TLR7 is associated with development of systemic lupus erythematosus (SLE), but the underlying mechanisms are incompletely understood. Although TLRs are known to activate type I IFN (T1IFN) signaling, the role of T1IFN and IFN-γ signaling in differential regulation of TLR7-mediated Ab-forming cell (AFC) and germinal center (GC) responses, and SLE development has never been directly investigated. Using TLR7-induced and TLR7 overexpression models of SLE, we report in this study a previously unrecognized indispensable role of TLR7-induced IFN-γ signaling in promoting AFC and GC responses, leading to autoreactive B cell and SLE development. T1IFN signaling in contrast, only modestly contributed to autoimmune responses and the disease process in these mice. TLR7 ligand imiquimod treated IFN-γ reporter mice show that CD4+ effector T cells including follicular helper T (Tfh) cells are the major producers of TLR7-induced IFN-γ. Transcriptomic analysis of splenic tissues from imiquimod-treated autoimmune-prone B6.Sle1b mice sufficient and deficient for IFN-γR indicates that TLR7-induced IFN-γ activates multiple signaling pathways to regulate TLR7-promoted SLE. Conditional deletion of Ifngr1 gene in peripheral B cells further demonstrates that TLR7-driven autoimmune AFC, GC and Tfh responses and SLE development are dependent on IFN-γ signaling in B cells. Finally, we show crucial B cell-intrinsic roles of STAT1 and T-bet in TLR7-driven GC, Tfh and plasma cell differentiation. Altogether, we uncover a nonredundant role for IFN-γ and its downstream signaling molecules STAT1 and T-bet in B cells in promoting TLR7-driven AFC, GC, and SLE development whereas T1IFN signaling moderately contributes to these processes.

Systemic lupus erythematosus (SLE) is a debilitating autoimmune disease mediated by Abs reactive to DNA- and RNA-associated self-antigens. Although extrafollicular Ab-forming cell (AFC) and follicular germinal center (GC) responses are traditionally thought to be involved mainly in anti-pathogen Ab responses, AFCs and GCs can also develop spontaneously in the absence of purposeful immunization or overt infection (1, 2). Dysregulation in spontaneous AFC (Spt-AFC) and Spt-GC responses in SLE patients and SLE-prone mice promotes autoreactive B cell survival and pathogenic autoantibody production (38). However, the mechanisms that drive Spt-AFC and spontaneous GC (Spt-GC) responses which can lead to the development of autoreactive B cells, autoantibodies and SLE are incompletely understood.

Genome-wide association studies revealed multiple risk alleles in the TLR pathway that are strongly associated with SLE development (9, 10). Overexpression and overactivity of TLR7 are associated with development of SLE in humans and in mouse models (1113). Recent collaborative works also have highlighted the significance of TLR7 in promoting extrafollicular double negative B cell populations involved in SLE pathogenesis (8). TLR7 gene escape from X chromosome inactivation enhances the B cell response to TLR7 stimulation (14). Studies in the MRL-lpr model highlight the opposing inflammatory and regulatory roles for TLR7 and TLR9, respectively, in extrafollicular autoreactive B cell responses and SLE development (15). Using a variety of SLE mouse models, we and others discovered that TLR7 signaling specifically in B cells promoted SLE development through formation of Spt-AFCs and Spt-GCs (11, 16, 17). These data highlight the importance of TLR7 signaling in promoting autoimmune AFC and GC responses, leading to the development of autoreactive B cells, autoantibody production and SLE; however, mechanisms of TLR7-mediated autoimmunity and SLE development are not clearly defined.

Type I IFN (T1IFN) and type II (IFN-γ) IFN signaling are both implicated in SLE (9, 18). An IFN-α signature is associated with human SLE (19, 20). Lupus-prone mice deficient in IFN-αR have reduced anti-nuclear Abs (ANAs) and renal disease (21). Exogenous IFN-α treatment accelerates the SLE phenotype in female (NZW × NZB) F1 mice with an early development of anti-dsDNA Abs and nephritis (22). The administration of IFN-α in lupus-prone mice also leads to enhanced short-lived AFC and GC responses (23), and an altered B cell selection in the GCs (24). We and others recently showed a B cell–intrinsic role of T1IFN signaling in loss of tolerance and autoreactive B cell development in the AFC and GC pathways in lupus-prone mice (25, 26). SLE patients also showed an increased production of IFN-γ (27, 28), and blockade of IFN-γ normalizes IFN regulated gene expression in SLE patients (29). MRL/lpr and NZW/NZBF1 lupus-prone mice deficient in IFN-γ have reduced autoantibody production and renal disease (3035). We and others discovered a B cell–intrinsic requirement of IFN-γ signaling in Spt-AFC and Spt-GC responses and SLE development (4, 36). These data indicate that both T1IFN and IFN-γ signaling contribute to autoreactive B cell development in the AFC and GC pathways. However, the mechanisms and relative contribution of T1IFN and IFN-γ signaling in TLR7-mediated AFC, GC, and autoimmunity development have never been directly investigated using a TLR7-induced model, especially when T1IFN signaling is believed to be strongly associated with TLR signaling and SLE development.

T1IFN and IFN-γ signaling mediate signals through STAT1. Interestingly, we previously reported a B cell–intrinsic requirement of STAT1 in the Spt-GC responses (36) which we also showed dependent on TLR7 signaling in B cells (16). However, how STAT1 downstream of T1IFN and IFN-γ signaling in B cells controls TLR7-mediated AFC and GC responses, promoting development of autoreactive B cells and SLE, remain unanswered. In this study, we used TLR7-induced and -overexpression models of SLE to dissect the relative contribution of T1IFN and IFN-γ signaling in driving autoimmune AFC and GC responses, promoting development of autoreactive B cells, autoantibody production, and SLE. We found that IFN-γ and STAT1 signaling in B cells played an indispensable role in TLR7-promoted AFC and GC responses and SLE development, whereas global T1IFN signaling only moderately contributed to these processes. Using a TLR7-induced SLE model we show that TLR7 not only increased expression of T1IFN genes but also the IFN-γ gene in splenic tissue compared with untreated mice. Our data from IFN-γ reporter mice and RNA sequencing (RNA-seq) analysis indicate that TLR7-driven and T cell–produced IFN-γ activates multiple signaling pathways to regulate TLR7-promoted SLE. Together, our data show that TLR7 drives autoreactive B cell development in the AFC and GC pathways and promotes SLE by activating T1IFN and IFN-γ signaling in which IFN-γ signaling plays an indispensable role, which cannot be overcome by the activation of other downstream pathways of TLR7 signaling.

C57BL/6J (B6), B6.129S7-Ifngr1tm1Agt/J (IFN-γR1−/−), B6(Cg)-Ifnar1tm1.2Ees/J (IFN-αR1−/−), C.129S4(B6)-Ifngtm3.1Lky/J mice (IFN-γ eYFP), B6.129S(Cg)-Stat1tm1Dlv/J (STAT1−/−), B6.129S-Stat1tm1Mam/Mmjax (STAT1fl/fl), C57BL/6N-Ifngr1tm1.1Rds/J (IFN-γRfl/fl), B6.129-Tbx21tm2Srnr/J (T-betfl/fl), and B6.SB-Yaa/J (B6.yaa) mice were originally purchased from The Jackson Laboratory and bred in house. The B6.Sle1b mice (congenic for the Sle1b sublocus) were described previously (37). B6.Sle1bYaa mice were generated by crossing B6.Yaa mice to B6.Sle1b mice. B6.CD23Cre (B6.Cg-Tg(Fcer2a-cre)5Mbu, provided by Dr. Meinrad Busslinger, were crossed to B6.Sle1b autoimmune mice in house. All animal studies were conducted at Penn State Hershey Medical Center in accordance with the guidelines approved by our Institutional Animal Care and Use Committee. Animals were housed in a specific pathogen-free barrier facility.

For epicutaneous imiquimod (IMQ) treatment, 5% IMQ cream (Glenmark Pharmaceuticals) was applied on the ears of mice 3 times weekly for 4–12 wk, depending on the experiments as previously described (38, 39). For PCR array and RNA-seq sample preparation, mice were treated for 4 wk. To study the systemic autoimmune responses, such as AFC, GC, follicular helper T (Tfh), and Ab responses, mice were treated for 8 wk. To evaluate immune complex deposition and kidney pathology, mice were treated for 12 wk (three times a wk for 8 wk, followed by once a week for another 4 wk).

Flow cytometric analysis of total mouse splenocytes was performed using the following Abs: B220-BV605 (RA3-6B2), CD4-AF700 (RMP4-5), CD44-allophycocyanin (IM7), CD62L-PECy7 (MEK-14), PD1-PE (29F.1A12), NK1.1-biotin (PK136), Ly6G-PB (1A8), and MHCII-PE-Cy7 (M5/114.15.2) (BioLegend); GL7-FITC (GL-7), CD95- PeCy7, CXCR5-biotin (2G8), IFN-γR-biotin (GR20), STAT1-PE (1/STAT1), CD11b-AF700 (M1/70), and CD11c-FITC (HL3) (BD Biosciences); Ly6C-allophycocyanin (HK1.4), PDC-PE (eBio927), and CD8α (clone 53–6.7) (eBioscience). All cells were stained with fixable viability dye-eFluor780 (Invitrogen) prior to surface staining. Stained cells were analyzed using the BD LSR II flow cytometer (BD Biosciences). Data were acquired using FACSDiva software (BD Biosciences) and analyzed using FlowJo software (Tree Star).

Mouse spleens or kidneys were embedded in OCT compound and snap frozen over liquid nitrogen. Five-micrometer-thin sections were cut on a cryostat, mounted on ColorFrost Plus microscope slides (Thermo Fisher Scientific), and fixed in cold acetone for 20 min. The following Abs and reagents were used for immunofluorescence staining of mouse spleen sections for GCs: PE–anti-CD4 (GK1.5; BioLegend), FITC-GL7 (RA3-6B2; BD Biosciences), and allophycocyanin–anti-IgD (11-26c2a; BD Biosciences). Kidney sections were stained for C3 using FITC–anti-C3 (ICL Laboratories) or Biotin–anti-IgG (Jackson ImmunoResearch), followed by streptavidin (SA)–PE. ANA reactivity was detected by indirect immunofluorescence staining of HEp-2 cell slides using sera from indicated mice at a 1:50 dilution and probed with FITC-rat anti-mouse κ (H139-52.1). The images of stained spleen and kidney sections were captured using the Leica DM4000 fluorescence microscope and analyzed using a Leica application suite–advanced fluorescence software (LAS-AF; Leica Microsystems). For the measurement of the GC area, a total of at least 10 randomly selected GCs (GL-7+) per mouse spleen section from at least five mice of each genotype were measured for total area (square micrometers) using the LAS-AF quantitation tool.

