Immunological tolerance is constantly being maintained in the periphery by dendritic cells processing material from apoptotic cells (ACs) in the steady-state. Although research has focused on the uptake of ACs by phagocytes, tolerogenic signals exposed by the ACs are much less well defined. In this article, we show that the annexin (Anx) family members AnxA5 and AnxA13 translocate to the surface of ACs to function as redundant tolerogenic signals in vitro and in vivo. Exposure of bone marrow–derived dendritic cells to AnxA5 or AnxA13 in vitro resulted in the inhibition of both proinflammatory cytokine secretion and the upregulation of costimulatory molecules upon TLR stimulation. The highly conserved Anx core domain was sufficient to mediate these effects, whereas recognition by N-formyl peptide receptor family members was dispensable. In vivo, coinjection of OVA-expressing and Anx-expressing ACs prevented induction of Ag-specific CD8+ T cells. Moreover, mice immunized with Anx-expressing ACs became refractory to an antigenic challenge. These results suggest that several Anxs contribute to AC-induced suppression of dendritic cell activation. Therefore, manipulating Anx-mediated immunosuppression may prove beneficial for patients with cancer or autoimmune diseases and chronic inflammatory disorders.

One crucial mechanism of peripheral tolerance induction is the uptake, processing, and presentation of apoptotic cell (AC)-derived Ags by professional phagocytes (1). In the steady-state, immature dendritic cells (DCs) continuously engulf ACs and present self-Ags from peripheral tissues in the draining lymph nodes (2, 3). In the absence of inflammation or infection, the presentation of self-Ags to naive T cells induces tolerance (46). Defects in AC uptake, AC processing, or tolerization of phagocytes after AC uptake have been implicated in the development of autoimmune diseases and chronic inflammatory disorders (710). Although a plethora of eat-me signals and their uptake receptors on phagocytes are well characterized, little is known about molecules on ACs that mediate DC tolerization. Recently, we identified the early exposure of the cytosolic protein annexin (Anx)1 (AnxA1) as a tolerogenic signal on the surface of ACs (11).

AnxA1 belongs to an evolutionarily conserved family of proteins that binds to negatively charged phospholipids in a Ca2+-dependent manner (12). Lipid binding is mediated by the C-terminal core domain, which is highly conserved among all Anx family members (12, 13). The N-terminal domain is unique for each Anx and is proposed to mediate specific functions of individual Anxs (12, 14). Peptides corresponding to the AnxA1 N terminus were shown to bind to members of the N-formyl peptide receptor (FPR) family, resulting in a reduction of neutrophil transmigration in several models of acute and chronic inflammation (1518). Downstream signaling induced by binding of AnxA1 N-terminal peptides to FPR family members causes activation of ERK but not of p38 or JNK (19, 20). Additional anti-inflammatory properties of AnxA1 have been attributed to the inhibition of cytosolic phospholipase A2 (cPLA2) and secretory phospholipase A2 (sPLA2) activity (2123). Inhibition of sPLA2 is mediated by the antiflammin-2 (AF-2) sequence, which also negatively regulates polymorphonuclear leukocyte adhesion to activated endothelium by binding to human FPR2 (24).

The presence of multiple Anx family members in all higher eukaryotes suggests a fundamental role for Anxs in cell biology. However, mice deficient in individual Anx family members have no severe phenotype, suggesting that several Anxs have (partially) overlapping functions (12, 25). In fact, functional redundancy of Anxs was proved in the context of membrane trafficking, inhibition of phospholipase A2 activity, and blood coagulation (12). Whether tolerance induction, which we showed to be mediated by AnxA1, is a property shared by several Anx family members and, therefore, represents an additional redundant function of Anxs is unknown (11).

In this article, we describe two novel tolerogenic signals on the surface of ACs, AnxA5 and AnxA13, promoting the development of DCs with a tolerogenic phenotype (i.e., DCs with an impaired upregulation of costimulatory molecules and with low proinflammatory cytokine secretion upon TLR stimulation). These effects were mediated by the Anx core domain and were independent of N-terminal peptides of AnxA1 or the AF-2 sequence. In line with this, AnxA1, AnxA5, and AnxA13 inhibit the induction of Ag-specific CD8+ T cell responses in vivo. Importantly, mice immunized with AnxA5- or AnxA13-expressing ACs showed a significantly reduced population of CD8+ T cells specific for AC-associated Ags and became refractory to an antigenic challenge. Based on these results, we propose that several Anxs contribute to AC-induced immunosuppression in a functionally redundant manner and, thus, contribute to peripheral tolerance induction.

Tlr4−/−, AnxA1−/−, wild-type (WT), OT-I, and lymphoproliferation (lpr) mice (all on a C57BL/6 background) were maintained and bred under specific pathogen–free conditions. Tlr4−/− mice were kindly provided by S. Akira (Osaka University, Osaka, Japan), S. Uematsu (Osaka University), and L. Gissmann (German Cancer Research Center). AnxA1−/− mice were purchased from B & K Universal. OT-I mice were kindly provided by N. Garbi (University of Bonn, Bonn, Germany) and G. Hämmerling (German Cancer Research Center).

Bone marrow (BM)-derived DCs (BMDCs) were prepared from male or female mice between 6 and 12 wk of age, as described (26, 27). In brief, BM cells were flushed from tibias and femurs, and RBCs were lysed by brief exposure to 0.168 M NH4Cl. Cells were washed twice with RPMI 1640. For differentiation of BM precursors to BMDCs using recombinant murine GM-CSF, 1 × 106 cells were seeded at a density of 1 × 106 cells/ml in RPMI 1640 complete medium (10% FCS, 10 U/ml penicillin/streptomycin, 300 mg/l l-glutamine, 20 ng/ml GM-CSF [Immunotools]) in a 24-well plate. After 2 d, the medium was replaced by fresh medium. After 4 d, half of the medium was removed and replaced by fresh medium. Experiments were conducted 7–8 d after differentiation. For differentiation of BM precursors to BMDCs using recombinant human Flt3L, cells were seeded at a density of 3 × 106 cells/ml in RPMI 1640 complete medium (10% FCS, 10 U/ml penicillin/streptomycin, 300 mg/l l-glutamine, 300 ng/ml Flt3L [eBioscience]) in a 100-mm petri dish for 8–10 d. Additional Flt3L was added to the culture at day 5 or 6. Differentiation of BM precursors to BMDCs was monitored by flow cytometric analysis of cells using mAbs against CD11c and MHC class II.

mAbs and reagents used for FACS were purchased from BD Biosciences (anti-CD3–FITC, anti-CD4–FITC, anti-CD8–PE, anti-CD8–allophycocyanin, anti-CD40–PE, anti-CD45R–PerCP–Cy5.5, anti-CD86–PE, anti-Thy1.1–PE, and streptavidin-allophycocyanin), eBioscience (anti-CD14–allophycocyanin, anti-CD44–PerCP–Cy5.5, anti-CD80–FITC, anti-PD-L1–PE, anti-PD-L2–FITC), Caltag (anti-CD11c–PE, anti-MHC class II–FITC), Immunotools (anti-CD62L–PE), or QIAGEN (anti-His–biotin). Anti-human AnxA1 (DAC5) and anti-MHC class I–biotin (Y3) Abs were generated in our laboratory and the laboratory of G. Hämmerling (11). Appropriate isotype controls were purchased from BD Biosciences or eBioscience. Phosphatidylserine exposure and cellular membrane integrity were analyzed by staining with AnxV-FITC (Immunotools) and 7-aminoactinomycin D (7-AAD; Sigma-Aldrich), respectively. Stained cells were analyzed on a FACSCanto (BD Biosciences) with FlowJo software (TreeStar). For detection of FPR family members, cells were stained with the fluorescently labeled FPR ligand WKYMVM-FAM (Phoenix Pharmaceuticals) or Formyl-Nle-Leu-Phe-Nle-Tyr-Lys-fluorescein (Life Technologies).

Anti-murine AnxA1 (8D10) and anti-human AnxA1 (DAC5) Abs were generated in our laboratory (11). Commercially available Abs for detection of MAPK, α-tubulin, and β-actin were purchased as follows: AnxA5 (AF399; R&D Systems), AnxA13 (AF4149; R&D Systems), p-ERK (E10; Cell Signaling), ERK1 (MK12; BD), p-JNK (G9; Cell Signaling), JNK1/3 (C17; Santa Cruz Biotechnology), p-p38 (D3F9; Cell Signaling), p38α (5F11; Cell Signaling), α-tubulin (B-5-1-2; Sigma-Aldrich), and β-actin (AC-15; Abcam). Cytokine concentrations in supernatants were determined by ELISA for murine TNF-α, IL-12p40, IL-6, and IL-10 (PeproTech), according to the manufacturers’ instructions. Boc-1 and Boc-2 were purchased from MP Biomedicals. fMLF was purchased from Sigma-Aldrich.

