1
|
Krawczyk MC, Pan L, Zhang AJ, Zhang Y. Lymphocyte deficiency alters the transcriptomes of oligodendrocytes, but not astrocytes or microglia. PLoS One 2023; 18:e0279736. [PMID: 36827449 PMCID: PMC9956607 DOI: 10.1371/journal.pone.0279736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 12/14/2022] [Indexed: 02/26/2023] Open
Abstract
Though the brain was long characterized as an immune-privileged organ, findings in recent years have shown extensive communications between the brain and peripheral immune cells. We now know that alterations in the peripheral immune system can affect the behavioral outputs of the central nervous system, but we do not know which brain cells are affected by the presence of peripheral immune cells. Glial cells including microglia, astrocytes, oligodendrocytes, and oligodendrocyte precursor cells (OPCs) are critical for the development and function of the central nervous system. In a wide range of neurological and psychiatric diseases, the glial cell state is influenced by infiltrating peripheral lymphocytes. However, it remains largely unclear whether the development of the molecular phenotypes of glial cells in the healthy brain is regulated by lymphocytes. To answer this question, we acutely purified each type of glial cell from immunodeficient Rag2-/- mice. Interestingly, we found that the transcriptomes of microglia, astrocytes, and OPCs developed normally in Rag2-/- mice without reliance on lymphocytes. In contrast, there are modest transcriptome differences between the oligodendrocytes from Rag2-/- and control mice. Furthermore, the subcellular localization of the RNA-binding protein Quaking, is altered in oligodendrocytes. These results demonstrate that the molecular attributes of glial cells develop largely without influence from lymphocytes and highlight potential interactions between lymphocytes and oligodendrocytes.
Collapse
Affiliation(s)
- Mitchell C. Krawczyk
- Department of Psychiatry and Biobehavioral Sciences, Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, Los Angeles, California, United States of America
| | - Lin Pan
- Department of Psychiatry and Biobehavioral Sciences, Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, Los Angeles, California, United States of America
| | - Alice J. Zhang
- Department of Psychiatry and Biobehavioral Sciences, Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, Los Angeles, California, United States of America
| | - Ye Zhang
- Department of Psychiatry and Biobehavioral Sciences, Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, Los Angeles, California, United States of America
- Brain Research Institute, University of California Los Angeles (UCLA), Los Angeles, Los Angeles, California, United States of America
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles (UCLA), Los Angeles, Los Angeles, California, United States of America
- Molecular Biology Institute, University of California Los Angeles (UCLA), Los Angeles, Los Angeles, California, United States of America
| |
Collapse
|
2
|
Current concepts on communication between the central nervous system and peripheral immunity via lymphatics: what roles do lymphatics play in brain and spinal cord disease pathogenesis? Biol Futur 2021; 72:45-60. [PMID: 34554497 DOI: 10.1007/s42977-021-00066-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 01/12/2021] [Indexed: 12/25/2022]
Abstract
The central nervous system (CNS) lacks conventional lymphatics within the CNS parenchyma, yet still maintains fluid homeostasis and immunosurveillance. How the CNS communicates with systemic immunity has thus been a topic of interest for scientists in the past century, which has led to several theories of CNS drainage routes. In addition to perineural routes, rediscoveries of lymphatics surrounding the CNS in the meninges revealed an extensive network of lymphatics, which we now know play a significant role in fluid homeostasis and immunosurveillance. These meningeal lymphatic networks exist along the superior sagittal sinus and transverse sinus dorsal to the brain, near the cribriform plate below the olfactory bulbs, at the base of the brain, and surrounding the spinal cord. Inhibition of one or all of these lymphatic networks can reduce CNS autoimmunity in a mouse model of multiple sclerosis (MS), while augmenting these lymphatic networks can improve immunosurveillance, immunotherapy, and clearance in glioblastoma, Alzheimer's disease, traumatic brain injury, and cerebrovascular injury. In this review, we will provide historical context of how CNS drainage contributes to immune surveillance, how more recently published studies fit meningeal lymphatics into the context of CNS homeostasis and neuroinflammation, identify the complex dualities of lymphatic function during neuroinflammation and how therapeutics targeting lymphatic function may be more complicated than currently appreciated, and conclude by identifying some unresolved questions and controversies that may guide future research.
Collapse
|
3
|
Hsu M, Laaker C, Sandor M, Fabry Z. Neuroinflammation-Driven Lymphangiogenesis in CNS Diseases. Front Cell Neurosci 2021; 15:683676. [PMID: 34248503 PMCID: PMC8261156 DOI: 10.3389/fncel.2021.683676] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 05/05/2021] [Indexed: 11/13/2022] Open
Abstract
The central nervous system (CNS) undergoes immunosurveillance despite the lack of conventional antigen presenting cells and lymphatic vessels in the CNS parenchyma. Additionally, the CNS is bathed in a cerebrospinal fluid (CSF). CSF is continuously produced, and consequently must continuously clear to maintain fluid homeostasis despite the lack of conventional lymphatics. During neuroinflammation, there is often an accumulation of fluid, antigens, and immune cells to affected areas of the brain parenchyma. Failure to effectively drain these factors may result in edema, prolonged immune response, and adverse clinical outcome as observed in conditions including traumatic brain injury, ischemic and hypoxic brain injury, CNS infection, multiple sclerosis (MS), and brain cancer. Consequently, there has been renewed interest surrounding the expansion of lymphatic vessels adjacent to the CNS which are now thought to be central in regulating the drainage of fluid, cells, and waste out of the CNS. These lymphatic vessels, found at the cribriform plate, dorsal dural meninges, base of the brain, and around the spinal cord have each been implicated to have important roles in various CNS diseases. In this review, we discuss the contribution of meningeal lymphatics to these processes during both steady-state conditions and neuroinflammation, as well as discuss some of the many still unknown aspects regarding the role of meningeal lymphatics in neuroinflammation. Specifically, we focus on the observed phenomenon of lymphangiogenesis by a subset of meningeal lymphatics near the cribriform plate during neuroinflammation, and discuss their potential roles in immunosurveillance, fluid clearance, and access to the CSF and CNS compartments. We propose that manipulating CNS lymphatics may be a new therapeutic way to treat CNS infections, stroke, and autoimmunity.
