1
|
Zhang X, Zhang Y, Wu T, He H, Peng R, Jin K, Mo H, Qu F, Tang J, Zhou Y, Yang Y, Zhou Z, Fan J, Li J, Liu Z. Fish decay-accelerating factor (DAF) regulates intestinal complement pathway and immune response to bacterial challenge. FISH & SHELLFISH IMMUNOLOGY 2024; 151:109741. [PMID: 38964436 DOI: 10.1016/j.fsi.2024.109741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/25/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024]
Abstract
Decay-accelerating factor (DAF) is an essential member of the complement regulatory protein family that plays an important role in immune response and host homeostasis in mammals. However, the immune function of DAF has not been well characterized in bony fish. In this study, a complement regulatory protein named CiDAF was firstly characterized from Ctenopharyngodon idella and its potential roles were investigated in intestine following bacterial infection. Similar to mammalian DAFs, CiDAF has multiple complement control protein (CCP) functional domains, suggesting the evolutionary conservation of DAFs. CiDAF was broadly expressed in all tested tissues, with a relatively high expression level detected in the spleen and kidney. In vivo immune challenge experiments revealed that CiDAF strongly responded to bacterial pathogens (Aeromonas hydrophila and Aeromonas veronii) and PAMPs (lipopolysaccharide (LPS) or muramyl dipeptide (MDP)) challenges. In vitro RNAi experiments indicated that knockdown of CiDAF could upregulate the expression of complement genes (C4b, C5 and C7) and inflammatory cytokines (TNF-α, IL-1β and IL-8). Moreover, 2000 ng/mL of CiDAF agonist progesterone effectively alleviated LPS- or MDP-induced intestinal inflammation by regulating expression of complement factors, TLR/PepT1 pathway genes and inflammatory cytokines. Overall, these findings revealed that CiDAF may act as a negative regulator of intestinal complement pathway and immune response to bacterial challenge in grass carp.
Collapse
Affiliation(s)
- Xia Zhang
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Department of Biological and Chemical Engineering, Changsha University, Changsha, 410022, China; State Key Laboratory of Developmental Biology of Freshwater Fish, Department of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Yuhan Zhang
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Department of Biological and Chemical Engineering, Changsha University, Changsha, 410022, China
| | - Ting Wu
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Department of Biological and Chemical Engineering, Changsha University, Changsha, 410022, China
| | - Hao He
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Department of Biological and Chemical Engineering, Changsha University, Changsha, 410022, China
| | - Ran Peng
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Department of Biological and Chemical Engineering, Changsha University, Changsha, 410022, China; State Key Laboratory of Developmental Biology of Freshwater Fish, Department of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Kelan Jin
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Department of Biological and Chemical Engineering, Changsha University, Changsha, 410022, China
| | - Huilan Mo
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Department of Biological and Chemical Engineering, Changsha University, Changsha, 410022, China
| | - Fufa Qu
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Department of Biological and Chemical Engineering, Changsha University, Changsha, 410022, China.
| | - Jianzhou Tang
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Department of Biological and Chemical Engineering, Changsha University, Changsha, 410022, China
| | - Yonghua Zhou
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Department of Biological and Chemical Engineering, Changsha University, Changsha, 410022, China
| | - Yalin Yang
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Zhigang Zhou
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Junde Fan
- Yueyang Yumeikang Biotechnology Co., Ltd., Yueyang, 414100, China
| | - Jianzhong Li
- State Key Laboratory of Developmental Biology of Freshwater Fish, Department of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Zhen Liu
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Department of Biological and Chemical Engineering, Changsha University, Changsha, 410022, China.
| |
Collapse
|
2
|
Microglial Pruning: Relevance for Synaptic Dysfunction in Multiple Sclerosis and Related Experimental Models. Cells 2021; 10:cells10030686. [PMID: 33804596 PMCID: PMC8003660 DOI: 10.3390/cells10030686] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/12/2021] [Accepted: 03/17/2021] [Indexed: 12/11/2022] Open
Abstract
Microglia, besides being able to react rapidly to a wide range of environmental changes, are also involved in shaping neuronal wiring. Indeed, they actively participate in the modulation of neuronal function by regulating the elimination (or “pruning”) of weaker synapses in both physiologic and pathologic processes. Mounting evidence supports their crucial role in early synaptic loss, which is emerging as a hallmark of several neurodegenerative diseases, including multiple sclerosis (MS) and its preclinical models. MS is an inflammatory, immune-mediated pathology of the white matter in which demyelinating lesions may cause secondary neuronal death. Nevertheless, primitive grey matter (GM) damage is emerging as an important contributor to patients’ long-term disability, since it has been associated with early and progressive cognitive decline (CD), which seriously worsens the quality of life of MS patients. Widespread synapse loss even in the absence of demyelination, axon degeneration and neuronal death has been demonstrated in different GM structures, thus raising the possibility that synaptic dysfunction could be an early and possibly independent event in the neurodegenerative process associated with MS. This review provides an overview of microglial-dependent synapse elimination in the neuroinflammatory process that underlies MS and its experimental models.
