51
|
Pender MP, Csurhes PA, Smith C, Douglas NL, Neller MA, Matthews KK, Beagley L, Rehan S, Crooks P, Hopkins TJ, Blum S, Green KA, Ioannides ZA, Swayne A, Aftab BT, Hooper KD, Burrows SR, Thompson KM, Coulthard A, Khanna R. Epstein-Barr virus-specific T cell therapy for progressive multiple sclerosis. JCI Insight 2018; 3:124714. [PMID: 30429369 DOI: 10.1172/jci.insight.124714] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 10/16/2018] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Increasing evidence indicates a role for EBV in the pathogenesis of multiple sclerosis (MS). EBV-infected autoreactive B cells might accumulate in the CNS because of defective cytotoxic CD8+ T cell immunity. We sought to determine the feasibility and safety of treating progressive MS patients with autologous EBV-specific T cell therapy. METHODS An open-label phase I trial was designed to treat 5 patients with secondary progressive MS and 5 patients with primary progressive MS with 4 escalating doses of in vitro-expanded autologous EBV-specific T cells targeting EBV nuclear antigen 1, latent membrane protein 1 (LMP1), and LMP2A. Following adoptive immunotherapy, we monitored the patients for safety and clinical responses. RESULTS Of the 13 recruited participants, 10 received the full course of T cell therapy. There were no serious adverse events. Seven patients showed improvement, with 6 experiencing both symptomatic and objective neurological improvement, together with a reduction in fatigue, improved quality of life, and, in 3 patients, reduced intrathecal IgG production. All 6 patients receiving T cells with strong EBV reactivity showed clinical improvement, whereas only 1 of the 4 patients receiving T cells with weak EBV reactivity showed improvement (P = 0.033, Fisher's exact test). CONCLUSION EBV-specific adoptive T cell therapy was well tolerated. Clinical improvement following treatment was associated with the potency of EBV-specific reactivity of the administered T cells. Further clinical trials are warranted to determine the efficacy of EBV-specific T cell therapy in MS. TRIAL REGISTRATION Australian New Zealand Clinical Trials Registry, ACTRN12615000422527. FUNDING MS Queensland, MS Research Australia, Perpetual Trustee Company Ltd., and donations from private individuals who wish to remain anonymous.
Collapse
Affiliation(s)
- Michael P Pender
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Department of Neurology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia.,QIMR Berghofer Centre for Immunotherapy and Vaccine Development, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Peter A Csurhes
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,The University of Queensland Centre for Clinical Research, Brisbane, Queensland, Australia
| | - Corey Smith
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Nanette L Douglas
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Department of Neurology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Michelle A Neller
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Katherine K Matthews
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Leone Beagley
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Sweera Rehan
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Pauline Crooks
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Tracey J Hopkins
- Internal Medicine Day Treatment Unit, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Stefan Blum
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Department of Neurology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Kerryn A Green
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Department of Neurology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Zara A Ioannides
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Department of Neurology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Andrew Swayne
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Department of Neurology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Blake T Aftab
- Preclinical Science and Translational Medicine, Atara Biotherapeutics, South San Francisco, California, USA
| | - Kaye D Hooper
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Department of Neurology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Scott R Burrows
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,QIMR Berghofer Centre for Immunotherapy and Vaccine Development, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Kate M Thompson
- Department of Psychology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia.,Faculty of Health and Behavioural Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Alan Coulthard
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Department of Medical Imaging, Royal Brisbane and Women's Hospital, Brisbane,Queensland, Australia
| | - Rajiv Khanna
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,QIMR Berghofer Centre for Immunotherapy and Vaccine Development, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
52
|
Gut microbiota, cannabinoid system and neuroimmune interactions: New perspectives in multiple sclerosis. Biochem Pharmacol 2018; 157:51-66. [PMID: 30171835 DOI: 10.1016/j.bcp.2018.08.037] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 08/22/2018] [Indexed: 02/07/2023]
Abstract
The gut microbiota plays a fundamental role on the education and function of the host immune system. Immunological dysregulation is the cause of numerous human disorders such as autoimmune diseases and metabolic disorders frequently associated with inflammatory processes therefore is critical to explore novel mechanisms involved in maintaining the immune system homeostasis. The cannabinoid system and related bioactive lipids participate in multiple central and peripheral physiological processes that affect metabolic, gastrointestinal and neuroimmune regulatory mechanisms displaying a modulatory role and contributing to the maintenance of the organism's homeostasis. In this review, we gather the knowledge on the gut microbiota-endocannabinoids interactions and their impact on autoimmune disorders such as inflammatory bowel disease, rheumatoid arthritis and particularly, multiple sclerosis (MS) as the best example of a CNS autoimmune disorder. Furthermore, we contribute to this field with new data on changes in many elements of the cannabinoid system in a viral model of MS after gut microbiota manipulation by both antibiotics and probiotics. Finally, we highlight new therapeutic opportunities, under an integrative view, targeting the eCBS and the commensal microbiota in the context of neuroinflammation and MS.
Collapse
|
53
|
Michel L. Environmental factors in the development of multiple sclerosis. Rev Neurol (Paris) 2018; 174:372-377. [PMID: 29735167 DOI: 10.1016/j.neurol.2018.03.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 03/06/2018] [Accepted: 03/08/2018] [Indexed: 10/17/2022]
Abstract
Epidemiology of Multiple Sclerosis (MS) has been intensively studied and we know now that its occurrence result from the combined action of genetic and environmental factors. There are significant geographic and temporal variations in MS incidence and the risk associated with the development of MS may be affected by many potential factors (including infections, climate, diet, etc.). But none of these factors has been identified as "causal". The accumulation of these different agents as well as their interactions probably contribute to the development of the disease.
Collapse
Affiliation(s)
- L Michel
- Department of Neurology, Hôpital Pontchaillou, 2, rue Henri-Le-Guilloux, 35000 Rennes, France.
| |
Collapse
|
54
|
Abstract
Vasculitides are disorders characterized by inflammation of the vessel walls, often caused by autoimmunity, but sometimes as a result of microbial invasion. Almost all types of microbes including bacteria, viruses, protozoa and fungi have been incriminated in the pathogenesis of vasculitis. Accurate etiological diagnosis is important since immunosuppressive treatment may lead to further deterioration if infection is the cause of vasculitis. Clinical features sometimes provide clues to the etiology. Further evaluation requires a focused and cost-effective plan of laboratory investigation. The investigations aim at establishing the diagnosis of vasculitis and identify the causative organism. An accurate diagnosis of vasculitis optimally requires histological examination and imaging. For infection-associated vasculitis, the identification of the organism requires studies of stained specimens, cultures, and/or detection of antigens and antibodies. Ideally, the treatment involves administration of an appropriate antimicrobial. In non-self-limiting types of vasculitides, glucocorticoids are needed when the symptoms are progressive, with vital organs involvement, and sometimes, when there is no antimicrobial agent of proven efficacy against the incriminated agent. Additional immunosuppressive agents or interventions must be considered when the disease is severe and/or post-infective immune mechanisms are involved in the pathogenesis, e.g., severe HBV- or HCV-associated vasculitides. Available preventative vaccinations are also crucial. The incidence of HBV-associated vasculitides dramatically decreased following HBV vaccination campaigns, and other infection-associated vasculitides may as well in the future.
Collapse
Affiliation(s)
- Syed A Haq
- BSM Medical University, Dhaka, Bangladesh
| | | |
Collapse
|
55
|
Abstract
Primary Sjögren syndrome (pSS) is a prototypical autoimmune disease. The involvement of B cells in the pathogenesis of pSS has long been suspected on the basis of clinical observations that include the presence of serum autoantibodies, hypergammaglobulinaemia, increased levels of free light chains and increased risk of B cell lymphoma. Moreover, the composition of the B cell subset is altered in pSS. In this Review, we discuss the mechanisms that support the increased activation of B cells in pSS, including genetic and epigenetic factors and environmental triggers that promote B cell activation via the innate immune system. B cell activating factor (BAFF, also known as TNF ligand superfamily member 13B) is at the crossroads of this process. An important role also exists for the target tissue (exocrine glands, namely the salivary and lachrymal glands), which promotes local B cell activation. This continuous stimulation of B cells is the main driver of lymphomatous escape. Identification of the multiple steps that support B cell activation has led to the development of promising targeted therapies that will hopefully lead to the development of an efficient therapeutic strategy for pSS.
Collapse
|
56
|
Dendrou CA, Fugger L. Immunomodulation in multiple sclerosis: promises and pitfalls. Curr Opin Immunol 2017; 49:37-43. [PMID: 28926740 DOI: 10.1016/j.coi.2017.08.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 08/26/2017] [Indexed: 12/31/2022]
Abstract
Multiple sclerosis (MS) afflicts about 2.5 million people globally and poses a major personal and socioeconomic burden. The recognition of MS as an inflammatory disease, characterized by infiltration of immune cells into the central nervous system, has spurred research into the autoimmune etiology of the condition and has provided the rationale for its treatment through immunomodulation. Experience with immunotherapies in MS to date has suggested a disparity between the observed immune cell infiltration and the progressive loss of neurons. However, recent clinical efforts are providing new insights into progressive MS that once again place the immune system at center stage. This article reviews the main mechanisms of MS immunopathogenesis, and the benefits, risks and challenges of immunomodulatory treatments for the disease.
Collapse
Affiliation(s)
- Calliope A Dendrou
- Nuffield Department of Medicine, The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Lars Fugger
- Oxford Centre for Neuroinflammation, Nuffield Department of Clinical Neurosciences, Division of Clinical Neurology and MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK.
| |
Collapse
|
57
|
Benito-León J, Laurence M. The Role of Fungi in the Etiology of Multiple Sclerosis. Front Neurol 2017; 8:535. [PMID: 29085329 PMCID: PMC5650687 DOI: 10.3389/fneur.2017.00535] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 09/25/2017] [Indexed: 01/07/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disorder of the central nervous system. Infectious triggers of MS are being actively investigated. Substantial evidence supports the involvement of the Epstein-Barr virus (EBV), though other viruses, bacteria, protists, and fungi are also being considered. Many links between fungi and diseases involving chronic inflammation have been found recently. Evidence linking MS and fungi is reviewed here. The HLA-DRB1*15 allele group is the most important genetic risk factor of MS, and is a risk factor in several other conditions linked to fungal infections. Many biomarkers of MS are consistent with fungal infections, such as IL-17, chitotriosidase, and antibodies against fungi. Dimethyl fumarate (DMF), first used as an industrial fungicide, was recently repurposed to reduce MS symptoms. Its mechanisms of action in MS have not been firmly established. The low risk of MS during childhood and its moderate association with herpes simplex virus type 2 suggest genital exposure to microbes (including fungi) should be investigated as a possible trigger. Molecular and epidemiological evidence support a role for infections such as EBV in MS. Though fungal infections have not been widely studied in MS, many lines of evidence are consistent with a fungal etiology. Future microbiome and serological studies should consider fungi as a possible risk factor for MS, and future clinical studies should consider the effect of fungicides other than DMF on MS symptoms.
