Skip to main content
Journal of Leukocyte Biology logoLink to Journal of Leukocyte Biology
. 2013 Oct;94(4):759–768. doi: 10.1189/jlb.0712351

Nucleotide receptor P2RX7 stimulation enhances LPS-induced interferon-β production in murine macrophages

M L Gavala *, Y-P Liu †,, L Y Lenertz *, L Zeng †,, J B Blanchette *, A G Guadarrama *, L C Denlinger , P J Bertics *,1, J A Smith †,‡,2
PMCID: PMC3774844  PMID: 23911869

LPS-induced IFN-β is enhanced in macrophages upon activation of P2RX7, potentially related to an increase in active IRF-3.

Keywords: IRF-3, extracellular ATP, enhanceosome, type I IFN, endotoxin

Abstract

Stimulation of P2RX7 with extracellular ATP potentiates numerous LPS-induced proinflammatory events, including cytokine induction in macrophages, but the molecular mechanisms underlying this process are not well defined. Although P2RX7 ligation has been proposed to activate several transcription factors, many of the LPS-induced mediators affected by P2RX7 activation are not induced by P2RX7 agonists alone, suggesting a complementary role for P2RX7 in transcriptional regulation. Type I IFN production, whose expression is tightly controlled by multiple transcription factors that form an enhanceosome, is critical for resistance against LPS-containing bacteria. The effect of purinergic receptor signaling on LPS-dependent type I IFN is unknown and would be of great relevance to a diverse array of inflammatory conditions. The present study demonstrates that stimulation of macrophages with P2RX7 agonists substantially enhances LPS-induced IFN-β expression, and this enhancement is ablated in macrophages that do not express functional P2RX7 or when the MAPK MEK1/2 pathways are inhibited. Potentiation of LPS-induced IFN-β expression following P2RX7 stimulation is likely transcriptionally regulated, as this enhancement is observed at the IFN-β promoter level. Furthermore, P2RX7 stimulation is able to increase the phosphorylation and subsequent IFN-β promoter occupancy of IRF-3, a transcription factor that is critical for IFN-β transcription by TLR agonists. This newly discovered role for P2RX7 in IFN regulation may have implications in antimicrobial defense, which has been linked to P2RX7 activation in other studies.

Introduction

Recent immunological studies have revealed an intriguing role for purinergic receptors as danger sensors of immune cells that influence the magnitude of a mounted innate immune response, depending on the concentration and duration of extracellular ATP exposure [13]. In particular, extracellular ATP-induced activation of the ionotropic P2RX7 nucleotide receptor has been implicated in the modulation of host immune response to pathogens and inflammatory disease progression [1, 46]. Studies conducted in P2RX7−/− mice found reduced incidence and severity of anticollagen-induced arthritis symptoms compared with littermate controls [7]. Results from further animal studies suggest that targeting P2RX7 may be useful in the treatment of arthritis and spinal cord injury [811]. Moreover, it has been observed that patients with rheumatoid arthritis have increased P2RX7 expression on their blood monocytes compared with nonarthritic controls [12]. Furthermore, mutations in P2RX7 have been linked to increased susceptibility to infectious diseases, which has been ascribed, at least in part, to impaired macrophage function [6]. Thus, P2RX7 activation has been linked to numerous diseases, but the intracellular signaling mechanisms that are induced are incompletely understood.

Even though P2RX7 initiates many processes on its own, nucleotide stimulation of P2RX7 also displays a striking capacity to modulate endotoxin/LPS-induced signaling [1317]. Early purinergic signaling studies found that administration of 2-methylthioadenosine-5′-O-triphosphate, a partial P2RX7 agonist, protected mice against lethal endotoxic shock [18]. Moreover, P2RX7 ligation increases LPS-induced ROS, and NO production [15, 1921] plays a critical role in the processing and release of IL-converting enzyme (caspase-1)-dependent cytokines, IL-1β and IL-18, and augments production of TNF-α and IL-6 in macrophages [5, 7, 22]. Interestingly, PBMCs from human subjects that possess a polymorphism that impairs P2RX7 function produced less proinflammatory (e.g., TNF-α) but more anti-inflammatory (e.g., IL-10) cytokines in response to LPS stimulation [23]. Although it is known that stimulation of P2RX7 can potentiate numerous LPS-induced proinflammatory events, particularly in macrophages, the molecular mechanisms underlying this process are not well defined.

It has been suggested that the enhancement of LPS-induced IL-1β and IL-18 release by nucleotides involves the formation of the inflammasome [22]. Although the inflammasome theory explains the release of preformed cytokines within endocytic vesicles [24] or microvesicles [25] in the cell, it does not address the enhancement of LPS-induced mRNA and protein expression after longer stimulation with nucleotides. For instance, activation of P2RX7 increases the mRNA and/or protein expression of TNF-α, iNOS, and COX-2 in LPS-primed macrophages [17, 19, 21, 26]. In terms of intracellular signaling events, it has been observed that costimulation of macrophages with P2RX7 agonists and LPS results in prolonged IκBα degradation and increased NF-κB-binding activity, decreased ERK1/2 activation, and increased JNK and p38 MAPK activation [13]. Although P2RX7 activates multiple transcription factors [27, 28], many of the LPS-induced mediators that P2RX7 has been shown to modulate are not induced by P2RX7 stimulation alone [16, 17]. One explanation for the potentiation of LPS-induced mediator expression following P2RX7 stimulation could be the activation of an enhanceosome, a complex of transcription factors that induce gene expression when stimulated together.

One gene, whose transcription is tightly regulated by enhanceosome formation and is induced by LPS stimulation in macrophages, is IFN-β. Similarly to the actions of P2RX7, type I IFNs (IFN-α and -β), produced by macrophages, play an important role in antimicrobial immunity by acting in an autocrine manner to enhance phagocytosis and the induction of iNOS [29]. IFN-β is the primary type I IFN produced by macrophages following LPS stimulation [3032]. It is worth noting that P2RX7 stimulation and IFN-β play important roles in inflammatory responses to pathogenic bacteria [18, 33]. Interestingly, IFN-β null mice are completely resistant to LPS-induced septic shock, and inhibition of IFN-β has been proposed as a potential treatment for sepsis [34, 35].

The majority of the signaling events induced by LPS is mediated through TLR4. Stimulation of TLR4 leads to the activation of MyD88- and TRIF-dependent signaling cascades, leading to the activation of NF-κB and IRF-3, respectively, which are both critical transcription factors in the induction of IFN-β expression [30, 36]. Although P2RX7 agonists have been demonstrated to enhance the release of inflammatory cytokines induced via the MyD88-dependent pathway (e.g., IL-1β, TNF-α) [5, 7, 22], the effect of P2RX7 signaling on TRIF-dependent signaling or the induction of type I IFN by LPS is entirely unknown. Considering that P2RX7 is able to modulate other LPS-initiated signaling events and shares many immunological processes with IFN-β, a role for P2RX7 stimulation in the modulation of LPS-induced IFN-β expression was investigated. We found that activation of P2RX7 significantly enhanced the expression and release of LPS-induced IFN-β from macrophages, potentially through the activation of IRF-3.

MATERIALS AND METHODS

Materials

Unless otherwise specified, reagents for cell culture were purchased from Mediatech (Herndon, VA, USA). All nucleotides and LPS (Escherichia coli, serotype 0111:B4) were obtained from Sigma Chemical (St. Louis, MO, USA). Antibodies directed against FosB, β-tubulin, and pIRF-3 (against Ser 396; Cat. #4947) for immunoblotting were purchased from Cell Signaling Technology (Danvers, MA, USA); anti-pIRF-3 (against Ser 386; Cat. #2562-1) for immunofluorescence was purchased from Epitomics (Burlingame, CA, USA); anti-IRF-3 antibody was purchased from ProSci (Poway, CA, USA); Alexa Fluor 488 goat anti-rabbit IgG and ProLong Gold antifade reagent with DAPI were purchased from Invitrogen (Grand Island, NY, USA); anti-β-actin, anti-IRF-3 (for ChIP; Cat. #SC-9082x), and HRP-conjugated anti-rabbit IgG antibodies were purchased from Santa Cruz Biotechnology (Santa Cruz, CA, USA). The active MEK1/2 inhibitor U0126 was purchased from Promega (Madison, WI, USA). The p38 inhibitor SB203580 was from Cell Signaling Technology. MRT67307 was a generous gift of Dr. Philip Cohen (University of Dundee, Scotland).

