Necroptosis is a form of cell death associated with inflammation; however, the biological consequences of chronic necroptosis are unknown. Necroptosis is mediated by RIPK1, RIPK3, and MLKL kinases but in hematopoietic cells RIPK1 has anti-inflammatory roles and functions to prevent necroptosis. Here we interrogate the consequences of chronic necroptosis on immune homeostasis by deleting Ripk1 in mouse dendritic cells. We demonstrate that deregulated necroptosis results in systemic inflammation, tissue fibrosis, and autoimmunity. We show that inflammation and autoimmunity are prevented upon expression of kinase inactive RIPK1 or deletion of RIPK3 or MLKL. We provide evidence that the inflammation is not driven by microbial ligands, but depends on the release of danger-associated molecular patterns and MyD88-dependent signaling. Importantly, although the inflammation is independent of type I IFN and the nucleic acid sensing TLRs, blocking these pathways rescues the autoimmunity. These mouse genetic studies reveal that chronic necroptosis may underlie human fibrotic and autoimmune disorders.

This article is featured in In This Issue, p.373

Receptor interacting protein kinase (RIPK) 1 is a key component of the necroptotic and apoptotic cell death pathways, and is important for the optimal activation of the NF-κB and MAPK pathways. TNF normally induces NF-κB and MAPK activation, but under certain conditions can induce apoptosis or, when caspases are inhibited, stimulate necroptosis. Necroptosis is an inflammatory form of cell death triggered by death ligands such as TNF, FasL, TRAIL, and type I and type II IFNs or by activation of pathogen recognition receptors including TLR 3 or 4 (1). RIPK1 initiates the necroptotic kinase cascade by phosphorylating and activating RIPK3, which then activates the pseudo-kinase mixed lineage kinase domain-like (MLKL) (2, 3). MLKL phosphorylation results in its translocation to the plasma membrane and changes in membrane permeability (4), resulting in the release of danger-associated molecular patterns (DAMPs) such as HMGB1, ATP, and mitochondrial DNA (5). These DAMPs activate TLRs on macrophages and dendritic cells (DCs) to induce and amplify proinflammatory cytokine and chemokine production. In some cell types, RIPK1 kinase activity is crucial for the activation of necroptosis, as the kinase inhibitor Necrostatin-1 prevents necroptosis (6) and RIPK1 kinase inactive mice, Ripk1D138N, are resistant to necroptosis and TNF-induced shock in vivo (7, 8). RIPK1 has also been shown to have essential kinase-independent scaffold functions that mediate cell survival due to effects on the canonical (9) or noncanonical NF-κB pathways, depending on the cell type (10).

Complete RIPK1 deficiency results in postnatal lethality (9) driven by an increased sensitivity to both RIPK3-dependent necroptosis and caspase-8–dependent apoptosis, whereby compound deletion of both Caspase-8 and Ripk3 or Caspase-8 and Mlkl or the deletion of Ripk3 and TNFR type 1 (Tnfr1) rescues RIPK1-associated lethality (1113). Whether RIPK1 triggers apoptosis and/or necroptosis is cell type and context dependent. For example, RIPK1 is essential for necroptosis in murine embryonic fibroblasts and bone marrow–derived macrophages (8, 14) but functions to negatively regulate RIPK3 and MLKL in hematopoietic stem and progenitor cells and keratinocytes (11, 15, 16). Ripk1 deletion in intestinal epithelial cells sensitizes to both TNF-mediated apoptosis and necroptosis (16). These findings reveal that RIPK1 can positively or negatively regulate necroptosis or apoptosis depending on cellular context.

DCs are critical to maintain immune homeostasis and to generate successful responses to infection. Given the important roles DCs have in maintaining tolerance, we examined the in vivo consequences of DC necroptosis on immune homeostasis by deleting Ripk1 in DCs. We found that RIPK1-deficient DCs exhibit normal responses to TNF- and FasL-induced apoptosis, but have an increased sensitivity to necroptosis in vitro. Mice with a DC RIPK1 deficiency develop inflammation and autoimmunity, characterized by splenomegaly, lymphadenopathy, tissue fibrosis, and production of anti‐nuclear autoantibodies (ANAs). We demonstrate that inflammation and autoimmunity associated with a DC RIPK1 deficiency are rescued by expression of kinase inactive RIPK1 or an absence of RIPK3, MLKL, or the TLR adapter MyD88, thereby implicating necroptosis as the mediator of inflammation, and MyD88-dependent TLR signaling as an amplifier of DAMP signaling. Importantly, autoantibody production but not inflammation was prevented by an absence of the type I IFNR or UNC93B1-dependent TLR 3, 7, and 9 signaling, thereby genetically separating signals that trigger DAMP release, inflammation, and fibrosis from those responsible for autoimmunity. This study provides genetic evidence that cell death and inflammation precede the development of autoimmunity and suggests that necroptosis kinase inhibitors may be useful in at-risk individuals to prevent autoimmune disease.

Ripk1 conditional mice (Ripk1fl/fl) (16) were crossed with CD11cCre (Itgax-cre) (17), Ripk3−/− mice (a gift from V. Dixit, Genentech, San Francisco), Mlkl−/− (4), Ifngr1−/− (18), Tnfr1−/− (19), MyD88−/− (20), Ifnar1−/− (21), and Unc93b13d/3d (22) mice. B6.Cg-Tg(TcraTcrb)425Cbn/J (OT-II) and Gt(ROSA)26Sortm9(CAG-tdTomato)Hze mice were obtained from Jackson Laboratory. All animal procedures used in this study were approved by the University of Massachusetts Medical School Institutional Animal Care and Use Committee. For antibiotic treatment, ampicillin (1 mg/ml), neomycin (1 mg/ml), ciprofloxacin (0.5 mg/ml), meropenem (0.5 mg/ml), and Grape Kool-Aid (20 mg/ml) were added to drinking water from 2 d after birth. Following weaning, ciprofloxacin was substituted with vancomycin (0.5 mg/ml). When littermate controls were not used, sex-matched control mice carrying the CD11cCre transgene were cohoused with experimental mice. For LPS-induced endotoxic shock experiments, age and sex-matched mice were i.p. injected with 5 mg/kg LPS from Escherichia coli (Sigma) and re-extracted using phenol chloroform as previously described (23).

Bone marrow–derived DCs (BMDCs) were generated by culturing bone marrow cell suspensions in 20 ng/ml recombinant GM-CSF (PeproTech) for 10 d. For necroptosis assays, BMDCs were treated with 0.1 μM SMAC mimetic (ChemieTek) and 10 μM zVAD (Enzo). For apoptosis assays, BMDCs were treated with cycloheximide (0.5 μg/ml), TNF-α (10 ng/ml), IFN-γ (10 ng/ml), or with FasL and control vesicles purified from N2-mFasL and N2-neo cell supernatant (diluted 1/40), as previously described (24). Splenic DCs were isolated from mice following treatment of the Flt3L producing melanoma line B16, using a CD11c positive selection kit (Stemcell Technologies).

To examine T cell proliferation, purified CD11c+ splenic DCs from mice of the indicated genotypes were incubated with OVA323–339 or control OVA257–264 peptide for 1 h. CD4+ T cells were isolated from spleens of OT-II mice using CD4 positive selection beads (Invitrogen). Isolated CD4+ cells were labeled with 0.5 μM CFSE (Invitrogen) and incubated with splenic DCs for 72 h. CFSE staining was examined in viable CD4+ cells by flow cytometry.

ANAs were detected by immunofluorescence on HEp-2 slides (Abs) as previously described (25).

Tissues were fixed in 10% formalin (Fisher Scientific). Slides were stained with H&E or Masson’s trichrome. Images were taken on an Olympus BX41 microscope using an Evolution MP 5.0 Mega-Pixel Camera (MediaCybernetics) and QCapture Pro software (QImaging).

Single-cell suspensions were stained with cell-surface Abs and DAPI (Molecular Probes) was used to distinguish between live and dead cells. Samples were run on a BD LSRII flow cytometer (BD Biosciences) and analyzed using FlowJo software (Tree Star). Cell populations were determined as follows: erythrocytes, CD71 Ter119+; erythroblasts, CD71+ Ter119+; neutrophils, CD11b+ Ly-6Cint Gr-1+; monocytes, CD11b+, Ly-6Chi and Gr-1lo; CD4 T lymphocytes, CD4+ CD8; CD8 T lymphocytes, CD8+ CD4; B lymphocytes, B220+ CD4 CD8; germinal center B cells, B220+ Fas+ GL7+; progenitors, lineagec-Kit+. DCs were gated according to (26).

Serum cytokines were measured using an 11-plex protein/peptide multiplex analysis (Luminex Technology) conducted by the National Mouse Metabolic Phenotyping Center at the University of Massachusetts Medical School. Chemokines and cytokines below the level of detection were assigned the value of zero. Serum Flt3L levels were measured by ELISA (R&D Systems). IFN-β ELISA kit was used as described previously (27).

RNA was prepared using TRIzol (Invitrogen) or with the RNeasy Mini kit (Qiagen). Each RNA sample was adjusted to contain the same quantity using the Nanodrop ND-1000 spectrophotometer (Thermo Scientific). RNA was used for quantitative RT-PCR using Power SYBR Green PCR Master Mix (Applied Biosystems) with the following primer pairs: β-actin, sense, 5′-TGG CAT AGA GGT CTT TAC GGA-3′, antisense, 5′-TTG AAC ATG GCA TTG TTA CCA A-3′; Iκbα, sense, 5′-TGA AGG ACG AGG AGT ACG AGC-3′, antisense, 5′-TTC GTG GAT GAT TGC CAA GTG-3′; A20, sense, 5′-CTT TCT TCA TGT CCG TGA ACA CT-3′, antisense, 5′-TTC AGG GCC TAG CTT CGA GT-3′; cIAP1, sense, 5′-TGT GGC CTG ATG TTG GAT AAC-3′, antisense, 5′-GGT GAC GAA TGT GCA AAT CTA CT-3′; cIAP2, sense, 5′-GCT GTG GCC TAA TGC TAG ACA-3′, antisense, 5′-GGA CAA TCT TGA TTT GCT CGG AA-3′; XIAP, sense, 5′-CGA GCT GGG TTT CTT TAT ACC G-3′, antisense, 5′-GCA ATT ATG GGA TAT TCT CCT GT-3′.

An anti-mouse albumin ELISA was used to measure urine protein (Bethyl Laboratories).

Cell lysates were prepared in RIPA buffer (150 mM NaCl, 50 mM Tris-HCl [pH 7.5], 1% NP40, 0.25% deoxycholate, 0.1% SDS, 1 mM EDTA), supplemented with protease inhibitors (Roche Applied Science), 1 mM DTT, 1 mM Na3VaO4, and 1 mM PMSF and boiled with SDS reducing sample buffer. Lysates were run on 4–12% Bis-Tris gels (Invitrogen). Membranes were probed with phospho-MLKL (Abcam), cIAP1 (Enzo Life Sciences), cIAP2 (R&D Systems), RIPK1 (BD Transduction), RIPK3 (Prosci), XIAP (MBL), β-Actin (Sigma-Aldrich), Erk 1/2 (Cell Signaling Technology), MLKL (a gift from J. Han) or phospho-IκBα (Cell Signaling Technology) Abs.

