101
|
Neyt K, GeurtsvanKessel CH, Deswarte K, Hammad H, Lambrecht BN. Early IL-1 Signaling Promotes iBALT Induction after Influenza Virus Infection. Front Immunol 2016; 7:312. [PMID: 27579026 PMCID: PMC4985557 DOI: 10.3389/fimmu.2016.00312] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 08/02/2016] [Indexed: 11/13/2022] Open
Abstract
Inducible bronchus-associated lymphoid tissue (iBALT) is a long lasting tertiary lymphoid tissue that can be induced following influenza A virus (IAV) infection. Previous studies have shown that iBALT structures containing germinal center (GC) B cells protect against repeated infection by contributing locally to the cellular and humoral immune response. If we are to exploit this in vaccination strategies, we need a better understanding on how iBALT structures are induced. One hypothesis is that the strength of the initial innate response dictates induction of iBALT. In the present study, we investigated the role of interleukin (IL)-1 and IL-1R signaling on iBALT formation. Mice lacking the IL-1R had a delayed viral clearance and, thus, a prolonged exposure to viral replication, leading to increased disease severity, compared to wild-type mice. Contradictorily, iBALT formation following clearance of the virus was heavily compromised in Il1r1−/− mice. Quantification of gene induction after IAV infection demonstrated induction of IL-1α and to a much lesser extent of IL-1β. Administration of recombinant IL-1α to the lungs of wild-type mice, early but not late, after IAV infection led to more pronounced iBALT formation and an increased amount of GC B cells in the lungs. Bone marrow chimeric mice identified the stromal compartment as the crucial IL-1 responsive cell for iBALT induction. Mechanistically, Q-PCR analysis of lung homogenates revealed a strongly diminished production of CXCL13, a B cell-attracting chemokine, in Il1r−/− mice during the early innate phase of IAV infection. These experiments demonstrate that appropriate innate IL-1α–IL-1R signaling is necessary for IAV clearance and at the same time instructs the formation of organized tertiary lymphoid tissues through induction of CXCL13 early after infection. These findings are discussed in the light of recent insights on the pathogenesis of tertiary lymphoid organ formation in the lung in various diseases where the IL-1 axis is hyperactive, such as rheumatoid arthritis and COPD.
Collapse
Affiliation(s)
- Katrijn Neyt
- Laboratory of Immunoregulation, VIB Inflammation Research Center, Ghent, Belgium; Department of Respiratory Medicine, Ghent University, Ghent, Belgium
| | | | - Kim Deswarte
- Laboratory of Immunoregulation, VIB Inflammation Research Center, Ghent, Belgium; Department of Respiratory Medicine, Ghent University, Ghent, Belgium
| | - Hamida Hammad
- Laboratory of Immunoregulation, VIB Inflammation Research Center, Ghent, Belgium; Department of Respiratory Medicine, Ghent University, Ghent, Belgium
| | - Bart N Lambrecht
- Laboratory of Immunoregulation, VIB Inflammation Research Center, Ghent, Belgium; Department of Respiratory Medicine, Ghent University, Ghent, Belgium; Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands
| |
Collapse
|
102
|
Agahozo MC, Peferoen L, Baker D, Amor S. CD20 therapies in multiple sclerosis and experimental autoimmune encephalomyelitis - Targeting T or B cells? Mult Scler Relat Disord 2016; 9:110-7. [PMID: 27645355 DOI: 10.1016/j.msard.2016.07.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 07/06/2016] [Accepted: 07/19/2016] [Indexed: 01/03/2023]
Abstract
MS is widely considered to be a T cell-mediated disease although T cell immunotherapy has consistently failed, demonstrating distinct differences with experimental autoimmune encephalomyelitis (EAE), an animal model of MS in which T cell therapies are effective. Accumulating evidence has highlighted that B cells also play key role in MS pathogenesis. The high frequency of oligoclonal antibodies in the CSF, the localization of immunoglobulin in brain lesions and pathogenicity of antibodies originally pointed to the pathogenic role of B cells as autoantibody producing plasma cells. However, emerging evidence reveal that B cells also act as antigen presenting cells, T cell activators and cytokine producers suggesting that the strong efficacy of anti-CD20 antibody therapy observed in people with MS may reduce disease progression by several different mechanisms. Here we review the evidence and mechanisms by which B cells contribute to disease in MS compared to findings in the EAE model.
Collapse
Affiliation(s)
- Marie Colombe Agahozo
- Pathology Department, VU Medical Centre, VU University of Amsterdam, The Netherlands
| | - Laura Peferoen
- Pathology Department, VU Medical Centre, VU University of Amsterdam, The Netherlands
| | - David Baker
- Neuroimmunolgy Unit, Blizard Institute, Barts and the London School of Medicine & Dentistry Queen Mary University of London, United Kingdom
| | - Sandra Amor
- Pathology Department, VU Medical Centre, VU University of Amsterdam, The Netherlands; Neuroimmunolgy Unit, Blizard Institute, Barts and the London School of Medicine & Dentistry Queen Mary University of London, United Kingdom.
| |
Collapse
|
103
|
Serafini B, Rosicarelli B, Veroni C, Zhou L, Reali C, Aloisi F. RORγt Expression and Lymphoid Neogenesis in the Brain of Patients with Secondary Progressive Multiple Sclerosis. J Neuropathol Exp Neurol 2016; 75:877-88. [PMID: 27413074 DOI: 10.1093/jnen/nlw063] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Indexed: 12/16/2022] Open
Abstract
Ectopic B-cell follicle-like structures (ELS) are found in the meninges of patients with secondary progressive multiple sclerosis (SPMS). Because cells expressing the transcriptional regulator retinoic acid receptor-related orphan receptor-γt (RORγt) and producing interleukin 17 (IL17), e.g. T helper 17 cells and lymphoid tissue inducer (LTi) cells, have been implicated in the formation of ELS, we studied RORγt and IL17 expression in brain tissue from patients with SPMS an assessed their relationships to immune infiltrates and meningeal ELS. By immunohistochemistry, small numbers of RORγt-positive cells were detected in the meninges of 6 of 12 SPMS cases analyzed. RORγt-positive cells were localized in B-cell follicles or aggregates and nearby diffuse meningeal infiltrates, and predominantly co-expressed CD3. Only a few RORγt-positive, CD3-negative cells were observed, suggesting the presence of group 3 innate lymphoid cells, which comprise the LTi cell subset. Some IL17-positive cells, co-expressing in part RORγt and predominantly CD3, were found in meningeal B-cell follicles from 4 SPMS cases. Rare RORγt-positive and IL17-positive cells were detected in white matter. Gene expression analysis of laser dissected meningeal infiltrates and white matter lesions confirmed low frequencies and virtual absence of RORγt and IL17 signals, respectively. Thus, there is selective migration or survival of RORγt-positive cells in MS patient meninges and an association of these cells with ELS.
Collapse
Affiliation(s)
- Barbara Serafini
- From the Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, Rome, Italy (BS, BR, CV, FA) and GlaxoSmithKline Shanghai Research and Development Center, Zhangjiang Hi-Tech Park, TAU, Pudong, China, Neuroscience Shanghai (LZ, CR)
| | - Barbara Rosicarelli
- From the Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, Rome, Italy (BS, BR, CV, FA) and GlaxoSmithKline Shanghai Research and Development Center, Zhangjiang Hi-Tech Park, TAU, Pudong, China, Neuroscience Shanghai (LZ, CR)
| | - Caterina Veroni
- From the Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, Rome, Italy (BS, BR, CV, FA) and GlaxoSmithKline Shanghai Research and Development Center, Zhangjiang Hi-Tech Park, TAU, Pudong, China, Neuroscience Shanghai (LZ, CR)
| | - Ling Zhou
- From the Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, Rome, Italy (BS, BR, CV, FA) and GlaxoSmithKline Shanghai Research and Development Center, Zhangjiang Hi-Tech Park, TAU, Pudong, China, Neuroscience Shanghai (LZ, CR)
| | - Camilla Reali
- From the Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, Rome, Italy (BS, BR, CV, FA) and GlaxoSmithKline Shanghai Research and Development Center, Zhangjiang Hi-Tech Park, TAU, Pudong, China, Neuroscience Shanghai (LZ, CR)
| | - Francesca Aloisi
- From the Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, Rome, Italy (BS, BR, CV, FA) and GlaxoSmithKline Shanghai Research and Development Center, Zhangjiang Hi-Tech Park, TAU, Pudong, China, Neuroscience Shanghai (LZ, CR).
| |
Collapse
|
104
|
Zhang Q, Cao DL, Zhang ZJ, Jiang BC, Gao YJ. Chemokine CXCL13 mediates orofacial neuropathic pain via CXCR5/ERK pathway in the trigeminal ganglion of mice. J Neuroinflammation 2016; 13:183. [PMID: 27401148 PMCID: PMC4940825 DOI: 10.1186/s12974-016-0652-1] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 07/04/2016] [Indexed: 12/30/2022] Open
Abstract
Background Trigeminal nerve damage-induced neuropathic pain is a severely debilitating chronic orofacial pain syndrome. Spinal chemokine CXCL13 and its receptor CXCR5 were recently demonstrated to play a pivotal role in the pathogenesis of spinal nerve ligation-induced neuropathic pain. Whether and how CXCL13/CXCR5 in the trigeminal ganglion (TG) mediates orofacial pain are unknown. Methods The partial infraorbital nerve ligation (pIONL) was used to induce trigeminal neuropathic pain in mice. The expression of ATF3, CXCL13, CXCR5, and phosphorylated extracellular signal-regulated kinase (pERK) in the TG was detected by immunofluorescence staining and western blot. The effect of shRNA targeting on CXCL13 or CXCR5 on pain hypersensitivity was checked by behavioral testing. Results pIONL induced persistent mechanical allodynia and increased the expression of ATF3, CXCL13, and CXCR5 in the TG. Inhibition of CXCL13 or CXCR5 by shRNA lentivirus attenuated pIONL-induced mechanical allodynia. Additionally, pIONL-induced neuropathic pain and the activation of ERK in the TG were reduced in Cxcr5−/− mice. Furthermore, MEK inhibitor (PD98059) attenuated mechanical allodynia and reduced TNF-α and IL-1β upregulation induced by pIONL. TNF-α inhibitor (Etanercept) and IL-1β inhibitor (Diacerein) attenuated pIONL-induced orofacial pain. Finally, intra-TG injection of CXCL13 induced mechanical allodynia, increased the activation of ERK and the production of TNF-α and IL-1β in the TG of WT mice, but not in Cxcr5−/− mice. Pretreatment with PD98059, Etanercept, or Diacerein partially blocked CXCL13-induced mechanical allodynia, and PD98059 also reduced CXCL13-induced TNF-α and IL-1β upregulation. Conclusions CXCL13 and CXCR5 contribute to orofacial pain via ERK-mediated proinflammatory cytokines production. Targeting CXCL13/CXCR5/ERK/TNF-α and IL-1β pathway in the trigeminal ganglion may offer effective treatment for orofacial neuropathic pain.