Collapse
Affiliation(s)
- Julián Benito-León
- Department of Neurology, University Hospital “12 de Octubre”, Madrid, Spain
- Department of Medicine, Faculty of Medicine, Complutense University, Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | | |
Collapse
|
58
|
Ren MW, Du Y, Ren S, Tang CY, He JF. Epstein-Barr virus-encoded small RNAs in idiopathic orbital inflammatory pseudotumor tissues: a comparative case series. Int J Ophthalmol 2017; 10:1268-1272. [PMID: 28861354 DOI: 10.18240/ijo.2017.08.14] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Accepted: 05/24/2017] [Indexed: 11/23/2022] Open
Abstract
AIM To investigate the positive rate and types of cells that express Epstein-Barr virus-encoded small RNAs (EBERs) and to determine the distribution of EBER-expressing cells in idiopathic orbital inflammatory pseudotumor (IOIP) tissues. METHODS We retrospectively examined 40 archived paraffin specimens from two teaching hospitals in Southern China between January 2007 and January 2015 that were pathologically determined to exhibit IOIP. Eleven concurrent paraffin specimens of thyroid-associated ophthalmopathy (TAO) composed the control group. In situ hybridization was performed to detect EBERs. Immunohistochemistry was employed to detect CD3, CD20, Vimentin, and smooth muscle actin (SMA), and the positive rate, types of positive cells, and distribution and location of EBERs were evaluated. RESULTS The positive expression rate of EBERs was 47.5% (19/40) in the IOIP group, which was significantly higher than that in the TAO group [0 (0/11), P=0.011]. In the IOIP group, the lymphocyte infiltrative subtype, fibrotic subtype, and mixed subtype exhibited EBER-positive rates of 57.1% (12/21), 12.5% (1/8), and 54.5% (6/11), respectively, and no significant differences were found between these subtypes (P=0.085). Positive signals of EBERs were mainly present in medium-small lymphocytes between or around follicles and in the nuclei of activated immunoblasts (14/19). CONCLUSION The positive rate, types, and distribution of EBER-expressing cells in IOIP have been documented. These findings are conducive for a better understanding of the underlying mechanisms of Epstein-Barr virus infection in IOIP pathogenesis.
Collapse
Affiliation(s)
- Min-Wei Ren
- Department of Ophthalmology, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China.,Department of Ophthalmology, Liuzhou People's Hospital, Liuzhou 545006, Guangxi Zhuang Autonomous Region, China
| | - Yi Du
- Department of Ophthalmology, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Shan Ren
- Department of Ophthalmology, Liuzhou People's Hospital, Liuzhou 545006, Guangxi Zhuang Autonomous Region, China
| | - Cheng-Ye Tang
- Department of Ophthalmology, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Jian-Feng He
- Department of Ophthalmology, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
59
|
Burnard S, Lechner-Scott J, Scott RJ. EBV and MS: Major cause, minor contribution or red-herring? Mult Scler Relat Disord 2017; 16:24-30. [DOI: 10.1016/j.msard.2017.06.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 06/05/2017] [Accepted: 06/09/2017] [Indexed: 10/19/2022]
|
60
|
Laurence M, Benito-León J. Epstein–Barr virus and multiple sclerosis: Updating Pender's hypothesis. Mult Scler Relat Disord 2017; 16:8-14. [DOI: 10.1016/j.msard.2017.05.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 05/14/2017] [Accepted: 05/26/2017] [Indexed: 10/19/2022]
|
61
|
't Hart BA, Dunham J, Faber BW, Laman JD, van Horssen J, Bauer J, Kap YS. A B Cell-Driven Autoimmune Pathway Leading to Pathological Hallmarks of Progressive Multiple Sclerosis in the Marmoset Experimental Autoimmune Encephalomyelitis Model. Front Immunol 2017; 8:804. [PMID: 28744286 PMCID: PMC5504154 DOI: 10.3389/fimmu.2017.00804] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 06/26/2017] [Indexed: 12/20/2022] Open
Abstract
The absence of pathological hallmarks of progressive multiple sclerosis (MS) in commonly used rodent models of experimental autoimmune encephalomyelitis (EAE) hinders the development of adequate treatments for progressive disease. Work reviewed here shows that such hallmarks are present in the EAE model in marmoset monkeys (Callithrix jacchus). The minimal requirement for induction of progressive MS pathology is immunization with a synthetic peptide representing residues 34–56 from human myelin oligodendrocyte glycoprotein (MOG) formulated with a mineral oil [incomplete Freund’s adjuvant (IFA)]. Pathological aspects include demyelination of cortical gray matter with microglia activation, oxidative stress, and redistribution of iron. When the peptide is formulated in complete Freund’s adjuvant, which contains mycobacteria that relay strong activation signals to myeloid cells, oxidative damage pathways are strongly boosted leading to more intensive pathology. The proven absence of immune potentiating danger signals in the MOG34–56/IFA formulation implies that a narrow population of antigen-experienced T cells present in the monkey’s immune repertoire is activated. This novel pathway involves the interplay of lymphocryptovirus-infected B cells with MHC class Ib/Caja-E restricted CD8+ CD56+ cytotoxic T lymphocytes.
Collapse
Affiliation(s)
- Bert A 't Hart
- Department of Immunobiology, Biomedical Primate Research Center, Rijswijk, Netherlands.,Department of Neuroscience, University of Groningen, University Medical Center, Groningen, Netherlands
| | - Jordon Dunham
- Department of Immunobiology, Biomedical Primate Research Center, Rijswijk, Netherlands.,Department of Neuroscience, University of Groningen, University Medical Center, Groningen, Netherlands
| | - Bart W Faber
- Department of Parasitology, Biomedical Primate Research Center, Rijswijk, Netherlands
| | - Jon D Laman
- Department of Neuroscience, University of Groningen, University Medical Center, Groningen, Netherlands.,MS Center Noord-Nederland, Groningen, Netherlands
| | - Jack van Horssen
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, Netherlands
| | - Jan Bauer
- Department of Neuroimmunology, Brain Research Institute, Medical University Vienna, Vienna, Austria
| | - Yolanda S Kap
- Department of Immunobiology, Biomedical Primate Research Center, Rijswijk, Netherlands
| |
Collapse
|
62
|
Morandi E, Jagessar SA, 't Hart BA, Gran B. EBV Infection Empowers Human B Cells for Autoimmunity: Role of Autophagy and Relevance to Multiple Sclerosis. THE JOURNAL OF IMMUNOLOGY 2017; 199:435-448. [PMID: 28592428 DOI: 10.4049/jimmunol.1700178] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 05/05/2017] [Indexed: 11/19/2022]
Abstract
The efficacy of B cell depletion therapy in multiple sclerosis indicates their central pathogenic role in disease pathogenesis. The B lymphotropic EBV is a major risk factor in multiple sclerosis, via as yet unclear mechanisms. We reported in a nonhuman primate experimental autoimmune encephalomyelitis model that an EBV-related lymphocryptovirus enables B cells to protect a proteolysis-sensitive immunodominant myelin oligodendrocyte glycoprotein (MOG) epitope (residues 40-48) against destructive processing. This facilitates its cross-presentation to autoaggressive cytotoxic MHC-E-restricted CD8+CD56+ T cells. The present study extends these observations to intact human B cells and identifies a key role of autophagy. EBV infection upregulated APC-related markers on B cells and activated the cross-presentation machinery. Although human MOG protein was degraded less in EBV-infected than in uninfected B cells, induction of cathepsin G activity by EBV led to total degradation of the immunodominant peptides MOG35-55 and MOG1-20 Inhibition of cathepsin G or citrullination of the arginine residue within an LC3-interacting region motif of immunodominant MOG peptides abrogated their degradation. Internalized MOG colocalized with autophagosomes, which can protect from destructive processing. In conclusion, EBV infection switches MOG processing in B cells from destructive to productive and facilitates cross-presentation of disease-relevant epitopes to CD8+ T cells.
Collapse
Affiliation(s)
- Elena Morandi
- Division of Clinical Neuroscience, University of Nottingham School of Medicine, Nottingham NG7 2UH, United Kingdom
| | - S Anwar Jagessar
- Department of Immunobiology, Biomedical Primate Research Centre, Rijswijk 2288, the Netherlands
| | - Bert A 't Hart
- Department of Immunobiology, Biomedical Primate Research Centre, Rijswijk 2288, the Netherlands.,Department of Neuroscience, University Medical Center, University of Groningen, Groningen 9700, the Netherlands; and
| | - Bruno Gran
- Division of Clinical Neuroscience, University of Nottingham School of Medicine, Nottingham NG7 2UH, United Kingdom; .,Department of Neurology, Nottingham University Hospitals NHS Trust, Nottingham NG7 2UH, United Kingdom
| |
Collapse
|
63
|
Karakayali B, Yilmaz S, Çakir D, Günes PG, Güven S, Islek I. Henoch-Schonlein purpura associated with primary active Epstein-Barr virus infection: a case report. Pan Afr Med J 2017; 27:29. [PMID: 28761605 PMCID: PMC5516656 DOI: 10.11604/pamj.2017.27.29.10481] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 04/28/2017] [Indexed: 11/11/2022] Open
Abstract
Henoch-Schönlein purpura (HSP) is the most common form of childhood vasculitis. Various viral and bacterial infections, drugs, vaccines, food allergy and even insect bites have been considered as triggering factors in pathogenesis of HSP. Epstein-Barr virus (EBV) infection, which is associated with HSP, have been rarely reported. Herein we present HSP patient possibly caused by EBV infection. A 8-year old boy was admitted to our department with fever, rashes on legs and arms and intermittent mild abdominal pain. Multiple purpuric rashes were on his extremities, abdomen and buttock. Laboratory investigations revealed that monospot test was positive, EBV serology tests; Anti-EA-D Ig G: 3+, Anti-VCA gp125 Ig G: 3+, Anti-VCA p19 Ig M: 2+, Anti EBNA-1 Ig M: negative, Anti EBNA-1 Ig M: negative, Anti EBNA-1 Ig G: negative. The patient was interpreted as the primary active acute EBV infection. A skin biopsy showed leucocytoclastic vasculitis. The other viral and bacterial investigations were negative. The patient was diagnosed as HSP vasculitis according to EULAR criteria and treated with intravenous hydration and ibuprofen. He was discharged after 15 days with normal laboratory findings and physical examination. We think that EBV infection may be stimulant factor for autoimmune reactions and may cause HSP vasculitis. Hence, it may be useful to investigate the EBV infection in etiology of HSP cases.
