Summary
Systemic lupus erythematosus (SLE) is a common autoimmune disorder with a complex and poorly understood immunopathogenesis. However, a pathogenic role for the T helper type 17 (Th17) axis was demonstrated by many studies, while regulatory T cells (Tregs) were shown to mediate protection. Recently, we and others characterized a novel and independent T cell population expressing both the Treg characteristic transcription factor forkhead box protein 3 (FoxP3) and the Th17‐defining retinoic acid receptor‐related orphan nuclear receptor γt (RORγt). Studies in a model of acute glomerulonephritis unveiled potent regulatory, but also proinflammatory, functions of RORγt+FoxP3+ Tregs. This bi‐functional nature prompted us to suggest the name ‘biTregs’. Importantly, the pathogenic biTreg effects were dependent upon expression of RORγt. We thus aimed to evaluate the contribution of RORγt+FoxP3+ biTregs to pristane‐induced SLE and explored the therapeutic potential of interference with RORγt activation. Our analyses revealed expansion of IL‐17 producing biTregs in a distinctive time–course and organ‐specific pattern, coincident with the development of autoimmunity and tissue injury. Importantly, specific ablation of RORγt activation in endogenous biTregs resulted in significant amelioration of pristane‐induced pulmonary vasculitis and lupus nephritis. As potential mechanisms underlying the observed protection, we found that secretion of IL‐17 by biTregs was abrogated completely in FoxP3Cre × RORCfl/fl mice. Furthermore, Tregs showed a more activated phenotype after cell‐specific inactivation of RORγt signalling. Finally, and remarkably, biTregs were found to potently suppress anti‐inflammatory Th2 immunity in a RORγt‐dependent manner. Our study thus identifies biTregs as novel players in SLE and advocates RORγt‐directed interventions as promising therapeutic strategies.
Keywords: immunology, lupus nephritis, lymphocytes, systemic lupus, transcription factors
Introduction
Systemic lupus erythematosus (SLE) is a complex and relatively common autoimmune disorder, which can affect multiple organs including skin, joints, lungs, kidneys and the central nervous system 1, 2. As SLE causes high morbidity and mortality in a rather young collective of patients, the search for new therapeutic strategies is a priority for scientists worldwide 3, 4. Despite intensive research, the events which lead to development of SLE and eventually cause the associated organ pathologies still remain widely elusive 5, 6. However, a central role for CD4+ T helper cells in disease pathogenesis has been demonstrated by multiple studies in mice and humans, including our own 7, 8, 9, 10, 11. Recently, we and others particularly highlighted the importance of the T helper type 17/interleukin‐17 (Th17/IL‐17) axis for systemic autoimmunity and lupus nephritis 10, 12, 13. Importantly, mice deficient in IL‐17A or F were shown to be protected from disease in three independent studies, using the murine model of pristane‐induced SLE 14, 15, 16. Furthermore, recent studies could identify a specialized regulatory T cell subset, which is tailor‐made for down‐regulation of pathogenic Th17 responses 17, 18. Lack of these T regulatory 17 (Treg17) cells resulted in much increased SLE associated mortality and organ pathologies 19. A crucial role for both, Th17 cells and Tregs in SLE is therefore evident. Interestingly, we and others could recently describe an intriguing novel T cell subset, expressing the unusual combination of the Treg master transcription factor forkhead box protein 3 (FoxP3), together with the Th17 master regulator retinoic acid receptor‐related orphan nuclear receptor γt (RORγt) 20, 21, 22. Functional characterization revealed that these RORγt FoxP3 double‐positive T cells displayed both regulatory as well as proinflammatory functions in a model of acute crescentic glomerulonephritis 22. RORγt+FoxP3+ T cells were shown to produce the proinflammatory cytokine IL‐17, but at the same time they secreted high amounts of immunoregulatory IL‐10, transforming growth factor (TGF)‐β and IL‐35. Furthermore, they potently suppressed T effector cell (Teff) responses in vitro, identifying them as Tregs 20, 22. Given their bi‐functional properties, we proposed to name these RORγt+FoxP3+ T cells ‘biTregs’. In accordance with their unique cytokine profile and transcription factor expression, biTregs showed both pro‐ and anti‐inflammatory in‐vivo functions in crescentic glomerulonephritis 22. Exogenous biTreg transfer ameliorated renal inflammation to a similar extent as conventional RORγt‐negative Tregs, although the suppressive mechanisms seem to be different. Conversely, endogenous biTregs displayed additional proinflammatory functions. Interestingly, these proinflammatory biTreg functions depended upon activation of RORγt 22. This is clinically highly relevant, as multiple pharmaceutical companies are currently developing RORγt blocking agents and some Phases I and II studies are already ongoing 23, 24, 25. Furthermore, a landmark study which was published during preparation of this paper confirmed the biological relevance of biTregs and surprisingly identified them as potent down‐regulators of anti‐inflammatory Th2 immunity 26. This effect, which might explain some of the proinflammatory biTreg functions, also depended upon activation of the transcription factor RORγt. Given their multiple immune modulatory functions, biTregs are highly likely to contribute to development of SLE. Because, however, nothing is known to date about the clinical relevance of biTregs, we decided to study their role in SLE. A major hallmark of human SLE is activation of the Type I interferon pathway, as has just recently been highlighted again 27. We thus chose to use the pristane model of SLE, which is currently the only available murine model characterized by a strong interferon signature 28. In particular, our study addressed the following aspects: (1) characterization of biTreg dynamics in the different organ systems affected during the course of SLE, (2) analysis of the role of RORγt in biTregs with special focus on IL‐17 secretion and regulation of Th2 immunity and (3) assessment of the contribution of biTregs to organ pathologies in pristane‐induced SLE.
Materials and methods
Animals
LoxP‐site flanked RORCfl/fl mice were obtained from the Jackson Laboratory (Bar Harbor, ME, USA). FoxP3YFP‐Cre mice were a kind gift from Alexander Y. Rudensky (Memorial Sloan‐Kettering Cancer Center, New York, NY, USA). All animals used in this study were on a C57BL/6 background and were raised under specific pathogen‐free conditions at our animal facility.
Animal experiments and functional studies
Pristane disease was induced in 8–10‐week‐old male or female FoxP3Cre × RORCfl/fl mice and matched FoxP3Cre × RORCwt/wt (referred to as FoxP3Cre) littermate controls by single intraperitoneal (i.p.) injection of 500 µl of pristane oil (2,6,10,14‐tetramethylpentadecane; Sigma‐Aldrich, St Louis, MO, USA) 14, 19. Organs were harvested between 1 week and 9 months after injection, as indicated. Animal experiments were performed according to national and institutional animal care and ethical guidelines and were approved by local committees (approval codes 37/11, 45/12, 73/14 and 07/15).
