much longer history. This review has two overall aims. The first is to outline long-standing areas of research where there are unanswered questions, such as control of cerebrospinal fluid (CSF) secretion and blood flow. The second aim is to review research over the past 10 years where the focus has shifted to the idea that there are choroid plexuses located in each of the brain's ventricles that make specific contributions to brain development and function through molecules they generate for delivery via the CSF. These factors appear to be particularly important for aspects of normal brain growth. Most research carried out during the twentieth century dealt with the choroid plexus, a brain barrier interface making critical contributions to the composition and stability of the brain's internal environment throughout life. More recent research in the twenty-first century has shown the importance of choroid plexus-generated CSF in neurogenesis, influence of sex and other hormones on choroid plexus function, and choroid plexus involvement in circadian rhythms and sleep. The advancement of technologies to facilitate delivery of brain-specific therapies via the CSF to treat neurological disorders is a rapidly growing area of research. Conversely, understanding the basic mechanisms and implications of how maternal drug exposure during pregnancy impacts the developing brain represents another key area of research.
Collapse
Affiliation(s)
- Norman R Saunders
- Department of Neuroscience, The Alfred Centre, Monash University, Melbourne, Victoria, Australia
| | | | - Ryann M Fame
- Department of Pathology, Boston Children's Hospital, Boston, Massachusetts
| | - Maria K Lehtinen
- Department of Pathology, Boston Children's Hospital, Boston, Massachusetts
| | - Shane A Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York, New York
- Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York, New York
- Department of Ophthalmology, NYU Grossman School of Medicine, New York, New York
- Parekh Center for Interdisciplinary Neurology, NYU Grossman School of Medicine, New York, New York
| |
Collapse
|
9
|
Thompson D, Brissette CA, Watt JA. The choroid plexus and its role in the pathogenesis of neurological infections. Fluids Barriers CNS 2022; 19:75. [PMID: 36088417 PMCID: PMC9463972 DOI: 10.1186/s12987-022-00372-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 08/27/2022] [Indexed: 11/10/2022] Open
Abstract
The choroid plexus is situated at an anatomically and functionally important interface within the ventricles of the brain, forming the blood-cerebrospinal fluid barrier that separates the periphery from the central nervous system. In contrast to the blood-brain barrier, the choroid plexus and its epithelial barrier have received considerably less attention. As the main producer of cerebrospinal fluid, the secretory functions of the epithelial cells aid in the maintenance of CNS homeostasis and are capable of relaying inflammatory signals to the brain. The choroid plexus acts as an immunological niche where several types of peripheral immune cells can be found within the stroma including dendritic cells, macrophages, and T cells. Including the epithelia cells, these cells perform immunosurveillance, detecting pathogens and changes in the cytokine milieu. As such, their activation leads to the release of homing molecules to induce chemotaxis of circulating immune cells, driving an immune response at the choroid plexus. Research into the barrier properties have shown how inflammation can alter the structural junctions and promote increased bidirectional transmigration of cells and pathogens. The goal of this review is to highlight our foundational knowledge of the choroid plexus and discuss how recent research has shifted our understanding towards viewing the choroid plexus as a highly dynamic and important contributor to the pathogenesis of neurological infections. With the emergence of several high-profile diseases, including ZIKA and SARS-CoV-2, this review provides a pertinent update on the cellular response of the choroid plexus to these diseases. Historically, pharmacological interventions of CNS disorders have proven difficult to develop, however, a greater focus on the role of the choroid plexus in driving these disorders would provide for novel targets and routes for therapeutics.
Collapse
Affiliation(s)
- Derick Thompson
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, USA
| | - Catherine A Brissette
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, USA
| | - John A Watt
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, USA.
| |
Collapse
|
10
|
Frederico B, Martins I, Chapela D, Gasparrini F, Chakravarty P, Ackels T, Piot C, Almeida B, Carvalho J, Ciccarelli A, Peddie CJ, Rogers N, Briscoe J, Guillemot F, Schaefer AT, Saúde L, Reis e Sousa C. DNGR-1-tracing marks an ependymal cell subset with damage-responsive neural stem cell potential. Dev Cell 2022; 57:1957-1975.e9. [PMID: 35998585 PMCID: PMC9616800 DOI: 10.1016/j.devcel.2022.07.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 05/16/2022] [Accepted: 07/20/2022] [Indexed: 01/19/2023]
Abstract
Cells with latent stem ability can contribute to mammalian tissue regeneration after damage. Whether the central nervous system (CNS) harbors such cells remains controversial. Here, we report that DNGR-1 lineage tracing in mice identifies an ependymal cell subset, wherein resides latent regenerative potential. We demonstrate that DNGR-1-lineage-traced ependymal cells arise early in embryogenesis (E11.5) and subsequently spread across the lining of cerebrospinal fluid (CSF)-filled compartments to form a contiguous sheet from the brain to the end of the spinal cord. In the steady state, these DNGR-1-traced cells are quiescent, committed to their ependymal cell fate, and do not contribute to neuronal or glial lineages. However, trans-differentiation can be induced in adult mice by CNS injury or in vitro by culture with suitable factors. Our findings highlight previously unappreciated ependymal cell heterogeneity and identify across the entire CNS an ependymal cell subset wherein resides damage-responsive neural stem cell potential.
Collapse
Affiliation(s)
- Bruno Frederico
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.
| | - Isaura Martins
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Diana Chapela
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal; TechnoPhage, SA, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| | - Francesca Gasparrini
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Probir Chakravarty
- Bioinformatics and Biostatistics, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Tobias Ackels
- Sensory Circuits and Neurotechnology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Cécile Piot
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Bruna Almeida
- Experimental Histopathology, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Joana Carvalho
- Experimental Histopathology, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Alessandro Ciccarelli
- Advanced Light Microscopy, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Christopher J Peddie
- Electron Microscopy, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Neil Rogers
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - James Briscoe
- Developmental Dynamic Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - François Guillemot
- Neural Stem Cell Biology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Andreas T Schaefer
- Sensory Circuits and Neurotechnology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK; Department of Neuroscience, Physiology &Pharmacology, University College London, London, UK
| | - Leonor Saúde
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular e Instituto de Histologia e Biologia do Desenvolvimento, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Caetano Reis e Sousa
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.
| |
Collapse
|
11
|
Piacente F, Bottero M, Benzi A, Vigo T, Uccelli A, Bruzzone S, Ferrara G. Neuroprotective Potential of Dendritic Cells and Sirtuins in Multiple Sclerosis. Int J Mol Sci 2022; 23:ijms23084352. [PMID: 35457169 PMCID: PMC9025744 DOI: 10.3390/ijms23084352] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/06/2022] [Accepted: 04/11/2022] [Indexed: 12/04/2022] Open
Abstract
Myeloid cells, including parenchymal microglia, perivascular and meningeal macrophages, and dendritic cells (DCs), are present in the central nervous system (CNS) and establish an intricate relationship with other cells, playing a crucial role both in health and in neurological diseases. In this context, DCs are critical to orchestrating the immune response linking the innate and adaptive immune systems. Under steady-state conditions, DCs patrol the CNS, sampling their local environment and acting as sentinels. During neuroinflammation, the resulting activation of DCs is a critical step that drives the inflammatory response or the resolution of inflammation with the participation of different cell types of the immune system (macrophages, mast cells, T and B lymphocytes), resident cells of the CNS and soluble factors. Although the importance of DCs is clearly recognized, their exact function in CNS disease is still debated. In this review, we will discuss modern concepts of DC biology in steady-state and during autoimmune neuroinflammation. Here, we will also address some key aspects involving DCs in CNS patrolling, highlighting the neuroprotective nature of DCs and emphasizing their therapeutic potential for the treatment of neurological conditions. Recently, inhibition of the NAD+-dependent deac(et)ylase sirtuin 6 was demonstrated to delay the onset of experimental autoimmune encephalomyelitis, by dampening DC trafficking towards inflamed LNs. Thus, a special focus will be dedicated to sirtuins’ role in DCs functions.
