Fas, a member of the death receptor family, plays a central role in initiating cell death, a biological process crucial for immune homeostasis. However, the immunological and pathophysiological impacts to which enhanced Fas signaling gives rise remain to be fully understood. Here we demonstrate that TGF-β–activated kinase 1 (TAK1) works as a negative regulator of Fas signaling in macrophages. Upon Fas engagement with high concentrations of FasL, mouse primary macrophages underwent cell death, and, surprisingly, Fas stimulation led to proteolytic cleavage of gasdermin (GSDM) family members GSDMD and GSDME, a hallmark of pyroptosis, in a manner dependent on caspase enzymatic activity. Remarkably, TAK1-deficient macrophages were highly sensitive to even low concentrations of FasL. Mechanistically, TAK1 negatively modulated RIPK1 kinase activity to protect macrophages from excessive cell death. Intriguingly, mice deficient for TAK1 in macrophages (TAK1mKO mice) spontaneously developed tissue inflammation, and, more important, the emergence of inflammatory disease symptoms was markedly diminished in TAK1mKO mice harboring a catalytically inactive RIPK1. Taken together, these findings not only revealed an unappreciated role of TAK1 in Fas-induced macrophage death but provided insight into the possibility of perturbation of immune homeostasis driven by aberrant cell death.

This article is featured in Top Reads, p.

The TNF receptor (TNFR) superfamily forms a large group of cytokine receptors, all of which contribute to exertion of diverse biological functions, and, for this reason, some of them now serve as biomarkers and therapeutic targets (1, 2). Among them, the death receptor family is well known to induce programmed cell death, essential for embryogenesis and adult organ homeostasis. Fas (also known as CD95), the best characterized member of the death receptor family, plays a central role in immune homeostasis. A molecular mechanism by which Fas signaling induces apoptosis, a form of programmed cell death, has been well described (3). Engagement of Fas with FasL, a specific ligand for Fas, facilitates assembly of a complex of proteins, termed the “death-inducing signaling complex,” at the cytoplasmic tail of Fas (4). The Fas death-inducing signaling complex is composed of different death domain– and death effector domain–containing proteins: Fas, Fas-associated death domain, caspase-8, and c-FLIP. Its formation leads to autoproteolytic processing of caspase-8, and then the ensuing active caspase-8 dimerization enables initiation of apoptosis via processing of target proteins, including caspase-3 (4, 5). Now it is widely appreciated that failure of Fas-induced cell death causes serious immune dysregulation. In fact, humans with genetic mutations in Fas, FasL, caspase-8, and caspase-10 develop autoimmune lymphoproliferative syndrome, which is mainly characterized by accumulation of TCRαβ+CD3+CD4CD8 T cells, lymphadenopathy, splenomegaly, and autoimmunity, and mice with naturally occurring mutations in Fas (lpr mice) and FasL (gld mice) also show analogous phenotypes as in the case of human patients (6, 7). However, it has hitherto remained unknown whether a large excess of Fas-induced cell death gives rise to the immunological and pathophysiological impacts.

TGF-β–activated kinase 1 (TAK1), a member of the MAPK kinase kinase family encoded by Map3k7, participates in the various immunoreceptor-mediated signal transductions. It mainly functions as an important signaling factor to activate the MAPK and inhibitor of NF-κB kinase/NF-κB pathways, both of which are important to evoke innate and adaptive immune responses (811). We and other groups previously demonstrated that macrophages deficient in TAK1 or macrophages subjected to pharmacological inhibition of TAK1 underwent massive cell death upon TNFR1 or TLR stimulation, and, remarkably, these works highlighted the presence of novel cell death machinery dependent on the gasdermin family, executioners of pyroptosis, operating in downstream pathways of caspase-8 (1216). The findings not only identified a novel aspect of TAK1 function to restrain the appearance of such proinflammatory cell death but allowed us to consider the following two possibilities. The first is whether Fas signaling in macrophages triggers caspase-8–dependent cell death, followed by inflammation, and the second is whether and how TAK1 is associated with the Fas signaling pathway.

Here we show that TAK1 potentially functions as a negative regulator of Fas signaling in macrophages. Mouse primary macrophages underwent cell death only when engaged with high concentrations of FasL and displayed proteolytic cleavage of gasdermin D (GSDMD) and GSDME, members of the gasdermin family, in a manner dependent on caspase enzymatic activity. Remarkably, TAK1-deficient macrophages exhibited high sensitivity to even low concentrations of FasL, which is attributed to increased kinase activity of receptor-interacting serine/threonine-protein kinase 1 (RIPK1). Surprisingly, mice deficient in TAK1 in macrophages spontaneously developed tissue inflammation, indicative of autoinflammation, and, importantly, the symptom was markedly diminished by crossing to mice harboring a catalytically inactive RIPK1. These in vivo data also provided novel insight into the possibility of perturbation of immune homeostasis driven by aberrant cell death.

Map3k7flox/flox or Map3k7flox/–, Lyz2Cre/+, Tnf−/−, Ripk1D138N/D138N, and Ticam1−/− mice, all of which are on a C57BL/6 background, have been described previously (8, 15, 17). All the mice were maintained at the Shinshu University animal facility under specific pathogen-free (SPF) conditions. The animal protocol for this research was approved by the committee for animal experiments of Shinshu University.

