1
|
Veremeyko T, Barteneva NS, Vorobyev I, Ponomarev ED. The Emerging Role of Immunoglobulins and Complement in the Stimulation of Neuronal Activity and Repair: Not as Simple as We Thought. Biomolecules 2024; 14:1323. [PMID: 39456256 PMCID: PMC11506258 DOI: 10.3390/biom14101323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/20/2024] [Accepted: 09/30/2024] [Indexed: 10/28/2024] Open
Abstract
Neurologic disorders such as traumatic brain injury, multiple sclerosis, Alzheimer's disease, and drug-resistant epilepsy have a high socioeconomic impact around the world. Current therapies for these disorders are often not effective. This creates a demand for the development of new therapeutic approaches to treat these disorders. Recent data suggest that autoreactive naturally occurring immunoglobulins produced by subsets of B cells, called B1 B cells, combined with complement, are actively involved in the processes of restoration of neuronal functions during pathological conditions and remyelination. The focus of this review is to discuss the possibility of creating specific therapeutic antibodies that can activate and fix complement to enhance neuronal survival and promote central nervous system repair after injuries associated with many types of neurodegenerative diseases.
Collapse
Affiliation(s)
- Tatyana Veremeyko
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Astana 010000, Kazakhstan; (T.V.); (N.S.B.); (I.V.)
| | - Natasha S. Barteneva
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Astana 010000, Kazakhstan; (T.V.); (N.S.B.); (I.V.)
| | - Ivan Vorobyev
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Astana 010000, Kazakhstan; (T.V.); (N.S.B.); (I.V.)
- Laboratory of Cell Motility, National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan
| | - Eugene D. Ponomarev
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Astana 010000, Kazakhstan; (T.V.); (N.S.B.); (I.V.)
- Department of Biomedical Sciences, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong
| |
Collapse
|
2
|
Saez-Calveras N, Brewster AL, Stuve O. The validity of animal models to explore the pathogenic role of the complement system in multiple sclerosis: A review. Front Mol Neurosci 2022; 15:1017484. [PMID: 36311030 PMCID: PMC9606595 DOI: 10.3389/fnmol.2022.1017484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/26/2022] [Indexed: 11/26/2022] Open
Abstract
Animal models of multiple sclerosis (MS) have been extensively used to characterize the disease mechanisms in MS, as well as to identify potential pharmacologic targets for this condition. In recent years, the immune complement system has gained increased attention as an important effector in the pathogenesis of MS. Evidence from histological, serum, and CSF studies of patients supports an involvement of complement in both relapsing-remitting and progressive MS. In this review, we discuss the history and advances made on the use of MS animal models to profile the effects of the complement system in this condition. The first studies that explored the complement system in the context of MS used cobra venom factor (CVF) as a complement depleting agent in experimental autoimmune encephalomyelitis (EAE) Lewis rats. Since then, multiple mice and rat models of MS have revealed a role of C3 and the alternative complement cascade in the opsonization and phagocytosis of myelin by microglia and myeloid cells. Studies using viral vectors, genetic knockouts and pharmacologic complement inhibitors have also shown an effect of complement in synaptic loss. Antibody-mediated EAE models have revealed an involvement of the C1 complex and the classical complement as an effector of the humoral response in this disease. C1q itself may also be involved in modulating microglia activation and oligodendrocyte differentiation in these animals. In addition, animal and in vitro models have revealed that multiple complement factors may act as modulators of both the innate and adaptive immune responses. Finally, evidence gathered from mice models suggests that the membrane attack complex (MAC) may even exert protective roles in the chronic stages of EAE. Overall, this review summarizes the importance of MS animal models to better characterize the role of the complement system and guide future therapeutic approaches in this condition.
Collapse
Affiliation(s)
- Nil Saez-Calveras
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Neurology Section, Parkland Hospital, Dallas, TX, United States
| | - Amy L. Brewster
- Department of Biological Sciences, Southern Methodist University, Dallas, TX, United States
| | - Olaf Stuve
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Neurology Section, VA North Texas Health Care System, Dallas, TX, United States
- Peter O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, United States
- *Correspondence: Olaf Stuve,
| |
Collapse
|
3
|
Saez-Calveras N, Stuve O. The role of the complement system in Multiple Sclerosis: A review. Front Immunol 2022; 13:970486. [PMID: 36032156 PMCID: PMC9399629 DOI: 10.3389/fimmu.2022.970486] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 07/21/2022] [Indexed: 11/13/2022] Open
Abstract
The complement system has been involved in the pathogenesis of multiple neuroinflammatory and neurodegenerative conditions. In this review, we evaluated the possible role of complement activation in multiple sclerosis (MS) with a focus in progressive MS, where the disease pathogenesis remains to be fully elucidated and treatment options are limited. The evidence for the involvement of the complement system in the white matter plaques and gray matter lesions of MS stems from immunohistochemical analysis of post-mortem MS brains, in vivo serum and cerebrospinal fluid biomarker studies, and animal models of Experimental Autoimmune Encephalomyelitis (EAE). Complement knock-out studies in these animal models have revealed that this system may have a “double-edge sword” effect in MS. On the one hand, complement proteins may aid in promoting the clearance of myelin degradation products and other debris through myeloid cell-mediated phagocytosis. On the other, its aberrant activation may lead to demyelination at the rim of progressive MS white matter lesions as well as synapse loss in the gray matter. The complement system may also interact with known risk factors of MS, including as Epstein Barr Virus (EBV) infection, and perpetuate the activation of CNS self-reactive B cell populations. With the mounting evidence for the involvement of complement in MS, the development of complement modulating therapies for this condition is appealing. Herein, we also reviewed the pharmacological complement inhibitors that have been tested in MS animal models as well as in clinical trials for other neurologic diseases. The potential use of these agents, such as the C5-binding antibody eculizumab in MS will require a detailed understanding of the role of the different complement effectors in this disease and the development of better CNS delivery strategies for these compounds.
Collapse
Affiliation(s)
- Nil Saez-Calveras
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Olaf Stuve
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Neurology Section, Veterans Affairs (VA) North Texas Health Care System, Dallas, TX, United States
- *Correspondence: Olaf Stuve,
| |
Collapse
|
4
|
Nicaise AJ, McDonald A, Sears ER, Sturgis T, Kaplan BLF. TCDD Inhibition of IgG1 Production in Experimental Autoimmune Encephalomyelitis (EAE) and In Vitro. Antibodies (Basel) 2022; 11:4. [PMID: 35076460 PMCID: PMC8788515 DOI: 10.3390/antib11010004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/02/2021] [Accepted: 12/31/2021] [Indexed: 02/04/2023] Open
Abstract
The environmental contaminant 2,3,7,8-tetrachlorodibenzo-para-dioxin (TCDD) is a ligand for the aryl hydrocarbon receptor (AhR). TCDD is well-characterized to produce immunotoxicity, including suppression of antibody production. Previously we showed that TCDD inhibited myelin oligodendrocyte glycoprotein (MOG) peptide-specific IgG and attenuated disease in experimental autoimmune encephalomyelitis (EAE) model in mice. Thus, the purpose of this study was to characterize the effects of TCDD on IgG subclasses in EAE and in vitro and assess effects in B cells derived from various tissues. TCDD modestly suppressed intracellular IgG expression in splenocytes (SPLC), but not bone marrow (BM) or lymph node (LN) cells. To further understand TCDD's effects on IgG, we utilized LPS and LPS + IL-4 in vitro to stimulate IgG3 and IgG1 production, respectively. TCDD preferentially suppressed IgG1+ cell surface expression, especially in SPLC. However, TCDD was able to suppress IgG1 and IgG3 secretion from SPLC and B cells, but not BM cells. Lastly, we revisited the EAE model and determined that TCDD suppressed MOG-specific IgG1 production. Together these data show that the IgG1 subclass of IgG is a sensitive target of suppression by TCDD. Part of the pathophysiology of EAE involves production of pathogenic antibodies that can recruit cytolytic cells to destroy MOG-expressing cells that comprise myelin, so inhibition of IgG1 likely contributes to TCDD's EAE disease attenuation.
Collapse
Affiliation(s)
| | | | | | | | - Barbara L. F. Kaplan
- Center for Environmental Health Sciences, Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Starkville, MS 39762, USA; (A.J.N.); (A.M.); (E.R.S.); (T.S.)
| |
Collapse
|
5
|
Xiao C, Lu D, Chen J, Chen X, Lin H, Huang M, Cheng S, Wang Y, Liu Q, Zheng H. Human Olfactory Mesenchymal Stem Cells Are a Novel Candidate for Neurological Autoimmune Disease. Front Pharmacol 2021; 12:770884. [PMID: 34955841 PMCID: PMC8702423 DOI: 10.3389/fphar.2021.770884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 11/08/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Human olfactory mesenchymal stem cells (OMSC) have become a novel therapeutic option for immune disorder or demyelinating disease due to their immunomodulatory and regenerative potentials. However, the immunomodulatory effects of OMSC still need to be elucidated, and comparisons of the effects of different MSCs are also required in order to select an optimal cell source for further applications. Results: In animal experiments, we found neural functional recovery and delayed EAE attack in the OMSC treatment group. Compared with umbilical cord-derived mesenchymal stem cells (UMSC) treatment group and the control group, the OMSC treatment group had a better neurological improvement, lower serum levels of IFN-γ, and a lower proportion of CD4+IFN-γ+ T splenic lymphocyte. We also observed OMSC effectively suppressed CD4+IFN-γ+ T cell proportion in vitro when co-cultured with human peripheral blood-derived lymphocytes. The OMSC-mediated immunosuppressive effect on human CD4+IFN-γ+ T cells was attenuated by blocking cyclooxygenase activity. Conclusion: Our results suggest that OMSC treatment delayed the onset and promoted the neural functional recovery in the EAE mouse model possibly by suppressing CD4+IFN-γ+ T cells. OMSC transplantation might become an alternative therapeutic option for neurological autoimmune disease.