Cell culture

Murine RAW 264.7 macrophages were obtained from American Type Culture Collection (Manassas, VA, USA), whereas P2RX7-defective RAW cells (denoted SF) were generated as described previously [37]. Both RAW 264.7 cell lines were maintained in RPMI, supplemented with 5% cosmic calf serum, 2 mM sodium pyruvate, 2 mM L-glutamine, and 100 U/ml penicillin/streptomycin. All cells were grown in 10 cm tissue-culture dishes at 37°C in a humidified atmosphere with 5% CO2.

Isolation and culture of bone marrow-derived macrophages

The bone marrow cells of the tibiae, femurs, pelvic, and humerus bones of C57BL/6 mice were rinsed out with DMEM (Invitrogen) cell maintenance media, collected in a cell strainer, and layered over Histopaque 1083 (Sigma Chemical), twice, to isolate mononuclear cells. Cells were maintained in DMEM media, supplemented with 10% FCS, 20 ng/ml human rM-CSF, or CMG14-12 supernatant (as M-CSF source [38]), and plated in 10-cm Petri dishes at a density of 5 × 106 cells/plate. Three days after isolation, cells were lifted and plated at a density of 2.5 × 105 cells/well in a 12-well plate in CMG14-12-supplemented complete media. Six days after isolation, cells were treated as described.

qPCR

RAW 264.7 macrophages were plated at a density of 7.5 × 105 cells/well in six- or 12-well tissue-culture plates and incubated at 37°C, the day before each experiment. Following treatment and subsequent cell lysis in Trizol (Invitrogen), mRNA was isolated and converted to cDNA, according to the manufacturer's instructions. Primers directed toward murine IFN-β, FosB, or 18S (loading control) were designed using Beacon Design software (Premier Biosoft, Palo Alto, CA, USA). IFN-β primers are: F-ACT AGA GGA AAA GCA AGA GGA AAG, R-CCA CCA TCC AGG CGT AGC; FosB primers: F-TTC CAG ACC AGC CAA GAC, R-ACC GAA GAA GTG TAC GAA GG; 18S primers: F-GGA CAC GGA CAG GAT TGA CAG, R-ATC GCT CCA CCA ACT AAG AAC G. PCR reactions were detected by iQ SYBR Green supermix dye (Bio-Rad, Hercules, CA, USA) and were performed on a MyiQ real-time thermal cycler (Bio-Rad; 57–58°C annealing temp, 40 cycles).

ELISA

RAW 264.7 macrophages were plated at a density of 4 × 105 cells/well in 12-well tissue-culture plates and incubated at 37°C the day before treatment. Following treatment, the supernatants were centrifuged to remove cell debris and analyzed for IFN-β via ELISA, according to the manufacturer's specifications (PBL Laboratories, Piscataway, NJ, USA).

IFN-β luciferase promoter activation assay

The murine IFN-β promoter sequence (+10 from start site to −329) was inserted into a firefly luciferase reporter construct (Promega) and 2 μg of this plasmid was transfected, along with 0.2 μg of a pRL Renilla luciferase control vector (Promega)/2 × 106 RAW 264.7 macrophages/cuvette via Amaxa electroporation. The cells were plated subsequently at 5 × 105 cells/well in a 24-well plate and incubated at 37°C overnight. The day after electroporation, the cells were treated and then lysed to quantify luminescence using the Dual-Glo luciferase assay system (Promega), according to the manufacturer's specifications.

FosB siRNA

RAW 264.7 macrophages were transfected with SMARTpool ON-TARGET siRNA, directed toward FosB or control siRNA (Dharmacon, Lafayette, CO, USA) using Amaxa Nucleofector Kit V and an Amaxa electroporator (Lonza, Walkersville, MD, USA), according to the manufacturer's specifications. Electroporated cells were plated in six-well plates (1×106 cells/well), and 24 h after transfection, the cells were treated as specified and lysed in Trizol for mRNA processing.

Immunoblotting

RAW 264.7 macrophages were plated at a density of 3 × 105 cells/well in 24-well tissue-culture plates and incubated at 37°C the day before each experiment. Following treatment and subsequent cell lysis with SDS sample buffer (10 mM Tris, 1 mM EDTA, 0.5 mM Na3VO4, 1 mM DTT, 1% SDS, and 10% glycerol), the proteins were resolved on SDS-PAGE gels and transferred to PVDF membranes. Immunoblotting was performed by incubating the membranes with commercially available antisera, according to the manufacturer's protocols, and were visualized using the EpiChemi II darkroom (UVP, Upland, CA, USA), equipped with a 12-bit cooled, charge-coupled device camera.

Immunocytochemistry

RAW 264.7 cells (2×105/4 ml media) were plated in 60-mm dishes containing 20 mm × 20 mm coverslips and incubated at 37°C for 24 h. Cells were then stimulated as described. After stimulation, the media were removed, and the coverslip was rinsed with 0.1 M phosphate buffer (pH 7.4). The cells were fixed for 20 min with 4% PFA/PBS and then permeabilized with PBS containing 0.2% Triton X-100. To detect pIRF-3, the coverslips were blocked with 10% goat serum in 0.2% Triton X-100 PBS for 1 h and then incubated overnight with 1:400 anti-pIRF-3 antibody, diluted in 1% BSA-PBS. The coverslips were washed three times with 0.2% Triton X-100 PBS and incubated with 1:2000 Alexa Fluor 488 goat anti-rabbit IgG in 1% BSA-PBS for 1 h. The samples were washed three times with PBS and mounted on slides with ProLong Gold antifade reagent with DAPI. Staining was visualized with a Nikon 80i upright confocal microscope under 60× magnification. The antibody specificity of the anti-pIRF-3 antibody for immunocytochemistry was verified using macrophages from IRF-3−/− mice [39].

EMSA

RAW 264.7 cells were plated at 1.5 × 106 cells/well in six-well tissue-culture plates and incubated for 18–24 h before the start of the experiment. After the cells were treated, nuclear extracts were prepared using the NE-PER Nuclear and Cytoplasmic Extraction Reagent kit (Thermo Scientific, Waltham, MA, USA), according to the manufacturer's instructions. Biotin-labeled complementary oligonucleotides of the murine IFN-β promoter (5′-ATG ACA GAG GAA AAC TGA AAG GGA GAA CTG AAA GTG GGA AAT TCC T-3′; nt −151 to −106) were purchased from Integrated DNA Technologies (Coralville, IA, USA). Binding reactions were performed as described previously [40] using the LightShift Chemiluminescent EMSA kit (Thermo Scientific). Reactions were loaded onto native (6%) polyacrylamide gels and transferred to nylon (Biodyne B; Pierce, Thermo Scientific) membranes (45 min at 100 V). The migration of the labeled oligonucleotide was detected using HRP-conjugated streptavidin (LightShift Chemiluminescent EMSA kit), visualized on the EpiChemi II darkroom, and image processing and analyses were performed using ImageJ 1.33u.

ChIP

RAW 264.7 macrophages were plated at a density of 6 × 106 cells/well in 10-cm tissue-culture plates and incubated at 37°C, 24 h before treatment. After treatment, cells were processed using the EZ-Magna ChIP A kit (EMD Millipore, Billerica, MA, USA), according to the manufacturer's instructions. Protein/DNA complexes were immunoprecipitated with anti-IRF-3 (Santa Cruz Biotechnology; Cat. #SC-9082x), and purified, pulled-down DNA was amplified and quantified using qPCR with the following murine IFN-β primer sequence: F-GAA AGG GAG AAC TGA AAG, R-AGT GAG AAT GAT CTT CCT. IFN-β levels were normalized to input, and normal rabbit IgG (supplied in kit) was used as a control for nonspecific immunoprecipitation.