Collapse
Affiliation(s)
- Francesco Piacente
- Department of Experimental Medicine (DIMES), University of Genova, Viale Benedetto XV, 1, 16132 Genoa, Italy; (F.P.); (A.B.)
| | - Marta Bottero
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy; (M.B.); (T.V.); (A.U.); (G.F.)
| | - Andrea Benzi
- Department of Experimental Medicine (DIMES), University of Genova, Viale Benedetto XV, 1, 16132 Genoa, Italy; (F.P.); (A.B.)
| | - Tiziana Vigo
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy; (M.B.); (T.V.); (A.U.); (G.F.)
| | - Antonio Uccelli
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy; (M.B.); (T.V.); (A.U.); (G.F.)
| | - Santina Bruzzone
- Department of Experimental Medicine (DIMES), University of Genova, Viale Benedetto XV, 1, 16132 Genoa, Italy; (F.P.); (A.B.)
- Correspondence: ; Tel.: +39-(0)10-353-8150
| | - Giovanni Ferrara
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy; (M.B.); (T.V.); (A.U.); (G.F.)
| |
Collapse
|
12
|
Olate-Briones A, Escalona E, Salazar C, Herrada MJ, Liu C, Herrada AA, Escobedo N. The meningeal lymphatic vasculature in neuroinflammation. FASEB J 2022; 36:e22276. [PMID: 35344212 DOI: 10.1096/fj.202101574rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 03/08/2022] [Accepted: 03/14/2022] [Indexed: 12/13/2022]
Abstract
The lymphatic vasculature is a unidirectional network of lymphatic endothelial cells, whose main role is to maintain fluid homeostasis along with the absorption of dietary fat in the gastrointestinal organs and management and coordination of immune cell trafficking into lymph nodes during homeostasis and under inflammatory conditions. In homeostatic conditions, immune cells, such as dendritic cells, macrophages, or T cells can enter into the lymphatic vasculature and move easily through the lymph reaching secondary lymph nodes where immune cell activation or peripheral tolerance can be modulated. However, under inflammatory conditions such as pathogen infection, increased permeabilization of lymphatic vessels allows faster immune cell migration into inflamed tissues following a chemokine gradient, facilitating pathogen clearance and the resolution of inflammation. Interestingly, since the re-discovery of lymphatic vasculature in the central nervous system, known as the meningeal lymphatic vasculature, the role of these lymphatics as a key player in several neurological disorders has been described, with emphasis on the neurodegenerative process. Alternatively, less has been discussed about meningeal lymphatics and its role in neuroinflammation. In this review, we discuss current knowledge about the anatomy and function of the meningeal lymphatic vasculature and specifically analyze its contribution to different neuroinflammatory processes, highlighting the potential therapeutic target of meningeal lymphatic vasculature in these pathological conditions.
Collapse
Affiliation(s)
- Alexandra Olate-Briones
- Lymphatic Vasculature and Inflammation Research Laboratory, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Talca, Chile
| | - Emilia Escalona
- Lymphatic Vasculature and Inflammation Research Laboratory, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Talca, Chile
| | - Celia Salazar
- Lymphatic Vasculature and Inflammation Research Laboratory, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Talca, Chile
| | | | - Chaohong Liu
- Department of Microbiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Andrés A Herrada
- Lymphatic Vasculature and Inflammation Research Laboratory, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Talca, Chile
| | - Noelia Escobedo
- Lymphatic Vasculature and Inflammation Research Laboratory, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Talca, Chile
| |
Collapse
|
13
|
Wang S, van de Pavert SA. Innate Lymphoid Cells in the Central Nervous System. Front Immunol 2022; 13:837250. [PMID: 35185929 PMCID: PMC8852840 DOI: 10.3389/fimmu.2022.837250] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 01/17/2022] [Indexed: 12/19/2022] Open
Abstract
Immune cells are present within the central nervous system and play important roles in neurological inflammation and disease. As relatively new described immune cell population, Innate Lymphoid Cells are now increasingly recognized within the central nervous system and associated diseases. Innate Lymphoid Cells are generally regarded as tissue resident and early responders, while conversely within the central nervous system at steady-state their presence is limited. This review describes the current understandings on Innate Lymphoid Cells in the central nervous system at steady-state and its borders plus their involvement in major neurological diseases like ischemic stroke, Alzheimer's disease and Multiple Sclerosis.
Collapse
Affiliation(s)
| | - Serge A. van de Pavert
- Aix-Marseille Université, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre d’Immunologie de Marseille-Luminy (CIML), Marseille, France
| |
Collapse
|
14
|
Rickenbach C, Gericke C. Specificity of Adaptive Immune Responses in Central Nervous System Health, Aging and Diseases. Front Neurosci 2022; 15:806260. [PMID: 35126045 PMCID: PMC8812614 DOI: 10.3389/fnins.2021.806260] [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: 10/31/2021] [Accepted: 12/29/2021] [Indexed: 12/25/2022] Open
Abstract
The field of neuroimmunology endorses the involvement of the adaptive immune system in central nervous system (CNS) health, disease, and aging. While immune cell trafficking into the CNS is highly regulated, small numbers of antigen-experienced lymphocytes can still enter the cerebrospinal fluid (CSF)-filled compartments for regular immune surveillance under homeostatic conditions. Meningeal lymphatics facilitate drainage of brain-derived antigens from the CSF to deep cervical lymph nodes to prime potential adaptive immune responses. During aging and CNS disorders, brain barriers and meningeal lymphatic functions are impaired, and immune cell trafficking and antigen efflux are altered. In this context, alterations in the immune cell repertoire of blood and CSF and T and B cells primed against CNS-derived autoantigens have been observed in various CNS disorders. However, for many diseases, a causal relationship between observed immune responses and neuropathological findings is lacking. Here, we review recent discoveries about the association between the adaptive immune system and CNS disorders such as autoimmune neuroinflammatory and neurodegenerative diseases. We focus on the current challenges in identifying specific T cell epitopes in CNS diseases and discuss the potential implications for future diagnostic and treatment options.
Collapse
Affiliation(s)
- Chiara Rickenbach
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland
| | - Christoph Gericke
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland
| |
Collapse
|
15
|
Bala N, McGurk AI, Zilch T, Rup AN, Carter EM, Leddon SA, Fowell DJ. T cell activation niches-Optimizing T cell effector function in inflamed and infected tissues. Immunol Rev 2021; 306:164-180. [PMID: 34859453 PMCID: PMC9218983 DOI: 10.1111/imr.13047] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 11/06/2021] [Indexed: 12/29/2022]
Abstract
Successful immunity to infection, malignancy, and tissue damage requires the coordinated recruitment of numerous immune cell subsets to target tissues. Once within the target tissue, effector T cells rely on local chemotactic cues and structural cues from the tissue matrix to navigate the tissue, interact with antigen-presenting cells, and release effector cytokines. This highly dynamic process has been "caught on camera" in situ by intravital multiphoton imaging. Initial studies revealed a surprising randomness to the pattern of T cell migration through inflamed tissues, behavior thought to facilitate chance encounters with rare antigen-bearing cells. Subsequent tissue-wide visualization has uncovered a high degree of spatial preference when it comes to T cell activation. Here, we discuss the basic tenants of a successful effector T cell activation niche, taking cues from the dynamics of Tfh positioning in the lymph node germinal center. In peripheral tissues, steady-state microanatomical organization may direct the location of "pop-up" de novo activation niches, often observed as perivascular clusters, that support early effector T cell activation. These perivascular activation niches appear to be regulated by site-specific chemokines that coordinate the recruitment of dendritic cells and other innate cells for local T cell activation, survival, and optimized effector function.
