Abstract
The linear ubiquitin chain assembly complex (LUBAC) plays pivotal roles in regulating lymphocyte activation, inflammation, and cell death. This is highlighted by the fact that patients with mutations in LUBAC catalytic subunit HOIP suffer from autoinflammation combined with immunodeficiency. Although defective development of T and B cells resulting from HOIP deficiency in adaptive immunity can explain immunodeficiency, the pathogenesis of autoinflammation is not clear. In this study, we found that dendritic cell (DC)–specific deletion of HOIP resulted in spontaneous inflammation, indicating the essential role of HOIP in maintaining DC homeostasis. Although HOIP deficiency in DCs did not affect TNF-α–induced NF-κB activation, it enhanced TNF-α–induced apoptosis and necroptosis. However, crossing HoipDC KO mice with TNFR1-knockout mice surprisingly could not rescue the systematic inflammation, suggesting that the autoinflammation is not due to the effect of HOIP on TNF-α signaling. In contrast, treatment of HoipDC KO mice with antibiotics reduced the inflammation, implying that TLR signaling may contribute to the inflammatory phenotype found in HoipDC KO mice. Consistently, we found that LPS induced more cell death and significantly higher levels of IL-1α and IL-1β in HoipDC KO cells. Importantly, MyD88 deficiency rescued the inflammatory phenotype in HoipDC KO mice. Together, these findings reveal the indispensable function of HOIP in maintaining DC homeostasis, and MyD88-dependent proinflammatory signal plays a substantial role in the pathogenesis of human autoinflammation associated with HOIP mutations.
Introduction
The linear ubiquitin chain assembly complex (LUBAC), composed of three core subunits, heme-oxidized IRP2 ubiquitin ligase 1 (HOIL-1), Shank-associated RH domain-interacting protein (SHARPIN), and the catalytically active HOIL-1–interacting protein (HOIP), is the only E3 ligase identified so far that can generate linear ubiquitin linkages (1–3). Linear ubiquitylation is crucial for multiple immune receptor signaling pathways, including TNF, TLRs, TRAIL, NOD-like receptors, and T and B cell receptors (4–9). The essence of LUBAC and linear ubiquitylation has been demonstrated by embryonic lethality of mice with deficiency of LUBAC components HOIP, HOIL-1, or LUBAC activity, which is caused by aberrant TNFR1-mediated signaling (10, 11). Defect in SHARPIN triggers severe dermatitis and systemic inflammation in adulthood (12, 13). In humans, patients with loss-of-function mutations in HOIP or HOIL-1 causes multiorgan autoinflammation, combined immunodeficiency, and recurrent viral and bacterial infections (14–16). At cellular level, B cell–specific ablation of HOIP results in B-1 cell development defect and defective Ab response to thymus-dependent and independent Ags in mice (8, 17). And mice with T cell–specific HOIP ablation display a strong defect in late thymocyte differentiation, regulatory T cell development, and homeostasis (7). Although defects in B cell and T cell development resulting from LUBAC deficiency may partially account for the immunodeficiency in patients with loss-of-function mutations in HOIP or HOIL-1, the source of autoinflammation is still not well understood, and dysregulated LUBAC activity is expected to have cell-specific effects on many cellular pathways and functions. Whether deficiency of LUBAC in innate immune cells, especially dendritic cells (DCs), is involved in the pathogenesis of autoinflammation or immunodeficiency is not clear.
DCs are potent APCs that initiate lymphocyte activation and critical for maintaining immune tolerance (18, 19). DCs are ubiquitously distributed in all tissues and surveil the environment to initiate immune response against foreign Ags or tolerance to self-antigens, especially in the intestine where microbiota and foreign Ags are ample. Therefore, strict regulations of DC activities are required for proper execution of immune response. The differentiation of DCs from common DC progenitors (CDPs) depends on the cytokines like fms-related tyrosine receptor kinase 3 ligand (Flt-3L) and GM-CSF (20), whereas homeostasis of DCs is maintained by processes like apoptosis to limit immune response after Ag presentation to T cells (21). Dysregulation of apoptosis in DCs results in the development of systemic autoimmunity (18, 22–24).
Given the indispensable role of DCs in linking innate and adaptive systems and dysregulation of DCs resulting in the development of systemic autoimmunity, we sought to examine the function of HOIP in DCs and explore whether loss of LUBAC function in DCs has any relationship with autoinflammation of patients. Using DC-specific conditional knockout mice, we found that HOIP was essential for maintenance of DC homeostasis and prevention of systemic inflammation. In knockout mice, DC numbers were decreased in the spleen because of uncontrolled DC death. Importantly, dysregulation of DC death resulted in the appearance of chronic systemic inflammation characterized by increased levels of serum proinflammatory cytokines and expansions in myeloid and B cells. We further identified a critical role for the intestinal microbiota and MyD88-dependent signals in the development of inflammation in mice with DC-specific HOIP deficiency. Our study uncovers that aberrant DC death via deficiency of LUBAC in DCs may underpin the pathogenesis of autoinflammation in LUBAC-mutated patients, and MyD88-dependent proinflammatory signal plays a substantial role in the autoinflammation associated with HOIP mutations.
