Bacterial LPS induces the release of ATP from immune cells. Accruing evidence suggests that extracellular ATP participates in the inflammatory response as a proinflammatory mediator by activating the inflammasome complex, inducing secretion of cytokines (IL-1, IL-18) and cell damaging agents such as oxygen radicals, cationic proteins, and metalloproteases. It is not known whether ATP can also act as a proinflammatory mediator by inhibiting production of molecules down-modulating the immune response. Here, we show that extracellular ATP impairs in an IL-10-dependent fashion the expression of the tolerogenic soluble and membrane-bound HLA-G Ag in human monocytes. The effect of ATP was mimicked by BzATP (3′-O-(4-benzoyl)benzoyl-ATP) and greatly reduced by pretreatment with oATP (periodate-oxidized ATP), KN-62 (1-[N,O-bis(5-isoquinoline-sulfonyl)-N-methyl-l-tyrosyl]-4-phenylpiperazine), and an anti-P2X7 mAb, thus pointing to a specific role of the P2X7 receptor. The effect of ATP was time- and dose-dependent and was not due to a decrease in expression of IL-10 receptor. Inhibition by ATP was reverted by supplementation of culture medium with exogenous IL-10. Due to the well-known immunosuppressive activity of IL-10 and soluble HLA-G, this novel effect of ATP might be relevant for the pathophysiology and therapy of inflammatory disorders.

Bacterial LPS, the major constituent of the cell wall of Gram-negative bacteria, stimulates production of a plethora of inflammatory mediators (1, 2). Extracellular ATP is known to trigger or increase the secretion of cytokines whose synthesis is induced by LPS (3), therefore acting as a proinflammatory mediator. Accordingly, bacterial or viral infections, mechanical stress, or cell-damaging events induce ATP release (3, 4). Once in the extracellular milieu, ATP binds to and activates a class of ubiquitous plasma membrane receptors named P2 (5). P2 receptors are subdivided into P2X and P2Y subtypes.

P2X receptors are multimeric ligand-gated ion channels selective for monovalent and divalent cations originally identified in excitatory cells but later also found in many other cells. P2X subunits have intracellular amino- and carboxyl-termini, two transmembrane domains, and a large extracellular loop (6). P2Y receptors are ubiquitous G protein-coupled receptors whose activation triggers generation of inositol 1,4,5-trisphosphate, release of Ca2+ from intracellular stores, and for some members of the subfamily, increase in intracellular cAMP (7).

HLA-G Ags are currently defined as nonclassical HLA class Ib molecules, differing from classical HLA-A, -B, and -C class I molecules for the reduced allelic polymorphism (8), a restricted tissue distribution, and the occurrence of alternative mRNA splicing generating membrane-bound (HLA-G1–G4) or soluble (HLA-G5–G7) HLA-G isoforms (9). The HLA-G Ags were first observed in cytotrophoblast cells (10), suggesting a functional role for these molecules in the induction and maintenance of maternal tolerogenic conditions toward the semiallogenic fetus. HLA-G production was detected in physiological conditions in the thymus and in CD14+ peripheral blood monocytes following activation of cells by IL-10, IFNs, and hormones (11, 12). Further investigations confirmed that HLA-G Ags, mainly in their HLA-G1 membrane-bound and soluble isoforms generated by proteolitic shedding (sHLA-G1) or alternative splicing (HLA-G5), mediate inhibition of NK and CD8+ lymphocyte cytotoxic activity, but also affect CD4+ functions and dendritic cell maturation.

Recently, several investigations have shown abnormal HLA-G expression in autoimmune diseases, cancer, virally infected cells, and transplanted organs, confirming the hypothesis of a tolerogenic role for these HLA class Ib molecules (13, 14, 15, 16, 17, 18, 19). In particular, increased sHLA-G levels in cerebrospinal fluids have been associated with the absence of clinical activity in multiple sclerosis (20), while in solid organ transplantation high sHLA-G serum concentrations were detected in patients with a reduced number of rejection episodes (19). A reduced sHLA-G production was suggested to play a role in the persistence of inflammatory conditions in asthma (21). However, HLA-G expression may also mediate negative clinical outcome in cancer where increased membrane-bound or sHLA-G molecules have been associated with poor prognosis (17). These observations suggest that maintenance of physiological levels of sHLA-G relates to the delicate equilibrium that regulates the inflammatory/antiinflammatory response.

The immunomodulatory effect of ATP strongly depends on the nucleotide concentration: low extracellular ATP levels induce a Th2-skewing dendritic cell phenotype and are immunosuppressive (22, 23), while high levels trigger the release of IL-1 family cytokines, as well as that of IL-6 and TNF-α (24, 25, 26), and are therefore proinflammatory. The strong immunostimulatory effect of elevated ATP concentrations may be mediated by release of proinflammatory cytokines or reactive oxygen species or by inhibition of production of immunosuppressive cytokines (23, 27).

In the present report we investigated the effect of ATP on sHLA-G production induced by LPS-activated human monocytes. These data show that ATP acts as a potent inhibitor of soluble and membrane-bound HLA-G production via down-regulation of IL-10 release, thus further supporting the role of ATP as a proinflammatory mediator.

Ficoll was obtained from Cederlane Laboratories; Iscove’s medium was from Biochrom; CD hybridoma AGt and RPMI 1640 medium were from Invitrogen; LPS and KN-62 (1-[N,O-bis(5-isoquinoline-sulfonyl)-N-methyl-l-tyrosyl]-4-phenylpiperazine)4 were purchased from Calbiochem-Novabiochem; PPADS (pyridoxal-phosphate-6-azophenyl-2′,4′-disulfonic acid) was from Sigma-Aldrich; anti-human IL-10 mAb was from eBioscience; anti-dinitrophenyl hapten mAb and anti-β2-microglobulin HRP-conjugated mAb was from Dako; ATP and periodate-oxidized ATP (oATP) were from Sigma-Aldrich; recombinant IL-10 was from PeproTech; anti-human CD14-FITC-conjugated and CD14-PE-conjugated Abs were from Cymbus Biotechnology; anti-human IL-10R-PE-conjugated Abs were from R&D Systems; MEM-G9 and 5A6G7 mAbs were from Exbio; adenosine receptor agonist (SCH58261) was from Sigma-Aldrich; and the anti-human P2X7 L4 mAb was provided by Dr. James S. Wiley (University of Sydney, Penrith Hospital, Penrith, Australia).

Informed consent for blood drawing was obtained from twenty blood donors. PBMCs were isolated by Ficoll centrifugation and suspended at the concentration of 1 × 106/ml in Iscove’s medium containing 10% FCS plus antibiotics (penicillin/streptomycin) and maintained at 37°C in a humidified atmosphere with 5% CO2. The supernatants were centrifuged, to eliminate cellular debris, and stored at −80°C.

Monocytes were obtained from peripheral blood of healthy volunteers. Peripheral mononuclear cells were separated by Ficoll gradient centrifugation. Cells were further separated with anti-CD14 mAb-coated microbeads using MACS single-use separation columns from Miltenyi Biotec. To compare the sHLA-G secretion in PBMCs and monocyte cultures, the monocytes were suspended at the concentration of 1 × 104/ml corresponding to the average monocyte concentration (10%) present in 1 × 106/ml PBMC cultures. The cells were cultured in RPMI 1640 medium containing 10% FCS, 1% glutamine, 50 IU/ml penicillin, and 50 IU/ml streptomycin, and maintained at 37°C in a humidified atmosphere with 5% CO2.

