Skip to main content

Main menu

  • Home
  • Articles
    • Current Issue
    • Next in The JI
    • Archive
    • Brief Reviews
      • Neuroimmunology: To Sense and Protect
    • Pillars of Immunology
    • Translating Immunology
    • Most Read
    • Top Downloads
    • Annual Meeting Abstracts
  • COVID-19/SARS/MERS Articles
  • Info
    • About the Journal
    • For Authors
    • Journal Policies
    • Influence Statement
    • For Advertisers
  • Editors
  • Submit
    • Submit a Manuscript
    • Instructions for Authors
    • Journal Policies
  • Subscribe
    • Journal Subscriptions
    • Email Alerts
    • RSS Feeds
    • ImmunoCasts
  • More
    • Most Read
    • Most Cited
    • ImmunoCasts
    • AAI Disclaimer
    • Feedback
    • Help
    • Accessibility Statement
  • Other Publications
    • American Association of Immunologists
    • ImmunoHorizons

User menu

  • Subscribe
  • My alerts
  • Log in
  • Log out

Search

  • Advanced search
The Journal of Immunology
  • Other Publications
    • American Association of Immunologists
    • ImmunoHorizons
  • Subscribe
  • My alerts
  • Log in
  • Log out
The Journal of Immunology

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Next in The JI
    • Archive
    • Brief Reviews
    • Pillars of Immunology
    • Translating Immunology
    • Most Read
    • Top Downloads
    • Annual Meeting Abstracts
  • COVID-19/SARS/MERS Articles
  • Info
    • About the Journal
    • For Authors
    • Journal Policies
    • Influence Statement
    • For Advertisers
  • Editors
  • Submit
    • Submit a Manuscript
    • Instructions for Authors
    • Journal Policies
  • Subscribe
    • Journal Subscriptions
    • Email Alerts
    • RSS Feeds
    • ImmunoCasts
  • More
    • Most Read
    • Most Cited
    • ImmunoCasts
    • AAI Disclaimer
    • Feedback
    • Help
    • Accessibility Statement
  • Follow The Journal of Immunology on Twitter
  • Follow The Journal of Immunology on RSS

Regulation of TLR2 Expression by Prostaglandins in Brain Glia

Hee Jung Yoon, Sae-Bom Jeon, In-Hoo Kim and Eun Jung Park
J Immunol June 15, 2008, 180 (12) 8400-8409; DOI: https://doi.org/10.4049/jimmunol.180.12.8400
Hee Jung Yoon
*Immune and Cell Therapy Branch, National Cancer Center, Goyang, Korea;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Sae-Bom Jeon
*Immune and Cell Therapy Branch, National Cancer Center, Goyang, Korea;
†Neuroscience Graduate Program, School of Medicine, Ajou University, Suwon, Korea; and
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
In-Hoo Kim
‡Molecular Imaging and Therapy Branch, National Cancer Center, Goyang, Korea
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Eun Jung Park
*Immune and Cell Therapy Branch, National Cancer Center, Goyang, Korea;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

TLR have emerged as important primary sensors for diverse stimuli and are increasingly implicated in various diseases. However, the molecular mechanisms underlying the regulation of the TLR system remain poorly understood. In this study, we report that some PGs may control TLR-mediated inflammatory events through modulation of TLR2 expression in brain immune cells. We first found that 15-deoxy-Δ12,14-PG J2 (15d-PGJ2) markedly altered the expression of TLR2 but not TLR4, TLR1, and TLR9 at the message and protein levels in activated glia. Down-regulation of TLR2 expression and downstream events of TLR2 activation, including phagocytosis by 15d-PGJ2, were also observed in cells treated with representative TLR2 ligands such as lipoteichoic acid and Pam3CSK4. We further revealed that certain 15d-PGJ2-related PGs such as 15d-PGD2 and PGD2 also suppressed the ligand-stimulated increase of TLR2 expression, whereas PGE2 and arachidonic acids did not. Interestingly, TLR2 expression was down-regulated even when such PGs were added at several hours after stimulator treatment. These findings appear to be independent of peroxisome proliferator-activated receptor γ and D prostanoid receptors (DPs) because potent synthetic peroxisome proliferator-activated receptor γ agonists, selective DP1 agonist, or DP2 agonist did not mimic the effects of such PGs on TLR2 expression. Taken together, our results suggest that 15d-PGJ2, 15d-PGD2, and PGD2 may play notable roles as modulators of the TLR2-mediated inflammatory events, and provide new insight into the resolution of inflammation in the brain.