Materials and Methods
Mice
All mice used in this study were on the C57BL/6 background at 7–9 wk of age unless otherwise noted. HoipDC KO mice were generated by crossing Itgax-Cre (CD11c-Cre) transgenic mice with Hoipflox/flox mice. Hoipflox/flox mice were generated by Crispr-Cas9 technique and backcrossed to C57BL/6 for at least 10 generations, as previously described (25). Itgax-Cre, Tnfrl−/−, and Casp8fl3-4 mice were purchased from The Jackson Laboratory and bred in the facility. Myd88−/−mice and Mlkl−/−mice were generously provided by Dr. Xiaoyu Hu at Tsinghua and Dr. Xiaodong Wang from the National Institute of Biological Sciences, respectively. For antibiotic treatment of HoipDC KO mice, broad-spectrum antibiotic mixture of 1 mg/ml ampicillin, 1 mg/ml neomycin, 0.5 mg/ml ciprofloxacin, and 0.5 mg/ml meropenem was added to the drinking water starting from day 2 of birth. Following weaning, mice were maintained on the same antibiotic mixture, except with 0.5 mg/ml vancomycin in place of ciprofloxacin until analysis (26). Mice treated with antibiotics were analyzed at ∼7 wk of age. All mice were housed in the specific pathogen–free animal facilities in Tsinghua University. All experiments used cohoused mice. All mouse experiments were performed in compliance with institutional guidelines and according to the protocol approved by the Institutional Animal Care and Use Committee of Tsinghua University.
Histology
Mouse spleens were fixed with 4% formaldehyde overnight and then dehydrated and embedded in paraffin. The 5-μm tissue sections were stained with H&E or Masson’s trichrome according to standard procedures. Stained sections were scanned using an Olympus microscope (IX73).
Flow cytometry
Spleens and lymph nodes were mechanically dissociated to obtain single-cell suspensions, and splenocytes were depleted of RBCs with ACK lysis buffer. Surface staining was completed in cold FACS buffer (PBS containing 1% FCS, 0.05% sodium azide). Samples were incubated with anti-CD16/CD32 prior to staining with the indicated fluorochrome-conjugated Abs. Samples were run on LSR Fortessa cytometers (BD Biosciences) and analyzed using FlowJo software (Tree Star). Cell populations were determined as follows: conventional DCs (cDCs), CD11chigh MHC class II+ (MHC II+); plasmacytoid DCs, CD11clow Siglec-H+; cDC1 cells, CD11chigh MHC II+ CD8+ Sirp1α−; cDC2 cells, CD11chigh MHC II+ CD8− Sirp1α+; pre-DCs, lin− c-kitint Flt-3+ CD11c+; CD115+ CDPs, lin− c-kitint Flt-3+ CD127− CD115+; neutrophils, CD11b+ Ly6Clow Ly6Ghigh; monocytes, CD11b+ Ly6Chigh Ly6Glow; B lymphocytes, CD19+ or B220+; follicular B lymphocytes, CD19+ CD93− CD23+ CD21low; germinal center B lymphocytes, CD19+ IgDlow CD95+ GL7+; CD4 T lymphocytes, CD4+ CD8−; CD8 T lymphocytes, CD8+ CD4−; and activated T lymphocytes, CD44high CD62Llow. See Supplemental Table I for Abs used in this study.
ELISA
Blood was collected from cardiac puncture, and different proteins in the serum were analyzed by sandwich ELISA. The levels of KC (MultiSciences), MCP-1 (MultiSciences), TNF-α (eBioscience), IFN-γ (eBioscience), IL-6 (eBioscience), IL-1β (eBioscience), and IL-1α (Abcam) were determined using ELISA kits. Abs to dsDNA (Thermo Fisher Scientific) were measured as previously described (27).
DC enrichment
Bone marrow was flushed from femurs and tibias using a needle and syringe, and RBCs were lysed. Bone marrow–derived DCs (BMDCs) were generated by culturing bone marrow cell suspensions in complete RPMI 1640 supplemented with 10% FCS, l-glutamine, penicillin-streptomycin, sodium pyruvate, 2-ME, and 20 ng/ml recombinant GM-CSF (PeproTech) for 8 d. BMDCs were harvested and replated in complete RPMI 1640 for stimulation. Alternatively, CD11c+ BMDCs were enriched by sorting with FACS Aria (BD Biosciences).
Real-time quantitative PCR
Total RNA was extracted by TRIzol (Invitrogen), and reverse transcription was performed using RevertAid First Strand cDNA Synthesis Kit (Thermo Fisher Scientific) according to the manufacturer’s instructions. An ABI 7500 Real-Time PCR system (Applied Biosystems) and Power SYBR Green PCR Master Mix (Genestar) were used for real-time quantitative PCR. Results were normalized to Gapdh, and quantification was carried out using the 2-ΔΔCt method. Melting curves were confirmed to ensure amplification of a single product. Primers for Hoip, which are located in exon3 and exon6, were designed. Primer pairs are as follows: Gapdh, sense, 5′-AAC AGC AAC TCC CAC TCT TC -3′, antisense, 5′-CCT GTT GCT GTA GCC GTA TT-3′; Il1a, sense, 5′-CGA AGA CTA CAG TTC TGC CAT T-3′, antisense, 5′-GAC GTT TCA GAG GTT CTC AGA G-3′; Il1b, sense, 5′-GCA ACT GTT CCT GAA CTC AAC T-3′, antisense, 5′-ATC TTT TGG GGT CCG TCA ACT-3′; and Hoip, sense, 5′-ACC GAG CTC AGT TTG CTG TT-3′, antisense, 5′-AGG AAA CAG GGA CCA GGA GT-3′.