Generation of mouse bone marrow–derived macrophages (BMDMs) was performed according to a protocol described previously (15). The BMDMs were cultured overnight, and the culture medium was replaced with Opti-MEM (Thermo Fisher), followed by treatment with the indicated cytokines. For the cell death assay, BMDMs were seeded at 5 × 104 cells per well in 96-well plates and were treated with FasL (MegaFasL, Adipogen). Six hours after stimulation, the proportion of dead cells was assessed by measuring lactate dehydrogenase (LDH) in culture supernatants using the Cytotoxicity LDH Assay Kit-WST (Dojindo) according to the manufacturer’s instructions. For immunoblot analysis, BMDMs were seeded at 1–1.5 × 106 cells per well in 6-well plates and were treated with FasL or TNF-α (PeproTech) in the absence or presence of 50 μM Z-VAD-FMK (Selleck). To prepare cell lysates, BMDMs were extensively washed with ice-cold 1× PBS and were lysed with TNE-NS lysis buffer (20 mM Tris-Cl, pH 7.5, 150 mM NaCl, 5 mM EDTA, 1% Nonidet P-40, and 0.1% SDS) supplemented with a protease inhibitor mixture (Roche) and a phosphatase inhibitor mixture (nacalai tesque). For detection of phosphorylated mixed lineage kinase domain-like protein (MLKL), BMDMs were lysed with 1× SDS sample buffer, and cell lysates after sonication were boiled. Proteins in cell culture supernatants were precipitated with methanol/chloroform for concentration and were suspended in 1× SDS sample buffer. Crude proteins were subjected to immunoblotting using specific Abs to detect target proteins. Specific proteins were visualized with Immobilon Chemiluminescent HRP Substrate (Millipore). The Abs used in this study are as follows: p-RIPK1(Ser166) (catalog no. 31122, Cell Signaling Technology), cleaved caspase-8 (Asp387) (catalog no. 9429, Cell Signaling Technology), c-FLIP (catalog no. 56343, Cell Signaling Technology), caspase-3 (catalog no. 9662, Cell Signaling Technology), poly(ADP-ribose) polymerase (PARP; catalog no. 9542, Cell Signaling Technology), TAK1 (catalog no. 5206, Cell Signaling Technology), cleaved caspase-1 (Asp296) (catalog no. 89332, Cell Signaling Technology), p-MLKL (Ser345) (catalog no. 37333, Cell Signaling Technology), p-IκBα (Ser32/36) (catalog no. 9246, Cell Signaling Technology), phosphorylated stress-activated protein kinase/JNK (Thr183/Tyr185) (catalog no. 9251, Cell Signaling Technology), stress-activated protein kinase/JNK (catalog no. 9252, Cell Signaling Technology), p-p38 (Thr180/Tyr182) (catalog no. 9211, Cell Signaling Technology), IκBα (sc-371, Santa Cruz Biotechnology), p38α (sc-81621, Santa Cruz), GSDMD (ab209845, Abcam), GSDME (ab215191, Abcam), RIPK1 (catalog no. 610459, BD Biosciences), caspase-8 (cat ALX-804-447-C100, Enzo), MLKL (SAB1302339, Sigma-Aldrich), high mobility group box 1 (HMGB1; catalog no. 651402, BioLegend), and GAPDH (catalog no. M171-3, MBL).

Six- to 10-wk-old mice were used to isolate cells from each tissue. To collect cells in the peritoneal cavity, mice were injected and extensively washed with 5 ml of 1× PBS/2% FBS in the cavity. Peritoneal cells were sampled and used for experiments. To collect cells from the liver, lung, and visceral adipose tissue, the isolated tissues were chopped into little pieces with scalpels and were enzymatically digested for 30 min at 37°C with gentle shaking in RPMI 1640 supplemented with 0.5 mg/ml collagenase IV (Sigma-Aldrich) and 50 U/ml DNase I (Wako). Preparation of colonic lamina propria cells were performed as described previously (18). Briefly, the isolated colon was cut and opened longitudinally. After extensively washing out luminal contents with 1× PBS, the tissue was incubated for 30 min at 37°C with vigorous shaking in 1× PBS (calcium and magnesium free) supplemented with 5 mM EDTA and 2 mM DTT to remove the epithelial cell layer. The remaining colon was chopped into little pieces with scalpels, and these pieces were enzymatically digested for 30 min at 37°C with gentle shaking in RPMI 1640 supplemented with 2% FBS, 0.5 mg/ml collagenase IV, and 50 U/ml DNase I. Discrete cells from each tissue after the treatment were extensively washed with 1× PBS and suspended in 1× PBS/2% FBS, then stored at 4°C.

Single-cell suspensions prepared from each tissue were used for Ab staining. The following fluorochrome-conjugated Abs were used in this study: CD11b (M1/70), CD11c (N418), CD64 (X54-5/7.1), CD301a (LOM-8.7), CX3CR1 (SA011F11), I-A/I-E (MHC class II [MHC II], M5/114.15.2), Ly6C (HK1.4), Ly6G (1A8), F4/80 (BM8), and Tim4 (RMT4-54), all from BioLegend; Siglec-F (E50-24400) and CD16/32 (2.4G2) from BD Biosciences; and CD45 (30-F11) from eBioscience. Propidium iodide (Sigma-Aldrich) was used for dead cell staining. Stained cells were assessed by flow cytometry using the FACSCelesta device (BD Biosciences), and the data were analyzed with Kaluza software (Beckman Coulter).

For histopathological examinations, the livers from 6- to 10-wk-old mice were fixed by immersion in 10% buffered formalin and embedded in paraffin. Tissue sections with 4-μm thickness were stained with H&E. For serological analysis, blood was sampled from the retro-orbital sinus. Sera were collected by centrifugation and were assessed by measurement of aspartate aminotransferase, alanine aminotransferase, and LDH. As criteria for peritonitis, we evaluated the disappearance of peritoneal macrophages, the infiltration of inflammatory cells (neutrophils and monocytes), and MHC II expression in large peritoneal macrophages.

An unpaired two-tailed t test was used for evaluation of statistical significance. The p Values <0.05 were considered significant. Statistical analyses were performed using GraphPad Prism software.

We previously demonstrated that TAK1-deficient macrophages evoked cell death in response to TNF-α and TLR ligands, indicating that TAK1 is necessary for the maintenance of macrophage survival in TNFR1 and TLR signaling (15). However, a lot of research has demonstrated that Fas signaling can induce cell death in various types of cells, even in the presence of TAK1. As expected, ligation of Fas with recombinant hexameric FasL (19) induced cell death in C57BL/6-derived mouse BMDMs, but, curiously, higher concentrations of FasL were required to achieve this despite its potent cytotoxic activity (Fig. 1A). In addition, immunoblot analysis showed Fas-induced proteolytic cleavage of caspase-8, caspase-3, and PARP in mouse BMDMs, all of which play a critical role in inducing apoptosis. Interestingly, a minor but detectable level of Ser166 phosphorylation of RIPK1, an autophosphorylation site essential for modulation of its kinase activity (20, 21), was also observed (Fig. 1B). Surprisingly, upon ligation with FasL, proteolytic cleavage of the gasdermin family members GSDMD and GSDME was detected, which was almost completely blocked by pretreatment with Z-VAD-FMK, a pan-caspase inhibitor (Fig. 1B, 1C). This means that the Fas-induced gasdermin family processing takes place in a caspase enzymatic activity–dependent manner. Given that these unique characteristics resemble the former reports (1216) and gasdermin processing is a well-defined feature for the induction of pyroptosis (22), our data demonstrate that Fas signaling possesses an ability to trigger pyroptosis in mouse macrophages, even though the strength of the Fas signal seems usually to be suppressed.

