AUTREV-01775; No of Pages 7 Autoimmunity Reviews xxx (2015) xxx–xxx
Contents lists available at ScienceDirect
Autoimmunity Reviews journal homepage: www.elsevier.com/locate/autrev
Review
Systemic sclerosis: New evidence re-enforces the role of B cells Lazaros I. Sakkas a,b,⁎, Dimitrios P. Bogdanos a,c a b c
Department of Rheumatology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa 41 110, Greece Center for Molecular Medicine, Old Dominion University, Norfolk, VA, USA Division of Transplantation Immunology and Mucosal Biology, Kings College School of Medicine, London SE5 9RS, UK
a r t i c l e
i n f o
Article history: Received 4 October 2015 Accepted 17 October 2015 Available online xxxx Keywords: Systemic sclerosis Autoantibodies B cells Bregs Fibrosis Vasoconstriction
a b s t r a c t Systemic sclerosis (SSc) is characterized by widespread fibrosis, microangiopathy (vasospasms and stenosis), and formation of autoantibodies. T cell activation has been shown to contribute to fibrosis and microvasculopathy in SSc. However, recent evidence suggests that B cells are also likely to contribute in the pathogenesis of the disease. B cells are hyperactivated in SSc, as indicated by the overexpression of the stimulatory CD19 receptor and impairment of the inhibitory CD22 receptor. They lead to the production of many autoantibodies, some of which induce collagen production and vasoconstriction. They promote fibroblast collagen production through cell contact. Furthermore, B cells can function as antigen-presenting cells to T cells and induce dendritic cell maturation that promotes profibrotic Th2 response. Lately, interleukin (IL)-10-producing B regulatory cells, which induce generation of T regulatory cells and can ameliorate autoimmune diseases, were found to be reduced in SSc, favoring autoaggression of B cells in this disease. Finally, B cell depletion with rituximab improves or stabilizes skin fibrosis and lung function. These finding suggest that new therapeutic strategies targeting B cell function(s) can be developed for SSc. © 2015 Elsevier B.V. All rights reserved.
Contents 1. Introduction . . . . . . . . . . . . . . . . . . . . . . . 2. B cells are hyperactivated in SSc . . . . . . . . . . . . . . 3. B cells can produce profibrotic cytokines . . . . . . . . . . 4. Autoantibodies promote fibrosis/inflammation in SSc . . . . 5. IL-10-producing regulatory B cells (Bregs) are decreased in SSc 6. B cells promote profibrotic Th2 response . . . . . . . . . . 7. B cells in models of human SSc . . . . . . . . . . . . . . 8. B cell-based treatment in SSc . . . . . . . . . . . . . . . 9. Future perspectives . . . . . . . . . . . . . . . . . . . Take-home messages . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
1. Introduction Systemic sclerosis (SSc) is chronic multisystem disease characterized by widespread fibrosis, autoantibodies, and microangiopathy. Fibrosis involves the skin and internal organs, including lung, gastrointestinal tract, and heart. Microangiopathy is characterized by vasospastic episodes and fibrointimal proliferation causing vascular ⁎ Corresponding author at: Department of Rheumatology, Medical School, University of Thessaly, Biopolis, Larissa 41 110, Greece. E-mail address:
[email protected] (L.I. Sakkas).
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
0 0 0 0 0 0 0 0 0 0 0
stenosis that leads to digital ulcers, pulmonary arterial hypertension, and scleroderma renal crisis. The etiopathogenesis of SSc is incompletely understood [1,2]. Environmental factors play a major role [3], since in a twin study the concordance rate of SSc in monozygotic twins is low and equal to that in dizygotic twin [4]. Fibrosis of the skin and internal organs is the hallmark of the disease, and for years the pathogenesis of SSc has been evolved around fibroblasts. However, the evolution of SSc in the skin, an easily accessible organ to study, has shown that there are lymphocytic infiltrates early in the disease process before even histological fibrosis [5]. In the early inflammatory skin stage, there is also apoptosis of endothelial cells. In
http://dx.doi.org/10.1016/j.autrev.2015.10.005 1568-9972/© 2015 Elsevier B.V. All rights reserved.
Please cite this article as: Sakkas LI, Bogdanos DP, Systemic sclerosis: New evidence re-enforces the role of B cells, Autoimmun Rev (2015), http:// dx.doi.org/10.1016/j.autrev.2015.10.005
2
L.I. Sakkas, D.P. Bogdanos / Autoimmunity Reviews xxx (2015) xxx–xxx
fact, endothelial cells were the first cell type to undergo apoptosis in UCD 200/206 chicken, an avian model of scleroderma [6]. Autoantibodies and microangiopathy, as detected by nailfoldcapillaroscopy, may occur years before clinical skin fibrosis and are now included in the new classification criteria for SSc [7–9]. These findings alluded to the possibility that immune cells might activate fibroblasts to produce excess collagen. T cells and macrophages are the predominant mononuclear cell infiltrates in the skin of SSc [5,10,11]. T cells in SSc skin are activated. They express the early activation antigen CD69 [11], and suction blisters in SSc skin contain elevated levels of soluble IL-2 receptor [12]. T cells in SSc are of TH2 type producing the profibrotic cytokines IL-4 and IL-13 [1]. The finding that T cells in SSc skin lesions exhibit oligoclonal expansion and certain T cell clones persist in skin lesions over time indicates an antigen-driven immune response and further re-enforces the concept of T cell involvement in SSc pathogenesis [1,13]. In recent years, the role of B cells in the pathogenesis of SSc has become increasingly apparent. B cells can promote fibrosis by cytokines, autoantibodies, and cell–cell contact.