Collapse
Affiliation(s)
- Martin Hsu
- Neuroscience Training Program, University of Wisconsin Madison, Madison, WI, United States
| | - Collin Laaker
- Neuroscience Training Program, University of Wisconsin Madison, Madison, WI, United States
| | - Matyas Sandor
- Department of Pathology and Laboratory Medicine, University of Wisconsin Madison, Madison, WI, United States
| | - Zsuzsanna Fabry
- Department of Pathology and Laboratory Medicine, University of Wisconsin Madison, Madison, WI, United States
| |
Collapse
|
4
|
Regional Distribution of CNS Antigens Differentially Determines T-Cell Mediated Neuroinflammation in a CX3CR1-Dependent Manner. J Neurosci 2018; 38:7058-7071. [PMID: 29959236 DOI: 10.1523/jneurosci.0366-18.2018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 05/15/2018] [Accepted: 05/18/2018] [Indexed: 01/29/2023] Open
Abstract
T cells continuously sample CNS-derived antigens in the periphery, yet it is unknown how they sample and respond to CNS antigens derived from distinct brain areas. We expressed ovalbumin (OVA) neoepitopes in regionally distinct CNS areas (Cnp-OVA and Nes-OVA mice) to test peripheral antigen sampling by OVA-specific T cells under homeostatic and neuroinflammatory conditions. We show that antigen sampling in the periphery is independent of regional origin of CNS antigens in both male and female mice. However, experimental autoimmune encephalomyelitis (EAE) is differentially influenced in Cnp-OVA and Nes-OVA female mice. Although there is the same frequency of CD45high CD11b+ CD11c+ CX3CL1+ myeloid cell-T-cell clusters in neoepitope-expressing areas, EAE is inhibited in Nes-OVA female mice and accelerated in CNP-OVA female mice. Accumulation of OVA-specific T cells and their immunomodulatory effects on EAE are CX3C chemokine receptor 1 (CX3CR1) dependent. These data show that despite similar levels of peripheral antigen sampling, CNS antigen-specific T cells differentially influence neuroinflammatory disease depending on the location of cognate antigens and the presence of CX3CL1/CX3CR1 signaling.SIGNIFICANCE STATEMENT Our data show that peripheral T cells similarly recognize neoepitopes independent of their origin within the CNS under homeostatic conditions. Contrastingly, during ongoing autoimmune neuroinflammation, neoepitope-specific T cells differentially influence clinical score and pathology based on the CNS regional location of the neoepitopes in a CX3CR1-dependent manner. Altogether, we propose a novel mechanism for how T cells respond to regionally distinct CNS derived antigens and contribute to CNS autoimmune pathology.
Collapse
|
5
|
Tanabe S, Yamashita T. The role of immune cells in brain development and neurodevelopmental diseases. Int Immunol 2018; 30:437-444. [DOI: 10.1093/intimm/dxy041] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Accepted: 06/14/2018] [Indexed: 12/13/2022] Open
Affiliation(s)
- Shogo Tanabe
- Department of Molecular Neuroscience, World Premier International Immunology Frontier Research Center, Osaka University, Suita-shi, Osaka, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, World Premier International Immunology Frontier Research Center, Osaka University, Suita-shi, Osaka, Japan
- Graduate School of Medicine, Osaka University, Suita-shi, Osaka, Japan
- Graduate School of Frontier Biosciences, Osaka University, Suita-shi, Osaka, Japan
| |
Collapse
|
6
|
Brombacher TM, Nono JK, De Gouveia KS, Makena N, Darby M, Womersley J, Tamgue O, Brombacher F. IL-13-Mediated Regulation of Learning and Memory. THE JOURNAL OF IMMUNOLOGY 2017; 198:2681-2688. [PMID: 28202615 DOI: 10.4049/jimmunol.1601546] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 01/17/2017] [Indexed: 11/19/2022]
Abstract
The role of proinflammatory cytokines in cognitive function has been investigated with both beneficial and possible detrimental effects, depending on the cytokine. More recently, the type 2 IL-4 has been demonstrated to play a role in cognition. In this study, using the Morris water maze task, we demonstrate that IL-13-deficient mice are significantly impaired in working memory as well as attenuated reference memory, both functions essential for effective complex learning. During the learning process, wild-type mice increased the number of CD4+ T cells in the meninges and production of IL-13, whereas neither Morris water maze-trained IL-4 nor trained IL-13-deficient mice were able to increase CD4+ T cells in the meninges. Mechanistically, we showed that IL-13 is able to stimulate primary astrocytes to produce brain-derived neurotrophic factor, which does foster cognitive functions. Moreover, Morris water maze-trained wild-type mice were able to increase astrocyte-produced glial fibrillary acidic protein in the hippocampus, which was impaired in Morris water maze-trained IL-4- and IL-13-deficient mice. Collectively, this study strongly suggests that the Th2 cytokines, not only IL-4 but also IL-13, are involved in cognitive functions by stimulating astrocytes from the meninges and hippocampus. These results may be important for future development of therapeutic approaches associated with neurologic disorders such as Parkinson disease-associated dementia and HIV-associated dementia among others.