Statistical analysis

The Student's two-tailed paired t-test or Tukey-Kramer method (ANOVA) was used to calculate the statistical differences between samples. Significance levels were set at P values < 0.05.

RESULTS

P2RX7 agonists augment LPS-induced IFN-β expression in murine macrophages

P2RX7 signaling and IFN-β have been implicated in inflammatory responses to bacterial infection. Even though P2RX7 is known to regulate other LPS-induced inflammatory cytokines, the effect of purinergic receptor signaling on type I IFN production has not been described previously. To test the hypothesis that extracellular adenine nucleotides exert an influence on LPS-induced IFN-β in macrophages, we used a well-characterized murine macrophage cell line RAW 264.7. These cells were costimulated with LPS and ATP or the potent P2RX7 agonist BzATP, and IFN-β mRNA induction was quantified using real-time qPCR (Fig. 1A and B). In the presence of extracellular nucleotides and LPS, RAW 264.7 macrophages produced significantly higher levels of IFN-β mRNA than in response to LPS alone, supporting a role for P2RX7 in the augmentation of LPS-induced IFN-β expression. P2RX7 stimulation alone did not induce detectable IFN-β expression. The transcriptional effect of purinergic receptor signaling on IFN-β appeared relatively cytokine-specific, as BzATP did not augment mRNA for IL-6 or TNF-α mRNA production during this time frame (data not shown). The ability of BzATP to potentiate LPS-induced IFN-β mRNA expression was also observed in primary murine bone marrow-derived macrophages (Fig. 1C). To determine the kinetics of IFN-β mRNA expression, a time course was performed. After 1 h of treatment, BzATP attenuates LPS-induced IFN-β expression (P=0.006), but by 2 h, BzATP augments LPS-induced IFN-β mRNA expression, and this synergy is sustained up to 4 h (Fig. 1D). To examine whether the ability of P2RX7 to enhance LPS-induced IFN-β mRNA reflects increases in IFN-β protein production, IFN-β release was monitored. As shown in Fig. 1E, BzATP significantly increased the amount of IFN-β released from macrophages in response to LPS. To determine if P2RX7 activation is able to potentiate the expression of IFN-β induced by other TLRs, RAW 264.7 macrophages were stimulated with the TLR3 synthetic agonist Poly I:C in the absence or presence of BzATP. Although neither stimulus alone induces robust IFN-β, the combination of Poly I:C and BzATP induces significant IFN-β expression in macrophages (Fig. 1F), without causing cell death (data not shown).

Figure 1. P2RX7 agonists augment LPS-induced IFN-β expression in murine macrophages.

Figure 1.

RAW 264.7 macrophages (A, B, and D–F) or bone marrow-derived murine macrophages (C) were treated with 2.5 mM HEPES (vehicle), 1 mM ATP (A), 100 μM BzATP (E and F), 250 μM BzATP (B–D), LPS (1 μg/ml for A, B, and D and 10 ng/ml for C and E), 30 μg/ml Poly I:C (F), or nucleotide + TLR agonist, together for 3 h (A–C), 4 h (E), 6 h (F), or the time-points indicated (D). To determine IFN-β mRNA expression (A–D and F), cells were lysed in Trizol, and the levels of IFN-β mRNA were measured using qRT-PCR. Experimental results were normalized to 18S, and IFN-β mRNA expression was plotted as an average of experimental replicates (mean±sem). (E) Supernatants were spun down and analyzed for IFN-β via ELISA. The results from four independent experiments were summarized and represent relative IFN-β protein expression (mean±sem). ND, Not detected; *P < 0.05; **P < 0.01; ***P < 0.001. BzATP+PolyI:C vs. all other conditions. Each panel represents data from at least three independent experiments.

BzATP-induced enhancement of LPS-induced IFN-β mRNA requires functional P2RX7

Macrophages express the mRNA for many other purinergic receptors, in addition to P2RX7 [26]. Although BzATP is a potent agonist of P2RX7, BzATP also activates several other P2Rs at high concentrations (e.g., P2RX1 and P2RY11) [41]. Therefore, to ascertain further whether the enhancement of LPS-induced IFN-β mRNA expression by BzATP is mediated through endogenous P2RX7, a nonfunctional P2RX7 cell line was used. Specifically, “SF” RAW 264.7 macrophages possess an endogenous mutant P2RX7 gene containing a serine-to-phenylalanine mutation in the second transmembrane domain (SF) that confers attenuated P2RX7 protein expression and function [15, 37, 42, 43], while retaining the ability to respond to other stimuli to the same extent as the P2RX7 WT-expressing cells. As shown in Fig. 2, BzATP treatment of macrophages containing the SF P2RX7 mutant gene did not enhance LPS-induced IFN-β mRNA expression (P=0.62). Of note, LPS-induced IFN-β mRNA expression trended lower in SF versus WT P2RX7-expressing macrophages, but this difference was not statistically significant (P=0.26). These mutant P2RX7 RAW cells were not generally defective in responding to LPS, as they produce as much TNF-α in response to LPS as RAW macrophages expressing WT P2X7 (data not shown).

Figure 2. BzATP-induced enhancement of LPS-induced IFN-β mRNA requires functional P2RX7.

Figure 2.

RAW 264.7 macrophages endogenously expressing WT or nonfunctional P2RX7 (SF) were treated with vehicle, 1 μg/mL LPS, or 1 μg/ml LPS + 300 μM BzATP for 3 h. IFN-β and 18S mRNA levels were assessed by qPCR. The results from at least five independent experiments were averaged and represent relative IFN-β mRNA expression (mean±sem); *P = 0.01.

Inhibition of the MEK1/2 pathway prevents the P2RX7 agonist from enhancing LPS-induced IFN-β expression

To begin investigating the mechanism by which P2RX7 activation augments LPS-mediated IFN-β expression, kinase inhibitor experiments were conducted to elucidate further the signal transduction pathways that distinguish LPS-induced IFN-β from BzATP-enhanced, LPS-induced IFN-β. Induction of IFN-β by TLR4 agonists, such as LPS, requires the activation of the TRIF/TRAM pathway for complete activation of the enhanceosome [44]. To ascertain if the TRIF/TRAM pathway was involved in P2RX7 agonist-induced, increased IFN-β expression, we chose to inhibit IKKϵ and TBK-1, as the TRIF/TRAM complex activates IKKϵ/TBK-1 directly, and these kinases are known to regulate IRF-3 activation and subsequent IFN-β expression in response to LPS [45]. As expected, pretreatment of RAW 264.7 macrophages with the IKKϵ/TBK-1 inhibitor MRT67307 [46] inhibited LPS-induced IFN-β mRNA expression (Fig. 3A), as well as BzATP + LPS-induced IFN-β expression.

Figure 3. Effect of kinase inhibition on P2RX7 agonist-potentiated, LPS-induced IFN-β expression.

Figure 3.

RAW 264.7 macrophages were pretreated for 30 min (A) or 15 min (B and C) with vehicle (DMSO) or the indicated kinase inhibitor (10 μM). The cells were subsequently treated with vehicle (2.5 mM HEPES), 250 μM BzATP, 1 μg/ml LPS, or BzATP + LPS together for 3 h. IFN-β and 18S mRNA levels were assessed by qPCR. The results of at least three independent experiments were collated and are represented as relative IFN-β mRNA expression (with LPS treatment set to 1; mean±sem); *P < 0.05; **P < 0.01; ***P < 0.001.

It is known that LPS and P2RX7 agonists are able to induce the activation of p38 MAPK, and there is evidence that p38 activation is involved in bacterially induced IFN-β expression [47]. Thus, we examined the effect of the p38 inhibitor SB203580 on IFN-β expression. We found that p38 inhibition was able to significantly attenuate LPS-induced and LPS + BzATP-induced IFN-β expression (Fig. 3B).

