1
|
Ushio A, Matsuda-Lennikov M, Kalle-Youngoue F, Shimizu A, Abdelmaksoud A, Kelly MC, Ishimaru N, Takahama Y. Functionally diverse thymic medullary epithelial cells interplay to direct central tolerance. Cell Rep 2024; 43:114072. [PMID: 38581680 PMCID: PMC11079940 DOI: 10.1016/j.celrep.2024.114072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 02/24/2024] [Accepted: 03/21/2024] [Indexed: 04/08/2024] Open
Abstract
Medullary thymic epithelial cells (mTECs) are essential for the establishment of self-tolerance in T cells. Promiscuous gene expression by a subpopulation of mTECs regulated by the nuclear protein Aire contributes to the display of self-genomic products to newly generated T cells. Recent reports have highlighted additional self-antigen-displaying mTEC subpopulations, namely Fezf2-expressing mTECs and a mosaic of self-mimetic mTECs including thymic tuft cells. In addition, a functionally different subset of mTECs produces chemokine CCL21, which attracts developing thymocytes to the medullary region. Here, we report that CCL21+ mTECs and Aire+ mTECs non-redundantly cooperate to direct self-tolerance to prevent autoimmune pathology by optimizing the deletion of self-reactive T cells and the generation of regulatory T cells. We also detect cooperation for self-tolerance between Aire and Fezf2, the latter of which unexpectedly regulates thymic tuft cells. Our results indicate an indispensable interplay among functionally diverse mTECs for the establishment of central self-tolerance.
Collapse
Affiliation(s)
- Aya Ushio
- Thymus Biology Section, Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; Department of Oral Molecular Pathology, Graduate School of Biomedical Sciences, Tokushima University, Kuramoto, Tokushima 770-8504, Japan
| | - Mami Matsuda-Lennikov
- Thymus Biology Section, Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Felix Kalle-Youngoue
- Thymus Biology Section, Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21701, USA
| | - Akihide Shimizu
- Thymus Biology Section, Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Abdalla Abdelmaksoud
- Center for Cancer Research Collaborative Bioinformatics Resource, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael C Kelly
- Single Cell Analysis Facility, Cancer Research Technology Program, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Naozumi Ishimaru
- Department of Oral Molecular Pathology, Graduate School of Biomedical Sciences, Tokushima University, Kuramoto, Tokushima 770-8504, Japan
| | - Yousuke Takahama
- Thymus Biology Section, Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
2
|
Yura Y, Hamada M. Outline of Salivary Gland Pathogenesis of Sjögren's Syndrome and Current Therapeutic Approaches. Int J Mol Sci 2023; 24:11179. [PMID: 37446355 DOI: 10.3390/ijms241311179] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 06/30/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Sjögren's syndrome (SS) is an autoimmune disease characterized by the involvement of exocrine glands such as the salivary and lacrimal glands. The minor salivary glands, from which tissue samples may be obtained, are important for the diagnosis, evaluation of therapeutic efficacy, and genetic analyses of SS. In the onset of SS, autoantigens derived from the salivary glands are recognized by antigen-presenting dendritic cells, leading to the activation of T and B cells, cytokine production, autoantibody production by plasma cells, the formation of ectopic germinal centers, and the destruction of salivary gland epithelial cells. A recent therapeutic approach with immune checkpoint inhibitors for malignant tumors enhances the anti-tumor activity of cytotoxic effector T cells, but also induces SS-like autoimmune disease as an adverse event. In the treatment of xerostomia, muscarinic agonists and salivary gland duct cleansing procedure, as well as sialendoscopy, are expected to ameliorate symptoms. Clinical trials on biological therapy to attenuate the hyperresponsiveness of B cells in SS patients with systemic organ involvement have progressed. The efficacy of treatment with mesenchymal stem cells and chimeric antigen receptor T cells for SS has also been investigated. In this review, we will provide an overview of the pathogenesis of salivary gland lesions and recent trends in therapeutic approaches for SS.
Collapse
Affiliation(s)
- Yoshiaki Yura
- Department of Oral & Maxillofacial Oncology and Surgery, Osaka University Graduate School of Dentistry, Osaka 565-0871, Japan
| | - Masakazu Hamada
- Department of Oral & Maxillofacial Oncology and Surgery, Osaka University Graduate School of Dentistry, Osaka 565-0871, Japan
| |
Collapse
|
3
|
Sisto M, Ribatti D, Lisi S. Molecular Mechanisms Linking Inflammation to Autoimmunity in Sjögren's Syndrome: Identification of New Targets. Int J Mol Sci 2022; 23:13229. [PMID: 36362017 PMCID: PMC9658723 DOI: 10.3390/ijms232113229] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/26/2022] [Accepted: 10/26/2022] [Indexed: 10/15/2023] Open
Abstract
Sjögren's syndrome (SS) is a systemic autoimmune rheumatic disorder characterized by the lymphocytic infiltration of exocrine glands and the production of autoantibodies to self-antigens. The involvement of the exocrine glands drives the pathognomonic manifestations of dry eyes (keratoconjunctivitis sicca) and dry mouth (xerostomia) that define sicca syndrome. To date, the molecular mechanisms mediating pathological salivary gland dysfunction in SS remain to be elucidated, despite extensive studies investigating the underlying cause of this disease, hampering the development of novel therapeutic strategies. Many researchers have identified a multifactorial pathogenesis of SS, including environmental, genetic, neuroendocrine, and immune factors. In this review, we explore the latest developments in understanding the molecular mechanisms involved in the pathogenesis of SS, which have attracted increasing interest in recent years.
Collapse
Affiliation(s)
- Margherita Sisto
- Department of Translational Biomedicine and Neuroscience (DiBraiN), Section of Human Anatomy and Histology, University of Bari “Aldo Moro”, Piazza Giulio Cesare 1, I-70124 Bari, Italy
| | | | | |
Collapse
|
4
|
Sato M, Arakaki R, Tawara H, Nagao R, Tanaka H, Tamura K, Kawahito Y, Otsuka K, Ushio A, Tsunematsu T, Ishimaru N. Disturbed natural killer cell homeostasis in the salivary gland enhances autoimmune pathology via IFN-γ in a mouse model of primary Sjögren's syndrome. Front Med (Lausanne) 2022; 9:1036787. [PMID: 36388880 PMCID: PMC9643684 DOI: 10.3389/fmed.2022.1036787] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 10/12/2022] [Indexed: 07/22/2023] Open
Abstract
OBJECTIVE Innate lymphoid cells (ILCs), including natural killer (NK) cells, ILC1, ILC2, lymphoid tissue-inducer (LTi) cells, and ILC3 cell, play a key role in various immune responses. Primary Sjögren's syndrome (pSS) is an autoimmune disease characterized by chronic inflammation of exocrine glands, such as the lacrimal and salivary glands (SGs). The role of NK cells among ILCs in the pathogenesis of pSS is still unclear. In this study, the characteristics and subsets of NK cells in the salivary gland (SG) tissue were analyzed using a murine model of pSS. METHODS Multiple phenotypes and cytotoxic signature of the SG NK cells in control and pSS model mice were evaluated by flow cytometric analysis. Intracellular expression of interferon-γ (IFN-γ) among T cells and NK cells from the SG tissues was compared by in vitro experiments. In addition, pathological analysis was performed using anti-asialo-GM1 (ASGM1) antibody (Ab)-injected pSS model mice. RESULTS The number of conventional NK (cNK) cells in the SG of pSS model mice significantly increased compared with that in control mice at 6 weeks of age. The production level of IFN-γ was significantly higher in SG NK cells than in SG T cells. The depletion of NK cells by ASGM1 Ab altered the ratio of tissue resident NK (rNK) cells to cNK cells, which inhibited the injury to SG cells with the recovery of saliva secretion in pSS model mice. CONCLUSION The results indicate that SG cNK cells may enhance the autoreactive response in the target organ by upregulating of IFN-γ, whereas SG rNK cells protect target cells against T cell cytotoxicity. Therefore, the activation process and multiple functions of NK cells in the target organ could be helpful to develop potential markers for determining autoimmune disease activity and target molecules for incurable immune disorders.
Collapse
|
5
|
Baradaran A, Rostamian A, Mottaghi P, Salesi M, Karimifar M, Mansouri V. Evaluation of diagnostic accuracy of alpha-fodrin antibody in Iranian patients with Sjogren's disease. Adv Biomed Res 2022; 11:64. [PMID: 36124018 PMCID: PMC9482376 DOI: 10.4103/abr.abr_260_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 04/11/2021] [Accepted: 06/12/2021] [Indexed: 11/04/2022] Open
Abstract
Background: Materials and Methods: Results: Conclusions:
Collapse
|
6
|
Autoimmune Epithelitis and Chronic Inflammation in Sjögren's Syndrome-Related Dry Eye Disease. Int J Mol Sci 2021; 22:ijms222111820. [PMID: 34769250 PMCID: PMC8584177 DOI: 10.3390/ijms222111820] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 10/07/2021] [Accepted: 10/09/2021] [Indexed: 12/18/2022] Open
Abstract
Autoimmune epithelitis and chronic inflammation are one of the characteristic features of the immune pathogenesis of Sjögren’s syndrome (SS)-related dry eye disease. Autoimmune epithelitis can cause the dysfunction of the excretion of tear fluid and mucin from the lacrimal glands and conjunctival epithelia and meibum from the meibomian glands. The lacrimal gland and conjunctival epithelia express major histocompatibility complex class II or human leukocyte antigen-DR and costimulatory molecules, acting as nonprofessional antigen-presenting cells for T cell and B cell activation in SS. Ocular surface epithelium dysfunction can lead to dry eye disease in SS. Considering the mechanisms underlying SS-related dry eye disease, this review highlights autoimmune epithelitis of the ocular surface, chronic inflammation, and several other molecules in the tear film, cornea, conjunctiva, lacrimal glands, and meibomian glands that represent potential targets in the treatment of SS-related dry eye disease.
Collapse
|
7
|
Huang W, Tourmouzis K, Perry H, Honkanen RA, Rigas B. Animal models of dry eye disease: Useful, varied and evolving (Review). Exp Ther Med 2021; 22:1394. [PMID: 34650642 PMCID: PMC8506913 DOI: 10.3892/etm.2021.10830] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/10/2021] [Indexed: 02/07/2023] Open
Abstract
Dry eye disease (DED), which is a prevalent disease that still lacks successful treatment options, remains a major challenge in ophthalmology. Multiple animal models of DED have been used to decipher its pathophysiology and to develop novel treatments. These models use mice, rats, rabbits, cats, dogs and non-human primates. Each model assesses aspects of DED by focusing on elements of the lacrimal functional unit, which controls the homeostasis of the tear film. The present review outlines representative DED animal models and assesses their contribution to the study of DED. Murine models are the most extensively used, followed by rabbit models; the latter offer the advantage of larger eyes, a favorable biochemical profile for drug studies, experimental ease and relatively low cost, contrasting with non-human primates, which, although closer to humans, are not as accessible and are expensive. No comprehensive ‘ideal’ animal model encompassing all aspects of human DED exists nor is it feasible. Investigators often choose an animal model based on their experimental needs and the following four features of a given model: The size of the eye, its biochemical composition, the available research reagents and cost. As research efforts in DED expand, more refined animal models are needed to supplement the enormous contribution made to date by existing models.