FIGURE 1.

Fas stimulation triggers caspase-dependent proteolytic cleavage of the gasdermin family in mouse BMDMs. (A) Cell death assay of mouse BMDMs. The BMDMs from wild-type C57BL/6 mice were left untreated or treated with the indicated concentrations of FasL for 6 h. The graph depicts the percentage of LDH release in the culture supernatant as a cell death index. Data were derived from two independent experiments. Bars are shown as mean values. (B and C) Immunoblot analysis. Wild-type BMDMs were left untreated or treated with FasL (200 ng/ml) for 2 h. Especially, in (C), wild-type BMDMs were left untreated or pretreated with Z-VAD-FMK (Z) for 1 h, followed by addition of FasL. GAPDH was employed as a loading control for cell lysates. Data are representative of three independent experiments. WCL, whole-cell lysates.

FIGURE 1.

Fas stimulation triggers caspase-dependent proteolytic cleavage of the gasdermin family in mouse BMDMs. (A) Cell death assay of mouse BMDMs. The BMDMs from wild-type C57BL/6 mice were left untreated or treated with the indicated concentrations of FasL for 6 h. The graph depicts the percentage of LDH release in the culture supernatant as a cell death index. Data were derived from two independent experiments. Bars are shown as mean values. (B and C) Immunoblot analysis. Wild-type BMDMs were left untreated or treated with FasL (200 ng/ml) for 2 h. Especially, in (C), wild-type BMDMs were left untreated or pretreated with Z-VAD-FMK (Z) for 1 h, followed by addition of FasL. GAPDH was employed as a loading control for cell lysates. Data are representative of three independent experiments. WCL, whole-cell lysates.

Close modal

Because it is unknown whether and how TAK1 is associated with the Fas-mediated signaling pathway, we sought to explore a potential role of TAK1 in Fas signaling. To this end, we used BMDMs from mice with myeloid-specific deletion of TAK1 on a TNF-α–deficient background (Lyz2Cre/+Map3k7flox/floxTnf−/− and Lyz2Cre/+Map3k7flox/–Tnf−/− mice, referred to hereafter as TAK1mKO mice) and Map3k7flox/floxTnf−/− mice as control mice (referred to hereafter as TAK1WT mice) (15). Surprisingly, TAK1-deficient BMDMs from TAK1mKO mice exhibited high sensitivity to even low concentrations of FasL compared with control BMDMs from TAK1WT mice (Fig. 2A). Immunoblot analysis also demonstrated that TAK1-deficient BMDMs possessed increased Fas signaling strength, as illustrated by enhanced processing of caspase-8, GSDMD, and GSDME (Fig. 2B). Furthermore, culture supernatants in the FasL-treated TAK1-deficient BMDMs included intracellular proteins such as caspase-3, caspase-1, and HMGB1, a well-characterized inflammatory mediator belonging to damage-associated molecular pattern molecules (23) (Fig. 2C). Fas-mediated activation of the RIPK1/RIPK3/MLKL signaling pathway, a hallmark of programmed necrosis, termed “necroptosis” (24), was detectable in TAK1-deficient BMDMs only when in the presence of Z-VAD-FMK (Fig. 2D). Taken together, our data indicate a novel negative regulatory role of TAK1 to limit Fas-induced macrophage death.

FIGURE 2.

Enhanced susceptibility to Fas-induced death in TAK1-deficient macrophages. (A) Cell death assay analogous to (Fig. 1. The BMDMs were left untreated or treated with the indicated concentrations of FasL for 6 h. The graph represents mean ± SEM from three independent experiments (n = 3 in each mouse group). (BD) Immunoblot analysis. In (B) and (C), the BMDMs were left untreated or treated with FasL [20 ng/ml in (B) and 20 and 50 ng/ml in (C)] for 2 h (B) or 4 h (C). In (D), TAK1-deficient BMDMs were left untreated or pretreated with Z-VAD-FMK (Z) for 1 h, followed by addition of FasL (F; 50 ng/ml) or TNF-α (T; 10 ng/ml). GAPDH was employed as a loading control for cell lysates. Data are representative of three independent experiments. **p < 0.01 and ***p < 0.001 (unpaired two-tailed t tests). SN, supernatants; WCL, whole-cell lysates.

FIGURE 2.

Enhanced susceptibility to Fas-induced death in TAK1-deficient macrophages. (A) Cell death assay analogous to (Fig. 1. The BMDMs were left untreated or treated with the indicated concentrations of FasL for 6 h. The graph represents mean ± SEM from three independent experiments (n = 3 in each mouse group). (BD) Immunoblot analysis. In (B) and (C), the BMDMs were left untreated or treated with FasL [20 ng/ml in (B) and 20 and 50 ng/ml in (C)] for 2 h (B) or 4 h (C). In (D), TAK1-deficient BMDMs were left untreated or pretreated with Z-VAD-FMK (Z) for 1 h, followed by addition of FasL (F; 50 ng/ml) or TNF-α (T; 10 ng/ml). GAPDH was employed as a loading control for cell lysates. Data are representative of three independent experiments. **p < 0.01 and ***p < 0.001 (unpaired two-tailed t tests). SN, supernatants; WCL, whole-cell lysates.