Treatment of macrophages with P2RX7 agonists leads to the activation of the MAPKs, MEK1/2 and subsequently, ERK1/2 [37]. To determine if the MEK/ERK MAPK pathway participates in augmenting IFN-β expression after P2RX7 activation, the pharmacological MEK1/2 inhibitor, U0126, was used. U0126 previously has been shown to attenuate viral-induced IFN-β release via antagonism of IRF-3 activation [48]. When macrophages were pretreated with U0126, the ability of BzATP to enhance LPS-induced IFN-β expression was ablated (Fig. 3C). Interestingly, U0126 actually slightly enhanced LPS-induced IFN-β production, which has been observed previously [49, 50]. These data suggest that P2RX7 stimulation augments LPS-induced IFN-β via a MEK1/2-dependent pathway, external or accessory to canonical TLR4-IFN-β signaling.

Potentiation of LPS-induced IFN-β production by the P2RX7 agonist is observed at the promoter level

To decipher further how P2RX7 activation is enhancing the ability of LPS to induce IFN-β expression, the role of P2RX7 in activating the IFN-β promoter was investigated. Macrophages were transfected with a luciferase reporter construct, driven by the IFN-β promoter and treated with LPS ± BzATP for 6 h. As shown in Fig. 4A, treatment of macrophages with BzATP significantly enhances LPS-induced luciferase activity, supporting a role for P2RX7 in amplifying IFN-β expression at the transcriptional level. To investigate the effect of P2RX7 activation on the transcriptional enhancement of IFN-β expression, EMSAs were performed. Cotreatment of RAW 264.7 macrophages with LPS and BzATP significantly enhances nuclear protein binding to the IFN-β promoter (Fig. 4B). These data introduce the possibility that P2RX7 alters LPS-induced promoter activity through modulation of the ifnb1 enhanceosome.

Figure 4. Potentiation of LPS-induced IFN-β production by P2RX7 agonist is observed at the IFN-β promoter level.

Figure 4.

(A) The IFN-β promoter sequence was inserted into a luciferase reporter construct and transfected into RAW 264.7 macrophages. Cells were subsequently treated with vehicle (2.5 mM HEPES), 250 μM BzATP, 10 ng/ml LPS, or BzATP+LPS for 6 h. After treatment, cells were lysed, and luminescence was quantified. Data from three independent experiments were combined and graphed ± sem, with the luminescence values for the vehicle-treated samples set to 1; *P < 0.05; **P < 0.01; ***P < 0.001. (B) RAW 264.7 macrophages were treated with vehicle (2.5 mM HEPES), 250 μM BzATP, 1 μg/ml LPS, or BzATP+LPS for the times indicated, and nuclear fractions were collected for nonradioactive EMSA, using the biotin-labeled IFN-β promoter sequence to cause a gel shift. A representative gel shift is shown, and data from four independent experiments were collated and are represented as relative nuclear protein binding to the IFN-β promoter (with 30 min vehicle treatment set to 1; mean±sem); *P < 0.05; **P < 0.005 compared with time-matched, vehicle-treated band density; ϕP < 0.005 between LPS and LPS + Bz-treated band density.

Costimulation of murine macrophages with LPS and a P2RX7 agonist induces robust FosB expression, but it is not related to IFN-β augmentation

We discovered recently that activation of P2RX7 in macrophages leads to the robust expression of the AP-1 family transcription factor FosB, and this expression was downstream of the MEK/ERK signaling cascade [43]. Considering that the promoter of IFN-β has an AP-1-binding site that is important for the assembly of the enhanceosome, we investigated if P2RX7 agonist-induced FosB was upstream of IFN-β expression. We first examined the expression of FosB mRNA and protein after P2RX7 agonist and LPS treatment. As shown in Fig. 5A, treatment of macrophages with BzATP or LPS induces FosB mRNA expression, and the combination of BzATP and LPS induces an enhanced induction of FosB mRNA expression compared with either treatment alone. This enhanced expression of FosB after BzATP + LPS treatment was also observed at the protein level (Fig. 5B). To determine if this induction of FosB was upstream of IFN-β expression, siRNA directed against FosB (or control siRNA) was transfected into macrophages, followed by treatment with BzATP and LPS. Although the FosB siRNA and not the control siRNA was able to knock down BzATP/LPS-induced FosB expression at mRNA (Fig. 5B) and protein levels (data not shown), FosB knockdown had no effect on BzATP/LPS-induced IFN-β expression (Fig. 5C).

Figure 5. Costimulation of murine macrophages with LPS and a P2RX7 agonist induces FosB expression but is not related to IFN-β release.

Figure 5.

RAW 264.7 macrophages were treated with 2.5 mM HEPES (vehicle), 250 μM BzATP, 1 μg/ml (and 0.1 μg/ml for B), LPS, or BzATP + LPS together for the time-points indicated. (A) Cells were lysed, and FosB mRNA levels were measured using qPCR. Experimental results were normalized to 18S. Data are representative of two independent experiments. (B) Cells were lysed in sample buffer after treatment and immunoblotted for FosB and anti-β-tubulin (loading control). Data are representative of three independent experiments. (C) Twenty-four hours prior to 3 h treatment with LPS ± BzATP, cells were transfected with control siRNA or siRNA directed toward murine FosB, and levels of FosB and IFN-β mRNA were measured. The results of three independent experiments were collated and are represented as relative FosB or IFN-β mRNA expression; *P = 0.01.

Activation of IRF-3 is up-regulated after P2RX7 activation

IRF-3 is a transcription factor critical for the activation of the IFN-β enhanceosome [51, 52]. Although the ability of LPS to induce IRF-3 activation is documented [52, 53], the ability of the P2RX7 agonists to activate this transcription factor is unknown. To detect IRF-3 activation, macrophages were treated with LPS ± BzATP, and pIRF-3 was monitored. In Fig. 6A, treated macrophages were subjected to immunoblotting using an antibody against the S396 pIRF-3, which at S396 was not reliably detectable after BzATP treatment. However, BzATP treatment was able to significantly enhance LPS-induced pIRF-3 (P=0.009; n=3; Fig. 6A). To determine whether P2RX7 agonist-induced enhancement of pIRF-3 translated into increased IRF-3 occupancy on the IFN-β promoter, ChIP was performed. As shown in Fig. 6B, treatment of macrophages with LPS + BzATP induces increased IRF-3 occupancy on the IFN-β promoter over LPS treatment, alone after 1 h and 2 h of treatment. The results demonstrate for the first time that stimulation of P2RX7 is able to activate IRF-3 and may explain the enhanced IFN-β expression.

Figure 6. Treatment of RAW 264.7 macrophages with a P2RX7 agonist promotes pIRF-3.

Figure 6.

RAW 264.7 macrophages were treated with 2.5 mM HEPES (vehicle), 250 μM BzATP, 1 μg/ml LPS (A), 100 ng/ml LPS (B), or BzATP + LPS together for 2 h (A) or the times indicated (B). (A) Lysed cells were immunoblotted for pIRF-3 and total IRF-3 (loading control). A representative immunoblot displaying pIRF-3/IRF-3 is shown. The results of three independent experiments were collated and are represented as relative pIRF-3 (normalized to IRF-3; mean±sem); *P < 0.01. (B) Cross-linked protein/DNA complexes were immunoprecipitated with an IRF-3 antibody, and the collected DNA was amplified with primers against the IFN-β promoter. IFN-β levels were normalized to their respective input control, and the results from three independent experiments were averaged and plotted (mean±sem); *P < 0.05; **P < 0.005.

To determine which signaling mediators were upstream of BzATP-mediated IRF-3 activation, macrophages were pretreated with MRT67307, SB203580, and U0126 before BzATP treatment, and S386 pIRF-3 was monitored by immunofluorescence. Interestingly, BzATP and LPS treatment appeared to enhance pIRF-3 at this residue (Fig. 7A). Furthermore, BzATP-induced pIRF-3 S386 was decreased in the presence off all of the inhibitors. When these same inhibitors were used to measure the phosphorylation of S396 by immunoblotting, MRT67307 was able to significantly attenuate LPS and LPS + BzATP-induced pIRF-3, but SB203580 was not able to inhibit either. Consistent with Fig. 3C, U0126 was only able to attenuate LPS + BzATP but not LPS-induced pIRF-3 (Fig. 7B). Cell viability assays were performed as described previously [43] to confirm that these effects were not a result of toxicity (data not shown). Together, these data suggest that P2RX7 signals through MAPK pathways and TBK-1/IKKϵ to activate IRF-3 and thus, augment LPS-induced IFN-β expression.

