We previously showed that the T cell activation inhibitor, mitochondrial (Tcaim) is highly expressed in grafts of tolerance-developing transplant recipients and that the encoded protein is localized within mitochondria. In this study, we show that CD11c+ dendritic cells (DCs), as main producers of TCAIM, downregulate Tcaim expression after LPS stimulation or in vivo alloantigen challenge. LPS-stimulated TCAIM-overexpressing bone marrow–derived DC (BMDCs) have a reduced capacity to induce proliferation of and cytokine expression by cocultured allogeneic T cells; this is not due to diminished upregulation of MHC or costimulatory molecules. Transcriptional profiling also revealed normal LPS-mediated upregulation of the majority of genes involved in TLR signaling. However, TCAIM BMDCs did not induce Il2 mRNA expression upon LPS stimulation in comparison with Control-BMDCs. In addition, TCAIM overexpression abolished LPS-mediated Ca2+ influx and mitochondrial reactive oxygen species formation. Addition of IL-2 to BMDC–T cell cocultures restored the priming capacity of TCAIM BMDCs for cocultured allogeneic CD8+ T cells. Furthermore, BMDCs of IL-2–deficient mice showed similarly abolished LPS-induced T cell priming as TCAIM-overexpressing wild type BMDCs. Thus, TCAIM interferes with TLR4 signaling in BMDCs and subsequently impairs their T cell priming capacity, which supports its role for tolerance induction.

T cell activation inhibitor, mitochondrial (Tcaim; also known as Toag-1) is highly expressed in graft and blood of tolerance-developing allogeneic kidney and heart graft recipients. In contrast, 3–5 days prior to graft rejection Tcaim mRNA is downregulated up to 10-fold in both compartments (1, 2). TCAIM is highly conserved among different species, but no homologies to protein families of known functions were detectable. The protein contains a mitochondrial leader sequence and a J-domain within the N- and C terminus, respectively. So far little is known about the specific function of TCAIM. It is highly expressed in CD11c+ dendritic cells (DCs) and CD4+ T cells (1). Keeren et al. (3) showed that TCAIM is exclusively localized within mitochondria. Increasing data identified mitochondria to be crucial for innate immune signaling. West et al. (4) demonstrated that TNF receptor associated factor (TRAF6) translocates to mitochondria in macrophages after TLR1/2/4 stimulation. Subsequently, TRAF6 ubiquitinates the mitochondrial protein evolutionarily conserved signaling intermediate in Toll pathway (ECSIT) and thereby induces mitochondrial reactive oxygen species (mROS) production (4). ECSIT is involved in complex I assembly (5), which is the main producer of ROS (6). mROS potentiate MAPK signaling by inactivating MAPK phosphatases (7). Consequently, protein-modification by ROS can result in a variety of functional changes, such as activation, inactivation, or multimerization of proteins (8).

As professional Ag-presenting cells, DCs communicate between innate and adaptive immunity. Upon activation within the tissue, they migrate to draining lymph nodes. Within the lymph nodes, DCs activate Ag-specific T cells because of their high expression levels of costimulatory proteins (e.g., CD86, CD80), MHC class II (MHC-II) molecules and increased secretion of proinflammatory cytokines (9). It has also been reported that DCs are able to produce IL-2 early upon TLR-mediated activation (10, 11). This might explain the high T cell priming capacity of DCs, because they are supposed to be the only IL-2–producing cells in the myeloid compartment (12, 13). Il2 expression is initiated upon activation of NFAT, which in turn is controlled via dephosphorylation by calcineurin. In DCs, calcineurin is activated by an increased intracellular Ca2+ concentration, initiated by CD14 signaling or Dectin-1 ligand binding (1416).

We studied the effect of Tcaim overexpression on the priming capacity of LPS-matured murine bone marrow–derived DCs (TCAIM-BMDCs) as compared with control transduced BMDCs (Control-BMDCs). We showed that LPS- but not polyinosinic-polycytidylic acid (poly-IC)–stimulated TCAIM-BMDCs have a reduced priming capacity for both CD4+ and CD8+ T cells. Interestingly, this was not due to reduced LPS-mediated up-regulation of MHC-II and CD86 expression or IL-6 production. In contrast, TCAIM abolished LPS-mediated Ca2+ influx, mROS formation and diminished Il2 expression. LPS-mediated Il2 expression was instrumental for the priming capacity of BMDCs. Addition of IL-2 restored the LPS-induced T cell priming capacity of TCAIM-BMDCs for CD8+ T cells. Thus, TCAIM interferes with crucial LPS-signaling steps at the mitochondria and may thereby inhibit inflammation.

Male BALB/c (H-2d) and C57BL/6 (H-2b) mice were purchased from Charles River Laboratories. Mice were 6–8 wk of age and were allowed free access to food and water. The mice were kept according to German Guidelines on the use of animals in research (from the Berliner Sentsverwaltung).

C57BL/6 mice received a transplant from tail skin of BALB/c mice as previously reported (17). After 2 and 7 d, spleens and lymph nodes were collected and DC subset sorted on a FACSAria II cell sorter (BD Biosciences, Heidelberg, Germany) according to the gating strategy depicted in Supplemental Fig. 1.

BMDCs were produced using a standard protocol as described by Inaba et al. (18). Briefly, C57BL/6 bone marrow cells were cultured in six-well non–tissue-treated culture plates (Falcon) in complete medium (RPMI 1640 with stable glutamine supplemented with 10% superior FBS and 1% penicillin–streptomycin; Biochrome). Recombinant mouse GM-CSF (6 ng/ml; Miltenyi Biotec) was added, and medium was changed every other day.

Immature DCs were transduced on day 6 with a TCAIM encoding adenovirus (TCAIM-BMDCs) or a control virus encoding Egfp (Control-BMDCs). To increase the transduction-rate 8 μg/ml Polybrene (Sigma-Aldrich) was added to the mixture. The mixture was centrifuged for 77 min, 3500 rpm, and 14°C. Cells were placed carefully for an additional 120 min in the incubator (37°C, 5% CO2, and 90% humidity). The maturation was induced on day 7 by adding 1 μg/ml LPS or 50 μg/ml poly-IC for 24 h (both from Sigma-Aldrich).

Lymph node cells of BALB/c mice were harvested. The organs were homogenized through a 100-μm cell strainer. The cell suspension was filtered using a 40-μm cell strainer after centrifugation. Cells were used as alloreactive responder cells in cocultures with LPS-stimulated transduced C57BL/6 BMDCs. To quantify the proliferation of CD4+ or CD8+ T cells a maximum of 2 × 107 cells was stained with 10 μM Cell Proliferation Dye eFluor 450 (CPD450; eBioscience) for 20 min at room temperature in the dark. Cells were washed once. After mixing BMDCs (4 × 104) and CPD450-labeled responder cells (5 × 105) in a 96-well plate, cells were incubated for 5 d. In some cocultures, recombinant mouse IL-2 (Peprotech) was added in increasing concentrations (20 and 200 ng/ml).

Coculture: intracellular cytokine staining.

Cells were washed once with medium and restimulated for 4 h at 37°C with 1 μg/ml ionomycin and 10 ng/ml PMA (Biotrend Chemikalien). After 2 h, 2 μg/ml Brefeldin A (Sigma-Aldrich) was added. Cells were stained for live dead using Fixable Viability Dye eFluor 506 (eBioscience) and subsequently for surface expression of CD3e, CD4, and CD8a at 4°C for 20 min. Cells were fixed and permeabilized with the BD Biosciences Cytofix/Cytoperm Fixation/Permeabilization Kit. For intracellular staining following Abs were used: CD3e-FITC, CD4-Pacific Blue, CD8a-allophycocyanin-Cy7, IFNγ-allophycocyanin (all from BioLegend), and the staining was performed for 30 min at 4°C. Samples were measured on an LSR II (BD Biosciences) and analyzed using the FlowJo software (Tree Star).

Coculture: surface staining and proliferation determination.

On day 5 of coculture CPD450-labeled lymph node cells were harvested carefully. They were stained for live dead and then for surface expression of CD3e, CD4, CD8a, and CD44 as described above (CD3e-FITC, CD4-Alexa Fluor 700, CD8a-allophycocyanin-Cy7, and CD44-PeCy7; all from BioLegend). Responder cell proliferation was measured by the dilution of CPD450 intensity. Surface expression and proliferation was measured on an LSR Fortessa (BD Biosciences) and analyzed using FlowJo software.