Collapse
Affiliation(s)
- Burcu Karakayali
- University of Health Sciences, Umraniye Research and Training Hospital, Department of Pediatrics, Istanbul, Turkey
| | - Sila Yilmaz
- University of Health Sciences, Umraniye Research and Training Hospital, Department of Pediatrics, Istanbul, Turkey
| | - Deniz Çakir
- University of Health Sciences, Umraniye Research and Training Hospital, Department of Pediatrics, Istanbul, Turkey
| | - Pembe Gül Günes
- Haydarpasa Numune Research and Training Hospital, Department of Pathology, Istanbul, Turkey
| | - Sirin Güven
- University of Health Sciences, Umraniye Research and Training Hospital, Department of Pediatrics, Istanbul, Turkey
| | - Ismail Islek
- University of Health Sciences, Umraniye Research and Training Hospital, Department of Pediatrics, Istanbul, Turkey
| |
Collapse
|
64
|
Oh SH, You CR, Kim EO, Bae SH, Choi JY, Yoon SK, Choi SW. A Case of Ulcerative Colitis Following Acute Hepatitis Induced by Epstein-Barr Virus Infection. THE KOREAN JOURNAL OF GASTROENTEROLOGY 2017; 68:104-8. [PMID: 27554218 DOI: 10.4166/kjg.2016.68.2.104] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Epstein-Barr virus (EBV) infection varies in its clinical manifestations and severity. EBV can be a causative agent of hepatitis and may have a role in the pathogenesis of chronic autoimmune diseases including inflammatory bowel disease. A 24-year-old woman was admitted to our hospital, presenting with fever and elevated liver enzymes. She was diagnosed with acute hepatitis and EBV infection according to serologic tests and liver biopsy. Within two months, she was re-admitted to our hospital, presenting with hematochezia and lower abdominal pain. She was diagnosed with ulcerative colitis. In situ hybridization for EBV was positive in initial liver biopsy and colon biopsy. Here we report an unusual case of acute EBV hepatitis followed at a short interval by ulcerative colitis.
Collapse
Affiliation(s)
- Seung Hyun Oh
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Chan Ran You
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Eun Ok Kim
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Si Hyun Bae
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jong Young Choi
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seung Kew Yoon
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sang Wook Choi
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
65
|
Eriksen W. The spread of EBV to ectopic lymphoid aggregates may be the final common pathway in the pathogenesis of ME/CFS. Med Hypotheses 2017; 102:8-15. [PMID: 28478837 DOI: 10.1016/j.mehy.2017.02.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 02/02/2017] [Accepted: 02/26/2017] [Indexed: 12/22/2022]
Abstract
According to the hypothesis presented here, myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) develops over 3 steps: Step 1 is characterized by the aggregation of lymphoid cells in dorsal root ganglia or other nervous structures. The cause of this formation of ectopic lymphoid aggregates may be an acute infection, asymptomatic reactivations of a common neurotropic virus, exposure to a neurotoxin, or physical injury to peripheral nerves. In step 2, Epstein-Barr virus (EBV)-infected lymphocytes or monocytes bring EBV from the circulation to one or several of these lymphoid aggregates, whereupon cell-to-cell transmission of EBV and proliferation of latently EBV-infected lymphocytes lead to the presence of many EBV-infected cells in the lymphoid aggregates. The EBV-infected cells in the aggregates ignite an inflammation in the surrounding nervous tissue. This local inflammation elicits, in turn, a wave of glial cell activation that spreads from the EBV-infected area to parts of the nervous system that are not EBV-infected, disturbing the neuron-glial interaction in both the peripheral - and central nervous system. In step 3, immune cell exhaustion contributes to a consolidation of the pathological processes. There might be a cure: Infusions of autologous EBV-specific T-lymphocytes can perhaps remove the EBV-infected cells from the nervous system.
Collapse
Affiliation(s)
- Willy Eriksen
- Domain for Mental and Physical Health, Norwegian Institute of Public Health, Box 4404 Nydalen, 0403 Oslo, Norway.
| |
Collapse
|
66
|
Pender MP, Csurhes PA, Burrows JM, Burrows SR. Defective T-cell control of Epstein-Barr virus infection in multiple sclerosis. Clin Transl Immunology 2017; 6:e126. [PMID: 28197337 PMCID: PMC5292561 DOI: 10.1038/cti.2016.87] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 12/02/2016] [Accepted: 12/02/2016] [Indexed: 02/07/2023] Open
Abstract
Mounting evidence indicates that infection with Epstein–Barr virus (EBV) has a
major role in the pathogenesis of multiple sclerosis (MS). Defective elimination of
EBV-infected B cells by CD8+ T cells might cause MS by allowing
EBV-infected autoreactive B cells to accumulate in the brain. Here we undertake a
comprehensive analysis of the T-cell response to EBV in MS, using flow cytometry and
intracellular IFN-γ staining to measure T-cell responses to EBV-infected
autologous lymphoblastoid cell lines and pools of human leukocyte antigen
(HLA)-class-I-restricted peptides from EBV lytic or latent proteins and
cytomegalovirus (CMV), in 95 patients and 56 EBV-seropositive healthy subjects. In 20
HLA-A2+ healthy subjects and 20 HLA-A2+ patients
we also analysed CD8+ T cells specific for individual peptides,
measured by binding to HLA-peptide complexes and production of IFN-γ,
TNF-α and IL-2. We found a decreased CD8+ T-cell response to
EBV lytic, but not CMV lytic, antigens at the onset of MS and at all subsequent
disease stages. CD8+ T cells directed against EBV latent antigens
were increased but had reduced cytokine polyfunctionality indicating T-cell
exhaustion. During attacks the EBV-specific CD4+ and
CD8+ T-cell populations expanded, with increased functionality of
latent-specific CD8+ T cells. With increasing disease duration,
EBV-specific CD4+ and CD8+ T cells progressively
declined, consistent with T-cell exhaustion. The anti-EBNA1 IgG titre correlated
inversely with the EBV-specific CD8+ T-cell frequency. We postulate
that defective CD8+ T-cell control of EBV reactivation leads to an
expanded population of latently infected cells, including autoreactive B cells.
Collapse
Affiliation(s)
- Michael P Pender
- School of Medicine, The University of Queensland, Brisbane, Queensland, Australia; Department of Neurology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia; Cellular ImmunoIogy Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Peter A Csurhes
- School of Medicine, The University of Queensland, Brisbane, Queensland, Australia; Cellular ImmunoIogy Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia; The University of Queensland Centre for Clinical Research, Brisbane, Queensland, Australia
| | - Jacqueline M Burrows
- Cellular ImmunoIogy Laboratory, QIMR Berghofer Medical Research Institute , Brisbane, Queensland, Australia
| | - Scott R Burrows
- School of Medicine, The University of Queensland, Brisbane, Queensland, Australia; Cellular ImmunoIogy Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
67
|
Jones GW, Hill DG, Jones SA. Understanding Immune Cells in Tertiary Lymphoid Organ Development: It Is All Starting to Come Together. Front Immunol 2016; 7:401. [PMID: 27752256 PMCID: PMC5046062 DOI: 10.3389/fimmu.2016.00401] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 09/21/2016] [Indexed: 01/28/2023] Open
Abstract
Tertiary lymphoid organs (TLOs) are frequently observed in tissues affected by non-resolving inflammation as a result of infection, autoimmunity, cancer, and allograft rejection. These highly ordered structures resemble the cellular composition of lymphoid follicles typically associated with the spleen and lymph node compartments. Although TLOs within tissues show varying degrees of organization, they frequently display evidence of segregated T and B cell zones, follicular dendritic cell networks, a supporting stromal reticulum, and high endothelial venules. In this respect, they mimic the activities of germinal centers and contribute to the local control of adaptive immune responses. Studies in various disease settings have described how these structures contribute to either beneficial or deleterious outcomes. While the development and architectural organization of TLOs within inflamed tissues requires homeostatic chemokines, lymphoid and inflammatory cytokines, and adhesion molecules, our understanding of the cells responsible for triggering these events is still evolving. Over the past 10–15 years, novel immune cell subsets have been discovered that have more recently been implicated in the control of TLO development and function. In this review, we will discuss the contribution of these cell types and consider the potential to develop new therapeutic strategies that target TLOs.
Collapse
Affiliation(s)
- Gareth W Jones
- Division of Infection and Immunity, Systems Immunity URI, The School of Medicine, Cardiff University , Cardiff , UK
| | - David G Hill
- Division of Infection and Immunity, Systems Immunity URI, The School of Medicine, Cardiff University , Cardiff , UK
| | - Simon A Jones
- Division of Infection and Immunity, Systems Immunity URI, The School of Medicine, Cardiff University , Cardiff , UK
| |
Collapse
|
68
|
Farina A, Farina GA. Fresh Insights into Disease Etiology and the Role of Microbial Pathogens. Curr Rheumatol Rep 2016; 18:1. [PMID: 26700911 DOI: 10.1007/s11926-015-0552-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Pathogens have been implicated in the initiation and/or promotion of systemic sclerosis (scleroderma, SSc); however, no evidence was found to substantiate the direct contribution to this disease in past years. Recently, significant advances have been made in understanding the role of the innate immune system in SSc pathogenesis, supporting the idea that pathogens might interact with host innate immune-regulatory responses in SSc. In light of these findings, we review the studies that identified the presence of pathogens in SSc, along with studies on pathogens implicated in driving the innate immune dysregulation in SSc. The goal of this review is to illustrate how these pathogens, specifically viruses, may play important role both as triggers of the innate immune system, and critical players in the development of SSc disease.