The mouse (m)AnxA1-pET41a, mAnxA1 mAF-2-pET41a, mAnxA1 ΔN-pET41a, mAnxA1 ΔN mAF-2-pET41a, mAnxA5-pET41a, and mAnxA13-pET41a plasmids were generated by cloning of mAnxA1, mAnxA1 mAF-2, mAnxA1 ΔN, mAnxA1 ΔN mAF-2, mAnxA5, or mAnxA13, respectively, into a modified version of pET41a harboring a C-terminal FLAG tag, a PreScission Protease cleavage site, and a protein A tag. Recombinant proteins were expressed in the Escherichia coli strain BL21 (DE3) pLysS (Promega). Removal of LPS during protein purification was achieved by washing with TBS containing 0.1% Triton X-114 (Sigma-Aldrich). Residual LPS concentrations in the protein preparations were determined by the Limulus Amebocyte Lysate Assay (Lonza). The mAnxA1-pAc5.1 V5/HisA, mAnxA5-pAc5.1 V5/HisA, and mAnxA13 pAc5.1/V5-HisA plasmids were generated by cloning mAnxA1, mAnxA5, and mAnxA13, respectively, into pAc5.1/V5-HisA (Life Technologies). Drosophila melanogaster Schneider 2 (S2) cells were transfected with Ca3(PO4)2, according to the manufacturer’s instructions (Life Technologies). To generate stably transfected S2 cell lines, cotransfection with pCoHygro was performed, and cells were selected using Hygromycin B (PAA). D. melanogaster S2 cells were cultured in Schneider’s insect medium (Sigma-Aldrich) supplemented with 10% FCS. Human Jurkat T cells and the human T-ALL cell line CEM were cultured in RPMI 1640 medium (Sigma-Aldrich) supplemented with 10% FCS.

RNA was isolated from cells using the RNAqueous-Micro Kit, according to the manufacturer’s instructions (Ambion). RNA was quantified by detection of SYBR Green incorporation using the ABI Prism 7500 sequence detector system (Applied Biosystems). Expression levels were normalized to GAPDH. The following primer sequences were used: Fpr1 5′-GCC GTC ACC ATG CTC ACT GTC A-3′ (fwd) and 5′-AAC CCG CAA AGG ACG GCT GG-3′ (rev); Fpr2 5′-AGC TGG TTG TGC AGA CAA AAT GGA-3′ (fwd) and 5′-TGC CCA GCA CAC CAA GGA AG-3′ (rev); Fpr3 5′-CCT TCC CGA GTT CTT ACA GG-3′ (fwd) and 5′-CAC TAA ACT GCA TCT CTT TGA G-3′ (rev); and GAPDH 5′-ACT CCA CTC ACG GCA AAT TCA-3′ (fwd) and 5′-GCC TCA CCC CAT TTG ATG TT-3′ (rev).

Murine neutrophils were isolated from BM by positive selection using anti-Ly6G MicroBeads (Miltenyi Biotec), according to the manufacturer’s instructions. Neutrophils were cultured in RPMI 1640 medium supplemented with 10% FCS at a density of 2 × 106 cells/ml. The purity of cells was controlled by flow cytometric analysis using anti-Gr-1–FITC (Miltenyi Biotec). Cells were >90% positive for Gr-1. Murine splenocytes were isolated from spleens and filtered through a 40-μm cell strainer (BD Biosciences). Splenocytes were cultured in RPMI 1640 medium supplemented with 10% FCS.

To generate early ACs, Jurkat T cells or CEM cells were UV-C irradiated (50 mJ/cm2, Stratalinker 1800; Stratagene) in six-well plates at a cell density of 1 × 106 cells/ml. Subsequently, cells were cultured at 37°C for 2–6 h. To generate early apoptotic splenocytes, cells were UV-C irradiated (15 mJ/cm2) in six-well plates at a cell density of 1 × 106 cells/ml. Subsequently, cells were cultured in media at 37°C for 3 h. Primary murine neutrophils spontaneously undergo apoptosis in cell culture and show characteristics of early ACs after 1 d. S2 cells were irradiated with 250 mJ/cm2 at a cell density of 1 × 106 cells/ml in 100-mm petri dishes and used after overnight (o/n) incubation.

For analysis of OVA-specific, endogenous T cells, a mixture of 5 × 106 apoptotic, membrane-anchored OVA (mOVA)-expressing S2 cells (aS2 mOVA) together with 5 × 106 AnxA1-, AnxA5-, or AnxA13-expressing, apoptotic S2 cells (aS2 AnxA1, aS2 AnxA5, and aS2 AnxA13, respectively) or mock-transfected apoptotic S2 cells (aS2 mock) were injected i.v. into female C57BL/6 WT mice. Six to eight days after immunization, single-cell suspensions of mesenteric lymph nodes were analyzed for OVA-specific CD8+ T cells using PE-labeled Kb/SIINFEKL pentamers, according to the manufacturer’s instructions (ProImmune). For the analysis of OVA-specific T cell proliferation and activity, 1 × 106 CFSE-labeled OT-I T cells were injected i.v. into female C57BL/6 WT mice. The next day, mice were immunized i.v. with a mixture of 0.5 × 106 aS2 mOVA, together with 1 × 106 aS2 AnxA5, aS2 AnxA13, or aS2 mock. After 12–13 d, mice were challenged i.p. with 50 μg OVA protein (InvivoGen) in 125 μl PBS emulsified in 125 μl IFA (Sigma-Aldrich). On day 6 after challenge, splenic cell suspensions were analyzed by flow cytometry and prepared for ELISPOT assays in vitro. For determination of absolute OT-I T cell numbers/spleen, we analyzed an exact aliquot of each spleen using Trucount Beads (Becton Dickinson), according to the manufacturer’s instructions.

IFN-γ ELISPOT assays were performed with 1.5 × 106 splenic cells/well and 1 μM the OVA-derived, MHC class I–dependent peptide SIINFEKL (Axxora). Splenic cell suspensions were stimulated o/n, and IFN-γ ELISPOTs were developed according to the manufacturer’s instructions (BD Pharmingen).

A total of 1 × 105 BMDCs was incubated with recombinant protein (125–500 nM) or with apoptotic Jurkat T cells (2–4 × 105 cells), apoptotic neutrophils (1 × 105–1 × 106 cells), apoptotic splenocytes (1 × 105–1 × 106 cells), or apoptotic S2 cells (5 × 105–1 × 106) for 6–8 h. Depending on the mouse background, BMDCs were subsequently stimulated with LPS (Sigma-Aldrich), CpG 1668, or CpG 2395 (InvivoGen). Cytokine concentrations in the supernatants were analyzed by ELISA 12–16 h after TLR stimulation. Surface molecule expression was analyzed 48 h after TLR stimulation by flow cytometry.

Statistical analysis of data was performed by one-way ANOVA, followed by the Bonferroni posttest for multiple comparisons. If not otherwise indicated, the significance of the difference compared with CpG-stimulated cells is depicted. The p values < 0.05 were considered statistically significant.

All animal studies were approved by the veterinary authorities (Regierungspräsidium Karlsruhe) of Baden-Württemberg (G-96 06, G-173/11).

We previously showed that AnxA1 on the surface of ACs negatively regulates DC activation, as evidenced by low expression of costimulatory molecules and low proinflammatory cytokine secretion upon TLR stimulation (11). To investigate the extent to which known anti-inflammatory sequences of AnxA1—the N terminus and the AF-2 sequence—contribute to this tolerogenic effect, we generated recombinant full-length AnxA1 and AnxA1 mutants (Fig. 1A, Supplemental Fig. 1A–D). LPS was carefully removed during purification of recombinant proteins by addition of Triton X-114, and all in vitro experiments were performed using BMDCs from Tlr4−/− mice to exclude effects induced by endotoxin tolerance. In addition, a control protein, serpin 8b, purified in an identical fashion as AnxA1, was inactive in our functional assays (11). Surprisingly, deletion of the N-terminal domain, mutation of the AF-2 sequence, or a combination of both did not affect AnxA1-induced inhibition of proinflammatory cytokine secretion upon stimulation with the TLR9 ligand CpG (Fig. 1B, Supplemental Fig. 1E). This was not due to an absence of FPR expression, because FPR family members involved in AnxA1 recognition are expressed in BMDCs on the mRNA level and on the protein level (Fig. 1C, 1D). To further study the involvement of FPR family members in AnxA1-induced DC tolerization, the pan-specific FPR antagonists Boc-1 and Boc-2 were used in subsequent experiments. The presence of Boc-1 and Boc-2 did not abrogate inhibition of TLR-induced TNF-α secretion by AnxA1 (Fig. 1E, Supplemental Fig. 1F, 1G). In line with this, the prototypic FPR ligand fMLF, but not AnxA1, led to activation of MAPK (Fig. 1F, 1G). In conclusion, FPR family members do not contribute to the recognition of AnxA1 by BMDCs in the context of suppression of DC activation upon TLR stimulation. Instead, the AnxA1 core domain is sufficient to mediate DC tolerization, independent of the AF-2 sequence and the AnxA1 N-terminal domain.

FIGURE 1.