Splenocytes and lymph node cells collected from naive C57BL/6 mice or transplant recipient mice for sorting of DC subsets were stained for surface expression of a lineage mixture (CD3/CD19/CD49b/F4-80-PB), CD11c-PerCP, B220-AF700, CD199-PE-Cy7, CD64-allophycocyanin, Ly6C-FITC, CD4-BV785, CD8a-BV650 (all from BioLegend), and CD103-PE (eBioscience).

BMDCs were harvested with ice-cold PBS and stained for surface expression of CD11c, IA(b) and CD86 (CD11c-allophycocyanin, IA(b)-PE and CD86-Biotin, Streptavidin-PerCP) (all from eBioscience). The cells were measured on a FACSCalibur (BD Biosciences) and analyzed using FlowJo software.

The cytokine concentrations in the supernatants taken 6, 12, and 24 h after 1 μg/ml LPS or 50 μg/ml poly-IC stimulation were determined using a Cytometric Bead Assay from Bender MedSystem according to the FlowCytomix Mouse/Rat Basic Kit instructions. We measured TNF-α and IL-6 on a FACSCalibur (BD Biosciences) and quantified the concentrations using the FlowCytomix Pro software (Bender Medsystem). As the sensitivity for mouse IL-2 with the CBA technology is low, we used the Luminex xMAP Technology. Supernatants were treated exactly according to the Milliplex MAP Kit protocol (Millipore) and were analyzed with the Bio-Plex200 System (Bio-Rad).

BMDCs were stimulated for 24 h with 1 μg/ml LPS or not. After a washing step with prewarmed PBS, BMDCs were incubated with 150 nM tetramethylrhodamine-methyl ester (TMRM) (Invitrogen) in PBS containing 2% BSA for 15 min at room temperature in the dark. After a washing step with a 150-pM TMRM solution, cells were stained with CD11c-allophycocyanin and measured within 30 min on a FACSCalibur (BD Biosciences). Data were analyzed with FlowJo software.

BMDCs were stimulated for 1 and 3 h with 1 μg/ml LPS or not. After a washing step with prewarmed PBS, BMDCs were incubated with 5 μM final concentration MitoSox (Molecular Probes) in HBSS containing 1 nM calcium, 1 mM magnesium. After a washing step with PBS/10% FCS cells were additionally stained with CD11c-allophycocyanin and measured within 30 min on a FACSCalibur (BD Biosciences). Data were analyzed with FlowJo software.

BMDCs were resuspended in prewarmed cell loading medium (HBSS containing 1 nM calcium, 1 mM magnesium, 1% FBS, and 0.5% BSA) and 4 mM probenecid (Santa Cruz). Cells were incubated for 30 min at 37°C with 2 μg/ml Indo-1 AM (Life Technologies). After two washing steps with cell loading medium at room temperature, cells were stained at 4°C with CD11c-allophycocyanin. Cells were rested and warmed to room temperature before measuring. The LSRFortessa (BD Biosciences) was used to quantify first the calcium flux without stimulus. Afterwards, the same cells were stimulated with 10 μg/ml LPS or 1 μg/ml ionomycin and immediately recorded.

After a surface staining step (CD11c-allophycocyanin), BMDCs were washed twice with cold PBS. Cell pellets were resuspended in 1× Annexin-V binding Buffer. Cells were stained with Annexin-V-PE and 7-aminoactinomycin D according to the protocol (Annexin-V-PE Apoptosis Detection Kit I; BD Biosciences) and measured within 1 h using an LSRFortessa (BD Biosciences).

BMDCs were stimulated 2 d after transduction with 1 μg/ml LPS for 1 and 12 h. RNA was extracted using NucleoSpin RNA II (Macherey-Nagel) following manufacturer’s instructions. In total, 3 μg RNA was used for cDNA synthesis according to the QuantiTect Reverse Transcription (Qiagen) manual. The Mouse Toll-Like Receptor Signaling Pathway Microarray (Qiagen) was performed following the instructions with some modifications. cDNA synthesis reaction mix (60 μl) was diluted with 60 μl RNase-free H2O. Diluted cDNA was mixed with additional H2O and SYBR Green PCR Master Mix (Life Technologies). The experimental mixture (25 μl) was pipetted to each well of the 96-well PCR array. A two-step cycling program was started using an ABI 7500 Sequence Detection System. Raw data were analyzed with 7500 System Software Version 1.3.1.2.1 (ABI). Mouse hypoxanthine phosphoribosyltransferase (mHprt) was used as a housekeeping gene.

All steps of the analysis were performed in R 2.15.0. The quantitative real-time PCR (QRT-PCR) data were normalized by subtracting the corresponding Ct-values of mHprt as housekeeping gene. The resulting ΔCt values of all target genes present on the arrays were used in a principle component analysis. The calculations were performed by a single value decomposition of the centered, not-scaled data matrix. The two first principle components are shown in Fig. 6B. Variances over all samples were calculated for each target gene. Highly variable genes with a variance ≥4 were selected and used in subsequent clustering; therefore, data were scaled and a hierarchical clustering was performed using euclidean distances and a complete linkage.

FIGURE 6.

Gene array analysis reveals an abolished Il2 induction in TCAIM-BMDCs. (A) Analysis of gene expression using a mouse TLR signaling pathway PCR array in Tcaim overexpressing BMDCs upon LPS activation. Unsupervised heat map of three independent experiments, in which TCAIM (T) overexpressing and control-transduced BMDCs (C) were left unstimulated or stimulated for 1 and 12 h with 1 μg/ml LPS. From the 84 genes analyzed, the expression of the 16 genes shown was regulated upon LPS stimulation, normalized to hypoxanthine-guanine phosphoribosyltransferase (HPRT), when setting the variance threshold (v > 4) high. (B) Depicted is a principal component analysis of three independent experiments. Shown are the single experiment gene expression results normalized to HPRT of TCAIM-BMDCs (triangle) or Control-BMDCs (circle) at the following time points: 1 h (orange) and 12 h (red) LPS stimulated or unstimulated (blue). (C) Il2 mRNA expression after LPS and poly-IC stimulation. TCAIM- and Control-BMDCs were stimulated with 1 μg/ml LPS or 50 μg/ml poly-IC for the indicated time points (0, 1, and 12 h). Shown are the Il2 mRNA expression results of n = 4–8 independent experiments, as mean ± SEM. **p < 0.01, ***p < 0.001. (D) IL-2 protein secretion into supernatants of LPS-stimulated TCAIM- and Control-BMDCs. Shown are the results of eight independent experiments as mean ± SEM. (E) Indo-1–labeled TCAIM- or Control-BMDCs were stimulated with 10 μg/ml LPS or 1 μg/ml ionomycin. Ca2+ influx is illustrated as the ratio of Indo-1 violet and Indo-1 blue over time. Depicted are exemplary histograms of pregated CD11c+ BMDCs. (F) Summary of peak influx levels of three independent Ca2+ influx measurements.

FIGURE 6.

Gene array analysis reveals an abolished Il2 induction in TCAIM-BMDCs. (A) Analysis of gene expression using a mouse TLR signaling pathway PCR array in Tcaim overexpressing BMDCs upon LPS activation. Unsupervised heat map of three independent experiments, in which TCAIM (T) overexpressing and control-transduced BMDCs (C) were left unstimulated or stimulated for 1 and 12 h with 1 μg/ml LPS. From the 84 genes analyzed, the expression of the 16 genes shown was regulated upon LPS stimulation, normalized to hypoxanthine-guanine phosphoribosyltransferase (HPRT), when setting the variance threshold (v > 4) high. (B) Depicted is a principal component analysis of three independent experiments. Shown are the single experiment gene expression results normalized to HPRT of TCAIM-BMDCs (triangle) or Control-BMDCs (circle) at the following time points: 1 h (orange) and 12 h (red) LPS stimulated or unstimulated (blue). (C) Il2 mRNA expression after LPS and poly-IC stimulation. TCAIM- and Control-BMDCs were stimulated with 1 μg/ml LPS or 50 μg/ml poly-IC for the indicated time points (0, 1, and 12 h). Shown are the Il2 mRNA expression results of n = 4–8 independent experiments, as mean ± SEM. **p < 0.01, ***p < 0.001. (D) IL-2 protein secretion into supernatants of LPS-stimulated TCAIM- and Control-BMDCs. Shown are the results of eight independent experiments as mean ± SEM. (E) Indo-1–labeled TCAIM- or Control-BMDCs were stimulated with 10 μg/ml LPS or 1 μg/ml ionomycin. Ca2+ influx is illustrated as the ratio of Indo-1 violet and Indo-1 blue over time. Depicted are exemplary histograms of pregated CD11c+ BMDCs. (F) Summary of peak influx levels of three independent Ca2+ influx measurements.