Morphological studies
Glomerular abnormalities were determined in 50 glomeruli per mouse in a blinded manner, as published previously 14, 29. These included glomerular hypercellularity, crescent formation, fibrinoid necrosis, segmental proliferation, hyalinosis and capillary wall thickening. Lung tissue was perfused with 500 µl formalin and fixed overnight, washed with ethanol, paraffin‐embedded and periodic acid‐Schiff (PAS)‐stained 30. Leucocyte infiltration, granuloma formation, alveolar‐wall thickening and alveolar haemorrhage were assessed in a semi‐quantitative score according to the percentage of tissue affected (0–25% = 1, 25–50% = 2, 50–75% = 3, >75% = 4). Mean scores from 15 high‐power fields were calculated. Numbers of pulmonary granulomas were determined in 10 low‐power fields (lpf, magnification × 100) per tissue section; their size was quantified using the Zeiss Axio Vision software (Carl Zeiss, Jena, Germany). For immunohistochemistry, tissue was stained with antibodies against CD3 (A0452; Dako, Hamburg, Germany), F4/80 (BM8; BMA Biomedicals, Hiddenhausen, Germany), MAC2 (M3/38; Cedarlane‐Laboratories, Burlington, ON, Canada), granulocyte‐differentiation antigen‐1 (GR‐1) (NIMP‐R14; Hycult Biotech, Uden, the Netherlands), FoxP3 (FJK‐16s; eBioscience, San Diego, CA, USA) or KI67 (D3B5; Cell Signaling, Danvers, MA, USA) and developed with a polymer‐based secondary antibody–alkaline phosphatase kit (POLAP; Zytomed, Berlin, Germany), as published previously. Fifty glomerular cross‐sections (gcs) per kidney and 30 high‐power fields (hpf, magnification × 400) per kidney and lung section were counted in a blinded fashion.
Isolation of leucocytes from various tissues
Spleens were harvested in Hanks’ balanced salt solution (HBSS) and passed through 70‐µm nylon meshes. After lysis of erythrocytes, cells were washed and passed over 40‐µm meshes, counted and resuspended in phosphate‐buffered saline (PBS) for either culture or fluorescence activated cell sorter (FACS) analysis. Kidneys were minced and incubated in digestion medium [RPMI‐1640 medium containing 10% fetal calf serum (FCS), 1% HEPES, 1% penicillin/streptomycin, 8 µg/ml collagenase D and 0·4 µg/ml DNase] at 37°C for 40 min. Kidney tissue was then homogenized using a gentle magnetic affinity cell sorter (MACS) dissociator (Miltenyi Biotech, Bergisch Gladbach, Germany) and purified using Percoll‐gradient centrifugation 31. The peritoneal cavity was washed with 5 ml ice‐cold PBS to harvest cells at the indicated time‐points after pristane administration 19. For analysis of leucocytes from lungs, organs were removed, minced and incubated in digestion medium at 37°C for 45 min. Tissues were then homogenized using a gentle MACS dissociator (Miltenyi Biotech), filtered over a 70‐µm mesh and purified using Percoll‐gradient centrifugation 30. Peripheral blood was drawn into ethylenediamine tetraacetic acid (EDTA)‐coated tubes and red blood cell lysis was performed.
Systemic humoral immune responses
Circulating anti‐ds‐DNA and anti‐U1‐ribonucleoprotein (RNP) antibodies from serum were analysed by ELISA at the indicated dilutions after coating microtitre plates with either poly‐L‐lysine (Sigma‐Aldrich) and calf thymus DNA (Worthington) or U1‐RNP (Arotec). For analysis of total non‐antigen‐specific immunoglobulins, ELISA plates were precoated with anti‐mouse immunoglobulin (Ig)G antibodies (Jackson Immuno Research). For all analyses, serum samples were applied at the dilutions indicated. The following secondary antibodies were used for detection: total IgG, IgG1 (both Southern Biotech), IgG2b (Invitrogen), IgG2c (Bethyl) and IgG3 (Jackson Immuno Research). For detection of IgE, sera were diluted 1 : 200 and a commercially available ELISA was utilized (Biolegend, San Diego, CA, USA), following the manufacturer's instructions.
Analyses of cytokines from serum and spleen cell cultures
Splenocytes (4 × 106 cells/ml) were cultured under standard conditions in the presence of 1 µg/ml anti‐CD3 (eBioscience, San Diego, CA, USA) and supernatants were harvested after 72 h. Commercially available ELISAs were used for detection of IFN‐γ and IL‐17A (Biolegend). Serum was obtained from mice at 5 and 9 months after pristane induction and was analysed for levels of IL‐17 and IFN‐γ by ELISA (Biolegend) 19.
Flow cytometry
Cells were stained with fluorochrome‐labelled antibodies against CD45, CD3, CD4, CD8, CD19, CD44, CD69, CD62L, CCR6, γδTCR, inducible T cell co‐stimulator (ICOS), glucocorticoid‐induced TNFR family‐related gene (GITR), CD103, cytotoxic T lymphocyte‐associated protein 4 (CTLA‐4), CXCR3 (eBioscience), C‐X‐C chemokine receptor type 5 (CXCR5), programmed death 1 (PD‐1), IgD, IgM, CD138 and PD‐L1 (Biolegend). For intracellular and intranuclear staining, samples were processed using a commercial intranuclear staining kit (FoxP3 kit; eBioscience). Fluorochrome‐labelled antibodies against IL‐4, IL‐5, IL‐13, IL‐17, IFN‐γ, FoxP3 (eBioscience), RORγt, KI67 (both BD Biosciences, Heidelberg, Germany) and Gata‐3 (Biolegend) were employed, as published recently 19, 22. For cytokine staining, cells were activated with phorbol myristate acetate (PMA) (50 ng/ml; Sigma‐Aldrich) and ionomycin (1 µg/ml; Calbiochem‐Merck, Temecula, CA, USA) for 3.5 h. After 30 min of incubation, brefeldin A (10 µg/ml; Sigma‐Aldrich) was added. LIVE/DEAD staining (Invitrogen Molecular Probes, Eugene, OR, USA) was used to exclude dead cells during flow cytometry. Experiments were performed on a BD LSRII Cytometer (Becton Dickinson, Heidelberg, Germany).
Treg suppression assay
CD4+ spleen cells were enriched using magnetic‐activated cell sorting according to the manufacturer's protocol (MACS CD4+ T cell Kit II; Miltenyi Biotec). Tregs and Teff cells were isolated by FACS sorting (performed on a BD ARIAIII Cytometer; Becton Dickinson). A total of 1 × 105 CD45+CD4+yellow fluorescent protein (YFP)− effector T cells from FoxP3Cre mice were then cultured for 72 h in anti‐CD3 monoclonal antibody (mAb) (5 µg/ml; BD Biosciences) precoated 96‐well plates either alone or in co‐culture with CD45+CD4+YFP+ Tregs from FoxP3Cre or FoxP3Cre × RORCfl/fl mice at the ratios indicated. Suppressive capacity was determined by IL‐2 ELISA performed from the supernatants, as published recently 17, 22, 32. For analyses of IFN‐γ, IL‐10, IL‐4, IL‐13 and TNF‐α in the supernatants, cytometric bead array assays were performed using a commercial kit (LEGENDplexTM mouse Th cytokine mix and match subpanel; BioLegend). To assess Teff cell proliferation, cells from the above culture experiments were harvested and analysed for their KI67 expression by FACS.
Quantitative real‐time polymerase chain reaction (PCR) analysis
Total RNA of renal cortex and spleen tissue was isolated according to a standard Trizol protocol and purified utilizing a Nucleospin kit (Macherey & Nagel, Düren, Germany). Real‐time PCR was performed after cDNA transcription as described previously (all primer sequences are available upon request) and results were normalized to expression of 18S rRNA 17.
Statistical analyses
Results are expressed as mean ± standard error of the mean (s.e.m.). Groups were compared by Student's t‐test. A P‐value < 0·05 was considered statistically significant.