Table 1 Autoantibody targets in patients with SSc. Autoantibody target
Comments
Ref
Topoisomerase I Centromere RNA polymerase III Th/To Platelet-derived growth factor receptor (PDGFR) Endothelial cell
Associated with dcSSc Associated with lcSSc Associated with dcSSc Associated with scleroderma renal crisis Stimulatory, inducing fibroblast activation and collagen production Induce endothelial cell activation and/or apoptosis Activate endothelial cells Stimulatory, strong predictors of digital ulcers Stimulatory, associated with pre-eclampsia and malignant hypertension
[32] [33] [35] [36] [37]
Mimicry targets, cross-reactive with endothelial cells Inhibit the inhibitory CD22 signals Inhibit collagenolyticactivity, thus increased intracellular matrix Increase fibroblast collagen production
[52]
ICAM-1 Endothelin type A receptor (ETAR) Angiotensin II type 1 receptor (AT1R) hCMV-derived UL94 CD22 Matrix metalloproteinase I
Fibrillin
[41] [42] [47] [44,48]
[23] [39]
[50]
2. B cells are hyperactivated in SSc B cells infiltrates in SSc lesions are variable[14–18], yet there is convincing evidence for the participation of B cells in the pathogenesis of the disease. B cells in SSc are hyperactive, as exemplified by the presence of hyper-γ-globulinemia, autoantibodies, and increased serum free light chains of immunoglobulins [19]. B cells from patients with SSc overexpress the B cell stimulatory receptor CD19 by 54% in patients with early SSc and by 28% in patients with long-standing disease [20]. Overexpression of CD19 in SSc, although to a lesser degree, was also reported earlier [21]. It should be mentioned that small increase (15–29%) in B cell CD19 expression in transgenic mice–induced autoantibody production [21]. B cell activating factor (BAFF), a B cell survival factor, is increased in SSc [22]. In contrast, the function of CD22, an inhibitory B cell molecule, is inhibited by anti-CD22 autoantibodies present in patients with SSc [23]. B cells are also activated in a model of systemic sclerosis, namely, chronic graft-versus-host disease (cGVHD), a condition that develops after allogeneic hematopoietic stem cell transplantation, and shares clinical and serological features with human SSc [24] and in tight-skin (TSK) mice, another model of SSc [25]. 3. B cells can produce profibrotic cytokines Activated B cells can produce profibrotic cytokines IL-6, and TGFβ. IL-6 can induce fibroblast collagen production [26] through the bone morphogenetic protein (BMP) antagonist gremlin and the canonical TGFβ signaling [27]. Serum IL-6 was elevated in SSc patients [28–30], and skin IL-6 expression was also increased [30]. TGFb, increased in scleroderma, is a powerful profibrotic cytokine [31]. 4. Autoantibodies promote fibrosis/inflammation in SSc The hyperactivity of B cells in SSc is exemplified by the presence of a plethora of autoantibodies. Some of autoantibodies found in patients with SSc promote fibrosis and/or pro-inflammatory response. These autoantibodies, and their specific action, are shown in Table 1. Antitopoisomerase I [32] and anti-centromere antibodies are diseasespecific antibodies associated with diffuse cutaneous (dcSSc) and limited cutaneous disease (lcSSc), respectively. More interestingly, anti-Topoisomerase I and anti-centromere antibodies were associated more closely with clinical manifestations than cutaneous SSc subsets were [33]. Anti-topoisomerase I antibodies recognize their target that is released from apoptotic endothelial cells and binds to fibroblast membrane [32]. Anti-fibroblast antibodies induce fibroblast production of
chemokines CCL2 (monocyte chemoattractant protein-1) and CXCL8 (interleukin-8) [34]. Anti-RNA polymerase III antibodies are associated with dcSSc [35] and anti-Th/To antibodies are associated with lcSSc and scleroderma renal crisis in some studies [36]. Autoantibodies against platelet-derived growth factor receptor (PDGFR) stimulated activation and collagen production in normal human fibroblasts [37]. Stimulatory anti-PDGFR autoantibodies inducing collagen production were also found cGVHD [38]. Anti-matrix metalloproteinase(MMP)-1 antibodies and anti-MMP-3 antibodies, found in SSc, are functional and inhibit MMP-1 and MMP-3 collagenase activity, respectively, thus reducing extracellular matrix breakdown and promoting accumulation of extracellular matrix [39,40]. Anti-endothelial cell antibodies cause endothelial cell apoptosis [41] and/or endothelial cell activation. Also, anti-ICAM-1 antibodies, frequently detected in SSc patients, cause increased endothelial cell production of reactive oxygen species (ROS) and expression of VCAM-1 [42]. Anti-endothelin-1 type A receptor (ETAR) autoantibodies and anti-angiotensin II type 1 receptor (AT1R) autoantibodies, detected in most SSc patients, are agonist antibodies and cause fibrosis and vasoconstriction [43–45]. They activate vascular endothelial cell production of TGFβ (a powerful profibrotic cytokine), IL-8, and vascular cell adhesion molecule-1 (VCAM-1) [43,46]. SSc-IgG with anti-ETAR and anti-AT1R antibodies, but not normal IgG, when