Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Cytosolic Phospholipase A2α and Eicosanoids Regulate Expression of Genes in Macrophages Involved in Host Defense and Inflammation

  • Saritha Suram,

    Affiliation Department of Pediatrics, National Jewish Health, Denver, Colorado, United States of America

  • Lori J. Silveira,

    Affiliation Division of Biostatistics and Bioinformatics, National Jewish Health, Denver, Colorado, United States of America

  • Spencer Mahaffey,

    Affiliation Department of Pediatrics, National Jewish Health, Denver, Colorado, United States of America

  • Gordon D. Brown,

    Affiliation Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom

  • Joseph V. Bonventre,

    Affiliation Renal Division, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America

  • David L. Williams,

    Affiliation Department of Surgery, James H. Quillen College of Medicine, Johnson City, Tennessee, United States of America

  • Neil A. R. Gow,

    Affiliation Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom

  • Donna L. Bratton,

    Affiliation Department of Pediatrics, National Jewish Health, Denver, Colorado, United States of America

  • Robert C. Murphy,

    Affiliation Department of Pharmacology, University of Colorado Denver, Aurora, Colorado, United States of America

  • Christina C. Leslie

    lesliec@njhealth.org

    Affiliations Department of Pediatrics, National Jewish Health, Denver, Colorado, United States of America, Department of Pharmacology, University of Colorado Denver, Aurora, Colorado, United States of America, Department of Pathology, University of Colorado Denver, Aurora, Colorado, United States of America

Abstract

The role of Group IVA cytosolic phospholipase A2 (cPLA2α) activation in regulating macrophage transcriptional responses to Candida albicans infection was investigated. cPLA2α releases arachidonic acid for the production of eicosanoids. In mouse resident peritoneal macrophages, prostacyclin, prostaglandin E2 and leukotriene C4 were produced within minutes of C. albicans addition before cyclooxygenase 2 expression. The production of TNFα was lower in C. albicans-stimulated cPLA2α+/+ than cPLA2α-/- macrophages due to an autocrine effect of prostaglandins that increased cAMP to a greater extent in cPLA2α+/+ than cPLA2α-/- macrophages. For global insight, differential gene expression in C. albicans-stimulated cPLA2α+/+ and cPLA2α-/- macrophages (3 h) was compared by microarray. cPLA2α+/+ macrophages expressed 86 genes at lower levels and 181 genes at higher levels than cPLA2α-/- macrophages (≥2-fold, p<0.05). Several pro-inflammatory genes were expressed at lower levels (Tnfα, Cx3cl1, Cd40, Ccl5, Csf1, Edn1, CxCr7, Irf1, Irf4, Akna, Ifnγ, several IFNγ-inducible GTPases). Genes that dampen inflammation (Socs3, Il10, Crem, Stat3, Thbd, Thbs1, Abca1) and genes involved in host defense (Gja1, Csf3, Trem1, Hdc) were expressed at higher levels in cPLA2α+/+ macrophages. Representative genes expressed lower in cPLA2α+/+ macrophages (Tnfα, Csf1) were increased by treatment with a prostacyclin receptor antagonist and protein kinase A inhibitor, whereas genes expressed at higher levels (Crem, Nr4a2, Il10, Csf3) were suppressed. The results suggest that C. albicans stimulates an autocrine loop in macrophages involving cPLA2α, cyclooxygenase 1-derived prostaglandins and increased cAMP that globally effects expression of genes involved in host defense and inflammation.

Introduction

The oxygenated metabolites of arachidonic acid comprise a large family of bioactive lipids that have diverse roles in regulating homeostatic processes and in modulating inflammation and immune responses [1]. The production of eicosanoids is initiated by the release of arachidonic acid that is metabolized through the 5-lipoxygenase pathway to leukotrienes and by cyclooxygenases (COX) to prostanoids and thromboxane. Eicosanoids are secreted and act locally in an autocrine or paracrine fashion through interaction with specific G-protein coupled receptors (GPCR) to exert their biological effects [24]. Leukotrienes are pro-inflammatory mediators but prostaglandins (PG) have pro- and anti-inflammatory effects depending on the cell type-specific GPCR-dependent signal transduction pathways that are triggered [1].

Macrophages are an important source of eicosanoids that are produced rapidly in response to stimulation by bacterial and fungal pathogens [58]. Resident tissue macrophages are a first line of defense against invading microorganisms that are recognized by pattern recognition receptors that engage microbial surface structures. We have used resident mouse peritoneal macrophages (RPM) to study the regulation of eicosanoid production in response to the model fungal agonist zymosan, cell wall particles of Saccharomyces cerevisiae [911]. Zymosan stimulates activation of the Group IVA cytosolic phospholipase A2 (cPLA2α), the first key regulatory enzyme in RPM that releases arachidonic acid for eicosanoid production [12]. To identify the pattern recognition receptors on RPM that mediate cPLA2α activation and eicosanoid production, the more medically relevant fungal pathogen Candida albicans was studied [13,14]. We found a role for dectin-1 and -2 that engage β-glucan and mannans on the C. albicans cell wall that, together with a MyD88-dependent pathway, promote cPLA2α activation and eicosanoid production [13,14]. Although C. albicans is a normal commensal organism, it is an opportunistic pathogen that is a leading cause of mycoses particularly in the immunocompromised and critically ill [15]. There has been considerable interest in elucidating the mechanisms regulating immune responses to C. albicans because of the prevalence of fungal infections [16].

Eicosanoids affect immune regulation by modulating cellular differentiation, phagocytic potential, migration and cytokine/chemokine production [5,1719]. The types and balance of cytokines produced during the early responses of innate immune cells to infection influence the macrophage phenotype, differentiation of lymphocytes and adaptive immune responses [2023]. In this study, we compared cPLA2α+/+ and cPLA2α-/- RPM to investigate the functional consequences of cPLA2α activation and the effect of endogenously produced eicosanoids on gene expression in response to C. albicans. Our results demonstrate that C. albicans-stimulated cPLA2α activation and the early production of prostanoids promotes an autocrine pathway in RPM that affects the expression of genes involved in host defense and to dampen inflammation.

Materials and Methods

Ethics Statement

The work with mice in this study was approved by the National Jewish Health Institutional Animal Care and Use Committee (IACUC) and conducted in accordance with their guidelines.

Materials

DMEM was from Cambrex BioScience. FBS (Gemini Bio-Products) was heat inactivated at 56°C for 30 min before use. Human serum albumin was obtained from Intergen. Polyclonal antibodies to murine COX1 and COX2, the protein kinase A inhibitor H-89, the COX inhibitor NS-398, the IP receptor antagonist CAY10441, the IP receptor agonist iloprost and the EP2 receptor agonist butaprost were from Cayman Chemical Co. Antibodies to β-actin were from Cell Signaling. The stable cAMP analogue 8-Br-cAMP was from Santa Cruz Biotechnology, Inc. The mouse TNFα cytoset ELISA kit was from Invitrogen. cAMP was quantified in macrophage lysates using the cAMP Biotrak EIA (non-acetylation protocol) from GE Healthcare according to the manufacturer’s protocol. RNA was isolated using the on-column DNase treatment with the RNeasy mini kit from Qiagen.

Mouse Strains

Pathogen-free Balb/c mice were obtained from Harlan Sprague Dawley. cPLA2α-/- mice were generated as previously described and backcrossed onto a Balb/c background for 10 generations [24]. The TLR4 mutant mouse strain C3H/HeJ and control strain C3H/HeOuJ were obtained from The Jackson Laboratory. TLR2-/- (C57BL/6) and MyD88-/- mice (C57BL/6/129) were generated as previously described [25]. MyD88+/- C57BL/6/129 mice were crossed to generate MyD88-/- mice and MyD88+/+ littermate controls. C57BL/6 control mice were obtained from The Jackson Laboratory. Dectin-1-/- mice (129sv/ev) were produced as described previously [26], and age and strain matched controls obtained from Taconic. Mice were used for macrophage isolation at 7-12 wk of age.

C. albicans Strains and Culture

C. albicans (ATCC 10261) was used for experiments unless otherwise indicated. The C. albicans Capmr1∆ null mutant defective in glycosylation, the re-integrant strain (Capmr1∆+CaPMR1) and parental wild-type control were generated as previously described [27]. C. albicans strains were grown on Sabouraud dextrose agar plates and maintained at 4°C.

RPM Infection

The day before the experiment, the strains were streaked onto fresh Sabouraud dextrose agar plates and incubated overnight at 37°C. C. albicans was scraped from the plate and washed twice in endotoxin-free PBS. Live C. albicans at a multiplicity of infection (moi) of 2 was used for all experiments.

RPM Isolation

RPM were obtained by peritoneal lavage as previously described [13]. Cells were plated at a density of 5 x 105/cm2 (48 well plate) and incubated for 2 h at 37°C in a humidified atmosphere of 5% CO2 in air. After washing the cultures to remove non-adherent cells, the adherent macrophages were incubated in DMEM containing 10% heat inactivated FBS, 100 µg/ml streptomycin sulfate, 100 units/ml penicillin G, 0.29 mg/ml glutamine for 16-18 h at 37°C. The cells were washed twice with serum-free DMEM containing 0.1% human serum albumin (stimulation medium) and then infected with C. albicans.

C. albicans Uptake and Killing assays

The ability of cPLA2α+/+ and cPLA2α-/- RPM to bind and internalize C. albicans was compared using an in vitro recognition assay as described previously with modifications [26]. RPM were incubated for 30 min with Alex Fluor 488-labeled C. albicans (m.o.i. 10) prepared as described [28]. RPM were washed 3 times with stimulation media and incubated further for 1 h. Cells were lysed with 3% Triton X-100 and the fluorescence intensity was measured. The killing assay involved incubating cPLA2α+/+ and cPLA2α-/- RPM with C. albicans (m.o.i. 5) for 30 min, followed by 3 washes and further incubation for 1 and 4 h. Cells were lysed with 3% Triton X-100 and the lysates streaked on Sabouraud dextrose agar plates to measure colony forming units (CFU).

Cytokine Measurement

The culture medium was removed at the indicated times after infection of RPM with C. albicans and stored at -80°C for cytokine measurement and eicosanoid analysis (see below). TNFα in the culture medium was quantified by ELISA and by Luminex assay, which gave similar results.

Mass Spectrometry Eicosanoid Analysis

The samples of culture media were thawed and mixed with an equal volume of cold methanol. Just prior to analysis they were diluted in water to a final methanol concentration of <15% and then extracted using a solid phase extraction cartridge (Strata Polymeric Reversed Phase 60 mg/ml, Phenomenex, Torrance, CA). The eluate (1 ml of methanol) was dried and reconstituted in 75 µl of HPLC solvent A (8.3 mM acetic acid buffered to pH 5.7 with NH4OH) and 25 µl of solvent B (acetonitrile/methanol, 65/35, v/v). An aliquot of each sample (50 µl) was injected into an HPLC and metabolites separated on a C18 column (Ascentis 15 cm x 2.1 mm, 5 µm, Supelco) eluted at a flow rate of 200 µl/min with a linear gradient from 25% to 75% solvent B in 13 min then increased to 98% in 2 min and held for 11 min. The HPLC system was directly interfaced into the electrospray ionization source of a triple quadrapole mass spectrometer (Sciex API 3000, PE-Sciex, Thornhill Ontario, Canada). Mass spectrometric analyses were performed in the negative ion mode using multiple reaction monitoring (MRM) for specific analytes. Deuterated internal standards were detected using the following transitions: m/z 355→275 for [d4]PGE2, m/z 373→167 for [d4]6-keto-PGF1α, mz 311→213 and mz 629→272 for [d4]LTC4. Eicosanoids were detected centered in specific retention time (RT) windows using the following transitions and limits of quantitation: PGE2, RT 9.3 min, m/z 351→271, 8 pg/ml; 6-keto-PGF1α, RT 6.4 min, m/z 369→163, 40 pg/ml and LTC4, RT 10.1 min, m/z 624→272, 40 pg/ml. MRM chromatograms using a similar analytic scheme have previously been described [29]. Quantitative results were calculated by determining the ratio of the signal of an analyte to that for an internal standard and comparing to a standard isotope dilution curve [30].

