1
|
Quan K, Wang H, Su P, Xu Y, Yao X. Decoding B Cells in Autoimmune Diseases Through ScRNA + BCR-Seq: Current Knowledge and Future Directions. Cells 2025; 14:539. [PMID: 40214492 PMCID: PMC11988620 DOI: 10.3390/cells14070539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 04/01/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025] Open
Abstract
The combined application of single-cell RNA sequencing (scRNA-seq) and single-cell B-cell receptor sequencing (scBCR-seq) offers a multidimensional perspective for dissecting the immunopathological mechanisms of B cells in autoimmune diseases. This review systematically summarizes the principles of these techniques, the analytical framework, and their key applications in diseases such as systemic lupus erythematosus et. al. It reveals the dynamic correlations between the transcriptome of B-cell subsets and B-cell receptor (BCR) clones. Furthermore, we focus on the potential roles of dual BCR B cells and B/T biphenotypic cells in autoimmunity, emphasizing their exacerbation of disease progression through abnormal clonal expansion and autoantibody secretion. By sorting through cutting-edge advancements and bottleneck issues, this article aims to propel the innovation of multi-omics research and precision treatment paradigms for autoimmune diseases.
Collapse
Affiliation(s)
| | | | | | | | - Xinsheng Yao
- Department of Immunology, Center of Immuno-Molecular Engineering, Innovation & Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi 563002, China; (K.Q.); (H.W.); (P.S.); (Y.X.)
| |
Collapse
|
2
|
Stramazzo I, Mangino G, Capriello S, Romeo G, Ferrari SM, Fallahi P, Bagaglini MF, Centanni M, Virili C. CD20 + T lymphocytes in isolated Hashimoto's thyroiditis and type 3 autoimmune polyendocrine syndrome: a pilot study. J Endocrinol Invest 2024; 47:2865-2871. [PMID: 38642306 PMCID: PMC11473566 DOI: 10.1007/s40618-024-02370-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 03/24/2024] [Indexed: 04/22/2024]
Abstract
BACKGROUND CD20+ T cells represent up to 5% of circulating T lymphocytes. These cells have been shown to produce higher levels of IL-17A and IFN-γ than those of CD20- T lymphocytes. Some reports described the role of CD20+ T cells in autoimmune disorders such as multiple sclerosis and rheumatoid arthritis possibly due to their ability to produce these inflammatory cytokines. This study is aimed at describing the behavior of CD20+ T lymphocytes in the most frequent autoimmune disorder, i.e., Hashimoto's thyroiditis (HT), presenting isolated or associated to further autoaggressive disorders in a frame of poly-autoimmunity. METHODS The study group encompasses 65 HT patients: 23 presenting in isolated form (IT) and 42 with an associated non-endocrine autoimmune disorder [16 with chronic atrophic gastritis (CAG), 15 with nonsegmental vitiligo (VIT), and 11 with celiac disease (CD)]. Twenty healthy donors act as control group (HD). Chronic use of interfering drugs, severe or chronic disorders, and pregnancy and lactation were used as exclusion criteria. Whole blood samples (100 µl) were stained with fluorescent-labeled antibodies (anti-CD45, anti-CD3, anti-CD19, anti-CD16, anti-CD56, anti-CD4, anti-CD8, anti-CD20). Red blood cells were then lysed by adding 1 ml of hypotonic buffer, and samples were acquired on a Flow Cytometer. RESULTS CD3+CD8+CD20+ T lymphocytes' percentages, were significantly higher in the whole group of autoimmune patients compared to healthy donors (p = 0.0145). Dividing HT patients based on the type of presentation of autoimmune thyroiditis, CAG group showed the highest percentage of these cells as compared to HD and CD (p = 0.0058). IT patients showed higher percentages of CD3+ CD8+CD20+ cells than those of HD patients although not reaching statistical significance. However, dividing IT group based on thyroid function, hypothyroid patients showed higher CD8+CD20+ cell percentages than those of HD and euthyroid patients (p = 0.0111). Moreover, in IT patients, these cells were negatively correlated with FT4 levels (p = 0.0171; r = -0.4921). CONCLUSIONS These preliminary findings indicate that CD8+CD20+ T cells are activated in patients with autoimmune thyroiditis and may behave differently according to the presence of poly-autoimmunity and hypothyroidism.
Collapse
Affiliation(s)
- Ilaria Stramazzo
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Giorgio Mangino
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Silvia Capriello
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Giovanna Romeo
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | | | - Poupak Fallahi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | | | - Marco Centanni
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy.
| | - Camilla Virili
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| |
Collapse
|
3
|
Pinho ACO, Barbosa P, Lazaro A, Tralhão JG, Pereira MJ, Paiva A, Laranjeira P, Carvalho E. Identification and characterization of circulating and adipose tissue infiltrated CD20 +T cells from subjects with obesity that undergo bariatric surgery. Immunol Lett 2024; 269:106911. [PMID: 39147242 DOI: 10.1016/j.imlet.2024.106911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 08/10/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024]
Abstract
T cells play critical roles in adipose tissue (AT) inflammation. The role of CD20+T cell in AT dysfunction and their contributing to insulin resistance (IR) and type 2 diabetes progression, is not known. The aim was to characterize CD20+T cells in omental (OAT), subcutaneous (SAT) and peripheral blood (PB) from subjects with obesity (OB, n = 42), by flow cytometry. Eight subjects were evaluated before (T1) and 12 months post (T2) bariatric/metabolic surgery (BMS). PB from subjects without obesity (nOB, n = 12) was also collected. Higher percentage of CD20+T cells was observed in OAT, compared to PB or SAT, in OB-T1. CD20 expression by PB CD4+T cells was inversely correlated with adiposity markers, while follicular-like CD20+T cells were positively correlated with impaired glucose tolerance (increased HbA1c). Notably, among OB-T1, IR establishment was marked by a lower percentage and absolute number of PB CD20+T cells, compared nOB. Obesity was associated with higher percentage of activated CD20+T cells; however, OAT-infiltrated CD20+T cells from OB-T1 with diabetes displayed the lowest activation. CD20+T cells infiltrating OAT from OB-T1 displayed a phenotype towards IFN-γ-producing Th1 and Tc1 cells. After BMS, the percentage of PB CD4+CD20+T cells increased, with reduced Th1 and increased Th17 phenotype. Whereas in OAT the percentage of CD20+T cells with Th1/17 and Tc1/17 phenotypes increased. Interestingly, OAT from OB pre/post BMS maintained higher frequency of effector memory CD20+T cells. In conclusion, CD20+T cells may play a prominent role in obesity-related AT inflammation.
Collapse
Affiliation(s)
- Aryane Cruz Oliveira Pinho
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal; CIBB - Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504, Coimbra, Portugal; Department of Life Sciences, Faculty of Science and Technology, University of Coimbra, 3000-456, Coimbra, Portugal
| | - Pedro Barbosa
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal; CIBB - Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504, Coimbra, Portugal; Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
| | - André Lazaro
- General Surgery Unit, Centro Hospitalar e Universitário de Coimbra University of Coimbra, 3000-075, Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548, Coimbra, Portugal
| | - José G Tralhão
- General Surgery Unit, Centro Hospitalar e Universitário de Coimbra University of Coimbra, 3000-075, Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Maria João Pereira
- Department of Medical Sciences, Clinical Diabetology and Metabolism, Uppsala University, Uppsala, Sweden
| | - Artur Paiva
- CIBB - Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504, Coimbra, Portugal; Flow Cytometry Unit, Clinical Pathology Department, Hospitais da Universidade de Coimbra, Unidade Local de Saúde de Coimbra, 3000-076, Coimbra, Portugal; Instituto Politécnico de Coimbra, ESTESC-Coimbra Health School, Ciências Biomédicas Laboratoriais, 3046-854, Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR), Group of Environmental Genetics of Oncobiology (CIMAGO), Faculty of Medicine (FMUC), University of Coimbra, 3000-548, Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), 3000-061, Coimbra, Portugal
| | - Paula Laranjeira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal; CIBB - Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504, Coimbra, Portugal; Flow Cytometry Unit, Clinical Pathology Department, Hospitais da Universidade de Coimbra, Unidade Local de Saúde de Coimbra, 3000-076, Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR), Group of Environmental Genetics of Oncobiology (CIMAGO), Faculty of Medicine (FMUC), University of Coimbra, 3000-548, Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), 3000-061, Coimbra, Portugal.
| | - Eugenia Carvalho
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal; CIBB - Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504, Coimbra, Portugal; Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal; APDP-Portuguese Diabetes Association, Lisbon, Portugal.
| |
Collapse
|
4
|
Arneth B. Current Knowledge about CD3 +CD20 + T Cells in Patients with Multiple Sclerosis. Int J Mol Sci 2024; 25:8987. [PMID: 39201672 PMCID: PMC11354236 DOI: 10.3390/ijms25168987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 08/16/2024] [Accepted: 08/18/2024] [Indexed: 09/03/2024] Open
Abstract
Multiple sclerosis (MS) is a disease of the central nervous system (CNS) characterized by inflammation and autoimmune responses. This review explores the participation of T cells, particularly certain CD3+CD20+ T cells, in the clinical manifestations of MS and highlights their presence in diagnosed patients. These T cells show aberrant expression of CD20, normally considered a B-cell marker. In this review, relevant journal articles available in PubMed and CINAHL were identified by employing diverse search terms, such as MS, CD3+CD20+ T cells, the incidence and significance of CD3+CD20+ T cells in MS patients, and the impact of rituximab treatment. The search was limited to articles published in the ten-year period from 2014 to 2024. The results of this review suggest that most scholars agree on the presence of CD3+CD20+ T cells in cerebrospinal fluid. Emerging concepts relate to the fundamental role of CD20-expressing T cells in determining the target and efficacy of MS therapeutics and the presence of T cells in the cerebrospinal fluid of MS patients. The results clearly show that CD20+ T cells indicate disease chronicity and high disease activity.