Collapse
Affiliation(s)
- Wei Huang
- Department of Ophthalmology, Stony Brook University, Stony Brook, NY 11794, USA.,Department of Ophthalmology, Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | | | - Henry Perry
- Ophthalomology Consultants of Long Island, Westbury, NY 11590, USA
| | - Robert A Honkanen
- Department of Ophthalmology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Basil Rigas
- Department of Preventive Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
8
|
Fritzler MJ, Choi MY, Satoh M, Mahler M. Autoantibody Discovery, Assay Development and Adoption: Death Valley, the Sea of Survival and Beyond. Front Immunol 2021; 12:679613. [PMID: 34122443 PMCID: PMC8191456 DOI: 10.3389/fimmu.2021.679613] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 05/04/2021] [Indexed: 01/08/2023] Open
Abstract
Dating to the discovery of the Lupus Erythematosus (LE) cell in 1948, there has been a dramatic growth in the discovery of unique autoantibodies and their cognate targets, all of which has led to the availability and use of autoantibody testing for a broad spectrum of autoimmune diseases. Most studies of the sensitivity, specificity, commutability, and harmonization of autoantibody testing have focused on widely available, commercially developed and agency-certified autoantibody kits. However, this is only a small part of the spectrum of autoantibody tests that are provided through laboratories world-wide. This manuscript will review the wider spectrum of testing by exploring the innovation pathway that begins with autoantibody discovery followed by assessment of clinical relevance, accuracy, validation, and then consideration of regulatory requirements as an approved diagnostic test. Some tests are offered as "Research Use Only (RUO)", some as "Laboratory Developed Tests (LDT)", some enter Health Technology Assessment (HTA) pathways, while others are relegated to a "death valley" of autoantibody discovery and become "orphan" autoantibodies. Those that achieve regulatory approval are further threatened by the business world's "Darwinian Sea of Survival". As one example of the trappings of autoantibody progression or failure, it is reported that more than 200 different autoantibodies have been described in systemic lupus erythematosus (SLE), a small handful (~10%) of these have achieved regulatory approval and are widely available as commercial diagnostic kits, while a few others may be available as RUO or LDT assays. However, the vast majority (90%) are orphaned and languish in an autoantibody 'death valley'. This review proposes that it is important to keep an inventory of these "orphan autoantibodies" in 'death valley' because, with the increasing availability of multi-analyte arrays and artificial intelligence (MAAI), some can be rescued to achieve a useful role in clinical diagnostic especially in light of patient stratification and precision medicine.
Collapse
Affiliation(s)
- Marvin J Fritzler
- Department of Medicine, Cumming School of Medicine, Calgary, AB, Canada
| | - May Y Choi
- Department of Medicine, Cumming School of Medicine, Calgary, AB, Canada
| | - Minoru Satoh
- Department of Clinical Nursing, School of Health Sciences, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Michael Mahler
- Research and Development, Inova Diagnostics, San Diego, CA, United States
| |
Collapse
|
9
|
Chen W, Wang Q, Zhou B, Zhang L, Zhu H. Lipid Metabolism Profiles in Rheumatic Diseases. Front Pharmacol 2021; 12:643520. [PMID: 33897433 PMCID: PMC8064727 DOI: 10.3389/fphar.2021.643520] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 02/18/2021] [Indexed: 12/25/2022] Open
Abstract
Rheumatic diseases are a group of chronic autoimmune disorders that involve multiple organs or systems and have high mortality. The mechanisms of these diseases are still ill-defined, and targeted therapeutic strategies are still challenging for physicians. Recent research indicates that cell metabolism plays important roles in the pathogenesis of rheumatic diseases. In this review, we mainly focus on lipid metabolism profiles (dyslipidaemia, fatty acid metabolism) and mechanisms in rheumatic diseases and discuss potential clinical applications based on lipid metabolism profiles.
Collapse
Affiliation(s)
- Weilin Chen
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China.,Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, China
| | - Qi Wang
- Department of Radiology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Bin Zhou
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lihua Zhang
- Department of Rheumatology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Honglin Zhu
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China.,Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, China
| |
Collapse
|
10
|
Saito I. Pathology of salivary gland dysfunction and restoration of function. Pathol Int 2021; 71:304-315. [PMID: 33751738 DOI: 10.1111/pin.13079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 01/15/2021] [Indexed: 11/27/2022]
Abstract
In this review, the author shows that simultaneous multiple disorders caused by reactivation of Epstein-Barr virus can lead to salivary gland disorders as part of Sjogren's syndrome (SS). Therefore, clinicians must differentiate SS from other diseases when diagnosing and treating salivary gland disorders. In particular, the author explains how microbial infection in SS overcomes immunological tolerance, leading to pathological changes, and how cytokine overexpression and endocrine disrupters contribute to glandular tissue injury. Also, the author suggests that involvement of reactive oxygen species is a common pathogenesis of salivary gland disorders and SS, so regulation of oxidative stress is an effective treatment for both. The results of clinical studies on restoring salivary gland function and regenerating salivary glands with tissue stem cells may provide clues on elucidating the cause of SS.
Collapse
Affiliation(s)
- Ichiro Saito
- Department of Pathology, Tsurumi University School of Dental Medicine, Kanagawa, Japan
| |
Collapse
|
11
|
Role of the Innate Immunity Signaling Pathway in the Pathogenesis of Sjögren's Syndrome. Int J Mol Sci 2021; 22:ijms22063090. [PMID: 33803026 PMCID: PMC8002742 DOI: 10.3390/ijms22063090] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/13/2021] [Accepted: 03/15/2021] [Indexed: 12/11/2022] Open
Abstract
Sjögren’s syndrome (SS) is a systemic autoimmune disease characterized by chronic inflammation of the salivary and lacrimal glands and extra-glandular lesions. Adaptive immune response including T- and B-cell activation contributes to the development of SS. However, its pathogenesis has not yet been elucidated. In addition, several patients with SS present with the type I interferon (IFN) signature, which is the upregulation of the IFN-stimulated genes induced by type I IFN. Thus, innate immune responses including type I IFN activity are associated with SS pathogenesis. Recent studies have revealed the presence of activation pattern recognition receptors (PRRs) including Toll-like receptors, RNA sensor retinoic acid-inducible gene I and melanoma differentiation-associated gene 5, and inflammasomes in infiltrating and epithelial cells of the salivary glands among patients with SS. In addition, the activation of PRRs via the downstream pathway such as the type I IFN signature and nuclear factor kappa B can directly cause organ inflammation, and it is correlated with the activation of adaptive immune responses. Therefore, this study assessed the role of the innate immune signal pathway in the development of inflammation and immune abnormalities in SS.
Collapse
|
12
|
Studying Sjögren's syndrome in mice: What is the best available model? J Oral Biol Craniofac Res 2021; 11:245-255. [PMID: 33665074 DOI: 10.1016/j.jobcr.2020.12.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 12/04/2020] [Accepted: 12/05/2020] [Indexed: 01/18/2023] Open
Abstract
Sjögren's syndrome (SS) is a common autoimmune disease characterized by lymphocytic infiltration and destruction of exocrine glands. The disease manifests primarily in the salivary and lacrimal glands, but other organs are also involved, leading to dry mouth, dry eyes, and other extra-glandular manifestations. Studying the disease in humans is entailed with many limitations and restrictions; therefore, the need for a proper mouse model is mandatory. SS mouse models are categorized, depending on the disease emergence into spontaneous or experimentally manipulated models. The usefulness of each mouse model varies depending on the SS features exhibited by that model; each SS model has advanced our understanding of the disease pathogenesis. In this review article, we list all the available murine models which have been used to study SS and we comment on the characteristics exhibited by each mouse model to assist scientists to select the appropriate model for their specific studies. We also recommend a murine strain that is the most relevant to the ideal SS model, based on our experience acquired during previous and current investigations.
Collapse
|
13
|
Yamane K, Nakamura H, Hamasaki M, Minei Y, Aibara N, Shimizu T, Kawakami A, Nakashima M, Kuroda N, Ohyama K. Immune complexome analysis reveals the presence of immune complexes and identifies disease-specific immune complex antigens in saliva samples from patients with Sjögren's syndrome. Clin Exp Immunol 2021; 204:212-220. [PMID: 33432580 DOI: 10.1111/cei.13574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 12/28/2020] [Accepted: 12/29/2020] [Indexed: 11/28/2022] Open
Abstract
Sjögren's syndrome (SS) is a chronic autoimmune disease that mainly damages the salivary and lacrimal glands. Immune complex (IC) formation triggers local inflammation through IC deposition and decreased antigen function. Some ICs can leak from the lesion and into the saliva, but no salivary ICs have been reported to date. We used immune complexome analysis to comprehensively identify antigens incorporated into IC (IC-antigens) in saliva samples from patients with SS (n = 9) or with xerostomia (n = 7). Neutrophil defensin 1 (67%), small proline-rich protein 2D (67%), myeloperoxidase (44%), neutrophil elastase (44%), cathepsin G (33%), nuclear mitotic apparatus 1 (33%) and phosphatidylinositol 4-phosphate 3-kinase C2 domain-containing subunit gamma (33%) were identified as new IC-antigens specifically and frequently detected in the saliva of SS patients. Of these, neutrophil defensin 1, myeloperoxidase, neutrophil elastase and cathepsin G are neutrophil intracellular proteins, which suggests that repeated destruction of neutrophils due to abnormal autoimmunity may be involved in the pathogenesis of SS. We also analyzed serum samples from three SS patients. There was little overlap of IC-antigens between two of the samples (fewer than 30% of the IC-antigens in the saliva samples), suggesting that many ICs are formed locally and independently of the circulation. In addition, we found that four SS-specific salivary antigens show sequence homology with several proteins of oral microbiomes but no antigen has homology with Epstein-Barr virus proteins. The homology between some IC-antigens and oral microbiome proteins may indicate the impact of oral infection on local autoimmunity through molecular mimicry theory.
Collapse
Affiliation(s)
- K Yamane
- School of Pharmaceutical Sciences, Nagasaki University, Nagasaki, Japan
| | - H Nakamura
- Department of Immunology and Rheumatology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - M Hamasaki
- School of Pharmaceutical Sciences, Nagasaki University, Nagasaki, Japan
| | - Y Minei
- School of Pharmaceutical Sciences, Nagasaki University, Nagasaki, Japan
| | - N Aibara
- Unit of Medical Pharmacy, Department of Pharmacy Practice, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - T Shimizu
- Department of Immunology and Rheumatology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - A Kawakami
- Department of Immunology and Rheumatology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - M Nakashima
- Unit of Medical Pharmacy, Department of Pharmacy Practice, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - N Kuroda
- Course of Pharmaceutical Sciences, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - K Ohyama
- Unit of Medical Pharmacy, Department of Pharmacy Practice, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
14
|
Tanaka T, Warner BM, Odani T, Ji Y, Mo YQ, Nakamura H, Jang SI, Yin H, Michael DG, Hirata N, Suizu F, Ishigaki S, Oliveira FR, Motta ACF, Ribeiro-Silva A, Rocha EM, Atsumi T, Noguchi M, Chiorini JA. LAMP3 induces apoptosis and autoantigen release in Sjögren's syndrome patients. Sci Rep 2020; 10:15169. [PMID: 32939030 PMCID: PMC7494869 DOI: 10.1038/s41598-020-71669-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 08/10/2020] [Indexed: 12/16/2022] Open
Abstract
Primary Sjögren's syndrome (pSS) is a complex autoimmune disease characterized by dysfunction of secretory epithelia with only palliative therapy. Patients present with a constellation of symptoms, and the diversity of symptomatic presentation has made it difficult to understand the underlying disease mechanisms. In this study, aggregation of unbiased transcriptome profiling data sets of minor salivary gland biopsies from controls and Sjögren's syndrome patients identified increased expression of lysosome-associated membrane protein 3 (LAMP3/CD208/DC-LAMP) in a subset of Sjögren's syndrome cases. Stratification of patients based on their clinical characteristics suggested an association between increased LAMP3 expression and the presence of serum autoantibodies including anti-Ro/SSA, anti-La/SSB, anti-nuclear antibodies. In vitro studies demonstrated that LAMP3 expression induces epithelial cell dysfunction leading to apoptosis. Interestingly, LAMP3 expression resulted in the accumulation and release of intracellular TRIM21 (one component of SSA), La (SSB), and α-fodrin protein, common autoantigens in Sjögren's syndrome, via extracellular vesicles in an apoptosis-independent mechanism. This study defines a clear role for LAMP3 in the initiation of apoptosis and an independent pathway for the extracellular release of known autoantigens leading to the formation of autoantibodies associated with this disease.ClinicalTrials.gov Identifier: NCT00001196, NCT00001390, NCT02327884.
