Using flow cytometry, we investigated the effect of TLR agonists on human polymorphonuclear neutrophil (PMN) apoptosis in whole blood. LPS (TLR4), peptidoglycan (TLR2), R-848 (TLR7/8), and CpG-DNA (TLR9) were equally effective at delaying spontaneous apoptosis of PMN, while PamCSK4 (TLR1/2), macrophage-activating lipopeptide-2 (TLR2/6), flagellin (TLR5), and loxoribine (TLR7) were less effective or inactive. TLR agonists found to delay apoptosis also extended the functional life span of PMN. Analysis of signaling pathways revealed that the antiapoptotic effect of TLR agonists required NF-κB and PI3K activation. Furthermore, analysis of intact cells by flow cytometry showed that TLR agonists delaying PMN apoptosis increased phosphorylation of Akt, a major target of PI3K. This effect was associated with a PI3K-dependent increase in heat shock protein 27 phosphorylation, which has been reported to play a key role in PMN survival. Finally, the TLR-induced delay in PMN apoptosis was associated with increased levels of Mcl-1 and A1, which are antiapoptotic members of the Bcl-2 family. These effects were reversed by PI3K and NF-κB inhibitors, respectively. TLR activation also led to PI3K-dependent phosphorylation of the proapoptotic protein Bad. Taken together, our results strongly suggest a role of NF-κB and PI3K in TLR-induced PMN survival, leading to modulation of Bcl-2 family molecules.

Polymorphonuclear neutrophils (PMN) 2 are key components of the first line of defense against microbial pathogens (1). They contribute to the early innate response by rapidly migrating into inflamed tissues, where their activation triggers microbicidal mechanisms such as release of proteolytic enzymes and antimicrobial peptides, and rapid production of reactive oxygen species (ROS). PMN activation is initiated upon recognition of Ab- or complement-opsonized particles (2). PMN also directly recognize microbial products via pattern recognition receptors such as TLR (3). Ten human TLRs have so far been identified, mediating responses to pathogen-associated molecular patterns (PAMPs) shared by many microorganisms. Human PMN have been reported to express all TLRs except TLR3 (4). TLRs are members of the IL-1R superfamily and share a common activation pathway mediated by their Toll/IL-1R signaling domain, resulting in activation of NF-κB and MAPK (5). Despite these shared pathways, TLRs probably show differences in their rate, intensity, or efficiency of activation, involving unidentified mechanisms. Selective pathways are reported to be triggered by some TLRs; in particular, TLR2, TLR4, and TLR9 can activate the PI3K pathway (6, 7). Activation of cell signaling cascades triggers immune responses leading to pathogen eradication.

PMN are usually short-lived immune cells, but the prolongation of their life span is critical in their efficiency against pathogens (8). PMN activation and survival is likely to be tightly regulated, as the cytotoxic substances they release can damage adjacent healthy tissue (9). Many inflammatory mediators, including cytokines, regulate cell survival by interfering with apoptosis (10, 11, 12, 13). Regulation of PMN survival has been widely studied but remains to be fully elucidated. In particular, the impact on PMN apoptosis of PAMPs recognized by the different TLRs has rarely been investigated. LPS, a TLR4 ligand, and peptidoglycan (PGN), a TLR2 ligand, delay PMN apoptosis through poorly known mechanisms (14, 15). No data on modulation of Bcl-2 family proteins after TLR activation have been reported.

In this study we report the first analysis of the differential effects of TLR agonists on apoptosis of human PMN studied in whole blood to minimize PMN activation during their isolation and to mimic physiological conditions. We used PGN from Staphylococcus aureus (a TLR2-selective stimulus), synthetic palmitoylated mimics of bacterial lipopeptides (Pam3CSK4, a TLR1/2 heterodimer stimulus), macrophage-activating lipopeptide-2 (MALP-2, a TLR2/6 heterodimer stimulus), purified LPS (a TLR4 stimulus), bacterial flagellin (a TLR5 stimulus), loxoribine (a guanosine analog and TLR7-selective stimulus), an imidazoquinoline pharmaceutical (R848, a TLR7/8 heterodimer stimulus) and unmethylated CpG-DNA (a TLR9 stimulus). We also examined mechanisms downstream of TLR signaling pathways leading to PMN survival by studying the participation of NF-κB, PI3K/Akt, and MAPK pathways, and modulation of Bcl-2 family proteins.

The reagents used and their sources were as follows: ultrapurified LPS from Escherichia coli serotype R515 (LPS), purified flagellin from Salmonella typhimurium, and synthetic MALP-2 (Alexis); PGN from S. aureus, R-848, the guanosine analog loxoribine, and a synthetic palmitoylated mimic of bacterial lipopeptides (Pam3CSK4) (Invivogen); unmethylated CpG-DNA (HyCult Biotechnology); hydroethidine (HE; Fluka); fMLP and rottlerin (Sigma-Aldrich); cycloheximide, diphenyleneiodonium (DPI), SN50, SB203580, PD98059, genistein, wortmannin, and GF109203X (Calbiochem); 3,3′-dihexyloxacarbocyanine iodide (DiOC6) (Molecular Probes); allophycocyanin-conjugated annexin V, 7-aminoactinomycin D (7-AAD), FITC-anti-CD15, allophycocyanin-anti-CD15, anti-Akt (protein kinase B α (PKBα)), anti-phospho-Akt (S472/S473), FITC-conjugated anti-active caspase-3, FITC-conjugated anti-Bad mAbs, and FITC-conjugated goat anti-rabbit Ab (BD Pharmingen, BD Biosciences); PE-conjugated anti-CD11b mAb, PE-anti-CD45 mAb (Immunotech); FITC-conjugated goat anti-mouse Ab (Nordic Immunology); anti-phospho-IκB kinase α (IKKα) (Ser180)/IKKβ (Ser181), anti-heat shock protein (Hsp) 27 mAbs, anti-phospho-Bad (Ser136) and anti-phospho-Hsp27 (Ser82) rabbit polyclonal Abs (Cell Signaling Technology); anti-Mcl-1 mAb and anti-A1/Bfl-1 rabbit polyclonal Ab (Santa Cruz Biotechnology); anti-TLR2 mAb, anti-TLR3 mAb, anti-TLR5, and anti-TLR7 polyclonal Abs (Imgenex).

One milliliter samples of fresh blood from healthy donors, collected onto lithium heparinate (10 U/ml) were incubated in 24-well tissue cultures plates at 37°C with 5% CO2 for various times with PBS or the following TLR agonists: LPS (0.001–10 ng/ml), PGN (0.01–1 μg/ml), R-848 (0.01–10 μg/ml), CpG-DNA (10–100 μg/ml), Pam3CSK4 (10–500 ng/ml), MALP-2 (0.01–10 ng/ml), flagellin (0.01–100 ng/ml), or loxoribine (0.1–1000 μM). Cycloheximide (10 μg/ml) and rhGM-CSF (1000 pg/ml) were used as proapoptotic (16) and antiapoptotic (12) controls, respectively.

In some experiments, samples were pretreated for 20 min with 10 μM DPI, an inhibitor of NADPH oxidase, or for 1 h with the NF-κB inhibitor SN50 (100 μg/ml) or kinase inhibitors at optimal concentrations previously determined in whole blood (wortmannin, 2500 nM; GF109203X, 5 μM; genistein, 100 μM; PD98059, 50 μM; SB203580, 25 μM; rottlerin, 10 μM).

Human PMN were isolated in LPS-free conditions by Dextran sedimentation and Ficoll-Hypaque centrifugation of freshly drawn blood; PMN were further purified by negative selection with pan anti-human HLA class II-coated magnetic beads (Miltenyi Biotec) to deplete B lymphocytes, activated T lymphocytes, and monocytes, as previously described (17). Less than 0.5% of cells were positive by nonspecific esterase staining, and flow cytometry showed the absence of CD45+/CD14high, CD45+/CD3+, or CD45+/CD19+ cells; this showed that the PMN were highly purified, without contaminating monocytes.

Apoptosis was quantified by staining with annexin V and a vital dye (7-AAD) (18). After pretreatment of whole blood with TLR agonists for 2–48 h, samples (100 μl) were washed twice in PBS, incubated on ice with FITC-anti-CD15 and PE-anti-CD45 for 15 min, and then with allophycocyanin-annexin V for 15 min. After dilution in PBS (500 μl), samples were incubated with 7-AAD at room temperature for 15 min and analyzed immediately by flow cytometry. PMN were identified on the CD15/side scatter (SSC) dot plot (see Fig. 1,B); use of the combination of allophycocyanin-annexin V and 7-AAD differentiates between early apoptotic PMN (annexin V+, 7-AAD) and late apoptotic PMN (annexin V+, 7-AAD+) (see Fig. 1,C). Fig. 1 A (after CD45 gating) shows an accurate distribution of the different leukocyte populations on the forward scatter (FSC)/SSC dot plot after 8 h of incubation. We checked that the percentage of apoptotic PMN, determined by flow cytometry on whole blood, correlated with the percentage obtained by morphological assessment on cytospin preparation stained with May-Grünwald Giemsa (not shown).

FIGURE 1.

Study of PMN apoptosis in whole blood by flow cytometry. Whole blood samples were incubated with PBS in 24-well tissue cultures plates at 37°C with 5% CO2 for 2, 4, 8, 12, 15, and 24 h. Samples were then incubated at 4°C for 15 min with FITC-anti-CD15 and PE-anti-CD45 Abs and stained with annexin V and 7-AAD as described in Materials and Methods. A, Morphological parameters of leukocytes after gating on CD45+ cells. The fluorescence of anti-CD15 Ab was used to identify PMN as CD15+ cells and to gate out other cells, erythrocytes, and debris. A gate was drawn around the PMN population (B). Fluorescence analysis was performed on this gate. The combination of allophycocyanin-annexin V and 7-AAD distinguished between early apoptotic cells (annexin V+, 7-AAD), late apoptotic cells (annexin V+, 7-AAD+), necrotic cells (annexin V, 7-AAD+), and viable cells (unstained) (C). D, The kinetics of PMN apoptosis in whole blood (♦, total annexin V+; □, annexin V+, 7-AAD) (n = 3).