Figure 7. Effect of kinase inhibition on pIRF-3 expression.

Figure 7.

RAW 264.7 macrophages were pretreated for 30 min [for MRT67307 (MRT)] or 15 min [for SB203580 (SB) or U0126] with vehicle (DMSO) or 10 μM of the indicated kinase inhibitor. The cells were subsequently treated with vehicle (2.5 mM HEPES), 250 μM BzATP, 1 μg/ml LPS, or BzATP + LPS together for 2 h. (A) Cells were fixed and stained for pIRF-3 (green); DAPI stain (nuclei; blue). (B) Cells were lysed in sample buffer after treatment and immunoblotted for pIRF-3 and total β-actin (loading control). A representative immunoblot displaying pIRF-3 is shown. The results of at least three independent experiments were collated and are represented as relative pIRF-3 (normalized to β-actin; mean±sem); *P < 0.05.

DISCUSSION

This present study is the first to demonstrate that LPS-induced IFN-β is enhanced in macrophages upon activation of P2RX7, at the mRNA and protein level. The potentiation of IFN-β expression was not exclusive to TLR4 agonists, as the induction of IFN-β expression by a TLR3 agonist was also augmented in the presence of the P2RX7 agonist BzATP. These data suggest that the P2RX7 agonist-induced enhancement of LPS-triggered IFN-β expression occurs at the promoter level of the IFN-β gene and is related to an increase in active IRF-3.

Type I IFNs can exert biological effects at low levels of expression [54]. For instance, low levels of IFN-α/β prime cells to respond to other cytokines, such as IFN-γ and IL-6, possibly by recruiting commonly used signaling molecules to the cell membrane in proximity to other cytokine receptor complexes [54]. Therefore, the increased IFN-β released from macrophages after LPS and P2RX7 cotreatment is likely to have a biological impact. It has been shown that IFN-β plays a role in mediating anti-inflammatory IL-10 expression in macrophages [55]. To begin to determine potential endpoints that could be downstream of LPS + P2RX7 agonist cotreatment, the induction of IL-10 mRNA expression in macrophages was measured. Although LPS and BzATP alone were able to induce increased IL-10 mRNA expression, the combination of these treatments was additive at most and not synergistic compared with the induction of IFN-β (Supplemental Fig. 1).

This study demonstrates that P2RX7 agonist-enhanced, LPS-induced IFN-β expression is abrogated by multiple kinase inhibitors (Fig. 3), but the effect of the MEK1/2 antagonist U0126 (Fig. 3C) distinguished LPS-induced versus LPS + P2RX7 agonist-induced IFN-β expression, suggesting that the MEK/ERK1/2 cascade is uniquely involved in the ability of P2RX7 to up-regulate the expression of IFN-β in macrophages. It is known that treatment of macrophages with LPS leads to the activation of ERK1/2, and this has been shown to be MyD88- but not TRIF-dependent [56]. As TLR4-induced IFN-β expression occurs via a MyD88-independent pathway, it is not surprising that LPS-induced ERK activation is not involved in IFN-β expression (Fig. 3C) and has been described by others [57].

As a result of the importance of the AP-1 component of the IFN-β enhanceosome and our recent observation that P2RX7 stimulation induces a robust activation of the AP-1 protein FosB (which is downstream of P2RX7-induced MEK/ERK activation), we first hypothesized that FosB may be involved in the increased IFN-β expression observed after LPS treatment. Although our data show that cotreatment of macrophages with the LPS and P2RX7 agonist induces FosB mRNA and protein expression (Fig. 5), our RNA interference data do not support a role for FosB in IFN-β expression. Therefore, our data raised the possibility that another U0126-dependent transcription factor is contributing to P2RX7-LPS synergistic IFN-β induction.

It has been noted previously by our group [13] and others [27, 58] that P2RX7 stimulation leads to the activation of NF-κB, and this could also be a mechanism by which nucleotide signaling enhances LPS-induced IFN-β expression, as there are NF-κB-binding elements (for a p50/p65 NF-κB heterodimer) in the enhanceosome region of the IFN-β promoter [59]. In macrophages, LPS induces the formation of p65/p50 NF-κB heterodimers, whereas ATP induces p65 NF-κB homodimers [58, 60]. Intriguingly, p50 and p105 NF-κB subunits, which lack transcriptional activation domains, inhibit LPS-induced IFN-β expression, purportedly by activating the MEK/ERK signaling cascade and preventing IRF-3 recruitment to the IFN-β promoter [60, 61]. We observed previously that U0126 pretreatment of macrophages leads to a sustained loss of IκBα expression in response to LPS [13]. As IκBα degradation is necessary for NF-κB translocation into the nucleus and subsequent gene activation, the sustained loss of IκBα expression suggests that NF-κB is transcriptionally active longer, and this may account for the increase in LPS-induced IFN-β expression upon U0126 pretreatment, shown in Fig. 4. However, as U0126 ablated the enhancement of LPS-induced IFN-β expression induced by P2RX7 activation, these data suggest that the U0126-sensitive portion of P2RX7 signaling may not proceed via NF-κB to enhance IFN-β expression.

Our study is the first to demonstrate that P2RX7 activation alone induces the pIRF-3 at serine 386, which is essential for the IRF-3 interaction with the transcription coactivator CREB-binding protein and subsequent transcriptional activation [62]. Our Western blot data also support a role for P2RX7 in enhancing LPS-dependent phosphorylation of serine 396 on IRF-3. IRF-3 is known to have two activation clusters, with one cluster including S386 and the other including S396, and there are data to support a role for phosphorylation of both of these serines in IRF-3-initiated gene transcription [6365]. IRF-3 plays a critical role in the activation of the IFN-β enhanceosome induced by bacterial and viral stimuli [36, 66]. Therefore, it is plausible that the enhancement of IFN-β by P2RX7 agonists is attributed to an increase in pIRF-3. Increased IRF-3 occupancy on the IFN-β promoter under LPS and BzATP costimulation (Fig. 6B) further supports a role for enhanced IRF-3 transcriptional activation in elevated LPS-induced IFN-β expression after P2RX7 activation in macrophages. The differential sensitivity of LPS- and BzATP-induced pIRF-3 to the MEK1/2 inhibitor U0126 suggests that P2RX7 stimulation supplements TLR4-triggered IFN-β induction by MAPK-dependent activation of IRF-3.

Previous studies have supported a role for purinergic receptor stimulation and IFN-β in inflammatory responses to pathogenic bacteria. One could envision a scenario in which macrophages encounter TLR ligands (exogenous or endogenous) in the setting of tissue damage and platelet degranulation, where extracellular nucleotides would be abundant. P2RX7 stimulation would then act as a costimulatory “danger signal” for macrophages that augments subsequent cytokine production. Our studies reveal that these two systems may be tightly linked. These findings may also have important physiological ramifications in the context of viral infections, where large amounts of type I IFNs are produced, and substantial ATP is released after lytic cell death [3, 67]. Interestingly, low P2RX7 activity has been associated with an increased risk of viral-induced/associated asthma exacerbations [68]. Considering the established antiviral properties of type I IFNs and the observation that IFN-β−/− mice are more susceptible to viral infection [69], it is possible that decreased P2RX7 activity would lead to lower IFN-β levels during a viral infection and consequently, less viral clearance. Our observation that P2RX7 activation potentiates TLR3 agonist-induced IFN-β expression (Fig. 1F) supports a role for extracellular nucleotides in modulating host responses to viral exposure that trigger the activation of this TLR. Further work into the role of P2RX7 in the context of antiviral signaling in macrophages is warranted.

The understanding of the role of extracellular nucleotides in amplifying the inflammatory response has broad implications for multiple diseases, including cancer, atherosclerosis, asthma, and autoimmune diseases. As P2RX7 is involved in the synthesis and release of multiple inflammatory mediators, it has been hypothesized that the production of a pharmacological inhibitor against P2RX7 will reduce the magnitude of an inflammatory response under physiological conditions where an immune reaction is unfavorable. As such, small molecule inhibitors of P2RX7 have been in clinical trials as treatment for rheumatoid arthritis [811, 28, 70]. IFN-β is present in the synovial fluid of arthritic patients [71] and based on our observations, may be, in part, a result of the activation of P2RX7 in the inflammed joints.

