Length Defective regulation of autoreactive IL-6-producing transitional B lymphocytes is associated with disease in patients with systemic sclerosis

Background: Systemic Sclerosis (SSc) has the highest case-specific mortality of any rheumatic disease and has no effective therapy. A clear manifestation of SSc is the presence of auto-antibodies. However, the origin of autoantibody-producing B lymphocytes and mechanisms of their activation, auto-antibody production and role remain unclear. Objective: To identify mechanisms that contribute to pathogenic B cell generation, involvement in SSc and assess the altered distribution and function of B cells in patients. Methods: Multi-colour flow cytometry was used to determine B cell subset distribution, cytokine production and tolerance induction in SSc patients and healthy controls. Cytokine production following stimulation of the cells ex vivo was carried out by multiplex analysis. Results: The study reveals a range of defects in B lymphocyte tolerance and cytokine production in SSc. Evidence is provided for altered distribution of transitional B cell subsets, increased production of IL-6 and IL-8 and defective tolerance induction in SSc B cells. In addition, the study reveals that B cells in SSc have a reduced ability to produce IL-10 when stimulated through innate immune pathways. In contrast to healthy individuals, tolerance checkpoints in SSc patients fail to suppress the emergence of B cells that produce autoantibodies with specificity to the Scl-70 antigen, a specificity strongly associated with SSc. These defects are paralleled by altered intracellular signalling and apoptosis following B cell receptor (BCR) engagement. A cc ep te d A rt ic le This article is protected by copyright. All rights reserved. Conclusion: The study provides new insights into mechanisms underlying defective B lymphocyte responses in patients with SSc and their contribution to pathology. INTRODUCTION Systemic sclerosis (SSc) is a severe inflammatory disease characterized by excessive extracellular matrix (ECM) deposition in the skin and visceral organs (1). It has complex pathogenesis with two major hallmarks: autoimmunity and inflammation leading to widespread damage to blood vessels and progressive interstitial and perivascular fibrosis (2). A key feature of autoimmunity in SSc is high levels of auto-antibodies (auto-Abs) to nuclear proteins including topoisomerase I enzyme, centromere and RNA polymerase and endothelial cell and platelet derived growth factor receptor (1, 3). Evidence for pathogenic roles of these auto-Abs comes from studies revealing the association between their specificity and which tissues and organs are involved. Importantly also, these auto-Abs appear prior to disease onset. Furthermore, B lymphocytes accumulate at sites of disease, around small vessels in the skin (4) and in alveolar interstitium in patients with lung involvement (5). In addition to producing auto-Abs, B lymphocytes contribute to fibrosis through producing interleukin 6 (IL-6) (6). The role played by B lymphocytes in SSc is supported by studies of disease pathogenesis in mouse models (7-9) and the success of treating patients with rituximab, a chimeric monoclonal antibody against human CD20 (10, 11). In the tight-skin mouse model of SSc that is associated with constitutive CD19-mediated signaling (7, 12), inhibition of CD19 expression abrogates auto-Ab production, IL-6 production by B cells and ameliorate skin fibrosis (7, 8). Similar effects were noted when CD19 expression was suppressed in the bleomycin-treated mouse model of SSc (9). Furthermore, SSc patients with interstitial lung A cc ep te d A rt ic le This article is protected by copyright. All rights reserved. and diffuse disease who are positive for anti-Scl70 auto-Abs benefit significantly from treatment with rituximab (10). Thus, no patient receiving rituximab exhibited lung function deterioration, whereas 5 out of 6 placebo-treated patients had a worsening of their disease. Additionally, skin thickening, collagen deposition in the skin and health quality improved in rituximab-treated patients but not in placebo-treated patients (10-14). Furthermore, rituximab reduced the level of plasma IL-6, activity index, depleted skin B lymphocytes and reduced dermal myofibroblasts and hyalinised collagen in the skin (15). The cause of defective B cell responses in SSc remains unclear. However, defective tolerance and deregulation of autoreactive B cell responses are potential causes of B cell-mediated pathology. High affinity self-reactive B cells are normally deleted in the bone marrow but some that recognize self with low affinity or those that do not encounter self-antigens in the bone marrow escape censure and migrate to the periphery as