Collapse
Affiliation(s)
- Tsutomu Tanaka
- National Institute of Dental and Craniofacial Research, National Institutes of Health, NIH 10 Center Dr., Bethesda, MD, 20892, USA
| | - Blake M Warner
- National Institute of Dental and Craniofacial Research, National Institutes of Health, NIH 10 Center Dr., Bethesda, MD, 20892, USA
| | - Toshio Odani
- National Institute of Dental and Craniofacial Research, National Institutes of Health, NIH 10 Center Dr., Bethesda, MD, 20892, USA
| | - Youngmi Ji
- National Institute of Dental and Craniofacial Research, National Institutes of Health, NIH 10 Center Dr., Bethesda, MD, 20892, USA
| | - Ying-Qian Mo
- National Institute of Dental and Craniofacial Research, National Institutes of Health, NIH 10 Center Dr., Bethesda, MD, 20892, USA
| | - Hiroyuki Nakamura
- National Institute of Dental and Craniofacial Research, National Institutes of Health, NIH 10 Center Dr., Bethesda, MD, 20892, USA
| | - Shyh-Ing Jang
- National Institute of Dental and Craniofacial Research, National Institutes of Health, NIH 10 Center Dr., Bethesda, MD, 20892, USA
| | - Hongen Yin
- National Institute of Dental and Craniofacial Research, National Institutes of Health, NIH 10 Center Dr., Bethesda, MD, 20892, USA
| | - Drew G Michael
- National Institute of Dental and Craniofacial Research, National Institutes of Health, NIH 10 Center Dr., Bethesda, MD, 20892, USA
| | - Noriyuki Hirata
- Division of Cancer Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Futoshi Suizu
- Division of Cancer Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Satoko Ishigaki
- Division of Cancer Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Fabiola Reis Oliveira
- Department of Clinical Medicine, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Ana Carolina F Motta
- Department of Stomatology, Public Health and Forensic Dentistry, School of Dentistry of Ribeirão Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Alfredo Ribeiro-Silva
- Department of Pathology and Legal Medicine, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Eduardo M Rocha
- Department of Ophthalmology, Otorhinolaryngology, Head and Neck Surgery, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Tatsuya Atsumi
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Masayuki Noguchi
- Division of Cancer Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - John A Chiorini
- National Institute of Dental and Craniofacial Research, National Institutes of Health, NIH 10 Center Dr., Bethesda, MD, 20892, USA.
| |
Collapse
|
15
|
Gao Y, Chen Y, Zhang Z, Yu X, Zheng J. Recent Advances in Mouse Models of Sjögren's Syndrome. Front Immunol 2020; 11:1158. [PMID: 32695097 PMCID: PMC7338666 DOI: 10.3389/fimmu.2020.01158] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 05/11/2020] [Indexed: 12/15/2022] Open
Abstract
Sjögren's syndrome (SS) is a complex rheumatoid disease that mainly affects exocrine glands, resulting in xerostomia (dry mouth) and xerophthalmia (dry eye). SS is characterized by autoantibodies, infiltration into exocrine glands, and ectopic expression of MHC II molecules on glandular epithelial cells. In contrast to the well-characterized clinical and immunological features, the etiology and pathogenesis of SS remain largely unknown. Animal models are powerful research tools for elucidating the pathogenesis of human diseases. To date, many mouse models of SS, including induced models, in which disease is induced in mice, and genetic models, in which mice spontaneously develop SS-like disease, have been established. These mouse models have provided new insight into the pathogenesis of SS. In this review, we aim to provide a comprehensive overview of recent advances in the field of experimental SS.
Collapse
Affiliation(s)
- Yunzhen Gao
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, China
| | - Yan Chen
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, China
| | - Zhongjian Zhang
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, China
| | - Xinhua Yu
- Priority Area Asthma & Allergy, Research Center Borstel, Airway Research Center North (ARCN), Members of the German Center for Lung Research (DZL), Borstel, Germany
| | - Junfeng Zheng
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
16
|
Mourão LC, Cardoso-Oliveira GP, Braga ÉM. Autoantibodies and Malaria: Where We Stand? Insights Into Pathogenesis and Protection. Front Cell Infect Microbiol 2020; 10:262. [PMID: 32596165 PMCID: PMC7300196 DOI: 10.3389/fcimb.2020.00262] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 05/04/2020] [Indexed: 12/16/2022] Open
Abstract
Autoantibodies are frequently reported in patients with malaria, but whether they contribute to protection or to pathology is an issue of debate. A large body of evidence indicates that antibodies against host-self components are associated to malaria clinical outcomes such as cerebral malaria, renal dysfunction and anemia. Nonetheless, self-reactive immunoglobulins induced during an infection can also mediate protection. In light of these controversies, we summarize here the latest findings in our understanding of autoimmune responses in malaria, focusing on Plasmodium falciparum and Plasmodium vivax. We review the main targets of self-antibody responses in malaria as well as the current, but still limited, knowledge of their role in disease pathogenesis or protection.
Collapse
Affiliation(s)
| | | | - Érika Martins Braga
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
17
|
Nakamura H, Shimizu T, Kawakami A. Role of Viral Infections in the Pathogenesis of Sjögren's Syndrome: Different Characteristics of Epstein-Barr Virus and HTLV-1. J Clin Med 2020; 9:jcm9051459. [PMID: 32414149 PMCID: PMC7290771 DOI: 10.3390/jcm9051459] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/06/2020] [Accepted: 05/06/2020] [Indexed: 12/12/2022] Open
Abstract
Viruses are possible pathogenic agents in several autoimmune diseases. Sjögren’s syndrome (SS), which involves exocrine dysfunction and the appearance of autoantibodies, shows salivary gland- and lacrimal gland-oriented clinical features. Epstein-Barr virus (EBV) is the most investigated pathogen as a candidate that directly induces the phenotype found in SS. The reactivation of the virus with various stimuli induced a dysregulated form of EBV that has the potential to infect SS-specific B cells and plasma cells that are closely associated with the function of an ectopic lymphoid structure that contains a germinal center (GC) in the salivary glands of individuals with SS. The involvement of human T-cell leukemia virus type 1 (HTLV-1) in SS has been epidemiologically established, but the disease concept of HTLV-1-associated SS remains unexplained due to limited evidence from basic research. Unlike the cell-to-cell contact between lymphocytes, biofilm-like structures are candidates as the mode of HTLV-1 infection of salivary gland epithelial cells (SGECs). HTLV-1 can infect SGECs with enhanced levels of inflammatory cytokines and chemokines that are secreted from SGECs. Regardless of the different targets that viruses have with respect to affinitive lymphocytes, viruses are involved in the formation of pathological alterations with immunological modifications in SS.
Collapse
|
18
|
Bufotalin ameliorates experimental Sjögren's syndrome development by inhibiting Th17 generation. Naunyn Schmiedebergs Arch Pharmacol 2020; 393:1977-1985. [PMID: 31950221 DOI: 10.1007/s00210-020-01817-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 01/08/2020] [Indexed: 12/13/2022]
Abstract
Chronic inflammatory autoimmune disease Sjögren's syndrome (SS) is characterized by the reduced secretion of exocrine glands, suggesting strategies targeting inflammation to be a potential option for SS therapy. Bufotalin, an active constituent of Bufadienolides, exerts potent antitumor effects with unknown effects on autoimmune diseases including SS. This study aims to investigate whether bufotalin possesses therapeutic potentials to SS and the underlying mechanisms. The experimental Sjögren's syndrome (ESS) murine model was constructed by SG-immunization and murine naïve CD4+ T cells were cultured under Th17 polarization conditions with or without low doses of bufotalin treatment. Saliva flow rate was measured, and flow cytometry was applied to analyze T cell subpopulations. ELISA was conducted to determine the levels of targeted inflammatory cytokines. Bufotalin-treated ESS mice showed higher saliva flow rates, lower serum levels of autoantibodies (anti-M3R and anti-SSA IgG), lower serum levels of pro-inflammatory cytokines, as well as lower Th17 cell population from spleens and cervical lymph nodes. Additionally, in vitro study showed that bufotalin inhibits Th17 polarization and secretion of cytokines IL-17 and IFN-γ. Bufotalin at a low dose significantly ameliorates ESS development, possibly via inhibiting pro-inflammatory Th17 population and secretion of inflammatory cytokines during ESS pathogenesis.
Collapse
|
19
|
Otsuka K, Yamada A, Saito M, Ushio A, Sato M, Kisoda S, Shao W, Tsunematsu T, Kudo Y, Arakaki R, Ishimaru N. Achaete-Scute Homologue 2–Regulated Follicular Helper T Cells Promote Autoimmunity in a Murine Model for Sjögren Syndrome. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:2414-2427. [DOI: 10.1016/j.ajpath.2019.08.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 08/12/2019] [Accepted: 08/26/2019] [Indexed: 02/06/2023]
|
20
|
Zinkevičienė A, Dumalakienė I, Mieliauskaitė D, Vilienė R, Narkevičiūtė I, Girkontaitė I. sICAM-1 as potential additional parameter in the discrimination of the Sjögren syndrome and non-autoimmune sicca syndrome: a pilot study. Clin Rheumatol 2019; 38:2803-2809. [PMID: 31152257 DOI: 10.1007/s10067-019-04621-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 05/17/2019] [Accepted: 05/21/2019] [Indexed: 12/29/2022]
Abstract
OBJECTIVE Both Sjögren's syndrome (SS) and non-autoimmune sicca syndrome (nSS) can show symptoms of dry eyes and a dry mouth, and objective reductions in tear and saliva production. Dry eyes and dry mouth are frequent but they are distinct pathological entities that require diagnostic discrimination. METHODS The aim of present study was to compare the serum levels of sICAM-1, TFF3, RANTES, adiponectin, and FGF in primary (pSS), secondary due to rheumatoid arthritis (sSS), non-autoimmune sicca syndrome (nSS), and healthy groups. The serum levels of selected molecules were determined by enzyme-linked immunosorbent assay (ELISA) in 29 patients with pSS, 30 with sSS, 17 with nSS, and 15 healthy subjects. RESULTS sICAM-1 was significantly elevated in pSS and sSS patients compared with nSS group. Levels of FGF, TFF3, and RANTES were significantly increased in pSS, sSS, and nSS patients compared with healthy controls. No significant correlations were found between the levels of measured molecules and the clinical parameters. CONCLUSIONS Our study showed that sICAM-1 might be useful as an additional parameter for differential diagnosis of SS and nSS, and TFF could be additional diagnostic marker for SS diagnosis. KEY POINTS • sICAM-1 was significantly elevated in Sjögren syndrome patients compared with non-autoimmune sicca syndrome group. • TFF was significantly elevated in Sjögren syndrome patients compared with healthy controls. • They might be useful as additional parameters for differential diagnosis.
Collapse
Affiliation(s)
- Auksė Zinkevičienė
- Department of Immunology, State Research Institute Centre for Innovative Medicine, Santariškių g. 5, LT-08406, Vilnius, Lithuania. .,Department of Experimental, Preventive and Clinical Medicine, State Research Institute Centre for Innovative Medicine, Santariškių g. 5, LT-08406, Vilnius, Lithuania.
| | - Irena Dumalakienė
- Department of Immunology, State Research Institute Centre for Innovative Medicine, Santariškių g. 5, LT-08406, Vilnius, Lithuania
| | - Diana Mieliauskaitė
- Department of Experimental, Preventive and Clinical Medicine, State Research Institute Centre for Innovative Medicine, Santariškių g. 5, LT-08406, Vilnius, Lithuania
| | - Rita Vilienė
- Department of Immunology, State Research Institute Centre for Innovative Medicine, Santariškių g. 5, LT-08406, Vilnius, Lithuania
| | - Ieva Narkevičiūtė
- Department of Immunology, State Research Institute Centre for Innovative Medicine, Santariškių g. 5, LT-08406, Vilnius, Lithuania
| | - Irutė Girkontaitė
- Department of Immunology, State Research Institute Centre for Innovative Medicine, Santariškių g. 5, LT-08406, Vilnius, Lithuania
| |
Collapse
|
21
|
Ohyama K. [Clinical Pharmaceutical Research Based on New Proteome Analysis Based on Chromatographic Separation]. YAKUGAKU ZASSHI 2019; 139:505-509. [PMID: 30930377 DOI: 10.1248/yakushi.18-00167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Comprehensive identification of antigens in immune complexes (IC-antigens) is beneficial to provide insights into pathophysiology and could form the basis for novel diagnostic and treatment strategies for many immune-related diseases. Immune complexome analysis is a method for comprehensively identifying and profiling IC-antigens in biological fluids (such as serum and cerebrospinal fluid). We applied this strategy to the analysis of circulating ICs in autoimmune diseases (rheumatoid arthritis, Sjögren's syndrome, systemic scleroderma, and systemic lupus erythematosus), infectious diseases, and cancers. Fluorogenic derivatization-liquid chromatography-tandem mass spectrometry (FD-LC-MS/MS) consists of fluorogenic derivatization of proteins, followed by HPLC of the derivatized proteins, isolation of the proteins differentially expressed in a certain group, enzymatic digestion of the isolated proteins followed by LC-tandem MS using a database-searching algorithm for protein identification. We have applied this method to understand the cardioprotective effect of pre-administration of docetaxel in adriamycin/docetaxel combination anti-cancer therapy, and the cellular processes that are affected by non-steroidal anti-inflammatory drugs (NSAIDs) in mouse stomach tissue during ulcer formation.