Western Blots

To prepare lysates for western blots, cell monolayers were washed twice in ice cold PBS and then scraped in lysis buffer: 50 mM Hepes, pH 7.4, 150 mM sodium chloride, 10% glycerol, 1% Triton X-100, 1 mM EGTA, 1 mM EDTA, 200 µM sodium vanadate, 10 mM tetrasodium pyrophosphate, 100 mM sodium fluoride, 300 nM p-nitrophenyl phosphate, 1 mM phenylmethylsulfonylfluoride, 10 µg/ml leupeptin, and 10 µg/ml aprotinin. After incubation on ice for 30 min, lysates were centrifuged at 15,000 rpm for 15 min and protein concentration in the supernatant determined by the bicinchoninic acid method. Lysates were boiled for 5 min after addition of Laemmli electrophoresis sample buffer, and then proteins were separated on 10% SDS-polyacrylamide gels. After transfer to nitrocellulose membrane, samples were incubated in blocking buffer (20 mM Tris-HCl, pH 7.6, 137 mM NaCl, 0.05% Tween (TTBS)) containing 5% nonfat milk for 1 h, and then incubated overnight at 4°C with primary antibodies in TTBS. The membranes were incubated with anti-rabbit IgG horseradish peroxidase antibody (1:5000) in TTBS for 30 min at room temperature. The immunoreactive proteins were detected using the Amersham ECL system.

Microarray Analysis

RPM cultured and stimulated with C. albicans for 3 h as described above were washed twice with endotoxin-free PBS and total RNA isolated. Template RNA quality was assessed with the Agilent Bioanalyzer 2100 and an Agilent Nano RNA 6000 kit per the Agilent protocol. RNA quality ranged from a RNA Integrity Number (RIN) of 8.1 to 10.0. An Agilent Quick Amp Labeling kit was used to generate Cy3 labeled RNA. Yields of 3.7-6.8 µg were obtained with specific activities of 7.5-9.4 pmol/µg. Fragmentation followed by hybridization was performed (Agilent Gene Expression Hybridization Kit) on Agilent Whole Mouse Genome kit 4x44 microarray slides at 65°C for 16 hr. Slides were washed according to the Agilent Quick Amp Labeling Kit protocol and scanned immediately on an Agilent G2505B scanner. The microarray results were log base 2 transformed and data normalization was applied using the 75% percentile shift method to adjust for experimental variability. Boxplots of resulting expression were examined for consistency and all quality control metrics were within acceptable ranges. Filtering was performed to exclude gene expression probes that did not reach a relative expression value of ≥35 across all groups. Microarray samples were grouped by unstimulated cPLA2α-/- RPM, C. albicans-stimulated cPLA2α-/- RPM, unstimulated cPLA2α+/+ RPM and C. albicans-stimulated cPLA2α+/+ RPM. Differences between C. albicans-stimulated cPLA2α+/+ and C. albicans-stimulated cPLA2α-/- RPM were compared using Student’s unpaired t-tests, while comparisons for unstimulated cPLA2α+/+ and C. albicans-stimulated cPLA2α+/+ RPM were evaluated using paired t-tests. For evaluating differential gene expression between C. albicans-stimulated cPLA2α+/+ and C. albicans-stimulated cPLA2α-/- RPM, genes that were not significantly affected by C. albicans treatment (p<0.05) in both cPLA2α+/+ and cPLA2α-/- RPM were excluded from the analysis. All processing and analyses were performed in Genespring GX 11.5 (Agilent Technologies, Santa Clara, CA). The data were analyzed using the DAVID bioinformatics resource to evaluate the functional clustering of genes [31]. The complete microarray results can be accessed in the Gene Expression Omnibus (GEO; www.ncbi.nlm.nih.gov/geo/) of the National Center for Biotechnology Information using the GEO Series accession number GSE46533.

Real-time PCR

RPM were isolated from cPLA2α+/+ and cPLA2α-/- mice, cultured as described above, and RNA isolated at 1, 3 and 6 h after stimulation with C. albicans. RNA concentration and purity were determined by UV spectrophotometry, and RNA integrity verified using an Agilent Bioanalyzer 2100. cDNA was synthesized from RNA (200 ng) using RT2 First Strand kit (SA Biosciences). Real-time PCR was performed using RT2 qPCR Mastermix and custom-made RT2 Profiler PCR Array System according to the manufacturer’s protocol using the StepOnePlus Real-Time PCR System (Applied Biosystems). PCR arrays in a 96-well format were used containing pre-validated primers tested for efficiency (SA Biosciences). The RT2 Profiler PCR Array System included a reverse transcription control preloaded into the primer buffer of the RT2 First Strand cDNA synthesis kit that measured the relative efficiency of the reverse transcription for all the samples. A genomic DNA control and a positive PCR control were also included in the system. The RT2 Profiler PCR Array data were normalized to two housekeeping genes Gapdh and Hprt and the relative gene expression level (2^(-ΔCt) was calculated using the formula ΔCt= Ct (gene of interest)-Ct (housekeeping gene). The data were analyzed on the PCR array data analysis SA Biosciences web portal (http://pcrdataanalysis.sabiosciences.com/pcr/arrayanalysis.php).

Real-time PCR was also performed with cDNA synthesized with random hexamer primers (Superscript III polymerase, Invitrogen) using TaqMan fast universal PCR master mix. TaqMan assay probes used were:

  1. Csf1 (1Mm00432686_m1),
  2. Csf3 (Mm00438335_g1),
  3. Tnf (Mm99999068_m1),
  4. Il10 (Mm00439614_m1),
  5. Nr4a2 (Mm00443060_m1),
  6. Crem (Mm00516346_m1),
  7. Stat3 (Mm01219775_m1) and

Gapdh (Mm99999915_g1). The housekeeping gene Gapdh and a calibrator containing mRNA from unstimulated cPLA2α+/+ and cPLA2α-/- RPM were used for normalization. Threshold cycle values (CT) were determined and used for ∆∆CT analysis of gene expression [32].

Results

The production of eicosanoids by RPM is initiated by the activation of cPLA2α, which occurs rapidly in response to C. albicans or zymosan due to post-translational processes [912]. The major arachidonic acid metabolites produced by RPM in response to C. albicans and zymosan are PGI2, PGE2, and LTC4, and their production is dependent on cPLA2α activation to provide arachidonic acid substrate [1214]. As shown in Figure 1A, eicosanoids were produced most rapidly during the first 30 min after C. albicans addition. Prostaglandin production occurred before the increase in COX2 expression stimulated by C. albicans, which was detected 3 h after addition of C. albicans but not at 1 h (Figure 1B). In contrast, COX1 was constitutively expressed in RPM and expression was not affected by C. albicans infection. Microarray data also confirmed that COX2 expression was very low compared to COX1 in unstimulated cPLA2α+/+ RPM, but there was a significant increase in expression of COX2 (Ptgs2) but not COX1 (Ptgs1) in cPLA2α+/+ RPM treated with C. albicans for 3 h (Table 1). The results suggest that cPLA2α-mediated release of arachidonic acid couples to COX1 for early production of prostaglandins.

thumbnail
Figure 1. Time course of C. albicans-stimulated eicosanoid production.

(A) RPM were incubated with C. albicans for the indicated times. The culture medium from unstimulated (open squares) or C. albicans-stimulated (closed squares) RPM was analyzed for eicosanoids by mass spectrometry. The data are the average of triplicate samples (±S.D.) from a representative experiment. (B) Cell lysates from unstimulated RPM (US) or RPM stimulated with C. albicans (CA) for 1, 3 and 6 h were analyzed for COX1 and COX2 expression by Western blotting. Sample loading was evaluated by probing with antibodies to β-actin.

https://doi.org/10.1371/journal.pone.0069002.g001

Official SymbolEntrez_Gene_IDUnstimulatedC. albicans-treated
Mean ExpressionMean Expression
Ptgs2*1922567 ± 1411243 ± 2938
Ptgs1192244694 ± 27312027 ± 655
Ptger219217204 ± 18177 ± 65
Ptger419219257 ± 24248 ± 50
Ptgir*19222589 ± 2171168 ± 179

Table 1. Relative expression values of cyclooxygenases and prostaglandin receptors in RPM.

cPLA2α+/+ RPM were stimulated with C. albicans for 3 h and gene expression determined by microarray analysis. The * denotes a significant (p<0.05) increase in expression by C. albicans treatment.
CSV
Download CSV

Role of cPLA2α in regulating TNFα production

The initial focus was to determine if cPLA2α activation regulates TNFα production in C. albicans-stimulated RPM since prostaglandins can suppress production of this pro-inflammatory cytokine as we reported for L. monocytogenes-stimulated RPM [8,33,34]. First we investigated if TNFα production was mediated by similar PRRs that promote cPLA2α activation in response to C. albicans. We reported that dectin-1 and MyD88, but not TLR2 or TLR4, play a role in the activation of cPLA2α in response to C. albicans [13,14]. We found that production of TNFα 6 h after addition of C. albicans was reduced in dectin-1-/- and MyD88-/- RPM compared to dectin-1+/+ and MyD88+/+ RPM (Figure 2A and 2B). The requirement for MyD88 suggested a role for TLRs. A comparison of RPM from TLR2+/+ and TLR2-/- mice showed that TNFα production was not mediated by TLR2 (data not shown). However, TLR4 partially contributed to C. albicans-mediated TNFα production, which was reduced by approximately 50% in TLR4-/- RPM (Figure 2C). Since mannans of C. albicans cell wall engage TLR4 we tested the ability of the C. albicans glycosylation mutant (Capmr1∆ null mutant), which is devoid of phosphomannans and has defective N- and O-linked mannans, to stimulate TNFα production in TLR4+/+ and TLR4-/- RPM [27,35]. Compared to TLR4+/+ RPM treated with wild type C. albicans, TNFα production in TLR4+/+ RPM treated with Capmr1∆ null mutant was reduced by about 50% similar to the level observed in TLR4-/- RPM stimulated with wild type C. albicans (Figure 2C). TNFα production by TLR4+/+ RPM was restored when the CaPMR1 gene was reintegrated into the mutant strain (Capmr1+CaPMR1). Therefore PRRs on RPM that engage cell wall mannans and β-glucans contribute to TNFα production. Since cPLA2α+/+ and cPLA2α-/- RPM were used to determine the role of cPLA2α in regulating gene expression in response to C. albicans (as described below), we compared their levels of expression of PRRs involved in C. albicans recognition. Microarray data showed that cPLA2α+/+ and cPLA2α-/- RPM express similar levels of PRRs Clec7a (dectin-1), Clec4n (dectin-2), Tlr4 and Tlr2 (Gene Expression Onmibus, www.ncbi.nlm.nih.gov.geo/, GSE46533). We also compared the ability of cPLA2α+/+ and cPLA2α-/- RPM to bind and internalize C. albicans. Results of a recognition assay demonstrated no differences in the uptake of Alex Fluor-labeled C. albicans by cPLA2α+/+ and cPLA2α-/- RPM (data not shown). A C. albicans killing assay was also carried out by incubating RPM with C. albicans and then measuring the recovery of CFU from RPM after further incubation for 1 and 4 h. There were no differences in CFU recovered at 1 h in WT and cPLA2α-/- RPM. However at 4 h there was a small but significantly higher level of C. albicans CFU recovered from cPLA2α-/- RPM. In three independent experiments the CFU in cPLA2α-/- RPM was 172%±32%, p<0.002 compared to cPLA2α+/+ RPM (100%). The results suggest that the cPLA2α+/+ RPM have a slightly greater ability to kill internalized C. albicans.

thumbnail
Figure 2. Role of PRRs in regulating C. albicans-stimulated TNFα production.