Close modal

We next sought to explore the molecular mechanism by which TAK1 negatively controls Fas-induced macrophage death. Through immunoblot-based screening, we discovered that ligation of Fas with a low concentration of FasL led to hyperphosphorylation of RIPK1 Ser166 in TAK1-deficient but not control BMDMs (Fig. 3A). Moreover, TAK1-deficient BMDMs showed accelerated protein reduction of full-length c-FLIP (c-FLIPL), even though the cleaved c-FLIP was detected equally between both types of macrophages (Fig. 3A). These data suggest that TAK1 somehow regulates the dynamics of both RIPK1 and c-FLIP in Fas signaling. To further address this issue, we used TAK1mKO mice harboring a catalytically inactive RIPK1 mutant (RIPK1D138N) (Lyz2Cre/+Map3k7flox/floxRipk1D138N/D138NTnf−/−, referred to hereafter as TAK1mKORIPK1D138N/D138N mice) (15, 17). Remarkably, TAK1-deficient BMDMs expressing the RIPK1D138N mutant showed resistance against cell death triggered by lower concentrations of FasL (Fig. 3B). Moreover, the presence of the RIPK1 mutant in TAK1-deficient macrophages effectively abrogated proteolytic cleavage of the effector molecules responding to Fas signaling and release of intracellular HMGB1 (Fig. 3C, 3D). Collectively, these data confirmed the role of TAK1 to target and antagonize RIPK1, whose kinase activity can affect the macrophage responsiveness to FasL.

FIGURE 3.

TAK1 restrains Fas-induced macrophage death by modulating RIPK1 kinase activity. (A, C, and D) Immunoblot analysis. The BMDMs were left untreated or treated with FasL [50 ng/ml in (A) and 20 ng/ml in (C) and (D)] for 2 h (C) or 4 h (D). GAPDH was employed as a loading control for cell lysates. Data are representative of three independent experiments. (B) Cell death assay analogous to (Fig. 1. The BMDMs were left untreated or treated with the indicated concentrations of FasL for 6 h. The graph represents mean ± SEM from four independent experiments (n = 4 in each mouse group). ***p < 0.001 and ****p < 0.0001 (unpaired two-tailed t tests). LE, long exposure; SE, short exposure; SN, supernatants; WCL, whole-cell lysates.

FIGURE 3.

TAK1 restrains Fas-induced macrophage death by modulating RIPK1 kinase activity. (A, C, and D) Immunoblot analysis. The BMDMs were left untreated or treated with FasL [50 ng/ml in (A) and 20 ng/ml in (C) and (D)] for 2 h (C) or 4 h (D). GAPDH was employed as a loading control for cell lysates. Data are representative of three independent experiments. (B) Cell death assay analogous to (Fig. 1. The BMDMs were left untreated or treated with the indicated concentrations of FasL for 6 h. The graph represents mean ± SEM from four independent experiments (n = 4 in each mouse group). ***p < 0.001 and ****p < 0.0001 (unpaired two-tailed t tests). LE, long exposure; SE, short exposure; SN, supernatants; WCL, whole-cell lysates.

Close modal

From further observation of TAK1mKO mice, we found that compared with TAK1WT mice, 44% of TAK1mKO mice among all we observed (22 out of 50 mice) were apparently ill from 6 wk after birth in the SPF environment, as characterized by growth retardation, ruffled fur, and lethargic behavior. These characteristic features of the mice strongly resembled those of mice with sepsis. Remarkably, flow cytometric analysis showed that peritoneal resident macrophages (defined as large peritoneal macrophages [LPMs]: CD11b+F4/80hi) (25) were conspicuously absent in apparently ill TAK1mKO mice (Fig. 4A, 4B). Alternatively, the CD11b+F4/80−/dimLy6C+ cell population was abnormally accumulated in the peritoneal cavities of the TAK1mKO mice (Fig. 4C) and was identified as a mixed population of monocytes and neutrophils from detailed analysis (Fig. 4C, 4D). These strongly resembled the symptoms of acute peritonitis, as exemplified by macrophage disappearance in parallel with the recruitment of neutrophils and monocytes in the peritoneal cavity (2527). Consistent with this phenotype, histological analysis identified the frequent presence of inflammatory infiltration in the livers of sick TAK1mKO mice (Fig. 4E4H). In addition, flow cytometric analysis revealed increased numbers of CD45+ leukocytes, a decrease in the percentage of tissue-resident macrophages, mainly Kupffer cells, and then accumulation of neutrophils and monocytes in the liver (Fig. 4I and Supplemental Fig. 1). Furthermore, serological analysis showed significantly elevated levels of aspartate aminotransferase, alanine aminotransferase, and LDH (Fig. 4J4L). Collectively, these data suggest that TAK1mKO mice developed hepatic injury. In addition to the peritoneal cavity and liver, a lower percentage of tissue-resident macrophages was observed in the lungs and visceral adipose tissues but not the colons of the sick TAK1mKO mice (Supplemental Fig. 2). Taken together, these data demonstrate that TAK1 in macrophages has an ability to protect mice from spontaneous tissue inflammation.

FIGURE 4.

Spontaneous tissue inflammation in TAK1mKO mice. (AD) Flow cytometric analysis of the leukocytes in the peritoneal cavities of TAK1WT and visibly ill TAK1mKO mice. (A and C) CD11b+/F4/80hi resident LPMs in CD45+ propidium iodide–negative (PI) cells (A) and Ly6C+/Ly6G monocytes and Ly6C+/Ly6G+ neutrophils in CD45+CD11b+PI cells (C). The plots are representative of five independent experiments. (B and D) The proportions of LPMs in CD45+PI cells and those of both monocytes and neutrophils in CD45+CD11b+PI cells (left). The absolute numbers of LPMs, monocytes, and neutrophils (right). Dots represent individual mice. Results are pooled from five independent experiments (n = 5 in each mouse group). (EH) H&E-stained sections of the livers of TAK1WT and visibly ill TAK1mKO mice. (E and G) TAK1WT mice. (F and H) TAK1mKO mice. In (E) and (F), images of the portal area are shown. In (G) and (H), images of the hepatic parenchyma are shown. Arrowheads in (H) represent microabscess. Scale bars, 100 μm in (E) and (F), 50 μm in (G) and (H). The images are representative of nine independent experiments (n = 9 in each mouse group). (I) Absolute numbers of CD45+ leukocytes in the whole livers of TAK1WT and visibly ill TAK1mKO mice were determined by flow cytometric analysis. Dots represent individual mice. Results are pooled from nine independent experiments (n = 9 for TAK1WT mice, and n = 10 for TAK1mKO mice). (JL) Serological analysis of TAK1WT and visibly ill TAK1mKO mice. Dots represent individual mice. Results are pooled from two independent experiments (n = 6 for TAK1WT mice, and n = 7 for TAK1mKO mice). All the graphs represent mean ± SEM. *p < 0.05, **p < 0.01, and ***p < 0.001 (unpaired two-tailed t tests).