Collapse
Affiliation(s)
- Kaname Ohyama
- Graduate School of Biomedical Sciences, Nagasaki University
| |
Collapse
|
22
|
Saito M, Otsuka K, Ushio A, Yamada A, Arakaki R, Kudo Y, Ishimaru N. Unique Phenotypes and Functions of Follicular Helper T Cells and Regulatory T Cells in Sjögren's Syndrome. Curr Rheumatol Rev 2019; 14:239-245. [PMID: 28124612 PMCID: PMC6225342 DOI: 10.2174/1573397113666170125122858] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 01/13/2017] [Accepted: 01/23/2017] [Indexed: 02/07/2023]
Abstract
Sjogren’s syndrome (SS) is a T cell-mediated autoimmune disease of the systemic exocrine glands, such as salivary and lacrimal glands. A variety of T-cell subpopulations maintain immune tolerance in the thymus and periphery through complex immune responses including cellular and humoral immunity. The T-cell subpopulations exhibiting abnormal or unique phenotypes and impaired functionality have been reported to play important roles in the cellular mechanisms of autoimmunity in SS patients and animal models of SS. In this review, we focused on follicular helper T cells related to antibody production and regulatory T cells to control immune tolerance in the pathogenesis of SS. The unique roles of these T-cell subpopulations in the process of the onset or development of SS have been demonstrated in this review of recent publications. The clinical application of these T-cell subpopulations will be helpful for the development of new techniques for diagnosis or treatment of SS in the future.
Collapse
Affiliation(s)
- Masako Saito
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8504, Japan
| | - Kunihiro Otsuka
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8504, Japan
| | - Aya Ushio
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8504, Japan
| | - Akiko Yamada
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8504, Japan
| | - Rieko Arakaki
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8504, Japan
| | - Yasusei Kudo
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8504, Japan
| | - Naozumi Ishimaru
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8504, Japan
| |
Collapse
|
23
|
Affiliation(s)
- Toshio Odani
- Adeno-Associated Virus Biology Section, Molecular Physiology and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - John A. Chiorini
- Adeno-Associated Virus Biology Section, Molecular Physiology and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
24
|
Shimizu T, Nakamura H, Takatani A, Umeda M, Horai Y, Kurushima S, Michitsuji T, Nakashima Y, Kawakami A. Activation of Toll-like receptor 7 signaling in labial salivary glands of primary Sjögren's syndrome patients. Clin Exp Immunol 2018; 196:39-51. [PMID: 30446998 DOI: 10.1111/cei.13242] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/05/2018] [Indexed: 12/31/2022] Open
Abstract
The aim of this study was to determine the expressions of Toll-like receptors (TLRs) 7-9 and type I interferon (IFN) signal in labial salivary glands (LSGs) and cultured salivary gland epithelial cells (SGECs) from primary Sjögren's syndrome (pSS) patients. We performed an immunohistochemistry analysis of LSGs from 11 patients with pSS as defined by American-European Consensus Group classification criteria and five healthy subjects. The pSS patients' SGECs were analyzed by immunofluorescence and western blotting. IFN-α expression was examined by immunosorbent assay and flow cytometry. Mononuclear cells (MNCs) from pSS patients' LSGs showed TLR-7-dominant expression. B cells, plasma cells and plasmacytoid dendritic cells (pDCs) co-expressed with TLR-7. Myeloid differentiation primary response gene 88 (MyD88), tumor necrosis factor receptor-associated factor 6 (TRAF6) and interferon regulatory factor 7 (IRF7) co-expressed with the pDC marker CD303 in LSGs. Ducts from pSS patients dominantly expressed TLR-7, and TLR-7 in the ducts co-expressed with MyD88, TRAF6 and IRF7. Type I IFNs including IFN-α and IFN-β were detected in MNCs and ducts in pSS patients' LSGs. Increased TRAF6 expression and the nuclear translocation of IRF7 in SGECs were detected by immunofluorescence following loxoribine (a TLR-7 ligand) stimulation despite IFN-β pretreatment. Western blotting showed increased TRAF6 expression in SGECs following IFN-β and loxoribine stimulation. Although no increase in IFN-α was detected in supernatant from stimulated SGECs, the IFN-α in supernatant from stimulated peripheral blood pDCs from pSS patients was significantly increased. Our findings suggest that TLR-7 is dominantly expressed in both MNCs and ducts with downstream signals for type I IFNs, indicating that TLR7-dominant innate immunity is related to the development of sialadenitis in pSS.
Collapse
Affiliation(s)
- T Shimizu
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - H Nakamura
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - A Takatani
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - M Umeda
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Y Horai
- Clinical Research Center, National Hospital Organization Nagasaki Medical Center, Omura, Japan
| | - S Kurushima
- Department of Rheumatology, Sasebo Chuo Hospital, Sasebo, Japan
| | - T Michitsuji
- Department of General and Internal Medicine, National Hospital Organization Nagasaki Medical Center, Omura, Japan
| | - Y Nakashima
- Department of Rheumatology, Sasebo City Medical Center, Sasebo, Japan
| | - A Kawakami
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
25
|
Ushio A, Arakaki R, Otsuka K, Yamada A, Tsunematsu T, Kudo Y, Aota K, Azuma M, Ishimaru N. CCL22-Producing Resident Macrophages Enhance T Cell Response in Sjögren's Syndrome. Front Immunol 2018; 9:2594. [PMID: 30467506 PMCID: PMC6236111 DOI: 10.3389/fimmu.2018.02594] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 10/22/2018] [Indexed: 12/28/2022] Open
Abstract
Macrophages (MΦs) are critical regulators of immune response and serve as a link between innate and acquired immunity. The precise mechanism of involvement of tissue-resident MΦs in the pathogenesis of autoimmune diseases is not clear. Here, using a murine model for Sjögren's syndrome (SS), we investigated the role of tissue-resident MΦs in the onset and development of autoimmunity. Two unique populations of CD11bhigh and CD11blow resident MΦs were observed in the target tissue of the SS model. Comprehensive gene expression analysis of chemokines revealed effective production of CCL22 by the CD11bhigh MΦs. CCL22 upregulated the migratory activity of CD4+ T cells by increasing CCR4, a receptor of CCL22, on T cells in the SS model. In addition, CCL22 enhanced IFN-γ production of T cells of the SS model, thereby suggesting that CCL22 may impair the local immune tolerance in the target organ of the SS model. Moreover, administration of anti-CCL22 antibody suppressed autoimmune lesions in the SS model. Finally, histopathological analysis revealed numerous CCL22-producing MΦs in the minor salivary gland tissue specimens of the SS patients. CCL22-producing tissue-resident MΦs may control autoimmune lesions by enhancing T cell response in the SS model. These results suggest that specific chemokines and their receptors may serve as novel therapeutic or diagnostic targets for SS.
Collapse
Affiliation(s)
- Aya Ushio
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Rieko Arakaki
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Kunihiro Otsuka
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Akiko Yamada
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Takaaki Tsunematsu
- Department of Pathology and Laboratory Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Yasusei Kudo
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Keiko Aota
- Department of Oral Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Masayuki Azuma
- Department of Oral Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Naozumi Ishimaru
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| |
Collapse
|
26
|
Free fatty acids may be involved in the pathogenesis of oral-related and cardiovascular diseases. J Oral Biosci 2018. [DOI: 10.1016/j.job.2018.04.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
27
|
Modulation of Apoptosis by Cytotoxic Mediators and Cell-Survival Molecules in Sjögren's Syndrome. Int J Mol Sci 2018; 19:ijms19082369. [PMID: 30103522 PMCID: PMC6121505 DOI: 10.3390/ijms19082369] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 08/08/2018] [Accepted: 08/10/2018] [Indexed: 12/11/2022] Open
Abstract
The pathogenesis of Sjögren’s syndrome (SS) involves multiple factors including genetic background, cell death, and exocrine dysfunction. We here discuss apoptotic control in exocrine glands in SS by showing various pro- and anti-apoptotic pathways. Although the membrane-bound and soluble form of the Fas/Fas ligand system is a leading player with activation of the death domain and caspase 8/3 cleavage, the role of soluble Fas/FasL (including its polymorphism) in apoptosis is controversial. The tumor necrosis factor related apoptosis-inducing ligand (TRAIL)-mediated apoptosis of salivary gland epithelial cells (SGECs) involves a mitochondrial pathway that includes caspase 9 cleavage. The involvement of innate immunity cells such as toll-like receptors (TLRs) has been investigated; TLR2-4 and TLR7-9 are associated with the induction of inflammation in exocrine glands of SS patients. TLR3 has the potential to induce the apoptosis of SS patients’ SGECs. Linkage of epidermal growth factor (EGF) was shown in exocrine glands in SS, and it inhibited the Fas/FasL system with the help of cell-survival factors. TLR3 has dual actions to cause inflammation as well as apoptosis, which are inhibited by EGF. In conclusion, apoptosis in exocrine glands of SS patients is tightly controlled by balance of pro-apoptotic signals and growth factor.
Collapse
|
28
|
Minagi HO, Ikai K, Araie T, Sakai M, Sakai T. Benefits of long-term pilocarpine due to increased muscarinic acetylcholine receptor 3 in salivary glands. Biochem Biophys Res Commun 2018; 503:1098-1102. [PMID: 29953856 DOI: 10.1016/j.bbrc.2018.06.125] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 06/21/2018] [Indexed: 01/10/2023]
Abstract
Hypofunction of the salivary gland causes several life-disrupting side effects such as dental caries, oral candidiasis, loss of taste, and swallowing disorders. No satisfactory therapy has been established to treat salivary hypofunction. Pilocarpine represents a potential treatment for dry mouth due to Sjögren's syndrome (SS). Although subjective improvement was consistently observed with pilocarpine therapy, the mechanism was unclear. In this study, we investigated the mechanism of recovery in salivation following treatment with pilocarpine. We first examined the effectiveness of pilocarpine in SS patients as quantified by the Saxon test and the visual analogue scale average. We found that salivation ability and subjective symptoms improved by continuous administration of pilocarpine. These results demonstrated that long-term medication for dry mouth patients was more effective. However, as the mechanism remained unclear, molecular biological mechanisms were analyzed based on the effects of continuous administration of pilocarpine using model mice. In the molecular biological analysis, continuous administration of pilocarpine was effective in both ICR and SS model mice. Gene and protein expression of muscarinic acetylcholine receptor 3 (M3R) increased in salivary glands following continuous administration of pilocarpine compared with single administration. Therefore, continuous administration of pilocarpine effectively induced M3R expression, thereby activating salivation.
Collapse
Affiliation(s)
- Hitomi Ono Minagi
- Department of Oral-facial Disorders, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Kazuki Ikai
- Department of Oral-facial Disorders, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Takafumi Araie
- Department of Oral-facial Disorders, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Manabu Sakai
- Division for Clinical Laboratory, Osaka University Dental Hospital, Osaka, Japan
| | - Takayoshi Sakai
- Department of Oral-facial Disorders, Osaka University Graduate School of Dentistry, Osaka, Japan.
| |
Collapse
|
29
|
MicroRNA in Sjögren's Syndrome: Their Potential Roles in Pathogenesis and Diagnosis. J Immunol Res 2018; 2018:7510174. [PMID: 29977932 PMCID: PMC6011049 DOI: 10.1155/2018/7510174] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 05/20/2018] [Indexed: 12/23/2022] Open
Abstract
Sjögren's syndrome (SS) or sicca syndrome was described by Swedish ophthalmologist Sjögren in the year 1933 for the first time. The etiology of the SS is multifunctional and includes a combination of genetic predisposition and environmental as well as epigenetic factors. It is an autoimmune disease characterized by features of systemic autoimmunity, dysfunction, and inflammation in the exocrine glands (mainly salivary and lacrimal glands) and lymphocytic infiltration of exocrine glands. In fact, the involvement of lacrimal and salivary glands results in the typical features of dry eye and salivary dysfunction (xerostomia). Only in one-third of the patients also present systemic extraglandular manifestations. T cells were originally considered to play the initiating role in the autoimmune process, while B cells were restricted to autoantibody production. In recent years, it is understood that the roles of B cells are multiple. Moreover, autoantibodies and blood B cell analysis are major contributors to a clinical diagnosis of Sjögren's syndrome. Recently, there has been rising interest in microRNA implication in autoimmunity. Unfortunately, to date, there are only a few studies that have investigated their participation in SS etiopathogenesis. The purpose of this work is to gather the data present in the literature to clarify this complex topic.