injected to mice, caused obliterative vasculopathy [45]. Anti-ETAR autoantibodies are strong predictors of digital ulcers in patients with SSc [47]. Anti-AT1R autoantibodies are associated with hypertensive entities, such as pre-eclampsia, and severe kidney transplant rejection with malignant hypertension [44]. Furthermore, pregnant mice develop pre-eclampsia when injected with affinity-purified anti-AT1R autoantibodies from women with pre-eclampsia [48]. It should be noted that angiotensin II increased fibroblast production of TGFβ and collagen via its type 1 receptor [49]. Anti-fibrillin antibodies activate fibroblast collagen production [50]. Another autoantibody found in SSc targets CD22, a major inhibitory B cell receptor. Anti-CD22 autoantibodies inhibit CD22 signals and are associated with enhanced skin fibrosis, as detected by the Rodnan skin score [23] (Fig. 1 and Table 1). The targets of autoantibodies in SSc, apart from contributing to disease pathogenesis, may also serve as mimicry targets of immune response to foreign agents. For instance, anti-topoisomerase I monoclonal antibodies recognize a pentapeptide of topoisomerase I from SSc patients that shares homology with human CMV-derived UL70 protein [51]. Also, many SSc patients have antibodies against human CMVderived UL94 protein that shares homology with NAG-2 (tetraspan novel antigen-2), expressed on endothelial cell surface. Furthermore,
Please cite this article as: Sakkas LI, Bogdanos DP, Systemic sclerosis: New evidence re-enforces the role of B cells, Autoimmun Rev (2015), http:// dx.doi.org/10.1016/j.autrev.2015.10.005
L.I. Sakkas, D.P. Bogdanos / Autoimmunity Reviews xxx (2015) xxx–xxx
Breg
B cell
3
B cell anti -CD22
a-fibrillin
a-ETAR a-AT1R
a-PDGFR
a-ECs a-ICAM1 a-MMP
Inhibition of inhibitory CD22
a-fib fibroblast
ECs
CXCL8 TGFβ VCAM-1
collagen
apoptosis ROS VCAM-1
CCL2 CXCL8
vasoconstriction
Extracellular Matrix
Fig. 1. Autoantibodies promote B cell hyperactivity, fibrosis, and vasoconstriction. In a hypothetical model, Bregs inhibit B cell hyperactivity through Tregs. ECs: endothelial cells; fib: fibroblast; MMP: metalloproteinase I; ETAR: endothelin A receptor; AT1T: angiotensin II type 1 receptor; ROS: reactive oxygen species.
anti-UL94 peptide antibodies bind to NAG-2 on endothelial cells and induce their apoptosis [52]. 5. IL-10-producing regulatory B cells (Bregs) are decreased in SSc Regulatory B cells (Bregs) are recently described subsets of B cells that downregulate inflammation, by inhibiting the differentiation of TH1 and TH17 cells and by inducing Tregs, and ameliorate autoimmune diseases through cytokines, such as IL10, TGFβ, and antigendependent-, CD40-dependent interaction with T cells and the help of IL-21 or IL-35[53–55]. Although the phenotypes of Bregs are still evolving, a subset of Bregs mediates their regulatory function through IL-10 production (B10 cells) [56]. B10 cells are reported to be mainly
CD24(hi) CD38(-) CD27(+) memory B regs (blue)
included within CD19 + CD24highCD38high (transitional Bregs) or CD19 + CD24highCD27+ (memory Bregs) phenotypes [57,58]. New data showing that B10 cells are decreased in SSc re-enforce the role of B cells in this disease. In SSc, we showed that phenotypic memory Bregs were decreased particularly in long-standing disease and in SScassociated pulmonary fibrosis (SSc-PF) [20]. IL-10-producing Bregs were also impaired particularly in SSc-PF (Fig. 2). Furthermore, after B cell receptor and TLR9 stimulation of naive and memory B cells STAT3 and p38MAPK activation was impaired. Another study also reported reduced IL-10-producing Bregs in SSc and an inverse correlation between Bregs and the titre of anti-topoisomerase I antibodies and anticentromere antibodies [59]. Similarly, in cGVHD, B cells were found to be activated [24] and Bregs to be deficient [60].
CD24(hi) CD38(hi) CD27(-) transitional B regs cells (green)
CD19
SSC
CD24
Healthy control
CD38
FSC
CD27
SSC
CD24
CD19
earlySSc
FSC
CD38
CD27
Fig. 2. Flow cytometry profile of memory (blue color) and transitional (green color) in a representative healthy individual and a patient with early SSc. Bregs are decreased in early SSc.
Please cite this article as: Sakkas LI, Bogdanos DP, Systemic sclerosis: New evidence re-enforces the role of B cells, Autoimmun Rev (2015), http:// dx.doi.org/10.1016/j.autrev.2015.10.005
4
L.I. Sakkas, D.P. Bogdanos / Autoimmunity Reviews xxx (2015) xxx–xxx
Induction of autoantibody production Anti-Topo I Anti-CEN
Naïve B cells
Naïve T cells
Naïve B cells
Effector Effector B cells T cells
Naïve T cells
Effector Effector B cells T cells
Bregs Decrease of Bregs and Tregs Tregs
Healthy Immunological Tolerance
SSc at diagnosis SSc with pulmonary fibrosis Loss of Immunological Tolerance
Fig. 3. Suggestive status of B cells and T cells in health and disease. Effector B cells and T cells are increased in SSc whereas Bregs and Tregs are decreased, particularly in long-standing disease and SSc-associated pulmonary fibrosis. The decreased number/function of Bregs correlates with high titre of disease-specific autoantibodies.