Close modal

RNA from BMDCs stimulated with LPS or poly-IC or from sorted DC subsets was isolated as described before. Up to 1 μg RNA was used for cDNA synthesis. QRT-PCR was done with TaqMan Universal PCR Master Mix (ABI) using FAM-TAMRA as reporter fluorescence and mHprt (MWG Biotech AG) as a normalizing control. The 7500 Real-Time PCR system was used to detect gene expression for mouse tcaim or IL-2 (mIl2). The primer sequences used to quantify mouse tcaim expression have been published previously (19), whereas the primer sequences used to quantify mouse mIl2 are as follows: Il2 sense 5′-TCGCCAGTCAAGAGCTTCAGACAAGCA-3′, anti-sense 5′-CATGCCGCAGAGGTCCAA-3′ (MWG Biotech AG). Primers applied for the detection of mHprt were reported previously (1). Each reaction was carried out in duplicate. For evaluation we used 7500 System SDS Software.

Transduced BMDCs were treated on day 7 overnight (14 h) with 1 μM Rotenone (Enzo Life Sciences). In the morning, complete media was exchanged and the BMDCs were stimulated with 1 μg/ml LPS for 24 h or left unstimulated for coculture experiments. For Il2 mRNA expression studies, Rotenone-treated cells were left untreated or stimulated with 1 μg/ml LPS for 1 h.

One day after transduction cells were seeded in a 96-well white-wall plate (Greiner Bio-One) at 70% confluency and incubated overnight. Recombinant baculovirus (30 μl) encoding a construct of a consensus sequence for one of the following transcription factors NF κ-light-chain-enhancer of activated B cells (NF-κB), NFAT, activating protein-1 (AP1) or CCAAT/enhancer binding proteins (C/EBP) followed by a luciferase reporter gene (Signosis) were added directly to the corresponding wells. Twenty-four hours later, cells were left untreated or were stimulated for 30 min and 3 h with 1 μg/ml LPS. After treatment, the media was carefully removed and cells were washed with 100 μl PBS. Cells were incubated with 40 μl passive lysis buffer (Promega) for 15 min at room temperature. Luciferase Assay Substrate (100 μl; Promega) was added and mixed gently. Luminescence was measured using Mithras LB 940 (Berthold Technologies) immediately after substrate addition.

Flow cytometry data were analyzed with FlowJo software version 9.4.3. The analyses were performed using R 2.15.0. We have used two-way ANOVA with multiple comparisons to analyze the data. Confidence intervals and adjusted p values were calculated using the Studentized range statistic (Tukey honest significant difference method). A p value < 0.05 was considered to be statistically significant (*p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001, ****p ≤ 0.0001). Data are expressed as mean ± SEM if not mentioned in the figure legends.

First, we determined whether inflammatory stimuli influence Tcaim expression in DCs. Therefore, BMDCs were stimulated with LPS for 1, 3, 6, 12 ,and 24 h, and their TCAIM mRNA expression was analyzed. As shown in Fig. 1A, LPS stimulation resulted in a transient significant reduction of Tcaim mRNA expression, reaching its maximum at 3 h after stimulation.

FIGURE 1.

DCs downregulate Tcaim expression following in vitro LPS stimulation or in vivo allogeneic skin transplant challenge. (A) Tcaim mRNA expression after LPS stimulation. BMDCs were stimulated with 1 μg/ml LPS for the indicated time points (0, 1, 3, 6, 12, and 24 h). Shown are the Tcaim mRNA expression results of n = 4–5 independent experiments, as mean ± SEM. (B) Changes in DC subset frequencies in spleen and lymph nodes following allogeneic skin transplantation (skin Tx). Indicated DC subsets were sorted from spleen and lymph nodes of naive C57BL/6 mice or mice, which received an allogeneic BALB/c skin transplant 2 or 7 d prior to sample collection. (C) Tcaim mRNA expression of sorted DC subsets. Shown are the frequencies and Tcaim mRNA expression results of n = 2–3 independent experiments, as mean ± SEM. Cells of three mice were pooled per experiment. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

FIGURE 1.

DCs downregulate Tcaim expression following in vitro LPS stimulation or in vivo allogeneic skin transplant challenge. (A) Tcaim mRNA expression after LPS stimulation. BMDCs were stimulated with 1 μg/ml LPS for the indicated time points (0, 1, 3, 6, 12, and 24 h). Shown are the Tcaim mRNA expression results of n = 4–5 independent experiments, as mean ± SEM. (B) Changes in DC subset frequencies in spleen and lymph nodes following allogeneic skin transplantation (skin Tx). Indicated DC subsets were sorted from spleen and lymph nodes of naive C57BL/6 mice or mice, which received an allogeneic BALB/c skin transplant 2 or 7 d prior to sample collection. (C) Tcaim mRNA expression of sorted DC subsets. Shown are the frequencies and Tcaim mRNA expression results of n = 2–3 independent experiments, as mean ± SEM. Cells of three mice were pooled per experiment. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

Close modal

Next, we investigated whether this also happens in vivo. To this end, we sorted different DC populations (CD4+, CD8+, CD4CD8, CD103+, monocyte-derived DCs [moDCs], and plasmacytoid DCs; gating strategy shown in Supplemental Fig. 1) from spleens and lymph nodes of naive C57BL/6 mice and mice 2 and 7 d after receiving a BALB/c skin transplant. CD64+ moDCs were included because they are supposed to most closely resemble BMDCs. As can be seen in Fig. 1B, allogeneic skin transplantation resulted in an early but transient increase in frequencies of plasmacytoid DCs, whereas we detected a continuous increase in CD4+, CD103+, and moDCs. Especially those three DC subsets showed also a downregulation in TCAIM transcription following transplantation (Fig. 1C). For CD103+ and moDCs this effect was transient, whereas in CD4+ DCs this effect was even more pronounced 7 d posttransplant.

To investigate whether the mitochondrial protein TCAIM affects BMDC function, we overexpressed Tcaim using adenoviral gene delivery, reaching a significant increase in TCAIM expression especially upon LPS stimulation (Supplemental Fig. 2).

Enhanced expression of Tcaim in BMDCs resulted in increased baseline proliferation of cocultured CD8+ T cells (Fig. 2). LPS and poly-IC stimulation of Control-BMDCs resulted in an increase of proliferating cocultured allogeneic CD4+ and especially CD8+ T cells (Fig. 2). TCAIM overexpression led to a reduced priming capacity of LPS-stimulated BMDCs (Fig. 2). We observed a substantially diminished proliferation and CD44 expression of cocultured CD4+ and CD8+ T cells (Fig. 2). Interestingly, this reduced priming capacity of TCAIM-BMDCs was specific for LPS-stimulation and not observed upon poly-IC stimulation.

FIGURE 2.

TCAIM-BMDCs fail to prime alloreactive T cells. (A) Representative dot plots showing CD44 expression and proliferation of CD4+ (left panel) and CD8+ T cells (right panel) cocultured for 5 d with LPS-or poly-IC-matured or immature TCAIM-BMDCs or Control-BMDCs. (B) The left panel summarizes the proliferation results of n = 4–8 independent experiments. Depicted are the mean ± SD of the proliferated cells (CPD negative/low) pregated on living CD3+CD4+ T cells. The right graph summarizes the proliferation of living CD3+CD8+ T cells. The mean ± SEM calculated from n = 3–6 independent experiments are shown. *p < 0.05, **p < 0.01, ****p < 0.0001.

FIGURE 2.

TCAIM-BMDCs fail to prime alloreactive T cells. (A) Representative dot plots showing CD44 expression and proliferation of CD4+ (left panel) and CD8+ T cells (right panel) cocultured for 5 d with LPS-or poly-IC-matured or immature TCAIM-BMDCs or Control-BMDCs. (B) The left panel summarizes the proliferation results of n = 4–8 independent experiments. Depicted are the mean ± SD of the proliferated cells (CPD negative/low) pregated on living CD3+CD4+ T cells. The right graph summarizes the proliferation of living CD3+CD8+ T cells. The mean ± SEM calculated from n = 3–6 independent experiments are shown. *p < 0.05, **p < 0.01, ****p < 0.0001.