transitional B cells. Migrant “transitional” B cells go through series of tolerance checkpoints and maturational phases to become mature B cells. This pathway was initially described in mice as immature B cells transiting to the spleen to mature into to B cell receptor(BCR) responsive cells to antigen engagement (16). Murine transitional B cells are distinguishable from mature cells based on the level of CD23, CD21 and the developmental marker CD24 expression (17). In humans, recent studies from a number of laboratories including ours have revealed that transitional human B cells (CD24CD38) do not constitute a single population but can be divided into 4 subsets (18). These subsets have subtle differences in maturational and tolerance status and capacity to produce IL-10 and IL-6 (18, 19). The current study was carried out to explore if defects in the maturation of, or tolerance induction in B cells at the transitional stage could relate to the emergence of high affinity pathogenic B cells in patients. Transitional B cells were studied for their ability to produce IL-6 as its production is a key mechanism by which B cells could promote fibrosis (6-8). In addition, the potential of transitional B cells in SSc to A cc ep te d A rt ic le This article is protected by copyright. All rights reserved. produce IL-10 was studied to assess their ability to be involved in immune regulation (19, 20). We used a multi-colour flow cytometry and a secretome profiling approach to characterize the distribution and functional characteristics of B cells in SSc to identify potential pathways through which pathogenic B cells could emerge and contribute to pathogenesis. METHODS Patients All patients fulfilled the 2013 EULAR/ACR criteria for SSc (21). Disease subsets were defined as patients with limited cutaneous SSc (lcSSc) when skin thickening was present distal to elbows and knees and as diffuse cutaneous SSc (dsSSc) when skin thickening affected both distal and proximal areas. Blood samples from 88 patients with SSc attending the clinics were obtained for the purpose of this study after their informed consents. Patients with interstitial lung disease were diagnosed based on characteristic changes visualised by high-resolution computed tomography (CT). Pulmonary arterial hypertension was diagnosed by right heart catheterization with mean pulmonary artery pressure of ≥25 mm Hg and normal pulmonary capillary wedge pressure. Scleroderma renal crisis (SRC) was defined as newonset systemic hypertension >150/85 mm Hg and a documented decrease in estimated glomerular filtration rate of ≥30% or confirmed features on renal biopsy (22). Blood samples from 17 healthy controls (HCs) (mean age 40.2±11.3 years; 25-60 years) were also included for comparisons. Some of the tested samples from both patients and controls were studied on multiple occasions. The study was approved by the London-Hampstead National Research Ethics Service Committee (REC reference: 6398) and was conducted in compliance with the Helsinki Declaration of 2013. A cc ep te d A rt ic le This article is protected by copyright. All rights reserved. B lymphocyte enrichment B lymphocytes were enriched from whole blood by negative selection using the EasySep Human B Cell Enrichment Kit (StemCell Technologies, Grenoble, France). All experiments presented in this study were carried out using freshly-enriched B lymphocytes from blood samples taken on the same day. The protocol of B cell enrichment involves using a cocktail of monoclonal antibodies (mAbs) with dual specificity for non-B cell components of blood mononuclear cells (MNCs) and red blood cells (RBCs). The cocktail aggregates all non-B cells with RBCs and these are then separated on Ficoll-Paque. The protocol is fast and purity of enriched B cells is >95%. Cell staining and flow cytometry Enriched B cells were stained with combinations of fluorochrome-conjugated antibodies (18). mAbs were purchased from the suppliers indicated as follows: APC-eFlour780-conjugated anti-human CD10 (Clone SN5c), PE-Cy5.5-anti-human CD19 (Clone: HIB19), PE-Cy7-antihuman CD32 (Clone 6C4), FITC-anti-human IgD (Clone: IA6-2), eFlour 450-anti-human CD21 (Clone: HB5) and PE-anti-human CD24 (Clone: eBioSN3) mAbs were all from eBioscience. Brilliant Violet 605-anti-human IgM (Clone: MHM-88), brilliant Violet 785anti-human CD19 (Clone: HIB19) and brilliant Violet 711-anti-human CD27 (Clone: O323) were from BioLegend. PerCP-Cy5.5-anti-CD38 mAb (Clone: HIT2) was from BD Biosciences. For intracellular cytokines, stained cells were fixed, permeabilized and stained with either APC-anti-human IL-6 mAb (Clone: MQ2-13A5), APC-anti-human IL-10 mAb (Clone: JES3-9D7) (both from BD Bioscience) and were assessed by LSR-Fortessa FACS machine and FacsDiva software.