Kidneys from 6-mo-old mice were fixed in 10% neutral buffered formalin and embedded in paraffin. Kidney sections were cut at 3- and 6-μm thickness for periodic acid-Schiff (PAS) and H&E staining, respectively. All images were obtained with an Olympus BX51 microscope and DP71 digital camera using cellSens Standard 1.12 imaging software (Olympus America, Center Valley, PA). Two pathologists blinded to the genotype of the mice examined kidney sections. One kidney section per mouse was evaluated: each glomerulus was examined at 400× magnification and scored from 0 (normal) to 4 (severe) based on glomerular size and lobulation, presence of karyorrhectic nuclear debris, capillary basement membrane thickening, and the degree of mesangial matrix expansion and mesangial cell proliferation (40). H&E sections were scored for overall glomerular damage and interstitial inflammation as described (41).

ELISPOT assays were performed as previously described (25, 36). Briefly, splenocytes in RPMI 1640 containing 10% FBS were plated at a concentration of 1 × 106 cells per well onto multiscreen 96-well filtration plates (Millipore) coated with salmon sperm dsDNA (Invitrogen), nucleosome (histone from Sigma Aldrich plated on a layer of dsDNA), or Smith/ribonucleoprotein (SmRNP; Arotec Diagnostics). Serially diluted (1:2) cells were incubated for 12 h at 37°C. dsDNA-, nucleosome-, and smRNP-specific AFCs were detected by biotinylated anti-κ Ab (Invitrogen) and SA–alkaline phosphatase (Vector Laboratories) or alkaline phosphatase-conjugated anti-mouse IgG (Molecular Probes). Plates were developed using the Vector Blue Alkaline phosphatase Substrate Kit III (Vector Laboratories). ELISPOTs were enumerated and analyzed using a computerized ELISPOT plate imaging/analysis system (Cellular Technology).

Serum autoantibodies were measured using standard ELISA protocols as described (3). Briefly, total IgG autoantibody titers were measured in ELISA plates coated with salmon sperm dsDNA, nucleosome or smRNP and detected with biotinylated secondary Ab followed by SA–alkaline phosphatase (Vector Laboratories). The plates were developed using p-nitrophenyl phosphate, disodium salt (Thermo Fisher Scientific) substrates for alkaline phosphatase and read at λ405 nm on Synergy H1 (BioTek Instruments).

RNA was extracted from spleen tissues by TRIzol as per the manufacturer’s instructions (Ambion). OD values of extracted RNA were measured using NanoDrop to confirm an A260:A280 ratio above 1.9. RNA integrity number was measured using BioAnalyzer (Agilent Technologies). RNA 6000 Pico Kit to confirm RIN above 7. The cDNA libraries were prepared using the Ovation RNA-Seq System V2 (NuGEN Technologies, San Carlos, CA) as per the manufacturer’s instructions. The unique barcode sequences were incorporated in the adaptors for multiplexed high-throughput sequencing. The final product was assessed for its size distribution and concentration using BioAnalyzer High Sensitivity DNA Kit (Agilent Technologies) and then loaded onto TruSeq SR v3 flow cells on an Illumina HiSeq 2500 (Illumina) and run for 100 cycles using a paired-read recipe (TruSeq SBS Kit v3; Illumina) according to the manufacturer’s instructions. Illumina CASAVA pipeline (released version 1.8; Illumina) was used to obtain demultiplexed sequencing reads (fastq files) passed the default purify filter. Additional quality filtering used FASTX-Toolkit (http://hannonlab.cshl.edu/fastx_toolkit) to keep only reads that have at least 80% of bases with a quality score of 20 or more (conducted by fastq_quality_filter function) and reads left with 10 bases or longer after being end trimmed with a base quality score of 20 (conducted by fastq_quality_trimmer function). A bowtie2 index was built for the mouse reference genome (GRCm38) using bowtie version 2.1.0. The RNA-seq reads were mapped using Tophat version 2.0.9 (42) supplied by Ensembl annotation file; GRCm38.78.gtf. Gene expression values were computed using fragments per kilo base per million mapped reads values. Differential gene expression was determined using Cuffdiff tool, which is available in Cufflinks version 2.2.1 supplied by GRCm38.78.gtf. Normalization was performed via the median of the geometric means of fragment counts across all libraries, as described (43). Statistical significance was assessed using a false discovery rate threshold of 0.05 to determine the differentially abundant genes. Genes that were expressed at ≥1.5-fold differences between control and test samples were selected for further analysis. The heatmaps of selected genes were generated via the pheatmap package in R using Euclidean distance and Ward clustering method.

The accession number for the RNA-seq data reported in this paper is in the Gene Expression Omnibus: GSE141433 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE141433).

The p values were calculated using unpaired, nonparametric, Mann–Whitney, Student t test (for comparison of two groups), or one-way or two-way ANOVA, with a follow-up Tukey multiple-comparison test (for comparison of more than two groups) or two-way ANOVA, with a follow-up Sidak multiple-comparison test (for comparison of two groups). The p values are as follows: *p < 0.05, **p < 0.01, ***p < 0.001, and ****p < 0.0001. GraphPad Prism 6 software was used.

To identify the mechanisms and determine the role of T1IFN and IFN-γ signaling in differential regulation of TLR7-mediated autoimmunity, we have used a TLR7-induced SLE model in which we treated 8 wk old female mice with TLR7 agonist IMQ as described (38). IMQ-treated B6 (B6-IMQ) and autoimmune-prone B6.Sle1b mice (Sle1b-IMQ) displayed more splenocytes compared with untreated control counterparts (Supplemental Fig. 1A). Overall percentage of B220+ B cells and CD4+ T cells was not significantly different in untreated and treated B6 and B6.Sle1b mice (Supplemental Fig. 1B, 1C), but the numbers of B cells and CD4+ T cells were higher in Sle1b-IMQ mice than three other groups (Supplemental Fig. 1D, 1E). In addition, Sle1b-IMQ mice had much higher percentage of splenic B220+GL7hiCD95hi GC B cells and CD4+CD44hiPD-1hiCXCR5hi Tfh cells than untreated B6.Sle1b mice (Fig. 1A, 1B). Flow cytometry gating strategy for GC B cells and Tfh cells is shown (Supplemental Fig. 1F). Elevated GC and Tfh responses in Sle1b-IMQ mice were strongly associated with increased numbers of dsDNA- and nucleosome-specific splenic AFCs, serum ANA reactivity, and autoantibody titers (Fig. 1C–F). Treated B6 mice (B6-IMQ) had a 2-fold higher GC and Tfh responses than untreated B6 mice but no autoantibody production. We then treated 8-wk-old mice with IMQ for 12 wk and found that Sle1b-IMQ mice had more severe nephritis than B6.Sle1b untreated mice and B6-treated mice (Fig. 1G, 1H). These elevated responses in Sle1b-IMQ mice were accompanied by an increased expression of IFN gene signatures in the splenic tissue (Fig. 1I), and elevated IFN-αR and IFN-γR expression on B cells upon TLR7 stimulation in vitro (Fig. 1J). Thus, Sle1b-IMQ mice represent a strong TLR7-driven systemic autoimmunity model with autoimmune phenotypes (elevated AFC, GC, Tfh, ANA, and nephritis) closely resembling human SLE.

FIGURE 1.

IMQ-induced mouse model of SLE. Eight- to ten-week-old C57BL/6 (B6) and B6.Sle1b mice were epicutaneously treated on the ears with TLR7 ligand IMQ for 8 wk. The percentages of splenic B220+GL7hiCD95hi GC B cells (A) and CD4+CD44hiPD-1hiCXCR5hi Tfh cells (B) from these mice were obtained through flow cytometric analysis. (C and D) The number of dsDNA- and nucleosome-specific AFCs in the spleens was quantified by ELISPOT assay. These data represent four mice of each group. ANA reactivity (E) and serum titers of dsDNA-, nucleosome-, and SmRNP-specific Abs (F) in these mice (six mice of each group) are shown. Representative images of kidney sections stained with PAS (G) and GN score (H) are shown. (I) B6.Sle1b mice were untreated or treated with IMQ for 4 wk, and the spleen tissues were processed for IFNs and IFN receptors PCR array. Arrows in the scatter plots represent upregulated genes upon IMQ treatment. (J) Expression of IFN-γR and IFN-αR on B cells before and after stimulation with R848 for 48 h. These data represent two to three experiments and each symbol indicates an individual mouse (A, B, and J) or a glomerulus (H). Statistical analysis was performed by one-way (A, B, and H) or two-way (C, D, and F) ANOVA, with a follow-up Tukey multiple-comparison test or unpaired, nonparametric Mann-Whitney Student t test (J). Error bars in (A), (B), and (H) represent mean ± SD, and error bars in (F), (C), and (D) represent mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. ns, not significant.

FIGURE 1.

IMQ-induced mouse model of SLE. Eight- to ten-week-old C57BL/6 (B6) and B6.Sle1b mice were epicutaneously treated on the ears with TLR7 ligand IMQ for 8 wk. The percentages of splenic B220+GL7hiCD95hi GC B cells (A) and CD4+CD44hiPD-1hiCXCR5hi Tfh cells (B) from these mice were obtained through flow cytometric analysis. (C and D) The number of dsDNA- and nucleosome-specific AFCs in the spleens was quantified by ELISPOT assay. These data represent four mice of each group. ANA reactivity (E) and serum titers of dsDNA-, nucleosome-, and SmRNP-specific Abs (F) in these mice (six mice of each group) are shown. Representative images of kidney sections stained with PAS (G) and GN score (H) are shown. (I) B6.Sle1b mice were untreated or treated with IMQ for 4 wk, and the spleen tissues were processed for IFNs and IFN receptors PCR array. Arrows in the scatter plots represent upregulated genes upon IMQ treatment. (J) Expression of IFN-γR and IFN-αR on B cells before and after stimulation with R848 for 48 h. These data represent two to three experiments and each symbol indicates an individual mouse (A, B, and J) or a glomerulus (H). Statistical analysis was performed by one-way (A, B, and H) or two-way (C, D, and F) ANOVA, with a follow-up Tukey multiple-comparison test or unpaired, nonparametric Mann-Whitney Student t test (J). Error bars in (A), (B), and (H) represent mean ± SD, and error bars in (F), (C), and (D) represent mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. ns, not significant.