The core domain mediates the tolerogenic effect of AnxA1. (A) Full-length AnxA1 and the AnxA1 mutants AnxA1ΔN, AnxA1 mAF-2, and AnxA1ΔN mAF-2. (B) GM-CSF–differentiated BMDCs from Tlr4−/− mice were incubated with the indicated concentrations of recombinant protein. Six to eight hours later, cells were stimulated with 40 nM CpG o/n. TNF-α concentrations in the supernatants were determined by ELISA. Data are mean ± SEM of three independent experiments. (C) Quantification of mRNA levels of FPR1, FPR2, and FPR3 relative to GAPDH in neutrophils (NΦ) and GM-CSF–differentiated BMDCs by quantitative RT-PCR. (D) Surface expression of FPR1, FPR2, and FPR3 on transiently transfected 293T or BMDCs was detected using the fluorescently labeled FPR ligands fMLF-fluorescein and WKYMVM-FAM. (E) GM-CSF–differentiated BMDCs from Tlr4−/− mice were incubated with the indicated concentrations of recombinant AnxA1 in the presence or absence of the FPR antagonists Boc-1 and/or Boc-2 or apoptotic Jurkat T cells (aJ, UV-C-irradiation 50 mJ/cm2, 4 h). Six to eight hours later, cells were stimulated with 40 nM CpG o/n. TNF-α concentrations in the supernatants were determined by ELISA. Data are mean ± SEM of three independent experiments. (F) Immunoblot analysis of MAPK activation of BMDCs after incubation with 500 nM of recombinant AnxA1 for the indicated times. Lysates of CpG- and LPS-stimulated BMDCs from Tlr4−/− and WT mice, respectively, served as positive controls. Data are representative of three independent experiments. (G) Immunoblot analysis of MAPK activation of BMDCs after incubation with fMLF for the indicated times. Lysates of CpG- and LPS-stimulated BMDCs from Tlr4−/− and WT mice, respectively, served as positive controls. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

FIGURE 1.

The core domain mediates the tolerogenic effect of AnxA1. (A) Full-length AnxA1 and the AnxA1 mutants AnxA1ΔN, AnxA1 mAF-2, and AnxA1ΔN mAF-2. (B) GM-CSF–differentiated BMDCs from Tlr4−/− mice were incubated with the indicated concentrations of recombinant protein. Six to eight hours later, cells were stimulated with 40 nM CpG o/n. TNF-α concentrations in the supernatants were determined by ELISA. Data are mean ± SEM of three independent experiments. (C) Quantification of mRNA levels of FPR1, FPR2, and FPR3 relative to GAPDH in neutrophils (NΦ) and GM-CSF–differentiated BMDCs by quantitative RT-PCR. (D) Surface expression of FPR1, FPR2, and FPR3 on transiently transfected 293T or BMDCs was detected using the fluorescently labeled FPR ligands fMLF-fluorescein and WKYMVM-FAM. (E) GM-CSF–differentiated BMDCs from Tlr4−/− mice were incubated with the indicated concentrations of recombinant AnxA1 in the presence or absence of the FPR antagonists Boc-1 and/or Boc-2 or apoptotic Jurkat T cells (aJ, UV-C-irradiation 50 mJ/cm2, 4 h). Six to eight hours later, cells were stimulated with 40 nM CpG o/n. TNF-α concentrations in the supernatants were determined by ELISA. Data are mean ± SEM of three independent experiments. (F) Immunoblot analysis of MAPK activation of BMDCs after incubation with 500 nM of recombinant AnxA1 for the indicated times. Lysates of CpG- and LPS-stimulated BMDCs from Tlr4−/− and WT mice, respectively, served as positive controls. Data are representative of three independent experiments. (G) Immunoblot analysis of MAPK activation of BMDCs after incubation with fMLF for the indicated times. Lysates of CpG- and LPS-stimulated BMDCs from Tlr4−/− and WT mice, respectively, served as positive controls. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

Close modal

The Anx core domain is highly conserved among Anx family members, and redundancy was reported for various functions of Anxs (12, 25, 2831). Therefore, we hypothesized that several Anxs might act as tolerogenic signals on the surface of ACs. To test this hypothesis, we first investigated whether other Anxs, in addition to AnxA1, translocate to the cell surface upon apoptosis induction. We focused on AnxA5, because it is upregulated in different tissues of AnxA1−/− mice and might compensate for the loss of AnxA1 (32), as well as on AnxA13, which represents the founding member of the family (33). The surface translocation of Anxs was first studied by ectopic expression of murine Anxs in D. melanogaster S2 cells. Stably transfected S2 cells showed surface exposure of AnxA1, AnxA5, or AnxA13 during early apoptosis when membrane integrity was still intact (Fig. 2A, Supplemental Fig. 2). Next, we induced apoptosis in the human T cell line CEM and monitored translocation of endogenous Anxs. In line with the findings observed in stably transfected S2 cells, AnxA5 and AnxA13, in addition to AnxA1, translocated to the surface of apoptotic CEM cells (Fig. 2B, 2C). Of note, protein amounts of Anxs in the cytosolic fraction remained almost unchanged, indicating that only a minor fraction of the cytosolic pool of Anxs translocates to the cell surface during apoptosis (Fig. 2B). In summary, AnxA1, as well as AnxA5 and AnxA13, are exposed on early ACs.

FIGURE 2.

AnxA5 and AnxA13 translocate to the cell surface upon induction of apoptosis. (A) Flow cytometric analysis of aS2. Sixteen hours after UV-C irradiation (250 mJ/cm2), mock- or AnxA1/5/13-transfected aS2 cells were early apoptotic (AnxV+/7-AAD) and stained using biotin-conjugated anti-6xHis streptavidin-allophycocyanin. (B) Immunoblot analysis of cytosolic and membrane (EDTA wash) fractions of viable (0 h) or early UV-C–irradiated apoptotic CEM cells (50 mJ/cm2, 2–6 h). (C) Quantification of cell viability of UV-C–irradiated early apoptotic CEM cells in (B) by flow cytometry using AnxV-FITC/7-AAD.

FIGURE 2.

AnxA5 and AnxA13 translocate to the cell surface upon induction of apoptosis. (A) Flow cytometric analysis of aS2. Sixteen hours after UV-C irradiation (250 mJ/cm2), mock- or AnxA1/5/13-transfected aS2 cells were early apoptotic (AnxV+/7-AAD) and stained using biotin-conjugated anti-6xHis streptavidin-allophycocyanin. (B) Immunoblot analysis of cytosolic and membrane (EDTA wash) fractions of viable (0 h) or early UV-C–irradiated apoptotic CEM cells (50 mJ/cm2, 2–6 h). (C) Quantification of cell viability of UV-C–irradiated early apoptotic CEM cells in (B) by flow cytometry using AnxV-FITC/7-AAD.

Close modal

Because AnxA5 and AnxA13 met the prerequisite to act as a tolerogenic signal, surface translocation during early apoptosis, we next studied whether they also, like AnxA1, regulate TLR-induced DC activation. For this, either purified recombinant proteins or stably transfected S2 cells exposing the respective Anx upon apoptosis induction were used (Fig. 2A, Supplemental Fig. 3A, 3B). Incubation of BMDCs with recombinant AnxA1, AnxA5, or AnxA13 significantly reduced the secretion of the proinflammatory cytokines TNF-α, IL-12p40, and IL-6 by TLR-stimulated BMDCs in a concentration-dependent manner (Fig. 3A–D). Importantly, this effect was independent of altered cell viability of BMDCs (Supplemental Fig. 3C). Similar to the results obtained using recombinant Anxs, aS2 AnxA1 or aS2 AnxA5 reduced TLR-induced TNF-α secretion. In contrast, aS2 mock did not modulate DC activation (Fig. 3E). Flow cytometric analysis did not reveal differences in phosphatidylserine externalization, indicating that apoptosis kinetics were comparable in mock- and Anx-transfected S2 cells (Supplemental Fig. 3D). The tolerogenic effect of Anxs was concentration dependent, because the inhibition of TLR-induced proinflammatory cytokine secretion increased with the ratio of apoptotic S2 cells/BMDCs and correlated with Anx expression levels in S2 cells (Fig. 3E, 3F, Supplemental Fig. 3E). CD8α+ DCs, which can be generated via differentiation of BM precursors using Flt3L in vitro, play a critical role in the presentation of self-Ags derived from ACs (34). Addition of recombinant Anxs to Flt3L-derived BMDCs reduced the TLR-induced secretion of TNF-α in a concentration-dependent manner, whereas the secretion of the anti-inflammatory cytokine IL-10 was not affected (Fig. 3G, 3H). These effects were recapitulated with Flt3L-derived BMDCs incubated with aS2 AnxA1 or aS2 AnxA5. Although the secretion of the proinflammatory cytokine TNF-α was reduced, Anxs had no influence on the secretion of IL-10 (Supplemental Fig. 3F, 3G). Taken together, our data show that AnxA5 and AnxA13 inhibit the TLR-induced secretion of proinflammatory cytokines by DCs, including CD8α+ DCs, whereas secretion of the immune-regulatory cytokine IL-10 was not affected.

FIGURE 3.

AnxA5 and AnxA13 inhibit proinflammatory cytokine secretion of BMDCs upon TLR stimulation. (A) AnxA1, AnxA5, and AnxA13. The N-terminal domain is unique for each Anx family member and is depicted in yellow. The highly conserved core domain is shown in blue. (BD) GM-CSF–differentiated BMDCs from Tlr4−/− mice were incubated with the indicated concentrations of recombinant protein. Six to eight hours later, cells were stimulated with 40 nM CpG o/n. TNF-α (B), IL-12p40 (C), or IL-6 (D) concentrations in the supernatants were determined by ELISA. Data are mean ± SEM of three independent experiments. (E and F) GM-CSF–differentiated BMDCs from Tlr4−/− mice were incubated with the indicated ratio of apoptotic Jurkat T cells (aJ) or aS2 AnxA1, aS2 AnxA5, or aS2 mock. Four to six hours later, cells were stimulated with 40 nM CpG o/n. TNF-α concentrations in the supernatants were determined by ELISA. Data are mean ± SEM of four independent experiments. (G and H) Flt3L-differentiated BMDCs from Tlr4−/− mice were incubated with the indicated concentrations of recombinant protein. Six to eight hours later, cells were stimulated with 40 nM CpG o/n. TNF-α (G) or IL-10 (H) concentrations in the supernatants were determined by ELISA. Data are mean ± SEM of two independent experiments. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

FIGURE 3.