Wild type (open bars) and Dectin-1-/- (A), MyD88-/- (B) and TLR4-/- (C) RPM (shaded bars) were incubated with C. albicans for 6 h. In panel C, RPM were stimulated with the parental wild type C. albicans (WT), the Capmr1∆ null mutant and the re-integrant strain (Capmr1∆+CaPMR1). The data are the average of 3 experiments ±S.E. (*p<0.05). Levels of TNFα in the culture medium were determined by ELISA.

https://doi.org/10.1371/journal.pone.0069002.g002

The role of cPLA2α activation and prostanoid production in regulating the production of TNFα in response to C. albicans was investigated by comparing RPM from cPLA2α+/+ and cPLA2α-/- mice, and by treating the macrophages with a cyclooxygenase inhibitor NS398 (Figure 3A). The production of TNFα was lower in cPLA2α+/+ RPM compared to cPLA2α-/- RPM measured 6 h after C. albicans addition. NS398 treatment enhanced TNFα production in cPLA2α+/+ but not in C. albicans-stimulated cPLA2α-/- RPM suggesting that prostanoids suppress TNFα expression. NS398 completely blocked production of PGE2 and PGI2 in RPM stimulated with C. albicans for 6 h (data not shown), and at the concentration used (10 µM) inhibits both murine COX1 and COX2 [36]. To further investigate the role of prostanoids in the autocrine regulation of TNFα production, RPM were treated with agonists for the PGE2 receptor EP2 (butaprost) and the PGI2 receptor IP (iloprost) (Figure 3B). Microarray data showed that RPM express the IP receptor (Ptgir), the EP2 (Ptger2) and EP4 (Ptger4) receptors (Table 1). The agonists had no effect on the levels of TNFα produced by cPLA2α+/+ RPM that produce endogenous prostaglandins in response to C. albicans (Figure 3B). However, the higher level of TNFα produced by C. albicans-stimulated cPLA2α-/- RPM, which do not produce endogenous prostaglandins, was reduced by the receptor agonists to the level produced by cPLA2α+/+ RPM. The data suggest that prostaglandins acting through the EP2 and IP receptors suppress TNFα production since it is enhanced by inhibiting prostaglandin production in C. albicans-stimulated cPLA2α+/+ RPM and suppressed by prostaglandin receptor agonists in cPLA2α-/- RPM.

thumbnail
Figure 3. Role of prostaglandins in regulating C. albicans-stimulated TNFα production.

cPLA2α+/+ and cPLA2α-/- RPM were incubated with (A) NS-398 (10 µM), or (B) iloprost (1 µM) or butaprost (10 µM) for 30 min followed by incubation with C. albicans for 6 h. Levels of TNFα in the culture medium were determined by ELISA. The data are the average of 3 experiments ±S.E. (*p<0.05).

https://doi.org/10.1371/journal.pone.0069002.g003

The EP2 and IP receptors mediate increases in cAMP, which is implicated in regulating Tnfα gene expression [37,38]. As shown in Figure 4A, the stable cAMP analogue 8-Br-cAMP suppressed C. albicans-stimulated TNFα production in cPLA2α-/- RPM, as observed for the prostanoid receptor agonists, but had no effect on the lower level of TNFα produced by cPLA2α+/+ RPM. The results suggest that prostaglandins produced by C. albicans-stimulated cPLA2α+/+ RPM act in an autocrine manner through prostaglandin receptors that increase cAMP to suppress TNFα production. This is supported by results showing that levels of cAMP were higher in cPLA2α+/+ RPM than cPLA2α-/- RPM within 5-30 min after C. albicans addition (Figure 4B).

thumbnail
Figure 4. cAMP production is enhanced by cPLA2α activation and suppresses TNFα production.

(A) cPLA2α+/+ and cPLA2α-/- RPM were incubated with 8-Br-cAMP (1 mM) for 30 min followed by incubation with C. albicans for 6 h. Levels of TNFα in the culture medium were determined by ELISA. (B) cPLA2α+/+ (WT, squares) and cPLA2α-/- RPM (KO, circles) were incubated with (closed symbols) or without (open symbols) C. albicans (CA) for the indicated times. Cell lysates were processed for cAMP determinations as described in Experimental Design. The data are the average of 3 experiments ±S.E. (*p<0.05). In panel B, CA treated WT vs. CA treated KO at 5 and 15 min are compared for significance.

https://doi.org/10.1371/journal.pone.0069002.g004

Effect of C. albicans on gene expression in RPM

We next determined the effect of C. albicans on global gene expression in RPM by microarray and then evaluated how cPLA2α activation modulates the transcriptional response. C. albicans stimulated an increase in expression of 427 genes (≥4.0-fold, p<0.05, n=3) in cPLA2α+/+ Balb/c RPM at 3 h. Relative expression levels for these genes and the fold change in response to C. albicans are shown in Table S1A. Many of the genes that increase in response to C. albicans represent the common host-response that is induced in many cell types by a variety of infectious agents [39]. The data were analyzed using the DAVID bioinformatics resource to evaluate the functional clustering of genes that were increased in RPM in response to C. albicans [31]. The most highly enriched clusters contained genes in apoptosis, cytokines, wound and inflammatory responses, regulation of phosphorylation and protein kinase activity, cell motion, vascular development, regulation of cytokine production, MAP kinase phosphatase activity, regulation of transcription and growth factor activity (Table 2). Csf3 was the most highly induced gene by C. albicans (>600-fold) (Table S1A). The cytokine CSF3 regulates the production and function of neutrophils and is important for host defense against C. albicans [40,41]. As discussed below, the expression of Csf3 was regulated by cPLA2 activation. There were 110 genes down-regulated in RPM at 3 h by C. albicans (≥4-fold, p<0.05, n=3) (Table S1B). The clusters for the down-regulated genes had very low enrichment scores compared to the up-regulated genes when subject to DAVID analysis (data not shown).

Annotation ClustersOfficial Symbol
ApoptosisBcl2l11, Cflar, Cd24a, Chac1, Ddit4, Egin3, Epha2, Rybp, Traf1, Traf5, Ahr, F2r, Csrnp1, Fem1b, Gzmb, Gadd45b, Gadd45g, Id1, Il6, Jmjd6, Malt1, Myc, Niacr1, Nfκb1, Nr4a2, Osm, Phlda1, Bnip3, Blcap, Ppp1r15a, Srgn, Siah2, Mcl1, Trib3, Trim69, Tnf, Tnfrsf12a, Tnfaip3, Unc5b, Zc3h12a
Cytokines, Response to wounding, and Inflammatory responseAreg, Cd24a, Kdm6b, Bmp2, Bmp6, Ccl2, Ccl3, Ccl4, Ccl7, Ccr1, Cxcl1, Cxcl2, Cxcl3, Csf2, Csf3, F2r, F3,Gdf15, Gja1, Hbegf, Id3, Il1a, Il1b, Il1f6, Il10, Il23a, Il6, Nfkbid, Olr1, Osm, Plaur, Plek, Procr, Proz, Slc7a2, Sphk1, Tnf, Tnfsf9
Regulation of phosphorylation and protein kinase activityCd24a, Adora2a, Bmp2, F2r, Cdkn1a, Cish, Dgkg, Dusp16, Edn1, Ereg, Gadd45b, Gadd45g, Il1b, Il6, Lrp8, Laper1, Nrg1, Osm, Reln, Spag9, Sphk, Spry2, Socs3, Trib3, Tnf
Cell motionCd24a, Epha4, Alcam, Ccr1, Cxcl2, Cxcl3, Egr2, Gja1, Hbegf, Il1b, Lrp8, Nrg1, Nr4a2, Plau, Pdpn, Pvr, Reln, Runx3, Zfand5, Tes, Tnf, Tnfrsf12a, Vegfa
Vascular developmentEpha2, Junb, Smad7, Edn1, Efnb2, Ereg, Gja1, Hbegf, Id1, Itgav, Il1b, Jmjd6, Pdpn, Prok2, Zfand5, Sphk1, Socs3, Tgm2, Tnfrsf12a, Vegfa, Zc3h12a, Zfp36l1
Regulation of cytokine productionCd24a, Cd83, Adora2a, Adora2b, F2r, Edn1, Ereg, Fst, Inhbb, Irf4, Il1a, Il1b, Il10, Il6, Nfκb1, Prok2, Rel, Srgn, Sphk1, Tnf
MAP kinase phosphatase activityDusp1, Dusp2, Dusp4, Dusp8, Dusp10, Dusp14, Dusp16
Regulation of transcription from RNA polymerase II promoterEaf1, Kdm6b, Mxi1, Pou3f1, Rybp, Skil, Atf4, Ahr, Bmp2, Csrnp1, Egr1, Egr2, Fosl1, Hes1, Inhba, Id1, Id2, Id3, Irf4, Il6, Jarid2, Med13, Myc, Nfκb1, Nufip1, Nr4a1, Nr4a2, Nr4a3, Osm, Plagl1, Pbx1, Sap30, Tnf, Vegfa
Growth factor activityAreg, Bmp2, Bmp6, Cxcl1, Csf2, Csf3, Ereg, Gdf15, Hbegf, Inhba, Inhbb, Il1b, Il6, Vegfa

Table 2. Functional annotation clusters of genes induced in C. albicans-stimulated RPM.

Genes expressed at higher levels (427 genes, ≥4-fold, <0.05) in C. albicans-stimulated cPLA2α+/+ RPM were analyzed using DAVID bioinformatics resource.
CSV
Download CSV

Genes expressed at lower levels in C. albicans-stimulated cPLA2α+/+ than cPLA2α-/- RPM

Differential gene expression was compared in cPLA2α+/+ and cPLA2α-/- RPM treated with C. albicans for 3 h. We chose to study the effect of C. albicans infection on gene expression at 3 h in order to evaluate the role of cPLA2α activation and eicosanoids in regulating early responses during the acute phase of infection. The regulation of gene expression at later times becomes more complicated due to autocrine effects of the products of early response genes that promote induction of a second wave of gene induction. In cPLA2α+/+ RPM, 86 genes were expressed at lower levels and 181 genes at higher levels than cPLA2α-/- RPM (≥2-fold, p<0.05, n=3) (Tables S2A and S2B, respectively). When genes expressed at lower levels in cPLA2α+/+ RPM were analyzed using DAVID, they grouped into functional clusters involving GTP binding, regulation of cytokine production/cytokine receptor interaction and regulation of proliferation (Table 3). The expression of genes for GTP binding proteins included several IFNγ-inducible GTPases (guanylate binding proteins (Gbp) 1, 2, 3, 5, 6 and 7; immunity-related GTPase family M members (Irgm) 1 and 2; IFNγ-inducible protein (Ifi) 47 and IFNγ-inducible GTPase (Iigp) 1). Some of these genes regulate host defense to microbial infection although their function is poorly understood [4245]. Several genes expressed lower in C. albicans-stimulated cPLA2α+/+ than cPLA2α-/- RPM in the cytokine cluster (Table 3) are pro-inflammatory such as the chemokine Cx3cl1 (fracktalkine), Cd40, Tnfα and Ifnγ [4648]. The lower expression of Ifnγ in cPLA2α+/+ RPM correlated with the reduced expression of the IFNγ regulated GTPases, although its level of expression in RPM was very low (Table S2A). Genes for the transcription factors, interferon regulatory factors (Irf) 1 and Irf4 (Cytokine cluster), and the AT-hook transcription factor (Akna) were also expressed at lower levels in cPLA2α+/+ than cPLA2α-/- RPM (Table S2A). AKNA promotes Cd40 expression suggesting a correlation between low expression of Akna and Cd40 in cPLA2α+/+ RPM [49]. AKNA functions in inflammation and cancer [50]. There was also a correlation with the lower expression of genes for guanylate binding proteins (Gbp) and Tnfα in cPLA2α+/+ RPM and their transcriptional regulator Irf1 [51]. IRF transcription factors play important roles in host defense and regulating immune responses [52].