Collapse
|
3
|
Schartz ND, Tenner AJ. The good, the bad, and the opportunities of the complement system in neurodegenerative disease. J Neuroinflammation 2020; 17:354. [PMID: 33239010 PMCID: PMC7690210 DOI: 10.1186/s12974-020-02024-8] [Citation(s) in RCA: 158] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 11/04/2020] [Indexed: 02/06/2023] Open
Abstract
The complement cascade is a critical effector mechanism of the innate immune system that contributes to the rapid clearance of pathogens and dead or dying cells, as well as contributing to the extent and limit of the inflammatory immune response. In addition, some of the early components of this cascade have been clearly shown to play a beneficial role in synapse elimination during the development of the nervous system, although excessive complement-mediated synaptic pruning in the adult or injured brain may be detrimental in multiple neurogenerative disorders. While many of these later studies have been in mouse models, observations consistent with this notion have been reported in human postmortem examination of brain tissue. Increasing awareness of distinct roles of C1q, the initial recognition component of the classical complement pathway, that are independent of the rest of the complement cascade, as well as the relationship with other signaling pathways of inflammation (in the periphery as well as the central nervous system), highlights the need for a thorough understanding of these molecular entities and pathways to facilitate successful therapeutic design, including target identification, disease stage for treatment, and delivery in specific neurologic disorders. Here, we review the evidence for both beneficial and detrimental effects of complement components and activation products in multiple neurodegenerative disorders. Evidence for requisite co-factors for the diverse consequences are reviewed, as well as the recent studies that support the possibility of successful pharmacological approaches to suppress excessive and detrimental complement-mediated chronic inflammation, while preserving beneficial effects of complement components, to slow the progression of neurodegenerative disease.
Collapse
Affiliation(s)
- Nicole D. Schartz
- Department of Molecular Biology and Biochemistry, University of California Irvine, 3205 McGaugh Hall, Irvine, CA 92697 USA
| | - Andrea J. Tenner
- Department of Molecular Biology and Biochemistry, University of California Irvine, 3205 McGaugh Hall, Irvine, CA 92697 USA
- Department of Neurobiology and Behavior, University of California Irvine, 3205 McGaugh Hall, Irvine, CA 92697 USA
- Department of Pathology and Laboratory Medicine, University of California Irvine, 3205 McGaugh Hall, Irvine, CA 92697 USA
| |
Collapse
|
4
|
Hammond JW, Bellizzi MJ, Ware C, Qiu WQ, Saminathan P, Li H, Luo S, Ma SA, Li Y, Gelbard HA. Complement-dependent synapse loss and microgliosis in a mouse model of multiple sclerosis. Brain Behav Immun 2020; 87:739-750. [PMID: 32151684 PMCID: PMC8698220 DOI: 10.1016/j.bbi.2020.03.004] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 03/04/2020] [Accepted: 03/05/2020] [Indexed: 12/14/2022] Open
Abstract
Multiple sclerosis (MS) is an inflammatory, neurodegenerative disease of the CNS characterized by both grey and white matter injury. Microglial activation and a reduction in synaptic density are key features of grey matter pathology that can be modeled with MOG35-55 experimental autoimmune encephalomyelitis (EAE). Complement deposition combined with microglial engulfment has been shown during normal development and in disease as a mechanism for pruning synapses. We tested whether there is excess complement production in the EAE hippocampus and whether complement-dependent synapse loss is a source of degeneration in EAE using C1qa and C3 knockout mice. We found that C1q and C3 protein and mRNA levels were elevated in EAE mice. Genetic loss of C3 protected mice from EAE-induced synapse loss, reduced microglial activation, decreased the severity of the EAE clinical score, and protected memory/freezing behavior after contextual fear conditioning. C1qa KO mice with EAE showed little to no change on these measurements compared to WT EAE mice. Thus, pathologic expression and activation of the early complement pathway, specifically at the level of C3, contributes to hippocampal grey matter pathology in the EAE.
Collapse
Affiliation(s)
- Jennetta W. Hammond
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642.,Department of Neurology, University of Rochester Medical
Center, 601 Elmwood Avenue, Rochester NY 14642.,Correspondence: Jennetta W. Hammond,
University of Rochester, Center for Neurotherapeutics Discovery, 601 Elmwood
Avenue, Box 645, Rochester, NY 14642, USA,
, Phone:
1-585-273-2872
| | - Matthew J. Bellizzi
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642.,Department of Neurology, University of Rochester Medical
Center, 601 Elmwood Avenue, Rochester NY 14642.,Department of Neuroscience, University of Rochester Medical
Center, 601 Elmwood Avenue, Rochester NY 14642
| | - Caroline Ware
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642.,Department of Neurology, University of Rochester Medical
Center, 601 Elmwood Avenue, Rochester NY 14642
| | - Wen Q. Qiu
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642.,Department of Neurology, University of Rochester Medical
Center, 601 Elmwood Avenue, Rochester NY 14642
| | - Priyanka Saminathan
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642.,Department of Microbiology and Immunology, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642
| | - Herman Li
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642
| | - Shaopeiwen Luo
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642
| | - Stefanie A. Ma
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642
| | - Yuanhao Li
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642
| | - Harris A. Gelbard
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642.,Department of Neurology, University of Rochester Medical
Center, 601 Elmwood Avenue, Rochester NY 14642.,Department of Neuroscience, University of Rochester Medical
Center, 601 Elmwood Avenue, Rochester NY 14642
| |
Collapse
|
5
|
Chen Y, Li R, Wu AM, Shu YQ, Lu ZQ, Hu XQ. The complement and immunoglobulin levels in NMO patients. Neurol Sci 2013; 35:215-20. [DOI: 10.1007/s10072-013-1481-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 06/11/2013] [Indexed: 10/26/2022]