Collapse
Affiliation(s)
- Qian Zhang
- Pain Research Laboratory, Institute of Nautical Medicine, Jiangsu Key Laboratory of Inflammation and Molecular Drug Target, Nantong University, Seyuan Road, Nantong, Jiangsu, 226019, China
| | - De-Li Cao
- Pain Research Laboratory, Institute of Nautical Medicine, Jiangsu Key Laboratory of Inflammation and Molecular Drug Target, Nantong University, Seyuan Road, Nantong, Jiangsu, 226019, China
| | - Zhi-Jun Zhang
- Pain Research Laboratory, Institute of Nautical Medicine, Jiangsu Key Laboratory of Inflammation and Molecular Drug Target, Nantong University, Seyuan Road, Nantong, Jiangsu, 226019, China.,Department of Human Anatomy, School of Medicine, Nantong University, Nantong, Jiangsu, 226001, China
| | - Bao-Chun Jiang
- Pain Research Laboratory, Institute of Nautical Medicine, Jiangsu Key Laboratory of Inflammation and Molecular Drug Target, Nantong University, Seyuan Road, Nantong, Jiangsu, 226019, China
| | - Yong-Jing Gao
- Pain Research Laboratory, Institute of Nautical Medicine, Jiangsu Key Laboratory of Inflammation and Molecular Drug Target, Nantong University, Seyuan Road, Nantong, Jiangsu, 226019, China. .,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, 226001, China.
| |
Collapse
|
105
|
Bugeja MJ, Booth DR, Bennetts BH, Heard RNS, Stewart GJ. An investigation of polymorphisms in the 4q13.3-21.1 CXC chemokine gene cluster for association with multiple sclerosis in Australians. Mult Scler 2016; 12:710-22. [PMID: 17262998 DOI: 10.1177/1352458506070964] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Susceptibility to multiple sclerosis (MS) is believed to result from the complex interaction of a number of genes, each with modest effect. Vital to the migration of cells to sites of inflammation, including the central nervous system, are chemokines, many of which are implicated in MS pathogenesis. Most of the CXC chemokine genes are encoded in a cluster on chromosome 4q13.3-21.1, which has been identified in several genome-wide screens as being potentially associated with MS. We conducted a two-stage analysis to investigate the chemokine gene cluster for association with MS. Initially, we sequenced the chemokine genes in several DNA pools to identify common polymorphisms, and then genotyped selected SNPs in 373 Australian MS trio families. We found no evidence that the CXC chemokine gene cluster is genetically associated with MS. However, the existence of common variants conferring small risk factors or rare variants with significant risk cannot be excluded.
Collapse
Affiliation(s)
- M J Bugeja
- The Institute for Immunology and Allergy Research, Westmead Millennium Institute, Westmead Campus, University of Sydney, Westmead, NSW 2145, Australia
| | | | | | | | | |
Collapse
|
106
|
Abstract
Death ligands induce apoptosis, which is a cell suicide program leading mainly to selective elimination of an organism's useless cells. Importantly, the dying cell is an active participant in its own demise (“cellular suicide”). Under physiological conditions, apoptosis is most often found during normal cell turnover and tissue homeostasis, embryogenesis, induction and maintenance of immune tolerance, development of the nervous system, and endocrine-dependent tissue atrophy. However, apoptotic processes have also been suggested to contribute to the pathology of the autoimmune demyelinating disease multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis. Here, apoptosis plays a double role. On one hand, impaired apoptosis may result in increased numbers or persistence of activated myelinspecific T cells. On the other hand, local tissue damage involves apoptosis of oligodendrocytes and neurons, leading to the clinical symptoms. In this article, an overview is given of the current knowledge of the roles of apoptosis-mediating and immune regulatory death ligands of the tumor necrosis factor (TNF) family (TNF, lymphotoxin-beta, OX40L [CD134L], CD154 [CD40L], CD95L, CD70 [CD27L], CD153 [CD30L], 4-1BBL [CD137L], TRAIL, TWEAK, BAFF, GITRL) in the pathogenesis of MS and of their implications for related therapeutic strategies.
Collapse
Affiliation(s)
- Orhan Aktas
- Institute of Neuroimmunology, Clinical and Experimental Neuroimmunology, Charité--Universitätsmedizin Berlin, Germany
| | | | | |
Collapse
|
107
|
Lokensgard JR, Mutnal MB, Prasad S, Sheng W, Hu S. Glial cell activation, recruitment, and survival of B-lineage cells following MCMV brain infection. J Neuroinflammation 2016; 13:114. [PMID: 27207308 PMCID: PMC4874004 DOI: 10.1186/s12974-016-0582-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 05/13/2016] [Indexed: 12/23/2022] Open
Abstract
Background Chemokines produced by reactive glia drive migration of immune cells and previous studies from our laboratory have demonstrated that CD19+ B cells infiltrate the brain. In this study, in vivo and in vitro experiments investigated the role of reactive glial cells in recruitment and survival of B-lineage cells in response to (murine cytomegalovirus) MCMV infection. Methods Flow cytometric analysis was used to assess chemokine receptor expression on brain-infiltrating B cells. Real-time RT-PCR and ELISA were used to measure chemokine levels. Dual-immunohistochemical staining was used to co-localize chemokine production by reactive glia. Primary glial cell cultures and migration assays were used to examine chemokine-mediated recruitment. Astrocyte: B cell co-cultures were used to investigate survival and proliferation. Results The chemokine receptors CXCR3, CXCR5, CCR5, and CCR7 were detected on CD19+ cells isolated from the brain during MCMV infection. In particular, CXCR3 was found to be elevated on an increasing number of cells over the time course of infection, and it was the primary chemokine receptor expressed at 60 days post infection Quite different expression kinetics were observed for CXCR5, CCR5, and CCR7, which were elevated on the highest number of cells early during infection and decreased by 14, 30, and 60 days post infection Correspondingly, elevated levels of CXCL9, CXCL10, and CXCL13, as well as CCL5, were found within the brains of infected animals, and only low levels of CCL3 and CCL19 were detected. Differential expression of CXCL9/CXCL10 and CXCL13 between microglia and astrocytes was apparent, and B cells moved towards supernatants from MCMV-infected microglia, but not astrocytes. Pretreatment with neutralizing Abs to CXCL9 and CXCL10 inhibited this migration. In contrast, neutralizing Abs to the ligand of CXCR5 (i.e., CXCL13) did not significantly block chemotaxis. Proliferation of brain-infiltrating B cells was detected at 7 days post infection and persisted through the latest time tested (60 days post infection). Finally, astrocytes produce BAFF (B cell activating factor of the TNF family) and promote proliferation of B cells via cell-to-cell contact. Conclusions CXCR3 is the primary chemokine receptor on CD19+ B cells persisting within the brain, and migration to microglial cell supernatants is mediated through this receptor. Correspondingly, microglial cells produce CXCL9 and CXCL10, but not CXCL13. Reactive astrocytes promote B cell proliferation.
Collapse
Affiliation(s)
- James R Lokensgard
- Neurovirology Laboratory, Department of Medicine, University of Minnesota, 3-220 LRB/MTRF, 2001 6th Street S.E., Minneapolis, MN, 55455, USA.
| | - Manohar B Mutnal
- Neurovirology Laboratory, Department of Medicine, University of Minnesota, 3-220 LRB/MTRF, 2001 6th Street S.E., Minneapolis, MN, 55455, USA
| | - Sujata Prasad
- Neurovirology Laboratory, Department of Medicine, University of Minnesota, 3-220 LRB/MTRF, 2001 6th Street S.E., Minneapolis, MN, 55455, USA
| | - Wen Sheng
- Neurovirology Laboratory, Department of Medicine, University of Minnesota, 3-220 LRB/MTRF, 2001 6th Street S.E., Minneapolis, MN, 55455, USA
| | - Shuxian Hu
- Neurovirology Laboratory, Department of Medicine, University of Minnesota, 3-220 LRB/MTRF, 2001 6th Street S.E., Minneapolis, MN, 55455, USA
| |
Collapse
|
108
|
Phares TW, DiSano KD, Stohlman SA, Segal BM, Bergmann CC. CXCL13 promotes isotype-switched B cell accumulation to the central nervous system during viral encephalomyelitis. Brain Behav Immun 2016; 54:128-139. [PMID: 26795429 PMCID: PMC4828287 DOI: 10.1016/j.bbi.2016.01.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 01/13/2016] [Accepted: 01/16/2016] [Indexed: 11/28/2022] Open
Abstract
Elevated CXCL13 within the central nervous system (CNS) correlates with humoral responses in several neuroinflammatory diseases, yet its role is controversial. During coronavirus encephalomyelitis CXCL13 deficiency impaired CNS accumulation of memory B cells and antibody-secreting cells (ASC) but not naïve/early-activated B cells. However, despite diminished germinal center B cells and follicular helper T cells in draining lymph nodes, ASC in bone marrow and antiviral serum antibody were intact in the absence of CXCL13. The data demonstrate that CXCL13 is not essential in mounting effective peripheral humoral responses, but specifically promotes CNS accumulation of differentiated B cells.
Collapse
Affiliation(s)
- Timothy W Phares
- Department of Neurosciences NC30, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH, USA.
| | - Krista D DiSano
- Department of Neurosciences NC30, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH, USA; School of Biomedical Sciences, Kent State University, Kent, OH, USA.
| | - Stephen A Stohlman
- Department of Neurosciences NC30, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH, USA.
| | - Benjamin M Segal
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Cornelia C Bergmann
- Department of Neurosciences NC30, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH, USA.
| |
Collapse
|
109
|
Sellebjerg F, Cadavid D, Steiner D, Villar LM, Reynolds R, Mikol D. Exploring potential mechanisms of action of natalizumab in secondary progressive multiple sclerosis. Ther Adv Neurol Disord 2016; 9:31-43. [PMID: 26788129 DOI: 10.1177/1756285615615257] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Multiple sclerosis (MS) is a common and chronic central nervous system (CNS) demyelinating disease and a leading cause of permanent disability. Patients most often present with a relapsing-remitting disease course, typically progressing over time to a phase of relentless advancement in secondary progressive MS (SPMS), for which approved disease-modifying therapies are limited. In this review, we summarize the pathophysiological mechanisms involved in the development of SPMS and the rationale and clinical potential for natalizumab, which is currently approved for the treatment of relapsing forms of MS, to exert beneficial effects in reducing disease progression unrelated to relapses in SPMS. In both forms of MS, active brain-tissue injury is associated with inflammation; but in SPMS, the inflammatory response occurs at least partly behind the blood-brain barrier and is followed by a cascade of events, including persistent microglial activation that may lead to chronic demyelination and neurodegeneration associated with irreversible disability. In patients with relapsing forms of MS, natalizumab therapy is known to significantly reduce intrathecal inflammatory responses which results in reductions in brain lesions and brain atrophy as well as beneficial effects on clinical measures, such as reduced frequency and severity of relapse and reduced accumulation of disability. Natalizumab treatment also reduces levels of cerebrospinal fluid chemokines and other biomarkers of intrathecal inflammation, axonal damage and demyelination, and has demonstrated the ability to reduce innate immune activation and intrathecal immunoglobulin synthesis in patients with MS. The efficacy of natalizumab therapy in SPMS is currently being investigated in a randomized, double-blind, placebo-controlled trial.