Cell viability assay
A total of 5 × 104 DCs per well in a 96-well format were treated with indicated stimulation. Effects on cell viability were measured using Cell Titer Glo, as described (28). Cell Titer Glo Luminescent Cell Viability Assay kit was from Promega.
Western blotting
Cells were collected with lysis buffer (50 mM HEPES, pH 7.4, 150 mM NaCl, 1% NP-40, 1 mM EDTA) containing 1 mM sodium orthovanadate, 1 mM sodium fluoride, 1 mM PMSF, and a protease inhibitor mixture (Roche). Total cell lysates were subjected to SDS-PAGE and transferred onto a PVDF membrane (Millipore). The membrane was sequentially probed with primary Abs and HRP-conjugated secondary Abs. ECL substrates (Pierce) were used to visualize the specific bands on the membrane.
For complex I immunoprecipitation, 2 × 107 treated BMDCs were lysed in lysis buffer (30 mM Tris-HCl, pH 7.4, 120 mM NaCl, 2 mM EDTA, 2 mM KCl, 10% glycerol, and 1% Triton X-100) containing 1 mM sodium orthovanadate, 1 mM sodium fluoride, 1 mM PMSF, and a protease inhibitor mixture (Roche). Each sample was incubated with anti-FLAG M2 beads (A2220; Sigma-Aldrich) at 4°C overnight. Agarose beads were washed four times with cold lysis buffer, and the precipitates were boiled for 10 min in SDS loading buffer. The input and immunoprecipitation samples were then subjected to SDS-PAGE and then blotted using the indicated Abs. See Supplemental Table for Abs used in this study.
Generation of bone marrow chimera mice
For bone marrow chimeras, 8-wk-old wild-type (WT) recipient mice were lethally irradiated by x-ray (5.5 Gy × 2) and received 5 × 106 bone marrow cells from indicated donors by i.v. injection. Chimeras were used for further experiments 8 wk after initial reconstitution.
Statistical analysis
All statistical analysis was performed using Prism 6 (GraphPad Software). Statistical significance was evaluated using Student t test or one-way ANOVA. Log-rank (Mantel–Cox) test was used to compare survival curves. Differences were considered statistically significant if *p < 0.05, **p < 0.01, ***p < 0.001, and ****p < 0.0001. Statistical parameters including number of biological replicates and repeat experiments, data dispersion, and precision measures (mean and SEM) are reported in the figure legends.
Results
HoipDC KO mice spontaneously develop systemic inflammation
To investigate the role of HOIP in DC homeostasis and effector functions, we generated mice that lack HOIP in the DCs by crossing Hoipflox/flox mice (Supplemental Fig. 1A) with mice expressing the Cre recombinase under the control of CD11c promoter (Itgax-Cre). Mice homozygous for DC-specific deletion of HOIP (Itgax-Cre Hoipflox/flox, termed “HoipDC KO”) was confirmed by PCR (Supplemental Fig. 1B). At mRNA expression level, the specific deletion of HOIP was confirmed in sorted DCs by quantitative RT-PCR compared with other immune cells of hematopoietic origin (Supplemental Fig. 1C). At protein level, the specific loss of HOIP expression in enriched CD11c+ in vitro–generated BMDC was verified by Western blot analysis compared with Itgax-wt Hoipflox/flox mice (termed “control”) (Supplemental Fig. 1D). In line with previous reports on other tissues and cells, analysis of TNFR1-signaling complex by TNF-α–Flag pulldown revealed that HOIP-deficient cells produced remarkably reduced levels of linear ubiquitination within the TNFR1 signaling complex (Supplemental Fig. 1E). Previous studies have reported that mouse embryonic fibroblasts with ablation of HOIP result in the impaired canonical NF-κB activation as exhibited by delayed phosphorylation and degradation of IκBα (10). Therefore, we examined NF-κB signaling in HOIP-deficient DCs. Surprisingly, this is not the case for HoipDC KO cells (Supplemental Fig. 1F). Consistently, NF-κB–targeted gene Ciap1 and A20 expression are not affected by HOIP deficiency in DCs (Supplemental Fig. 1G).
HoipDC KO mice were born at the expected Mendelian ratio and appeared healthy with no obvious abnormalities. However, by 6–8 wk of age, HoipDC KO mice spontaneously developed splenomegaly and lymphadenopathy (Fig. 1A–C). Histological analysis revealed that the splenic architecture was disrupted in HoipDC KO mice, and Masson trichrome staining revealed collagen deposits in the spleen, indicating fibrosis (Fig. 1D). Evidence of inflammation was observed in HoipDC KO mice, with elevated percentage and numbers of neutrophils and inflammatory monocytes in the spleens and mesenteric lymph nodes (mLNs) of HoipDC KO mice (Fig. 1E, 1F). We also detected elevated B cell numbers in the spleen and mLNs of HoipDC KO mice; further analysis revealed that follicular B cells, especially geminal center B cells, were accounted for the expansion of B cells (Supplemental Fig. 2A–C). Consistent with increased B cell expansion, a significant increase in serum anti-dsDNA IgG was observed in HoipDC KO mice compared with control mice (Fig. 1G), implicating autoimmunity was developed in HoipDC KO mice. Conversely, the absolute numbers of CD4 and CD8 T lymphocytes in spleen or mLNs have no differences, and there was no evidence of T cell activation in spleen or lymph nodes (Supplemental Fig. 2D, 2E). To further assess the inflammation in HoipDC KO mice, the levels of different proinflammatory cytokines and chemokines in the serum were measured. A significant increase in basal serum TNF-α, IFN-γ, KC, and MCP-1 was observed in HoipDC KO mice compared with control mice (Fig. 1H). To determine the ramifications of having increased inflammation and elevated cytokine levels, control or HoipDC KO mice were given a low dose of LPS (5 mg/kg). Under this circumstance, most of control mice survived, whereas all HoipDC KO mice succumbed to LPS-induced endotoxic shock within 48 h (Fig. 1I). Additionally, the levels of proinflammatory cytokines TNF-α, IL-1β, and IL-6 were significantly elevated in the sera of HoipDC KO mice compared with controls following LPS injection (Fig. 1J). Our data specifically demonstrate that HoipDC KO mice develop systemic inflammation characterized by increased numbers of neutrophils, inflammatory monocytes, B cells, and elevated levels of proinflammatory cytokines.