FIGURE 4.

Spontaneous tissue inflammation in TAK1mKO mice. (AD) Flow cytometric analysis of the leukocytes in the peritoneal cavities of TAK1WT and visibly ill TAK1mKO mice. (A and C) CD11b+/F4/80hi resident LPMs in CD45+ propidium iodide–negative (PI) cells (A) and Ly6C+/Ly6G monocytes and Ly6C+/Ly6G+ neutrophils in CD45+CD11b+PI cells (C). The plots are representative of five independent experiments. (B and D) The proportions of LPMs in CD45+PI cells and those of both monocytes and neutrophils in CD45+CD11b+PI cells (left). The absolute numbers of LPMs, monocytes, and neutrophils (right). Dots represent individual mice. Results are pooled from five independent experiments (n = 5 in each mouse group). (EH) H&E-stained sections of the livers of TAK1WT and visibly ill TAK1mKO mice. (E and G) TAK1WT mice. (F and H) TAK1mKO mice. In (E) and (F), images of the portal area are shown. In (G) and (H), images of the hepatic parenchyma are shown. Arrowheads in (H) represent microabscess. Scale bars, 100 μm in (E) and (F), 50 μm in (G) and (H). The images are representative of nine independent experiments (n = 9 in each mouse group). (I) Absolute numbers of CD45+ leukocytes in the whole livers of TAK1WT and visibly ill TAK1mKO mice were determined by flow cytometric analysis. Dots represent individual mice. Results are pooled from nine independent experiments (n = 9 for TAK1WT mice, and n = 10 for TAK1mKO mice). (JL) Serological analysis of TAK1WT and visibly ill TAK1mKO mice. Dots represent individual mice. Results are pooled from two independent experiments (n = 6 for TAK1WT mice, and n = 7 for TAK1mKO mice). All the graphs represent mean ± SEM. *p < 0.05, **p < 0.01, and ***p < 0.001 (unpaired two-tailed t tests).

Close modal

Intriguingly, even in visibly healthy TAK1mKO mice, we have observed the complicated phenotypes in the peritoneal cavities: 32% of the mice (16 out of 50 mice) were normal, 34% of the mice (17 out of 50 mice) showed the accumulation of neutrophils and monocytes along with macrophage disappearance, and the remaining 34% of the mice (17 out of 50 mice) exhibited the presence of LPMs with increased expression of MHC II (Supplemental Fig. 3A, 3B). This implies that some event to induce the local inflammation naturally occurs in the peritoneal cavities of TAK1mKO mice. In addition, the TAK1mKO mice bearing MHC II+ LPMs tended to have infiltrating neutrophils in the peritoneal cavities (Supplemental Fig. 3A). As expected, neither MHC II+ LPMs nor infiltrating neutrophils was present in the TAK1WT mice we tested (Fig. 4C and Supplemental Fig. 3B). On the basis of these objective criteria, we concluded that TAK1mKO mice are prone to develop peritonitis spontaneously.

Given that special attention has been paid to a functional interplay between cell death and inflammation (28, 29), we hypothesized that the disappearance of tissue-resident macrophages in TAK1mKO mice may result from cell death. Having observed the effectiveness of a kinase-defective RIPK1 mutant to block Fas-induced aberrant cell death in TAK1-deficient macrophages, we thus sought to assess whether the onset of peritonitis in TAK1mKO mice was prevented by abrogating RIPK1 kinase activity. As mentioned above, inflammatory manifestations in the peritoneal cavity were evident in 68% of visibly healthy TAK1mKO mice, and, remarkably, the sign of the disease was substantially reduced in TAK1mKORIPK1D138N/D138N mice (Fig. 5). On the other hand, TIR Toll/IL-1R (TIR) domain-containing adapter inducing IFN-β (TRIF) deficiency in TAK1mKO mice (referred to as TAK1mKOTRIF−/− mice) (15) had only a minor impact on the relief of peritonitis compared with a catalytically inactive RIPK1 (Fig. 5). These data suggest that the emergence of spontaneous tissue inflammation in TAK1mKO mice is mainly attributed to the hyperactivation of RIPK1 in macrophages.

FIGURE 5.

The catalytically inactive RIPK1 blocks aseptic peritonitis in TAK1mKO mice. The cells sampled from the peritoneal cavities of visibly healthy mouse groups were analyzed by flow cytometry. The incidence of peritonitis in each animal group was evaluated on the basis of criteria for peritonitis, as represented by (Fig. 4A, 4C, and Supplemental Fig. 1, and was depicted by pie charts. Results are pooled from more than 20 independent experiments.

FIGURE 5.

The catalytically inactive RIPK1 blocks aseptic peritonitis in TAK1mKO mice. The cells sampled from the peritoneal cavities of visibly healthy mouse groups were analyzed by flow cytometry. The incidence of peritonitis in each animal group was evaluated on the basis of criteria for peritonitis, as represented by (Fig. 4A, 4C, and Supplemental Fig. 1, and was depicted by pie charts. Results are pooled from more than 20 independent experiments.

Close modal

A growing body of research has documented that the death receptor Fas induces apoptosis by facilitating proteolytic cleavage of caspase-8 and its downstream effector caspase-3 in various cell types (3). In the present study, we found that Fas stimulation promotes proteolytic cleavage of not only caspase-3 and PARP but also gasdermin family members GSDMD and GSDME in a caspase enzymatic activity–dependent mechanism. This result was surprising but was expected, to an extent, from previous reports (1216, 30). Our data argue that Fas-to-caspase-8 signaling urges macrophages to gasdermin-mediated pyroptosis, even though we do not rule out any possibility of caspase-8/caspase-3–dependent apoptosis. Another important view is that in normal mouse macrophages, potent Fas engagement is required to induce cell death. In this respect, a past report may give insight into the current observation (31). We speculate that specific mechanisms not to readily kill macrophages in response to death receptor stimulation are present under physiological conditions; otherwise, the induction of unwanted macrophage death has the potential for breakdown of immune homeostasis (32). In support of this notion, to our knowledge, we demonstrated for the first time that TAK1-deficient macrophages underwent massive cell death in response to even low concentrations of FasL. This represents a negative regulatory role for TAK1 to modulate the Fas signaling threshold. Thus, these findings provide a new model to limit Fas-induced proinflammatory cell death.

