1
|
Taylor X, Cisternas P, You Y, You Y, Xiang S, Marambio Y, Zhang J, Vidal R, Lasagna-Reeves CA. A1 reactive astrocytes and a loss of TREM2 are associated with an early stage of pathology in a mouse model of cerebral amyloid angiopathy. J Neuroinflammation 2020; 17:223. [PMID: 32711525 PMCID: PMC7382050 DOI: 10.1186/s12974-020-01900-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 07/15/2020] [Indexed: 02/07/2023] Open
Abstract
Background Cerebral amyloid angiopathy (CAA) is typified by the cerebrovascular deposition of amyloid. The mechanisms underlying the contribution of CAA to neurodegeneration are not currently understood. Although CAA is highly associated with the accumulation of amyloid beta (Aβ), other amyloids are known to associate with the vasculature. Alzheimer’s disease (AD) is characterized by parenchymal Aβ deposition, intracellular accumulation of tau, and significant neuroinflammation. CAA increases with age and is present in 85–95% of individuals with AD. A substantial amount of research has focused on understanding the connection between parenchymal amyloid and glial activation and neuroinflammation, while associations between vascular amyloid pathology and glial reactivity remain understudied. Methods Here, we dissect the glial and immune responses associated with early-stage CAA with histological, biochemical, and gene expression analyses in a mouse model of familial Danish dementia (FDD), a neurodegenerative disease characterized by the vascular accumulation of Danish amyloid (ADan). Findings observed in this CAA mouse model were complemented with primary culture assays. Results We demonstrate that early-stage CAA is associated with dysregulation in immune response networks and lipid processing, severe astrogliosis with an A1 astrocytic phenotype, and decreased levels of TREM2 with no reactive microgliosis. Our results also indicate how cholesterol accumulation and ApoE are associated with vascular amyloid deposits at the early stages of pathology. We also demonstrate A1 astrocytic mediation of TREM2 and microglia homeostasis. Conclusion The initial glial response associated with early-stage CAA is characterized by the upregulation of A1 astrocytes without significant microglial reactivity. Gene expression analysis revealed that several AD risk factors involved in immune response and lipid processing may also play a preponderant role in CAA. This study contributes to the increasing evidence that brain cholesterol metabolism, ApoE, and TREM2 signaling are major players in the pathogenesis of AD-related dementias, including CAA. Understanding the basis for possible differential effects of glial response, ApoE, and TREM2 signaling on parenchymal plaques versus vascular amyloid deposits provides important insight for developing future therapeutic interventions.
Collapse
Affiliation(s)
- Xavier Taylor
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Neurosciences Research Building 214G, 320 West 15th Street, Indianapolis, IN, 46202, USA.,Department of Anatomy, Cell Biology, & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Pablo Cisternas
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Neurosciences Research Building 214G, 320 West 15th Street, Indianapolis, IN, 46202, USA.,Department of Anatomy, Cell Biology, & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Yanwen You
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Neurosciences Research Building 214G, 320 West 15th Street, Indianapolis, IN, 46202, USA.,Department of Anatomy, Cell Biology, & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Yingjian You
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Neurosciences Research Building 214G, 320 West 15th Street, Indianapolis, IN, 46202, USA.,Department of Anatomy, Cell Biology, & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Shunian Xiang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Yamil Marambio
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Neurosciences Research Building 214G, 320 West 15th Street, Indianapolis, IN, 46202, USA.,Department of Anatomy, Cell Biology, & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jie Zhang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Ruben Vidal
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Neurosciences Research Building 214G, 320 West 15th Street, Indianapolis, IN, 46202, USA.,Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Cristian A Lasagna-Reeves
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Neurosciences Research Building 214G, 320 West 15th Street, Indianapolis, IN, 46202, USA. .,Department of Anatomy, Cell Biology, & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
2
|
Harrington EP, Bergles DE, Calabresi PA. Immune cell modulation of oligodendrocyte lineage cells. Neurosci Lett 2019; 715:134601. [PMID: 31693930 DOI: 10.1016/j.neulet.2019.134601] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 10/25/2019] [Accepted: 10/28/2019] [Indexed: 01/02/2023]
Abstract
Chronic demyelination and the concomitant loss of trophic support and increased energy demands in axons are thought to contribute to neurodegeneration in a number of neurological diseases such as multiple sclerosis (MS). Adult oligodendrocyte precursor cells (OPCs) play an important role in these demyelinating diseases by generating new myelinating oligodendrocytes that may help limit axonal degeneration. Thus, promoting the differentiation of OPCs and functional integration of newly generated oligodendrocytes is a crucial avenue for the next generation of therapies. Evidence to date suggests that the immune system may both positively and negatively impact OPC differentiation and endogenous remyelination in disease. Inflammatory cytokines not only suppress OPC differentiation but may also directly affect other functions of OPCs. Recent studies have demonstrated that OPCs and oligodendrocytes in both human multiple sclerosis lesions and mouse models of demyelination can express an immunogenic transcriptional signature and upregulate antigen presenting genes. In inflammatory demyelinating mouse models OPCs are capable of presenting antigen and activating CD8 + T cells. Here we review the evidence for this new role of oligodendroglia as antigen presenting cells and how these inflammatory OPCs (iOPCs) and inflammatory oligodendrocytes (iOLs) may influence myelin repair and other disease processes.
Collapse
Affiliation(s)
- Emily P Harrington
- Department of Neurology, Johns Hopkins University School of Medicine, 600 N. Wolfe St., Pathology 509, Baltimore, MD, 21287, USA; The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, 725 N. Wolfe St., WBSB 1001, Baltimore, MD, 21205, USA
| | - Dwight E Bergles
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, 725 N. Wolfe St., WBSB 1001, Baltimore, MD, 21205, USA; The Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University School of Medicine, 600 N. Wolfe St., Pathology 509, Baltimore, MD, 21287, USA; The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, 725 N. Wolfe St., WBSB 1001, Baltimore, MD, 21205, USA.
| |
Collapse
|
3
|
Astrocyte Signaling in the Neurovascular Unit After Central Nervous System Injury. Int J Mol Sci 2019; 20:ijms20020282. [PMID: 30642007 PMCID: PMC6358919 DOI: 10.3390/ijms20020282] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 12/24/2018] [Accepted: 01/07/2019] [Indexed: 12/20/2022] Open
Abstract
Astrocytes comprise the major non-neuronal cell population in the mammalian neurovascular unit. Traditionally, astrocytes are known to play broad roles in central nervous system (CNS) homeostasis, including the management of extracellular ion balance and pH, regulation of neurotransmission, and control of cerebral blood flow and metabolism. After CNS injury, cell–cell signaling between neuronal, glial, and vascular cells contribute to repair and recovery in the neurovascular unit. In this mini-review, we propose the idea that astrocytes play a central role in organizing these signals. During CNS recovery, reactive astrocytes communicate with almost all CNS cells and peripheral progenitors, resulting in the promotion of neurogenesis and angiogenesis, regulation of inflammatory response, and modulation of stem/progenitor response. Reciprocally, changes in neurons and vascular components of the remodeling brain should also influence astrocyte signaling. Therefore, understanding the complex and interdependent signaling pathways of reactive astrocytes after CNS injury may reveal fundamental mechanisms and targets for re-integrating the neurovascular unit and augmenting brain recovery.
Collapse
|
4
|
Lively S, Lam D, Wong R, Schlichter LC. Comparing Effects of Transforming Growth Factor β1 on Microglia From Rat and Mouse: Transcriptional Profiles and Potassium Channels. Front Cell Neurosci 2018; 12:115. [PMID: 29780305 PMCID: PMC5946019 DOI: 10.3389/fncel.2018.00115] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 04/11/2018] [Indexed: 12/02/2022] Open
Abstract
The cytokine, transforming growth factor β1 (TGFβ1), is up-regulated after central nervous system (CNS) injuries or diseases involving microglial activation, and it has been proposed as a therapeutic agent for treating neuroinflammation. Microglia can produce and respond to TGFβ1. While rats and mice are commonly used for studying neuroinflammation, very few reports directly compare them. Such studies are important for improving pre-clinical studies and furthering translational progress in developing therapeutic interventions. After intracerebral hemorrhage (ICH) in the rat striatum, the TGFβ1 receptor was highly expressed on microglia/macrophages within the hematoma. We recently found species similarities and differences in response to either a pro-inflammatory (interferon-γ, IFN-γ, +tumor necrosis factor, TNF-α) or anti-inflammatory interleukin-4 (IL-4) stimulus. Here, we assessed whether rat and mouse microglia differ in their responses to TGFβ1. Microglia were isolated from Sprague-Dawley rats and C57BL/6 mice and treated with TGFβ1. We quantified changes in expression of >50 genes, in their morphology, proliferation, apoptosis and in three potassium channels that are considered therapeutic targets. Many inflammatory mediators, immune receptors and modulators showed species similarities, but notable differences included that, for some genes, only one species responded (e.g., Il4r, Il10, Tgfbr2, colony-stimulating factor receptor (Csf1r), Itgam, suppressor of cytokine signaling 1 (Socs1), toll-like receptors 4 (Tlr4), P2rx7, P2ry12), and opposite responses were seen for others (Tgfb1, Myc, Ifngr1). In rat only, TGFβ1 affected microglial morphology and proliferation, but there was no apoptosis in either species. In both species, TGFβ1 dramatically increased Kv1.3 channel expression and current (no effects on Kir2.1). KCa3.1 showed opposite species responses: the current was low in unstimulated rat microglia and greatly increased by TGFβ1 but higher in control mouse cells and decreased by TGFβ1. Finally, we compared TGFβ1 and IL10 (often considered similar anti-inflammatory stimuli) and found many different responses in both species. Overall, the numerous species differences should be considered when characterizing neuroinflammation and microglial activation in vitro and in vivo, and when targeting potassium channels.