AnxA5 and AnxA13 inhibit proinflammatory cytokine secretion of BMDCs upon TLR stimulation. (A) AnxA1, AnxA5, and AnxA13. The N-terminal domain is unique for each Anx family member and is depicted in yellow. The highly conserved core domain is shown in blue. (BD) GM-CSF–differentiated BMDCs from Tlr4−/− mice were incubated with the indicated concentrations of recombinant protein. Six to eight hours later, cells were stimulated with 40 nM CpG o/n. TNF-α (B), IL-12p40 (C), or IL-6 (D) concentrations in the supernatants were determined by ELISA. Data are mean ± SEM of three independent experiments. (E and F) GM-CSF–differentiated BMDCs from Tlr4−/− mice were incubated with the indicated ratio of apoptotic Jurkat T cells (aJ) or aS2 AnxA1, aS2 AnxA5, or aS2 mock. Four to six hours later, cells were stimulated with 40 nM CpG o/n. TNF-α concentrations in the supernatants were determined by ELISA. Data are mean ± SEM of four independent experiments. (G and H) Flt3L-differentiated BMDCs from Tlr4−/− mice were incubated with the indicated concentrations of recombinant protein. Six to eight hours later, cells were stimulated with 40 nM CpG o/n. TNF-α (G) or IL-10 (H) concentrations in the supernatants were determined by ELISA. Data are mean ± SEM of two independent experiments. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

Close modal

ACs and AnxA1 also regulate surface expression of costimulatory molecules on DCs (11, 35, 36). Therefore, the effect of recombinant AnxA5 and AnxA13 on the expression of coregulatory and MHC molecules was investigated 2 d after TLR stimulation. Incubation of BMDCs with AnxA1, AnxA5, or AnxA13 did not influence the surface expression of MHC class I molecules, but it slightly increased the expression of MHC class II molecules (Fig. 4A, 4B). Remarkably, AnxA1, AnxA5, and AnxA13 inhibited upregulation of the costimulatory molecules CD40, CD80, and CD86 upon TLR stimulation (Fig. 4C–E). In contrast, surface expression of the coinhibitory molecules programmed cell death ligand (PD-L)1 and PD-L2, as well as the cell viability of BMDCs, were not affected (Fig. 4F–H). In summary, AnxA1, AnxA5, and AnxA13 impair TLR-induced upregulation of costimulatory molecules without affecting surface expression of coinhibitory and MHC molecules.

FIGURE 4.

AnxA5 and AnxA13 specifically downregulate the expression of costimulatory molecules on BMDCs. (AG) GM-CSF–differentiated BMDCs from Tlr4−/− mice were incubated with 500 nM of the indicated recombinant protein. Six to eight hours later, cells were stimulated with 20 nM CpG. After 2 d, cells were stained with Abs against MHC I (A), MHC class II (B), CD40 (C), CD80 (D), CD86 (E), PD-L1 (F), and PD-L2 (G). Depicted are the mean fluorescence intensity (MFI) of individual mice and the group mean. (H) Quantification of cell viability of BMDCs of mouse 1 used in (A)–(G) by flow cytometry using AnxV-FITC/7-AAD. *p < 0.05, ****p < 0.0001.

FIGURE 4.

AnxA5 and AnxA13 specifically downregulate the expression of costimulatory molecules on BMDCs. (AG) GM-CSF–differentiated BMDCs from Tlr4−/− mice were incubated with 500 nM of the indicated recombinant protein. Six to eight hours later, cells were stimulated with 20 nM CpG. After 2 d, cells were stained with Abs against MHC I (A), MHC class II (B), CD40 (C), CD80 (D), CD86 (E), PD-L1 (F), and PD-L2 (G). Depicted are the mean fluorescence intensity (MFI) of individual mice and the group mean. (H) Quantification of cell viability of BMDCs of mouse 1 used in (A)–(G) by flow cytometry using AnxV-FITC/7-AAD. *p < 0.05, ****p < 0.0001.

Close modal

The phenotypes of BMDCs incubated with AnxA5 or AnxA13 resemble the phenotype induced by AnxA1. Therefore, we next tested whether recognition of the AnxA5, AnxA13, or the AnxA1 core domain is also independent of FPR family members. Indeed, analysis of MAPK activation revealed that MAPK was not activated in BMDCs after incubation with AnxA5, AnxA13, or the AnxA1 core domain (Fig. 5A). Furthermore, the pan-specific FPR antagonist Boc-2 did not abrogate inhibition of TLR-induced proinflammatory cytokine secretion by AnxA5, AnxA13, or the AnxA1 core domain (Fig. 5B). In conclusion, AnxA1, AnxA5, and AnxA13 induce the development of DCs with a tolerogenic phenotype, as characterized by low expression of costimulatory molecules and low proinflammatory cytokine secretion independent of FPR signaling.

FIGURE 5.

FPR family members are not involved in the recognition of AnxA5 and AnxA13 on BMDCs. (A) Immunoblot analysis of MAPK activation of BMDCs after incubation with 500 nM of the indicated recombinant protein for the indicated times. Lysates of CpG- and LPS-stimulated BMDCs from Tlr4−/− and WT mice, respectively, served as positive controls. Data are representative of three independent experiments. (B) GM-CSF–differentiated BMDCs from Tlr4−/− mice were incubated with the indicated concentrations of recombinant protein in the presence or absence of the FPR antagonist Boc-2 or apoptotic Jurkat T cells (aJ). Six to eight hours later, cells were stimulated with 40 nM CpG o/n. IL-12p40 concentrations in the supernatants were determined by ELISA. Data are mean ± SEM of three independent experiments. *p < 0.05, ****p < 0.0001.

FIGURE 5.

FPR family members are not involved in the recognition of AnxA5 and AnxA13 on BMDCs. (A) Immunoblot analysis of MAPK activation of BMDCs after incubation with 500 nM of the indicated recombinant protein for the indicated times. Lysates of CpG- and LPS-stimulated BMDCs from Tlr4−/− and WT mice, respectively, served as positive controls. Data are representative of three independent experiments. (B) GM-CSF–differentiated BMDCs from Tlr4−/− mice were incubated with the indicated concentrations of recombinant protein in the presence or absence of the FPR antagonist Boc-2 or apoptotic Jurkat T cells (aJ). Six to eight hours later, cells were stimulated with 40 nM CpG o/n. IL-12p40 concentrations in the supernatants were determined by ELISA. Data are mean ± SEM of three independent experiments. *p < 0.05, ****p < 0.0001.

Close modal

Several Anxs translocate to the surface of ACs to promote the development of tolerogenic DCs, and Anx single-knockout mice lack a severe phenotype. Therefore, we hypothesized that Anxs are redundant tolerogenic signals in vivo. Thus, deletion of AnxA1 should neither impair suppressive effects of ACs on AC-treated BMDCs nor provoke autoimmunity against self-Ags derived from ACs. In fact, the absence of AnxA1 does not lead to an altered phenotype of BMDCs after AC engulfment. Apoptotic neutrophils or splenocytes from WT and AnxA1−/− mice suppressed TLR-induced TNF-α secretion by BMDCs to a comparable extent. The level of suppression increased with an elevated ratio of ACs/BMDCs, as seen with apoptotic Jurkat T cells, which served as a positive control (Fig. 6A, Supplemental Fig. 4A, 4B). Importantly, neutrophils and splenocytes from WT and AnxA1−/− mice showed no difference in apoptosis kinetics upon irradiation with UV-C (Supplemental Fig. 4C).

FIGURE 6.

AnxA5 and AnxA13 suppress the induction of CD8+ T cell immune responses in vivo. (A) GM-CSF–differentiated BMDCs from WT mice were incubated with the indicated ratio of apoptotic Jurkat T cells (aJ) or apoptotic neutrophils (aNΦ, after 1 d in vitro culture). Four hours later, cells were stimulated with 1 ng/ml LPS o/n. TNF-α concentrations in the supernatants were determined by ELISA. Data are mean ± SEM of three independent experiments. (B) aS2 mOVA, together with aS2 AnxA1, aS2 AnxA5, or aS2 AnxA13 were injected i.v. into mice. Mice injected with aS2 mock served as a negative control. After 8 d, the induction of OVA-specific CD8+ T cells in the mesenteric lymph nodes was analyzed by flow cytometry. Errors bars represent mean ± SEM. (C and D) One day after transfer of 1 × 106 CFSE-labeled OT-I T cells, C57BL/6 WT mice were immunized i.v. with 0.5 × 106 aS2 mOVA together with 1 × 106 aS2 AnxA5, aS2 AnxA13, or aS2 mock. After 12–13 d, mice were challenged with 50 μg OVA protein in IFA, and spleens were analyzed 6 d after challenge by flow cytometry for cell numbers of transferred OT-I cells in spleens (C) and IFN-γ secreting CD8+ T cells by ELISPOT (D). n = 18 (aS2 mOVA/aS2 mock), n = 9 (aS2 mOVA/aS2 AnxA5 and aS2 mOVA/aS2 AnxA13). *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

FIGURE 6.