[1]  C. Denton,et al.  UK Scleroderma Study Group (UKSSG) guidelines on the diagnosis and management of scleroderma renal crisis. , 2016, Clinical and experimental rheumatology.

[2]  K. Chakravarty,et al.  BSR and BHPR guideline for the treatment of systemic sclerosis. , 2016, Rheumatology.

[3]  J. Pers,et al.  In-depth characterization of CD24(high)CD38(high) transitional human B cells reveals different regulatory profiles. , 2016, The Journal of allergy and clinical immunology.

[4]  A. Strasser,et al.  NFκB1 is essential to prevent the development of multiorgan autoimmunity by limiting IL-6 production in follicular B cells , 2016, Journal of Experimental Medicine.

[5]  D. Huscher,et al.  Effects and safety of rituximab in systemic sclerosis: an analysis from the European Scleroderma Trial and Research (EUSTAR) group , 2014, Annals of the rheumatic diseases.

[6]  K. Koelsch,et al.  B-Cell ELISPOT: For the Identification of Antigen-Specific Antibody-Secreting Cells. , 2015, Methods in molecular biology.

[7]  Oliver Distler,et al.  2013 classification criteria for systemic sclerosis: an American College of Rheumatology/European League against Rheumatism collaborative initiative. , 2013, Arthritis and rheumatism.

[8]  S. Alzabin,et al.  Incomplete response of inflammatory arthritis to TNFα blockade is associated with the Th17 pathway , 2012, Annals of the rheumatic diseases.

[9]  D. Gray,et al.  B cell depletion therapy ameliorates autoimmune disease through ablation of IL-6–producing B cells , 2012, The Journal of experimental medicine.

[10]  M. Neurath,et al.  IL-6 signaling in autoimmunity, chronic inflammation and inflammation-associated cancer. , 2011, Cytokine & growth factor reviews.

[11]  B. Berman Rituximab in diffuse cutaneous systemic sclerosis: an open-label clinical and histopathological study , 2011 .

[12]  D. Isenberg,et al.  CD19(+)CD24(hi)CD38(hi) B cells exhibit regulatory capacity in healthy individuals but are functionally impaired in systemic Lupus Erythematosus patients. , 2010, Immunity.

[13]  S. Tangye,et al.  Differential expression of CD21 identifies developmentally and functionally distinct subsets of human transitional B cells. , 2010, Blood.

[14]  C. Kalogeropoulou,et al.  Experience with rituximab in scleroderma: results from a 1-year, proof-of-principle study , 2009, Rheumatology.

[15]  T. Tsubata,et al.  The Development and Function of Regulatory B Cells Expressing IL-10 (B10 Cells) Requires Antigen Receptor Diversity and TLR Signals1 , 2009, The Journal of Immunology.

[16]  S. Lundy Killer B lymphocytes: the evidence and the potential , 2009, Inflammation Research.

[17]  P. Merkel,et al.  B cell depletion with rituximab in patients with diffuse cutaneous systemic sclerosis. , 2009, Arthritis and rheumatism.

[18]  T. Ohkawara,et al.  IL-6 blockade inhibits the induction of myelin antigen-specific Th17 cells and Th1 cells in experimental autoimmune encephalomyelitis , 2008, Proceedings of the National Academy of Sciences.

[19]  M. Fujimoto,et al.  Immunopathology and Infectious Disease CD 19 Regulates Skin and Lung Fibrosis via Toll-Like Receptor Signaling in a Model of Bleomycin-Induced Scleroderma , 2010 .

[20]  D. Gray,et al.  TLR-mediated stimulation of APC: Distinct cytokine responses of B cells and dendritic cells , 2007, European journal of immunology.