|
6
|
Hu X, Holers VM, Thurman JM, Schoeb TR, Ramos TN, Barnum SR. Therapeutic inhibition of the alternative complement pathway attenuates chronic EAE. Mol Immunol 2013; 54:302-8. [PMID: 23337717 PMCID: PMC3602149 DOI: 10.1016/j.molimm.2012.12.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2012] [Accepted: 12/14/2012] [Indexed: 12/13/2022]
Abstract
Previous studies from our laboratory using complement-mutant mice demonstrated that the alternative pathway is the dominant activation pathway responsible for complement-mediated pathology in demyelinating disease. Using a well-characterized inhibitory monoclonal antibody (mAb 1379) directed against mouse factor B, we assessed the therapeutic value of inhibiting the alternative complement pathway in experimental autoimmune encephalomyelitis (EAE), the animal model for multiple sclerosis. Administration of anti-factor B antibody to mice prior to the onset of clinical signs of active EAE had no affect on the onset or acute phase of disease, but significantly attenuated the chronic phase of disease resulting in reduced cellular infiltration, inflammation and demyelination in antibody-treated mice. Attenuation of the chronic phase of disease was long lasting even though antibody administration was terminated shortly after disease onset. Chronic disease was also attenuated in transferred EAE when anti-factor B antibody was administered before or after disease onset. Similar levels of disease attenuation were observed in transferred EAE using MOG-specific encephalitogenic T cells. These studies demonstrate the therapeutic potential for inhibition of factor B in the chronic phase of demyelinating disease, where treatment options are limited.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacology
- Complement Factor B/classification
- Complement Pathway, Alternative/drug effects
- Complement Pathway, Alternative/immunology
- Complement System Proteins/immunology
- Demyelinating Diseases/immunology
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Inflammation/immunology
- Mice
- Mice, Inbred C57BL
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Xianzhen Hu
- Department of Microbiology, University of Alabama at Birmingham, 845 19th St. S., Birmingham, AL 35294, USA
| | | | | | | | | | | |
Collapse
|
7
|
Inglis HR, Greer JM, McCombe PA. Gene expression in the spinal cord in female lewis rats with experimental autoimmune encephalomyelitis induced with myelin basic protein. PLoS One 2012; 7:e48555. [PMID: 23139791 PMCID: PMC3491034 DOI: 10.1371/journal.pone.0048555] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2012] [Accepted: 09/27/2012] [Indexed: 12/21/2022] Open
Abstract
Background Experimental autoimmune encephalomyelitis (EAE), the best available model of multiple sclerosis, can be induced in different animal strains using immunization with central nervous system antigens. EAE is associated with inflammation and demyelination of the nervous system. Micro-array can be used to investigate gene expression and biological pathways that are altered during disease. There are few studies of the changes in gene expression in EAE, and these have mostly been done in a chronic mouse EAE model. EAE induced in the Lewis with myelin basic protein (MBP-EAE) is well characterised, making it an ideal candidate for the analysis of gene expression in this disease model. Methodology/Principal Findings MBP-EAE was induced in female Lewis rats by inoculation with MBP and adjuvants. Total RNA was extracted from the spinal cords and used for micro-array analysis using AffimetrixGeneChip Rat Exon 1.0 ST Arrays. Gene expression in the spinal cords was compared between healthy female rats and female rats with MBP-EAE. Gene expression in the spinal cord of rats with MBP-EAE differed from that in the spinal cord of normal rats, and there was regulation of pathways involved with immune function and nervous system function. For selected genes the change in expression was confirmed with real-time PCR. Conclusions/Significance EAE leads to modulation of gene expression in the spinal cord. We have identified the genes that are most significantly regulated in MBP-EAE in the Lewis rat and produced a profile of gene expression in the spinal cord at the peak of disease.
Collapse
Affiliation(s)
- Hayley R. Inglis
- University of Queensland Centre for Clinical Research, Brisbane, Queensland, Australia
| | - Judith M. Greer
- University of Queensland Centre for Clinical Research, Brisbane, Queensland, Australia
| | - Pamela A. McCombe
- University of Queensland Centre for Clinical Research, Brisbane, Queensland, Australia
- * E-mail:
| |
Collapse
|
8
|
Hu X, Tomlinson S, Barnum SR. Targeted inhibition of complement using complement receptor 2-conjugated inhibitors attenuates EAE. Neurosci Lett 2012; 531:35-9. [PMID: 23079547 DOI: 10.1016/j.neulet.2012.10.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 10/04/2012] [Accepted: 10/07/2012] [Indexed: 01/27/2023]
Abstract
Multiple sclerosis (MS) is the most common autoimmune demyelinating disease, affecting millions of individuals worldwide. In the last two decades, many therapeutic options for the treatment of MS have become available, however they are limited in terms of effectiveness and some remain plagued by safety issues. The currently available treatment options target relapsing remitting forms of MS and are not effective against the more progressive forms of the disease. These limitations highlight a significant unmet treatment need for MS. In experimental autoimmune encephalomyelitis (EAE) studies from our laboratory, we have previously shown, using a number of complement mutant and transgenic mice, that inhibition of the alternative complement pathway and the C3 convertase confers significant protection from disease. We report here that targeted inhibition of complement activation using complement receptor 2 (CR2)-conjugated inhibitors significantly attenuates EAE. Administration of CR2-Crry (blocks all complement pathways at C3 activation) and CR2-fH (specifically blocks the alternative pathway) just prior to and during the onset of EAE blocks progression of both acute and chronic disease. These data indicate that inhibition of complement may offer an effective therapeutic approach to treating both acute and chronic forms of demyelinating disease through blocking the alternative pathway or complement convertases.