Annotation ClustersOfficial Symbol
GTP bindingRab33A, Rasd2, Gbp1, Gbp2, Gbp3, Gbp5, Gbp6, Gbp7, Irgm1, Irgm2, Ifi47, Iigp1, Ak4
Regulation of cytokine production, Cytokine receptor interactionCd40, Ccl5, Cx3cl1, Csf1, Infg, Irf1, Irf4, Il15ra, Ticam2, Il20rb, Tnfrsf14, Tnf
Regulation of proliferationCd40, Adm, Csf1, Edn1, Igf1, Infg, Il20rb, Lst1, Plau, Smo, Tnfrsf14, Tnf

Table 3. Functional annotation clusters of genes expressed at lower levels in C. albicans-stimulated cPLA2α+/+ than cPLA2α-/- RPM.

Genes expressed at lower levels (86 genes, ≥2-fold, <0.05) in cPLA2α+/+ than cPLA2α-/- RPM stimulated for 3 h with C. albicans were analyzed using DAVID bioinformatics resource.
CSV
Download CSV

cPLA2α+/+ RPM expressed lower mRNA levels of the chemokine Ccl5 (Cytokine cluster), which promotes the trafficking of cells to sites of inflammation [53]. PGE2 suppresses CCL5 production in macrophages and dendritic cells thus dampening inflammation and immune responses [54]. Colony stimulating factor 1 (Csf1, Cytokine cluster) was induced to a greater extent in cPLA2α-/- RPM (10-fold) than cPLA2α+/+ RPM (3-fold) (Table S2A). It promotes macrophage-lineage development but also recruits myeloid cells during inflammation and infection, and promotes their survival [55]. Another pro-inflammatory gene expressed at lower levels in cPLA2α+/+ RPM was the vasoactive peptide endothelin 1 (Edn1, Regulation of proliferation cluster), which stimulates myeloid and mast cells at sites of inflammation [56] (Table S2A). Overall the results implicate cPLA2α activation and eicosanoid production in suppressing the expression of pro-inflammatory genes, and transcription factors that regulate their expression.

We corroborated the microarray results by real-time PCR for representative genes expressed lower in cPLA2α+/+ than cPLA2α-/- RPM. Their expression was preferentially enhanced by C. albicans in cPLA2α-/- RPM compared to cPLA2α+/+ RPM suggesting that products of cPLA2α activation suppress their expression (Figure 5). Results of real time PCR showed that expression of these genes was transient in cPLA2α-/- RPM occurring maximally 3 h after stimulation with C. albicans.

thumbnail
Figure 5. Time course of expression of genes expressed at lower levels in C. albicans-stimulated cPLA2α+/+ than cPLA2α-/- RPM.

cPLA2α+/+ (WT, circles) and cPLA2α-/- (KO, triangles) RPM were incubated with (CA) or without (US) C. albicans for the indicated times. RNA was isolated and gene expression determined by real-time PCR using the RT2 Profiler PCR Array System (SA Bioscience) as described in Experimental Design. The data were normalized to the housekeeping genes Gapdh and Hprt. The results are the average of 3 experiments ±S.E. Gene expression in C. albicans infected WT at 3 h was compared to C. albicans infected KO at 3 h to determine significance (*p<0.05).

https://doi.org/10.1371/journal.pone.0069002.g005

Genes expressed at higher levels in C. albicans-stimulated cPLA2α+/+ than cPLA2α-/- RPM

A larger number of genes were expressed at higher levels in cPLA2α+/+ RPM than cPLA2α-/- RPM (181 genes, ≥2-fold, p<0.05, n=3) (Table S2B). From DAVID analysis, genes clustered in functional groups involving vascular development, embryonic morphogenesis, sexual reproduction, response to wounding, inflammatory and defense responses, growth factors and growth factor activity, DNA binding and transcription regulation, and disulfide bond (Table 4). Several genes in these clusters are associated with cancer development consistent with a role for prostaglandins in promoting carcinogenesis [57,58]. These include the Eph receptor A2 tyrosine kinase (EphA2, Vascular development cluster), the epidermal growth factor receptor (EGFR) ligands epiregulin (Ereg) and amphiregulin (Areg) (Growth factor cluster), the transmembrane glycoprotein podoplanin (Pdpn) and its transcriptional regulator the homeobox protein Prox1 (Vascular development cluster), the chemokine receptor 7 (CxCr7, Disulfide bond cluster), matrix metalloproteinase 13 (Mmp13, Embryonic morphogenesis cluster) and its transcriptional regulators Runx2 and nuclear receptor subfamily 4, group A, member 2 (Nr4a2). These genes expressed at higher levels in cPLA2α+/+ RPM promote angiogenesis, tumor growth and invasion, and are regulated by prostaglandins and cAMP [5966].

Annotation ClustersOfficial Symbol
Vascular developmentEph2, Chd7, Ereg, Foxc1, Gja1, Itgav, Lepr, Nus1, Pdpn, Prox1, S1pr1, Socs3, Zfp36l1
Embryonic morphogenesisEph2, Chd7, Chst11, Foxc1, Hes1, Il10, Mmp13, Pbx1, Prox1, Socs3, Spry2, Jag2
Sexual reproductionBcl6, Bcl2l11, Crem, Calca, Cadm1, Ereg, Fst, Foxc1, Jag2, Lepr, Pvrl3, Rgs2, Stat3
Response to wounding, Inflammatory and Defense responsesBmp6, Cd14, Calca, Ddah2, Entpd1, Gja1, Hdac5, Il1f6, Il10, Saa1, Saa2, Stat3, Thbd, Thbs1
Growth factors, GF activityAreg, Bmp6, Chst11, Csf3, Ereg, Foxc1, Gja1, Hgf, Inhbb, Jag2
DNA binding, Transcription regulationArid3b, Bcl6, Bach2, Gata2, Lhx8, Mxd1, Mxi1, Setbp1, Thap2, Crem, Chd7, Dedd2, Foxc1, Hes1, Hdac5, Lrrfip1, Nr4a2, Pbx1, Prox1, Runx2, Stat3, Fosl2, Sbno2, Tshz3, Tle1, Mafb, Zfp36, Zfp36l1
Disulfide bondNt53, Abca1, Cd14, Cd80, Edil3, Eph2, Gpr35, Areg, Antxr2, Bmp6, Calca, Cacna1d, Cadm1, Cbln3, CxCr7, Csf3, Entpd1, Ereg, Fst, Gja1, Havcr2, Hgf, Inhbb, Itgav, Il10, Jag2, Lepr, Lifr, Man1a, Mmp13, Mmp3, Mpzl1, Niacr1, Pla1a, Pvrl3, Ptger2, Lpar6, Ramp3, Sema6d, Tnfaip6, Thbd, Thbs1, Trem1, Tnfrsf9

Table 4. Functional annotation clusters of genes expressed at higher levels in C. albicans-stimulated cPLA2α+/+ than cPLA2α-/- RPM.

Genes expressed at higher levels (181 genes, ≥2-fold, <0.05) in cPLA2α+/+ than cPLA2α-/- RPM stimulated for 3 h with C. albicans were analyzed using DAVID bioinformatics resource.
CSV
Download CSV

Of particular interest were the large number of genes expressed at higher levels in C. albicans-stimulated cPLA2α+/+ RPM that function to dampen inflammation. C. albicans induced high expression of suppressor of cytokine signaling 3 (Socs3, Vascular development and Embryonic morphogenesis clusters) in cPLA2α+/+ RPM (16-fold) and to a lesser extent in cPLA2α-/- RPM (6-fold) (Table 4, Table S2B). SOCS proteins function as negative feedback inhibitory pathways to control immune cell activation and inflammation [67]. Socs3 expression is also regulated by STAT3 (Table 4, Sexual reproduction and response to wounding clusters), which was induced 4-fold in C. albicans-stimulated cPLA2α+/+ RPM but not significantly affected in cPLA2α-/- RPM (Table S2B). One of the most differentially expressed genes was Il10 (Embryonic morphogenesis cluster) that was induced 78-fold by C. albicans cPLA2α+/+ RPM and 7-fold in cPLA2α-/- RPM (Table 4, Table S2B). The expression of Il10 is regulated in macrophages by the transcription factor PBX1 [68], also expressed at higher levels in C. albicans-stimulated cPLA2α+/+ than cPLA2α-/- RPM. The anti-inflammatory response (AIR) in macrophages induced by IL10 is mediated by STAT3 through induction of the helicase family co-repressor, Strawberry notch homologue 2 (Sbno2) [6971]. Expression of Sbno2 (Table 4, DNA binding, Transcription regulation cluster) was increased in C. albicans-stimulated cPLA2α+/+ RPM but not in cPLA2α-/- RPM (Table S2B).

Several genes implicated in suppressing Tnfα expression were expressed at higher levels in cPLA2α+/+ than cPLA2α-/- RPM. One of these genes, the zinc finger protein 36, C3H type-like 1 (Zfp36l1, DNA binding cluster), was increased by C. albicans in cPLA2α+/+ but not cPLA2α-/- RPM (Table 4, Table S2B), and inhibits TNFα production in macrophages by destabilizing its mRNA [72]. The cAMP responsive element modulator (Crem, also called Icer) (Table 4, Sexual reproduction and DNA binding cluster), was highly induced in cPLA2α+/+ RPM (16-fold) in response to C. albicans but not significantly affected in cPLA2α-/- RPM (Table S2B). CREM suppresses expression of pro-inflammatory genes including Tnfα [73]. The anti-inflammatory and immunosuppressive neuropeptide calcitonin gene-related peptide (Calca, Sexual reproduction cluster) that is higher in cPLA2α+/+ than cPLA2α-/- RPM suppresses Tnfα through induction of Crem [74,75]. C. albicans also induces expression of the cAMP-regulated nuclear receptor Nr4a2 to a greater extent in cPLA2α+/+ RPM (81-fold) than in cPLA2α-/- RPM (10-fold) (Table S2B). NR4A2 suppresses Tnfα expression in microglia and astrocytes [76].

Several other genes that are expressed at higher levels in cPLA2α+/+ RPM than cPLA2α-/- RPM have diverse functions but also act to dampen inflammation (Table 4). Follistatin (Fst) (Sexual reproduction cluster) curbs inflammation by inactivating the inflammatory actions of activin [77]. Expression of the anti-inflammatory genes Thbs1 and Thbd are also expressed higher in C. albicans-stimulated cPLA2α+/+ RPM than cPLA2α-/- RPM (Table S2B) [78,79].