Collapse
Affiliation(s)
- Tiroyaone M Brombacher
- Cape Town Component, International Centre for Genetic Engineering and Biotechnology, Cape Town 7925, South Africa.,Division of Immunology, Institute of Infectious Disease and Molecular Medicine, Health Science Faculty, University of Cape Town, Cape Town 7925, South Africa.,South African Medical Research Council, Cape Town 7501, South Africa
| | - Justin K Nono
- Cape Town Component, International Centre for Genetic Engineering and Biotechnology, Cape Town 7925, South Africa.,Division of Immunology, Institute of Infectious Disease and Molecular Medicine, Health Science Faculty, University of Cape Town, Cape Town 7925, South Africa.,South African Medical Research Council, Cape Town 7501, South Africa.,Medical Research Centre, Institute of Medical Research and Medicinal Plant Studies, Ministry of Scientific Research and Innovation, Yaoundé, Cameroon; and
| | - Keisha S De Gouveia
- Cape Town Component, International Centre for Genetic Engineering and Biotechnology, Cape Town 7925, South Africa.,Division of Immunology, Institute of Infectious Disease and Molecular Medicine, Health Science Faculty, University of Cape Town, Cape Town 7925, South Africa.,South African Medical Research Council, Cape Town 7501, South Africa
| | - Nokuthula Makena
- Department of Human Biology, University of Cape Town, Cape Town 7925, South Africa
| | - Matthew Darby
- Cape Town Component, International Centre for Genetic Engineering and Biotechnology, Cape Town 7925, South Africa.,Division of Immunology, Institute of Infectious Disease and Molecular Medicine, Health Science Faculty, University of Cape Town, Cape Town 7925, South Africa.,South African Medical Research Council, Cape Town 7501, South Africa
| | - Jacqueline Womersley
- Department of Human Biology, University of Cape Town, Cape Town 7925, South Africa
| | - Ousman Tamgue
- Cape Town Component, International Centre for Genetic Engineering and Biotechnology, Cape Town 7925, South Africa.,Division of Immunology, Institute of Infectious Disease and Molecular Medicine, Health Science Faculty, University of Cape Town, Cape Town 7925, South Africa.,South African Medical Research Council, Cape Town 7501, South Africa
| | - Frank Brombacher
- Cape Town Component, International Centre for Genetic Engineering and Biotechnology, Cape Town 7925, South Africa; .,Division of Immunology, Institute of Infectious Disease and Molecular Medicine, Health Science Faculty, University of Cape Town, Cape Town 7925, South Africa.,South African Medical Research Council, Cape Town 7501, South Africa
| |
Collapse
|
7
|
Ritzel RM, Crapser J, Patel AR, Verma R, Grenier JM, Chauhan A, Jellison ER, McCullough LD. Age-Associated Resident Memory CD8 T Cells in the Central Nervous System Are Primed To Potentiate Inflammation after Ischemic Brain Injury. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 196:3318-30. [PMID: 26962232 PMCID: PMC4868658 DOI: 10.4049/jimmunol.1502021] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 02/05/2016] [Indexed: 01/17/2023]
Abstract
Aging is associated with an increase in basal inflammation in the CNS and an overall decline in cognitive function and poorer recovery following injury. Growing evidence suggests that leukocyte recruitment to the CNS is also increased with normal aging, but, to date, no systematic evaluation of these age-associated leukocytes has been performed. In this work, the effect of aging on CNS leukocyte recruitment was examined. Aging was associated with more CD45(high) leukocytes, primarily composed of conventional CD8(+) T cells. These results were strain independent and seen in both sexes. Intravascular labeling and immunohistology revealed the presence of parenchymal CD8(+) T cells in several regions of the brain, including the choroid plexus and meninges. These cells had effector memory (CD44(+)CD62L(-)) and tissue-resident phenotypes and expressed markers associated with TCR activation. Analysis of TCRvβ repertoire usage suggested that entry into the CNS is most likely stochastic rather than Ag driven. Correlational analyses revealed a positive association between CD8 T cell numbers and decreased proinflammatory function of microglia. However, the effects of cerebral ischemia and ex vivo stimulation of these cells dramatically increased production of TNF, IFN-γ, and MCP-1/CCL2. Taken together, we identified a novel population of resident memory, immunosurveillant CD8 T cells that represent a hallmark of CNS aging and appear to modify microglia homeostasis under normal conditions, but are primed to potentiate inflammation and leukocyte recruitment following ischemic injury.
Collapse
Affiliation(s)
- Rodney M Ritzel
- Neuroscience Department, University of Connecticut Health Center, Farmington, CT 06030
| | - Joshua Crapser
- Neuroscience Department, University of Connecticut Health Center, Farmington, CT 06030
| | - Anita R Patel
- Neuroscience Department, University of Connecticut Health Center, Farmington, CT 06030
| | - Rajkumer Verma
- Neuroscience Department, University of Connecticut Health Center, Farmington, CT 06030
| | - Jeremy M Grenier
- Immunology Department, University of Connecticut Health Center, Farmington, CT 06030; and
| | - Anjali Chauhan
- Neuroscience Department, University of Connecticut Health Center, Farmington, CT 06030
| | - Evan R Jellison
- Immunology Department, University of Connecticut Health Center, Farmington, CT 06030; and
| | - Louise D McCullough
- Neuroscience Department, University of Connecticut Health Center, Farmington, CT 06030; Department of Neurology, McGovern Medical School, University of Texas Health Science Center, Houston, TX 77370
| |
Collapse
|
8
|
Matrine improves cognitive impairment and modulates the balance of Th17/Treg cytokines in a rat model of Aβ1-42-induced Alzheimer's disease. Cent Eur J Immunol 2016; 40:411-9. [PMID: 26862304 PMCID: PMC4737738 DOI: 10.5114/ceji.2015.56961] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 08/17/2015] [Indexed: 11/29/2022] Open
Abstract
Matrine (MAT) has been reported for its anti-inflammatory and neuroprotective effects. However, little is known about its effects on Th17/Treg cytokines and cognitive impairment in Alzheimer's disease (AD). In the present study, we injected Aβ1-42 to the hippocampus of the rat to induce AD. Three groups of the AD rats were treated with MAT (25, 100 or 200 mg/kg/day, respectively) by intraperitoneal injection for 5 weeks. Levels of Th17 cell cytokines [interleukin (IL)-17A and IL-23] and regulatory T (Treg) cell cytokines [transforming growth factor β (TGF-β) and IL-35] in homogenates of the brain cortex and hippocampus were measured using enzyme-linked immunosorbent assay (ELISA) kits. The mRNA expressions of Th17 cell specific transcription factor RORγt and Treg cell specific transcription factor Foxp3 in the brain cortex and hippocampus were quantified by real-time RT-PCR. Learning and memory ability of the rats were evaluated by Morris water maze test and novel object recognition test. ELISA detections showed the AD rats had increased levels of IL-17A and IL-23 as well as decreased levels of TGF-β and IL-35. Matrine (100 and 200 mg/kg/day) significantly reversed the alternations of Th17/Treg cytokines induced by Aβ1-42 injection, decreased RORγt mRNA expression, increased Foxp3 mRNA expression and improved the learning and memory ability in the AD rats. The findings demonstrated that the AD rats had imbalance of Th17/Treg cytokines in the brain. MAT could dose-dependently restore the balance of Th17/Treg cytokines and attenuate the cognitive impairment in AD rats.