FIGURE 1.

Study of PMN apoptosis in whole blood by flow cytometry. Whole blood samples were incubated with PBS in 24-well tissue cultures plates at 37°C with 5% CO2 for 2, 4, 8, 12, 15, and 24 h. Samples were then incubated at 4°C for 15 min with FITC-anti-CD15 and PE-anti-CD45 Abs and stained with annexin V and 7-AAD as described in Materials and Methods. A, Morphological parameters of leukocytes after gating on CD45+ cells. The fluorescence of anti-CD15 Ab was used to identify PMN as CD15+ cells and to gate out other cells, erythrocytes, and debris. A gate was drawn around the PMN population (B). Fluorescence analysis was performed on this gate. The combination of allophycocyanin-annexin V and 7-AAD distinguished between early apoptotic cells (annexin V+, 7-AAD), late apoptotic cells (annexin V+, 7-AAD+), necrotic cells (annexin V, 7-AAD+), and viable cells (unstained) (C). D, The kinetics of PMN apoptosis in whole blood (♦, total annexin V+; □, annexin V+, 7-AAD) (n = 3).

Close modal

Some experiments were performed with highly purified PMN (1 × 106/ml) incubated with TLR agonists at optimal concentrations for 8 h and then with allophycocyanin-annexin V and 7-AAD before flow cytometry.

The mitochondrial apoptotic pathway was detected as a loss of the mitochondrial transmembrane potential (Δψm), measured as a reduction of the DiOC6 incorporation (19). After pretreatment of whole blood at 37°C for 8 h with PBS or TLR agonists, samples (100 μl) were loaded with DiOC6 (40 nM) for 15 min at 37°C and then stained with allophycocyanin-anti-CD15 for 30 min at 4°C. Samples were then suspended in PBS and analyzed immediately by flow cytometry.

Leukocytes (obtained after red cell lysis with FACS lysing solution) were fixed with 2% paraformaldehyde-PBS for 10 min at 37°C. After one wash with PBS, leukocytes were incubated in ice-cold PBS-90% methanol in the dark for 30 min at 4°C to permeabilize the membranes as previously described (20). After one wash with PBS-human serum albumin (HSA) (2%), cells were stained with FITC-conjugated anti-active caspase-3 for 1 h at room temperature. After one wash (400 × g for 5 min), cells were resuspended in 1% paraformaldehyde-PBS and analyzed by flow cytometry.

Superoxide anion () production was measured with a flow cytometric assay derived from the HE oxidation technique (21): HE diffuses into cells and, during the PMN oxidative burst, nonfluorescent intracellular HE is oxidized by to highly fluorescent ethidium (E+), that is trapped in the nucleus by intercalation into DNA. After the different treatments, whole blood (1 ml) was loaded for 15 min with HE (1500 ng/ml) at 37°C, followed by PBS or 10−6 M fMLP for 5 min. The reaction was stopped and RBC were lysed as described above. After one wash, leukocytes were resuspended in 1% paraformaldehyde-PBS.

After the different treatments, whole blood was incubated at 37°C with PBS or 10−6 M fMLP for 5 min. Samples (100 μl) were stained at 4°C for 30 min with FITC-anti-human CD11b.

After incubation of whole blood with TLR agonists or PBS for various times at 37°C, leukocytes were permeabilized in 90% methanol as previously described (20). Cells were then stained with anti-Akt, anti-Akt phosphospecific, anti-Hsp27, anti-Hsp27 phosphospecific, anti-Mcl-1, anti-A1/Bfl-1, or anti-Bad phosphospecific Abs for 1 h at room temperature and washed once in PBS-2% HSA. Samples were then incubated for 30 min with FITC-goat anti-mouse or anti-rabbit Ab. Bad content was studied by staining with FITC-conjugated anti-Bad. After one wash, leukocytes were resuspended in 1% paraformaldehyde-PBS and analyzed by flow cytometry.

We used a BD Immunocytometry Systems FACSCalibur. To measure apoptosis in whole blood, PMN were identified on the CD15/SSC dot plot and 2 × 105 events were counted per sample. In other experiments, FSC and SSC were used to identify the PMN population and to gate out other cells and debris; 10,000 events were counted per sample. All the results were obtained with a constant photomultiplier gain. The data were analyzed using CellQuest software (BD Biosciences), and mean fluorescence intensity (MFI) was used to quantify the responses. Caspase-3 activity was expressed in relative units (percentage of caspase-3high cells × MFI). Nonspecific Ab binding was determined on cells incubated with the same concentration of the corresponding isotype control or with nonimmune serum.

Suspensions of 40 × 106 PMN/ml in PBS buffer were treated with 2.7 mM diisopropylfluorophosphate for 20 min at 4°C and pelleted at 400 × g for 8 min at 4°C. The pellet was resuspended in CHAPS solubilization buffer containing 50 mM Tris, pH 7.5, 15 mM CHAPS, 1 mM EDTA and antiproteases. The cells were incubated on ice and the suspension was then centrifuged at 1500 × g for 5 min. Following SDS-PAGE on 10% acrylamide gels, proteins were transferred to nitrocellulose filters. The filters were incubated for 1 h at room temperature in 50 mM Tris, 150 mM NaCl, 0.1% Tween 20 (TBST) containing 5% (w/v) fat-free dried milk. Nitrocellulose membranes were incubated overnight with specific Abs against TLR2, TLR3, TLR5, and TLR7 at 1/500 dilution. Following five washes with TBST, the membranes were incubated with goat anti-mouse or goat anti-rabbit Abs conjugated to HRP. After five washes with TBST, revelation was performed with a chemiluminescence method (ECL; Amersham Life Sciences) following the manufacturer’s instructions.

Data are reported as means ± SEM. Comparisons were based on ANOVA and Tukey’s posthoc test, using Prism 3.0 software (Graph Pad Software).

Whole blood PMN cultured at 37°C died by apoptosis: ∼26 and 70% of cells were annexin V+ after 8 and 24 h, respectively (Fig. 1,D). As previously reported with isolated PMN (12, 16), apoptosis was accelerated by cycloheximide (65% of annexin V+ cells after 8 h), and delayed by GM-CSF (4% of annexin V+ cells after 8 h). As shown in Fig. 2, the percentage of total annexin V+ cells, and the percentage of annexin V+, 7-AAD cells, fell significantly, in a concentration-dependent manner after 8 h of treatment with all TLR agonists except flagellin (TLR5) and loxoribine (TLR7). We checked that environmental LPS did not contribute to the effect of the TLR agonists (except for the TLR4 agonist, which is purified LPS). In fact, the prolongation of PMN survival by PGN, R-848, CpG-DNA, Pam3CSK4, and MALP-2 was not modified by preincubation with a TLR4 neutralizing Ab, while the effect of LPS on PMN survival was completely abolished (not shown). PMN apoptosis, measured in terms of total annexin V+ PMN, fell to a similar extent after incubation for 8 h with LPS, PGN, R-848, and CpG-DNA at optimal concentrations. This inhibition was significantly stronger than that induced by Pam3CSK4 and MALP-2 (Table I). Similar levels of inhibition were found in the early stage of PMN apoptosis (annexin V+/7AAD cells; not shown). A kinetic study showed that LPS, PGN, R-848, and CpG-DNA induced ∼50% inhibition of PMN apoptosis after 2 h, rising to a maximum of 80% after 8 h. In samples treated with all the TLR agonists except flagellin and loxoribine, 80% of PMN were annexin V+ after 48 h incubation, compared with 24 h in PBS-treated samples, further showing that TLR agonists delay PMN apoptosis (Fig. 3).

FIGURE 2.

Concentration-dependent inhibition of PMN apoptosis by TLR agonists. Whole blood samples were incubated with PBS or TLR agonists at various concentrations in 24-well tissue cultures plates at 37°C with 5% CO2 for 8 h. Apoptosis was quantified by staining with allophycocyanin-annexin V and 7-AAD as described in the legend of Fig. 1. Results are expressed as the percentage of total annexin V+ cells (♦) and annexin V+ and 7-AAD cells (early apoptosis) (□). The proportion of necrotic cells (annexin V, 7-AAD+) was always lower than 2%. The percentage of spontaneous apoptotic circulating PMN (annexin V+) was 0.64 ± 0.14. Values are means ± SEM (n = 10). ∗, Significantly different from sample incubated with PBS (p < 0.05).

FIGURE 2.

Concentration-dependent inhibition of PMN apoptosis by TLR agonists. Whole blood samples were incubated with PBS or TLR agonists at various concentrations in 24-well tissue cultures plates at 37°C with 5% CO2 for 8 h. Apoptosis was quantified by staining with allophycocyanin-annexin V and 7-AAD as described in the legend of Fig. 1. Results are expressed as the percentage of total annexin V+ cells (♦) and annexin V+ and 7-AAD cells (early apoptosis) (□). The proportion of necrotic cells (annexin V, 7-AAD+) was always lower than 2%. The percentage of spontaneous apoptotic circulating PMN (annexin V+) was 0.64 ± 0.14. Values are means ± SEM (n = 10). ∗, Significantly different from sample incubated with PBS (p < 0.05).

Close modal
Table I.