Collapse
Affiliation(s)
- Xianzhen Hu
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | |
Collapse
|
9
|
Ramos TN, Darley MM, Weckbach S, Stahel PF, Tomlinson S, Barnum SR. The C5 convertase is not required for activation of the terminal complement pathway in murine experimental cerebral malaria. J Biol Chem 2012; 287:24734-8. [PMID: 22689574 DOI: 10.1074/jbc.c112.378364] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Cerebral malaria (CM) is the most severe manifestation of clinical malaria syndromes and has a high fatality rate especially in the developing world. Recent studies demonstrated that C5(-/-) mice are resistant to experimental CM (ECM) and that protection was due to the inability to form the membrane attack complex. Unexpectedly, we observed that C4(-/-) and factor B(-/-) mice were fully susceptible to disease, indicating that activation of the classical or alternative pathways is not required for ECM. C3(-/-) mice were also susceptible to ECM, indicating that the canonical C5 convertases are not required for ECM development and progression. Abrogation of ECM by treatment with anti-C9 antibody and detection of C5a in serum of C3(-/-) mice confirmed that C5 activation occurs in ECM independent of C5 convertases. Our data indicate that activation of C5 in ECM likely occurs via coagulation enzymes of the extrinsic protease pathway.
Collapse
Affiliation(s)
- Theresa N Ramos
- Department of Microbiology, University of Alabama, Birmingham, Alabama 35294, USA
| | | | | | | | | | | |
Collapse
|
10
|
Ingram G, Hakobyan S, Hirst CL, Harris CL, Loveless S, Mitchell JP, Pickersgill TP, Robertson NP, Morgan BP. Systemic complement profiling in multiple sclerosis as a biomarker of disease state. Mult Scler 2012; 18:1401-11. [PMID: 22354735 PMCID: PMC3697901 DOI: 10.1177/1352458512438238] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND There is increasing evidence of significant and dynamic systemic activation and upregulation of complement in multiple sclerosis (MS), which may contribute to disease pathogenesis. OBJECTIVE We aimed to investigate the pathological role of complement in MS and the potential role for complement profiling as a biomarker of MS disease state. METHODS Key components of the classical, alternative and terminal pathways of complement were measured in plasma and cerebrospinal fluid (CSF) of patients with MS in different clinical phases of disease and in matched controls. RESULTS Increased plasma levels of C3 (p<0.003), C4 (p<0.001), C4a (p<0.001), C1 inhibitor (p<0.001), and factor H (p<0.001), and reduced levels of C9 (p<0.001) were observed in MS patients compared with controls. Combined profiling of these analytes produced a statistical model with a predictive value of 97% for MS and 73% for clinical relapse when combined with selected demographic data. CSF-plasma correlations suggested that source of synthesis of these components was both systemic and central. CONCLUSION These data provide further evidence of alterations in both local and systemic expression and activation of complement in MS and suggest that complement profiling may be informative as a biomarker of MS disease, although further work is needed to determine its use in distinguishing MS from its differential.
Collapse
Affiliation(s)
- G Ingram
- Department of Neurology, University Hospital of Wales, Cardiff, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Blanchet L, Smolinska A, Attali A, Stoop MP, Ampt KAM, van Aken H, Suidgeest E, Tuinstra T, Wijmenga SS, Luider T, Buydens LMC. Fusion of metabolomics and proteomics data for biomarkers discovery: case study on the experimental autoimmune encephalomyelitis. BMC Bioinformatics 2011; 12:254. [PMID: 21696593 PMCID: PMC3225201 DOI: 10.1186/1471-2105-12-254] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Accepted: 06/22/2011] [Indexed: 01/22/2023] Open
Abstract
Background Analysis of Cerebrospinal Fluid (CSF) samples holds great promise to diagnose neurological pathologies and gain insight into the molecular background of these pathologies. Proteomics and metabolomics methods provide invaluable information on the biomolecular content of CSF and thereby on the possible status of the central nervous system, including neurological pathologies. The combined information provides a more complete description of CSF content. Extracting the full combined information requires a combined analysis of different datasets i.e. fusion of the data. Results A novel fusion method is presented and applied to proteomics and metabolomics data from a pre-clinical model of multiple sclerosis: an Experimental Autoimmune Encephalomyelitis (EAE) model in rats. The method follows a mid-level fusion architecture. The relevant information is extracted per platform using extended canonical variates analysis. The results are subsequently merged in order to be analyzed jointly. We find that the combined proteome and metabolome data allow for the efficient and reliable discrimination between healthy, peripherally inflamed rats, and rats at the onset of the EAE. The predicted accuracy reaches 89% on a test set. The important variables (metabolites and proteins) in this model are known to be linked to EAE and/or multiple sclerosis. Conclusions Fusion of proteomics and metabolomics data is possible. The main issues of high-dimensionality and missing values are overcome. The outcome leads to higher accuracy in prediction and more exhaustive description of the disease profile. The biological interpretation of the involved variables validates our fusion approach.