Collapse
Affiliation(s)
- Antonella Farina
- Institute Pasteur-Fondazione Cenci Bolognetti, Department of Experimental Medicine, "Sapienza", University of Rome, Rome, Italy.
| | - G Alessandra Farina
- Arthritis Center, Department of Rheumatology, Boston University, 72 East Concord Street, E501, Boston, MA, 02118, USA.
| |
Collapse
|
69
|
Maghzi H, Ataei B, Khorvash F, Yaran M, Maghzi AH. Association Between Acute Infectious Mononucleosis and Vitamin D Deficiency. Viral Immunol 2016; 29:398-400. [PMID: 27505106 DOI: 10.1089/vim.2016.0038] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Epstein-Barr virus and vitamin D both have been implicated in the pathogenesis of autoimmune diseases, especially multiple sclerosis (MS). Vitamin D influences both innate and adaptive immune responses and has been linked to increased susceptibility to other viral infections such as influenza. Here we aimed to examine the association between vitamin D and acute infectious mononucleosis (IM).This study is a case-control study that was conducted on IM patients and a control group of healthy individuals at infectious disease clinics of Isfahan University of Medical Sciences. Patients were recruited from January to December 2014. The viral capsid antigen (VCA) IgM titer and vitamin D levels were measured at the time of acute infection in IM patients. We also measured vitamin D levels in healthy controls recruited during the same period of time. A total number of 60 IM patients with the mean age of 23.26 ± 7.59 and a healthy control group with the mean age of 25.13 ± 6.72 were enrolled. In the IM patients, there was no significant association between 25(OH) D3 levels and VCA IgM titers (r = 0.190, p = 0.146). Mean 25(OH) D3 levels in IM patients were significantly lower than in the control group (15.61 ± 9.72 vs. 21.41 ± 12.64, p = 0.006). Our findings showed significantly lower vitamin D levels in IM patients at the time of infection than in the control group, providing some evidence that the two major risk factors for autoimmune diseases (e.g., MS) might not be independent risk factors.
Collapse
Affiliation(s)
- Helia Maghzi
- 1 Multiple Sclerosis Research Committee, Isfahan University of Medical Sciences , Isfahan, Iran
| | - Behrooz Ataei
- 2 Infectious Diseases and Tropical Medicine Research Center, Isfahan University of Medical Sciences , Isfahan, Iran
| | - Farzin Khorvash
- 3 Nosocomial Infection Research Center, Isfahan University of Medical Sciences , Isfahan, Iran
| | - Majid Yaran
- 4 Acquired Immunodeficiency Research Center, Isfahan University of Medical Sciences , Isfahan, Iran
| | - Amir-Hadi Maghzi
- 5 Department of Neurology, Cedars-Sinai Medical Center , Los Angeles, California
| |
Collapse
|
70
|
Abstract
BACKGROUND/OBJECTIVES Multiple sclerosis (MS) is an inflammatory disorder of the central nervous system. Many diseases are associated with Epstein-Barr virus (EBV) infection, such as infectious mononucleosis and many types of malignancies, and it is thought to be related to some diseases of autoimmune origin, such as rheumatoid arthritis, systemic lupus erythematosis, and others. The present study aimed to assess EBV in patients with MS. PATIENTS AND METHODS This case-control study was conducted from October 2012 to September 2013 on 75 MS patients and non-MS controls. Both were tested quantitatively for immunoglobulin G (IgG) antibodies against Epstein-Barr nuclear antigen-1 (EBNA1) and viral capsid antigen (VCA) using the enzyme linked immunosorbent assay technique. RESULTS Seventy MS patients (93.3%) were positive for EBNA1 IgG compared with 68 controls (90.7%). In MS patients, the mean EBNA1 IgG serum level was 310.91 (±131.05) U/ml; meanwhile, among controls the mean serum EBNA IgG level was 177.81 (±104.98) U/ml.All patients with MS were positive for VCA IgG, whereas only 60 (80.0%) controls were positive. In the MS group, the VCA IgG mean level was 302.19 (±152.11) U/ml compared with 167.94 (±111.79) U/ml in controls. The differences in the serum levels of both markers between the two groups were statistically significant (P<0.001). CONCLUSION AND RECOMMENDATIONS EBV proved to have a unique immunological pattern in MS patients when compared with non-MS controls. Further studies for more confirmation of the relation between EBV and MS on a large scale are recommended.
Collapse
|
71
|
Rezvani K, Champlin RE. Epstein-Barr Virus and B Cells in the Pathogenesis of Graft-Versus-Host Disease After Allogeneic Hematopoietic Stem-Cell Transplantation. J Clin Oncol 2016; 34:2201-2. [DOI: 10.1200/jco.2016.66.6099] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
|
72
|
Bhise V, Dhib-Jalbut S. Further understanding of the immunopathology of multiple sclerosis: impact on future treatments. Expert Rev Clin Immunol 2016; 12:1069-89. [PMID: 27191526 DOI: 10.1080/1744666x.2016.1191351] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The understanding of the immunopathogenesis of multiple sclerosis (MS) has expanded with more research into T-cell subtypes, cytokine contributors, B-cell participation, mitochondrial dysfunction, and more. Treatment options have rapidly expanded with three relatively recent oral therapy alternatives entering the arena. AREAS COVERED In the following review, we discuss current mechanisms of immune dysregulation in MS, how they relate to current treatments, and the impact these findings will have on the future of therapy. Expert commentary: The efficacy of these medications and understanding their mechanisms of actions validates the immunopathogenic mechanisms thought to underlie MS. Further research has exposed new targets, while new promising therapies have shed light on new aspects into the pathophysiology of MS.
Collapse
Affiliation(s)
- Vikram Bhise
- a Rutgers Biomedical and Health Sciences - Departments of Pediatrics , Robert Wood Johnson Medical School , New Brunswick , NJ , USA
| | - Suhayl Dhib-Jalbut
- b Rutgers Biomedical and Health Sciences - Departments of Neurology , Robert Wood Johnson Medical School , New Brunswick , NJ , USA
| |
Collapse
|
73
|
Abstract
Primary Sjögren's syndrome, a chronic inflammatory process, is among the most commonly occurring rheumatologic diseases. The clinical hallmark of this disease is exocrine gland dysfunction, resulting predominately in dry eyes and dry mouth. However, the disease often extends beyond the exocrine glands to seriously affect other organs systems, such as the lungs, kidneys, and nervous system. Moreover, patients with primary Sjögren's syndrome develop non-Hodgkin's B cell lymphoma at a substantially higher rate than the general population. New research has improved our understanding of disease mechanisms, with notable advances in our knowledge about the genetic susceptibility of disease, the molecular details of the chronic inflammatory response in the salivary glands, and the complex role of the type 1 interferon pathway. The pipeline of drugs under development for the treatment of primary Sjögren's syndrome is enriched with novel biologics and small molecular entities targeting the pathogenic process. Herein, we summarize the latest advances in elucidating the pathogenesis of primary Sjögren's syndrome and highlight new drugs in clinical development aiming to reverse the glandular dysfunction and favorably impact the systemic features of this disease.
Collapse
Affiliation(s)
- Nicholas Holdgate
- Department of Medicine, Division of Rheumatology and Immunology, School of Medicine, Duke University, Durham, NC, 27710, USA
| | - E. Wiliam St.Clair
- Department of Medicine, Division of Rheumatology and Immunology, School of Medicine, Duke University, Durham, NC, 27710, USA
| |
Collapse
|
74
|
Elansary M, El Haddad HE, Eldin UAAS, Hamdy A, Sherif MM. Seroprevalence and real-time PCR study of Epstein—Barr virus and the value of screening in pretransplant patients. THE EGYPTIAN JOURNAL OF INTERNAL MEDICINE 2016. [DOI: 10.4103/1110-7782.182947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
75
|
Hartwell BL, Martinez-Becerra FJ, Chen J, Shinogle H, Sarnowski M, Moore DS, Berkland C. Antigen-Specific Binding of Multivalent Soluble Antigen Arrays Induces Receptor Clustering and Impedes B Cell Receptor Mediated Signaling. Biomacromolecules 2016; 17:710-22. [PMID: 26771518 DOI: 10.1021/acs.biomac.5b01097] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A pressing need exists for autoimmune disease therapies that act in an antigen-specific manner while avoiding global immunosuppression. Multivalent soluble antigen arrays (SAgAPLP:LABL), designed to induce tolerance to a specific multiple sclerosis autoantigen, consist of a flexible hyaluronic acid (HA) polymer backbone cografted with multiple copies of autoantigen peptide (PLP) and cell adhesion inhibitor peptide (LABL). Previous in vivo studies revealed copresentation of both signals on HA was necessary for therapeutic efficacy. To elucidate therapeutic cellular mechanisms, in vitro studies were performed in a model B cell system to evaluate binding and specificity. Compared to HA and HA arrays containing only grafted PLP or LABL, SAgAPLP:LABL displaying both PLP and LABL exhibited greatly enhanced B cell binding. Furthermore, the binding avidity of SAgAPLP:LABL was primarily driven by the PLP antigen, determined via flow cytometry competitive dissociation studies. Fluorescence microscopy showed SAgAPLP:LABL induced mature receptor clustering that was faster than other HA arrays with only one type of grafted peptide. SAgAPLP:LABL molecules also reduced and inhibited IgM-stimulated signaling as discerned by a calcium flux assay. The molecular mechanisms of enhanced antigen-specific binding, mature receptor clustering, and dampened signaling observed in B cells may contribute to SAgAPLP:LABL therapeutic efficacy.