In summary, these studies suggest that P2RX7 signaling may act as a danger signal, by augmenting TLR4 agonist-induced IFN-β expression in macrophages. Our data also begin to elucidate the mechanisms by which purine nucleotides regulate the expression of this proinflammatory, pleiotropic cytokine. To our knowledge, this is the first investigation into the mechanism of how P2RX7 signaling modulates type I IFN responses.

Supplementary Material

Supplemental Data

ACKNOWLEDGMENTS

This work was supported by U.S. National Institutes of Health grants K08 AI081045, U19 AI070503, RO1 HL069116, and RO1 HL115118 and an American Heart Association Midwest Affiliate Postdoctoral Fellowship.

This manuscript is dedicated to the memory of our colleague Dr. Paul Bertics, who made many contributions to this publication prior to his untimely death in December 2011. The authors thank the former members of the Bertics lab for their critical review of the manuscript.

The online version of this paper, found at www.jleukbio.org, includes supplemental information.

−/−
deficient
BzATP
benzoyl ATP
ChIP
chromatin immunoprecipitation
F
forward
IRF-3
IFN regulatory factor-3
P2RX7
purinergic receptor P2X, ligand-gated ion channel 7
pIRF
phosphorylated IRF
Poly I:C
polyinosinic:polycytidylic acid
q
quantitative
R
reverse
SF
S342F
siRNA
small interfering RNA
TBK-1
TANK-binding kinase-1
TRAM
Toll/IL-1R-domain-containing adaptor-inducing IFN-β-related adapter molecule
TRIF
Toll/IL-1R-domain-containing adaptor-inducing IFN-β

AUTHORSHIP

All authors contributed to the conception and design of the study, interpretation of the data, and revision of the manuscript and provided final approval of the submitted version. P.J.B., L.C.D., and J.A.S. were responsible for obtaining funding. M.L.G. drafted the manuscript. M.L.G., Y.P.L., L.Y.L., L.Z., A.G.G., and J.B.B. contributed to data acquisition. M.L.G. and J.A.S. completed data analysis and interpretation.