Toll-like receptors, the mammalian homologs of the Drosophila Toll receptor, are key components of the immune system against pathogen attack. The TLRs are mainly expressed on the cells at the initial line of host defense, such as neutrophils, macrophages, and dendritic cells, where they act as primary sensors by recognizing a diverse range of endogenous and exogenous stimuli (1, 2, 3, 4, 5). To date, 13 mammalian TLRs have been identified and functionally characterized in mammals. Each TLR recognizes a distinct ligand and elicits different, but sometimes overlapping, immune responses (5). They use distinct combinations of adapter molecules and intracellular signaling molecules, thereby resulting in robust production or induction of inflammation-associated molecules that could influence both innate and adaptive immunity (6, 7).

Although the TLR-mediated inflammation is a critical response of the host to defend itself against pathological stimuli, excessive inflammatory signaling can be detrimental to the host, as is the case in endotoxin shock (8). Recent studies have shown that multiple mechanisms tightly regulate TLR-mediated responses to ensure appropriate response to pathogens and protect the host from a fatal response to pathogenic environments (5, 8). It has been reported that TLRs are negatively regulated at several levels, including down-regulation of TLR expression, sequestration of signaling molecules, blockade of their recruitment, degradation of target proteins, and inhibition of transcription (7). Several molecules, including A20, IL-1R-associated kinase-M, Toll-interacting protein, suppressor of cytokine signaling (SOCS)3 1, and PDZ-LIM domain protein 2, have been proposed to negatively regulate TLR signaling (9, 10, 11, 12, 13, 14). However, the precise mechanisms controlling the balance of TLR-mediated responses to pathogenic conditions are not yet fully understood.

Microglia and astrocytes are the resident immunoeffector cells of the CNS. Although these cells are quiescent under normal conditions, they are rapidly activated in response to pathological stimuli. Upon activation, astrocytes and microglia change their morphology, immunophenotype, and expression pattern of inflammatory mediators, which leads to immune and inflammatory responses (15, 16). These inflammatory mediators include PGs, a family of bioactive lipid mediators that are produced via the metabolism of arachidonic acid (17, 18). Arachidonic acid is converted to PGH2, which is subsequently converted to PGD2, PGE2, PGF2α, prostacyclin, or thromboxane A2. They bind to specific G protein-coupled receptors, designated D prostanoid receptors (DPs; for D PG receptor), EP, FP, IP, and TP receptor, respectively (19, 20).

PGs have long been considered to be potent proinflammatory regulators, and blockade of PG synthesis and activity has been a widely accepted treatment for inflammation-associated diseases. However, recent increasing reports have revealed that PGs can exert both toxic and paradoxically protective roles during inflammation (21). In particular, some PGs are shown to be increased during the resolution phase of inflammation and to alleviate inflammation in animal models (22, 23, 24). PGD2 and its metabolites reduced inflammatory events in several inflammation-associated disease models including asthma, allergic, and lung inflammation. Moreover, deficient production of PGs by genetic deficiency or by drugs such as cyclooxygenase (COX) inhibitors even worsened the inflammatory conditions (25, 26, 27, 28). In the CNS, PGD2 and 15-deoxy-Δ12,14-PG J2 (15d-PGJ2), a cyclopentenone PG derived from PGD2, showed protective effects on neuronal injury and suppressed the clinical features of several neuronal diseases, thus presenting as a novel therapeutic target for neurological diseases (29, 30, 31, 32, 33, 34). These opposing properties of some PGs have recently received much attention as targets for resolving inflammation.