Collapse
Affiliation(s)
- Brittany L Hartwell
- Bioengineering Graduate Program, University of Kansas , 1520 West 15th Street, Lawrence, Kansas 66045, United States
| | - Francisco J Martinez-Becerra
- Immunology Core Laboratory of the Kansas Vaccine Institute, University of Kansas 2030 Becker Drive, Lawrence, Kansas 66047, United States.,Department of Pharmaceutical Chemistry, University of Kansas 2095 Constant Avenue, Lawrence, Kansas 66047, United States
| | - Jun Chen
- Department of Pharmaceutical Chemistry, University of Kansas 2095 Constant Avenue, Lawrence, Kansas 66047, United States
| | - Heather Shinogle
- Microscopy and Analytical Imaging Laboratory, University of Kansas 1200 Sunnyside Avenue, Lawrence, Kansas 66045, United States
| | - Michelle Sarnowski
- Department of Chemical and Petroleum Engineering, University of Kansas 1530 West 15th Street, Lawrence, Kansas 66045, United States
| | - David S Moore
- Microscopy and Analytical Imaging Laboratory, University of Kansas 1200 Sunnyside Avenue, Lawrence, Kansas 66045, United States
| | - Cory Berkland
- Bioengineering Graduate Program, University of Kansas , 1520 West 15th Street, Lawrence, Kansas 66045, United States.,Department of Pharmaceutical Chemistry, University of Kansas 2095 Constant Avenue, Lawrence, Kansas 66047, United States.,Department of Chemical and Petroleum Engineering, University of Kansas 1530 West 15th Street, Lawrence, Kansas 66045, United States
| |
Collapse
|
76
|
Rolf L, Muris AH, Hupperts R, Damoiseaux J. Illuminating vitamin D effects on B cells--the multiple sclerosis perspective. Immunology 2016; 147:275-84. [PMID: 26714674 DOI: 10.1111/imm.12572] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 12/17/2015] [Accepted: 12/18/2015] [Indexed: 12/11/2022] Open
Abstract
Vitamin D is associated with many immune-mediated disorders. In multiple sclerosis (MS) a poor vitamin D status is a major environmental factor associated with disease incidence and severity. The inflammation in MS is primarily T-cell-mediated, but increasing evidence points to an important role for B cells. This has paved the way for investigating vitamin D effects on B cells. In this review we elaborate on vitamin D interactions with antibody production, T-cell-stimulating capacity and regulatory B cells. Although in vitro plasma cell generation and expression of co-stimulatory molecules are inhibited and the function of regulatory B cells is promoted, this is not supported by in vivo data. We speculate that differences might be explained by the B-cell-Epstein-Barr virus interaction in MS, the exquisite role of germinal centres in B-cell biology, and/or in vivo interactions with other hormones and vitamins that interfere with the vitamin D pathways. Further research is warranted to illuminate this tube-versus-body paradox.
Collapse
Affiliation(s)
- Linda Rolf
- School for Mental Health and Neuroscience, Maastricht University Medical Centre, Maastricht, The Netherlands.,Academic MS Centre Limburg, Zuyderland Medisch Centrum, Sittard, The Netherlands
| | - Anne-Hilde Muris
- School for Mental Health and Neuroscience, Maastricht University Medical Centre, Maastricht, The Netherlands.,Academic MS Centre Limburg, Zuyderland Medisch Centrum, Sittard, The Netherlands
| | - Raymond Hupperts
- School for Mental Health and Neuroscience, Maastricht University Medical Centre, Maastricht, The Netherlands.,Academic MS Centre Limburg, Zuyderland Medisch Centrum, Sittard, The Netherlands
| | - Jan Damoiseaux
- Central Diagnostic Laboratory, Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|
77
|
Close Encounters of the First Kind: Innate Sensors and Multiple Sclerosis. Mol Neurobiol 2016; 54:101-114. [PMID: 26732593 DOI: 10.1007/s12035-015-9665-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 12/17/2015] [Indexed: 10/22/2022]
Abstract
Although autoimmune diseases by definition imply adaptive immune system pathologies, growing evidence points to the relevance of innate receptors in modulating the initiation and progression of the autoreactive response. Multiple sclerosis (MS) is a chronic autoimmune disease characterised by central nervous system (CNS) demyelination, inflammation and axonal damage, in which the role of several pathogens such as herpes viruses have long been described as potential triggers. Encounters of these pathogens with altered innate receptors in susceptible individuals might drive pathological autoreactivity and inflammation, overcoming tolerance and causing subsequent CNS damage. In particular, functional and genetic studies reveal that Toll-like receptor (TLR) 2 and the Nod-like receptor (NLR) P3 could be involved in MS pathogenesis, whereas TLR3, the triggering receptor expressed on myeloid cells (TREM)-2 and the C-type lectin receptors (CLRs) MBL and MASP-3 would have a putative protective role. A better understanding of these interactions will provide important insights into the aetiopathogenesis of MS and could help design potential targets for novel therapies.
Collapse
|
78
|
Correale J, Gaitán MI. Multiple sclerosis and environmental factors: the role of vitamin D, parasites, and Epstein-Barr virus infection. Acta Neurol Scand 2016; 132:46-55. [PMID: 26046559 DOI: 10.1111/ane.12431] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/10/2015] [Indexed: 01/08/2023]
Abstract
Pathogenic mechanisms underlying multiple sclerosis development have yet to be clearly identified, but considerable evidence indicates that autoimmunity plays an important role in the etiology of the disease. It is generally accepted that autoimmune diseases like MS arise from complex interactions between genetic susceptibility and environmental factors. Although environmental factors unequivocally influencing MS development have yet to be established, accumulating evidence singles out several candidates, including sunlight-UV exposure or vitamin D deficiency, viral infections, hygiene, and cigarette smoking. Vitamin D deficiency has been associated with different autoimmune diseases. Several investigations indicate 125 (OH)2 vitamin D plays a critical role in shaping T-cell response and inducing T cells with immunosuppressive properties. Likewise, helminth infections represent another potential environmental factor exerting immunomodulatory properties. Both epidemiological and experimental data provide evidence to support autoimmune down-regulation secondary to parasite infections in patients with MS, through regulatory T- and B-cell action, with effects extending beyond simple response to an infectious agent. Finally, different epidemiological studies have demonstrated that Epstein-Barr virus infection confers added risk of developing MS. Proposed mechanisms responsible for this association include activation and expansion of self-reactive T and B cells, lower threshold for self-tolerance breakdown, and enhanced autoreactive B-cell survival, all to be discussed in this review. Understanding environmental factors influencing propensity to MS will lead to new and more effective approaches to prevent and treat the disease.
Collapse
Affiliation(s)
- J. Correale
- Department of Neurology; Institute for Neurological Research Dr. Raúl Carrea; FLENI; Buenos Aires Argentina
| | - M. I. Gaitán
- Department of Neurology; Institute for Neurological Research Dr. Raúl Carrea; FLENI; Buenos Aires Argentina
| |
Collapse
|
79
|
Xiao D, Ye X, Zhang N, Ou M, Guo C, Zhang B, Liu Y, Wang M, Yang G, Jing C. A meta-analysis of interaction between Epstein-Barr virus and HLA-DRB1*1501 on risk of multiple sclerosis. Sci Rep 2015; 5:18083. [PMID: 26656273 PMCID: PMC4676020 DOI: 10.1038/srep18083] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 11/11/2015] [Indexed: 11/09/2022] Open
Abstract
Infection with Epstein-Barr virus (EBV) and HLA-DRB1*1501-positivity is a risk factor for multiple sclerosis (MS), but whether an interaction between these two factors causes MS is unclear. We therefore conducted a meta-analysis on the effect of the interaction between HLA-DRB1*1501 and EBV infection on MS. Searches of PubMed, Web of Science, China National Knowledge Infrastructure (CNKI), and the Wanfan databases through February 2015 yielded 5 studies that met the criteria for inclusion in the meta-analysis. EBV infection and HLA-DRB1*1501-positivity were dichotomized. The additive (S) and multiplicative interaction indexes (OR) between EBV infection and HLA-DRB1*1501 and their 95% confidence intervals (95%CI) were calculated for each study and then combined in a meta-analysis. EBV infection was significantly associated with MS (OR = 2.60; 95%CI, 1.48–4.59). HLA-DRB1*1501 was associated with a significantly increased risk of MS (OR, 3.06; 95%CI, 2.30–4.08). An interaction effect between EBV infection and HLA-DRB1*1501 on MS was observed on the additive scale (S, 1.43; 95%CI, 1.05–1.95, P = 0.023), but no interaction effect was observed on the multiplicative scale (OR, 0.86, 95%CI, 0.59–1.26). This meta-analysis provides strong evidence that EBV alone, HLA-DRB1*1501 alone or their interaction is associated with an elevated risks of MS.
Collapse
Affiliation(s)
- Di Xiao
- Department of Epidemiology, School of Medicine, Jinan University, Guangzhou, China
| | - Xingguang Ye
- Department of Epidemiology, School of Medicine, Jinan University, Guangzhou, China
| | - Na Zhang
- Department of Epidemiology, School of Medicine, Jinan University, Guangzhou, China
| | - Meiling Ou
- Department of Epidemiology, School of Medicine, Jinan University, Guangzhou, China
| | - Congcong Guo
- Department of Epidemiology, School of Medicine, Jinan University, Guangzhou, China
| | - Baohuan Zhang
- Department of Epidemiology, School of Medicine, Jinan University, Guangzhou, China
| | - Yang Liu
- Department of Epidemiology, School of Medicine, Jinan University, Guangzhou, China
| | - Man Wang
- Department of Epidemiology, School of Medicine, Jinan University, Guangzhou, China
| | - Guang Yang
- Department of Parasitology, School of Medicine, Jinan University, No.601, Huangpu Avenue West, Guangzhou, Guangdong, China.,Key Laboratory of environmental exposure and health in Guangzhou, Jinan University, Guangzhou, Guangdong, China
| | - Chunxia Jing
- Department of Epidemiology, School of Medicine, Jinan University, Guangzhou, China.,Key Laboratory of environmental exposure and health in Guangzhou, Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
80
|
Márquez AC, Horwitz MS. The Role of Latently Infected B Cells in CNS Autoimmunity. Front Immunol 2015; 6:544. [PMID: 26579121 PMCID: PMC4623415 DOI: 10.3389/fimmu.2015.00544] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 10/09/2015] [Indexed: 11/16/2022] Open
Abstract
The onset of multiple sclerosis (MS) is caused by both genetic and environmental factors. Among the environmental factors, it is believed that previous infection with Epstein–Barr virus (EBV) may contribute in the development of MS. EBV has been associated with other autoimmune diseases, such as systemic lupus erythematous, and cancers like Burkitt’s lymphoma. EBV establishes a life-long latency in B cells with occasional reactivation of the virus throughout the individual’s life. The role played by B cells in MS pathology has been largely studied, yet is not clearly understood. In MS patients, Rituximab, a novel treatment that targets CD20+ B cells, has proven to have successful results in diminishing the number of relapses in remitting relapsing MS; however, the mechanism of how this drug acts has not been clearly established. In this review, we analyze the evidence of how B cells latently infected with EBV might be altering the immune system response and helping in the development of MS. We will also discuss how animal models, such as experimental autoimmune encephalomyelitis (EAE) and murine gammaherpesvirus-68 (γHV-68), can be used as powerful tools in the study of the relationship between EBV, MS, and B cells.