Reduced IL-10-producing Bregs were also associated with increased autoantibody titres in ANCA-associated vasculitis. In the latter disease, IL-10-producing Bregs were reduced in active disease and normalized in disease remission which was accompanied with reduction of ANCA titres [61]. Based on these findings, we propose a scheme of Bregs involvement in autoantibody production (Figs. 3 and 4). 6. B cells promote profibrotic Th2 response B cells, through cell contact, induce dendritic cell maturation that promotes profibrotic Th2 differentiation [62] (Fig. 5). They can also serve as antigen-presenting cells [63]. Although still evolving, there is increasing evidence suggesting that antigen presentation by B cells interplaying with dendritic cells favors Th2 response [64–66]. In human x-linked agammaglobulinemia, a primary deficiency of the B cell compartment, T cell response is preferentially Th1 [67].
Breg
7. B cells in models of human SSc There are various animal models of SSc that offer the sequence of events leading to fibrosis, as well as access to study the effect of manipulation of various cells and cytokines on animal manifestations. As already mentioned, in cGVHDB cells are activated [24] and Bregs are deficient [60]. Furthermore, Bregs derived from donor suppress cGVHD in mice [68]. Serum IL-6 was increased in murine cGVHD [69]. In tight-skin (TSK) mice, a genetic model of SSc characterized by skin fibrosis and autoantibodies, B cell activation is important for autoantibody production and fibrosis [25]. CD19 is overexpressed whereas CD22 function is impaired in TSK mice [21,25,70]. Small increase (15–29%) in the expression of B cell CD19 results in spontaneous autoantibody production [21,70]. In contrast, CD19 deficiency significantly decreased skin fibrosis and CD19 loss completely abrogated autoantibody production in TSK mice [25]. Serum BAFF levels were increased
Breg
CD4 T cell
CD4 T cell
Th2
B cell Treg
Th2
AutoAbs
B cell
AutoAbs
Treg
Healthy Individual
Systemic Sclerosis
Fig. 4. Bregs maintain Tregs which in turn inhibit activated T cells. In a hypothetical scheme, this leads to downregulation of B cells with subsequent reduction of autoantibodies production. In SSc, B regs are deficient and inadequate to sustain Tregs. This in turn facilitates Th2 activation and autoantibodies production.
Please cite this article as: Sakkas LI, Bogdanos DP, Systemic sclerosis: New evidence re-enforces the role of B cells, Autoimmun Rev (2015), http:// dx.doi.org/10.1016/j.autrev.2015.10.005
L.I. Sakkas, D.P. Bogdanos / Autoimmunity Reviews xxx (2015) xxx–xxx Activated B cell
DC
Th2
5
It is worth mentioning the therapeutic efficacy of high-dose intravenous immunoglobulin (IVIg) in SSc ([83] and reviewed in Cantarini et al [84]). IVIg has multiple actions on different immune cells and mediators including its effect through anti-idiotypic antibodies [85]. All evidence outlined above supports a role for B cell involvement in SSc (Table 2).
IL-13
9. Future perspectives
IL-4
Fibroblast activation Collagen accumulation
fibrosis Fig. 5. Activated B cells through cell contact promote dendritic cell (DC) maturation that favors Th2 response and leads to fibrosis.
in TSK mice, whereas a BAFF antagonist prevented skin fibrosis and autoantibody production [71]. Another model of SSc, bleomycin-induced fibrosis, is caused by subcutaneous injection of bleomycin and characterized by skin and lung fibrosis and autoantibodies. In this model, bleomycin induced hyaluronan production which activated B cells mainly through tolllike receptor 4, whereas CD19 deficiency resulted in attenuation of fibrosis and autoantibody production [72]. A similar effect of CD19 deficiency was found in a bleomycin-induced lung fibrosis mouse model, caused by intratracheal injection of bleomycin [73]. A new mice model of SSc develops after injection of topoisomerase I with complete Freud's adjuvant [74]. These mice develop skin and lung fibrosis along with Th2/Th17 response and increased IL-6 production. Furthermore, loss of IL-6 expression improved skin and lung fibrosis [74].
The finding of increased B cell CD19 expression in early SSc along with greatly reduced IL10-producing Bregs favors autoaggression of B cells and makes B cells an attractive therapeutic target for this disease. It also offers new insights for treatment strategies. For instance, Bregs derived from donor suppress the cGVHD in mice [68]. Also, ex vivo provision of CD40 and IL-21 receptor signals induced the development and expansion of B10 cells that, when infused into mice with established EAE, a model for multiple sclerosis, greatly inhibited the symptoms of the disease [86]. Similarly, ex vivo expansion of Bregs with IL-35 suppressed uveitis in mice by inhibiting TH1 and TH17 responses and promoting the expansion of Tregs [55]. Therefore, B cells are becoming an attractive target for intervention with different approaches in this frequently devastating disease. Take-home messages • Systemic sclerosis is a multisystem disease characterized by widespread fibrosis, microvasculopathy, and autoantibodies. • Autoantibodies may appear years before clinical disease and inflammatory infiltrates in skin appear before histological fibrosis • B cells may participate in disease pathogenesis through cytokines, autoantibodies, and B cell contact with fibroblasts or dendritic cells • The immunoregulatory Bregs producing IL-10 are decreased in SSc. • B cell depleting treatment decreased skin fibrosis and stabilized/ improved lung function.