Collapse
Affiliation(s)
- Borros Arneth
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Hospital of the Universities of Giessen and Marburg (UKGM), Justus Liebig University Giessen, Feulgenstr. 12, 35392 Giessen, Germany;
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Hospital of the Universities of Giessen and Marburg (UKGM), Philipps University Marburg, Baldinger Str., 35043 Marburg, Germany
| |
Collapse
|
5
|
Pan P, Pineda MA, Wang Y, Khan A, Nyirenda MH. Aberrant pro-inflammatory responses of CD20 + T cells in experimental arthritis. Cell Immunol 2023; 387:104717. [PMID: 37075620 DOI: 10.1016/j.cellimm.2023.104717] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/28/2023] [Accepted: 04/02/2023] [Indexed: 04/08/2023]
Abstract
CD20+ T cells comprise a highly inflammatory subset implicated in autoimmunity, including rheumatoid arthritis (RA). We sought to characterize the CD20+ T cell subset in the murine collagen-induced arthritis (CIA) model of RA and investigate the phenotype and functional relevance of CD3+CD20+ T cells in the lymph nodes and arthritic joints using flow cytometry and immunohistochemistry. We demonstrate that CD3+CD4+CD20+ and CD3+CD8+CD20+ T cells are expanded in the draining lymph nodes of CIA mice, produce increased levels of pro-inflammatory cytokines and are less susceptible to regulation by regulatory T cells. Notably, CD3+CD4+CD20+ and CD3+CD8+CD20+ T cells are enriched with CXCR5+PD-1+ T follicular helper cells and CXCR5-PD-1+ peripheral T helper cells, subsets of T cells implicated in promoting B-cell responses and antibody production within pathologically inflamed non-lymphoid tissues in RA. Our findings suggest CD20+ T cells are associated with inflammatory responses and may exacerbate pathology by promoting inflammatory B-cell responses.
Collapse
Affiliation(s)
- Piaopiao Pan
- University of Glasgow, School of Infection and Immunity, Glasgow, UK
| | - Miguel A Pineda
- Research into Inflammatory Arthritis Centre, Versus Arthritis (RACE-VA), Glasgow, Birmingham, Newcastle, and Oxford, UK; University of Glasgow, Centre for the Cellular Microenvironment, School of Molecular Biosciences, Glasgow, UK
| | - Yilin Wang
- University of Glasgow, School of Infection and Immunity, Glasgow, UK
| | - Aneesah Khan
- University of Glasgow, School of Infection and Immunity, Glasgow, UK
| | - Mukanthu H Nyirenda
- University of Glasgow, School of Infection and Immunity, Glasgow, UK; Research into Inflammatory Arthritis Centre, Versus Arthritis (RACE-VA), Glasgow, Birmingham, Newcastle, and Oxford, UK.
| |
Collapse
|
6
|
Bridges JM, Chatham WW. Biologic Response Modifiers. Clin Immunol 2023. [DOI: 10.1016/b978-0-7020-8165-1.00086-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
|
7
|
Fonseca Peixoto R, Ewerton Maia Rodrigues C, Henrique de Sousa Palmeira P, Cézar Comberlang Queiroz Davis Dos Santos F, Keesen de Souza Lima T, de Sousa Braz A. Immune hallmarks of rheumatoid arthritis management: A brief review. Cytokine 2022; 158:156007. [PMID: 35985174 DOI: 10.1016/j.cyto.2022.156007] [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: 05/14/2022] [Revised: 07/31/2022] [Accepted: 08/08/2022] [Indexed: 11/17/2022]
Abstract
The purpose of this review was to examine current evidence on immunomodulation mediated by conventional drugs and the use of novel biological agents for the treatment of rheumatoid arthritis (RA). Currently, treatment is focused on maximizing quality of life through sustained clinical remission and/or attenuating disease activity. To do so, disease-modifying antirheumatic drugs, especially methotrexate, are used alone or in combination with other drugs, including leflunomide, biological disease-modifying antirheumatic drugs (bDMARDs) and targeted synthetic disease-modifying antirheumatic drugs (tsDMARDs). The most recent strategies modulate the immune response of the individual RA patient using tsDMARDs such as JAK inhibitors and bDMARDs such as ig-CTLA-4, anti- IL6R, anti-TNF-α and anti-CD20. To better understand current immunopharmacological interventions, we also looked at documented mechanisms of RA-mediated immunomodulation, highlighting perspectives potentially boosting RA treatment.
Collapse
Affiliation(s)
- Rephany Fonseca Peixoto
- Laboratory of Immunology of Infectious Diseases, Department of Cellular and Molecular Biology, Federal University of Paraiba, João Pessoa, Paraíba 58051-900, Brazil
| | - Carlos Ewerton Maia Rodrigues
- Post‑Graduate Program in Medical Sciences, Medical School, University of Fortaleza (Unifor), Fortaleza, Brazil; Department of Internal Medicine, Federal University of Ceará, Brazil.
| | - Pedro Henrique de Sousa Palmeira
- Laboratory of Immunology of Infectious Diseases, Department of Cellular and Molecular Biology, Federal University of Paraiba, João Pessoa, Paraíba 58051-900, Brazil
| | | | - Tatjana Keesen de Souza Lima
- Laboratory of Immunology of Infectious Diseases, Department of Cellular and Molecular Biology, Federal University of Paraiba, João Pessoa, Paraíba 58051-900, Brazil
| | | |
Collapse
|
8
|
Lee AYS. CD20 + T cells: an emerging T cell subset in human pathology. Inflamm Res 2022; 71:1181-1189. [PMID: 35951029 PMCID: PMC9616751 DOI: 10.1007/s00011-022-01622-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 07/23/2022] [Accepted: 07/27/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction Although CD20 is classically a B cell marker, in the last three decades, dim expression has been noted on a subset of T cells as well that has been independently verified by a number of groups. Our understanding of these cells and their function is not well established. Methods A thorough review of original articles on CD20+T cells was undertaken of Pubmed by using combination of phrases including “CD20+”, “CD20-positive” and “T cells”. Articles in English were considered, and there was no time restriction. Results CD20+T cells express the standard T cell markers and, in comparison to CD20¯ T cells, appear to express greater inflammatory cytokines and markers of effector function. Although the ontogeny of these cells is still being established, the current theory is that CD20 may be acquired by trogocytosis from B cells. CD20+T cells may be found in healthy controls and in a wide range of pathologies including autoimmune diseases, haematological and non-haematological malignancies and human immunodeficiency virus (HIV) infections. One of the best studied diseases where these cells are found is multiple sclerosis (MS) where a number of therapeutic interventions, including anti-CD20 depletion, have been shown to effectively deplete these cells. Conclusion This review summarises the latest understanding of CD20+T cells, their presence in various diseases, their putative function and how they may be an ongoing target of CD20-depleting agents. Unfortunately, our understanding of these cells is still at its infancy and ongoing study in a wider range of pathologies is required.