Collapse
|
30
|
Immune Privilege and Eye-Derived T-Regulatory Cells. J Immunol Res 2018; 2018:1679197. [PMID: 29888291 PMCID: PMC5985108 DOI: 10.1155/2018/1679197] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 04/18/2018] [Indexed: 02/08/2023] Open
Abstract
Certain cellular components of the eye, such as neural retina, are unable to regenerate and replicate after destructive inflammation. Ocular immune privilege provides the eye with immune protection against intraocular inflammation in order to minimize the risk to vision integrity. The eye and immune system use strategies to maintain the ocular immune privilege by regulating the innate and adaptive immune response, which includes immunological ignorance, peripheral tolerance to eye-derived antigens, and intraocular immunosuppressive microenvironment. In this review, we summarize current knowledge regarding the molecular mechanism responsible for the development and maintenance of ocular immune privilege via regulatory T cells (Tregs), which are generated by the anterior chamber-associated immune deviation (ACAID), and ocular resident cells including corneal endothelial (CE) cells, ocular pigment epithelial (PE) cells, and aqueous humor. Furthermore, we examined the therapeutic potential of Tregs generated by RPE cells that express transforming growth factor beta (TGF-β), cytotoxic T lymphocyte-associated antigen-2 alpha (CTLA-2α), and retinoic acid for autoimmune uveoretinitis and evaluated a new strategy using human RPE-induced Tregs for clinical application in inflammatory ocular disease. We believe that a better understanding of the ocular immune privilege associated with Tregs might offer a new approach with regard to therapeutic interventions for ocular autoimmunity.
Collapse
|
31
|
Pathological Analysis of Ocular Lesions in a Murine Model of Sjögren's Syndrome. Int J Mol Sci 2017; 18:ijms18061209. [PMID: 28587293 PMCID: PMC5486032 DOI: 10.3390/ijms18061209] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 05/27/2017] [Accepted: 06/03/2017] [Indexed: 01/22/2023] Open
Abstract
Sjögren’s syndrome (SS) is a systemic autoimmune disease characterized by severe inflammation of exocrine glands such as the salivary and lacrimal glands. When it affects the lacrimal glands, many patients experience keratoconjunctivitis due to severely dry eyes. This study investigated the pathological and immunological characteristics of ocular lesions in a mouse model of SS. Corneal epithelial injury and hyperplasia were confirmed pathologically. The number of conjunctival mucin-producing goblet cells was significantly decreased in the SS model mice compared with control mice. Expression levels of transforming growth factor (TGF)-β, interleukin (IL)-6, tumor necrosis factor (TNF)-α, and C-X-C motif chemokine (CXCL) 12 were significantly higher in the corneal epithelium of the SS model mice than in control mice. Inflammatory lesions were observed in the Harderian, intraorbital, and extraorbital lacrimal glands in the SS model mice, suggesting that the ocular glands were targeted by an autoimmune response. The lacrimal glands of the SS model mice were infiltrated by cluster of differentiation (CD)4+ T cells. Real-time reverse transcription-polymerase chain reaction (RT-PCR) revealed significantly increased mRNA expression of TNF-α, TGF-β, CXCL9, and lysozyme in the extraorbital lacrimal glands of the SS model mice compared with control mice. These results add to the understanding of the complex pathogenesis of SS and may facilitate development of new therapeutic strategies.
Collapse
|
32
|
Gimenez-Molina Y, Villanueva J, Nanclares C, Lopez-Font I, Viniegra S, Francés MDM, Gandia L, Gil A, Gutiérrez LM. The Differential Organization of F-Actin Alters the Distribution of Organelles in Cultured When Compared to Native Chromaffin Cells. Front Cell Neurosci 2017; 11:135. [PMID: 28522964 PMCID: PMC5415619 DOI: 10.3389/fncel.2017.00135] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 04/21/2017] [Indexed: 11/17/2022] Open
Abstract
Cultured bovine chromaffin cells have been used extensively as a neuroendocrine model to study regulated secretion. In order to extend such experimental findings to the physiological situation, it is necessary to study mayor cellular structures affecting secretion in cultured cells with their counterparts present in the adrenomedullary tissue. F-actin concentrates in a peripheral ring in cultured cells, as witnessed by phalloidin–rodhamine labeling, while extends throughout the cytoplasm in native cells. This result is also confirmed when studying the localization of α-fodrin, a F-actin-associated protein. Furthermore, as a consequence of this redistribution of F-actin, we observed that chromaffin granules and mitochondria located into two different cortical and internal populations in cultured cells, whereas they are homogeneously distributed throughout the cytoplasm in the adrenomedullary tissue. Nevertheless, secretion from isolated cells and adrenal gland pieces is remarkably similar when measured by amperometry. Finally, we generate mathematical models to consider how the distribution of organelles affects the secretory kinetics of intact and cultured cells. Our results imply that we have to consider F-actin structural changes to interpret functional data obtained in cultured neuroendocrine cells.
Collapse
Affiliation(s)
- Yolanda Gimenez-Molina
- Instituto de Neurociencias, Centro Mixto del Consejo Superior de Investigaciones Científicas, Universidad Miguel HernándezAlicante, Spain
| | - José Villanueva
- Instituto de Neurociencias, Centro Mixto del Consejo Superior de Investigaciones Científicas, Universidad Miguel HernándezAlicante, Spain
| | - Carmen Nanclares
- Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de MadridMadrid, Spain
| | - Inmaculada Lopez-Font
- Instituto de Neurociencias, Centro Mixto del Consejo Superior de Investigaciones Científicas, Universidad Miguel HernándezAlicante, Spain.,Centro de Investigación Biomédica en Red de Enfermedades NeurodegenerativasAlicante, Spain
| | - Salvador Viniegra
- Instituto de Neurociencias, Centro Mixto del Consejo Superior de Investigaciones Científicas, Universidad Miguel HernándezAlicante, Spain
| | - Maria Del Mar Francés
- Instituto de Neurociencias, Centro Mixto del Consejo Superior de Investigaciones Científicas, Universidad Miguel HernándezAlicante, Spain
| | - Luis Gandia
- Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de MadridMadrid, Spain
| | - Amparo Gil
- Department Matemática Aplicada y Ciencias de la Computación, Universidad de CantabriaSantander, Spain
| | - Luis M Gutiérrez
- Instituto de Neurociencias, Centro Mixto del Consejo Superior de Investigaciones Científicas, Universidad Miguel HernándezAlicante, Spain
| |
Collapse
|
33
|
Shikama Y, Kudo Y, Ishimaru N, Funaki M. Potential Role of Free Fatty Acids in the Pathogenesis of Periodontitis and Primary Sjögren's Syndrome. Int J Mol Sci 2017; 18:ijms18040836. [PMID: 28420093 PMCID: PMC5412420 DOI: 10.3390/ijms18040836] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 04/07/2017] [Accepted: 04/11/2017] [Indexed: 12/15/2022] Open
Abstract
Clinical studies have shown that metabolic disorders such as type 2 diabetes and dyslipidemia are associated with increased risk of oral-related diseases, such as periodontitis and Sjögren’s syndrome. Although changes in the immune system are critical in both of these metabolic disorders and oral-related diseases, the mechanism underlying the interaction between these diseases remains largely unknown. Obesity and type 2 diabetes are known to be associated with higher concentrations of free fatty acids in blood. Among free fatty acids, saturated fatty acids such as palmitic acid have been demonstrated to induce inflammatory responses mainly via the innate immune systems, and to be involved in the pathogenesis of type 2 diabetes in tissues such as adipose tissue, liver, pancreas, and skeletal muscle. Here, we highlight recent advances in evidence for the potential involvement of palmitic acid in the pathogenesis of periodontitis and Sjögren’s syndrome, and discuss the possibility that improvement of the lipid profile could be a new strategy for the treatment of these diseases.
Collapse
Affiliation(s)
- Yosuke Shikama
- Department of Oral Disease Research, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu 474-8511, Japan.
| | - Yasusei Kudo
- Department of Oral Molecular Pathology, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8504, Japan.
| | - Naozumi Ishimaru
- Department of Oral Molecular Pathology, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8504, Japan.
| | - Makoto Funaki
- Clinical Research Center for Diabetes, Tokushima University Hospital, 2-50-1 Kuramoto-cho, Tokushima 770-8503, Japan.
| |
Collapse
|
34
|
Shen L, Suresh L. Autoantibodies, detection methods and panels for diagnosis of Sjögren's syndrome. Clin Immunol 2017; 182:24-29. [PMID: 28390965 DOI: 10.1016/j.clim.2017.03.017] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 03/31/2017] [Indexed: 01/10/2023]
Abstract
The presence of autoantibodies is one of several hallmarks of Sjögren's Syndrome, the detection of serum autoantibodies has a central role in the diagnosis and classification of Sjögren's syndrome. In this review, we will discuss autoantibodies that are helpful in the diagnosis of Sjögren's syndrome. This includes the traditional autoantibodies for disease classification (ANA, Anti-Ro/SSA, Anti-La/SSB, RF), autoantibodies identified from mouse models (Anti-SP1, Anti- PSP, Anti-CA6, and anti-alpha fodrin) and autoantibodies associated with other autoimmune disease (ACA, AMA, and Anti-CCP). We will also review the methods for the detection of autoantibodies and associated challenges for clinical results reporting. The significance of using an autoantibody panel for the diagnosis of SS will be also be reviewed.
Collapse
Affiliation(s)
- Long Shen
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen 361003, China; Autoimmune Division, Trinity Biotech, 60 Pineview Drive, Buffalo, NY 14228, USA.
| | - Lakshmanan Suresh
- Department of Oral Diagnostic Sciences, School of Dental Medicine, University at Buffalo, Buffalo, NY 14214, USA; Autoimmune Division, Trinity Biotech, 60 Pineview Drive, Buffalo, NY 14228, USA
| |
Collapse
|
35
|
Ohyama K. [Screening for Potential Drug Targets by Comprehensive Identification of Disease-specific Antigens Incorporated into Immune Complexes in Patients with Immunological Diseases]. YAKUGAKU ZASSHI 2016; 136:157-61. [PMID: 26831786 DOI: 10.1248/yakushi.15-00226-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Our immune system resembles an intelligent security system, which continually monitors for foreign invaders (infectious diseases); however, in some cases, this system recognizes healthy parts as something harmful or foreign and then attacks them (autoimmune diseases). The defining characteristics of an autoimmune disease are the existence of T- and B-cell autoreactivity against self proteins (autoantigens). In addition to autoimmune diseases, aberrant host proteins that occur during a certain state of diseases (e.g., cancer) can be recognized as an autoantigen. Immune complexes (ICs) are produced during an immune response and may reflect some aspects of an ongoing immune response. Therefore, the identity of antigens incorporated into ICs provides the information that in the future may aid in the development of diagnosis and treatment strategies for autoimmune diseases, infection, cancer, and transplantation therapy, and this information might be more relevant than information on free antigens. We developed a novel proteomic strategy (immune complexome analysis) in which ICs are separated from serum, followed by direct tryptic digestion and nano-liquid chromatography-tandem mass spectrometry for the identification and profiling of antigens in circulating ICs. We applied this strategy to the analysis of circulating ICs in autoimmune diseases (rheumatoid arthritis, anti-neutrophil cytoplasmic antibody-associated vasculitis, Takayasu's arteritis, mixed connective tissue disease, dermatomyositis, Sjögren's syndrome, systemic scleroderma, and systemic lupus erythematosus), infectious diseases and cancers. In this review, we mainly discuss the results for autoimmune diseases.