Collapse
Affiliation(s)
- Chongjun Xiao
- Department of Rehabilitation Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Di Lu
- The Biotherapy Center, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jinshuo Chen
- Department of Rehabilitation Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiaoyan Chen
- The Biotherapy Center, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Huizhu Lin
- The Biotherapy Center, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Mudan Huang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Shimei Cheng
- Department of Rehabilitation Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yuge Wang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Qiuli Liu
- The Biotherapy Center, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Haiqing Zheng
- Department of Rehabilitation Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
6
|
Complement component 3 from astrocytes mediates retinal ganglion cell loss during neuroinflammation. Acta Neuropathol 2021; 142:899-915. [PMID: 34487221 DOI: 10.1007/s00401-021-02366-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 08/13/2021] [Accepted: 08/27/2021] [Indexed: 12/14/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS) characterized by varying degrees of secondary neurodegeneration. Retinal ganglion cells (RGC) are lost in MS in association with optic neuritis but the mechanisms of neuronal injury remain unclear. Complement component C3 has been implicated in retinal and cerebral synaptic pathology that may precede neurodegeneration. Herein, we examined post-mortem MS retinas, and then used a mouse model, experimental autoimmune encephalomyelitis (EAE), to examine the role of C3 in the pathogenesis of RGC loss associated with optic neuritis. First, we show extensive C3 expression in astrocytes (C3+/GFAP+ cells) and significant RGC loss (RBPMS+ cells) in post-mortem retinas from people with MS compared to retinas from non-MS individuals. A patient with progressive MS with a remote history of optic neuritis showed marked reactive astrogliosis with C3 expression in the inner retina extending into deeper layers in the affected eye more than the unaffected eye. To study whether C3 mediates retinal degeneration, we utilized global C3-/- EAE mice and found that they had less RGC loss and partially preserved neurites in the retina compared with C3+/+ EAE mice. C3-/- EAE mice had fewer axonal swellings in the optic nerve, reflecting reduced axonal injury, but had no changes in demyelination or T cell infiltration into the CNS. Using a C3-tdTomato reporter mouse line, we show definitive evidence of C3 expression in astrocytes in the retina and optic nerves of EAE mice. Conditional deletion of C3 in astrocytes showed RGC protection replicating the effects seen in the global knockouts. These data implicate astrocyte C3 expression as a critical mediator of retinal neuronal pathology in EAE and MS, and are consistent with recent studies showing C3 gene variants are associated with faster rates of retinal neurodegeneration in human disease.
Collapse
|
7
|
Chen X, Yang X, Yuan P, Jin R, Bao L, Qiu X, Liu S, Liu T, Gooding JJ, Chen W, Liu G, Bai Y, Liu S, Jin Y. Modular immune-homeostatic microparticles promote immune tolerance in mouse autoimmune models. Sci Transl Med 2021; 13:13/584/eaaw9668. [PMID: 33692135 DOI: 10.1126/scitranslmed.aaw9668] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 03/14/2020] [Accepted: 01/26/2021] [Indexed: 12/29/2022]
Abstract
The therapeutic goal for autoimmune diseases is disease antigen-specific immune tolerance without nonspecific immune suppression. However, it is a challenge to induce antigen-specific immune tolerance in a dysregulated immune system. In this study, we developed immune-homeostatic microparticles (IHMs) that treat multiple mouse models of autoimmunity via induction of apoptosis in activated T cells and reestablishment of regulatory T cells. Specifically, in an experimental model of colitis, IHMs rapidly released monocyte chemotactic protein-1 after intravenous administration, which recruited activated T cells and then induced their apoptosis by conjugated Fas ligand on the IHM surface. This triggered professional macrophages to ingest apoptotic T cells and produce high quantities of transforming growth factor-β, which drove regulatory T cell differentiation. Furthermore, the modular design of IHMs allowed IHMs to be engineered with the autoantigen peptides that can reduce disease in an experimental autoimmune encephalomyelitis mouse model and a nonobese diabetic mouse model. This was accomplished by sustained release of the autoantigens after induction of T cell apoptosis and transforming growth factor-β production by macrophages, which promoted to establish an immune tolerant environment. Thus, IHMs may be an efficient therapeutic strategy for autoimmune diseases through induction of apoptosis and reestablishment of tolerant immune responses.
Collapse
Affiliation(s)
- Xin Chen
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiao Tong University, Xi'an, Shaanxi 710049, China.
| | - Xiaoshan Yang
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Pingyun Yuan
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiao Tong University, Xi'an, Shaanxi 710049, China
| | - Ronghua Jin
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiao Tong University, Xi'an, Shaanxi 710049, China
| | - Lili Bao
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Xinyu Qiu
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Siying Liu
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Tao Liu
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiao Tong University, Xi'an, Shaanxi 710049, China
| | - John Justin Gooding
- School of Chemistry, Australian Centre for NanoMedicine and ARC Australian Centre of Excellence in Convergent Bio-Nano Science and Technology, University of New South Wales, Sydney 2052, Australia
| | - WanJun Chen
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Guozhen Liu
- Graduate School of Biomedical Engineering, ARC Centre of Excellence for Nanoscale BioPhotonics and Australian Centre for NanoMedicine, Faculty of Engineering, University of New South Wales, Sydney 2052, Australia
| | - Yongkang Bai
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiao Tong University, Xi'an, Shaanxi 710049, China
| | - Shiyu Liu
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China.
| | - Yan Jin
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China.
| |
Collapse
|
8
|
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: 80] [Impact Index Per Article: 20.0] [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
|
9
|
Göbel K, Eichler S, Wiendl H, Chavakis T, Kleinschnitz C, Meuth SG. The Coagulation Factors Fibrinogen, Thrombin, and Factor XII in Inflammatory Disorders-A Systematic Review. Front Immunol 2018; 9:1731. [PMID: 30105021 PMCID: PMC6077258 DOI: 10.3389/fimmu.2018.01731] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 07/12/2018] [Indexed: 11/13/2022] Open
Abstract
Background The interaction of coagulation factors has been shown to go beyond their traditional roles in hemostasis and to affect the development of inflammatory diseases. Key molecular players, such as fibrinogen, thrombin, or factor XII have been mechanistically and epidemiologically linked to inflammatory disorders like multiple sclerosis (MS), rheumatoid arthritis (RA), and colitis. Objectives To systematically review the evidence for a role of coagulation factors, especially factor XII, fibrinogen, and thrombin in inflammatory disorders like MS, RA, and bowel disorders. Methods A systematic literature search was done in the PubMed database to identify studies about coagulation factors in inflammatory diseases. Original articles and reviews investigating the role of the kallikrein–kinin and the coagulation system in mouse and humans were included. Results We identified 43 animal studies dealing with inflammatory disorders and factors of the kallikrein–kinin or the coagulation system. Different immunological influences are described and novel molecular mechanisms linking coagulation and inflammation are reported. Conclusion A number of studies have highlighted coagulation factors to tip the balance between hemostasis and thrombosis and between protection from infection and extensive inflammation. To optimize the treatment of chronic inflammatory disorders by these factors, further studies are necessary.
Collapse
Affiliation(s)
- Kerstin Göbel
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Susann Eichler
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Heinz Wiendl
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Triantafyllos Chavakis
- Department of Clinical Pathobiochemistry, Laboratory Medicine, Institute for Clinical Chemistry, University Clinic Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | | | - Sven G Meuth
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany
| |
Collapse
|
10
|
Dietrich M, Helling N, Hilla A, Heskamp A, Issberner A, Hildebrandt T, Kohne Z, Küry P, Berndt C, Aktas O, Fischer D, Hartung HP, Albrecht P. Early alpha-lipoic acid therapy protects from degeneration of the inner retinal layers and vision loss in an experimental autoimmune encephalomyelitis-optic neuritis model. J Neuroinflammation 2018. [PMID: 29514678 PMCID: PMC5840773 DOI: 10.1186/s12974-018-1111-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Background In multiple sclerosis (MS), neurodegeneration is the main reason for chronic disability. Alpha-lipoic acid (LA) is a naturally occurring antioxidant which has recently been demonstrated to reduce the rate of brain atrophy in progressive MS. However, it remains uncertain if it is also beneficial in the early, more inflammatory-driven phases. As clinical studies are costly and time consuming, optic neuritis (ON) is often used for investigating neuroprotective or regenerative therapeutics. We aimed to investigate the prospect for success of a clinical ON trial using an experimental autoimmune encephalomyelitis-optic neuritis (EAE-ON) model with visual system readouts adaptable to a clinical ON trial. Methods Using an in vitro cell culture model for endogenous oxidative stress, we compared the neuroprotective capacity of racemic LA with the R/S-enantiomers and its reduced form. In vivo, we analyzed retinal neurodegeneration using optical coherence tomography (OCT) and the visual function by optokinetic response (OKR) in MOG35–55-induced EAE-ON in C57BL/6J mice. Ganglion cell counts, inflammation, and demyelination were assessed by immunohistological staining of retinae and optic nerves. Results All forms of LA provided equal neuroprotective capacities in vitro. In EAE-ON, prophylactic LA therapy attenuated the clinical EAE score and prevented the thinning of the inner retinal layer while therapeutic treatment was not protective on visual outcomes. Conclusions A prophylactic LA treatment is necessary to protect from visual loss and retinal thinning in EAE-ON, suggesting that a clinical ON trial starting therapy after the onset of symptoms may not be successful. Electronic supplementary material The online version of this article (10.1186/s12974-018-1111-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Michael Dietrich
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Niklas Helling
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Alexander Hilla
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Annemarie Heskamp
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Andrea Issberner
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Thomas Hildebrandt
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Zippora Kohne
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Patrick Küry
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Carsten Berndt
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Orhan Aktas
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Dietmar Fischer
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Philipp Albrecht
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.