Collapse
|
9
|
Abstract
Despite dramatic advances in surgical techniques, imaging and adjuvant radiotherapy or chemotherapy, the prognosis for patients with malignant glial tumors remains dismal. Based on the current knowledge regarding immune responses in the healthy CNS and glioma-bearing hosts, this review discusses dendritic cell-based immunotherapeutic approaches for malignant gliomas and the relevance of recent clinical trials and their outcomes. It is now recognized that the CNS is not an immunologically tolerated site and clearance of arising glioma cells is a routine physiologic function of the normal, noncompromised immune system. To escape from immune surveillance, however, clinically apparent gliomas develop complex mechanisms that suppress tumoricidal immune responses. Although the use of dendritic cells for the treatment of glioma patients may be the most appropriate approach, an effective treatment paradigm for these tumors may eventually require the use of several types of treatment. Additionally, given the heterogeneity of this disease process and an immune-refractory tumor cell population, the series use of rational multiple modalities that target disparate tumor characteristics may be the most effective therapeutic strategy to treat malignant gliomas.
Collapse
Affiliation(s)
- Yasuharu Akasaki
- Maxine Dunitz Neurosurgical Institute, Cedars-Sinai Medical Center, Suite 800 East, 8631 West 3 Street, Los Angeles, CA 90048, USA
| | | | | |
Collapse
|
10
|
Glioma stem cells and immunotherapy for the treatment of malignant gliomas. ISRN ONCOLOGY 2013; 2013:673793. [PMID: 23762610 PMCID: PMC3671309 DOI: 10.1155/2013/673793] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/03/2013] [Accepted: 03/27/2013] [Indexed: 02/06/2023]
Abstract
Stem cell research has led to the discovery of glioma stem cells (GSCs), and because these cells are resistant to chemotherapy and radiotherapy, analysis of their properties has been rapidly pursued for targeted treatment of malignant glioma. Recent studies have also revealed complex crosstalk between GSCs and their specialized environment (niche). Therefore, targeting not only GSCs but also their niche may be a principle for novel therapies of malignant glioma. One possible novel strategy for targeting GSCs and their niches is immunotherapy with different antitumor mechanism(s) from those of conventional therapy. Recent clinical studies of immunotherapy using peptide vaccines and antibodies have shown promising results. This review describes the recent findings related to GSCs and their niches, as well as immunotherapies for glioma, followed by discussion of immunotherapies that target GSCs for the treatment of malignant glioma.
Collapse
|
11
|
Harris MG, Fabry Z. Initiation and Regulation of CNS Autoimmunity: Balancing Immune Surveillance and Inflammation in the CNS. ACTA ACUST UNITED AC 2012. [DOI: 10.4236/nm.2012.33026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
12
|
Lambracht-Washington D, Qu BX, Fu M, Anderson LD, Stüve O, Eagar TN, Rosenberg RN. DNA immunization against amyloid beta 42 has high potential as safe therapy for Alzheimer's disease as it diminishes antigen-specific Th1 and Th17 cell proliferation. Cell Mol Neurobiol 2011; 31:867-74. [PMID: 21625960 DOI: 10.1007/s10571-011-9680-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2011] [Accepted: 02/23/2011] [Indexed: 10/18/2022]
Abstract
The pathogenesis of Alzheimer's disease (AD) has been strongly associated with the accumulation of amyloid beta (Aβ) peptides in brain, and immunotherapy targeting Aβ provides potential for AD prevention. A clinical trial in which AD patients were immunized with Aβ42 peptide was stopped when 6% of participants showed meningoencephalitis, apparently due to an inflammatory Th1 immune response. Previously, we and other have shown that Aβ42 DNA vaccination via gene gun generates a Th2 cellular immune response, which was shown by analyses of the respective antibody isotype profiles. We also determined that in vitro T cell proliferation in response to Aβ42 peptide re-stimulation was absent in DNA Aβ42 trimer-immunized mice when compared to Aβ42 peptide-immunized mice. To further characterize this observation prospectively and longitudinally, we analyzed the immune response in wild-type mice after vaccination with Aβ42 trimer DNA and Aβ42 peptide with Quil A adjuvant. Wild-type mice were immunized with short-term (1-3× vaccinations) or long-term (6× vacinations) immunization strategies. Antibody titers and isotype profiles of the Aβ42 specific antibodies, as well as cytokine profiles and cell proliferation studies from this longitudinal study were determined. Sufficient antibody titers to effectively reduce Aβ42, but an absent T cell proliferative response and no IFNγ or IL-17 secretion after Aβ42 DNA trimer immunization minimizes the risk of inflammatory activities of the immune system towards the self antigen Aβ42 in brain. Therefore, Aβ42 DNA trimer immunization has a high probability to be effective and safe to treat patients with early AD.
Collapse
|
13
|
Chlamydia pneumoniae infection enhances microglial activation in atherosclerotic mice. Neurobiol Aging 2010; 31:1766-73. [DOI: 10.1016/j.neurobiolaging.2008.09.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2006] [Revised: 09/14/2008] [Accepted: 09/30/2008] [Indexed: 11/22/2022]
|
14
|
Derecki NC, Cardani AN, Yang CH, Quinnies KM, Crihfield A, Lynch KR, Kipnis J. Regulation of learning and memory by meningeal immunity: a key role for IL-4. J Exp Med 2010; 207:1067-80. [PMID: 20439540 PMCID: PMC2867291 DOI: 10.1084/jem.20091419] [Citation(s) in RCA: 617] [Impact Index Per Article: 41.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2009] [Accepted: 04/01/2010] [Indexed: 12/25/2022] Open
Abstract
Proinflammatory cytokines have been shown to impair cognition; consequently, immune activity in the central nervous system was considered detrimental to cognitive function. Unexpectedly, however, T cells were recently shown to support learning and memory, though the underlying mechanism was unclear. We show that one of the steps in the cascade of T cell-based support of learning and memory takes place in the meningeal spaces. Performance of cognitive tasks led to accumulation of IL-4-producing T cells in the meninges. Depletion of T cells from meningeal spaces skewed meningeal myeloid cells toward a proinflammatory phenotype. T cell-derived IL-4 was critical, as IL-4(-/-) mice exhibited a skewed proinflammatory meningeal myeloid cell phenotype and cognitive deficits. Transplantation of IL-4(-/-) bone marrow into irradiated wild-type recipients also resulted in cognitive impairment and proinflammatory skew. Moreover, adoptive transfer of T cells from wild-type into IL-4(-/-) mice reversed cognitive impairment and attenuated the proinflammatory character of meningeal myeloid cells. Our results point to a critical role for T cell-derived IL-4 in the regulation of cognitive function through meningeal myeloid cell phenotype and brain-derived neurotrophic factor expression. These findings might lead to the development of new immune-based therapies for cognitive impairment associated with immune decline.