To find optimal LPS concentrations and incubation times we ran preliminary dose-dependency curves with PBMCs from four healthy subjects. PBMCs were cultured as described above and stimulated with four different LPS concentrations (2.5, 5.0, 10.0, and 20.0 ng/ml). The optimal sHLA-G release occurred after a 48-h incubation in the presence of 25 ng/ml LPS (see also Ref. 28). This experimental protocol was chosen to investigate the effect of ATP on IL-10 and sHLA-G levels in culture supernatants. Increasing ATP concentrations (0.025, 0.05, 0.1, 0.5, 1 mM) were added to the PBMC cultures to inhibit sHLA-G secretion. As the optimal concentration turned out to be 1 mM, all experiments were performed with this ATP concentration unless otherwise indicated. In some experiments, LPS- and ATP-treated cultures were supplemented with 20 ng/ml recombinant IL-10 to restore sHLA-G secretion (28). The P2X7 receptor blockers were used at the following concentrations: KN-62, 50 nM; PPADS, 200 μM; and oATP, 600 μM. The anti-P2X7 mAb was used at a concentration of 10 μg/ml. The indifferent anti-dinitrophenyl hapten mAb was used at a concentration of 7.5 ng/ml. The anti-human IL-10 mAb was used at 7.5 ng/ml.

Mild acid treatment was performed by resuspending the cells in 0.2 ml of 0.2 M citrate-phosphate buffer (pH 3.0) supplemented with 0.1% BSA (29). After a 1-min incubation on ice the sample was neutralized by adding an excess of cold 0.1% BSA in PBS (pH 7.2), centrifuged, and rinsed with PBS (pH 7.2).

Quantitation of apoptotic and necrotic cells was performed with the annexin V-FITC apoptosis detection kit from Bender MedSystems. PBMCs were centrifuged at 300 × g for 15 min, resuspended in 1× binding buffer (10 mmol/L HEPES (pH 7.4), 140 mmol/L NaCl, 2.5 mmol/L CaCl2), and stained with annexin V-FITC and propidium iodide. Analysis was conducted with a FACSCount flow cytometer (BD Biosciences) using standard settings, and the data analysis was performed with the CellQuest software (BD Biosciences). Unstained cells were classified as “live”; cells stained with annexin V only were classified as “apoptotic”; cells stained with both annexin V and propidium iodide were classified as “end-stage apoptotic/necrotic”. CD14+ cell viability and concentration in peripheral blood were evaluated by cytofluorometry (FACSVantage; cytofluorimetry), using propidium iodide staining and an anti-human CD14-PE-conjugated mAb. IL-10R1 expression was evaluated by cytofluorometry, using an anti-human IL-10R-PE-conjugated mAb. HLA-G membrane expression was assessed with a MEM-G9-FITC-conjugated mAb, specific for HLA-G H chain β2-microglobulin-associated, and MEM-G1 mAb, specific for HLA-G free H chain molecule. FITC- or PE-conjugated rabbit anti-mouse IgG mAbs (Dako) were used as isotype control.

Cell viability was evaluated by trypan blue uptake or lactate dehydrogenase (LDH) release as previously described (30).

Human choriocarcinoma trophoblastic cells (JEG-3) were used as positive control, as they constitutively express HLA-G Ags (31). These cells were cultured in RPMI 1640 medium (Sigma-Aldrich) supplemented with 10% FCS and maintained at 37°C in a sterile humid atmosphere under 5% CO2 and 95% O2.

Total cellular RNA was prepared from monocytes and JEG-3 cultures with TRIzol reagent (Invitrogen) following producer protocol. The mRNA samples were digested with DNase. The quality and quantity of mRNA samples have been assessed by a 1% agarose gel electrophoresis, followed by ethidium bromide staining. mRNA samples were immediately used for cDNA synthesis or stored frozen at −80°C until use.

Two micrograms of mRNA, prepared as previously described, was reverse transcribed for each sample using the SuperScript first-strand synthesis system (Invitrogen) according to the manufacturer’s instructions. β-actin was used as control for the mRNA content and JEG-3 cell mRNA as positive HLA-G control. Specific primers for HLA-G molecules (32) were used for HLA-G mRNA amplification. Amplification products were analyzed by ethidium bromide-stained agarose gel electrophoresis. Densitometric mRNA analysis was performed with GeneTools software in a Geliance 600 system (PerkinElmer).

sHLA-G1, obtained from the proteolytic cleavage of membrane-bound HLA-G1, and HLA-G5, which was latter generated by mRNA alternative splicing, were assayed in PBMC and monocyte culture supernatants in triplicate as reported in the Essen Workshop on sHLA-G quantification (33). Briefly, 96-microwell plates (Nunc-Immuno Plate; PolySorp) were coated with the capture mAb MEM-G9 (Exbio), which recognizes shed sHLA-G1, a β2-microglobulin-associated form obtained from the proteolytic cleavage of membrane-bound HLA-G1, and the secreted HLA-G5 isoform generated by alternative splicing. The mAb was used at the concentration of 20 μg/ml in 0.1 M carbonate buffer (pH 9.5). After a 1-h incubation at 37°C followed by overnight incubation at 4°C, plates were saturated with 100 μl of PBS containing 4% BSA and further incubated overnight at 4°C. Thereafter, 50 μl of undiluted samples were added to triplicate wells. Standards were run on each plate in the 1–35 ng/ml range. Dilution buffer was the negative control (blank). After incubation for 2 h at 37°C, plates were rinsed three times with PBS-Tween and incubated for 1 h at 37°C together with 50 μl of the detecting anti-β2-microglobulin HRP-conjugated mAb (Dako). Samples were rinsed five times with PBS-Tween, 50 μl of o-phenylenediamine peroxidase substrate (Sigma-Aldrich) was added to each well, and incubation was carried out for 15 min at room temperature. The concentration of sHLA-G1/HLA-G5 was measured as the mean of triplicate cultures by absorbance at 450 nm with an ELISA microplate reader 400 (Packard), after subtraction of negative control OD values. Standard calibration curves were generated with supernatants from transfected HeLa-G5 cells (provided by Dr. E. Weiss, Institut für Anthropologie und Genetik, Ludwig-Maximilians-Universität, München, Germany), which were cultured in CD hybridoma AGt medium (Invitrogen) added with 1% FCS and antibiotics. Culture supernatants were collected at cell confluence and concentrated by lyophilization procedure. Following depletion of albumin with an albumin depletion kit (Enchant life science kit; Pall), the purification of the sHLA-G proteins was conducted as previously described (34). The intra-assay coefficient of variation was 1.4%, and the inter-assay coefficient of variation was 4%. The limit of sensitivity was 1 ng/ml.

HLA-G5 molecules, generated by mRNA alternative splicing, were assayed in PBMC and monocyte culture supernatants in triplicate as reported in the Essen Workshop on sHLA-G quantification (33). This ELISA test differs from the ELISA assay for sHLA-G1/HLA-G5 for the capture mAb 5A6G7 (Exbio), a specific Ab for the intron-4-derived polypeptide sequence of HLA-G5, and for the detection anti-HLA class I W6/32 (Dako) biotinylated Ab. The intra-assay coefficient of variation was 2.0%, the interassay coefficient of variation was 3.5%. The limit of sensitivity was 1.0 ng/ml.

sHLA-G1 was expressed as the difference between sHLA-G1/HLA-G5 and HLA-G5 concentrations.