REFERENCES

  • 1. Burnstock G. (2006) Pathophysiology and therapeutic potential of purinergic signaling. Pharmacol. Rev. 58, 58–86 [DOI] [PubMed] [Google Scholar]
  • 2. Di Virgilio F. (2007) Purinergic signalling in the immune system. A brief update. Purinergic Signal. 3, 1–3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Khakh B. S., North R. A. (2006) P2X receptors as cell-surface ATP sensors in health and disease. Nature 442, 527–532 [DOI] [PubMed] [Google Scholar]
  • 4. Di Virgilio F. (2005) Purinergic mechanism in the immune system: a signal of danger for dendritic cells. Purinergic Signal. 1, 205–209 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Lister M. F., Sharkey J., Sawatzky D. A., Hodgkiss J. P., Davidson D. J., Rossi A. G., Finlayson K. (2007) The role of the purinergic P2X7 receptor in inflammation. J. Inflamm. (Lond.) 4, 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Sluyter R., Stokes L. (2011) Significance of P2X7 receptor variants to human health and disease. Recent Pat. DNA Gene Seq. 5, 41–54 [DOI] [PubMed] [Google Scholar]
  • 7. Labasi J. M., Petrushova N., Donovan C., McCurdy S., Lira P., Payette M. M., Brissette W., Wicks J. R., Audoly L., Gabel C. A. (2002) Absence of the P2X7 receptor alters leukocyte function and attenuates an inflammatory response. J. Immunol. 168, 6436–6445 [DOI] [PubMed] [Google Scholar]
  • 8. Dell'Antonio G., Quattrini A., Cin E. D., Fulgenzi A., Ferrero M. E. (2002) Relief of inflammatory pain in rats by local use of the selective P2X7 ATP receptor inhibitor, oxidized ATP. Arthritis Rheum. 46, 3378–3385 [DOI] [PubMed] [Google Scholar]
  • 9. Dell'Antonio G., Quattrini A., Dal Cin E., Fulgenzi A., Ferrero M. E. (2002) Antinociceptive effect of a new P(2Z)/P2X7 antagonist, oxidized ATP, in arthritic rats. Neurosci. Lett. 327, 87–90 [DOI] [PubMed] [Google Scholar]
  • 10. Peng W., Cotrina M. L., Han X., Yu H., Bekar L., Blum L., Takano T., Tian G. F., Goldman S. A., Nedergaard M. (2009) Systemic administration of an antagonist of the ATP-sensitive receptor P2X7 improves recovery after spinal cord injury. Proc. Natl. Acad. Sci. USA 106, 12489–12493 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Wang L., Jacobsen S. E., Bengtsson A., Erlinge D. (2004) P2 receptor mRNA expression profiles in human lymphocytes, monocytes and CD34+ stem and progenitor cells. BMC Immunol. 5, 16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Portales-Cervantes L., Nino-Moreno P., Doniz-Padilla L., Baranda-Candido L., Garcia-Hernandez M., Salgado-Bustamante M., Gonzalez-Amaro R., Portales-Perez D. (2010) Expression and function of the P2X(7) purinergic receptor in patients with systemic lupus erythematosus and rheumatoid arthritis. Hum. Immunol. 71, 818–825 [DOI] [PubMed] [Google Scholar]
  • 13. Aga M., Watters J. J., Pfeiffer Z. A., Wiepz G. J., Sommer J. A., Bertics P. J. (2004) Evidence for nucleotide receptor modulation of cross talk between MAP kinase and NF-κ B signaling pathways in murine RAW 264.7 macrophages. Am. J. Physiol. Cell. Physiol. 286, C923–C930 [DOI] [PubMed] [Google Scholar]
  • 14. Guerra A. N., Gavala M. L., Chung H. S., Bertics P. J. (2007) Nucleotide receptor signalling and the generation of reactive oxygen species. Purinergic Signal. 3, 39–51 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Pfeiffer Z. A., Guerra A. N., Hill L. M., Gavala M. L., Prabhu U., Aga M., Hall D. J., Bertics P. J. (2007) Nucleotide receptor signaling in murine macrophages is linked to reactive oxygen species generation. Free Radic. Biol. Med. 42, 1506–1516 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Solle M., Labasi J., Perregaux D. G., Stam E., Petrushova N., Koller B. H., Griffiths R. J., Gabel C. A. (2001) Altered cytokine production in mice lacking P2X(7) receptors. J. Biol. Chem. 276, 125–132 [DOI] [PubMed] [Google Scholar]
  • 17. Watters J., Sommer J., Fisette P., Pfeiffer Z., Aga M., Prabhu U., Guerra A., Denlinger L., Bertics P. (2001) P2X7 nucleotide receptor: modulation of LPS-induced macrophage signaling and mediator production. Drug Dev. Res. 53, 91–104 [Google Scholar]
  • 18. Proctor R. A., Denlinger L. C., Leventhal P. S., Daugherty S. K., van de Loo J. W., Tanke T., Firestein G. S., Bertics P. J. (1994) Protection of mice from endotoxic death by 2-methylthio-ATP. Proc. Natl. Acad. Sci. USA 91, 6017–6020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Hu Y., Fisette P. L., Denlinger L. C., Guadarrama A. G., Sommer J. A., Proctor R. A., Bertics P. J. (1998) Purinergic receptor modulation of lipopolysaccharide signaling and inducible nitric-oxide synthase expression in RAW 264.7 macrophages. J. Biol. Chem. 273, 27170–27175 [DOI] [PubMed] [Google Scholar]
  • 20. Lenertz L. Y., Gavala M. L., Hill L. M., Bertics P. J. (2009) Cell signaling via the P2X(7) nucleotide receptor: linkage to ROS production, gene transcription, and receptor trafficking. Purinergic Signal. 5, 175–187 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Tonetti M., Sturla L., Giovine M., Benatti U., De Flora A. (1995) Extracellular ATP enhances mRNA levels of nitric oxide synthase and TNF-α in lipopolysaccharide-treated RAW 264.7 murine macrophages. Biochem. Biophys. Res. Commun. 214, 125–130 [DOI] [PubMed] [Google Scholar]
  • 22. Ferrari D., Pizzirani C., Adinolfi E., Lemoli R. M., Curti A., Idzko M., Panther E., Di Virgilio F. (2006) The P2X7 receptor: a key player in IL-1 processing and release. J. Immunol. 176, 3877–3883 [DOI] [PubMed] [Google Scholar]
  • 23. Denlinger L. C., Angelini G., Schell K., Green D. N., Guadarrama A. G., Prabhu U., Coursin D. B., Bertics P. J., Hogan K. (2005) Detection of human P2X7 nucleotide receptor polymorphisms by a novel monocyte pore assay predictive of alterations in lipopolysaccharide-induced cytokine production. J. Immunol. 174, 4424–4431 [DOI] [PubMed] [Google Scholar]
  • 24. Andrei C., Margiocco P., Poggi A., Lotti L. V., Torrisi M. R., Rubartelli A. (2004) Phospholipases C and A2 control lysosome-mediated IL-1 β secretion: implications for inflammatory processes. Proc. Natl. Acad. Sci. USA 101, 9745–9750 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. MacKenzie A., Wilson H. L., Kiss-Toth E., Dower S. K., North R. A., Surprenant A. (2001) Rapid secretion of interleukin-1beta by microvesicle shedding. Immunity 15, 825–835 [DOI] [PubMed] [Google Scholar]
  • 26. Guerra A. N., Fisette P. L., Pfeiffer Z. A., Quinchia-Rios B. H., Prabhu U., Aga M., Denlinger L. C., Guadarrama A. G., Abozeid S., Sommer J. A., Proctor R. A., Bertics P. J. (2003) Purinergic receptor regulation of LPS-induced signaling and pathophysiology. J. Endotoxin Res. 9, 256–263 [DOI] [PubMed] [Google Scholar]
  • 27. Armstrong S., Korcok J., Sims S. M., Dixon S. J. (2007) Activation of transcription factors by extracellular nucleotides in immune and related cell types. Purinergic Signal. 3, 59–69 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Lenertz L. Y., Gavala M. L., Zhu Y., Bertics P. J. (2011) Transcriptional control mechanisms associated with the nucleotide receptor P2X7, a critical regulator of immunologic, osteogenic, and neurologic functions. Immunol. Res. 50, 22–38 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Bogdan C. (2000) The function of type I interferons in antimicrobial immunity. Curr. Opin. Immunol. 12, 419–424 [DOI] [PubMed] [Google Scholar]
  • 30. Barton G. M., Medzhitov R. (2003) Linking Toll-like receptors to IFN-α/β expression. Nat. Immunol. 4, 432–433 [DOI] [PubMed] [Google Scholar]
  • 31. Gessani S., Belardelli F., Pecorelli A., Puddu P., Baglioni C. (1989) Bacterial lipopolysaccharide and γ interferon induce transcription of β interferon mRNA and interferon secretion in murine macrophages. J. Virol. 63, 2785–2789 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Seong S. Y., Matzinger P. (2004) Hydrophobicity: an ancient damage-associated molecular pattern that initiates innate immune responses. Nat. Rev. Immunol. 4, 469–478 [DOI] [PubMed] [Google Scholar]
  • 33. Mancuso G., Midiri A., Biondo C., Beninati C., Zummo S., Galbo R., Tomasello F., Gambuzza M., Macri G., Ruggeri A., Leanderson T., Teti G. (2007) Type I IFN signaling is crucial for host resistance against different species of pathogenic bacteria. J. Immunol. 178, 3126–3133 [DOI] [PubMed] [Google Scholar]
  • 34. Karaghiosoff M., Steinborn R., Kovarik P., Kriegshauser G., Baccarini M., Donabauer B., Reichart U., Kolbe T., Bogdan C., Leanderson T., Levy D., Decker T., Muller M. (2003) Central role for type I interferons and Tyk2 in lipopolysaccharide-induced endotoxin shock. Nat. Immunol. 4, 471–477 [DOI] [PubMed] [Google Scholar]
  • 35. Kim J. H., Kim S. J., Lee I. S., Lee M. S., Uematsu S., Akira S., Oh K. I. (2009) Bacterial endotoxin induces the release of high mobility group box 1 via the IFN-β signaling pathway. J. Immunol. 182, 2458–2466 [DOI] [PubMed] [Google Scholar]
  • 36. Doyle S., Vaidya S., O'Connell R., Dadgostar H., Dempsey P., Wu T., Rao G., Sun R., Haberland M., Modlin R., Cheng G. (2002) IRF3 mediates a TLR3/TLR4-specific antiviral gene program. Immunity 17, 251–263 [DOI] [PubMed] [Google Scholar]
  • 37. Gavala M. L., Pfeiffer Z. A., Bertics P. J. (2008) The nucleotide receptor P2RX7 mediates ATP-induced CREB activation in human and murine monocytic cells. J. Leukoc. Biol. 84, 1159–1171 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Takeshita S., Kaji K., Kudo A. (2000) Identification and characterization of the new osteoclast progenitor with macrophage phenotypes being able to differentiate into mature osteoclasts. J. Bone Miner. Res. 15, 1477–1488 [DOI] [PubMed] [Google Scholar]