Collapse
Affiliation(s)
- Noël C. Derecki
- Department of Neuroscience and Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, Graduate Program in Neuroscience and Graduate Program in Immunology, University of Virginia, Charlottesville, VA 22908
| | - Amber N. Cardani
- Department of Neuroscience and Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, Graduate Program in Neuroscience and Graduate Program in Immunology, University of Virginia, Charlottesville, VA 22908
| | - Chun Hui Yang
- Department of Neuroscience and Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, Graduate Program in Neuroscience and Graduate Program in Immunology, University of Virginia, Charlottesville, VA 22908
| | - Kayla M. Quinnies
- Department of Neuroscience and Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, Graduate Program in Neuroscience and Graduate Program in Immunology, University of Virginia, Charlottesville, VA 22908
| | - Anastasia Crihfield
- Department of Neuroscience and Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, Graduate Program in Neuroscience and Graduate Program in Immunology, University of Virginia, Charlottesville, VA 22908
| | - Kevin R. Lynch
- Department of Neuroscience and Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, Graduate Program in Neuroscience and Graduate Program in Immunology, University of Virginia, Charlottesville, VA 22908
| | - Jonathan Kipnis
- Department of Neuroscience and Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, Graduate Program in Neuroscience and Graduate Program in Immunology, University of Virginia, Charlottesville, VA 22908
| |
Collapse
|
15
|
Derecki NC, Privman E, Kipnis J. Rett syndrome and other autism spectrum disorders--brain diseases of immune malfunction? Mol Psychiatry 2010; 15:355-63. [PMID: 20177406 PMCID: PMC3368984 DOI: 10.1038/mp.2010.21] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2010] [Accepted: 02/03/2010] [Indexed: 02/07/2023]
Abstract
Neuroimmunology was once referred to in terms of its pathological connotation only and was generally understood as covering the deleterious involvement of the immune system in various diseases and disorders of the central nervous system (CNS). However, our conception of the function of the immune system in the structure, function, and plasticity of the CNS has undergone a sea change after relevant discoveries over the past two decades, and continues to be challenged by more recent studies of neurodevelopment and cognition. This review summarizes the recent advances in understanding of immune-system participation in the development and functioning of the CNS under physiological conditions. Considering as an example Rett syndrome a devastating neurodevelopmental disease, we offer a hypothesis that might help to explain the part played by immune cells in its etiology, and hence suggests that the immune system might be a feasible therapeutic target for alleviation of some of the symptoms of this and other autism spectrum disorders.
Collapse
Affiliation(s)
- NC Derecki
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - E Privman
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
- Medical Scientist Training Program, University of Virginia, Charlottesville, VA, USA
| | - J Kipnis
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
- Medical Scientist Training Program, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
16
|
Ling C, Verbny YI, Banks MI, Sandor M, Fabry Z. In situ activation of antigen-specific CD8+ T cells in the presence of antigen in organotypic brain slices. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 180:8393-9. [PMID: 18523307 PMCID: PMC3338102 DOI: 10.4049/jimmunol.180.12.8393] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The activation of Ag-specific T cells locally in the CNS could potentially contribute to the development of immune-mediated brain diseases. We addressed whether Ag-specific T cells could be stimulated in the CNS in the absence of peripheral lymphoid tissues by analyzing Ag-specific T cell responses in organotypic brain slice cultures. Organotypic brain slice cultures were established 1 h after intracerebral OVA Ag microinjection. We showed that when OVA-specific CD8(+) T cells were added to Ag-containing brain slices, these cells became activated and migrated into the brain to the sites of their specific Ags. This activation of OVA-specific T cells was abrogated by the deletion of CD11c(+) cells from the brain slices of the donor mice. These data suggest that brain-resident CD11c(+) cells stimulate Ag-specific naive CD8(+) T cells locally in the CNS and may contribute to immune responses in the brain.
Collapse
Affiliation(s)
- Changying Ling
- Department of Pathology and Laboratory Medicine, University of Wisconsin Medical School, University of Wisconsin, Madison, WI 53706
| | - Yakov I. Verbny
- Department of Anesthesiology, University of Wisconsin, Madison, WI 53706
| | - Matthew I. Banks
- Department of Anesthesiology, University of Wisconsin, Madison, WI 53706
| | - Matyas Sandor
- Department of Pathology and Laboratory Medicine, University of Wisconsin Medical School, University of Wisconsin, Madison, WI 53706
| | - Zsuzsanna Fabry
- Department of Pathology and Laboratory Medicine, University of Wisconsin Medical School, University of Wisconsin, Madison, WI 53706
| |
Collapse
|
17
|
Reinke EK, Lee J, Zozulya A, Karman J, Muller WA, Sandor M, Fabry Z. Short-term sPECAM-Fc treatment ameliorates EAE while chronic use hastens onset of symptoms. J Neuroimmunol 2007; 186:86-93. [PMID: 17467062 PMCID: PMC1950937 DOI: 10.1016/j.jneuroim.2007.03.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2006] [Revised: 03/06/2007] [Accepted: 03/07/2007] [Indexed: 11/19/2022]
Abstract
The homophilic cell adhesion molecule PECAM-1 is a major participant in the migration of leukocytes across endothelium. We examined the ability of a chimeric soluble PECAM-1 fused to human IgG-Fc to impair leukocyte entry through the blood-brain barrier and reduce CNS autoimmunity. sPECAM-Fc impaired migration of lymphocytes across brain endothelial monolayers and diminished the severity of EAE, an experimental model of MS, when administered at the onset of symptoms. However, in mice transgenic for sPECAM-Fc, the chronically elevated levels of sPECAM-Fc hastened onset of EAE disease without significantly changing clinical score severity. Our data suggest that short-term treatment of diseases like MS with sPECAM-Fc has therapeutic potential.