Collapse
Affiliation(s)
- Kaname Ohyama
- Graduate School of Biomedical Sciences, Nagasaki University
| |
Collapse
|
36
|
Hernández-Molina G, Nuñez-Alvarez C, Avila-Casado C, Llorente L, Hernández-Hernández C, Calderillo ML, Marroquín V, Recillas-Gispert C, Romero-Díaz J, Sánchez-Guerrero J. Usefulness of IgA Anti-α-fodrin Antibodies in Combination with Rheumatoid Factor and/or Antinuclear Antibodies as Substitute Immunological Criterion in Sjögren Syndrome with Negative Anti-SSA/SSB Antibodies. J Rheumatol 2016; 43:1852-1857. [PMID: 27481899 DOI: 10.3899/jrheum.151315] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/20/2016] [Indexed: 11/22/2022]
Abstract
OBJECTIVE We aimed to evaluate the usefulness of anti-α-fodrin antibodies (AFA) in combination with rheumatoid factor (RF) and/or antinuclear antibodies (ANA) as an alternative immunological criterion for Sjögren syndrome (SS) among patients with negative anti-Ro/La serology. METHODS The study included 350 patients (100 with rheumatoid arthritis, systemic lupus erythematosus, and systemic sclerosis, and 50 with primary SS) randomly selected and assessed for SS. All patients were tested for ANA, RF, anti-SSA/SSB, and AFA antibodies. SS diagnosis was made on a clinical basis by 2 rheumatologists based on the 6-item screening questionnaire, Schirmer-I test, nonstimulated whole salivary flow rate, fluorescein staining test, autoantibodies, lip biopsy, and medical chart review. Non-SS was defined as lack of clinical diagnosis and not fulfilling the American-European Consensus Group classification criteria and the American College of Rheumatology (ACR) criteria. The ACR criteria were applied substituting the immunological criteria as follows: (1) RF plus ANA > 1:320, (2) RF plus AFA, (3) ANA > 1:320 plus AFA, (4) RF alone, and (5) 2 positive tests out of RF, ANA > 1:320, or AFA. We estimated the sensitivity, specificity, positive predictive value, negative predictive value, and likelihood ratio positivity with 95% CI for each criterion. RESULTS There were 236 patients (67%) who tested negative for anti-SSA/SSB antibodies, of whom 65 (27.5%) were clinically diagnosed as SS, and 149 (63%) with non-SS. RF + AFA and ANA + AFA performed similarly to RF + ANA > 1:320. The model 2 out of 3 of RF, ANA, or AFA improved the sensitivity from 56.9% to 70.7%, although the specificity decreased. CONCLUSION The combination AFA + RF, AFA + ANA > 1:320, or at least 2 out of 3, performed well as a proxy immunological test for patients with SS and negative Ro/La serology.
Collapse
Affiliation(s)
- Gabriela Hernández-Molina
- From the Department of Immunology and Rheumatology, the Dental Service, and the Ophthalmology Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico; Department of Pathology, University Health Network, Toronto General Hospital, and Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada.G. Hernández-Molina, MS, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Nuñez-Alvarez, PhD, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Avila-Casado, PhD, Department of Pathology, University Health Network, Toronto General Hospital, University of Toronto; L. Llorente, MD, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Hernández-Hernández, MD, Dental Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; M.L. Calderillo, MD, Dental Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; V. Marroquín, MD, Dental Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Recillas-Gispert, MD, Ophthalmology Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; J. Romero-Díaz, MS, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; J. Sánchez-Guerrero, MS, Mount Sinai Hospital and University Health Network, University of Toronto
| | - Carlos Nuñez-Alvarez
- From the Department of Immunology and Rheumatology, the Dental Service, and the Ophthalmology Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico; Department of Pathology, University Health Network, Toronto General Hospital, and Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada.G. Hernández-Molina, MS, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Nuñez-Alvarez, PhD, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Avila-Casado, PhD, Department of Pathology, University Health Network, Toronto General Hospital, University of Toronto; L. Llorente, MD, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Hernández-Hernández, MD, Dental Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; M.L. Calderillo, MD, Dental Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; V. Marroquín, MD, Dental Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Recillas-Gispert, MD, Ophthalmology Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; J. Romero-Díaz, MS, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; J. Sánchez-Guerrero, MS, Mount Sinai Hospital and University Health Network, University of Toronto
| | - Carmen Avila-Casado
- From the Department of Immunology and Rheumatology, the Dental Service, and the Ophthalmology Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico; Department of Pathology, University Health Network, Toronto General Hospital, and Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada.G. Hernández-Molina, MS, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Nuñez-Alvarez, PhD, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Avila-Casado, PhD, Department of Pathology, University Health Network, Toronto General Hospital, University of Toronto; L. Llorente, MD, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Hernández-Hernández, MD, Dental Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; M.L. Calderillo, MD, Dental Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; V. Marroquín, MD, Dental Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Recillas-Gispert, MD, Ophthalmology Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; J. Romero-Díaz, MS, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; J. Sánchez-Guerrero, MS, Mount Sinai Hospital and University Health Network, University of Toronto
| | - Luis Llorente
- From the Department of Immunology and Rheumatology, the Dental Service, and the Ophthalmology Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico; Department of Pathology, University Health Network, Toronto General Hospital, and Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada.G. Hernández-Molina, MS, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Nuñez-Alvarez, PhD, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Avila-Casado, PhD, Department of Pathology, University Health Network, Toronto General Hospital, University of Toronto; L. Llorente, MD, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Hernández-Hernández, MD, Dental Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; M.L. Calderillo, MD, Dental Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; V. Marroquín, MD, Dental Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Recillas-Gispert, MD, Ophthalmology Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; J. Romero-Díaz, MS, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; J. Sánchez-Guerrero, MS, Mount Sinai Hospital and University Health Network, University of Toronto
| | - Carlos Hernández-Hernández
- From the Department of Immunology and Rheumatology, the Dental Service, and the Ophthalmology Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico; Department of Pathology, University Health Network, Toronto General Hospital, and Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada.G. Hernández-Molina, MS, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Nuñez-Alvarez, PhD, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Avila-Casado, PhD, Department of Pathology, University Health Network, Toronto General Hospital, University of Toronto; L. Llorente, MD, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Hernández-Hernández, MD, Dental Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; M.L. Calderillo, MD, Dental Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; V. Marroquín, MD, Dental Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Recillas-Gispert, MD, Ophthalmology Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; J. Romero-Díaz, MS, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; J. Sánchez-Guerrero, MS, Mount Sinai Hospital and University Health Network, University of Toronto
| | - María Luisa Calderillo
- From the Department of Immunology and Rheumatology, the Dental Service, and the Ophthalmology Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico; Department of Pathology, University Health Network, Toronto General Hospital, and Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada.G. Hernández-Molina, MS, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Nuñez-Alvarez, PhD, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Avila-Casado, PhD, Department of Pathology, University Health Network, Toronto General Hospital, University of Toronto; L. Llorente, MD, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Hernández-Hernández, MD, Dental Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; M.L. Calderillo, MD, Dental Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; V. Marroquín, MD, Dental Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Recillas-Gispert, MD, Ophthalmology Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; J. Romero-Díaz, MS, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; J. Sánchez-Guerrero, MS, Mount Sinai Hospital and University Health Network, University of Toronto
| | - Verónica Marroquín
- From the Department of Immunology and Rheumatology, the Dental Service, and the Ophthalmology Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico; Department of Pathology, University Health Network, Toronto General Hospital, and Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada.G. Hernández-Molina, MS, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Nuñez-Alvarez, PhD, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Avila-Casado, PhD, Department of Pathology, University Health Network, Toronto General Hospital, University of Toronto; L. Llorente, MD, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Hernández-Hernández, MD, Dental Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; M.L. Calderillo, MD, Dental Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; V. Marroquín, MD, Dental Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Recillas-Gispert, MD, Ophthalmology Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; J. Romero-Díaz, MS, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; J. Sánchez-Guerrero, MS, Mount Sinai Hospital and University Health Network, University of Toronto
| | - Claudia Recillas-Gispert
- From the Department of Immunology and Rheumatology, the Dental Service, and the Ophthalmology Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico; Department of Pathology, University Health Network, Toronto General Hospital, and Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada.G. Hernández-Molina, MS, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Nuñez-Alvarez, PhD, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Avila-Casado, PhD, Department of Pathology, University Health Network, Toronto General Hospital, University of Toronto; L. Llorente, MD, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Hernández-Hernández, MD, Dental Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; M.L. Calderillo, MD, Dental Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; V. Marroquín, MD, Dental Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Recillas-Gispert, MD, Ophthalmology Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; J. Romero-Díaz, MS, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; J. Sánchez-Guerrero, MS, Mount Sinai Hospital and University Health Network, University of Toronto
| | - Juanita Romero-Díaz
- From the Department of Immunology and Rheumatology, the Dental Service, and the Ophthalmology Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico; Department of Pathology, University Health Network, Toronto General Hospital, and Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada.G. Hernández-Molina, MS, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Nuñez-Alvarez, PhD, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Avila-Casado, PhD, Department of Pathology, University Health Network, Toronto General Hospital, University of Toronto; L. Llorente, MD, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Hernández-Hernández, MD, Dental Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; M.L. Calderillo, MD, Dental Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; V. Marroquín, MD, Dental Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Recillas-Gispert, MD, Ophthalmology Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; J. Romero-Díaz, MS, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; J. Sánchez-Guerrero, MS, Mount Sinai Hospital and University Health Network, University of Toronto
| | - Jorge Sánchez-Guerrero
- From the Department of Immunology and Rheumatology, the Dental Service, and the Ophthalmology Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico; Department of Pathology, University Health Network, Toronto General Hospital, and Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada.G. Hernández-Molina, MS, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Nuñez-Alvarez, PhD, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Avila-Casado, PhD, Department of Pathology, University Health Network, Toronto General Hospital, University of Toronto; L. Llorente, MD, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Hernández-Hernández, MD, Dental Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; M.L. Calderillo, MD, Dental Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; V. Marroquín, MD, Dental Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; C. Recillas-Gispert, MD, Ophthalmology Service, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; J. Romero-Díaz, MS, Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; J. Sánchez-Guerrero, MS, Mount Sinai Hospital and University Health Network, University of Toronto.
| |
Collapse
|
37
|
Uomori K, Nozawa K, Ikeda K, Doe K, Yamada Y, Yamaguchi A, Fujishiro M, Kawasaki M, Morimoto S, Takamori K, Sekigawa I, Chan EKL, Takasaki Y. A re-evaluation of anti-NA-14 antibodies in patients with primary Sjögren's syndrome: Significant role of interferon-γ in the production of autoantibodies against NA-14. Autoimmunity 2016; 49:347-56. [PMID: 27328271 DOI: 10.1080/08916934.2016.1196676] [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] [Indexed: 10/21/2022]
Abstract
Novel autoantibodies against nuclear antigen of 14 kDa (NA-14)/Sjögren's syndrome nuclear antigen-1 (SSNA-1) are predominantly recognized in sera of patients with primary Sjögren's syndrome (pSS). However, the detailed characteristics of the anti-NA-14 antibody remain unknown. Here, we sought to clarify the characteristics of anti-SSNA-1/NA-14 antibodies and the mechanisms of autoantibody production using sera from patients with connective tissue diseases (including pSS), autoimmune sera reacting with standard autoantigens (SS-A/Ro and/or SS-B/La, ds DNA, Scl-70 and Jo-1), and normal healthy controls (NHCs). Anti-NA-14 antibodies were predominantly recognized in sera from patients with pSS and in autoimmune sera reacting with thSS-A/Ro and/or -SS-B/Lo. Indirect immunofluorescence analysis showed that NA-14 was strongly expressed in mitotic-phase cells. Patients with pSS having anti-NA-14 antibodies exhibited significant elevation of serum IP-10 and BAFF compared to that in patients with pSS without anti-NA-14 antibodies and NHCs. Thus, our data demonstrated that anti-NA-14 antibodies could be classified as novel autoantibodies reacting with mitosis-related autoantigens predominantly recognized in pSS. Moreover, interferon-γ played an important role in the production of anti-NA-14 autoantibodies as patients with pSS having anti-NA-14 antibodies exhibited increased serum levels of IP-10 and BAFF.