Several genes expressed higher in cPLA2α+/+ than cPLA2α-/- RPM are involved in host defense such as the gap junction protein, alpha 1 (Gja1, Vascular development cluster) (Table 4, Table S2B). GJA1 promotes phagocytosis and host survival to bacterial infection [80]. Csf3 (Growth factor cluster) is highly upregulated in response to C. albicans in cPLA2α+/+ RPM (640-fold) but to a lesser extent in cPLA2α-/- PM (140-fold) (Table S2B). The orphan receptor triggering receptor expression on myeloid cells (Trem1, Disulfide bond cluster) is upregulated to a greater extent in C. albicans-stimulated cPLA2α+/+ (11-fold) than cPLA2α-/- RPM (2.7-fold) (Table S2B). TREM1 couples with the signaling adaptor DAP12 and has complex effects to enhance or dampen responses to TLR activation [81]. Histidine decarboxylase (Hdc), the encodes the rate-limiting enzyme for histamine synthesis, is another highly differentially expressed gene that is 20-fold higher in cPLA2α+/+ than cPLA2α-/- RPM (Table S2B). Hdc is transcriptionally induced in myeloid cells in response to cytokines and TLR agonists leading to immediate secretion of newly synthesized histamine [82]. Prostaglandins induce Hdc expression and also greatly potentiate the vasoactive effects of histamine [8385].

The microarray results were corroborated by real-time PCR analysis for several representative genes expressed at higher levels in cPLA2α+/+ than cPLA2α-/- RPM (Figure 6). When analyzed 1-6 h after C. albicans addition, several early response genes (Crem, Nr4a2, Cxcr7) showed highest expression in cPLA2α+/+ RPM at 1 h. The early induction of the transcriptional regulators Crem and Nr4a2 due to cPLA2α activation suggests that they play a role in regulating gene expression to increases in cAMP. The expression of most genes peaked 3 h after C. albicans addition with some decreasing to near baseline by 6 h (Hdc, Map4k4, Il10, Stat3, Thbs1, Trem1) while others remained elevated (Csf3, Adamts9, Gja1).

thumbnail
Figure 6. Time course of expression of genes expressed at higher levels in C. albicans-stimulated cPLA2α+/+ than cPLA2α-/- RPM.

cPLA2α+/+ (WT, circles) and cPLA2α-/- (KO, triangles) RPM were incubated with (CA) or without (US) C. albicans for the indicated times. RNA was isolated and gene expression determined by real-time PCR using the RT2 Profiler PCR Array System (SA Bioscience) as described in Experimental Design. The data were normalized to the housekeeping genes Gapdh and Hprt. The results are the average of 3 experiments ±S.E. Gene expression in C. albicans infected WT at 3 h was compared to C. albicans infected KO at 3 h to determine significance (*p<0.05).

https://doi.org/10.1371/journal.pone.0069002.g006

Role of the IP receptor and PKA in regulating gene expression

We investigated the role of prostacyclin production (the prostanoid produced at the highest level in RPM) and PKA, the downstream mediator of cAMP, in regulating gene expression by treating cPLA2α+/+ RPM with the IP receptor antagonist CAY10441 and the PKA inhibitor H89 (Figure 7). Representative genes expressed at lower levels in C. albicans-stimulated cPLA2α+/+ than cPLA2α-/- RPM (Tnfα and Csf1) were enhanced by blocking the action of PGI2 and inhibiting PKA. In contrast, genes expressed at higher levels in cPLA2α+/+ than cPLA2α-/- RPM (Crem, Il10, Csf3, Nr4a2) were suppressed by the IP receptor antagonist and by the PKA inhibitor. The results suggest that cPLA2α-mediated prostaglandin production promotes an autocrine loop to increase cAMP and PKA activation for regulating expression of these genes.

thumbnail
Figure 7. Effect of IP receptor antagonist and PKA inhibitor on gene expression.

cPLA2α+/+ RPM were incubated with the IP receptor antagonist CAY10441 (1 µM) (light gray bars) and the PKA inhibitor H89 (10 µM) (black bars) for 30 min followed by stimulation with C. albicans for 3 h. RNA was isolated and gene expression determined by real-time PCR. Gene expression values are presented as the % of control values (set at 100%), which is C. albicans-stimulated RPM not treated with CAY10441 or H89. The results are the average of 3 experiments ±S.E. (*p<0.05).

https://doi.org/10.1371/journal.pone.0069002.g007

Discussion

In this study we describe the changes in gene expression that occur in RPM during infection with C. albicans, and how gene expression is influenced by the activation of cPLA2α and endogenously produced lipid mediators. Resident tissue macrophages are sentinel cells that are important in first sensing and responding to microbial invasion. Therefore our study investigates how cPLA2α activation modulates macrophage responses during the initial stages of infection to affect the balance of host defense and inflammation. The production of eicosanoids in RPM is dependent on cPLA2α activation to provide arachidonic acid [12,14]. They are released within minutes of activation by C. albicans to rapidly engage eicosanoid receptors for regulating transcriptional responses. Although there have been a number of studies investigating the effect of adding exogenous eicosanoids to cells, by comparing cPLA2α+/+ and cPLA2α-/- RPM we are probing the primary mechanism for production of eicosanoids in macrophages at levels expected to occur locally in tissues in response to microbial infection. Our analysis provides global insight into the extensive changes in gene expression that are initiated by activation of cPLA2α and endogenously produced eicosanoids in resident tissue macrophages early in response to microbial infection.

The recognition of C. albicans by macrophages is complex since the fungal cell wall contains several chemical components that differentially engage a number of receptors including a variety of TLRs and lectins [86]. These receptors promote unique signaling pathways that preferentially induce distinct cellular responses. In RPM C. albicans triggers rapid activation of mitogen-activated protein kinases and calcium mobilization necessary for cPLA2α activation through dectin-1, dectin-2 and MyD88 pathways [13,14]. The results of this study suggest that the differential expression of many genes observed in cPLA2α+/+ and cPLA2α-/- RPM is due to an autocrine loop involving cPLA2α, prostaglandins and increased cAMP production, which is significantly higher in C. albicans-stimulated cPLA2α+/+ than C. albicans-stimulated cPLA2α-/- RPM. This is illustrated by results showing that TNFα production is suppressed by prostaglandins through increases in cAMP. Expression of TNFα occurs in part through dectin-1 and TLR4 in RPM that activate NF-κB and transcription [86]. In RPM the rapid production of prostanoids, particularly PGI2 that acts through the IP receptor, increases cAMP and PKA activation that suppresses transcription by mechanisms that are not fully understood. In addition to TNFα we observed differential expression of several genes previously reported to be regulated by prostaglandins and increases in cAMP in a variety of cell types. These include Ccl5, Socs3, Il10, Gja1, Crem, Thbd, Abca1, Csf3, Trem1 [33,69,73,8793]. Similar to our results in C. albicans-stimulated RPM, an autocrine loop pathway involving cPLA2α, prostacyclin and cAMP has been shown to enhance expression of Areg, Ereg and Fst, Cre-dependent genes involved in vascular remodeling and angiogenesis [94]. This autocrine loop involving prostaglandins and cAMP is triggered in many cell types in response to a variety of agonists indicating that it is an important, widely used pathway for regulating gene expression.

The rapid increase in cAMP that occurs in C. albicans-stimulated cPLA2α+/+ RPM is consistent with functional coupling of cPLA2α activation and metabolism of arachidonic acid to prostanoids by constitutively expressed COX1 since the response occurs before the expression of COX2. A role for COX1 in mediating prostaglandin production in LPS-stimulated RPM has previously been reported [34]. COX1 provides prostaglandins that regulate normal physiological processes and can regulate the early phases of inflammation [17]. RPM express the EP2, EP4 and IP receptors that mediate increases in cAMP, and our results show that EP2 or IP receptor agonists suppress TNFα production. It is likely that PGI2 and PGE2 both contribute to the regulation of transcription through increases in cAMP. However, PGI2 is produced at higher amounts than PGE2 during the first 15-30 min after activation by C. albicans. We were not successful in testing the EP2 receptor antagonist due to adverse effects on RPM. Although not addressed in this study, other eicosanoids such as LTC4 and arachidonic acid itself released by RPM in response to C. albicans could also influence macrophage activation. Arachidonic acid has been shown to suppress the expression of the complement receptor immunoglobulin (CRIg) during maturation of human monocytes to macrophages resulting in a decrease in the phagocytosis of opsonized C. albicans [95]. LTC4 could act through the CYSLT1 and CYSLT2 receptors expressed on RPM. For example these receptors promote calcium mobilization that may influence transcriptional responses due to cross-talk with cAMP signaling, and by potentiating cPLA2α activation [96]. Leukotrienes have been shown to promote uptake of C. albicans by alveolar macrophages and to enhance fungicidal activity [97]. It is possible that leukotrienes contribute to the enhanced C. albicans killing we observed in cPLA2α+/+ RPM compared to cPLA2α-/- RPM.

Microbial pathogens engage PRRs on macrophages that induce extensive effects on gene expression as we observed in C. albicans-stimulated RPM [39]. A characteristic of the "common host response" is increased expression of a large number of pro-inflammatory cytokines and chemokines that is important for the recruitment and activation of myeloid cells during infection [39]. Pro-inflammatory host defense responses are balanced by the activation of negative feedback loops that are important in dampening inflammation and potential tissue damage [21]. Our data suggest that cPLA2α activation and lipid mediator production represents one of the negative feedback loops since cPLA2α+/+ RPM exhibit lower expression of select pro-inflammatory genes such as Tnfα, Csf1, Ccl5, Cd40, Cx3cl1, Edn, Ifnγ and several IFNγ regulated GTP binding proteins, and higher expression of anti-inflammatory genes such as Il10, Socs3, Stat3, Fst, Thbd, Thsp1, Calca and CxCr7 than cPLA2α-/- RPM. Historically there has been an emphasis on the role of prostaglandins in mediating the cardinal signs of inflammation that is supported by the clinical effects of non-steroidal anti-inflammatory drugs. However, prostaglandins play an important role in suppressing inflammation and immune responses by acting through prostanoid receptors that increase cAMP resulting in PKA activation as supported by our results [18]. This pathway has immunosuppressive effects by inhibiting the differentiation of antigen presenting cells, lymphocyte activation and production of Th1 cytokines.

Our results show that the activation of cPLA2α and coupling to COX1 is an early response to C. albicans infection of RPM that can regulate the amplitude and timing of inflammation and host defense mechanisms as exemplified by the decrease in expression of Tnfα and increase of Il10. ERK activation and calcium mobilization are the signaling cascades activated by PRRs that are important for promoting IL10 production [69,98]. These are the signals required for optimal cPLA2α activation and eicosanoid production [99]. This cytokine signature is also a characteristic of resolution phase macrophages that contribute to restoration of normal tissue function by dampening inflammatory signals and the clearance of apoptotic neutrophils [100,101]. Resolution phase macrophages are characterized by the expression of COX2, decreased TNFα and increased IL10 production controlled by cAMP production. Prostaglandins and increases in cAMP contribute to the resolution phase by enhancing the ability of macrophages to phagocytose apoptotic neutrophils [102,103]. Activated and apoptotic neutrophils produce lyso-phosphatidylserine that acts through the macrophage G2A receptor to trigger an autocrine loop involving cPLA2α activation, PGE2 production, EP2 receptor-dependent increases in cAMP and PKA activation to enhance efferocytosis [102,104]. Therefore cPLA2α activation and prostaglandin production play a role in balancing host defense responses and the extent of inflammation in both the initiation and resolution phases of infection.