Results
IL‐17+ biTregs expand during systemic pristane‐induced lupus
In order to characterize more clearly the pristane model of SLE, we initially performed time–course analyses of the multiple organ manifestations. After pristane injection, mice develop acute sterile peritonitis and non‐immune complex pulmonary capillaritis. Approximately 3 weeks later, immunologically active peritoneal and pulmonary granulomas become apparent. Subsequently, autoimmunity is established with increasing serum levels of various autoantibodies, which we found to be present already in low amounts at week 8. Deposition of immune complexes then leads to progressive development of lupus nephritis which, according to our analyses, became evident by light microscopy at approximately 5 months (data not shown). In Fig. 1a a schematic overview is depicted. In order to assess the potential role of biTregs, we analysed their frequencies in multiple organs before and at different time–points after injection of pristane. biTregs were found to be present in both peritoneal lavage cells and lungs of healthy animals. In acute antigen‐independent sterile peritonitis and pulmonary vasculitis, their population expanded only slightly (Fig. 1b,c). However, at later time‐points, coincident with development of peritoneal and pulmonary granulomas, biTreg percentages increased significantly (2·5 ± 0·27% of peritoneal FoxP3+ Tregs at baseline versus 5·56 ± 0·98% at 5 weeks and 7·57 ± 0·99% at 8 weeks; 5·5 ± 0·46% of pulmonary FoxP3+ Tregs at baseline, 18·96 ± 3·17% at 5 weeks and 21·4 ± 0·86% of FoxP3+ Tregs at week 8) (Fig. 1b,c). In contrast, spleens which are only mildly affected during pristane‐induced SLE showed merely a slight expansion of biTregs (Fig. 1d). Finally, we studied biTregs during the course of immune complex lupus nephritis. Interestingly, our analyses revealed a distinctive pattern of renal infiltration. While only few biTregs were detected at 2 months after pristane injection (1·3 ± 0·38% of FoxP3+ Tregs), their frequencies increased to reach a maximum at 5 months (10·62 ± 1·7% of FoxP3+ Tregs). Afterwards, biTreg proportions declined steadily and were almost back to baseline levels (0·3 ± 0·04% of FoxP3+ Tregs) at 9 months (1·3 ± 0·38% of FoxP3+ Tregs) (Fig. 1e). A significant proportion of biTregs produced IL‐17 in all examined organs at all time‐points (Fig. 1b–e). A representative FACS plot of pulmonary biTregs, including their IL‐17 secretion, is shown in Fig. 1f.
Figure 1.

(a) The temporal course of pristane‐induced inflammation and organ‐specific pathologies is shown. Intraperitoneal injection leads to development of acute innate mediated peritonitis and non‐immune complex pulmonary capillaritis. Subsequently peritoneal and pulmonary granulomas develop and autoantibody formation is initiated. During the following months, immune complex lupus nephritis develops. (b) Retinoic acid receptor‐related orphan nuclear receptor γt (RORγt)+forkhead box protein 3 (FoxP3)+ bi‐functional regulatory T cell (biTreg) percentages among FoxP3+ peritoneal lavage cells at the indicated time‐points after pristane injection (left) and interleukin (IL)‐17 production by biTregs (right). (c) Pulmonary (d) splenic and (e) renal biTreg percentages among FoxP3+ Tregs and percentages of IL‐17+ biTregs at the indicated time‐points after pristane injection. (f) A representative fluorescence activated cell sorter (FACS) plot from inflamed lungs at 5 weeks after pristane injection shows RORγt+FoxP3+ biTregs, RORγt+FoxP3– T helper type 17 (Th17) cells and FoxP3+RORγt– conventional Tregs (cTregs). IL‐17 production by biTregs is shown on the right. Numbers indicate percentages of FoxP3+ cells. Dotted lines indicate basal biTreg percentages in healthy mice. Bars show mean ± standard error of the mean (s.e.m.). *P < 0·05; **P < 0·01; ***P < 0·001 versus basal percentages.
RORγt activation in biTregs aggravates pulmonary capillaritis
In order to study the role of RORγt in biTregs, we generated FoxP3Cre × RORCfl/fl mice, which specifically lack RORγt activation in Tregs. As pristane‐injected mice develop severe, often lethal, haemorrhagic pulmonary capillaritis, we first studied lung pathology in FoxP3Cre × RORCfl/fl and FoxP3Cre control mice. At 3 weeks after pristane injection, no macroscopic or histological differences were noted (Supporting information, Fig. S1a,b). However, at later stages, when biTregs expand and pulmonary inflammation becomes granulomatous, we found striking protection of mice lacking RORγt in Tregs. Macroscopic pulmonary haemorrhage was reduced significantly in FoxP3Cre × RORCfl/fl mice (haemorrhage score 2·9 ± 0·3 versus 1·67 ± 0·3) (Fig. 2a). In line with this, histological signs of vasculitis were ameliorated (vasculitis score 1·63 ± 0·3 versus 0·77 ± 0·2) (Fig. 2a). Interestingly, granulomas were also reduced in numbers (6 ± 1·0 versus 2·5 ± 0·6) and size (41·23 ± 4·3 versus 26·88 ± 5·4 × 103 µm2) in mice with Treg‐specific deficiency of RORγt (Fig. 2b). Furthermore, our analyses revealed a significant reduction of infiltrating T cells, both by immunohistochemistry (17·16 ± 1·6 versus 11·52 ± 1·4 CD3+ cells/hpf) as well as FACS analysis (55·66 ± 1·5 versus 49·48 ± 2·4% CD4+ of total CD3+ cells) of lungs (Fig. 2c). Similarly, numbers of GR‐1‐positive neutrophils were also reduced in FoxP3Cre × RORCfl/fl mice (9·72 ± 0·7 versus 6·8 ± 0·4 GR‐1+ cells/hpf) (Fig. 2d).
Figure 2.

(a) Representative photographs of periodic acid‐Schiff (PAS)‐stained lungs from indicated mouse strains at 8 weeks after pristane injection (left). Quantification of macroscopic haemorrhage as well as vasculitis score is shown (right) (original magnification ×200). (b) Representative photographs of pulmonary granulomas and quantification of granuloma numbers and size (original magnification ×400). (c) Immunohistochemical staining for pan T cell marker CD3 (left). Quantification of pulmonary CD3+ T cell numbers by immunohistochemistry and CD4+ T helper cell percentages by fluorescence activated cell sorter (FACS) as indicated (original magnification ×200). (d) Immunohistochemical staining and quantification of infiltrating granulocyte‐differentiation antigen‐1 (GR‐1)+ neutrophils (original magnification ×200). A = alveolus; B = bronchus; V = vessel; arrows indicate pristane droplets. Squares represent individual animals, horizontal lines indicate means. Bars show mean ± standard error of the mean (s.e.m.). *P < 0·05; **P < 0·01.
Lupus nephritis is ameliorated in FoxP3Cre × RORCfl/fl mice
At later stages, pristane‐injected mice develop proliferative immune complex glomerulonephritis. We thus studied renal disease and found significant amelioration of lupus nephritis at 9 months in FoxP3Cre × RORCfl/fl mice, as evidenced by protection from glomerular injury (58·9 ± 6·3 versus 26·18 ± 4·4% abnormal glomeruli) (Fig. 3a). In line with this, glomerular cell proliferation, a hallmark of lupus nephritis, was also reduced (1·0 ± 0·07 versus 0·67 ± 0·07 KI67+ cells/gcs) (Fig. 3b). Importantly, protection from histological damage also resulted in less functional injury, as indicated by lower blood urea nitrogen (BUN) levels (24·4 ± 1·2 versus 19·6 ± 1·6 mg/dl) (Fig. 3c). Finally, we found that loss of RORγt activation in Tregs also protected from development of albuminuria (239·5 ± 20·4 versus 151·7 ± 18·7 mg/g albumin per creatinine) (Fig. 3d).