Collapse
Affiliation(s)
- Adrian Y S Lee
- Department of Clinical Immunology, Westmead Hospital, Hawkesbury Road, Westmead, NSW, 2145, Australia. .,Department of Immunopathology, ICPMR and NSW Health Pathology, Westmead Hospital, Westmead, NSW, Australia. .,Department of Medicine, Westmead Clinical School, The University of Sydney, Westmead, NSW, Australia.
| |
Collapse
|
9
|
Meinl E, Hohlfeld R. CD20 + T Cells as Pathogenic Players and Therapeutic Targets in MS. Ann Neurol 2021; 90:722-724. [PMID: 34585761 DOI: 10.1002/ana.26232] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 09/25/2021] [Accepted: 09/27/2021] [Indexed: 12/28/2022]
Affiliation(s)
- Edgar Meinl
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany.,Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Reinhard Hohlfeld
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany.,Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
10
|
Bone-sparing effects of rituximab and body composition analysis in a cohort of postmenopausal women affected by rheumatoid arthritis - retrospective study. Reumatologia 2021; 59:206-210. [PMID: 34538950 PMCID: PMC8436793 DOI: 10.5114/reum.2021.108430] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 08/06/2021] [Indexed: 12/04/2022] Open
Abstract
Objective Osteoporosis is the most common bone tissue disease and it is characterized by a reduced bone mineral density (BMD). The main physiopathological mechanisms converge on the uncoupling between bone formation and resorption, thus leading to an enhanced risk of fractures. Several papers have documented the inverse relationships linking high inflammatory cytokines, anti-citrullinated protein antibodies, rheumatoid factor, and BMD in rheumatoid arthritis (RA). Rituximab (RTX) is a chimeric monoclonal antibody directed against the CD20 receptor of B cells. Since the Food and Drug Administration approved it for RA in 2006, there have been many clinical experiences regarding its use. Nevertheless, few studies evaluate the effect of rituximab on BMD. RA is a disease characterized by immune dysfunction with high levels of inflammatory cytokines, autoantibodies, and it is reasonable that a B cell depleting therapy could restore a physiological cytokine balance, thus exerting an osteoprotective effect on the bone tissue. The purpose of this paper is to highlight any difference in BMD and to assess differences in body composition over a retrospective 18-month follow-up period after RTX treatment with a B cell depleting therapy. Material and methods We analyzed by dual energy X-ray absorptiometry BMD expressed as g/cm2 and body composition modifications over 18 months with RTX treatment of 20 postmenopausal RA patients. Results After eighteen months of therapy with RTX, a statistically significant increase in vertebral (L1–L4) BMD and the stability of femoral BMD were documented. Conclusions Rituximab is associated with an improvement of vertebral and preservation of femoral BMD, suggesting a bone-sparing effect due to B cell depletion. Furthermore, patients displayed a redistribution of fat masses toward the hip region.
Collapse
|
11
|
Quendt C, Ochs J, Häusser-Kinzel S, Häusler D, Weber MS. Proinflammatory CD20 + T Cells are Differentially Affected by Multiple Sclerosis Therapeutics. Ann Neurol 2021; 90:834-839. [PMID: 34516013 DOI: 10.1002/ana.26216] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 09/08/2021] [Accepted: 09/11/2021] [Indexed: 12/13/2022]
Abstract
The frequency of CD20+ T cells was reported to be increased in several inflammatory conditions. We report that in patients with multiple sclerosis (MS), CD20+ T cells display a distinct proinflammatory phenotype with pathogenic properties. Anti-CD20 treatment virtually extinguished CD20+ T cells, which might explain its broad effectiveness. Dimethyl fumarate dampened activity of differentiated CD20+ T cells, whereas fingolimod reduced their abundance only as part of its overall T cell suppressive capacity. Natalizumab increased the frequency of CD20+ effector T cells. Widely used MS therapeutics affect this proinflammatory T cell subset with assumed pathogenic potential in a surprisingly differential manner. ANN NEUROL 2021 ANN NEUROL 2021;90:834-839.
Collapse
Affiliation(s)
- Corinna Quendt
- Department of Neurology, University Medical Center, Göttingen, Germany
| | - Jasmin Ochs
- Institute of Neuropathology, University Medical Center, Göttingen, Germany
| | | | - Darius Häusler
- Institute of Neuropathology, University Medical Center, Göttingen, Germany
| | - Martin S Weber
- Department of Neurology, University Medical Center, Göttingen, Germany.,Institute of Neuropathology, University Medical Center, Göttingen, Germany.,Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Göttingen: Robert-Koch-Straße 40, Göttingen, Germany
| |
Collapse
|
12
|
Retamozo S, Sisó-Almirall A, Flores-Chávez A, Ramos-Casals M, Brito-Zerón P. An update of targeted therapeutic options for primary Sjögren syndrome: current status and future development. Expert Opin Pharmacother 2021; 22:2359-2371. [PMID: 34323636 DOI: 10.1080/14656566.2021.1951224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Introduction: Primary Sjögren syndrome (pSS) is a systemic autoimmune disease that may affect 3 in 1,000 people within the general population. The therapeutic scenario is complex, and no therapy has proved to be able to modify the natural course of the disease, nor to prevent the most severe systemic complications.Areas covered: Recently, the EULAR 2020 Recommendations for pSS have underlined the low level of evidence supporting efficacious therapeutic approaches, lacking a definition of specific treatment targets and being far from the 'disease modification' concept that is frequently used in other diseases. Herein, the authors review the status of current targeted therapies and provide the reader with their expert opinion.Expert opinion: The progress in discovering novel treatments for pSS seem to be focused on searching new biological therapies as highly-selective drugs that can be effective without the adverse effects related to the wide, nonspecific immunosuppression induced by the drugs currently used. Most likely, the more disruptive therapeutic approach in pSS that could be seen in a few years is the use of combination strategies targeting different etiopathogenic pathways.
Collapse
Affiliation(s)
- Soledad Retamozo
- Sjogren Syndrome Research Group (AGAUR), Department of Autoinmune Diseases, ICMiD, Hospital Clinic, Barcelona, Spain.,Rheumatology Department, Hospital Universitari Parc Taulí, Sabadell, Barcelona, Spain
| | - Antoni Sisó-Almirall
- Primary Care Centre Les Corts, Consorci d'Atenció Primària De Salut Barcelona Esquerra (CAPSBE), Barcelona, Spain.,Primary Healthcare Transversal Research Group, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | | | - Manuel Ramos-Casals
- Sjogren Syndrome Research Group (AGAUR), Department of Autoinmune Diseases, ICMiD, Hospital Clinic, Barcelona, Spain.,Laboratory of Autoimmune Diseases Josep Font, IDIBAPS-CELLEX, Barcelona, Spain
| | - Pilar Brito-Zerón
- Laboratory of Autoimmune Diseases Josep Font, IDIBAPS-CELLEX, Barcelona, Spain.,Autoimmune Diseases Unit, Department of Medicine, Hospital CIMA-Sanitas, Barcelona, Spain
| |
Collapse
|
13
|
Webendörfer M, Reinhard L, Stahl RAK, Wiech T, Mittrücker HW, Harendza S, Hoxha E. Rituximab Induces Complete Remission of Proteinuria in a Patient With Minimal Change Disease and No Detectable B Cells. Front Immunol 2021; 11:586012. [PMID: 33628202 PMCID: PMC7897659 DOI: 10.3389/fimmu.2020.586012] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 12/21/2020] [Indexed: 12/26/2022] Open
Abstract
Minimal change disease (MCD) is a common cause of nephrotic syndrome. Treatment with steroids is usually effective, but frequent relapses are therapeutic challenges. The anti-CD20 antibody rituximab has shown promising results for treatment of steroid-sensitive nephrotic syndrome. Since predictive biomarkers for treatment efficacy and the accurate rituximab dosage for effective induction of remission are unknown, measurement of CD19+ B cells in blood is often used as marker of successful B cell depletion and treatment efficacy. A male patient with relapsing MCD was successfully treated with rituximab, but developed relapse of proteinuria 1 year later, although no B cells were detectable in his blood. B and T cell populations in the patient's blood were analyzed before and after treatment with rituximab using FACS analysis. Rituximab binding to B and T cells were measured using Alexa Fluor 647 conjugated rituximab. We identified a population of CD20+ CD19- cells in the patient's blood, which consisted mostly of CD20+ CD3+ T cells. Despite the absence of B cells in the blood, the patient was again treated with rituximab. He developed complete remission of proteinuria and depletion of CD20+ T cells. In a control patient with relapsing MCD initial treatment with rituximab led to depletion of both CD20+ B and T cells. Rituximab induces remission of proteinuria in patients with MCD even if circulating B cells are absent. CD20+ T cells may play a role in the pathogenesis of MCD and might be a promising treatment target in patients with MCD.