| |
Collapse
|
11
|
Renner M, Stute G, Alzureiqi M, Reinhard J, Wiemann S, Schmid H, Faissner A, Dick HB, Joachim SC. Optic Nerve Degeneration after Retinal Ischemia/Reperfusion in a Rodent Model. Front Cell Neurosci 2017; 11:254. [PMID: 28878627 PMCID: PMC5572359 DOI: 10.3389/fncel.2017.00254] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 08/08/2017] [Indexed: 01/11/2023] Open
Abstract
Retinal ischemia is a common pathomechanism in many ocular disorders such as age-related macular degeneration (AMD), diabetic retinopathy, glaucoma or retinal vascular occlusion. Several studies demonstrated that ischemia/reperfusion (I/R) leads to morphological and functional changes of different retinal cell types. However, little is known about the ischemic effects on the optic nerve. The goal of this study was to evaluate these effects. Ischemia was induced by raising the intraocular pressure (IOP) in one eye of rats to 140 mmHg for 1 h followed by natural reperfusion. After 21 days, histological as well as quantitative real-time PCR (qRT-PCR) analyses of optic nerves were performed. Ischemic optic nerves showed an infiltration of cells and also degeneration with signs of demyelination. Furthermore, a migration and an activation of microglia could be observed histologically as well as on mRNA level. In regard to macroglia, a trend toward gliosis could be noted after ischemia induction by vimentin staining. Additionally, an up-regulation of glial fibrillary acidic protein (GFAP) mRNA was found in ischemic optic nerves. Counting of oligodendrocyte transcription factor 2 positive (Olig2+) cells revealed a decrease of oligodendrocytes in the ischemic group. Also, myelin basic protein (MBP) and myelin oligodendrocyte glycoprotein (MOG) mRNA expression was down-regulated after induction of I/R. On immunohistological level, a decrease of MOG was detectable in ischemic optic nerves as well. In addition, SMI-32 stained neurofilaments of longitudinal optic nerve sections showed a strong structural damage of the ischemic optic nerves in comparison to controls. Consequently, retinal ischemia impacts optic nerve degeneration. These findings could help to better understand the course of destruction in the optic nerve after an ischemic insult. Especially for therapeutic studies, the optic nerve is important because of its susceptibility to be damaged as a result to retinal ischemic injury and also its connecting function between the eye and the brain. So, future drug screenings should target not only the retina, but also the functionality and structure of the optic nerve. In the future, these results could lead to the development of new therapeutic strategies for treatment of ischemic injury.
Collapse
Affiliation(s)
- Marina Renner
- Experimental Eye Research, University Eye Hospital, Ruhr-University BochumBochum, Germany
| | - Gesa Stute
- Experimental Eye Research, University Eye Hospital, Ruhr-University BochumBochum, Germany
| | - Mohammad Alzureiqi
- Experimental Eye Research, University Eye Hospital, Ruhr-University BochumBochum, Germany
| | - Jacqueline Reinhard
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University BochumBochum, Germany
| | - Susanne Wiemann
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University BochumBochum, Germany
| | - Heiko Schmid
- Experimental Eye Research, University Eye Hospital, Ruhr-University BochumBochum, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University BochumBochum, Germany
| | - H Burkhard Dick
- Experimental Eye Research, University Eye Hospital, Ruhr-University BochumBochum, Germany
| | - Stephanie C Joachim
- Experimental Eye Research, University Eye Hospital, Ruhr-University BochumBochum, Germany
| |
Collapse
|
12
|
Mohammadzadeh A, Pourfathollah AA, Shahrokhi S, Fallah A, Tahoori MT, Amari A, Forouzandeh M, Soleimani M. Evaluation of AD-MSC (adipose-derived mesenchymal stem cells) as a vehicle for IFN-β delivery in experimental autoimmune encephalomyelitis. Clin Immunol 2016; 169:98-106. [DOI: 10.1016/j.clim.2016.06.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 06/22/2016] [Accepted: 06/29/2016] [Indexed: 02/08/2023]
|
13
|
Pacini G, Ieronymaki M, Nuti F, Sabatino G, Larregola M, Aharoni R, Papini AM, Rovero P. Epitope mapping of anti-myelin oligodendrocyte glycoprotein (MOG) antibodies in a mouse model of multiple sclerosis: microwave-assisted synthesis of the peptide antigens and ELISA screening. J Pept Sci 2015; 22:52-8. [PMID: 26663200 DOI: 10.1002/psc.2839] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 10/28/2015] [Accepted: 10/28/2015] [Indexed: 01/06/2023]
Abstract
The role of pathologic auto-antibodies against myelin oligodendrocyte glycoprotein (MOG) in multiple sclerosis is a highly controversial matter. As the use of animal models may enable to unravel the molecular mechanisms of the human disorder, numerous studies on multiple sclerosis are carried out using experimental autoimmune encephalomyelitis (EAE). In particular, the most extensively used EAE model is obtained by immunizing C57BL/6 mice with the immunodominant peptide MOG(35-55). In this scenario, we analyzed the anti-MOG antibody response in this model using the recombinant refolded extracellular domain of the protein, MOG(1-117). To assess the presence of a B-cell intramolecular epitope spreading mechanism, we tested also five synthetic peptides mapping the 1-117 sequence of MOG, including MOG(35-55). For this purpose, we cloned, expressed in Escherichia coli and on-column refolded MOG(1-117), and we applied an optimized microwave-assisted solid-phase synthetic strategy to obtain the designed peptide sequences. Subsequently, we set up a solid-phase immunoenzymatic assay testing both naïve and EAE mice sera and using MOG protein and peptides as antigenic probes. The results obtained disclose an intense IgG antibody response against both the recombinant protein and the immunizing peptide, while no response was observed against the other synthetic fragments, thus excluding the presence of an intramolecular epitope spreading mechanism. Furthermore, as the properly refolded recombinant probe is able to bind antibodies with greater efficiency compared with MOG(35-55), we hypothesize the presence of both linear and conformational epitopes on MOG(35-55) sequence.
Collapse
Affiliation(s)
- Giulia Pacini
- French-Italian Interdepartmental Laboratory of Peptide and Protein Chemistry and Biology - PeptLab, Florence, Italy and Cergy-Pontoise, France.,Department NeuroFarBa, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Via Ugo Schiff 6, Sesto Fiorentino, Florence, I-50019, Italy
| | - Matthaia Ieronymaki
- French-Italian Interdepartmental Laboratory of Peptide and Protein Chemistry and Biology - PeptLab, Florence, Italy and Cergy-Pontoise, France.,Department NeuroFarBa, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Via Ugo Schiff 6, Sesto Fiorentino, Florence, I-50019, Italy.,Laboratoire de Chimie Biologique EA4505, University of Cergy-Pontoise, 5 mail Gay-Lussac Neuville-sur-Oise, Cergy-Pontoise, 95000, France
| | - Francesca Nuti
- French-Italian Interdepartmental Laboratory of Peptide and Protein Chemistry and Biology - PeptLab, Florence, Italy and Cergy-Pontoise, France.,Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3/13, Sesto Fiorentino, Florence, I-50019, Italy
| | - Giuseppina Sabatino
- French-Italian Interdepartmental Laboratory of Peptide and Protein Chemistry and Biology - PeptLab, Florence, Italy and Cergy-Pontoise, France.,Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3/13, Sesto Fiorentino, Florence, I-50019, Italy
| | - Maud Larregola
- French-Italian Interdepartmental Laboratory of Peptide and Protein Chemistry and Biology - PeptLab, Florence, Italy and Cergy-Pontoise, France.,Laboratoire de Chimie Biologique EA4505, University of Cergy-Pontoise, 5 mail Gay-Lussac Neuville-sur-Oise, Cergy-Pontoise, 95000, France
| | - Rina Aharoni
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Anna Maria Papini
- French-Italian Interdepartmental Laboratory of Peptide and Protein Chemistry and Biology - PeptLab, Florence, Italy and Cergy-Pontoise, France.,Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3/13, Sesto Fiorentino, Florence, I-50019, Italy.,Laboratoire de Chimie Biologique EA4505, University of Cergy-Pontoise, 5 mail Gay-Lussac Neuville-sur-Oise, Cergy-Pontoise, 95000, France
| | - Paolo Rovero
- French-Italian Interdepartmental Laboratory of Peptide and Protein Chemistry and Biology - PeptLab, Florence, Italy and Cergy-Pontoise, France.,Department NeuroFarBa, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Via Ugo Schiff 6, Sesto Fiorentino, Florence, I-50019, Italy
| |
Collapse
|
14
|
Hundgeburth LC, Wunsch M, Rovituso D, Recks MS, Addicks K, Lehmann PV, Kuerten S. The complement system contributes to the pathology of experimental autoimmune encephalomyelitis by triggering demyelination and modifying the antigen-specific T and B cell response. Clin Immunol 2012; 146:155-64. [PMID: 23352967 DOI: 10.1016/j.clim.2012.12.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Revised: 12/07/2012] [Accepted: 12/09/2012] [Indexed: 12/31/2022]
Abstract
So far, studies of the human autoimmune disease multiple sclerosis (MS) have largely been hampered by the absence of a pathogenic B cell component in its animal model, experimental autoimmune encephalomyelitis (EAE). To overcome this shortcoming, we have previously introduced the myelin basic protein (MBP)-proteolipid protein (PLP) MP4-induced EAE, which is B cell and autoantibody-dependent. Here we show that MP4-immunized wild-type C57BL/6 mice displayed a significantly lower disease incidence when their complement system was transiently depleted by a single injection of cobra venom factor (CVF) prior to immunization. Considering the underlying pathomechanism, our data suggest that the complement system is crucial for MP4-specific antibodies to trigger CNS pathology. Demyelinated lesions in the CNS were colocalized with complement depositions. In addition, B cell deficient JHT mice reconstituted with MP4-reactive serum showed significantly attenuated clinical and histological EAE after depletion of complement by CVF. The complement system was also critically involved in the generation of the MP4-specific T and B cell response: in MP4-immunized wild-type mice treated with CVF the MP4-specific cytokine and antibody response was significantly attenuated compared to untreated wild-type mice. Taken together, we propose two independent mechanisms by which the complement system can contribute to the pathology of autoimmune encephalomyelitis. Our data corroborate the role of complement in triggering antibody-dependent demyelination and antigen-specific T cell immunity and also provide first evidence that the complement system can modify the antigen-specific B cell response in EAE and possibly MS.