Statistical analyses were performed using GraphPad Prism software, version 6.0. Kaplan–Meier survival curves were analyzed using a log-rank test with a 95% confidence interval. A two-sided p < 0.05 was considered statistically significant for Student t tests and two-way ANOVA tests, with *p < 0.05, **p < 0.01.

To determine the effects of DC necroptosis on immune homeostasis, mice containing a conditional allele of Ripk1 (16) were crossed to CD11cCre transgenic mice (hereafter referred to as Ripk1DC KO). Ripk1DC KO mice were born at the expected Mendelian ratios and developed to adulthood normally. These mice exhibit a specific loss of RIPK1 expression in CD11c+-enriched splenic DCs by Western blot analysis (Fig. 1A).

FIGURE 1.

RIPK1-deficient DCs are more sensitive to necroptosis. (A) Western blot depicting RIPK1 expression in CD11c+-enriched splenic DCs from CD11cCre and Ripk1DC KO mice. (B) RIPK1-deficient DCs are more susceptible to necroptosis. BMDCs were treated with 0.1 μM SMAC mimetics and 10 μM zVAD-fmk. Effects on cell viability were measured at 6 and 18 h using Cell Titer Glo. n = 6–8 independent samples. (C) Western blot depicting MLKL phosphorylation and total MLKL, RIPK1, and RIPK3 levels in BMDCs treated with SMAC mimetics and zVAD-fmk. Quantification of pMLKL relative to MLKL from three Western blots is shown. (D) Number of splenic conventional and plasmacytoid DCs. (E) Number of DCs in the bone marrow. (F) Serum Flt3L levels in 4-wk-old mice. (G) DCs were incubated with OVA323–339 peptide for 1 h then incubated with CFSE-labeled CD4+ OT-II T cells for 3 d. T cell proliferation was measured by flow cytometry. The percentage of proliferating CD4 T cells is shown from five independent samples. Error bars, mean ± SEM. Unpaired two-tailed Student t test (B and D–G). *p < 0.05.

FIGURE 1.

RIPK1-deficient DCs are more sensitive to necroptosis. (A) Western blot depicting RIPK1 expression in CD11c+-enriched splenic DCs from CD11cCre and Ripk1DC KO mice. (B) RIPK1-deficient DCs are more susceptible to necroptosis. BMDCs were treated with 0.1 μM SMAC mimetics and 10 μM zVAD-fmk. Effects on cell viability were measured at 6 and 18 h using Cell Titer Glo. n = 6–8 independent samples. (C) Western blot depicting MLKL phosphorylation and total MLKL, RIPK1, and RIPK3 levels in BMDCs treated with SMAC mimetics and zVAD-fmk. Quantification of pMLKL relative to MLKL from three Western blots is shown. (D) Number of splenic conventional and plasmacytoid DCs. (E) Number of DCs in the bone marrow. (F) Serum Flt3L levels in 4-wk-old mice. (G) DCs were incubated with OVA323–339 peptide for 1 h then incubated with CFSE-labeled CD4+ OT-II T cells for 3 d. T cell proliferation was measured by flow cytometry. The percentage of proliferating CD4 T cells is shown from five independent samples. Error bars, mean ± SEM. Unpaired two-tailed Student t test (B and D–G). *p < 0.05.

Close modal

To determine whether RIPK1 deletion altered DC survival, BMDCs were treated in vitro with necroptotic and apoptotic stimuli. Treatment with zVAD-fmk and SMAC mimetics primes cells for necroptosis by inhibiting caspase activation and inducing IAP degradation, respectively. Consistent with its negative regulatory roles in hematopoietic cells (11, 15), RIPK1-deficient DCs are more sensitive than controls to necroptotic death (Fig. 1B). In contrast, RIPK1 kinase inactive (Ripk1D138N) DCs remained resistant to zVAD and SMAC treatment, revealing a requirement for the kinase activity of RIPK1 in TNF-mediated necroptosis in DCs.

Necroptosis is thought to be mediated by a serial phosphorylation cascade whereby RIPK1 phosphorylates RIPK3, which then phosphorylates the effector pseudo-kinase MLKL (24). To confirm that necroptosis was induced in RIPK1-deficient DCs, we examined DC cultures for evidence of MLKL phosphorylation. MLKL phosphorylation was detected in RIPK1-deficient DCs treated with zVAD-fmk and SMAC mimetics at early time points, indicating that a RIPK1 deficiency predisposes DCs to necroptotic death (Fig. 1C). RIPK3 overexpression has been shown to bypass the requirement for RIPK1 to drive necroptotic death (28); however, no differences in RIPK3 or MLKL expression were observed in RIPK1-deficient DCs (Fig. 1C). In fact, we demonstrate that increasing concentrations of zVAD-fmk, in the absence of SMAC mimetics or exogenous death ligands, was sufficient to induce necroptosis in RIPK1-deficient DCs (Supplemental Fig. 1A). DCs deficient for both RIPK1 and RIPK3 were not susceptible to zVAD-fmk–induced cell death, confirming that caspase inhibition alone was sufficient to trigger necroptotic death in RIPK1-deficient DCs (Supplemental Fig. 1A). In contrast, RIPK1-deficient BMDCs responded normally to apoptotic stimuli including exposure to FasL, IFN-γ, or TNF-α with cycloheximide (Supplemental Fig. 1B, 1C).

The fact that necroptosis was induced by treatment with zVAD-fmk and that SMAC mimetics were not required suggested that IAP/XIAP expression may be reduced in RIPK1-deficient DCs. Reduced cIAP1/2 and XIAP expression has been shown previously to sensitize Ag-presenting cells to necroptosis (2931). We found cIAP2 and XIAP expression reduced in the absence of RIPK1; however, no significant changes in cIAP1 levels were observed (Supplemental Fig. 1D). Gene expression analysis revealed no differences in cIAP1/2 or XIAP mRNA levels in Ripk1DC KO DCs (Supplemental Fig. 1E). Therefore, these data suggest that a RIPK1 deficiency may lead to cIAP2/XIAP protein degradation and predispose DCs to necroptosis.

Because RIPK1-deficient DCs exhibited increased susceptibility to necroptosis in vitro (Fig. 1B), decreased numbers of DCs were expected in Ripk1DC KO mice. However, the loss of RIPK1 did not lead to alterations in the number of splenic conventional DCs (cDCs; CD11c+ MHC class II+) or plasmacytoid DCs (pDCs; CD11clo MHC class II+ Siglec H+) (Fig. 1D), even at 1 y of age (Supplemental Fig. 1F). Furthermore, no significant differences in the expression of activation markers CD80, CD86, or MHC class II were observed in cDCs or pDCs from Ripk1DC KO mice compared with CD11cCre controls (Supplemental Fig. 1G, 1H). However, significant increases in bone marrow cDCs were observed (Fig. 1E) as well as increases in serum Flt3L levels (Fig. 1F), suggesting that DC progenitor activity may be stimulated in response to DC loss in vivo (32). The loss of RIPK1 had no detectable effects on BMDC production, as normal numbers of BMDC were generated from the bone marrow in vitro, indicating that RIPK1 is not required for DC proliferation and/or differentiation (Supplemental Fig. 1I).

We treated RIPK1-deficient and control DCs with TNF to examine NF-κB activation. Although not statistically significant, the IκBα phosphorylation in response to TNF was consistently less robust in RIPK1-deficient DCs than controls (Supplemental Fig. 1J). Similarly, IκBα expression was slightly dampened in TNF-treated RIPK1-deficient DCs (Supplemental Fig. 1K). We then examined the ability of RIPK1-deficient DCs to induce CD4 T cell proliferation. We found that RIPK1-deficient DCs induced proliferation of CD4 T cells, although slightly fewer T cells proliferated in response to Ag-stimulated RIPK1-deficient DCs (Fig. 1G). These data demonstrate that RIPK1-deficient DCs are able to respond to TNF and to Ag stimulation.

A caspase-8 deficiency has been shown to enhance the assembly and activation of the NLRP3 inflammasome, a signaling complex that mediates the processing of proinflammatory cytokines such as IL-1β (33). IL-1β secretion generally requires exposure to two stimuli: first, a priming agent such as LPS, which triggers pro–IL-1β synthesis; and a second signal such as ATP, which processes pro–IL-1β to its active form (34). RIPK1 deletion in macrophages, as well cIAP, XIAP, or caspase-8 depletion in DCs, has been shown to stimulate IL-1β production in response to LPS without the need for a second signal (11, 31, 35). We therefore examined IL-1β production in RIPK1-deficient DCs stimulated with LPS only or in combination with ATP and found that IL-1β processing was not significantly altered in LPS-primed RIPK1-deficient DCs in vitro (Supplemental Fig. 1L).

Given the increased sensitivity of RIPK1-deficient DCs to necroptosis in vitro, we examined the in vivo effects of RIPK1 deficiency in DCs by examining Ripk1DC KO mice. Ripk1DC KO mice developed splenomegaly and lymphadenopathy by 16 wk of age (Fig. 2A–E). Evidence of inflammation was observed in Ripk1DC KO mice, with elevated numbers of neutrophils and inflammatory monocytes in the spleens of Ripk1DC KO mice (Fig. 2C). Conversely, no differences were detected in splenic B or T lymphocyte populations (Supplemental Fig. 2B, 2C). Histological analysis revealed that the splenic architecture was disrupted in Ripk1DC KO mice (Fig. 2D). Masson’s trichrome staining revealed collagen deposits in the spleen indicating fibrosis, which largely accounted for the increased spleen size. Masson’s trichrome staining also revealed evidence of fibrosis in skin and lungs of Ripk1DCKO mice but not controls (Supplemental Fig. 2A).

FIGURE 2.

Ripk1DC KO mice develop systemic inflammation and autoimmunity. (A) Representative images of spleens and lymph nodes from Ripk1DC KO mice. (B) Spleen weight and cellularity. (C) Number of inflammatory monocytes and neutrophils in the spleen. (D) H&E and Masson’s trichrome staining on spleen sections from age-matched CD11cCre and Ripk1DC KO mice. (E) Lymph node (LN) weight, cellularity, with numbers of DCs, inflammatory monocytes, neutrophils, and B cells in the lymph nodes. (F) Number of bone marrow erythroblasts. (G) Number of erythroblasts in the spleen. (H) Number of erythrocytes in the spleen. (I) Number of lineagec-Kit+ progenitors in the spleen. (J) Serum cytokine and chemokine levels in CD11cCre and Ripk1DC KO mice (n = 5 per genotype). Data shown from (A)–(J) are from 16-wk-old mice. (K) Survival curve of CD11cCre Ripk1fl/+ and Ripk1DC KO mice i.p. injected with 5 mg/kg LPS. (L) Serum cytokine and chemokine levels in CD11cCre and Ripk1DC KO mice before and 7 h after i.p. injection with 5 mg/kg LPS. (M) Representative images of HEp-2 cells stained with serum from 6-mo-old mice of the indicated genotypes. (N) Proportion of CD11cCre and Ripk1DC KO mice serum positive for cytoplasmic or nucleolar ANAs. n = 3–6 per genotype. Error bars, mean ± SEM. Scale bar, 100 μm. Unpaired two-tailed Student t test (B, C, E–J, and L), and log-rank test (K). *p < 0.05, **p < 0.01.