Collapse
Affiliation(s)
- Noor Bala
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Alexander I McGurk
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Tiago Zilch
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Anastasia N Rup
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Evan M Carter
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Scott A Leddon
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Deborah J Fowell
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| |
Collapse
|
16
|
Immune cell compartmentalization for brain surveillance and protection. Nat Immunol 2021; 22:1083-1092. [PMID: 34429552 DOI: 10.1038/s41590-021-00994-2] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 07/08/2021] [Indexed: 02/07/2023]
Abstract
For decades, it was commonly accepted that the brain is secluded from peripheral immune activity and is self-sufficient for its maintenance and repair. This simplistic perception was based on the presence of resident immune cells, the microglia, and barrier systems within the brain, and the assumption that the central nervous system (CNS) lacks lymphatic drainage. This view was revised with the discoveries that higher functions of the CNS, homeostasis and repair are supported by peripheral innate and adaptive immune cells. The findings of bone marrow-derived immune cells in specialized niches, and the renewed observation that a lymphatic drainage system exists within the brain, further contributed to this revised model. In this Review, we describe the immune niches within the brain, the contribution of professional immune cells to brain functions, the bidirectional relationships between the CNS and the immune system and the relevance of immune components to brain aging and neurodegenerative diseases.
Collapse
|
17
|
Fernández A, Quintana E, Velasco P, Moreno-Jimenez B, de Andrés B, Gaspar ML, Liste I, Vilar M, Mira H, Cano E. Senescent accelerated prone 8 (SAMP8) mice as a model of age dependent neuroinflammation. J Neuroinflammation 2021; 18:75. [PMID: 33736657 PMCID: PMC7977588 DOI: 10.1186/s12974-021-02104-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 02/03/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Aging and age-related diseases are strong risk factors for the development of neurodegenerative diseases. Neuroinflammation (NIF), as the brain's immune response, plays an important role in aged associated degeneration of central nervous system (CNS). There is a need for well characterized animal models that will allow the scientific community to understand and modulate this process. METHODS We have analyzed aging-phenotypical and inflammatory changes of brain myeloid cells (bMyC) in a senescent accelerated prone aged (SAMP8) mouse model, and compared with their senescence resistant control mice (SAMR1). We have performed morphometric methods to evaluate the architecture of cellular prolongations and determined the appearance of Iba1+ clustered cells with aging. To analyze specific constant brain areas, we have performed stereology measurements of Iba1+ cells in the hippocampal formation. We have isolated bMyC from brain parenchyma (BP) and choroid plexus plus meningeal membranes (m/Ch), and analyzed their response to systemic lipopolysaccharide (LPS)-driven inflammation. RESULTS Aged 10 months old SAMP8 mice present many of the hallmarks of aging-dependent neuroinflammation when compared with their SAMR1 control, i.e., increase of protein aggregates, presence of Iba1+ clusters, but not an increase in the number of Iba1+ cells. We have further observed an increase of main inflammatory mediator IL-1β, and an augment of border MHCII+Iba1+ cells. Isolated CD45+ bMyC from brain parenchyma (BP) and choroid plexus plus meningeal membranes (m/Ch) have been analyzed, showing that there is not a significant increase of CD45+ cells from the periphery. Our data support that aged-driven pro-inflammatory cytokine interleukin 1 beta (IL-1β) transcription is enhanced in CD45+BP cells. Furthermore, LPS-driven systemic inflammation produces inflammatory cytokines mainly in border bMyC, sensed to a lesser extent by the BP bMyC, showing that IL-1β expression is further augmented in aged SAMP8 compared to control SAMR1. CONCLUSION Our data validate the SAMP8 model to study age-associated neuroinflammatory events, but careful controls for age and strain are required. These animals show morphological changes in their bMyC cell repertoires associated to age, corresponding to an increase in the production of pro-inflammatory cytokines such as IL-1β, which predispose the brain to an enhanced inflammatory response after LPS-systemic challenge.
Collapse
Affiliation(s)
- Andrés Fernández
- Chronic Disease Programme, Neuroinflammation Unit, Instituto de Salud Carlos III, Carretera Majadahonda-Pozuelo, Km.2,2, Majadahonda, 28220, Madrid, Spain
| | - Elena Quintana
- Chronic Disease Programme, Neuroinflammation Unit, Instituto de Salud Carlos III, Carretera Majadahonda-Pozuelo, Km.2,2, Majadahonda, 28220, Madrid, Spain
| | - Patricia Velasco
- Chronic Disease Programme, Neuroinflammation Unit, Instituto de Salud Carlos III, Carretera Majadahonda-Pozuelo, Km.2,2, Majadahonda, 28220, Madrid, Spain
| | - Belén Moreno-Jimenez
- Chronic Disease Programme, Neuroinflammation Unit, Instituto de Salud Carlos III, Carretera Majadahonda-Pozuelo, Km.2,2, Majadahonda, 28220, Madrid, Spain
| | - Belén de Andrés
- Unidad de Inmunobiología, Instituto de Salud Carlos II, Madrid, Spain
| | | | - Isabel Liste
- Chronic Disease Programme, Neuroinflammation Unit, Instituto de Salud Carlos III, Carretera Majadahonda-Pozuelo, Km.2,2, Majadahonda, 28220, Madrid, Spain
| | - Marçal Vilar
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Helena Mira
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Eva Cano
- Chronic Disease Programme, Neuroinflammation Unit, Instituto de Salud Carlos III, Carretera Majadahonda-Pozuelo, Km.2,2, Majadahonda, 28220, Madrid, Spain.
| |
Collapse
|
18
|
Vincenti I, Merkler D. New advances in immune components mediating viral control in the CNS. Curr Opin Virol 2021; 47:68-78. [PMID: 33636592 DOI: 10.1016/j.coviro.2021.02.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/01/2021] [Accepted: 02/05/2021] [Indexed: 11/26/2022]
Abstract
Protective immune responses in the central nervous system (CNS) must act efficiently but need to be tightly controlled to avoid excessive damage to this vital organ. Under homeostatic conditions, the immune surveillance of the CNS is mediated by innate immune cells together with subsets of memory lymphocytes accumulating over lifetime. Accordingly, a wide range of immune responses can be triggered upon pathogen infection that can be associated with devastating clinical outcomes, and which most frequently are due to neurotropic viruses. Here, we discuss recent advances about our understanding of anti-viral immune responses with special emphasis on mechanisms operating in the various anatomical compartments of the CNS.