Decreased DC numbers and increased susceptibility of HOIP-deficient DCs to cell death
We further determined the effect of HOIP deficiency on DC population change. A significant decrease in the number of CD11chiMHC II+ cDCs was detected in the spleen of HoipDC KO mice compared with littermate control mice (Fig. 2A, 2B), whereas the number of plasmacytoid DCs in spleen was unchanged (Supplemental Fig. 3A, 3B). Further investigation of different subsets of splenic cDCs by examining CD8 and Sirp1α expression revealed that the percentage of CD8+Sirp1α− DC subset (termed “cDC1”) was diminished, whereas the CD8−Sirp1α+ subset (termed “cDC2”) was increased (Supplemental Fig. 3A, 3B). Apart from spleen, CD103+ migratory DCs from mLNs were examined (29). Interestingly, significant reduction of migratory CD103+ DCs in mLN was observed in HoipDC KO mice (Fig. 2A, 2B). To investigate whether developmental impairment within DC precursor populations was responsible for the reduction of DC numbers in peripheral lymphoid organs, we examined pre-DCs and CDPs in the bone marrow of HoipDC KO mice. HoipDC KO mice still exhibited comparable number of both pre-DCs and CDPs in the bone marrow (Supplemental Fig. 3C, 3D), indicating that the origin of DCs is not responsible for the decreased DCs in peripheral lymphoid organs.
We next determined the cause of DC reduction and whether the decrease in DCs was responsible for the systemic inflammation observed in the HoipDC KO mice. It has been shown that LUBAC plays a critical role in TNF-α–induced cell death by switching prosurvival TNFR1 signaling into Caspase-8–dependent apoptosis or Caspase-8–independent, RIPK3/MLKL-mediated necroptosis (1, 10, 11). Therefore, we assessed the ability of HOIP-deficient DCs to undergo cell death in response TNF-α stimulation. Apoptosis can be classified into RIPK1-independent apoptosis (RIA) and RIPK1-dependent apoptosis (RDA) according to the dependence on RIPK1 kinase activity. Thus, we investigated the effect of HOIP loss on RIA. With TNF-α/CHX stimulation, cell death induction was comparable between HOIP-deficient DCs and control DCs (Fig. 2C, 2D), indicating HOIP is not required for prevention of TNF-α–induced RIA in DCs.
Second mitochondrial–derived activator of caspase (Smac) is a mitochondrial protein that can promote caspase activation to induce RDA through inhibition of cIAP1/2 (30). We then assessed the RDA induced by TNF-α plus Smac-mimetics (BV-6), and interestingly, we found that HOIP-deficient DCs showed a higher level of cell death than control DCs (Fig. 2C, 2E), suggesting HOIP is involved in negative regulation of TNF-α–induced RDA. Moreover, when TNF-α–induced apoptosis was blocked by pan-caspase inhibitor z-VAD, which induces necroptosis, the viability of HOIP-deficient DCs was lower compared with control cells (Fig. 2C, 2F), indicating HOIP is required for prevention of necroptosis induced by TNF-α. Necroptosis induced by TNF-α is dependent on the activities of RIPK1, RIPK3, and the pseudo-kinase MLKL. We also investigated the effect of inhibiting RIPK1 kinase activity on the caspase activation and necroptosis in HOIP-deficient DCs. Blocking of RDA and necroptosis using RIPK1 kinase inhibitor, Necrostatin-1, could increase the survival of DCs from HoipDC KO mice (Fig. 2C–F).
TNFR1 deficiency exacerbates systemic inflammation of HoipDC KO mice
Because TNFR1 deficiency could prolong the survival of Hoip−/− and Hoil-1−/− embryos (10, 11) and HOIP-deficient DCs are sensitive to TNF-α–induced apoptosis and necroptosis, we hypothesized that TNF-α–induced cell death may cause the systemic inflammation in HoipDC KO mice. To address the role of TNFR1 signaling in inflammation in vivo, we crossed HOIPDC KO mice with Tnfrl−/− mice to generate HoipDC KO Tnfrl−/− mice. Surprisingly, TNFR1 deficiency did not rescue inflammation but resulted in significant weight loss of HoipDC KO Tnfrl−/− mice (Fig. 3A). In contrast to the accelerated TNF-α–induced cell death observed in vitro (Fig. 2E, 2F), HoipDC KO Tnfrl−/− mice develop more severe systemic inflammation compared with HoipDC KO mice, with significant increases in percentage and numbers of inflammatory monocytes and neutrophils in spleen and mLN (Fig. 3C–F). Besides, absolute number of DCs of HoipDC KO Tnfrl−/− mice were still reduced compared with HoipDC KO mice (Fig. 3G, 3H). Although HOIP is critical for restricting TNF-α–induced cell death in vitro, HoipDC KO Tnfrl−/− mice develop inflammation and cachexia, indicating that signaling other than TNFR1 may contribute to HOIP deficiency–associated inflammation.