FIGURE 2.

Ripk1DC KO mice develop systemic inflammation and autoimmunity. (A) Representative images of spleens and lymph nodes from Ripk1DC KO mice. (B) Spleen weight and cellularity. (C) Number of inflammatory monocytes and neutrophils in the spleen. (D) H&E and Masson’s trichrome staining on spleen sections from age-matched CD11cCre and Ripk1DC KO mice. (E) Lymph node (LN) weight, cellularity, with numbers of DCs, inflammatory monocytes, neutrophils, and B cells in the lymph nodes. (F) Number of bone marrow erythroblasts. (G) Number of erythroblasts in the spleen. (H) Number of erythrocytes in the spleen. (I) Number of lineagec-Kit+ progenitors in the spleen. (J) Serum cytokine and chemokine levels in CD11cCre and Ripk1DC KO mice (n = 5 per genotype). Data shown from (A)–(J) are from 16-wk-old mice. (K) Survival curve of CD11cCre Ripk1fl/+ and Ripk1DC KO mice i.p. injected with 5 mg/kg LPS. (L) Serum cytokine and chemokine levels in CD11cCre and Ripk1DC KO mice before and 7 h after i.p. injection with 5 mg/kg LPS. (M) Representative images of HEp-2 cells stained with serum from 6-mo-old mice of the indicated genotypes. (N) Proportion of CD11cCre and Ripk1DC KO mice serum positive for cytoplasmic or nucleolar ANAs. n = 3–6 per genotype. Error bars, mean ± SEM. Scale bar, 100 μm. Unpaired two-tailed Student t test (B, C, E–J, and L), and log-rank test (K). *p < 0.05, **p < 0.01.

Close modal

In contrast to the spleen, the lymph nodes exhibited increased cellularity, largely consisting of B cells, but also with increases in DCs, neutrophils, and inflammatory monocytes (Fig. 2E). No increases in lymph node T cells were observed, nor was there evidence of T cell activation in spleen or lymph nodes (Supplemental Fig. 2B, 2C).

The bone marrow appeared largely normal (Supplemental Fig. 2D); however, Ripk1DC KO mice had significantly decreased numbers of erythroblasts (Fig. 2F). Conversely, increased numbers of erythroblasts and lineage-negative, c-Kit+ progenitor cells were observed in the spleen (Fig. 2G, 2I) indicative of extramedullary hematopoiesis, potentially a result of the inflammation. Importantly, splenic erythrocyte numbers remained normal, indicating that the erythrocyte population did not contribute to spleen enlargement (Fig. 2H).

To further assess the inflammation in Ripk1DC KO mice, we measured the levels of proinflammatory cytokines in the serum. A significant increase in serum TNFα and IFNγ was observed in Ripk1DC KO mice compared with control mice (Fig. 2J, 2L). Although not significant, increases in serum IL-1β, IL-6, IL-12, KC, and MCP-1 were also observed in Ripk1DC KO mice. To determine the consequences of inflammation and elevated cytokine levels, control or Ripk1DC KO mice were given a low dose of LPS (5 mg/kg). Sixty percent of control mice succumbed to LPS administration with a median latency of 70 h, whereas all Ripk1DC KO mice succumbed to LPS-induced endotoxic shock within 30 h (Fig. 2K). Additionally, the levels of proinflammatory cytokines TNF-α, IL-1β, and IFN-γ were significantly elevated in the sera of Ripk1DC KO mice compared with controls (Fig. 2L). Thus, the proinflammatory cell death in Ripk1DC KO mice sensitizes the mice to LPS-induced endotoxic shock. Interestingly, DC loss is a clinical feature of septic patients, suggesting that DC necroptosis may underlie the hyperinflammatory syndrome and immune suppression in patients with severe sepsis (36).

The lymphadenopathy and splenomegaly observed in Ripk1DC KO mice suggested that these mice developed autoimmune disease. The B220+, CD3+, CD4, and CD8 T cells prevalent in autoimmune mouse models such as the Faslpr and Caspase-8−/− Ripk3−/− mice (37, 38) were not present in Ripk1DC KO mice (Supplemental Fig. 2E). However, ANAs were detected in the serum of Ripk1DC KO mice from 6 mo of age (Fig. 2M). To characterize the autoantibodies produced by these mice, we screened sera by immunofluorescent staining of HEp-2 cells. The staining intensity was similar to that observed with serum from autoimmune prone Faslpr mice. Abs reactive with RNA-associated autoantigens frequently exhibit a more speckled nuclear or cytoplasmic staining pattern, whereas Abs reactive with dsDNA or other chromatin components exhibit a homogenous nuclear stain as observed with sera from Fas-deficient mice. The staining pattern of the circulating ANAs in Ripk1DC KO mice was consistent with Abs targeting nucleolar components, with one of six mice examined exhibiting a cytoplasmic staining pattern (Fig. 2M, 2N). Antinucleolar and other speckled nuclear patterns are commonly found in TLR9-deficient or TLR7-overexpressing lupus-prone mice (39, 40). These data indicate that autoantibodies produced by Ripk1DC KO mice recognize RNA-associated autoantigens and not dsDNA.

Correlating with the presence of ANAs, we observed increased numbers of germinal center B cells in the lymph nodes of these mice (Supplemental Fig. 2F). Although ANAs were detected in Ripk1DC KO mice, the CD11c promoter can be active in some lymphoid cells (17), therefore we mated the Ripk1DC KO and CD11cCre mice with a Rosa26-loxP-Stop-loxP-tdTomato reporter line and assessed Tomato red expression in hematopoietic cells from spleen and lymph nodes (Supplemental Fig. 2G). Tomato red expression was detected in 86% of DCs and 26% of B cells isolated from spleen and lymph nodes of Ripk1DC KO but not control mice. To assess Ripk1 deletion in B cells, we sorted Tomato red–positive and –negative B cells from control and Ripk1DC KO mice and performed genomic PCR. We detected evidence of Ripk1 deletion in Tomato red–positive but not –negative B cells (Supplemental Fig. 2I). Notably, B cells isolated from mice with lymphadenopathy retained a floxed Ripk1 allele, indicating that these B cells likely expressed RIPK1. Based on this analysis and the fact that germinal center B cells expanded in the lymph node, we conclude that Ripk1 deletion in the CD11c-positive, age-associated B cell population associated with young lupus-prone or aged wild-type mice is not responsible for B cell expansion and autoantibody production in these mice (41). As is typical for mice on the C57BL/6 background (42), Ripk1DCKO mice did not develop proteinuria by 12 mo of age (Supplemental Fig. 2J). Together, these data suggest that a RIPK1 deficiency in DCs leads to systemic inflammation that triggers B cell expansion and autoantibody production.

Given the in vitro sensitivity of RIPK1-deficient DCs to necroptosis (Fig. 1B) and the lack of reliable and sensitive measures to detect necroptotic death in mice, we used a genetic approach to determine whether aberrant DC necroptosis is responsible for the inflammation and autoimmunity in Ripk1DC KO mice. To address the specific role of RIPK1 kinase-dependent cell death, Ripk1DC KO mice were crossed to mice expressing the kinase inactive RIPK1 (D138N) allele (8) to generate CD11cCre Ripk1fl/D138N (Ripk1DC KO/D138N) mice. Because the kinase activity of RIPK1 is not required for necroptosis in certain cell types (43, 44), we also generated Ripk1DC KO Ripk3−/− and Ripk1DC KO Mlkl−/− mice to provide additional genetic evidence that DC necroptosis results in chronic inflammation and a break in tolerance.

Introduction of the Ripk1D138N allele or deletion of Ripk3 protected RIPK1-deficient DCs from necroptosis in vitro (Fig. 3A). Similarly, the introduction of the Ripk1D138N allele or deletion of Ripk3 or Mlkl ameliorated the inflammatory and autoimmune disease associated with a DC RIPK1 deficiency, resulting in reductions in spleen and lymph node weight (Fig. 3B). Significant decreases in neutrophil and inflammatory monocyte infiltration were also observed in the spleens of these mice indicating that inflammation was reduced (Fig. 3C). Importantly, the splenic architecture was maintained in these mice with no detectable signs of fibrosis (Fig. 3D, Supplemental Fig. 3A). Interestingly, some differences were observed in Ripk1DC KO/D138N, Ripk1DC KO Ripk3−/−, and Ripk1DC KO Mlkl−/− mice. For example, although Ripk1DC KO/D138N and Ripk1DC KO Mlkl−/− mice showed a complete rescue of the lymphadenopathy, Ripk1DC KO Ripk3−/− mice displayed only a partial reduction in lymph node weight and cellularity, with increased numbers of inflammatory monocytes, neutrophils, and B cells in the lymph nodes (Fig. 3E). Collectively, these data demonstrate that chronic necroptosis drives inflammation in Ripk1DC KO mice. However, these data are also consistent with published work that reveals necroptosis-independent functions of RIPK3 (33, 35, 45).

FIGURE 3.

DC necroptosis underlies the systemic inflammation in Ripk1DC KO mice. (A) DCs from CD11cCre Ripk1fl/D138N (Ripk1DC KO/D138N) and Ripk1DC KO Ripk3−/− mice are protected from necroptosis in vitro. BMDCs were treated with SMAC mimetics and zVAD-fmk. Effects on cell viability were measured at 18 h using Cell Titer Glo. n = 3–6 samples. (B) Spleen and lymph node (LN) weights and lymph node cellularity from CD11cCre, Ripk1DC KO, Ripk1DC KO/D138N, Ripk1DC KO Ripk3−/−, and Ripk1DC KO Mlkl−/− mice. (C) Neutrophil and inflammatory monocyte numbers in the spleens of CD11cCre, Ripk1DC KO, Ripk1DC KO/D138N, Ripk1DC KO Ripk3−/−, and Ripk1DC KO Mlkl−/− mice. (D) H&E and Masson’s trichrome staining of spleens from Ripk1DC KO, Ripk1DC KO/D138N, Ripk1DC KO Ripk3−/−, and Ripk1DC KO Mlkl−/− mice. (E) Neutrophils, inflammatory monocytes, and B cells are elevated in the lymph nodes of Ripk1DC KO and Ripk1DC KO Ripk3−/− mice but not Ripk1DC KO/D138N or Ripk1DC KOMlkl−/− mice. (F) Proportion of mice serum positive for ANAs at 6 mo (n = 3–6 mice per genotype). Unless otherwise stated, all data shown are from 16-wk-old mice. CD11cCre and Ripk1DC KO mouse phenotyping data shown in (B), (C), (E), and (F) were shown in Fig. 2. Error bars, mean ± SEM. Scale bars, 100 μm. Unpaired two-tailed Student t test (A–C and E). *p < 0.05, **p < 0.01.