Collapse
Affiliation(s)
- Finn Sellebjerg
- Danish Multiple Sclerosis Center, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Luisa Maria Villar
- Department of Immunology, Ramón y Cajal University Hospital, Institute Ramón y Cajal for Biomedical Research, Madrid, Spain
| | - Richard Reynolds
- Division of Brain Sciences, Imperial College London, Hammersmith Hospital Campus, London, UK
| | | |
Collapse
|
110
|
Pikor NB, Prat A, Bar-Or A, Gommerman JL. Meningeal Tertiary Lymphoid Tissues and Multiple Sclerosis: A Gathering Place for Diverse Types of Immune Cells during CNS Autoimmunity. Front Immunol 2016; 6:657. [PMID: 26793195 PMCID: PMC4710700 DOI: 10.3389/fimmu.2015.00657] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 12/22/2015] [Indexed: 11/13/2022] Open
Abstract
Collections of leukocytes in the meningeal space have been documented in Multiple Sclerosis (MS). These meningeal aggregates, which in the context of other autoimmune diseases have often been termed tertiary lymphoid tissues (TLT), have been associated with sub-pial cortical damage and disease progression. However, the key molecular and cellular signals required for their formation and maintenance remain unclear. Herein, we review TLT structures in other disease states in order to provide a framework for understanding these structures in the MS meninges. We then assess the evidence that the meningeal compartment serves as an important nexus for immune cells as well as a location for drainage of antigen into cervical lymph nodes. Extrapolating what is known about the molecular and cellular cues that initiate the formation of leukocyte aggregates in non-lymphoid tissues, we speculate on what signals lead to the formation and maintenance of meningeal TLT structures. Referring to the animal model of MS [experimental autoimmune encephalomyelitis (EAE)], we also explore what is known about these structures in supporting B cell and T cell responses during neuroinflammation. Last, we examine the evidence that connects these structures to ongoing neuropathology. Collectively, our review points to the meningeal compartment as an important player in neuroinflammatory processes. Moreover, we hypothesize that in order to gain insights into pro- and anti-inflammatory properties of lymphocytes in MS, one must understand the cellular scaffolds that support lymphocyte retention within the meninges, thus highlighting the importance of non-immune cells (stromal cells) in the neuroinflammatory process.
Collapse
Affiliation(s)
- Natalia B Pikor
- Department of Immunology, University of Toronto , Toronto, ON , Canada
| | - Alexandre Prat
- Neuroimmunology Unit, Department of Neuroscience, Centre de Recherche de CHUM, Université de Montréal , Montreal, QC , Canada
| | - Amit Bar-Or
- Neuroimmunology Unit, Montreal Neurological Institute and Hospital, McGill University , Montreal, QC , Canada
| | | |
Collapse
|
111
|
Jiang BC, Cao DL, Zhang X, Zhang ZJ, He LN, Li CH, Zhang WW, Wu XB, Berta T, Ji RR, Gao YJ. CXCL13 drives spinal astrocyte activation and neuropathic pain via CXCR5. J Clin Invest 2016; 126:745-61. [PMID: 26752644 DOI: 10.1172/jci81950] [Citation(s) in RCA: 237] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 11/19/2015] [Indexed: 12/21/2022] Open
Abstract
Recent studies have implicated chemokines in microglial activation and pathogenesis of neuropathic pain. C-X-C motif chemokine 13 (CXCL13) is a B lymphocyte chemoattractant that activates CXCR5. Using the spinal nerve ligation (SNL) model of neuropathic pain, we found that CXCL13 was persistently upregulated in spinal cord neurons after SNL, resulting in spinal astrocyte activation via CXCR5 in mice. shRNA-mediated inhibition of CXCL13 in the spinal cord persistently attenuated SNL-induced neuropathic pain. Interestingly, CXCL13 expression was suppressed by miR-186-5p, a microRNA that colocalized with CXCL13 and was downregulated after SNL. Spinal overexpression of miR-186-5p decreased CXCL13 expression, alleviating neuropathic pain. Furthermore, SNL induced CXCR5 expression in spinal astrocytes, and neuropathic pain was abrogated in Cxcr5-/- mice. CXCR5 expression induced by SNL was required for the SNL-induced activation of spinal astrocytes and microglia. Intrathecal injection of CXCL13 was sufficient to induce pain hypersensitivity and astrocyte activation via CXCR5 and ERK. Finally, intrathecal injection of CXCL13-activated astrocytes induced mechanical allodynia in naive mice. Collectively, our findings reveal a neuronal/astrocytic interaction in the spinal cord by which neuronally produced CXCL13 activates astrocytes via CXCR5 to facilitate neuropathic pain. Thus, miR-186-5p and CXCL13/CXCR5-mediated astrocyte signaling may be suitable therapeutic targets for neuropathic pain.
Collapse
|
112
|
Lee JY, Biemond M, Petratos S. Axonal degeneration in multiple sclerosis: defining therapeutic targets by identifying the causes of pathology. Neurodegener Dis Manag 2015; 5:527-48. [DOI: 10.2217/nmt.15.50] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Current therapeutics in multiple sclerosis (MS) target the putative inflammation and immune attack on CNS myelin. Despite their effectiveness in blunting the relapse rate in MS patients, such therapeutics do not prevent MS disease progression. Importantly, specific clinical dilemma arises through inability to predict MS progression and thereby therapeutically target axonal injury during MS, limiting permanent disability. The current review identifies immune and neurobiological principles that govern the sequelae of axonal degeneration during MS disease progression. Defining the specific disease arbiters, inflammatory and autoimmune, oligodendrocyte dystrophy and degenerative myelin, we discuss a basis for a molecular mechanism in axons that may be targeted therapeutically, in spatial and temporal manner to limit axonal degeneration and thereby halt progression of MS.
Collapse
Affiliation(s)
- Jae Young Lee
- Department of Medicine, Central Clinical School, Monash University, Prahran VIC 3004, Australia
| | - Melissa Biemond
- Department of Medicine, Central Clinical School, Monash University, Prahran VIC 3004, Australia
| | - Steven Petratos
- Department of Medicine, Central Clinical School, Monash University, Prahran VIC 3004, Australia
| |
Collapse
|
113
|
Kannel K, Alnek K, Vahter L, Gross-Paju K, Uibo R, Kisand KV. Changes in Blood B Cell-Activating Factor (BAFF) Levels in Multiple Sclerosis: A Sign of Treatment Outcome. PLoS One 2015; 10:e0143393. [PMID: 26600308 PMCID: PMC4658115 DOI: 10.1371/journal.pone.0143393] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 11/04/2015] [Indexed: 01/21/2023] Open
Abstract
Multiple sclerosis (MS) is mediated primarily by autoreactive T cells. However, evidence suggesting the involvement of humoral immunity in brain diseases has increased interest in the role of B cells and their products during MS pathogenesis. The major survival factor for B cells, BAFF has been shown to play a role in several autoimmune conditions. Elevated BAFF levels have been reported in MS animal model and during MS relapse in patients. Moreover, disease-modifying treatments (DMT) reportedly influence blood BAFF levels in MS patients, but the significance of these changes remains unclear. The present study addresses how blood BAFF levels are associated with the clinical course of relapsing-remitting MS and the effectiveness of DMT and short-term steroid treatment. During a prospective longitudinal follow-up of 2.3 years, BAFF was measured in the blood of 170 MS patients in the stable phase and within 186 relapses. BAFF levels were significantly higher in MS patients compared to healthy controls. However, stable MS patients without relapses exhibited significantly higher BAFF levels than relapsing patients. Treatment with interferon-β and immunosuppressants raised BAFF blood levels. Interestingly, a similar effect was not seen in patients treated with glatiramer acetate. Short-term treatment with high doses of intravenous methylprednisolone did not significantly alter plasma BAFF levels in 65% of relapsing-remitting MS patients. BAFF were correlated weakly but significantly with monocyte and basophil counts, but not with other blood cell types (neutrophils, lymphocytes, or eosinophils) or inflammatory biomarkers. To our knowledge, this is the first report demonstrating that higher blood BAFF levels may reflect a more stable and effective MS treatment outcome. These results challenge hypotheses suggesting that elevated blood BAFF levels are associated with more severe disease presentation and could explain the recent failure of pharmaceutical trials targeting BAFF with soluble receptor for MS treatment.
Collapse
Affiliation(s)
- Karin Kannel
- West-Tallinn Central Hospital MS Centre, Tallinn, Estonia
| | - Kristi Alnek
- Department of Immunology, Institute of Biomedicine and Translational Medicine, Tartu University, Tartu, Estonia
| | - Liina Vahter
- West-Tallinn Central Hospital MS Centre, Tallinn, Estonia
- Institute of Psychology, Tallinn University, Tallinn, Estonia
| | - Katrin Gross-Paju
- West-Tallinn Central Hospital MS Centre, Tallinn, Estonia
- Institute of Clinical Medicine, Tallinn University of Technology, Tallinn, Estonia
| | - Raivo Uibo
- Department of Immunology, Institute of Biomedicine and Translational Medicine, Tartu University, Tartu, Estonia
| | - Kalle V. Kisand
- Department of Immunology, Institute of Biomedicine and Translational Medicine, Tartu University, Tartu, Estonia
- * E-mail:
| |
Collapse
|
114
|
Abstract
The chemokine, C-X-C motif ligand 13 (CXCL13), is constitutively expressed in lymphoid organs and controls the recruitment and compartmentalization of lymphocytes and antigen presenting cells within these specialized structures. Recent data, however, also find induction of this molecule during central nervous system (CNS) inflammation under a variety of circumstances. While its role(s) in the pathogenesis of neoplastic, infectious and autoimmune disorders of the CNS remain incompletely understood, several lines of evidence suggest that CXCL13 could become a relevant therapeutic target in at least some of these diseases. This review focuses on how CXCL13 contributes to the pathogenesis of selected CNS disorders involving both experimental animals and humans, paying particular attention to the issue of whether (and if so, how) blockade of this ligand or its receptor might benefit the host. Current blocking strategies largely involve the use of monoclonal antibodies, but an improved understanding of downstream signaling pathways makes small molecule inhibition a future possibility.
Collapse
Affiliation(s)
- Amanda K Huber
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - David N Irani
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
115
|
Hoorweg K, Narang P, Li Z, Thuery A, Papazian N, Withers DR, Coles MC, Cupedo T. A Stromal Cell Niche for Human and Mouse Type 3 Innate Lymphoid Cells. THE JOURNAL OF IMMUNOLOGY 2015; 195:4257-4263. [PMID: 26378073 DOI: 10.4049/jimmunol.1402584] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 08/22/2015] [Indexed: 11/19/2022]
Abstract
Adaptive immunity critically depends on the functional compartmentalization of secondary lymphoid organs. Mesenchymal stromal cells create and maintain specialized niches that support survival, activation, and expansion of T and B cells, and integrated analysis of lymphocytes and their niche has been instrumental in understanding adaptive immunity. Lymphoid organs are also home to type 3 innate lymphoid cells (ILC3), innate effector cells essential for barrier immunity. However, a specialized stromal niche for ILC3 has not been identified. A novel lineage-tracing approach now identifies a subset of murine fetal lymphoid tissue organizer cells that gives rise exclusively to adult marginal reticular cells. Moreover, both cell types are conserved from mice to humans and colocalize with ILC3 in secondary lymphoid tissues throughout life. In sum, we provide evidence that fetal stromal organizers give rise to adult marginal reticular cells and form a dedicated stromal niche for innate ILC3 in adaptive lymphoid organs.