Collapse
Affiliation(s)
- Lorenz C Hundgeburth
- Department of Anatomy I, University of Cologne, Joseph-Stelzmann-Str. 9, 50931 Cologne, Germany.
| | | | | | | | | | | | | |
Collapse
|
15
|
Yang J, Yan Y, Ma CG, Kang T, Zhang N, Gran B, Xu H, Li K, Ciric B, Zangaladze A, Curtis M, Rostami A, Zhang GX. Accelerated and enhanced effect of CCR5-transduced bone marrow neural stem cells on autoimmune encephalomyelitis. Acta Neuropathol 2012; 124:491-503. [PMID: 22526024 DOI: 10.1007/s00401-012-0989-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Revised: 04/10/2012] [Accepted: 04/13/2012] [Indexed: 01/02/2023]
Abstract
The suppressive effect of neural stem cells (NSCs) on experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS), has been reported. However, the migration of NSCs to inflammatory sites was relatively slow as was the onset of rather limited clinical benefit. Lack of, or low expression of particular chemokine receptors on NSCs could be an important factor underlying the slow migration of NSCs. To enhance the therapeutic effect of NSCs, in the present study we transduced bone marrow (BM)-derived NSCs with CCR5, a receptor for CCL3, CCL4, and CCL5, chemokines that are abundantly produced in CNS-inflamed foci of MS/EAE. After i.v. injection, CCR5-NSCs rapidly reached EAE foci in larger numbers, and more effectively suppressed CNS inflammatory infiltration, myelin damage, and clinical EAE than GFP-NSCs used as controls. CCR5-NSC-treated mice also exhibited augmented remyelination and neuron/oligodendrocyte repopulation compared to PBS- or GFP-NSC-treated mice. We inferred that the critical mechanism underlying enhanced effect of CCR5-transduced NSCs on EAE is the early migration of chemokine receptor-transduced NSCs into the inflamed foci. Such migration at an earlier stage of inflammation enables NSCs to exert more effective immunomodulation, to reduce the extent of early myelin/neuron damage by creating a less hostile environment for remyelinating cells, and possibly to participate in the remyelination/neural repopulation process. These features of BM-derived transduced NSCs, combined with their easy availability (the subject's own BM) and autologous properties, may lay the groundwork for an innovative approach to rapid and highly effective MS therapy.
Collapse
MESH Headings
- Animals
- Bone Marrow Cells/cytology
- Bone Marrow Cells/immunology
- Bone Marrow Cells/metabolism
- Cell Differentiation/immunology
- Chemotaxis, Leukocyte/immunology
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Female
- Immunohistochemistry
- Immunomodulation
- Mice
- Mice, Inbred C57BL
- Microscopy, Electron, Transmission
- Neural Stem Cells/cytology
- Neural Stem Cells/immunology
- Neural Stem Cells/metabolism
- Receptors, CCR5/immunology
- Receptors, CCR5/metabolism
- Transduction, Genetic
Collapse
Affiliation(s)
- Jingxian Yang
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Benkhoucha M, Molnarfi N, Santiago-Raber ML, Weber MS, Merkler D, Collin M, Lalive PH. IgG glycan hydrolysis by EndoS inhibits experimental autoimmune encephalomyelitis. J Neuroinflammation 2012; 9:209. [PMID: 22943418 PMCID: PMC3458989 DOI: 10.1186/1742-2094-9-209] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Accepted: 08/14/2012] [Indexed: 01/03/2023] Open
Abstract
Studies in experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis, have shown that B cells markedly influence the course of the disease, although whether their effects are protective or pathological is a matter of debate. EndoS hydrolysis of the IgG glycan has profound effects on IgG effector functions, such as complement activation and Fc receptor binding, suggesting that the enzyme could be used as an immunomodulatory therapeutic agent against IgG-mediated diseases. We demonstrate here that EndoS has a protective effect in myelin oligodendrocyte glycoprotein peptide amino acid 35–55 (MOG35-55)-induced EAE, a chronic neuroinflammatory demyelinating disorder of the central nervous system (CNS) in which humoral immune responses are thought to play only a minor role. EndoS treatment in chronic MOG35-55-EAE did not impair encephalitogenic T cell priming and recruitment into the CNS of mice, consistent with a primary role of EndoS in controlling IgG effector functions. In contrast, reduced EAE severity coincided with poor serum complement activation and deposition within the spinal cord, suggesting that EndoS treatment impairs B cell effector function. These results identify EndoS as a potential therapeutic agent against antibody-mediated CNS autoimmune disorders.
Collapse
Affiliation(s)
- Mahdia Benkhoucha
- Department of Pathology and Immunology, University of Geneva, 1211, Geneva, Switzerland.
| | | | | | | | | | | | | |
Collapse
|
17
|
Kindling-induced learning deficiency and possible cellular and molecular involved mechanisms. Neurol Sci 2012; 34:883-90. [DOI: 10.1007/s10072-012-1142-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Accepted: 06/12/2012] [Indexed: 10/28/2022]
|
18
|
Sherafat MA, Heibatollahi M, Mongabadi S, Moradi F, Javan M, Ahmadiani A. Electromagnetic field stimulation potentiates endogenous myelin repair by recruiting subventricular neural stem cells in an experimental model of white matter demyelination. J Mol Neurosci 2012; 48:144-53. [PMID: 22588976 DOI: 10.1007/s12031-012-9791-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2011] [Accepted: 04/26/2012] [Indexed: 04/28/2023]
Abstract
Electromagnetic fields (EMFs) may affect the endogenous neural stem cells within the brain. The aim of this study was to assess the effects of EMFs on the process of toxin-induced demyelination and subsequent remyelination. Demyelination was induced using local injection of lysophosphatidylcholine within the corpus callosum of adult female Sprague-Dawley rats. EMFs (60 Hz; 0.7 mT) were applied for 2 h twice a day for 7, 14, or 28 days postlesion. BrdU labeling and immunostaining against nestin, myelin basic protein (MBP), and BrdU were used for assessing the amount of neural stem cells within the tissue, remyelination patterns, and tracing of proliferating cells, respectively. EMFs significantly reduced the extent of demyelinated area and increased the level of MBP staining within the lesion area on days 14 and 28 postlesion. EMFs also increased the number of BrdU- and nestin-positive cells within the area between SVZ and lesion as observed on days 7 and 14 postlesion. It seems that EMF potentiates proliferation and migration of neural stem cells and enhances the repair of myelin in the context of demyelinating conditions.
Collapse
Affiliation(s)
- Mohammad Amin Sherafat
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | | | | | | |
Collapse
|
19
|
Mann MK, Ray A, Basu S, Karp CL, Dittel BN. Pathogenic and regulatory roles for B cells in experimental autoimmune encephalomyelitis. Autoimmunity 2012; 45:388-99. [PMID: 22443691 DOI: 10.3109/08916934.2012.665523] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A dual role of B cells in experimental autoimmune encephalomyelitis (EAE), the animal model of the human autoimmune disease multiple sclerosis (MS), has been established. In the first role, B cells contribute to the pathogenesis of EAE through the production of anti-myelin antibodies that contribute to demyelination. On the contrary, B cells have also been shown to have protective functions in that they play an essential role in the spontaneous recovery from EAE. In this review, we summarize studies conducted in a number of species demonstrating the conditions under which B cells are pathogenic in EAE. We also discuss the phenotype and anti-inflammatory mechanisms of regulatory B cells.
Collapse
Affiliation(s)
- Monica K Mann
- Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, Wisconsin 53201-2178, USA
| | | | | | | | | |
Collapse
|
20
|
Lee DH, Linker RA. The role of myelin oligodendrocyte glycoprotein in autoimmune demyelination: a target for multiple sclerosis therapy? Expert Opin Ther Targets 2012; 16:451-62. [DOI: 10.1517/14728222.2012.677438] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
21
|
C3-dependent mechanism of microglial priming relevant to multiple sclerosis. Proc Natl Acad Sci U S A 2012; 109:965-70. [PMID: 22219359 DOI: 10.1073/pnas.1111924109] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Microglial priming predisposes the brain to neurodegeneration and affects disease progression. The signal to switch from the quiescent to the primed state is unknown. We show that deleting the C3 convertase regulator complement receptor 1-related protein y (Crry) induces microglial priming. Mice that were double-knockout for Crry and either C3 or factor B did not show priming, demonstrating dependence on alternative pathway activation. Colocalization of C3b/iC3b and CR3 implicated the CR3/iC3b interaction in priming. Systemic lipopolysaccharide challenge overactivated primed microglia with florid expression of proinflammatory molecules, which were blocked by complement inhibition. Relevance for neurodegenerative disease is exemplified by human multiple sclerosis (MS) and by experimental autoimmune encephalomyelitis (EAE), a model of MS. In human MS, microglial priming was evident in perilesional white matter, in close proximity to C3b/iC3b deposits. EAE was accelerated and exacerbated in Crry-deficient mice, and was dependent on C activation. In summary, C3-dependent microglial priming confers susceptibility to other challenges. Our observations are relevant to progression in MS and other neurological diseases exacerbated by acute insults.