Percentage inhibition of PMN apoptosis by TLR agonistsa

Percentage Inhibitionb
Whole BloodIsolated PMN
PBS 
LPS (TLR4) 87.4 ± 2.4c 70.3 ± 7.8c 
PGN (TLR2) 81.8 ± 7.3c 69.1 ± 4.9c 
R-848 (TLR7/8) 83.5 ± 4.1c 66.7 ± 1.9c 
CpG-DNA (TLR9) 75.2 ± 4.0c 65.1 ± 3.2c 
Pam3CSK4 (TLR1/2) 60.2 ± 5.8cd 57.3 ± 2.3c 
MALP-2 (TLR2/6) 58.1 ± 6.6cd 49.3 ± 5.8cd 
Flagellin (TLR5) 16.1 ± 7.2 12.7 ± 5.4 
Loxoribine (TLR7) 10.6 ± 4.5 13.6 ± 3.2 
Percentage Inhibitionb
Whole BloodIsolated PMN
PBS 
LPS (TLR4) 87.4 ± 2.4c 70.3 ± 7.8c 
PGN (TLR2) 81.8 ± 7.3c 69.1 ± 4.9c 
R-848 (TLR7/8) 83.5 ± 4.1c 66.7 ± 1.9c 
CpG-DNA (TLR9) 75.2 ± 4.0c 65.1 ± 3.2c 
Pam3CSK4 (TLR1/2) 60.2 ± 5.8cd 57.3 ± 2.3c 
MALP-2 (TLR2/6) 58.1 ± 6.6cd 49.3 ± 5.8cd 
Flagellin (TLR5) 16.1 ± 7.2 12.7 ± 5.4 
Loxoribine (TLR7) 10.6 ± 4.5 13.6 ± 3.2 
a

Whole blood or isolated PMN were incubated in 24-well tissue cultures plates at 37°C with 5% CO2 for 8 h with PBS or TLR agonists at the following optimal concentrations: LPS, 10 ng/ml (TLR4); PGN, 1 μg/ml (TLR2); R-848, 10 μg/ml (TLR7/8); CpG-DNA, 100 μg/ml (TLR9); Pam3CSK4, 500 ng/ml (TLR1/2); MALP-2, 10 ng/ml (TLR2/6); flagellin, 100 ng/ml (TLR5); or loxoribine, 100 μM (TLR7). PMN were identified by using a FITC anti-CD15 Ab. Apoptosis was quantified by staining with allophycocyanin-annexin V and 7-AAD as described in Materials and Methods.

b

Results are expressed as the percentage inhibition of PMN apoptosis [1 − (% of total annexin V+ PMN in TLR agonist-treated sample/% of total annexin V+ PMN in PBS-treated sample)] × 100. Values are means ± SEM (n = 5).

c

Significantly different from sample incubated with PBS (percentage inhibition of PMN apoptosis = 0) and samples incubated with flagellin or loxoribine (p < 0.05).

d

Significantly different from samples incubated with LPS, PGN, R848, or CpG (p < 0.05).

FIGURE 3.

Kinetics of PMN apoptosis after treatment with LPS, PGN, R-848, CpG-DNA, Pam3CSK4, and MALP-2. Whole blood samples were incubated in 24-well tissue cultures plates at 37°C with 5% CO2 for various times with PBS or the following TLR agonists: LPS, 10 ng/ml (TLR4); PGN, 1 μg/ml (TLR2); R-848, 10 μg/ml (TLR7/8); CpG-DNA, 100 μg/ml (TLR9); Pam3CSK4, 500 ng/ml (TLR1/2); MALP-2, 10 ng/ml (TLR2/6). Apoptosis was quantified by staining with allophycocyanin-annexin V and 7-AAD as described in the legend of Fig. 1. Results are expressed as the percentage of total annexin V+ cells. Values are means ± SEM (n = 3). ∗, Significantly different from sample incubated with PBS (p < 0.05).

FIGURE 3.

Kinetics of PMN apoptosis after treatment with LPS, PGN, R-848, CpG-DNA, Pam3CSK4, and MALP-2. Whole blood samples were incubated in 24-well tissue cultures plates at 37°C with 5% CO2 for various times with PBS or the following TLR agonists: LPS, 10 ng/ml (TLR4); PGN, 1 μg/ml (TLR2); R-848, 10 μg/ml (TLR7/8); CpG-DNA, 100 μg/ml (TLR9); Pam3CSK4, 500 ng/ml (TLR1/2); MALP-2, 10 ng/ml (TLR2/6). Apoptosis was quantified by staining with allophycocyanin-annexin V and 7-AAD as described in the legend of Fig. 1. Results are expressed as the percentage of total annexin V+ cells. Values are means ± SEM (n = 3). ∗, Significantly different from sample incubated with PBS (p < 0.05).

Close modal

As cytokine production by contaminating monocytes has been implicated in TLR agonist-induced inhibition of PMN apoptosis at later times (22 h) but not at earlier times (4 h) (22), we examined the effect of 8 h of incubation with TLR agonists on highly purified PMN depleted of all monocytes as previously described (17). TLR agonist-induced inhibition of PMN apoptosis was similar whether apoptosis was studied in whole blood or in highly purified preparations, suggesting a direct effect of TLR agonists on PMN apoptosis. However, apoptosis inhibition was slightly less effective with purified PMN than with whole blood: the protective agonists induced 38–69% inhibition of PMN apoptosis in purified preparations and 46–87% in whole blood (Table I). To minimize cell activation during isolation and to better mimic physiological conditions, the following experiments were performed with PMN in their whole blood environment.

After 8 h of incubation, PBS-treated PMN exhibited a reduced capacity to produce superoxide anion () and to increase surface CD11b expression in response to fMLP relative to baseline (Fig. 4). This impairment was found with both annexin V and annexin V+ PMN, although annexin V+ cells were less potent than annexin V cells. In contrast, incubation for 8 h with the TLR agonists found to delay apoptosis increased production in the basal state and also in response to fMLP (LPS was the most potent agonist in terms of increasing production) (Fig. 4,A). This impact of the TLR agonists on ROS production did not differ significantly according to annexin V staining, although the stimulation index (MFI of fMLP-stimulated sample/MFI of unstimulated sample) was slightly lower in annexin V+ than annexin V populations. Similar results were observed concerning PMN surface CD11b expression (Fig. 4 B). Flagellin and loxoribine did not significantly modify PMN responses. This increased ROS production was not involved in the effects of TLR agonists on PMN apoptosis, as incubation with DPI, a NADPH oxidase inhibitor, altered neither the intensity nor the kinetics of apoptosis inhibition (not shown).

FIGURE 4.

TLR agonists extend the PMN functional life span. PMN functions were analyzed in whole blood immediately after sampling (T0h) or after 8 h of incubation (T8h) in 24-well tissue cultures plates at 37°C with 5% CO2, with PBS or LPS, 10 ng/ml (TLR4); PGN, 1 μg/ml (TLR2); R-848, 10 μg/ml (TLR7/8); CpG-DNA, 100 μg/ml (TLR9); Pam3CSK4, 500 ng/ml (TLR1/2); MALP2, 10 ng/ml (TLR2/6); flagellin, 100 ng/ml (TLR5); or loxoribine, 100 μM (TLR7). At T0h and T8h, whole blood was treated with PBS or fMLP (10−6 M) for 5 min. PMN oxidative burst (A) and CD11b expression (B) at the PMN surface were studied as described in Materials and Methods. Values are means ± SEM (n = 4). ∗, Significantly different from the T0h sample stimulated with fMLP (p < 0.05). †, Significantly different at T8h from the samples incubated for 8 h with PBS, flagellin, or loxoribine without fMLP stimulation (p < 0.05). ‡, Significantly different at T8h from the samples incubated for 8 h with PBS, flagellin, or loxoribine and stimulated with fMLP (p < 0.05).

FIGURE 4.

TLR agonists extend the PMN functional life span. PMN functions were analyzed in whole blood immediately after sampling (T0h) or after 8 h of incubation (T8h) in 24-well tissue cultures plates at 37°C with 5% CO2, with PBS or LPS, 10 ng/ml (TLR4); PGN, 1 μg/ml (TLR2); R-848, 10 μg/ml (TLR7/8); CpG-DNA, 100 μg/ml (TLR9); Pam3CSK4, 500 ng/ml (TLR1/2); MALP2, 10 ng/ml (TLR2/6); flagellin, 100 ng/ml (TLR5); or loxoribine, 100 μM (TLR7). At T0h and T8h, whole blood was treated with PBS or fMLP (10−6 M) for 5 min. PMN oxidative burst (A) and CD11b expression (B) at the PMN surface were studied as described in Materials and Methods. Values are means ± SEM (n = 4). ∗, Significantly different from the T0h sample stimulated with fMLP (p < 0.05). †, Significantly different at T8h from the samples incubated for 8 h with PBS, flagellin, or loxoribine without fMLP stimulation (p < 0.05). ‡, Significantly different at T8h from the samples incubated for 8 h with PBS, flagellin, or loxoribine and stimulated with fMLP (p < 0.05).

Close modal

Thus, TLR agonists found to delay apoptosis also extended the functional life span of PMN.

As flagellin (a TLR5 agonist) and loxoribine (a TLR7 agonist) did not delay PMN apoptosis, in contrast to the other TLR agonists, we checked that TLR5 and TLR7 were expressed in PMN. TLR2 and TLR3 were used as positive and negative controls, respectively (4). Fig. 5 shows that TLR2, TLR5, and TLR7 were expressed in PMN, contrary to TLR3.

FIGURE 5.

Expression of TLR5 and TLR7 by Western blotting. A total of 2.5 × 106 cell equivalents were loaded in each well. Following SDS-PAGE, the proteins were transferred to nitrocellulose membranes and then incubated with anti-human TLR-2, TLR-3, TLR-5, TLR-7 Abs at 1/500 dilution overnight. The Western blots were revealed as described in Materials and Methods.

FIGURE 5.

Expression of TLR5 and TLR7 by Western blotting. A total of 2.5 × 106 cell equivalents were loaded in each well. Following SDS-PAGE, the proteins were transferred to nitrocellulose membranes and then incubated with anti-human TLR-2, TLR-3, TLR-5, TLR-7 Abs at 1/500 dilution overnight. The Western blots were revealed as described in Materials and Methods.

Close modal

To identify the stage of the apoptotic program at which TLR agonists act to delay apoptosis, we measured the effects of TLR agonists on caspase-3 activity and Δψm. As shown in Table II caspase-3 activity was detected during spontaneous PMN apoptosis after 8 h incubation with PBS at 37°C, and was significantly reduced by the TLR agonists that delayed PMN apoptosis (LPS, PGN, R-848, CpG-DNA, Pam3CSK4, MALP-2). In contrast, flagellin and loxoribine did not significantly modify caspase-3 activity relative to PBS. The proportion of DiOC6low (loss of Δψm) cells was significantly reduced after treatment with the active TLR agonists, relative to both PBS, flagellin and loxoribine. Furthermore, both caspase-3 activity and the percentage of DiOC6low cells correlated with the percentage of annexin V+ cells.