Systemic inflammation in HoipDC KO mice is partially abrogated by antibiotic treatment
Because HoipDC KO mice have increased susceptibility to LPS-stimulated death (Fig. 1I) and multiple groups have reported that LPS can induce cell death (28, 31, 32), we examined whether a lower level of DCs in HoipDC KO mice was the result of HOIP-deficient DCs dying by LPS challenge. BMDCs generated from HoipDC KO or control mice were stimulated with LPS, cell death was examined by cell survival measurement, and increased cell death was observed in HoipDC KO cells (Fig. 4A). LPS has been reported as a strong inducer for both apoptosis and necroptosis; thus, we further investigated whether HOIP deficiency affects LPS-induced apoptosis. Because treatment with LPS in the presence of CHX, an inhibitor of protein synthesis, leads to activation of Caspase-3 and apoptosis via TLR4 (33), we stimulated BMDCs from HoipDC KO or control mice with LPS/CHX. With LPS/CHX stimulation, HOIP-deficient DCs exhibited higher level of cell death than control DCs (Fig. 4A, 4B), indicating that HOIP is required for prevention of apoptosis induced by LPS/CHX. Apart from LPS/CHX-induced apoptosis, apoptosis can also be induced by LPS in cells with the absence of inhibitor of apoptosis protein (IAP) (32). Thus, we used the LPS plus Smac-mimetics (BV-6) to inhibit IAP to induce apoptosis. With LPS/BV-6 stimulation, HOIP-deficient DCs exhibited decreased cell survival and enhanced Caspase-8 and Caspase-3 cleavage (Fig. 4A, 4C), suggesting that HOIP is involved in the negative regulation of apoptosis induced by LPS/BV-6. In addition to apoptosis, TLRs can activate necroptosis when Caspase-8 is inhibited by z-VAD (28, 34). Therefore, we used the LPS plus z-VAD to induce necroptosis. Remarkably, LPS/z-VAD induced more necroptosis in HOIP-deficient DCs (Fig. 4A, 4D), indicating that HOIP is required for prevention of necroptosis via TLR4. Interestingly, LPS-induced apoptosis and necroptosis were only partially rescued by the RIPK1 kinase inhibitor Necrostatin-1, implying that RIPK1 kinase-independent mechanism exists under LPS-induced cell death (Fig. 4C, 4D).
Because above data indicate that HOIP-deficient DCs were more sensitive to LPS-induced cell death, we hypothesized that reduced DCs in HoipDC KO mice could be the consequences of cell death induced directly by TLR ligands presented by commensal microbiota. To address the specific role of commensal microbiota in the development of inflammation in HoipDC KO mice, we depleted most of the commensal microbiota by treating 2-d-old mice with a broad spectrum of antibiotics for 7 wk. Following antibiotic treatment, HoipDC KO mice had reduced enlargement of spleens and mLNs (Fig. 4E). Furthermore, antibiotic treatment reduced the percentage and number of inflammatory monocytes and neutrophils in HoipDC KO mice (Fig. 4F, 4G). The reduction of DCs in HoipDC KO mice was partially recovered with depletion of commensal microbiota (Fig. 4H, 4I). Therefore, the systemic inflammation observed in untreated HoipDC KO mice was partially rescued in antibiotic-treated mice, suggesting the contribution of intestinal microbiota to the systemic inflammation in HoipDC KO mice.
DC death leads to systemic inflammation in HoipDC KO mice
To determine whether aberrant DC cell death is responsible for the systemic inflammation in HoipDC KO mice, we crossed HoipDC KO mice to Mlkl−/− mice to generate HoipDC KO Mlkl−/− mice, which would directly address the specific role of HOIP on MLKL-mediated necroptosis in DCs. We found that genetic ablation of MLKL alone failed to prevent systemic inflammation in HoipDC KO mice. Deletion of MLKL alone had little effect on inflammation of spleen and mLN, with HoipDC KO Mlkl−/− mice exhibiting splenomegaly and lymphadenopathy (Supplemental Fig. 4A, 4B). Besides, inflammatory cell infiltration of spleens and mLNs observed in HoipDC KO Mlkl−/− mice were comparable to HoipDC KO mice (Supplemental Fig. 4C, 4D).