Close modal

Although the induction of T1IFNs by TLR signaling and their roles in SLE pathogenesis have been well described (4446), we know little about how IFN-γ signaling may contribute to TLR-mediated SLE autoimmunity. Therefore, using the TLR7-induced Sle1b-IMQ model, in this study we determined the relative contribution of T1IFN and IFN-γ signaling in TLR7-mediated SLE autoimmune responses. Although IMQ-treated B6.Sle1b mice deficient in IFN-αR (Sle1b.IFN-αR−/−-IMQ) had lower GC B cell and Tfh cell responses than Sle1b-IMQ mice, these responses were diminished in treated B6.Sle1b mice deficient in IFN-γR (Sle1b.IFN-γR−/−-IMQ) (Fig. 2A, 2B). The frequency of GCs between Sle1b-IMQ and Sle1b.IFN-αR−/−-IMQ mice was similar, but GC sizes were smaller in Sle1b.IFN-αR−/−-IMQ mice than Sle1b-IMQ control mice, whereas a negligible number of very small GCs was observed in Sle1b.IFN-γR−/−-IMQ mice (Fig. 2C, 2D). In addition, the number of dsDNA- and nucleosome-specific splenic AFCs (Fig. 2E), serum dsDNA-, nucleosome- and SmRNP-specific Abs (Fig. 2F), and serum ANA reactivity (Fig. 2G) were moderately reduced in Sle1b.IFN-αR−/−-IMQ mice, whereas they were diminished in Sle1b.IFN-γR−/−-IMQ mice compared with Sle1b-IMQ control mice. Consistent with systemic autoimmune B and T cell responses and serum ANA reactivity, immune complex (IC) deposition (IgG and C3) and severe glomerulonephritis (GN) in B6.Sle1b-IMQ mice were moderately reduced in Sle1b.IFN-αR−/−-IMQ mice, whereas they were completely absent in Sle1b.IFN-γR−/−-IMQ mice (Fig. 2H–J).

FIGURE 2.

Differential roles of type I and II IFNs in TLR7-mediated AFC, GC, and Tfh responses and SLE development. Representative flow cytometry plots (A) and percentage (B) of B220+GL7hiCD95hi GC B cells and CD4+CD44hiPD-1hiCXCR5hi Tfh cells in IMQ treated B6.Sle1b, Sle1b.IFN-αR−/− and Sle1b.IFN-γR−/− female mice. (C) Representative spleen sections stained with GC B cell marker GL7 (green), naive B cell marker IgD (blue), and T cell marker CD4 (red). (D) Number of GCs per 5× field (top panel) and GC areas (bottom) were measured in 10 mice of each genotype. (E) Numbers of dsDNA- and nucleosome-specific splenic AFCs in these mice were quantified by ELISPOT assay. dsDNA-, nucleosome- and SmRNP-specific serum IgG titers (F) and serum ANA reactivity (G) were measured by ELISA and HEp-2 assays, respectively. Data in (F) and (G) represent six mice of each genotype. (H) Representative kidney sections from these mice were stained with anti-C3 (green) and anti-mouse IgG (red). Representative PAS-stained kidney sections (I) and GN score (J) from all three genotypes are shown. These data represent three experiments and each symbol indicates an individual mouse (B and E) or a GC (D) or a glomerulus (J). Statistical analysis was performed by one-way (B, D, E, and J) or two-way (F) ANOVA, with a follow-up Tukey multiple-comparison test. Error bars in (B), (D), (E), and (J) represent mean ± SD, and error bars in (F) represent mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. ns, not significant.

FIGURE 2.

Differential roles of type I and II IFNs in TLR7-mediated AFC, GC, and Tfh responses and SLE development. Representative flow cytometry plots (A) and percentage (B) of B220+GL7hiCD95hi GC B cells and CD4+CD44hiPD-1hiCXCR5hi Tfh cells in IMQ treated B6.Sle1b, Sle1b.IFN-αR−/− and Sle1b.IFN-γR−/− female mice. (C) Representative spleen sections stained with GC B cell marker GL7 (green), naive B cell marker IgD (blue), and T cell marker CD4 (red). (D) Number of GCs per 5× field (top panel) and GC areas (bottom) were measured in 10 mice of each genotype. (E) Numbers of dsDNA- and nucleosome-specific splenic AFCs in these mice were quantified by ELISPOT assay. dsDNA-, nucleosome- and SmRNP-specific serum IgG titers (F) and serum ANA reactivity (G) were measured by ELISA and HEp-2 assays, respectively. Data in (F) and (G) represent six mice of each genotype. (H) Representative kidney sections from these mice were stained with anti-C3 (green) and anti-mouse IgG (red). Representative PAS-stained kidney sections (I) and GN score (J) from all three genotypes are shown. These data represent three experiments and each symbol indicates an individual mouse (B and E) or a GC (D) or a glomerulus (J). Statistical analysis was performed by one-way (B, D, E, and J) or two-way (F) ANOVA, with a follow-up Tukey multiple-comparison test. Error bars in (B), (D), (E), and (J) represent mean ± SD, and error bars in (F) represent mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. ns, not significant.

Close modal

Next, we determined the role of IFN-γ signaling in SLE autoimmunity development in B6.Sle1b mice overexpressing TLR7 by breeding Sle1b.IFN-γR−/− mice to B6.Yaa mice that carry an extra copy of TLR7 to generate Sle1bYaaIFN-γR−/− mice. B6.Sle1b male mice expressing Yaa (Sle1bYaa) developed splenomegaly, which was reversed to a level observed in B6.Sle1b males in the absence of IFN-γR (Fig. 3A). Elevated GC B cell (Fig. 3B, 3D, 3E) and Tfh cell (Fig. 3C) responses in Sle1bYaa mice were significantly reduced in Sle1bYaaIFN-γR−/− mice. Similarly, the number of dsDNA- and nucleosome-specific splenic AFCs (Fig. 3F), serum dsDNA-, nucleosome- and SmRNP-specific Abs (Fig. 3G), and serum ANA reactivity (Fig. 3H) were significantly reduced in Sle1bYaaIFN-γR−/− mice compared with Sle1bYaa control mice. Finally, IC deposition in the kidney and the developed GN in Sle1bYaa mice were also diminished in Sle1bYaaIFN-γR−/− mice (Fig. 3I–K). Our data from TLR7-induced and -overexpression SLE models indicate that IFN-γ signaling plays a nonredundant role in TLR7-promoted AFC and GC responses and subsequent autoantibody production and SLE pathogenesis, whereas T1IFN signaling only moderately contributes to these responses.

FIGURE 3.

Essential role of IFN-γ in the Yaa mouse model of SLE. Representative images of spleen size (A), the percentages of B220+GL7hiCD95hi GC B cells (B), and CD4+CD44hiPD-1hiCXCR5hi Tfh cells (C) are shown. Histological images of splenic GCs (D) stained for GC B cell marker, GL7 (green), mature B cell marker, IgD (blue), and T cell marker, CD4 (red) and GC areas (E) are shown. Number of splenic dsDNA- and nucleosome-specific AFCs (F), serum Ab titers against dsDNA, nucleosome and SmRNP (G), and serum ANA reactivity (H) are illustrated. Data in (G) and (H) represent 7–10 mice of each group. Representative images of kidney sections stained with anti-C3 (green) and anti-IgG (red) (I). Representative images of PAS stained kidney sections (J) and GN score (K). These data were derived from 6- to 8-mo-old B6.Sle1b, Sle1bYaa and SlebYaaIFN-γR−/− mice and represent two to three experiments. Each symbol represents a mouse (B–F) or a glomerulus (K). Statistical analysis was performed by one-way (B, C, E, F, and K) or two-way (G) ANOVA, with a follow-up Tukey multiple-comparison test. Error bars in (B), (C), (E), (F), and (K) represent mean ± SD, and in error bars in (G) represent mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

FIGURE 3.

Essential role of IFN-γ in the Yaa mouse model of SLE. Representative images of spleen size (A), the percentages of B220+GL7hiCD95hi GC B cells (B), and CD4+CD44hiPD-1hiCXCR5hi Tfh cells (C) are shown. Histological images of splenic GCs (D) stained for GC B cell marker, GL7 (green), mature B cell marker, IgD (blue), and T cell marker, CD4 (red) and GC areas (E) are shown. Number of splenic dsDNA- and nucleosome-specific AFCs (F), serum Ab titers against dsDNA, nucleosome and SmRNP (G), and serum ANA reactivity (H) are illustrated. Data in (G) and (H) represent 7–10 mice of each group. Representative images of kidney sections stained with anti-C3 (green) and anti-IgG (red) (I). Representative images of PAS stained kidney sections (J) and GN score (K). These data were derived from 6- to 8-mo-old B6.Sle1b, Sle1bYaa and SlebYaaIFN-γR−/− mice and represent two to three experiments. Each symbol represents a mouse (B–F) or a glomerulus (K). Statistical analysis was performed by one-way (B, C, E, F, and K) or two-way (G) ANOVA, with a follow-up Tukey multiple-comparison test. Error bars in (B), (C), (E), (F), and (K) represent mean ± SD, and in error bars in (G) represent mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

Close modal

To investigate how IFN-γ may promote autoreactive B cell development into the AFC and GC pathways in TLR7-accelerated SLE, we analyzed the gene transcripts in spleens of IMQ-treated B6.Sle1b and Sle1b.IFN-γR−/− mice by RNA-seq. Clustering of differentially expressed genes showed a distinct transcriptional profile in Sle1b.IFN-γR−/−-IMQ mice compared with B6.Sle1b-IMQ control mice. We found that 130 and 213 genes were up and downregulated, respectively, in Sle1b.IFN-γR−/−-IMQ mice (p < 0.05; fold change ≥1.5; Fig. 4A). Functional classification of the differentially expressed genes with gene ontology analysis revealed many biological processes or the pathways that were downregulated (Fig. 4B) or upregulated (Fig. 4C) in the absence of IFN-γR signaling. Genes differentially regulated between B6.Sle1b-IMQ and B6.Sle1b.IFN-γR−/−-IMQ splenic tissues were associated with gene transcription regulation (Fig. 4D), ubiquitination/proteasome complex (Fig. 4E), cell proliferation and migration (Fig. 4F), autoimmunity (Fig. 4G), inflammation (Fig. 4H), cellular and oxidative stress (Fig. 4I), and cellular metabolism (Fig. 4J). These results indicate that IFN-γ signaling promotes TLR7-mediated autoimmunity by regulating multiple biological processes.

FIGURE 4.