Collapse
|
22
|
Castration Attenuates Myelin Repair Following Lysolecithin Induced Demyelination in Rat Optic Chiasm: An Evaluation Using Visual Evoked Potential, Marker Genes Expression and Myelin Staining. Neurochem Res 2011; 36:1887-95. [DOI: 10.1007/s11064-011-0510-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/13/2011] [Indexed: 10/18/2022]
|
23
|
Human RORγt+ TH17 cells preferentially differentiate from naive FOXP3+Treg in the presence of lineage-specific polarizing factors. Proc Natl Acad Sci U S A 2010; 107:19402-7. [PMID: 20962281 DOI: 10.1073/pnas.1008247107] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
RORγt(+) T(H)17 cells are a proinflammatory CD4(+) T-cell population associated with autoimmune tissue injury. In mice, priming of T(H)17 requires TGF-β, which alone directs the priming of FOXP3(+) regulatory T cells (Treg), in association with inflammatory cytokines. Priming of human T(H)17 cells from conventional naive CD4(+) T cells under similar conditions, however, has proved difficult to achieve. Here, we report that differentiation of human T(H)17 cells preferentially occurs from FOXP3(+) naive Treg (NTreg) in the presence of IL-2 and IL-1β and is increased by IL-23 and TGF-β. IL-1β-mediated differentiation correlated with IL-1RI expression in stimulated NTreg and was accompanied by induction of RORγt along with down-regulation of FOXP3. IL-17-secreting cells in NTreg cultures cosecreted TNF-α and IL-2 and contained distinct subpopulations cosecreting or not cosecreting IFN-γ and other T(H)17-associated cytokines. Polarized NTreg contained significant subpopulations of CCR6-expressing cells that were highly enriched in IL-17-secreting cells. Finally, analysis of CCR6 expression with respect to that of IL-1RI identified distinct IL-17-secreting subpopulations that had maintained or lost their suppressive functions. Together our results support the concept that priming of human T(H)17 from naive CD4(+) T cells preferentially takes place from FOXP3(+) Treg precursors in the presence of lineage-specific polarizing factors.
Collapse
|
24
|
Dunkelberger JR, Song WC. Role and mechanism of action of complement in regulating T cell immunity. Mol Immunol 2010; 47:2176-86. [PMID: 20603023 DOI: 10.1016/j.molimm.2010.05.008] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
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 19104, USA
| | | |
Collapse
|
25
|
Linker RA, Lee DH. Models of autoimmune demyelination in the central nervous system: on the way to translational medicine. EXPERIMENTAL & TRANSLATIONAL STROKE MEDICINE 2009; 1:5. [PMID: 20142992 PMCID: PMC2816864 DOI: 10.1186/2040-7378-1-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/22/2009] [Accepted: 10/21/2009] [Indexed: 11/10/2022]
Abstract
Multiple sclerosis (MS) is the most common neurologic disease of young adults. In the recent years, our understanding on disease pathomechanisms has considerably improved and new therapies have emerged. Yet a cure for this devastating disorder is still a far cry away and human resources on ex vivo specimens are limited. More than 70 years after its first description, experimental autoimmune encephalomyelitis (EAE) remains an important tool to understand concepts of T cell mediated autoimmunity as well as the roles of the innate and the humoral immune systems. Some EAE models also well reflect mechanisms of tissue damage including demyelination, axonal injury and also cortical changes. A limitation of the classical EAE model is a neglect of CD8 T cell mediated immune mechanisms. Moreover, well characterized models for primary progressive MS or demyelination patterns involving primary oligodendrocyte dystrophy are still not available. Yet many current therapeutic concepts including glatiramer acetate or natalizumab stem from their successful first application in EAE models. New strategies include the widespread use of conditional knockout mice to understand the cell-type specific function of single genes, innovative approaches to establish models on the roles of B cells and CD8 T cells as well as on the relation of inflammation to primary degeneration. In summary, EAE models continue to play an important role in neuroimmunology thereby also stimulating research in other fields of the neurosciences and immunobiology.
Collapse
Affiliation(s)
- Ralf A Linker
- Department of Neurology, St Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | | |
Collapse
|
26
|
Baranzini SE. Systems-based medicine approaches to understand and treat complex diseases. The example of multiple sclerosis. Autoimmunity 2009; 39:651-62. [PMID: 17178562 DOI: 10.1080/08916930601061686] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Systems medicine is an emerging concept that acknowledges the complexity of a multitude of non-linear interactions among molecular and physiological variables. Under this new paradigm, rather than a collection of symptoms, diseases are seen as the product of deviations from a robust steady state compatible with life. This concept requires the incorporation of mathematics and physics to the more classical arsenal of physiology and molecular biology with which physicians are trained today. This review explores the diverse types of information that can be accumulated towards the understanding of multiple sclerosis (MS), a complex autoimmune disease that targets the central nervous system (CNS). The challenge of data integration and modeling of dynamical systems is discussed in the context of disease susceptibility and response to treatment. A theoretical framework that supports the use of combination therapy is also presented.
Collapse
Affiliation(s)
- Sergio E Baranzini
- Department of Neurology, School of Medicine, University of California, San Francisco, 513 Parnassus Avenue Room S-256, San Francisco, CA 94143-0435, USA.
| |
Collapse
|
27
|
Ayyoub M, Deknuydt F, Raimbaud I, Dousset C, Leveque L, Bioley G, Valmori D. Human memory FOXP3+ Tregs secrete IL-17 ex vivo and constitutively express the T(H)17 lineage-specific transcription factor RORgamma t. Proc Natl Acad Sci U S A 2009; 106:8635-40. [PMID: 19439651 PMCID: PMC2688993 DOI: 10.1073/pnas.0900621106] [Citation(s) in RCA: 247] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2009] [Indexed: 11/18/2022] Open
Abstract
Recent studies have suggested a close relationship between CD4(+)FOXP3(+) regulatory T cells (Tregs) and proinflammatory IL-17-producing T helper cells (T(H)17) expressing the lineage-specific transcription factor RORgamma t. We report here the unexpected finding that human memory Tregs secrete IL-17 ex vivo and constitutively express RORgamma t. IL-17-secreting Tregs share some phenotypic and functional features with conventional T(H)17 cells, expressing high levels of CCR4 and CCR6 and low levels of CXCR3. However, unlike conventional T(H)17 cells, they express low levels of CD161 and mostly fail to cosecrete IL-22 and TNF-alpha ex vivo. Ex vivo secretion of IL-17 and constitutive expression of RORgamma t by human memory Tregs suggest that, in addition to their well-known suppressive functions, these cells likely play additional, as yet undescribed, proinflammatory functions.
Collapse
Affiliation(s)
- Maha Ayyoub
- Institut National de la Santé et de la Recherche Médicale, Unité 892, Centre René Gauducheau, 44800 Saint Herblain, France.
| | | | | | | | | | | | | |
Collapse
|
28
|
IL-1β and IL-2 convert human Treg into TH17 cells. Clin Immunol 2009; 131:298-307. [DOI: 10.1016/j.clim.2008.12.008] [Citation(s) in RCA: 134] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2008] [Revised: 12/11/2008] [Accepted: 12/20/2008] [Indexed: 01/11/2023]
|
29
|
Dong M, Liu R, Guo L, Li C, Tan G. Pathological findings in rats with experimental allergic encephalomyelitis. APMIS 2009; 116:972-84. [PMID: 19132994 DOI: 10.1111/j.1600-0463.2008.00726.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The aim of this study was to establish an animal model of experimental allergic encephalomyelitis (EAE) and examine the basic pathological changes, as well as expression and distribution of MMP-2 and MMP-9, in Wistar rats. Tissue sections were processed for HE staining, Weil myelin staining, and modified Bielschowsky staining. Expression and distribution of glial fibrillary acidic protein (GFAP), matrix metalloproteinase-2 (MMP-2) and matrix metalloproteinase-9 (MMP-9) were detected with immunohistochemistry. We divided the EAE into five types, depending on pathological characteristics and clinical manifestations: acute EAE, relapsing-remitting EAE, progressive EAE, benign EAE, and asymptomatic EAE. Rats with acute EAE suffered from quick, severe attacks with widespread inflammatory cells and axonal loss. No demyelination or astrocytic hyperplasia was found around the lesions. Rats with relapsing-remitting EAE broke down twice, with many perivascular cuffs and demyelinating plaques in lesions; hyperplastic and hypertrophic astrocytes characterized old lesions and axonal loss was evident. Rats suffering from progressive EAE exhibited continuous aggravation without improvement, accompanied by perivascular cuffs, demyelination, increased gliocytes and axonal damage. Rats with benign EAE recovered to a normal state with obviously decreased inflammatory cells and almost entirely unaffected myelin and axons. Rats with asymptomatic EAE also had various pathological changes that were not coincident with their clinical manifestations. Elevated expression of MMP-2 and MMP-9 was concordant in different types of EAE, but the extent differed in each type of EAE. MMP-2 and MMP-9 can be expressed in the form of vascular endothelial cells, meninges, or accumulated inflammatory cells. Multiple clinical courses of disease were demonstrated in Wistar rat EAE, with attributes similar to multiple sclerosis (MS) in clinical and pathological characteristics. Elevated expression of MMP-2 and MMP-9 may play a role in some aspects of pathological changes in EAE, for example, destroying the blood-brain barrier, degrading the myelin sheath, and damaging axons.
Collapse
Affiliation(s)
- Mei Dong
- Department of Neurology, The Second Affiliated Hospital, Hebei Medical University, Shijiazhuang, China
| | | | | | | | | |
Collapse
|
30
|
Ingram G, Hakobyan S, Robertson NP, Morgan BP. Complement in multiple sclerosis: its role in disease and potential as a biomarker. Clin Exp Immunol 2008; 155:128-39. [PMID: 19040603 DOI: 10.1111/j.1365-2249.2008.03830.x] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [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
|
31
|
Nizri E, Irony-Tur-Sinai M, Faranesh N, Lavon I, Lavi E, Weinstock M, Brenner T. Suppression of neuroinflammation and immunomodulation by the acetylcholinesterase inhibitor rivastigmine. J Neuroimmunol 2008; 203:12-22. [DOI: 10.1016/j.jneuroim.2008.06.018] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2008] [Revised: 06/10/2008] [Accepted: 06/10/2008] [Indexed: 11/25/2022]
|
32
|
Guan Y, Yu S, Zhao Z, Ciric B, Zhang GX, Rostami A. Antigen presenting cells treated in vitro by macrophage colony-stimulating factor and autoantigen protect mice from autoimmunity. J Neuroimmunol 2007; 192:68-78. [PMID: 18006080 DOI: 10.1016/j.jneuroim.2007.09.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2006] [Revised: 09/07/2007] [Accepted: 09/07/2007] [Indexed: 12/29/2022]
Abstract
Macrophage colony-stimulating factor (M-CSF) is a critical cytokine in the development of monocytic lineage and may have immunoregulatory properties. Here we show that peritoneal antigen presenting cells (APCs) treated with M-CSF produced decreased levels of proinflammatory cytokines IFN-gamma, TNF-alpha and IL-12. These APCs treated with M-CSF+autoantigen peptide significantly suppressed antigen-specific T cell proliferation, induced regulatory CD4(+) and CD8(+) T cells in vitro and in vivo, and significantly suppressed experimental autoimmune encephalomyelitis (EAE). Thus, in vitro treatment of APCs with M-CSF+autoantigen can be a novel therapeutic option for autoimmune diseases.