IL-10 concentration was determined in triplicate undiluted samples using the commercially available human IL-10 BioSource ELISA kit with a detection limit of 0.2 pg/ml.

Statistical analysis was performed with a StatView software package (SAS Institute). sHLA-G and IL-10 levels were analyzed by Mann-Whitney U test. Statistical significance was assumed for p < 0.05 (two-tailed).

Whether extracellular ATP might trigger release of immunosuppressive factors is still a debated issue (22). Fig. 1,A shows that under basal conditions, that is, in the absence of added stimuli, sHLA-G concentration in PBMCs supernatants was below the detection limit of the assay, while LPS addition caused a strong sHLA-G release. Incubation in the presence of LPS and ATP fully abrogated sHLA-G release (p < 0.0042 for LPS vs LPS plus ATP). To rule out possible toxic effects of ATP, we performed an ATP dose dependency (Fig. 1,B) and FACS analysis to assess cell death. The ATP inhibitory effect was dose-dependent with half-maximal dose at an ATP concentration of ∼200 μM and maximal effect at 1 mM. Annexin V and propidium iodide staining (Fig. 2,A) show that after a 48-h incubation in the presence of LPS plus ATP, 84.7% of the cells were vital, while 5.6% were apoptotic (Fig. 2,A, upper right) and 9.7% were necrotic (Fig. 2,A, lower right). This percentage of live cells did not significantly differ from that of LPS-treated (83.7%) or untreated control cells (79.1%), confirming that ATP-dependent inhibition of sHLA-G release was not due to cell death. Cell viability was also assessed by LDH release (Fig. 2,B) and trypan blue exclusion (Fig. 2,C). Furthermore, to rule out selective deletion of monocyte cells as a consequence of the ATP treatment, CD14+ cells were evaluated by cytofluorometry (Fig. 2 D). Even though PBMCs are sensitive to the cytotoxic effect of extracellular ATP, this effect is mitigated by incubation in culture medium, especially if calf serum is present, and all these controls provided a confirmation of no cell death induced by ATP treatment. As PBMCs express multiple P2 subtypes, we tested the effect of other nucleotides.

FIGURE 1.

Extracellular ATP inhibits secretion of sHLA-G from LPS-activated PBMCs. A, PMBCs were cultured in Iscove’s medium at 37°C for 48 h and left untreated (Cont) or incubated in the presence of 10 ng/ml LPS (LPS) or LPS plus 1 mM ATP (LPS + ATP). Supernatants were then withdrawn and assayed by ELISA for sHLA-G content, as reported in Materials and Methods. B, Dose dependence of ATP on sHLA-G secretion. Cells were incubated in the presence of LPS plus increasing ATP concentrations. After 48 h, supernatants were withdrawn and assayed for sHLA-G as reported before.

FIGURE 1.

Extracellular ATP inhibits secretion of sHLA-G from LPS-activated PBMCs. A, PMBCs were cultured in Iscove’s medium at 37°C for 48 h and left untreated (Cont) or incubated in the presence of 10 ng/ml LPS (LPS) or LPS plus 1 mM ATP (LPS + ATP). Supernatants were then withdrawn and assayed by ELISA for sHLA-G content, as reported in Materials and Methods. B, Dose dependence of ATP on sHLA-G secretion. Cells were incubated in the presence of LPS plus increasing ATP concentrations. After 48 h, supernatants were withdrawn and assayed for sHLA-G as reported before.

Close modal
FIGURE 2.

Inhibition of sHLA-G release by ATP is not due to the cytotoxic effect of the nucleotide. A, PMBCs were left untreated (Cont) or incubated in the presence of 10 ng/ml LPS (LPS) or LPS plus 1 mM ATP (LPS + ATP), and at the end of the incubation, cell viability was fluorometrically analyzed as reported in Materials and Methods. Data are expresses as dot plots. Percentage of apoptotic (upper right) and late apoptosis/necrotic cells (lower right) are shown. B, LDH test was performed as reported in Materials and Methods. C, The cells were also stained with trypan blue to evaluate the cell count. Left panel, Phase-contrast images of cultured PBMCs stained with trypan blue. Few PBMCs stained with trypan blue were observed. Scale bar, 20 μm. Right panel, Alive cell percentage (open bars) and dead cells (filled bars). D, The PBMCs were stained with annexin V-FITC-conjugated (FL1) and CD14-PE-conjugated (FL2) mAbs and the percentage of CD14+ gated cells was evaluated with or without ATP treatment.

FIGURE 2.

Inhibition of sHLA-G release by ATP is not due to the cytotoxic effect of the nucleotide. A, PMBCs were left untreated (Cont) or incubated in the presence of 10 ng/ml LPS (LPS) or LPS plus 1 mM ATP (LPS + ATP), and at the end of the incubation, cell viability was fluorometrically analyzed as reported in Materials and Methods. Data are expresses as dot plots. Percentage of apoptotic (upper right) and late apoptosis/necrotic cells (lower right) are shown. B, LDH test was performed as reported in Materials and Methods. C, The cells were also stained with trypan blue to evaluate the cell count. Left panel, Phase-contrast images of cultured PBMCs stained with trypan blue. Few PBMCs stained with trypan blue were observed. Scale bar, 20 μm. Right panel, Alive cell percentage (open bars) and dead cells (filled bars). D, The PBMCs were stained with annexin V-FITC-conjugated (FL1) and CD14-PE-conjugated (FL2) mAbs and the percentage of CD14+ gated cells was evaluated with or without ATP treatment.

Close modal

Fig. 3 A shows data obtained with two nucleotide concentrations (500 μM, black bars, or 1 mM, open bars), except for BzATP (3′-O-(4-benzoyl)benzoyl-ATP), which was used at a concentration of 100 μM (black bars) or 0500 μM (open bars). ADP, which is a preferential agonist at P2Y1, P2Y12, and P2Y13 receptors, UTP, which is a preferential agonist at P2Y2 and P2Y4, UDP, a preferential agonist at P2Y6, and UDP-glucose, a preferential agonist at P2Y14, were inactive, while BzATP, which is a preferential, although not exclusive, agonist at P2X7, was a potent inhibitor at concentrations as low as 100 μM. The strong effect of BzATP, and the lack of activity of all other nucleotides tested besides ATP, strongly suggested an involvement of the P2X7 receptor. To test this hypothesis, PBMCs were preincubated in the presence of the noncompetitive covalent P2X inhibitor oATP, the competitive P2X7 antagonist KN-62 (35), or the widely used blocker PPADS.

FIGURE 3.

ATP and BzATP inhibit sHLA-G secretion from LPS-activated PBMCs. A, PMBCs were treated as reported in Fig. 1 and then incubated in the presence of 500 μM (filled bars) or 1 mM (open bars) nucleotide, except for BzATP (100 μM, filled bars; 500 μM, open bars). Supernatants were then withdrawn and assayed by ELISA for sHLA-G content, as reported in Materials and Methods. B, P2X inhibitors counteract the effect of ATP on HLA-G secretion. Cells were preincubated with 600 μM oATP for 2 h and then rinsed and stimulated with nucleotides. Cells were also incubated with 50 nM KN-62 or 200 μM PPADS (which were left throughout the experiment) and then incubated with ATP or BzATP. Cells were also incubated with 10 μg/ml anti-P2X7 (L4) mAb or with 7.5 ng/ml anti-dinitrophenyl hapten mAb (CTRmAb, which were left throughout the experiment) and then incubated with ATP. C, Adenosine is not involved in the inhibition of sHLA-G secretion induced by ATP. Cells were incubated as reported in Fig. 1. Some cells were also kept in the presence of 2 U/ml ADA or 100 nM SCH58261 adenosine receptor agonist throughout the experiment.