Figure 3.

(a) Representative photographs of periodic acid‐Schiff (PAS)‐stained kidney sections at 9 months after pristane injection, as well as quantification of glomerular injury (original magnification ×400). (b) Representative photographs of immunohistochemical staining of KI67 and quantification of glomerular KI67+ cells (original magnification ×400). (c) Quantification of serum blood urea nitrogen (BUN) levels. (d) Quantification of urinary albumin/creatinine ratios at the indicated time‐points before and after injection of pristane. Bars show mean ± standard error of the mean (s.e.m.). Squares represent individual animals, horizontal lines indicate means. *P < 0·05; **P < 0·01.
Renal inflammatory cell infiltration is reduced in mice with RORγt‐deficient Tregs
Because infiltrating renal leucocytes are key players of renal tissue injury, we next analysed renal inflammatory cell infiltration. In line with amelioration of renal damage, immunohistochemistry revealed a significant reduction of both glomerular (2·9 ± 0·3 versus 1·84 ± 0·2 CD3+ cells/gcs) and interstitial (29·58 ± 1·1 versus 24·08 ± 1·5 CD3+ cells/hpf) T cells in FoxP3Cre × RORCfl/fl mice (Fig. 4a). Percentages of regulatory T cells, however, were not significantly different (0·022 ± 0·002 versus 0·017 ± 0·002 FoxP3+/CD3+ cells per hpf) (Fig. 4b). Similarly, we found unchanged proportions of renal Th1 and Th17 cells (Supporting information, Fig. S2a,b). Interestingly, however, percentages of total IL‐17+ renal leucocytes were significantly lower in kidneys of FoxP3Cre × RORCfl/fl mice (2·11 ± 0·16 versus 1·6 ± 0·07% IL‐17+ of CD45+ cells), indicating reduced influx of IL‐17+ populations different from Th17 cells (Fig. 4c). Furthermore, we also detected lower numbers of glomerular (3·74 ± 0·55 versus 2·2 ± 0·27 MAC‐2+ cells/gcs) (Fig. 4d) and interstitial (21·53 ± 2·1 versus 13·86 ± 1·2 F4/80+ cells/hpf) (Fig. 4e) macrophages in mice with Treg‐specific deletion of RORγt.
Figure 4.

Representative photographs and quantification of glomerular and interstitial renal (a) CD3+ T cell and (b) forkhead box protein 3 (FoxP3)+ regulatory T cell (Treg) infiltration at 9 months after pristane injection (original magnification ×400). (c) Fluorescence activated cell sorter (FACS) analysis of total renal infiltrating interleukin (IL)−17+ leucocytes (one of two sets shown). (d) Representative photographs and quantification of glomerular MAC2+ monocyte/macrophage infiltration (original magnification ×400). (e) Representative photographs and quantification of renal interstitial F4/80+ monocyte/macrophage infiltration (original magnification ×200). Squares represent individual animals, horizontal lines indicate means. *P < 0·05; **P < 0·01.
Humoral autoimmunity remains unaltered in FoxP3Cre × RORCfl/fl mice
Next, we wanted to explore the mechanisms leading to tissue protection from pristane‐induced SLE in FoxP3Cre × RORCfl/fl mice. We therefore assessed the effects of RORγt deactivation in biTregs on humoral immune responses. In this respect, we found unaltered levels of serum total IgG as well as identical amounts of all analysed subclasses at 9 months after pristane injection (Supporting information, Fig. S3a). Similarly, formation of IgG autoantibodies of all subclasses against dsDNA (Fig. 5a) as well as U1‐RNP was unchanged in FoxP3Cre × RORCfl/fl mice (Fig. 5b). In line with unchanged serum antibody levels, we documented a similar immune complex deposition in kidneys of pristane‐injected mice, as evidenced by complement C3 (0·84 ± 0·1 versus 0·59 ± 0·05 C3 deposition score) (Fig 5c) and mouse IgG (mIgG) staining (0·53 ± 0·06 versus 0·46 ± 0·05 mIgG deposition score) (Fig. 5d). Similarly, analyses of splenic B cell populations showed similar percentages of total B cells (16·7 ± 1·0 versus 16·38 ± 2·2% CD19+CD138– of CD45+ cells), switched memory B cells (9·24 ± 3·6 versus 14·55 ± 1·3% IgM–IgD– of CD19+CD138– cells) as well as plasma cells (1·34 ± 0·39 versus 0·52 ± 0·07% CD138high of CD45+ cells) in both strains of mice (Supporting information, Fig. S3b–d). Finally, we found that percentages of splenic T follicular helper cells (TfH) were also unaffected by abrogation of RORγt activation in Tregs (1·3 ± 0·29 versus 2·48 ± 0·85% CXCR5+PD‐1+ of CD4+FoxP3– cells) (Supporting information, Fig. S3e).
Figure 5.

(a) Quantification of immunoglobulin (Ig)G anti‐ds‐DNA antibodies, as well as the indicated IgG subclasses from serum of forkhead box protein 3 (FoxP3)Cre wild‐type and FoxP3Cre × retinoic acid receptor‐related orphan nuclear receptor (ROR)Cfl/fl mice in serial dilutions by enzyme‐linked immunosorbent assay (ELISA). (b) Quantification of serum anti‐U1‐ribonucleoprotein (RNP) autoantibodies of the indicated IgG subclasses in serial dilutions by ELISA. (c) Immunohistochemical staining and quantification of glomerular complement C3 deposition (original magnification ×400). (d) Immunohistochemical staining and quantification of glomerular mouse IgG (mIgG) deposition (original magnification ×400). Squares represent individual animals, horizontal lines indicate means.
IL‐17 secretion by biTregs is dependent upon RORγt
In a next step, we aimed to explore alterations of cellular immune responses. As IL‐17 is a known downstream target of RORγt, we analysed IL‐17 expression in FoxP3Cre × RORCfl/fl mice. Analysis of peritoneal lavage cells at 3 weeks after pristane injection confirmed specific and complete absence of RORγt in FoxP3+ Tregs (4·58 ± 1·5 versus 0·28 ± 0·3% biTregs of FoxP3+ cells) (Fig. 6a, left). In line with our hypothesis, IL‐17 production by Tregs was abrogated totally in FoxP3Cre × RORCfl/fl mice, indicating absolute dependency on RORγt (2·33 ± 0·48 versus 0·15 ± 0·06% IL‐17+ of FoxP3+ cells) (Fig. 6a, right). Similarly, analysis of vasculitic lungs showed absence of RORγt+ biTregs (4·06 ± 1·0 versus 0·55 ± 0·33% biTregs of FoxP3+ cells) as well as abolished IL‐17 production by Tregs (2·9 ± 0·74 versus 0·35 ± 0·19% IL‐17+ of FoxP3+ cells) (Fig. 6b). Finally, we analysed nephritic kidneys at 5 months after pristane injection and again found absence of biTregs in FoxP3Cre × RORCfl/fl mice (10·62 ± 1·7 versus 0·8 ± 0·17% biTregs of FoxP3+ cells) as well as abrogation of IL‐17 secretion by renal FoxP3+ Treg cells (0·92 ± 0·27 versus 0·24 ± 0·09% IL‐17+ of FoxP3+ cells) (Fig. 6c).
Figure 6.