Because Caspase-8–deficient mice are embryonically lethal because of sensitization to RIPK3- and MLKL-induced necroptosis (35–38), it is not possible to generate viable Casp8−/− HoipDC KO mice to specifically address the role of apoptosis in HoipDC KO mice. We crossed HoipDC KO Mlkl−/− to Casp8+/−mice to generate HoipDC KO Casp8−/− Mlkl−/− mice to concurrently block both the Caspase-8–mediated apoptosis and MLKL-mediated necroptosis in HoipDC KO mice. Remarkably, although HoipDC KO Casp8−/− Mlkl−/− mice developed splenomegaly and lymphadenopathy like Casp8−/− Mlkl−/− mice as previously reported (39), the systemic inflammation associated with DC-HOIP deficiency was significantly alleviated (Supplemental Fig. 4E–G). Reduced numbers of inflammatory cell populations correlated with decreased level of inflammatory cytokine TNF-α in the sera of HoipDC KO Casp8−/− Mlkl−/− mice (Supplemental Fig. 4H). Because deletion of Caspase-8 and MLKL are assumed to prevent both apoptosis and necroptosis, we expected that cDC numbers would be rescued in HoipDC KO Casp8−/− Mlkl−/− mice. Indeed, decreased numbers of cDCs in HoipDC KO mice were reversed in the spleens of HoipDC KO Casp8−/− Mlkl−/− mice (Supplemental Fig. 4I), indicating DC death results in the systemic inflammation in HoipDC KO mice.
Because systemic deletion of Caspase-8 and MLKL may alleviate systemic inflammation of HoipDC KO mice by suppressing cell death of cells other than DCs, including both hematopoietic and nonhematopoietic cells, we further investigate whether Caspase-8 and MLKL in hematopoietic cells mediate systemic inflammation in HoipDC KO mice. We transferred bone marrow cells from CD45.2+ control mice, CD45.2+ HoipDC KO mice, CD45.2+ Casp8−/− Mlkl−/− mice, and CD45.2+ HoipDC KO Casp8−/− Mlkl−/− mice into irradiated CD45.1+ WT recipient mice. In contrast to mice transferred with bone marrow cells from HoipDC KO mice, the inflammation in mice transferred with bone marrow cells from HoipDC KO Casp8−/− Mlkl−/− mice was significantly alleviated, as exhibited by decreased infiltration of neutrophils and inflammatory monocytes in mice (Fig. 5B–E). Furthermore, the reduction of DCs in mice transferred with bone marrow cells from HoipDC KO mice was reversed in mice transferred with bone marrow cells from HoipDC KO Casp8−/− Mlkl−/− mice (Fig. 5F, 5G). Thus, aberrant apoptosis and necroptosis lead to systemic inflammation in HoipDC KO mice.
MyD88-dependent signaling is crucial for systemic inflammation in HoipDC KO mice
Cell death is predicted to result in the release damage-associated molecular patterns (DAMPs) and cytokines that are capable of activating other innate cells, resulting in inflammation (40). MyD88 is an essential adaptor molecule for TLRs and IL-1R, and activation of MyD88-dependent TLR signaling elicits the production of several proinflammatory cytokines, including TNF-α, IL-1, and IL-6 (41, 42). Besides, MyD88 signaling can also be activated by endogenous DAMPs (e.g., IL-1, IL-18) (43, 44). To investigate the involvement of MyD88 signaling in systemic inflammation, we crossed HoipDC KO mice to Myd88−/−mice to generate HoipDC KO Myd88−/− mice. Interestingly, HoipDC KO Myd88−/− mice almost completely rescued the HOIP deficiency–associated enlargement of spleen and mLN (Fig. 6A). Furthermore, the inflammatory monocytes and neutrophils in the HoipDC KO Myd88−/− mice also reduced to similar cell numbers as control mice (Fig. 6B). Accordingly, the elevated levels of TNF-α in HoipDC KO mice were reduced in HoipDC KO Myd88−/− mice (Fig. 6C). However, analysis of DCs in HoipDC KO Myd88−/− mice revealed that the numbers of DCs were still significantly decreased compared with those in control and Myd88−/− mice (Fig. 6D). Consistent with decreased DCs in HoipDC KO Myd88−/− mice, BMDCs from HoipDC KO Myd88−/− mice are still susceptible to either TNF-α– or LPS-induced cell death (Fig. 6E). These results demonstrate that MyD88 is not responsible for the DC reduction because DCs still die in the absence of MyD88. However, MyD88 is required for the development of inflammation observed in HoipDC KO mice.
Because MyD88 deficiency is not responsible for DC reduction in HoipDC KO mice, we further assessed IL-1α in HoipDC KO mice and HoipDC KO Myd88−/− mice considering the essential function of MyD88 in IL-1R signal. Interestingly, an increase in basal serum level of IL-1α was observed in HoipDC KO mice (Fig. 6F). Furthermore, LPS challenge could significantly enhance IL-1α serum level, which could be abrogated by MyD88 deficiency (Fig. 6F). Consistently, increased secretion of IL-1α and IL-1β was detected in BMDCs from HoipDC KO mice upon LPS challenge, which was diminished with MyD88 deficiency (Fig. 6G). Further analysis of mRNA of Il1a and Il1b revealed that the enhanced secretion of IL-1α and IL-1β was not due to an enhanced transcription in HoipDC KO cells (Fig. 6H), and enhanced cleavage of procaspase-1 and pro–IL-1β was detected in BMDC upon LPS challenge (Fig. 6I), indicating a negative regulation of IL-1α/IL-1β secretion by HOIP. Together, these results indicate that MyD88-dependent signaling plays crucial roles in the development of systemic inflammation in HoipDC KO mice.