Transcriptome and gene ontology analysis of IMQ-treated Sle1b.IFN-γR−/− and B6.Sle1b splenic tissue RNA. B6.Sle1b and Sle1b.IFN-γR−/− mice were treated with IMQ for 4 wk and the splenic tissues were processed for RNA-seq. (A) Heat map showing the relative expression pattern of the top 343 differentially expressed genes between the groups. Fold-change values were visualized as log10 (FC). Gene ontology and pathway analysis for the genes that were downregulated (B) or upregulated (C) in Sle1b.IFN-γR−/−-IMQ mice (FC > 2 and p value <0.05). (DJ) Heat maps display differentially expressed genes that were categorized based on their functions in particular biological processes (FC > 1.5 and p value <0.05). Hierarchical-clustering analysis was conducted for the transcript levels of the differentially expressed genes (Ward clustering algorithm and Euclidean distance measure; n = 3, each sample was pooled from four mice of each genotype).

FIGURE 4.

Transcriptome and gene ontology analysis of IMQ-treated Sle1b.IFN-γR−/− and B6.Sle1b splenic tissue RNA. B6.Sle1b and Sle1b.IFN-γR−/− mice were treated with IMQ for 4 wk and the splenic tissues were processed for RNA-seq. (A) Heat map showing the relative expression pattern of the top 343 differentially expressed genes between the groups. Fold-change values were visualized as log10 (FC). Gene ontology and pathway analysis for the genes that were downregulated (B) or upregulated (C) in Sle1b.IFN-γR−/−-IMQ mice (FC > 2 and p value <0.05). (DJ) Heat maps display differentially expressed genes that were categorized based on their functions in particular biological processes (FC > 1.5 and p value <0.05). Hierarchical-clustering analysis was conducted for the transcript levels of the differentially expressed genes (Ward clustering algorithm and Euclidean distance measure; n = 3, each sample was pooled from four mice of each genotype).

Close modal

Considering the increased expression of the Ifn-γ gene in splenic tissue (Fig. 1I) and regulation of multiple signaling pathways by IFN-γ signaling (Fig. 4) in IMQ-treated mice, we next determined the cellular source of IFN-γ in TLR7-mediated autoimmunity. We treated IFN-γ–eYFP reporter mice with IMQ for 6 wk and found that CD4+ T cells were the major producers of IFN-γ, although CD8+ T cells and NK cells produced this cytokine to a certain extent (Fig. 5A). Of the CD4+CD44hi effector T cells, a higher frequency of pre-Tfh and Tfh cells produced IFN-γ than non-Tfh cells upon IMQ treatment (Fig. 5B–D). Dendritic cells, macrophages, eosinophils, monocytes, and neutrophils did not produce IFN-γ in these mice (Fig. 5E). These data together with increased IFN-γR expression on B cells upon TLR7 stimulation ex vivo (Fig. 1J) point to an important role for TLR7 in regulating the IFN-γ–IFN-γR pathway in B cells by inducing T cell IFN-γ production and upregulating IFN-γR on B cells.

FIGURE 5.

CD4+ T cells are the major IFN-γ producers upon TLR7 stimulation. Three- to four-month-old B6 (non-eYFP) and eYFP-IFN-γ reporter (B6.eYFP) mice were left untreated (UT) or treated with IMQ for 4 wk, and splenocytes were analyzed for eYFP expressing cells by flow cytometry. (A) Bar diagrams show the percentage of eYFP+ (IFN-γ+) total CD4+, CD8+, and NK1.1+ cells. These data represent six mice per group. (B) Flow cytometry gating strategy for CD4+CD44hi effector T cells that are subdivided into CD4+PD-1loCXCR5lo non-Tfh, CD4+PD-1intCXCR5int pre-Tfh, and CD4+PD-1hiCXCR5hi Tfh. (C) Representative flow cytometry dot plots from indicated mouse strains show frequency of eYFP-expressing (IFN-γ+) cells in non-Tfh, pre-Tfh, Tfh, and CD4 effector cells. (D) Bar diagrams show the percentage of IFN-γ+ cells in non-Tfh, pre-Tfh, and Tfh cells in UT and IMQ-treated B6.eYFP mice. These data represent five mice of each group. (E) Flow cytometry contour plots represent eYFP expression in the indicated myeloid cell populations. moMPs, monocyte-derived macrophages. Statistical analysis was performed by unpaired, nonparametric Mann-Whitney Student t test or two-way ANOVA, with a follow-up Sidak multiple-comparison test. Error bars in (A) and (D) represent mean ± SEM. **p < 0.01, ***p < 0.001, ****p < 0.0001. ns, not significant.

FIGURE 5.

CD4+ T cells are the major IFN-γ producers upon TLR7 stimulation. Three- to four-month-old B6 (non-eYFP) and eYFP-IFN-γ reporter (B6.eYFP) mice were left untreated (UT) or treated with IMQ for 4 wk, and splenocytes were analyzed for eYFP expressing cells by flow cytometry. (A) Bar diagrams show the percentage of eYFP+ (IFN-γ+) total CD4+, CD8+, and NK1.1+ cells. These data represent six mice per group. (B) Flow cytometry gating strategy for CD4+CD44hi effector T cells that are subdivided into CD4+PD-1loCXCR5lo non-Tfh, CD4+PD-1intCXCR5int pre-Tfh, and CD4+PD-1hiCXCR5hi Tfh. (C) Representative flow cytometry dot plots from indicated mouse strains show frequency of eYFP-expressing (IFN-γ+) cells in non-Tfh, pre-Tfh, Tfh, and CD4 effector cells. (D) Bar diagrams show the percentage of IFN-γ+ cells in non-Tfh, pre-Tfh, and Tfh cells in UT and IMQ-treated B6.eYFP mice. These data represent five mice of each group. (E) Flow cytometry contour plots represent eYFP expression in the indicated myeloid cell populations. moMPs, monocyte-derived macrophages. Statistical analysis was performed by unpaired, nonparametric Mann-Whitney Student t test or two-way ANOVA, with a follow-up Sidak multiple-comparison test. Error bars in (A) and (D) represent mean ± SEM. **p < 0.01, ***p < 0.001, ****p < 0.0001. ns, not significant.

Close modal

To determine how TLR7-induced and T cell–produced IFN-γ may signal in B cells for regulating autoimmune AFC, GC, and Tfh responses and promoting SLE development, we conditionally deleted the Ifngr1 gene in peripheral B cells in B6.Sle1b mice (designated Sle1bIFN-γRfl/fl.CD23Cre+). IMQ-treated Sle1bIFN-γRfl/fl.CD23Cre+ mice did not show splenomegaly that was observed in IMQ-treated Sle1bIFN-γRfl/fl control mice (Fig. 6A, 6B). Sle1bIFN-γRfl/fl.CD23Cre+ mice also had markedly reduced B220+GL-7hiCD95hi GC B cell and CD4+CD44hiCXCR5hiPD-1hi Tfh cell responses, including reduced frequency and size of GCs, compared with Sle1bIFN-γRfl/fl mice (Fig. 6C–G). Sle1bIFN-γRfl/fl.CD23Cre+ mice also had diminished serum ANA reactivity (Fig. 6H), reduced serum autoantibody titers (Fig. 6I), and splenic autoantibody-producing AFCs (Fig. 6J). Dampened AFC, GC, and Tfh responses in Sle1bIFN-γRfl/fl.CD23Cre+ mice were strongly associated with diminished IC deposition in the kidney and with significantly improved kidney inflammation such as GN, interstitial nephritis, and vasculitis score (Fig. 6K–M). These data highlight the nonredundant role of IFN-γ signaling in B cells in TLR7-mediated AFC, GC, and Tfh responses and the development of autoreactive B cells, autoantibodies, and lupus nephritis.

FIGURE 6.

B cell–intrinsic IFN-γ signaling is necessary for TLR7-mediated autoimmune responses and lupus nephritis development. (AM) Sle1bIFN-γRfl/fl and Sle1bIFN-γRfl/fl. CD23Cre+ mice were treated with IMQ for 12 wk. Spleen pictures (A) and their weight (B) are shown. Representative spleen sections were stained for GCs (C) with GL7 (green), CD4 (red), and IgD (blue); and the areas (D) and the frequency of GCs (E) were measured. The percentage of B220+GL7hiCD95hi GC B cells (F) and CD4+CD44hiPD-1hiCXCR5hi Tfh cells (G) were measured by flow cytometry. Serum ANA reactivity (H) and autoantibody titers (I) against dsDNA, nucleosome, and SmRNP are shown. Data in (H) and (I) represent six mice of each genotype. Numbers of splenic dsDNA-, nucleosome-, and SmRNP-specific AFCs were quantified by ELISPOT assay (J). (K) Representative kidney sections were stained with anti-C3 (green) and anti-IgG (red). Representative kidney sections were stained with PAS (L) or H&E (M) and scored for GN, interstitial nephritis, or inflammation in the vessels. These data represent two to three experiments,and each symbol indicates an individual mouse (B, F, G, J, and M) or a GC (D and E) or a glomerulus (L). Statistical analysis was performed by unpaired, nonparametric Mann-Whitney Student t test or two-way ANOVA, with a follow-up Sidak multiple-comparison test (I). Error bars in (B), (D)–(G), (J), (L), and (M) represent mean ± SD, and error bars in (I) represent mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

FIGURE 6.