The results also indicate that cPLA2α-mediated prostaglandin production enhances the expression of certain pro-inflammatory genes, such as Csf3, that are important for host defense against C. albicans infection by promoting neutrophil function [40,41]. Prostaglandins also contribute to Candidiasis protection by promoting the Th17 response [105,106]. IL17 regulates neutrophil recruitment and is important for host defense to mucocutaneous Candidiasis [107110]. However if pro-inflammatory responses go unchecked prostaglandins contribute to chronic inflammation that is characteristic of cancer, and vascular and autoimmune diseases [111]. The ability of prostaglandins to promote the development of Th17 differentiation and production of IL17 contributes to chronic inflammation associated with autoimmune diseases [111,112]. COX2 is overexpressed in cancers and prostaglandins promote cancer development by regulating angiogenesis, cell migration, adhesion and invasiveness in part through promoting receptor specific increases in cAMP [57,58]. Several of the genes that are differentially expressed in cPLA2α+/+ RPM and cPLA2α-/- RPM (i.e. Gdf15, Eph2, Ereg, Areg, Lepr, Nr4a2, Runx2, Mmp13, CxCr7, Pdpn, Prox1) are positively or negatively regulated in cancers compared to normal tissue as a result of prostaglandins [5963,113]. Therefore, eicosanoids have complex biological effects depending on the tissue context, the specific receptors expressed on cells in the local environment and the timing of their production contributing to both anti- and pro-inflammation responses. Results from this study support an important role for cPLA2α activation early in response to microbial infection in resident tissue macrophages that helps to balance the expression of genes important for host defense and genes that contribute to inflammation.

Supporting Information

Table S1.

S1A and S1B Genes (A) increased and (B) decreased by C. albicans in wild type RPM.

https://doi.org/10.1371/journal.pone.0069002.s001

(DOC)

Table S2.

S2A and S2B Genes expressed at (A) lower and (B) higher levels in C. albicans-stimulated cPLA2α+/+ than cPLA2α-/- RPM.

https://doi.org/10.1371/journal.pone.0069002.s002

(DOC)

Acknowledgments

We thank Dr. Robert Barkley and Charis Uhlson for mass spectrometry analysis.

Author Contributions

Conceived and designed the experiments: CCL SS. Performed the experiments: SS SM. Analyzed the data: CCL SS LJS RCM. Contributed reagents/materials/analysis tools: GDB JVB DLW NARG RCM. Wrote the manuscript: CCL SS DLB.