Collapse
Affiliation(s)
- Yangtai Guan
- Department of Neurology, Thomas Jefferson University, 300 JHN Building, 900 Walnut Street, Philadelphia, PA 19107, United States
| | | | | | | | | | | |
Collapse
|
33
|
McGeachy MJ, O'Connor R, Stephens LA, Anderton SM. Antigen-based therapy and immune-regulation in experimental autoimmune encephalomyelitis. Methods Mol Biol 2007; 380:313-26. [PMID: 17876101 DOI: 10.1007/978-1-59745-395-0_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Experimental autoimmune encephalomyelitis is a long-established mouse model of multiple sclerosis. The requirements for autoreactive T-cell activation in this disease have been characterized extensively and novel strategies for immune-intervention are being developed continually. Notably, identification of immunodominant T-cell epitopes allows the induction of T-cell tolerance with synthetic peptides. Several transgenic mouse lines that express transgenic T-cell receptors recognizing myelin autoantigenic epitopes have been developed. These allow adoptive transfer studies to analyse the activation of naive autoreactive T cells in vivo during the induction of tolerance vs immunity. More recently, our attention has focused on immune mechanisms underlying the natural recovery from disease. Sampling of the lymphoid cell infiltrate within the central nervous system has identified the accumulation of regulatory T cells in the target organ during this period of resolution.
Collapse
Affiliation(s)
- Mandy J McGeachy
- Institute of Immunology and Infection Research, Iniversity of Edinburgh, UK
| | | | | | | |
Collapse
|
34
|
Jégou JF, Chan P, Schouft MT, Griffiths MR, Neal JW, Gasque P, Vaudry H, Fontaine M. C3d binding to the myelin oligodendrocyte glycoprotein results in an exacerbated experimental autoimmune encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2007; 178:3323-31. [PMID: 17312184 DOI: 10.4049/jimmunol.178.5.3323] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The complement system is known to contribute to demyelination in multiple sclerosis and experimental autoimmune encephalomyelitis. However, there are few data concerning the natural adjuvant effect of C3d on the humoral response when it binds to myelin Ags. This study addresses the effect of C3d binding to the myelin oligodendrocyte glycoprotein (MOG) in the induction of experimental autoimmune encephalomyelitis in C57BL/6J mice. Immunization with human MOG coupled to C3d was found to accelerate the appearance of clinical signs of the disease and to enhance its severity compared with MOG-immunized mice. This finding was correlated with an increased infiltration of leukocytes into the central nervous system accompanied by increased complement activation and associated with areas of demyelination and axonal loss. Furthermore, B cell participation in the pathogenesis of the disease was determined by their increased capacity to act as APCs and to form germinal centers. Consistent with this, the production of MOG-specific Abs was found to be enhanced following MOG/C3d immunization. These results suggest that binding of C3d to self-Ags could increase the severity of an autoimmune disease by enhancing the adaptive autoimmune response.
Collapse
Affiliation(s)
- Jean-François Jégou
- INSERM U413, Institut Fédératif de Recherches Multidisciplinaires sur les Peptides 23, University of Rouen, Place Emile Blondel, Mont Saint-Aignan Cedex, France
| | | | | | | | | | | | | | | |
Collapse
|
35
|
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: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [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
- *Corresponding Author: Dr. Scott R. Barnum, Department of Microbiology, University of Alabama at Birmingham, 845 19 St. S., BBRB/842, Birmingham, AL 35294. E-mail address:
| |
Collapse
|
36
|
Rus H, Cudrici C, Niculescu F. C5b-9 complement complex in autoimmune demyelination: dual role in neuroinflammation and neuroprotection. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 586:139-51. [PMID: 16893070 DOI: 10.1007/0-387-34134-x_10] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Affiliation(s)
- Horea Rus
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD 21201, USA
| | | | | |
Collapse
|
37
|
Urich E, Gutcher I, Prinz M, Becher B. Autoantibody-mediated demyelination depends on complement activation but not activatory Fc-receptors. Proc Natl Acad Sci U S A 2006; 103:18697-702. [PMID: 17121989 PMCID: PMC1693725 DOI: 10.1073/pnas.0607283103] [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] [Indexed: 11/18/2022] Open
Abstract
The precise mechanisms leading to CNS inflammation and myelin destruction in both multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE) remain the subject of intense debate. In both MS and EAE, autoantibodies (autoAbs) are thought to be involved in tissue destruction through recruiting Fc receptor (FcR)-bearing cells or direct cytotoxic effects through the activation of the complement pathway. Whereas intrathecal immunoglobulin (Ig) production and Ig deposition in inflammatory lesions is a hallmark of MS, mice deficient in B cells and Igs develop severe EAE. Paradoxically, mice of the same genetic background but deficient in FcRgamma are EAE-resistant. We found that the functional expression of FcRgamma on systemic accessory cells, but not CNS-resident cells, appears to be vital for the development of CNS inflammation, independent of antigen-presenting cell function or Ab involvement. On the other hand, we found that the injection of antimyelin oligodendrocyte glycoprotein-Abs drastically worsens disease severity, inflammation, and demyelination. Using FcRgamma(-/-) and C1q(-/-) mice, we could definitively establish that the demyelinating capacity of such autoAb in vivo relies entirely on complement activation and is FcR-independent.
Collapse
MESH Headings
- Animals
- Autoantibodies/physiology
- Complement Activation/genetics
- Complement Activation/immunology
- Complement System Proteins/physiology
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Receptors, IgG/deficiency
- Receptors, IgG/genetics
- Receptors, IgG/physiology
Collapse
Affiliation(s)
- Eduard Urich
- *Neurology Department, Division of Neuroimmunology, University of Zurich, Y44-J38/J42, Winterthurerstrasse 190, 8057 Zurich, Switzerland; and
| | - Ilona Gutcher
- *Neurology Department, Division of Neuroimmunology, University of Zurich, Y44-J38/J42, Winterthurerstrasse 190, 8057 Zurich, Switzerland; and
| | - Marco Prinz
- Department of Neuropathology, Georg-August-University, University of Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany
| | - Burkhard Becher
- *Neurology Department, Division of Neuroimmunology, University of Zurich, Y44-J38/J42, Winterthurerstrasse 190, 8057 Zurich, Switzerland; and
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
38
|
Rus H, Cudrici C, Niculescu F, Shin ML. Complement activation in autoimmune demyelination: Dual role in neuroinflammation and neuroprotection. J Neuroimmunol 2006; 180:9-16. [PMID: 16905199 DOI: 10.1016/j.jneuroim.2006.07.009] [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: 04/26/2006] [Revised: 07/05/2006] [Accepted: 07/07/2006] [Indexed: 12/27/2022]
Abstract
Multiple sclerosis and its animal model experimental allergic encephalomyelitis are inflammatory demyelinating diseases of the central nervous system mediated by activated lymphocytes, macrophages/microglia and the complement system. Complement activation and the C5b-9 terminal complex contribute to the pathogenesis of these diseases through its role to promote demyelination. C5b-9 was also shown to protect oligodendrocytes from apoptosis both in vitro and in vivo. Our findings indicate that activation of complement and C5b-9 assembly plays a pro-inflammatory role in the acute phase, but may also be neuroprotective.
Collapse
Affiliation(s)
- Horea Rus
- Department of Neurology, Baltimore MD, USA.
| | | | | | | |
Collapse
|
39
|
Rus H, Cudrici C, David S, Niculescu F. The complement system in central nervous system diseases. Autoimmunity 2006; 39:395-402. [PMID: 16923539 DOI: 10.1080/08916930600739605] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
The activation of complement system is important factor in inflammatory, neurodegenerative and cerebrovascular diseases. CNS cells are able to synthesize complement components, and myelin and oligodendrocytes (OLG) are known to activate the classical pathway of complement in vitro in the absence of antibodies. Although activation of the complement system is known to promote tissue injury, recent evidence has also indicated that this process can have neuroprotective effects. In particular, terminal C5b-9 complexes enhance OLG survival both in vitro and in vivo. Complement activation may also reduce the accumulation of amyloid and degenerating neurons by promoting their clearance and suggest that certain inflammatory defense mechanisms in the brain may be beneficial in neurodegenerative disease. Complement system activation plays also an important role in brain damage after ischemic injury or head trauma. These findings strongly suggest that complement activation and membrane assembly of C5b-9 can play a role in injury but can also provide neuroprotection depending on the pathophysiological context.