(a) A representative fluorescence activated cell sorter (FACS) plot and quantification of peritoneal lavage bi‐functional regulatory T cells (biTregs) (left) as well as interleukin (IL)‐17 production by biTregs (right) at 3 weeks after pristane injection in forkhead box protein 3 (FoxP3)Cre wild‐type and FoxP3Cre × retinoic acid receptor‐related orphan nuclear receptor (ROR)Cfl/fl mice (right). (b) Quantification of pulmonary biTregs (left) and IL‐17 production (right) at 3 weeks after pristane injection in the indicated mouse strains. (c) A representative FACS plot and quantification of renal biTregs (left) and their IL‐17 production (right) at 5 months after pristane injection. Numbers in FACS plots indicate percentages of FoxP3+ cells. Squares represent individual animals, horizontal lines indicate means. *P < 0·05; **P < 0·01; ***P < 0·001.
RORγt deficiency alters the phenotype of Tregs
In order to evaluate whether RORγt also affects intrinsic Treg functions, we studied FoxP3 protein expression on the single‐cell level. Interestingly, we found significantly higher FoxP3 mean fluorescence intensities in Tregs from the blood of naive FoxP3Cre × RORCfl/fl mice [1376 ± 24 versus 1567 ± 36 FoxP3 mean fluorescence intensity (MFI)]. The same was true for Tregs from both peritoneal lavage cells (702·6 ± 79·8 versus 1212 ± 200·3 FoxP3 MFI) and lungs (500·7 ± 55·7 versus 778·5 ± 120 FoxP3 MFI) 3 weeks after pristane injection (Fig. 7a). Detailed analysis of Treg activation marker molecules revealed enhanced surface expression of CD103 (7·9 ± 1·0 versus 15·33 ± 1·7% of FoxP3+ cells), GITR (7·67 ± 0·68 versus 22·9 ± 1·1% of FoxP3+ cells) and ICOS (9·06 ± 0·91 versus 21·95 ± 2·14% of FoxP3+ cells) on RORγt‐deficient Tregs. Levels of CTLA‐4 and PD‐L1 remained unaffected (Fig. 7b). In congruence with elevated expression of activation markers, we found higher Treg activation (37·07 ± 2·6 versus 45·53 ± 0·86% CD69+CD62L– and 21·68 ±1·2 versus 36·57 ± 2·5% CD44+CD62L– of FoxP3+ cells) (Fig. 7c) as well as proliferative activity (77·09 ± 2·1 versus 83·43 ± 1·5% KI67+ of FoxP3+ cells) (Fig. 7d). In‐vitro suppression of cytokine production by Tregs, including IL‐2, Th1 characteristic IFN‐γ, Th2 prototype IL‐4 and IL‐13, as well as Th17‐associated TNF‐α, were unchanged in co‐culture experiments with Teff cells (Supporting information, Fig. S4a–d). Induction of IL‐10 production was also similar between the groups (Supporting information, Fig. S4e). Finally, suppression of Teff cell proliferation was unaffected in FoxP3Cre × RORCfl/fl mice (Supporting information, Fig. S4f).
Figure 7.

(a) Quantification of forkhead box protein 3 (FoxP3) mean fluorescence intensity (MFI) in regulatory T cells (Tregs) from blood of naive mice as well as peritoneal lavage and pulmonary Tregs at 3 weeks after pristane injection from indicated mouse strains (left) and a representative fluorescence activated cell sorter (FACS) plot of FoxP3 MFI in peritoneal lavage Tregs (right). (b) Analysis of the indicated surface markers on Tregs from blood of naive mice. (c) Quantification of activation markers on Tregs from blood of naive mice of the indicated genotypes. (d) Assessment of Treg proliferative activity by FACS analysis of KI67 expression in naive blood Tregs. Squares represent individual animals, horizontal lines indicate means. *P < 0·05; **P < 0·01; ***P < 0·001.
biTregs control Th2 immunity in a RORγt‐dependent manner
Next, we wanted to assess whether the observed absence of IL‐17 secretion and enhanced Treg activation in FoxP3Cre × RORCfl/fl mice might affect systemic immunity. We therefore analysed immune responses in the blood of naive mice. Remarkably, we found a spontaneous hyper‐Th2 phenotype with an increase in Gata3 (0·47 ± 0·04 versus 0·69 ± 0·05% Gata3+ among CD4+FoxP3– cells), IL‐5 (0·27 ± 0·09 versus 0·64 ± 0·08% IL‐5+ among CD4+FoxP3– cells) and IL‐13 (1·9 ± 0·41 versus 4·3 ± 0·29% IL‐13+ among CD4+FoxP3– cells)‐positive T helper cells in FoxP3Cre × RORCfl/fl animals (Fig. 8a). In line with this, we also found enhanced Th2 responses in peritoneal lavage cells (Fig. 8b, Supporting information, Fig. S5a) and lungs (Fig. 8c) during pristane‐induced inflammation. Analyses of the renal cellular infiltrate supported these findings and also showed broad skewing towards Th2 immunity (0·41 ± 0·08 versus 0·73 ± 0·1% IL‐4+, 0·95 ± 0·14 versus 2·0 ± 0·5% IL‐5+ and 4·14 ± 0·2 versus 6·62 ± 0·7% IL‐13+ among CD4+FoxP3– cells) (Fig. 8d). Conversely, Th1 and Th17 responses remained unaffected by knock‐out of RORγt in Tregs in all examined organs (Supporting information, Fig. S5b–f). Similarly, serum levels of IFN‐γ and IL‐17 were identical (Supporting information, Fig. S5g), as was splenic and renal IFN‐γ mRNA expression (Supporting information, Fig. S5h). Finally, we analysed humoral Th2 immunity and found strikingly elevated levels of IgE antibodies in serum of naive FoxP3Cre × RORCfl/fl mice (358 ± 26·8 versus 801·2 ± 110 ng/ml) as well as after disease induction (670·9 ± 116 versus 1633 ± 194 ng/ml) (Fig. 8e).
Figure 8.

(a) Quantification of T helper (Th) cells expressing the Th2 characteristic transcription factor Gata3 or the indicated Th2 cytokines in peripheral blood from naive mice by flow cytometry. (b) Flow cytometric analysis of peritoneal Th cells expressing Gata3 or the indicated Th2 prototype cytokines from peritoneal lavage cells 3 weeks after pristane injection. (c) Gata3 expression by T helper cells in lungs at 3 weeks after pristane injection. (d) Quantification of renal T helper cells expressing the indicated Th2 cytokines at 2 months after pristane injection. (e) Quantification of immunoglobulin (Ig)E levels by enzyme‐linked immunosorbent assay (ELISA) from serum of naive mice and at 5 months after pristane injection. Squares represent individual animals, horizontal lines indicate means. *P < 0·05; **P < 0·01; ***P < 0·001.