AnxA5 and AnxA13 suppress the induction of CD8+ T cell immune responses in vivo. (A) GM-CSF–differentiated BMDCs from WT mice were incubated with the indicated ratio of apoptotic Jurkat T cells (aJ) or apoptotic neutrophils (aNΦ, after 1 d in vitro culture). Four hours later, cells were stimulated with 1 ng/ml LPS o/n. TNF-α concentrations in the supernatants were determined by ELISA. Data are mean ± SEM of three independent experiments. (B) aS2 mOVA, together with aS2 AnxA1, aS2 AnxA5, or aS2 AnxA13 were injected i.v. into mice. Mice injected with aS2 mock served as a negative control. After 8 d, the induction of OVA-specific CD8+ T cells in the mesenteric lymph nodes was analyzed by flow cytometry. Errors bars represent mean ± SEM. (C and D) One day after transfer of 1 × 106 CFSE-labeled OT-I T cells, C57BL/6 WT mice were immunized i.v. with 0.5 × 106 aS2 mOVA together with 1 × 106 aS2 AnxA5, aS2 AnxA13, or aS2 mock. After 12–13 d, mice were challenged with 50 μg OVA protein in IFA, and spleens were analyzed 6 d after challenge by flow cytometry for cell numbers of transferred OT-I cells in spleens (C) and IFN-γ secreting CD8+ T cells by ELISPOT (D). n = 18 (aS2 mOVA/aS2 mock), n = 9 (aS2 mOVA/aS2 AnxA5 and aS2 mOVA/aS2 AnxA13). *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

Close modal

Next, AnxA1−/− mice were analyzed for signs of autoimmunity. CD62L/CD44 surface expression of different splenic T cell populations was comparable between WT and AnxA1−/− mice, indicating that there was no change in the activation status of peripheral T cells. In contrast, lpr mice, which served as a positive control for the development of systemic autoimmunity, clearly showed enhanced activation of splenic T cells (Supplemental Fig. 4D). Furthermore, splenomegaly and lymphadenopathy were absent in 5–6-mo-old AnxA1−/− mice (Supplemental Fig. 4E). For the abovementioned investigations (Supplemental Fig. 4D, 4E), cells from female and male mice were used, and no gender-specific differences were noticed. In conclusion, the lack of autoimmunity in AnxA1−/− mice and the unaltered suppressive capacity of ACs from AnxA1−/− mice indicated that AnxA5 and AnxA13 might substitute for the loss of AnxA1 and might act as tolerogenic signals in vivo.

Recently, our group showed that AnxA1 restrains the function and reduces the frequency of Ag-specific CD8+ T cells upon immunization of mice with xenogeneic ACs ectopically expressing AnxA1 (11). To investigate whether AnxA5 and AnxA13 also regulate the induction of Ag-specific T cell responses in vivo, aS2 mOVA were injected into mice, and the anti-OVA T cell response was monitored. Injection of aS2 mOVA induced a cell-mediated immune response and led to the development of OVA-specific CD8+ T cells (Fig. 6B, Supplemental Fig. 4F). Notably, coinjection of apoptotic S2 cells overexpressing AnxA1, AnxA5, or AnxA13 strongly inhibited the development of OVA-specific CD8+ T cells (Fig. 6B). We further analyzed the effect of Anxs on the surface of ACs with regard to the stimulation and fate of Ag-specific CD8+ T cells in a transfer experiment. We injected CFSE-labeled OVA-specific OT-I T cells into C57BL/6 WT mice and followed proliferation and expansion of this population with the help of the congenic marker Thy-1.1. We detected no difference in proliferation or OT-I population size 6 d after immunization (data not shown). However, when challenging with OVA protein 12 d after the primary immunization, we detected a significant reduction in the absolute number of OT-I cells in mice coimmunized with aS2 AnxA5 or aS2 AnxA13 (Fig. 6C). Importantly, the immune response in mice coimmunized with Anx-overexpressing apoptotic S2 cells was also functionally impaired, as evidenced by a severely reduced number of IFN-γ–secreting CD8+ T cells (Fig. 6D). Similar results were obtained by ELISA (data not shown). Thus, AnxA5 and AnxA13 negatively regulate the induction of Ag-specific CD8+ T cell responses and, therefore, act as tolerogenic signals on ACs in vivo.

Uptake and processing of self-Ags derived from ACs by DCs in the steady-state and subsequent presentation of self-peptides on MHC class I and MHC class II by tolerogenic DCs are important mechanisms to induce CD4+ and CD8+ T cell tolerance in the periphery (36, 37). Immunosuppression of phagocytes by ACs is mediated by tolerogenic signals on the surface of ACs, including growth arrest-specific gene 6, inactivated complement component 3b, thrombospondin 1, and AnxA1 (10, 11, 3840). In lymphoid organs, the subsequent interaction of tolerogenic DCs and autoreactive T cells induces anergy or deletion, or it may trigger conversion of naive CD4+ T cells into regulatory T cells (4146). Because the knowledge of tolerogenic signals on the surface of ACs is limited, and functional redundancy has been reported for different properties of Anxs, this study aimed at investigating whether Anxs act as redundant tolerogenic signals on ACs.

Upon induction of apoptosis, AnxA1 translocates to the surface of ACs and is recognized by phagocytosing DCs (11). Using a leukemia cell line and stably transfected apoptotic S2 cells, we showed that translocation upon induction of apoptosis is a common feature of AnxA1, AnxA5, and AnxA13 (Fig. 2). To our knowledge, this is the first report showing translocation of AnxA13 to the surface of ACs, whereas translocation of AnxA5 to the surface of rat cardiomyocytes upon treatment with staurosporine or H2O2 was reported previously (47). Externalization of AnxA5 also takes place under pathophysiological conditions like acute myocardial infarction or glomerulonephritis (48, 49). In both cases, passive release from necrotic cells cannot be ruled out. However, this study shows that Anxs are externalized at an early stage of apoptosis preceding the loss of membrane integrity (Fig. 2).

AnxA5 and AnxA13 induce the development of a tolerogenic DC phenotype, as we showed previously for AnxA1 (11). Soluble and membrane-bound AnxA5 and AnxA13 inhibit the TLR-induced secretion of proinflammatory cytokines (i.e., IL-12p40, IL-6, and TNF-α), whereas the secretion of IL-10 is not affected (Fig. 3B–H). TLR-induced upregulation of the costimulatory molecules CD40, CD80, and CD86 was inhibited by Anx pretreatment, whereas surface levels of MHC class I and II molecules and coinhibitory molecules were not altered (Fig. 4A–G). Thus, the phenotype of DCs obtained after Anx incubation resembles the one of DCs after uptake of ACs or upon incubation with AC-derived tolerogenic signals (10, 38, 39, 5052). Unaltered expression of MHC molecules on DCs is a prerequisite for efficient presentation of self-peptides and, thus, is required for deletion or tolerization of autoreactive T cells in the draining lymph nodes (2). In addition, low expression of costimulatory molecules, especially CD40, was described as a decisive feature of tolerogenic DCs (53, 54). Inhibition of TLR-induced upregulation of costimulatory molecules was observed after incubation of BMDCs with the tolerogenic signals growth arrest-specific gene 6 or inactivated complement component 3b or upon ligation of receptors that recognize tolerogenic signals, including Mer tyrosine kinase, complement receptor 3, or CD36 (10, 38, 39, 51). Importantly, deletion and functional inactivation of autoreactive CD8+ T cells require the absence of CD40L-induced signaling, as well as the expression of PD-1 ligands PD-L1 and PD-L2, neither of which was affected by Anx pretreatment of BMDCs (46).

Similar to AC uptake or ligation of Mer tyrosine kinase, CD36, or complement receptor 3, preincubation of BMDCs with Anxs results in inhibition of TLR-induced secretion of proinflammatory cytokines, including TNF-α, IL-6, and IL-12p40 (Fig. 3B–H) (10, 38, 39, 50). The low secretion of the Th1-promoting cytokines TNF-α and IL-12p40, as well as of the Th17-promoting cytokine IL-6, implies that Th1 and Th17 priming of naive CD4+ T cells by Anx-treated BMDCs is unlikely.

Tolerance induction by AnxA1, AnxA5, and AnxA13 is independent of DC apoptosis, recognition of Anxs by FPR family members, or inhibition of phospholipase A2 activity. cPLA2 was described to be the key enzyme for signal transduction of inflammation (55, 56). Inhibition of cPLA2 is mediated by specific interaction of cPLA2 and aa 275–346 of AnxA1. Other Anxs, including AnxA5, do not inhibit cPLA2 activity in vitro (57). Because AnxA5 and AnxA13 promote the development of tolerogenic DCs (Figs. 3B–H, 4A–G), inhibition of cPLA2 is not required for suppression of DC activation by Anxs. High amounts of AnxA1 released into the inflammatory fluid accelerate apoptosis of neutrophils and monocytes, which contributes to the resolution of inflammation (5860). AnxA5 can have proapoptotic or antiapoptotic effects, depending on the cell type investigated (47, 61). Importantly, preincubation of BMDCs with AnxA1, AnxA5, or AnxA13 did not influence apoptosis progression, ruling out DC apoptosis as a mechanism for tolerance induction by Anxs (Fig. 4H, Supplemental Fig. 3C).