Collapse
Affiliation(s)
- Ilena Vincenti
- University of Geneva, Department of Pathology and Immunology, Geneva, Switzerland
| | - Doron Merkler
- University of Geneva, Department of Pathology and Immunology, Geneva, Switzerland; Division of Clinical Pathology, Geneva University Hospital, 1211 Geneva, Switzerland.
| |
Collapse
|
19
|
Autophagy Pathways in CNS Myeloid Cell Immune Functions. Trends Neurosci 2020; 43:1024-1033. [DOI: 10.1016/j.tins.2020.09.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/17/2020] [Accepted: 09/04/2020] [Indexed: 02/06/2023]
|
20
|
Gallizioli M, Miró-Mur F, Otxoa-de-Amezaga A, Cugota R, Salas-Perdomo A, Justicia C, Brait VH, Ruiz-Jaén F, Arbaizar-Rovirosa M, Pedragosa J, Bonfill-Teixidor E, Gelderblom M, Magnus T, Cano E, Del Fresno C, Sancho D, Planas AM. Dendritic Cells and Microglia Have Non-redundant Functions in the Inflamed Brain with Protective Effects of Type 1 cDCs. Cell Rep 2020; 33:108291. [PMID: 33086061 PMCID: PMC7578563 DOI: 10.1016/j.celrep.2020.108291] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 09/02/2020] [Accepted: 09/29/2020] [Indexed: 01/14/2023] Open
Abstract
Brain CD11c+ cells share features with microglia and dendritic cells (DCs). Sterile inflammation increases brain CD11c+ cells, but their phenotype, origin, and functions remain largely unknown. We report that, after cerebral ischemia, microglia attract DCs to the inflamed brain, and astroglia produce Flt3 ligand, supporting development and expansion of CD11c+ cells. CD11c+ cells in the inflamed brain are a complex population derived from proliferating microglia and infiltrating DCs, including a major subset of OX40L+ conventional cDC2, and also cDC1, plasmacytoid, and monocyte-derived DCs. Despite sharing certain morphological features and markers, CD11c+ microglia and DCs display differential expression of pattern recognition receptors and chemokine receptors. DCs excel CD11c- and CD11c+ microglia in the capacity to present antigen through MHCI and MHCII. Of note, cDC1s protect from brain injury after ischemia. We thus reveal aspects of the dynamics and functions of brain DCs in the regulation of inflammation and immunity.
Collapse
Affiliation(s)
- Mattia Gallizioli
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona 08036, Spain; Area of Neurosciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona 08036, Spain
| | - Francesc Miró-Mur
- Area of Neurosciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona 08036, Spain; Fundació Clínic, Barcelona 08036, Spain
| | - Amaia Otxoa-de-Amezaga
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona 08036, Spain; Area of Neurosciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona 08036, Spain
| | - Roger Cugota
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona 08036, Spain
| | - Angélica Salas-Perdomo
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona 08036, Spain; Fundació Clínic, Barcelona 08036, Spain
| | - Carles Justicia
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona 08036, Spain; Area of Neurosciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona 08036, Spain
| | - Vanessa H Brait
- Area of Neurosciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona 08036, Spain
| | - Francisca Ruiz-Jaén
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona 08036, Spain; Area of Neurosciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona 08036, Spain
| | - Maria Arbaizar-Rovirosa
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona 08036, Spain; Area of Neurosciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona 08036, Spain
| | - Jordi Pedragosa
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona 08036, Spain; Area of Neurosciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona 08036, Spain
| | - Ester Bonfill-Teixidor
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona 08036, Spain
| | - Mathias Gelderblom
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg 20251, Germany
| | - Tim Magnus
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg 20251, Germany
| | - Eva Cano
- Neuroinflammation Unit, Unidad Funcional de Investigación de Enfermedades Crónicas, Instituto de Salud Carlos III, Majadahonda, Madrid 28222, Spain
| | - Carlos Del Fresno
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain
| | - David Sancho
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain
| | - Anna M Planas
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona 08036, Spain; Area of Neurosciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona 08036, Spain.
| |
Collapse
|
21
|
Thompson D, Sorenson J, Greenmyer J, Brissette CA, Watt JA. The Lyme disease bacterium, Borrelia burgdorferi, stimulates an inflammatory response in human choroid plexus epithelial cells. PLoS One 2020; 15:e0234993. [PMID: 32645014 PMCID: PMC7347220 DOI: 10.1371/journal.pone.0234993] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 06/05/2020] [Indexed: 11/19/2022] Open
Abstract
The main functions of the choroid plexus (CP) are the production of cerebral spinal fluid (CSF), the formation of the blood-CSF barrier, and regulation of immune response. This barrier allows for the exchange of specific nutrients, waste, and peripheral immune cells between the blood stream and CSF. Borrelia burgdorferi (Bb), the causative bacteria of Lyme disease, is associated with neurological complications including meningitis-indeed, Bb has been isolated from the CSF of patients. While it is accepted that B. burgdorferi can enter the central nervous system (CNS) of patients, it is unknown how the bacteria crosses this barrier and how the pathogenesis of the disease leads to the observed symptoms in patients. We hypothesize that during infection Borrelia burgdorferi will induce an immune response conducive to the chemotaxis of immune cells and subsequently lead to a pro-inflammatory state with the CNS parenchyma. Primary human choroid plexus epithelial cells were grown in culture and infected with B. burgdorferi strain B31 MI-16 for 48 hours. RNA was isolated and used for RNA sequencing and RT-qPCR validation. Secreted proteins in the supernatant were analyzed via ELISA. Transcriptome analysis based on RNA sequencing determined a total of 160 upregulated genes and 98 downregulated genes. Pathway and biological process analysis determined a significant upregulation in immune and inflammatory genes specifically in chemokine and interferon related pathways. Further analysis revealed downregulation in genes related to cell to cell junctions including tight and adherens junctions. These results were validated via RT-qPCR. Protein analysis of secreted factors showed an increase in inflammatory chemokines, corresponding to our transcriptome analysis. These data further demonstrate the role of the CP in the modulation of the immune response in a disease state and give insight into the mechanisms by which Borrelia burgdorferi may disseminate into, and act upon, the CNS. Future experiments aim to detail the impact of B. burgdorferi on the blood-CSF-barrier (BCSFB) integrity and inflammatory response within animal models.
Collapse
Affiliation(s)
- Derick Thompson
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| | - Jordyn Sorenson
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| | - Jacob Greenmyer
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| | - Catherine A. Brissette
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| | - John A. Watt
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| |
Collapse
|
22
|
Miró-Mur F, Urra X, Ruiz-Jaén F, Pedragosa J, Chamorro Á, Planas AM. Antigen-Dependent T Cell Response to Neural Peptides After Human Ischemic Stroke. Front Cell Neurosci 2020; 14:206. [PMID: 32719588 PMCID: PMC7348665 DOI: 10.3389/fncel.2020.00206] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 06/11/2020] [Indexed: 12/20/2022] Open
Abstract
Ischemic stroke causes brain tissue damage and may release central nervous system (CNS)-specific peptides to the periphery. Neural antigen presentation in the lymphoid tissue could prime immune cells and result in adaptive immune response. However, autoimmune responses against neural antigens are not commonly uncovered after stroke. We studied the brain tissue of nine fatal stroke cases and the blood of a cohort of 13 patients and 11 controls. Flow cytometry carried out in three of the brain samples showed CD8 and CD4 T cells in the cerebrospinal fluid (CSF) of the ventricles in the patient deceased 1 day poststroke, T cells with an activated phenotype in the CSF of the patient that died at day 6, and T cells in the ischemic brain tissue in the patient deceased 140 days after stroke onset. Immunohistochemistry showed higher T cell numbers in the core of the lesion of the patient deceased 18 days post-stroke than in the patients deceased from 1 to 5 days post-stroke. In blood samples, we studied whether lymphocytes were primed in the periphery against neural antigens at sequential times (on admission, day 5, and day 90) after stroke. T lymphocytes of stroke patients produced IFN-γ and TNF-α and responded to MBP peptides by increasing their production of TNF-α and IL-10 at admission, but not at later time points. In contrast, IL-4 producing T cells showed progressive increases. Higher percentages of TNF-α producing T lymphocytes at admission were independently associated with poorer outcomes at 90 days. However, we did not detect T cell responses to neural-antigen stimulation 90 days post-stroke. Altogether the results suggest acute T cell priming in the periphery in acute stroke, T cell trafficking from the CSF to the ischemic brain tissue, and the existence of active mechanisms preventing autoreactivity.