Collapse
Affiliation(s)
- Horea Rus
- Department of Neurology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA.
| | | | | | | |
Collapse
|
40
|
Zhu YN, Zhong XG, Feng JQ, Yang YF, Fu YF, Ni J, Liu QF, Tang W, Zhao WM, Zuo JP. Periplocoside E inhibits experimental allergic encephalomyelitis by suppressing interleukin 12-dependent CCR5 expression and interferon-gamma-dependent CXCR3 expression in T lymphocytes. J Pharmacol Exp Ther 2006; 318:1153-62. [PMID: 16751252 DOI: 10.1124/jpet.106.105445] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Periplocoside E (PSE) was found to inhibit primary T-cell activation in our previous study. Now we examined the effect and mechanisms of PSE on the central nervous system (CNS) demyelination in experimental allergic encephalomyelitis (EAE). C57BL/6 mice immunized with myelin oligodendrocyte glyco-protein (MOG) were treated with PSE following immunization and continued throughout the study. The effect on the progression of EAE and other relevant parameters were assessed. PSE reduced the incidence and severity of EAE. Spinal cord histopathology analysis showed that the therapeutic effect of PSE was associated with reduced mononuclear cell infiltration and CNS inflammation. As reverse transcription-polymerase chain reaction analysis showed, PSE decreased the CD4(+), CD8(+), and CD11b(+) cell infiltration. T cells from lymph nodes of MOG-immunized mice expressed enhanced levels of CCR5 and CXCR3 mRNA compared with T cells from normal mice. However, CCR5 and CXCR3 expressions were suppressed in T cells from PSE-treated mice. In vitro study also showed PSE inhibited interferon (IFN)-gamma-dependent CXCR3 expression in T cells through suppressing T-cell receptor (TCR) ligation-induced IFN-gamma production, whereas it inhibited interleukin (IL)-12-dependent CCR5 expression through suppressing IL-12 reactivity in TCR-triggered T cells. As a result, the initial influx of T cells into CNS was inhibited in PSE-treated mice. The consequent activation of macrophages/microglia cells was inhibited in spinal cord from PSE-treated mice as determination of chemokine expressions (CCL2, CCL3, CCL4, CCL5, CXCL9, and CXCL10). Consistently, the secondary influx of CD4(+), CD8(+), and CD11b(+) cells was decreased in spinal cords from PSE-treated mice. These findings suggest the potential therapeutic effect of PSE on multiple sclerosis.
Collapse
Affiliation(s)
- Yi-Na Zhu
- Laboratory of Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Barnum SR, Szalai AJ. Complement and demyelinating disease: no MAC needed? ACTA ACUST UNITED AC 2006; 52:58-68. [PMID: 16443278 DOI: 10.1016/j.brainresrev.2005.12.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2005] [Revised: 12/09/2005] [Accepted: 12/15/2005] [Indexed: 12/22/2022]
Abstract
It has long been accepted that the complement system participates in the onset, evolution, and exacerbation of demyelinating disease, and it is widely suspected that this is accomplished mainly via destruction of nervous tissue by membrane attack complex (MAC)-mediated lysis of oligodendrocytes and neurons. However, recent studies using mutant mice indicate the MAC may not be so important. For example, mice lacking C5 and mice lacking the C5a receptor both develop experimental autoimmune encephalomyelitis (EAE) with the same frequency and intensity as their wild type counterparts. Also, transgenic mice that express C5a exclusively in the central nervous system (CNS) develop EAE that is not remarkably different from that in non-transgenic littermates. Since C5 is required for formation of the MAC, development of fulminant EAE in the absence of this complement protein demonstrates that non-complement-mediated mechanisms of CNS damage are operating. Paradoxically, mice lacking C3, mice lacking the C3a receptor, and mice lacking the complement receptor type 3 develop attenuated EAE, while mice that express C3a exclusively in the CNS develop severe and often fulminant EAE. Based on these newer data, we posit that C3-derived biologically active fragments, rather than C5 and the MAC, are central players in the pathophysiology of complement in EAE.
Collapse
Affiliation(s)
- Scott R Barnum
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | | |
Collapse
|
42
|
Linker RA, Rott E, Hofstetter HH, Hanke T, Toyka KV, Gold R. EAE in beta-2 microglobulin-deficient mice: axonal damage is not dependent on MHC-I restricted immune responses. Neurobiol Dis 2005; 19:218-28. [PMID: 15837577 DOI: 10.1016/j.nbd.2004.12.017] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2004] [Revised: 12/06/2004] [Accepted: 12/08/2004] [Indexed: 11/17/2022] Open
Abstract
There is accumulating evidence that CD8-positive (CD8+) T-cells and MHC-I expression may also play a role in neurodegeneration associated with multiple sclerosis (MS). We investigated the role of MHC-I and CD8+ T-cells by studying experimental autoimmune encephalomyelitis (EAE) in beta-2 microglobulin knockout mice induced by myelin oligodendrocyte glycoprotein (MOG) peptide 35-55 or whole rat myelin basic protein (rMBP). For both encephalitogens and even after reconstitution of the immune system with MHC-I-positive bone marrow and transfer of mature CD8+ T-cells (iMHC-I+ CD8+ beta2m-/- mice), the disease course in beta2m-/- mice was significantly more severe with a 10-fold increased mortality in the beta2m-/- mice as compared to wild-type C57BL/6 mice. EAE in beta2m-/- mice caused more severe demyelination after immunization with MOG than with rMBP and axonal damage was more marked with rMBP as well as MOG even in iMHC-I+ CD8+ beta2m-/- mice. Immunocytochemical analysis of spinal cord tissue revealed a significant increase in macrophage and microglia infiltration in beta2m-/- and iMHC-I+ CD8+ beta2m-/- mice. The different pattern of T-cell infiltration was underscored by a 2.5-fold increase in CD4-positive (CD4+) T-cells in beta2m-/- mice after induction of MOG 35-55 EAE. We conclude that lack of functional MHC-I molecules and CD8+ T-cells aggravates autoimmune tissue destruction in the CNS. Enhanced axonal damage speaks for pathways of tissue damage independent of CD8+ T-cells and neuronal MHC-I expression.
Collapse
MESH Headings
- Animals
- Axons/immunology
- Axons/metabolism
- Axons/pathology
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Genes, MHC Class I
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- T-Lymphocytes/pathology
- beta 2-Microglobulin/deficiency
- beta 2-Microglobulin/genetics
- beta 2-Microglobulin/physiology
Collapse
Affiliation(s)
- Ralf A Linker
- Department of Neurology, Clinical Research Group for Multiple Sclerosis, Julius-Maximilians-Universität Würzburg, Josef-Schneider-Str. 11, D-97080 Würzburg, Germany
| | | | | | | | | | | |
Collapse
|
43
|
Linker RA, Sendtner M, Gold R. Mechanisms of axonal degeneration in EAE—lessons from CNTF and MHC I knockout mice. J Neurol Sci 2005; 233:167-72. [PMID: 15949503 DOI: 10.1016/j.jns.2005.03.021] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The major pathological hallmarks of multiple sclerosis (MS) comprise inflammation, demyelination with associated gliosis and axonal damage, which most likely correlates with persisting disability. Axonal damage can occur by several mechanisms. This article focuses on myelin disintegration and direct immune attack on axons by CD8-positive T-cells as two possible scenarios for axonal injury. As protoypic models, we investigated experimental autoimmune encephalomyelitis (EAE) in ciliary neurotrophic factor gene knockout mice (CNTF-/- mice) with severe myelin pathology and EAE in beta-2 microglobulin gene knockout mice (beta2m-/- mice) lacking CD8-positive T-cells. The results from these studies indicate that the trigger attack for axonal injury even in a well-defined experimental design can be multi-faceted. No single factor seems to be absolutely necessary for the initiation of the process, but they rather act in concert and orchestrate tissue destruction, inflammation and regeneration. Some mechanisms of primary or secondary axonal damage may be shared between inflammatory and degenerative diseases of the nervous system, thereby establishing a link which might be of importance for future therapeutic strategies.
Collapse
Affiliation(s)
- Ralf A Linker
- Department of Neurology, Clinical Research Group for Multiple Sclerosis, University of Würzburg, Josef-Schneider-Strasse 11, D-97080 Würzburg, Germany
| | | | | |
Collapse
|
44
|
Kim TS, Perlman S. Virus-specific antibody, in the absence of T cells, mediates demyelination in mice infected with a neurotropic coronavirus. THE AMERICAN JOURNAL OF PATHOLOGY 2005; 166:801-9. [PMID: 15743792 PMCID: PMC1602352 DOI: 10.1016/s0002-9440(10)62301-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Mice infected with mouse hepatitis virus strain JHM develop an inflammatory demyelinating disease in the central nervous system with many similarities to human multiple sclerosis. The mouse disease is primarily immune-mediated because demyelination is not detected in JHM-infected mice lacking T or B cells but does occur after transfer of JHM-specific T cells. Although less is known about the ability of antibodies to mediate demyelination, the presence of oligoclonally expanded B cells and high concentrations of antibodies (against self or infectious agents) in the central nervous system of many multiple sclerosis patients suggests that antibodies may also contribute to myelin destruction. Here, we show that anti-JHM antibodies, in the absence of T or B cells, caused demyelination in JHM-infected mice. Anti-JHM antibody was detected adjacent to areas of demyelination, consistent with a direct interaction between antibody and infected cells. Demyelination was reduced by 85 to 90% in infected RAG1(-/-) mice lacking normal expression of activating Fc receptors (FcRgamma(-/-)) and by approximately 76% when complement was depleted by treatment with cobra venom factor. These data demonstrate that JHM-specific antibodies are sufficient to cause demyelination and that myelin destruction in the presence of anti-virus antibodies results from a combination of complement- and Fc receptor-dependent mechanisms.
Collapse
Affiliation(s)
- Taeg S Kim
- Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA 52242, USA
| | | |
Collapse
|
45
|
Breij ECW, Heijnen P, Vloet R, Saito T, van de Winkel JGJ, Dijkstra CD, Amor S, Verbeek S. The FcRγ Chain Is Not Essential for Induction of Experimental Allergic Encephalomyelitis (EAE) or Anti-Myelin Antibody-Mediated Exacerbation of EAE. J Neuropathol Exp Neurol 2005; 64:304-11. [PMID: 15835266 DOI: 10.1093/jnen/64.4.304] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Macrophages are considered essential mediators in multiple sclerosis (MS) pathogenesis, presumably through myelin phagocytosis and release of inflammatory mediators. Macrophages and microglia express activating Fcgamma receptors (FcgammaRI and FcgammaRIII), which depend on the FcRgamma chain for surface expression and signaling. In MS lesions, crosslinking of FcgammaR by immunoglobulins (IgG) directed against myelin may enhance myelin phagocytosis and inflammation. We studied the role of FcgammaR and anti-myelin antibodies in MOG35-55-induced experimental allergic encephalomyelitis (EAE) in C57BL/6 mice, a model of MS-like disease. Incidence and severity of EAE were similar in FcRy chain-/- (FcRgamma-/-) and wild-type (wt) mice, albeit with delayed onset in FcRgamma-/- mice. This demonstrates that the FcRy chain is not essential for induction of EAE, but that FcRgamma signaling may contribute to the preclinical phase. The role of FcgammaR in antibody-mediated demyelination was addressed by injection of anti-myelin antibodies (Z12 mAb) at onset of MOG35-55-induced EAE. Injection of Z12 mAb rapidly reduced survival time in both wt and FcRgamma-/- mice, demonstrating that antibody-mediated exacerbation of EAE is independent of the FcRgamma chain. Interestingly, Z12-induced exacerbation of inflammation and demyelination persisted longer in wt than FcRgamma-/- mice, suggesting that IgG-FcgammaR interactions may contribute to a sustained pathologic effect of anti-myelin antibodies in the CNS.