Close modal

In addition, we analyzed cocultured T cells for their cytokine production after restimulation with PMA–ionomycin. Again LPS and to a reduced extent also poly-IC stimulation of Control-BMDCs increased the frequencies of IFN-γ expressing cocultured allogeneic CD4+ and, even more pronounced, CD8+ T cells (Fig. 3A). We observed reduced IFN-γ production of cocultured CD4+ and CD8+ T cells with LPS-stimulated TCAIM-BMDCs compared with Control-BMDCs. This reached significance when applying the Wilcoxon test (p < 0.05), but did not reach significance when correcting for multiple comparisons because of low numbers (n = 4–5). In contrast, poly-IC stimulation of TCAIM-BMDCs induced a similar increase in IFN-γ expressing cocultured allogeneic CD8+ T cells. CD4+ and CD8+ T cells cocultured with LPS- or poly-IC–stimulated TCAIM- and Control-BMDCs showed no altered IL-17 and TNF-α production compared with cocultures with unstimulated BMDCs (Fig. 3B, 3C).

FIGURE 3.

TCAIM-BMDCs induce fewer cytokine producing alloreactive T cells. (A) The left panel depicts the results of n = 3–5 independent experiments for CD4+ T cells and n = 3–4 independent tests for CD8+ T cells cocultured with allogeneic BMDCs. Depicted is the percentage of IFN-γ–producing living CD3+CD4+ or CD8+ T cells. The results are shown as mean ± SEM; p < 0.05 (Wilcoxon-test) for CD4+ and CD8+ cocultured with LPS-stimulated TCAIM-BMDCs versus LPS-stimulated Control-BMDCs. In addition, the percentages of (B) IL-17– and (C) TNF-α–producing living CD3+CD4+ (left panels) or CD8+ (right panels) T cells are shown. The mean ± SEM calculated from n = 3–5 independent experiments is shown.

FIGURE 3.

TCAIM-BMDCs induce fewer cytokine producing alloreactive T cells. (A) The left panel depicts the results of n = 3–5 independent experiments for CD4+ T cells and n = 3–4 independent tests for CD8+ T cells cocultured with allogeneic BMDCs. Depicted is the percentage of IFN-γ–producing living CD3+CD4+ or CD8+ T cells. The results are shown as mean ± SEM; p < 0.05 (Wilcoxon-test) for CD4+ and CD8+ cocultured with LPS-stimulated TCAIM-BMDCs versus LPS-stimulated Control-BMDCs. In addition, the percentages of (B) IL-17– and (C) TNF-α–producing living CD3+CD4+ (left panels) or CD8+ (right panels) T cells are shown. The mean ± SEM calculated from n = 3–5 independent experiments is shown.

Close modal

Next, we tested whether an altered surface expression and cytokine secretion of TCAIM-BMDCs explains the diminished priming capacity. No differences in upregulation of MHC-II or CD86 upon LPS and poly-IC stimulation between TCAIM-BMDCs and Control-BMDCs were detectable. In contrast, baseline CD86 expression was even slightly increased in unstimulated TCAIM-BMDCs (Fig. 4A, 4B). In addition, LPS-induced IL-6 secretion was not affected by TCAIM overexpression (Fig. 4C). TCAIM-BMDCs did also release TNF-α upon LPS stimulation; however, we observed reduced TNF-α secretion by 42% in comparison with Control-BMDCs. Poly-IC–induced TNF-α and IL-6 production was only slightly lower in TCAIM-BMDC cultures as compared with Control-BMDCs (Fig. 4C).

FIGURE 4.

Tcaim overexpression does not affect LPS-mediated MHC-II and CD86 upregulation. (A) Representative dot plots showing the expression of MHC-II and CD86 of CD11c+ immature and 24 h LPS- or poly-IC–matured TCAIM-BMDCs or Control-BMDCs analyzed by flow cytometry. (B) MHC-II (left panel) and CD86 (right panel) expression in the percentage of CD11c+ cells stimulated with LPS (1 μg/ml) or poly-IC (50 μg/ml) for 24 h or left unstimulated TCAIM-BMDCs and Control-BMDCs of n = 5–6 independent experiments are shown, depicted as mean ± SD. *p < 0.05, **p < 0.01, ***p < 0.001. (C) Effect on cytokine release upon Tcaim overexpression. The graphs show the cytokine production of LPS-stimulated (1 μg/ml) or poly-IC-stimulated (50 μg/ml) stimulated BMDCs analyzed using a cytometric bead array. Supernatants were taken 0, 6, 12, and 24 h after stimulation. The left figure displays the TNF-α secretion of n = 2–7 independent experiments, whereas the right graph shows the IL-6 release of n = 1–9 independent experiments. Results are depicted as mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

FIGURE 4.

Tcaim overexpression does not affect LPS-mediated MHC-II and CD86 upregulation. (A) Representative dot plots showing the expression of MHC-II and CD86 of CD11c+ immature and 24 h LPS- or poly-IC–matured TCAIM-BMDCs or Control-BMDCs analyzed by flow cytometry. (B) MHC-II (left panel) and CD86 (right panel) expression in the percentage of CD11c+ cells stimulated with LPS (1 μg/ml) or poly-IC (50 μg/ml) for 24 h or left unstimulated TCAIM-BMDCs and Control-BMDCs of n = 5–6 independent experiments are shown, depicted as mean ± SD. *p < 0.05, **p < 0.01, ***p < 0.001. (C) Effect on cytokine release upon Tcaim overexpression. The graphs show the cytokine production of LPS-stimulated (1 μg/ml) or poly-IC-stimulated (50 μg/ml) stimulated BMDCs analyzed using a cytometric bead array. Supernatants were taken 0, 6, 12, and 24 h after stimulation. The left figure displays the TNF-α secretion of n = 2–7 independent experiments, whereas the right graph shows the IL-6 release of n = 1–9 independent experiments. Results are depicted as mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

Close modal

As a mitochondrial protein TCAIM might also interfere with cell survival and thereby affect the T cell priming capacity of DCs. Therefore, we analyzed the percentage of apoptotic or necrotic cells in unstimulated and LPS-stimulated TCAIM- and Control-BMDCs. As shown in Fig. 5A, TCAIM overexpression did not affect cell viability of BMDCs. In addition, recovery of BMDCs in absolute cell numbers was not different between TCAIM-BMDCs and Control-BMDCs (TCAIM-BMDCs: 55% ± 5.7; Control-BMDCs: 55.5% ± 3.4).

FIGURE 5.

Tcaim overexpression abolishes release of mROS, which regulate T cell priming capacity of BMDCs. (A) Effect of Tcaim overexpression on apoptosis induction. Dot plots of unstimulated or 24 h LPS-stimulated (1 μg/ml) BMDCs stained for Annexin-V and 7-actinomycin D are depicted. Results of seven independent experiments are shown as mean ± SEM. The cells were pregated as CD11c+. (B) Effect on ΔΨ upon Tcaim overexpression. ΔΨ was measured 24 h after LPS-stimulation in CD11c+ TCAIM- and Control-BMDCs using TMRM. Results are depicted from five independent experiments as means ± SEM. (C) MROS produced by unstimulated or LPS (1 μg/ml) stimulated TCAIM- and Control-BMDCs after indicated time points (0, 1, and 3 h) are depicted as means ± SEM. Results of 10 independent experiments are shown. *p < 0.05, **p < 0.01.

FIGURE 5.

Tcaim overexpression abolishes release of mROS, which regulate T cell priming capacity of BMDCs. (A) Effect of Tcaim overexpression on apoptosis induction. Dot plots of unstimulated or 24 h LPS-stimulated (1 μg/ml) BMDCs stained for Annexin-V and 7-actinomycin D are depicted. Results of seven independent experiments are shown as mean ± SEM. The cells were pregated as CD11c+. (B) Effect on ΔΨ upon Tcaim overexpression. ΔΨ was measured 24 h after LPS-stimulation in CD11c+ TCAIM- and Control-BMDCs using TMRM. Results are depicted from five independent experiments as means ± SEM. (C) MROS produced by unstimulated or LPS (1 μg/ml) stimulated TCAIM- and Control-BMDCs after indicated time points (0, 1, and 3 h) are depicted as means ± SEM. Results of 10 independent experiments are shown. *p < 0.05, **p < 0.01.

Close modal

ATP and mROS production are dependent on mitochondrial membrane potential ΔΨ. To determine whether LPS-stimulated TCAIM-BMDCs show an altered ΔΨ, we used TMRM. TMRM is a lipophilic cation that accumulates ΔΨ-dependent in negatively charged mitochondria (20, 21). Fig. 5B shows the percentages of TMRM accumulation in unstimulated and LPS-stimulated TCAIM- and Control-BMDCs. Tcaim overexpression showed no effect on the mitochondrial membrane potential.

In addition, we investigated whether Tcaim overexpression alters mROS release (Fig. 5C). We measured mROS in TCAIM-BMDCs and Control-BMDCs, which were left unstimulated or stimulated with LPS for 1 h and 3 h. TCAIM-BMDCs showed a significantly diminished mROS production compared with Control-BMDCs. As described by West et al. (4, 22), we could not detect mROS induction upon poly-IC stimulation (data not shown).