Collapse
Affiliation(s)
- Lionel Blanchet
- Radboud University Nijmegen, Institute for Molecules and Materials, Heyendaalseweg, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Abstract
Complement is a part of the innate immune system that contributes to first-line host defense. It is also implicated in a number of human inflammatory conditions and has attracted interest as a potential therapeutic target. Understanding the basic biology of complement and its mechanism(s) of action is imperative for developing complement-based treatments for infectious and autoimmune diseases. One of the exciting new developments in this regard is the revelation that complement plays an important role in T cell immunity. In this review, we highlight recent published studies implicating complement in models of CD4+ and CD8+ T cell immune responses, and discuss its potential mechanism(s) action in these processes. We also comment on issues that may impact data interpretation and draw attention to their consideration in future studies.
Collapse
Affiliation(s)
- Jason R Dunkelberger
- Institute for Translational Medicine and Therapeutics and Department of Pharmacology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Wen-Chao Song
- Institute for Translational Medicine and Therapeutics and Department of Pharmacology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
13
|
Abstract
The complement (C) system plays a central role in innate immunity and bridges innate and adaptive immune responses. A fine balance of C activation and regulation mediates the elimination of invading pathogens and the protection of the host from excessive C deposition on healthy tissues. If this delicate balance is disrupted, the C system may cause injury and contribute to the pathogenesis of various diseases, including neurodegenerative disorders and neuropathies. Here we review evidence indicating that C factors and regulators are locally synthesized in the nervous system and we discuss the evidence supporting the protective or detrimental role of C activation in health, injury, and disease of the nerve.
Collapse
Affiliation(s)
- V Ramaglia
- Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | |
Collapse
|
14
|
Deletion of both the C3a and C5a receptors fails to protect against experimental autoimmune encephalomyelitis. Neurosci Lett 2009; 467:234-6. [PMID: 19850104 DOI: 10.1016/j.neulet.2009.10.045] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2009] [Revised: 10/12/2009] [Accepted: 10/13/2009] [Indexed: 11/21/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune disease in which inflammation, leukocyte infiltration, and ultimately, demyelination occur as a result of innate and adaptive immune-mediated mechanisms. The pathophysiological role of the complement system, a major component of innate immunity, in the development and progression of experimental autoimmune encephalomyelitis (EAE), the animal model for MS has been extensively examined. Previous studies from our lab have shown that the complement receptor for the anaphylatoxin C3a, but not for C5a plays an important role in EAE. Based on the important contributions of the complement anaphylatoxin receptors to other inflammatory conditions in the CNS, we reasoned that deletion of both receptors may reveal underlying interactions between them that are important to EAE pathology. We performed EAE in C3aR/C5aR double knockout mice (C3aR/C5aR(-/-)) and observed delayed onset of disease but no attenuation of disease severity compared to wild type mice. Interestingly there was trend toward greater infiltration of CD4(+), but not CD8(+) T cells, in C3aR/C5aR(-/-) mice with EAE, suggesting altered trafficking of these cells. Antigen-specific T cells isolated from C3aR/C5aR(-/-) mice during acute EAE produced elevated levels of TNF-alpha, but markedly reduced levels of IFN-gamma and IL-12 compared to wild type mice. It remains unclear how the changes in these disease parameters contribute to the loss of the protective effect seen in C3aR(-/-) mice, however our data indicate a level of cross-modulation between the C3aR and C5aR during EAE.
Collapse
|
15
|
Saikali P, Cayrol R, Vincent T. Anti-aquaporin-4 auto-antibodies orchestrate the pathogenesis in neuromyelitis optica. Autoimmun Rev 2009; 9:132-5. [PMID: 19389490 DOI: 10.1016/j.autrev.2009.04.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2009] [Accepted: 04/15/2009] [Indexed: 10/20/2022]
Abstract
NMO-IgG, the auto-antibody specific to the aquaporin-4 (AQP4) water channel associated with the autoimmune inflammatory disease neuromyelitis optica (NMO), is considered to be an accurate serum biomarker and is thought to be an important contributor to NMO pathology. In this review, we summarize recent evidences from our group and others indicating that NMO-IgG can be implicated at several levels in the immuno-pathology of NMO. NMO-IgG/anti-AQP4 antibodies may compromise the integrity of the blood-brain barrier and consequently facilitate and enhance the perivascular inflammation characteristic of NMO. Lastly, NMO-IgG can induce astrocyte injury which may lead to the accumulation of excitatory/toxic molecules and accordingly damage oligodendrocytes and compromise myelin integrity.