Collapse
Affiliation(s)
- Francesc Miró-Mur
- Functional Unit of Cerebrovascular Diseases, Hospital Clinic, Barcelona, Spain
| | - Xabier Urra
- Functional Unit of Cerebrovascular Diseases, Hospital Clinic, Barcelona, Spain.,Area of Neuroscience, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Francisca Ruiz-Jaén
- Area of Neuroscience, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Jordi Pedragosa
- Area of Neuroscience, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain.,Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain
| | - Ángel Chamorro
- Functional Unit of Cerebrovascular Diseases, Hospital Clinic, Barcelona, Spain.,Area of Neuroscience, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Anna M Planas
- Area of Neuroscience, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain.,Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain
| |
Collapse
|
23
|
Petersen N, Torz L, Jensen KHR, Hjortø GM, Spiess K, Rosenkilde MM. Three-Dimensional Explant Platform for Studies on Choroid Plexus Epithelium. Front Cell Neurosci 2020; 14:108. [PMID: 32431599 PMCID: PMC7214744 DOI: 10.3389/fncel.2020.00108] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 04/08/2020] [Indexed: 12/12/2022] Open
Abstract
The choroid plexus (CP) plays a major role in controlling the entry of substances and immune cells into the brain as it forms the blood-cerebrospinal fluid barrier (BCSFB) in the brain ventricles. Dysregulated immune cell trafficking through the epithelial cell (EC) layer of CP is central for the pathogenesis of infectious diseases in the brain and many neurodegenerative disorders. In vitro studies elucidating the function of the CP have so far been limited to the monolayer culture of CP ECs. To mimic immune cell migration across the CP barrier, a three-dimensional model would be advantageous. Here, we present an in vitro platform for studies of the immune cell trafficking based on CP explants/organoids. The explants were generated from fragments of mouse CPs in Matrigel, where the cells formed luminal spaces and could be maintained in culture for at least 8 weeks. We demonstrate expression of the major CP markers in the explants, including transthyretin and aquaporin 1 as well as ZO1 and ICAM-1, indicating a capacity for secretion of cerebrospinal fluid (CSF) and presence of tight junctions. CP explants displayed CP-like cell polarization and formed an intact EC barrier. We also show that the expression of transthyretin, transferrin, occludin and other genes associated with various functions of CP was maintained in the explants at similar levels as in native CP. By using dendritic cells and neutrophils, we show that the migration activity of immune cells and their interactions with CP epithelium can be monitored by microscopy. Thereby, the three-dimensional CP explant model can be used to study the cellular and molecular mechanisms mediating immune cell migration through CP epithelium and other functions of choroid EC. We propose this platform can potentially be used in the search for therapeutic targets and intervention strategies to improve control of (drug) substances and (immune) cell entry into the central nervous system.
Collapse
Affiliation(s)
- Natalia Petersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lola Torz
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kristian H Reveles Jensen
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Gertrud Malene Hjortø
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Katja Spiess
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mette Marie Rosenkilde
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
24
|
Immune cell regulation of glia during CNS injury and disease. Nat Rev Neurosci 2020; 21:139-152. [DOI: 10.1038/s41583-020-0263-9] [Citation(s) in RCA: 146] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2020] [Indexed: 12/13/2022]
|
25
|
Dixon GA, Pérez CA. Multiple Sclerosis and the Choroid Plexus: Emerging Concepts of Disease Immunopathophysiology. Pediatr Neurol 2020; 103:65-75. [PMID: 31780202 DOI: 10.1016/j.pediatrneurol.2019.08.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 08/11/2019] [Accepted: 08/15/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND The coexistence of multiple sclerosis and intracranial neoplasms is very rare, and whether this occurrence can be explained by a causal relationship or by coincidence remains a matter of debate. Possible roles of the choroid plexus as a site of tumor cell invasion and lymphocyte infiltration into the central nervous system have been hypothesized in recent studies. METHODS We describe a 13-year-old boy with concurrent multiple sclerosis and choroid plexus papilloma, then review the published literature with a focus on the pathophysiologic mechanisms of neuroinflammation in multiple sclerosis and the potential role of the choroid plexus in this process. RESULTS A growing body of evidence suggests that both physical and functional dysregulation of the choroid plexus may be a common mechanism underlying the pathophysiology of central nervous system inflammation. CONCLUSIONS In multiple sclerosis, the choroid plexus could act as a gateway for lymphocyte entry from the peripheral blood into the central nervous system at its earlier stages. However, future studies are needed to identify whether structural alterations of the choroid plexus play a role in the pathophysiology of multiple sclerosis and to provide suitable models to determine their consequences.
Collapse
Affiliation(s)
- Grant A Dixon
- McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas
| | - Carlos A Pérez
- Division of Multiple Sclerosis and Neuroimmunology, Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas.
| |
Collapse
|
26
|
Sie C, Perez LG, Kreutzfeldt M, Potthast M, Ohnmacht C, Merkler D, Huber S, Krug A, Korn T. Dendritic Cell Accumulation in the Gut and Central Nervous System Is Differentially Dependent on α4 Integrins. THE JOURNAL OF IMMUNOLOGY 2019; 203:1417-1427. [PMID: 31399516 DOI: 10.4049/jimmunol.1900468] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 07/13/2019] [Indexed: 12/11/2022]
Abstract
Homing of pathogenic CD4+ T cells to the CNS is dependent on α4 integrins. However, it is uncertain whether α4 integrins are also required for the migration of dendritic cell (DC) subsets, which sample Ags from nonlymphoid tissues to present it to T cells. In this study, after genetic ablation of Itga4 in DCs and monocytes in mice via the promoters of Cd11c and Lyz2 (also known as LysM), respectively, the recruitment of α4 integrin-deficient conventional and plasmacytoid DCs to the CNS was unaffected, whereas α4 integrin-deficient, monocyte-derived DCs accumulated less efficiently in the CNS during experimental autoimmune encephalomyelitis in a competitive setting than their wild-type counterparts. In a noncompetitive setting, α4 integrin deficiency on monocyte-derived DCs was fully compensated. In contrast, in small intestine and colon, the fraction of α4 integrin-deficient CD11b+CD103+ DCs was selectively reduced in steady-state. Yet, T cell-mediated inflammation and host defense against Citrobacter rodentium were not impaired in the absence of α4 integrins on DCs. Thus, inflammatory conditions can promote an environment that is indifferent to α4 integrin expression by DCs.
Collapse
Affiliation(s)
- Christopher Sie
- Abteilung für Experimentelle Neuroimmunologie, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany.,Klinik für Neurologie, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany
| | - Laura Garcia Perez
- I. Medizinische Klinik und Poliklinik, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Mario Kreutzfeldt
- Division of Clinical Pathology, Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland
| | - Maria Potthast
- Center of Allergy and Environment, Helmholtz Center and Technical University of Munich, 80802 Munich, Germany
| | - Caspar Ohnmacht
- Center of Allergy and Environment, Helmholtz Center and Technical University of Munich, 80802 Munich, Germany
| | - Doron Merkler
- Division of Clinical Pathology, Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland
| | - Samuel Huber
- I. Medizinische Klinik und Poliklinik, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Anne Krug
- Institute for Immunology, Biomedical Center, Ludwig Maximilians University of Munich, 82152 Planegg-Martinsried, Germany; and
| | - Thomas Korn
- Abteilung für Experimentelle Neuroimmunologie, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany; .,Klinik für Neurologie, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany.,Munich Cluster for Systems Neurology, SyNergy, 81377 Munich, Germany
| |
Collapse
|
27
|
De Laere M, Berneman ZN, Cools N. To the Brain and Back: Migratory Paths of Dendritic Cells in Multiple Sclerosis. J Neuropathol Exp Neurol 2019; 77:178-192. [PMID: 29342287 PMCID: PMC5901086 DOI: 10.1093/jnen/nlx114] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Migration of dendritic cells (DC) to the central nervous system (CNS) is a critical event in the pathogenesis of multiple sclerosis (MS). While up until now, research has mainly focused on the transmigration of DC through the blood-brain barrier, experimental evidence points out that also the choroid plexus and meningeal vessels represent important gateways to the CNS, especially in early disease stages. On the other hand, DC can exit the CNS to maintain immunological tolerance to patterns expressed in the CNS, a process that is perturbed in MS. Targeting trafficking of immune cells, including DC, to the CNS has demonstrated to be a successful strategy to treat MS. However, this approach is known to compromise protective immune surveillance of the brain. Unravelling the migratory paths of regulatory and pathogenic DC within the CNS may ultimately lead to the design of new therapeutic strategies able to selectively interfere with the recruitment of pathogenic DC to the CNS, while leaving host protective mechanisms intact.