FIGURE 3.

DC necroptosis underlies the systemic inflammation in Ripk1DC KO mice. (A) DCs from CD11cCre Ripk1fl/D138N (Ripk1DC KO/D138N) and Ripk1DC KO Ripk3−/− mice are protected from necroptosis in vitro. BMDCs were treated with SMAC mimetics and zVAD-fmk. Effects on cell viability were measured at 18 h using Cell Titer Glo. n = 3–6 samples. (B) Spleen and lymph node (LN) weights and lymph node cellularity from CD11cCre, Ripk1DC KO, Ripk1DC KO/D138N, Ripk1DC KO Ripk3−/−, and Ripk1DC KO Mlkl−/− mice. (C) Neutrophil and inflammatory monocyte numbers in the spleens of CD11cCre, Ripk1DC KO, Ripk1DC KO/D138N, Ripk1DC KO Ripk3−/−, and Ripk1DC KO Mlkl−/− mice. (D) H&E and Masson’s trichrome staining of spleens from Ripk1DC KO, Ripk1DC KO/D138N, Ripk1DC KO Ripk3−/−, and Ripk1DC KO Mlkl−/− mice. (E) Neutrophils, inflammatory monocytes, and B cells are elevated in the lymph nodes of Ripk1DC KO and Ripk1DC KO Ripk3−/− mice but not Ripk1DC KO/D138N or Ripk1DC KOMlkl−/− mice. (F) Proportion of mice serum positive for ANAs at 6 mo (n = 3–6 mice per genotype). Unless otherwise stated, all data shown are from 16-wk-old mice. CD11cCre and Ripk1DC KO mouse phenotyping data shown in (B), (C), (E), and (F) were shown in Fig. 2. Error bars, mean ± SEM. Scale bars, 100 μm. Unpaired two-tailed Student t test (A–C and E). *p < 0.05, **p < 0.01.

Close modal

We predicted that the release of DAMPs from necroptotic DCs drive the autoimmunity observed in Ripk1DC KO mice. Therefore, we examined autoantibody production in Ripk1DC KO/D138N, Ripk1DC KO Ripk3−/−, and Ripk1DC KO Mlkl−/− mice. We were unable to detect ANAs in the serum of these mice (Fig. 3F, Supplemental Fig. 3B), suggesting that necroptotic DCs release DAMPs including RNA and other nucleolar components that trigger B cell proliferation, differentiation, and autoantibody production. The ability of a RIPK1 kinase inactive D138N allele, an MLKL deficiency and to some extent a RIPK3 deficiency, to rescue disease in Ripk1DC KO mice provides genetic evidence that DC necroptosis underlies the inflammation and autoimmunity.

TNF-α and IFN-γ are known to induce necroptosis (1) and elevated levels of TNF-α and IFN-γ were observed in the serum of Ripk1DC KO mice (Fig. 2J), implicating TNF-α and/or IFN-γ as inducers of DC necroptosis in Ripk1DC KO mice. The increased sensitivity of RIPK1-deficient BMDCs to SMAC- and zVAD-induced necroptosis in vitro was lost upon compound deletion of Tnfr1, but not Ifngr1 (Fig. 4A), further implicating TNF-α as the DC necroptosis-inducing ligand in Ripk1DC KO mice.

FIGURE 4.

Ripk1DC KO mice develop inflammation that is partially TNF dependent. (A) BMDCs from Ripk1DC KO Tnfr1−/− but not Ripk1DC KO Ifngr−/− mice are protected from necroptosis in vitro. BMDCs were treated with SMAC mimetics and zVAD-fmk. Death measured at 20 h by Cell Titer Glo. n = 3. (B) Spleen and lymph node (LN) weights and lymph node cellularity of mice of the indicated genotypes. (C) Neutrophils and inflammatory monocyte numbers in the spleens of mice with the indicated genotypes. (D) H&E and Masson’s trichrome staining of spleen sections from Ripk1DC KO Tnfr1−/−, Ripk1DC KO Ifngr−/−, and control mice. (E) Numbers of neutrophils, inflammatory monocytes, and B cells in the lymph nodes of mice of the indicated genotypes. (F) Proportion of mice serum positive for ANAs at 6 mo (n = 3–6 mice per genotype). CD11cCre and Ripk1DC KO mouse phenotyping data used in (B), (C), (E), and (F) were shown in Fig. 2. Error bars, mean ± SEM. Scale bar, 100 μm. Unpaired two-tailed Student t test (A–C and E). *p < 0.05, **p < 0.01.

FIGURE 4.

Ripk1DC KO mice develop inflammation that is partially TNF dependent. (A) BMDCs from Ripk1DC KO Tnfr1−/− but not Ripk1DC KO Ifngr−/− mice are protected from necroptosis in vitro. BMDCs were treated with SMAC mimetics and zVAD-fmk. Death measured at 20 h by Cell Titer Glo. n = 3. (B) Spleen and lymph node (LN) weights and lymph node cellularity of mice of the indicated genotypes. (C) Neutrophils and inflammatory monocyte numbers in the spleens of mice with the indicated genotypes. (D) H&E and Masson’s trichrome staining of spleen sections from Ripk1DC KO Tnfr1−/−, Ripk1DC KO Ifngr−/−, and control mice. (E) Numbers of neutrophils, inflammatory monocytes, and B cells in the lymph nodes of mice of the indicated genotypes. (F) Proportion of mice serum positive for ANAs at 6 mo (n = 3–6 mice per genotype). CD11cCre and Ripk1DC KO mouse phenotyping data used in (B), (C), (E), and (F) were shown in Fig. 2. Error bars, mean ± SEM. Scale bar, 100 μm. Unpaired two-tailed Student t test (A–C and E). *p < 0.05, **p < 0.01.

Close modal

To address the roles of TNF-α and IFN-γ in inflammation and autoimmunity, we generated Ripk1DC KO mice that lack either TNF-R1 or IFN-γR1. Surprisingly, the absence of TNF-α or IFN-γ signaling had little effect on spleen inflammation, with both Ripk1DC KO Tnfr1−/− and Ripk1DC KO Ifngr1−/− mice exhibiting splenic fibrosis and significant increases in inflammatory cell infiltration (Fig. 4B–D). Consistent with the accelerated TNF-mediated necroptosis observed in vitro (Fig. 4A), we found the lymphadenopathy significantly ameliorated in Ripk1DC KO Tnfr1−/− mice but not in Ripk1DC KO Ifngr1−/− mice (Fig. 4B, 4C). The decrease in lymph node size observed in Ripk1DC KO Tnfr1−/− mice appeared due to decreases in B cell number (Fig. 4E; p = 0.075), indicating that TNF-R1 signaling is required for optimal germinal center B cell expansion in lymph nodes. Serum ANAs, however, were not detected in either mouse strain, indicating that loss of TNF-α or IFN-γ signaling is sufficient to prevent ANA development (Fig. 4F, Supplemental Fig. 3C).

Necroptotic death is predicted to result in the release of cytokines, chemokines, and DAMPs that activate TLRs and NOD-like receptors to promote inflammation (5). We examined the contribution of MyD88, a key component of TLR signaling (46), to the inflammatory disease observed in Ripk1DC KO mice. At 8 wk of age Ripk1DC KO Myd88−/− mice exhibited significant decreases in spleen and lymph node size compared with Ripk1DC KO mice, together with decreases in neutrophil and inflammatory monocyte infiltrates in the spleen and lymph nodes (Fig. 5A–C). In addition, the number of B cells in the lymph nodes was comparable to controls. These data strongly suggest that the inflammation observed in Ripk1DC KO mice is exacerbated by MyD88-dependent TLR signaling, which is likely acting to amplify inflammation in response to the release of DAMPs from necroptotic RIPK1-deficient DCs. Notably, a TLR-MyD88 pathway also drives the B cell expansion observed in the lymph nodes of Ripk1DC KO mice.

FIGURE 5.

MyD88 is required for the development of inflammation in Ripk1DC KO mice. (A) Spleen and lymph node (LN) weights and lymph node cellularity of Ripk1DC KO and Ripk1DC KO Myd88−/− mice at 8 wk of age compared with CD11cCre controls. (B) Neutrophils and inflammatory monocytes are elevated in the spleens of Ripk1DC KO and Ripk1DC KO Myd88−/− mice compared with CD11cCre controls. (C) Neutrophils, inflammatory monocytes, and B cells are increased in the lymph nodes of Ripk1DC KO and Ripk1DC KO Myd88−/− mice compared with CD11cCre controls. (D) Depletion of microbiota does not alter the inflammation of Ripk1DC KO mice. Spleen and lymph node weights and lymph node cellularity of Ripk1DC KO mice treated with broad-spectrum antibiotics in the drinking water for 12 wk. (E) Neutrophils, inflammatory monocytes, and B cells remain elevated in the lymph nodes of antibiotic-treated Ripk1DC KO mice. (F) Neutrophils and inflammatory monocytes remain elevated in the spleens of Ripk1DC KO mice treated with antibiotics. (G) Survival curve of mice of the indicated genotypes with or without treatment with broad-spectrum antibiotics in the drinking water. n = 3–6 per group. Error bars, mean ± SEM. Unpaired two-tailed Student t test (A–F), log-rank test (G). *p < 0.05, **p < 0.01.

FIGURE 5.

MyD88 is required for the development of inflammation in Ripk1DC KO mice. (A) Spleen and lymph node (LN) weights and lymph node cellularity of Ripk1DC KO and Ripk1DC KO Myd88−/− mice at 8 wk of age compared with CD11cCre controls. (B) Neutrophils and inflammatory monocytes are elevated in the spleens of Ripk1DC KO and Ripk1DC KO Myd88−/− mice compared with CD11cCre controls. (C) Neutrophils, inflammatory monocytes, and B cells are increased in the lymph nodes of Ripk1DC KO and Ripk1DC KO Myd88−/− mice compared with CD11cCre controls. (D) Depletion of microbiota does not alter the inflammation of Ripk1DC KO mice. Spleen and lymph node weights and lymph node cellularity of Ripk1DC KO mice treated with broad-spectrum antibiotics in the drinking water for 12 wk. (E) Neutrophils, inflammatory monocytes, and B cells remain elevated in the lymph nodes of antibiotic-treated Ripk1DC KO mice. (F) Neutrophils and inflammatory monocytes remain elevated in the spleens of Ripk1DC KO mice treated with antibiotics. (G) Survival curve of mice of the indicated genotypes with or without treatment with broad-spectrum antibiotics in the drinking water. n = 3–6 per group. Error bars, mean ± SEM. Unpaired two-tailed Student t test (A–F), log-rank test (G). *p < 0.05, **p < 0.01.