Collapse
Affiliation(s)
- Kerim Hoorweg
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Priyanka Narang
- Centre for Immunology and Infection, Department of Biology and Hull York Medical School, University of York, York, UK
| | - Zhi Li
- Centre for Immunology and Infection, Department of Biology and Hull York Medical School, University of York, York, UK
| | - Anne Thuery
- Centre for Immunology and Infection, Department of Biology and Hull York Medical School, University of York, York, UK
| | - Natalie Papazian
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - David R Withers
- Medical Research Council Centre for Immune Regulation, College of Medical and Dental Sciences, University of Birmingham, Birmingham, U.K
| | - Mark C Coles
- Centre for Immunology and Infection, Department of Biology and Hull York Medical School, University of York, York, UK
| | - Tom Cupedo
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
116
|
Blockade of B-cell activating factor with TACI-IgG effectively reduced Th1 and Th17 cells but not memory T cells in experimental allergic encephalomyelitis mice. Cent Eur J Immunol 2015; 40:142-8. [PMID: 26557026 PMCID: PMC4637387 DOI: 10.5114/ceji.2015.52826] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Accepted: 04/09/2015] [Indexed: 12/15/2022] Open
Abstract
B-cell activating factor (BAFF) is regarded as a new therapeutic target in autoimmune diseases such as systemic lupus erythematosus (SLE) and multiple sclerosis (MS). Along with other researchers, we have demonstrated that BAFF inhibitor atacicept (TACI-IgG) suppresses lupus and experimental allergic encephalomyelitis (EAE) by reducing the mature B-cell number but not memory B cells. It is however unclear whether TACI-Ig affects pathogenic T cells and memory T cells. In the present study, we found that blocking BAFF with TACI-IgG effectively reduces the pathogenic Th1 and Th17 cells in EAE mice. However, TACI-IgG did not reduce memory CD62L+CD44hiCD4+ and CD62L+CD44hiCD8+ T cells in EAE mice. When interleukin (IL)-15 was neutralized, memory CD62L+CD44hi T cells were significantly reduced in TACI-IgG-treated EAE mice. These results suggest that TACI-IgG is effective in effective controlling Th1 and Th17 cells, but it also increases IL-15 to upregulate memory T cells in EAE mice. The study provides hints for the clinical application of the combination of BAFF- and IL-15-specific therapeutic agents.
Collapse
|
117
|
Fan X, Lin C, Han J, Jiang X, Zhu J, Jin T. Follicular Helper CD4+ T Cells in Human Neuroautoimmune Diseases and Their Animal Models. Mediators Inflamm 2015; 2015:638968. [PMID: 26300592 PMCID: PMC4537760 DOI: 10.1155/2015/638968] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Accepted: 02/16/2015] [Indexed: 01/27/2023] Open
Abstract
Follicular helper CD4(+) T (TFH) cells play a fundamental role in humoral immunity deriving from their ability to provide help for germinal center (GC) formation, B cell differentiation into plasma cells and memory cells, and antibody production in secondary lymphoid tissues. TFH cells can be identified by a combination of markers, including the chemokine receptor CXCR5, costimulatory molecules ICOS and PD-1, transcription repressor Bcl-6, and cytokine IL-21. It is difficult and impossible to get access to secondary lymphoid tissues in humans, so studies are usually performed with human peripheral blood samples as circulating counterparts of tissue TFH cells. A balance of TFH cell generation and function is critical for protective antibody response, whereas overactivation of TFH cells or overexpression of TFH-associated molecules may result in autoimmune diseases. Emerging data have shown that TFH cells and TFH-associated molecules may be involved in the pathogenesis of neuroautoimmune diseases including multiple sclerosis (MS), neuromyelitis optica (NMO)/neuromyelitis optica spectrum disorders (NMOSD), and myasthenia gravis (MG). This review summarizes the features of TFH cells, including their development, function, and roles as well as TFH-associated molecules in neuroautoimmune diseases and their animal models.
Collapse
Affiliation(s)
- Xueli Fan
- Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun 130021, China
| | - Chenhong Lin
- Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun 130021, China
| | - Jinming Han
- Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun 130021, China
| | - Xinmei Jiang
- Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun 130021, China
| | - Jie Zhu
- Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun 130021, China
- Department of Neurobiology, Care Sciences and Society, Karolinska Institute, 14186 Stockholm, Sweden
| | - Tao Jin
- Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun 130021, China
| |
Collapse
|
118
|
Abstract
While over the past decades T cells have been considered key players in the pathogenesis of multiple sclerosis (MS), it has only recently become evident that B cells have a major contributing role. Our understanding of the role of B cells has evolved substantially following the clinical success of B cell-targeting therapies and increasing experimental evidence for significant B cell involvement. Rather than mere antibody-producing cells, it is becoming clear that they are team players with the capacity to prime and regulate T cells, and function both as pro- and anti-inflammatory mediators. However, despite tremendous efforts, the target antigen(s) of B cells in MS have yet to be identified. The first part of this review summarizes the clinical evidence and results from animal studies pointing to the relevance of B cells in the pathogenesis of MS. The second part gives an overview of the currently known potential autoantigen targets. The third part recapitulates and critically appraises the currently available B cell-directed therapies.
Collapse
|
119
|
Ferraro D, Galli V, Vitetta F, Simone AM, Bedin R, Del Giovane C, Morselli F, Filippini MM, Nichelli PF, Sola P. Cerebrospinal fluid CXCL13 in clinically isolated syndrome patients: Association with oligoclonal IgM bands and prediction of Multiple Sclerosis diagnosis. J Neuroimmunol 2015; 283:64-9. [PMID: 26004159 DOI: 10.1016/j.jneuroim.2015.04.011] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 04/21/2015] [Accepted: 04/22/2015] [Indexed: 12/12/2022]
Abstract
Cerebrospinal fluid (CSF) CXCL13 was shown to correlate with markers of intrathecal inflammation and CSF oligoclonal IgM bands (IgMOB) have been associated with a more severe Multiple Sclerosis (MS) course. We correlated CSF CXCL13 levels with clinical, MRI and CSF parameters, including CSF IgMOB, in 110 Clinically Isolated Syndrome (CIS) patients. CSF CXCL13 levels correlated with CSF cell count, total protein, IgG Index and with the presence of CSF IgGOB and IgMOB. CSF CXCL13 levels ≥15.4 pg/ml showed a good positive predictive value and specificity for a MS diagnosis and for a clinical relapse within one year from onset.
Collapse
Affiliation(s)
- Diana Ferraro
- Neurology Unit, Department of Biomedical, Metabolic and Neurosciences, University of Modena and Reggio Emilia, Via Pietro Giardini, 1355, 41126 Modena, Italy.
| | - Veronica Galli
- Neuroimmunology Laboratory, Department of Biomedical, Metabolic and Neurosciences, University of Modena and Reggio Emilia, Via Pietro Giardini, 1355, 41126 Modena, Italy.
| | - Francesca Vitetta
- Neurology Unit, Department of Biomedical, Metabolic and Neurosciences, University of Modena and Reggio Emilia, Via Pietro Giardini, 1355, 41126 Modena, Italy.
| | - Anna Maria Simone
- Neurology Unit, Department of Biomedical, Metabolic and Neurosciences, University of Modena and Reggio Emilia, Via Pietro Giardini, 1355, 41126 Modena, Italy.
| | - Roberta Bedin
- Neuroimmunology Laboratory, Department of Biomedical, Metabolic and Neurosciences, University of Modena and Reggio Emilia, Via Pietro Giardini, 1355, 41126 Modena, Italy.
| | - Cinzia Del Giovane
- Department of Diagnostic and Clinical Medicine and Public Health, University of Modena and Reggio Emilia, Via del Pozzo, 70, 41100 Modena, Italy.
| | - Franca Morselli
- Neurology Unit, Department of Biomedical, Metabolic and Neurosciences, University of Modena and Reggio Emilia, Via Pietro Giardini, 1355, 41126 Modena, Italy.
| | - Maria Maddalena Filippini
- Neurology Unit, Department of Biomedical, Metabolic and Neurosciences, University of Modena and Reggio Emilia, Via Pietro Giardini, 1355, 41126 Modena, Italy.
| | - Paolo Frigio Nichelli
- Neurology Unit, Department of Biomedical, Metabolic and Neurosciences, University of Modena and Reggio Emilia, Via Pietro Giardini, 1355, 41126 Modena, Italy.
| | - Patrizia Sola
- Neurology Unit, Department of Biomedical, Metabolic and Neurosciences, University of Modena and Reggio Emilia, Via Pietro Giardini, 1355, 41126 Modena, Italy.
| |
Collapse
|
120
|
Calabrese M, Magliozzi R, Ciccarelli O, Geurts JJG, Reynolds R, Martin R. Exploring the origins of grey matter damage in multiple sclerosis. Nat Rev Neurosci 2015; 16:147-58. [PMID: 25697158 DOI: 10.1038/nrn3900] [Citation(s) in RCA: 307] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Multiple sclerosis is characterized at the gross pathological level by the presence of widespread focal demyelinating lesions of the myelin-rich white matter. However, it is becoming clear that grey matter is not spared, even during the earliest phases of the disease. Furthermore, grey matter damage may have an important role both in physical and cognitive disability. Grey matter pathology involves both inflammatory and neurodegenerative mechanisms, but the relationship between the two is unclear. Histological, immunological and neuroimaging studies have provided new insight in this rapidly expanding field, and form the basis of the most recent hypotheses on the pathogenesis of grey matter damage.
Collapse
Affiliation(s)
- Massimiliano Calabrese
- Advanced Neuroimaging Laboratory of Neurology B, Department of Neurological and Movement Sciences, University Hospital Verona, Piazzale Ludovico Antonio Scuro 10, 37134, Verona, Italy
| | - Roberta Magliozzi
- 1] Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London W12 0NN, UK. [2] Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome, Italy
| | - Olga Ciccarelli
- 1] National Institute for Health Research, University College London/University College London Hospitals NHS Foundation Trust (NIHR UCL/UCLH) Biomedical Research Centre, 149 Tottenham Court Road, London W1T 7DN, UK. [2] Queen Square Multiple Sclerosis Centre, University College London, Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Jeroen J G Geurts
- Section of Clinical Neuroscience, Department of Anatomy and Neurosciences, VU University Medical Center, van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands
| | - Richard Reynolds
- Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London W12 0NN, UK
| | - Roland Martin
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University Hospital Zurich, University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| |
Collapse
|
121
|
Parker Harp CR, Archambault AS, Sim J, Ferris ST, Mikesell RJ, Koni PA, Shimoda M, Linington C, Russell JH, Wu GF. B cell antigen presentation is sufficient to drive neuroinflammation in an animal model of multiple sclerosis. THE JOURNAL OF IMMUNOLOGY 2015; 194:5077-84. [PMID: 25895531 DOI: 10.4049/jimmunol.1402236] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 03/20/2015] [Indexed: 11/19/2022]
Abstract
B cells are increasingly regarded as integral to the pathogenesis of multiple sclerosis, in part as a result of the success of B cell-depletion therapy. Multiple B cell-dependent mechanisms contributing to inflammatory demyelination of the CNS have been explored using experimental autoimmune encephalomyelitis (EAE), a CD4 T cell-dependent animal model for multiple sclerosis. Although B cell Ag presentation was suggested to regulate CNS inflammation during EAE, direct evidence that B cells can independently support Ag-specific autoimmune responses by CD4 T cells in EAE is lacking. Using a newly developed murine model of in vivo conditional expression of MHC class II, we reported previously that encephalitogenic CD4 T cells are incapable of inducing EAE when B cells are the sole APC. In this study, we find that B cells cooperate with dendritic cells to enhance EAE severity resulting from myelin oligodendrocyte glycoprotein (MOG) immunization. Further, increasing the precursor frequency of MOG-specific B cells, but not the addition of soluble MOG-specific Ab, is sufficient to drive EAE in mice expressing MHCII by B cells alone. These data support a model in which expansion of Ag-specific B cells during CNS autoimmunity amplifies cognate interactions between B and CD4 T cells and have the capacity to independently drive neuroinflammation at later stages of disease.