Table II.

Caspase-3 activity and mitochondrial membrane integritya

Caspase-3 Activity (Relative Units)bDiOC6low Cells (Percentage)c
PBS 290.3 ± 38.6 39.5 ± 4.5 
LPS (TLR4) 66.5 ± 19.8b 14.5 ± 1.5b 
PGN (TLR2) 73.1 ± 27.6b 16.5 ± 2.5b 
R-848 (TLR7/8) 66.6 ± 23.4b 11.7 ± 0.8b 
CpG-DNA (TLR9) 97.2 ± 28.8b 17.0 ± 1.5b 
Pam3CSK4 (TLR1/2) 123.1 ± 15.5b 19.2 ± 2.1b 
MALP-2 (TLR2/6) 154.1 ± 12.4b 17.0 ± 1.2b 
Flagellin (TLR5) 228.4 ± 25.6 31.0 ± 4.3 
Loxoribine (TLR7) 235.2 ± 18.2 34.3 ± 1.8 
Caspase-3 Activity (Relative Units)bDiOC6low Cells (Percentage)c
PBS 290.3 ± 38.6 39.5 ± 4.5 
LPS (TLR4) 66.5 ± 19.8b 14.5 ± 1.5b 
PGN (TLR2) 73.1 ± 27.6b 16.5 ± 2.5b 
R-848 (TLR7/8) 66.6 ± 23.4b 11.7 ± 0.8b 
CpG-DNA (TLR9) 97.2 ± 28.8b 17.0 ± 1.5b 
Pam3CSK4 (TLR1/2) 123.1 ± 15.5b 19.2 ± 2.1b 
MALP-2 (TLR2/6) 154.1 ± 12.4b 17.0 ± 1.2b 
Flagellin (TLR5) 228.4 ± 25.6 31.0 ± 4.3 
Loxoribine (TLR7) 235.2 ± 18.2 34.3 ± 1.8 
a

Whole blood samples were incubated for 8 h in 24-well tissue cultures plates at 37°C with 5% CO2, with either PBS or the following TLR agonists: LPS, 10 ng/ml (TLR4); PGN, 1 μg/ml (TLR2); R-848, 10 μg/ml (TLR7/8); CpG-DNA; 100 μg/ml (TLR9); Pam3CSK4, 500 ng/ml (TLR1/2); MALP2, 10 ng/ml (TLR2/6); flagellin, 100 ng/ml (TLR5), or loxoribine, 100 μM (TLR7). Caspase-3 activity (b) and retention of DiOC6 (c) by PMN were measured as described in Materials and Methods. Caspase-3 activity is expressed in relative units (percentage of caspase-3high cells × MFI) and retention of DiOC6 in percentage of DiOC6low cells. Values are means ± SEM (n = 3).

b

Significantly different from sample incubated with PBS and samples incubated with flagellin or loxoribine (p < 0.05).

These results suggest that apoptosis was delayed, at least in part, through an effect at or upstream of the mitochondrial membrane permeabilization.

NF-κB activation has been implicated in PMN survival (23), and particularly in LPS-induced PMN survival. However, the involvement of the NF-κB pathway in the effects of TLR agonists that delay PMN apoptosis has not been comparatively investigated.

To determine the involvement of the NF-κB pathway in whole blood, we first used an inhibitor-based approach. The NF-κB inhibitor (SN50) had no significant effect on spontaneous apoptosis: the percentage of annexin V+ cells was 24.2 ± 4.5 and 25.1 ± 3.8 in samples incubated with PBS and SN50, respectively. As shown in Fig. 6,A, SN50 (100 μg/ml), which inhibits nuclear translocation of NF-κB, significantly reduced TLR-mediated inhibition of PMN apoptosis at 8 h, suggesting a role of NF-κB. The control peptide SN50M did not significantly alter PMN apoptosis, whatever the TLR agonist used. Another NF-κB inhibitor, the sesquiterpene lactone parthenolide (10 μM) also inhibited TLR-induced inhibition of PMN apoptosis (not shown). To confirm these findings, we used flow cytometry and a mouse anti-phospho-IKK mAb, and found that the protective agonists significantly induced IKK phosphorylation relative to PBS-treated samples (Fig. 6 B).

FIGURE 6.

Involvement of the NF-κB signaling pathway in TLR agonist-induced PMN survival. A, Effect of an NF-κB inhibitor (SN50) on TLR agonist-induced PMN survival. Whole blood samples were incubated in 24-well tissue cultures plates at 37°C with 5% CO2 with SN50 (100 μg/ml) or PBS for 1 h. Samples were then treated with PBS or the following TLR agonists: LPS, 10 ng/ml (TLR4); PGN, 1 μg/ml (TLR2); R-848, 10 μg/ml (TLR7/8); CpG-DNA, 100 μg/ml (TLR9); Pam3CSK4, 500 ng/ml (TLR1/2); MALP-2, 10 ng/ml (TLR2/6). Apoptosis was quantified as described in the legend of Fig. 1. Results are expressed as the percentage inhibition of PMN apoptosis [1 − (% of total annexin V+ PMN in TLR agonist-treated sample/% of total annexin V+ PMN in PBS-treated sample)] × 100. The SN50M control peptide did not significantly alter apoptosis whatever the TLR agonist used. Values are means ± SEM (n = 3). ∗, Significantly different from sample incubated with PBS (p < 0.05). B, Effect of TLR agonists on IKK phosphorylation. Samples were incubated in 24-well tissue cultures plates at 37°C with 5% CO2 for 5 min with PBS or with TLR agonists: LPS, 10 ng/ml (TLR4); PGN, 1 μg/ml (TLR2); R-848, 10 μg/ml (TLR7/8); CpG-DNA, 100 μg/ml (TLR9); Pam3CSK4, 500 ng/ml (TLR1/2); or MALP-2, 10 ng/ml (TLR2/6); flagellin, 100 ng/ml (TLR5); or loxoribine, 100 μM (TLR7). Phospho-IKK content was then measured by flow cytometry on methanol-permeabilized cells as described in Materials and Methods. Results are MFIs. Values obtained with an irrelevant Ab of the same isotype or with nonimmune serum were subtracted. ∗, Significantly different from samples incubated with PBS, flagellin, or loxoribine (p < 0.05).

FIGURE 6.

Involvement of the NF-κB signaling pathway in TLR agonist-induced PMN survival. A, Effect of an NF-κB inhibitor (SN50) on TLR agonist-induced PMN survival. Whole blood samples were incubated in 24-well tissue cultures plates at 37°C with 5% CO2 with SN50 (100 μg/ml) or PBS for 1 h. Samples were then treated with PBS or the following TLR agonists: LPS, 10 ng/ml (TLR4); PGN, 1 μg/ml (TLR2); R-848, 10 μg/ml (TLR7/8); CpG-DNA, 100 μg/ml (TLR9); Pam3CSK4, 500 ng/ml (TLR1/2); MALP-2, 10 ng/ml (TLR2/6). Apoptosis was quantified as described in the legend of Fig. 1. Results are expressed as the percentage inhibition of PMN apoptosis [1 − (% of total annexin V+ PMN in TLR agonist-treated sample/% of total annexin V+ PMN in PBS-treated sample)] × 100. The SN50M control peptide did not significantly alter apoptosis whatever the TLR agonist used. Values are means ± SEM (n = 3). ∗, Significantly different from sample incubated with PBS (p < 0.05). B, Effect of TLR agonists on IKK phosphorylation. Samples were incubated in 24-well tissue cultures plates at 37°C with 5% CO2 for 5 min with PBS or with TLR agonists: LPS, 10 ng/ml (TLR4); PGN, 1 μg/ml (TLR2); R-848, 10 μg/ml (TLR7/8); CpG-DNA, 100 μg/ml (TLR9); Pam3CSK4, 500 ng/ml (TLR1/2); or MALP-2, 10 ng/ml (TLR2/6); flagellin, 100 ng/ml (TLR5); or loxoribine, 100 μM (TLR7). Phospho-IKK content was then measured by flow cytometry on methanol-permeabilized cells as described in Materials and Methods. Results are MFIs. Values obtained with an irrelevant Ab of the same isotype or with nonimmune serum were subtracted. ∗, Significantly different from samples incubated with PBS, flagellin, or loxoribine (p < 0.05).

Close modal

However, the NF-κB inhibitors only partially reversed TLR agonist-induced inhibition of PMN apoptosis, suggesting the involvement of other signaling pathways.

TLRs may activate many other signaling pathways, a number of which have already been implicated in the regulation of PMN life span, including MAPKs (24, 25), PI3K (26, 27), and tyrosine kinases (28).

To investigate the signaling pathways involved in TLR agonist-induced inhibition of apoptosis, we first examined the effects of various kinase inhibitors on PMN in whole blood incubated with TLR agonists for 8 h. We checked that kinase inhibitors alone did not alter PMN apoptosis; in particular, the percentage of annexin V+ cells was 25.3 ± 5.2 and 23.2 ± 4.7 in samples incubated with PBS and wortmannin, respectively. As shown in Fig. 7,A, only wortmannin (a PI3K inhibitor) attenuated LPS-dependent inhibition of apoptosis. Similar effect was observed with a second PI3K inhibitor, LY2940002 (25 μM) (not shown). Blockade of both the NF-κB and PI3K pathways potentiated this effect (18% apoptosis inhibition with LPS + wortmannin + SN50). GF109203X (a protein kinase C (PKC) inhibitor), genistein (a broadly specific tyrosine kinase inhibitor), PD98059 (a MEK1/2 kinase inhibitor), SB203580 (a p38MAPK inhibitor), and rottlerin (a PKCδ inhibitor) had no effects on LPS-induced PMN survival whatever the concentrations used. Similar results were observed with the other TLR agonists that delayed neutrophil apoptosis (not shown). As shown in Fig. 7 B, wortmannin has a similar inhibitory effect on LPS, PGN, R-848, CpG-DNA, Pam3CSK4, and MALP-2-induced PMN survival. Wortmannin also inhibited the effect of TLR agonists on caspase-3 activity, and reduced the percentage of DiOC6low cells (not shown).