Close modal

The MyD88 dependency raised the possibility that pathogenic or commensal bacteria trigger and/or exacerbate inflammation in mice with DC RIPK1 deficiency. To examine this, we treated Ripk1DC KO mice with broad-spectrum antibiotics for 12 wk. We found that antibiotic treatment had no detectable effect on the development of inflammatory disease in Ripk1DC KO mice (Fig. 5D–F). This suggests that the MyD88-dependent inflammation in Ripk1DC KO mice is independent of pathogenic or commensal bacteria and likely a consequence of endogenous DAMPs.

The Ripk1DC KO Myd88−/− mice began to lose weight and became moribund, succumbing with an average latency of 11.5 wk (Fig. 5G). Treatment with broad-spectrum antibiotics significantly prolonged the survival of Ripk1DC KO Myd88−/− mice (Fig. 5G). The heightened susceptibility of Ripk1DC KO Myd88−/− mice to bacterial infection suggests that a combined total MyD88 deficiency with a DC RIPK1 deficiency interfered with the animals’ innate and adaptive antibacterial immune responses.

Given the recent data implicating type I IFNs in necroptosis and known roles for type I IFNs in ANA production and autoimmunity (47, 48), we hypothesized that type I IFN may contribute to autoinflammation and autoimmunity observed in Ripk1DC KO mice. Although we were unable to detect IFN-α or IFN-β in the serum of Ripk1DC KO mice (Supplemental Table I), we tested this hypothesis genetically by generating Ripk1DC KO Ifnar1−/− mice. The absence of the type I IFNR had no effect on the splenomegaly or lymphadenopathy associated with a DC RIPK1 deficiency, ruling out a critical role for type I IFN-induced necroptosis in the organ inflammation (Fig. 6). Although inflammation remained unaffected, an absence of type I IFN signaling prevented ANA development in five of six Ripk1DC KO Ifnar1−/− mice examined (Fig. 6E, Supplemental Fig. 3D). One Ripk1DC KO Ifnar1−/− mouse exhibited weak ANA staining (Supplemental Fig. 3D). Surprisingly, B cell numbers remained increased in the lymph nodes of Ripk1DC KO Ifnar1−/− mice. Overall, these data reveal that autoimmunity but not inflammation is largely driven by or dependent on type I IFN signaling. These data provide genetic evidence that inflammation precedes autoimmunity, and suggests that type I IFNs mediate B cell differentiation into Ab-secreting plasma cells.

FIGURE 6.

Autoimmunity, but not inflammation, in Ripk1DC KO mice is type I IFN dependent. (A) Spleen and lymph node (LN) weights and lymph node cellularity from mice of the indicated genotypes. (B) Number of splenic neutrophils and inflammatory monocytes from mice of the indicated genotypes. (C) H&E and Masson’s trichrome staining on spleen sections from mice of the indicated genotypes. (D) Number of neutrophils, inflammatory monocytes, and B cells in the lymph nodes from mice of the indicated genotypes. (E) Proportion of mice that are serum positive for cytoplasmic or nucleolar ANAs. n = 3–6 mice per genotype. All data shown are from 6-mo-old mice. Error bars, mean ± SEM. Scale bar, 100 μm. Unpaired two-tailed Student t test (A, B, and D). *p < 0.05, **p < 0.01.

FIGURE 6.

Autoimmunity, but not inflammation, in Ripk1DC KO mice is type I IFN dependent. (A) Spleen and lymph node (LN) weights and lymph node cellularity from mice of the indicated genotypes. (B) Number of splenic neutrophils and inflammatory monocytes from mice of the indicated genotypes. (C) H&E and Masson’s trichrome staining on spleen sections from mice of the indicated genotypes. (D) Number of neutrophils, inflammatory monocytes, and B cells in the lymph nodes from mice of the indicated genotypes. (E) Proportion of mice that are serum positive for cytoplasmic or nucleolar ANAs. n = 3–6 mice per genotype. All data shown are from 6-mo-old mice. Error bars, mean ± SEM. Scale bar, 100 μm. Unpaired two-tailed Student t test (A, B, and D). *p < 0.05, **p < 0.01.

Close modal

Given that the ANAs generated in Ripk1DC KO mice largely target the nucleolar components of the cell (Fig. 2N), we examined the role of the nucleic acid sensing TLRs as drivers of the autoimmune disease. We used the Unc93b13d/3d mouse, which lacks functional UNC93B1 required for the intracellular trafficking of TLRs 3, 7, and 9 from the endoplasmic reticulum to the endosome (22). These TLRs sense nucleolar components such as ssRNA, dsRNA, and unmethylated DNA and TLR 7 is required for the development of autoimmunity in lupus-prone mouse strains (22, 49, 50). An UNC93B1 deficiency had no detectable effects on splenomegaly, Masson’s trichrome staining, or on splenic inflammatory cell infiltrates, indicating that the nucleic acid TLRs do not mediate the inflammation and tissue fibrosis (Fig. 7). The size and cellularity of the lymph nodes, however, were significantly reduced in Ripk1DC KO Unc93b13d/3d mice (Fig. 7A). Importantly, the UNC93B1-dependent reduction in lymph node size correlated with significant reductions in B cell number, whereas neutrophils and inflammatory monocytes were not significantly altered (Fig. 7D). No ANAs were detected in serum from Ripk1DC KO Unc93b13d/3d mice (Fig. 7E, Supplemental Fig. 3E), revealing that nucleic acid sensing TLRs are required for the generation of autoantibodies.

FIGURE 7.

TLR 3, 7 and 9 signaling drives B cell expansion and autoimmunity in Ripk1DC KO. (A) Spleen and lymph node (LN) weights from mice of the indicated genotypes. (B) Splenic neutrophils and inflammatory monocyte numbers from mice of the indicated genotypes. (C) H&E and Masson’s trichrome staining on spleen sections from mice of the indicated genotypes. (D) Number of neutrophils, inflammatory monocytes and B cells in the lymph nodes from mice of the indicated genotypes. (E) Proportion of mice that are serum positive for cytoplasmic or nucleolar ANAs at 6 mo. n = 3–6 per genotype. All data shown are from 6-mo-old mice. CD11cCre and Ripk1DC KO mouse phenotyping data shown in Fig. 7 were shown in Fig. 6. Error bars, mean ± SEM. Scale bar, 100 μm. Unpaired two-tailed Student t test (A, B, and D). *p < 0.05, **p < 0.01.

FIGURE 7.

TLR 3, 7 and 9 signaling drives B cell expansion and autoimmunity in Ripk1DC KO. (A) Spleen and lymph node (LN) weights from mice of the indicated genotypes. (B) Splenic neutrophils and inflammatory monocyte numbers from mice of the indicated genotypes. (C) H&E and Masson’s trichrome staining on spleen sections from mice of the indicated genotypes. (D) Number of neutrophils, inflammatory monocytes and B cells in the lymph nodes from mice of the indicated genotypes. (E) Proportion of mice that are serum positive for cytoplasmic or nucleolar ANAs at 6 mo. n = 3–6 per genotype. All data shown are from 6-mo-old mice. CD11cCre and Ripk1DC KO mouse phenotyping data shown in Fig. 7 were shown in Fig. 6. Error bars, mean ± SEM. Scale bar, 100 μm. Unpaired two-tailed Student t test (A, B, and D). *p < 0.05, **p < 0.01.

Close modal

These genetic data reveal that the inflammation, tissue fibrosis, and autoimmunity is dependent on the presence of RIPK1 kinase activity, RIPK3, MLKL, and MyD88 (Figs. 3, 5, 6). Necroptotic cell death and MyD88-dependent TLRs mediate the inflammatory disease; however, the nucleic acid sensing TLRs do not contribute to tissue fibrosis or inflammation. Although type I and II IFNs induce necroptosis in vitro and are thought to underlie TNF-induced necroptosis in vivo (12, 51), neither IFN-γ or IFN-α/β are major drivers of inflammatory disease in this model. By contrast, the B cell expansion is partially TNF-R1–dependent, and requires both MyD88 and UNC93B1. Surprisingly, type I IFN signaling is not required for B cell expansion in the lymph node, but proves essential for the differentiation of these cells into Ab-secreting cells and for ANA production. These in vivo genetic studies delineate the cell death and innate immune pathways as well as the temporal order of events that break tolerance and give rise to autoimmunity. These findings reveal that DC necroptosis is sufficient to give rise to inflammation and autoimmunity.

Herein we demonstrate that the loss of RIPK1 primes DCs for RIPK3- and MLKL-dependent necroptosis leading to inflammation, tissue fibrosis, and autoimmunity. We show that in DCs, RIPK1 negatively regulates necroptosis but has no detectable effects on apoptosis. Genetically blocking necroptosis in DCs prevents the development of inflammation and autoimmunity in Ripk1DC KO mice, revealing a crucial role for RIPK1 in DC survival and maintenance of immune homeostasis.

The proinflammatory cytokine TNF is considered the classical activator of necroptosis. We show that in vitro RIPK1-deficient DCs undergo TNF-mediated necroptosis (Figs. 1B, 4A); however, an absence of TNF-R1 had no detectable effects on splenic inflammation or tissue fibrosis (Fig. 4). These findings were unexpected as pulmonary fibrosis patients display elevated levels of TNF-α and mice that overexpress TNF in the lung develop progressive pulmonary fibrosis (52). Splenic fibrosis was prevented by expression of kinase inactive RIPK1 or an absence of RIPK3 or MLKL (Fig. 3D), implicating necroptosis in the fibrotic response. An absence of TNF-R1 signaling significantly ameliorated the lymphadenopathy in Ripk1DC KO mice (Fig. 4B), potentially due to TNF-R1 effects on follicular DC networks and/or germinal center formation (53, 54).

Because an absence of TNF, IFN-γ, or type I IFN signaling had no effect on inflammation, we speculate that ligand-independent DC necroptosis drives the inflammation observed in Ripk1DC KO mice. A RIPK1 deficiency in DCs stimulates RIPK3-MLKL–dependent necroptosis in vivo; however, no detectable increases in RIPK3 and/or MLKL expression were observed in vitro (Fig. 1C). The necroptotic sensitivity of RIPK1-deficient DCs could be explained by decreased expression of cIAP2 and XIAP (Supplemental Fig. 1D). Consistent with this hypothesis, cIAP1 and cIAP2 depletion in macrophages and monocytes have been show to stimulate necroptosis (29, 30). The IAP proteins regulate RIPK1 polyubiquitination and reductions in polyubiquitinated RIPK1 stimulate the formation of RIPK1-containing death complexes in the cytosol. Thus, a RIPK1 deficiency, which may result in IAP degradation (10, 16, 55), may sensitize DCs to necroptosis by unleashing RIPK3 and by promoting necrosome or complex 2b (RIPK3/MLKL) formation. XIAP loss in DCs also alters RIPK1 polyubiquitination and promotes RIPK3-mediated cell death by as yet unclear mechanisms (31).