B cell–intrinsic IFN-γ signaling is necessary for TLR7-mediated autoimmune responses and lupus nephritis development. (AM) Sle1bIFN-γRfl/fl and Sle1bIFN-γRfl/fl. CD23Cre+ mice were treated with IMQ for 12 wk. Spleen pictures (A) and their weight (B) are shown. Representative spleen sections were stained for GCs (C) with GL7 (green), CD4 (red), and IgD (blue); and the areas (D) and the frequency of GCs (E) were measured. The percentage of B220+GL7hiCD95hi GC B cells (F) and CD4+CD44hiPD-1hiCXCR5hi Tfh cells (G) were measured by flow cytometry. Serum ANA reactivity (H) and autoantibody titers (I) against dsDNA, nucleosome, and SmRNP are shown. Data in (H) and (I) represent six mice of each genotype. Numbers of splenic dsDNA-, nucleosome-, and SmRNP-specific AFCs were quantified by ELISPOT assay (J). (K) Representative kidney sections were stained with anti-C3 (green) and anti-IgG (red). Representative kidney sections were stained with PAS (L) or H&E (M) and scored for GN, interstitial nephritis, or inflammation in the vessels. These data represent two to three experiments,and each symbol indicates an individual mouse (B, F, G, J, and M) or a GC (D and E) or a glomerulus (L). Statistical analysis was performed by unpaired, nonparametric Mann-Whitney Student t test or two-way ANOVA, with a follow-up Sidak multiple-comparison test (I). Error bars in (B), (D)–(G), (J), (L), and (M) represent mean ± SD, and error bars in (I) represent mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

Close modal

We next asked how much of a role STAT1 that functions downstream of IFN-γ signaling played in TLR7-driven GC, Tfh, and IgG Ab responses under a scenario in which several pathways (NF-κB, IRF5/7, and P38/MAPK) can be potentially activated by the strong in vivo TLR7 stimulation. We first treated B6 and B6.STAT1−/− mice with IMQ and found that STAT1 is absolutely required for the development of TLR7-driven GC B cell, Tfh cell, and plasma cell responses (Fig. 7A–C). Next, by using conditional knockout mice with peripheral B cells deficient in STAT1 (STAT1fl/flCD23Cre+), we found the requirement of B cell–intrinsic STAT1 in TLR7-mediated GC, Tfh, plasma cell, and IgG Ab responses (Fig. 7D–F). Consistent with these data, B cells stimulated with TLR7 ligand had much higher STAT1 expression than unstimulated B cells (Fig. 7G). Similar to the increased STAT1 expression, T-bet downstream of STAT1 was also upregulated in B cells stimulated with TLR7 ligand (Fig. 7H). To solidify the importance of B cell–intrinsic T-bet in TLR7 driven GC, Tfh, and Ab responses, we treated T-betfl/flCD23Cre+ mice where peripheral B cells are deficient in T-bet. We observed a crucial B cell–intrinsic role of T-bet in TLR7-mediated GC, Tfh, and IgG Ab responses (Fig. 7I–K). These data indicate that STAT1 and T-bet downstream of TLR7 and IFN-γ signaling play a significant role in TLR7-driven GC, Tfh, and IgG Ab responses, which cannot be overcome by other pathways potentially activated by TLR7 stimulation.

FIGURE 7.

B cell–intrinsic roles of STAT1 and T-bet in TLR7-mediated GC, Tfh, and Ab responses. (A) Representative histological images of splenic GCs in B6 and B6.STAT1−/− mice treated with IMQ for 8 wk. Spleen sections were stained for GC B cell marker, GL7 (green), mature B cell marker, IgD (blue), and T cell marker, CD4 (red). (B) Flow cytometry gating strategy and (C) the percentage of GC B cells, Tfh, and plasma cells in B6 and B6.STAT1−/− mice are shown. (D and E) B6.STAT1fl/fl control mice and STAT1fl/flCD23Cre+ conditional knockout mice were treated with IMQ for 8 wk before performing similar analyses as in (B) and (C). (F) Serum IgG and IgG2c Abs from mice described in (D) and (E) were measured by ELISA. STAT1 (G) and T-bet (H) expression in unstimulated (media) and TLR7 stimulated B cells were measured by intracellular staining (ICS) and flow cytometry analysis. (I) Representative histological images of splenic GCs, (J) percentage of GC B cells and Tfh, and (K) total serum IgG and IgG2c Abs in IMQ-treated B6.STAT1fl/fl and STAT1fl/flCD23Cre+ mice were analyzed as in (A)–(C) and (F). Statistical analysis was performed by unpaired, nonparametric Mann-Whitney Student t test (C, E, G, H, and J) or two-way ANOVA, with a follow-up Sidak multiple-comparison test (F and K). Error bars in (C), (E), (G), (H), and (J) represent mean ± SD, and error bars in (F) and (K) represent mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

FIGURE 7.

B cell–intrinsic roles of STAT1 and T-bet in TLR7-mediated GC, Tfh, and Ab responses. (A) Representative histological images of splenic GCs in B6 and B6.STAT1−/− mice treated with IMQ for 8 wk. Spleen sections were stained for GC B cell marker, GL7 (green), mature B cell marker, IgD (blue), and T cell marker, CD4 (red). (B) Flow cytometry gating strategy and (C) the percentage of GC B cells, Tfh, and plasma cells in B6 and B6.STAT1−/− mice are shown. (D and E) B6.STAT1fl/fl control mice and STAT1fl/flCD23Cre+ conditional knockout mice were treated with IMQ for 8 wk before performing similar analyses as in (B) and (C). (F) Serum IgG and IgG2c Abs from mice described in (D) and (E) were measured by ELISA. STAT1 (G) and T-bet (H) expression in unstimulated (media) and TLR7 stimulated B cells were measured by intracellular staining (ICS) and flow cytometry analysis. (I) Representative histological images of splenic GCs, (J) percentage of GC B cells and Tfh, and (K) total serum IgG and IgG2c Abs in IMQ-treated B6.STAT1fl/fl and STAT1fl/flCD23Cre+ mice were analyzed as in (A)–(C) and (F). Statistical analysis was performed by unpaired, nonparametric Mann-Whitney Student t test (C, E, G, H, and J) or two-way ANOVA, with a follow-up Sidak multiple-comparison test (F and K). Error bars in (C), (E), (G), (H), and (J) represent mean ± SD, and error bars in (F) and (K) represent mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

Close modal

Cellular and molecular mechanisms by which TLR7 overexpression or overactivity promotes autoreactive B cell development in the extrafollicular AFC and follicular GC pathways that are activated in SLE remain largely unknown. In this study, using the TLR7-induced and TLR7-overexpression models of SLE, as well as an IFN-γ reporter mouse model, we demonstrate that TLR7 overactivation drives development of autoreactive B cells in both the AFC and GC pathways and promotes SLE by driving predominantly IFN-γ production by T cells and mediating IFN-γ signaling in B cells. TLR7-driven and T cell–produced IFN-γ controls gene transcription in multiple cellular pathways important for immune cell survival, proliferation, metabolism, and autoimmunity.

Several groups, including ours, previously identified a B cell–intrinsic role of TLR7-MyD88 signaling in promoting autoimmune AFC and GC responses, leading to SLE pathogenesis in the mouse models (5, 11, 16, 17, 47). We and others also reported B cell–intrinsic roles of T1IFN and IFN-γ signaling in regulating SLE-associated AFC, GC, and autoantibody responses in SLE-prone mice (4, 25, 36). Although TLR7 signaling is believed to drive T1IFN production and activation of T1IFN signaling in SLE (19, 45, 48, 49), how TLR7-driven IFN-γ production and activation of IFN-γ signaling in B cells may promote TLR7-mediated autoimmune AFC and GC responses and SLE development has previously not been addressed. Using the TLR7-induced SLE model and B cell–specific conditional deletion of the Ifngr1 gene, we found that IFN-γ signaling in B cells was essential for TLR7-mediated AFC, GC, and Tfh responses and GN development, whereas global T1IFN signaling deficiency moderately reduced these responses and was associated with less kidney pathology. Using IFN-γ reporter mice, we further established that TLR7 induced IFN-γ production primarily by CD4+ T cells, including pre-Tfh and Tfh. Together, our data demonstrate that TLR7-driven and T cell–produced IFN-γ act nonredundantly to activate IFN-γ signaling in B cells, promoting development of autoreactive B cells and SLE pathogenesis.

Although MRL/lpr and NZW/NZBF1 lupus-prone mice deficient in IFN-γ or treated with anti–IFN-γ blocking therapy had reduced autoantibody production and renal disease (3035), the role of IFN-γ signaling in autoreactive B cell development in either the extrafollicular or the GC pathways in these mice was not tested. David Rawlings’ and our groups previously discovered the role of IFN-γ signaling in regulating SLE-associated GC responses; however, the link between TLR7 and IFN-γ signaling in autoreactive B cell expansion in the GC pathway was not directly tested in these studies using a TLR7-induced SLE mouse model (4, 36). In this study, we find a significantly reduced number of autoantibody-producing AFCs and dampened GC responses in the spleens of TLR7-induced SLE-prone mice. Our current data highlight the importance of B cell–intrinsic IFN-γ signaling downstream of TLR7 signaling in regulating autoreactive B cell development in both the extrafollicular and GC pathways, leading to TLR7-promoted autoantibody production and SLE development.

Synergy between TLR-MyD88 and IFN-γ–STAT1 signaling in potentiating inflammatory responses in macrophages has previously been described (50). IFN-γ–STAT1 signaling was found to aid TLR-mediated inflammatory cytokine production by regulating cellular metabolism (51) and gene transcription through chromatin remodeling of the inflammatory gene promoter and enhancer regions (52). MyD88 was also shown to directly interact with IFN-γR to stabilize the IFN-γ–induced cytokine and chemokine mRNA (53). In addition, STAT1 was shown to interact with cell cycle regulators cyclin D1 and Cdk4 (54). Consistent with these previous reports, we found that global and B cell–specific deficiency of STAT1 resulted in reduced GC B cell and Tfh responses upon treatment with a TLR7 ligand, highlighting the importance of STAT1 in B cells in TLR7-mediated GC and Tfh responses. Published data and our current findings collectively suggest that TLR7-MyD88 and IFN-γ–STAT1 signaling may synergize to activate multiple biological processes in B cells that are important for mediating SLE-associated AFC, GC, and Tfh responses in TLR7-accelerated SLE autoimmunity.

Although T-bet has been associated with age-associated B cells and class switching to the IgG2a/c isotype (5557), we found significantly reduced TLR7-driven GC and Tfh responses in B cell conditional T-bet–deficient mice. Rubtsova et al. (58) recently showed a B cell–intrinsic role of T-bet in B cell differentiation into age-associated B cells, Spt-GC B cells, and plasma cells and which subsequently increased kidney pathology. In addition, CD11chiT-bet+ B cells have recently been described in SLE patients (59). An earlier report showed that TLR7 stimulation of B cells upregulated T-bet and that T-bet expression was elevated in 564Igi autoreactive B cells (60). Rubtsova et al. (61) similarly found that BCR and TLR7 signaling synergize with IFN-γ signaling to drive T-bet expression in B cells. The Cancro group recently reported that T-bet expression is induced in B cells by TLR signaling but not to BCR and/or anti-CD40 stimulation alone (62). Published data and our current observation together suggest that TLR and IFN-γ–STAT1 signaling cooperate to drive T-bet expression in B cells, which is important for GC, Tfh, and Th1-biased IgG2c Ab responses. Consistent with this idea, we found upregulation of STAT1 and T-bet in TLR7 stimulated B cells, implicating the involvement of both T-bet and STAT1 in TLR7-mediated AFC, GC, and SLE autoimmune responses.