Collapse
Affiliation(s)
- Esther C W Breij
- Department of Molecular Cell Biology and Immunology, VU Medical Center, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Morgan BP, Griffiths M, Khanom H, Taylor SM, Neal JW. Blockade of the C5a receptor fails to protect against experimental autoimmune encephalomyelitis in rats. Clin Exp Immunol 2005; 138:430-8. [PMID: 15544619 PMCID: PMC1809229 DOI: 10.1111/j.1365-2249.2004.02646.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Complement activation contributes to inflammation and tissue damage in human demyelinating diseases and in rodent models of demyelination. Inhibitors of complement activation ameliorate disease in the rat model antibody-dependent experimental autoimmune encephalomyelitis and rats unable to generate the membrane attack complex of complement develop inflammation without demyelination. The role of the highly active chemotactic and anaphylactic complement-derived peptide C5a in driving inflammation and pathology in rodent models of demyelination has been little explored. Here we have used a small molecule C5a receptor antagonist, AcF-[OPdChaWR], to examine the effects of C5a receptor blockade in rat models of brain inflammation and demyelination. C5a receptor antagonist therapy completely blocked neutrophil response to C5a in vivo but had no effect on clinical disease or resultant pathology in either inflammatory or demyelinating rat models. We conclude that C5a is not required for disease induction or perpetuation in these strongly complement-dependent disease models.
Collapse
Affiliation(s)
- B P Morgan
- Complement Biology Group, Department of Medical Biochemistry and Immunology and Department of Pathology, UWCM, Cardiff, UK.
| | | | | | | | | |
Collapse
|
47
|
Raivich G, Banati R. Brain microglia and blood-derived macrophages: molecular profiles and functional roles in multiple sclerosis and animal models of autoimmune demyelinating disease. ACTA ACUST UNITED AC 2005; 46:261-81. [PMID: 15571769 DOI: 10.1016/j.brainresrev.2004.06.006] [Citation(s) in RCA: 170] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/01/2004] [Indexed: 12/23/2022]
Abstract
Microglia and macrophages, one a brain-resident, the other a mostly hematogenous cell type, represent two related cell types involved in the brain pathology in multiple sclerosis and its autoimmune animal model, the experimental allergic encephalomyelitis. Together, they perform a variety of different functions: they are the primary sensors of brain pathology, they are rapidly recruited to sites of infection, trauma or autoimmune inflammation in experimental allergic encephalomyelitis and multiple sclerosis and they are competent presenters of antigen and interact with T cells recruited to the inflamed CNS. They also synthesise a variety of molecules, such as cytokines (TNF, interleukins), chemokines, accessory molecules (B7, CD40), complement, cell adhesion glycoproteins (integrins, selectins), reactive oxygen radicals and neurotrophins, that could exert a damaging or a protective effect on adjacent axons, myelin and oligodendrocytes. The current review will give a detailed summary on their cellular response, describe the different classes of molecules expressed and their attribution to the blood derived or brain-resident macrophages and then discuss how these molecules contribute to the neuropathology. Recent advances using chimaeric and genetically modified mice have been particularly telling about the specific, overlapping and nonoverlapping roles of macrophages and microglia in the demyelinating disease. Interestingly, they point to a crucial role of hematogenous macrophages in initiating inflammation and myelin removal, and that of microglia in checking excessive response and in the induction and maintenance of remission.
Collapse
Affiliation(s)
- Gennadij Raivich
- Department of Anatomy, Obstetrics and Gynaecology, Perinatal Brain Repair Centre, University College London, Chenies Mews 86-96 WC1E 6HX London, UK.
| | | |
Collapse
|
48
|
Liu J, Miwa T, Hilliard B, Chen Y, Lambris JD, Wells AD, Song WC. The complement inhibitory protein DAF (CD55) suppresses T cell immunity in vivo. ACTA ACUST UNITED AC 2005; 201:567-77. [PMID: 15710649 PMCID: PMC2213052 DOI: 10.1084/jem.20040863] [Citation(s) in RCA: 187] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Decay-accelerating factor ([DAF] CD55) is a glycosylphosphatidylinositol-anchored membrane inhibitor of complement with broad clinical relevance. Here, we establish an additional and unexpected role for DAF in the suppression of adaptive immune responses in vivo. In both C57BL/6 and BALB/c mice, deficiency of the Daf1 gene, which encodes the murine homologue of human DAF, significantly enhanced T cell responses to active immunization. This phenotype was characterized by hypersecretion of interferon (IFN)-γ and interleukin (IL)-2, as well as down-regulation of the inhibitory cytokine IL-10 during antigen restimulation of lymphocytes in vitro. Compared with wild-type mice, Daf1−/− mice also displayed markedly exacerbated disease progression and pathology in a T cell–dependent experimental autoimmune encephalomyelitis (EAE) model. However, disabling the complement system in Daf1−/− mice normalized T cell secretion of IFN-γ and IL-2 and attenuated disease severity in the EAE model. These findings establish a critical link between complement and T cell immunity and have implications for the role of DAF and complement in organ transplantation, tumor evasion, and vaccine development.
Collapse
Affiliation(s)
- Jianuo Liu
- Institute for Translational Medicine and Therapeutics and Department of Pharmacology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Rus H, Cudrici C, Niculescu F. C5b-9 complement complex in autoimmune demyelination and multiple sclerosis: dual role in neuroinflammation and neuroprotection. Ann Med 2005; 37:97-104. [PMID: 16026117 DOI: 10.1080/07853890510007278] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Complement system activation plays an important role in innate and acquired immunity. Activation of complement leads to the formation of C5b-9 terminal complex. While C5b-9 can promote cell lysis, sublytic assembly of C5b-9 on plasma membranes induces cell cycle activation and survival. Multiple sclerosis (MS) and its animal model experimental allergic encephalomyelitis (EAE) are inflammatory demyelinating diseases of the central nervous system (CNS) mediated by activated lymphocytes, macrophages/microglia and the complement system. Complement activation may contribute to the pathogenesis of these diseases through its dual role: the ability of activated terminal complex C5b-9 to promote demyelination and the capacity of sublytic C5b-9 to protect oligodendrocytes (OLG) from apoptosis. By inducing EAE in C5-deficient mice, we showed that complement C5 promotes remyelination and protects oligodendrocytes from apoptotic cell death. These findings indicate that activation of complement C5b-9 plays a pro-inflammatory role in the acute phase of the disease, but may also be neuroprotective during the chronic phase of the disease.
Collapse
Affiliation(s)
- Horea Rus
- University of Maryland at Baltimore, School of Medicine, Department of Neurology, 655 W. Baltimore Street, BRB 12-016, Baltimore, MD 21201, USA.
| | | | | |
Collapse
|
50
|
Alt C, Duvefelt K, Franzén B, Yang Y, Engelhardt B. Gene and protein expression profiling of the microvascular compartment in experimental autoimmune encephalomyelitis in C57Bl/6 and SJL mice. Brain Pathol 2005; 15:1-16. [PMID: 15779231 PMCID: PMC8095736 DOI: 10.1111/j.1750-3639.2005.tb00094.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Dysfunction of the blood-brain barrier (BBB) is a hallmark of inflammatory diseases of the central nervous system (CNS) such as multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE). The molecular mechanisms leading to BBB breakdown are not well understood. In order to find molecules involved in this process, we used oligonucleotide microarrays and proteomics to analyze gene and protein expression of the microvascular compartment isolated from brains of C57Bl/6 and SJL/N mice afflicted with EAE and the microvascular compartment isolated from healthy controls. Out of the 6500 known genes and expressed sequence tags (ESTs) studied, expression of 288 genes was found to be changed. Of these genes 128 were altered in the microvascular compartment in both EAE models. Six proteins were identified to be present at altered levels. In addition to the expected increased expression of genes coding for molecules involved in leukocyte recruitment, genes not yet ascribed to EAE pathogenesis were identified. Thus, proteomics and gene array screens of the microvascular compartment are valid approaches, that can be used to define novel candidate molecules involved in EAE pathogenesis at the level of the BBB.
Collapse
MESH Headings
- Animals
- Blood-Brain Barrier/physiology
- Brain/blood supply
- Cerebrovascular Circulation/genetics
- Electrophoresis, Gel, Two-Dimensional
- Electrophoresis, Polyacrylamide Gel
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Gene Expression
- Gene Expression Profiling
- Image Processing, Computer-Assisted
- Immunohistochemistry
- In Situ Hybridization
- Mice
- Mice, Inbred C57BL
- Mice, Inbred Strains
- Microcirculation/physiology
- Oligonucleotide Array Sequence Analysis
- Protein Array Analysis
- Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
Collapse
Affiliation(s)
- Carsten Alt
- Max‐Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Kristina Duvefelt
- Gene and Protein Technology, Molecular Sciences, Local Discovery Research Area CNS & Pain Control, Sweden
- Division of Neurology, Neurotec, Karolinska Institute, Huddinge University Hospital, Stockholm, Sweden (present address)
| | - Bo Franzén
- Gene and Protein Technology, Molecular Sciences, Local Discovery Research Area CNS & Pain Control, Sweden
| | - Yang Yang
- Analytical Development, AstraZeneca R&D Södertälje, Sweden
| | - Britta Engelhardt
- Max‐Planck Institute for Molecular Biomedicine, Münster, Germany
- Theodor‐Kocher Institute, University of Bern, Switzerland
| |
Collapse
|