FIGURE 7.

Effect of kinase inhibitors on TLR agonist-induced PMN survival. A, Whole blood samples were first incubated in 24-well tissue cultures plates at 37°C with 5% CO2 with PBS, PI3K inhibitor (wortmannin: 2500 nM), MEK1/2 inhibitor (PD98059: 50 μM), PKC inhibitor (GF109203X: 5 μM), tyrosine kinase inhibitor (genistein: 100 μM), PKCδ inhibitor (rottlerin: 10 μM), or p38MAPK inhibitor (SB203580: 25 μM) for 1 h and then with LPS 10 ng/ml for 8 h. Results are expressed as the percentage inhibition as described in the legend of Fig. 6,A. Values are means ± SEM (n = 3). ∗, Significantly different from sample incubated with PBS instead of kinase inhibitors (p < 0.05). B, Samples were incubated in 24-well tissue cultures plates at 37°C with 5% CO2 with PBS or PI3K inhibitor (wortmannin: 2500 nM) for 1 h and then for 8 h with TLR agonists delaying apoptosis, namely LPS, 10 ng/ml (TLR4); PGN, 1 μg/ml (TLR2); R-848, 10 μg/ml (TLR7/8); CpG-DNA, 100 μg/ml (TLR9); Pam3CSK4, 500 ng/ml (TLR1/2); or MALP-2, 10 ng/ml (TLR2/6). Apoptosis was quantified and the results are expressed as the percentage inhibition of apoptosis as described in the legend of Fig. 6 A. Values are means ± SEM (n = 3). ∗, Significantly different from sample incubated with PBS instead of wortmannin (p < 0.05).

FIGURE 7.

Effect of kinase inhibitors on TLR agonist-induced PMN survival. A, Whole blood samples were first incubated in 24-well tissue cultures plates at 37°C with 5% CO2 with PBS, PI3K inhibitor (wortmannin: 2500 nM), MEK1/2 inhibitor (PD98059: 50 μM), PKC inhibitor (GF109203X: 5 μM), tyrosine kinase inhibitor (genistein: 100 μM), PKCδ inhibitor (rottlerin: 10 μM), or p38MAPK inhibitor (SB203580: 25 μM) for 1 h and then with LPS 10 ng/ml for 8 h. Results are expressed as the percentage inhibition as described in the legend of Fig. 6,A. Values are means ± SEM (n = 3). ∗, Significantly different from sample incubated with PBS instead of kinase inhibitors (p < 0.05). B, Samples were incubated in 24-well tissue cultures plates at 37°C with 5% CO2 with PBS or PI3K inhibitor (wortmannin: 2500 nM) for 1 h and then for 8 h with TLR agonists delaying apoptosis, namely LPS, 10 ng/ml (TLR4); PGN, 1 μg/ml (TLR2); R-848, 10 μg/ml (TLR7/8); CpG-DNA, 100 μg/ml (TLR9); Pam3CSK4, 500 ng/ml (TLR1/2); or MALP-2, 10 ng/ml (TLR2/6). Apoptosis was quantified and the results are expressed as the percentage inhibition of apoptosis as described in the legend of Fig. 6 A. Values are means ± SEM (n = 3). ∗, Significantly different from sample incubated with PBS instead of wortmannin (p < 0.05).

Close modal

One mechanism involved in PI3K prevention of apoptosis is activation of the PKB/Akt pathway (29). We therefore studied the phospho-Akt content of intact PMN treated in whole blood, by means of flow cytometry with a mouse anti-human-phospho-Akt mAb. As shown in Fig. 8,A, incubation of whole blood with LPS significantly increased Akt phosphorylation after as little as 2 min, as compared with PBS. Similar results were observed with PGN, R-848, CpG-DNA, Pam3CSK4, and MALP-2 (Fig. 8 B). In contrast, pretreatment with flagellin and loxoribine did not modify Akt phosphorylation. Total Akt content, measured with a mouse anti-human Akt mAb in the same conditions, was not modified by treatment with TLR agonists (not shown).

FIGURE 8.

Effect of TLR agonists on intracellular Akt and Hsp27 phosphorylation. Whole blood samples were preincubated at 37°C in a water bath with gentle horizontal agitation for 15 min with PBS or wortmannin and then treated for 1–10 min with PBS or the following TLR agonists: LPS, 10 ng/ml (TLR4); PGN, 1 μg/ml (TLR2); R-848, 10 μg/ml (TLR7/8); CpG-DNA, 100 μg/ml (TLR9); Pam3CSK4, 500 ng/ml (TLR1/2); MALP-2, 10 ng/ml (TLR2/6); flagellin, 100 ng/ml (TLR5); or loxoribine, 100 μM (TLR7). Phospho-Akt (A and B) and phospho-Hsp27 (C and D) content was then measured by flow cytometry on methanol-permeabilized cells as described in Materials and Methods. Results are MFIs. Values obtained with an irrelevant Ab of the same isotype or with nonimmune serum were subtracted. A and C, The time course of the LPS action on intracellular expression of phospho-Akt and phospho-Hsp27. Values are means ± SEM (n = 4). ∗, Significantly different from samples incubated with PBS (p < 0.05). B and D, The differential effects of TLR agonists on intracellular expression of phospho-Akt and phospho-Hsp27 at optimal incubation times (2 min for phospho-Akt and 5 min for phospho-Hsp27) and the inhibitory effect of wortmannin. Values are means ± SEM (n = 4). ∗, Significantly different from samples incubated with PBS, flagellin, and loxoribine (p < 0.05). †, Not significantly different from samples incubated with PBS, flagellin, and loxoribine (p < 0.05).

FIGURE 8.

Effect of TLR agonists on intracellular Akt and Hsp27 phosphorylation. Whole blood samples were preincubated at 37°C in a water bath with gentle horizontal agitation for 15 min with PBS or wortmannin and then treated for 1–10 min with PBS or the following TLR agonists: LPS, 10 ng/ml (TLR4); PGN, 1 μg/ml (TLR2); R-848, 10 μg/ml (TLR7/8); CpG-DNA, 100 μg/ml (TLR9); Pam3CSK4, 500 ng/ml (TLR1/2); MALP-2, 10 ng/ml (TLR2/6); flagellin, 100 ng/ml (TLR5); or loxoribine, 100 μM (TLR7). Phospho-Akt (A and B) and phospho-Hsp27 (C and D) content was then measured by flow cytometry on methanol-permeabilized cells as described in Materials and Methods. Results are MFIs. Values obtained with an irrelevant Ab of the same isotype or with nonimmune serum were subtracted. A and C, The time course of the LPS action on intracellular expression of phospho-Akt and phospho-Hsp27. Values are means ± SEM (n = 4). ∗, Significantly different from samples incubated with PBS (p < 0.05). B and D, The differential effects of TLR agonists on intracellular expression of phospho-Akt and phospho-Hsp27 at optimal incubation times (2 min for phospho-Akt and 5 min for phospho-Hsp27) and the inhibitory effect of wortmannin. Values are means ± SEM (n = 4). ∗, Significantly different from samples incubated with PBS, flagellin, and loxoribine (p < 0.05). †, Not significantly different from samples incubated with PBS, flagellin, and loxoribine (p < 0.05).

Close modal

Hsp27 has been reported to associate with Akt (30). Furthermore, phosphorylation of Hsp27 by Akt results in its dissociation from Akt and may participate to the Hsp27-induced delay in PMN apoptosis (31). We therefore studied the effects of TLR agonists on Hsp27 phosphorylation by means of flow cytometry. Phospho-Hsp27 content was significantly increased after 5 min incubation with LPS (Fig. 8,C) and with the other TLR agonists which increased Akt phosphorylation (Fig. 8,D). No modification of total Hsp27 content was observed. The TLR agonist-induced increase in phospho-Hsp27 and phospho-Akt content was completely reversed by pretreatment of whole blood with wortmannin (Fig. 8, B and D).

It is now generally accepted that blood PMN do not express the classical antiapoptotic protein Bcl-2 (32). In contrast, there is a very close correlation between levels of the antiapoptotic protein Mcl-1 and PMN survival (32). As previously reported with isolated PMN (33), we found that Mcl-1 levels fell during constitutive PMN apoptosis in whole blood (Fig. 9,A). Treatment of whole blood with LPS significantly increased Mcl-1 levels after 1 h as compared with PBS controls; values peaked at 2 h (Fig. 9,A). Similar results were observed with PGN, R-848, CpG-DNA, Pam3CSK4, and MALP-2 (Fig. 9 B). The TLR-induced increase in Mcl-1 was significantly reversed by 30 min preincubation with wortmannin (not shown).

FIGURE 9.

Effect of TLR agonists on intracellular Mcl-1, A1, and phospho-Bad expression. Whole blood samples were preincubated at 37°C in a water bath with gentle horizontal agitation for 5–240 min with PBS or the following TLR agonists: LPS, 10 ng/ml (TLR4); PGN, 1 μg/ml (TLR2); R-848, 10 μg/ml (TLR7/8); CpG-DNA, 100 μg/ml (TLR9); Pam3CSK4, 500 ng/ml (TLR1/2); MALP-2, 10 ng/ml (TLR2/6); flagellin, 100 ng/ml (TLR5); or loxoribine, 100 μM (TLR7). Mcl-1 (A and B) and A1 (C and D) and phospho-Bad (E and F) content was then measured by flow cytometry on methanol-permeabilized cells as described in Materials and Methods. Results are MFIs. Values obtained with an irrelevant Ab of the same isotype or with nonimmune serum were subtracted. A, C, and E, The time course of the LPS action on intracellular expression of Mcl-1, A1, and phospho-Bad. Values are means ± SEM (n = 4). ∗, Significantly different from samples incubated with PBS (p < 0.05). B, D, and F, The differential effects of TLR agonists on intracellular expression of Mcl-1, A1, and phospho-Bad at optimal incubation times (120 min for Mcl-1 and A1 and 60 min for phospho-Bad). Values are means ± SEM (n = 4). ∗, Significantly different from samples incubated with PBS, flagellin, or loxoribine (p < 0.05).