Collapse
Affiliation(s)
- Starlee Lively
- Krembil Research Institute, Genes and Development Division, University Health Network, Toronto, ON, Canada
| | - Doris Lam
- Krembil Research Institute, Genes and Development Division, University Health Network, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Raymond Wong
- Krembil Research Institute, Genes and Development Division, University Health Network, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Lyanne C Schlichter
- Krembil Research Institute, Genes and Development Division, University Health Network, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
5
|
Menzel F, Kaiser N, Haehnel S, Rapp F, Patties I, Schöneberg N, Haimon Z, Immig K, Bechmann I. Impact of X-irradiation on microglia. Glia 2017; 66:15-33. [DOI: 10.1002/glia.23239] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 09/05/2017] [Accepted: 09/18/2017] [Indexed: 12/20/2022]
Affiliation(s)
| | - Nicole Kaiser
- Institute of Anatomy, Leipzig University; Leipzig Germany
| | - Susann Haehnel
- Institute of Anatomy, Leipzig University; Leipzig Germany
| | - Felicitas Rapp
- Institute of Anatomy, Leipzig University; Leipzig Germany
| | - Ina Patties
- Department of Radiation Therapy; Leipzig University; Leipzig Germany
| | | | - Zhana Haimon
- Department of Immunology; Weizmann Institute of Science; Rehovot Israel
| | - Kerstin Immig
- Institute of Anatomy, Leipzig University; Leipzig Germany
| | - Ingo Bechmann
- Institute of Anatomy, Leipzig University; Leipzig Germany
| |
Collapse
|
6
|
Westmoreland SV, Rosen J, MacKey J, Romsey C, Xia DL, Visvesvera GS, Mansfield KG. Necrotizing Meningoencephalitis and Pneumonitis in a Simian Immunodeficiency Virus—infected Rhesus Macaque due to Acanthamoeba. Vet Pathol 2016; 41:398-404. [PMID: 15232140 DOI: 10.1354/vp.41-4-398] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Free-living amoebae of the genus Acanthamoeba can cause a fatal disease of the brain in humans called granulomatous amoebic encephalitis. We present a case of meningoencephalitis and pneumonitis in a simian immunodeficiency virus (SIV)-infected rhesus macaque caused by Acanthamoeba sp. The animal became ill 176 days after intravenous inoculation with SIVmac251 after a short history of weight loss and a sudden onset of hind limb paresis and abnormal head movements. Histopathologic examination of hematoxylin and eosin-stained tissues revealed multifocal to coalescing necrotizing neutrophilic meningoencephalitis and pneumonitis. Immunofluorescence and polymerase chain reaction were used to identify the genus of amoeba as Acanthamoeba. Immunohistochemistry of immune cell markers was used to characterize the animal's immune response to the opportunistic amoebic infection with features of both innate and adaptive cell-mediated immunity. Although not previously reported, the potential transmission to humans, either through environmental contamination or contact with an infected animal, makes this disease a threat to laboratory animal care staff and pathologists.
Collapse
Affiliation(s)
- S V Westmoreland
- Division of Comparative Pathology, New England Primate Research Center, Harvard Medical School, One Pine Hill Drive, Southborough, MA 01772, USA.
| | | | | | | | | | | | | |
Collapse
|
7
|
Caravagna C, Jaouën A, Debarbieux F, Rougon G. Overview of Innovative Mouse Models for Imaging Neuroinflammation. ACTA ACUST UNITED AC 2016; 6:131-147. [PMID: 27248431 DOI: 10.1002/cpmo.5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Neuroinflammation demands a comprehensive appraisal in situ to gain in-depth knowledge on the roles of particular cells and molecules and their potential roles in therapy. Because of the lack of appropriate tools, direct visualization of cells has been poorly investigated up to the present. In this context, reporter mice expressing cell-specific fluorescent proteins, combined with multiphoton microscopy, provide a window into cellular processes in living animals. In addition, the ability to collect multiple fluorescent colors from the same sample makes in vivo microscopy uniquely useful for characterizing many parameters from the same area, supporting powerful correlative analyses. Here, we present an overview of the advantages and limitations of this approach, with the purpose of providing insight into the neuroinflammation field. We also provide a review of existing fluorescent mouse models and describe how these models have been used in studies of neuroinflammation. Finally, the potential for developing advanced genetic tools and imaging resources is discussed. © 2016 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Céline Caravagna
- Aix Marseille Université, CNRS, Institut de Neurosciences de la Timone, Marseille, France.,Aix Marseille Université, Centre Européen de Recherche en Imagerie Médicale, Marseille, France
| | - Alexandre Jaouën
- Aix Marseille Université, CNRS, Institut de Neurosciences de la Timone, Marseille, France.,Aix Marseille Université, Centre Européen de Recherche en Imagerie Médicale, Marseille, France
| | - Franck Debarbieux
- Aix Marseille Université, CNRS, Institut de Neurosciences de la Timone, Marseille, France.,Aix Marseille Université, Centre Européen de Recherche en Imagerie Médicale, Marseille, France.,These authors contributed equally to this work
| | - Geneviève Rougon
- Aix Marseille Université, CNRS, Institut de Neurosciences de la Timone, Marseille, France.,Aix Marseille Université, Centre Européen de Recherche en Imagerie Médicale, Marseille, France.,These authors contributed equally to this work
| |
Collapse
|
8
|
Immig K, Gericke M, Menzel F, Merz F, Krueger M, Schiefenhövel F, Lösche A, Jäger K, Hanisch UK, Biber K, Bechmann I. CD11c-positive cells from brain, spleen, lung, and liver exhibit site-specific immune phenotypes and plastically adapt to new environments. Glia 2014; 63:611-25. [DOI: 10.1002/glia.22771] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 11/06/2014] [Indexed: 12/23/2022]
Affiliation(s)
- Kerstin Immig
- Institute of Anatomy, Leipzig University; Leipzig Germany
| | - Martin Gericke
- Institute of Anatomy, Leipzig University; Leipzig Germany
| | | | - Felicitas Merz
- Institute of Anatomy, Leipzig University; Leipzig Germany
| | - Martin Krueger
- Institute of Anatomy, Leipzig University; Leipzig Germany
| | | | - Andreas Lösche
- IZKF-FACS-Core Unit; Leipzig University; Leipzig Germany
| | - Kathrin Jäger
- IZKF-FACS-Core Unit; Leipzig University; Leipzig Germany
| | | | - Knut Biber
- Department of Psychiatry and Psychotherapy; Section of Molecular Psychiatry, University of Freiburg; Freiburg Germany
| | - Ingo Bechmann
- Institute of Anatomy, Leipzig University; Leipzig Germany
| |
Collapse
|
9
|
Martínez-Canabal A. Potential neuroprotective role of transforming growth factor β1 (TGFβ1) in the brain. Int J Neurosci 2014; 125:1-9. [PMID: 24628581 DOI: 10.3109/00207454.2014.903947] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
TGFβ1 is a growth factor that is known to be expressed in most neurodegenerative diseases and after vascular accidents in the brain. TGFβ1 downregulates the activity of activated microglia and promotes astrogliosis. It also prevents cell death by a known mechanism dependant on astrocytes and the secretion of the plasminogen activator inhibitor 1 (PAI-1). This mechanism can provide light on what is the mechanism of action of TGFβ1 as a protective factor and it can provide the pharmacological principles in which this pathway could be used with therapeutic purposes. TGFβ1 is upregulated in most neurodegenerative diseases, however, its expression appears dramatically blocked in Huntington's disease, the fastest of those diseases in progress after the onset. This fact suggests that TGFβ1 slows down the neurodegenerative process, preventing tissue damage and neural apoptotic death. However, the exact details of TGFβ1 action are still unknown and the physiological roles on the diseases are still mysterious. Interestingly, all the data regarding the roles of TGFβ1 in health and disease have been also confirmed with the use of transgenic knockouts and TGFβ1 overexpressing mice. What possibly came as a surprise from the study of TGFβ1 overexpressing models is that combining its neuroprotective and antiproliferative effects, this cytokine generates a significant disruption in the hippocampal circuitry with its consequent learning and memory deficit.
Collapse
Affiliation(s)
- Alonso Martínez-Canabal
- Department of Molecular Neuropathology, Cell Physiology Institute (IFC), Department of Cell Biology, Faculty of Sciences, National Autonomous University of Mexico (UNAM). Ciudad Universitaria, Circuito exterior S/N, Coyoacan, 04510 Mexico D.F. Mexico
| |
Collapse
|
10
|
Acevedo G, Padala NK, Ni L, Jonakait GM. Astrocytes inhibit microglial surface expression of dendritic cell-related co-stimulatory molecules through a contact-mediated process. J Neurochem 2013; 125:575-87. [PMID: 23439211 DOI: 10.1111/jnc.12221] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2013] [Revised: 02/08/2013] [Accepted: 02/08/2013] [Indexed: 12/25/2022]
Abstract
Murine microglia cultured in isolation were treated sequentially with granulocyte/monocyte colony-stimulating factor (GM-CSF) (5 days) and lipopolysaccharide (LPS) (2 days) to elicit a mature dendritic cell-like (DC-like) phenotype. Examined by flow cytometry microglia thus isolated show high surface expression of CD11c together with the co-stimulatory molecules CD40, CD80, and CD86 that are necessary for T-cell activation. In contrast, microglia co-cultured with astrocytes fail to achieve a mature DC-like phenotype. Contact with the astrocytic environment is necessary for the inhibition. Failure was not because of a more rapid degradation of protein. Bone marrow-derived cells, like microglia, were prevented by astrocytes from attaining a mature DC phenotype. Although GM-CSF pre-treatment substantially increases mRNA of co-stimulatory molecules and major histocompatibility complex (MHC) Class II in isolated microglia, co-cultured microglia await treatment with LPS to up-regulate them. In contrast, western blot and immunocytochemical analysis revealed that it is not a failure of transcription or translation, nor is it a more rapid degradation of mRNA that is responsible for the low surface expression; rather microglia co-cultured with astrocytes produce mRNA and protein but do not traffic the protein onto the cell surface.
Collapse
|
11
|
Rodgers JM, Miller SD. Cytokine control of inflammation and repair in the pathology of multiple sclerosis. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2012; 85:447-68. [PMID: 23239947 PMCID: PMC3516888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Cytokines are secreted signaling proteins that play an essential role in propagating and regulating immune responses during experimental autoimmune encephalomyelitis (EAE), the mouse model of the neurodegenerative, autoimmune disease multiple sclerosis (MS). EAE pathology is driven by a myelin-specific T cell response that is activated in the periphery and mediates the destruction of myelin upon T cell infiltration into the central nervous system (CNS). Cytokines provide cell signals both in the immune and CNS compartment, but interestingly, some have detrimental effects in the immune compartment while having beneficial effects in the CNS compartment. The complex nature of these signals will be reviewed.
Collapse
Affiliation(s)
- Jane M. Rodgers
- Department of Microbiology-Immunology, Feinberg School of Medicine,
Northwestern University, Chicago, Illinois
| | - Stephen D. Miller
- Department of Microbiology-Immunology, Feinberg School of Medicine,
Northwestern University, Chicago, Illinois
| |
Collapse
|
12
|
Abstract
Recent research has overcome the old paradigms of the brain as an immunologically privileged organ, and of the exclusive role of neurotransmitters and neuropeptides as signal transducers in the central nervous system. Growing evidence suggests that the signal proteins of the immune system - the cytokines - are also involved in modulation of behavior and induction of psychiatric symptoms. This article gives an overview on the nature of cytokines and the proposed mechanisms of immune-to-brain interaction. The role of cytokines in psychiatric symptoms, syndromes, and disorders like sickness behavior, major depression, and schizophrenia are discussed together with recent immunogenetic findings.