N-terminal peptides and the AF-2 sequence in the core domain were implicated in mediating anti-inflammatory effects of AnxA1 (15, 21, 62, 63). However, several points make it unlikely that recognition by FPR family members contributes to the tolerogenic effect of Anxs on BMDCs. First, the Anx core domain, which is highly conserved among Anx family members, is sufficient to inhibit TLR-induced cytokine secretion of BMDCs (Fig. 1B). Second, mutation of the AF-2 sequence, the only other peptide sequence binding to FPR family members apart from the AnxA1 N terminus, and ablation of the N-terminal domain of AnxA1 do not abrogate AnxA1-induced suppression of TLR-induced DC activation (Fig. 1B) (24). Third, AnxA5 and AnxA13, which have not been shown to bind to FPR family members, possess suppressive activity comparable to AnxA1 (Fig. 3B–D). Fourth, pan-specific antagonists of FPR family members do not abrogate the suppressive effect elicited by Anxs (Figs. 1E, 5B). Finally, phosphorylation of ERK, induced upon binding of N-terminal peptides of AnxA1 to FPR family members, could not be detected in BMDCs upon treatment with AnxA1, AnxA5, or AnxA13 (Figs. 1F, 5A). The lower levels of FPR family members expressed on DCs and/or different signaling thresholds of DCs compared to neutrophils may explain the differential effects noted for the individual domains of Anxs, depending on the cell type or biological setting (Fig. 1C). The data presented in this article support the existence of a different, hitherto unknown receptor involved in the recognition of Anxs on DCs.

Our results show that several Anx family members translocate to the surface of ACs and induce tolerogenic signaling upon recognition by their cognate receptor on DCs. The absence of severe phenotypes in mice deficient in individual Anx family members and almost identical tissue-specific expression patterns of most similar Anx genes suggest that Anxs have redundant functions in vivo (12, 25, 2831). In fact, redundant functions for different Anxs have been reported in the context of membrane trafficking, inhibition of phospholipase activity, and blood coagulation in vitro (12). In a noninflammatory or noninfectious setting and on a mouse background not prone to develop autoimmunity, AnxA1−/− mice lack symptoms of autoimmune or chronic inflammatory diseases (Fig. 6). This is likely due to upregulation of other, equally suppressive Anx family members in AnxA1−/− mice (Fig. 6) (32). By analyzing AnxA1, AnxA5, and AnxA13 we chose representative Anxs that are distributed equally through the phylogenetic tree of vertebrate Anxs, including AnxA13 as the founding member of this protein family (13). As typical representatives of the Anx family, the core domains of murine AnxA1, AnxA5, and AnxA13 show a comparatively high mean amino acid similarity of 55–60% to all other Anx core domains. Therefore, we hypothesize that the immune-suppressive activity residing in the Anx core domain, as identified in our study, is likely shared by more, if not most, members of the Anx family. If so, the ubiquitous expression of the Anx family as a whole (13) could afford protection from autoimmunity to virtually any tissue of the organism.

By analyzing the fate of transferred Ag-specific T cells, we observed that AnxA5 and AnxA13 exert a tolerogenic effect on the surface of ACs, which leads to a significant reduction in the population size and activity of Ag-specific CD8+ T cells. Regarding CD8+ T cell activity, AnxA5 and AnxA13 recapitulate the effect of AnxA1, as analyzed in our previous study (11). Moreover, these results further elucidate the Anx-mediated mechanism of immune suppression in vivo and suggest a mechanism similar to deletional tolerance, as described by Steinman and colleagues (64).

A better understanding of tolerogenic signals on ACs provides new opportunities to interfere with peripheral tolerance induction for the treatment of autoimmune diseases, chronic inflammatory diseases, and cancer. The clearance of apoptotic tumor cells by tumor-associated macrophages or DCs is associated with the release of anti-inflammatory cytokines and tolerance induction to tumor Ags (65). The immunoregulatory environment established by the tumor and the presentation of tumor Ags in the absence of danger signals may account for the failure of most anticancer chemotherapies to promote curative T cell immunity (66). One powerful approach to counteract the tolerogenic properties of the tumor is to induce immunogenic cell death in which alarmins are released during apoptosis (67). However, the release of alarmins has severe side effects, including promotion of angiogenesis, resistance to chemotherapeutics, chronic inflammation, development of gout, and renal failure (65). Our data can guide the development of an alternative approach: interference with the presentation of tolerogenic signals, such as Anxs, or blockade of those on dying tumor cells with the goal to maximize the impact of cancer immunotherapy.

We thank S. Akira, S. Uematsu, and L. Gissmann for providing Tlr4−/− mice; U. Rescher (University of Münster, Münster, Germany) for providing a eukaryotic FPR1 overexpression plasmid; and N. Garbi and G. Hämmerling for providing OT-I mice and biotin-labeled Y3. We also thank H. Sauter for excellent secretarial assistance.

This work was supported in part by the Helmholtz Alliance on Immunotherapy.

The online version of this article contains supplemental material.

Abbreviations used in this article:

7-AAD

7-aminoactinomycin D

AC

apoptotic cell

AF-2

antiflammin-2

Anx

annexin

aS2 AnxA1

AnxA1-expressing apoptotic S2 cell

aS2 AnxA5

AnxA5-expressing apoptotic S2 cell

aS2 AnxA13

AnxA13-expressing apoptotic S2 cell

aS2 mock

mock-transfected apoptotic S2 cell

aS2 mOVA

apoptotic, membrane-anchored OVA–expressing S2 cell

BM

bone marrow

BMDC

BM-derived DC

cPLA2

cytosolic phospholipase A2

DC

dendritic cell

FPR

N-formyl peptide receptor

lpr

lymphoproliferation

m

mouse

mOVA

membrane-anchored OVA

o/n

overnight

PD-L

programmed cell death ligand

sPLA2

secretory phospholipase A2

S2

Schneider 2

WT

wild-type.