Collapse
Affiliation(s)
- Chelsea R Parker Harp
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110
| | - Angela S Archambault
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110
| | - Julia Sim
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110
| | - Stephen T Ferris
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Robert J Mikesell
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110
| | - Pandelakis A Koni
- Cancer Immunology, Inflammation, and Tolerance Program, Cancer Center and Department of Medicine, Georgia Regents University, Augusta, GA 30912
| | - Michiko Shimoda
- Department of Dermatology, University of California at Davis School of Medicine, Sacramento, CA 95817; and
| | | | - John H Russell
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110
| | - Gregory F Wu
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110;
| |
Collapse
|
122
|
Palanichamy A, Apeltsin L, Kuo TC, Sirota M, Wang S, Pitts SJ, Sundar PD, Telman D, Zhao LZ, Derstine M, Abounasr A, Hauser SL, von Büdingen HC. Immunoglobulin class-switched B cells form an active immune axis between CNS and periphery in multiple sclerosis. Sci Transl Med 2015; 6:248ra106. [PMID: 25100740 DOI: 10.1126/scitranslmed.3008930] [Citation(s) in RCA: 177] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
In multiple sclerosis (MS), lymphocyte--in particular B cell--transit between the central nervous system (CNS) and periphery may contribute to the maintenance of active disease. Clonally related B cells exist in the cerebrospinal fluid (CSF) and peripheral blood (PB) of MS patients; however, it remains unclear which subpopulations of the highly diverse peripheral B cell compartment share antigen specificity with intrathecal B cell repertoires and whether their antigen stimulation occurs on both sides of the blood-brain barrier. To address these questions, we combined flow cytometric sorting of PB B cell subsets with deep immune repertoire sequencing of CSF and PB B cells. Immunoglobulin (IgM and IgG) heavy chain variable (VH) region repertoires of five PB B cell subsets from MS patients were compared with their CSF Ig-VH transcriptomes. In six of eight patients, we identified peripheral CD27(+)IgD(-) memory B cells, CD27(hi)CD38(hi) plasma cells/plasmablasts, or CD27(-)IgD(-) B cells that had an immune connection to the CNS compartment. Pinpointing Ig class-switched B cells as key component of the immune axis thought to contribute to ongoing MS disease activity strengthens the rationale of current B cell-targeting therapeutic strategies and may lead to more targeted approaches.
Collapse
Affiliation(s)
| | | | - Tracy C Kuo
- Rinat-Pfizer Inc., 230 East Grand Avenue, South San Francisco, CA 94080, USA
| | - Marina Sirota
- Rinat-Pfizer Inc., 230 East Grand Avenue, South San Francisco, CA 94080, USA
| | - Shengzhi Wang
- Department of Neurology, UCSF, San Francisco, CA 94148 USA
| | - Steven J Pitts
- Rinat-Pfizer Inc., 230 East Grand Avenue, South San Francisco, CA 94080, USA
| | - Purnima D Sundar
- Rinat-Pfizer Inc., 230 East Grand Avenue, South San Francisco, CA 94080, USA
| | - Dilduz Telman
- Rinat-Pfizer Inc., 230 East Grand Avenue, South San Francisco, CA 94080, USA
| | - Lora Z Zhao
- Rinat-Pfizer Inc., 230 East Grand Avenue, South San Francisco, CA 94080, USA
| | - Mia Derstine
- Department of Neurology, UCSF, San Francisco, CA 94148 USA
| | - Aya Abounasr
- Department of Neurology, UCSF, San Francisco, CA 94148 USA
| | | | | |
Collapse
|
123
|
Oxombre B, Lee-Chang C, Duhamel A, Toussaint M, Giroux M, Donnier-Maréchal M, Carato P, Lefranc D, Zéphir H, Prin L, Melnyk P, Vermersch P. High-affinity σ1 protein agonist reduces clinical and pathological signs of experimental autoimmune encephalomyelitis. Br J Pharmacol 2015; 172:1769-82. [PMID: 25521311 PMCID: PMC4376455 DOI: 10.1111/bph.13037] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 10/16/2014] [Accepted: 11/16/2014] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND AND PURPOSE Selective agonists of the sigma-1 receptor (σ1 protein) are generally reported to protect against neuronal damage and modulate oligodendrocyte differentiation. Human and rodent lymphocytes possess saturable, high-affinity binding sites for compounds binding to the σ1 protein and potential immunomodulatory properties have been described for σ1 protein ligands. Experimental autoimmune encephalomyelitis (EAE) is recognized as a valuable model of the inflammatory aspects of multiple sclerosis (MS). Here, we have assessed the role of a σ1 protein agonist, containing the tetrahydroisoquinoline-hydantoin structure, in EAE. EXPERIMENTAL APPROACH EAE was induced in SJL/J female mice by active immunization with myelin proteolipid protein (PLP)139-151 peptide. The σ1 protein agonist was injected i.p. at the time of immunization (day 0). Disease severity was assessed clinically and by histopathological evaluation of the CNS. Phenotyping of B-cell subsets and regulatory T-cells were performed by flow cytometry in spleen and cervical lymph nodes. KEY RESULTS Prophylactic treatment of EAE mice with the σ1 protein agonist prevented mononuclear cell accumulation and demyelination in brain and spinal cord and increased T2 B-cells and regulatory T-cells, resulting in an overall reduction in the clinical progression of EAE. CONCLUSIONS AND IMPLICATIONS This σ1 protein agonist, containing the tetrahydroisoquinoline-hydantoin structure, decreased the magnitude of inflammation in EAE. This effect was associated with increased proportions of B-cell subsets and regulatory T-cells with potential immunoregulatory functions. Targeting of the σ1 protein might thus provide new therapeutic opportunities in MS.
Collapse
MESH Headings
- Animals
- B-Lymphocytes/immunology
- Brain/drug effects
- Brain/pathology
- Cytokines/blood
- Encephalomyelitis, Autoimmune, Experimental/blood
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Female
- Immunoglobulin G/blood
- Lymph Nodes/drug effects
- Lymph Nodes/immunology
- Mice
- Multiple Sclerosis/blood
- Multiple Sclerosis/drug therapy
- Multiple Sclerosis/immunology
- Multiple Sclerosis/pathology
- Myelin Proteolipid Protein/immunology
- Neuroprotective Agents/pharmacology
- Neuroprotective Agents/therapeutic use
- Peptide Fragments/immunology
- Receptors, sigma/agonists
- Spinal Cord/drug effects
- Spinal Cord/pathology
- Spleen/drug effects
- Spleen/immunology
- T-Lymphocytes, Regulatory/immunology
- Sigma-1 Receptor
Collapse
Affiliation(s)
- B Oxombre
- Université de LilleLille, France
- UDSL, EA2686-LIRIC, UFR MédecineLille, France
| | - C Lee-Chang
- Université de LilleLille, France
- UDSL, EA2686-LIRIC, UFR MédecineLille, France
| | - A Duhamel
- Université de LilleLille, France
- UDSL, EA 2694, UFR MédecineLille, France
| | - M Toussaint
- Université de LilleLille, France
- CNRS UMR8161Lille, France
| | - M Giroux
- Université de LilleLille, France
- UDSL, EA2686-LIRIC, UFR MédecineLille, France
- Centre Hospitalier Régional et Universitaire de Lille, Pôle de neurologie–Service de Neurologie DLille, France
| | - M Donnier-Maréchal
- Université de LilleLille, France
- UDSL, EA 4481, UFR PharmacieLille, France
| | - P Carato
- Université de LilleLille, France
- UDSL, EA 4481, UFR PharmacieLille, France
| | - D Lefranc
- Université de LilleLille, France
- UDSL, EA2686-LIRIC, UFR MédecineLille, France
| | - H Zéphir
- Université de LilleLille, France
- UDSL, EA2686-LIRIC, UFR MédecineLille, France
- Centre Hospitalier Régional et Universitaire de Lille, Pôle de neurologie–Service de Neurologie DLille, France
| | - L Prin
- Université de LilleLille, France
- UDSL, EA2686-LIRIC, UFR MédecineLille, France
- Centre Hospitalier Régional et Universitaire de Lille, Pôle d'immunologie–Centre de Biologie Pathologie et GénétiqueLille, France
| | - P Melnyk
- Université de LilleLille, France
- CNRS UMR8161Lille, France
- UDSL, EA 4481, UFR PharmacieLille, France
- Inserm UMR-S1172, Jean-Pierre Aubert Research CenterLille, France
| | - P Vermersch
- Université de LilleLille, France
- UDSL, EA2686-LIRIC, UFR MédecineLille, France
- UDSL, EA 4481, UFR PharmacieLille, France
| |
Collapse
|
124
|
von Büdingen HC, Palanichamy A, Lehmann-Horn K, Michel BA, Zamvil SS. Update on the autoimmune pathology of multiple sclerosis: B-cells as disease-drivers and therapeutic targets. Eur Neurol 2015; 73:238-246. [PMID: 25824054 DOI: 10.1159/000377675] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 02/01/2015] [Indexed: 01/10/2023]
Abstract
BACKGROUND Collectively, research on the role of B-cells in the pathogenesis of multiple sclerosis (MS) illustrates how translational medicine has given rise to promising therapeutic approaches for one of the most debilitating chronic neurological diseases in young adults. First described in 1935, the experimental autoimmune/allergic encephalomyelitis model is a key animal model that has provided the foundation for important developments in targeted therapeutics. SUMMARY While additional B-cell therapies for MS are presently being developed by the pharmaceutical industry, much remains to be understood about the role played by B-cells in MS. The goal of this review is to summarize how B-cells may contribute to MS pathogenesis and thereby provide a basis for understanding why B-cell depletion is so effective in the treatment of this disease. Key Messages: B-cells are key players in the pathogenesis of MS, and their depletion via B-cell-targeted therapy ameliorates disease activity. CLINICAL IMPLICATIONS In 2008, data from the first CD20-targeting B-cell depleting therapeutic trials using rituximab in MS were published. Since then, there has been a large body of evidence demonstrating the effectiveness of B-cell depletion mediated via anti-CD20 antibodies. Intense research efforts focusing on the immunopathological relevance of B-cells has gained significant momentum and given rise to a constellation of promising therapeutic agents for this complex B-cell-driven disease, including novel anti-CD20 antibodies, as well as agents targeting CD19 and BAFF-R.
Collapse
|
125
|
Intrathecal IgG synthesis: a resistant and valuable target for future multiple sclerosis treatments. Mult Scler Int 2015; 2015:296184. [PMID: 25653878 PMCID: PMC4306411 DOI: 10.1155/2015/296184] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Revised: 12/15/2014] [Accepted: 12/16/2014] [Indexed: 01/02/2023] Open
Abstract
Intrathecal IgG synthesis is a key biological feature of multiple sclerosis (MS). When acquired early, it persists over time. A growing body of evidence suggests that intrathecal Ig-secreting cells may be pathogenic either by a direct action of toxic IgG or by locally secreting bystander toxic products. Intrathecal IgG synthesis depends on the presence of CNS lymphoid organs, which are strongly linked at anatomical level to cortical subpial lesions and at clinical level to the impairment slope in progressive MS. As a consequence, targeting CNS lymphoid lesions could be a valuable new target in MS, especially during the progressive phase. As intrathecal IgGs are end-products of these lymphoid lesions, intrathecal IgG synthesis may be considered as a specific marker of the persistence of these inflammatory lesions. Here we review the effect upon intrathecal IgG synthesis of all drugs ever used in MS. Except for steroids, all these therapeutic strategies, including rituximab, failed to decrease intrathecal IgG synthesis, with the exception of a questionable incomplete action of natalizumab. Thus, IgG synthesis is a robust marker of persistent intrathecal inflammation and its complete normalization should be one of the goals in future therapeutic strategies.