8. B cell-based treatment in SSc References B cell depleting therapy with anti-CD20 monoclonal antibody (rituximab) in SSc case series improved skin fibrosis and stabilized/ improved lung involvement (function) [15,16,75–79]. The improvement of skin fibrosis by rituximab was associated with attenuation of PDGFR in the skin [80]. Similarly, B cell depletion with anti-CD20 monoclonal antibody in neonatal TSK mice reduced skin fibrosis [81]. Loss of CD19 abrogated autoantibody production and attenuated skin fibrosis [25], whereas BAFF antagonist attenuated skin fibrosis in TSK mice [71]. Bregs derived from donor suppress cGVHD in a mouse model of SSc [68]. Neutralization of IL-6 by tocilizumab (anti-IL-6 receptor monoclonal antibody) improved skin thickness in SSc patients in an observational study [82]. Similarly, anti-IL-6R treatment or immunization with IL-6 peptide reduced skin thickness in bleomycin-induced mouse model of SSc [30]. Anti-IL-6R monoclonal antibody also attenuated murine cGVHD [69]. Table 2 Evidence for B cell involvement in the pathogenesis of SSc. B cells are hyperactivated. B cells can produce cytokines that promote fibrosis/inflammation. B cells produce autoantibodies that promote fibrosis/vasoconstriction/ inflammation. B cell-fibroblast contact promotes fibrosis. B cells through dendritic cell maturation promotes profibrotic TH2 response. Bregs are reduced in SSc. B cell-eliminating therapies reduce/stabilize fibrosis in SSc. B cells are critical for skin fibrosis and autoantibody production in human cGVHD, and animal models of SSc. TSK: tight-skin mice; cGVHD: chronic graft-versus-host disease.
[1] Sakkas LI, Chikanza IC, Platsoucas CD. Mechanisms of disease: the role of immune cells in the pathogenesis of systemic sclerosis. Nat Clin Pract Rheumatol 2006;2: 679–85. [2] Elhai M, Avouac J, Kahan A, Allanore Y. Systemic sclerosis at the crossroad of polyautoimmunity. Autoimmun Rev 2013;12:1052–7. [3] Marie I, Gehanno JF, Bubenheim M, et al. Prospective study to evaluate the association between systemic sclerosis and occupational exposure and review of the literature. Autoimmun Rev 2014;13:151–6. [4] Feghali-Bostwick C, Medsger Jr TA, Wright TM. Analysis of systemic sclerosis in twins reveals low concordance for disease and high concordance for the presence of antinuclear antibodies. Arthritis Rheum 2003;48:1956–63. [5] Prescott RJ, Freemont AJ, Jones CJ, Hoyland J, Fielding P. Sequential dermal microvascular and perivascular changes in the development of scleroderma. J Pathol 1992; 166:255–63. [6] Sgonc R, Gruschwitz MS, Boeck G, Sepp N, Gruber J, Wick G. Endothelial cell apoptosis in systemic sclerosis is induced by antibody-dependent cell-mediated cytotoxicity via CD95. Arthritis Rheum 2000;43:2550–62. [7] van den Hoogen F, Khanna D, Fransen J, et al. 2013 classification criteria for systemic sclerosis: an American College of Rheumatology/European League against Rheumatism collaborative initiative. Arthritis Rheum 2013;65:2737–47. [8] van den Hoogen F, Khanna D, Fransen J, et al. 2013 classification criteria for systemic sclerosis: an American college of rheumatology/European league against rheumatism collaborative initiative. Ann Rheum Dis 2013;72:1747–55. [9] Sakkas LI, Simopoulou T, Katsiari C, Bogdanos D, Chikanza IC. Early systemic sclerosis-opportunities for treatment. Clin Rheumatol 2015;34:1327–31. [10] Kraling BM, Maul GG, Jimenez SA. Mononuclear cellular infiltrates in clinically involved skin from patients with systemic sclerosis of recent onset predominantly consist of monocytes/macrophages. Pathobiology 1995;63:48–56. [11] Kalogerou A, Gelou E, Mountantonakis S, Settas L, Zafiriou E, Sakkas L. Early T cell activation in the skin from patients with systemic sclerosis. Ann Rheum Dis 2005;64: 1233–5. [12] Sondergaard K, Stengaard-Pedersen K, Zachariae H, Heickendorff L, Deleuran M, Deleuran B. Soluble intercellular adhesion molecule-1 (sICAM-1) and soluble interleukin-2 receptors (sIL-2R) in scleroderma skin. Br J Rheumatol 1998;37: 304–10. [13] Sakkas LI, Xu B, Artlett CM, Lu S, Jimenez SA, Platsoucas CD. Oligoclonal T cell expansion in the skin of patients with systemic sclerosis. J Immunol 2002;168:3649–59.