Collapse
Affiliation(s)
- Maximilian Webendörfer
- III. Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Linda Reinhard
- III. Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Rolf A. K. Stahl
- III. Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thorsten Wiech
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hans-Willi Mittrücker
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sigrid Harendza
- III. Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Elion Hoxha
- III. Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
14
|
CD80 Insights as Therapeutic Target in the Current and Future Treatment Options of Frequent-Relapse Minimal Change Disease. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6671552. [PMID: 33506028 PMCID: PMC7806396 DOI: 10.1155/2021/6671552] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 12/26/2020] [Indexed: 12/14/2022]
Abstract
Minimal change disease (MCD) is the most common cause of idiopathic nephrotic syndrome in children, and it is well known for its multifactorial causes which are the manifestation of the disease. Proteinuria is an early consequence of podocyte injury and a typical sign of kidney disease. Steroid-sensitive patients react well with glucocorticoids, but there is a high chance of multiple relapses. CD80, also known as B7-1, is generally expressed on antigen-presenting cells (APCs) in steroid-sensitive MCD patients. Various glomerular disease models associated with proteinuria demonstrated that the detection of CD80 with the increase of urinary CD80 was strongly associated closely with frequent-relapse MCD patients. The role of CD80 in MCD became controversial because one contradicts finding. This review covers the treatment alternatives for MCD with the insight of CD80 as a potential therapeutic target. The promising effectiveness of CD20 (rituximab) antibody and CD80 inhibitor (abatacept) encourages further investigation of CD80 as a therapeutic target in frequent-relapse MCD patients. Therapeutic-based antibody towards CD80 (galiximab) had never been investigated in MCD or any kidney-related disease; hence, the role of CD80 is still undetermined. A new therapeutic approach towards MCD is essential to provide broader effective treatment options besides the general immunosuppressive agents with gruesome adverse effects.
Collapse
|
15
|
Moser T, Akgün K, Proschmann U, Sellner J, Ziemssen T. The role of TH17 cells in multiple sclerosis: Therapeutic implications. Autoimmun Rev 2020; 19:102647. [PMID: 32801039 DOI: 10.1016/j.autrev.2020.102647] [Citation(s) in RCA: 174] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 03/08/2020] [Indexed: 12/13/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS) where immunopathology is thought to be mediated by myelin-reactive CD4+ T helper (TH) cells. The TH cells most commonly implicated in the pathogenesis of the disease are of TH1 and TH17 lineage, which are defined by the production of interferon-γ and interleukin-17, respectively. Moreover, there is emerging evidence for the involvement of TH17.1 cells, which share the hallmarks of TH1 and TH17 subsets. In this review, we summarise current knowledge about the potential role of TH17 subsets in the initiation and progression of the disease and put a focus on their response to approved immunomodulatory MS drugs. In this regard, TH17 cells are abundant in peripheral blood, cerebrospinal fluid and brain lesions of MS patients, and their counts and inflammatory mediators are further increased during relapses. Fingolimod and alemtuzumab induce a paramount decrease in central memory T cells, which harbour the majority of peripheral TH17 cells, while the efficacy of natalizumab, dimethyl fumarate and importantly hematopoietic stem cell therapy correlates with TH17.1 cell inhibition. Interestingly, also CD20 antibodies target highly inflammatory TH cells and hamper TH17 differentiation by IL-6 reductions. Moreover, recovery rates of TH cells best correlate with long-term efficacy after therapeutical immunodepletion. We conclude that central memory TH17.1 cells play a pivotal role in MS pathogenesis and they represent a major target of MS therapeutics.
Collapse
Affiliation(s)
- Tobias Moser
- Center of Clinical Neuroscience, University Hospital Carl Gustav Carus, Dresden University of Technical, Fetscherstrasse 74, 01307 Dresden, Germany; Department of Neurology, Christian Doppler Medical Center, Paracelsus Medical University, Ignaz-Harrer-Straße 79, 5020 Salzburg, Austria
| | - Katja Akgün
- Center of Clinical Neuroscience, University Hospital Carl Gustav Carus, Dresden University of Technical, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Undine Proschmann
- Center of Clinical Neuroscience, University Hospital Carl Gustav Carus, Dresden University of Technical, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Johann Sellner
- Department of Neurology, Christian Doppler Medical Center, Paracelsus Medical University, Ignaz-Harrer-Straße 79, 5020 Salzburg, Austria; Department of Neurology, Landesklinikum Mistelbach-Gänserndorf, Liechtensteinstrasse 67, 3120 Mistelbach, Austria; Department of Neurology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Strasse 22, 81675 München, Germany
| | - Tjalf Ziemssen
- Center of Clinical Neuroscience, University Hospital Carl Gustav Carus, Dresden University of Technical, Fetscherstrasse 74, 01307 Dresden, Germany.
| |
Collapse
|
16
|
Fan NW, Dohlman TH, Foulsham W, McSoley M, Singh RB, Chen Y, Dana R. The role of Th17 immunity in chronic ocular surface disorders. Ocul Surf 2020; 19:157-168. [PMID: 32470612 DOI: 10.1016/j.jtos.2020.05.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/06/2020] [Accepted: 05/14/2020] [Indexed: 12/23/2022]
Abstract
Th17 cells have been implicated in the pathogenesis of numerous inflammatory and autoimmune conditions. At the ocular surface, Th17 cells have been identified as key effector cells in chronic ocular surface disease. Evidence from murine studies indicates that following differentiation and expansion, Th17 cells migrate from the lymphoid tissues to the eye, where they release inflammatory cytokines including, but not limited to, their hallmark cytokine IL-17A. As the acute phase subsides, a population of long-lived memory Th17 cells persist, which predispose hosts both to chronic inflammation and severe exacerbations of disease; of great interest is the small subset of Th17/1 cells that secrete both IL-17A and IFN-γ in acute-on-chronic disease exacerbation. Over the past decade, substantial progress has been made in deciphering how Th17 cells interact with the immune and neuroimmune pathways that mediate chronic ocular surface disease. Here, we review (i) the evidence for Th17 immunity in chronic ocular surface disease, (ii) regulatory mechanisms that constrain the Th17 immune response, and (iii) novel therapeutic strategies targeting Th17 cells.
Collapse
Affiliation(s)
- Nai-Wen Fan
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, 02114, USA; Department of Ophthalmology, Taipei Veterans General Hospital, Taipei, Taiwan; Faculty of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Thomas H Dohlman
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, 02114, USA
| | - William Foulsham
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, 02114, USA
| | - Matthew McSoley
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, 02114, USA; University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Rohan Bir Singh
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, 02114, USA
| | - Yihe Chen
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, 02114, USA
| | - Reza Dana
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
17
|
A prospective cross-sectional study of serum IL-17A in antisynthetase syndrome. Clin Rheumatol 2020; 39:2763-2771. [PMID: 32152919 DOI: 10.1007/s10067-020-05013-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/20/2020] [Accepted: 02/28/2020] [Indexed: 12/16/2022]
Abstract
OBJECTIVES The pro-inflammatory interleukin (IL)-17A serum has been characterized in several systemic autoimmune diseases, but not in antisynthetase syndrome (ASS). Therefore, the present study aims firstly to assess the serum level of the IL-17A in patients with ASS, comparing with healthy individuals, and secondly to analyze prospectively this IL in patients with refractory ASS undergoing rituximab treatment. MATERIALS AND METHODS A cross-sectional, single-center study that included 64 patients with ASS who were age-, gender-, and ethnicity-matched to 64 healthy individuals. Disease status was measured by the International Myositis Assessment and Clinical Studies Group (IMACS) set scores. Secondarily, the patients with refractory disease treated with rituximab were prospectively followed for 12 months. The IL-17A was assessed by the ELISA method. RESULTS The mean age of the patients was 44.8 ± 11.8 years, with a predominance of female gender and Caucasian. The median serum IL-17A level was higher in ASS patients compared with healthy individuals: 9.7 (9.1-10.4) vs. 7.7 (5.7-9.0) pg/mL, respectively, and P < 0.001. However, the demographical, clinical, and laboratory data indicates that disease status did not correlate with serum levels of the IL-17A in ASS patients. Prospectively, 16 patients received rituximab, and there was a drop of IL-17A serum level over the first year of treatment in these patients: from 9.7 (9.1-10.6) to 9.0 (8.2-9.7) pg/mL (P = 0.01). CONCLUSIONS Our study demonstrated that patients with ASS have increased serum levels of the IL-17A compared with healthy controls. In addition, the patients with refractory ASS treated with rituximab showed a reduction of the serum levels of the IL-17A. Key Points • Patients with ASS have increased serum levels of the IL-17A. • Patients with refractory ASS treated with rituximab showed a reduction of the serum levels of the IL-17A.