Collapse
|
126
|
DeLuca GC, Yates RL, Beale H, Morrow SA. Cognitive impairment in multiple sclerosis: clinical, radiologic and pathologic insights. Brain Pathol 2015; 25:79-98. [PMID: 25521179 PMCID: PMC8029470 DOI: 10.1111/bpa.12220] [Citation(s) in RCA: 142] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 10/15/2014] [Indexed: 01/18/2023] Open
Abstract
Cognitive impairment is a common and debilitating feature of multiple sclerosis (MS) that has only recent gained considerable attention. Clinical neuropsychological studies have made apparent the multifaceted nature of cognitive troubles often encountered in MS and continue to broaden our understanding of its complexity. Radiographic studies have started to decipher the neuroanatomic substrate of MS-related cognitive impairment and have shed light onto its pathogenesis. Where radiographic studies have been limited by inadequate resolution or non-specificity, pathological studies have come to the fore. This review aims to provide an overview of the nature of cognitive impairment typically seen in MS and to explore the literature on imaging and pathological studies relevant to its evolution. In particular, the relative contributions of gray (i.e., cerebral cortex, hippocampus, thalamus and basal ganglia) and white matter to MS-related cognitive impairment will be discussed and the importance of interconnectivity between structures highlighted. The pressing need for longitudinal studies combining standardized neuropsychometric, paraclinical and radiographic outcomes obtained during life with post-mortem tissue analysis after death is presented.
Collapse
Affiliation(s)
- Gabriele C. DeLuca
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Richard L. Yates
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Harry Beale
- Oxford Medical SchoolUniversity of OxfordOxfordUK
| | - Sarah A. Morrow
- Department of Clinical Neurological SciencesThe University of Western OntarioLondonCanada
| |
Collapse
|
127
|
Bonnan M. [Meningeal tertiary lymphoid organs: Major actors in intrathecal autoimmunity]. Rev Neurol (Paris) 2014; 171:65-74. [PMID: 25555848 DOI: 10.1016/j.neurol.2014.08.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2014] [Revised: 07/24/2014] [Accepted: 08/29/2014] [Indexed: 12/29/2022]
Abstract
Multiple sclerosis (MS) is characterized by an intrathecal synthesis of immunoglobulins synthesized by B-cell clones and by a brain infiltrate of clonal T-cells. The clonal maturation of these lymphocytes takes place in tertiary lymphoid organs (TLO) developed in the intrathecal compartment. TLO are acquired lymphoid organs able to develop in the vicinity of the inflammatory sites, where they mount a complete antigen-driven immune response. We here review TLO pathophysiology in animal models of MS and human MS. Several pieces of evidence suggest that intrathecal TLO may play a major role in the clinical impairment. Potential therapeutic applications are examined.
Collapse
Affiliation(s)
- M Bonnan
- Service de neurologie, hôpital F.-Mitterrand, 4, boulevard Hauterive, 64000 Pau, France.
| |
Collapse
|
128
|
Miyazaki Y, Niino M. Molecular targeted therapy against B cells in multiple sclerosis. ACTA ACUST UNITED AC 2014. [DOI: 10.1111/cen3.12160] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Yusei Miyazaki
- Department of Clinical Research; Hokkaido Medical Center; Sapporo Japan
- Department of Neurology; Hokkaido Medical Center; Sapporo Japan
| | - Masaaki Niino
- Department of Clinical Research; Hokkaido Medical Center; Sapporo Japan
| |
Collapse
|
129
|
Clarkson BD, Walker A, Harris MG, Rayasam A, Sandor M, Fabry Z. CCR2-dependent dendritic cell accumulation in the central nervous system during early effector experimental autoimmune encephalomyelitis is essential for effector T cell restimulation in situ and disease progression. THE JOURNAL OF IMMUNOLOGY 2014; 194:531-41. [PMID: 25505278 DOI: 10.4049/jimmunol.1401320] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Dendritic cells (DCs)--although absent from the healthy CNS parenchyma--rapidly accumulate within brain and spinal cord tissue during neuroinflammation associated with experimental autoimmune encephalomyelitis (EAE; a mouse model of multiple sclerosis). Yet, although DCs have been appreciated for their role in initiating adaptive immune responses in peripheral lymphoid organ tissues, how DCs infiltrate the CNS and contribute to ongoing neuroinflammation in situ is poorly understood. In this study, we report the following: 1) CD11c(+) bone marrow-derived DCs and CNS-infiltrating DCs express chemokine receptor CCR2; 2) compared with CCR2(+/+) cells, adoptively transferred CCR2(-/-) bone marrow-derived DCs or DC precursors do not accumulate in the CNS during EAE, despite abundance in blood; 3) CCR2(-/-) DCs show less accumulation in the inflamed CNS in mixed bone marrow chimeras, when compared with CCR2(+/+) DCs; and 4) ablation of CCR2(+/+) DCs during EAE clinical onset delays progression and attenuates cytokine production by infiltrating T cells. Whereas the role of CCR2 in monocyte migration into the CNS has been implicated previously, the role of CCR2 in DC infiltration into the CNS has never been directly addressed. Our data suggest that CCR2-dependent DC recruitment to the CNS during ongoing neuroinflammation plays a crucial role in effector T cell cytokine production and disease progression, and signify that CNS-DCs and circulating DC precursors might be key therapeutic targets for suppressing ongoing neuroinflammation in CNS autoimmune diseases.
Collapse
Affiliation(s)
- Benjamin D Clarkson
- Department of Pathology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792; Department of Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792; Graduate Training Program of Cellular and Molecular Pathology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792; and
| | - Alec Walker
- Department of Pathology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792; Department of Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792
| | - Melissa G Harris
- Department of Pathology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792; Department of Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792
| | - Aditya Rayasam
- Graduate Training Program of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792
| | - Matyas Sandor
- Department of Pathology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792; Department of Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792
| | - Zsuzsanna Fabry
- Department of Pathology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792; Department of Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792;
| |
Collapse
|
130
|
Anti-CXCL13 antibody can inhibit the formation of gastric lymphoid follicles induced by Helicobacter infection. Mucosal Immunol 2014; 7:1244-54. [PMID: 24646940 DOI: 10.1038/mi.2014.14] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2013] [Accepted: 02/09/2014] [Indexed: 02/04/2023]
Abstract
Helicobacter suis infects the stomachs of both animals and humans, and can induce gastric mucosa-associated lymphoid tissue (MALT) lymphomas. It is known that CXC chemokine ligand 13 (CXCL13) is highly expressed in the Helicobacter-infected mice and gastric MALT lymphoma patients, but the pathway that links the activation of CXCL13 and the formation of gastric MALT lymphomas remains unclear. In this study, we examined whether CXCL13 neutralization would interfere with the formation of gastric lymphoid follicles including B cells, CD4+T cells, dendritic cells (DCs), and follicular DCs (FDCs) in germinal centers to determine the role of CXCL13 in the formation of B-cell aggregates after H. suis infection. Moreover, the expression of genes associated with the lymphoid follicle formation was also effectively suppressed by anti-CXCL13 antibody treatment. These results suggest that the upregulation of CXCL13 has an important role in the development of gastric MALT lymphomas and highlight the potential of anti-CXCL13 antibody for protection against Helicobacter-induced gastric diseases.
Collapse
|
131
|
Haugen M, Frederiksen JL, Degn M. B cell follicle-like structures in multiple sclerosis—With focus on the role of B cell activating factor. J Neuroimmunol 2014; 273:1-7. [DOI: 10.1016/j.jneuroim.2014.05.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 04/21/2014] [Accepted: 05/16/2014] [Indexed: 12/21/2022]
|
132
|
Berrih-Aknin S. Myasthenia Gravis: paradox versus paradigm in autoimmunity. J Autoimmun 2014; 52:1-28. [PMID: 24934596 DOI: 10.1016/j.jaut.2014.05.001] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 05/07/2014] [Indexed: 12/12/2022]
Abstract
Myasthenia Gravis (MG) is a paradigm of organ-specific autoimmune disease (AID). It is mediated by antibodies that target the neuromuscular junction. The purpose of this review is to place MG in the general context of autoimmunity, to summarize the common mechanisms between MG and other AIDs, and to describe the specific mechanisms of MG. We have chosen the most common organ-specific AIDs to compare with MG: type 1 diabetes mellitus (T1DM), autoimmune thyroid diseases (AITD), multiple sclerosis (MS), some systemic AIDs (systemic lupus erythematous (SLE), rheumatoid arthritis (RA), Sjogren's syndrome (SS)), as well as inflammatory diseases of the gut and liver (celiac disease (CeD), Crohn's disease (CD), and primary biliary cirrhosis (PBC)). Several features are similar between all AIDs, suggesting that common pathogenic mechanisms lead to their development. In this review, we address the predisposing factors (genetic, epigenetic, hormones, vitamin D, microbiota), the triggering components (infections, drugs) and their interactions with the immune system [1,2]. The dysregulation of the immune system is detailed and includes the role of B cells, Treg cells, Th17 and cytokines. We particularly focused on the role of TNF-α and interferon type I whose role in MG is very analogous to that in several other AIDS. The implication of AIRE, a key factor in central tolerance is also discussed. Finally, if MG is a prototype of AIDS, it has a clear specificity compared to the other AIDS, by the fact that the target organ, the muscle, is not the site of immune infiltration and B cell expansion, but exclusively that of antibody-mediated pathogenic mechanisms. By contrast, the thymus in the early onset subtype frequently undergoes tissue remodeling, resulting in the development of ectopic germinal centers surrounded by high endothelial venules (HEV), as observed in the target organs of many other AIDs.
Collapse
Affiliation(s)
- Sonia Berrih-Aknin
- Sorbonne Universités, UPMC Univ Paris 06, Myology Research Center UM76, F-75013 Paris, France; INSERM U974, F-75013 Paris, France; CNRS FRE 3617, F-75013 Paris, France; Institute of Myology, F-75013 Paris, France.
| |
Collapse
|
133
|
Progression from IgD+ IgM+ to isotype-switched B cells is site specific during coronavirus-induced encephalomyelitis. J Virol 2014; 88:8853-67. [PMID: 24872583 DOI: 10.1128/jvi.00861-14] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
UNLABELLED Various infections in the central nervous system (CNS) trigger B cell accumulation; however, the relative dynamics between viral replication and alterations in distinct B cell subsets are largely unknown. Using a glia-tropic coronavirus infection, which is initiated in the brain but rapidly spreads to and predominantly persists in the spinal cord, this study characterizes longitudinal changes in B cell subsets at both infected anatomical sites. The phase of T cell-dependent, antibody-independent control of infectious virus was associated with a similar recruitment of naive/early-activated IgD(+) IgM(+) B cells into both the brain and spinal cord. This population was progressively replaced by CD138(-) IgD(-) IgM(+) B cells, isotype-switched CD138(-) IgD(-) IgM(-) memory B cells (B(mem)), and CD138(+) antibody-secreting cells (ASC). A more rapid transition to B(mem) and ASC in spinal cord than in brain was associated with higher levels of persisting viral RNA and transcripts encoding factors promoting B cell migration, differentiation, and survival. The results demonstrate that naive/early-activated B cells are recruited early during coronavirus CNS infection but are subsequently replaced by more differentiated B cells. Furthermore, viral persistence, even at low levels, is a driving force for accumulation of isotype-switched B(mem) and ASC. IMPORTANCE Acute and chronic human CNS infections are associated with an accumulation of heterogeneous B cell subsets; however, their influence on viral load and disease is unclear. Using a glia-tropic coronavirus model, we demonstrate that the accumulation of B cells ranging from early-activated to isotype-switched differentiation stages is both temporally and spatially orchestrated. Acutely infected brains and spinal cords indiscriminately recruit a homogeneous population of early-activated B cells, which is progressively replaced by diverse, more differentiated subsets. The latter process is accelerated by elevated proinflammatory responses associated with viral persistence. The results imply that early-recruited B cells do not have antiviral function but may contribute to the inflammatory environment or act as antigen-presenting cells. Moreover, CNS viral persistence is a driving force promoting differentiated B cells with protective potential.