Collapse
Affiliation(s)
- Markus J Schwarz
- Psychiatric Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| |
Collapse
|
13
|
Costello DA, Lyons A, Denieffe S, Browne TC, Cox FF, Lynch MA. Long term potentiation is impaired in membrane glycoprotein CD200-deficient mice: a role for Toll-like receptor activation. J Biol Chem 2011; 286:34722-32. [PMID: 21835925 PMCID: PMC3186410 DOI: 10.1074/jbc.m111.280826] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Indexed: 12/26/2022] Open
Abstract
The membrane glycoprotein CD200 is expressed on several cell types, including neurons, whereas expression of its receptor, CD200R, is restricted principally to cells of the myeloid lineage, including microglia. The interaction between CD200 and CD200R maintains microglia and macrophages in a quiescent state; therefore, CD200-deficient mice express an inflammatory phenotype exhibiting increased macrophage or microglial activation in models of arthritis, encephalitis, and uveoretinitis. Here, we report that lipopolysaccharide (LPS) and Pam(3)CysSerLys(4) exerted more profound effects on release of the proinflammatory cytokines, interleukin (IL)-1β, IL-6, and tumor necrosis factor-α (TNFα), in glia prepared from CD200(-/-) mice compared with wild type mice. This effect is explained by the loss of CD200 on astrocytes, which modulates microglial activation. Expression of Toll-like receptors 4 and 2 (TLR4 and -2) was increased in glia prepared from CD200(-/-) mice, and the evidence indicates that microglial activation, assessed by the increased numbers of CD11b(+) cells that stained positively for both MHCII and CD40, was enhanced in CD200(-/-) mice compared with wild type mice. These neuroinflammatory changes were associated with impaired long term potentiation (LTP) in CA1 of hippocampal slices prepared from CD200(-/-) mice. One possible explanation for this is the increase in TNFα in hippocampal tissue prepared from CD200(-/-) mice because TNFα application inhibited LTP in CA1. Significantly, LPS and Pam(3)CysSerLys(4), at concentrations that did not affect LTP in wild type mice, inhibited LTP in slices prepared from CD200(-/-) mice, probably due to the accompanying increase in TLR2 and TLR4. Thus, the neuroinflammatory changes that result from CD200 deficiency have a negative impact on synaptic plasticity.
Collapse
Affiliation(s)
- Derek A Costello
- Department of Physiology and the Trinity College Institute of Neuroscience, Trinity College, Dublin 2, Ireland.
| | | | | | | | | | | |
Collapse
|
14
|
Grace PM, Rolan PE, Hutchinson MR. Peripheral immune contributions to the maintenance of central glial activation underlying neuropathic pain. Brain Behav Immun 2011; 25:1322-32. [PMID: 21496480 DOI: 10.1016/j.bbi.2011.04.003] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Revised: 04/04/2011] [Accepted: 04/04/2011] [Indexed: 10/18/2022] Open
Abstract
Recent evidence implicates an adaptive immune response in the central nervous system (CNS) mechanisms of neuropathic pain. This review identifies how neuropathic pain alters CNS immune privilege to facilitate T cell infiltration. Once in the CNS, T cells may interact with the local antigen presenting cells, microglia, via the major histocompatibility complex and the costimulatory molecules CD40 and B7. In this way, T cells may contribute to the maintenance of neuropathic pain through pro-inflammatory interactions with microglia and by facilitating the activation of astrocytes in the spinal dorsal horn. Based on the evidence presented in this review, we suggest that this bidirectional, pro-inflammatory system of neurons, glia and T cells in neuropathic pain should be renamed the pentapartite synapse, and identifies the latest member as a potential disease-modifying therapeutic target.
Collapse
Affiliation(s)
- Peter M Grace
- Discipline of Pharmacology, School of Medical Sciences, University of Adelaide, Adelaide, SA 5005, Australia.
| | | | | |
Collapse
|
15
|
Huang YH, Airas L, Schwab N, Wiendl H. Janus head: the dual role of HLA-G in CNS immunity. Cell Mol Life Sci 2011; 68:407-16. [PMID: 21086150 PMCID: PMC11114849 DOI: 10.1007/s00018-010-0582-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2010] [Accepted: 10/22/2010] [Indexed: 10/25/2022]
Abstract
The central nervous system (CNS) is considered an immune-privileged organ that maintains an adaptable immune surveillance system. Dysregulated immune function within the CNS contributes to the development of brain tumor growth, and robust immune activation results in excessive inflammation. Human lymphocyte antigen-G (HLA-G) proteins with tolerogenic immunoreactivity have been implicated in various pathophysiological processes including immune surveillance, governing homeostasis and immune regulation. In this review, we describe the wealth of evidence for the involvement of HLA-G in the CNS under physiological and pathological conditions. Further, we review regulatory functions that may be applicable as beneficial strategies in the therapeutic manipulation of immune-mediated CNS immune responses. Additionally, we try to understand how this molecule cooperates with other CNS-resident cells to maintain normal immune homeostasis, while still facilitating the development of the appropriate immune responses.
Collapse
Affiliation(s)
- Yu-Hwa Huang
- Department of Neurology, Inflammatory Disorders of the Nervous System and Neurooncology, University of Müenster, Domagkstr. 13, 48149 Müenster, Germany
| | - Laura Airas
- Department of Neurology, MediCity Research Laboratory, Turku University Hospital, Turku, Finland
| | - Nicholas Schwab
- Department of Neurology, Inflammatory Disorders of the Nervous System and Neurooncology, University of Müenster, Domagkstr. 13, 48149 Müenster, Germany
| | - Heinz Wiendl
- Department of Neurology, Inflammatory Disorders of the Nervous System and Neurooncology, University of Müenster, Domagkstr. 13, 48149 Müenster, Germany
| |
Collapse
|
16
|
Chastain EML, Duncan DS, Rodgers JM, Miller SD. The role of antigen presenting cells in multiple sclerosis. BIOCHIMICA ET BIOPHYSICA ACTA 2011; 1812:265-74. [PMID: 20637861 PMCID: PMC2970677 DOI: 10.1016/j.bbadis.2010.07.008] [Citation(s) in RCA: 188] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2010] [Revised: 07/06/2010] [Accepted: 07/07/2010] [Indexed: 12/25/2022]
Abstract
Multiple sclerosis (MS) is a debilitating T cell mediated autoimmune disease of the central nervous system (CNS). Animal models of MS, such as experimental autoimmune encephalomyelitis (EAE) and Theiler's murine encephalomyelitis virus-induced demyelinating disease (TMEV-IDD) have given light to cellular mechanisms involved in the initiation and progression of this organ-specific autoimmune disease. Within the CNS, antigen presenting cells (APC) such as microglia and astrocytes participate as first line defenders against infections or inflammation. However, during chronic inflammation they can participate in perpetuating the self-destructive environment by secretion of inflammatory factors and/or presentation of myelin epitopes to autoreactive T cells. Dendritic cells (DC) are also participants in the presentation of antigen to T cells, even within the CNS. While the APCs alone are not solely responsible for mediating the destruction to the myelin sheath, they are critical players in perpetuating the inflammatory milieu. This review will highlight relevant studies which have provided insight to the roles played by microglia, DCs and astrocytes in the context of CNS autoimmunity.
Collapse
Affiliation(s)
- Emily M L Chastain
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | | | | |
Collapse
|
17
|
Lynch AM, Murphy KJ, Deighan BF, O'Reilly JA, Gun'ko YK, Cowley TR, Gonzalez-Reyes RE, Lynch MA. The impact of glial activation in the aging brain. Aging Dis 2010; 1:262-278. [PMID: 22396865 PMCID: PMC3295033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2010] [Revised: 09/01/2010] [Accepted: 09/02/2010] [Indexed: 05/31/2023] Open
Abstract
The past decade or so has witnessed a rekindling of interest in glia requiring a re-evaluation of the early descriptions of astrocytes as merely support cells, and microglia as adopting either a resting state or an activated state in a binary fashion. We now know that both cell types contribute to the optimal functioning of neurons in the healthy brain, and that altered function of either cell impacts on neuronal function and consequently cognitive function. The evidence indicates that both astrocytic and microglial phenotype change with age and that the shift from the resting state is associated with deterioration in synaptic function. In this review, we consider the rapidly-expanding array of functions attributed to these cells and focus on evaluating the changes in cell activation that accompany ageing.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Marina A. Lynch
- Correspondence should be addressed to: Dr. MA Lynch, Trinity College Institute of Neuroscience, Trinity College, Dublin 2, Ireland.
| |
Collapse
|
18
|
Becker KJ. Sensitization and tolerization to brain antigens in stroke. Neuroscience 2008; 158:1090-7. [PMID: 18706487 DOI: 10.1016/j.neuroscience.2008.07.027] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2008] [Revised: 07/14/2008] [Accepted: 07/16/2008] [Indexed: 12/22/2022]
Abstract
Despite encounter of novel brain antigens by the systemic immune system following stroke, autoimmune responses to these antigens do not seem to occur. In rats, a systemic inflammatory response at the time of stroke, however, provokes changes that increase the likelihood of developing detrimental autoimmunity. These findings may help to explain why infections in the post-stroke period are associated with worse outcome. In addition, data suggest that the immune response can be manipulated in an antigen specific fashion to improve stroke outcome. Together these data argue that the nature of the post-ischemic immune response influences neurological recovery from stroke.
Collapse
Affiliation(s)
- K J Becker
- University of Washington School of Medicine, Harborview Medical Center, Box 359775, 325 9th Avenue, Seattle, WA 98104-2499, USA.
| |
Collapse
|
19
|
Immunosuppression after traumatic or ischemic CNS damage: it is neuroprotective and illuminates the role of microglial cells. Prog Neurobiol 2007; 84:211-33. [PMID: 18262323 DOI: 10.1016/j.pneurobio.2007.12.001] [Citation(s) in RCA: 125] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2007] [Revised: 11/15/2007] [Accepted: 12/11/2007] [Indexed: 01/08/2023]
Abstract
Acute traumatic and ischemic events in the central nervous system (CNS) invariably result in activation of microglial cells as local representatives of the immune system. It is still under debate whether activated microglia promote neuronal survival, or whether they exacerbate the original extent of neuronal damage. Protagonists of the view that microglial cells cause secondary damage have proposed that inhibition of microglial activation by immunosuppression is beneficial after acute CNS damage. It is the aim of this review to analyse the effects of immunosuppressants on isolated microglial cells and neurons, and to scrutinize the effects of immunosuppression in different in vivo models of acute CNS trauma or ischemia. It is found that the immunosuppressants cytosine-arabinoside, different steroids, cyclosporin A, FK506, rapamycin, mycophenolate mofetil, and minocycline all have direct inhibitory effects on microglial cells. These effects are mainly exerted by inhibiting microglial proliferation or microglial secretion of neurotoxic substances such as proinflammatory cytokines and nitric oxide. Furthermore, immunosuppression after acute CNS trauma or ischemia results in improved structure preservation and, mostly, in enhanced function. However, all investigated immunosuppressants also have direct effects on neurons, and some immunosuppressants affect other glial cells such as astrocytes. In summary, it is safe to conclude that immunosuppression after acute CNS trauma or ischemia is neuroprotective. Furthermore, circumferential evidence indicates that microglial activation after traumatic or ischemic CNS damage is not beneficial to adjacent neurons in the immediate aftermath of such acute lesions. Further experiments with more specific agents or genetic approaches that specifically inhibit microglial cells are needed in order to fully answer the question of whether microglial activation is "good or bad".