Discussion
Our study aimed to evaluate the role of the newly identified RORγt+FoxP3+ biTregs 20, 22, 26, 33 in systemic lupus erythematosus. As nothing is known about the occurrence and biology of these multi‐potent cells in SLE, we started with a thorough analysis of the temporal and organ‐specific pattern of biTreg expansion. In the first 2 weeks after pristane injection mice develop sterile peritonitis, which is mediated by innate immunity 34. During this acute, non‐autoantigen‐dependent inflammation, biTreg levels remained at baseline. Another early and antigen‐independent manifestation of pristane is acute pulmonary vasculitis with diffuse alveolar haemorrhage 35. Similarly to peritonitis, we did not find relevant expansion of biTregs during this innate phase of pulmonary inflammation. However, the persistence of inflammatory responses after pristane injection leads to chronically progressive development of autoimmunity and formation of peritoneal and pulmonary lymphogranulomas, which became apparent from approximately 3 weeks post‐injection. Interestingly, during this stage of disease, biTreg percentages increased significantly in both organs, indicating a role for establishment of granulomas and autoimmunity. In addition, these findings suggest that biTreg expansion occurs in an antigen‐dependent manner, which is in full agreement with a recent report 26. With increasing time after pristane injection, various types of autoantibodies develop 36, 37. These are deposited in the kidneys and result in progressive immune complex‐dependent lupus nephritis. During this process, renal biTregs showed a very distinct and concerted time–course. Their population started to expand early, in parallel with increasing renal inflammation and reached a maximum at approximately 5 months. Subsequently, biTreg percentages decreased slowly and were almost back to baseline levels at 9 months after pristane injection. Interestingly, this distinguished time–course of renal biTregs in chronically developing lupus nephritis parallels their dynamics in the nephrotoxic nephritis model of acute glomerulonephritis 22. In contrast to the massive peritoneal, pulmonary and renal biTreg expansion, they increased only slightly in spleens. This concurs with the fact that pristane injection results only in a mild splenic inflammatory response. In summary, biTregs thus seem to be early mediators of adaptive inflammation and tissue injury during pristane‐induced SLE. Importantly, and in accordance with previous findings by us and others 21, 22, 38, 39, a robust fraction of biTregs produces IL‐17 in all organs and at all investigated time‐points. Given the observed expansion of biTregs in all affected organs, we hypothesized that biTregs might play key functional roles during SLE development. As our earlier data suggest that activation of RORγt mediates the pathogenic functions of biTregs 22, we generated mice with selective deficiency of RORγt in FoxP3+ Tregs. In a first step, we wanted to explore whether abrogation of RORγt activation would result in protection from pulmonary vasculitis, which is a rare but life‐threatening complication of SLE in humans 40. Analysis of lungs at an early time‐point, before development of autoimmunity and expansion of biTregs, showed similar degrees of injury. However, at later stages, FoxP3Cre × RORCfl/fl mice were protected significantly from pulmonary vasculitis in terms of haemorrhage, leucocyte infiltration and histological damage. Furthermore, pulmonary granulomas were reduced both in size and numbers. Next, we aimed to study the development of lupus nephritis, which is one of the most severe organ manifestations of SLE in humans and associates with a bad prognosis. In this respect, we found that FoxP3Cre × RORCfl/fl mice were protected significantly, as evidenced by less histological injury and glomerular cell proliferation. In line with this, functional parameters of renal injury as serum BUN levels and albuminuria were also reduced significantly. Furthermore, we found much decreased renal proinflammatory leucocyte infiltration in mice lacking RORγt activation in Tregs. Interestingly, frequencies of IL‐17‐producing total renal leucocytes were also reduced, despite unaltered renal Th17 responses. This indicates effects of Treg‐specific RORγt deletion on IL‐17+ leucocyte populations different from Th17 cells as, for example, γδTcells and Tregs themselves. In order to explore the mechanisms by which abrogation of RORγt activation in biTregs protects from SLE organ manifestations, we next assessed development of humoral autoimmunity. However, our analyses revealed unchanged levels of total serum IgG as well as all measured subclasses in FoxP3Cre × RORCfl/fl mice. Similarly, serum levels of SLE characteristic anti‐ds‐DNA, and anti‐U1‐RNP autoantibodies of all subclasses remained unaltered. Consistent with unaffected serum antibodies, we found the same extent of renal complement C3 and IgG deposition in both groups of mice. In congruence, analyses of splenic B cell subpopulations, plasma cells and T follicular helper cells showed no difference.
Given the unchanged humoral autoimmunity, we next explored T cell immune responses. Strikingly, and in line with the importance of RORγt for Th17 cells 41, proinflammatory IL‐17 expression by biTregs was abrogated completely in FoxP3Cre × RORCfl/fl mice. This observation might, at least in part, account for the observed amelioration of SLE‐induced tissue injury. In order to identify further potential mechanisms of protection, we explored whether RORγt would also affect Treg suppression and activity. Similar to our previous data, analysing Tregs from RORC pan‐knock‐out mice 22, we found unchanged in‐vitro suppressive capacity. In vivo, however, Treg‐selective deletion of RORγt resulted in enhanced levels of FoxP3 protein as well as increased expression of Treg activation markers. These findings indicate that RORγt expression in Tregs impairs their in‐vivo regulatory function. Therefore, the high activation status of RORγt+ biTregs, which we and others have described previously 22, 33, 42, 43, does not seem to be induced by RORγt. Rather, additional transcription factors, which are co‐activated in RORγt+ biTregs as, for example, interferon regulatory factor (IRF)4 26, runt‐related transcription (Runx)1, Runx3 42 or unknown others, might be responsible. Next, we wanted to explore whether these observed Treg alterations would result in any effects on systemic immunity. Interestingly, we found significantly elevated type 2 immune responses in naive and pristane‐challenged FoxP3Cre × RORCfl/fl mice, both systemically and in all inflamed target organs. Th1 and Th17 responses, in contrast, remained unaltered. Importantly, these observations are in full agreement with a very recent report, which was published during preparation of this paper 26. This emergence of biTregs as potent regulators of type 2 immunity is particularly remarkable, as the mechanisms regulating Th2 responses remain widely unknown to date. In this regard, our data do not indicate a direct effect of biTregs on suppression of Th2 cells, as generation of Th2 immunity was not altered in our in‐vitro co‐culture assays of Teff with RORγt‐deficient Tregs. Rather, as suggested by Ohnmacht et al., indirect mechanisms, involving interactions between Tregs and dendritic cells, seem to be important 26.
Taken together, we found that biTreg selective deletion of RORγt resulted in abrogation of their IL‐17 secretion, enhancement of Th2 immunity and increased Treg activation. Importantly, these changes were associated with protection from pulmonary vasculitis and lupus nephritis. It remains unclear, however, to what extent each of these three RORγt‐dependent immune alterations contribute to disease amelioration. While it is likely that protection results from a mixed phenotype, involving all three mechanisms, future studies will need to address this aspect in more detail.
In summary, our study provides the first evidence, to our knowledge, for a crucial role of the newly defined biTregs in SLE. Our findings thus further support research into biTreg biology and favour RORγt‐directed interventions as novel therapeutic options for SLE.
Disclosure
None.
Supporting information
Additional Supporting information may be found in the online version of this article at the publisher's web‐site:
Fig. S1. (a) Quantification of macroscopic haemorrhage as well as vasculitis score is shown as indicated. (b) Quantification of granuloma numbers and size. Squares represent individual animals, horizontal lines indicate means. All comparisons between the groups P > 0·05 not significant (n.s.).
Fig. S2. (a) Representative fluorescence activated cell sorter (FACS) plots and (b) quantification of CD45+CD3+CD4+ forkhead box protein 3 (FoxP3)– T effector cells (Teff) secreting the indicated cytokines from kidneys of mice at 9 months after pristane injection. Numbers in fluorescence activated cell sorter (FACS) plots indicate percentages. Squares represent individual animals, horizontal lines indicate means. All comparisons between the groups P > 0·05 not significant (n.s.).
Fig. S3. (a) Levels of total immunoglobulin (Ig)G and the indicated IgG subclasses were determined by enzyme‐linked immunosorbent assay (ELISA) from serum of mice at 9 months after pristane injection at serial dilutions. (b) Percentages of splenic total B cells, (c) switched‐memory B cells (sMB) and (d) plasma cells were not different between the groups at 9 months after pristane injection. (e) Frequencies of splenic T follicular helper cells (TfH) were similar at 9 months after pristane injection. All comparisons between the groups P > 0·05 not significant (n.s.).