FIGURE 9.

Effect of TLR agonists on intracellular Mcl-1, A1, and phospho-Bad expression. Whole blood samples were preincubated at 37°C in a water bath with gentle horizontal agitation for 5–240 min with PBS or the following TLR agonists: LPS, 10 ng/ml (TLR4); PGN, 1 μg/ml (TLR2); R-848, 10 μg/ml (TLR7/8); CpG-DNA, 100 μg/ml (TLR9); Pam3CSK4, 500 ng/ml (TLR1/2); MALP-2, 10 ng/ml (TLR2/6); flagellin, 100 ng/ml (TLR5); or loxoribine, 100 μM (TLR7). Mcl-1 (A and B) and A1 (C and D) and phospho-Bad (E and F) content was then measured by flow cytometry on methanol-permeabilized cells as described in Materials and Methods. Results are MFIs. Values obtained with an irrelevant Ab of the same isotype or with nonimmune serum were subtracted. A, C, and E, The time course of the LPS action on intracellular expression of Mcl-1, A1, and phospho-Bad. Values are means ± SEM (n = 4). ∗, Significantly different from samples incubated with PBS (p < 0.05). B, D, and F, The differential effects of TLR agonists on intracellular expression of Mcl-1, A1, and phospho-Bad at optimal incubation times (120 min for Mcl-1 and A1 and 60 min for phospho-Bad). Values are means ± SEM (n = 4). ∗, Significantly different from samples incubated with PBS, flagellin, or loxoribine (p < 0.05).

Close modal

The only other antiapoptotic factor so far implicated in PMN survival is A1/Bfl-1. In contrast to Mcl-1, A1 levels did not fall during spontaneous PMN apoptosis. However, the A1 level increased significantly after incubation with TLR agonists that delayed apoptosis. This effect appeared as early as 1 h and was maximal after 2 h; flagellin and loxoribine had no effect (Fig. 9, C and D). As synthesis of the antiapoptotic protein A1 depends on NF-κB activation (34), we analyzed the effect of SN50 on TLR-induced A1 levels. We found that the TLR agonist-induced increase in A1 was reversed by sample preincubation with the NF-κB inhibitor SN50. In fact, MFI of the sample pretreated with PBS and then incubated with LPS was 209.0 ± 12.7, while MFI of the sample pretreated with SN50 and then incubated with LPS was 149.2 ± 5 and did not differ from that of the sample incubated with PBS alone. Similar results were observed with the other TLR agonists (not shown). Preincubation with wortmannin only partially reversed the effect of TLR agonists on A1 levels (not shown).

Most reports show that levels of proapoptotic proteins in PMN remain fairly constant when the cells age naturally and when their apoptosis is experimentally delayed or accelerated (32). Bad phosphorylation by Akt/PKB can induce an antiapoptotic effect by Bad dissociation from antiapoptotic proteins of the Bcl-2 family, also resulting in delayed apoptosis (35). We therefore examined whether TLR agonist-induced phosphorylation of Akt was associated with Bad phosphorylation. As shown in Fig. 9,E, levels of phospho-Bad increased significantly during incubation (30 and 60 min) with LPS, while total Bad content was unaffected (not shown). Similar results were observed with other TLR agonists that delayed apoptosis (Fig. 9 F). This increase was completely reversed by preincubation with wortmannin (not shown).

Our results show that all TLR agonists except for flagellin (TLR5) and loxoribine (TLR7) delay spontaneous apoptosis of human PMN and extend their functional life span in whole blood. The antiapoptotic action of TLR agonists required activation of NF-κB and PI3K. Furthermore, flow cytometry of intact cells showed that the TLR agonists which delayed PMN apoptosis induced phosphorylation of Akt, a major target of PI3K, as well as Hsp27 phosphorylation. Finally, the TLR-induced delay in PMN apoptosis was associated with increased levels of Mcl-1 and A1, both of which are antiapoptotic members of the Bcl-2 family. These effects were reversed by PI3K and NF-κB inhibitors, respectively. In addition, TLR activation led to PI3K-dependent phosphorylation of the proapoptotic protein Bad, which is known to result in delayed PMN apoptosis.

One particularity of this study is that we analyzed PMN apoptosis by flow cytometry in whole blood. This avoided PMN isolation procedures, which have been shown to induce surface expression of molecules that are not detected in whole blood and may thereby alter PMN responses (36). Such artifacts might contribute to the acceleration of spontaneous apoptosis that we observed after PMN purification. In addition, analysis of TLR agonist-induced modulation of PMN survival in whole blood mimics physiological conditions more closely than the use of isolated cells. In particular, interactions between cellular elements have been reported to be important in maintaining PMN viability (37). However, our data do not rule out a contribution of PBMC to the kinetics of spontaneous PMN apoptosis and to the TLR agonist-induced delay in PMN apoptosis. Release of survival factors such as cytokines by activated monocytes (22, 38) could contribute to the stronger agonist-induced inhibition of PMN apoptosis that we observed in whole blood as compared with highly purified PMN.

In keeping with previous data (12, 14, 15, 39), we found that LPS delayed PMN apoptosis in whole blood. We also extended this observation to PGN, R-848, CpG-DNA, Pam3CSK, and MALP-2. MALP-2 (TLR2/6) and Pam3CSK4 (TLR1/2) had milder effects on PMN survival. TLR1 and TLR6 act as coreceptors for TLR2 and have been reported to inhibit cellular responsiveness to activating ligands (40, 41). The lack of effect of flagellin and loxoribine could be related to a lack of triggering of their signaling pathways, itself possibly due to weak expression of TLR5 and TLR7, in keeping with previous reports that the corresponding mRNAs are weakly expressed in PMN (41, 42).

We observed, for the first time, that treatment of PMN in whole blood with TLR agonists which strongly delayed PMN apoptosis at 8 h maintained and even enhanced PMN functions, both in the basal state and in response to a bacterial product (fMLP), as compared with values obtained at baseline (T0h) and with untreated samples at 8 h. TLRs play a central role in innate immunity by mediating PAMP recognition, as reflected by the increased susceptibility to infections of children with deficient TLR transduction (43). The prolonged functional life span induced by LPS, PGN, R-848, CpG-DNA and, to a lesser degree, Pam3CSK and MALP-2 may represent a crucial enhancement of PMN defenses against microbial pathogens. Furthermore, our data, obtained with whole blood, suggest that in pathological situations such as sepsis the delayed PMN apoptosis induced by TLR agonists at the systemic level could potentiate inflammatory reactions and lead to vessel damage.

Many groups have investigated the signaling pathways involved in PMN survival, but the specific roles of individual pathways involved in the response to individual TLR ligands remain to be clarified. We first used NF-κB inhibitors commonly used to inhibit NF-κB nuclear translocation (44, 45). The NF-κB control peptide SN50M had only minor effects on TLR-induced PMN survival, whereas the active SN50 NF-κB inhibitor significantly prevented TLR agonist-induced survival. Similar effects were observed with a second NF-κB inhibitor, the sesquiterpene lactone parthenolide. In addition, using an anti-phosphorylated-IKK Ab, we found that TLR-agonists protecting against apoptosis induced IKK phosphorylation, which permits IκB phosphorylation, leading to its degradation and NF-κB nuclear translocation (46). These data are in keeping with those previously reported on LPS-treated isolated PMN (15, 23). In this study, we extend this observation to all TLR agonists capable of delaying PMN apoptosis, pointing to a major role of NF-κB in TLR-mediated PMN survival.

Our results also demonstrate that TLR agonists delay PMN apoptosis via a PI3K-dependent pathway, although our data do not rule out the possibility that PI3K inhibitors could also act by preventing, at least in part, the generation of survival signals by other cells in whole blood. In contrast, tyrosine kinases, MAPK and PKC do not appear to be involved. The lipid products of PI3K–predominantly phosphatidylinositol 3,4,5-triphosphate–induce translocation of Akt/PKB to the plasma membrane, where it is phosphorylated and activated by phosphatidyl-inositol 3,4,5-phosphate-dependent protein kinase (PDK1), and this pathway has been forwarded as a major mediator downstream of PI3K (29, 47). Akt/PKB activity has been shown to prevent apoptosis induced by cytokines and growth factors, cellular stress, chemotherapeutic agents, and irradiation (48). Our data showing that TLR agonists capable of delaying PMN apoptosis increase PI3K-dependent Akt phosphorylation strongly suggest a central role of Akt in this effect. Furthermore, we show for the first time that the same TLR agonists induce a PI3K-dependent increase in Hsp27 phosphorylation. It has recently been shown that Hsp27 phosphorylation by Akt leads to disruption of the Akt-Hsp27 interaction, and it has been suggested that released Hsp27 may promote independent survival signals (31). It has also been postulated that Hsp27 inhibits apoptosis through inactivation of caspase-3, caspase-9, and inhibition of cytochrome c release (49, 50).

Akt activation could also explain, at least in part, the role of NF-κB in TLR-induced PMN survival. Indeed, it was recently reported that Akt modulates NF-κB-dependent transcription in TLR2-stimulated PMN by modifying phosphorylation of the p65 subunit (7). Nevertheless, Akt is necessary but not sufficient for NF-κB activation by TLR (51).