TLR7 can potentially activate multiple signaling pathways (e.g., NF-κB, IRF5/7, and p38/MAPK) to promote autoimmunity (63). Both canonical and noncanonical NF-κB pathways have been shown to be important for foreign Ag-driven AFC and GC responses (64, 65), although the role of NF-κB in spontaneous or TLR7-mediated AFC and GC responses in SLE is unclear. Altered NF-κB activity was previously reported in B cells and T cells from SLE patients (66). IRF5 deficiency abrogates disease development in the FcγRIIB−/− Yaa model of SLE (67). Despite the potential activation of these pathways in TLR7-induced and TLR7-overexpression models of SLE, we found that IFN-γR and STAT1 signaling in B cells played an important role in TLR7-mediated AFC and GC responses. Given the importance of both TLR7 and IFN-γ–STAT1 signaling in autoimmune AFC and GC responses, we postulate that IFN-γ–STAT1 signaling in B cells may co-operate with the NF-κB or IRF5/7 pathway to orchestrate a genetic program required for autoimmune AFC and GC responses in TLR7-accelerated SLE. Cooperation of NF-κB and STAT1 in the transcriptional control of anti-microbial genes in response to in vitro Listeria monocytogenes infection has previously been described (68).

Our RNA-seq data derived from splenic tissues of IMQ-treated Sle1b.IFN-γR−/− and B6.Sle1b mice revealed differential expression of multiple genes and pathways involved in the autoimmune response. Specifically, multiple biological processes are disrupted in the absence of IFN-γR signaling such as cell proliferation and migration, inflammation, and cellular metabolism that can contribute to the development of autoimmunity. In support of the overall reduction in the number of autoantibody-producing AFCs in Sle1b.IFN-γR1−/− mice, the xbp-1 gene that encodes for the transcription factor (XBP-1) and is required for plasma cell differentiation and autoimmune responses (69, 70), is downregulated in these mice. IL-6 signaling is involved in autoimmune GC formation and autoimmune disease progression likely by increasing the production of IFN-γ (71, 72). Interestingly, il6r gene expression is downregulated in Sle1b.IFN-γR1−/− mice. In addition, the relationship between IL-6 and IFN-γ signaling in autoimmunity is further supported by the upregulation of the Smyd2_TSS3427 gene in Sle1b.IFN-γR−/− mice, a gene that encodes a histone methyltransferase that negatively regulates IL-6 production (73). Finally, genes encoding chemokine receptor Cxcr3 and its ligand Cxcl9 that play an important role in autoimmunity development (7476) are downregulated in the absence of IFN-γR signaling. Overall, our data indicate that TLR7 stimulation induces expression of genes that are associated with autoimmune responses in an IFN-γR signaling dependent manner.

In summary, our data define a previously unrecognized indispensable role for IFN-γ signaling and its downstream transcription factors STAT1 and T-bet in B cells in promoting TLR7-accelerated AFC and GC responses and SLE autoimmunity. In contrast, we find moderate effects of T1IFN signaling on these processes. Our current T1IFN-signaling data reflect the moderate efficacy of the clinical trials for anti–IFN-α or anti–IFN-αR therapy in SLE patients. Future treatment approaches could aim to reduce TLR7 signaling in combination with T1IFN or IFN-γ signaling to a threshold that can prevent or treat SLE patients and maintain protection against pathogenic infection.

We thank the Pennsylvania State University Hershey Medical Center flow cytometry core facility for assistance. We thank the Pennsylvania State University Hershey Medical Center Department of Comparative Medicine for animal housing and care. We thank Dr. Meinrad Busslinger for providing CD23Cre mice.

This work was supported by National Institutes of Health RO1AI091670 to Z.S.M.R., Lupus Research Alliance Award 548931 to Z.S.M.R., and the Finkelstein Memorial Award to S.B.C.

The sequences presented in this article have been submitted to the Gene Expression Omnibus (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE141433) under accession number GSE141433.

The online version of this article contains supplemental material.

Abbreviations used in this article:

AFC

Ab-forming cell

ANA

anti-nuclear Ab

GC

germinal center

GN

glomerulonephritis

IMQ

imiquimod

PAS

periodic acid-Schiff

RNA-seq

RNA sequencing

SA

streptavidin

SLE

systemic lupus erythematosus

SmRNP

Smith/ribonucleoprotein

Spt-AFC

spontaneous AFC

Spt-GC

spontaneous GC

Tfh

follicular helper T

T1IFN

type I IFN.