Collapse
Affiliation(s)
- Philippe Saikali
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, Québec, Canada
| | | | | |
Collapse
|
16
|
Griffiths MR, Neal JW, Fontaine M, Das T, Gasque P. Complement factor H, a marker of self protects against experimental autoimmune encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2009; 182:4368-77. [PMID: 19299737 DOI: 10.4049/jimmunol.0800205] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The CNS innate immune response is a "double-edged sword" representing a fine balance between protective antipathogen responses and detrimental neurocytotoxic effects. Hence, it is important to identify the key regulatory mechanisms involved in the control of CNS innate immunity and which could be harnessed to explore novel therapeutic avenues. In analogy to the newly described neuroimmune regulatory proteins also known as "don't eat me" signals (CD200, CD47, CD22, fractalkine, semaphorins), we herein identify the key role of complement regulator factor H (fH) in controlling neuroinflammation initiated in an acute mouse model of Ab-dependent experimental autoimmune encephalomyelitis. Mouse fH was found to be abundantly expressed by primary cultured neurons and neuronal cell lines (N1E115 and Neuro2a) at a level comparable to BV2 microglia and CLTT astrocytes. Mouse neurons expressed other complement regulators crry and low levels of CD55. In the brain, the expression of fH was localized to neuronal bodies and axons, endothelial cells, microglia but not oligodendrocytes and myelin sheaths and was dramatically reduced in inflammatory experimental autoimmune encephalomyelitis settings. When exogenous human fH was administered to disease Ab-dependent experimental autoimmune encephalomyelitis animals, there was a significant decrease in clinical score, inflammation, and demyelination, as compared with PBS-injected animals. We found that the accumulation of human fH in the brain parenchyma protected neurons from complement opsonization, axonal injury, and leukocyte infiltration. Our data argue for a key regulatory activity of fH in neuroprotection and provide novel therapeutic avenues for CNS chronic inflammatory diseases.
Collapse
Affiliation(s)
- Mark R Griffiths
- Department of Medical Biochemistry, Cardiff University, United Kingdom
| | | | | | | | | |
Collapse
|
17
|
Ingram G, Hakobyan S, Robertson NP, Morgan BP. Complement in multiple sclerosis: its role in disease and potential as a biomarker. Clin Exp Immunol 2009; 155:128-39. [PMID: 19040603 PMCID: PMC2675242 DOI: 10.1111/j.1365-2249.2008.03830.x] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2008] [Indexed: 01/27/2023] Open
Abstract
Multiple sclerosis (MS) is a common inflammatory disease of the central nervous system with a poorly defined and complex immunopathogenesis. Although initiated by reactive T cells, persistent inflammation is evident throughout the disease course. A contribution from complement has long been suspected, based on the results of pathological and functional studies which have demonstrated complement activation products in MS brain and biological fluids. However, the extent and nature of complement activation and its contribution to disease phenotype and long-term outcome remain unclear. Furthermore, functional polymorphisms in components and regulators of the complement system which cause dysregulation, and are known to contribute to other autoimmune inflammatory disorders, have not been investigated to date in MS in any detail. In this paper we review evidence from pathological, animal model and human functional and genetic studies, implicating activation of complement in MS. We also evaluate the potential of complement components and regulators and their polymorphic variants as biomarkers of disease, and suggest appropriate directions for future research.
Collapse
Affiliation(s)
- G Ingram
- Department of Neurosciences, Cardiff University, Heath Park, Cardiff, UK
| | | | | | | |
Collapse
|
18
|
Alexander JJ, Anderson AJ, Barnum SR, Stevens B, Tenner AJ. The complement cascade: Yin-Yang in neuroinflammation--neuro-protection and -degeneration. J Neurochem 2008; 107:1169-87. [PMID: 18786171 DOI: 10.1111/j.1471-4159.2008.05668.x] [Citation(s) in RCA: 136] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The complement cascade has long been recognized to play a key role in inflammatory and degenerative diseases. It is a 'double edged' sword as it is necessary to maintain health, yet can have adverse effects when unregulated, often exacerbating disease. The contrasting effects of complement, depending on whether in a setting of health or disease, is the price paid to achieve flexibility in scope and degree of a protective response for the host from infection and injury. Loss or even decreased efficiency of critical regulatory control mechanisms can result in aggravated inflammation and destruction of self-tissue. The role of the complement cascade is poorly understood in the nervous system and neurological disorders. Novel studies have demonstrated that the expression of complement proteins in brain varies in different cell types and the effects of complement activation in various disease settings appear to differ. Understanding the functioning of this cascade is essential, as it has therapeutic implications. In this review, we will attempt to provide insight into how this complex cascade functions and to identify potential strategic targets for therapeutic intervention in chronic diseases as well as acute injury in the CNS.
Collapse
|
19
|
Smith SS, Ludwig M, Wohler JE, Bullard DC, Szalai AJ, Barnum SR. Deletion of both ICAM-1 and C3 enhances severity of experimental autoimmune encephalomyelitis compared to C3-deficient mice. Neurosci Lett 2008; 442:158-60. [PMID: 18634851 PMCID: PMC2556246 DOI: 10.1016/j.neulet.2008.07.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2008] [Revised: 07/01/2008] [Accepted: 07/02/2008] [Indexed: 12/19/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune disease characterized by central nervous system (CNS) inflammation and leukocyte infiltration, demyelination of neurons, and blood-brain barrier breakdown. The development of experimental autoimmune encephalomyelitis (EAE), the animal model for MS is dependent on a number of components of the immune system including complement and adhesion molecules. Previous studies in our lab have examined the role of C3, the central complement component, and intercellular adhesion molecule-1 (ICAM-1) a key cell adhesion molecule involved in leukocyte trafficking to sites of inflammation including the CNS. In these studies we demonstrated that myelin oligodendrocyte glycoprotein (MOG)-induced EAE is markedly attenuated in both ICAM-1(-/-) and C3(-/-) mice. Given the pivotal role that these proteins play in EAE, we hypothesized that EAE in ICAM-1(-/-) and C3(-/-) double mutant mice would likely fail to develop. Unexpectedly, EAE in ICAM-1(-/-)xC3(-/-) mice was only modestly attenuated compared to wild type mice and significantly worse than C3(-/-) mice. Leukocyte infiltration was commensurate with disease severity between the three groups of mice. Spinal cord T cells from ICAM-1(-/-)xC3(-/-) mice produced the highest levels of IFN-gamma and TNF-alpha, despite reduced disease severity compared to wild type mice. The mechanisms behind the elevated EAE severity in ICAM-1(-/-)xC3(-/-) mice may relate to altered homing of leukocytes or processing of self-antigens in the double mutant background.