Please cite this article as: Sakkas LI, Bogdanos DP, Systemic sclerosis: New evidence re-enforces the role of B cells, Autoimmun Rev (2015), http:// dx.doi.org/10.1016/j.autrev.2015.10.005
6
L.I. Sakkas, D.P. Bogdanos / Autoimmunity Reviews xxx (2015) xxx–xxx
[14] Whitfield ML, Finlay DR, Murray JI, et al. Systemic and cell type-specific gene expression patterns in scleroderma skin. Proc Natl Acad Sci U S A 2003;100:12319–24. [15] Daoussis D, Liossis SN, Tsamandas AC, et al. Experience with rituximab in scleroderma: results from a 1-year, proof-of-principle study. Rheumatology (Oxford) 2010; 49:271–80. [16] Bosello S, De Santis M, Lama G, et al. B cell depletion in diffuse progressive systemic sclerosis: safety, skin score modification and IL-6 modulation in an up to thirty-six months follow-up open-label trial. Arthritis Res Ther 2010;12:R54. [17] Lafyatis R, O'Hara C, Feghali-Bostwick CA, Matteson E. B cell infiltration in systemic sclerosis-associated interstitial lung disease. Arthritis Rheum 2007;56:3167–8. [18] Hussein MR, Hassan HI, Hofny ER, et al. Alterations of mononuclear inflammatory cells, CD4/CD8+ T cells, interleukin 1beta, and tumour necrosis factor alpha in the bronchoalveolar lavage fluid, peripheral blood, and skin of patients with systemic sclerosis. J Clin Pathol 2005;58:178–84. [19] Lanteri A, Sobanski V, Langlois C, et al. Serum free light chains of immunoglobulins as biomarkers for systemic sclerosis characteristics, activity and severity. Autoimmun Rev 2014;13:974–80. [20] Mavropoulos A, Simopoulou T, Varna A, et al. B regulatory cells are decreased and functionally impaired in patients with systemic sclerosis. Arthritis Rheum 2015. http://dx.doi.org/10.1002/art.39437 (in press). [21] Sato S, Hasegawa M, Fujimoto M, Tedder TF, Takehara K. Quantitative genetic variation in CD19 expression correlates with autoimmunity. J Immunol 2000;165: 6635–43. [22] Matsushita T, Hasegawa M, Yanaba K, Kodera M, Takehara K, Sato S. Elevated serum BAFF levels in patients with systemic sclerosis: enhanced BAFF signaling in systemic sclerosis B lymphocytes. Arthritis Rheum 2006;54:192–201. [23] Odaka M, Hasegawa M, Hamaguchi Y, et al. Autoantibody-mediated regulation of B cell responses by functional anti-CD22 autoantibodies in patients with systemic sclerosis. Clin Exp Immunol 2010;159:176–84. [24] Allen JL, Fore MS, Wooten J, et al. B cells from patients with chronic GVHD are activated and primed for survival via BAFF-mediated pathways. Blood 2012;120: 2529–36. [25] Saito E, Fujimoto M, Hasegawa M, et al. CD19-dependent B lymphocyte signaling thresholds influence skin fibrosis and autoimmunity in the tight-skin mouse. J Clin Invest 2002;109:1453–62. [26] Duncan MR, Berman B. Stimulation of collagen and glycosaminoglycan production in cultured human adult dermal fibroblasts by recombinant human interleukin 6. J Invest Dermatol 1991;97:686–92. [27] O'Reilly S, Ciechomska M, Cant R, van Laar JM. Interleukin-6 (IL-6) trans signaling drives a STAT3-dependent pathway that leads to hyperactive transforming growth factor-beta (TGF-beta) signaling promoting SMAD3 activation and fibrosis via Gremlin protein. J Biol Chem 2014;289:9952–60. [28] Sato S, Hasegawa M, Takehara K. Serum levels of interleukin-6 and interleukin-10 correlate with total skin thickness score in patients with systemic sclerosis. J Dermatol Sci 2001;27:140–6. [29] Scala E, Pallotta S, Frezzolini A, et al. Cytokine and chemokine levels in systemic sclerosis: relationship with cutaneous and internal organ involvement. Clin Exp Immunol 2004;138:540–6. [30] Desallais L, Avouac J, Frechet M, et al. Targeting IL-6 by both passive or active immunization strategies prevents bleomycin-induced skin fibrosis. Arthritis Res Ther 2014;16:R157. [31] Snapper CM, Waegell W, Beernink H, Dasch JR. Transforming growth factor-beta 1 is required for secretion of IgG of all subclasses by LPS-activated murine B cells in vitro. J Immunol 1993;151:4625–36. [32] Henault J, Robitaille G, Senecal JL, Raymond Y. DNA topoisomerase I binding to fibroblasts induces monocyte adhesion and activation in the presence of antitopoisomerase I autoantibodies from systemic sclerosis patients. Arthritis Rheum 2006;54:963–73. [33] Walker UA, Tyndall A, Czirjak L, et al. Clinical risk assessment of organ manifestations in systemic sclerosis: a report from the EULAR Scleroderma Trials And Research group database. Ann Rheum Dis 2007;66:754–63. [34] Fineschi S, Goffin L, Rezzonico R, et al. Antifibroblast antibodies in systemic sclerosis induce fibroblasts to produce profibrotic chemokines, with partial exploitation of toll-like receptor 4. Arthritis Rheum 2008;58:3913–23. [35] Villalta D, Imbastaro T, Di Giovanni S, et al. Diagnostic accuracy and predictive value of extended autoantibody profile in systemic sclerosis. Autoimmun Rev 2012;12: 114–20. [36] Mahler M, Fritzler MJ, Satoh M. Autoantibodies to the mitochondrial RNA processing (MRP) complex also known as Th/To autoantigen. Autoimmun Rev 2015;14:254–7. [37] Baroni SS, Santillo M, Bevilacqua F, et al. Stimulatory autoantibodies to the PDGF receptor in systemic sclerosis. N Engl J Med 2006;354:2667–76. [38] Svegliati S, Olivieri A, Campelli N, et al. Stimulatory autoantibodies to PDGF receptor in patients with extensive chronic graft-versus-host disease. Blood 2007;110: 237–41. [39] Sato S, Hayakawa I, Hasegawa M, Fujimoto M, Takehara K. Function blocking autoantibodies against matrix metalloproteinase-1 in patients with systemic sclerosis. J Invest Dermatol 2003;120:542–7. [40] Nishijima C, Hayakawa I, Matsushita T, et al. Autoantibody against matrix metalloproteinase-3 in patients with systemic sclerosis. Clin Exp Immunol 2004; 138:357–63. [41] Bordron A, Dueymes M, Levy Y, et al. The binding of some human antiendothelial cell antibodies induces endothelial cell apoptosis. J Clin Invest 1998;101:2029–35. [42] Wolf SI, Howat S, Abraham DJ, Pearson JD, Lawson C. Agonistic anti-ICAM-1 antibodies in scleroderma: activation of endothelial pro-inflammatory cascades. Vascul Pharmacol 2013;59:19–26.