Collapse
|
20
|
Abstract
A major causative factor in the paralysis that often follows an acute injury to the central nervous system (CNS) is the paradoxical inability of the CNS to tolerate its own mechanism of self-repair. The dismal result is often a wider spread of damage (part of the inevitable "secondary" or "delayed" degeneration) rather than contribution toward a cure. Ever since the phenomenon of posttraumatic damage spread in the CNS was first recognized, neuroscientists have attempted to identify the players in this destructive process and have sought ways to neutralize or bypass them with the object of rescuing any neurons that are still viable. This approach is collectively termed neuroprotection. In this chapter, we present a view of experimental paradigms used to study neuroprotection.
Collapse
Affiliation(s)
- Michal Schwartz
- Department of Neurobiology, The Weizmann Institute of Science, Rehovot, Israel
| | | |
Collapse
|
21
|
Miguel-Hidalgo JJ, Nithuairisg S, Stockmeier C, Rajkowska G. Distribution of ICAM-1 immunoreactivity during aging in the human orbitofrontal cortex. Brain Behav Immun 2007; 21:100-11. [PMID: 16824729 PMCID: PMC2921168 DOI: 10.1016/j.bbi.2006.05.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2006] [Revised: 04/19/2006] [Accepted: 05/09/2006] [Indexed: 10/24/2022] Open
Abstract
Neurological and psychiatric alterations during aging are associated with increased cerebrovascular disturbances and inflammatory markers such as Intercellular Adhesion Molecule-1 (ICAM-1). We investigated whether the distribution of ICAM-1 immunoreactivity (ICAM-1-I) in histological sections from the left orbitofrontal cortex (ORB) was altered during normal aging. Postmortem tissue from the ORB of nine younger (27-54 years old) and 10 older (60-86) human subjects was collected. Cryostat sections were immunostained only with antibodies to ICAM-1 or together with an antibody to glial fibrillary acidic protein (GFAP). The total area fraction of ICAM-1-I, and the fraction of vascular and extravascular ICAM-1-I were quantified in the gray matter. Furthermore, we examined the association of extravascular ICAM-1-I to GFAP immunoreactive (GFAP-IR) astrocytes. In all subjects, brain blood vessels were similarly ICAM-1 immunoreactive, and in some subjects there was a variable number of extravascular patches of ICAM-1-I. The area fraction of ICAM-1-I was 120% higher (p<.0001) in the old subjects than in the young subjects. This increase localized mostly to the extravascular ICAM-1-I in register with GFAP-IR astrocytes. A much smaller, also age-dependent increase occurred in vascular ICAM-1-I. Our results indicate a dramatic increase in extravascular ICAM-1-I associated to GFAP-IR astrocytes in the ORB in normal aging. This increase may contribute to an enhanced risk for brain inflammatory processes during aging, although a role of extravascular ICAM-1 as a barrier to further inflammation cannot be ruled out.
Collapse
Affiliation(s)
- Jose Javier Miguel-Hidalgo
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216, USA.
| | | | | | | |
Collapse
|
22
|
Mutlu L, Brandt C, Kwidzinski E, Sawitzki B, Gimsa U, Mahlo J, Aktas O, Nitsch R, van Zwam M, Laman JD, Bechmann I. Tolerogenic effect of fiber tract injury: reduced EAE severity following entorhinal cortex lesion. Exp Brain Res 2006; 178:542-53. [PMID: 17091291 DOI: 10.1007/s00221-006-0758-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2006] [Accepted: 10/10/2006] [Indexed: 12/25/2022]
Abstract
Despite transient, myelin-directed adaptive immune responses in regions of fiber tract degeneration, none of the current models of fiber tract injuries evokes disseminated demyelination, implying effective mechanisms maintaining or re-establishing immune tolerance. In fact, we have recently detected CD95L upregulation accompanied by apoptosis of leukocytes in zones of axonal degeneration induced by entorhinal cortex lesion (ECL), a model of layer-specific axonal degeneration. Moreover, infiltrating monocytes readily transformed into ramified microglia exhibiting a phenotype of immature (CD86+/CD80-) antigen-presenting cells. We now report the appearance of the axonal antigen neurofilament-light along with increased T cell apoptosis and enhanced expression of the pro-apoptotic gene Bad in cervical lymph nodes after ECL. In order to test the functional significance of such local and systemic depletory/regulatory mechanisms on subsequent immunity to central nervous system antigens, experimental autoimmune encephalomyelitis was induced by proteolipid protein immunization 30 days after ECL. In three independent experiments, we found significantly diminished disease scores and infiltrates in lesioned compared to sham-operated SJL mice. This is consistent with a previous meta-statistical analysis (Goodin et al. in Neurology 52:1737-1745, 1999) rejecting the O-hypothesis that brain trauma causes or exacerbates multiple sclerosis. Conversely, brain injuries may involve long-term tolerogenic effects towards brain antigens.
Collapse
Affiliation(s)
- Leman Mutlu
- Institute of Cell Biology and Neurobiology, Charité, 10098, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Schmitt KRL, Kern C, Berger F, Ullrich O, Hendrix S, Abdul-Khaliq H. Methylprednisolone attenuates hypothermia- and rewarming-induced cytotoxicity and IL-6 release in isolated primary astrocytes, neurons and BV-2 microglia cells. Neurosci Lett 2006; 404:309-14. [PMID: 16860472 DOI: 10.1016/j.neulet.2006.05.064] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2006] [Revised: 04/18/2006] [Accepted: 05/27/2006] [Indexed: 10/24/2022]
Abstract
Brain protection is crucial during neonatal and pediatric cardiac surgery. The major methods for brain protection are the administration of steroids and deep hypothermia. Therefore, we have investigated the impact of methylprednisolone (MP) administration and deep hypothermia on neonatal mouse astrocytes, neurons and BV-2 microglia cells. Brain cells were pretreated with MP (100 mM) and incubated according to a deep hypothermia protocol mimicking temperature changes during cardiac surgery in children: deep hypothermia (2 h at 17 degrees C, phase 1), slow rewarming (2 h up to 37 degrees C, phase 2), and normothermia (20 h at 37 degrees C, phase 3). In all brain-related cell types cytotoxicity was investigated as well as the release of the pro-inflammatory cytokine interleukin-6 (IL-6), which plays a major role in neuroprotection and neuroregeneration. Deep hypothermia induces substantial cytotoxicity and the secretion of IL-6 by astrocytes, BV-2 microglia cells and neurons. MP administration has no influence on the cell survival and IL-6 release of normothermic astrocytes, BV-2 microglia cells and neurons, while hypothermia-induced cytotoxicity and IL-6 secretion are significantly suppressed by MP. These data suggest that MP increases cell survival after deep hypothermia but also suppresses important neuroprotective and regenerative processes induced by IL-6. Hence, more specific immune modulation than that provided by MP may be needed to protect the brain during neonatal and pediatric cardiac surgery.
Collapse
Affiliation(s)
- Katharina R L Schmitt
- Department for Congenital Heart Disease and Pediatric Cardiology, German Heart Institute Berlin, Deutsches Herzzentrum Berlin, Augustenburger Platz 1, 13353 Berlin, Germany.
| | | | | | | | | | | |
Collapse
|
24
|
Becher B, Bechmann I, Greter M. Antigen presentation in autoimmunity and CNS inflammation: how T lymphocytes recognize the brain. J Mol Med (Berl) 2006; 84:532-43. [PMID: 16773356 DOI: 10.1007/s00109-006-0065-1] [Citation(s) in RCA: 165] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2005] [Accepted: 03/02/2006] [Indexed: 12/23/2022]
Abstract
The central nervous system (CNS) is traditionally viewed as an immune privileged site in which overzealous immune cells are prevented from doing irreparable damage. It was believed that immune responses occurring within the CNS could potentially do more damage than the initial pathogenic insult itself. However, virtually every aspect of CNS tissue damage, including degeneration, tumors, infection, and of course autoimmunity, involves a significant cellular inflammatory component. While the blood-brain barrier (BBB) inhibits diffusion of hydrophilic (immune) molecules across brain capillaries, activated lymphocytes readily pass the endothelial layer of postcapillary venules without difficulty. In classic neuro-immune diseases such as multiple sclerosis or acute disseminated encephalomyelitis, it is thought that neuroantigen-reactive lymphocytes, which have escaped immune tolerance, now invade the CNS and are responsible for tissue damage, demyelination, and axonal degeneration. The developed animal model for these disorders, experimental autoimmune encephalomyelitis (EAE), reflects many aspects of the human conditions. Studies in EAE proved that auto-reactive encephalitogenic T helper (Th) cells are responsible for the onset of the disease. Th cells recognize their cognate antigen (Ag) only when presented by professional Ag-presenting cells in the context of major histocompatibility complex class II molecules. The apparent target structures of EAE immunity are myelinating oligodendrocytes, which are not capable of presenting Ag to invading encephalitogenic T cells. A compulsory third party is thus required to mediate between the attacking T cells and the myelin-expressing target. This review will discuss the recent advances in this field of research and we will discuss the journey of an auto-reactive T cell from its site of activation into perivascular spaces and further into the target tissue.
Collapse
Affiliation(s)
- Burkhard Becher
- Neurology Department, Division for Neuroimmunology, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland.
| | | | | |
Collapse
|
25
|
Bechmann I. Failed central nervous system regeneration: a downside of immune privilege? Neuromolecular Med 2006; 7:217-28. [PMID: 16247182 DOI: 10.1385/nmm:7:3:217] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2005] [Accepted: 07/07/2005] [Indexed: 12/25/2022]
Abstract
Immunity is required to eliminate dangerous or degenerated material and to support regeneration, but also causes significant parenchymal damage. In the eye and the brain, in which cornea and lens poorly regenerate and neurons are hardly replaceable, early transplantation experiments demonstrated remarkable tolerance to various grafts. This "immunologically privileged status" (Billingham and Boswell, 1953) may reflect evolutionary pressure to downmodulate certain actions of immune cells within particularly vulnerable tissues. As an example, tolerating certain "neurotrophic" viruses may often be a more successful strategy for survival than the elimination of all infected neurons. While several constitutive and inducible signals maintaining or re-establishing immune tolerance within the brain have been identified, it has also become evident that the resulting anti-inflammatory environment limits certain beneficial effects of neuroinflammation such as neurotrophin secretion or glutamate buffering by T-cells and the clearance of growth-inhibiting myelin or amyloid. Following spinal cord injury, the costs and benefits of neuroinflammation seem to come close because enhancing as well as suppressing innate or adaptive immunity caused amelioration and aggravation of functional regeneration in similar experiments. Evaluating such balances has also begun in (animal models of) Alzheimer's disease, central nervous system trauma, and stroke, and the appreciation of the beneficial side of neuroinflammation has caused a rethinking of the ill-defined use of immune suppressants. As dual roles for individual molecules have been recognized (Merrill and Benveniste, 1996), we are uncovering an already fine-tuned system, but the challenge remains to further support beneficial immune cascades without causing additional damage, and vice versa.