Collapse
Affiliation(s)
- Ana Citlali Márquez
- Department of Microbiology and Immunology, The University of British Columbia , Vancouver, BC , Canada
| | - Marc Steven Horwitz
- Department of Microbiology and Immunology, The University of British Columbia , Vancouver, BC , Canada
| |
Collapse
|
81
|
Ball RJ, Avenell A, Aucott L, Hanlon P, Vickers MA. Systematic review and meta-analysis of the sero-epidemiological association between Epstein-Barr virus and rheumatoid arthritis. Arthritis Res Ther 2015; 17:274. [PMID: 26416719 PMCID: PMC4587583 DOI: 10.1186/s13075-015-0755-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 08/12/2015] [Indexed: 12/17/2022] Open
Abstract
Introduction Infection with Epstein-Barr virus (EBV) has been suggested to contribute to the pathogenesis of autoimmune diseases, including rheumatoid arthritis (RA). We sought to determine whether prior infection with the virus occurs more frequently in patients with RA compared to controls. Methods We performed a systematic review and meta-analyses of studies that reported the prevalence of anti-EBV antibodies in the sera of cases with RA and controls by searching Medline and Embase databases from 1946 to 2014, with no language restriction. Mantel-Haenszel odds ratios for the detection of anti-EBV antibodies were calculated, and meta-analyses conducted. Quality assessments were performed using a modified version of the Newcastle-Ottawa scale. Results Twenty-three studies were included. Quality assessment found most studies reported acceptable selection criteria but poor descriptions of how cases and controls were recruited. When all studies were included, there was a statistically significant higher seroprevalence of anti-VCA IgG in patients with RA compared to controls with an odds ratio (OR) of 1.61 (95 % confidence interval (CI) 1.05–2.46, p = 0.03), which is a similar-sized summary OR to that reported for systemic lupus erythematosus (SLE). However, when studies were restricted to those reporting more plausible levels of exposure to EBV in the control groups, no significant association was apparent, OR 1.47 (95 % CI 0.88–2.46, p = 0.14). Using anti-EBNA 1 or anti-EA IgG as markers of previous infection also did not yield significant associations (OR 1.05, 95 % CI 0.68–1.61, p = 0.82; OR 2.2, 95 % CI 0.86–5.65, p = 0.10 respectively). Conclusions Overall, these findings do not demonstrate an association between EBV seroprevalence and RA and therefore do not support the hypothesis that prior infection with EBV predisposes to the development of RA. This contrasts with meta-analyses that indicate EBV infection is associated with multiple sclerosis and SLE. Electronic supplementary material The online version of this article (doi:10.1186/s13075-015-0755-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Robert J Ball
- Health Services Research Unit, Division of Applied Health Sciences, University of Aberdeen, Foresterhill, AB25 2ZD, Aberdeen, UK.
| | - Alison Avenell
- Research Health Services Research Unit, Division of Applied Health Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK.
| | - Lorna Aucott
- School of Medicine and Dentistry, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK.
| | - Peter Hanlon
- Health Services Research Unit, Division of Applied Health Sciences, University of Aberdeen, Foresterhill, AB25 2ZD, Aberdeen, UK.
| | - Mark A Vickers
- Division of Applied Medicine, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK. .,Blood Transfusion Centre, Foresterhill Road, Aberdeen, AB25 2ZW, UK.
| |
Collapse
|
82
|
Farjam M, Zhang GX, Ciric B, Rostami A. Emerging immunopharmacological targets in multiple sclerosis. J Neurol Sci 2015; 358:22-30. [PMID: 26440421 DOI: 10.1016/j.jns.2015.09.346] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 09/09/2015] [Accepted: 09/10/2015] [Indexed: 10/23/2022]
Abstract
Inflammatory demyelination of the central nervous system (CNS) is the hallmark of multiple sclerosis (MS), a chronic debilitating disease that affects more than 2.5 million individuals worldwide. It has been widely accepted, although not proven, that the major pathogenic mechanism of MS involves myelin-reactive T cell activation in the periphery and migration into the CNS, which subsequently triggers an inflammatory cascade that leads to demyelination and axonal damage. Virtually all MS medications now in use target the immune system and prevent tissue damage by modulating neuroinflammatory processes. Although current therapies such as commonly prescribed disease-modifying medications decrease the relapse rate in relapsing-remitting MS (RRMS), the prevention of long-term accumulation of deficits remains a challenge. Medications used for progressive forms of MS also have limited efficacy. The need for therapies that are effective against disease progression continues to drive the search for novel pharmacological targets. In recent years, due to a better understanding of MS immunopathogenesis, new approaches have been introduced that more specifically target autoreactive immune cells and their products, thus increasing specificity and efficacy, while reducing potential side effects such as global immunosuppression. In this review we describe several immunopharmacological targets that are currently being explored for MS therapy.
Collapse
Affiliation(s)
- Mojtaba Farjam
- Non-communicable Diseases Research Center, Department of Medical Pharmacology, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Guang-Xian Zhang
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Bogoljub Ciric
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Abdolmohamad Rostami
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
83
|
Latent virus infection upregulates CD40 expression facilitating enhanced autoimmunity in a model of multiple sclerosis. Sci Rep 2015; 5:13995. [PMID: 26356194 PMCID: PMC4564856 DOI: 10.1038/srep13995] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 08/13/2015] [Indexed: 02/07/2023] Open
Abstract
Epstein-Barr virus (EBV) has been identified as a putative environmental trigger of multiple sclerosis (MS) by multiple groups working worldwide. Previously, we reported that when experimental autoimmune encephalomyelitis (EAE) was induced in mice latently infected with murine γ-herpesvirus 68 (γHV-68), the murine homolog to EBV, a disease more reminiscent of MS developed. Specifically, MS-like lesions developed in the brain that included equal numbers of IFN-γ producing CD4+ and CD8+ T cells and demyelination, none of which is observed in MOG induced EAE. Herein, we demonstrate that this enhanced disease was dependent on the γHV-68 latent life cycle and was associated with STAT1 and CD40 upregulation on uninfected dendritic cells. Importantly, we also show that, during viral latency, the frequency of regulatory T cells is reduced via a CD40 dependent mechanism and this contributes towards a strong T helper 1 response that resolves in severe EAE disease pathology. Latent γ-herpesvirus infection established a long-lasting impact that enhances subsequent adaptive autoimmune responses.
Collapse
|
84
|
Rigante D, Esposito S. Infections and Systemic Lupus Erythematosus: Binding or Sparring Partners? Int J Mol Sci 2015; 16:17331-17343. [PMID: 26230690 PMCID: PMC4581196 DOI: 10.3390/ijms160817331] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 07/13/2015] [Accepted: 07/24/2015] [Indexed: 11/23/2022] Open
Abstract
Extensive work on experimental animal models clearly demonstrates that infectious agents can break immunological tolerance to self-antigens and induce autoimmune disorders, mainly systemic lupus erythematosus (SLE). The establishment of a causative link between infections and autoimmunity has been largely studied in a host of clinical studies, proving the role of infectious agents in the induction, as well as in the progression or exacerbation of SLE. However, we are far from a plain understanding of microbial-host interactions in the pathogenesis of SLE. Much serological, molecular and geoepidemiological evidence supports the relationship of different environmental infectious triggers in the inception of SLE-related autoimmune phenomena with adjuvant effects. The promotion of autoimmune responses through bystander activation or epitope spreading via multiple inflammatory pathways has been confirmed in animal models. Different viruses have been implicated in SLE pathogenesis, particularly Epstein-Barr virus, but also parvovirus B19, cytomegalovirus and retroviruses. SLE patients usually have an impaired immune response towards Epstein-Barr virus and dysregulation of the viral latency period. Furthermore, the accumulation of endogenous retroviral products might trigger the production of interferon and anti-DNA antibodies. In addition, protozoan infections might even protect from autoimmune processes and rescind an ongoing B cell activation. Herein, we discuss which type of infections induce, exacerbate or inhibit autoimmune disorders and analyze the principal infection-induced immunological mechanisms influencing the development of SLE.
Collapse
Affiliation(s)
- Donato Rigante
- Institute of Pediatrics, Università Cattolica Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli, 00168 Rome, Italy.
| | - Susanna Esposito
- Pediatric Highly Intensive Care Unit, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy.
| |
Collapse
|
85
|
Mortazavi H, Hejazi P, Khamesipour A, Mohebali M, Ehsani AH, Mohammadi Y, Farahani IV, Amirzargar AA. Frequency of seropositivity against infectious agents amongst pemphigus vulgaris patients: a case-control study on Strongyloides stercoralis, Helicobacter pylori, Toxoplasma gondii, Leishmania major, and Epstein-Barr virus. Int J Dermatol 2015; 54:e458-65. [PMID: 26175264 DOI: 10.1111/ijd.12869] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Revised: 08/04/2014] [Accepted: 09/14/2014] [Indexed: 01/03/2023]
Abstract
BACKGROUND Environmental and genetic factors may contribute to the pathogenesis of pemphigus vulgaris (PV) as an autoimmune disease. We aimed to determine rates of seropositivity for immunoglobulin G (IgG) antibodies against a number of infectious agents in untreated and treated PV patients and in healthy individuals. METHODS Eighty-two newly diagnosed untreated PV patients (34 men and 48 women; mean ± standard deviation [SD] age: 44.18 ± 14.43 years) and 36 previously diagnosed patients under immunosuppressive therapy (16 men and 20 women; mean ± SD age: 38.53 ± 9.96 years) were enrolled in the study. The clinical diagnosis of PV was confirmed by histopathology and direct immunofluorescence findings. As a control group, 131 healthy individuals (68 men and 63 women; mean ± SD age: 42.56 ± 19.69 years) were recruited. In all patients and controls, serum IgG antibodies against Strongyloides stercoralis, Helicobacter pylori, Epstein-Barr virus (EBV) capsid antigen, and Leishmania major were measured using enzyme-linked immunosorbent assays. The indirect immunofluorescence test was used to detect IgG antibodies against Toxoplasma gondii. RESULTS Newly-diagnosed untreated PV patients had significantly higher rates of seropositivity of IgG antibodies against S. stercoralis and H. pylori compared with the control group (69.5% vs. 16.0% [P < 0.001] and 79.3% vs. 59.5% [P = 0.004], respectively). For the other agents, namely T. gondii, L. major, and EBV capsid antigen, the differences between groups in seropositivity for IgG antibodies were not statistically significant. CONCLUSIONS Significant associations between S. stercoralis and H. pylori seropositivity rates and untreated disease led to the hypothesis that these pathogenic agents may contribute to the pathogenesis of PV.