Collapse
Affiliation(s)
- Emily K. Reinke
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison WI
- Neuroscience Training Program University of Wisconsin-Madison, Madison, WI
| | - JangEun Lee
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison WI
- Cellular and Molecular Pathology Training Program University of Wisconsin-Madison; Madison, WI
| | - Alla Zozulya
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison WI
| | - Jozsef Karman
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison WI
- Cellular and Molecular Pathology Training Program University of Wisconsin-Madison; Madison, WI
| | - William A. Muller
- Department of Pathology, Weill Medical College of Cornell University, New York, NY 10021
| | - Matyas Sandor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison WI
- Cellular and Molecular Pathology Training Program University of Wisconsin-Madison; Madison, WI
| | - Zsuzsanna Fabry
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison WI
- Cellular and Molecular Pathology Training Program University of Wisconsin-Madison; Madison, WI
- Neuroscience Training Program University of Wisconsin-Madison, Madison, WI
| |
Collapse
|
18
|
Ling C, Sandor M, Suresh M, Fabry Z. Traumatic injury and the presence of antigen differentially contribute to T-cell recruitment in the CNS. J Neurosci 2006; 26:731-41. [PMID: 16421293 PMCID: PMC6675378 DOI: 10.1523/jneurosci.3502-05.2006] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
T-cell recruitment into the brain is critical in inflammatory and autoimmune diseases of the CNS. We use intracerebral antigen microinjection and tetramer technology to track antigen-specific CD8+ T-cells in the CNS and to clarify the contribution of antigen deposition or traumatic injury to the accumulation of T-cells in the brain. We demonstrate that, after intracerebral microinjection of ovalbumin, ovalbumin-specific CD8+ T-cells expand systemically and then migrate into the brain where they complete additional proliferation cycles. T-cells in the brain are activated and respond to in vitro secondary antigen challenge. CD8+ T-cells accumulate and persist in sites of antigen in the brain without replenishment from the periphery. Persistent survival of CD8+ T-cells at sites of cognate antigen is significantly reduced by blocking CD154 molecules. A small traumatic injury itself does not lead to recruitment of CD8+ T-cells into the brain but attracts activated antigen-specific CD8+ T-cells from cognate antigen injection sites. This process is presumably antigen independent and cannot be inhibited by blocking CD154 molecules. These data show that activated antigen-specific CD8+ T-cells accumulate in the CNS at both cognate antigen-containing and traumatic injury sites after intracerebral antigen delivery. The accumulation of activated antigen-specific T-cells at traumatic injury sites, in addition to antigen-containing areas, could amplify local inflammatory processes in the CNS. Combination therapies in neuroinflammatory diseases to block both of these processes should be considered.
Collapse
Affiliation(s)
- Changying Ling
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison Medical School, Madison, Wisconsin 53706, USA
| | | | | | | |
Collapse
|
19
|
Karman J, Ling C, Sandor M, Fabry Z. Initiation of Immune Responses in Brain Is Promoted by Local Dendritic Cells. THE JOURNAL OF IMMUNOLOGY 2004; 173:2353-61. [PMID: 15294948 DOI: 10.4049/jimmunol.173.4.2353] [Citation(s) in RCA: 177] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The contribution of dendritic cells (DCs) to initiating T cell-mediated immune response in and T cell homing into the CNS has not yet been clarified. In this study we show by confocal microscopy and flow cytometry that cells expressing CD11c, CD205, and MHC class II molecules and containing fluorescently labeled, processed Ag accumulate at the site of intracerebral Ag injection. These cells follow a specific pattern upon migrating out of the brain. To track their pathway out of the CNS, we differentiated DCs from bone marrow of GFP-transgenic mice and injected them directly into brains of naive C57BL/6 mice. We demonstrate that DCs migrate from brain to cervical lymph nodes, a process that can be blocked by fixation or pertussis toxin treatment of the DCs. Injection of OVA-loaded DCs into brain initiates a SIINFEKL (a dominant OVA epitope)-specific T cell response in lymph nodes and spleen, as measured by specific tetramer and LFA-1 activation marker staining. Additionally, a fraction of activated SIINFEKL-specific T cells home to the CNS. Specific T cell homing to the CNS, however, cannot be induced by i.v. injection of OVA-loaded DCs alone. These data suggest that brain-emigrant DCs are sufficient to support activated T cells to home to the tissue of DC origination. Thus, initiation of immune reactivity against CNS Ags involves the migration of APCs from nervous tissue to peripheral lymphoid tissues, similarly to that in other organs.
Collapse
Affiliation(s)
- Jozsef Karman
- Cellular and Molecular Pathology Program, University of Wisconsin, Madison 53706, USA
| | | | | | | |
Collapse
|
20
|
Ling C, Sandor M, Fabry Z. In situ processing and distribution of intracerebrally injected OVA in the CNS. J Neuroimmunol 2003; 141:90-8. [PMID: 12965258 DOI: 10.1016/s0165-5728(03)00249-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Drainage and retention of brain-derived antigens are important factors in initiating and regulating immune responses in the central nervous system (CNS). We investigated distribution, immunological processing and retention of intracerebrally infused protein antigen, ovalbumin (OVA), and the subsequent recruitment of CD8(+) T cells into the CNS. We found that protein antigens infused into the CNS can drain rapidly into the cervical lymph node and initiate antigen-specific immune response in the periphery. A portion of the antigens are also retained by CD11b/MAC-1(+) cells in the brain parenchyma where they are recognized by antigen-specific CD8(+) T cells.