Collapse
Affiliation(s)
- Ingo Bechmann
- Center for Anatomy, Institute of Cell Biology and Neurobiology, Department of Experimental Neuroimmunology, Charité-Universitätsmedizin Berlin, Schumannstr, 20/21 10098 Berlin, Germany.
| |
Collapse
|
26
|
Kort JJ, Kawamura K, Fugger L, Weissert R, Forsthuber TG. Efficient presentation of myelin oligodendrocyte glycoprotein peptides but not protein by astrocytes from HLA-DR2 and HLA-DR4 transgenic mice. J Neuroimmunol 2006; 173:23-34. [PMID: 16386804 DOI: 10.1016/j.jneuroim.2005.11.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2005] [Accepted: 11/16/2005] [Indexed: 10/25/2022]
Abstract
The role of astrocytes in the pathogenesis of multiple sclerosis (MS) is not well understood. Astrocytes may modulate the activity of pathogenic T cells by presenting myelin antigens in combination with pro- or anti-inflammatory signals. Astrocytes have been shown to present myelin basic protein (MBP) and proteolipid protein (PLP) to T cells, but it has remained unresolved whether astrocytes present myelin oligodendrocyte glycoprotein (MOG), which has been implicated as an important autoantigen in MS. Here, we asked whether astrocytes presented MOG to T cells. To closer model presentation of human MOG by astrocytes in MS patients, we generated astrocytes from transgenic mice expressing the MS-associated MHC class II alleles HLA-DR2 (DRB1*1501) and HLA-DR4 (DRB1*0401). The results show that IFN-gamma-activated HLA-DR2 and HLA-DR4 expressing astrocytes efficiently presented immunodominant and subdominant MOG peptides to T cells. The hierarchy of the presented MOG epitopes was comparable to that of professional APCs, including dendritic cells and microglia. Importantly, astrocytes were poor at processing and presenting native MOG protein. Furthermore, astrocytes induced a mixed Th1/Th2 cytokine response in MOG-specific T cells, whereas dendritic cells induced a predominantly Th1 cell response. Collectively, the results suggest that astrocytes may modulate anti-MOG T cell responses in the CNS.
Collapse
Affiliation(s)
- Jens J Kort
- Institute of Pathology, School of Medicine, Case Western Reserve University, Cleveland, USA
| | | | | | | | | |
Collapse
|
27
|
Rasley A, Tranguch SL, Rati DM, Marriott I. Murine glia express the immunosuppressive cytokine, interleukin-10, following exposure toBorrelia burgdorferi orNeisseria meningitidis. Glia 2006; 53:583-92. [PMID: 16419089 DOI: 10.1002/glia.20314] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
There is growing appreciation that resident glial cells can initiate and/or regulate inflammation following trauma or infection in the central nervous system (CNS). We have previously demonstrated the ability of microglia and astrocytes, resident glial cells of the CNS, to respond to bacterial pathogens by rapid production of inflammatory mediators. However, inflammation within the brain parenchyma is notably absent during some chronic bacterial infections in humans and nonhuman primates. In the present study, we demonstrate the ability of the immunosuppressive cytokine, interleukin-10 (IL-10), to inhibit inflammatory immune responses of primary microglia and astrocytes to B. burgdorferi and N. meningitidis, two disparate gram negative bacterial species that can cross the blood-brain barrier in humans. Importantly, we demonstrate that these organisms induce the delayed production of significant quantities of IL-10 by both microglia and astrocytes. Furthermore, we demonstrate that such production occurs independent of the actions of bacterial lipopolysaccharide and is secondary to the autocrine or paracrine actions of other glia-derived soluble mediators. The late onset of IL-10 production by resident glia following activation, the previously documented expression of specific receptors for this cytokine on microglia and astrocytes, and the ability of IL-10 to inhibit bacterially induced immune responses by these cells, suggest a mechanism by which resident glial cells can limit potentially damaging inflammation within the CNS in response to invading pathogens, and could explain the suppression of inflammation seen within the brain parenchyma during chronic bacterial infections.
Collapse
Affiliation(s)
- Amy Rasley
- Department of Biology, University of North Carolina at Charlotte, 28223, USA
| | | | | | | |
Collapse
|
28
|
Hinkerohe D, Smikalla D, Haghikia A, Heupel K, Haase CG, Dermietzel R, Faustmann PM. Effects of cytokines on microglial phenotypes and astroglial coupling in an inflammatory coculture model. Glia 2005; 52:85-97. [PMID: 15920725 DOI: 10.1002/glia.20223] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cytokines play an important role in the onset, regulation, and propagation of immune and inflammatory responses within the central nervous system (CNS). The main source of cytokines in the CNS are microglial cells. Under inflammatory conditions, microglial cells are capable of producing pro- and antiinflammatory cytokines, which convey essential impact on the glial and neuronal environment. One paramount functional feature of astrocytes is their ability to form a functionally coupled syncytium. The structural link, which is responsible for the syncytial behavior of astrocytes, is provided by gap junctions. The present study was performed to evaluate the influence of inflammation related cytokines on an astroglial/microglial inflammatory model. Primary astrocytic cultures of newborn rats were cocultured with either 5% (M5) or 30% (M30) microglial cells and were incubated with the following proinflammatory cytokines: tumor necrosis factor-alpha (TNF-alpha), interleukin-1beta (IL-1beta), interleukin-6 (IL-6), interferon-gamma (IFN-gamma), and the antiinflammatory cytokines transforming growth factor-beta1 (TGF-beta1) and IFN-beta. Under these conditions, i.e., incubation with the inflammatory cytokines and the high fraction of microglia (M30), microglial cells revealed a significant increase of activated round phagocytotic cells accompanied by a reduction of astroglial connexin 43 (Cx43) expression, a reduced functional coupling together with depolarization of the membrane resting potential (MRP). When the antiinflammatory mediator TGF-beta1 was added to proinflammatory altered M30 cocultures, a reversion of microglial activation and reconstitution of functional coupling together with recovery of the astroglial MRP was achieved. Finally IFN-beta, added to M5 cocultures was able to prevent the effects of the proinflammatory cytokines TNF-alpha, IL-1beta, and IFN-gamma.
Collapse
Affiliation(s)
- Daniel Hinkerohe
- Department of Neuroanatomy and Molecular Brain Research, Ruhr University Bochum, Bochum, Germany
| | | | | | | | | | | | | |
Collapse
|
29
|
Rambach G, Hagleitner M, Mohsenipour I, Lass-Flörl C, Maier H, Würzner R, Dierich MP, Speth C. Antifungal activity of the local complement system in cerebral aspergillosis. Microbes Infect 2005; 7:1285-95. [PMID: 16027023 DOI: 10.1016/j.micinf.2005.04.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2004] [Revised: 03/30/2005] [Accepted: 04/22/2005] [Indexed: 10/25/2022]
Abstract
Dissemination of aspergillosis into the central nervous system is associated with nearly 100% mortality. To study the reasons for the antifungal immune failure we analyzed the efficacy of cerebral complement to combat the fungus Aspergillus. Incubation of Aspergillus in non-inflammatory cerebrospinal fluid (CSF) revealed that complement levels were sufficient to obtain a deposition on the surface, but opsonization was much weaker than in serum. Consequently complement deposition from normal CSF on fungal surface stimulated a very low phagocytic activity of microglia, granulocytes, monocytes and macrophages compared to stimulation by conidia opsonized in serum. Similarly, opsonization of Aspergillus by CSF was not sufficient to induce an oxidative burst in infiltrating granulocytes, whereas conidia opsonized in serum induced a clear respiratory signal. Thus, granulocytes were capable of considerably reducing the viability of serum-opsonized Aspergillus conidia, but not of conidia opsonized in CSF. The limited efficacy of antifungal attack by cerebral complement can be partly compensated by enhanced synthesis, leading to elevated complement concentrations in CSF derived from a patient with cerebral aspergillosis. This inflammatory CSF was able to induce (i) a higher complement deposition on the Aspergillus surface than non-inflammatory CSF, (ii) an accumulation of complement activation products and (iii) an increase in phagocytic and killing activity of infiltrating granulocytes. However, levels and efficacy of the serum-derived complement were not reached. These data indicate that low local complement synthesis and activation may represent a central reason for the insufficient antifungal defense in the brain and the high mortality rate of cerebral aspergillosis.
Collapse
Affiliation(s)
- Günter Rambach
- Department of Hygiene, Microbiology and Social Medicine, Innsbruck Medical University, and Ludwig-Boltzmann-Institute for AIDS Research, Fritz-Pregl-Str. 3, 6020 Innsbruck, Austria
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Pearse DD, Pereira FC, Stolyarova A, Barakat DJ, Bunge MB. Inhibition of tumour necrosis factor-alpha by antisense targeting produces immunophenotypical and morphological changes in injury-activated microglia and macrophages. Eur J Neurosci 2005; 20:3387-96. [PMID: 15610171 DOI: 10.1111/j.1460-9568.2004.03799.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Microglia respond in a stereotypical pattern to a diverse array of pathological states. These changes are coupled to morphological and immunophenotypical alterations and the release of a variety of reactive species, trophic factors and cytokines that modify both microglia and their cellular environment. We examined whether a microglial-produced cytokine, tumour necrosis factor-alpha (TNF-alpha), was involved in the maintenance of microglial activation after spinal cord injury by selective inhibition using TNF-alpha antisense deoxyoligonucleotides (ASOs). Microglia and macrophages harvested from 3 d post-contused rat spinal cord were large and rounded (86.3 +/- 9.6%). They were GSA-IB4-positive (GSA-IB4(+)) (Griffonia simplicifolia lectin, microglia specific; 94.8 +/- 5.1%), strongly OX-42 positive (raised against a type 3 complement/integrin receptor, CD11b; 78.9 +/- 9.1%), ED-1 positive (a lysosomal marker shown to correlate well with immune cell activation; 97.2 +/- 2.6%) and IIA positive (antibody recognizes major histocompatibility complex II; 57.2 +/- 5.6%), indicative of fully activated cells, for up to 48 h after plating. These cells also secreted significant amounts of TNF-alpha (up to 436 pg/microg total protein, 16 h). Fluoroscein isothiocyanate-labelled TNF-alpha ASOs (5, 50 and 200 nm) added to the culture medium were taken up very efficiently into the cells (> 90% cells) and significantly reduced TNF-alpha production by up to 92% (26.5 pg/microg total protein, 16 h, 200 nm TNF-alpha ASOs). Furthermore, few of the treated cells at this time were round (5.4 +/- 2.7%), having become predominantly spindle shaped (74.9 +/- 6.3%) or stellate (21.4 +/- 2.7%); immunophenotypically, although all of them remained GSA-IB4 positive (91.6 +/- 6.2%), many were weakly OX-42 positive and few expressed either ED-1 (12.9 +/- 2.5%) or IIA (19.8 +/- 7.4%). Thus, the secretion of TNF-alpha early in spinal cord injury may be involved in autoactivating microglia/macrophages. However, at the peak of microglial activation after injury, the activation state of microglia/macrophages is not stable and this process may still be reversible by blocking TNF-alpha.