Fig. S4. (a–f) In‐vitro suppression assays were performed by co‐culturing wild‐type CD4+ T effector cells (Teff) with regulatory T cells (Tregs) from forkhead box protein 3 (FoxP3)Cre × retinoic acid receptor‐related orphan nuclear receptor (ROR)Cfl/fl mice or FoxP3Cre controls at the indicated ratios (n = 4 per group). (a) Cytokine levels of interleukin (IL)−2 were analysed in co‐culture supernatants by enzyme‐linked immunosorbent assay (ELISA). Cytokine levels of (b) Th1 cytokine interferon (IFN)‐γ, (c) T helper type 2 (Th2) cytokines IL‐4 and IL‐13, (d) Th17‐associated cytokine tumour necrosis factor (TNF)‐α and (e) IL‐10 were analysed in co‐culture supernatants by cytometric bead array. (f) Suppression of Teff proliferation was analysed by quantification of KI67 expression. Dotted lines represent Teff alone without Tregs (n = 4). All comparisons between the groups P > 0·05 not significant (n.s.).
Fig. S5. (a) Representative fluorescence activated cell sorter (FACS) plots of expression of Gata3 and the indicated T helper type 2 (Th2) cytokines in T effector cells (Teff) of peritoneal lavage cells at 3 weeks after pristane injection. (b–e) Fluorescence activated cell sorter (FACS) analyses of indicated Th1 and Th17 cytokine and transcription factor expression in Teff from (b) blood of naive mice, (c) peritoneal lavage cells at 3 weeks, (d) pulmonary leucocytes at 3 weeks and (e) renal infiltrating leucocytes at 8 weeks after pristane injection. (f) Enzyme‐linked immunosorbent assay (ELISA) analysis of spleen cell cytokine production at 9 months. (g) ELISA analyses of serum cytokine levels at the indicated time‐points after pristane injection. (h) Interferon (IFN)‐γ mRNA expression levels in spleens and kidneys at 9 months after pristane injection. Numbers in FACS plots indicate percentages. Squares represent individual animals, horizontal lines indicate means. Bars represent mean ± standard error of the mean (s.e.m.). All comparisons between the groups P > 0·05 not significant (n.s.).
Acknowledgements
We thank M. Schaper and M. Reszka for their excellent technical help. This work was supported by grants from the Deutsche Forschungsgemeinschaft (STE 1822/2‐1, KFO 228 STE 1822/3‐1 to O. M. S. and SFB 1192 to O. M. S. and M. A. K.) and Deutsche Gesellschaft fuer Nephrologie to M. A. K.
References
- 1. Bagavant H, Fu SM. Pathogenesis of kidney disease in systemic lupus erythematosus. Curr Opin Rheumatol 2009; 21:489–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Tsokos GC. Systemic lupus erythematosus. N Engl J Med 2011; 365:2110–21. [DOI] [PubMed] [Google Scholar]
- 3. Bomback AS, Appel GB. Updates on the treatment of lupus nephritis. J Am Soc Nephrol 2010; 21:2028–35. [DOI] [PubMed] [Google Scholar]
- 4. Anders HJ, Appel GB. Lupus nephritis: implications of the new ACR lupus nephritis guidelines. Nat Rev Nephrol 2012; 8:500–1. [DOI] [PubMed] [Google Scholar]
- 5. Liu Z, Davidson A. Taming lupus – a new understanding of pathogenesis is leading to clinical advances. Nat Med 2012; 18:871–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Gottschalk TA, Tsantikos E, Hibbs ML. Pathogenic inflammation and its therapeutic targeting in systemic lupus erythematosus. Front Immunol 2015; 6:550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Theofilopoulos AN, Koundouris S, Kono DH, Lawson BR. The role of IFN‐gamma in systemic lupus erythematosus: a challenge to the Th1/Th2 paradigm in autoimmunity. Arthritis Res 2001; 3:136–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Shah K, Lee WW, Lee SH et al Dysregulated balance of Th17 and Th1 cells in systemic lupus erythematosus. Arthritis Res Ther 2010; 12:R53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Kurts C, Panzer U, Anders HJ, Rees AJ. The immune system and kidney disease: basic concepts and clinical implications. Nat Rev Immunol 2013; 13:738–53. [DOI] [PubMed] [Google Scholar]
- 10. Steinmetz OM, Turner JE, Paust HJ et al CXCR3 mediates renal Th1 and Th17 immune response in murine lupus nephritis. J Immunol 2009; 183:4693–704. [DOI] [PubMed] [Google Scholar]
- 11. Shin MS, Lee N, Kang I. Effector T‐cell subsets in systemic lupus erythematosus: update focusing on Th17 cells. Curr Opin Rheumatol 2011; 23:444–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Zhang Z, Kyttaris VC, Tsokos GC. The role of IL‐23/IL‐17 axis in lupus nephritis. J Immunol 2009; 183:3160–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Pisitkun P, Ha HL, Wang H et al Interleukin‐17 cytokines are critical in development of fatal lupus glomerulonephritis. Immunity 2012; 37:1104–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Summers SA, Odobasic D, Khouri MB et al Endogenous interleukin (IL)‐17A promotes pristane‐induced systemic autoimmunity and lupus nephritis induced by pristane. Clin Exp Immunol 2014; 176:341–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Amarilyo G, Lourenco EV, Shi FD, La Cava A. IL‐17 promotes murine lupus. J Immunol 2014; 193:540–3. [DOI] [PubMed] [Google Scholar]
- 16. Riedel JH, Paust HJ, Krohn S et al IL‐17F promotes tissue injury in autoimmune kidney diseases. J Am Soc Nephrol 2016; 26:3666–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Kluger MA, Luig M, Wegscheid C et al Stat3 programs Th17‐specific regulatory T cells to control GN. J Am Soc Nephrol 2014; 25:1291–302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Chaudhry A, Rudra D, Treuting P et al CD4+ regulatory T cells control TH17 responses in a Stat3‐dependent manner. Science 2009; 326:986–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Kluger MA, Melderis S, Nosko A et al Treg17 cells are programmed by Stat3 to suppress Th17 responses in systemic lupus. Kidney Int 2016; 89:158–66. [DOI] [PubMed] [Google Scholar]
- 20. Lochner M, Peduto L, Cherrier M et al In vivo equilibrium of proinflammatory IL‐17+ and regulatory IL‐10+ Foxp3+ RORgamma t+ T cells. J Exp Med 2008; 205:1381–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Ayyoub M, Deknuydt F, Raimbaud I et al Human memory FOXP3+ Tregs secrete IL‐17 ex vivo and constitutively express the T(H)17 lineage‐specific transcription factor RORgamma t. Proc Natl Acad Sci USA 2009; 106:8635–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Kluger MA, Meyer MC, Nosko A et al RORgammat+Foxp3+ cells are an independent bifunctional regulatory T cell lineage and mediate crescentic GN. J Am Soc Nephrol 2016; 27:454–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Huh JR, Englund EE, Wang H et al Identification of potent and selective diphenylpropanamide RORgamma inhibitors. ACS Med Chem Lett 2013; 4:79–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Kojetin DJ, Burris TP. REV‐ERB and ROR nuclear receptors as drug targets. Nat Rev Drug Discov 2014; 13:197–216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Abdel‐Magid AF. RORc modulators for the treatment of autoimmune diseases. ACS Med Chem Lett 2015; 6:958–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Ohnmacht C, Park JH, Cording S et al Mucosal immunology. The microbiota regulates type 2 immunity through RORgammat(+) T cells. Science 2015; 349:989–93. [DOI] [PubMed] [Google Scholar]