Discussion
Investigating the mechanism underpinning the autoinflammation is critical to understand the pathogenesis of patients with loss-of-function mutations in LUBAC components. Our study evaluated the impact of LUBAC deficiency on DC homeostasis by genetically ablating HOIP, the catalytically active component of LUBAC, in mouse DCs. Intriguingly, LUBAC deficiency in DCs results in the spontaneous development of systemic inflammation resembled to inflammation in patients. Additionally, this elevated systemic inflammation of HoipDC KO mice was distinct from other tissue-specific HOIP deficiency mice that exhibited no inflammation or localized inflammation (7, 8, 17, 25, 45, 46). In contrast to mice with T cell–specific or B cell–specific HOIP deficiency, both mice exhibit no obvious systemic inflammation; HoipDC KO mice spontaneously develop systemic inflammation at the age of 6–8 wk. Thus, LUBAC in DCs is indispensable for maintaining homeostasis and prevention of autoinflammation.
Previous studies showed that cell death can be the cause of inflammation and inflammation-associated diseases (47), and the absence of HOIP in other cell types caused marked sensitivity to cell death (8, 25, 45, 46). The results presented in this article provide additional evidence in support of cell death as an etiology of inflammation-associated diseases. Our study found that HOIP-deficient DCs were more susceptible to both apoptotic and necroptotic cell death in response to TNF-α or LPS stimulation.
Thus, the observed autoinflammatory phenotype in HoipDC KO mice presumably results from elevated levels of cell death, especially necroptosis (47, 48), upon LUBAC deficiency. In mice with HOIP deficiency, concomitant blocking of both the Caspase-8–mediated apoptosis and MLKL-mediated necroptosis by systemic deletion of both Caspase-8 and MLKL prevents cell death in Hoip−/− embryos and embryonic lethality, resulting in viable Hoip−/− Casp8−/− Mlkl−/− mice (11). In mice with keratinocyte-specific HOIP deficiency, systemic deletion of both Caspase-8 and MLKL could prevent cell death–associated dermatitis and postnatal lethality (46). Interestingly, our study found that concomitant blocking of both the Caspase-8–mediated apoptosis and MLKL-mediated necroptosis alleviates the systemic inflammation associated with a DC-HOIP deficiency, and DC numbers in HoipDC KO mice were reversed to the amount of WT mice by codeletion of Caspsase-8 and MLKL, suggesting that cell death of DCs is responsible for the inflammatory phenotype of HoipDC KO mice. Because various cell types from Casp8−/− Mlkl−/− mice were resistant to diverse necroptotic cell death stimuli, and Casp8−/− Mlkl−/− mice rapidly developed splenomegaly, lymphadenopathy, and autoimmune manifestations, bone marrow chimeric mice were employed to specifically delete Caspase-8 and MLKL in hematopoietic cells to confirm that preventing cell death in a hematopoietic system can rescue the inflammation in HoipDC KO mice. In the future, it would be better to delete Caspase-8 and MLKL in a DC-specific manner by using Itgax-Cre Hoipflox/flox Casp8flox/flox Mlklflox/flox to fully substantiate the involvement of DC cell death in systemic inflammation of HoipDC KO mice.
Apart from Caspase-8 and MLKL, a recent study reported that LUBAC interacts with Caspase-1 to generate linear ubiquitination on the CARD domain of Caspase-1 to regulate its function, and depletion of HOIP or Sharpin resulted in heightened Caspase-1 activation, IL-1β secretion, and pyroptosis (49, 50). Our study found that enhanced IL-1α and IL-1β secretion were detected in HoipDC KO cells upon LPS challenge, indicating the involvement of Caspase-1 in the regulation of cell death of HoipDC KO cells in response to LPS. Thus, it would be worth trying to explore the role of Caspase-1 in HoipDC KO cell death and following inflammation in HoipDC KO mice. Because simultaneous Ripk3 deletion and Casp8 heterozygosity suppressed dermatitis of Sharpincpdm/cpdm mice (51), and genetic ablation of Caspase-1 from Sharpincpdm/cpdm mice significantly reduced skin inflammation (52), concomitant prevention of pyroptosis, apoptosis, and necroptosis (PANoptosis) is likely to completely rescue inflammation in HoipDC KO mice.
Intriguingly, although both TNF-α and TLR4 stimulation can induce the cell death of HOIP-deficient DCs in vitro, the absence of TNFR1 signaling and TLR4 stimulation in vivo led to distinct results. Unexpectedly, unlike observations in keratinocyte-specific HOIP deficiency mice (25, 46) and HOIP-deficient embryos(10), the systemic inflammation in HoipDC KO mice could not be rescued by depleting TNFR1. One possibility is that TNFR1 signaling from non-DCs might also be involved in the development of inflammation in HoipDC KO mice. Therefore, it would be interesting to investigate the role of DC-specific TNFR1 signaling in aberrant cell death and inflammation driven by HOIP deficiency in DCs. Of note, although RIPK1-deficient DCs undergo TNF-α–mediated necroptosis in vitro, the absence of TNFR1 had no detectable effects on splenic inflammation in Ripk1DC KO mice (53). Contrary to other findings that TNF-α promotes inflammation, however, our study shows that TNFR1 deficiency exacerbates the systemic inflammation of HoipDC KO mice as they developed symptoms of cachexia. Thus, the detailed molecular mechanism of TNFR1 in DC death and following inflammation needs to be further investigated.