FIGURE 3.

ATP and BzATP inhibit sHLA-G secretion from LPS-activated PBMCs. A, PMBCs were treated as reported in Fig. 1 and then incubated in the presence of 500 μM (filled bars) or 1 mM (open bars) nucleotide, except for BzATP (100 μM, filled bars; 500 μM, open bars). Supernatants were then withdrawn and assayed by ELISA for sHLA-G content, as reported in Materials and Methods. B, P2X inhibitors counteract the effect of ATP on HLA-G secretion. Cells were preincubated with 600 μM oATP for 2 h and then rinsed and stimulated with nucleotides. Cells were also incubated with 50 nM KN-62 or 200 μM PPADS (which were left throughout the experiment) and then incubated with ATP or BzATP. Cells were also incubated with 10 μg/ml anti-P2X7 (L4) mAb or with 7.5 ng/ml anti-dinitrophenyl hapten mAb (CTRmAb, which were left throughout the experiment) and then incubated with ATP. C, Adenosine is not involved in the inhibition of sHLA-G secretion induced by ATP. Cells were incubated as reported in Fig. 1. Some cells were also kept in the presence of 2 U/ml ADA or 100 nM SCH58261 adenosine receptor agonist throughout the experiment.

Close modal

Fig. 3 B shows that oATP induced a complete (with BzATP as an agonist) or near complete (with ATP as an agonist) reversal of inhibition of sHLA-G secretion. KN-62 produced an ∼50–60% reversal of ATP or BzATP inhibitory effect, while PPADS was ineffective on ATP or BzATP inhibitory effect. To further support the role of the P2X7 receptor we used the specific L4 mouse anti-human P2X7 mAb raised against the extracellular domain of the P2X7 receptor. This mAb was previously shown to have a potent blocking effect of P2X7 function (36, 37). Addition of the L4 mAb caused an almost complete reversal of ATP inhibitory effect, while an indifferent mAb was without effect.

The ATP inhibitory effect might be due to accumulation of adenosine, a powerful immunosuppressant generated by the hydrolysis of extracellular ATP by ectonucleotidases (38). Thus, we tested the effect of the adenosine degrading enzyme adenosine deaminase (ADA) and of the adenosine receptor agonist SCH58261. Fig. 3 C shows that ADA and the adenosine receptor agonist did not reverse the inhibitory effect of ATP.

We routinely determined sHLA-G release after a 48-h incubation in the presence of the various stimuli or inhibitors. We then asked if shorter incubations in the presence of ATP were effective to inhibit sHLA-G release. We selected 15 min and 16 h as intermediate time points. At time 0, PBMCs were incubated in the presence of LPS plus ATP. At time of 15 min or time of 16 h, samples were rinsed to remove ATP and supplemented again with LPS-containing incubation solution. All samples were finally collected at 48 h. Fig. 4 shows that sHLA-G secretion was reduced by 30% by an ATP pulse of 15 min, and fully abrogated by an ATP pulse of 16 h.

FIGURE 4.

An ATP pulse is able to reduce sHLA-G secretion induced by LPS. Cells were pulsed with ATP for 15 min (ATP (15 min) + LPS) or 16 h (ATP (16 h) + LPS) then rinsed and then LPS was added. At the end of the incubation (48 h), supernatants were withdrawn and sHLA-G content was measured by ELISA.

FIGURE 4.

An ATP pulse is able to reduce sHLA-G secretion induced by LPS. Cells were pulsed with ATP for 15 min (ATP (15 min) + LPS) or 16 h (ATP (16 h) + LPS) then rinsed and then LPS was added. At the end of the incubation (48 h), supernatants were withdrawn and sHLA-G content was measured by ELISA.

Close modal

HLA-G is normally present in two isoforms identified as HLA-G1 and HLA-G5 originating from an alternative mRNA splicing (9). HLA-G1 is a membrane-bound molecule while HLA-G5 lacks the hydrophobic transmembrane domain. Fig. 5,A shows that LPS causes release of both HLA-G1 (filled bars) and HLA-G5 (open bars) isoforms, although the G5 form was predominant, and ATP inhibited release of both. Monocytes, that is, CD14+ cells, are the main source of sHLA-G in the blood. Therefore, we isolated monocytes from blood and verified the inhibitory effect of the nucleotide on this purified cell population. Fig. 5,B shows that ATP potently inhibited HLA-G release (▴), and this effect was not due to deletion of this cell population (▪; see also Fig. 2,D). The ATP dose-response curve was similar to that observed in PBMCs, strongly indicating monocytes as a candidate target for the ATP-induced response also in PBMCs. Fig. 5 C shows that also in purified human monocytes the great majority of the HLA-G molecules belonged to the G5 type and that ATP-induced inhibition was effective on both G5 and sG1 isoforms.

FIGURE 5.

HLA-G5 is the main target of ATP inhibitory effects. A, HLA-G content from PBMC supernatants was subjected to ELISA, which was performed by using two Abs recognizing the HLA-G1 and HLA-G5 isoforms (see Materials and Methods). B, Monocytes are target of ATP-mediated effect on HLA-G release. Monocytes were separated from PBMCs by Ficoll gradient (see Materials and Methods), suspended at the concentration of 1 × 104/ml to have the same proportion as in the PBMC cultures, and incubated as reported in Fig. 1 sHLA-G levels and CD14 expression were evaluated. C, HLA-G content from monocyte supernatants was subjected to ELISA, which was performed by using two Abs recognizing the HLA-G1 and HLA-G5 isoforms (see Materials and Methods).

FIGURE 5.

HLA-G5 is the main target of ATP inhibitory effects. A, HLA-G content from PBMC supernatants was subjected to ELISA, which was performed by using two Abs recognizing the HLA-G1 and HLA-G5 isoforms (see Materials and Methods). B, Monocytes are target of ATP-mediated effect on HLA-G release. Monocytes were separated from PBMCs by Ficoll gradient (see Materials and Methods), suspended at the concentration of 1 × 104/ml to have the same proportion as in the PBMC cultures, and incubated as reported in Fig. 1 sHLA-G levels and CD14 expression were evaluated. C, HLA-G content from monocyte supernatants was subjected to ELISA, which was performed by using two Abs recognizing the HLA-G1 and HLA-G5 isoforms (see Materials and Methods).

Close modal

The effects of ATP on HLA-G membrane expression were analyzed by cytofluorometry with the MEM-G9 mAb, specific for β2-microglobulin-associated HLA-G H chain. Stimulation with LPS induced a strong HLA-G membrane expression, which was fully abolished by ATP treatment (Fig. 6,A). To check if HLA-G molecules could be expressed as free heavy chains, cells were also stained with MEM-G1 mAb, specific for HLA-G free H chain. No staining was revealed after LPS and LPS plus ATP treatment. Positive staining with MEM-G1 was obtained treating LPS-activated monocytes with mild acid that dissociates β2-microglobulin from HLA-G H chain (29). Fig. 6 B shows that ATP also reduced HLA-G expression at the transcription level.

FIGURE 6.