Collapse
|
18
|
Nasonov EL, Beketova TV, Ananyeva LP, Vasilyev VI, Solovyev SK, Avdeeva AS. PROSPECTS FOR ANTI-B-CELL THERAPY IN IMMUNO-INFLAMMATORY RHEUMATIC DISEASES. RHEUMATOLOGY SCIENCE AND PRACTICE 2019. [DOI: 10.14412/1995-4484-2019-3-40] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- E L. Nasonov
- V.A. Nasonova Research Institute of Rheumatology; I.M. Sechenov First Moscow State Medical University (Sechenov University), Ministry of Health of Russia
| | | | | | | | | | | |
Collapse
|
19
|
Skarlis C, Marketos N, Mavragani CP. Biologics in Sjögren's syndrome. Pharmacol Res 2019; 147:104389. [DOI: 10.1016/j.phrs.2019.104389] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 08/01/2019] [Accepted: 08/05/2019] [Indexed: 12/20/2022]
|
20
|
Mielle J, Tison A, Cornec D, Le Pottier L, Daien C, Pers JO. B cells in Sjögren's syndrome: from pathophysiology to therapeutic target. Rheumatology (Oxford) 2019; 60:2545-2560. [PMID: 30770916 DOI: 10.1093/rheumatology/key332] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 09/04/2018] [Indexed: 12/12/2022] Open
Abstract
Biological abnormalities associated with B lymphocytes are a hallmark of patients with primary Sjögren's syndrome. Those patients present abnormal distribution of B lymphocytes in peripheral blood and B cells in exocrine glands. B cells produce auto-antibodies, cytokines and present antigens but can also suppressive functions. In this review, we will summarize current knowledge on B cells in primary Sjögren's syndrome patients, demonstrate their critical role in the immunopathology of the disease and describe the past and current trials targeting B cells.
Collapse
Affiliation(s)
- Julie Mielle
- Departement of Rheumatology, UMR5535, Inflammation and Cancer, University of Montpellier and Teaching hospital of Montpellier, Montpellier, France
| | - Alice Tison
- UMR1227, Lymphocytes B et Autoimmunité, Université de Brest, Inserm, France.,Service de Rhumatologie, CHU de Brest, Brest, France
| | - Divi Cornec
- UMR1227, Lymphocytes B et Autoimmunité, Université de Brest, Inserm, France.,Service de Rhumatologie, CHU de Brest, Brest, France
| | | | - Claire Daien
- Departement of Rheumatology, UMR5535, Inflammation and Cancer, University of Montpellier and Teaching hospital of Montpellier, Montpellier, France
| | | |
Collapse
|
21
|
Chen Q, Yuan S, Sun H, Peng L. CD3 +CD20 + T cells and their roles in human diseases. Hum Immunol 2019; 80:191-194. [PMID: 30639700 DOI: 10.1016/j.humimm.2019.01.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 12/30/2018] [Accepted: 01/10/2019] [Indexed: 02/07/2023]
Abstract
CD3+CD20+ T cells are a population of CD3+ T cells co-expressing CD20 that make up to ∼3-5% of the CD3+ T-cell compartment in the peripheral blood of human beings. In healthy individuals, CD3+CD20+ T cells are heterogeneous for containing a lower proportion of CD4+ cells, but produce higher levels of IL-17A and/or IFN-γ than those of CD3+CD20- T cells. Recently, emerging studies have shown a pathogenic behavior of CD3+CD20+ T cells in autoimmune diseases and CD20+ T-cell malignancies, and patients with the diseases may benefit from anti-CD20 immunotherapy to deplete these cells. However, CD3+CD20+ T cells may also play a protective role in ovarian cancer and HIV infection for their strong propensity to IFN-γ production. In this review, we will describe the current knowledge about CD3+CD20+ T-cell biology, and discuss their functional roles in autoimmune diseases as well as cancer and infectious diseases.
Collapse
Affiliation(s)
- Qin Chen
- Department of Dermatology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Shunling Yuan
- East China Military Material Purchasing Bureau, Shanghai 200433, China
| | - Hongwu Sun
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China.
| | - Liusheng Peng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
22
|
|
23
|
Bertelli R, Bonanni A, Caridi G, Canepa A, Ghiggeri GM. Molecular and Cellular Mechanisms for Proteinuria in Minimal Change Disease. Front Med (Lausanne) 2018; 5:170. [PMID: 29942802 PMCID: PMC6004767 DOI: 10.3389/fmed.2018.00170] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 05/15/2018] [Indexed: 12/15/2022] Open
Abstract
Minimal Change Disease (MCD) is a clinical condition characterized by acute nephrotic syndrome, no evident renal lesions at histology and good response to steroids. However, frequent recurrence of the disease requires additional therapies associated with steroids. Such multi-drug dependence and frequent relapses may cause disease evolution to focal and segmental glomerulosclerosis (FSGS) over time. The differences between the two conditions are not well defined, since molecular mechanisms may be shared by the two diseases. In some cases, genetic analysis can make it possible to distinguish MCD from FSGS; however, there are cases of overlap. Several hypotheses on mechanisms underlying MCD and potential molecular triggers have been proposed. Most studies were conducted on animal models of proteinuria that partially mimic MCD and may be useful to study glomerulosclerosis evolution; however, they do not demonstrate a clear-cut separation between MCD and FSGS. Puromycin Aminonucleoside and Adriamycin nephrosis are models of glomerular oxidative damage, characterized by loss of glomerular basement membrane polyanions resembling MCD at the onset and, at more advanced stages, by glomerulosclerosis resembling FSGS. Also Buffalo/Mna rats present initial lesions of MCD, subsequently evolving to FSGS; this mechanism of renal damage is clearer since this rat strain inherits the unique characteristic of overexpressing Th2 cytokines. In Lipopolysaccharide nephropathy, an immunological condition of renal toxicity linked to B7-1(CD80), mice develop transient proteinuria that lasts a few days. Overall, animal models are useful and necessary considering that they reproduce the evolution from MCD to FSGS that is, in part, due to persistence of proteinuria. The role of T/Treg/Bcells on human MCD has been discussed. Many cytokines, immunomodulatory mechanisms, and several molecules have been defined as a specific cause of proteinuria. However, the hypothesis of a single cell subset or molecule as cause of MCD is not supported by research and an interactive process seems more logical. The implication or interactive role of oxidants, Th2 cytokines, Th17, Tregs, B7-1(CD80), CD40/CD40L, c-Mip, TNF, uPA/suPAR, Angiopoietin-like 4 still awaits a definitive confirmation. Whole genome sequencing studies could help to define specific genetic features that justify a definition of MCD as a “clinical-pathology-genetic entity.”
Collapse
Affiliation(s)
| | | | | | - Alberto Canepa
- Nephrology, Dialysis, Transplantation Unit, Integrated Department of Pediatrics and Hemato-Oncology Sciences, Istituto Giannina Gaslini IRCCS, Genoa, Italy
| | - G M Ghiggeri
- Laboratory of Molecular Nephrology, Genoa, Italy.,Nephrology, Dialysis, Transplantation Unit, Integrated Department of Pediatrics and Hemato-Oncology Sciences, Istituto Giannina Gaslini IRCCS, Genoa, Italy
| |
Collapse
|
24
|
Del Papa N, Vitali C. Management of primary Sjögren's syndrome: recent developments and new classification criteria. Ther Adv Musculoskelet Dis 2018; 10:39-54. [PMID: 29387177 DOI: 10.1177/1759720x17746319] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 11/12/2017] [Indexed: 12/14/2022] Open
Abstract
For many years primary Sjögren's syndrome (pSS) has been considered an orphan disease, since no specific therapies were recognized as being capable of contrasting the development and progression of this disorder. The treatment of oral and ocular features, as well as of the systemic organ involvement, has been entrusted to the joint management of different subspecialty physicians, like ophthalmologists, otolaryngologists, dentists and rheumatologists. These latter subspecialty doctors are usually more involved in the treatment of systemic extraglandular involvement and, to do it, they have long been using the conventional therapies borrowed by the treatment schedules adopted in other systemic autoimmune diseases. The increasing knowledge of the biological pathways that are operative in patients with pSS, and the parallel development of molecular biology technology, have allowed the production and availability of a number of biological agents able to positively act on different disease mechanisms, and thus are candidates for testing in therapeutic trials. Meanwhile, the scientific community has made a great effort to develop new accurate and validated classification criteria and outcome measures to be applied in the selection of patients to be included and monitored in therapeutic studies. Some of the new-generation biotechnological agents have been tested in a number of open-label and randomized controlled trials that have produced in many cases inconclusive or contradictory results. Behind the differences in trial protocols, adopted outcome measures and predefined endpoints, reasons for such unsatisfactory results can be found in the large heterogeneity of clinical subtypes in the examined cohorts. The future challenge for a substantial advancement in the therapeutic approach to pSS could be to identify the pathologic mechanisms, outcome tools and biomarkers that characterize the different subsets of the disease in order to test carefully selected target therapies with the highest probability of success in each different clinical phenotype.