In this report, we provide new insight into the anti-inflammatory actions of 15d-PGJ2, PGD2, and 15d-PGD2 based on their regulatory effects on TLR2 signaling in brain resident immune cells. We find that these PGs may regulate TLR2-mediated inflammatory responses by modulating TLR2 expression, but not TLR4, TLR1, and TLR9, independent of peroxisome proliferator-activated receptor γ (PPARγ) and DPs in activated glia. Our findings suggest not only a novel molecular explanation for the anti-inflammatory actions of these PGs but also suggest a possible new therapeutic intervention for TLR2-associated inflammatory diseases.

Materials and Methods

Reagents

Salmonella typhimurium LPS was purchased from Sigma-Aldrich. Purified lipoteichoic acid (LTA)-streptavidin and CpG oligodeoxynucleotide (ODN2216) were purchased from InvivoGen. Pam3Cys-Ser-(Lys)4 (Pam3CSK4) was purchased from Calbiochem. 15d-PGJ2, 15d-PGD2, PGD2, PGE2, DK-PGD2, BW245C, pioglitazone, and rosiglitazone were obtained from Cayman Chemical. DMEM and MEM were obtained from Invitrogen Life Technologies. The Abs used in this study included mouse anti-α-tubulin (Sigma-Aldrich), mouse anti-TLR2 (R&D Systems), TLR1 (Abcam), and rabbit anti-inducible NO synthase (anti-iNOS; Santa Cruz Biotechnology).

Cell culture

Primary microglia and astrocytes were cultured from the cerebral cortices of 1- to 3-day-old ICR mice or Sprague-Dawley rats (Samtaco). Briefly, cortices were triturated into single cells in MEM (Sigma-Aldrich) containing 10% FBS (HyClone), plated in 75-cm2 T flasks (1 hemisphere/flask for mice, 0.5 hemisphere/flask for rat) and incubated for 2 wk. The microglia were detached from the flasks by mild shaking and applied to a nylon mesh to remove astrocytes and cell clumps. Cells were plated in 6-well plates (5 × 105 cells/well), 60-mm2 dishes (8 × 105 cells/dish), or 100-mm2 dishes (2 × 106 cells/dish). One hour later, the cells were washed to remove unattached cells before being used in experiments. Following removal of the microglia, primary astrocytes were prepared using trypsinization. The BV2 murine microglia was cultured in DMEM (Invitrogen Life Technologies) supplemented with 5% FBS (HyClone). Mouse peritoneal macrophages were obtained from sterile lavage of the peritoneal cavity with cold PBS (pH 7.4). The lavage fluid was centrifuged at 1000 × g for 10 min. The cell pellet was resuspended in DMEM containing 10% FBS, followed by plating in a 60-mm2 dishes, and then incubation at 37°C. Nonadherent cells were removed by repeated washing after 2 h. The adherent cells (macrophages) were cultured overnight.

RT-PCR analysis

Total RNA was extracted using RNAzol B (Tel-Test) and cDNA was prepared using reverse transcriptase that originated from avian myeloblastosis virus (Takara), according to the manufacturer’s instructions. PCR was performed with 25 cycles of sequential reactions: 94°C for 30 s, 55°C for 30 s, and 72°C for 30 s. Oligonucleotide primers were purchased from Bioneer. The sequences of PCR primers were as follows: reverse (R): 5′-GTA GCC CAC GTC GTA GCA AA-3′, forward (F): 5′-TCC CTC AAG ATT GTC AGC AA-3′ for TLR2; (R) 5′-AAA CGC AAA CCT TAC CAG-3′, (F) 5′-CCG AGA ACC GCT CAA C-3′ for TLR1; and (R) 5′-GCC ATC TCC TGC TCG AAG TCT AG-3′, (F) 5′-CAT GTT TGA GAC CTT CAA CAC CCC-3′ for actin. PCR products were separated by electrophoresis in a 1.5% agarose gel and detected under UV light.