To reveal the underlying mechanism for the diminished T cell priming capacity of TCAIM-BMDCs, we studied their LPS-induced transcription using a TLR signaling pathway PCR Array. TCAIM-BMDCs and Control-BMDCs were stimulated for 1 or 12 h with LPS or were left unstimulated. We visualized the results of three independent experiments using an unsupervised heat map analysis, meaning that the sample arrangement occurred respectively to expression similarities and not to groups. A clustering of all unstimulated, 1-h LPS-stimulated, or 12-h LPS-stimulated samples implied that the stimulatory effect of LPS exceeds generally the effect of Tcaim overexpression (Fig. 6A). A principal component analysis, in which triplicates of TCAIM- and Control-BMDCs of three stimulation time points are depicted, also showed a uniform distribution of TCAIM- and Control-BMDCs according to their stimulation (Fig. 6B). Surprisingly, of the 84 genes analyzed, only expression of Il2 was significantly downregulated in TCAIM-BMDCs compared with Control-BMDCs. To verify the array results, we performed QRT-PCR on additional samples. LPS stimulation of Control-BMDCs resulted in an up to 20-fold upregulation of Il2 mRNA expression (Fig. 6C). Already in unstimulated BMDCs, TCAIM overexpression reduced the basal Il2 transcription. LPS-induced Il2 mRNA expression was significantly decreased in TCAIM-BMDCs compared with Control-BMDCs. Il2 mRNA expression was not increased upon poly-IC stimulation in TCAIM- and Control-BMDCs compared with unstimulated cells. Furthermore, we detected a time-dependent increase in IL-2 protein secretion in supernatants of LPS-stimulated Control-BMDCs, which we did not observe in supernatants of LPS-stimulated TCAIM-BMDCs (Fig. 6D).

Il2 transcription in T cells and in DCs is regulated by intracellular Ca2+ increase with subsequent NFAT/NF-κB/AP1 activation; therefore, we analyzed whether TCAIM overexpression affects LPS-induced Ca2+ signaling. Fig. 6E shows the intracellular Ca2+ influx after LPS and ionomycin stimulation of TCAIM-BMDCs and Control-BMDCs. In Control-BMDCs and TCAIM-BMDCs, we observed a rapid increase of intracellular Ca2+ after ionomycin stimulation, which was lower in TCAIM-BMDCs. TCAIM-BMDCs were completely unable to increase free cytosolic Ca2+ upon LPS stimulation. This effect was highly reproducible (Fig. 6F).

Next we questioned whether the addition of recombinant mouse IL-2 (rmIL-2) would restore the diminished T cell priming capacity of TCAIM-BMDCs. We added 20 or 200 ng/ml rmIL-2 to the coculture of LPS-stimulated transduced BMDCs and allogeneic lymph node cells. We observed a dose-dependent induction of CD8+ T cell proliferation in cocultures with LPS-matured TCAIM-BMDCs (Fig. 7A), whereas the addition of rmIL-2 increased but did not normalize proliferation of cocultured CD4+ T cells. The proliferation in cocultures with LPS-stimulated Control-BMDCs was not affected by the addition of rmIL-2.

FIGURE 7.

The addition of IL-2 can restore T cell priming capacity of TCAIM-BMDCs. (A) Representative dot plots showing CD44 expression and proliferation (CPD eFluor450), 5 d after cocultures of 5 × 105 BALB/c lymph node cells and 4 × 104 LPS-matured C57BL/6 TCAIM-BMDCs and Control-BMDCs upon the addition of 20 or 200 ng/ml recombinant mouse IL-2, are depicted. (B) Representative dot plots showing CD44 expression and IFN-γ production of living CD3+CD4+ or CD8+ T cells after PMA/ionomycin restimulation; 5 × 105 BALB/c lymph node cells were cocultured for 5 d with 4 × 104 unstimulated or LPS-matured IL-2+/− heterozygous or IL-2–deficient Control-BMDCs and TCAIM-BMDCs.

FIGURE 7.

The addition of IL-2 can restore T cell priming capacity of TCAIM-BMDCs. (A) Representative dot plots showing CD44 expression and proliferation (CPD eFluor450), 5 d after cocultures of 5 × 105 BALB/c lymph node cells and 4 × 104 LPS-matured C57BL/6 TCAIM-BMDCs and Control-BMDCs upon the addition of 20 or 200 ng/ml recombinant mouse IL-2, are depicted. (B) Representative dot plots showing CD44 expression and IFN-γ production of living CD3+CD4+ or CD8+ T cells after PMA/ionomycin restimulation; 5 × 105 BALB/c lymph node cells were cocultured for 5 d with 4 × 104 unstimulated or LPS-matured IL-2+/− heterozygous or IL-2–deficient Control-BMDCs and TCAIM-BMDCs.

Close modal

Last, we tested whether IL-2 production by BMDCs determines their T cell priming capacity. Therefore, BMDCs generated from bone marrow of IL-2–deficient and IL-2+/− heterozygous control mice were cocultured with allogeneic lymph node cells. Indeed, IL-2–deficient BMDCs failed to increase their T cell priming capacity upon LPS stimulation (Fig. 7B). This failure was more pronounced for priming of CD8+ T cells as compared with CD4+ T cells.

Rotenone, a systemic complex I inhibitor, was used to inhibit the mROS production in BMDCs (23), as complex I is one important source of mROS (6). To evaluate the effect of Rotenone on Il2 mRNA expression, we treated the cells overnight with 1 μM Rotenone or control medium and then stimulated them for 1 h with LPS or left them unstimulated. Overnight incubation with Rotenone had no effect on Il2 mRNA expression of LPS-stimulated TCAIM-BMDCs (Fig. 8A). In contrast, Il2 mRNA expression is considerably attenuated in LPS-stimulated Rotenone-treated Control-BMDCS. Furthermore, Rotenone-treated LPS-matured Control-BMDCs had a diminished T cell priming capacity in comparison with untreated LPS-matured Control-BMDCs (Fig. 8B). This effect was more prominent for cocultured CD8+ T cells than for CD4+ T cells. In contrast, Rotenone-treatment of LPS-stimulated TCAIM-BMDCs did not reduce their residual low T cell priming capacity.

FIGURE 8.

Inhibition of mROS generating complex I reduces Il2 expression and T cell priming capacity of LPS-stimulated BMDCs. (A) Expression of Il2 mRNA after overnight Rotenone (1 μM) treatment in unstimulated BMDCs and following 1 h LPS (1 μg/ml) stimulation. Mean ± SD from five independent experiments are shown. (B) Dot plots of proliferation and CD44 expression of alloreactive CD4+ and CD8+ lymph node cells cocultured for 5 d with overnight Rotenone-treated (1 μM) and 24 h LPS-matured (1 μg/ml) TCAIM-BMDCs and Control-BMDCs. The cells are pregated for living CD3+CD4+ or CD3+CD8+ T cells. (C) TCAIM alters transcription factor binding activity of AP1, NF-κB, and C/EBPβ. TCAIM- and Control-BMDCs were incubated with baculoviruses encoding a construct of a consensus sequence for one of the following transcription factors: NF-κB, NFAT, AP1, or C/BEP followed by a luciferase reporter gene. Twenty-four hours later, cells were left untreated or were stimulated for 30 min and 3 h with 1 μg/ml LPS and luciferase activity was measured. Shown are the means of two independent experiments.

FIGURE 8.

Inhibition of mROS generating complex I reduces Il2 expression and T cell priming capacity of LPS-stimulated BMDCs. (A) Expression of Il2 mRNA after overnight Rotenone (1 μM) treatment in unstimulated BMDCs and following 1 h LPS (1 μg/ml) stimulation. Mean ± SD from five independent experiments are shown. (B) Dot plots of proliferation and CD44 expression of alloreactive CD4+ and CD8+ lymph node cells cocultured for 5 d with overnight Rotenone-treated (1 μM) and 24 h LPS-matured (1 μg/ml) TCAIM-BMDCs and Control-BMDCs. The cells are pregated for living CD3+CD4+ or CD3+CD8+ T cells. (C) TCAIM alters transcription factor binding activity of AP1, NF-κB, and C/EBPβ. TCAIM- and Control-BMDCs were incubated with baculoviruses encoding a construct of a consensus sequence for one of the following transcription factors: NF-κB, NFAT, AP1, or C/BEP followed by a luciferase reporter gene. Twenty-four hours later, cells were left untreated or were stimulated for 30 min and 3 h with 1 μg/ml LPS and luciferase activity was measured. Shown are the means of two independent experiments.