Collapse
Affiliation(s)
- Nicoletta Del Papa
- Day Hospital of Rheumatology, Department of Rheumatology, ASST G. Pini-CTO, via Pini 3, 20122 Milan, Italy
| | | |
Collapse
|
25
|
Retamozo S, Flores-Chavez A, Consuegra-Fernández M, Lozano F, Ramos-Casals M, Brito-Zerón P. Cytokines as therapeutic targets in primary Sjögren syndrome. Pharmacol Ther 2017; 184:81-97. [PMID: 29092775 DOI: 10.1016/j.pharmthera.2017.10.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Primary Sjögren syndrome (SjS) is a systemic autoimmune disease that may affect 1 in 1000 people (overwhelmingly women) and that can be a serious disease with excess mortality due to severe organ-specific involvements and the development of B cell lymphoma; systemic involvement clearly marks the disease prognosis, and strongly suggests the need for closer follow-up and more robust therapeutic management. Therapy is established according to the organ involved and severity. As a rule, the management of systemic SjS should be organ-specific, with glucocorticoids and immunosuppressive agents limited to potentially-severe involvements; unfortunately, the limited evidence available for these drugs, together with the potential development of serious adverse events, makes solid therapeutic recommendations difficult. The emergence of biological therapies has increased the therapeutic armamentarium available to treat primary SjS. Biologics currently used in SjS patients are used off-label and are overwhelmingly agents targeting B cells, but the most recent studies are moving on into the evaluation of targeting specific cytokines involved in the SjS pathogenesis. The most recent etiopathogenic advances in SjS are shedding some light in the search for new highly-selective biological therapies without the adverse effects of the standard drugs currently used (corticosteroids and immunosuppressant drugs). This review summarizes the potential pharmacotherapeutic options targeting the main cytokine families involved in the etiopathogenesis of primary SjS and analyzes potential insights for developing new therapies.
Collapse
Affiliation(s)
- Soledad Retamozo
- Sjögren Syndrome Research Group (AGAUR), Laboratory of Autoimmune Diseases Josep Font, CELLEX-IDIBAPS, Spain; Hospital Privado Universitario de Córdoba, Instituto Universitario de Ciencias Biomédicas de Córdoba (IUCBC), Córdoba, Argentina; Instituto de Investigaciones en Ciencias de la Salud, Universidad Nacional de Córdoba, Consejo Nacional de Investigaciones Científicas y Técnicas (INICSA-UNC-CONICET), Córdoba, Argentina; Department of Autoimmune Diseases, ICMiD, Hospital Clínic Barcelona, Spain
| | - Alejandra Flores-Chavez
- Sjögren Syndrome Research Group (AGAUR), Laboratory of Autoimmune Diseases Josep Font, CELLEX-IDIBAPS, Spain; Biomedical Research Unit 02, Clinical Epidemiology Research Unit, UMAE, Specialties Hospital, Western Medical Center, Mexican Institute for Social Security (IMSS), Guadalajara, Mexico; Postgraduate Program of Medical Science, University Center for Biomedical Research (CUIB), University of Colima, Colima, Mexico; Department of Autoimmune Diseases, ICMiD, Hospital Clínic Barcelona, Spain
| | - Marta Consuegra-Fernández
- Immunoreceptors del Sistema Innat I Adaptatiu, Institut D'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Francisco Lozano
- Immunoreceptors del Sistema Innat I Adaptatiu, Institut D'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; Servei d'Immunologia, Centre de Diagnòstic Biomèdic, Hospital Clínic de Barcelona, Barcelona, Spain; Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain.
| | - Manuel Ramos-Casals
- Sjögren Syndrome Research Group (AGAUR), Laboratory of Autoimmune Diseases Josep Font, CELLEX-IDIBAPS, Spain; Department of Medicine, University of Barcelona, Barcelona, Spain; Department of Autoimmune Diseases, ICMiD, Hospital Clínic Barcelona, Spain.
| | - Pilar Brito-Zerón
- Sjögren Syndrome Research Group (AGAUR), Laboratory of Autoimmune Diseases Josep Font, CELLEX-IDIBAPS, Spain; Autoimmune Diseases Unit, Department of Medicine, Hospital CIMA-Sanitas, Barcelona, Spain; Department of Autoimmune Diseases, ICMiD, Hospital Clínic Barcelona, Spain
| |
Collapse
|
26
|
The value of rituximab treatment in primary Sjögren's syndrome. Clin Immunol 2017; 182:62-71. [PMID: 28478105 DOI: 10.1016/j.clim.2017.05.002] [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: 03/10/2017] [Accepted: 05/02/2017] [Indexed: 12/12/2022]
Abstract
The rationale for B cell depletion therapy with rituximab in primary Sjögren's syndrome relies upon the well-established role of B cell hyperactivity in immunopathogenesis. In line with this notion, several biomarkers of B cell activity are significantly affected by treatment, both in the target organs and periphery. In contrast to most biological outcomes, clinical outcomes are not consistent between studies. Although two large RCTs did not meet their primary endpoint, several beneficial clinical effects of treatment have been shown. As discussed in this review, differences in study design and patient characteristics could explain the variation in results. Interestingly, a newly developed composite endpoint of subjective and objective outcomes did show a significant effect of rituximab in one of the large RCTs. Response predictors need to be identified to define more targeted inclusion criteria and achieve precision medicine. The positive effects seen on biological and clinical parameters warrant future studies to investigate this promising treatment modality.
Collapse
|
27
|
Brito-Zerón P, Retamozo S, Gheitasi H, Ramos-Casals M. Treating the Underlying Pathophysiology of Primary Sjögren Syndrome: Recent Advances and Future Prospects. Drugs 2017; 76:1601-1623. [PMID: 27844414 DOI: 10.1007/s40265-016-0659-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Sjögren Syndrome (SS) is a systemic autoimmune disease with a wide clinical spectrum that extends from sicca symptoms of the mucosal surfaces to extra-glandular systemic manifestations. Understanding of the pathophysiology of primary SS has advanced over recent years, and this, in turn, has presented new targeted treatment options. We provide a brief, up-to-date description of the pathophysiology of SS and the main etiopathogenic pathways implicated in the disease process and review clinical evidence in support of new treatment options targeting these pathways, highlighting successes and failures, and concluding with a summary of gaps in knowledge and where future research should be focused. Direct and indirect B-cell targeted therapies are currently the most promising biological agents in primary SS, especially for systemic involvement, but other pathways (T-cell co-stimulation, cytokine-based therapies, intracellular pathways and gene therapies) are under development. The next 10 years may witness a disruptive therapeutic scenario in primary SS.
Collapse
Affiliation(s)
- Pilar Brito-Zerón
- Autoimmune Diseases Unit, Department of Medicine, Hospital CIMA-Sanitas, Barcelona, Spain.,Sjögren Syndrome Research Group (AGAUR), Laboratory of Autoimmune Diseases Josep Font, CELLEX-IDIBAPS, Barcelona, Spain.,Department of Autoimmune Diseases, ICMiD, Hospital Clínic, C/Villarroel, 170, 08036, Barcelona, Spain
| | - Soledad Retamozo
- Sjögren Syndrome Research Group (AGAUR), Laboratory of Autoimmune Diseases Josep Font, CELLEX-IDIBAPS, Barcelona, Spain.,Centro Médico de Córdoba, Hospital Privado, Córdoba, Argentina
| | - Hoda Gheitasi
- Sjögren Syndrome Research Group (AGAUR), Laboratory of Autoimmune Diseases Josep Font, CELLEX-IDIBAPS, Barcelona, Spain
| | - Manuel Ramos-Casals
- Sjögren Syndrome Research Group (AGAUR), Laboratory of Autoimmune Diseases Josep Font, CELLEX-IDIBAPS, Barcelona, Spain. .,Department of Autoimmune Diseases, ICMiD, Hospital Clínic, C/Villarroel, 170, 08036, Barcelona, Spain. .,Department of Medicine, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
28
|
Madanchi N, Bitzan M, Takano T. Rituximab in Minimal Change Disease: Mechanisms of Action and Hypotheses for Future Studies. Can J Kidney Health Dis 2017; 4:2054358117698667. [PMID: 28540057 PMCID: PMC5433659 DOI: 10.1177/2054358117698667] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 11/23/2016] [Indexed: 12/13/2022] Open
Abstract
Treatment with rituximab, a monoclonal antibody against the B-lymphocyte surface protein CD20, leads to the depletion of B cells. Recently, rituximab was reported to effectively prevent relapses of glucocorticoid-dependent or frequently relapsing minimal change disease (MCD). MCD is thought to be T-cell mediated; how rituximab controls MCD is not understood. In this review, we summarize key clinical studies demonstrating the efficacy of rituximab in idiopathic nephrotic syndrome, mainly MCD. We then discuss immunological features of this disease and potential mechanisms of action of rituximab in its treatment based on what is known about the therapeutic action of rituximab in other immune-mediated disorders. We believe that studies aimed at understanding the mechanisms of action of rituximab in MCD will provide a novel approach to resolve the elusive immune pathophysiology of MCD.