Collapse
Affiliation(s)
- Maxime De Laere
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp
| | - Zwi N Berneman
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp.,Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital (UZA), Edegem, Belgium
| | - Nathalie Cools
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp
| |
Collapse
|
28
|
Yan J, Zhao Q, Gabrusiewicz K, Kong LY, Xia X, Wang J, Ott M, Xu J, Davis RE, Huo L, Rao G, Sun SC, Watowich SS, Heimberger AB, Li S. FGL2 promotes tumor progression in the CNS by suppressing CD103 + dendritic cell differentiation. Nat Commun 2019; 10:448. [PMID: 30683885 PMCID: PMC6347641 DOI: 10.1038/s41467-018-08271-x] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 12/19/2018] [Indexed: 12/20/2022] Open
Abstract
Few studies implicate immunoregulatory gene expression in tumor cells in arbitrating brain tumor progression. Here we show that fibrinogen-like protein 2 (FGL2) is highly expressed in glioma stem cells and primary glioblastoma (GBM) cells. FGL2 knockout in tumor cells did not affect tumor-cell proliferation in vitro or tumor progression in immunodeficient mice but completely impaired GBM progression in immune-competent mice. This impairment was reversed in mice with a defect in dendritic cells (DCs) or CD103+ DC differentiation in the brain and in tumor-draining lymph nodes. The presence of FGL2 in tumor cells inhibited granulocyte-macrophage colony-stimulating factor (GM-CSF)-induced CD103+ DC differentiation by suppressing NF-κB, STAT1/5, and p38 activation. These findings are relevant to GBM patients because a low level of FGL2 expression with concurrent high GM-CSF expression is associated with higher CD8B expression and longer survival. These data provide a rationale for therapeutic inhibition of FGL2 in brain tumors.
Collapse
Affiliation(s)
- Jun Yan
- Center for Brain Disorders Research, Capital Medical University, Beijing, 100069, China
- Beijing Institute for Brain Disorders, Beijing, 100069, China
- Department of Pediatrics-Research, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Qingnan Zhao
- Department of Pediatrics-Research, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Konrad Gabrusiewicz
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Ling-Yuan Kong
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Xueqing Xia
- Department of Pediatrics-Research, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jian Wang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Martina Ott
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jingda Xu
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - R Eric Davis
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Longfei Huo
- Department of Pediatrics-Research, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Ganesh Rao
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Shao-Cong Sun
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Stephanie S Watowich
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Amy B Heimberger
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Shulin Li
- Department of Pediatrics-Research, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
29
|
Antigen-presenting cell diversity for T cell reactivation in central nervous system autoimmunity. J Mol Med (Berl) 2018; 96:1279-1292. [PMID: 30386908 DOI: 10.1007/s00109-018-1709-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 10/19/2018] [Accepted: 10/24/2018] [Indexed: 12/13/2022]
Abstract
Autoreactive T cells are considered the major culprits in the pathogenesis of many autoimmune diseases like multiple sclerosis (MS). Upon activation in the lymphoid organs, autoreactive T cells migrate towards the central nervous system (CNS) and target the myelin sheath-forming oligodendrocytes, resulting in detrimental neurological symptoms. Despite the availability of extensively studied systems like the experimental autoimmune encephalomyelitis (EAE) model, our understanding of this disease and the underlying pathogenesis is still elusive. One vividly discussed subject represents the T cell reactivation in the CNS. In order to exert their effector functions in the CNS, autoreactive T cells must encounter antigen-presenting cells (APCs). This interaction provides an antigen-restricted stimulus in the context of major histocompatibility complex class II (MHC-II) and other co-stimulatory molecules. Peripherally derived dendritic cells (DCs), B cells, border-associated macrophages (BAM), CNS-resident microglia, and astrocytes have the capacity to express molecules required for antigen presentation under inflammatory conditions. Also, endothelial cells can fulfill these prerequisites in certain situations. Which of these cells in fact act as APCs for T cell reactivation and to which extent they can exert this function has been studied intensively, but unfortunately with no firm conclusion. In this review, we will summarize the findings that support or question the antigen presenting capacities of the mentioned cell types of CNS-localized T cell reactivation.
Collapse
|
30
|
Giles DA, Duncker PC, Wilkinson NM, Washnock-Schmid JM, Segal BM. CNS-resident classical DCs play a critical role in CNS autoimmune disease. J Clin Invest 2018; 128:5322-5334. [PMID: 30226829 DOI: 10.1172/jci123708] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 09/11/2018] [Indexed: 12/19/2022] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is an inflammatory demyelinating disease of the central nervous system (CNS), induced by the adoptive transfer of myelin-reactive CD4+ T cells into naive syngeneic mice. It is widely used as a rodent model of multiple sclerosis (MS). The development of EAE lesions is initiated when transferred CD4+ T cells access the CNS and are reactivated by local antigen-presenting cells (APCs) bearing endogenous myelin peptide/MHC class II complexes. The identity of the CNS-resident, lesion-initiating APCs is widely debated. Here we demonstrate that classical dendritic cells (cDCs) normally reside in the meninges, brain, and spinal cord in the steady state. These cells are unique among candidate CNS APCs in their ability to stimulate naive, as well as effector, myelin-specific T cells to proliferate and produce proinflammatory cytokines directly ex vivo. cDCs expanded in the meninges and CNS parenchyma in association with disease progression. Selective depletion of cDCs led to a decrease in the number of myelin-primed donor T cells in the CNS and reduced the incidence of clinical EAE by half. Based on our findings, we propose that cDCs, and the factors that regulate them, be further investigated as potential therapeutic targets in MS.
Collapse
Affiliation(s)
- David A Giles
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology.,Graduate Program in Immunology, and.,Medical Scientist Training Program, University of Michigan, Ann Arbor, Michigan, USA
| | - Patrick C Duncker
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology.,Graduate Program in Immunology, and
| | | | | | - Benjamin M Segal
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology.,Graduate Program in Immunology, and.,Neurology Service, VA Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
| |
Collapse
|
31
|
Strominger I, Elyahu Y, Berner O, Reckhow J, Mittal K, Nemirovsky A, Monsonego A. The Choroid Plexus Functions as a Niche for T-Cell Stimulation Within the Central Nervous System. Front Immunol 2018; 9:1066. [PMID: 29868025 PMCID: PMC5962702 DOI: 10.3389/fimmu.2018.01066] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 04/27/2018] [Indexed: 12/20/2022] Open
Abstract
The choroid plexus (CP) compartment in the ventricles of the brain comprises fenestrated vasculature and, therefore, it is permeable to blood-borne mediators of inflammation. Here, we explored whether T-cell activation in the CP plays a role in regulating central nervous system (CNS) inflammation. We show that CD4 T cells injected into the lateral ventricles adhere to the CP, transmigrate across its epithelium, and undergo antigen-specific activation and proliferation. This process is enhanced following peripheral immune stimulation and significantly impacts the immune signaling induced by the CP. Ex vivo studies demonstrate that T-cell harboring the CP through its apical surface is a chemokine- and adhesion molecule-dependent process. We suggest that, within the CNS, the CP serves an immunological niche, which rapidly responds to peripheral inflammation and, thereby, promotes two-way T-cell trafficking that impact adaptive immunity in the CNS.