Collapse
Affiliation(s)
- Damien D Pearse
- The Miami Project to Cure Paralysis, University of Miami School of Medicine, PO Box 016960 (R-48), Miami, FL 33101, USA.
| | | | | | | | | |
Collapse
|
31
|
Bechmann I, Goldmann J, Kovac AD, Kwidzinski E, Simbürger E, Naftolin F, Dirnagl U, Nitsch R, Priller J. Circulating monocytic cells infiltrate layers of anterograde axonal degeneration where they transform into microglia. FASEB J 2005; 19:647-9. [PMID: 15671154 DOI: 10.1096/fj.04-2599fje] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
In this study, we demonstrate the infiltration of blood-derived monocytic cells and their morphologic transformation into microglia in zones of acute, anterograde (Wallerian) axonal degeneration induced by entorhinal cortex lesion (ECL). ECL was performed in mice which had received green fluorescent protein (GFP)-transduced bone marrow grafts allowing identification of blood-derived elements within the brain. While in the unlesioned hemisphere GFP+ cells were restricted to perivascular and leptomeningeal sites, many round fluorescent cells appeared in hippocampal zones of axonal degeneration at 24 h post lesion (hpl). Within 72 hpl, these GFP+ cells acquired ramified, microglia-like morphologies, which persisted for at least 7 days post ECL. Differentiation of GFP+ cells into glial fibrillary acidic protein (GFAP)+ astrocytes was never observed. To exclude that this recruitment is an artifact of irradiation or bone marrow transplantation, the fluorescent cell tracker 6-carboxylfluorescein diacetate (CFDA) was injected into spleens of normal mice 1 day before ECL. Again, fluorescent cells appeared at the lesion site and along the layers of axonal degeneration at 48 hpl and CFDA+/MAC-1+, cells exhibited amoeboid and ramified morphologies. Thus, blood-derived cells infiltrate not only the site of mechanical lesion, but also the layers of anterograde axonal degeneration, where they readily transform into microglia-like elements. A role for infiltrating leukocytes in facilitating or modulating postlesional plasticity, e.g., by phagocytosis of growth-inhibiting myelin should now be considered. Moreover, monocytic cells may serve as vehicles to transport therapeutic substances such as neurotrophic factors or caspase inhibitors to zones of axonal degeneration.
Collapse
Affiliation(s)
- Ingo Bechmann
- Institute of Cell Biology and Neurobiology, Charité University Hospital, Berlin, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Heppner FL, Greter M, Marino D, Falsig J, Raivich G, Hövelmeyer N, Waisman A, Rülicke T, Prinz M, Priller J, Becher B, Aguzzi A. Experimental autoimmune encephalomyelitis repressed by microglial paralysis. Nat Med 2005; 11:146-52. [PMID: 15665833 DOI: 10.1038/nm1177] [Citation(s) in RCA: 567] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2004] [Accepted: 12/02/2004] [Indexed: 12/23/2022]
Abstract
Although microglial activation occurs in inflammatory, degenerative and neoplastic central nervous system (CNS) disorders, its role in pathogenesis is unclear. We studied this question by generating CD11b-HSVTK transgenic mice, which express herpes simplex thymidine kinase in macrophages and microglia. Ganciclovir treatment of organotypic brain slice cultures derived from CD11b-HSVTK mice abolished microglial release of nitrite, proinflammatory cytokines and chemokines. Systemic ganciclovir administration to CD11b-HSVTK mice elicited hematopoietic toxicity, which was prevented by transfer of wild-type bone marrow. In bone marrow chimeras, ganciclovir blocked microglial activation in the facial nucleus upon axotomy and repressed the development of experimental autoimmune encephalomyelitis. We conclude that microglial paralysis inhibits the development and maintenance of inflammatory CNS lesions. The microglial compartment thus provides a potential therapeutic target in inflammatory CNS disorders. These results validate CD11b-HSVTK mice as a tool to study the impact of microglial activation on CNS diseases in vivo.
Collapse
Affiliation(s)
- Frank L Heppner
- Institute of Neuropathology, University Hospital Zurich, CH-8091 Zurich, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Speth C, Dierich MP, Sopper S. HIV-infection of the central nervous system: the tightrope walk of innate immunity. Mol Immunol 2005; 42:213-28. [PMID: 15488609 DOI: 10.1016/j.molimm.2004.06.018] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Infection of the central nervous system (CNS) by HIV is a frequent and sometimes very early event in the course of HIV pathogenesis. Possible consequences are diverse symptoms of neurological dysfunction, but also the establishment of a lifelong latent viral reservoir in the brain. Whereas in the periphery innate and adaptive immunity are equal partners, the blood-brain barrier (BBB) with its restricted access of peripheral immune effectors shifts this balance in favour of the local innate immunity. Four main elements of cerebral innate immunity are discussed in the present article, including two cell types with immunological functions and two soluble immune systems: (1) the stimulation of microglial cells as the predominant brain-resident immune cell and the main local reservoir for the virus; (2) the reaction of astrocytes in response to viral infection; (3) the activation of the local complement system as important soluble immune cascade; and (4) the role of chemokines and cytokines which help to conduct and cross-link the interplay between the different immune elements. These components of the cerebral innate immunity do not act separately from each other but form a functional immunity network. A dual role of these components with both harmful and protective effects further enhances the complexity of the mutual interactions.
Collapse
Affiliation(s)
- Cornelia Speth
- Institute of Hygiene and Social Medicine, Medical University Innsbruck and Ludwig-Boltzmann-Institute for AIDS Research, Fritz-Pregl-Str. 3, A-6020 Innsbruck, Austria.
| | | | | |
Collapse
|
34
|
Mutlu LK, Woiciechowsky C, Bechmann I. Inflammatory response after neurosurgery. Best Pract Res Clin Anaesthesiol 2004; 18:407-24. [PMID: 15212336 DOI: 10.1016/j.bpa.2003.12.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Investigation into the inflammatory response in the central nervous system (CNS) is a rapidly growing field, and a vast amount of information on this topic has accumulated over the past two decades. Inflammation is a particularly interesting issue in the (traditionally non-regenerating) CNS, owing to its dual role in worsening or improving regeneration and functional outcome in certain circumstances. This paper reviews the current literature on the interactions between the immune system and the CNS in physiological and pathological states. The first part will provide an overview of the cellular and molecular components of CNS inflammation, this being followed by a discussion of the concept of systemic immunodepression after neurotrauma and neurosurgery. Finally, the delicate balance of immune responses in the CNS, with an emphasis on the beneficial effects of inflammation and possible therapeutic options, will be discussed.
Collapse
Affiliation(s)
- Leman K Mutlu
- Department of Cell and Neurobiology, Institute of Anatomy, Humboldt University Medical School-Charité, Berlin, Germany
| | | | | |
Collapse
|
35
|
Schultz J, Schwarz A, Neidhold S, Burwinkel M, Riemer C, Simon D, Kopf M, Otto M, Baier M. Role of interleukin-1 in prion disease-associated astrocyte activation. THE AMERICAN JOURNAL OF PATHOLOGY 2004; 165:671-8. [PMID: 15277240 PMCID: PMC1618583 DOI: 10.1016/s0002-9440(10)63331-7] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Prion-induced chronic neurodegeneration has a substantial inflammatory component, and the activation of glia cells may play an important role in disease development and progression. However, the functional contribution of cytokines to the development of the gliosis in vivo was never systematically studied. We report here that the expression of interleukin-1beta (IL-1beta), IL-1beta-converting enzyme, and IL-1 receptor type 1 (IL-1RI) is up-regulated in a murine scrapie model. The scrapie-induced gliosis in IL-1RI(-/-) mice was characterized by an attenuated activation of astrocytes in the asymptomatic stage of the disease and a reduced expression of CXCR3 ligands. Furthermore, the accumulation of the misfolded isoform of the prion protein PrP(Sc) was significantly delayed in the IL-1RI(-/-) mice. These observations indicate that IL-1 is a driver of the scrapie-associated astrocytosis and possibly the accompanying amyloid deposition. In addition, scrapie-infected IL-1RI-deficient (IL-1RI(-/-)) mice showed a delayed disease onset and significantly prolonged survival times suggesting that an anti-inflammatory therapeutical approach to suppress astrocyte activation and/or glial IL-1 expression may help to delay disease onset in established prion infections of the central nervous system.
Collapse
Affiliation(s)
- Julia Schultz
- Project "Neurodegenerative Diseases," Robert-Koch-Institute, Nordufer 20, 13353 Berlin, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Speth C, Williams K, Hagleitner M, Westmoreland S, Rambach G, Mohsenipour I, Schmitz J, Würzner R, Lass-Flörl C, Stoiber H, Dierich MP, Maier H. Complement synthesis and activation in the brain of SIV-infected monkeys. J Neuroimmunol 2004; 151:45-54. [PMID: 15145603 DOI: 10.1016/j.jneuroim.2004.02.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2003] [Revised: 01/12/2004] [Accepted: 02/19/2004] [Indexed: 11/25/2022]
Abstract
Complement is one of the most critical defence tools against cerebral infections, but uncontrolled complement biosynthesis and activation can induce profound brain tissue damage. To clarify the role of complement in the pathogenesis of AIDS-associated neurological disorders, we analysed the synthesis of complement in the brains of SIV-infected rhesus macaques. Using immunohistochemical staining we could show that the cerebral synthesis of complement factors C1q and C3 was strongly upregulated in SIV-infected monkeys compared to the spontaneous synthesis in uninfected control monkeys. Astrocytes, neurons, microglia, infiltrating macrophages and multinuclear giant cells all contribute to the high amounts of C1q and C3 in the brain. Secreted C1q and C3 are also deposited on the membrane of neurons, a prerequisite for formation of the membrane-driven lytic membrane attack complex. The membrane deposition thus might suggest complement-induced lysis of bystander neurons as a potential mechanism for cell damage during viral infection of the brain.
Collapse
Affiliation(s)
- Cornelia Speth
- Institute of Hygiene and Social Medicine, Innsbruck Medical University and Ludwig-Bolthmann-Institute for AIDS Research, Fritz-Pregl-Str. 3, Innsbruck A-6020, Austria.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Hanbury R, Ling ZD, Wuu J, Kordower JH. GFAP knockout mice have increased levels of GDNF that protect striatal neurons from metabolic and excitotoxic insults. J Comp Neurol 2003; 461:307-16. [PMID: 12746870 DOI: 10.1002/cne.10667] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
In response to injury and degeneration, astrocytes hypertrophy, extend processes, and increase production of glial fibrillary acidic protein (GFAP), an intermediate filament protein located within their cytoplasm. The present study tested the hypothesis that GFAP expression alters the vulnerability of neurons to excitotoxic and metabolic insult induced by 3-nitroproprionic acid (3-NP), an irreversible inhibitor of mitochondrial complex II activity or the excitotoxin quinolinic acid (QA). In this respect, adult GFAP knockout mice (KO) and wild-type control mice (WT) received unilateral intrastriatal injections of 3-NP (200 nmol/microl) or QA (100 nmol/microl) and were killed 1, 2, or 4 weeks later. Lesion volume and neuronal counts were quantified using unbiased stereologic principles. For both QA and 3-NP lesions, a significant decrease in lesion volume and an increase in striatal projection neurons were seen in GFAP KO mice compared with WT mice. Enzyme-linked immunoassay analysis revealed increased basal levels of glial cell derived neurotrophic factor (GDNF) relative to WT mice. In contrast, no differences were observed in the expression of ciliary neurotrophic factor or nerve growth factor. These data strongly suggest that the expression of GFAP is implicated with the production of GDNF to a degree that confers neuroprotection after an excitotoxic or metabolic insult.