Close modal

To obtain insight into the molecular mechanisms of TCAIM-mediated reduction in IL-2 production and T cell priming capacity of BMDCs, we have performed preliminary experiments studying LPS-induced transcription factors activation. TCAIM- and Control-BMDCs were incubated with baculoviruses encoding luciferase constructs with transcription factor binding sites for NFAT, NF-κB, AP1, and C/EBPβ. The following day, BMDCs were stimulated with LPS for 30 min and 3 h, and transcription factor binding activity was determined with luciferase assay (Fig. 8C). We detected a low NFAT binding activity in BMDCs, but we did not observe a difference between TCAIM-BMDCs and Control-BMDCs. In contrast, we detected a LPS-induced increase in NF-κB and AP1 binding activity, which was especially lower for NF-κB in TCAIM-BMDCs. Strikingly, TCAIM-BMDCs displayed a transient increase in C/EBPβ binding activity, which we did not observe for Control-BMDCs.

In this study, we show that increased expression of TCAIM in murine BMDCs leads to an impaired LPS-induced priming capacity for allogeneic CD4+, but even more CD8+ T cells. This was not due to abolished LPS-mediated upregulation of MHC-II or CD86 expression. Release of inflammatory cytokines and mediators such as IL-6, IL-12, or IL-1 was not affected. However, LPS-stimulated TCAIM-BMDCs were characterized by a reduced Ca2+ influx, mROS formation, and Il2 expression. Adding IL-2 normalized proliferation of cocultured CD8+ T cells. Furthermore, IL-2 deficient BMDCs failed to increase priming of cocultured CD8+ T cells upon LPS stimulation. Interestingly, inhibiting mROS producing mitochondrial complex I did mimic Tcaim overexpression as it resulted in reduced LPS-mediated Il2 expression and T cell priming capacity.

Mitochondria play an important and decisive role in cell signaling. Especially their role in immune cell signaling has gained much attention in recent years (24, 25). Apart from regulating apoptosis, mitochondrial release of ROS is instrumental for redox-sensitive signaling pathways such as activation of MAP kinases, NF-κB, and AP1 as well as mitochondria are indispensable for Ca2+ signaling (22, 2629).

Mitochondria are efficient Ca2+ buffers (30). Because of close proximity to the plasma membrane, mitochondria can take up large amounts of inflowing Ca2+, which prevents the inactivation of calcium release activated Ca2+ (CRAC) channels (31).

Ca2+ is an important second messenger in DCs and its influx occurs upon PLC-γ activation transiently by depleting intracellular stores, such as the endoplasmic reticulum (ER) or through opening of Ca2+ channels in the plasma membrane. Ca2+ influx is induced by certain but not all microbial products, such as LPS, CpG, or peptidoglycan, resulting in a calcineurin-dependent NFAT activation, subsequent nuclear translocation, and NF-κB activation by degradation of the NF-κB inhibitor IκB (31). Applying CRAC channel inhibitors results in a less mature phenotype (32) and a reduced IL-2 secretion (33).

TCAIM as a mitochondrial protein containing a J-domain could interfere with mitochondrial Ca2+ uptake, as J-domain proteins are described to be involved in mitochondrial translocases (34, 35). Mitochondrial calcium uptake can be mediated by the mitochondrial calcium uniporter. This transport is supported by the mitochondrial membrane potential ΔΨ and leads to the production of mROS (30). Inhibition of the prolonged Ca2+ uptake after LPS-stimulation in TCAIM-BMDCs by disturbing the Ca2+ transport into mitochondria could explain the reduced mROS production. Furthermore, mitochondria play an active part in regulating inositol triphosphate–mediated Ca2+ release from the ER (36, 37). Mitochondrial Ca2+ uptake exerts positive feedback effects on the inositol triphosphate receptor-mediated ER Ca2+ mobilization (38). Thus, mitochondria can affect both Ca2+ release from the ER and Ca2+ entry across the plasma membrane, thereby shaping the size and duration of the intracellular Ca2+ signal (39). This could explain that TCAIM-BMDCs showed abolished LPS-mediated and reduced ionomycin-mediated increase in Ca2+.

Interestingly, we could observe a stronger effect of TCAIM overexpression on the induction of proliferation and cytokine production by cocultured CD8+ T cells than by CD4+ T cells.

Shumilina et al. (31) described that the calcium influx at the immunologic synapse is dependent on the type of T cell contacting the DC. When the DC interacted with alloreactive CD8+ T cells, the Ca2+ concentration rose within DCs. In contrast, when the DC formed a synapse with alloreactive CD4+ T cells, only an increase of Ca2+ in the T cells was detectable (31, 40, 41). Thus, the more profound defect in proliferation and IFN-γ by cocultured CD8+ T cells is perhaps due to the completely abolished calcium influx in TCAIM-BMDCs.

It is well known that during priming of naive CD8+ T cells presence of CD4+ T cells is critical (42, 43). Wolkers et al. showed that in the absence of CD4+ T cells, the addition of exogenous IL-2 markedly increased the secondary expansion of and IFN-γ production by CD8+ T cells (44). The maximal expansion was achieved when using IL-2 concentrations between 10 and 100 ng/ml (44). This finding is in accordance with our data, because in our cocultures with LPS-stimulated TCAIM-BMDCs CD4+, T cells were not activated compared with cocultures with LPS-stimulated Control-BMDCs, but the addition of 20 to 200 ng/ml IL-2 could normalize proliferation of CD8+ T cells.

Considering that TCAIM-BMDCs showed a reduced Ca2+ influx and thereby a nearly abolished IL-2 secretion after LPS-stimulation, it is not surprising that it affected especially the priming of CD8+ T cells and to a lower extent of CD4+ T cells (45). Indeed, the addition of IL-2 to cocultures of TCAIM-BMDCs with allogeneic T cells is able to restore proliferation of CD8+ T cells to nearly normal levels, whereas the effect was not as evident for CD4+ T cells.

As pointed out earlier, Ca2+ influx accelerates the generation of mROS (30). Furthermore, triggering of certain TLRs in immune cells results in formation of mROS important for bactericidal activity and for regulation of redox-sensitive signaling pathways regulating cytokine release and Ag-presenting capacity (22, 26). It has been shown that ROS production in DCs upon communication with cocultured T cells determines their Ag-presentation capacity (46, 47). In those studies, scavenging of ROS by antioxidants, resulted in reduced release of inflammatory cytokines such as IL-6, but also abrogated induction of MHC and costimulatory molecules. We showed that the Tcaim overexpression in BMDCs, which completely abolished LPS-induced Ca2+ influx and impaired mROS production, was associated with diminished T cell priming capacity. In contrast to the findings above, this finding was not associated with reduced surface expression of MHC and costimulatory molecules or production of IL-6. This may be due to a more specific action of TCAIM on mROS, whereas scavenging antioxidants can also act on other enzymes generating ROS.

The effects of Tcaim overexpression on the T cell priming capacity were limited to LPS stimulation and were not observed upon poly-IC stimulation. This result further supports our hypothesis that TCAIM effects are mediated by inhibiting Ca2+ influx and mROS production, as we and others have observed that stimulation of BMDCs with TLR3 ligands such as poly-IC do not elicit either Ca2+ influx or mROS production (4, 15, 48, 49).

In conjunction with the diminished LPS-mediated Ca2+ influx and mROS production, we observed a reduced NF-κB but increased C/EBPβ binding activity. This observation might explain, on the molecular level, why TCAIM-BMDCs show reduced LPS-induced IL-2 expression, because NF-κB is well known to control Il2 transcription positively (50, 51). In contrast, C/EBPβ was shown to influence Il2 transcription negatively, at least in T cells (52).

Our investigations did not completely explain how increased Tcaim expression abolishes the priming capacity of BMDCs for cocultured allogeneic CD4+ T cells, as addition of IL-2 did enhance but not normalize T cell proliferation. In addition to testing IL-2, we tested other γ-chain signaling cytokines, such as IL-15. We could neither detect an LPS-induced increase of IL-15 expression nor did addition of IL-15 restore T cell proliferation (data not shown).

Thus, the mitochondrial protein TCAIM diminishes the T cell priming capacity of BMDCs, in particular for CD8+ T cells, by inhibiting LPS-mediated Ca2+ influx, mROS formation, NF-κB, and especially Il2 expression.

We thank Amiya Patra (Institute of Pathology, Department of Molecular Pathology, Julius Maximilians-University of Würzburg) for providing the IL-2+/− heterozygous and IL-2 deficient mice and Desiree Kunkel (Berlin Brandenburg Center for Regenerative Therapies) for support with flow-sorting DC subsets.