Collapse
Affiliation(s)
- Kaori Uomori
- a Department of Internal Medicine and Rheumatology , Faculty of Medicine, Juntendo University , Tokyo , Japan
| | - Kazuhisa Nozawa
- a Department of Internal Medicine and Rheumatology , Faculty of Medicine, Juntendo University , Tokyo , Japan
| | - Keigo Ikeda
- b Juntendo University Urayasu Hospital, Juntendo University Graduate School of Medicine , Urayasu , Japan , and
| | - Kentaro Doe
- a Department of Internal Medicine and Rheumatology , Faculty of Medicine, Juntendo University , Tokyo , Japan
| | - Yusuke Yamada
- a Department of Internal Medicine and Rheumatology , Faculty of Medicine, Juntendo University , Tokyo , Japan
| | - Ayako Yamaguchi
- a Department of Internal Medicine and Rheumatology , Faculty of Medicine, Juntendo University , Tokyo , Japan
| | - Maki Fujishiro
- b Juntendo University Urayasu Hospital, Juntendo University Graduate School of Medicine , Urayasu , Japan , and
| | - Mikiko Kawasaki
- b Juntendo University Urayasu Hospital, Juntendo University Graduate School of Medicine , Urayasu , Japan , and
| | - Shinji Morimoto
- b Juntendo University Urayasu Hospital, Juntendo University Graduate School of Medicine , Urayasu , Japan , and
| | - Kenji Takamori
- b Juntendo University Urayasu Hospital, Juntendo University Graduate School of Medicine , Urayasu , Japan , and
| | - Iwao Sekigawa
- b Juntendo University Urayasu Hospital, Juntendo University Graduate School of Medicine , Urayasu , Japan , and
| | - Edward K L Chan
- c Department of Oral Biology , University of Florida , Gainesville , FL , USA
| | - Yoshinari Takasaki
- a Department of Internal Medicine and Rheumatology , Faculty of Medicine, Juntendo University , Tokyo , Japan
| |
Collapse
|
38
|
Park YS, Gauna AE, Cha S. Mouse Models of Primary Sjogren's Syndrome. Curr Pharm Des 2016; 21:2350-64. [PMID: 25777752 DOI: 10.2174/1381612821666150316120024] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 03/13/2015] [Indexed: 01/03/2023]
Abstract
Sjogren's syndrome (SjS) is a chronic autoimmune disorder characterized by immune cell infiltration and progressive injury to the salivary and lacrimal glands. As a consequence, patients with SjS develop xerostomia (dry mouth) and keratoconjunctivitis sicca (dry eyes). SjS is the third most common rheumatic autoimmune disorder, affecting 4 million Americans with over 90% of patients being female. Current diagnostic criteria for SjS frequently utilize histological examinations of minor salivary glands for immune cell foci, serology for autoantibodies, and dry eye evaluation by corneal or conjunctival staining. SjS can be classified as primary or secondary SjS, depending on whether it occurs alone or in association with other systemic rheumatic conditions, respectively. Clinical manifestations typically become apparent when the disease is relatively advanced in SjS patients, which poses a challenge for early diagnosis and treatment of SjS. Therefore, SjS mouse models, because of their close resemblance to the human SjS, have been extremely valuable to identify early disease markers and to investigate underlying biological and immunological dysregulations. However, it is important to bear in mind that no single mouse model has duplicated all aspects of SjS pathogenesis and clinical features, mainly due to the multifactorial etiology of SjS that includes numerous susceptibility genes and environmental factors. As such, various mouse models have been developed in the field to try to recapitulate SjS. In this review, we focus on recent mouse models of primary SjS xerostomia and describe them under three categories of spontaneous, genetically engineered, and experimentally induced models. In addition, we discuss future perspectives highlighting pros and cons of utilizing mouse models and current demands for improved models.
Collapse
Affiliation(s)
| | | | - Seunghee Cha
- Department of Oral and Maxillofacial Diagnostic Sciences, University of Florida College of Dentistry, Gainesville, FL32610, USA.
| |
Collapse
|
39
|
Gramlich OW, Teister J, Neumann M, Tao X, Beck S, von Pein HD, Pfeiffer N, Grus FH. Immune response after intermittent minimally invasive intraocular pressure elevations in an experimental animal model of glaucoma. J Neuroinflammation 2016; 13:82. [PMID: 27090083 PMCID: PMC4836145 DOI: 10.1186/s12974-016-0542-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 04/07/2016] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Elevated intraocular pressure (IOP), as well as fluctuations in IOP, is a main risk factor for glaucoma, but its pathogenic effect has not yet been clarified. Beyond the multifactorial pathology of the disease, autoimmune mechanisms seem to be linked to retinal ganglion cell (RGC) death. This study aimed to identify if intermittent IOP elevations in vivo (i) elicit neurodegeneration, (ii) provokes an immune response and (iii) whether progression of RGC loss can be attenuated by the B lymphocyte inhibitor Belimumab. METHODS Using an intermittent ocular hypertension model (iOHT), Long Evans rats (n = 21) underwent 27 unilateral simulations of a fluctuating pressure profile. Nine of these animals received Belimumab, and additional seven rats served as normotensive controls. Axonal density was analyzed in PPD-stained optic nerve cross-sections. Retinal cross-sections were immunostained against Brn3a, Iba1, and IgG autoantibody depositions. Serum IgG concentration and IgG reactivities were determined using ELISA and protein microarrays. Data was analyzed using ANOVA and Tukey HSD test (unequal N) or student's independent t test by groups. RESULTS A wavelike IOP profile led to a significant neurodegeneration of optic nerve axons (-10.6 %, p < 0.001) and RGC (-19.5 %, p = 0.02) in iOHT eyes compared with fellow eyes. Belimumab-treated animals only showed slightly higher axonal survival and reduced serum IgG concentration (-29 %) after iOHT. Neuroinflammatory events, indicated by significantly upregulated microglia activation and IgG autoantibody depositions, were shown in all injured retinas. Significantly elevated serum autoantibody immunoreactivities against glutathione-S-transferase, spectrin, and transferrin were observed after iOHT and were negatively correlated to the axon density. CONCLUSIONS Intermittent IOP elevations are sufficient to provoke neurodegeneration in the optic nerve and the retina and elicit changes of IgG autoantibody reactivities. Although the inhibition of B lymphocyte activation failed to ameliorate axonal survival, the correlation between damage and changes in the autoantibody reactivity suggests that autoantibody profiling could be useful as a biomarker for glaucoma.
Collapse
Affiliation(s)
- Oliver W Gramlich
- Experimental Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany.,Glaucoma Cell Biology Laboratory, Department of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, IA, 62242, USA
| | - Julia Teister
- Experimental Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Mareike Neumann
- Experimental Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Xue Tao
- Experimental Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Sabine Beck
- Experimental Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Harald D von Pein
- Department of Neuropathology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, Mainz, 55131, Germany
| | - Norbert Pfeiffer
- Experimental Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Franz H Grus
- Experimental Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany.
| |
Collapse
|
40
|
Xerostomia: current streams of investigation. Oral Surg Oral Med Oral Pathol Oral Radiol 2016; 122:53-60. [PMID: 27189896 DOI: 10.1016/j.oooo.2016.03.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 01/13/2016] [Accepted: 03/04/2016] [Indexed: 12/12/2022]
Abstract
Xerostomia is the subjective feeling of dry mouth, and it is often related to salivary hypofunction. Besides medication-related salivary hypofunction, Sjögren syndrome and head-and-neck radiation are two common etiologies that have garnered considerable attention. Approaches to treating and/or preventing salivary hypofunction in patients with these conditions will likely incorporate gene therapy, stem cell therapy, and tissue engineering. Advances in these disciplines are central to current research in the cure for xerostomia and will be key to eventual treatment.
Collapse
|
41
|
Primary Sjögren's syndrome. Best Pract Res Clin Rheumatol 2016; 30:189-220. [DOI: 10.1016/j.berh.2016.04.003] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Revised: 04/04/2016] [Accepted: 04/09/2016] [Indexed: 12/13/2022]
|
42
|
Yamada A, Ushio A, Arakaki R, Tsunematsu T, Kudo Y, Hayashi Y, Ishimaru N. Impaired expansion of regulatory T cells in a neonatal thymectomy-induced autoimmune mouse model. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:2886-97. [PMID: 26343329 DOI: 10.1016/j.ajpath.2015.07.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 06/17/2015] [Accepted: 07/28/2015] [Indexed: 11/16/2022]
Abstract
Neonatal thymectomy in certain mouse strains is known to induce organ-specific autoimmunity due to impaired functions of T cells, including Foxp3(+) regulatory T (Treg) cells in the thymus. The precise mechanism underlying the induction of autoimmunity by neonatal thymectomy remains unclear. One possibility is that depletion of Treg cells breaks down peripheral tolerance. We examined the functions of Treg cells by using a murine Sjögren syndrome model of NFS/sld mice that underwent neonatal thymectomy. The ratio of Treg cells to effector memory phenotype T cells in thymectomy mice was significantly lower than that of nonthymectomy mice. In addition, in vitro induction of peripherally induced Treg cells by transforming growth factor-β (TGF-β) using naive T cells from Sjögren syndrome model mice was severely impaired. The mRNA expression of TGF-β receptor I and II and Smad3 and -4 in the TGF-β-induced signal transduction pathway of Treg cells in this Sjögren syndrome model were lower than those of control mice. In addition, Treg cells in this Sjögren syndrome model exhibited an interferon-γ-producing Th1-like phenotype that resembled effector T cells. In conclusion, these results suggest that abnormal expansion and differentiation of Treg cells and inflammatory cytokines produced by Treg cells contribute to the development of autoimmunity.
Collapse
Affiliation(s)
- Akiko Yamada
- Department of Oral Molecular Pathology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Aya Ushio
- Department of Oral Molecular Pathology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Rieko Arakaki
- Department of Oral Molecular Pathology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Takaaki Tsunematsu
- Department of Oral Molecular Pathology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Yasusei Kudo
- Department of Oral Molecular Pathology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Yoshio Hayashi
- Department of Oral Molecular Pathology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Naozumi Ishimaru
- Department of Oral Molecular Pathology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan.
| |
Collapse
|
43
|
Lee KE, Kang JH, Lee JW, Wen L, Park DJ, Kim TJ, Park YW, Lee SS. Anti-centromere antibody-positive Sjögren's syndrome: A distinct clinical subgroup? Int J Rheum Dis 2015; 18:776-82. [DOI: 10.1111/1756-185x.12684] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Kyung-Eun Lee
- Division of Rheumatology; Department of Internal Medicine; Chonnam National University Medical School & Hospital; Gwangju Korea
| | - Ji-Hyoun Kang
- Division of Rheumatology; Department of Internal Medicine; Chonnam National University Medical School & Hospital; Gwangju Korea
| | - Jeong-Won Lee
- Division of Rheumatology; Department of Internal Medicine; Chonnam National University Medical School & Hospital; Gwangju Korea
| | - Lihui Wen
- Division of Rheumatology; Department of Internal Medicine; Chonnam National University Medical School & Hospital; Gwangju Korea
| | - Dong-Jin Park
- Division of Rheumatology; Department of Internal Medicine; Chonnam National University Medical School & Hospital; Gwangju Korea
| | - Tae-Jong Kim
- Division of Rheumatology; Department of Internal Medicine; Chonnam National University Medical School & Hospital; Gwangju Korea
| | - Yong-Wook Park
- Division of Rheumatology; Department of Internal Medicine; Chonnam National University Medical School & Hospital; Gwangju Korea
| | - Shin-Seok Lee
- Division of Rheumatology; Department of Internal Medicine; Chonnam National University Medical School & Hospital; Gwangju Korea
| |
Collapse
|
44
|
Corsiero E, Sutcliffe N, Pitzalis C, Bombardieri M. Accumulation of self-reactive naïve and memory B cell reveals sequential defects in B cell tolerance checkpoints in Sjögren's syndrome. PLoS One 2014; 9:e114575. [PMID: 25535746 PMCID: PMC4275206 DOI: 10.1371/journal.pone.0114575] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 11/11/2014] [Indexed: 11/18/2022] Open
Abstract
Sjögren's syndrome (SS) is an autoimmune disease characterised by breach of self-tolerance towards nuclear antigens resulting in high affinity circulating autoantibodies. Although peripheral B cell disturbances have been described in SS, with predominance of naïve and reduction of memory B cells, the stage at which errors in B cell tolerance checkpoints accumulate in SS is unknown. Here we determined the frequency of self- and poly-reactive B cells in the circulating naïve and memory compartment of SS patients. Single CD27-IgD+ naïve, CD27+IgD+ memory unswitched and CD27+IgD- memory switched B cells were sorted by FACS from the peripheral blood of 7 SS patients. To detect the frequency of polyreactive and autoreactive clones, paired Ig VH and VL genes were amplified, cloned and expressed as recombinant monoclonal antibodies (rmAbs) displaying identical specificity of the original B cells. IgVH and VL gene usage and immunoreactivity of SS rmAbs were compared with those obtained from healthy donors (HD). From a total of 353 VH and 293 VL individual sequences, we obtained 114 rmAbs from circulating naïve (n = 66) and memory (n = 48) B cells of SS patients. Analysis of the Ig V gene repertoire did not show significant differences in SS vs. HD B cells. In SS patients, circulating naïve B cells (with germline VH and VL genes) displayed a significant accumulation of clones autoreactive against Hep-2 cells compared to HD (43.1% vs. 25%). Moreover, we demonstrated a progressive increase in the frequency of circulating anti-nuclear naïve (9.3%), memory unswitched (22.2%) and memory switched (27.3%) B cells in SS patients. Overall, these data provide novel evidence supporting the existence of both early and late defects in B cell tolerance checkpoints in patients with SS resulting in the accumulation of autoreactive naïve and memory B cells.