References

  1. 1. Funk CD (2001) Prostaglandins and leukotrienes: advances in eicosanoid biology. Science 294: 1871-1875. doi:https://doi.org/10.1126/science.294.5548.1871. PubMed: 11729303.
  2. 2. Evans JF (2003) The cysteinyl leukotriene receptors. Prostaglandins Leukot Essent Fatty Acids 69: 117-122. doi:https://doi.org/10.1016/S0952-3278(03)00072-3. PubMed: 12895594.
  3. 3. Toda A, Yokomizo T, Shimizu T (2002) Leukotriene B4 receptors. Prostaglandins Other Lipid Mediat 68-69: 575-585. doi:https://doi.org/10.1016/S0090-6980(02)00056-4. PubMed: 12432944.
  4. 4. Woodward DF, Jones RL, Narumiya S (2011) International Union of Basic and Clinical Pharmacology. LXXXIII: Classification of prostanoid receptors, updating 15 years of progress. Pharmocol Rev 63: 471-538. doi:https://doi.org/10.1124/pr.110.003517.
  5. 5. Peters-Golden M, Canetti C, Mancuso P, Coffey MJ (2005) Leukotrienes: Underappreciated mediators of innate immune responses. J Immunol 173: 589-594. PubMed: 15634873.
  6. 6. Alvarez Y, Valera I, Municio C, Hugo E, Padrón F et al. (2010) Eicosanoids in the innate immune response: TLR and non-TLR routes. Mediat Inflamm, 2010, Article ID 201929, 14 pages. . PubMed: 20689730.
  7. 7. Norris PC, Reichart D, Dumlao DS, Glass CK, Dennis EA (2011) Specificity of eicosanoid production depends on the TLR-4-stimulated macrophage phenotype. J Leukoc Biol 90: 563-574. doi:https://doi.org/10.1189/jlb.0311153. PubMed: 21653236.
  8. 8. Noor S, Goldfine H, Tucker DE, Suram S, Lenz LL et al. (2008) Activation of cytosolic phospholipase A2α in resident peritoneal macrophages by Listeria monocytogenes involves listeriolysin O and TLR2. J Biol Chem 283: 4744-4755. PubMed: 18083708.
  9. 9. Qiu Z-H, de Carvalho MS, Leslie CC (1993) Regulation of phospholipase A2 activation by phosphorylation in mouse peritoneal macrophages. J Biol Chem 268: 24506-24513. PubMed: 8227003.
  10. 10. Qiu Z-H, Gijón MA, de Carvalho MS, Spencer DM, Leslie CC (1998) The role of calcium and phosphorylation of cytosolic phospholipase A2 in regulating arachidonic acid release in macrophages. J Biol Chem 273: 8203-8211. doi:https://doi.org/10.1074/jbc.273.14.8203. PubMed: 9525925.
  11. 11. Qiu Z-H, Leslie CC (1994) Protein kinase C-dependent and -independent pathways of mitogen-activated protein kinase activation in macrophages by stimuli that activate phospholipase A2. J Biol Chem 269: 19480-19487. PubMed: 8034717.
  12. 12. Gijón MA, Spencer DM, Siddiqi AR, Bonventre JV, Leslie CC (2000) Cytosolic phospholipase A2 is required for macrophage arachidonic acid release by agonists that do and do not mobilize calcium. Novel role of mitogen-activated protein kinase pathways in cytosolic phospholipase A2 regulation. J Biol Chem 275: 20146-20156.
  13. 13. Suram S, Brown GD, Ghosh M, Gordon S, Loper R et al. (2006) Regulation of cytosolic phospholipase A2 activation and cyclooxygeanse 2 expression in macrophages by the β-glucan receptor. J Biol Chem 9: 5506-5514.
  14. 14. Suram S, Gangelhoff TA, Taylor PR, Rosas M, Brown GD et al. (2010) Pathways regulating cytosolic phospholipase A2 activation and eicosanoid production in macrophages by Candida albicans. J Biol Chem 285: 30676-30685. doi:https://doi.org/10.1074/jbc.M110.143800. PubMed: 20643646.
  15. 15. Pfaller MA, Diekema DJ (2007) Epidemiology of invasive candidiasis: a persistent public health problem. Clin Microbiol Rev 20: 133-163. doi:https://doi.org/10.1128/CMR.00029-06. PubMed: 17223626.
  16. 16. Cheng SC, Joosten LA, Kullberg BJ, Netea MG (2012) Interplay between Candida albicans and the mammalian innate host defense. Infect Immun 80: 1304-1313. doi:https://doi.org/10.1128/IAI.06146-11. PubMed: 22252867.
  17. 17. Ricciotti E, Fitzgerald GA (2011) Prostaglandins and inflammation. Arterioscler Thromb Vasc Biol 31: 986-1000. doi:https://doi.org/10.1161/ATVBAHA.110.207449. PubMed: 21508345.
  18. 18. Kalinski P (2012) Regulation of immune responses by prostaglandin E2. J Immunol 18: 21-28. PubMed: 22187483.
  19. 19. Serhan CN (2007) Resolution phase of inflammation: Novel endogenous anti-inflammatory and proresolving lipid mediators and pathways. Annu Rev Immunol 25: 101-137. doi:https://doi.org/10.1146/annurev.immunol.25.022106.141647. PubMed: 17090225.
  20. 20. Goldszmid RS, Trinchieri G (2012) The price of immunity. Nat Immunol 13: 932-938. doi:https://doi.org/10.1038/ni.2422. PubMed: 22990891.
  21. 21. Ivashkiv LB (2011) Inflammatory signaling in macrophages: Transitions from acute to tolerant and alternative activation states. Eur J Immunol 41: 2470-2525. doi:https://doi.org/10.1002/eji.201141988. PubMed: 21952798.
  22. 22. Mosser DM, Edwards JP (2008) Exploring the full spectrum of macrophage activation. Nat Rev Immunol 8: 958-969. doi:https://doi.org/10.1038/nri2448. PubMed: 19029990.
  23. 23. Silva MT (2010) Neutrophils and macrophages work in concert as inducers and effectors of adaptive immunity against extracellular and intracellular microbial pathogens. J Leukoc Biol 87: 805-813. doi:https://doi.org/10.1189/jlb.1109767. PubMed: 20110444.
  24. 24. Bonventre JV, Huang Z, Taheri MR, O’Leary E, Li E et al. (1997) Reduced fertility and postischaemic brain injury in mice deficient in cytosolic phospholipase A2. Nature 390: 622-625. doi:https://doi.org/10.1038/37635. PubMed: 9403693.
  25. 25. Takeuchi O, Hoshino K, Kawai T, Sanjo H, Takada H et al. (1999) Differential roles of TLR2 and TLR4 in recognition of Gram-negative and Gram-positive bacterial cell wall components. Immunity 11: 443-451. doi:https://doi.org/10.1016/S1074-7613(00)80119-3. PubMed: 10549626.
  26. 26. Taylor PR, Tsoni SV, Willment JA, Dennehy KM, Rosas M et al. (2007) Dectin-1 is required for β-glucan recognition and control of fungal infection. Nat Immunol 8: 31-38. doi:https://doi.org/10.1038/ni1408. PubMed: 17159984.
  27. 27. Bates S, MacCallum DM, Bertram G, Munro CA, Hughes HB et al. (2005) Candida albicans Pmr1p, a secretory pathway P-type Ca2+/Mn2+-ATPase, is required for glycosylation and virulence. J Biol Chem 280: 23408-23415. doi:https://doi.org/10.1074/jbc.M502162200. PubMed: 15843378.
  28. 28. de Pedro MA, Young KD, Höltje J-V, Schwarz H (2003) Branching of Escherichia coli cells arises from multiple sites of inert peptidoglycan. J Bacteriol 185: 1147-1152. doi:https://doi.org/10.1128/JB.185.4.1147-1152.2003. PubMed: 12562782.
  29. 29. Gijón MA, Zarini S, Murphy RC (2007) Biosynthesis of eicosanoids and transcellular metabolism of leukotrienes in murine bone marrow cells. J Lipid Res 48: 716-725. PubMed: 17179116.
  30. 30. Hall LM, Murphy RC (1998) Electrospray mass spectrometric analysis of 5-hydroperoxy and 5-hydroxyeicosatetraenoic acids generated by lipid peroxidation of red blood cell ghost phospholipids. J Am Soc Mass Spectrom 9: 527-532. doi:https://doi.org/10.1016/S1044-0305(98)00013-0. PubMed: 9879367.
  31. 31. Huang DW, Sherman BT, Lempicki RA (2009) Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc 4: 44-57. PubMed: 19131956.
  32. 32. Livak KJ, Schmittgen TD (2001) Analysis of relative gene expression data using real time quantitative PCR and the 2-ΔΔCT method. Methods 25: 402-408. doi:https://doi.org/10.1006/meth.2001.1262. PubMed: 11846609.
  33. 33. Shinomiya S, Naraba H, Ueno A, Utsunomiya I, Maruyama T et al. (2001) Regulation of TNFα and interleukin-10 production by prostaglandins I2 and E2: studies with prostaglandin receptor-deficient mice and prostaglandin E-receptor subtype-selective synthetic agonists. Biochem Pharmacol 61: 1153-1160. doi:https://doi.org/10.1016/S0006-2952(01)00586-X. PubMed: 11301049.
  34. 34. Rouzer CA, Kingsley PJ, Wang H, Zhang H, Morrow JD et al. (2004) Cyclooxygenase-1-dependent prostaglandin synthesis modulates tumor necrosis factor-α secretion in lipopolysaccharide-challenged murine resident peritoneal macrophages. J Biol Chem 279: 34256-34268. doi:https://doi.org/10.1074/jbc.M402594200. PubMed: 15181007.
  35. 35. Netea MG, Gow NAR, Munro CA, Bates PA, Collins C et al. (2006) Immune sensing of Candida albicans requires cooperative recognition of mannans and glucans by lectin and Toll-like receptors. J Clin Invest 6: 1642-1650. PubMed: 16710478.
  36. 36. Chulada PC, Langenbach R (1997) Differential inhibition of murine prostaglandin synthase-1 and -2 by nonsteroidal anti-inflammatory drugs using exogenous and endogenous sources of arachidonic acid. J Pharmacol Exp Ther 280: 606-613. PubMed: 9023270.
  37. 37. Spengler RN, Spengler ML, Lincoln P, Remick DG, Strieter RM et al. (1989) Dynamics of dibutyryl cyclic AMP- and prostaglandin E2-mediated suppression of lipopolysaccharide-induced tumor necrosis factor alpha gene expression. Infect Immun 57: 2837-2841. PubMed: 2547721.
  38. 38. Stafford JB, Marnett LJ (2008) Prostaglandin E2 inhibits tumor necrosis factor-alpha RNA through PKA type I. Biochem Biophys Res Commun 366: 104-109. doi:https://doi.org/10.1016/j.bbrc.2007.11.091. PubMed: 18060853.
  39. 39. Jenner RG, Young RA (2005) Insights into host responses against pathogens from transcriptional profiling. Nat Rev Microbiol 3: 281-294. doi:https://doi.org/10.1038/nrmicro1126. PubMed: 15806094.
  40. 40. Basu S, Quilici C, Zhang HH, Grail D, Dunn AR (2008) Mice lacking both G-CSF and IL-6 are more susceptible to Candida albicans infection: critical role of neutrophils in defense against Candida albicans. Growth Factors 26: 23-34. doi:https://doi.org/10.1080/08977190801987513. PubMed: 18365876.
  41. 41. Vonk AG, Netea MG, van der Meer JWM, Kullberg BJ (2006) Host defence against disseminated Candida albicans infection and implications for antifungal immunotherapy. Expert Opin Biol Ther 6: 891-903. doi:https://doi.org/10.1517/14712598.6.9.891. PubMed: 16918256.
  42. 42. Degrandi D, Konermann C, Beuter-Gunia C, Kresse A, Würthner J et al. (2007) Extensive characterization of IFN-induced GTPases mGBP1 to mGBP10 involved in host defense. J Immunol 179: 7729-7740. PubMed: 18025219.
  43. 43. Vestal DJ, Jeyaratnam JA (2011) The guanylate-binding proteins: Emerging insights into the biochemical properties and functions of this family of large interferon-induced guanosine triphosphatase. J Interferon Cytokine Res 31: 89-97. doi:https://doi.org/10.1089/jir.2010.0102. PubMed: 21142871.
  44. 44. Kim B-H, Shenoy AR, Kumar P, Das R, Tiwari S et al. (2011) A family of IFN-γ-inducible 65-kD GTPases protects against bacterial infection. Science 332: 717-721. doi:https://doi.org/10.1126/science.1201711. PubMed: 21551061.
  45. 45. Hunn JP, Feng CG, Sher A, Howard JC (2011) The immunity-related GTPases in mammals: a fast-evolving cell-autonomous resistance system against intracellular pathogens. Mamm Genome 22: 43-54. doi:https://doi.org/10.1007/s00335-010-9293-3. PubMed: 21052678.
  46. 46. D’Haese JG, Friess H, Ceyhan GO (2012) Therapeutic potential of the chemokine-receptor duo fractalkine/CX3CR1: an update. Expert Opin Ther Targets 16: 613-618. doi:https://doi.org/10.1517/14728222.2012.682574. PubMed: 22530606.
  47. 47. Suttles J, Stout RD (2009) Macrophage CD40 signaling: A pivotal regulator of disease protection and pathogenesis. Semin Immunol 21: 257-264. doi:https://doi.org/10.1016/j.smim.2009.05.011. PubMed: 19540774.
  48. 48. Hu X, Ivashkiv LB (2009) Cross-regulation of signaling pathways by interferon-γ: Implications for immune responses and autoimmune diseases. Immunity 31: 539-550. doi:https://doi.org/10.1016/j.immuni.2009.09.002. PubMed: 19833085.
  49. 49. Siddiqa A, Sims-Mourtada JC, Guzman-Rojas L, Rangel R, Guret C et al. (2001) Regulation of CD40 and CD40 ligand by the AT-hook transcription factor AKNA. Nature 410: 383-387. doi:https://doi.org/10.1038/35066602. PubMed: 11268217.
  50. 50. Moliterno AR, Resar LMS (2011) AKNA: Another AT-hook transcription factor "hooking-up" with inflammation. Cell Res 21: 1528-1530. doi:https://doi.org/10.1038/cr.2011.96. PubMed: 21670742.
  51. 51. Briken V, Ruffner H, Schultz U, Schwarz A, Reis LF et al. (1995) Interferon regulatory factor 1 is required for mouse Gbp gene activation by gamma interferon. Mol Cell Biol 15: 975-982. PubMed: 7823961.
  52. 52. Savitsky D, Tamura T, Yanai H, Taniguchi T (2010) Regulation of immunity and oncogenesis by the IRF transcription factor family. Cancer Immunol Immunother 59: 480-510. PubMed: 20049431.
  53. 53. Wong MM, Fish EN (2003) Chemokines: attractive mediators of the immune response. Semin Immunol 15: 5-14. doi:https://doi.org/10.1016/S1044-5323(02)00123-9. PubMed: 12495636.
  54. 54. Qian X, Zhang J, Liu J (2011) Tumor-secreted PGE2 inhibits CCL5 production in activated macrophages through cAMP/PKA signaling pathway. J Biol Chem 286: 2111-2120. doi:https://doi.org/10.1074/jbc.M110.154971. PubMed: 21097507.
  55. 55. Hamilton JA (2008) Colony-stimulating factors in inflammation and autoimmunity. Nat Rev Immunol 8: 533-544. doi:https://doi.org/10.1038/nri2356. PubMed: 18551128.
  56. 56. Teder P, Noble PW (2000) A cytokine reborn? Endothelin-1 in pulmonary inflammation and fibrosis. Am J Respir Cell Mol Biol 23: 7-10. doi:https://doi.org/10.1165/ajrcmb.23.1.f192. PubMed: 10873147.
  57. 57. Wang D, DuBois RN (2010) Eicosanoids and cancer. Nat Rev Cancer 10: 181-193. doi:https://doi.org/10.1038/nrc2809. PubMed: 20168319.
  58. 58. Menter DG, DuBois RN (2012) Prostaglandins in cancer cell adhesion, migration, and invasion. Int J Cell Biol, 2012: 2012: 723419. PubMed: 22505934.
  59. 59. Du X, Baldwin C, Hooker E, Glorion P, Lemay S (2011) Basal and Src kinase-mediated activation of the EphA2 promoter requires a cAMP-responsive element but is CREB-independent. J Cell Biochem 112: 1268-1276. doi:https://doi.org/10.1002/jcb.23018. PubMed: 21344481.