[43] Kill A, Tabeling C, Undeutsch R, et al. Autoantibodies to angiotensin and endothelin receptors in systemic sclerosis induce cellular and systemic events associated with disease pathogenesis. Arthritis Res Ther 2014;16:R29. [44] Xia Y, Kellems RE. Angiotensin receptor agonistic autoantibodies and hypertension: preeclampsia and beyond. Circ Res 2013;113:78–87. [45] Becker MO, Kill A, Kutsche M, et al. Vascular receptor autoantibodies in pulmonary arterial hypertension associated with systemic sclerosis. Am J Respir Crit Care Med 2014;190:808–17. [46] Riemekasten G, Philippe A, Nather M, et al. Involvement of functional autoantibodies against vascular receptors in systemic sclerosis. Ann Rheum Dis 2011;70:530–6. [47] Avouac J, Riemekasten G, Meune C, Ruiz B, Kahan A, Allanore Y. Autoantibodies against Endothelin 1 Type A receptor are strong predictors of digital ulcers in systemic sclerosis. J Rheumatol 2015;42:1801–7. [48] Zhou CC, Zhang Y, Irani RA, et al. Angiotensin receptor agonistic autoantibodies induce pre-eclampsia in pregnant mice. Nat Med 2008;14:855–62. [49] Kawaguchi Y, Takagi K, Hara M, et al. Angiotensin II in the lesional skin of systemic sclerosis patients contributes to tissue fibrosis via angiotensin II type 1 receptors. Arthritis Rheum 2004;50:216–26. [50] Zhou X, Tan FK, Milewicz DM, Guo X, Bona CA, Arnett FC. Autoantibodies to fibrillin1 activate normal human fibroblasts in culture through the TGF-beta pathway to recapitulate the “scleroderma phenotype”. J Immunol 2005;175:4555–60. [51] Muryoi T, Kasturi KN, Kafina MJ, et al. Antitopoisomerase I monoclonal autoantibodies from scleroderma patients and tight skin mouse interact with similar epitopes. J Exp Med 1992;175:1103–9. [52] Lunardi C, Bason C, Navone R, et al. Systemic sclerosis immunoglobulin G autoantibodies bind the human cytomegalovirus late protein UL94 and induce apoptosis in human endothelial cells. Nat Med 2000;6:1183–6. [53] Flores-Borja F, Bosma A, Ng D, et al. CD19 + CD24hiCD38hi B cells maintain regulatory T cells while limiting TH1 and TH17 differentiation. Sci Transl Med 2013;5 [173ra123]. [54] Shen P, Roch T, Lampropoulou V, et al. IL-35-producing B cells are critical regulators of immunity during autoimmune and infectious diseases. Nature 2014;507:366–70. [55] Wang RX, Yu CR, Dambuza IM, et al. Interleukin-35 induces regulatory B cells that suppress autoimmune disease. Nat Med 2014;20:633–41. [56] Tedder TF. B10 cells: a functionally defined regulatory B cell subset. J Immunol 2015; 194:1395–401. [57] Blair PA, Norena LY, Flores-Borja F, et al. CD19(+)CD24(hi)CD38(hi) B cells exhibit regulatory capacity in healthy individuals but are functionally impaired in systemic Lupus Erythematosus patients. Immunity 2010;32:129–40. [58] Iwata Y, Matsushita T, Horikawa M, et al. Characterization of a rare IL-10-competent B-cell subset in humans that parallels mouse regulatory B10 cells. Blood 2011;117: 530–41. [59] Matsushita T, Hamaguchi Y, Hasegawa M, Takehara K, Fujimoto M. Decreased levels of regulatory B cells in patients with systemic sclerosis: association with autoantibody production and disease activity. Rheumatology (Oxford) 2015 (Epub ahead of print) PMID:26350483. [60] Khoder A, Sarvaria A, Alsuliman A, et al. Regulatory B cells are enriched within the IgM memory and transitional subsets in healthy donors but are deficient in chronic GVHD. Blood 2014;124:2034–45. [61] Aybar LT, McGregor JG, Hogan SL, et al. Reduced CD5(+) CD24(hi) CD38(hi) and interleukin-10(+) regulatory B cells in active anti-neutrophil cytoplasmic autoantibody-associated vasculitis permit increased circulating autoantibodies. Clin Exp Immunol 2015;180:178–88. [62] Maddur MS, Sharma M, Hegde P, et al. Human B cells induce dendritic cell maturation and favour Th2 polarization by inducing OX-40 ligand. Nat Commun 2014;5: 4092. [63] Beland K, Marceau G, Labardy A, Bourbonnais S, Alvarez F. Depletion of B cells induces remission of autoimmune hepatitis in mice through reduced antigen presentation and help to T cells. Hepatology 2015;62:1511–23. [64] Crawford A, Macleod M, Schumacher T, Corlett L, Gray D. Primary T cell expansion and differentiation in vivo requires antigen presentation by B cells. J Immunol 2006;176:3498–506. [65] Leon B, Ballesteros-Tato A, Lund FE. Dendritic cells and B cells: unexpected