Collapse
Affiliation(s)
- Changying Ling
- Department of Pathology and Laboratory Medicine, University of Wisconsin, 6130 MSC, Madison, WI 53706, USA
| | | | | |
Collapse
|
21
|
Giuliani F, Goodyer CG, Antel JP, Yong VW. Vulnerability of human neurons to T cell-mediated cytotoxicity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 171:368-79. [PMID: 12817020 DOI: 10.4049/jimmunol.171.1.368] [Citation(s) in RCA: 158] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Axonal and neuronal loss occurs in inflammatory diseases of the CNS such as multiple sclerosis. The cause of the loss remains unclear. We report that polyclonally activated T cells align along axons and soma of cultured human neurons leading to substantial neuronal death. This occurs in an allogeneic and syngeneic manner in the absence of added Ag, requires T cells to be activated, and is mediated through cell contact-dependent mechanisms involving FasL, LFA-1, and CD40 but not MHC class I. Activated CD4(+) and CD8(+) T cell subsets are equally neuronal cytotoxic. In contrast to neurons, other CNS cell types (oligodendrocytes and astrocytes) are not killed by T cells. These results demonstrate for the first time the high and selective vulnerability of human neurons to T cells, and suggest that when enough activated T cells accumulate in the CNS, neuronal cytotoxicity can result through Ag-independent non-MHC class I mechanisms.
Collapse
Affiliation(s)
- Fabrizio Giuliani
- Department of Oncology, University of Calgary, Calgary, Alberta, Canada
| | | | | | | |
Collapse
|
22
|
Pachter JS, de Vries HE, Fabry Z. The blood-brain barrier and its role in immune privilege in the central nervous system. J Neuropathol Exp Neurol 2003; 62:593-604. [PMID: 12834104 DOI: 10.1093/jnen/62.6.593] [Citation(s) in RCA: 192] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The blood-brain barrier (BBB) provides both anatomical and physiological protection for the central nervous system (CNS), strictly regulating the entry of many substances and blood borne cells into the nervous tissue. Increased understanding of how the unique microenvironment in the CNS influences the BBB is crucial for developing novel therapeutic approaches to CNS diseases. In this review, we discuss those characteristics of the BBB that play an important role in maintaining immune privilege in the CNS, as well as factors that regulate immune cell invasion through the BBB and thereby modulate immune responses in the nervous tissue. In general, immune cell invasion across the BBB is highly restricted and carefully regulated. A florid invasion of activated white blood cells can create a predominantly proinflammatory local environment in the CNS, leading to immune-mediated diseases of the nervous tissue. Recent developments in cellular and molecular biological methods have allowed closer analysis of BBB function, and led to an improved understanding of the active role of the BBB in immune-mediated diseases of the CNS.
Collapse
Affiliation(s)
- Joel S Pachter
- University of Connecticut Health Center, Farmington, Connecticut, USA
| | | | | |
Collapse
|
23
|
Walker PR, Calzascia T, de Tribolet N, Dietrich PY. T-cell immune responses in the brain and their relevance for cerebral malignancies. BRAIN RESEARCH. BRAIN RESEARCH REVIEWS 2003; 42:97-122. [PMID: 12738053 DOI: 10.1016/s0165-0173(03)00141-3] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
In order that cellular immune responses afford protection without risk to sensitive normal tissue, they must be adapted to individual tissues of the body. Nowhere is this more critical than for the brain, where various passive and active mechanisms maintain a state of immune privilege that can limit high magnitude immune responses. Nevertheless, it is now clear that immune responses are induced to antigens in the brain, including those expressed by cerebral malignancies. We discuss hypotheses of how this can occur, although details such as which antigen presenting cells are involved remain to be clarified. Antitumor responses induced spontaneously are insufficient to eradicate malignant astrocytomas; many studies suggest that this can be explained by a combination of low level immune response induction and tumor mediated immunosuppression. A clinical objective currently pursued is to use immunotherapy to ameliorate antitumour immunity. This will necessitate a high level immune response to ensure sufficient effector cells reach the tumor bed, focused cytotoxicity to eradicate malignant cells with little collateral damage to critical normal cells, and minimal inflammation. To achieve these aims, priority should be given to identifying more target antigens in astrocytoma and defining those cells present in the brain parenchyma that are essential to maintain antitumour effector function without exacerbating inflammation. If we are armed with better understanding of immune interactions with brain tumor cells, we can realistically envisage that immunotherapy will one day offer hope to patients with currently untreatable neoplastic diseases of the CNS.
Collapse
Affiliation(s)
- Paul R Walker
- Laboratory of Tumour Immunology, Division of Oncology, Geneva University Hospital, 24 rue Micheli-du-Crest, 1211 Geneva 14, Switzerland.
| | | | | | | |
Collapse
|
24
|
Fee D, Crumbaugh A, Jacques T, Herdrich B, Sewell D, Auerbach D, Piaskowski S, Hart MN, Sandor M, Fabry Z. Activated/effector CD4+ T cells exacerbate acute damage in the central nervous system following traumatic injury. J Neuroimmunol 2003; 136:54-66. [PMID: 12620643 DOI: 10.1016/s0165-5728(03)00008-0] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
CD4(+) helper T cells (Th) have been demonstrated to participate in the chronic phase of traumatic injury repair in the central nervous system (CNS). Here, we show that CD4(+) T cells can also contribute to the severity of the acute phase of CNS traumatic injury. We compared the area of tissue damage and the level of cellular apoptosis in aseptic cerebral injury (ACI) sites of C57BL/6 wild type and RAG1(-/-) immunodeficient mice. We demonstrate that ACI is attenuated in RAG1(-/-) mice compared to C57BL/6 animals. Adoptive transfer of CD4(+)CD62L(low)CD44(high) activated/effector T cells 24 h prior to ACI into RAG1(-/-) mice resulted in a significantly enhanced acute ACI that was comparable to ACI in the C57BL/6 animals. Adoptive transfer of CD4(+)CD62L(high)CD44(low) naive/non-activated T cells did not increase ACI in the brains of RAG1(-/-) mice. T cell inhibitory agents, cyclosporin A (CsA) and FK506, significantly decreased ACI-induced acute damage in C57BL/6 mice. These results suggest a previously undescribed role for activated/effector CD4(+) T cells in exacerbating ACI-induced acute damage in the CNS and raise a novel possibility for acute treatment of sterile traumatic brain injury.