PI3K/Akt activation could be an alternative pathway to the IL-1R-associated kinase/IKK/NF-κB pathway, leading to independent modulation of Bcl-2 family proteins (52). Increased levels of Mcl-1 have been implicated in PMN survival induced by proinflammatory cytokines such as GM-CSF, IL-1, TNF-α, and IL-15 (12, 33, 53, 54). We found that PMN treatment for 1 h with TLR agonists capable of delaying neutrophil apoptosis prevented the loss of Mcl-1. In keeping with previous data demonstrating that increased Mcl-1 translation depends on the PI3K/Akt pathway (55), we found that the TLR-induced Mcl-1 elevation was reversed by pretreatment with wortmannin. The only other antiapoptotic gene product so far implicated in PMN survival is A1. Previous studies have been restricted to mRNA, and show that A1 transcripts are cytokine-regulated in human PMN (34). Using a novel method–flow cytometry–to investigate A1 protein expression by intact permeabilized PMN, we showed for the first time that PMN activation by TLR agonists is associated with an increased intracellular content of A1 protein. In keeping with previous data (56), this effect was regulated by NF-κB and partially inhibited by wortmannin. Finally, kinetic analysis showed an early increase in Mcl-1 and A1 levels and a return to control value by 240 min. These results strongly suggest that this early increase in Mcl-1 and A1 levels inhibits the cellular apoptosis machinery and gives an advantage to TLR agonist-stimulated cells.

Rapid regulation of PMN survival could be achieved by posttranslational protein modifications. In particular, increased Bad (Bcl-xL/Bcl-2-associated death promoter homologue) phosphorylation has been implicated in PMN survival induced by GM-CSF (13, 24). Phosphorylated Bad interacts with 14-3-3 protein, and the resulting Bad sequestration diminishes Bad binding to diverse antiapoptotic Bcl-2 proteins anchored to the mitochondrial membrane (57). Increased amounts of antiapoptotic proteins are then free to bind to Bax and to prevent its proapoptotic activity, leading to cell survival. Recently, it was reported that underphosphorylated Bad interacts with all antiapoptotic Bcl-2 family members, and particularly A1 and Mcl-1 (58). Our results demonstrating that TLR agonists which inhibit PMN apoptosis increase Bad phosphorylation from 30 to 60 min strongly suggest that this phenomenon is involved in PMN survival induced by TLR activation. In keeping with previous data showing that Bad phosphorylation on serine 136 is induced by phospho-Akt (59), we observed that TLR agonist-induced Bad phosphorylation was PI3K-dependent.

Taken together, our findings demonstrate that relatively specific agonists of TLR, namely LPS (TLR4), PGN (TLR2), R-848 (TLR7/8), CpG-DNA (TLR9), Pam3CSK4 (TLR1/2), MALP-2 (TLR2/6), with the exception of flagellin (TLR5) and loxoribine (TLR7), are able to delay PMN apoptosis and extend the PMN functional life span in whole blood. Our results also point to the involvement of the PI3K/Akt and NF-κB pathways in PMN survival induced by TLR activation. PI3K-dependent phosphorylation of Akt may be strongly involved in the increased levels of the antiapoptotic protein Mcl-1 and the increased phosphorylation of the proapoptotic protein Bad. The antiapoptotic action of TLR agonists may be facilitated by Akt-dependent phosphorylation of Hsp27. In addition, NF-κB activation may lead to increased levels of the antiapoptotic protein A1. To our knowledge, this is the first report that Bcl-2 family proteins are modulated upon TLR activation. Ongoing studies may identify new therapeutic targets for regulating TLR-induced PMN survival and functions in inflammatory disorders in which these cells contribute to bystander tissue damage.

The authors have no financial conflict of interest.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

2

Abbreviations used in this paper: PMN, polymorphonuclear neutrophil; ROS, reactive oxygen species; PAMP, pathogen-associated molecular pattern; PGN, peptidoglycan; MALP-2, macrophage-activating lipopeptide-2; HE, hydroethidine; DPI, diphenyleneiodonium; DiOC6, 3,3′-dihexyloxacarbocyanine; 7-AAD, 7-aminoactinomycin D; Hsp, heat shock protein; FSC, forward scatter; SSC, side scatter; MFI, mean fluorescence intensity; PKB, protein kinase B; PKC, protein kinase C; IKK, IκB kinase; Δψm, mitochondrial transmembrane potential.