Collapse
Affiliation(s)
- Itai Strominger
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.,Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva, Israel.,The National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Yehezqel Elyahu
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.,Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva, Israel.,The National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Omer Berner
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.,Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva, Israel.,The National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Jensen Reckhow
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.,Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva, Israel.,The National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Kritika Mittal
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.,Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva, Israel.,The National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Anna Nemirovsky
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.,Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva, Israel.,The National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Alon Monsonego
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.,Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva, Israel.,The National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| |
Collapse
|
32
|
Yao WR, Li D, Yu L, Wang FJ, Xing H, Yang GB. The levels of DNGR-1 and its ligand-bearing cells were altered after human and simian immunodeficiency virus infection. Immunol Res 2018; 65:869-879. [PMID: 28478499 DOI: 10.1007/s12026-017-8925-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Dendritic cell NK lectin Group Receptor-1 (DNGR-1), also known as C-type lectin domain family 9, member A (CLEC9A), is a member of C-type lectin receptor superfamily expressed primarily on dendritic cells (DC) that excel in cross-presentation of exogenous antigens. To find out whether and how it is affected in human immunodeficiency virus infections or acquired immunodeficiency syndromes (HIV/AIDS), DNGR-1 expression and DNGR-1-binding cells in simian/human immunodeficiency virus (SHIV) and simian immunodeficiency virus (SIV)-infected rhesus macaques and antiretroviral therapy (ART)-treated AIDS patients were examined by real-time RT-PCR, flow cytometry, and confocal microscopy. DNGR-1 expression was observed in both lymphoid and non-lymphoid tissues including gut-associated lymphoid tissues (GALT) of rhesus macaques. DNGR-1 mRNA levels were significantly reduced in the blood while significantly elevated in the GALT of SHIV/SIV-infected rhesus macaques. DNGR-1 transcription levels were also significantly reduced in the blood of ART-treated AIDS patients irrespective of viral status. White blood cells with exposed DNGR-1 ligands were significantly increased in ART-treated AIDS patients, while significantly decreased in SIV-infected rhesus macaques. These data indicate that DNGR-1 expression, and by extension, the function of cross-presentation of antigens associated with dead/damaged cells might be compromised in HIV/SIV infection, which might play a role in HIV/AIDS pathogenesis and should be taken into consideration in therapeutic AIDS vaccine development.
Collapse
Affiliation(s)
- Wen-Rong Yao
- National Center for AIDS/STD Control and Prevention, China CDC, 155 Changbai Road, Changping District, Beijing, 102206, People's Republic of China
| | - Dong Li
- National Center for AIDS/STD Control and Prevention, China CDC, 155 Changbai Road, Changping District, Beijing, 102206, People's Republic of China
| | - Lei Yu
- National Center for AIDS/STD Control and Prevention, China CDC, 155 Changbai Road, Changping District, Beijing, 102206, People's Republic of China
| | - Feng-Jie Wang
- National Center for AIDS/STD Control and Prevention, China CDC, 155 Changbai Road, Changping District, Beijing, 102206, People's Republic of China
| | - Hui Xing
- National Center for AIDS/STD Control and Prevention, China CDC, 155 Changbai Road, Changping District, Beijing, 102206, People's Republic of China
| | - Gui-Bo Yang
- National Center for AIDS/STD Control and Prevention, China CDC, 155 Changbai Road, Changping District, Beijing, 102206, People's Republic of China.
| |
Collapse
|
33
|
Herz J, Filiano AJ, Wiltbank AT, Yogev N, Kipnis J. Myeloid Cells in the Central Nervous System. Immunity 2017; 46:943-956. [PMID: 28636961 PMCID: PMC5657250 DOI: 10.1016/j.immuni.2017.06.007] [Citation(s) in RCA: 240] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 05/17/2017] [Accepted: 06/02/2017] [Indexed: 02/07/2023]
Abstract
The central nervous system (CNS) and its meningeal coverings accommodate a diverse myeloid compartment that includes parenchymal microglia and perivascular macrophages, as well as choroid plexus and meningeal macrophages, dendritic cells, and granulocytes. These myeloid populations enjoy an intimate relationship with the CNS, where they play an essential role in both health and disease. Although the importance of these cells is clearly recognized, their exact function in the CNS continues to be explored. Here, we review the subsets of myeloid cells that inhabit the parenchyma, meninges, and choroid plexus and discuss their roles in CNS homeostasis. We also discuss the role of these cells in various neurological pathologies, such as autoimmunity, mechanical injury, neurodegeneration, and infection. We highlight the neuroprotective nature of certain myeloid cells by emphasizing their therapeutic potential for the treatment of neurological conditions.
Collapse
Affiliation(s)
- Jasmin Herz
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Anthony J Filiano
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA.
| | - Ashtyn T Wiltbank
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Nir Yogev
- Gutenberg Research Fellowship Group of Neuroimmunology, Focus Program Translational Neuroscience and Immunotherapy, Rhine Main Neuroscience Network, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Jonathan Kipnis
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA; Gutenberg Research Fellowship Group of Neuroimmunology, Focus Program Translational Neuroscience and Immunotherapy, Rhine Main Neuroscience Network, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany.
| |
Collapse
|
34
|
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: 11.1] [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
|
35
|
Yao WR, Yu L, Li D, Yang GB. Molecular cloning and characterization of DNGR-1 in rhesus macaques. Mol Immunol 2017; 87:217-226. [PMID: 28511091 DOI: 10.1016/j.molimm.2017.04.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 03/22/2017] [Accepted: 04/26/2017] [Indexed: 02/07/2023]
Abstract
DC, NK lectin group receptor-1 (DNGR-1), also known as C-type lectin domain family 9 member A (CLEC9A), is a promising target for immunological therapeutics and vaccination against tumors and viruses. However, little is known about its property in rhesus macaques. In this study, we cloned rhesus macaque DNGR-1 cDNA, and found that its coding region could encode a 241-amino acid polypeptide with 91.7% sequence identity and similar antigenicity to that of humans. Both free and cell surface rhesus macaque DNGR-1 expressed in vitro could bind to apoptotic/dead cells induced by serum deprivation or freeze-thaw, and to pyroptotic cells stimulated with PMA and LPS. We also demonstrated that rhesus macaque DNGR-1 mRNA was present in all the examined tissues, with the highest in lymph nodes, spleen, blood, and thymus. The expression of DNGR-1 that is highly similar to that of humans warranted the usefulness of rhesus macaques in testing human therapeutics and vaccines targeting DNGR-1, especially those for HIV/AIDS.
Collapse
Affiliation(s)
- Wen-Rong Yao
- National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, 155 Changbai Road, Changping District, Beijing 102206, China
| | - Lei Yu
- National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, 155 Changbai Road, Changping District, Beijing 102206, China
| | - Dong Li
- National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, 155 Changbai Road, Changping District, Beijing 102206, China
| | - Gui-Bo Yang
- National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, 155 Changbai Road, Changping District, Beijing 102206, China.
| |
Collapse
|
36
|
Abstract
T cells are required for immune surveillance of the central nervous system (CNS); however, they can also induce severe immunopathology in the context of both viral infections and autoimmunity. The mechanisms that are involved in the priming and recruitment of T cells to the CNS are only partially understood, but there has been renewed interest in this topic since the 'rediscovery' of lymphatic drainage from the CNS. Moreover, tissue-resident memory T cells have been detected in the CNS and are increasingly recognized as an autonomous line of host defence. In this Review, we highlight the main mechanisms that are involved in the priming and CNS recruitment of CD4+ T cells, CD8+ T cells and regulatory T cells. We also consider the plasticity of T cell responses in the CNS, with a focus on viral infection and autoimmunity.