The deletion of other death receptor pathway components in DCs such as caspase-8 or Fas-associated death domain protein (termed FADD) reveal similar but distinct disease phenotypes. For example, although the deletion of FADD sensitized the DCs to necroptosis leading to inflammation in vivo, the inflammation was driven by commensal bacteria (56). Yet, depleting commensal bacteria in Ripk1DC KO mice had no effect on disease severity or kinetics (Fig. 5D–F). A caspase-8 DC deficiency also results in chronic inflammation that was rescued upon deletion of RIPK3. However, the effects on inflammatory disease were not attributed to necroptosis, but rather to requirements for caspase-8 and RIPK3 in inflammasome activation and IL-1β production (33, 57). In contrast to caspase-8–deficient DCs, we found RIPK1-deficient DCs are not primed to produce higher levels of IL-1β when stimulated with LPS in vitro (Supplemental Fig. 1L). Collectively these data suggest that commensal bacteria or inflammasome activation are not responsible for disease in Ripk1DC KO mice.

Mice deficient for caspase-8 are protected from embryonic lethality by concomitant loss of RIPK3 or MLKL, revealing that caspase-8 prevents necroptosis during development (38, 58, 59). The Caspase-8−/− Ripk3−/− and Caspase-8−/− Mlkl−/− mice are viable and fertile but develop autoimmune disease due to the expansion of CD3+, B220+, CD4, and CD8 T cells, due to impaired Fas-mediated apoptosis. We have shown that RIPK1 functions in certain cell types to prevent RIPK3 and MLKL activation; however, this unusual T cell population was not detected in autoimmune Ripk1DC KO mice nor did we find any evidence of T cell activation (Supplemental Fig. 2).

Ripk1DC KO mice develop autoimmunity, which is prevented when necroptosis is blocked. We show that autoimmunity is type I IFN dependent and that inhibiting TLR3, 7, and/or 9 signaling largely prevents ANA production. Interestingly, in our model, blocking the nucleic acid sensing TLRs (likely TLR7/9), but not type I IFNs, inhibits B cell expansion in the lymph nodes. The fact that B cell numbers were not altered in Ifnar1−/− mice was unexpected, as TLR7/9 play central roles in autoimmune disease and one of the main downstream effects of TLR7/9 signaling is IFN-α/β production (60). B cells may directly respond to RNA or DNA/immune complexes through the BCR which can directly bind ssRNA or dsDNA and transport it to the endosome to activate TLR7 or TLR9, respectively (61, 62). Collectively, these data indicate that endosomal TLR7 and/or TLR9 cooperate with the BCR to mediate the expansion of autoreactive B cells via a pathway that does not require IFN-α/β.

The frequency of Ripk1DC KO mice that produce ANAs targeting nucleolar components correlate with data showing that anti-RNA autoantibodies are common in systemic lupus erythematosus (SLE) patients (63, 64). Our studies suggest that ANA production in SLE patients may also be a consequence of increased necroptosis, which likely provides a continuous source of RNA autoantigens. Finally, these data demonstrate that chronic necroptosis (in the absence of DC or T cell activation) is sufficient for germinal center B cell expansion and ANA production.

A theme that has emerged from the study of a wide range of autoimmune diseases is the appearance of disease-specific autoantibodies before evidence of clinical disease. In rheumatoid arthritis, Abs reactive with citrullinated histones can be detected years before any evidence of joint inflammation and IFN-γ levels correlate with the appearance of serum autoantibodies years prior to the clinical diagnosis of SLE (6568). These genetic studies reveal that necroptotic cell death triggers inflammation and precedes the development of autoimmunity. Tissue fibrosis observed in the spleen, lungs, and skin of Ripk1DC KO mice was prevented by expression of kinase inactive RIPK1 or an absence of RIPK3 or MLKL, implicating, to our knowledge, for the first time, this form of inflammatory cell death in the fibrotic response. Fibrosis was unaffected by an absence of TNF-R1, type I or II IFN signaling, but appeared rescued by an MyD88 deficiency. Our data also show that necroptosis and inflammation are not sufficient for the development of autoimmunity; ANA production requires the presence of functional nucleic acid–sensing TLRs and the type I IFNR.

Recently, the inflammation associated with a RIPK1 deficiency in keratinocytes has been shown to be dependent on the receptor interacting protein homotypic interacting motif of RIPK1, MLKL, and the putative nucleic acid sensor Z-DNA binding protein 1, ZBP1, also known as DNA-dependent activator of IFN regulatory factors (69, 70). These findings suggest that the ZBP1-RIPK3-MLKL necroptotic pathway may be constitutively activated in Ripk1DC KO mice. Although initially identified as a dsDNA sensor capable of activating the NF-κB and IRF3 transcription factors (71), ZBP1 has recently been shown to recognize RNA viruses and induce necroptosis (72). Interestingly, ZBP1 expression is increased in PBMCs of lupus patients (73) raising the intriguing possibility that ZBP1-mediated necroptosis may contribute to SLE and other autoimmune or fibrotic disorders.

We thank Dr. Ben Croker, Dr. Shruti Sharma, and Dr. Kristen Nundel for invaluable advice and discussion. We thank Dr. Dave Garlick for histopathological analysis. We thank Jason McGowan and Stacy Cote for help with ANA experiments. We acknowledge the support of the University of Massachusetts Flow Cytometry Core and the Mouse Metabolic Phenotyping Center.

This work was supported by National Institutes of Health Grant AI075118 (to M.A.K.).

The online version of this article contains supplemental material.

Abbreviations used in this article:

ANA

anti‐nuclear autoantibody

BMDC

bone marrow–derived DC

DAMP

danger-associated molecular pattern

DC

dendritic cell

MLKL

mixed lineage kinase domain-like

RIPK

receptor interacting protein kinase

SLE

systemic lupus erythematosus; Tnfr1, TNFR type 1.