Collapse
Affiliation(s)
- Rose Hanbury
- Research Center for Brain Repair and Department of Neurological Sciences, Rush Presbyterian Medical Center, Chicago, Illinois 60612, USA
| | | | | | | |
Collapse
|
38
|
Eyüpoglu IY, Bechmann I, Nitsch R. Modification of microglia function protects from lesion-induced neuronal alterations and promotes sprouting in the hippocampus. FASEB J 2003; 17:1110-1. [PMID: 12692086 DOI: 10.1096/fj.02-0825fje] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Primary neuronal destruction in the central nervous system triggers rapid changes in glial morphology and function, after which activated glial cells contribute to secondary neuronal changes. Here we show that, after entorhinal cortex lesion, activation of microglia, but not other glial cells, leads to massive secondary dendritic changes of deafferentiated hippocampal neurons. Blocking of microglial activation in vivo reduced this secondary neuronal damage and enhanced regenerative axonal sprouting. In contrast, abolishing astrocytes or oligodendroglia did not result in specific neuronal changes. Furthermore, primary damage leads to an interleukin 1beta up-regulation, which is attenuated by the immuno-modulator transforming growth factor beta1, whereas tumor necrosis factor alpha is not affected. Modification of microglial activity following denervation of the hippocampus protects neurons from secondary dendritic alterations and therefore enables their reinnervation. These data render activated microglia a putative therapeutic target during the course of axonal degeneration.
Collapse
Affiliation(s)
- Ilker Y Eyüpoglu
- Institute of Anatomy, Department of Cell and Neurobiology, Humboldt University Hospital (Charité), 10098 Berlin, Germany.
| | | | | |
Collapse
|
39
|
Faustmann PM, Haase CG, Romberg S, Hinkerohe D, Szlachta D, Smikalla D, Krause D, Dermietzel R. Microglia activation influences dye coupling and Cx43 expression of the astrocytic network. Glia 2003; 42:101-8. [PMID: 12655594 DOI: 10.1002/glia.10141] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Under inflammatory conditions, activated microglia are capable of producing proinflammatory cytokines that are reported to influence cell-to-cell communication. The present study was performed to evaluate the influence of microglial activation on the coupling efficiency of the astroglial network. Primary astrocyte cultures of newborn rats were cocultured with either 5% (M5) or 30% (M30) microglia. Microglial activation (rounded phagocytotic phenotype) was investigated using the monoclonal anti-ED1 antibody, and immunofluorescence with a polyclonal anti-Cx43 antibody was used to study astroglial Cx43 expression and distribution. Functional coupling of astrocytes was evaluated by monitoring the transfer of microinjected Lucifer yellow into neighboring cells. The data obtained can be summarized as follows: astroglia/M30 cocultures contained significantly fewer resting microglia and significantly more activated microglia than the M5 cocultures; significantly reduced astroglial Cx43 staining was found in M30 cocultures concurrently with a reduced number of dye coupled astrocytes; and the positive correlation of percent activated microglia with reduced astroglial Cx43 expression was highly significant, indicating that the degree of intercellular communication in the astroglial network may be modulated by the activation of microglia under in vitro conditions.
Collapse
Affiliation(s)
- Pedro M Faustmann
- Department of Neuroanatomy and Molecular Brain Research, Ruhr-University Bochum, Bochum, Germany
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Eyüpoglu IY, Savaskan NE, Bräuer AU, Nitsch R, Heimrich B. Identification of neuronal cell death in a model of degeneration in the hippocampus. BRAIN RESEARCH. BRAIN RESEARCH PROTOCOLS 2003; 11:1-8. [PMID: 12697257 DOI: 10.1016/s1385-299x(02)00186-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Neuronal cell death and microglial changes are both hallmarks of neurodegenerative disorders. Therefore, analysis of degenerating neurons related to microglial changes are addressed in many studies of neurosciences. Here we compared different lesion models and two markers for neurodegeneration (Fluoro-Jade and propidium iodide) in an in vivo as well as an in vitro approach. Fluoro-Jade is a specific and selective marker to identify neurons undergoing degeneration. We also tested this marker to analyze neurodegeneration in organotypic hippocampal slice cultures. We could show that activation of microglia is followed by neuronal cell death. Most degeneration markers, such as propidium iodide, only stain the neuronal cell body excluding the axonal and dendritic processes. Fluoro-Jade is able to stain the distal portion as well as the proximal portion of the dissected axon including the axotomized neuron, as so called anterograde and retrograde degeneration after axotomy. To analyze the specificity of Fluoro-Jade, we used primary microglial and BV-2 cells, a well-described murine microglial cell line. Treatment of microglial and BV-2 cells with an excess of L-glutamate induces cell death which could be detected by propidium iodide staining, but not by Fluoro-Jade, demonstrating its specificity to monitor neuronal cell death.
Collapse
Affiliation(s)
- Ilker Y Eyüpoglu
- Institute of Anatomy, Department of Cell- and Neurobiology, Charité Campus Miffe, Humboldt University Medical School Charité, 10098 Berlin, Germany.
| | | | | | | | | |
Collapse
|
41
|
Abstract
A successful outcome for the host of virus infection of the central nervous system (CNS) requires the elimination of the virus without damage to essential non-renewable cells, such as neurons. As a result, inflammatory responses must be tightly controlled, and many unique mechanisms seem to contribute to this control. In addition to being important causes of human disease, RNA viruses that infect the CNS provide useful models in which to study immune responses in the CNS. Recent work has shown the importance of innate immune responses in the CNS in controlling virus infection. And advances have been made in assessing the relative roles of cytotoxic T cells, antibodies and cytokines in the clearance of viruses from neurons, glial cells and meningeal cells.
Collapse
Affiliation(s)
- Diane E Griffin
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland 21205, USA.
| |
Collapse
|
42
|
Badie B, Bartley B, Schartner J. Differential expression of MHC class II and B7 costimulatory molecules by microglia in rodent gliomas. J Neuroimmunol 2002; 133:39-45. [PMID: 12446006 DOI: 10.1016/s0165-5728(02)00350-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
To assess the immune function of microglia and macrophages in brain tumors, the expression of MHC class II and B7 costimulatory molecules in three rodent glioma models was examined. Microglia and macrophages, which accounted for 5-12% of total cells, expressed B7.1 and MHC class II molecules in the C6 and 9L tumors, but not RG2 gliomas. Interestingly, the expression of B7.1 and MHC class II molecules by microglia and macrophage was associated with an increase in the number of tumor-infiltrating lymphocytes in C6 and 9L tumors. B7.2 expression, which was present at low levels on microglia and macrophages in normal brain, did not significantly change in tumors. Interestingly, the expression of all three surface antigens increased after microglia were isolated from intracranial C6 tumors and cultured for a short period of time. We conclude that microglia immune activity may be suppressed in gliomas and directly correlates to the immunogenecity of experimental brain tumors.
Collapse
Affiliation(s)
- Behnam Badie
- Neuro-Oncology Laboratory, K3/805 Clinical Science Center, Department of Neurological Surgery, University of Wisconsin, School of Medicine, Madison, WI 53792-3232, USA.
| | | | | |
Collapse
|
43
|
Wirjatijasa F, Dehghani F, Blaheta RA, Korf HW, Hailer NP. Interleukin-4, interleukin-10, and interleukin-1-receptor antagonist but not transforming growth factor-beta induce ramification and reduce adhesion molecule expression of rat microglial cells. J Neurosci Res 2002; 68:579-87. [PMID: 12111847 DOI: 10.1002/jnr.10254] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The activity of microglial cells is strictly controlled in order to maintain central nervous system (CNS) immune privilege. We hypothesized that several immunomodulatory factors present in the CNS parenchyma, i.e., the Th2-derived cytokines interleukin (IL)-4 and IL-10, interleukin-1-receptor-antagonist (IL-1-ra), or transforming growth factor (TGF)-beta can modulate microglial morphology and functions. Microglial cells were incubated with IL-4, IL-10, IL-1-ra, TGF-beta, or with astrocyte conditioned media (ACM) and were analyzed for morphological changes, expression of intercellular adhesion molecule (ICAM)-1, and secretion of IL-1beta or tumor necrosis factor (TNF)-alpha. Whereas untreated controls showed an amoeboid morphology both Th2-derived cytokines, IL-1-ra, and ACM induced a morphological transformation to the ramified phenotype. In contrast, TGF-beta-treated microglial cells showed an amoeboid morphology. Even combined with the neutralizing antibodies against IL-4, IL-10, or TGF-beta ACM induced microglial ramification. Furthermore, ACM did not contain relevant amounts of IL-4 and IL-10, as measured by enzyme-linked immunosorbent assay (ELISA). Flow cytometry showed that lipopolysaccharide (LPS)-induced ICAM-1-expression on microglial cells was strongly suppressed by ACM, significantly modulated by IL-4, IL-10, or IL-1-ra, but not influenced by TGF-beta. The LPS-induced secretion of IL-1beta and TNF-alpha was only reduced after application of ACM, whereas IL-4 or IL-10 did not inhibit IL-1beta- or TNF-alpha secretion. TGF-beta enhanced IL-1beta- but not TNF-alpha secretion. In summary, we demonstrate that IL-4, IL-10, and IL-1-ra induce microglial ramification and reduce ICAM-1-expression, whereas the secretion of proinflammatory cytokines is not prevented. TGF-beta has no modulating effects. Importantly, unidentified astrocytic factors that are not identical with IL-4, IL-10, or TGF-beta possess strong immunomodulatory properties.
Collapse
|
44
|
Melchior B, Rémy S, Nerrière-Daguin V, Heslan JM, Soulillou JP, Brachet P. Temporal analysis of cytokine gene expression during infiltration of porcine neuronal grafts implanted into the rat brain. J Neurosci Res 2002; 68:284-92. [PMID: 12111858 DOI: 10.1002/jnr.10216] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
A large array of evidence supports the involvement of infiltrating T lymphocytes in the rejection process of intracerebral neuronal xenografts. Little is known, however, about the molecular mechanisms that drive the recruitment of this cell type. In the present work, we used real-time RT-PCR methodology to investigate the kinetics of cytokine gene expression during the infiltration of fetal porcine neurons (PNEU) implanted into the striatum of LEW.1A rats. T lymphocyte infiltration was followed by measuring the intracerebral levels of transcripts encoding the beta chain of the T cell receptor. These transcripts remained barely detectable until the fourth week (28 days) postimplantation, when a sudden accumulation occurred. Their kinetics, which support previous immunohistochemical observations, indicate that alphabetaT lymphocyte recruitment occurs rapidly after a delay of several weeks in this experimental model. Infiltration of PNEU grafts by T lymphocytes was accompanied by a concomitant, dramatic augmentation of transcripts coding for monocyte chemotactic protein-1 and RANTES (for regulated on activation, normal T cell expressed and secreted), two chemokines targeting this cell type, among others. Likewise, a sudden accumulation of transcripts of proinflammatory lymphokines [interleukin (IL)-1alpha, tumor necrosis factor-alpha, IL-6] as well as Th1 cytokines (IL-2, interferon-gamma) was also detected. In contrast, IL-4, -10, and -13 mRNA remained barely detectable at the different time points. No significant changes were noticed for IL-12 or transforming growth factor-beta transcripts. These data support the concept that T lymphocyte infiltration of PNEU grafts is actively promoted by a local production of chemokines and proinflammatory lymphokines and is based on a Th1 polarization.