ATP reduces the transcription of HLA-G and inhibits the production of membrane HLA-G molecules from LPS-activated monocytes. A, Monocytes were treated with LPS (LPS) or LPS + 1 mM ATP (LPS + ATP), then stained with CD14-PE-conjugated and MEM-G9-FITC-conjugated mAb to evaluate HLA-G H chain-β2-microglobulin-associated membrane expression or with MEM-G1 mAb to evaluate HLA-G free H chain membrane expression. LPS and 0.2 M citrate-phosphate buffer (pH 3.0) plus 0.1% BSA treatment (see Materials and Methods) (LPS + MILD ACID) was used as positive control to confirm the MEM-G1 staining of HLA-G free H chain. Isotype control was performed with FITC- or PE-conjugated rabbit anti-mouse IgG. B, mRNA was extracted from monocytes and amplified as previously described (see Materials and Methods). β-actin expression was used as mRNA concentration control. HLA-G mRNA expression in JEG-3 was used as positive control (PC); untreated (Ctr), LPS (LPS), and LPS plus 1 mM ATP-treated (LPS + ATP) monocytes were tested. M, DNA molecular weight marker IX (Roche Diagnostics); NC, negative control obtained without cDNA addition. The values obtained after densitometric analysis were indicated as arbitrary units (a.u.).

FIGURE 6.

ATP reduces the transcription of HLA-G and inhibits the production of membrane HLA-G molecules from LPS-activated monocytes. A, Monocytes were treated with LPS (LPS) or LPS + 1 mM ATP (LPS + ATP), then stained with CD14-PE-conjugated and MEM-G9-FITC-conjugated mAb to evaluate HLA-G H chain-β2-microglobulin-associated membrane expression or with MEM-G1 mAb to evaluate HLA-G free H chain membrane expression. LPS and 0.2 M citrate-phosphate buffer (pH 3.0) plus 0.1% BSA treatment (see Materials and Methods) (LPS + MILD ACID) was used as positive control to confirm the MEM-G1 staining of HLA-G free H chain. Isotype control was performed with FITC- or PE-conjugated rabbit anti-mouse IgG. B, mRNA was extracted from monocytes and amplified as previously described (see Materials and Methods). β-actin expression was used as mRNA concentration control. HLA-G mRNA expression in JEG-3 was used as positive control (PC); untreated (Ctr), LPS (LPS), and LPS plus 1 mM ATP-treated (LPS + ATP) monocytes were tested. M, DNA molecular weight marker IX (Roche Diagnostics); NC, negative control obtained without cDNA addition. The values obtained after densitometric analysis were indicated as arbitrary units (a.u.).

Close modal

Since the main stimulus for HLA-G release is IL-10, we tested whether ATP-mediated inhibition of sHLA-G release was due to inhibition of IL-10 release. Fig. 7,A shows that LPS-stimulated IL-10 secretion was significantly reduced by ATP but does not formally prove that sHLA-G decrease depended on decreased IL-10 secretion. Fig. 7,B shows that an anti-IL-10 mAb (•), but not an indifferent mAb (□), strongly inhibited sHLA-G release, strongly suggesting that sHLA-G release is closely dependent on IL-10 secretion. In further support of the role of IL-10, recombinant human IL-10 restored fully (0.5 mM ATP) or partially (1 mM ATP) sHLA-G production (Fig. 7,C). Addition of recombinant human IL-10 was also effective in restoring membrane expression of HLA-G (Fig. 7 C).

FIGURE 7.

ATP decreases IL-10 and sHLA-G production. A, Monocytes were stimulated as in Fig. 6 A and supernatants analyzed by ELISA for IL-10 content. B, Addition of anti-IL-10 Ab (•), but not an indifferent MoAb (□), strongly inhibited sHLA-G release. Monocytes were stimulated with 10 ng/ml LPS and supernatants were analyzed for sHLA-G content by ELISA. C, Recombinant IL-10 counteracts the effect of ATP on sHLA-G secretion and membrane HLA-G expression in monocyte cultures. Monocytes were stimulated with ATP, the culture supernatants were analyzed by ELISA for sHLA-G content (left panel), and monocytes were stained with MEM-G9-FITC-conjugated moAb to evaluate HLA-G membrane expression (right panel).

FIGURE 7.

ATP decreases IL-10 and sHLA-G production. A, Monocytes were stimulated as in Fig. 6 A and supernatants analyzed by ELISA for IL-10 content. B, Addition of anti-IL-10 Ab (•), but not an indifferent MoAb (□), strongly inhibited sHLA-G release. Monocytes were stimulated with 10 ng/ml LPS and supernatants were analyzed for sHLA-G content by ELISA. C, Recombinant IL-10 counteracts the effect of ATP on sHLA-G secretion and membrane HLA-G expression in monocyte cultures. Monocytes were stimulated with ATP, the culture supernatants were analyzed by ELISA for sHLA-G content (left panel), and monocytes were stained with MEM-G9-FITC-conjugated moAb to evaluate HLA-G membrane expression (right panel).

Close modal

Although ATP had a clear effect on IL-10 release, it could not be excluded that nucleotide also affected monocyte (i.e., CD14+ cells) responsiveness to IL-10 by decreasing cell surface expression of the IL-10 receptor (IL-10R1). To rule out this possibility, we correlated ATP-mediated inhibition of sHLA-G and IL-10 release to IL-10R1 subunit expression and to CD14+ cell number. Fig. 8 shows that while sHLA-G and IL-10 concentrations dramatically declined during a 48-h incubation in the presence of increasing ATP doses, IL-10 R1+ and CD14+ cells showed a negligible reduction, suggesting that ATP caused neither down-modulation of the IL-10 receptor nor deletion of monocyte cells.

FIGURE 8.

IL-10 R1 is not down-regulated by ATP. ATP decreases IL-10 and sHLA-G production. PBMCs were treated as in Fig. 1 and examined for IL-10 production, sHLA-G secretion, and CD14 and IL-10 R1 expression as reported in Materials and Methods.

FIGURE 8.

IL-10 R1 is not down-regulated by ATP. ATP decreases IL-10 and sHLA-G production. PBMCs were treated as in Fig. 1 and examined for IL-10 production, sHLA-G secretion, and CD14 and IL-10 R1 expression as reported in Materials and Methods.

Close modal

Release of ATP by many different cell types is a common event. Under certain conditions associated with inflammation, high ATP concentrations (100–500 μM) can accumulate into the tissue microenvironment, thus stimulating P2 receptors (4). Given the different sensitivity to ATP of cloned P2 subtypes, the wide range of ATP concentrations that might be present at inflammatory sites confers to purinergic signaling a remarkable plasticity. Extracellular ATP has a potent immunomodulatory activity that, depending on the dose, the time, and the biochemical composition of the inflammatory microenvironment, can be either proinflammatory or antiinflammatory (27). A few P2 receptors are responsible for the proinflammatory activity of ATP and other nucleotides: P2Y2 and P2Y12 mediate chemotaxis of inflammatory cells (39, 40, 41), P2Y6 supports phagocytosis (42), and P2Y11 supports differentiation of dendritic cells (43). However, there is no doubt that the key P2 receptor that is responsible for the proinflammatory effect of ATP in vitro and in vivo is P2X7. This receptor is one of the most potent triggers of the inflammasome, and thus of IL-1β processing and secretion (44), and it causes TNF release, NO (45) and O2− generation (46) and mediates inflammatory pain (47) and cell death (48).