1
Pasparakis
,
M.
,
P.
Vandenabeele
.
2015
.
Necroptosis and its role in inflammation.
Nature
517
:
311
320
.
2
Sun
,
L.
,
H.
Wang
,
Z.
Wang
,
S.
He
,
S.
Chen
,
D.
Liao
,
L.
Wang
,
J.
Yan
,
W.
Liu
,
X.
Lei
,
X.
Wang
.
2012
.
Mixed lineage kinase domain-like protein mediates necrosis signaling downstream of RIP3 kinase.
Cell
148
:
213
227
.
3
Zhao
,
J.
,
S.
Jitkaew
,
Z.
Cai
,
S.
Choksi
,
Q.
Li
,
J.
Luo
,
Z. G.
Liu
.
2012
.
Mixed lineage kinase domain-like is a key receptor interacting protein 3 downstream component of TNF-induced necrosis.
Proc. Natl. Acad. Sci. USA
109
:
5322
5327
.
4
Murphy
,
J. M.
,
P. E.
Czabotar
,
J. M.
Hildebrand
,
I. S.
Lucet
,
J. G.
Zhang
,
S.
Alvarez-Diaz
,
R.
Lewis
,
N.
Lalaoui
,
D.
Metcalf
,
A. I.
Webb
, et al
.
2013
.
The pseudokinase MLKL mediates necroptosis via a molecular switch mechanism.
Immunity
39
:
443
453
.
5
Kaczmarek
,
A.
,
P.
Vandenabeele
,
D. V.
Krysko
.
2013
.
Necroptosis: the release of damage-associated molecular patterns and its physiological relevance.
Immunity
38
:
209
223
.
6
Degterev
,
A.
,
J.
Hitomi
,
M.
Germscheid
,
I. L.
Ch’en
,
O.
Korkina
,
X.
Teng
,
D.
Abbott
,
G. D.
Cuny
,
C.
Yuan
,
G.
Wagner
, et al
.
2008
.
Identification of RIP1 kinase as a specific cellular target of necrostatins.
Nat. Chem. Biol.
4
:
313
321
.
7
Newton
,
K.
,
D. L.
Dugger
,
K. E.
Wickliffe
,
N.
Kapoor
,
M. C.
de Almagro
,
D.
Vucic
,
L.
Komuves
,
R. E.
Ferrando
,
D. M.
French
,
J.
Webster
, et al
.
2014
.
Activity of protein kinase RIPK3 determines whether cells die by necroptosis or apoptosis.
Science
343
:
1357
1360
.
8
Polykratis
,
A.
,
N.
Hermance
,
M.
Zelic
,
J.
Roderick
,
C.
Kim
,
T.-M.
Van
,
T. H.
Lee
,
F. K. M.
Chan
,
M.
Pasparakis
,
M. A.
Kelliher
.
2014
.
Cutting edge: RIPK1 Kinase inactive mice are viable and protected from TNF-induced necroptosis in vivo.
J. Immunol.
193
:
1539
1543
.
9
Kelliher
,
M. A.
,
S.
Grimm
,
Y.
Ishida
,
F.
Kuo
,
B. Z.
Stanger
,
P.
Leder
.
1998
.
The death domain kinase RIP mediates the TNF-induced NF-kappaB signal.
Immunity
8
:
297
303
.
10
Gentle
,
I. E.
,
W. W.
Wong
,
J. M.
Evans
,
A.
Bankovacki
,
W. D.
Cook
,
N. R.
Khan
,
U.
Nachbur
,
J.
Rickard
,
H.
Anderton
,
M.
Moulin
, et al
.
2011
.
In TNF-stimulated cells, RIPK1 promotes cell survival by stabilizing TRAF2 and cIAP1, which limits induction of non-canonical NF-kappaB and activation of caspase-8. [Published erratum appears in 2016 J. Biol. Chem. 291: 2547.]
J. Biol. Chem.
286
:
13282
13291
.
11
Rickard
,
J. A.
,
J. A.
O’Donnell
,
J. M.
Evans
,
N.
Lalaoui
,
A. R.
Poh
,
T.
Rogers
,
J. E.
Vince
,
K. E.
Lawlor
,
R. L.
Ninnis
,
H.
Anderton
, et al
.
2014
.
RIPK1 regulates RIPK3-MLKL-driven systemic inflammation and emergency hematopoiesis.
Cell
157
:
1175
1188
.
12
Dillon
,
C. P.
,
R.
Weinlich
,
D. A.
Rodriguez
,
J. G.
Cripps
,
G.
Quarato
,
P.
Gurung
,
K. C.
Verbist
,
T. L.
Brewer
,
F.
Llambi
,
Y.-N.
Gong
, et al
.
2014
.
RIPK1 blocks early postnatal lethality mediated by caspase-8 and RIPK3.
Cell
157
:
1189
1202
.
13
Kaiser
,
W. J.
,
L. P.
Daley-Bauer
,
R. J.
Thapa
,
P.
Mandal
,
S. B.
Berger
,
C.
Huang
,
A.
Sundararajan
,
H.
Guo
,
L.
Roback
,
S. H.
Speck
, et al
.
2014
.
RIP1 suppresses innate immune necrotic as well as apoptotic cell death during mammalian parturition.
Proc. Natl. Acad. Sci. USA
111
:
7753
7758
.
14
Silke
,
J.
,
J. A.
Rickard
,
M.
Gerlic
.
2015
.
The diverse role of RIP kinases in necroptosis and inflammation.
Nat. Immunol.
16
:
689
697
.
15
Roderick
,
J. E.
,
N.
Hermance
,
M.
Zelic
,
M. J.
Simmons
,
A.
Polykratis
,
M.
Pasparakis
,
M. A.
Kelliher
.
2014
.
Hematopoietic RIPK1 deficiency results in bone marrow failure caused by apoptosis and RIPK3-mediated necroptosis.
Proc. Natl. Acad. Sci. USA
111
:
14436
14441
.
16
Dannappel
,
M.
,
K.
Vlantis
,
S.
Kumari
,
A.
Polykratis
,
C.
Kim
,
L.
Wachsmuth
,
C.
Eftychi
,
J.
Lin
,
T.
Corona
,
N.
Hermance
, et al
.
2014
.
RIPK1 maintains epithelial homeostasis by inhibiting apoptosis and necroptosis.
Nature
513
:
90
94
.
17
Caton
,
M. L.
,
M. R.
Smith-Raska
,
B.
Reizis
.
2007
.
Notch-RBP-J signaling controls the homeostasis of CD8- dendritic cells in the spleen.
J. Exp. Med.
204
:
1653
1664
.
18
Huang
,
S.
,
W.
Hendriks
,
A.
Althage
,
S.
Hemmi
,
H.
Bluethmann
,
R.
Kamijo
,
J.
Vilcek
,
R. M.
Zinkernagel
,
M.
Aguet
.
1993
.
Immune response in mice that lack the interferon-gamma receptor.
Science
259
:
1742
1745
.
19
Peschon
,
J. J.
,
D. S.
Torrance
,
K. L.
Stocking
,
M. B.
Glaccum
,
C.
Otten
,
C. R.
Willis
,
K.
Charrier
,
P. J.
Morrissey
,
C. B.
Ware
,
K. M.
Mohler
.
1998
.
TNF receptor-deficient mice reveal divergent roles for p55 and p75 in several models of inflammation.
J. Immunol.
160
:
943
952
.
20
Adachi
,
O.
,
T.
Kawai
,
K.
Takeda
,
M.
Matsumoto
,
H.
Tsutsui
,
M.
Sakagami
,
K.
Nakanishi
,
S.
Akira
.
1998
.
Targeted disruption of the MyD88 gene results in loss of IL-1- and IL-18-mediated function.
Immunity
9
:
143
150
.
21
Müller
,
U.
,
U.
Steinhoff
,
L. F.
Reis
,
S.
Hemmi
,
J.
Pavlovic
,
R. M.
Zinkernagel
,
M.
Aguet
.
1994
.
Functional role of type I and type II interferons in antiviral defense.
Science
264
:
1918
1921
.
22
Tabeta
,
K.
,
K.
Hoebe
,
E. M.
Janssen
,
X.
Du
,
P.
Georgel
,
K.
Crozat
,
S.
Mudd
,
N.
Mann
,
S.
Sovath
,
J.
Goode
, et al
.
2006
.
The Unc93b1 mutation 3d disrupts exogenous antigen presentation and signaling via Toll-like receptors 3, 7 and 9.
Nat. Immunol.
7
:
156
164
.
23
Hirschfeld
,
M.
,
Y.
Ma
,
J. H. J.
Weis
,
S. N.
Vogel
,
J. J.
Weis
.
2000
.
Cutting edge: repurification of lipopolysaccharide eliminates signaling through both human and murine toll-like receptor 2.
J. Immunol.
165
:
618
622
.
24
Hohlbaum
,
A. M.
,
M. S.
Gregory
,
S.-T.
Ju
,
A.
Marshak-Rothstein
.
2001
.
Fas ligand engagement of resident peritoneal macrophages in vivo induces apoptosis and the production of neutrophil chemotactic factors.
J. Immunol.
167
:
6217
6224
.
25
Bossaller
,
L.
,
V. A. K.
Rathinam
,
R.
Bonegio
,
P.- I.
Chiang
,
P.
Busto
,
A. R.
Wespiser
,
D. R.
Caffrey
,
Q.-Z.
Li
,
C.
Mohan
,
K. A.
Fitzgerald
, et al
.
2013
.
Overexpression of membrane-bound Fas ligand (CD95L) exacerbates autoimmune disease and renal pathology in pristane-induced lupus.
J. Immunol.
191
:
2104
2114
.
26
Dong
,
M. B.
,
M. J.
Rahman
,
K. V.
Tarbell
.
2016
.
Flow cytometric gating for spleen monocyte and DC subsets: differences in autoimmune NOD mice and with acute inflammation.
J. Immunol. Methods
432
:
4
12
.
27
Sharma
,
S.
,
A. M.
Campbell
,
J.
Chan
,
S. A.
Schattgen
,
G. M.
Orlowski
,
R.
Nayar
,
A. H.
Huyler
,
K.
Nündel
,
C.
Mohan
,
L. J.
Berg
, et al
.
2015
.
Suppression of systemic autoimmunity by the innate immune adaptor STING.
Proc. Natl. Acad. Sci. USA
112
:
E710
E717
.
28
Moujalled
,
D. M.
,
W. D.
Cook
,
T.
Okamoto
,
J.
Murphy
,
K. E.
Lawlor
,
J. E.
Vince
,
D. L.
Vaux
.
2013
.
TNF can activate RIPK3 and cause programmed necrosis in the absence of RIPK1.
Cell Death Dis.
4
:
e465
.
29
McComb
,
S.
,
H. H.
Cheung
,
R. G.
Korneluk
,
S.
Wang
,
L.
Krishnan
,
S.
Sad
.
2012
.
cIAP1 and cIAP2 limit macrophage necroptosis by inhibiting Rip1 and Rip3 activation.
Cell Death Differ.
19
:
1791
1801
.
30
Müller-Sienerth
,
N.
,
L.
Dietz
,
P.
Holtz
,
M.
Kapp
,
G. U.
Grigoleit
,
C.
Schmuck
,
H.
Wajant
,
D.
Siegmund
.
2011
.
SMAC mimetic BV6 induces cell death in monocytes and maturation of monocyte-derived dendritic cells.
PLoS One
6
:
e21556
.
31
Yabal
,
M.
,
N.
Müller
,
H.
Adler
,
N.
Knies
,
C. J.
Groß
,
R. B.
Damgaard
,
H.
Kanegane
,
M.
Ringelhan
,
T.
Kaufmann
,
M.
Heikenwälder
, et al
.
2014
.
XIAP restricts TNF- and RIP3-dependent cell death and inflammasome activation.
Cell Reports
7
:
1796
1808
.
32
Birnberg
,
T.
,
L.
Bar-On
,
A.
Sapoznikov
,
M. L.
Caton
,
L.
Cervantes-Barragán
,
D.
Makia
,
R.
Krauthgamer
,
O.
Brenner
,
B.
Ludewig
,
D.
Brockschnieder
, et al
.
2008
.
Lack of conventional dendritic cells is compatible with normal development and T cell homeostasis, but causes myeloid proliferative syndrome.
Immunity
29
:
986
997
.
33
Kang
,
T.-B.
,
S.-H.
Yang
,
B.
Toth
,
A.
Kovalenko
,
D.
Wallach
.
2013
.
Caspase-8 blocks kinase RIPK3-mediated activation of the NLRP3 inflammasome.
Immunity
38
:
27
40
.
34
Croker
,
B. A.
,
J. A.
O’Donnell
,
M.
Gerlic
.
2014
.
Pyroptotic death storms and cytopenia.
Curr. Opin. Immunol.
26
:
128
137
.
35
Vince
,
J. E.
,
W. W.
Wong
,
I.
Gentle
,
K. E.
Lawlor
,
R.
Allam
,
L.
O’Reilly
,
K.
Mason
,
O.
Gross
,
S.
Ma
,
G.
Guarda
, et al
.
2012
.
Inhibitor of apoptosis proteins limit RIP3 kinase-dependent interleukin-1 activation.
Immunity
36
:
215
227
.
36
Angus
,
D. C.
,
T.
van der Poll
.
2013
.
Severe sepsis and septic shock.
N. Engl. J. Med.
369
:
840
851
.
37
Laouar
,
Y.
,
S.
Ezine
.
1994
.
In vivo CD4+ lymph node T cells from lpr mice generate CD4-CD8-B220+TCR-beta low cells.