Collapse
Affiliation(s)
- Hossein Mortazavi
- Autoimmune Bullous Diseases Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Department of Dermatology, Razi Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Pardis Hejazi
- Department of Dermatology, Razi Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Khamesipour
- Center for Research and Training in Skin Diseases and Leprosy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Mohebali
- Department of Medical Parasitology and Mycology, School of Public Health, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Center for Research of Endemic Parasites of Iran (CREPI), School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Hooshang Ehsani
- Department of Dermatology, Razi Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Younes Mohammadi
- Department of Epidemiology and Biostatistics, School of Public Health, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Iman Vasheghani Farahani
- Department of Industrial and Systems Engineering, North Carolina State University, Raleigh, NC, 27695, USA
| | - Ali Akbar Amirzargar
- Molecular Immunology Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
86
|
Epstein-Barr virus in peripheral blood is associated with response to rituximab therapy in rheumatoid arthritis patients. Clin Rheumatol 2015; 34:1485-8. [PMID: 26076687 DOI: 10.1007/s10067-015-2992-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 06/07/2015] [Indexed: 02/05/2023]
Abstract
Autoreactive B cells infected by Epstein-Barr virus (EBV) are suspected to be involved in the etiology of various human chronic autoimmune diseases. This motivated us to study the relationship between peripheral blood EBV load at baseline and treatment response to B cell-depleting therapy in rheumatoid arthritis (RA) patients. Thirty-five RA patients who started treatment with rituximab (RTX) in a routine clinical setting were assessed for baseline disease activity using disease activity score using 28 joint counts (DAS28) (erythrocyte sedimentation rate [ESR]). Treatment response was evaluated 3-7 months after RTX. EBV load in baseline whole blood (WB) samples was determined using quantitative PCR. EBV DNA was detected in 16/35 (46 %) of the WB samples. In these 16 EBV-positive patients, the median viral load was 3.15 (2.68-4.00) log copies/ml. Good/moderate European League Against Rheumatism (EULAR) response was observed in 16/16 of the EBV DNA-positive vs 13/19 EBV DNA-negative patients, p = 0.022. Significant response (DAS28 change >1.2) was observed in 14/16 of the EBV DNA-positive vs 10/19 EBV DNA-negative patients, p = 0.035. The decline in DAS28 after RTX was 2.10 (1.03-4. 78) in the EBV DNA-positive vs 1.47 (-0.7-4.70) in the EBV DNA-negative patients, p = 0.13. EBV load at baseline significantly correlated with change in DAS28 after RTX (τB = -0.261, p = 0.042). Our results suggest that the presence of EBV genome in WB could serve as a predictive marker to RTX therapy in RA.
Collapse
|
87
|
Pender MP, Khanna R. Epstein-Barr virus-specific adoptive immunotherapy: a new horizon for multiple sclerosis treatment? Immunotherapy 2015; 6:659-61. [PMID: 25186599 DOI: 10.2217/imt.14.43] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Michael P Pender
- The University of Queensland, School of Medicine, Brisbane, Queensland, Australia
| | | |
Collapse
|
88
|
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease that is characterized by the development of autoantibodies and immunologic attack of different organ systems, including the skin. This review aims to provide an overview of some of the pathogenic processes that may be important in the development of SLE, specifically cutaneous lupus erythematosus, and then illustrates how therapies might be tailored to modify these processes and treat disease.
Collapse
Affiliation(s)
- Mark G Kirchhof
- Department of Dermatology and Skin Science, University of British Columbia, 835 West 10th Avenue, Vancouver, British Columbia V5Z 4E8, Canada
| | - Jan P Dutz
- Department of Dermatology and Skin Science, University of British Columbia, 835 West 10th Avenue, Vancouver, British Columbia V5Z 4E8, Canada; Child and Family Research Institute, University of British Columbia, 950 West 28th Avenue, Vancouver, British Columbia V5Z 4H4, Canada.
| |
Collapse
|
89
|
Song J, Chen KC. Spectacle: fast chromatin state annotation using spectral learning. Genome Biol 2015; 16:33. [PMID: 25786205 PMCID: PMC4355146 DOI: 10.1186/s13059-015-0598-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 01/26/2015] [Indexed: 11/10/2022] Open
Abstract
Epigenomic data from ENCODE can be used to associate specific combinations of chromatin marks with regulatory elements in the human genome. Hidden Markov models and the expectation-maximization (EM) algorithm are often used to analyze epigenomic data. However, the EM algorithm can have overfitting problems in data sets where the chromatin states show high class-imbalance and it is often slow to converge. Here we use spectral learning instead of EM and find that our software Spectacle overcame these problems. Furthermore, Spectacle is able to find enhancer subtypes not found by ChromHMM but strongly enriched in GWAS SNPs. Spectacle is available at https://github.com/jiminsong/Spectacle.
Collapse
|
90
|
Abstract
Although a role of EBV in autoimmunity is biologically plausible and evidence of altered immune responses to EBV is abundant in several autoimmune diseases, inference on causality requires the determination that disease risk is higher in individuals infected with EBV than in those uninfected and that in the latter it increases following EBV infection. This determination has so far been possible only for multiple sclerosis (MS) and, to some extent, for systemic lupus erythematosus (SLE), whereas evidence is either lacking or not supportive for other autoimmune conditions. In this chapter, we present the main epidemiological findings that justify the conclusion that EBV is a component cause of MS and SLE and possible mechanisms underlying these effects.
Collapse
|
91
|
Severa M, Rizzo F, Giacomini E, Salvetti M, Coccia EM. IFN-β and multiple sclerosis: cross-talking of immune cells and integration of immunoregulatory networks. Cytokine Growth Factor Rev 2014; 26:229-39. [PMID: 25498525 DOI: 10.1016/j.cytogfr.2014.11.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 11/05/2014] [Indexed: 12/31/2022]
Abstract
Multiple sclerosis (MS) is characterized by autoimmune inflammation affecting the central nervous system and subsequent neurodegeneration. Historically, damage was thought to be mediated exclusively by auto-antigen-activated pro-inflammatory T cells. However, more recently, we are gaining increasing knowledge on the pathogenic role played in MS by B cells, dendritic cells and monocytes. IFN-β therapy was one the first approved therapy for MS for its ability to reduce relapse rate and MRI lesion activity and to significantly decrease risk of disability progression. IFN-β-mediated mechanisms of action, even if not completely understood, mainly rely on its multifaceted pleiotropic effects resulting in sustained anti-inflammatory properties directed toward almost every immune cell type. Here, we will discuss in detail literature data characterizing the pathogenic activity of the different immune cell subsets involved in MS pathogenesis and how IFN-β therapy regulates their function by modulating bystander responses. We believe that the effectiveness of this drug in MS treatment, even if in use for a long time, can unveil new insights on this disease and still teach a lesson to researchers in the MS field.
Collapse
Affiliation(s)
- Martina Severa
- Department of Infectious, Parasitic and Immune-mediated Diseases, Istituto Superiore di Sanità, Rome, Italy.
| | - Fabiana Rizzo
- Department of Infectious, Parasitic and Immune-mediated Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Elena Giacomini
- Department of Infectious, Parasitic and Immune-mediated Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Marco Salvetti
- Centre for Experimental Neurological Therapies (CENTERS) - Neurology and Department of Neurosciences, Mental Health and Sensory Organs; Sapienza, University of Rome, S. Andrea Hospital Site, Italy
| | - Eliana M Coccia
- Department of Infectious, Parasitic and Immune-mediated Diseases, Istituto Superiore di Sanità, Rome, Italy.
| |
Collapse
|
92
|
Pender MP, Burrows SR. Epstein-Barr virus and multiple sclerosis: potential opportunities for immunotherapy. Clin Transl Immunology 2014; 3:e27. [PMID: 25505955 PMCID: PMC4237030 DOI: 10.1038/cti.2014.25] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Revised: 09/30/2014] [Accepted: 09/30/2014] [Indexed: 01/04/2023] Open
Abstract
Multiple sclerosis (MS) is a common chronic inflammatory demyelinating disease of the central nervous system (CNS) causing progressive disability. Many observations implicate Epstein–Barr virus (EBV) in the pathogenesis of MS, namely universal EBV seropositivity, high anti-EBV antibody levels, alterations in EBV-specific CD8+ T-cell immunity, increased spontaneous EBV-induced transformation of peripheral blood B cells, increased shedding of EBV from saliva and accumulation of EBV-infected B cells and plasma cells in the brain. Several mechanisms have been postulated to explain the role of EBV in the development of MS including cross-reactivity between EBV and CNS antigens, bystander damage to the CNS by EBV-specific CD8+ T cells, activation of innate immunity by EBV-encoded small RNA molecules in the CNS, expression of αB-crystallin in EBV-infected B cells leading to a CD4+ T-cell response against oligodendrocyte-derived αB-crystallin and EBV infection of autoreactive B cells, which produce pathogenic autoantibodies and provide costimulatory survival signals to autoreactive T cells in the CNS. The rapidly accumulating evidence for a pathogenic role of EBV in MS provides ground for optimism that it might be possible to prevent and cure MS by effectively controlling EBV infection through vaccination, antiviral drugs or treatment with EBV-specific cytotoxic CD8+ T cells. Adoptive immunotherapy with in vitro-expanded autologous EBV-specific CD8+ T cells directed against viral latent proteins was recently used to treat a patient with secondary progressive MS. Following the therapy, there was clinical improvement, decreased disease activity on magnetic resonance imaging and reduced intrathecal immunoglobulin production.