1
Luzina
,
I. G.
,
S. P.
Atamas
,
C. E.
Storrer
,
L. C.
daSilva
,
G.
Kelsoe
,
J. C.
Papadimitriou
,
B. S.
Handwerger
.
2001
.
Spontaneous formation of germinal centers in autoimmune mice.
J. Leukoc. Biol.
70
:
578
584
.
2
Domeier
,
P. P.
,
S. L.
Schell
,
Z. S.
Rahman
.
2017
.
Spontaneous germinal centers and autoimmunity.
Autoimmunity
50
:
4
18
.
3
Wong
,
E. B.
,
T. N.
Khan
,
C.
Mohan
,
Z. S.
Rahman
.
2012
.
The lupus-prone NZM2410/NZW strain-derived Sle1b sublocus alters the germinal center checkpoint in female mice in a B cell-intrinsic manner.
J. Immunol.
189
:
5667
5681
.
4
Jackson
,
S. W.
,
H. M.
Jacobs
,
T.
Arkatkar
,
E. M.
Dam
,
N. E.
Scharping
,
N. S.
Kolhatkar
,
B.
Hou
,
J. H.
Buckner
,
D. J.
Rawlings
.
2016
.
B cell IFN-γ receptor signaling promotes autoimmune germinal centers via cell-intrinsic induction of BCL-6.
J. Exp. Med.
213
:
733
750
.
5
Hua
,
Z.
,
A. J.
Gross
,
C.
Lamagna
,
N.
Ramos-Hernández
,
P.
Scapini
,
M.
Ji
,
H.
Shao
,
C. A.
Lowell
,
B.
Hou
,
A. L.
DeFranco
.
2014
.
Requirement for MyD88 signaling in B cells and dendritic cells for germinal center anti-nuclear antibody production in Lyn-deficient mice.
J. Immunol.
192
:
875
885
.
6
Cappione
,
A.
 III
,
J. H.
Anolik
,
A.
Pugh-Bernard
,
J.
Barnard
,
P.
Dutcher
,
G.
Silverman
,
I.
Sanz
.
2005
.
Germinal center exclusion of autoreactive B cells is defective in human systemic lupus erythematosus.
J. Clin. Invest.
115
:
3205
3216
.
7
Vinuesa
,
C. G.
,
I.
Sanz
,
M. C.
Cook
.
2009
.
Dysregulation of germinal centres in autoimmune disease.
Nat. Rev. Immunol.
9
:
845
857
.
8
Jenks
,
S. A.
,
K. S.
Cashman
,
E.
Zumaquero
,
U. M.
Marigorta
,
A. V.
Patel
,
X.
Wang
,
D.
Tomar
,
M. C.
Woodruff
,
Z.
Simon
,
R.
Bugrovsky
, et al
.
2018
.
Distinct effector B cells induced by unregulated toll-like receptor 7 contribute to pathogenic responses in systemic lupus erythematosus.
Immunity
49
:
725
739.e6
.
9
Celhar
,
T.
,
A. M.
Fairhurst
.
2014
.
Toll-like receptors in systemic lupus erythematosus: potential for personalized treatment.
Front. Pharmacol.
5
:
265
.
10
Mohan
,
C.
,
C.
Putterman
.
2015
.
Genetics and pathogenesis of systemic lupus erythematosus and lupus nephritis.
Nat. Rev. Nephrol.
11
:
329
341
.
11
Walsh
,
E. R.
,
P.
Pisitkun
,
E.
Voynova
,
J. A.
Deane
,
B. L.
Scott
,
R. R.
Caspi
,
S.
Bolland
.
2012
.
Dual signaling by innate and adaptive immune receptors is required for TLR7-induced B-cell-mediated autoimmunity.
Proc. Natl. Acad. Sci. USA
109
:
16276
16281
.
12
Hwang
,
S. H.
,
H.
Lee
,
M.
Yamamoto
,
L. A.
Jones
,
J.
Dayalan
,
R.
Hopkins
,
X. J.
Zhou
,
F.
Yarovinsky
,
J. E.
Connolly
,
M. A.
Curotto de Lafaille
, et al
.
2012
.
B cell TLR7 expression drives anti-RNA autoantibody production and exacerbates disease in systemic lupus erythematosus-prone mice.
J. Immunol.
189
:
5786
5796
.
13
Bubier
,
J. A.
,
T. J.
Sproule
,
O.
Foreman
,
R.
Spolski
,
D. J.
Shaffer
,
H. C.
Morse
III
,
W. J.
Leonard
,
D. C.
Roopenian
.
2009
.
A critical role for IL-21 receptor signaling in the pathogenesis of systemic lupus erythematosus in BXSB-Yaa mice.
Proc. Natl. Acad. Sci. USA
106
:
1518
1523
.
14
Souyris
,
M.
,
C.
Cenac
,
P.
Azar
,
D.
Daviaud
,
A.
Canivet
,
S.
Grunenwald
,
C.
Pienkowski
,
J.
Chaumeil
,
J. E.
Mejía
,
J. C.
Guéry
.
2018
.
TLR7 escapes X chromosome inactivation in immune cells.
Sci. Immunol.
3
:
eaap8855
.
15
Christensen
,
S. R.
,
J.
Shupe
,
K.
Nickerson
,
M.
Kashgarian
,
R. A.
Flavell
,
M. J.
Shlomchik
.
2006
.
Toll-like receptor 7 and TLR9 dictate autoantibody specificity and have opposing inflammatory and regulatory roles in a murine model of lupus.
Immunity
25
:
417
428
.
16
Soni
,
C.
,
E. B.
Wong
,
P. P.
Domeier
,
T. N.
Khan
,
T.
Satoh
,
S.
Akira
,
Z. S.
Rahman
.
2014
.
B cell-intrinsic TLR7 signaling is essential for the development of spontaneous germinal centers.
J. Immunol.
193
:
4400
4414
.
17
Jackson
,
S. W.
,
N. E.
Scharping
,
N. S.
Kolhatkar
,
S.
Khim
,
M. A.
Schwartz
,
Q. Z.
Li
,
K. L.
Hudkins
,
C. E.
Alpers
,
D.
Liggitt
,
D. J.
Rawlings
.
2014
.
Opposing impact of B cell-intrinsic TLR7 and TLR9 signals on autoantibody repertoire and systemic inflammation.
J. Immunol.
192
:
4525
4532
.
18
Pollard
,
K. M.
,
D. M.
Cauvi
,
C. B.
Toomey
,
K. V.
Morris
,
D. H.
Kono
.
2013
.
Interferon-γ and systemic autoimmunity.
Discov. Med.
16
:
123
131
.
19
Kiefer
,
K.
,
M. A.
Oropallo
,
M. P.
Cancro
,
A.
Marshak-Rothstein
.
2012
.
Role of type I interferons in the activation of autoreactive B cells.
Immunol. Cell Biol.
90
:
498
504
.
20
Wilson
,
L. E.
,
D.
Widman
,
S. H.
Dikman
,
P. D.
Gorevic
.
2002
.
Autoimmune disease complicating antiviral therapy for hepatitis C virus infection.
Semin. Arthritis Rheum.
32
:
163
173
.
21
Agrawal
,
H.
,
N.
Jacob
,
E.
Carreras
,
S.
Bajana
,
C.
Putterman
,
S.
Turner
,
B.
Neas
,
A.
Mathian
,
M. N.
Koss
,
W.
Stohl
, et al
.
2009
.
Deficiency of type I IFN receptor in lupus-prone New Zealand mixed 2328 mice decreases dendritic cell numbers and activation and protects from disease.
J. Immunol.
183
:
6021
6029
.
22
Mathian
,
A.
,
A.
Weinberg
,
M.
Gallegos
,
J.
Banchereau
,
S.
Koutouzov
.
2005
.
IFN-alpha induces early lethal lupus in preautoimmune (New Zealand Black x New Zealand White) F1 but not in BALB/c mice.
J. Immunol.
174
:
2499
2506
.
23
Ramanujam
,
M.
,
P.
Kahn
,
W.
Huang
,
H.
Tao
,
M. P.
Madaio
,
S. M.
Factor
,
A.
Davidson
.
2009
.
Interferon-alpha treatment of female (NZW x BXSB)F(1) mice mimics some but not all features associated with the Yaa mutation.
Arthritis Rheum.
60
:
1096
1101
.
24
Moisini
,
I.
,
W.
Huang
,
R.
Bethunaickan
,
R.
Sahu
,
P. G.
Ricketts
,
M.
Akerman
,
T.
Marion
,
M.
Lesser
,
A.
Davidson
.
2012
.
The Yaa locus and IFN-α fine-tune germinal center B cell selection in murine systemic lupus erythematosus.
J. Immunol.
189
:
4305
4312
.
25
Domeier
,
P. P.
,
S. B.
Chodisetti
,
S. L.
Schell
,
Y. I.
Kawasawa
,
M. J.
Fasnacht
,
C.
Soni
,
Z. S. M.
Rahman
.
2018
.
B-cell-intrinsic type 1 interferon signaling is crucial for loss of tolerance and the development of autoreactive B cells.
Cell Rep.
24
:
406
418
.
26
Hamilton
,
J. A.
,
Q.
Wu
,
P.
Yang
,
B.
Luo
,
S.
Liu
,
H.
Hong
,
J.
Li
,
M. R.
Walter
,
E. N.
Fish
,
H. C.
Hsu
,
J. D.
Mountz
.
2017
.
Cutting edge: endogenous IFN-β regulates survival and development of transitional B cells.
J. Immunol.
199
:
2618
2623
.
27
Harigai
,
M.
,
M.
Kawamoto
,
M.
Hara
,
T.
Kubota
,
N.
Kamatani
,
N.
Miyasaka
.
2008
.
Excessive production of IFN-gamma in patients with systemic lupus erythematosus and its contribution to induction of B lymphocyte stimulator/B cell-activating factor/TNF ligand superfamily-13B.
J. Immunol.
181
:
2211
2219
.
28
Csiszár
,
A.
,
G.
Nagy
,
P.
Gergely
,
T.
Pozsonyi
,
E.
Pócsik
.
2000
.
Increased interferon-gamma (IFN-gamma), IL-10 and decreased IL-4 mRNA expression in peripheral blood mononuclear cells (PBMC) from patients with systemic lupus erythematosus (SLE).
Clin. Exp. Immunol.
122
:
464
470
.
29
Welcher
,
A. A.
,
M.
Boedigheimer
,
A.
Kivitz
,
Z.
Amoura
,
J.
Buyon
,
A.
Rudinskaya
,
K.
Latinis
,
K.
Chiu
,
K. S.
Oliner
,
M. A.
Damore
, et al.
2015
.
Blockade of interferon-γ normalizes interferon-regulated gene expression and serum CXCL10 levels in patients with systemic lupus erythematosus.
Arthritis Rheumatol.
67
:
2713
2722
.
30
Jacob
,
C. O.
,
P. H.
van der Meide
,
H. O.
McDevitt
.
1987
.
In vivo treatment of (NZB X NZW)F1 lupus-like nephritis with monoclonal antibody to gamma interferon.
J. Exp. Med.
166
:
798
803
.
31
Ozmen
,
L.
,
D.
Roman
,
M.
Fountoulakis
,
G.
Schmid
,
B.
Ryffel
,
G.
Garotta
.
1995
.
Experimental therapy of systemic lupus erythematosus: the treatment of NZB/W mice with mouse soluble interferon-gamma receptor inhibits the onset of glomerulonephritis.
Eur. J. Immunol.
25
:
6
12
.
32
Haas
,
C.
,
B.
Ryffel
,
M.
Le Hir
.
1998
.
IFN-gamma receptor deletion prevents autoantibody production and glomerulonephritis in lupus-prone (NZB x NZW)F1 mice.
J. Immunol.
160
:
3713
3718
.
33
Lawson
,
B. R.
,
G. J.
Prud’homme
,
Y.
Chang
,
H. A.
Gardner
,
J.
Kuan
,
D. H.
Kono
,
A. N.
Theofilopoulos
.
2000
.
Treatment of murine lupus with cDNA encoding IFN-gammaR/Fc.
J. Clin. Invest.
106
:
207
215
.
34
Schwarting
,
A.
,
T.
Wada
,
K.
Kinoshita
,
G.
Tesch
,
V. R.
Kelley
.
1998
.
IFN-gamma receptor signaling is essential for the initiation, acceleration, and destruction of autoimmune kidney disease in MRL-Fas(lpr) mice.
J. Immunol.
161
:
494
503
.
35
Balomenos
,
D.
,
R.
Rumold
,
A. N.
Theofilopoulos
.
1998
.
Interferon-gamma is required for lupus-like disease and lymphoaccumulation in MRL-lpr mice.
J. Clin. Invest.
101
:
364
371
.
36
Domeier
,
P. P.
,
S. B.
Chodisetti
,
C.
Soni
,
S. L.
Schell
,
M. J.
Elias
,
E. B.
Wong
,
T. K.
Cooper
,
D.
Kitamura
,
Z. S.
Rahman
.
2016
.
IFN-γ receptor and STAT1 signaling in B cells are central to spontaneous germinal center formation and autoimmunity.
J. Exp. Med.
213
:
715
732
.
37
Morel
,
L.
,
B. P.
Croker
,
K. R.
Blenman
,
C.
Mohan
,
G.
Huang
,
G.
Gilkeson
,
E. K.
Wakeland
.
2000
.
Genetic reconstitution of systemic lupus erythematosus immunopathology with polycongenic murine strains.
Proc. Natl. Acad. Sci. USA
97
:
6670
6675
.
38
Yokogawa
,
M.
,
M.
Takaishi
,
K.
Nakajima
,
R.
Kamijima
,
C.
Fujimoto
,
S.
Kataoka
,
Y.
Terada
,
S.
Sano
.
2014
.
Epicutaneous application of toll-like receptor 7 agonists leads to systemic autoimmunity in wild-type mice: a new model of systemic Lupus erythematosus.