partners in Th2 development. J Immunol 2014;193:1531–7. [66] Dwyer DF, Woodruff MC, Carroll MC, Austen KF, Gurish MF. B cells regulate CD4+ T cell responses to papain following B cell receptor-independent papain uptake. J Immunol 2014;193:529–39. [67] Amedei A, Romagnani C, Benagiano M, et al. Preferential Th1 profile of T helper cell responses in X-linked (Bruton's) agammaglobulinemia. Eur J Immunol 2001;31: 1927–34. [68] Le Huu D, Matsushita T, Jin G, et al. Donor-derived regulatory B cells are important for suppression of murine sclerodermatous chronic graft-versus-host disease. Blood 2013;121:3274–83. [69] Le Huu D, Matsushita T, Jin G, et al. IL-6 blockade attenuates the development of murine sclerodermatous chronic graft-versus-host disease. J Invest Dermatol 2012;132: 2752–61. [70] Asano N, Fujimoto M, Yazawa N, et al. B Lymphocyte signaling established by the CD19/CD22 loop regulates autoimmunity in the tight-skin mouse. Am J Pathol 2004;165:641–50. [71] Matsushita T, Fujimoto M, Hasegawa M, et al. BAFF antagonist attenuates the development of skin fibrosis in tight-skin mice. J Invest Dermatol 2007;127:2772–80. [72] Yoshizaki A, Iwata Y, Komura K, et al. CD19 regulates skin and lung fibrosis via Tolllike receptor signaling in a model of bleomycin-induced scleroderma. Am J Pathol 2008;172:1650–63. [73] Komura K, Yanaba K, Horikawa M, et al. CD19 regulates the development of bleomycin-induced pulmonary fibrosis in a mouse model. Arthritis Rheum 2008; 58:3574–84.
Please cite this article as: Sakkas LI, Bogdanos DP, Systemic sclerosis: New evidence re-enforces the role of B cells, Autoimmun Rev (2015), http:// dx.doi.org/10.1016/j.autrev.2015.10.005
L.I. Sakkas, D.P. Bogdanos / Autoimmunity Reviews xxx (2015) xxx–xxx [74] Asano Y, Sato S. Animal models of scleroderma: current state and recent development. Curr Rheumatol Rep 2013;15:382. [75] Jordan S, Distler JH, Maurer B, et al. Effects and safety of rituximab in systemic sclerosis: an analysis from the European Scleroderma Trial and Research (EUSTAR) group. Ann Rheum Dis 2015;74:1188–94. [76] Giuggioli D, Lumetti F, Colaci M, Fallahi P, Antonelli A, Ferri C. Rituximab in the treatment of patients with systemic sclerosis. Our experience and review of the literature. Autoimmun Rev 2015;14:1072–8. [77] Smith V, Van Praet JT, Vandooren B, et al. Rituximab in diffuse cutaneous systemic sclerosis: an open-label clinical and histopathological study. Ann Rheum Dis 2010; 69:193–7. [78] Lafyatis R, Kissin E, York M, et al. B cell depletion with rituximab in patients with diffuse cutaneous systemic sclerosis. Arthritis Rheum 2009;60:578–83. [79] McGonagle D, Tan AL, Madden J, et al. Successful treatment of resistant sclerodermaassociated interstitial lung disease with rituximab. Rheumatology (Oxford) 2008;47: 552–3. [80] Daoussis D, Tsamandas AC, Liossis SN, et al. B-cell depletion therapy in patients with diffuse systemic sclerosis associates with a significant decrease in PDGFR
[81]
[82]
[83]
[84]
[85] [86]
7
expression and activation in spindle-like cells in the skin. Arthritis Res Ther 2012;14:R145. Hasegawa M, Hamaguchi Y, Yanaba K, et al. B-lymphocyte depletion reduces skin fibrosis and autoimmunity in the tight-skin mouse model for systemic sclerosis. Am J Pathol 2006;169:954–66. Shima Y, Kuwahara Y, Murota H, et al. The skin of patients with systemic sclerosis softened during the treatment with anti-IL-6 receptor antibody tocilizumab. Rheumatology (Oxford) 2010;49:2408–12. Molina V, Haj-Yahia S, Solodeev I, Levy Y, Blank M, Shoenfeld Y. Immunomodulation of experimental pulmonary fibrosis by intravenous immunoglobulin (IVIG). Autoimmunity 2006;39:711–7. Cantarini L, Rigante D, Vitale A, et al. Intravenous immunoglobulins (IVIG) in systemic sclerosis: a challenging yet promising future. Immunol Res 2015;61: 326–37. Blank M, Bashi T, Shoenfeld Y. Idiotype-specific intravenous immunoglobulin (IVIG) for therapy of autoimmune diseases. Methods Mol Biol 2014;1060:353–61. Yoshizaki A, Miyagaki T, DiLillo DJ, et al. Regulatory B cells control T-cell autoimmunity through IL-21-dependent cognate interactions. Nature 2012;491:264–8.
Please cite this article as: Sakkas LI, Bogdanos DP, Systemic sclerosis: New evidence re-enforces the role of B cells, Autoimmun Rev (2015), http:// dx.doi.org/10.1016/j.autrev.2015.10.005