Collapse
MESH Headings
- Animals
- Antigens, CD/metabolism
- Cell Proliferation/drug effects
- Complement C3/deficiency
- Complement C3/genetics
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/physiopathology
- Gene Expression Regulation/drug effects
- Gene Expression Regulation/genetics
- Intercellular Adhesion Molecule-1/genetics
- Intercellular Adhesion Molecule-1/physiology
- Leukocytes/metabolism
- Mice
- Mice, Knockout
- Myelin Proteins
- Myelin-Associated Glycoprotein/adverse effects
- Myelin-Oligodendrocyte Glycoprotein
- Severity of Illness Index
- Spinal Cord/pathology
- T-Lymphocytes/drug effects
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- Sherry S. Smith
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Michael Ludwig
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Jillian E. Wohler
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Daniel C. Bullard
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Alex J. Szalai
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Scott R. Barnum
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35294
| |
Collapse
|
20
|
Ramaglia V, Daha M, Baas F. The complement system in the peripheral nerve: Friend or foe? Mol Immunol 2008; 45:3865-77. [DOI: 10.1016/j.molimm.2008.06.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2008] [Revised: 06/09/2008] [Accepted: 06/13/2008] [Indexed: 12/21/2022]
|
21
|
Templeton SP, Perlman S. Pathogenesis of acute and chronic central nervous system infection with variants of mouse hepatitis virus, strain JHM. Immunol Res 2008; 39:160-72. [PMID: 17917063 PMCID: PMC7090838 DOI: 10.1007/s12026-007-0079-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 01/12/2023]
Abstract
Infection of mice with variants of mouse hepatitis virus, strain JHM (MHV-JHM), provide models of acute and chronic viral infection of the central nervous system (CNS). Through targeted recombination and reverse genetic manipulation, studies of infection with MHV-JHM variants have identified phenotypic differences and examined the effects of these differences on viral pathogenesis and anti-viral host immune responses. Studies employing recombinant viruses with a modified spike (S) glycoprotein of MHV-JHM have identified the S gene as a major determinant of neurovirulence. However, the association of S gene variation and neurovirulence with host ability to generate anti-viral CD8 T cell responses is not completely clear. Partially protective anti-viral immune responses may result in persistent infection and chronic demyelinating disease characterized by myelin removal from axons of the CNS and associated with dense macrophage/microglial infiltration. Demyelinating disease during MHV-JHM infection is immune-mediated, as mice that lack T lymphocytes fail to develop disease despite succumbing to encephalitis with high levels of infectious virus in the CNS. However, the presence of T lymphocytes or anti-viral antibody can induce disease in infected immunodeficient mice. The mechanisms by which these immune effectors induce demyelination share an ability to activate and recruit macrophages and microglia, thus increasing the putative role of these cells in myelin destruction.
Collapse
Affiliation(s)
- Steven P Templeton
- Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA 52242, USA
| | | |
Collapse
|
22
|
Briggs DT, Martin CB, Ingersoll SA, Barnum SR, Martin BK. Astrocyte-specific expression of a soluble form of the murine complement control protein Crry confers demyelination protection in the cuprizone model. Glia 2007; 55:1405-15. [PMID: 17674370 DOI: 10.1002/glia.20551] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Complement has been implicated as a potential effector mechanism in neurodegeneration; yet the precise role of complement in this process remains elusive. In this report, we have utilized the cuprizone model of demyelination-remyelination to examine the contribution of complement to disease. C1q deposition was observed in the corpus callosum of C57BL/6 mice during demyelination, suggesting complement activation by apoptotic oligodendrocyte debris. Simultaneously, these mice lost expression of the rodent complement regulatory protein, Crry. A soluble CNS-specific form of the Crry protein (sCrry) expressed in a transgenic mouse under the control of an astrocyte-specific promoter was induced in the corpus callosum during cuprizone treatment. Expression of this protein completely protected the mice from demyelination. Interestingly, sCrry mice had low levels of demyelination at later times when control mice were remyelinating. Although the sCrry transgenic mice had lower levels of demyelination, there was no decrease in overall cellularity, however there were decreased numbers of microglia in the sCrry mice relative to controls. Strikingly, sCrry mice had early recovery of mature oligodendrocytes, although they later disappeared. TUNEL staining suggested that production of the sCrry protein in the transgenic mice protected from a late apoptosis event at 3 weeks of cuprizone treatment. Our data suggest complement provides some protection of mature oligodendrocytes during cuprizone treatment but may be critical for subsequent remyelination events. These data suggest that temporal restriction of complement inhibition may be required in some disease settings.