1
Mueller
D. L.
2010
.
Mechanisms maintaining peripheral tolerance.
Nat. Immunol.
11
:
21
27
.
2
Huang
F. P.
,
Platt
N.
,
Wykes
M.
,
Major
J. R.
,
Powell
T. J.
,
Jenkins
C. D.
,
MacPherson
G. G.
.
2000
.
A discrete subpopulation of dendritic cells transports apoptotic intestinal epithelial cells to T cell areas of mesenteric lymph nodes.
J. Exp. Med.
191
:
435
444
.
3
Gregory
C. D.
,
Devitt
A.
.
2004
.
The macrophage and the apoptotic cell: an innate immune interaction viewed simplistically?
Immunology
113
:
1
14
.
4
Bedoui
S.
,
Whitney
P. G.
,
Waithman
J.
,
Eidsmo
L.
,
Wakim
L.
,
Caminschi
I.
,
Allan
R. S.
,
Wojtasiak
M.
,
Shortman
K.
,
Carbone
F. R.
, et al
.
2009
.
Cross-presentation of viral and self antigens by skin-derived CD103+ dendritic cells.
Nat. Immunol.
10
:
488
495
.
5
Bursch
L. S.
,
Wang
L.
,
Igyarto
B.
,
Kissenpfennig
A.
,
Malissen
B.
,
Kaplan
D. H.
,
Hogquist
K. A.
.
2007
.
Identification of a novel population of Langerin+ dendritic cells.
J. Exp. Med.
204
:
3147
3156
.
6
del Rio
M. L.
,
Rodriguez-Barbosa
J. I.
,
Kremmer
E.
,
Förster
R.
.
2007
.
CD103− and CD103+ bronchial lymph node dendritic cells are specialized in presenting and cross-presenting innocuous antigen to CD4+ and CD8+ T cells.
J. Immunol.
178
:
6861
6866
.
7
A-Gonzalez
N.
,
Bensinger
S. J.
,
Hong
C.
,
Beceiro
S.
,
Bradley
M. N.
,
Zelcer
N.
,
Deniz
J.
,
Ramirez
C.
,
Díaz
M.
,
Gallardo
G.
, et al
.
2009
.
Apoptotic cells promote their own clearance and immune tolerance through activation of the nuclear receptor LXR.
Immunity
31
:
245
258
.
8
Scott
R. S.
,
McMahon
E. J.
,
Pop
S. M.
,
Reap
E. A.
,
Caricchio
R.
,
Cohen
P. L.
,
Earp
H. S.
,
Matsushima
G. K.
.
2001
.
Phagocytosis and clearance of apoptotic cells is mediated by MER.
Nature
411
:
207
211
.
9
Kawane
K.
,
Ohtani
M.
,
Miwa
K.
,
Kizawa
T.
,
Kanbara
Y.
,
Yoshioka
Y.
,
Yoshikawa
H.
,
Nagata
S.
.
2006
.
Chronic polyarthritis caused by mammalian DNA that escapes from degradation in macrophages.
Nature
443
:
998
1002
.
10
Wallet
M. A.
,
Sen
P.
,
Flores
R. R.
,
Wang
Y.
,
Yi
Z.
,
Huang
Y.
,
Mathews
C. E.
,
Earp
H. S.
,
Matsushima
G.
,
Wang
B.
,
Tisch
R.
.
2008
.
MerTK is required for apoptotic cell-induced T cell tolerance.
J. Exp. Med.
205
:
219
232
.
11
Weyd
H.
,
Abeler-Dörner
L.
,
Linke
B.
,
Mahr
A.
,
Jahndel
V.
,
Pfrang
S.
,
Schnölzer
M.
,
Falk
C. S.
,
Krammer
P. H.
.
2013
.
Annexin A1 on the surface of early apoptotic cells suppresses CD8+ T cell immunity.
PLoS ONE
8
:
e62449
.
12
Gerke
V.
,
Moss
S. E.
.
2002
.
Annexins: from structure to function.
Physiol. Rev.
82
:
331
371
.
13
Moss
S. E.
,
Morgan
R. O.
.
2004
.
The annexins.
Genome Biol.
5
:
219
.
14
Raynal
P.
,
Pollard
H. B.
.
1994
.
Annexins: the problem of assessing the biological role for a gene family of multifunctional calcium- and phospholipid-binding proteins.
Biochim. Biophys. Acta
1197
:
63
93
.
15
Walther
A.
,
Riehemann
K.
,
Gerke
V.
.
2000
.
A novel ligand of the formyl peptide receptor: annexin I regulates neutrophil extravasation by interacting with the FPR.
Mol. Cell
5
:
831
840
.
16
Ernst
S.
,
Lange
C.
,
Wilbers
A.
,
Goebeler
V.
,
Gerke
V.
,
Rescher
U.
.
2004
.
An annexin 1 N-terminal peptide activates leukocytes by triggering different members of the formyl peptide receptor family.
J. Immunol.
172
:
7669
7676
.
17
Strausbaugh
H. J.
,
Rosen
S. D.
.
2001
.
A potential role for annexin 1 as a physiologic mediator of glucocorticoid-induced L-selectin shedding from myeloid cells.
J. Immunol.
166
:
6294
6300
.
18
Perretti
M.
,
Dalli
J.
.
2009
.
Exploiting the Annexin A1 pathway for the development of novel anti-inflammatory therapeutics.
Br. J. Pharmacol.
158
:
936
946
.
19
Hayhoe
R. P.
,
Kamal
A. M.
,
Solito
E.
,
Flower
R. J.
,
Cooper
D.
,
Perretti
M.
.
2006
.
Annexin 1 and its bioactive peptide inhibit neutrophil-endothelium interactions under flow: indication of distinct receptor involvement.
Blood
107
:
2123
2130
.
20
Pupjalis
D.
,
Goetsch
J.
,
Kottas
D. J.
,
Gerke
V.
,
Rescher
U.
.
2011
.
Annexin A1 released from apoptotic cells acts through formyl peptide receptors to dampen inflammatory monocyte activation via JAK/STAT/SOCS signalling.
EMBO Mol. Med.
3
:
102
114
.
21
Wallner
B. P.
,
Mattaliano
R. J.
,
Hession
C.
,
Cate
R. L.
,
Tizard
R.
,
Sinclair
L. K.
,
Foeller
C.
,
Chow
E. P.
,
Browing
J. L.
,
Ramachandran
K. L.
, et al
.
1986
.
Cloning and expression of human lipocortin, a phospholipase A2 inhibitor with potential anti-inflammatory activity.
Nature
320
:
77
81
.
22
Davidson
F. F.
,
Lister
M. D.
,
Dennis
E. A.
.
1990
.
Binding and inhibition studies on lipocortins using phosphatidylcholine vesicles and phospholipase A2 from snake venom, pancreas, and a macrophage-like cell line.
J. Biol. Chem.
265
:
5602
5609
.
23
Kim
S. W.
,
Rhee
H. J.
,
Ko
J.
,
Kim
Y. J.
,
Kim
H. G.
,
Yang
J. M.
,
Choi
E. C.
,
Na
D. S.
.
2001
.
Inhibition of cytosolic phospholipase A2 by annexin I. Specific interaction model and mapping of the interaction site.
J. Biol. Chem.
276
:
15712
15719
.
24
Kamal
A. M.
,
Hayhoe
R. P.
,
Paramasivam
A.
,
Cooper
D.
,
Flower
R. J.
,
Solito
E.
,
Perretti
M.
.
2006
.
Antiflammin-2 activates the human formyl-peptide receptor like 1.
ScientificWorldJournal
6
:
1375
1384
.
25
Farber
S. A.
,
De Rose
R. A.
,
Olson
E. S.
,
Halpern
M. E.
.
2003
.
The zebrafish annexin gene family.
Genome Res.
13
(
6A
):
1082
1096
.
26
Inaba
K.
,
Inaba
M.
,
Romani
N.
,
Aya
H.
,
Deguchi
M.
,
Ikehara
S.
,
Muramatsu
S.
,
Steinman
R. M.
.
1992
.
Generation of large numbers of dendritic cells from mouse bone marrow cultures supplemented with granulocyte/macrophage colony-stimulating factor.
J. Exp. Med.
176
:
1693
1702
.
27
Naik
S. H.
,
Proietto
A. I.
,
Wilson
N. S.
,
Dakic
A.
,
Schnorrer
P.
,
Fuchsberger
M.
,
Lahoud
M. H.
,
O’Keeffe
M.
,
Shao
Q. X.
,
Chen
W. F.
, et al
.
2005
.
Cutting edge: generation of splenic CD8+ and CD8- dendritic cell equivalents in Fms-like tyrosine kinase 3 ligand bone marrow cultures.
J. Immunol.
174
:
6592
6597
.
28
Brachvogel
B.
,
Dikschas
J.
,
Moch
H.
,
Welzel
H.
,
von der Mark
K.
,
Hofmann
C.
,
Pöschl
E.
.
2003
.
Annexin A5 is not essential for skeletal development.
Mol. Cell. Biol.
23
:
2907
2913
.
29
Hawkins
T. E.
,
Roes
J.
,
Rees
D.
,
Monkhouse
J.
,
Moss
S. E.
.
1999
.
Immunological development and cardiovascular function are normal in annexin VI null mutant mice.
Mol. Cell. Biol.
19
:
8028
8032
.
30
Herr
C.
,
Smyth
N.
,
Ullrich
S.
,
Yun
F.
,
Sasse
P.
,
Hescheler
J.
,
Fleischmann
B.
,
Lasek
K.
,
Brixius
K.
,
Schwinger
R. H.
, et al
.
2001
.
Loss of annexin A7 leads to alterations in frequency-induced shortening of isolated murine cardiomyocytes.
Mol. Cell. Biol.
21
:
4119
4128
.
31
Ling
Q.
,
Jacovina
A. T.
,
Deora
A.
,
Febbraio
M.
,
Simantov
R.
,
Silverstein
R. L.
,
Hempstead
B.
,
Mark
W. H.
,
Hajjar
K. A.
.
2004
.
Annexin II regulates fibrin homeostasis and neoangiogenesis in vivo.
J. Clin. Invest.
113
:
38
48
.
32
Hannon
R.
,
Croxtall
J. D.
,
Getting
S. J.
,
Roviezzo
F.
,
Yona
S.
,
Paul-Clark
M. J.
,
Gavins
F. N.
,
Perretti
M.
,
Morris
J. F.
,
Buckingham
J. C.
,
Flower
R. J.
.
2003
.
Aberrant inflammation and resistance to glucocorticoids in annexin 1−/− mouse.
FASEB J.
17
:
253
255
.
33
Iglesias
J. M.
,
Morgan
R. O.
,
Jenkins
N. A.
,
Copeland
N. G.
,
Gilbert
D. J.
,
Fernandez
M. P.
.
2002
.
Comparative genetics and evolution of annexin A13 as the founder gene of vertebrate annexins.
Mol. Biol. Evol.
19
:
608
618
.
34
Iyoda
T.
,
Shimoyama
S.
,
Liu
K.
,
Omatsu
Y.
,
Akiyama
Y.
,
Maeda
Y.
,
Takahara
K.
,
Steinman
R. M.
,
Inaba
K.
.
2002
.
The CD8+ dendritic cell subset selectively endocytoses dying cells in culture and in vivo.
J. Exp. Med.
195
:
1289
1302
.
35
Ip
W. K.
,
Lau
Y. L.
.
2004
.
Distinct maturation of, but not migration between, human monocyte-derived dendritic cells upon ingestion of apoptotic cells of early or late phases.