The potent IL-1β-releasing activity of P2X7 would be by itself sufficient to justify its proinflammatory action; however, it is well known that most potent proinflammatory agents act by inducing a mix of responses that include stimulation of release of immunostimulating agents and inhibition of generation of immunodepressive factors, with the end result being amplification of inflammation.

The effect of ATP on IL-10 release is rather controversial, as it is reported that this nucleotide may cause either stimulation (23, 49, 50) or inhibition of IL-10 release (51), or have no effect (22). Such contrasting results are likely dependent on the different ATP doses and experimental protocols used, as there is increasing evidence that low (<100 mM) ATP concentrations down-modulate while high (>100 mM) concentrations stimulate inflammation (27). Few reports are available on the effect of ATP on the other major immunosuppressant TGF-β. A study in kidney mesangial cells showed that TGF-β secretion is stimulated by ATP (52), but no additional data are available in other cells relevant for immunity and inflammation.

One of the most potent recently characterized endogenous immunosuppressants is HLA-G. Human leukocyte Ag G is a nonclassical human class I molecule characterized by a limited polymorphism and a restricted tissue distribution with a broad inhibitory, immunosuppressive activity. High expression of HLA-G1 by the cytotrophoblast is thought to protect the fetus from the cytotoxic activity of maternal NK cells (10, 11). This hypothesis is corroborated by several studies showing alterations in blood levels of sHLA-G (sHLA-G1) and HLA-G5 in various pregnancy complications.

The immunosuppressive activity of HLA-G can also be unveiled outside the reproductive system. sHLA-G causes apoptosis of CD8+ T and NK cells, presumably via a Fas-FasL-dependent mechanism, inhibits proliferation of allo-specific CD4+, may enhance generation of suppressor CD4+ cells, and inhibits differentiation of immature into mature dendritic cells (13, 14, 15). The intracellular pathways responsible for this inhibitory effect are poorly known. However, there is evidence for a modified nuclear translocation of NF-κB and expression of costimulatory molecules (53).

Under physiological conditions, in the absence of factors that perturb body homeostasis, tissue and blood levels of HLA-G are very low. On the contrary, several disease states such as cancer, viral infection, and transplants potently up-regulate HLA-G expression and release. In organ transplantation there is a clear correlation between increased sHLA-G serum level and a diminished number of rejection episodes (19). Moreover, in autoimmune diseases nonactive clinical conditions in relapsing-remitting multiple sclerosis patients correlates with the presence of sHLA-G in cerebrospinal fluid (20), while impairment in the production of these antiinflammatory molecules is related to persistence of severe asthma (21). Finally, high HLA-G expression in rheumatoid arthritis and ulcerative colitis suggests that this molecule might have a broader involvement in inflammation (54, 55). Overall, these data suggest that HLA-G is a widely distributed immunoregulatory molecule with a strong immunosuppressive activity. Paradoxically, one of the most potent stimuli for HLA-G release is LPS. However, the paradox is only apparent, as many proinflammatory factors also set in motion feedback, inhibitory mechanisms devised to limit the amplification of inflammation.

In view of its potent proinflammatory action, we postulated that ATP should depress rather than stimulate IL-10 secretion. Interestingly, not only our prediction was fulfilled, but we found that the inhibitory effect of ATP extended to other potent endogenous immunosuppressants of HLA-G. The effect is dramatic, as in the presence of 1 mM ATP, LPS-stimulated soluble and membrane-bound HLA-G/β2-microglobulin-associated production was fully obliterated, while IL-10 release was decreased by ∼50%. Although ATP did not fully block IL-10 release, we think that inhibition of HLA-G was due to depression of IL-10 secretion, as supplementation of the ATP-treated cultures with recombinant IL-10 fully restored HLA-G expression. This may suggest that a threshold IL-10 concentration is necessary to efficiently trigger HLA-G release. We also wondered whether the ATP effect might at least in part be due to down-modulation of the IL-10 receptor. However, analysis of ATP-treated monocytes by cytofluorometry showed no reduction in plasma membrane expression of this receptor. Another possible reason for the sHLA-G decrease might be a reduction in the overall CD14+ cell population. This does not seem to be the case, as in the different subjects the extent of CD14+ cell decrease caused by an ATP dose (1 mM) that fully abrogated sHLA-G shedding ranged between 20% and 25%. Thus, cell death does not seem to be a cause for ATP-dependent inhibition of LPS-stimulated sHLA-G production. Finally, restoration by exogenous IL-10 of HLA-G production strongly suggests that the ATP effect was not irreversible.

These observations validate the paradigm stemming from several previous studies in the field that support a chief function for ATP as a negative modulator of inflammation at low concentrations, an effect mainly mediated via P2Y receptors, and on the other hand a strong proinflammatory effect for ATP at high concentrations, very likely acting at the low-affinity P2X7 receptor (27, 56). An observation made in microglia adds a further level of complexity to this picture. In this cell type, LPS-stimulated release of IL-10 was shown to be dependent on the release of ATP and the ensuing autocrine/paracrine stimulation via P2Y receptors (57). Since LPS causes release of small ATP amounts, this observation is in keeping with the tolerogenic activity of low doses of extracellular ATP and supports data showing IL-10 releasing activity of exogenous ATP.

In conclusion, our data add further support to the hypothesis that extracellular ATP is a crucial immunomodulating agent.

The authors have no financial conflicts 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.

1

This work was supported by grants from the Italian Ministry of Education, University and Scientific Research, the Italian Association for Cancer Research, the Italian Space Agency, the Regione Emilia-Romagna (“Progetto Università-Regione” and “Moniter”), Telethon of Italy, and institutional funds from the University of Ferrara.

4

Abbreviations used in this paper: KN-62, 1-[N,O-bis(5-isoquinoline-sulfonyl)-N-methyl-l-tyrosyl]-4-phenylpiperazine; ADA, adenosine deaminase; BzATP, 3′-O-(4-benzoyl)benzoyl-ATP; oATP, periodate-oxidized ATP; PPADS, pyridoxal-phosphate-6-azophenyl-2′,4′-disulfonic acid; LDH, lactate dehydrogenase.