This work was supported by the Deutsche Forschungsgemeinschaft (Transregio TR52).

The online version of this article contains supplemental material.

Abbreviations used in this article:

AP1

activating protein-1

BMDC

bone marrow–derived DC

CPD

cell proliferation dye

CRAC

calcium release activated channel

DC

dendritic cell

ER

endoplasmic reticulum

ESCIT

evolutionarily conserved signaling intermediate in Toll pathway

MHC-II

MHC class II

mHprt

mouse hypoxanthine phosphoribosyltransferase

moDC

monocyte-derived DC

mROS

mitochondrial reactive oxygen species

QRT-PCR

quantitative real-time PCR

rmIL-2

recombinant mouse IL-2

TCAIM

T cell activation inhibitor, mitochondrial

TMRM

tetramethylrhodamine-methyl ester.

1
Sawitzki
B.
,
Bushell
A.
,
Steger
U.
,
Jones
N.
,
Risch
K.
,
Siepert
A.
,
Lehmann
M.
,
Schmitt-Knosalla
I.
,
Vogt
K.
,
Gebuhr
I.
, et al
.
2007
.
Identification of gene markers for the prediction of allograft rejection or permanent acceptance.
Am. J. Transplant.
7
:
1091
1102
.
2
Gajanayake
T.
,
Sawitzki
B.
,
Matozan
K.
,
Korchagina
E. Y.
,
Lehmann
M.
,
Volk
H. D.
,
Rieben
R.
.
2008
.
Dextran sulfate facilitates anti-CD4 mAb-induced long-term rat cardiac allograft survival after prolonged cold ischemia.
Am. J. Transplant.
8
:
1151
1162
.
3
Keeren
K.
,
Friedrich
M.
,
Gebuhr
I.
,
Philipp
S.
,
Sabat
R.
,
Sterry
W.
,
Brandt
C.
,
Meisel
C.
,
Grütz
G.
,
Volk
H. D.
,
Sawitzki
B.
.
2009
.
Expression of tolerance associated gene-1, a mitochondrial protein inhibiting T cell activation, can be used to predict response to immune modulating therapies.
J. Immunol.
183
:
4077
4087
.
4
West
A. P.
,
Brodsky
I. E.
,
Rahner
C.
,
Woo
D. K.
,
Erdjument-Bromage
H.
,
Tempst
P.
,
Walsh
M. C.
,
Choi
Y.
,
Shadel
G. S.
,
Ghosh
S.
.
2011
.
TLR signalling augments macrophage bactericidal activity through mitochondrial ROS.
Nature
472
:
476
480
.
5
Vogel
R. O.
,
Janssen
R. J.
,
van den Brand
M. A.
,
Dieteren
C. E.
,
Verkaart
S.
,
Koopman
W. J.
,
Willems
P. H.
,
Pluk
W.
,
van den Heuvel
L. P.
,
Smeitink
J. A.
,
Nijtmans
L. G.
.
2007
.
Cytosolic signaling protein Ecsit also localizes to mitochondria where it interacts with chaperone NDUFAF1 and functions in complex I assembly.
Genes Dev.
21
:
615
624
.
6
Li
R.
,
Jen
N.
,
Yu
F.
,
Hsiai
T. K.
.
2011
.
Assessing mitochondrial redox status by flow cytometric methods: vascular response to fluid shear stress.
Curr. Protoc. Cytom.
Chapter 9
:
37
.
7
Bulua
A. C.
,
Simon
A.
,
Maddipati
R.
,
Pelletier
M.
,
Park
H.
,
Kim
K. Y.
,
Sack
M. N.
,
Kastner
D. L.
,
Siegel
R. M.
.
2011
.
Mitochondrial reactive oxygen species promote production of proinflammatory cytokines and are elevated in TNFR1-associated periodic syndrome (TRAPS).
J. Exp. Med.
208
:
519
533
.
8
Tal
M. C.
,
Iwasaki
A.
.
2011
.
Mitoxosome: a mitochondrial platform for cross-talk between cellular stress and antiviral signaling.
Immunol. Rev.
243
:
215
234
.
9
Banchereau
J.
,
Steinman
R. M.
.
1998
.
Dendritic cells and the control of immunity.
Nature
392
:
245
252
.
10
Granucci
F.
,
Andrews
D. M.
,
Degli-Esposti
M. A.
,
Ricciardi-Castagnoli
P.
.
2002
.
IL-2 mediates adjuvant effect of dendritic cells.
Trends Immunol.
23
:
169
171
.
11
Sauma
D.
,
Michea
P.
,
Lennon-Duménil
A. M.
,
Fierro
A.
,
Morales
J.
,
Rosemblatt
M.
,
Bono
M. R.
.
2004
.
Interleukin-4 selectively inhibits interleukin-2 secretion by lipopolysaccharide-activated dendritic cells.
Scand. J. Immunol.
59
:
183
189
.
12
Granucci
F.
,
Vizzardelli
C.
,
Pavelka
N.
,
Feau
S.
,
Persico
M.
,
Virzi
E.
,
Rescigno
M.
,
Moro
G.
,
Ricciardi-Castagnoli
P.
.
2001
.
Inducible IL-2 production by dendritic cells revealed by global gene expression analysis.
Nat. Immunol.
2
:
882
888
.
13
Zelante
T.
,
Fric
J.
,
Wong
A. Y.
,
Ricciardi-Castagnoli
P.
.
2012
.
Interleukin-2 production by dendritic cells and its immuno-regulatory functions.
Front. Immunol.
3
:
161
.
14
Zanoni
I.
,
Granucci
F.
.
2012
.
Regulation and dysregulation of innate immunity by NFAT signaling downstream of pattern recognition receptors (PRRs).
Eur. J. Immunol.
42
:
1924
1931
.
15
Zanoni
I.
,
Ostuni
R.
,
Capuano
G.
,
Collini
M.
,
Caccia
M.
,
Ronchi
A. E.
,
Rocchetti
M.
,
Mingozzi
F.
,
Foti
M.
,
Chirico
G.
, et al
.
2009
.
CD14 regulates the dendritic cell life cycle after LPS exposure through NFAT activation.
Nature
460
:
264
268
.
16
Rogers
N. C.
,
Slack
E. C.
,
Edwards
A. D.
,
Nolte
M. A.
,
Schulz
O.
,
Schweighoffer
E.
,
Williams
D. L.
,
Gordon
S.
,
Tybulewicz
V. L.
,
Brown
G. D.
,
Reis e Sousa
C.
.
2005
.
Syk-dependent cytokine induction by Dectin-1 reveals a novel pattern recognition pathway for C type lectins.
Immunity
22
:
507
517
.
17
Schliesser
U.
,
Chopra
M.
,
Beilhack
A.
,
Appelt
C.
,
Vogel
S.
,
Schumann
J.
,
Panov
I.
,
Vogt
K.
,
Schlickeiser
S.
,
Olek
S.
, et al
.
2013
.
Generation of highly effective and stable murine alloreactive Treg cells by combined anti-CD4 mAb, TGF-β, and RA treatment.
Eur. J. Immunol.
43
:
3291
3305
.
18
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
.
19
Schumann
J.
,
Stanko
K.
,
Woertge
S.
,
Appelt
C.
,
Schumann
M.
,
Kühl
A. A.
,
Panov
I.
,
Schliesser
U.
,
Vogel
S.
,
Ahrlich
S.
, et al
.
2014
.
The mitochondrial protein TCAIM regulates activation of T cells and thereby promotes tolerance induction of allogeneic transplants.
Am. J. Transplant.
14
:
2723
2735
.
20
Perry
S. W.
,
Norman
J. P.
,
Barbieri
J.
,
Brown
E. B.
,
Gelbard
H. A.
.
2011
.
Mitochondrial membrane potential probes and the proton gradient: a practical usage guide.
Biotechniques
50
:
98
115
.
21
Scaduto
R. C.
 Jr.
,
Grotyohann
L. W.
.
1999
.
Measurement of mitochondrial membrane potential using fluorescent rhodamine derivatives.
Biophys. J.
76
:
469
477
.
22
West
A. P.
,
Shadel
G. S.
,
Ghosh
S.
.
2011
.
Mitochondria in innate immune responses.
Nat. Rev. Immunol.
11
:
389
402
.
23
Wallace
K. B.
2008
.
Mitochondrial off targets of drug therapy.