Our data clearly showed that hyperphosphorylation of RIPK1 was crucial for an increased sensitivity to Fas-induced death of TAK1-deficient macrophages. This indicates the requirement of TAK1 in targeting and antagonizing RIPK1, as described previously (3335). Because mouse BMDMs expressing a catalytically inactive RIPK1 mutant had no effect on the induction of cell death by Fas stimulation (Supplemental Fig. 4A), the data indicate that the RIPK1 kinase activity per se is dispensable for Fas-induced cell death in normal macrophages. In addition, we observed an accelerated reduction in c-FLIPL protein in FasL-treated TAK1-deficient macrophages, and this resembles that of a past study (36). The mutual regulation between c-FLIP and caspase-8 is well appreciated, and c-FLIP is also known as one of prosurvival genes induced by NF-κB activation (5). However, Fas ligation did not lead to robust activation of NF-κB and MAPK signaling pathways even in TAK1-sufficient BMDMs (Supplemental Fig. 4B). For this reason, the dynamics of c-FLIPL in TAK1-deficient macrophages is most likely affected by hyperactivation of RIPK1, thereby promoting caspase-8 activity that is directed to c-FLIPL for proteolytic cleavage, whereas the involvement of NF-κB–dependent events is unlikely. Therefore, it is plausible that TAK1 limits Fas-to-caspase-8–driven macrophage death by restraining the RIPK1 kinase activity in a manner independent of NF-κB–induced de novo protein synthesis.

It is now known that the RIPK1 kinase activity is associated with the induction of not only caspase-8–mediated apoptosis but also RIPK3/MLKL-mediated necroptosis under certain circumstances (37, 38). Nevertheless, we failed to detect Fas-induced RIPK3/MLKL activation in TAK1-deficient macrophages in the absence of caspase inhibitor, indicating that necroptosis does not occur in this situation. One conceivable reason is that active caspase-8 targets and antagonizes RIPK3 as well as upstream RIPK1 to downregulate their activities, as supported by past works (39, 40).

In the present study, we provided strong evidence that macrophage TAK1 plays a unique role in suppressing tissue inflammation. Considering our previous report that Lyz2Cre/+Map3k7flox/flox mice showed neither gross abnormalities nor spontaneous tissue inflammation in the SPF environment (15), the unanticipated phenotypes of TAK1mKO mice offered an opportunity for consideration of TAK1 function in macrophages in vivo. Our present views are as follows. TAK1-deficient macrophages in Lyz2Cre/+Map3k7flox/flox mice may die during fetal stages or soon after birth by the action of TNF-α. Meanwhile, TAK1-deficient macrophages in TAK1mKO mice can persist in vivo after birth. However, as the mice grow up, they are threatened by some intrinsic factors that may lead the macrophages to death. Frequent macrophage disappearance in TAK1mKO mice may reflect a result of macrophage death, and inflammation may be caused by damage-associated molecular patterns (e.g., HMGB1) derived from dead macrophages themselves. Taking our former and current in vitro studies into account (15), this scenario is feasible. Therefore, we focused on a causal relationship between aberrant cell death in TAK1-deficient macrophages after death receptor and/or TLR stimulation and spontaneous tissue inflammation in TAK1mKO mice. Although we have yet to establish its direct link, the characteristic features of TAK1mKO mice remind us of the relevance of death receptor–induced cell death and tissue inflammation. Indeed, our current data from TAK1mKORIPK1D138N/D138N mice are highly suggestive. In the meantime, our previous (15) and present studies suggest that there are only a few relationships of TLR-to-TRIF signaling with disease onset, if any. Future studies will be needed to unequivocally answer this question.

In summary, this study highlighted not only a potential role of TAK1 in regulating Fas-induced macrophage death but also the possibility of perturbation of immune homeostasis driven by aberrant proinflammatory cell death. On the basis of our results, we propose here that TAK1 deficiency in myeloid lineage cells is associated with autoinflammatory disease. Our findings may be applicable as supporting information to understand pathologies of unidentified autoinflammatory diseases and emerging infectious diseases (41, 42).

We are grateful to S. Akira (Osaka University) for providing Lyz2Cre/+Map3k7flox/flox mice and V. Dixit (Genentech) for providing Ripk1D138N/D138N mice. We also thank the staff of the Division of Animal Research and Division of Instrumental Research in Research Center for Supports to Advanced Science at Shinshu University for helpful support.

This work was supported by Japan Society for the Promotion of Science KAKENHI Grant JP21K06868 and Kobayashi International Scholarship Foundation (H.S.).

The online version of this article contains supplemental material.

Abbreviations used in this article:

BMDM

bone marrow–derived macrophage

c-FLIPL

full-length c-FLIP

GSDMD

gasdermin D

HMGB1

high mobility group box 1

LDH

lactate dehydrogenase

LPM

large peritoneal macrophage

MLKL

mixed lineage kinase domain-like protein

PARP

poly(ADP-ribose) polymerase

RIPK1

receptor-interacting serine/threonine-protein kinase 1

SPF

specific pathogen-free

TAK1

TGF-β–activated kinase 1

TNFR

TNF receptor

TRIF

Toll/IL-1R (TIR) domain-containing adapter inducing IFN-β

1.
Croft
M.
,
C. A.
Benedict
,
C. F.
Ware
.
2013
.
Clinical targeting of the TNF and TNFR superfamilies.
Nat. Rev. Drug Discov.
12
:
147
168
.
2.
Dostert
C.
,
M.
Grusdat
,
E.
Letellier
,
D.
Brenner
.
2019
.
The TNF Family of Ligands and Receptors: Communication Modules in the Immune System and Beyond.
Physiol. Rev.
99
:
115
160
.
3.
Strasser
A.
,
P. J.
Jost
,
S.
Nagata
.
2009
.
The many roles of FAS receptor signaling in the immune system.
Immunity
30
:
180
192
.
4.
Lavrik
I. N.
,
P. H.
Krammer
.
2012
.
Regulation of CD95/Fas signaling at the DISC.
Cell Death Differ.
19
:
36
41
.
5.
Tummers
B.
,
D. R.
Green
.
2017
.
Caspase-8: regulating life and death.
Immunol. Rev.
277
:
76
89
.
6.
Bidère
N.
,
H. C.
Su
,
M. J.
Lenardo
.
2006
.
Genetic disorders of programmed cell death in the immune system.
Annu. Rev. Immunol.
24
:
321
352
.
7.
Rieux-Laucat
F.
,
A.
Magérus-Chatinet
,
B.
Neven
.
2018
.
The Autoimmune Lymphoproliferative Syndrome with Defective FAS or FAS-Ligand Functions.
J. Clin. Immunol.
38
:
558
568
.
8.
Sato
S.
,
H.
Sanjo
,
K.
Takeda
,
J.
Ninomiya-Tsuji
,
M.
Yamamoto
,
T.
Kawai
,
K.
Matsumoto
,
O.
Takeuchi
,
S.
Akira
.
2005
.
Essential function for the kinase TAK1 in innate and adaptive immune responses.
Nat. Immunol.
6
:
1087
1095
.
9.
Oeckinghaus
A.
,
S.
Ghosh
.
2009
.
The NF-kappaB family of transcription factors and its regulation.
Cold Spring Harb. Perspect. Biol.
1
:
a000034
.
10.
Rincón
M.
,
R. J.
Davis
.
2009
.
Regulation of the immune response by stress-activated protein kinases.
Immunol. Rev.
228
:
212
224
.
11.
Chen
Z. J.
2012
.
Ubiquitination in signaling to and activation of IKK.
Immunol. Rev.
246
:
95
106
.
12.
Orning
P.
,
D.
Weng
,
K.
Starheim
,
D.
Ratner
,
Z.
Best
,
B.
Lee
,
A.
Brooks
,
S.
Xia
,
H.
Wu
,
M. A.
Kelliher
, et al
2018
.
Pathogen blockade of TAK1 triggers caspase-8-dependent cleavage of gasdermin D and cell death.
Science
362
:
1064
1069
.
13.
Sarhan
J.
,
B. C.
Liu
,
H. I.
Muendlein
,
P.
Li
,
R.
Nilson
,
A. Y.
Tang
,
A.
Rongvaux
,
S. C.
Bunnell
,
F.
Shao
,
D. R.
Green
,
A.
Poltorak
.
2018
.
Caspase-8 induces cleavage of gasdermin D to elicit pyroptosis during Yersinia infection.
Proc. Natl. Acad. Sci. USA
115
:
E10888
E10897
.
14.
Chen
K. W.
,
B.
Demarco
,
R.
Heilig
,
K.
Shkarina
,
A.
Boettcher
,
C. J.
Farady
,
P.
Pelczar
,
P.
Broz
.
2019
.
Extrinsic and intrinsic apoptosis activate pannexin-1 to drive NLRP3 inflammasome assembly.
EMBO J.
38
:
e101638
.
15.
Sanjo
H.
,
J.
Nakayama
,
T.
Yoshizawa
,
H. J.
Fehling
,
S.
Akira
,
S.
Taki
.
2019
.
Cutting Edge: TAK1 Safeguards Macrophages against Proinflammatory Cell Death.
J. Immunol.
203
:
783
788
.
16.
Malireddi
R. K. S.
,
P.
Gurung
,
S.
Kesavardhana
,
P.
Samir
,
A.
Burton
,
H.
Mummareddy
,
P.
Vogel
,
S.
Pelletier
,
S.
Burgula
,
T. D.
Kanneganti
.
2020
.
Innate immune priming in the absence of TAK1 drives RIPK1 kinase activity-independent pyroptosis, apoptosis, necroptosis, and inflammatory disease.
J. Exp. Med.
217
:
e20191644
.
17.
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
.
18.
Sanjo
H.
,
S.
Tokumaru
,
S.
Akira
,
S.
Taki
.
2015
.
Conditional Deletion of TAK1 in T Cells Reveals a Pivotal Role of TCRαβ+ Intraepithelial Lymphocytes in Preventing Lymphopenia-Associated Colitis.
PLoS One
10
:
e0128761
.
19.
Holler
N.
,
A.
Tardivel
,
M.
Kovacsovics-Bankowski
,
S.
Hertig
,
O.
Gaide
,
F.
Martinon
,
A.
Tinel
,
D.
Deperthes
,
S.
Calderara
,
T.
Schulthess
, et al
2003
.
Two adjacent trimeric Fas ligands are required for Fas signaling and formation of a death-inducing signaling complex.
Mol. Cell. Biol.
23
:
1428
1440
.
20.
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
.
21.
Laurien
L.
,
M.
Nagata
,
H.
Schünke
,
T.
Delanghe
,
J. L.
Wiederstein
,
S.
Kumari
,
R.
Schwarzer
,
T.
Corona
,
M.
Krüger
,
M. J. M.
Bertrand
, et al
2020
.
Autophosphorylation at serine 166 regulates RIP kinase 1-mediated cell death and inflammation.
Nat. Commun.
11
:
1747
.
22.
Shi