Collapse
|
134
|
Esen N, Rainey-Barger EK, Huber AK, Blakely PK, Irani DN. Type-I interferons suppress microglial production of the lymphoid chemokine, CXCL13. Glia 2014; 62:1452-62. [PMID: 24829092 PMCID: PMC4143141 DOI: 10.1002/glia.22692] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 04/28/2014] [Accepted: 04/29/2014] [Indexed: 12/23/2022]
Abstract
Lymphoid chemokines are crucial for the development and maintenance of lymphoid organs, but their ectopic expression in non-lymphoid tissues is implicated in both local response to infection and chronic organ-specific autoimmunity. Production of one such chemokine, C-X-C motif ligand 13 (CXCL13), within the central nervous system (CNS) has been linked to the pathogenesis of multiple sclerosis (MS), although little is known about factors controlling its expression in different neural cell types and across a range of disease states. We provoked acute neuroinflammation in experimental animals without causing any associated demyelination using neuroadapted Sindbis virus (NSV) to better understand the sources and regulators of this chemokine in the CNS. We found that mice genetically deficient in the transcription factor, interferon (IFN) regulatory factor-7 (IRF7), made significantly higher CXCL13 protein levels in the CNS compared with wild-type (WT) controls. Microglia proved to be the main producer of CXCL13 in the brain during infection of both WT and IRF7−/− mice, and primary microglia cultured in vitro generated CXCL13 following stimulation with either virus particles or synthetic Toll-like receptor (TLR) ligands. Microglia cultured from IRF7−/− mice selectively overproduced CXCL13, and manipulation of extracellular type-I IFN levels demonstrated the existence of a negative feedback loop whereby type-I IFN receptor signaling specifically suppressed microglial CXCL13 release. Since IFN-β is used to treat patients with relapsing-remitting MS and yet acts through unknown mechanisms, we speculate that suppressed lymphoid chemokine production by microglia could contribute to its therapeutic effects.
Collapse
Affiliation(s)
- Nilufer Esen
- Department of Neurology, University of Michigan Medical School, Ann Arbor, Michigan
| | | | | | | | | |
Collapse
|
135
|
The role of lymphotoxin signaling in the development of autoimmune pancreatitis and associated secondary extra-pancreatic pathologies. Cytokine Growth Factor Rev 2014; 25:125-37. [DOI: 10.1016/j.cytogfr.2014.01.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 12/23/2013] [Accepted: 01/02/2014] [Indexed: 12/24/2022]
|
136
|
Ma N, Xing C, Xiao H, He Y, Han G, Chen G, Hou C, Marrero B, Wang Y, Zhang S, Shen B, Li Y, Wang R. BAFF Suppresses IL-15 Expression in B Cells. THE JOURNAL OF IMMUNOLOGY 2014; 192:4192-201. [DOI: 10.4049/jimmunol.1302132] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
137
|
BAFF maintains T-cell survival by inducing OPN expression in B cells. Mol Immunol 2014; 57:129-37. [DOI: 10.1016/j.molimm.2013.08.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 08/23/2013] [Accepted: 08/24/2013] [Indexed: 01/10/2023]
|
138
|
Schreiner B, Bailey SL, Miller SD. T-cell response dynamics in animal models of multiple sclerosis: implications for immunotherapies. Expert Rev Clin Immunol 2014; 3:57-72. [DOI: 10.1586/1744666x.3.1.57] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
139
|
The experimental autoimmune encephalomyelitis (EAE) model of MS: utility for understanding disease pathophysiology and treatment. HANDBOOK OF CLINICAL NEUROLOGY 2014; 122:173-89. [PMID: 24507518 DOI: 10.1016/b978-0-444-52001-2.00008-x] [Citation(s) in RCA: 342] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
While no single model can exactly recapitulate all aspects of multiple sclerosis (MS), animal models are essential in understanding the induction and pathogenesis of the disease and to develop therapeutic strategies that limit disease progression and eventually lead to effective treatments for the human disease. Several different models of MS exist, but by far the best understood and most commonly used is the rodent model of experimental autoimmune encephalomyelitis (EAE). This model is typically induced by either active immunization with myelin-derived proteins or peptides in adjuvant or by passive transfer of activated myelin-specific CD4+ T lymphocytes. Mouse models are most frequently used because of the inbred genotype of laboratory mice, their rapid breeding capacity, the ease of genetic manipulation, and availability of transgenic and knockout mice to facilitate mechanistic studies. Although not all therapeutic strategies for MS have been developed in EAE, all of the current US Food and Drug Administration (FDA)-approved immunomodulatory drugs are effective to some degree in treating EAE, a strong indicator that EAE is an extremely useful model to study potential treatments for MS. Several therapies, such as glatiramer acetate (GA: Copaxone), and natalizumab (Tysabri), were tested first in the mouse model of EAE and then went on to clinical trials. Here we discuss the usefulness of the EAE model in understanding basic disease pathophysiology and developing treatments for MS as well as the potential drawbacks of this model.
Collapse
|
140
|
Soellner IA, Rabe J, Mauri V, Kaufmann J, Addicks K, Kuerten S. Differential aspects of immune cell infiltration and neurodegeneration in acute and relapse experimental autoimmune encephalomyelitis. Clin Immunol 2013; 149:519-29. [DOI: 10.1016/j.clim.2013.10.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2013] [Revised: 10/17/2013] [Accepted: 10/19/2013] [Indexed: 12/11/2022]
|
141
|
Chaitanya GV, Omura S, Sato F, Martinez NE, Minagar A, Ramanathan M, Guttman BW, Zivadinov R, Tsunoda I, Alexander JS. Inflammation induces neuro-lymphatic protein expression in multiple sclerosis brain neurovasculature. J Neuroinflammation 2013; 10:125. [PMID: 24124909 PMCID: PMC3854084 DOI: 10.1186/1742-2094-10-125] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 09/24/2013] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is associated with ectopic lymphoid follicle formation. Podoplanin+ (lymphatic marker) T helper17 (Th17) cells and B cell aggregates have been implicated in the formation of tertiary lymphoid organs (TLOs) in MS and experimental autoimmune encephalitis (EAE). Since podoplanin expressed by Th17 cells in MS brains is also expressed by lymphatic endothelium, we investigated whether the pathophysiology of MS involves inductions of lymphatic proteins in the inflamed neurovasculature. METHODS We assessed the protein levels of lymphatic vessel endothelial hyaluronan receptor and podoplanin, which are specific to the lymphatic system and prospero-homeobox protein-1, angiopoietin-2, vascular endothelial growth factor-D, vascular endothelial growth factor receptor-3, which are expressed by both lymphatic endothelium and neurons. Levels of these proteins were measured in postmortem brains and sera from MS patients, in the myelin proteolipid protein (PLP)-induced EAE and Theiler's murine encephalomyelitis virus (TMEV) induced demyelinating disease (TMEV-IDD) mouse models and in cell culture models of inflamed neurovasculature. RESULTS AND CONCLUSIONS Intense staining for LYVE-1 was found in neurons of a subset of MS patients using immunohistochemical approaches. The lymphatic protein, podoplanin, was highly expressed in perivascular inflammatory lesions indicating signaling cross-talks between inflamed brain vasculature and lymphatic proteins in MS. The profiles of these proteins in MS patient sera discriminated between relapsing remitting MS from secondary progressive MS and normal patients. The in vivo findings were confirmed in the in vitro cell culture models of neuroinflammation.
Collapse
Affiliation(s)
- Ganta Vijay Chaitanya
- Department of Molecular & Cellular Physiology, School of Medicine, Louisiana State University Health-Shreveport, 1501 Kings Highway, Shreveport, LA, 71130, USA
| | - Seiichi Omura
- Department of Microbiology and Immunology, School of Medicine, Louisiana State University Health-Shreveport, 1501 Kings Highway, Shreveport, LA, 71130, USA
| | - Fumitaka Sato
- Department of Microbiology and Immunology, School of Medicine, Louisiana State University Health-Shreveport, 1501 Kings Highway, Shreveport, LA, 71130, USA
| | - Nicholas E Martinez
- Department of Microbiology and Immunology, School of Medicine, Louisiana State University Health-Shreveport, 1501 Kings Highway, Shreveport, LA, 71130, USA
| | - Alireza Minagar
- Department of Neurology, School of Medicine, Louisiana State University Health-Shreveport, 1501 Kings Highway, Shreveport, LA, 71130, USA
| | - Murali Ramanathan
- Department of Neurology, State University of New York, Buffalo, NY, USA
- Department of Pharmaceutical Sciences, State University of New York, Buffalo, NY, USA
| | | | - Robert Zivadinov
- Department of Neurology, State University of New York, Buffalo, NY, USA
- Buffalo Neuroimaging Analysis Center, State University of New York, Buffalo, NY, USA
| | - Ikuo Tsunoda
- Department of Microbiology and Immunology, School of Medicine, Louisiana State University Health-Shreveport, 1501 Kings Highway, Shreveport, LA, 71130, USA
| | - Jonathan S Alexander
- Department of Molecular & Cellular Physiology, School of Medicine, Louisiana State University Health-Shreveport, 1501 Kings Highway, Shreveport, LA, 71130, USA
| |
Collapse
|
142
|
Gasperini C, Haggiag S, Ruggieri S. Drugs in clinical development for multiple sclerosis: focusing on anti-CD20 antibodies. Expert Opin Investig Drugs 2013; 22:1243-53. [PMID: 23855792 DOI: 10.1517/13543784.2013.820275] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS), traditionally considered to be an autoimmune disease. Despite the standard of care for patients with MS is significantly improved in recent years, there is still room for improvement in terms of effectiveness and also compliance. AREAS COVERED The continuous improvements of our understanding of the pathophysiological changes that occur in MS have translated into many novel therapeutic agents at different stages of development. A number of therapies for MS are in advanced development and likely to be available soon. Along with these, we have also seen the appearance of a group of drugs considered together as a consequence of their similar design: the monoclonal antibodies (mAbs). Here, the focus will be on reviewing results that have emerged from a better clarification of MS pathogenesis to clinical trials of different anti-CD20 mAbs. EXPERT OPINION The decision to switch established patients from well-known drugs to either new formulations or new agents will be made on balancing efficacy and tolerability of the existing treatments. Safety seems increasingly likely to become a key factor.
Collapse
Affiliation(s)
- Claudio Gasperini
- Multiple Sclerosis Center, S Camillo-Forlanini, Neurosciences , Circonvallazione Gianicolense 87, Rome 00152 , Italy
| | | | | |
Collapse
|
143
|
Minagar A, Barnett MH, Benedict RHB, Pelletier D, Pirko I, Sahraian MA, Frohman E, Zivadinov R. The thalamus and multiple sclerosis: modern views on pathologic, imaging, and clinical aspects. Neurology 2013; 80:210-9. [PMID: 23296131 DOI: 10.1212/wnl.0b013e31827b910b] [Citation(s) in RCA: 258] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The paired thalamic nuclei are gray matter (GM) structures on both sides of the third ventricle that play major roles in cortical activation, relaying sensory information to the higher cortical centers that influence cognition. Multiple sclerosis (MS) is an immune-mediated disease of the human CNS that affects both the white matter (WM) and GM. A number of clinical observations as well as recent neuropathologic and neuroimaging studies have clearly demonstrated extensive involvement of the thalamus, basal ganglia, and neocortex in patients with MS. Modern MRI techniques permit visualization of GM lesions and measurement of atrophy. These contemporary methods have fundamentally altered our understanding of the pathophysiologic nature of MS. Evidence confirms the contention that GM injury can be detected in the earliest phases of MS, and that iron deposition and atrophy of deep gray nuclei are closely related to the magnitude of inflammation. Extensive involvement of GM, and particularly of the thalamus, is associated with a wide range of clinical manifestations including cognitive decline, motor deficits, fatigue, painful syndromes, and ocular motility disturbances in patients with MS. In this review, we characterize the neuropathologic, neuroimaging, and clinical features of thalamic involvement in MS. Further, we underscore the contention that neuropathologic and neuroimaging correlative investigations of thalamic derangements in MS may elucidate not heretofore considered pathobiological underpinnings germane to understanding the ontogeny, magnitude, and progression of the disease process.