Collapse
Affiliation(s)
- Elisa Corsiero
- Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, United Kingdom
- * E-mail: (EC); (MB)
| | - Nurhan Sutcliffe
- Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, United Kingdom
| | - Costantino Pitzalis
- Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, United Kingdom
| | - Michele Bombardieri
- Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, United Kingdom
- * E-mail: (EC); (MB)
| |
Collapse
|
45
|
Proteomic profiling of antigens in circulating immune complexes associated with each of seven autoimmune diseases. Clin Biochem 2014; 48:181-5. [PMID: 25438073 DOI: 10.1016/j.clinbiochem.2014.11.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 10/10/2014] [Accepted: 11/11/2014] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Immune complexes (ICs) trigger humoral immune responses. Therefore, the identification of constituent antigens within ICs would have very different clinical significance than identification of free antigens. DESIGN AND METHODS Here, we applied immune complexome analysis of serum to the study of seven major autoimmune diseases-anti-neutrophil cytoplasmic antibody-associated vasculitis, Takayasu's arteritis, mixed connective tissue disease, dermatomyositis, Sjögren's syndrome, systemic scleroderma, and systemic lupus erythematosus-and healthy donors to comprehensively identify antigens incorporated into circulating ICs and to find disease-specific antigens. RESULTS We identified 468 distinct IC-associated antigens using this method. Importantly, 62 of those antigens were disease-specific antigens, and there were at least three disease-specific antigens for each of the seven autoimmune diseases. Of the disease-specific antigens identified, coiled-coil domain-containing protein 158 and spectrin were identified as potential autoantigens important to SSc and SS pathogenesis, respectively; notable titin and spectrin autoantibodies are reportedly found in SSc and SS patients, respectively. CONCLUSION Immune complexome analysis may be generally applicable to the study of the relationship between ICs and autoimmune diseases in animals and humans.
Collapse
|
46
|
Kern J, Drutel R, Leanhart S, Bogacz M, Pacholczyk R. Reduction of T cell receptor diversity in NOD mice prevents development of type 1 diabetes but not Sjögren's syndrome. PLoS One 2014; 9:e112467. [PMID: 25379761 PMCID: PMC4224485 DOI: 10.1371/journal.pone.0112467] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 10/09/2014] [Indexed: 12/23/2022] Open
Abstract
Non-obese diabetic (NOD) mice are well-established models of independently developing spontaneous autoimmune diseases, Sjögren’s syndrome (SS) and type 1 diabetes (T1D). The key determining factor for T1D is the strong association with particular MHCII molecule and recognition by diabetogenic T cell receptor (TCR) of an insulin peptide presented in the context of I-Ag7 molecule. For SS the association with MHCII polymorphism is weaker and TCR diversity involved in the onset of the autoimmune phase of SS remains poorly understood. To compare the impact of TCR diversity reduction on the development of both diseases we generated two lines of TCR transgenic NOD mice. One line expresses transgenic TCRβ chain originated from a pathogenically irrelevant TCR, and the second line additionally expresses transgenic TCRαmini locus. Analysis of TCR sequences on NOD background reveals lower TCR diversity on Treg cells not only in the thymus, but also in the periphery. This reduction in diversity does not affect conventional CD4+ T cells, as compared to the TCRmini repertoire on B6 background. Interestingly, neither transgenic TCRβ nor TCRmini mice develop diabetes, which we show is due to lack of insulin B:9–23 specific T cells in the periphery. Conversely SS develops in both lines, with full glandular infiltration, production of autoantibodies and hyposalivation. It shows that SS development is not as sensitive to limited availability of TCR specificities as T1D, which suggests wider range of possible TCR/peptide/MHC interactions driving autoimmunity in SS.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Autoantibodies/immunology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/immunology
- Flow Cytometry
- Genetic Variation/immunology
- Insulin/genetics
- Insulin/immunology
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, Knockout
- Mice, Transgenic
- Molecular Sequence Data
- Peptide Fragments/genetics
- Peptide Fragments/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Salivary Glands/immunology
- Salivary Glands/metabolism
- Sjogren's Syndrome/genetics
- Sjogren's Syndrome/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Xerostomia/immunology
Collapse
Affiliation(s)
- Joanna Kern
- Center for Biotechnology and Genomic Medicine, Georgia Regents University, Augusta, Georgia, United States of America
| | - Robert Drutel
- Center for Biotechnology and Genomic Medicine, Georgia Regents University, Augusta, Georgia, United States of America
| | - Silvia Leanhart
- Center for Biotechnology and Genomic Medicine, Georgia Regents University, Augusta, Georgia, United States of America
| | - Marek Bogacz
- Center for Biotechnology and Genomic Medicine, Georgia Regents University, Augusta, Georgia, United States of America
| | - Rafal Pacholczyk
- Center for Biotechnology and Genomic Medicine, Georgia Regents University, Augusta, Georgia, United States of America
| |
Collapse
|
47
|
Aromatase controls Sjögren syndrome-like lesions through monocyte chemotactic protein-1 in target organ and adipose tissue-associated macrophages. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 185:151-61. [PMID: 25447050 DOI: 10.1016/j.ajpath.2014.09.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 09/02/2014] [Accepted: 09/09/2014] [Indexed: 02/06/2023]
Abstract
Several autoimmune diseases are known to develop in postmenopausal women. However, the mechanism by which estrogen deficiency influences autoimmunity is unknown. Aromatase is an enzyme that converts androgens to estrogens. Herein, we used female aromatase gene knockout (ArKO) mice as a model of estrogen deficiency to investigate the molecular mechanism that underlies the onset and development of autoimmunity. Histological analyses showed that inflammatory lesions in the lacrimal and salivary glands of ArKO mice increased with age. Adoptive transfer of spleen cells or bone marrow cells from ArKO mice into recombination activating gene 2 knockout mice failed to induce the autoimmune lesions. Expression of mRNA encoding proinflammatory cytokines and monocyte chemotactic protein-1 increased in white adipose tissue of ArKO mice and was significantly higher than that in wild-type mice. Moreover, an increased number of inflammatory M1 macrophages was observed in white adipose tissue of ArKO mice. A significantly increased monocyte chemotactic protein-1 mRNA expression of the salivary gland tissue in ArKO was found together with adiposity. Furthermore, the autoimmune lesions in a murine model of Sjögren syndrome were exacerbated by administration of an aromatase inhibitor. These results suggest that aromatase may play a key role in the pathogenesis of Sjögren syndrome-like lesions by controlling the target organ and adipose tissue-associated macrophage.
Collapse
|
48
|
Arakaki R, Eguchi H, Yamada A, Kudo Y, Iwasa A, Enkhmaa T, Hotta F, Mitamura-Aizawa S, Mitamura Y, Hayashi Y, Ishimaru N. Anti-inflammatory effects of rebamipide eyedrop administration on ocular lesions in a murine model of primary Sjögren's syndrome. PLoS One 2014; 9:e98390. [PMID: 24866156 PMCID: PMC4035282 DOI: 10.1371/journal.pone.0098390] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 05/01/2014] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Topical therapy is effective for dry eye, and its prolonged effects should help in maintaining the quality of life of patients with dry eye. We previously reported that the oral administration of rebamipide (Reb), a mucosal protective agent, had a potent therapeutic effect on autoimmune lesions in a murine model of Sjögren's syndrome (SS). However, the effects of topical treatment with Reb eyedrops on the ocular lesions in the murine model of SS are unknown. METHODS AND FINDING Reb eyedrops were administered to the murine model of SS aged 4-8 weeks four times daily. Inflammatory lesions of the extraorbital and intraorbital lacrimal glands and Harderian gland tissues were histologically evaluated. The direct effects of Reb on the lacrimal glands were analyzed using cultured lacrimal gland cells. Tear secretions of Reb-treated mice were significantly increased compared with those of untreated mice. In addition to the therapeutic effect of Reb treatment on keratoconjunctivitis, severe inflammatory lesions of intraorbital lacrimal gland tissues in this model of SS were resolved. The mRNA expression levels of IL-10 and mucin 5Ac in conjunctival tissues from Reb-treated mice was significantly increased compared with those of control mice. Moreover, lactoferrin production from lacrimal gland cells was restored by Reb treatment. CONCLUSION Topical Reb administration had an anti-inflammatory effect on the ocular autoimmune lesions in the murine model of SS and a protective effect on the ocular surfaces.
Collapse
Affiliation(s)
- Rieko Arakaki
- Department of Oral Molecular Pathology, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Hiroshi Eguchi
- Department of Ophthalmology, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Akiko Yamada
- Department of Oral Molecular Pathology, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Yasusei Kudo
- Department of Oral Molecular Pathology, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Akihiko Iwasa
- Department of Oral Molecular Pathology, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Tserennadmid Enkhmaa
- Department of Ophthalmology, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Fumika Hotta
- Department of Ophthalmology, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Sayaka Mitamura-Aizawa
- Department of Ophthalmology, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Yoshinori Mitamura
- Department of Ophthalmology, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Yoshio Hayashi
- Department of Oral Molecular Pathology, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Naozumi Ishimaru
- Department of Oral Molecular Pathology, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| |
Collapse
|
49
|
Chin MS, Hooper LC, Hooks JJ, Detrick B. Identification of α-fodrin as an autoantigen in experimental coronavirus retinopathy (ECOR). J Neuroimmunol 2014; 272:42-50. [PMID: 24864013 PMCID: PMC7112846 DOI: 10.1016/j.jneuroim.2014.05.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 03/19/2014] [Accepted: 05/04/2014] [Indexed: 11/29/2022]
Abstract
The coronavirus, mouse hepatitis virus (MHV), JHM strain induces a biphasic disease in BALB/c mice that consists of an acute retinitis followed by progression to a chronic retinal degeneration with autoimmune reactivity. Retinal degeneration resistant CD-1 mice do not develop either the late phase or autoimmune reactivity. A mouse RPE/choroid DNA expression library was screened using sera from virus infected BALB/c mice. Two clones were identified, villin-2 protein and α-fodrin protein. α-Fodrin protein was used for further analysis and western blot reactivity was seen only in sera from virus infected BALB/c mice. CD4 T cells were shown to specifically react with MHV antigens and with α-fodrin protein. These studies clearly identified both antibody and CD4 T cell reactivities to α-fodrin in sera from virus infected, retinal degenerative susceptible BALB/c mice.
Collapse
Affiliation(s)
- Marian S Chin
- Immunology and Virology Section, Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, United States
| | - Laura C Hooper
- Immunology and Virology Section, Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, United States
| | - John J Hooks
- Immunology and Virology Section, Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, United States
| | - Barbara Detrick
- Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| |
Collapse
|
50
|
Donate A, Voigt A, Nguyen CQ. The value of animal models to study immunopathology of primary human Sjögren's syndrome symptoms. Expert Rev Clin Immunol 2014; 10:469-81. [PMID: 24506531 PMCID: PMC5769146 DOI: 10.1586/1744666x.2014.883920] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Sjögren's syndrome (SjS) is a complex chronic autoimmune disease of multifactorial etiology that results in eventual loss of secretory function in the exocrine glands. The challenges towards finding a therapeutic prevention or treatment for SjS are due primarily to a lack of understanding in the pathophysiological and clinical progression of the disease. In order to circumnavigate this problem, there is a need for appropriate animal models that resemble the major phenotypes of human SjS and deliver a clear underlying biological or molecular mechanism capable of defining various aspects for the disease. Here, we present an overview of SjS mouse models that are providing insight into the autoimmune process of SjS and advance our focus on potential diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Amy Donate
- Department of Infectious Diseases and Pathology, University of Florida College of Veterinary Medicine, 2015 SW 16 Ave, Gainesville, Florida 32611, USA
| | - Alexandria Voigt
- Department of Infectious Diseases and Pathology, University of Florida College of Veterinary Medicine, 2015 SW 16 Ave, Gainesville, Florida 32611, USA
| | - Cuong Q. Nguyen
- Department of Infectious Diseases and Pathology, University of Florida College of Veterinary Medicine, 2015 SW 16 Ave, Gainesville, Florida 32611, USA
- Center for Orphan Autoimmune Disorders, University of Florida College of Dentistry, 1600 SW Archer Rd, Gainesville, Florida 32610, USA
- Department of Oral Biology, University of Florida College of Dentistry, 1600 SW Archer Rd, Gainesville, Florida 32610, USA
| |
Collapse
|