J. Immunol.
173
:
189
196
.
36
Stuart
L. M.
,
Lucas
M.
,
Simpson
C.
,
Lamb
J.
,
Savill
J.
,
Lacy-Hulbert
A.
.
2002
.
Inhibitory effects of apoptotic cell ingestion upon endotoxin-driven myeloid dendritic cell maturation.
J. Immunol.
168
:
1627
1635
.
37
Liu
K.
,
Iyoda
T.
,
Saternus
M.
,
Kimura
Y.
,
Inaba
K.
,
Steinman
R. M.
.
2002
.
Immune tolerance after delivery of dying cells to dendritic cells in situ.
J. Exp. Med.
196
:
1091
1097
.
38
Skoberne
M.
,
Somersan
S.
,
Almodovar
W.
,
Truong
T.
,
Petrova
K.
,
Henson
P. M.
,
Bhardwaj
N.
.
2006
.
The apoptotic-cell receptor CR3, but not alphavbeta5, is a regulator of human dendritic-cell immunostimulatory function.
Blood
108
:
947
955
.
39
Urban
B. C.
,
Willcox
N.
,
Roberts
D. J.
.
2001
.
A role for CD36 in the regulation of dendritic cell function.
Proc. Natl. Acad. Sci. USA
98
:
8750
8755
.
40
Krispin
A.
,
Bledi
Y.
,
Atallah
M.
,
Trahtemberg
U.
,
Verbovetski
I.
,
Nahari
E.
,
Zelig
O.
,
Linial
M.
,
Mevorach
D.
.
2006
.
Apoptotic cell thrombospondin-1 and heparin-binding domain lead to dendritic-cell phagocytic and tolerizing states.
Blood
108
:
3580
3589
.
41
Adler
A. J.
,
Marsh
D. W.
,
Yochum
G. S.
,
Guzzo
J. L.
,
Nigam
A.
,
Nelson
W. G.
,
Pardoll
D. M.
.
1998
.
CD4+ T cell tolerance to parenchymal self-antigens requires presentation by bone marrow-derived antigen-presenting cells.
J. Exp. Med.
187
:
1555
1564
.
42
Akbari
O.
,
DeKruyff
R. H.
,
Umetsu
D. T.
.
2001
.
Pulmonary dendritic cells producing IL-10 mediate tolerance induced by respiratory exposure to antigen.
Nat. Immunol.
2
:
725
731
.
43
Hawiger
D.
,
Inaba
K.
,
Dorsett
Y.
,
Guo
M.
,
Mahnke
K.
,
Rivera
M.
,
Ravetch
J. V.
,
Steinman
R. M.
,
Nussenzweig
M. C.
.
2001
.
Dendritic cells induce peripheral T cell unresponsiveness under steady state conditions in vivo.
J. Exp. Med.
194
:
769
779
.
44
Janssen
E. M.
,
Droin
N. M.
,
Lemmens
E. E.
,
Pinkoski
M. J.
,
Bensinger
S. J.
,
Ehst
B. D.
,
Griffith
T. S.
,
Green
D. R.
,
Schoenberger
S. P.
.
2005
.
CD4+ T-cell help controls CD8+ T-cell memory via TRAIL-mediated activation-induced cell death.
Nature
434
:
88
93
.
45
Kawahata
K.
,
Misaki
Y.
,
Yamauchi
M.
,
Tsunekawa
S.
,
Setoguchi
K.
,
Miyazaki
J.
,
Yamamoto
K.
.
2002
.
Generation of CD4(+)CD25(+) regulatory T cells from autoreactive T cells simultaneously with their negative selection in the thymus and from nonautoreactive T cells by endogenous TCR expression.
J. Immunol.
168
:
4399
4405
.
46
Probst
H. C.
,
McCoy
K.
,
Okazaki
T.
,
Honjo
T.
,
van den Broek
M.
.
2005
.
Resting dendritic cells induce peripheral CD8+ T cell tolerance through PD-1 and CTLA-4.
Nat. Immunol.
6
:
280
286
.
47
Monceau
V.
,
Belikova
Y.
,
Kratassiouk
G.
,
Charue
D.
,
Camors
E.
,
Communal
C.
,
Trouvé
P.
,
Russo-Marie
F.
,
Charlemagne
D.
.
2004
.
Externalization of endogenous annexin A5 participates in apoptosis of rat cardiomyocytes.
Cardiovasc. Res.
64
:
496
506
.
48
Kaneko
N.
,
Matsuda
R.
,
Hosoda
S.
,
Kajita
T.
,
Ohta
Y.
.
1996
.
Measurement of plasma annexin V by ELISA in the early detection of acute myocardial infarction.
Clin. Chim. Acta
251
:
65
80
.
49
Matsuda
R.
,
Kaneko
N.
,
Horikawa
Y.
,
Chiwaki
F.
,
Shinozaki
M.
,
Ieiri
T.
,
Suzuki
T.
,
Ogawa
N.
.
2001
.
Localization of annexin V in rat normal kidney and experimental glomerulonephritis.
Res. Exp. Med. (Berl.)
200
:
77
92
.
50
Sen
P.
,
Wallet
M. A.
,
Yi
Z.
,
Huang
Y.
,
Henderson
M.
,
Mathews
C. E.
,
Earp
H. S.
,
Matsushima
G.
,
Baldwin
A. S.
 Jr.
,
Tisch
R. M.
.
2007
.
Apoptotic cells induce Mer tyrosine kinase-dependent blockade of NF-kappaB activation in dendritic cells.
Blood
109
:
653
660
.
51
Verbovetski
I.
,
Bychkov
H.
,
Trahtemberg
U.
,
Shapira
I.
,
Hareuveni
M.
,
Ben-Tal
O.
,
Kutikov
I.
,
Gill
O.
,
Mevorach
D.
.
2002
.
Opsonization of apoptotic cells by autologous iC3b facilitates clearance by immature dendritic cells, down-regulates DR and CD86, and up-regulates CC chemokine receptor 7.
J. Exp. Med.
196
:
1553
1561
.
52
Voll
R. E.
,
Herrmann
M.
,
Roth
E. A.
,
Stach
C.
,
Kalden
J. R.
,
Girkontaite
I.
.
1997
.
Immunosuppressive effects of apoptotic cells.
Nature
390
:
350
351
.
53
Fujii
S.
,
Liu
K.
,
Smith
C.
,
Bonito
A. J.
,
Steinman
R. M.
.
2004
.
The linkage of innate to adaptive immunity via maturing dendritic cells in vivo requires CD40 ligation in addition to antigen presentation and CD80/86 costimulation.
J. Exp. Med.
199
:
1607
1618
.
54
Joffre
O.
,
Nolte
M. A.
,
Spörri
R.
,
Reis e Sousa
C.
.
2009
.
Inflammatory signals in dendritic cell activation and the induction of adaptive immunity.
Immunol. Rev.
227
:
234
247
.
55
Serhan
C. N.
,
Brain
S. D.
,
Buckley
C. D.
,
Gilroy
D. W.
,
Haslett
C.
,
O’Neill
L. A.
,
Perretti
M.
,
Rossi
A. G.
,
Wallace
J. L.
.
2007
.
Resolution of inflammation: state of the art, definitions and terms.
FASEB J.
21
:
325
332
.
56
Serhan
C. N.
,
Haeggström
J. Z.
,
Leslie
C. C.
.
1996
.
Lipid mediator networks in cell signaling: update and impact of cytokines.
FASEB J.
10
:
1147
1158
.
57
Kim
K. M.
,
Kim
D. K.
,
Park
Y. M.
,
Kim
C. K.
,
Na
D. S.
.
1994
.
Annexin-I inhibits phospholipase A2 by specific interaction, not by substrate depletion.
FEBS Lett.
343
:
251
255
.
58
Solito
E.
,
Mulla
A.
,
Morris
J. F.
,
Christian
H. C.
,
Flower
R. J.
,
Buckingham
J. C.
.
2003
.
Dexamethasone induces rapid serine-phosphorylation and membrane translocation of annexin 1 in a human folliculostellate cell line via a novel nongenomic mechanism involving the glucocorticoid receptor, protein kinase C, phosphatidylinositol 3-kinase, and mitogen-activated protein kinase.
Endocrinology
144
:
1164
1174
.
59
Solito
E.
,
de Coupade
C.
,
Canaider
S.
,
Goulding
N. J.
,
Perretti
M.
.
2001
.
Transfection of annexin 1 in monocytic cells produces a high degree of spontaneous and stimulated apoptosis associated with caspase-3 activation.
Br. J. Pharmacol.
133
:
217
228
.
60
Solito
E.
,
Kamal
A.
,
Russo-Marie
F.
,
Buckingham
J. C.
,
Marullo
S.
,
Perretti
M.
.
2003
.
A novel calcium-dependent proapoptotic effect of annexin 1 on human neutrophils.
FASEB J.
17
:
1544
1546
.
61
Gidon-Jeangirard
C.
,
Solito
E.
,
Hofmann
A.
,
Russo-Marie
F.
,
Freyssinet
J. M.
,
Martínez
M. C.
.
1999
.
Annexin V counteracts apoptosis while inducing Ca(2+) influx in human lymphocytic T cells.
Biochem. Biophys. Res. Commun.
265
:
709
715
.
62
Miele
L.
,
Cordella-Miele
E.
,
Facchiano
A.
,
Mukherjee
A. B.
.
1988
.
Novel anti-inflammatory peptides from the region of highest similarity between uteroglobin and lipocortin I.
Nature
335
:
726
730
.
63
Perretti
M.
,
Chiang
N.
,
La
M.
,
Fierro
I. M.
,
Marullo
S.
,
Getting
S. J.
,
Solito
E.
,
Serhan
C. N.
.
2002
.
Endogenous lipid- and peptide-derived anti-inflammatory pathways generated with glucocorticoid and aspirin treatment activate the lipoxin A4 receptor.
Nat. Med.
8
:
1296
1302
.
64
Bonifaz
L.
,
Bonnyay
D.
,
Mahnke
K.
,
Rivera
M.
,
Nussenzweig
M. C.
,
Steinman
R. M.
.
2002
.
Efficient targeting of protein antigen to the dendritic cell receptor DEC-205 in the steady state leads to antigen presentation on major histocompatibility complex class I products and peripheral CD8+ T cell tolerance.
J. Exp. Med.
196
:
1627
1638
.
65
Haynes
N. M.
,
van der Most
R. G.
,
Lake
R. A.
,
Smyth
M. J.
.
2008
.
Immunogenic anti-cancer chemotherapy as an emerging concept.
Curr. Opin. Immunol.
20
:
545
557
.
66
van der Most
R. G.
,
Currie
A. J.
,
Robinson
B. W.
,
Lake
R. A.
.
2008
.
Decoding dangerous death: how cytotoxic chemotherapy invokes inflammation, immunity or nothing at all.
Cell Death Differ.
15
:
13
20
.
67
Garg
A. D.
,
Nowis
D.
,
Golab
J.
,
Vandenabeele
P.
,
Krysko
D. V.
,
Agostinis
P.
.
2010
.
Immunogenic cell death, DAMPs and anticancer therapeutics: an emerging amalgamation.
Biochim. Biophys. Acta
1805
:
53
71
.

The authors have no financial conflicts of interest.

Supplementary data