Collapse
MESH Headings
- Animals
- Apoptosis/physiology
- Astrocytes/immunology
- Astrocytes/metabolism
- Brain/immunology
- Brain/metabolism
- Brain/physiopathology
- Chelating Agents
- Complement System Proteins/immunology
- Complement System Proteins/metabolism
- Corpus Callosum/immunology
- Corpus Callosum/metabolism
- Corpus Callosum/physiopathology
- Cuprizone
- Cytoprotection/genetics
- Demyelinating Autoimmune Diseases, CNS/chemically induced
- Demyelinating Autoimmune Diseases, CNS/genetics
- Demyelinating Autoimmune Diseases, CNS/immunology
- Disease Models, Animal
- Glial Fibrillary Acidic Protein/genetics
- Gliosis/genetics
- Gliosis/immunology
- Gliosis/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Nerve Regeneration/genetics
- Oligodendroglia/immunology
- Receptors, Complement/genetics
- Receptors, Complement/immunology
- Receptors, Complement/metabolism
- Receptors, Complement 3b
- Time Factors
Collapse
Affiliation(s)
- Dustin T Briggs
- Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa 52242, USA
| | | | | | | | | |
Collapse
|
23
|
Ramaglia V, King RHM, Nourallah M, Wolterman R, de Jonge R, Ramkema M, Vigar MA, van der Wetering S, Morgan BP, Troost D, Baas F. The membrane attack complex of the complement system is essential for rapid Wallerian degeneration. J Neurosci 2007; 27:7663-72. [PMID: 17634361 PMCID: PMC6672891 DOI: 10.1523/jneurosci.5623-06.2007] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The complement (C) system plays an important role in myelin breakdown during Wallerian degeneration (WD). The pathway and mechanism involved are, however, not clear. In a crush injury model of the sciatic nerve, we show that C6, necessary for the assembly of the membrane attack complex (MAC), is essential for rapid WD. At 3 d after injury, pronounced WD occurred in wild-type animals, whereas the axons and myelin of C6-deficient animals appeared intact. Macrophage recruitment and activation was inhibited in C6-deficient rats. However, 7 d after injury, the distal part of the C6-deficient nerves appeared degraded. As a consequence of a delayed WD, more myelin breakdown products were present than in wild-type nerves. Reconstitution of the C6-deficient animals with C6 restored the wild-type phenotype. Treatment with rhC1INH (recombinant human complement 1 inhibitor) blocked deposition of activated C-cleaved products after injury. These experiments demonstrate that the classical pathway of the complement system is activated after acute nerve trauma and that the entire complement cascade, including MAC deposition, is essential for rapid WD and efficient clearance of myelin after acute peripheral nerve trauma.
Collapse
Affiliation(s)
| | - Rosalind Helen Mary King
- Department of Clinical Neurosciences, Royal Free and University College Medical School, London NW3 2PF, United Kingdom
| | - Michelle Nourallah
- Department of Clinical Neurosciences, Royal Free and University College Medical School, London NW3 2PF, United Kingdom
| | | | | | - Marja Ramkema
- Department of Neuropathology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Miriam Ann Vigar
- Department of Medical Biochemistry and Immunology, School of Medicine, Cardiff University, Cardiff CF14 4XN, United Kingdom, and
| | | | - Brian Paul Morgan
- Department of Medical Biochemistry and Immunology, School of Medicine, Cardiff University, Cardiff CF14 4XN, United Kingdom, and
| | - Dirk Troost
- Department of Neuropathology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Frank Baas
- Neurogenetics Laboratory
- Department of Neurology, and
| |
Collapse
|
24
|
Szalai AJ, Hu X, Adams JE, Barnum SR. Complement in experimental autoimmune encephalomyelitis revisited: C3 is required for development of maximal disease. Mol Immunol 2007; 44:3132-6. [PMID: 17353050 PMCID: PMC1986644 DOI: 10.1016/j.molimm.2007.02.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2007] [Revised: 02/02/2007] [Accepted: 02/05/2007] [Indexed: 10/23/2022]
Abstract
Complement per se has been shown to play an important role in demyelinating disease but controversy remains regarding the role of C3 in the development and progression of experimental autoimmune encephalomyelitis (EAE), the animal model for multiple sclerosis. In this study, we used C3(-/-) mice to confirm previous findings that C3 is required for full development of EAE. Furthermore, C3(+/-) mice (with serum C3 levels 50% that of wild-type mice) developed EAE with a severity intermediate between wild-type and C3(-/-) mice. Importantly transfer of wild-type encephalitogenic T cells to C3(-/-) mice resulted in attenuated EAE. C3(-/-) mice with EAE had fewer CD4(+) and CD8(+) T cells in the CNS and 50% fewer of these cells produced IFN-gamma compared to wild-type mice. When treated with anti-CD3 antibody, CD4(+) T cells from wild-type and C3(-/-) mice had similar activation profiles as judged by IFN-gamma production and CD25 and CD69 expression, indicating there is no gross or intrinsic defect in T cells from C3(-/-) mice. T cells from primed C3(-/-) mice proliferated comparably to that of control T cells on re-stimulation with MOG peptide. Our results confirm a requirement for C3 for maximal development of EAE and suggest that receptors for C3-derived activation fragments might be a viable therapeutic target for prevention and treatment demyelinating disease.
Collapse
Affiliation(s)
- Alexander J. Szalai
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Xianzhen Hu
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Jillian E. Adams
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Scott R. Barnum
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35294
| |
Collapse
|