1
Lu, Y. C., W. C. Yeh, P. S. Ohashi.
2008
. LPS/TLR4 signal transduction pathway.
Cytokine
42
:
145
-151.
2
Herzum, I., H. Renz.
2008
. Inflammatory markers in SIRS, sepsis and septic shock.
Curr. Med. Chem.
15
:
581
-587.
3
Ferrari, D., P. Chiozzi, S. Falzoni, S. Hanau, F. Di Virgilio.
1997
. Purinergic modulation of interleukin-1β release from microglial cells stimulated with bacterial endotoxin.
J. Exp. Med.
185
:
579
-582.
4
Pellegatti, P., L. Raffaghello, G. Bianchi, F. Piccardi, V. Pistoia, F. Di Virgilio.
2008
. Increased level of extracellular ATP at tumor sites: in vivo imaging with plasma membrane luciferase.
PLoS ONE
3
:
e2599
5
Abbracchio, M. P., G. Burnstock.
1994
. Purinoceptors: are there families of P2X and P2Y purinoceptors?.
Pharmacol. Ther.
64
:
445
-475.
6
North, R. A., A. Surprenant.
2000
. Pharmacology of cloned P2X receptor.
Annu. Rev. Pharmacol. Toxicol.
40
:
563
-580.
7
von Kügelgen, I., A. Wetter.
2000
. Molecular pharmacology of P2Y-receptors.
Naunyn-Schmiedebergs Arch. Pharmacol.
362
:
310
-323.
8
Moscoso, J., J. I. Serrano-Vela, B. Perez-Saborido, E. Moreno, A. Amaiz-Villena.
2007
. A novel HLA-G allele (HLA-G*0108) with an α-3 domain amino acid change.
Tissue Antigens
70
:
171
-173.
9
Clements, C. S., L. Kjer-Nielsen, L. Kostenko, J. McCluskey, J. Rossjohn.
2006
. The production, purification and crystallization of a soluble form of the nonclassical MHC HLA-G: the essential role of cobalt.
Acta Crystallogr. Sect. F Struct. Biol. Cryst. Commun.
62
:
70
-73.
10
Kovats, S., E. K. Main, C. Librach, M. Stubblebine, S. J. Fisher, R. A. DeMars.
1990
. HLA class I antigen, HLA-G, expressed in human trophoblasts.
Science
248
:
220
-223.
11
Lefebvre, S., P. Moreau, V. Guiard, E. C. Ibrahim, F. Adrian-Cabestre, C. Menier, J. Dausset, E. D. Carosella, P. Paul.
1999
. Molecular mechanisms controlling constitutive and IFN-γ-inducible HLA-G expression in various cell types.
J. Reprod. Immunol.
43
:
213
-224.
12
Yie, S. M., R. Xiao, C. L. Librach.
2006
. Progesterone regulates HLA-G gene expression through a novel progesterone response element.
Hum. Reprod.
21
:
2538
-2544.
13
Rouas-Freiss, N., R. E. Marchal, M. Kirszenbaum, J. Dausset, E. D. Carosella.
1997
. The α1 domain of HLA-G1 and HLA-G2 inhibits cytotoxicity induced by natural killer cells: is HLA-G the public ligand for natural killer cell inhibitory receptors?.
Proc. Natl. Acad. Sci. USA
94
:
5249
-5254.
14
Fournel, S., M. Aguerre-Girr, X. Huc, F. Lenfant, A. Alam, A. Toubert, A. Bensussan, P. Le Bouteiller.
2000
. Cutting edge: soluble HLA-G1 triggers CD95/CD95 ligand-mediated apoptosis in activated CD8+ cells by interacting with CD8.
J. Immunol.
164
:
6100
-6104.
15
Bainbridge, D. R., S. A. Ellis, I. L. Sargent.
2000
. HLA-G suppresses proliferation of CD4+ T-lymphocytes.
J. Reprod. Immunol.
48
:
17
-26.
16
Aractingi, S., N. Briand, C. Le Danff, M. Viguier, H. Bachelez, L. Michel, L. Dubertret, E. D. Carosella.
2001
. HLA-G and NK receptor are expressed in psoriatic skin: a possible pathway for regulating infiltrating T cells?.
Am. J. Pathol.
159
:
71
-77.
17
Yie, S. M., H. Yang, S. R. Ye, K. Li, D. D. Dong, X. M. Lin.
2007
. Expression of human leucocyte antigen G (HLA-G) is associated with prognosis in non-small cell lung cancer.
Lung Cancer
58
:
267
-274.
18
Megert, F., C. Prehaud, M. Lafage, P. Moreau, N. Rouas-Freiss, E. D. Carosella, M. Lafon.
2007
. Modulation of HLA-G and HLA-E expression in human neuronal cells after rabies virus or herpes virus simplex type 1 infections.
Hum. Immunol.
68
:
294
-302.
19
Luque, J., M. I. Torres, M. D. Aumente, J. M. Lozano, G. García-Jurado, R. González, M. R. Alvarez-López, J. M. Arizón, J. Peña.
2006
. sHLA-G levels in the monitoring of immunosuppressive therapy and rejection following heart transplantation.
Transplant Immunol.
17
:
70
-73.
20
Fainardi, E., R. Rizzo, L. Melchiorri, M. Castellazzi, E. Paolino, M. R. Tola, E. Granieri, O. R. Baricordi.
2006
. Intrathecal synthesis of soluble HLA-G and HLA-I molecules are reciprocally associated to clinical and MRI activity in patients with multiple sclerosis.
Mult. Scler.
12
:
2
-12.
21
Rizzo, R., C. E. Mapp, L. Melchiorri, P. Maestrelli, A. Visentin, S. Ferretti, I. Bononi, D. Miotto, O. R. Baricordi.
2005
. Defective production of soluble HLA-G molecules by peripheral blood monocytes in patients with asthma.
J. Allergy Clin. Immunol.
115
:
508
-513.
22
la Sala, A., D. Ferrari, S. Corinti, A. Cavani, F. Di Virgilio, G. Girolomoni.
2001
. Extracellular ATP induces a distorted maturation of dendritic cells and inhibits their capacity to initiate Th1 responses.
J. Immunol.
166
:
1611
-1617.
23
Wilkin, F., P. Stordeur, M. Goldman, J. M. Boeynaems, B. Robaye.
2002
. Extracellular adenine nucleotides modulate cytokine production by human monocyte-derived dendritic cells: dual effect on IL-12 and stimulation of IL-10.
Eur. J. Immunol.
32
:
2409
-2417.
24
Ferrari, D., M. Villalba, P. Chiozzi, S. Falzoni, P. Ricciardi-Castagnoli, F. Di Virgilio.
1996
. Mouse microglial cells express a plasma membrane pore gated by extracellular ATP.
J. Immunol.
156
:
1531
-1539.
25
Solini, A., P. Chiozzi, A. Morelli, R. Fellin, F. Di Virgilio.
1999
. Human primary fibroblasts in vitro express a purinergic P2X7 receptor coupled to ion fluxes, microvesicle formation and IL-6 release.
J. Cell Sci.
112
:
297
-305.
26
Suzuki, T., I. Hide, K. Ido, S. Kohsaka, K. Inoue, Y. Nakata.
2004
. Production and release of neuroprotective tumor necrosis factor by P2X7 receptor-activated microglia.
J. Neurosci.
24
:
1
-7.
27
Bours, M. J., E. L. Swennen, F. Di Virgilio, B. N. Cronstein, P. C. Dagnelie.
2006
. Adenosine 5′-triphosphate and adenosine as endogenous signaling molecules in immunity and inflammation.
Pharmacol. Ther.
112
:
358
-404.
28
Rizzo, R., T. V. Hviid, M. Stignani, A. Balboni, M. T. Grappa, L. Melchiorri, O. R. Baricordi.
2005
. The HLA-G genotype is associated with IL-10 levels in activated PBMCs.
Immunogenetics
57
:
172
-181.
29
Poláková, K., D. Kuba, G. Russ.
2004
. The 4H84 monoclonal antibody detecting β2m free nonclassical HLA-G molecules also binds to free heavy chains of classical HLA class I antigens present on activated lymphocytes.