  60. 60. Gupta GP, Nguyen DX, Chiang AC, Bos PD, Kim JY et al. (2007) Mediators of vascular remodelling co-opted for sequential steps in lung metastasis. Nature 446: 765-770. doi:https://doi.org/10.1038/nature05760. PubMed: 17429393.
  61. 61. Oshima H, Popivanova BK, Oguma K, Kong D, Ishikawa TO et al. (2011) Activation of epidermal growth factor receptor signaling by the prostaglandin E2 receptor EP4 pathway during gastric tumorigenesis. Cancer Sci 102: 713-719. doi:https://doi.org/10.1111/j.1349-7006.2011.01847.x. PubMed: 21205091.
  62. 62. Raica M, Cimpean AM, Ribatti D (2008) The role of podoplanin in tumor progression and metastasis. Anticancer Res 28: 2997-3006. PubMed: 19031946.
  63. 63. Sun X, Cheng G, Hao M, Zheng J, Zhou X et al. (2010) CXCL12/ CXCR4/ CXCR7 chemokine axis and cancer progression. Cancer Metastasis Rev 29: 709-722. doi:https://doi.org/10.1007/s10555-010-9256-x. PubMed: 20839032.
  64. 64. Mix KS, McMahon K, McMorrow JP, Walkenhorst DE, Smyth AM et al. (2012) Orphan nuclear receptor NR4A2 induces synoviocytes proliferation, invasion, and matrix metalloproteinase 13 transcription. Arthritis Rheum 64: 2126-2136. doi:https://doi.org/10.1002/art.34399. PubMed: 22275273.
  65. 65. Selvamurugan N, Shimizu E, Lee MS, Liu T, Li H et al. (2009) Identification and characterization of Runx2 phosphorylation sites involved in matrix metalloproteinase-13 promoter activation. FEBS Lett 583: 1141-1146. doi:https://doi.org/10.1016/j.febslet.2009.02.040. PubMed: 19264160.
  66. 66. Holla VR, Mann JR, Shi Q, DuBois RN (2006) Prostaglandin E2 regulates the nuclear receptor NR4A2 in colorectal cancer. J Biol Chem 281: 2676-2682. PubMed: 16293616.
  67. 67. Yoshimura A, Naka T, Kubo M (2007) SOCS proteins, cytokine signalling and immune regulation. Nat Immunol 7: 454-465. doi:https://doi.org/10.1038/nri2093. PubMed: 17525754.
  68. 68. Chung EY, Liu J, Homma Y, Zhang Y, Brendolan A et al. (2007) Interleukin-10 expression in macrophages during phagocytosis of apoptotic cells is mediated by homeodomain proteins Pbx1 and Prep-1. Immunity 37: 952-964.
  69. 69. Saraiva M, O’Garra A (2010) The regulation of IL-10 production by immune cells. Nat Immunol 10: 170-181. doi:https://doi.org/10.1038/nri2711. PubMed: 20154735.
  70. 70. El Kasmi KC, Smith AM, Williams L, Neale G, Panopolous A et al. (2007) A transcriptional repressor and corepressor induced by the STAT3-regulated anti-inflammatory signaling pathway. J Immunol 179: 7215-7219. PubMed: 18025162.
  71. 71. Hutchins AP, Poulain S, Miranda-Saavedra D (2012) Genome-wide analysis of STAT3 binding in vivo predicts effectors of the anti-inflammatory response in macrophages. Blood 119: e110-e119. doi:https://doi.org/10.1182/blood-2011-09-381483. PubMed: 22323479.
  72. 72. Carballo E, Lai WS, Blackshear PJ (1998) Feeback inhibition of macrophage tumor necrosis factor-α production by tristetraprolin. Science 281: 1001-1005. doi:https://doi.org/10.1126/science.281.5379.1001. PubMed: 9703499.
  73. 73. Feng CG, Beller EM, Bagga S, Boyce JA (2006) Human mast cells express multiple EP receptors for prostaglandin E2 that differentially modulate activation responses. Blood 107: 3243-3250. doi:https://doi.org/10.1182/blood-2005-07-2772. PubMed: 16357326.
  74. 74. Harzenetter MD, Novotny AR, Gais P, Molina CA, Altmayr F et al. (2007) Negative regulation of TLR responses by the neuropeptide CGRP is mediated by the transcriptional repressor ICER. J Immunol 179: 607-615. PubMed: 17579082.
  75. 75. Ma W, Dumont Y, Vercauteren F, Quirion R (2010) Lipopolysaccharide induces calcitonin gene-related peptide in the RAW264.7 macrophage cell line. Immunol 130: 399-409. doi:https://doi.org/10.1111/j.1365-2567.2009.03239.x. PubMed: 20141542.
  76. 76. Saijo K, Winner B, Carson CT, Collier JG, Boyer L et al. (2009) A Nurr1/CoREST pathway in microglia and astocytes protects dopaminergic neurons from inflammation-induced death. Cell 137: 47-59. doi:https://doi.org/10.1016/j.cell.2009.01.038. PubMed: 19345186.
  77. 77. de Kretser DM, O’Hehir RE, Hardy CL, Hedger MP (2012) The roles of activin A and its binding protein, follistatin, in inflammation and tissue repair. Mol Cell Endocrinol 359: 101-106. doi:https://doi.org/10.1016/j.mce.2011.10.009. PubMed: 22037168.
  78. 78. Okamoto T, Tanigami H, Suzuki K, Shimaoka M (2012) Thrombomodulin: A bifunctional modulator of inflammation and coagulation in sepsis. Crit Care Med Practice, 2012: 2012: 614545. PubMed: 22482044.
  79. 79. Doyen V, Rubio M, Braun D, Nakajima T, Abe J et al. (2003) Thrombospondin 1 is an autocrine negative regulator of human dendritic cell activation. J Exp Med 198: 1277-1283. doi:https://doi.org/10.1084/jem.20030705. PubMed: 14568985.
  80. 80. Anand RJ, Dai S, Gribar SC, Richardson W, Kohler JW et al. (2008) A role for connexin43 in macrophage phagocytosis and host survival after bacterial peritoneal infection. J Immunol 181: 8534-8543. PubMed: 19050272.
  81. 81. Dower K, Ellis DK, Saraf K, Jelinsky SA, Lin L-L (2008) Innate immune responses to TREM-1 activation: Overlap, divergence, and positive and negative cross-talk with bacterial lipopolysaccharide. J Immunol 180: 3520-3534. PubMed: 18292579.
  82. 82. Schneider E, Rolli-Derkinderen M, Arock M, Dy M (2002) Trends in histamine research: new functions during immune responses and hematopoiesis. Trends Immunol 23: 255-263. doi:https://doi.org/10.1016/S1471-4906(02)02215-9. PubMed: 12102747.
  83. 83. Miyazaki T, Ohgoh M, Ohmori E, Yamamoto J, Emoto S et al. (1992) Synergistic effects of cyclic AMP and Ca2+ ionophore A23187 on de novo synthesis of histidine decarboxylase in masocytoma P-815 cells. Biochim Biophys Acta 1133: 179-186. doi:https://doi.org/10.1016/0167-4889(92)90067-L. PubMed: 1310051.
  84. 84. Huang Z-L, Sato Y, Mochizuki T, Okada T, Qu W-M et al. (2003) Prostaglandin E2 activates the histaminergic system via the EP4 receptor to induce wakefulness in rats. J Neurosci 23: 5975-5983. PubMed: 12853415.
  85. 85. Tilley SL, Coffman TM, Koller BH (2001) Mixed messages: modulation of inflammation and immune responses by prostaglandins and thromboxanes. J Clin Invest 108: 15-23. doi:https://doi.org/10.1172/JCI13416. PubMed: 11435451.
  86. 86. Netea MG, Brown GD, Kullberg BJ, Gow NAR (2008) An integrated model of the recognition of Candida albicans by the innate immune system. Nat Rev Microbiol 6: 67-78. doi:https://doi.org/10.1038/nrmicro1815. PubMed: 18079743.
  87. 87. Barclay JL, Anderson ST, Waters MJ, Curlewis JD (2007) Characterization of the SOCS3 promoter response to prostaglandin E2 in T47D cells. Mol Endocrinol 21: 2516-2528. doi:https://doi.org/10.1210/me.2007-0030. PubMed: 17636039.
  88. 88. Xia X, Batra N, Shi Q, Bonewald LF, Sprague E et al. (2010) Prostaglandin promotion of osteocyte gap junction function through transciptional regulation of connexin 43 by glycogen synthase kinase 3/β-catenin signaling. Mol Cell Biol 30: 206-219. doi:https://doi.org/10.1128/MCB.01844-08. PubMed: 19841066.
  89. 89. Alvarez Y, Municio C, Alonso S, Sánchez Crespo M, Fernández N (2009) The induction of IL-10 by zymosan in dendritic cells depends on CREB activation by the coactivators CREB-binding protein and TORC2 and autocrine PGE2. J Immunol 183: 1471-1479. doi:https://doi.org/10.4049/jimmunol.0900312. PubMed: 19564345.
  90. 90. Kainoh M, Maruyama I, Nishio S, Nakadate T (1991) Enhancement by beraprost sodium, a stable analogue of prostacyclin, in thrombomodulin expression on membrane surface of cultured vascular endothelial cells via increase in cyclic AMP level. Biochem Pharmacol 41: 1135-1140. doi:https://doi.org/10.1016/0006-2952(91)90651-K. PubMed: 1706920.
  91. 91. Le Goff W, Zheng P, Brubaker G, Smith JD (2006) Identification of the cAMP-responsive enhancer of the murine ABCA1 gene. Requirement for CREB1 and STAT3/4 elements. Arterioscler Thromb Vasc Biol 26: 527-533.
  92. 92. Clarke DL, Belvisi MG, Smith SJ, Hardaker E, Yacoub MH et al. (2005) Prostanoid receptor expression by human airway smooth muscle cells and regulation of the secretion of granulocyte colony-stimulating factor. Am J Physiol Lung Cell Mol Physiol 288: L238-L250. PubMed: 15640521.
  93. 93. Murakami Y, Kohsaka H, Kitasato H, Akahoshi T (2007) Lipopolysaccharide-induced up-regulation of triggering receptor expressed on myeloid cells-1 expression on macrophages is regulated by endogenous prostaglandin E2. J Immunol 178: 1144-1150. PubMed: 17202378.
  94. 94. Deacon K, Knox AJ (2010) Endothelin-1 (ET-1) increases the expression of remodeling genes in vascular smooth muscle through linked calcium and cAMP pathways: role of a phospholipase A2 (cPLA2)/cyclooxygenase-2 (COX-2)/prostacyclin receptor-dependent autocrine loop. J Biol Chem 285: 25913-25927. doi:https://doi.org/10.1074/jbc.M110.139485. PubMed: 20452970.
  95. 95. Gorgani NN, Thathaisong U, Mukaro VR, Poungpair O, Tirimacco A et al. (2011) Regulation of CRIg expression and phagocytosis in human macrophages by arachidonate, dexamethasone, and cytokines. Am J Pathol 179: 1310-1318. doi:https://doi.org/10.1016/j.ajpath.2011.05.021. PubMed: 21741936.
  96. 96. Kanaoka Y, Boyce JA (2004) Cysteinyl leukotrienes and their receptors: Cellular distribution and function in immune and inflammatory responses. J Immunol 173: 1503-1510. PubMed: 15265876.
  97. 97. Morato-Marques M, Campos MR, Kane S, Rangel AP, Lewis C et al. (2011) Leukotrienes target F-actin/cofilin-1 to enhance alveolar macrophage anti-fungal activity. J Biol Chem 286: 28902-28913. doi:https://doi.org/10.1074/jbc.M111.235309. PubMed: 21715328.
  98. 98. Kelly EK, Wang L, Ivashkiv LB (2010) Calcium-activated pthways and oxidative burst mediate zymosan-induced signaling and IL-10 production in human macrophages. J Immunol 184: 5545-5552. doi:https://doi.org/10.4049/jimmunol.0901293. PubMed: 20400701.
  99. 99. Ghosh M, Tucker DE, Burchett SA, Leslie CC (2006) Properties of the Group IV phospholipase A2 family. Prog Lipid Res 45: 487-510. doi:https://doi.org/10.1016/j.plipres.2006.05.003. PubMed: 16814865.
  100. 100. Bystrom J, Evans I, Newson J, Stables M, Toor I et al. (2008) Resolution-phase macrophages possess a unique inflammatory phenotype that is controlled by cAMP. Blood 112: 4117-4127. doi:https://doi.org/10.1182/blood-2007-12-129767. PubMed: 18779392.
  101. 101. Medeiros AI, Serezani CH, Lee SP, Peters-Golden M (2009) Efferocytosis impairs pulmonary macrophage and lung antibacterial function via PGE2/EP2 signaling. J Exp Med 16: 61-68. PubMed: 19124657.
  102. 102. Frasch SC, Fernandez-Boyanapalli RF, Berry KZ, Leslie CC, Bonventre JV et al. (2011) Signaling via macrophage G2A enhances efferocytosis of dying neutrophils by augmentation of Rac activity. J Biol Chem 286: 12108-12122. doi:https://doi.org/10.1074/jbc.M110.181800. PubMed: 21297111.
  103. 103. Frasch SC, Bratton DL (2012) Emerging roles of lysophosphatidylserine in resolution of inflammation. Prog Lipid Res 51: 199-207. doi:https://doi.org/10.1016/j.plipres.2012.03.001. PubMed: 22465125.
  104. 104. Frasch SC, Berry KZ, Fernandez-Boyanapalli R, Jin HS, Leslie CC et al. (2008) NADPH oxidase-dependent generation of lysophosphatidylserine enhances clearance of activated and dying neutrophils via G2A. J Biol Chem 283: 33736-33749. doi:https://doi.org/10.1074/jbc.M807047200. PubMed: 18824544.
  105. 105. Smeekens SP, van de Veerdonk FL, van der Meer JW, Kullberg BJ, Joosten LA et al. (2010) The Candida Th17 response is dependent on mannan- and beta-glucan-induced prostaglandin E2. Int Immunol 22: 889-895. doi:https://doi.org/10.1093/intimm/dxq442. PubMed: 21059767.
  106. 106. Gagliardi MC, Teloni R, Mariotti S, Bromuro C, Chiani P et al. (2010) Endogenous PGE2 promotes the induction of human Th17 responses by fungal β-glucan. J Leukoc Biol 88: 947-954. doi:https://doi.org/10.1189/jlb.0310139. PubMed: 20807707.
  107. 107. Ouyang W, Kolls JK, Zheng Y (2008) The biological functions of T helper 17 cell effector cytokines in inflammation. Immunity 28: 454-467. doi:https://doi.org/10.1016/j.immuni.2008.03.004. PubMed: 18400188.
  108. 108. Ferwerda B, Ferwerda G, Plantinga TS, Willment JA, van Spriel AB et al. (2009) Human dectin-1 deficiency and mucocutaneous fungal infections. N Engl J Med 361: 1760-1767. doi:https://doi.org/10.1056/NEJMoa0901053. PubMed: 19864674.
  109. 109. Conti HR, Shen F, Nayyar N, Stocum E, Sun JN et al. (2009) Th17 cells and IL-17 receptor signaling are essential for mucosal host defense against oral candidiasis. J Exp Med 206: 299-311. doi:https://doi.org/10.1084/jem.20081463. PubMed: 19204111.
  110. 110. Puel A, Cypowyj S, Bustamante J, Wright JF, Liu L et al. (2011) Chronic mucocutaneous candidiasis in humans with inborn errors of interleukin-17 immunity. Science 332: 65-68. doi:https://doi.org/10.1126/science.1200439. PubMed: 21350122.
  111. 111. Aoki T, Narumiya S (2012) Prostaglandins and chronic inflammation. Trends Pharmacol Sci 33: 304-311. doi:https://doi.org/10.1016/j.tips.2012.02.004. PubMed: 22464140.
  112. 112. Yao C, Sakata D, Esaki Y, Li Y, Matsuoka T et al. (2009) Prostaglandin E2-EP4 signaling promotes immune inflammation through TH1 cell differentiation and TH17 cell expansion. Nat Med 15: 633-640. doi:https://doi.org/10.1038/nm.1968. PubMed: 19465928.
  113. 113. Iguchi G, Chrysovergis K, Lee S-H, Baek SJ, Langenbach R et al. (2009) A reciprocal relationship exists between non-steroidal anti-inflammatory drug-activated gene-1 (NAG-1) and cyclooxygenase-2. Cancer Lett 282: 152-158. doi:https://doi.org/10.1016/j.canlet.2009.03.006. PubMed: 19375854.