Collapse
MESH Headings
- Animals
- Antigens, CD
- Antigens, Neoplasm
- Antigens, Surface
- Avian Proteins
- Basigin
- Blood Proteins
- Brain Tissue Transplantation/adverse effects
- Brain Tissue Transplantation/methods
- Cells, Cultured
- Chemokine CCL2/metabolism
- Chemokine CCL5/metabolism
- Chemotaxis, Leukocyte/immunology
- Cytokines/genetics
- Fetus
- Gene Expression Regulation/immunology
- Graft Rejection/immunology
- Graft Rejection/metabolism
- Graft Rejection/physiopathology
- Immunohistochemistry
- Interferon-gamma/metabolism
- Interleukin-2 Receptor alpha Subunit
- Interleukins/metabolism
- Kinetics
- Male
- Membrane Glycoproteins/metabolism
- RNA, Messenger/immunology
- RNA, Messenger/metabolism
- Rats
- Rats, Inbred Lew
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Interleukin/metabolism
- Swine
- T-Lymphocytes/cytology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Time Factors
- Transforming Growth Factor beta/metabolism
- Tumor Necrosis Factor-alpha/metabolism
Collapse
Affiliation(s)
- Benoît Melchior
- Institut National de la Santé et de la Recherche Médicale, Unité 437, Centre Hospitalier Universitaire, Nantes, France
| | | | | | | | | | | |
Collapse
|
45
|
Speth C, Schabetsberger T, Mohsenipour I, Stöckl G, Würzner R, Stoiber H, Lass-Flörl C, Dierich MP. Mechanism of human immunodeficiency virus-induced complement expression in astrocytes and neurons. J Virol 2002; 76:3179-88. [PMID: 11884542 PMCID: PMC136041 DOI: 10.1128/jvi.76.7.3179-3188.2002] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The cerebral complement system is hypothesized to contribute to neurodegeneration in the pathogenesis of AIDS-associated neurological disorders. Our former results have shown that the human immunodeficiency virus (HIV) strongly induces the synthesis of complement factor C3 in astrocytes. This upregulation explains in vivo data showing elevated complement levels in the cerebrospinal fluid of patients with AIDS-associated neurological symptoms. Since inhibition of complement synthesis and activation in the brain may represent a putative therapeutic goal to prevent virus-induced damage, we analyzed in detail the mechanisms of HIV-induced modulation of C3 expression. HIV-1 increased the C3 levels in astrocyte culture supernatants from 30 to up to 400 ng/ml; signal transduction studies revealed that adenylate cyclase activation with upregulation of cyclic AMP is the central signaling pathway to mediate that increase. Furthermore, activity of protein kinase C is necessary for HIV induction of C3, since inhibition of protein kinase C by prolonged exposure to the phorbol ester tetradecanoyl phorbol acetate partly abolished the HIV effect. The cytokines tumor necrosis factor alpha and gamma interferon were not involved in mediating the HIV-induced C3 upregulation, since neutralizing antibodies had no effect. Besides whole HIV virions, the purified viral proteins Nef and gp41 are biologically active in upregulating C3, whereas Tat, gp120, and gp160 were not able to modulate C3 synthesis. Further experiments revealed that neurons were also able to respond on incubation with HIV with increased C3 synthesis, although the precise pattern was slightly different from that in astrocytes. This strengthens the hypothesis that HIV-induced complement synthesis represents an important mechanism for the pathogenesis of AIDS in the brain.
Collapse
Affiliation(s)
- Cornelia Speth
- Institute of Hygiene and Social Medicine, University of Innsbruck Ludwig Boltzmann Institute for AIDS Research, Innsbruck, Austria.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Togo T, Akiyama H, Iseki E, Kondo H, Ikeda K, Kato M, Oda T, Tsuchiya K, Kosaka K. Occurrence of T cells in the brain of Alzheimer's disease and other neurological diseases. J Neuroimmunol 2002; 124:83-92. [PMID: 11958825 DOI: 10.1016/s0165-5728(01)00496-9] [Citation(s) in RCA: 339] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
We investigated the occurrence of T cells in the brain parenchyma of Alzheimer's disease (AD), non-AD degenerative dementias and controls by semi-quantitative analysis of immunohistochemically stained tissue sections. In all cases, we found at least some T cells. The number of T cells was increased in the majority of AD cases compared with other cases. The phenotype of T cells in the AD brain indicates that they are activated but are not fully differentiated. Antigen-triggered clonal expansion is not likely to take place. Local inflammatory conditions might cause accumulation and activation of T cells in the AD brain.
Collapse
Affiliation(s)
- Takashi Togo
- Tokyo Institute of Psychiatry, 2-1-8 Kamikitazawa, Setagaya-ku, Tokyo 156-8585, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Ullrich O, Diestel A, Eyüpoglu IY, Nitsch R. Regulation of microglial expression of integrins by poly(ADP-ribose) polymerase-1. Nat Cell Biol 2001; 3:1035-42. [PMID: 11781564 DOI: 10.1038/ncb1201-1035] [Citation(s) in RCA: 150] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Excitotoxic brain lesions initially result in the primary destruction of brain parenchyma, after which microglial cells migrate towards the sites of injury. At these sites, the cells produce large quantities of oxygen radicals and cause secondary damage that accounts for most of the loss of brain function. Here we show that this microglial migration is strongly controlled in living brain tissue by expression of the integrin CD11a, regulated by the nuclear enzyme poly(ADP-ribose) polymerase-1 (PARP-1) through the formation of a nuclear PARP-NF-kappaB-protein complex. Downregulation of PARP or CD11a by transfection with antisense DNA abrogated microglial migration almost completely and prevented neurons from secondary damage.
Collapse
Affiliation(s)
- O Ullrich
- Department of Cell- and Neurobiology, Institute of Anatomy. Medical Faculty (Charité), Humboldt-University Berlin, Schumannstrasse 20/21, 10098 Berlin, Germany.
| | | | | | | |
Collapse
|
48
|
Schwarz MJ, Chiang S, Müller N, Ackenheil M. T-helper-1 and T-helper-2 responses in psychiatric disorders. Brain Behav Immun 2001; 15:340-70. [PMID: 11782103 DOI: 10.1006/brbi.2001.0647] [Citation(s) in RCA: 143] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The expanding field of psychoneuroimmunology has markedly increased knowledge about the interference of the central nervous system and the immune system. Immunological abnormalities in psychiatric patients have been repeatedly described in the last century. Modern concepts of immunology and the growing knowledge of psychoneuroimmunology may help in understanding the distinct immunological mechanisms in psychiatric disorders. One of these concepts regarding the adaptive immune system is the discrimination between Th1-like cell-mediated and Th2-like antibody-related immune responses. This article systematically describes alterations of Th1- or Th2-specific parameters in the major psychiatric disorders schizophrenia, major depression, and Alzheimer's disease. There are several hints of associations of these two distinct arms of immune response with subgroups of schizophrenia and major depression. The immunological research in Alzheimer's disease has already led to a preclinical model of immunotherapy. Categorization of immune parameters may also help to identify a possible immune-related pathophysiology in psychotic and affective disorders, resulting in specific treatment strategies.
Collapse
Affiliation(s)
- M J Schwarz
- Psychiatric Hospital, University of Munich, Nussbaumstr. 7, D-80336 Munich, Germany
| | | | | | | |
Collapse
|
49
|
Gobin SJ, Montagne L, Van Zutphen M, Van Der Valk P, Van Den Elsen PJ, De Groot CJ. Upregulation of transcription factors controlling MHC expression in multiple sclerosis lesions. Glia 2001; 36:68-77. [PMID: 11571785 DOI: 10.1002/glia.1096] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The expression of major histocompatibility complex (MHC) class I and class II in the CNS has received considerable interest because of its importance in neurodegenerative or inflammatory diseases, such as multiple sclerosis (MS). However, at the moment nothing is known about the expression patterns of transcription factors controlling MHC expression in MS lesions. Here, we performed an extensive immunohistochemical analysis on MS affected postmortem brain tissue to determine the cellular localization and distribution of different MHC-controlling transcription factors. We show that phagocytic macrophages in active demyelinating MS lesions displayed a moderate to strong immunostaining of the MHC-specific transcription factors RFX and CIITA, as well as the general transcription factors NF-kappaB, IRF1, STAT1, USF, and CREB, which was congruent with a strongly enhanced expression of HLA-DR, HLA-DQ, HLA-DP, and HLA class I. In the normal-appearing white matter (NAWM), clusters of activated microglial cells forming preactive lesions displayed an overall stronger expression level of these transcription factors, combined with a strong to intense level of MHC class I and class II immunostaining. In general, astrocytes and oligodendrocytes either did not express, or weakly expressed, these transcription factors, correlating with a lack of MHC class II and weak MHC class I expression. Together, the elevated expression level of transcription factors governing expression of MHC class I and class II molecules in activated microglial cells and phagocytic macrophages strongly suggests a general state of microglial cell activation in MS lesions.
Collapse
Affiliation(s)
- S J Gobin
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands.
| | | | | | | | | | | |
Collapse
|
50
|
Gimsa U, Wolf SA, Haas D, Bechmann I, Nitsch R. Th2 cells support intrinsic anti-inflammatory properties of the brain. J Neuroimmunol 2001; 119:73-80. [PMID: 11525802 DOI: 10.1016/s0165-5728(01)00343-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
In experimental autoimmune encephalomyelitis (EAE), Th1 cells are responsible for disease induction while Th2 cells can be protective. To address the mechanisms of this differential behavior, we utilized organotypic murine entorhinal-hippocampal slice cultures to analyze interactions between myelin basic protein-specific Th1 and Th2 cells with microglial cells. While both Th1 and Th2 cells induced CD40 expression, only Th1 cells induced intercellular adhesion molecule-1 (ICAM-1) expression on microglia. Moreover, Th2 cells prevented or even reversed Th1-induced ICAM-1 upregulation. Evidently, Th2 cells could diminish Th1-induced inflammatory reactions and actively support the resting state of microglia, which could be one mechanism of Th2-mediated remission of neuroinflammation during EAE.
Collapse
Affiliation(s)
- U Gimsa
- Department of Cell and Neurobiology, Institute of Anatomy, Humboldt-University Clinic Charité, D-10098, Berlin, Germany.
| | | | | | | | | |
Collapse
|