Trends Pharmacol. Sci.
29
:
361
366
.
24
Cloonan
S. M.
,
Choi
A. M.
.
2013
.
Mitochondria: sensors and mediators of innate immune receptor signaling.
Curr. Opin. Microbiol.
16
:
327
338
.
25
Galluzzi
L.
,
Kepp
O.
,
Kroemer
G.
.
2012
.
Mitochondria: master regulators of danger signalling.
Nat. Rev. Mol. Cell Biol.
13
:
780
788
.
26
Collins
Y.
,
Chouchani
E. T.
,
James
A. M.
,
Menger
K. E.
,
Cochemé
H. M.
,
Murphy
M. P.
.
2012
.
Mitochondrial redox signalling at a glance.
J. Cell Sci.
125
:
801
806
.
27
Emre
Y.
,
Hurtaud
C.
,
Nübel
T.
,
Criscuolo
F.
,
Ricquier
D.
,
Cassard-Doulcier
A. M.
.
2007
.
Mitochondria contribute to LPS-induced MAPK activation via uncoupling protein UCP2 in macrophages.
Biochem. J.
402
:
271
278
.
28
Olson
M. L.
,
Chalmers
S.
,
McCarron
J. G.
.
2012
.
Mitochondrial organization and Ca2+ uptake.
Biochem. Soc. Trans.
40
:
158
167
.
29
Quintana
A.
,
Hoth
M.
.
2012
.
Mitochondrial dynamics and their impact on T cell function.
Cell Calcium
52
:
57
63
.
30
Feissner
R. F.
,
Skalska
J.
,
Gaum
W. E.
,
Sheu
S. S.
.
2009
.
Crosstalk signaling between mitochondrial Ca2+ and ROS.
Front Biosci (Landmark Ed)
14
:
1197
1218
.
31
Shumilina
E.
,
Huber
S. M.
,
Lang
F.
.
2011
.
Ca2+ signaling in the regulation of dendritic cell functions.
Am. J. Physiol. Cell Physiol.
300
:
C1205
C1214
.
32
Matzner
N.
,
Zemtsova
I. M.
,
Nguyen
T. X.
,
Duszenko
M.
,
Shumilina
E.
,
Lang
F.
.
2008
.
Ion channels modulating mouse dendritic cell functions.
J. Immunol.
181
:
6803
6809
.
33
Di Sabatino
A.
,
Rovedatti
L.
,
Kaur
R.
,
Spencer
J. P.
,
Brown
J. T.
,
Morisset
V. D.
,
Biancheri
P.
,
Leakey
N. A.
,
Wilde
J. I.
,
Scott
L.
, et al
.
2009
.
Targeting gut T cell Ca2+ release-activated Ca2+ channels inhibits T cell cytokine production and T-box transcription factor T-bet in inflammatory bowel disease.
J. Immunol.
183
:
3454
3462
.
34
Frazier
A. E.
,
Dudek
J.
,
Guiard
B.
,
Voos
W.
,
Li
Y.
,
Lind
M.
,
Meisinger
C.
,
Geissler
A.
,
Sickmann
A.
,
Meyer
H. E.
, et al
.
2004
.
Pam16 has an essential role in the mitochondrial protein import motor.
Nat. Struct. Mol. Biol.
11
:
226
233
.
35
Truscott
K. N.
,
Voos
W.
,
Frazier
A. E.
,
Lind
M.
,
Li
Y.
,
Geissler
A.
,
Dudek
J.
,
Müller
H.
,
Sickmann
A.
,
Meyer
H. E.
, et al
.
2003
.
A J-protein is an essential subunit of the presequence translocase-associated protein import motor of mitochondria.
J. Cell Biol.
163
:
707
713
.
36
Putney
J. W.
 Jr.
,
Thomas
A. P.
.
2006
.
Calcium signaling: double duty for calcium at the mitochondrial uniporter.
Curr. Biol.
16
:
R812
R815
.
37
Rizzuto
R.
,
Pozzan
T.
.
2006
.
Microdomains of intracellular Ca2+: molecular determinants and functional consequences.
Physiol. Rev.
86
:
369
408
.
38
Pacher
P.
,
Sharma
K.
,
Csordás
G.
,
Zhu
Y.
,
Hajnóczky
G.
.
2008
.
Uncoupling of ER-mitochondrial calcium communication by transforming growth factor-beta.
Am. J. Physiol. Renal Physiol.
295
:
F1303
F1312
.
39
Rizzuto
R.
,
Duchen
M. R.
,
Pozzan
T.
.
2004
.
Flirting in little space: the ER/mitochondria Ca2+ liaison.
Sci. STKE
2004
:
re1
.
40
Delon
J.
,
Bercovici
N.
,
Raposo
G.
,
Liblau
R.
,
Trautmann
A.
.
1998
.
Antigen-dependent and -independent Ca2+ responses triggered in T cells by dendritic cells compared with B cells.
J. Exp. Med.
188
:
1473
1484
.
41
Gardella
S.
,
Andrei
C.
,
Lotti
L. V.
,
Poggi
A.
,
Torrisi
M. R.
,
Zocchi
M. R.
,
Rubartelli
A.
.
2001
.
CD8(+) T lymphocytes induce polarized exocytosis of secretory lysosomes by dendritic cells with release of interleukin-1beta and cathepsin D.
Blood
98
:
2152
2159
.
42
Wilson
E. B.
,
Livingstone
A. M.
.
2008
.
Cutting edge: CD4+ T cell-derived IL-2 is essential for help-dependent primary CD8+ T cell responses.
J. Immunol.
181
:
7445
7448
.
43
Zhang
S.
,
Zhang
H.
,
Zhao
J.
.
2009
.
The role of CD4 T cell help for CD8 CTL activation.
Biochem. Biophys. Res. Commun.
384
:
405
408
.
44
Wolkers
M. C.
,
Bensinger
S. J.
,
Green
D. R.
,
Schoenberger
S. P.
,
Janssen
E. M.
.
2011
.
Interleukin-2 rescues helpless effector CD8+ T cells by diminishing the susceptibility to TRAIL mediated death.
Immunol. Lett.
139
:
25
32
.
45
Vukcevic
M.
,
Zorzato
F.
,
Spagnoli
G.
,
Treves
S.
.
2010
.
Frequent calcium oscillations lead to NFAT activation in human immature dendritic cells.
J. Biol. Chem.
285
:
16003
16011
.
46
Maemura
K.
,
Zheng
Q.
,
Wada
T.
,
Ozaki
M.
,
Takao
S.
,
Aikou
T.
,
Bulkley
G. B.
,
Klein
A. S.
,
Sun
Z.
.
2005
.
Reactive oxygen species are essential mediators in antigen presentation by Kupffer cells.
Immunol. Cell Biol.
83
:
336
343
.
47
Matsue
H.
,
Edelbaum
D.
,
Shalhevet
D.
,
Mizumoto
N.
,
Yang
C.
,
Mummert
M. E.
,
Oeda
J.
,
Masayasu
H.
,
Takashima
A.
.
2003
.
Generation and function of reactive oxygen species in dendritic cells during antigen presentation.
J. Immunol.
171
:
3010
3018
.
48
Abarca-Rojano
E.
,
Muñiz-Hernández
S.
,
Moreno-Altamirano
M. M.
,
Mondragón-Flores
R.
,
Enriquez-Rincón
F.
,
Sánchez-García
F. J.
.
2009
.
Re-organization of mitochondria at the NK cell immune synapse.
Immunol. Lett.
122
:
18
25
.
49
Bugeon
L.
,
Gardner
L. M.
,
Rose
A.
,
Gentle
M.
,
Dallman
M. J.
.
2008
.
Cutting edge: Notch signaling induces a distinct cytokine profile in dendritic cells that supports T cell-mediated regulation and IL-2-dependent IL-17 production.
J. Immunol.
181
:
8189
8193
.
50
Arendt
C. W.
,
Albrecht
B.
,
Soos
T. J.
,
Littman
D. R.
.
2002
.
Protein kinase C-theta; signaling from the center of the T-cell synapse.
Curr. Opin. Immunol.
14
:
323
330
.
51
Jain
J.
,
Loh
C.
,
Rao
A.
.
1995
.
Transcriptional regulation of the IL-2 gene.
Curr. Opin. Immunol.
7
:
333
342
.
52
Berberich-Siebelt
F.
,
Klein-Hessling
S.
,
Hepping
N.
,
Santner-Nanan
B.
,
Lindemann
D.
,
Schimpl
A.
,
Berberich
I.
,
Serfling
E.
.
2000
.
C/EBPbeta enhances IL-4 but impairs IL-2 and IFN-gamma induction in T cells.
Eur. J. Immunol.
30
:
2576
2585
.

The authors have no financial conflicts of interest.

Supplementary data