Unlike TNFR1 signaling, TLR ligands from commensal microbiota are likely the source of signals to induce the cell death of HOIP-deficient DCs and inflammation of HoipDC KO mice, and depletion of microbiota by administration of broad-spectrum antibiotics partly abrogated the inflammation in HoipDC KO mice. In HOIP-deficient DCs, TLR4 stimulation can induce both enhanced apoptosis and necroptosis. Thus, the explanation for the involvement of intestinal microbiota in the pathogenesis of HoipDC KO mice could be that TLR ligands directly induce the cell death of HOIP-deficient DCs and consequently release DAMPs to induce inflammation. Of note, although a study by Sharma et al. (54) has demonstrated that depletion of gut microbiota by administration of antibiotics partially rescued skin inflammation and systemic inflammation of Sharpincpdm/cpdm mice, the detailed mechanism is not clear. Our results provide additional evidence in support of the role of commensal microbiota in the pathogenesis of inflammation associated with LUBAC component mutation and a possible mechanism for the regulation of TLR signal by LUBAC in DCs to prevent cell death and inflammation.
MyD88 is an essential adaptor for both TLRs and IL-1R, and previous studies have shown that MyD88 is involved in inflammation triggered by necroptotic cells (22, 55). Our data show that the inflammatory phenotype in HoipDC KO mice was absent in HoipDC KO Myd88−/− mice. However, the absence of MyD88 did not rescue the death of DCs in HoipDC KO mice as shown by decreased splenic DCs and susceptible death of BMDCs generated from HoipDC KO Myd88−/− mice. Thus, the abrogation of inflammation by MyD88 is not through prevention of DC cell death directly but relies on other indirect mechanisms.
Considering MyD88 mediates signal transduction for both most TLRs and IL-1R, the absence of inflammatory responses in the HoipDC KO Myd88−/− mice may be the outcome of defective TLR signaling or IL-1R signaling. Because administration of broad-spectrum antibiotics partially rescued the inflammation of HoipDC KO mice and MyD88 deficiency abrogated the secretion of IL-1α/IL-1β induced by LPS, one explanation for the abrogation of inflammation in the HoipDC KO Myd88−/− mice could be defective TLR signaling. Apart from TLR signaling, IL-1 signaling has been reported to be responsible for the inflammation induced by necrotic cells; thus mice deficiency of MyD88 failed to recruit neutrophils in response to dead cells (55). In our study, deficiency of MyD88 may abrogate the IL-1 signaling induced by cell death, contributing to the prevention of inflammation in HoipDC KO mice. In the case of HoipDC KO mice, we suggest that commensal microbes initiate innate immune signaling via the TLR/MyD88 axis, which leads to IL-1 production. Besides, TLR ligands from microbes can also induce cell death of DCs to release DAMPs like IL-1α. All above signaling via the IL-1R/MyD88 axis results in the development of chronic inflammation. Considering the essential function of MyD88 for both TLR4 and IL-1R, separately investigating the role of TLR4 deletion and IL-1R deletion in inflammation of HoipDC KO mice would be worth trying in the future.
It would therefore be tempting to speculate that HOIP deficiency in the DCs could mimic the conditions in the patients with loss-of-function mutations of LUBAC components and could possibly serve as a model for its etiology. The pathologies found in the HoipDC KO mice appear to be driven by aberrant cell death, which is often seen in LUBAC-mutated patients. However, blocking of TNFR1 signaling by anti-TNF treatment in LUBAC-mutated patients may not be as effective as several other autoinflammatory and autoimmune disorders, including rheumatoid arthritis and psoriasis (56, 57), as deficiency of TNFR1 signaling could not alleviate disease development. Our findings that commensal microbiota and MyD88-dependent signaling are required for development of inflammation in HoipDC KO mice suggest that modulation of microbiota may serve as a therapeutic method to attenuate the clinical autoinflammatory diseases because of mutations in the LUBAC complex. In conclusion, our findings suggest that LUBAC is essential for regulating DC survival, that disturbance of this regulation results in the development of systemic inflammation resembling autoinflammation in LUBAC-mutated patients, and that modulation of commensal microbiota may serve as a therapeutic method for these patients.
Acknowledgements
We thank Drs. Xiaodong Wang and Xiaoyu Hu for the generous sharing of Mlkl−/−mice and Myd88−/−mice, respectively.
Footnotes
This work was partially supported by the National Key Research and Development Program of China (Grant 2019YFA0508502 to X.L.), the National Natural Science Foundation of China (Grants 31930039, 81630058, 91942303, and 31821003 [to X.L.] and 81971469 and 31670904 [to X.Z.]), and annual funding from the Tsinghua-Peking Center for Life Sciences.
X.W., X.Z., and X.L. designed the study and wrote the manuscript. X.W. performed the experiments and analyzed the data. Y.T. provided mice and intellectual input. S.Z. helped to perform FACS for mice phenotype analysis. X.Z. provided reagents and gave intellectual advice.
The online version of this article contains supplemental material.
Abbreviations used in this article
- BMDC
bone marrow–derived DC
- cDC
conventional DC
- CDP
common DC progenitor
- DAMP
damage-associated molecular pattern
- DC
dendritic cell
- HOIL-1
heme-oxidized IRP2 ubiquitin ligase 1
- HOIP
HOIL-1–interacting protein
- LUBAC
linear ubiquitin chain assembly complex
- MHC II+
MHC class II+
- mLN
mesenteric lymph node
- RDA
RIPK1-dependent apoptosis
- RIA
RIPK1-independent apoptosis
- SHARPIN
Sharpin, Shank-associated RH domain-interacting protein
- Smac
second mitochondrial–derived activator of caspase
- WT
wild-type
References
Disclosures
The authors have no financial conflicts of interest.