Hum. Immunol.
65
:
157
-162.
30
Falzoni, S., M. Munerati, D. Ferrari, S. Spisani, S. Moretti, F. Di Virgilio.
1995
. The purinergic P2Z receptor of human macrophage cells: characterization and possible physiological role.
J. Clin. Invest.
95
:
1207
-1216.
31
Ringler, G. E., J. F. Strauss.
1990
. In vitro systems for the study of human placental endocrine function.
Endocr. Rev.
11
:
105
-123.
32
Yao, Y. Q., D. H. Barlow, I. L. Sargent.
2005
. Differential expression of alternatively spliced transcripts of HLA-G in human preimplantation embryos and inner cell masses.
J. Immunol.
175
:
8379
-8385.
33
Rebmann, V., J. LeMaoult, N. Rouas-Freiss, E. D. Carosella, H. Grosse-Wilde.
2007
. Quantification and identification of soluble HLA-G isoforms.
Tissue Antigens
69
:
143
-149.
34
Le Friec, G., B. Laupeze, O. Fardel, Y. Sebti, C. Pangault, V. Guilloux, A. Beauplet, R. Fauchet, L. Amiot.
2003
. Soluble HLA-G inhibits human dendritic cell-triggered allogeneic T-cell proliferation without altering dendritic differentiation and maturation processes.
Hum. Immunol.
64
:
752
-761.
35
Baraldi, P. G., F. Di Virgilio, R. Romagnoli.
2004
. Agonists and antagonists acting at P2X7 receptor.
Curr. Top. Med. Chem.
4
:
1707
-1717.
36
Buell, G., I. P. Chessell, A. D. Michel, G. Collo, M. Salazzo, S. Herren, D. Gretener, C. Grahames, R. Kaur, M. H. Kosco-Vilbois, P. P. Humphrey.
1998
. Blockade of human P2X7 receptor function with a monoclonal antibody.
Blood
92
:
3521
-3528.
37
Falzoni, S., P. Chiozzi, D. Ferrari, G. Buell, F. Di Virgilio.
2000
. P2X7 receptor and polykarion formation.
Mol. Biol. Cell
11
:
3169
-3176.
38
Zimmermann, H., N. Braun, B. Kegel, P. Heine.
1998
. New insights into molecular structure and function of ectonucleotidases in the nervous system.
Neurochem. Int.
32
:
421
-425.
39
Honda, S., Y. Sasaki, K. Ohsawa, Y. Imai, Y. Nakamura, K. Inoue, S. Kohsaka.
2001
. Extracellular ATP or ADP induce chemotaxis of cultured microglia through Gi/o-coupled P2Y receptors.
J. Neurosci.
21
:
1975
-1982.
40
Haynes, S. E., G. Hollopeter, G. Yang, D. Kurpius, M. E. Dailey, W. B. Gan, D. Julius.
2006
. The P2Y12 receptor regulates microglial activation by extracellular nucleotides.
Nat. Neurosci.
9
:
1512
-1519.
41
Ohsawa, K., Y. Irino, Y. Nakamura, C. Akazawa, K. Inoue, S. Kohsaka.
2007
. Involvement of P2X4 and P2Y12 receptors in ATP-induced microglial chemotaxis.
Glia
55
:
604
-616.
42
Koizumi, S., Y. Shigemoto-Mogami, K. Nasu-Tada, Y. Shinozaki, K. Ohsawa, M. Tsuda, B.V. Joshi, K.A. Jacobson, S. Kohsaka, K. Inoue.
2007
. UDP acting at P2Y6 receptors is a mediator of microglial phagocytosis.
Nature
446
:
1091
-1095.
43
Schnurr, M., T. Toy, P. Stoitzner, P. Cameron, A. Shin, T. Beecroft, I. D. Davis, J. Cebon, E. Maraskovsky.
2003
. ATP gradients inhibit the migratory capacity of specific human dendritic cell types: implications for P2Y11 receptor signaling.
Blood
102
:
613
-620.
44
Di Virgilio, F..
2007
. Liaisons dangereuses: P2X7 and the inflammasome.
Trends Pharmacol. Sci.
28
:
465
-472.
45
Hu, Y., P. L. Fisette, L. C. Denlinger, A. G. Guadarrama, J. A. Sommer, R. A. Proctor, P. J. Bertics.
1998
. Purinergic receptor modulation of lipopolysaccharide signaling and inducible nitric-oxide synthase expression in RAW 264.7 macrophages.
J. Biol. Chem.
273
:
27170
-27175.
46
Suh, B. C., J. S. Kim, U. Namgung, H. Ha, K. T. Kim.
2001
. P2X7 nucleotide receptor mediation of membrane pore formation and superoxide generation in human promyelocytes and neutrophils.
J. Immunol.
166
:
6754
-6763.
47
Chessell, I. P., J. P. Hatcher, C. Bountra, A. D. Michel, J. P. Hughes, P. Green, J. Egerton, M. Murfin, J. Richardson, W. L. Peck, et al
2005
. Disruption of the P2X7 purinoceptor gene abolishes chronic inflammatory and neuropathic pain.
Pain
114
:
386
-396.
48
Di Virgilio, F., P. Chiozzi, S. Falzoni, D. Ferrari, J. M. Sanz, V. Venketaramanm, O. R. Baricordi.
1998
. Cytolytic P2X purinoceptors.
Cell Death Differ.
5
:
191
-199.
49
Haskó, G., D. G. Kuhel, A. L. Salzman, C. Szabó.
2000
. ATP suppression of interleukin-12 and tumour necrosis factor-α release from macrophages.
Br. J. Pharmacol.
129
:
909
-914.
50
Swennen, E. L., A. Bast, P. C. Dagnelie.
2005
. Immunoregulatory effects of adenosine 5′-triphosphate on cytokine release from stimulated whole blood.
Eur. J. Immunol.
35
:
852
-858.
51
Duhant, X., L. Schandené, C. Bruyns, N. S. Gonzalez, M. Goldman, J. M. Boeynaems, D. Communi.
2002
. Extracellular adenine nucleotides inhibit the activation of human CD4+ T lymphocytes.
J. Immunol.
169
:
15
-21.
52
Solini, A., C. Iacobini, C. Ricci, P. Chiozzi, L. Amadio, F. Pricci, U. Di Mario, F. Di Virgilio, G. Pugliese.
2005
. Purinergic modulation of mesangial extracellular matrix production: role in diabetic and other glomerular diseases.
Kidney Int.
67
:
875
-885.
53
Yu, Y., Y. Wang, M. Feng.
2008
. Human leukocyte antigen-G1 inhibits natural killer cytotoxicity through blocking the activating signal transduction pathway and formation of activating immunologic synapse.
Hum. Immunol.
69
:
16
-54.
54
Rizzo, R., M. Rubini, M. Govoni, M. Padovan, L. Melchiorri, M. Stignani, S. Carturan, S. Ferretti, F. Trotta, O. R. Baricordi.
2006
. HLA-G 14-bp polymorphism regulates the methotrexate response in rheumatoid arthritis.
Pharmacogenet. Genomics
16
:
615
-623.
55
Rizzo, R., L. Melchiorri, L. Simone, M. Stignani, A. Marzola, S. Gullini, O. R. Baricordi.
2008
. Different production of soluble HLA-G antigens by peripheral blood mononuclear cells in ulcerative colitis and Crohn’s disease: a noninvasive diagnostic tool?.
Inflamm. Bowel Dis.
14
:
100
-105.
56
Di Virgilio, F., P. Chiozzi, D. Ferrari, S. Falzoni, J. M. Sanz, A. Morelli, M. Torboli, G. Bolognesi, O. R. Baricordi.
2001
. Nucleotide receptors: an emerging family of regulatory molecules in blood cells.
Blood
97
:
587
-600.
57
Seo, D. R., K. Y. Kim, Y. B. Lee.
2004
. Interleukin-10 expression in lipopolysaccharide-activated microglia is mediated by extracellular ATP in an autocrine fashion.
Neuroreport
15
:
1157
-1161.