[21]  E. Matteson,et al.  B cell infiltration in systemic sclerosis-associated interstitial lung disease. , 2007, Arthritis and rheumatism.

[22]  G. Kelsoe,et al.  T-Independent Activation-Induced Cytidine Deaminase Expression, Class-Switch Recombination, and Antibody Production by Immature/Transitional 1 B Cells1 , 2007, The Journal of Immunology.

[23]  L. Wysocki,et al.  Identification of anergic B cells within a wild-type repertoire. , 2006, Immunity.

[24]  M. Fujimoto,et al.  B-lymphocyte depletion reduces skin fibrosis and autoimmunity in the tight-skin mouse model for systemic sclerosis. , 2006, The American journal of pathology.

[25]  H. Weiner,et al.  Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells , 2006, Nature.

[26]  S. Tangye,et al.  Expansion of Functionally Immature Transitional B Cells Is Associated with Human-Immunodeficient States Characterized by Impaired Humoral Immunity1 , 2006, The Journal of Immunology.

[27]  M. Fujimoto,et al.  B lymphocytes and systemic sclerosis , 2005, Current opinion in rheumatology.

[28]  J. Varga,et al.  Scleroderma: from cell and molecular mechanisms to disease models. , 2005, Trends in immunology.

[29]  C. Goodnow,et al.  Cellular and genetic mechanisms of self tolerance and autoimmunity , 2005, Nature.

[30]  V. Pascual,et al.  Defective B cell tolerance checkpoints in systemic lupus erythematosus , 2005, The Journal of experimental medicine.

[31]  E. Meffre,et al.  Impaired early B cell tolerance in patients with rheumatoid arthritis , 2005, The Journal of experimental medicine.

[32]  M. Fujimoto,et al.  B Lymphocyte signaling established by the CD19/CD22 loop regulates autoimmunity in the tight-skin mouse. , 2004, The American journal of pathology.

[33]  David Botstein,et al.  Systemic and cell type-specific gene expression patterns in scleroderma skin , 2003, Proceedings of the National Academy of Sciences of the United States of America.

[34]  T. Medsger,et al.  Analysis of systemic sclerosis in twins reveals low concordance for disease and high concordance for the presence of antinuclear antibodies. , 2003, Arthritis and rheumatism.

[35]  Ruslan Medzhitov,et al.  Toll Pathway-Dependent Blockade of CD4+CD25+ T Cell-Mediated Suppression by Dendritic Cells , 2003, Science.

[36]  H. Ploegh,et al.  Predominant Autoantibody Production by Early Human B Cell Precursors , 2003 .

[37]  P. Ohashi,et al.  Making and breaking tolerance. , 2002, Current opinion in immunology.

[38]  David Gray,et al.  B cells regulate autoimmunity by provision of IL-10 , 2002, Nature Immunology.

[39]  M. Fujimoto,et al.  CD19-dependent B lymphocyte signaling thresholds influence skin fibrosis and autoimmunity in the tight-skin mouse. , 2002, The Journal of clinical investigation.

[40]  D. Rawlings,et al.  Transitional B Lymphocyte Subsets Operate as Distinct Checkpoints in Murine Splenic B Cell Development1 , 2002, The Journal of Immunology.

[41]  S. Shinton,et al.  Resolution of Three Nonproliferative Immature Splenic B Cell Subsets Reveals Multiple Selection Points During Peripheral B Cell Maturation1 , 2001, The Journal of Immunology.

[42]  P. Sideras,et al.  B Cell Development in the Spleen Takes Place in Discrete Steps and Is Determined by the Quality of B Cell Receptor–Derived Signals , 1999, The Journal of experimental medicine.

[43]  G. Köhler,et al.  Transitional B cells are the target of negative selection in the B cell compartment , 1995, The Journal of experimental medicine.

[44]  W. Earnshaw,et al.  Autoantibodies to topoisomerase I (Scl-70): analysis by gel diffusion, immunoblot, and enzyme-linked immunosorbent assay. , 1990, Clinical immunology and immunopathology.

[45]  T. Medsger,et al.  Scleroderma (systemic sclerosis): classification, subsets and pathogenesis. , 1988, The Journal of rheumatology.