  • 39. Liu Y. P., Zeng L., Tian A., Bomkamp A., Rivera D., Gutman D., Barber G. N., Olson J. K., Smith J. A. (2012) Endoplasmic reticulum stress regulates the innate immunity critical transcription factor IRF3. J. Immunol. 189, 4630–4639 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Zhao W., Wang L., Zhang M., Wang P., Zhang L., Yuan C., Qi J., Qiao Y., Kuo P. C., Gao C. (2011) Peroxisome proliferator-activated receptor γ negatively regulates IFN-β production in Toll-like receptor (TLR) 3- and TLR4-stimulated macrophages by preventing interferon regulatory factor 3 binding to the IFN-β promoter. J. Biol. Chem. 286, 5519–5528 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Di Virgilio F., Baricordi O. R., Romagnoli R., Baraldi P. G. (2005) Leukocyte P2 receptors: a novel target for anti-inflammatory and anti-tumor therapy. Curr. Drug Targets Cardiovasc. Haematol. Disord. 5, 85–99 [DOI] [PubMed] [Google Scholar]
  • 42. Denlinger L. C., Sommer J. A., Parker K., Gudipaty L., Fisette P. L., Watters J. W., Proctor R. A., Dubyak G. R., Bertics P. J. (2003) Mutation of a dibasic amino acid motif within the C terminus of the P2X7 nucleotide receptor results in trafficking defects and impaired function. J. Immunol. 171, 1304–1311 [DOI] [PubMed] [Google Scholar]
  • 43. Gavala M. L., Hill L. M., Lenertz L. Y., Karta M. R., Bertics P. J. (2010) Activation of the transcription factor FosB/activating protein-1 (AP-1) is a prominent downstream signal of the extracellular nucleotide receptor P2RX7 in monocytic and osteoblastic cells. J. Biol. Chem. 285, 34288–34298 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Yamamoto M., Sato S., Mori K., Hoshino K., Takeuchi O., Takeda K., Akira S. (2002) Cutting edge: a novel Toll/IL-1 receptor domain-containing adapter that preferentially activates the IFN-β promoter in the Toll-like receptor signaling. J. Immunol. 169, 6668–6672 [DOI] [PubMed] [Google Scholar]
  • 45. Fitzgerald K. A., Rowe D. C., Barnes B. J., Caffrey D. R., Visintin A., Latz E., Monks B., Pitha P. M., Golenbock D. T. (2003) LPS-TLR4 signaling to IRF-3/7 and NF-κB involves the Toll adapters TRAM and TRIF. J. Exp. Med. 198, 1043–1055 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Clark K., Peggie M., Plater L., Sorcek R. J., Young E. R., Madwed J. B., Hough J., McIver E. G., Cohen P. (2011) Novel cross-talk within the IKK family controls innate immunity. Biocheml. J. 434, 93–104 [DOI] [PubMed] [Google Scholar]
  • 47. Reimer T., Schweizer M., Jungi T. W. (2007) Stimulation-specific contribution of p38 and JNK to IFN-β gene expression in human macrophages. J. Interferon Cytokine Res. 27, 751–755 [DOI] [PubMed] [Google Scholar]
  • 48. Wang F., Ma Y., Barrett J. W., Gao X., Loh J., Barton E., Virgin H. W., McFadden G. (2004) Disruption of Erk-dependent type I interferon induction breaks the myxoma virus species barrier. Nat. Immunol. 5, 1266–1274 [DOI] [PubMed] [Google Scholar]
  • 49. Kaiser F., Cook D., Papoutsopoulou S., Rajsbaum R., Wu X., Yang H. T., Grant S., Ricciardi-Castagnoli P., Tsichlis P. N., Ley S. C., O'Garra A. (2009) TPL-2 negatively regulates interferon-β production in macrophages and myeloid dendritic cells. J. Exp. Med. 206, 1863–1871 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Yang H. T., Wang Y., Zhao X., Demissie E., Papoutsopoulou S., Mambole A., O'Garra A., Tomczak M. F., Erdman S. E., Fox J. G., Ley S. C., Horwitz B. H. (2011) NF-κB1 inhibits TLR-induced IFN-β production in macrophages through TPL-2-dependent ERK activation. J. Immunol. 186, 1989–1996 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Servant M. J., Tenoever B., Lin R. (2002) Overlapping and distinct mechanisms regulating IRF-3 and IRF-7 function. J. Interferon Cytokine Res. 22, 49–58 [DOI] [PubMed] [Google Scholar]
  • 52. Solis M., Romieu-Mourez R., Goubau D., Grandvaux N., Mesplede T., Julkunen I., Nardin A., Salcedo M., Hiscott J. (2007) Involvement of TBK1 and IKKϵ in lipopolysaccharide-induced activation of the interferon response in primary human macrophages. Eur. J. Immunol. 37, 528–539 [DOI] [PubMed] [Google Scholar]
  • 53. Reimer T., Brcic M., Schweizer M., Jungi T. W. (2008) poly(I: C) and LPS induce distinct IRF3 and NF-κB signaling during type-I IFN and TNF responses in human macrophages. J. Leukoc. Biol. 83, 1249–1257 [DOI] [PubMed] [Google Scholar]
  • 54. Taniguchi T., Takaoka A. (2001) A weak signal for strong responses: interferon-α/β revisited. Nat. Rev. Mol. Cell Biol. 2, 378–386 [DOI] [PubMed] [Google Scholar]
  • 55. Chang E. Y., Guo B., Doyle S. E., Cheng G. (2007) Cutting edge: involvement of the type I IFN production and signaling pathway in lipopolysaccharide-induced IL-10 production. J. Immunol. 178, 6705–6709 [DOI] [PubMed] [Google Scholar]
  • 56. Watts B. A., III, George T., Sherwood E. R., Good D. W. (2011) Basolateral LPS inhibits NHE3 and HCOFormula absorption through TLR4/MyD88-dependent ERK activation in medullary thick ascending limb. Am. J. Physiol. Cell. Physiol. 301, C1296–C1306 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Koide N., Ito H., Mu M. M., Sugiyama T., Hassan F., Islam S., Mori I., Yoshida T., Yokochi T. (2005) Inhibition of extracellular signal-regulated kinase 1/2 augments nitric oxide production in lipopolysaccharide-stimulated RAW264.7 macrophage cells. FEMS Immunol. Med. Microbiol. 45, 213–219 [DOI] [PubMed] [Google Scholar]
  • 58. Ferrari D., Chiozzi P., Falzoni S., Dal Susino M., Collo G., Buell G., Di Virgilio F. (1997) ATP-mediated cytotoxicity in microglial cells. Neuropharmacology 36, 1295–1301 [DOI] [PubMed] [Google Scholar]
  • 59. Edwards M. R., Slater L., Johnston S. L. (2007) Signalling pathways mediating type I interferon gene expression. Microbes Infect. 9, 1245–1251 [DOI] [PubMed] [Google Scholar]
  • 60. Cheng C. S., Feldman K. E., Lee J., Verma S., Huang D. B., Huynh K., Chang M., Ponomarenko J. V., Sun S. C., Benedict C. A., Ghosh G., Hoffmann A. (2011) The specificity of innate immune responses is enforced by repression of interferon response elements by NF-κB p50. Sci. Signl. 4, ra11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Zhao X., Ross E. J., Wang Y., Horwitz B. H. (2012) Nfkb1 inhibits LPS-induced IFN-β and IL-12 p40 production in macrophages by distinct mechanisms. PloS One 7, e32811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Takahasi K., Horiuchi M., Fujii K., Nakamura S., Noda N. N., Yoneyama M., Fujita T., Inagaki F. (2010) Ser386 phosphorylation of transcription factor IRF-3 induces dimerization and association with CBP/p300 without overall conformational change. Genes Cells 15, 901–910 [DOI] [PubMed] [Google Scholar]
  • 63. Chen W., Srinath H., Lam S. S., Schiffer C. A., Royer W. E., Jr., Lin K. (2008) Contribution of Ser386 and Ser396 to activation of interferon regulatory factor 3. J. Mol. Biol. 379, 251–260 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Mori M., Yoneyama M., Ito T., Takahashi K., Inagaki F., Fujita T. (2004) Identification of Ser-386 of interferon regulatory factor 3 as critical target for inducible phosphorylation that determines activation. J. Biol. Chem. 279, 9698–9702 [DOI] [PubMed] [Google Scholar]
  • 65. Servant M. J., Grandvaux N., tenOever B. R., Duguay D., Lin R., Hiscott J. (2003) Identification of the minimal phosphoacceptor site required for in vivo activation of interferon regulatory factor 3 in response to virus and double-stranded RNA. J. Biol. Chem. 278, 9441–9447 [DOI] [PubMed] [Google Scholar]
  • 66. Sakaguchi S., Negishi H., Asagiri M., Nakajima C., Mizutani T., Takaoka A., Honda K., Taniguchi T. (2003) Essential role of IRF-3 in lipopolysaccharide-induced interferon-β gene expression and endotoxin shock. Biochem. Biophys. Res. Commun. 306, 860–866 [DOI] [PubMed] [Google Scholar]
  • 67. Shi L., Manthei D. M., Guadarrama A. G., Lenertz L. Y., Denlinger L. C. (2012) Rhinovirus-induced IL-1β release from bronchial epithelial cells is independent of functional P2X7. Am. J. Respir. Cell Mol. Biol. 47, 363–371 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Denlinger L. C., Shi L., Guadarrama A., Schell K., Green D., Morrin A., Hogan K., Sorkness R. L., Busse W. W., Gern J. E. (2009) Attenuated P2X7 pore function as a risk factor for virus-induced loss of asthma control. Am. J. Respir. Crit. Care Med. 179, 265–270 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Deonarain R., Alcami A., Alexiou M., Dallman M. J., Gewert D. R., Porter A. C. (2000) Impaired antiviral response and α/β interferon induction in mice lacking β interferon. J. Virol. 74, 3404–3409 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Guile S. D., Alcaraz L., Birkinshaw T. N., Bowers K. C., Ebden M. R., Furber M., Stocks M. J. (2009) Antagonists of the P2X(7) receptor. From lead identification to drug development. J. Med. Chem. 52, 3123–3141 [DOI] [PubMed] [Google Scholar]
  • 71. Van Holten J., Smeets T. J., Blankert P., Tak P. P. (2005) Expression of interferon β in synovial tissue from patients with rheumatoid arthritis: comparison with patients with osteoarthritis and reactive arthritis. Ann. Rheum. Dis. 64, 1780–1782 [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplemental Data

Articles from Journal of Leukocyte Biology are provided here courtesy of The Society for Leukocyte Biology

RESOURCES