1
Babior, B. M..
1984
. Oxidants from phagocytes: agents of defense and destruction.
Blood
64
:
959
.
2
Greenberg, S., S. Grinstein.
2002
. Phagocytosis and innate immunity.
Curr. Opin. Immunol.
14
:
136
.
3
Medzhitov, R., P. Preston-Hurlburt, C. A. Janeway, Jr.
1997
. A human homologue of the Drosophila Toll protein signals activation of adaptive immunity.
Nature
388
:
394
.
4
Hayashi, F., T. K. Means, A. D. Luster.
2003
. Toll-like receptors stimulate human neutrophil function.
Blood
102
:
2660
.
5
Beutler, B..
2000
. TLR4: central component of the sole mammalian LPS sensor.
Curr. Opin. Immunol.
12
:
20
.
6
Yum, H. K., J. Arcaroli, J. Kupfner, R. Shenkar, J. M. Penninger, T. Sasaki, K. Y. Yang, J. S. Park, E. Abraham.
2001
. Involvement of phosphoinositide 3-kinases in neutrophil activation and the development of acute lung injury.
J. Immunol.
167
:
6601
.
7
Strassheim, D., K. Asehnoune, J. S. Park, J. Y. Kim, Q. He, D. Richter, K. Kuhn, S. Mitra, E. Abraham.
2004
. Phosphoinositide 3-kinase and Akt occupy central roles in inflammatory responses of Toll-like receptor 2-stimulated neutrophils.
J. Immunol.
172
:
5727
.
8
Haslett, C., J. S. Savill, M. K. Whyte, M. Stern, I. Dransfield, L. C. Meagher.
1994
. Granulocyte apoptosis and the control of inflammation.
Philos. Trans. R. Soc. London Biol. Sci.
345
:
327
.
9
Savill, J., I. Dransfield, C. Gregory, C. Haslett.
2002
. A blast from the past: clearance of apoptotic cells regulates immune responses.
Nat. Rev. Immunol.
2
:
965
.
10
Colotta, F., F. Re, N. Polentarutti, S. Sozzani, A. Mantovani.
1992
. Modulation of granulocyte survival and programmed cell death by cytokines and bacterial products.
Blood
80
:
2012
.
11
Murray, J., J. A. Barbara, S. A. Dunkley, A. F. Lopez, X. Van Ostade, A. M. Condliffe, I. Dransfield, C. Haslett, E. R. Chilvers.
1997
. Regulation of neutrophil apoptosis by tumor necrosis factor-α: requirement for TNFR55 and TNFR75 for induction of apoptosis in vitro.
Blood
90
:
2772
.
12
Moulding, D. A., J. A. Quayle, C. A. Hart, S. W. Edwards.
1998
. Mcl-1 expression in human neutrophils: regulation by cytokines and correlation with cell survival.
Blood
92
:
2495
.
13
Cowburn, A. S., K. A. Cadwallader, B. J. Reed, N. Farahi, E. R. Chilvers.
2002
. Role of PI3-kinase-dependent Bad phosphorylation and altered transcription in cytokine-mediated neutrophil survival.
Blood
100
:
2607
.
14
Klein, J. B., A. Buridi, P. Y. Coxon, M. J. Rane, T. Manning, R. Kettritz, K. R. McLeish.
2001
. Role of extracellular signal-regulated kinase and phosphatidylinositol-3 kinase in chemoattractant and LPS delay of constitutive neutrophil apoptosis.
Cell. Signal.
13
:
335
.
15
Sabroe, I., L. R Prince, E. C. Jones, M. J. Horsburgh, S. J. Foster, S. N. Vogel, S. K. Dower, M. K. Whyte.
2003
. Selective roles for Toll-like receptor (TLR)2 and TLR4 in the regulation of neutrophil activation and life span.
J. Immunol.
170
:
5268
.
16
Stringer, R. E., C. A. Hart, S. W. Edwards.
1996
. Sodium butyrate delays neutrophil apoptosis: role of protein biosynthesis in neutrophil survival.
Br. J. Haematol.
92
:
169
.
17
Grenier, A., M. Dehoux, A. Boutten, M. Arce-Vicioso, G. Durand, M. A. Gougerot-Pocidalo, S. Chollet-Martin.
1999
. Oncostatin M production and regulation by human polymorphonuclear neutrophils.
Blood
93
:
1413
.
18
Herault, O., P. Colombat, J. Domenech, M. Degenne, J. L. Bremond, L. Sensebe, M. C. Bernard, C. Binet.
1999
. A rapid single-laser flow cytometric method for discrimination of early apoptotic cells in a heterogenous cell population.
Br. J. Haematol.
104
:
530
.
19
Shapiro, H. M..
2000
. Membrane potential estimation by flow cytometry.
Methods
21
:
271
.
20
Elbim, C., H. Reglier, M. Fay, C. Delarche, V. Andrieu, J. El Benna, M. A. Gougerot-Pocidalo.
2001
. Intracellular pool of IL-10 receptors in specific granules of human neutrophils: differential mobilization by proinflammatory mediators.
J. Immunol.
166
:
5201
.
21
Rothe, G., G. Valet.
1990
. Flow cytometric analysis of respiratory burst activity in phagocytes with hydroethidine and 2′,7′-dichlorofluorescin.
J. Leukocyte Biol.
47
:
440
.
22
Sabroe, I., E. C. Jones, L. R. Usher, M. K. Whyte, S. K. Dower.
2002
. Toll-like receptor (TLR)2 and TLR4 in human peripheral blood granulocytes: a critical role for monocytes in leukocyte lipopolysaccharide responses.
J. Immunol.
168
:
4701
.
23
Ward, C., E. R. Chilvers, M. F. Lawson, J. G. Pryde, S. Fujihara, S. N. Farrow, C. Haslett, A. G. Rossi.
1999
. NF-κB activation is a critical regulator of human granulocyte apoptosis in vitro.
J. Biol. Chem.
274
:
4309
.
24
Klein, J. B., M. J. Rane, J. A. Scherzer, P. Y. Coxon, R. Kettritz, J. M. Mathiesen, A. Buridi, K. R. McLeish.
2000
. Granulocyte-macrophage colony-stimulating factor delays neutrophil constitutive apoptosis through phosphoinositide 3-kinase and extracellular signal-regulated kinase pathways.
J. Immunol.
164
:
4286
.
25
Alvarado-Kristensson, M., F. Melander, K. Leandersson, L. Ronnstrand, C. Wernstedt, T. Andersson.
2004
. P38-MAPK signals survival by phosphorylation of caspase-8 and caspase-3 in human neutrophils.
J. Exp. Med.
199
:
449
.
26
Alvarado-Kristensson, M., M. I. Porn-Ares, S. Grethe, D. Smith, L. Zheng, T. Andersson.
2002
. P38 Mitogen-activated protein kinase and phosphatidylinositol 3-kinase activities have opposite effects on human neutrophil apoptosis.
FASEB J.
16
:
129
.
27
Wang, K., D. Scheel-Toellner, S. H. Wong, R. Craddock, J. Caamano, A. N. Akbar, M. Salmon, J. M. Lord.
2003
. Inhibition of neutrophil apoptosis by type 1 IFN depends on cross-talk between phosphoinositol 3-kinase, protein kinase C-δ, and NF-κB signaling pathways.
J. Immunol.
171
:
1035
.
28
Sweeney, J. F., P. K. Nguyen, G. M. Omann, D. B. Hinshaw.
1998
. Lipopolysaccharide protects polymorphonuclear leukocytes from apoptosis via tyrosine phosphorylation-dependent signal transduction pathways.
J. Surg. Res.
74
:
64
.
29
Kennedy, S. G., A. J. Wagner, S. D. Conzen, J. Jordan, A. Bellacosa, P. N. Tsichlis, N. Hay.
1997
. The PI3-kinase/Akt signaling pathway delivers an anti-apoptotic signal.
Genes Dev.
11
:
701
.
30
Konishi, H., H. Matsuzaki, M. Tanaka, Y. Takemura, S. Kuroda, Y. Ono, U. Kikkawa.
1997
. Activation of protein kinase B (Akt/RAC-protein kinase) by cellular stress and its association with heat shock protein Hsp27.
FEBS Lett.
410
:
493
.
31
Rane, M. J., Y. Pan, S. Singh, D. W. Powell, R. Wu, T. Cummins, Q. Chen, K. R. McLeish, J. B. Klein.
2003
. Heat shock protein 27 controls apoptosis by regulating Akt activation.
J. Biol. Chem.
278
:
27828
.
32
Akgul, C., D. A. Moulding, S. W. Edwards.
2001
. Molecular control of neutrophil apoptosis.
FEBS Lett.
487
:
318
.
33
Pelletier, M., C. Ratthe, D. Girard.
2002
. Mechanisms involved in interleukin-15-induced suppression of human neutrophil apoptosis: role of the anti-apoptotic Mcl-1 protein and several kinases including Janus kinase-2, p38 mitogen-activated protein kinase and extracellular signal-regulated kinases-1/2.
FEBS Lett.
532
:
164
.
34
Chuang, P. I., E. Yee, A. Karsan, R. K. Winn, J. M. Harlan.
1998
. A1 is a constitutive and inducible Bcl-2 homologue in mature human neutrophils.
Biochem. Biophys. Res. Commun.
249
:
361
.
35
Perianayagam, M. C., V. S. Balakrishnan, B. J. Pereira, B. L. Jaber.
2004
. C5a delays apoptosis of human neutrophils via an extracellular signal-regulated kinase and Bad-mediated signalling pathway.
Eur. J. Clin. Invest.
34
:
50
.
36
Macey, M. G., D. A. McCarthy, S. Vordermeier, A. C. Newland, K. A. Brown.
1995
. Effects of cell purification methods on CD11b and L-selectin expression as well as the adherence and activation of leucocytes.
J. Immunol. Methods.
181
:
211
.
37
Hodge, G., S. Hodge, P. Han.
2000
. Increased levels of apoptosis of leukocyte subsets in cultured PBMCs compared to whole blood as shown by annexin V binding: relevance to cytokine production.
Cytokine
12
:
1763
.
38
Prince, L. R., L. Allen, E. C. Jones, P. G. Hellewell, S. K. Dower, M. K. B. Whyte, I. Sabroe.
2004
. The role of interleukin-1β in direct and Toll-like receptor 4-mediated neutrophil activation and survival.
Am. J. Pathol.
165
:
1819
.
39
Preshaw, P. M., R. E. Schifferle, J. D. Walters.
1999
. Porphyromonas gingivalis lipopolysaccharide delays human polymorphonuclear leukocyte apoptosis in vitro.
J. Periodontal Res.
34
:
197
.
40
Hajjar, A. M., D. S. O’Mahony, A. Ozinsky, D. M. Underhill, A. Aderem, S. J. Klebanoff, C. B. Wilson.
2001
. Cutting edge: functional interactions between Toll-like receptor (TLR) 2 and TLR1 or TLR6 in response to phenol-soluble modulin.
J. Immunol.
166
:
15
.
41
Zarember, K. A., P. J. Godowski.
2002
. Tissue expression of human Toll-like receptors and differential regulation of Toll-like receptor mRNAs in leukocytes in response to microbes, their products, and cytokines.
J. Immunol.
168
:
554
.
42
Nagase, H., S. Okugawa, Y. Ota, M. Yamaguchi, H. Tomizawa, K. Matsushima, K. Ohta, K. Yamamoto, K. Hirai.
2003
. Expression and function of Toll-like receptors in eosinophils: activation by Toll-like receptor 7 ligand.
J. Immunol.
171
:
3977
.
43
Picard, C., A. Puel, M. Bonnet, C. L. Ku, J. Bustamante, K. Yang, C. Soudais, S. Dupuis, J. Feinberg, C. Fieschi, et al
2003
. Pyogenic bacterial infections in humans with IRAK-4 deficiency.
Science
299
:
2076
.
44
Lin, Y. Z., S. Y. Yao, R. A. Veach, T. R. Torgerson, J. Hawinger.
1995
. Inhibition of nuclear translocation of transcription factor NF-κB by a synthetic peptide containing a cell membrane-permeable motif and nuclear localization sequence.
J. Biol. Chem.
270
:
14255
.
45
Hehner, S. P., M. Heinrich, P. M. Bork, M. Vogt, F. Ratter, V. Lehman, K. Schulze-Osthoff, W. Dröge, M. L. Schmitz.
1998
. Sesquiterpene lactones specifically inhibit activation of NF-κB by preventing the degradation of IκB-α and IκB-β.
J. Biol. Chem.
273
:
1288
.
46
Yamamoto, Y., R. B. Gaynor.
2004
. IκB kinases: key regulators of the NF-κB pathway.
TRENDS Biochem. Sci.
29
:
72
.
47
Toker, A., L. C. Cantley.
1997
. Signalling through the lipid products of phosphoinositide-3-OH kinase.
Nature
387
:
673
.
48
Downward, J..
1998
. Mechanisms and consequences of activation of protein kinase B/Akt.
Curr. Opin. Cell. Biol.
10
:
262
.
49
Garrido, C., J. M. Bruey, A. Fromentin, A. Hammann, A. P. Arrigo, E. Solary.
1999
. HSP27 inhibits cytochrome c-dependent activation of procaspase-9.
FASEB J.
13
:
2061
.
50
Paul, C., F. Manero, S. Gonin, C. Kretz-Remy, S. Virot, A. P. Arrigo.
2002
. Hsp27 as a negative regulator of cytochrome C release.
Mol. Cell. Biol.
22
:
816
.
51
Li, X., J. C. Tupper, D. D. Bannerman, R. K. Winn, C. J. Rhodes, J. M. Harlan.
2003
. Phosphoinositide 3 kinase mediates Toll-like receptor 4-induced activation of NF-κB in endothelial cells.
Infect. Immun.
71
:
4414
.
52
Takeshita, F., I. Gursel, K. J. Ishii, K. Suzuki, M. Gursel, D. M. Klinman.
2004
. Signal transduction pathways mediated by the interaction of CpG DNA with Toll-like receptor 9.
Semin. Immunol.
16
:
17
.
53
Epling-Burnette, P. K., B. Zhong, F. Bai, K. Jiang, R. D. Bailey, R. Garcia, R. Jove, J. Y. Djeu, T. P. Loughran, Jr, S. Wei.
2001
. Cooperative regulation of Mcl-1 by Janus kinase/Stat and phosphatidylinositol 3-kinase contribute to granulocyte-macrophage colony-stimulating factor-delayed apoptosis in human neutrophils.
J. Immunol.
166
:
7486
.
54
Derouet, M., L. Thomas, A. Cross, R. J. Moots, S. W. Edwards.
2004
. Granulocyte macrophage colony-stimulating factor signaling and proteasome inhibition delay neutrophil apoptosis by increasing the stability of Mcl-1.
J. Biol. Chem.
279
:
26915
.
55
Schubert, K. M., V. Duronio.
2001
. Distinct roles for extracellular-signal-regulated protein kinase (ERK) mitogen-activated protein kinases and phosphatidylinositol 3-kinase in the regulation of Mcl-1 synthesis.
Biochem. J.
356
:
473
.
56
Lee, H. H., H. Dadgostar, Q. Cheng, J. Shu, G. Cheng.
1999
. NF-κB-mediated up-regulation of Bcl-x and Bfl-1/A1 is required for CD40 survival signaling in B lymphocytes.
Proc. Natl. Acad. Sci. USA
96
:
9136
.
57
Hsu, S. Y., A. Kaipia, L. Zhu, A. J. Hsueh.
1997
. Interference of BAD (Bcl-xL/Bcl-2-associated death promoter)-induced apoptosis in mammalian cells by 14-3-3 isoforms and P11.
Mol. Endocrinol.
11
:
1858
.
58
Bae, J., S. Y. Hsu, C. P. Leo, K. Zell, A. J. Hsueh.
2001
. Underphosphorylated BAD interacts with diverse antiapoptotic Bcl-2 family proteins to regulate apoptosis.
Apoptosis
6
:
319
.
59
del Peso, L., M. Gonzalez-Garcia, C. Page, R. Herrera, G. Nunez.
1997
. Interleukin-3-induced phosphorylation of BAD through the protein kinase Akt.
Science
278
:
687
.