J.
,
W.
Gao
,
F.
Shao
.
2017
.
Pyroptosis: Gasdermin-Mediated Programmed Necrotic Cell Death.
Trends Biochem. Sci.
42
:
245
254
.
23.
Andersson
U.
,
K. J.
Tracey
.
2011
.
HMGB1 is a therapeutic target for sterile inflammation and infection.
Annu. Rev. Immunol.
29
:
139
162
.
24.
Orozco
S.
,
A.
Oberst
.
2017
.
RIPK3 in cell death and inflammation: the good, the bad, and the ugly.
Immunol. Rev.
277
:
102
112
.
25.
Ghosn
E. E.
,
A. A.
Cassado
,
G. R.
Govoni
,
T.
Fukuhara
,
Y.
Yang
,
D. M.
Monack
,
K. R.
Bortoluci
,
S. R.
Almeida
,
L. A.
Herzenberg
,
L. A.
Herzenberg
.
2010
.
Two physically, functionally, and developmentally distinct peritoneal macrophage subsets.
Proc. Natl. Acad. Sci. USA
107
:
2568
2573
.
26.
Barth
M. W.
,
J. A.
Hendrzak
,
M. J.
Melnicoff
,
P. S.
Morahan
.
1995
.
Review of the macrophage disappearance reaction.
J. Leukoc. Biol.
57
:
361
367
.
27.
Vega-Pérez
A.
,
L. H.
Villarrubia
,
C.
Godio
,
A.
Gutiérrez-González
,
L.
Feo-Lucas
,
M.
Ferriz
,
N.
Martínez-Puente
,
J.
Alcaín
,
A.
Mora
,
G.
Sabio
, et al
2021
.
Resident macrophage-dependent immune cell scaffolds drive anti-bacterial defense in the peritoneal cavity.
Immunity
54
:
2578
2594.e5
.
28.
Wallach
D.
,
T. B.
Kang
,
C. P.
Dillon
,
D. R.
Green
.
2016
.
Programmed necrosis in inflammation: Toward identification of the effector molecules.
Science
352
:
aaf2154
.
29.
Newton
K.
,
V. M.
Dixit
,
N.
Kayagaki
.
2021
.
Dying cells fan the flames of inflammation.
Science
374
:
1076
1080
.
30.
Donado
C. A.
,
A. B.
Cao
,
D. P.
Simmons
,
B. A.
Croker
,
P. J.
Brennan
,
M. B.
Brenner
.
2020
.
A Two-Cell Model for IL-1β Release Mediated by Death-Receptor Signaling.
Cell Rep.
31
:
107466
.
31.
Scaffidi
C.
,
S.
Fulda
,
A.
Srinivasan
,
C.
Friesen
,
F.
Li
,
K. J.
Tomaselli
,
K. M.
Debatin
,
P. H.
Krammer
,
M. E.
Peter
.
1998
.
Two CD95 (APO-1/Fas) signaling pathways.
EMBO J.
17
:
1675
1687
.
32.
Poon
I. K.
,
C. D.
Lucas
,
A. G.
Rossi
,
K. S.
Ravichandran
.
2014
.
Apoptotic cell clearance: basic biology and therapeutic potential.
Nat. Rev. Immunol.
14
:
166
180
.
33.
Dondelinger
Y.
,
M. A.
Aguileta
,
V.
Goossens
,
C.
Dubuisson
,
S.
Grootjans
,
E.
Dejardin
,
P.
Vandenabeele
,
M. J.
Bertrand
.
2013
.
RIPK3 contributes to TNFR1-mediated RIPK1 kinase-dependent apoptosis in conditions of cIAP1/2 depletion or TAK1 kinase inhibition.
Cell Death Differ.
20
:
1381
1392
.
34.
Lamothe
B.
,
Y.
Lai
,
M.
Xie
,
M. D.
Schneider
,
B. G.
Darnay
.
2013
.
TAK1 is essential for osteoclast differentiation and is an important modulator of cell death by apoptosis and necroptosis.
Mol. Cell. Biol.
33
:
582
595
.
35.
Geng
J.
,
Y.
Ito
,
L.
Shi
,
P.
Amin
,
J.
Chu
,
A. T.
Ouchida
,
A. K.
Mookhtiar
,
H.
Zhao
,
D.
Xu
,
B.
Shan
, et al
2017
.
Regulation of RIPK1 activation by TAK1-mediated phosphorylation dictates apoptosis and necroptosis.
Nat. Commun.
8
:
359
.
36.
Guo
X.
,
H.
Yin
,
Y.
Chen
,
L.
Li
,
J.
Li
,
Q.
Liu
.
2016
.
TAK1 regulates caspase 8 activation and necroptotic signaling via multiple cell death checkpoints.
Cell Death Dis.
7
:
e2381
.
37.
Delanghe
T.
,
Y.
Dondelinger
,
M. J. M.
Bertrand
.
2020
.
RIPK1 Kinase-Dependent Death: A Symphony of Phosphorylation Events.
Trends Cell Biol.
30
:
189
200
.
38.
Schwarzer
R.
,
L.
Laurien
,
M.
Pasparakis
.
2020
.
New insights into the regulation of apoptosis, necroptosis, and pyroptosis by receptor interacting protein kinase 1 and caspase-8.
Curr. Opin. Cell Biol.
63
:
186
193
.
39.
Lin
Y.
,
A.
Devin
,
Y.
Rodriguez
,
Z. G.
Liu
.
1999
.
Cleavage of the death domain kinase RIP by caspase-8 prompts TNF-induced apoptosis.
Genes Dev.
13
:
2514
2526
.
40.
Feng
S.
,
Y.
Yang
,
Y.
Mei
,
L.
Ma
,
D. E.
Zhu
,
N.
Hoti
,
M.
Castanares
,
M.
Wu
.
2007
.
Cleavage of RIP3 inactivates its caspase-independent apoptosis pathway by removal of kinase domain.
Cell. Signal.
19
:
2056
2067
.
41.
Manthiram
K.
,
Q.
Zhou
,
I.
Aksentijevich
,
D. L.
Kastner
.
2017
.
The monogenic autoinflammatory diseases define new pathways in human innate immunity and inflammation. [Published erratum appears in 2017 Nat. Immunol. 18: 1271.]
Nat. Immunol.
18
:
832
842
.
42.
Baker
R. E.
,
A. S.
Mahmud
,
I. F.
Miller
,
M.
Rajeev
,
F.
Rasambainarivo
,
B. L.
Rice
,
S.
Takahashi
,
A. J.
Tatem
,
C. E.
Wagner
,
L. F.
Wang
, et al
2022
.
Infectious disease in an era of global change.
Nat. Rev. Microbiol.
20
:
193
205
.

The authors have no financial conflicts of interest.

Supplementary data