Collapse
Affiliation(s)
- Dominic Fee
- Department of Neurology, University of Wisconsin-Madison, 53706, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
Unlike most bodily organs, the central nervous system (CNS) exists behind a blood-tissue barrier designed to minimize the passage of cells and macromolecules into the neural parenchyma. Yet, the CNS is routinely and effectively surveyed by the immune system. This review examines the mechanisms and participants in this immunological surveillance mechanism. The nature of the healthy blood-brain barrier, factors modifying it, and its central position in determining the number and nature of leukocytes permitted to enter, are considered. In addition the role in surveillance played by lymphatic drainage, migrating T and B lymphocytes, and elements of the monocyte/macrophage/microglia family are considered. While all these participants are known to be important in responding to a CNS antigen and/or establishing a site of inflammation in the nervous system, they also are major elements in maintaining the homeostasis of the CNS and permitting the necessary immunological surveillance of that organ.
Collapse
Affiliation(s)
- W F Hickey
- Department of Pathology, Dartmouth Medical School, Lebanon, New Hampshire 03756, USA
| |
Collapse
|
26
|
Abstract
Characterising the factors that control the entry of leucocytes into tissue in response to inflammatory or microbial insult continues to generate considerable interest. Of all the tissues studied it is probably that of the CNS which is the most fascinating because of the specialised properties of its blood vessel walls, which constitute the blood-brain barrier (BBB). In health, very few leucocytes penetrate the BBB but in disorders such as MS the barrier becomes compromised with the result that there is an intense infiltration of the CNS by T lymphocytes whose subsequent activity appears to underlie the onset and progression of disease. The purpose of this article is to summarise and assess recent literature pertaining to how lymphocytes bind to cerebral endothelial cells, migrate across the blood vessel walls and enter the CNS parenchyma. Particular emphasis is devoted to the cellular and molecular aspects of these events and addressing the questions of whether certain subsets of circulating T lymphocytes are more favourably disposed than others to CNS infiltration and whether entry is dependent upon the initial expression of distinct groups of adhesion molecules and upon the generation of chemotactic factors. This article also focuses upon identifying the key stages of lymphocyte migration across the BBB and their susceptibility to antagonism by therapeutic agents. It is intended that the review will provide a useful source of information and offer additional insights into the mechanisms controlling lymphocyte passage across the BBB during pathological disturbance.
Collapse
MESH Headings
- Animals
- Antigen Presentation
- Astrocytes/pathology
- Autoimmune Diseases/blood
- Autoimmune Diseases/immunology
- Autoimmune Diseases/pathology
- Blood-Brain Barrier
- Brain/blood supply
- Brain/immunology
- Callithrix
- Cell Adhesion
- Cell Adhesion Molecules/physiology
- Chemokines/physiology
- Chemotaxis, Leukocyte/drug effects
- Chemotaxis, Leukocyte/physiology
- Cytokines/physiology
- Drug Design
- Drug Evaluation, Preclinical
- Encephalomyelitis, Autoimmune, Experimental/blood
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Endothelium, Vascular/immunology
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Extracellular Matrix Proteins/physiology
- Forecasting
- Humans
- Immunologic Memory
- Immunophenotyping
- Immunosuppressive Agents/pharmacology
- Immunosuppressive Agents/therapeutic use
- Interferon-gamma/metabolism
- Interleukin-2/metabolism
- Lymphocyte Activation
- Metalloendopeptidases/physiology
- Mice
- Microcirculation
- Multiple Sclerosis/blood
- Multiple Sclerosis/immunology
- Multiple Sclerosis/pathology
- Myelin Sheath/immunology
- Myelin Sheath/pathology
- Rats
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocyte Subsets/pathology
Collapse
Affiliation(s)
- K A Brown
- Department of Immunobiology, Guy's Hospital, London, UK
| |
Collapse
|
27
|
Affiliation(s)
- P R Walker
- Laboratory of Tumor Immunology, Division of Oncology, University Hospital, Rue Micheli-du-Crest 24, 1211 Geneva 14, Switzerland.
| | | |
Collapse
|
28
|
Qing Z, Sandor M, Radvany Z, Sewell D, Falus A, Potthoff D, Muller WA, Fabry Z. Inhibition of antigen-specific T cell trafficking into the central nervous system via blocking PECAM1/CD31 molecule. J Neuropathol Exp Neurol 2001; 60:798-807. [PMID: 11487054 DOI: 10.1093/jnen/60.8.798] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Trafficking of antigen-specific T cells into the central nervous system (CNS) is an important initiating step in inflammation in the brain. In spite of the extensive knowledge about the role of adhesion molecules in T cell migration across peripheral vessels, the mechanism of the entry of antigen-specific T cells into the CNS is not known. This work was designed to study the regulatory roles of adhesion molecules in antigen-specific T cell migration into the CNS. Antigen-specific T cells were tracked in an in vivo migration assay using T cell receptor (TCR) transgenic mice having 95% of T cells specific for a defined antigen. pigeon cytochrome c (PCC). TCR transgenic mice were cannulated intraventricularly (IVT) for PCC antigen infusion and cerebrospinal fluid (CSF) sampling. Upon PCC infusion into the CNS, the number of alpha/beta TCR+ Vbeta3+ Mac1- cells in the CSF was characterized in the presence or absence of anti-adhesion molecule reagents. We found that antibodies against VCAM-1 (CD106), VLA-4 (CD49d/CD29), ICAM-1 (CD54), and LFA-1 (CD11a/CD18) did not influence the increased number of antigen-specific T cells in the CSF However, upon intravenous (i.v.) injection, anti-PECAM-1 (CD31) antibody or PECAM-Ig chimeric molecule inhibited the trafficking of alpha/beta TCR+ Vbeta3+ Mac1- cells into the CNS. The expression of PECAM-1 (CD31) was also up-regulated on antigen-specific T cells in a time-dependent manner in vitro upon antigenic stimulation. The antigen-induced activation of T cells in vivo was measured by CD44 and LFA-1 expression and found to be comparable between mPECAMIg-treated mice and wild-type serum control-treated groups. This indicates that CD31 inhibition of antigen-specific T cell accumulation in the CNS is probably not due to a functional inhibition of these cells. Finally, adoptive transfer of CFSE-labeled AND transgenic cells into naïve animals resulted in the accumulation of these cells in the CNS upon PCC IVT immunization that was also inhibited by mPECAMIg treatment. Hence, PECAM-1 (CD31) might play an important role in regulating antigen-specific T cells trafficking in CNS inflammatory diseases.
Collapse
Affiliation(s)
- Z Qing
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, 53706, USA
| | | | | | | | | | | | | | | |
Collapse
|