Collapse
Affiliation(s)
- Nima Madanchi
- Division of Nephrology, Department of Medicine, McGill University Health Centre, Montreal, Quebec, Canada
| | - Martin Bitzan
- Division of Nephrology, Department of Pediatrics, Montreal Children's Hospital, McGill University Health Centre, Montreal, Quebec, Canada
| | - Tomoko Takano
- Division of Nephrology, Department of Medicine, McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
29
|
Abstract
Primary Sjögren's syndrome (SS) is a common chronic autoimmune disease characterized by lymphocytic infiltration of exocrine glands, mainly salivary and lacrimal, resulting in oral and ocular dryness, although virtually any organ system can be affected. SS-related systemic manifestations are classified as either related to the presence of periepithelial infiltrates in exocrine and parenchymal organs or resulting from immunocomplex deposition due to B cell hyperactivity with increased risk for B cell lymphoma development. Activation of both innate and adaptive immune pathways contributes to disease pathogenesis, with prominent interferon (IFN) signatures identified in both peripheral blood and affected salivary gland tissues. Recently, LINE-1 genomic repeat elements have been proposed as potential triggers of type I IFN pathway activation in SS through activation of Toll-like receptor-dependent and -independent pathways. In view of the increasingly appreciated variability of SS, elucidation of distinct operating pathways in relation to diverse clinical phenotypes and selection of the optimal therapeutic intervention remain major challenges. Inhibition of cathepsin S molecules, blockade of costimulation through administration of abatacept and inhibitors of B7-related molecules and CD40, blockade of B cell function and B cell survival factors, and disruption of the formation of ectopic germinal centers are considered the main therapeutic targets. Well-controlled multicenter clinical trials are ongoing and data are awaited.
Collapse
Affiliation(s)
- Clio P Mavragani
- Department of Physiology, School of Medicine and Joint Academic Rheumatology Program, National and Kapodistrian University of Athens, Athens, Greece;
| |
Collapse
|
30
|
Verstappen GM, Kroese FGM, Meiners PM, Corneth OB, Huitema MG, Haacke EA, van der Vegt B, Arends S, Vissink A, Bootsma H, Abdulahad WH. B Cell Depletion Therapy Normalizes Circulating Follicular Th Cells in Primary Sjögren Syndrome. J Rheumatol 2016; 44:49-58. [PMID: 28042126 DOI: 10.3899/jrheum.160313] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2016] [Indexed: 12/14/2022]
Abstract
OBJECTIVE To assess the effect of B cell depletion therapy on effector CD4+ T cell homeostasis and its relation to objective measures of disease activity in patients with primary Sjögren syndrome (pSS). METHODS Twenty-four patients with pSS treated with rituximab (RTX) and 24 healthy controls (HC) were included. Frequencies of circulating effector CD4+ T cell subsets were examined by flow cytometry at baseline and 16, 24, 36, and 48 weeks after the first RTX infusion. Th1, Th2, follicular Th (TFH), and Th17 cells were discerned based on surface marker expression patterns. Additionally, intracellular cytokine staining was performed for interferon-γ, interleukin (IL)-4, IL-21, and IL-17 and serum levels of these cytokines were analyzed. RESULTS In patients with pSS, frequencies of circulating TFH cells and Th17 cells were increased at baseline compared with HC, whereas frequencies of Th1 and Th2 cells were unchanged. B cell depletion therapy resulted in a pronounced decrease in circulating TFH cells, whereas Th17 cells were only slightly lowered. Frequencies of IL-21-producing and IL-17-producing CD4+ T cells and serum levels of IL-21 and IL-17 were also reduced. Importantly, the decrease in circulating TFH cells was associated with lower systemic disease activity over time, as measured by the European League Against Rheumatism Sjögren's Syndrome Disease Activity Index scores and serum IgG levels. CONCLUSION B cell depletion therapy in patients with pSS results in normalization of the elevated levels of circulating TFH cells. This reduction is associated with improved objective clinical disease activity measures. Our observations illustrate the pivotal role of the crosstalk between B cells and TFH cells in the pathogenesis of pSS.
Collapse
Affiliation(s)
- Gwenny M Verstappen
- From the departments of Rheumatology and Clinical Immunology, Oral and Maxillofacial Surgery, and Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen; the Department of Pulmonary Medicine, Erasmus MC, Rotterdam, the Netherlands. .,G.M. Verstappen, MSc, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; F.G. Kroese, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; P.M. Meiners, MD, PhD, DMD, Oral and Maxillofacial Surgery, University of Groningen, University Medical Center Groningen; O.B. Corneth, PhD, Department of Pulmonary Medicine, Erasmus MC; M.G. Huitema, BSc, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; E.A. Haacke, MD, departments of Rheumatology and Clinical Immunology, and departments of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen; B. van der Vegt, MD, PhD, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen; S. Arends, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; A. Vissink, DMD, MD, PhD, Oral and Maxillofacial Surgery, University of Groningen, University Medical Center Groningen; H. Bootsma, MD, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; W.H. Abdulahad, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen.
| | - Frans G M Kroese
- From the departments of Rheumatology and Clinical Immunology, Oral and Maxillofacial Surgery, and Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen; the Department of Pulmonary Medicine, Erasmus MC, Rotterdam, the Netherlands.,G.M. Verstappen, MSc, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; F.G. Kroese, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; P.M. Meiners, MD, PhD, DMD, Oral and Maxillofacial Surgery, University of Groningen, University Medical Center Groningen; O.B. Corneth, PhD, Department of Pulmonary Medicine, Erasmus MC; M.G. Huitema, BSc, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; E.A. Haacke, MD, departments of Rheumatology and Clinical Immunology, and departments of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen; B. van der Vegt, MD, PhD, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen; S. Arends, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; A. Vissink, DMD, MD, PhD, Oral and Maxillofacial Surgery, University of Groningen, University Medical Center Groningen; H. Bootsma, MD, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; W.H. Abdulahad, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen
| | - Petra M Meiners
- From the departments of Rheumatology and Clinical Immunology, Oral and Maxillofacial Surgery, and Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen; the Department of Pulmonary Medicine, Erasmus MC, Rotterdam, the Netherlands.,G.M. Verstappen, MSc, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; F.G. Kroese, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; P.M. Meiners, MD, PhD, DMD, Oral and Maxillofacial Surgery, University of Groningen, University Medical Center Groningen; O.B. Corneth, PhD, Department of Pulmonary Medicine, Erasmus MC; M.G. Huitema, BSc, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; E.A. Haacke, MD, departments of Rheumatology and Clinical Immunology, and departments of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen; B. van der Vegt, MD, PhD, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen; S. Arends, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; A. Vissink, DMD, MD, PhD, Oral and Maxillofacial Surgery, University of Groningen, University Medical Center Groningen; H. Bootsma, MD, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; W.H. Abdulahad, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen
| | - Odilia B Corneth
- From the departments of Rheumatology and Clinical Immunology, Oral and Maxillofacial Surgery, and Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen; the Department of Pulmonary Medicine, Erasmus MC, Rotterdam, the Netherlands.,G.M. Verstappen, MSc, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; F.G. Kroese, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; P.M. Meiners, MD, PhD, DMD, Oral and Maxillofacial Surgery, University of Groningen, University Medical Center Groningen; O.B. Corneth, PhD, Department of Pulmonary Medicine, Erasmus MC; M.G. Huitema, BSc, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; E.A. Haacke, MD, departments of Rheumatology and Clinical Immunology, and departments of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen; B. van der Vegt, MD, PhD, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen; S. Arends, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; A. Vissink, DMD, MD, PhD, Oral and Maxillofacial Surgery, University of Groningen, University Medical Center Groningen; H. Bootsma, MD, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; W.H. Abdulahad, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen
| | - Minke G Huitema
- From the departments of Rheumatology and Clinical Immunology, Oral and Maxillofacial Surgery, and Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen; the Department of Pulmonary Medicine, Erasmus MC, Rotterdam, the Netherlands.,G.M. Verstappen, MSc, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; F.G. Kroese, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; P.M. Meiners, MD, PhD, DMD, Oral and Maxillofacial Surgery, University of Groningen, University Medical Center Groningen; O.B. Corneth, PhD, Department of Pulmonary Medicine, Erasmus MC; M.G. Huitema, BSc, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; E.A. Haacke, MD, departments of Rheumatology and Clinical Immunology, and departments of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen; B. van der Vegt, MD, PhD, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen; S. Arends, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; A. Vissink, DMD, MD, PhD, Oral and Maxillofacial Surgery, University of Groningen, University Medical Center Groningen; H. Bootsma, MD, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; W.H. Abdulahad, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen
| | - Erlin A Haacke
- From the departments of Rheumatology and Clinical Immunology, Oral and Maxillofacial Surgery, and Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen; the Department of Pulmonary Medicine, Erasmus MC, Rotterdam, the Netherlands.,G.M. Verstappen, MSc, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; F.G. Kroese, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; P.M. Meiners, MD, PhD, DMD, Oral and Maxillofacial Surgery, University of Groningen, University Medical Center Groningen; O.B. Corneth, PhD, Department of Pulmonary Medicine, Erasmus MC; M.G. Huitema, BSc, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; E.A. Haacke, MD, departments of Rheumatology and Clinical Immunology, and departments of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen; B. van der Vegt, MD, PhD, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen; S. Arends, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; A. Vissink, DMD, MD, PhD, Oral and Maxillofacial Surgery, University of Groningen, University Medical Center Groningen; H. Bootsma, MD, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; W.H. Abdulahad, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen
| | - Bert van der Vegt
- From the departments of Rheumatology and Clinical Immunology, Oral and Maxillofacial Surgery, and Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen; the Department of Pulmonary Medicine, Erasmus MC, Rotterdam, the Netherlands.,G.M. Verstappen, MSc, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; F.G. Kroese, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; P.M. Meiners, MD, PhD, DMD, Oral and Maxillofacial Surgery, University of Groningen, University Medical Center Groningen; O.B. Corneth, PhD, Department of Pulmonary Medicine, Erasmus MC; M.G. Huitema, BSc, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; E.A. Haacke, MD, departments of Rheumatology and Clinical Immunology, and departments of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen; B. van der Vegt, MD, PhD, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen; S. Arends, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; A. Vissink, DMD, MD, PhD, Oral and Maxillofacial Surgery, University of Groningen, University Medical Center Groningen; H. Bootsma, MD, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; W.H. Abdulahad, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen
| | - Suzanne Arends
- From the departments of Rheumatology and Clinical Immunology, Oral and Maxillofacial Surgery, and Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen; the Department of Pulmonary Medicine, Erasmus MC, Rotterdam, the Netherlands.,G.M. Verstappen, MSc, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; F.G. Kroese, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; P.M. Meiners, MD, PhD, DMD, Oral and Maxillofacial Surgery, University of Groningen, University Medical Center Groningen; O.B. Corneth, PhD, Department of Pulmonary Medicine, Erasmus MC; M.G. Huitema, BSc, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; E.A. Haacke, MD, departments of Rheumatology and Clinical Immunology, and departments of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen; B. van der Vegt, MD, PhD, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen; S. Arends, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; A. Vissink, DMD, MD, PhD, Oral and Maxillofacial Surgery, University of Groningen, University Medical Center Groningen; H. Bootsma, MD, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; W.H. Abdulahad, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen
| | - Arjan Vissink
- From the departments of Rheumatology and Clinical Immunology, Oral and Maxillofacial Surgery, and Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen; the Department of Pulmonary Medicine, Erasmus MC, Rotterdam, the Netherlands.,G.M. Verstappen, MSc, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; F.G. Kroese, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; P.M. Meiners, MD, PhD, DMD, Oral and Maxillofacial Surgery, University of Groningen, University Medical Center Groningen; O.B. Corneth, PhD, Department of Pulmonary Medicine, Erasmus MC; M.G. Huitema, BSc, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; E.A. Haacke, MD, departments of Rheumatology and Clinical Immunology, and departments of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen; B. van der Vegt, MD, PhD, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen; S. Arends, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; A. Vissink, DMD, MD, PhD, Oral and Maxillofacial Surgery, University of Groningen, University Medical Center Groningen; H. Bootsma, MD, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; W.H. Abdulahad, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen
| | - Hendrika Bootsma
- From the departments of Rheumatology and Clinical Immunology, Oral and Maxillofacial Surgery, and Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen; the Department of Pulmonary Medicine, Erasmus MC, Rotterdam, the Netherlands.,G.M. Verstappen, MSc, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; F.G. Kroese, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; P.M. Meiners, MD, PhD, DMD, Oral and Maxillofacial Surgery, University of Groningen, University Medical Center Groningen; O.B. Corneth, PhD, Department of Pulmonary Medicine, Erasmus MC; M.G. Huitema, BSc, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; E.A. Haacke, MD, departments of Rheumatology and Clinical Immunology, and departments of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen; B. van der Vegt, MD, PhD, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen; S. Arends, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; A. Vissink, DMD, MD, PhD, Oral and Maxillofacial Surgery, University of Groningen, University Medical Center Groningen; H. Bootsma, MD, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; W.H. Abdulahad, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen
| | - Wayel H Abdulahad
- From the departments of Rheumatology and Clinical Immunology, Oral and Maxillofacial Surgery, and Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen; the Department of Pulmonary Medicine, Erasmus MC, Rotterdam, the Netherlands.,G.M. Verstappen, MSc, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; F.G. Kroese, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; P.M. Meiners, MD, PhD, DMD, Oral and Maxillofacial Surgery, University of Groningen, University Medical Center Groningen; O.B. Corneth, PhD, Department of Pulmonary Medicine, Erasmus MC; M.G. Huitema, BSc, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; E.A. Haacke, MD, departments of Rheumatology and Clinical Immunology, and departments of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen; B. van der Vegt, MD, PhD, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen; S. Arends, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; A. Vissink, DMD, MD, PhD, Oral and Maxillofacial Surgery, University of Groningen, University Medical Center Groningen; H. Bootsma, MD, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen; W.H. Abdulahad, PhD, departments of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen
| |
Collapse
|
31
|
Benchabane S, Boudjelida A, Toumi R, Belguendouz H, Youinou P, Touil-Boukoffa C. A case for IL-6, IL-17A, and nitric oxide in the pathophysiology of Sjögren's syndrome. Int J Immunopathol Pharmacol 2016; 29:386-97. [PMID: 27207443 DOI: 10.1177/0394632016651273] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 04/26/2016] [Indexed: 12/31/2022] Open
Abstract
Sjögren's syndrome (SS) is an autoimmune epithelitis characterized by mononuclear cell (MNC) infiltration of the lacrimal and salivary glands (SG), as well as the presence of serum autoantibodies. This condition is a growing public health concern in Algeria. Herein, we sought to determine if the levels of interleukin (IL)-6, IL-17A, and nitric oxide (NO), were correlated with the extent of MNC infiltration. The expression of inducible NO synthase (NOS2) and CD68 was measured in the SG of all patients, but not in those of the normal controls (NCs). We included 44 primary Sjögren's syndrome (pSS) patients and 15 NCs in this study; we found that the expression of NOS2 and CD68 was elevated in all of the SG of SS patients. Additionally, the serum and saliva levels of IL-6, IL-17A, and NO were higher in the pSS patients, compared with the NCs. Furthermore, the NOS2-induced excess NO was associated with the extent of the MNC infiltration, and thereby with tissue injury. It is also important to note that there were correlations between the levels of IL-6, IL-17A, and NO. Such findings indicate that through the effects of NO, IL-17A participates in the pathophysiology of the disease. With the purpose of improving both the diagnosis and prognosis, IL-6, IL-17A, and NO should be assayed in the serum and saliva of patients suspected of SS.
Collapse
Affiliation(s)
- Sarah Benchabane
- University of Sciences, Laboratory of Cellular and Molecular Biology, Cyokines and NO Synthases Group, Faculty of Biological Sciences, Algeria
| | | | - Ryma Toumi
- University of Sciences, Laboratory of Cellular and Molecular Biology, Cyokines and NO Synthases Group, Faculty of Biological Sciences, Algeria
| | - Houda Belguendouz
- University of Sciences, Laboratory of Cellular and Molecular Biology, Cyokines and NO Synthases Group, Faculty of Biological Sciences, Algeria
| | - Pierre Youinou
- Laboratory of Excellence (Labex) IGO, and INSERM ERI29, European University of Brit-tany, Brest, France
| | - Chafia Touil-Boukoffa
- University of Sciences, Laboratory of Cellular and Molecular Biology, Cyokines and NO Synthases Group, Faculty of Biological Sciences, Algeria
| |
Collapse
|