- 27. Banchereau R, Hong S, Cantarel B et al Personalized immunomonitoring uncovers molecular networks that stratify lupus patients. Cell 2016; 165:1548–50. [DOI] [PubMed] [Google Scholar]
- 28. Zhuang H, Szeto C, Han S, Yang L, Reeves WH. Animal models of interferon signature positive lupus. Front Immunol 2015; 6:291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Summers SA, Hoi A, Steinmetz OM et al TLR9 and TLR4 are required for the development of autoimmunity and lupus nephritis in pristane nephropathy. J Autoimmun 2010; 35:291–8. [DOI] [PubMed] [Google Scholar]
- 30. Turner JE, Morrison PJ, Wilhelm C et al IL‐9‐mediated survival of type 2 innate lymphoid cells promotes damage control in helminth‐induced lung inflammation. J Exp Med 2013; 210:2951–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Luig M, Kluger MA, Goerke B et al Inflammation‐induced IL‐6 functions as a natural brake on macrophages and limits GN. J Am Soc Nephrol 2015; 26:1597–607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Nosko A, Kluger MA, Diefenhardt P et al T‐Bet enhances regulatory T cell fitness and directs control of Th1 responses in crescentic GN. J Am Soc Nephrol 2016. doi: 10.1681/ASN.2009070741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Yang BH, Hagemann S, Mamareli P et al Foxp3(+) T cells expressing RORgammat represent a stable regulatory T‐cell effector lineage with enhanced suppressive capacity during intestinal inflammation. Mucosal Immunol 2016; 9:444–57. [DOI] [PubMed] [Google Scholar]
- 34. Reeves WH, Lee PY, Weinstein JS, Satoh M, Lu L. Induction of autoimmunity by pristane and other naturally occurring hydrocarbons. Trends Immunol 2009; 30:455–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Chowdhary VR, Grande JP, Luthra HS, David CS. Characterization of haemorrhagic pulmonary capillaritis: another manifestation of pristane‐induced lupus. Rheumatology 2007; 46:1405–10. [DOI] [PubMed] [Google Scholar]
- 36. Richards HB, Satoh M, Jennette JC, Okano T, Kanwar YS, Reeves WH. Disparate T cell requirements of two subsets of lupus‐specific autoantibodies in pristane‐treated mice. Clin Exp Immunol 1999; 115:547–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Satoh M, Richards HB, Shaheen VM et al Widespread susceptibility among inbred mouse strains to the induction of lupus autoantibodies by pristane. Clin Exp Immunol 2000; 121:399–405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Hovhannisyan Z, Treatman J, Littman DR, Mayer L. Characterization of interleukin‐17‐producing regulatory T cells in inflamed intestinal mucosa from patients with inflammatory bowel diseases. Gastroenterology 2011; 140:957–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Chung AY, Li Q, Blair SJ et al Oral interleukin‐10 alleviates polyposis via neutralization of pathogenic T‐regulatory cells. Cancer Res 2014; 74:5377–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Andrade C, Mendonca T, Farinha F et al Alveolar hemorrhage in systemic lupus erythematosus: a cohort review. Lupus 2016; 25:75–80. [DOI] [PubMed] [Google Scholar]
- 41. Steinmetz OM, Summers SA, Gan PY, Semple T, Holdsworth SR, Kitching AR. The Th17‐defining transcription factor RORgammat promotes glomerulonephritis. J Am Soc Nephrol 2011; 22:472–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Li L, Patsoukis N, Petkova V, Boussiotis VA. Runx1 and Runx3 are involved in the generation and function of highly suppressive IL‐17‐producing T regulatory cells. PLOS ONE 2012; 7:e45115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Vocanson M, Rozieres A, Hennino A et al Inducible costimulator (ICOS) is a marker for highly suppressive antigen‐specific T cells sharing features of TH17/TH1 and regulatory T cells. J Allergy Clin Immunol 2010; 126:280–9. 289.e1‐7. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Additional Supporting information may be found in the online version of this article at the publisher's web‐site:
Fig. S1. (a) Quantification of macroscopic haemorrhage as well as vasculitis score is shown as indicated. (b) Quantification of granuloma numbers and size. Squares represent individual animals, horizontal lines indicate means. All comparisons between the groups P > 0·05 not significant (n.s.).
Fig. S2. (a) Representative fluorescence activated cell sorter (FACS) plots and (b) quantification of CD45+CD3+CD4+ forkhead box protein 3 (FoxP3)– T effector cells (Teff) secreting the indicated cytokines from kidneys of mice at 9 months after pristane injection. Numbers in fluorescence activated cell sorter (FACS) plots indicate percentages. Squares represent individual animals, horizontal lines indicate means. All comparisons between the groups P > 0·05 not significant (n.s.).
Fig. S3. (a) Levels of total immunoglobulin (Ig)G and the indicated IgG subclasses were determined by enzyme‐linked immunosorbent assay (ELISA) from serum of mice at 9 months after pristane injection at serial dilutions. (b) Percentages of splenic total B cells, (c) switched‐memory B cells (sMB) and (d) plasma cells were not different between the groups at 9 months after pristane injection. (e) Frequencies of splenic T follicular helper cells (TfH) were similar at 9 months after pristane injection. All comparisons between the groups P > 0·05 not significant (n.s.).
Fig. S4. (a–f) In‐vitro suppression assays were performed by co‐culturing wild‐type CD4+ T effector cells (Teff) with regulatory T cells (Tregs) from forkhead box protein 3 (FoxP3)Cre × retinoic acid receptor‐related orphan nuclear receptor (ROR)Cfl/fl mice or FoxP3Cre controls at the indicated ratios (n = 4 per group). (a) Cytokine levels of interleukin (IL)−2 were analysed in co‐culture supernatants by enzyme‐linked immunosorbent assay (ELISA). Cytokine levels of (b) Th1 cytokine interferon (IFN)‐γ, (c) T helper type 2 (Th2) cytokines IL‐4 and IL‐13, (d) Th17‐associated cytokine tumour necrosis factor (TNF)‐α and (e) IL‐10 were analysed in co‐culture supernatants by cytometric bead array. (f) Suppression of Teff proliferation was analysed by quantification of KI67 expression. Dotted lines represent Teff alone without Tregs (n = 4). All comparisons between the groups P > 0·05 not significant (n.s.).
Fig. S5. (a) Representative fluorescence activated cell sorter (FACS) plots of expression of Gata3 and the indicated T helper type 2 (Th2) cytokines in T effector cells (Teff) of peritoneal lavage cells at 3 weeks after pristane injection. (b–e) Fluorescence activated cell sorter (FACS) analyses of indicated Th1 and Th17 cytokine and transcription factor expression in Teff from (b) blood of naive mice, (c) peritoneal lavage cells at 3 weeks, (d) pulmonary leucocytes at 3 weeks and (e) renal infiltrating leucocytes at 8 weeks after pristane injection. (f) Enzyme‐linked immunosorbent assay (ELISA) analysis of spleen cell cytokine production at 9 months. (g) ELISA analyses of serum cytokine levels at the indicated time‐points after pristane injection. (h) Interferon (IFN)‐γ mRNA expression levels in spleens and kidneys at 9 months after pristane injection. Numbers in FACS plots indicate percentages. Squares represent individual animals, horizontal lines indicate means. Bars represent mean ± standard error of the mean (s.e.m.). All comparisons between the groups P > 0·05 not significant (n.s.).