Collapse
Affiliation(s)
- Michael P Pender
- School of Medicine, The University of Queensland , Brisbane, QLD, Australia ; Department of Neurology, Royal Brisbane and Women's Hospital , Brisbane, QLD, Australia ; QIMR Berghofer Medical Research Institute , Brisbane, QLD, Australia
| | - Scott R Burrows
- School of Medicine, The University of Queensland , Brisbane, QLD, Australia ; QIMR Berghofer Medical Research Institute , Brisbane, QLD, Australia
| |
Collapse
|
93
|
Non-stiff anti-amphiphysin syndrome: Clinical manifestations and outcome after immunotherapy. J Neuroimmunol 2014; 274:209-14. [DOI: 10.1016/j.jneuroim.2014.07.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 07/13/2014] [Accepted: 07/16/2014] [Indexed: 12/29/2022]
|
94
|
Croia C, Astorri E, Murray-Brown W, Willis A, Brokstad KA, Sutcliffe N, Piper K, Jonsson R, Tappuni AR, Pitzalis C, Bombardieri M. Implication of Epstein-Barr Virus Infection in Disease-Specific Autoreactive B Cell Activation in Ectopic Lymphoid Structures of Sjögren's Syndrome. Arthritis Rheumatol 2014; 66:2545-57. [DOI: 10.1002/art.38726] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 05/22/2014] [Indexed: 12/11/2022]
Affiliation(s)
- Cristina Croia
- William Harvey Research Institute, Queen Mary University of London; London UK
| | - Elisa Astorri
- William Harvey Research Institute, Queen Mary University of London; London UK
| | | | - Amanda Willis
- Barts and The London School of Medicine and Dentistry, Queen Mary University of London; London UK
| | | | - Nurhan Sutcliffe
- Barts and The London School of Medicine and Dentistry, Queen Mary University of London; London UK
| | - Kim Piper
- Barts and The London School of Medicine and Dentistry, Queen Mary University of London; London UK
| | | | - Anwar R. Tappuni
- Barts and The London School of Medicine and Dentistry, Queen Mary University of London; London UK
| | - Costantino Pitzalis
- William Harvey Research Institute, Queen Mary University of London; London UK
| | - Michele Bombardieri
- William Harvey Research Institute, Queen Mary University of London; London UK
| |
Collapse
|
95
|
Pitzalis C, Jones GW, Bombardieri M, Jones SA. Ectopic lymphoid-like structures in infection, cancer and autoimmunity. Nat Rev Immunol 2014; 14:447-62. [PMID: 24948366 DOI: 10.1038/nri3700] [Citation(s) in RCA: 520] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Ectopic lymphoid-like structures often develop at sites of inflammation where they influence the course of infection, autoimmune disease, cancer and transplant rejection. These lymphoid aggregates range from tight clusters of B cells and T cells to highly organized structures that comprise functional germinal centres. Although the mechanisms governing ectopic lymphoid neogenesis in human pathology remain poorly defined, the presence of ectopic lymphoid-like structures within inflamed tissues has been linked to both protective and deleterious outcomes in patients. In this Review, we discuss investigations in both experimental model systems and patient cohorts to provide a perspective on the formation and functions of ectopic lymphoid-like structures in human pathology, with particular reference to the clinical implications and the potential for therapeutic targeting.
Collapse
Affiliation(s)
- Costantino Pitzalis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London, School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Gareth W Jones
- Cardiff Institute for Infection and Immunity, The School of Medicine, Cardiff University, The Tenovus Building, Heath Campus, Cardiff CF14 4XN, Wales, UK
| | - Michele Bombardieri
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London, School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Simon A Jones
- Cardiff Institute for Infection and Immunity, The School of Medicine, Cardiff University, The Tenovus Building, Heath Campus, Cardiff CF14 4XN, Wales, UK
| |
Collapse
|
96
|
Pender MP, Csurhes PA, Pfluger CM, Burrows SR. Deficiency of CD8+ effector memory T cells is an early and persistent feature of multiple sclerosis. Mult Scler 2014; 20:1825-32. [PMID: 24842963 PMCID: PMC4361480 DOI: 10.1177/1352458514536252] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Patients with multiple sclerosis (MS) have a deficiency of circulating CD8+ T cells, which might impair control of Epstein-Barr virus (EBV) and predispose to MS by allowing EBV-infected autoreactive B cells to accumulate in the central nervous system. Based on the expression of CD45RA and CD62L, CD4+ T cells and CD8+ T cells can be subdivided into four subsets with distinct homing and functional properties, namely: naïve, central memory, effector memory (EM) and effector memory re-expressing CD45RA (EMRA) cells. OBJECTIVE Our aim was to determine which memory subsets are involved in the CD8+ T cell deficiency and how these relate to clinical course. METHODS We used flow cytometry to analyze the memory phenotypes of T cells in the blood of 118 MS patients and 112 healthy subjects. RESULTS MS patients had a decreased frequency of EM (CD45RA(-)CD62L(-)) and EMRA (CD45RA(+)CD62L(-)) CD8+ T cells, which was present at the onset of disease and persisted throughout the clinical course. The frequencies of CD4+ EM and EMRA T cells were normal. CONCLUSION Deficiency of effector memory CD8+ T cells is an early and persistent feature of MS and might underlie the impaired CD8+ T cell control of EBV.
Collapse
Affiliation(s)
- Michael P Pender
- The University of Queensland, School of Medicine, Brisbane, Australia/Royal Brisbane and Women's Hospital, Australia/QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Peter A Csurhes
- The University of Queensland, School of Medicine, Brisbane, Australia/The University of Queensland Centre for Clinical Research, Brisbane, Australia
| | - Casey Mm Pfluger
- The University of Queensland, School of Medicine, Brisbane, Australia/The University of Queensland Centre for Clinical Research, Brisbane, Australia
| | - Scott R Burrows
- The University of Queensland, School of Medicine, Brisbane, Australia/QIMR Berghofer Medical Research Institute, Brisbane, Australia
| |
Collapse
|
97
|
Getts DR, Chastain EML, Terry RL, Miller SD. Virus infection, antiviral immunity, and autoimmunity. Immunol Rev 2014; 255:197-209. [PMID: 23947356 DOI: 10.1111/imr.12091] [Citation(s) in RCA: 210] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 05/08/2013] [Indexed: 12/12/2022]
Abstract
As a group of disorders, autoimmunity ranks as the third most prevalent cause of morbidity and mortality in the Western World. However, the etiology of most autoimmune diseases remains unknown. Although genetic linkage studies support a critical underlying role for genetics, the geographic distribution of these disorders as well as the low concordance rates in monozygotic twins suggest that a combination of other factors including environmental ones are involved. Virus infection is a primary factor that has been implicated in the initiation of autoimmune disease. Infection triggers a robust and usually well-coordinated immune response that is critical for viral clearance. However, in some instances, immune regulatory mechanisms may falter, culminating in the breakdown of self-tolerance, resulting in immune-mediated attack directed against both viral and self-antigens. Traditionally, cross-reactive T-cell recognition, known as molecular mimicry, as well as bystander T-cell activation, culminating in epitope spreading, have been the predominant mechanisms elucidated through which infection may culminate in an T-cell-mediated autoimmune response. However, other hypotheses including virus-induced decoy of the immune system also warrant discussion in regard to their potential for triggering autoimmunity. In this review, we discuss the mechanisms by which virus infection and antiviral immunity contribute to the development of autoimmunity.
Collapse
Affiliation(s)
- Daniel R Getts
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | | | | | | |
Collapse
|
98
|
Fissolo N. Epstein-Barr virus-specific adoptive immunotherapy for progressive multiple sclerosis. Mult Scler 2014; 20:1545. [PMID: 24622348 DOI: 10.1177/1352458514527025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Nicolás Fissolo
- Department of Neurology and Neuroimmunology, Centre d'Esclerosi Múltiple de Catalunya, Cemcat, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| |
Collapse
|
99
|
Mifsud EJ, Tan ACL, Jackson DC. TLR Agonists as Modulators of the Innate Immune Response and Their Potential as Agents Against Infectious Disease. Front Immunol 2014; 5:79. [PMID: 24624130 PMCID: PMC3939722 DOI: 10.3389/fimmu.2014.00079] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 02/13/2014] [Indexed: 12/28/2022] Open
Abstract
Immunotherapies that can either activate or suppress innate immune responses are being investigated as treatments against infectious diseases and the pathology they can cause. The objective of these therapies is to elicit protective immune responses thereby limiting the harm inflicted by the pathogen. The Toll-like receptor (TLR) signaling pathway plays critical roles in numerous host immune defenses and has been identified as an immunotherapeutic target against the consequences of infectious challenge. This review focuses on some of the recent advances being made in the development of TLR-ligands as potential prophylactic and/or therapeutic agents.
Collapse
Affiliation(s)
- Edin J. Mifsud
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Amabel C. L. Tan
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - David C. Jackson
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| |
Collapse
|
100
|
Lucchesi D, Pitzalis C, Bombardieri M. EBV and other viruses as triggers of tertiary lymphoid structures in primary Sjögren's syndrome. Expert Rev Clin Immunol 2014; 10:445-55. [PMID: 24564506 DOI: 10.1586/1744666x.2014.892417] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Sjögren's syndrome (SS) is an autoimmune disease that targets salivary (SG) and lachrymal glands, leading to exocrine dysfunction. Several viruses have been associated with SS, although the role of persistent viral infections in triggering and/or perpetuating the disease is still a matter of controversy. Together with exocrine dysfunction, SS is characterised by the production of autoantibodies and the presence of lymphomonocytic periductal aggregates in the SG, which in 30/40% of the patients display features of tertiary lymphoid structures (TLS) supporting an ectopic germinal centre response. Here we first review i) the relevance of TLS in SS and ii) the evidence in support of a role for viruses in SS insurgence and/or persistence; next, iii) we review recent data which links viral infection with TLS formation in the SG and suggests that viral-host interactions within TLS favour breach of tolerance and development of autoimmunity in SS.
Collapse
Affiliation(s)
- Davide Lucchesi
- Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | | | | |
Collapse
|