Collapse
|
37
|
Klein RS, Garber C, Howard N. Infectious immunity in the central nervous system and brain function. Nat Immunol 2017; 18:132-141. [PMID: 28092376 DOI: 10.1038/ni.3656] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 12/02/2016] [Indexed: 11/09/2022]
Abstract
Inflammation is emerging as a critical mechanism underlying neurological disorders of various etiologies, yet its role in altering brain function as a consequence of neuroinfectious disease remains unclear. Although acute alterations in mental status due to inflammation are a hallmark of central nervous system (CNS) infections with neurotropic pathogens, post-infectious neurologic dysfunction has traditionally been attributed to irreversible damage caused by the pathogens themselves. More recently, studies indicate that pathogen eradication within the CNS may require immune responses that interfere with neural cell function and communication without affecting their survival. In this Review we explore inflammatory processes underlying neurological impairments caused by CNS infection and discuss their potential links to established mechanisms of psychiatric and neurodegenerative diseases.
Collapse
Affiliation(s)
- Robyn S Klein
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA.,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA.,Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Charise Garber
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nicole Howard
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA.,Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
38
|
Worbs T, Hammerschmidt SI, Förster R. Dendritic cell migration in health and disease. Nat Rev Immunol 2016; 17:30-48. [PMID: 27890914 DOI: 10.1038/nri.2016.116] [Citation(s) in RCA: 536] [Impact Index Per Article: 67.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Dendritic cells (DCs) are potent and versatile antigen-presenting cells, and their ability to migrate is key for the initiation of protective pro-inflammatory as well as tolerogenic immune responses. Recent comprehensive studies have highlighted the importance of DC migration in the maintenance of immune surveillance and tissue homeostasis, and also in the pathogenesis of a range of diseases. In this Review, we summarize the anatomical, cellular and molecular factors that regulate the migration of different DC subsets in health and disease. In particular, we focus on new insights concerning the role of migratory DCs in the pathogenesis of diseases of the skin, intestine, lung, and brain, as well as in autoimmunity and atherosclerosis.
Collapse
Affiliation(s)
- Tim Worbs
- Institute of Immunology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Swantje I Hammerschmidt
- Institute of Immunology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| |
Collapse
|
39
|
Sie C, Korn T. Dendritic cells in central nervous system autoimmunity. Semin Immunopathol 2016; 39:99-111. [PMID: 27888330 DOI: 10.1007/s00281-016-0608-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 11/13/2016] [Indexed: 02/01/2023]
Abstract
Dendritic cells (DCs) operate at the intersection of the innate and adaptive immune systems. DCs can promote or inhibit adaptive immune responses against neuroantigens. While DC intrinsic properties, i.e., their maturation state or the subset they belong to, are important determinants of the outcome of an autoimmune reaction, tissue-specific cues might also be relevant for the function of DCs. Thus, a better understanding of the performance of distinct DC subsets in specific anatomical niches, not only in lymphoid tissue but also in non-lymphoid tissues such as the meninges, the choroid plexus, and the inflamed CNS parenchyma, will be instrumental for the design of immune intervention strategies to chronic inflammatory diseases that do not put at risk basic surveillance functions of the immune system in the CNS. Here, we will review modern concepts of DC biology in steady state and during autoimmune neuroinflammation.
Collapse
Affiliation(s)
- Christopher Sie
- Klinikum rechts der Isar, Department of Neurology and Department of Experimental Neuroimmunology, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - Thomas Korn
- Klinikum rechts der Isar, Department of Neurology and Department of Experimental Neuroimmunology, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany. .,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
40
|
Dando SJ, Naranjo Golborne C, Chinnery HR, Ruitenberg MJ, McMenamin PG. A case of mistaken identity: CD11c-eYFP(+) cells in the normal mouse brain parenchyma and neural retina display the phenotype of microglia, not dendritic cells. Glia 2016; 64:1331-49. [PMID: 27189804 DOI: 10.1002/glia.23005] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 04/25/2016] [Accepted: 04/25/2016] [Indexed: 12/13/2022]
Abstract
Under steady-state conditions the central nervous system (CNS) is traditionally thought to be devoid of antigen presenting cells; however, putative dendritic cells (DCs) expressing enhanced yellow fluorescent protein (eYFP) are present in the retina and brain parenchyma of CD11c-eYFP mice. We previously showed that these mice carry the Crb1(rd8) mutation, which causes retinal dystrophic lesions; therefore we hypothesized that the presence of CD11c-eYFP(+) cells within the CNS may be due to pathology associated with the Crb1(rd8) mutation. We generated CD11c-eYFP Crb1(wt/wt) mice and compared the distribution and immunophenotype of CD11c-eYFP(+) cells in CD11c-eYFP mice with and without the Crb1(rd8) mutation. The number and distribution of CD11c-eYFP(+) cells in the CNS was similar between CD11c-eYFP Crb1(wt/wt) and CD11c-eYFP Crb1(rd8/rd8) mice. CD11c-eYFP(+) cells were distributed throughout the inner retina, and clustered in brain regions that receive input from the external environment or lack a blood-brain barrier. CD11c-eYFP(+) cells within the retina and cerebral cortex of CD11c-eYFP Crb1(wt/wt) mice expressed CD11b, F4/80, CD115 and Iba-1, but not DC or antigen presentation markers, whereas CD11c-eYFP(+) cells within the choroid plexus and pia mater expressed CD11c, I-A/I-E, CD80, CD86, CD103, DEC205, CD8α and CD135. The immunophenotype of CD11c-eYFP(+) cells and microglia within the CNS was similar between CD11c-eYFP Crb1(wt/wt) and CD11c-eYFP Crb1(rd8/rd8) mice; however, CD11c and I-A/I-E expression was significantly increased in CD11c-eYFP Crb1(rd8/rd8) mice. This study demonstrates that the overwhelming majority of CNS CD11c-eYFP(+) cells do not display the phenotype of DCs or their precursors and are most likely a subpopulation of microglia. GLIA 2016. GLIA 2016;64:1331-1349.
Collapse
Affiliation(s)
- Samantha J Dando
- Department of Anatomy and Developmental Biology and Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Cecilia Naranjo Golborne
- Department of Anatomy and Developmental Biology and Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Holly R Chinnery
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Marc J Ruitenberg
- School of Biomedical Sciences, the University of Queensland, Brisbane, Queensland, Australia.,Queensland Brain Institute, the University of Queensland, Brisbane, Queensland, Australia
| | - Paul G McMenamin
- Department of Anatomy and Developmental Biology and Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
41
|
Ludewig P, Gallizioli M, Urra X, Behr S, Brait VH, Gelderblom M, Magnus T, Planas AM. Dendritic cells in brain diseases. Biochim Biophys Acta Mol Basis Dis 2015; 1862:352-67. [PMID: 26569432 DOI: 10.1016/j.bbadis.2015.11.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 11/05/2015] [Accepted: 11/05/2015] [Indexed: 12/25/2022]
Affiliation(s)
- Peter Ludewig
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mattia Gallizioli
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain
| | - Xabier Urra
- Functional Unit of Cerebrovascular Diseases, Hospital Clínic, Barcelona, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Sarah Behr
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Vanessa H Brait
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Mathias Gelderblom
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tim Magnus
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anna M Planas
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain.
| |
Collapse
|