Collapse
Affiliation(s)
- Alireza Minagar
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | | | | | | | | | | | | | | |
Collapse
|
144
|
Phares TW, Stohlman SA, Bergmann CC. Intrathecal humoral immunity to encephalitic RNA viruses. Viruses 2013; 5:732-52. [PMID: 23435240 PMCID: PMC3640523 DOI: 10.3390/v5020732] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 02/10/2013] [Accepted: 02/11/2013] [Indexed: 12/21/2022] Open
Abstract
The nervous system is the target for acute encephalitic viral infections, as well as a reservoir for persisting viruses. Intrathecal antibody (Ab) synthesis is well documented in humans afflicted by infections associated with neurological complications, as well as the demyelinating disease, multiple sclerosis. This review focuses on the origin, recruitment, maintenance, and biological relevance of Ab-secreting cells (ASC) found in the central nervous system (CNS) following experimental neurotropic RNA virus infections. We will summarize evidence for a highly dynamic, evolving humoral response characterized by temporal alterations in B cell subsets, proliferation, and differentiation. Overall local Ab plays a beneficial role via complement-independent control of virus replication, although cross or self-reactive Ab to CNS antigens may contribute to immune-mediated pathogenesis during some infections. Importantly, protective Ab exert anti-viral activity not only by direct neutralization, but also by binding to cell surface-expressed viral glycoproteins. Ab engagement of viral glycoproteins blocks budding and mediates intracellular signaling leading to restored homeostatic and innate functions. The sustained Ab production by local ASC, as well as chemokines and cytokines associated with ASC recruitment and retention, are highlighted as critical components of immune control.
Collapse
Affiliation(s)
- Timothy W Phares
- Departments of Neurosciences NC30, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA.
| | | | | |
Collapse
|
145
|
Grogan JL, Ouyang W. A role for Th17 cells in the regulation of tertiary lymphoid follicles. Eur J Immunol 2013; 42:2255-62. [PMID: 22949324 DOI: 10.1002/eji.201242656] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Immune responses propagate in secondary lymphoid organs (SLOs), such as the spleen and lymph nodes. These highly organized structures are typified by distinct B-cell follicles and T-cell zones, and are orchestrated by interactions between the TNF superfamily molecules expressed on hematopoietic cells and their receptors on mesenchymal cells and the subsequent cytokines and chemokines that are elicited. During chronic immune responses, cellular effectors of the immune response can infiltrate target tissue and organize anatomically into de novo B-cell follicles and T-cell areas, a phenomenon called lymphoid neogenesis or the formation of tertiary lymphoid organs (TLOs). Critical to the development of SLOs are lymphoid-tissue inducer (LTi) cells, that is innate lymphoid cells that arise from common precursor cells within the fetal liver. Of interest, Th17 cells, a subset of CD4(+) T cells most associated with autoimmune pathogenesis, share many developmental and effector markers with LTi cells. Here, we compare and contrast LTi and Th17 cells, and review recent evidence that Th17 cells and Th17 cytokines, such as IL-17 and IL-22, contribute to the development of ectopic lymphoid structures in chronic-ally inflamed tissue.
Collapse
Affiliation(s)
- Jane L Grogan
- Department of Immunology, Genentech Inc., South San Francisco, CA 94080, USA.
| | | |
Collapse
|
146
|
Astrocyte-derived CXCL10 drives accumulation of antibody-secreting cells in the central nervous system during viral encephalomyelitis. J Virol 2013; 87:3382-92. [PMID: 23302888 DOI: 10.1128/jvi.03307-12] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Microbial infections of the central nervous system (CNS) are often associated with local accumulation of antibody (Ab)-secreting cells (ASC). By providing a source of Ab at the site of infection, CNS-localized ASC play a critical role in acute viral control and in preventing viral recrudescence. Following coronavirus-induced encephalomyelitis, the CNS accumulation of ASC is chemokine (C-X-C motif) receptor 3 (CXCR3) dependent. This study demonstrates that CNS-expressed CXCR3 ligand CXCL10 is the critical chemokine regulating ASC accumulation. Impaired ASC recruitment in CXCL10(-/-) but not CXCL9(-/-) mice was consistent with reduced CNS IgG and κ-light chain mRNA and virus-specific Ab. Moreover, the few ASC recruited to the CNS in CXCL10(-/-) mice were confined to the vasculature, distinct from the parenchymal localization in wild-type and CXCL9(-/-) mice. However, neither CXCL9 nor CXCL10 deficiency diminished neutralizing serum Ab, supporting a direct role for CXCL10 in ASC migration. T cell accumulation, localization, and effector functions were also not affected in either CXCL9(-/-) or CXCL10(-/-) mice, consistent with similar control of infectious virus. There was also no evidence for dysregulation of chemokines or cytokines involved in ASC regulation. The distinct roles of CXCL9 and CXCL10 in ASC accumulation rather coincided with their differential localization. While CXCL10 was predominantly expressed by astrocytes, CXCL9 expression was confined to the vasculature/perivascular spaces. These results suggest that CXCL10 is critical for two phases: recruitment of ASC to the CNS vasculature and ASC entry into the CNS parenchyma.
Collapse
|
147
|
Abstract
Daclizumab is a humanized monoclonal antibody of IgG1 subtype that binds to the Tac epitope on the interleukin-2 (IL-2) receptor α-chain (CD25), thus, effectively blocking the formation of the high-affinity IL-2 receptor. Because the high-affinity IL-2 receptor signaling promotes expansion of activated T cells in vitro, daclizumab was designed as a therapy that selectively inhibits T-cell activation. Assuming the previous statement, daclizumab received regulatory approval as add-on therapy to standard immunosuppressive regimen for the prevention of acute allograft rejection in renal transplantation. Based on its putative mechanism of action (MOA), daclizumab represented an ideal therapy for T-cell-mediated autoimmune diseases and was subsequently tested in inflammatory uveitis and multiple sclerosis (MS). In both of these diseases, daclizumab therapy significantly inhibited target organ inflammation. Mechanistic studies in MS demonstrated that the MOA of daclizumab is surprisingly broad and that the drug exerts unexpected effects on multiple components of the innate immune system. Specifically, daclizumab dramatically expands and activates immunoregulatory CD56(bright) NK cells, which gain access to the intrathecal compartment in MS and can kill autologous activated T cells. Daclizumab also blocks trans-presentation of IL-2 by mature dendritic cells to primed T cells, resulting in profound inhibition of antigen-specific T cells. Finally, daclizumab modulates the development of innate lymphoid cells. In conclusion, daclizumab therapy, which is currently in phase III testing for inflammatory MS, has a unique MOA that does not limit migration of immune cells into the intrathecal compartment, but rather provides multifactorial immunomodulatory effects with resultant inhibition of MS-related inflammation.
Collapse
Affiliation(s)
- Bibiana Bielekova
- Neuroimmunological Diseases Unit (NDU), Neuroimmunology Branch (NIB), National Institute of Neurological Disorders and Stroke (NINDS), Bethesda, MD 20892, USA.
| |
Collapse
|
148
|
Christy AL, Walker ME, Hessner MJ, Brown MA. Mast cell activation and neutrophil recruitment promotes early and robust inflammation in the meninges in EAE. J Autoimmun 2012; 42:50-61. [PMID: 23267561 DOI: 10.1016/j.jaut.2012.11.003] [Citation(s) in RCA: 134] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Revised: 11/07/2012] [Accepted: 11/12/2012] [Indexed: 02/06/2023]
Abstract
The meninges are often considered inert tissues that house the CSF and provide protection for the brain and spinal cord. Yet emerging data demonstrates that they are also active sites of immune responses. Furthermore, the blood-CSF barrier surrounding meningeal blood vessels, together with the blood-brain barrier (BBB), is postulated to serve as a gateway for the pathological infiltration of immune cells into the CNS in multiple sclerosis (MS). Our previous studies using mast cell-deficient (Kit(W/Wv)) mice demonstrated that mast cells resident in the dura mater and pia mater exacerbate experimental autoimmune encephalomyelitis (EAE), a rodent model of MS, by facilitating CNS inflammatory cell influx. Here we examined the underlying mechanisms that mediate these effects. We demonstrate that there are dramatic alterations in immune associated gene expression in the meninges in pre-clinical disease, including those associated with mast cell and neutrophil function. Meningeal mast cells are activated within 24 h of disease induction, but do not directly compromise CNS vascular integrity. Rather, through production of TNF, mast cells elicit an early influx of neutrophils, cells known to alter vascular permeability, into the meninges. These data add to the growing evidence that inflammation in the meninges precedes CNS immune cell infiltration and establish that mast cells are among the earliest participants in these disease-initiating events. We hypothesize that mast cell-dependent neutrophil recruitment and activation in the meninges promotes early breakdown of the local BBB and CSF-blood barrier allowing initial immune cell access to the CNS.
Collapse
Affiliation(s)
- Alison L Christy
- Northwestern University, Feinberg School of Medicine, Department of Microbiology and Immunology, Chicago, IL, USA
| | | | | | | |
Collapse
|
149
|
Lisak RP, Ragheb S. The role of B cell-activating factor in autoimmune myasthenia gravis. Ann N Y Acad Sci 2012; 1274:60-7. [DOI: 10.1111/j.1749-6632.2012.06842.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
150
|
Tertiary lymphoid organ development coincides with determinant spreading of the myelin-specific T cell response. Acta Neuropathol 2012; 124:861-73. [PMID: 22842876 DOI: 10.1007/s00401-012-1023-3] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Revised: 07/08/2012] [Accepted: 07/18/2012] [Indexed: 01/07/2023]
Abstract
While the role of T cells has been studied extensively in multiple sclerosis (MS), the pathogenic contribution of B cells has only recently attracted major attention, when it was shown that B cell aggregates can develop in the meninges of a subset of MS patients and were suggested to be correlates of late-stage and more aggressive disease in this patient population. However, whether these aggregates actually exist has subsequently been questioned and their functional significance has remained unclear. Here, we studied myelin basic protein (MBP)-proteolipid protein (PLP)-induced experimental autoimmune encephalomyelitis (EAE), which is one of the few animal models for MS that is dependent on B cells. We provide evidence that B cell aggregation is reflective of lymphoid neogenesis in the central nervous system (CNS) in MBP-PLP-elicited EAE. B cell aggregation was present already few days after disease onset. With disease progression CNS B cell aggregates increasingly displayed the phenotype of tertiary lymphoid organs (TLOs). Our results further imply that these TLOs were not merely epiphenomena of the disease, but functionally active, supporting intrathecal determinant spreading of the myelin-specific T cell response. Our data suggest that the CNS is not a passive "immune-privileged" target organ, but rather a compartment, in which highly active immune responses can perpetuate and amplify the autoimmune pathology and thereby autonomously contribute to disease progression.
Collapse
|