Arthritis Rheumatol.
66
:
694
706
.
39
Liu
,
Y.
,
N. L.
Seto
,
C.
Carmona-Rivera
,
M. J.
Kaplan
.
2018
.
Accelerated model of lupus autoimmunity and vasculopathy driven by toll-like receptor 7/9 imbalance.
Lupus Sci. Med.
5
: e000259.
40
Soni
,
C.
,
P. P.
Domeier
,
E. B.
Wong
,
T. N.
Shwetank
,
T. N.
Khan
,
M. J.
Elias
,
S. L.
Schell
,
A. E.
Lukacher
,
T. K.
Cooper
,
Z. S.
Rahman
.
2015
.
Distinct and synergistic roles of FcγRIIB deficiency and 129 strain-derived SLAM family proteins in the development of spontaneous germinal centers and autoimmunity.
J. Autoimmun.
63
:
31
46
.
41
Corradetti
,
C.
,
N. R.
Jog
,
M.
Cesaroni
,
M.
Madaio
,
R.
Caricchio
.
2018
.
Estrogen receptor α signaling exacerbates immune-mediated nephropathies through alteration of metabolic activity.
J. Immunol.
200
:
512
522
.
42
Trapnell
,
C.
,
S. L.
Salzberg
.
2009
.
How to map billions of short reads onto genomes.
Nat. Biotechnol.
27
:
455
457
.
43
Anders
,
S.
,
W.
Huber
.
2010
.
Differential expression analysis for sequence count data.
Genome Biol.
11
:
R106
.
44
Thibault
,
D. L.
,
A. D.
Chu
,
K. L.
Graham
,
I.
Balboni
,
L. Y.
Lee
,
C.
Kohlmoos
,
A.
Landrigan
,
J. P.
Higgins
,
R.
Tibshirani
,
P. J.
Utz
.
2008
.
IRF9 and STAT1 are required for IgG autoantibody production and B cell expression of TLR7 in mice.
J. Clin. Invest.
118
:
1417
1426
.
45
Green
,
N. M.
,
A.
Laws
,
K.
Kiefer
,
L.
Busconi
,
Y. M.
Kim
,
M. M.
Brinkmann
,
E. H.
Trail
,
K.
Yasuda
,
S. R.
Christensen
,
M. J.
Shlomchik
, et al
.
2009
.
Murine B cell response to TLR7 ligands depends on an IFN-beta feedback loop.
J. Immunol.
183
:
1569
1576
.
46
Thibault
,
D. L.
,
K. L.
Graham
,
L. Y.
Lee
,
I.
Balboni
,
P. J.
Hertzog
,
P. J.
Utz
.
2009
.
Type I interferon receptor controls B-cell expression of nucleic acid-sensing toll-like receptors and autoantibody production in a murine model of lupus.
Arthritis Res. Ther.
11
:
R112
.
47
Teichmann
,
L. L.
,
D.
Schenten
,
R.
Medzhitov
,
M.
Kashgarian
,
M. J.
Shlomchik
.
2013
.
Signals via the adaptor MyD88 in B cells and DCs make distinct and synergistic contributions to immune activation and tissue damage in lupus.
Immunity
38
:
528
540
.
48
Sakata
,
K.
,
S.
Nakayamada
,
Y.
Miyazaki
,
S.
Kubo
,
A.
Ishii
,
K.
Nakano
,
Y.
Tanaka
.
2018
.
Up-regulation of TLR7-mediated IFN-α production by plasmacytoid dendritic cells in patients with systemic lupus erythematosus.
Front. Immunol.
9
:
1957
.
49
Yasuda
,
K.
,
C.
Richez
,
J. W.
Maciaszek
,
N.
Agrawal
,
S.
Akira
,
A.
Marshak-Rothstein
,
I. R.
Rifkin
.
2007
.
Murine dendritic cell type I IFN production induced by human IgG-RNA immune complexes is IFN regulatory factor (IRF)5 and IRF7 dependent and is required for IL-6 production.
J. Immunol.
178
:
6876
6885
.
50
Hu
,
X.
,
L. B.
Ivashkiv
.
2009
.
Cross-regulation of signaling pathways by interferon-gamma: implications for immune responses and autoimmune diseases.
Immunity
31
:
539
550
.
51
Su
,
X.
,
Y.
Yu
,
Y.
Zhong
,
E. G.
Giannopoulou
,
X.
Hu
,
H.
Liu
,
J. R.
Cross
,
G.
Rätsch
,
C. M.
Rice
,
L. B.
Ivashkiv
.
2015
.
Interferon-γ regulates cellular metabolism and mRNA translation to potentiate macrophage activation.
Nat. Immunol.
16
:
838
849
.
52
Qiao
,
Y.
,
E. G.
Giannopoulou
,
C. H.
Chan
,
S. H.
Park
,
S.
Gong
,
J.
Chen
,
X.
Hu
,
O.
Elemento
,
L. B.
Ivashkiv
.
2013
.
Synergistic activation of inflammatory cytokine genes by interferon-γ-induced chromatin remodeling and toll-like receptor signaling.
Immunity
39
:
454
469
.
53
Sun
,
D.
,
A.
Ding
.
2006
.
MyD88-mediated stabilization of interferon-gamma-induced cytokine and chemokine mRNA.
Nat. Immunol.
7
:
375
381
.
54
Dimco
,
G.
,
R. A.
Knight
,
D. S.
Latchman
,
A.
Stephanou
.
2010
.
STAT1 interacts directly with cyclin D1/Cdk4 and mediates cell cycle arrest. [Published erratum appears in 2016 Cell Cycle 15: 1387–1388.]
Cell Cycle
9
:
4638
4649
.
55
Rubtsov
,
A. V.
,
K.
Rubtsova
,
A.
Fischer
,
R. T.
Meehan
,
J. Z.
Gillis
,
J. W.
Kappler
,
P.
Marrack
.
2011
.
Toll-like receptor 7 (TLR7)-driven accumulation of a novel CD11c+ B-cell population is important for the development of autoimmunity.
Blood
118
:
1305
1315
.
56
Rubtsova
,
K.
,
A. V.
Rubtsov
,
M. P.
Cancro
,
P.
Marrack
.
2015
.
Age-associated B cells: a T-bet-dependent effector with roles in protective and pathogenic immunity.
J. Immunol.
195
:
1933
1937
.
57
Myles
,
A.
,
P. J.
Gearhart
,
M. P.
Cancro
.
2017
.
Signals that drive T-bet expression in B cells.
Cell. Immunol.
321
:
3
7
.
58
Rubtsova
,
K.
,
A. V.
Rubtsov
,
J. M.
Thurman
,
J. M.
Mennona
,
J. W.
Kappler
,
P.
Marrack
.
2017
.
B cells expressing the transcription factor T-bet drive lupus-like autoimmunity.
J. Clin. Invest.
127
:
1392
1404
.
59
Wang
,
S.
,
J.
Wang
,
V.
Kumar
,
J. L.
Karnell
,
B.
Naiman
,
P. S.
Gross
,
S.
Rahman
,
K.
Zerrouki
,
R.
Hanna
,
C.
Morehouse
, et al
Autoimmunity Molecular Medicine Team
.
2018
.
IL-21 drives expansion and plasma cell differentiation of autoreactive CD11chiT-bet+ B cells in SLE.
Nat. Commun.
9
:
1758
.
60
Berland
,
R.
,
L.
Fernandez
,
E.
Kari
,
J. H.
Han
,
I.
Lomakin
,
S.
Akira
,
H. H.
Wortis
,
J. F.
Kearney
,
A. A.
Ucci
,
T.
Imanishi-Kari
.
2006
.
Toll-like receptor 7-dependent loss of B cell tolerance in pathogenic autoantibody knockin mice.
Immunity
25
:
429
440
.
61
Rubtsova
,
K.
,
A. V.
Rubtsov
,
L. F.
van Dyk
,
J. W.
Kappler
,
P.
Marrack
.
2013
.
T-box transcription factor T-bet, a key player in a unique type of B-cell activation essential for effective viral clearance.
Proc. Natl. Acad. Sci. USA
110
:
E3216
E3224
.
62
Naradikian
,
M. S.
,
A.
Myles
,
D. P.
Beiting
,
K. J.
Roberts
,
L.
Dawson
,
R. S.
Herati
,
B.
Bengsch
,
S. L.
Linderman
,
E.
Stelekati
,
R.
Spolski
, et al
.
2016
.
Cutting edge: IL-4, IL-21, and IFN-γ interact to govern T-bet and CD11c expression in TLR-activated B cells.
J. Immunol.
197
:
1023
1028
.
63
Suthers
,
A. N.
,
S.
Sarantopoulos
.
2017
.
TLR7/TLR9- and B cell receptor-signaling crosstalk: promotion of potentially dangerous B cells.
Front. Immunol.
8
:
775
.
64
De Silva
,
N. S.
,
M. M.
Anderson
,
A.
Carette
,
K.
Silva
,
N.
Heise
,
G.
Bhagat
,
U.
Klein
.
2016
.
Transcription factors of the alternative NF-κB pathway are required for germinal center B-cell development.
Proc. Natl. Acad. Sci. USA
113
:
9063
9068
.
65
Heise
,
N.
,
N. S.
De Silva
,
K.
Silva
,
A.
Carette
,
G.
Simonetti
,
M.
Pasparakis
,
U.
Klein
.
2014
.
Germinal center B cell maintenance and differentiation are controlled by distinct NF-κB transcription factor subunits.
J. Exp. Med.
211
:
2103
2118
.
66
Wong
,
H. K.
,
G. M.
Kammer
,
G.
Dennis
,
G. C.
Tsokos
.
1999
.
Abnormal NF-kappa B activity in T lymphocytes from patients with systemic lupus erythematosus is associated with decreased p65-RelA protein expression.
J. Immunol.
163
:
1682
1689
.
67
Richez
,
C.
,
K.
Yasuda
,
R. G.
Bonegio
,
A. A.
Watkins
,
T.
Aprahamian
,
P.
Busto
,
R. J.
Richards
,
C. L.
Liu
,
R.
Cheung
,
P. J.
Utz
, et al
.
2010
.
IFN regulatory factor 5 is required for disease development in the FcgammaRIIB-/-Yaa and FcgammaRIIB-/- mouse models of systemic lupus erythematosus.
J. Immunol.
184
:
796
806
.
68
Wienerroither
,
S.
,
P.
Shukla
,
M.
Farlik
,
A.
Majoros
,
B.
Stych
,
C.
Vogl
,
H.
Cheon
,
G. R.
Stark
,
B.
Strobl
,
M.
Müller
,
T.
Decker
.
2015
.
Cooperative transcriptional activation of antimicrobial genes by STAT and NF-κB pathways by concerted recruitment of the mediator complex.
Cell Rep.
12
:
300
312
.
69
Savic
,
S.
,
L.
Ouboussad
,
L. J.
Dickie
,
J.
Geiler
,
C.
Wong
,
G. M.
Doody
,
S. M.
Churchman
,
F.
Ponchel
,
P.
Emery
,
G. P.
Cook
, et al
.
2014
.
TLR dependent XBP-1 activation induces an autocrine loop in rheumatoid arthritis synoviocytes.
J. Autoimmun.
50
:
59
66
.
70
Reimold
,
A. M.
,
N. N.
Iwakoshi
,
J.
Manis
,
P.
Vallabhajosyula
,
E.
Szomolanyi-Tsuda
,
E. M.
Gravallese
,
D.
Friend
,
M. J.
Grusby
,
F.
Alt
,
L. H.
Glimcher
.
2001
.
Plasma cell differentiation requires the transcription factor XBP-1.
Nature
412
:
300
307
.
71
Arkatkar
,
T.
,
S. W.
Du
,
H. M.
Jacobs
,
E. M.
Dam
,
B.
Hou
,
J. H.
Buckner
,
D. J.
Rawlings
,
S. W.
Jackson
.
2017
.
B cell-derived IL-6 initiates spontaneous germinal center formation during systemic autoimmunity.
J. Exp. Med.
214
:
3207
3217
.
72
Jones
,
B. E.
,
M. D.
Maerz
,
J. H.
Buckner
.
2018
.
IL-6: a cytokine at the crossroads of autoimmunity.
Curr. Opin. Immunol.
55
:
9
14
.
73
Xu
,
G.
,
G.
Liu
,
S.
Xiong
,
H.
Liu
,
X.
Chen
,
B.
Zheng
.
2015
.
The histone methyltransferase Smyd2 is a negative regulator of macrophage activation by suppressing interleukin 6 (IL-6) and tumor necrosis factor α (TNF-α) production.
J. Biol. Chem.
290
:
5414
5423
.
74
Menke
,
J.
,
G. C.
Zeller
,
E.
Kikawada
,
T. K.
Means
,
X. R.
Huang
,
H. Y.
Lan
,
B.
Lu
,
J.
Farber
,
A. D.
Luster
,
V. R.
Kelley
.
2008
.
CXCL9, but not CXCL10, promotes CXCR3-dependent immune-mediated kidney disease.
J. Am. Soc. Nephrol.
19
:
1177
1189
.
75
Liao
,
X.
,
T.
Pirapakaran
,
X. M.
Luo
.
2016
.
Chemokines and chemokine receptors in the development of lupus nephritis.
Mediators Inflamm.
2016
: 6012715.
76
Bauer
,
J. W.
,
E. C.
Baechler
,
M.
Petri
,
F. M.
Batliwalla
,
D.
Crawford
,
W. A.
Ortmann
,
K. J.
Espe
,
W.
Li
,
D. D.
Patel
,
P. K.
Gregersen
,
T. W.
Behrens
.
2006
.
Elevated serum levels of interferon-regulated chemokines are biomarkers for active human systemic lupus erythematosus.
PLoS Med.
3
: e491.

The authors have no financial conflicts of interest.

Supplementary data