J. Immunol.
153
:
3948
3955
.
38
Kaiser
,
W. J.
,
J. W.
Upton
,
A. B.
Long
,
D.
Livingston-Rosanoff
,
L. P.
Daley-Bauer
,
R.
Hakem
,
T.
Caspary
,
E. S.
Mocarski
.
2011
.
RIP3 mediates the embryonic lethality of caspase-8-deficient mice.
Nature
471
:
368
372
.
39
Kono
,
D. H.
,
M. K.
Haraldsson
,
B. R.
Lawson
,
K. M.
Pollard
,
Y. T.
Koh
,
X.
Du
,
C. N.
Arnold
,
R.
Baccala
,
G. J.
Silverman
,
B. A.
Beutler
,
A. N.
Theofilopoulos
.
2009
.
Endosomal TLR signaling is required for anti-nucleic acid and rheumatoid factor autoantibodies in lupus.
Proc. Natl. Acad. Sci. USA
106
:
12061
12066
.
40
Christensen
,
S. R.
,
M.
Kashgarian
,
L.
Alexopoulou
,
R. A.
Flavell
,
S.
Akira
,
M. J.
Shlomchik
.
2005
.
Toll-like receptor 9 controls anti-DNA autoantibody production in murine lupus.
J. Exp. Med.
202
:
321
331
.
41
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
.
42
Yang
,
H.-C.
,
Y.
Zuo
,
A. B.
Fogo
.
2010
.
Models of chronic kidney disease.
Drug Discov. Today Dis. Models
7
:
13
19
.
43
He
,
S.
,
Y.
Liang
,
F.
Shao
,
X.
Wang
.
2011
.
Toll-like receptors activate programmed necrosis in macrophages through a receptor-interacting kinase-3-mediated pathway.
Proc. Natl. Acad. Sci. USA
108
:
20054
20059
.
44
Kaiser
,
W. J.
,
H.
Sridharan
,
C.
Huang
,
P.
Mandal
,
J. W.
Upton
,
P. J.
Gough
,
C. A.
Sehon
,
R. W.
Marquis
,
J.
Bertin
,
E. S.
Mocarski
.
2013
.
Toll-like receptor 3-mediated necrosis via TRIF, RIP3, and MLKL.
J. Biol. Chem.
288
:
31268
31279
.
45
Moriwaki
,
K.
,
S.
Balaji
,
T.
McQuade
,
N.
Malhotra
,
J.
Kang
,
F. K.
Chan
.
2014
.
The necroptosis adaptor RIPK3 promotes injury-induced cytokine expression and tissue repair.
Immunity
41
:
567
578
.
46
Beutler
,
B.
,
Z.
Jiang
,
P.
Georgel
,
K.
Crozat
,
B.
Croker
,
S.
Rutschmann
,
X.
Du
,
K.
Hoebe
.
2006
.
Genetic analysis of host resistance: Toll-like receptor signaling and immunity at large.
Annu. Rev. Immunol.
24
:
353
389
.
47
Banchereau
,
J.
,
V.
Pascual
.
2006
.
Type I interferon in systemic lupus erythematosus and other autoimmune diseases.
Immunity
25
:
383
392
.
48
Thapa
,
R. J.
,
S.
Nogusa
,
P.
Chen
,
J. L.
Maki
,
A.
Lerro
,
M.
Andrake
,
G. F.
Rall
,
A.
Degterev
,
S.
Balachandran
.
2013
.
Interferon-induced RIP1/RIP3-mediated necrosis requires PKR and is licensed by FADD and caspases.
Proc. Natl. Acad. Sci. USA
110
:
E3109
E3118
.
49
Brinkmann
,
M. M.
,
E.
Spooner
,
K.
Hoebe
,
B.
Beutler
,
H. L.
Ploegh
,
Y. M.
Kim
.
2007
.
The interaction between the ER membrane protein UNC93B and TLR3, 7, and 9 is crucial for TLR signaling.
J. Cell Biol.
177
:
265
275
.
50
Kim
,
Y.-M.
,
M. M.
Brinkmann
,
M.-E.
Paquet
,
H. L.
Ploegh
.
2008
.
UNC93B1 delivers nucleotide-sensing toll-like receptors to endolysosomes.
Nature
452
:
234
238
.
51
Legarda
,
D.
,
S. J.
Justus
,
R. L.
Ang
,
N.
Rikhi
,
W.
Li
,
T. M.
Moran
,
J.
Zhang
,
E.
Mizoguchi
,
M.
Zelic
,
M. A.
Kelliher
, et al
.
2016
.
CYLD proteolysis protects macrophages from TNF-mediated auto-necroptosis induced by LPS and licensed by Type I IFN.
Cell Reports
15
:
2449
2461
.
52
Wynn
,
T. A.
2011
.
Integrating mechanisms of pulmonary fibrosis.
J. Exp. Med.
208
:
1339
1350
.
53
Pasparakis
,
M.
,
L.
Alexopoulou
,
V.
Episkopou
,
G.
Kollias
.
1996
.
Immune and inflammatory responses in TNF alpha-deficient mice: a critical requirement for TNF alpha in the formation of primary B cell follicles, follicular dendritic cell networks and germinal centers, and in the maturation of the humoral immune response.
J. Exp. Med.
184
:
1397
1411
.
54
Matsumoto
,
M.
,
S.
Mariathasan
,
M. H.
Nahm
,
F.
Baranyay
,
J. J.
Peschon
,
D. D.
Chaplin
.
1996
.
Role of lymphotoxin and the type I TNF receptor in the formation of germinal centers.
Science
271
:
1289
1291
.
55
Kim
,
J. Y.
,
M.
Morgan
,
D. G.
Kim
,
J. Y.
Lee
,
L.
Bai
,
Y.
Lin
,
Z. G.
Liu
,
Y. S.
Kim
.
2011
.
TNFα induced noncanonical NF-κB activation is attenuated by RIP1 through stabilization of TRAF2.
J. Cell Sci.
124
:
647
656
.
56
Young
,
J. A.
,
T. H.
He
,
B.
Reizis
,
A.
Winoto
.
2013
.
Commensal microbiota are required for systemic inflammation triggered by necrotic dendritic cells.
Cell Reports
3
:
1932
1944
.
57
Cuda
,
C. M.
,
A. V.
Misharin
,
A. K.
Gierut
,
R.
Saber
,
G. K.
Haines
III
,
J.
Hutcheson
,
S. M.
Hedrick
,
C.
Mohan
,
G. S.
Budinger
,
C.
Stehlik
,
H.
Perlman
.
2014
.
Caspase-8 acts as a molecular rheostat to limit RIPK1- and MyD88-mediated dendritic cell activation.
J. Immunol.
192
:
5548
5560
.
58
Oberst
,
A.
,
C. P.
Dillon
,
R.
Weinlich
,
L. L.
McCormick
,
P.
Fitzgerald
,
C.
Pop
,
R.
Hakem
,
G. S.
Salvesen
,
D. R.
Green
.
2011
.
Catalytic activity of the caspase-8-FLIP(L) complex inhibits RIPK3-dependent necrosis.
Nature
471
:
363
367
.
59
Alvarez-Diaz
,
S.
,
C. P.
Dillon
,
N.
Lalaoui
,
M. C.
Tanzer
,
D. A.
Rodriguez
,
A.
Lin
,
M.
Lebois
,
R.
Hakem
,
E. C.
Josefsson
,
L. A.
O’Reilly
, et al
.
2016
.
The Pseudokinase MLKL and the Kinase RIPK3 Have Distinct Roles in Autoimmune Disease Caused by Loss of Death-Receptor-Induced Apoptosis.
Immunity
45
:
513
526
.
60
Rifkin
,
I. R.
,
E. A.
Leadbetter
,
L.
Busconi
,
G.
Viglianti
,
A.
Marshak-Rothstein
.
2005
.
Toll-like receptors, endogenous ligands, and systemic autoimmune disease.
Immunol. Rev.
204
:
27
42
.
61
Lau
,
C. M.
,
C.
Broughton
,
A. S.
Tabor
,
S.
Akira
,
R. A.
Flavell
,
M. J.
Mamula
,
S. R.
Christensen
,
M. J.
Shlomchik
,
G. A.
Viglianti
,
I. R.
Rifkin
,
A.
Marshak-Rothstein
.
2005
.
RNA-associated autoantigens activate B cells by combined B cell antigen receptor/Toll-like receptor 7 engagement.
J. Exp. Med.
202
:
1171
1177
.
62
Leadbetter
,
E. A.
,
I. R.
Rifkin
,
A. M.
Hohlbaum
,
B. C.
Beaudette
,
M. J.
Shlomchik
,
A.
Marshak-Rothstein
.
2002
.
Chromatin-IgG complexes activate B cells by dual engagement of IgM and Toll-like receptors.
Nature
416
:
603
607
.
63
Hua
,
J.
,
K.
Kirou
,
C.
Lee
,
M. K.
Crow
.
2006
.
Functional assay of type I interferon in systemic lupus erythematosus plasma and association with anti-RNA binding protein autoantibodies.
Arthritis Rheum.
54
:
1906
1916
.
64
Sherer
,
Y.
,
A.
Gorstein
,
M. J.
Fritzler
,
Y.
Shoenfeld
.
2004
.
Autoantibody explosion in systemic lupus erythematosus: more than 100 different antibodies found in SLE patients.
Semin. Arthritis Rheum.
34
:
501
537
.
65
Sokolove
,
J.
,
R.
Bromberg
,
K. D.
Deane
,
L. J.
Lahey
,
L. A.
Derber
,
P. E.
Chandra
,
J. D.
Edison
,
W. R.
Gilliland
,
R. J.
Tibshirani
,
J. M.
Norris
, et al
.
2012
.
Autoantibody epitope spreading in the pre-clinical phase predicts progression to rheumatoid arthritis. [Published erratum appears in 2012 PLoS One. 7.]
.
PLoS One
7
:
e35296
.
66
Baechler
,
E. C.
,
F. M.
Batliwalla
,
G.
Karypis
,
P. M.
Gaffney
,
W. A.
Ortmann
,
K. J.
Espe
,
K. B.
Shark
,
W. J.
Grande
,
K. M.
Hughes
,
V.
Kapur
, et al
.
2003
.
Interferon-inducible gene expression signature in peripheral blood cells of patients with severe lupus.
Proc. Natl. Acad. Sci. USA
100
:
2610
2615
.
67
Bennett
,
L.
,
A. K.
Palucka
,
E.
Arce
,
V.
Cantrell
,
J.
Borvak
,
J.
Banchereau
,
V.
Pascual
.
2003
.
Interferon and granulopoiesis signatures in systemic lupus erythematosus blood.
J. Exp. Med.
197
:
711
723
.
68
Kirou
,
K. A.
,
C.
Lee
,
S.
George
,
K.
Louca
,
M. G. E.
Peterson
,
M. K.
Crow
.
2005
.
Activation of the interferon-alpha pathway identifies a subgroup of systemic lupus erythematosus patients with distinct serologic features and active disease.
Arthritis Rheum.
52
:
1491
1503
.
69
Newton
,
K.
,
K. E.
Wickliffe
,
A.
Maltzman
,
D. L.
Dugger
,
A.
Strasser
,
V. C.
Pham
,
J. R.
Lill
,
M.
Roose-Girma
,
S.
Warming
,
M.
Solon
, et al
.
2016
.
RIPK1 inhibits ZBP1-driven necroptosis during development.
Nature
540
:
129
133
.
70
Lin
,
J.
,
S.
Kumari
,
C.
Kim
,
T.-M.
Van
,
L.
Wachsmuth
,
A.
Polykratis
,
M.
Pasparakis
.
2016
.
RIPK1 counteracts ZBP1-mediated necroptosis to inhibit inflammation.
Nature
540
:
124
128
.
71
Takaoka
,
A.
,
Z.
Wang
,
M. K.
Choi
,
H.
Yanai
,
H.
Negishi
,
T.
Ban
,
Y.
Lu
,
M.
Miyagishi
,
T.
Kodama
,
K.
Honda
, et al
.
2007
.
DAI (DLM-1/ZBP1) is a cytosolic DNA sensor and an activator of innate immune response.
Nature
448
:
501
505
.
72
Thapa
,
R. J.
,
J. P.
Ingram
,
K. B.
Ragan
,
S.
Nogusa
,
D. F.
Boyd
,
A. A.
Benitez
,
H.
Sridharan
,
R.
Kosoff
,
M.
Shubina
,
V. J.
Landsteiner
, et al
.
2016
.
DAI senses influenza A virus genomic RNA and activates RIPK3-dependent cell death.
Cell Host Microbe
20
:
674
681
.
73
Zhang
,
W.
,
Q.
Zhou
,
W.
Xu
,
Y.
Cai
,
Z.
Yin
,
X.
Gao
,
S.
Xiong
.
2013
.
DNA-dependent activator of interferon-regulatory factors (DAI) promotes lupus nephritis by activating the calcium pathway.
J. Biol. Chem.
288
:
13534
13550
.

The authors have no financial conflicts of interest.

Supplementary data