Western blot analysis

Cells were washed twice with cold PBS, and then lysed in ice-cold modified RIPA buffer (50 mM Tris-HCl (pH 7.4), 1% Nonidet P-40, 0.25% sodium deoxycholate, 150 mM NaCl, 1 mM Na3VO4 and 1 mM NaF) containing protease inhibitors (2 mM PMSF, 100 μg/ml leupeptin, 10 μg/ml pepstatin, 1 μg/ml aprotinin, and 2 mM EDTA). The lysates were centrifuged at 12,000 × g for 10 min at 4°C and the supernatant was collected. Proteins were separated by SDS-PAGE and transferred to a nitrocellulose membrane, which was incubated with primary Abs and peroxidase-conjugated secondary Abs (Vector Laboratories). The results were visualized using an ECL system (Sigma-Aldrich).

Flow cytometric analysis

Cells were washed twice with PBS containing 1% FBS, collected, and stained with FITC-conjugated anti-mouse TLR2, PE-conjugated anti-mouse TLR4/MD2 Ab, or isotype control Ab (eBioscience) for 1 h at 4°C. After washing, the cells were analyzed with a FACSVantage (BD Biosciences). To stain intracellular TLR9, IFN-γ (5 U/ml)-primed cells were treated with CpG (5 μM) in the presence or absence of 15d-PGJ2, fixed in 2% paraformaldehyde, and washed with ice-cold FACS staining buffer containing 2.5% saponin on ice for 20 min. Then, the cells were stained with FITC-conjugated anti-mouse TLR9 (eBioscience) for 30 min at 4°C. The data analyses were done with CellQuest software (BD Bioscience) and Win-MDI software (The Scripps Research Institute, La Jolla, CA).

Phagocytosis assay

BV2 cells were plated in 60-mm2 dishes (2.5 × 104 cells/dish) or were left untreated with LPS, purified LTA, or Pam3CSK4 for 18 h. Phagocytic capacity was measured by incubating cells with FITC-conjugated phagocytic beads (8 × 106 beads/ml; FluoSpheres polystyrene microspheres; Molecular Probes) at 37°C. Subsequently, cells were washed three times with PBS and then gently removed from the wells using cell scrapers. To visualize the effect of PGs on phagocytosis, cells containing FITC-conjugated phagocytic beads were analyzed by fluorescence microscopy (Zeiss).

Immunofluorescence and confocal microscopy

BV2 cells were plated onto 12-mm round coverslips (Fisher Scientific), and then untreated or treated with LPS, LTA, or Pam3CSK4 in the presence or absence of 15d-PGJ2, and fixed with ice-cold 100% methanol. The cells were permeabilized with PBS containing 0.1% Triton X-100 and blocked with 1% BSA (Sigma-Aldrich). The samples were then washed with PBS containing 0.1% Triton X-100, and incubated overnight at 4°C with TLR2 Ab and then secondary Ab. 4′,6′-Diamidino-2-phenylindole hydrochloride (DAPI) was used to stain nuclei. Images were obtained with a Carl Zeiss LSM510 confocal microscope, and were analyzed by LSM software (release 3.2).

Statistical analysis

All data were expressed as the mean ± SD and analyzed by one-way ANOVA followed by post-hoc comparisons (Student-Newman-Keuls test) using Statistical Package for Social Sciences 8.0 (SPSS).

Results

Ligand-dependent change of TLR2 expression is notably modulated by 15d-PGJ2 in activated microglia

In an effort to explore the regulatory mechanism underlying TLR-mediated inflammation, we investigated the effect of 15d-PGJ2 on TLR-associated inflammatory events in brain immune cells. First, we examined the action of 15d-PGJ2 on cell surface expression of TLR2 and 4 because their expression is strictly regulated in response to endogenous and exogenous stimuli (35, 36). BV2 murine microglial cells were treated with Salmonella typhimurium LPS in the presence or absence of 2.5 μM 15d-PGJ2, and the cell surface expression levels of TLR2 and TLR4 were determined by flow cytometric analysis. When cells were stimulated with LPS, the cell surface expression levels of both TLR2 and TLR4 were markedly altered in BV2 microglial cells, with LPS treatment stimulating the up- and down-regulation of TLR2 and TLR4, respectively. Interestingly, LPS-stimulated enhancement of TLR2 expression was significantly attenuated by pretreatment with 15d-PGJ2. 15d-PGJ2 considerably reduced the LPS-stimulated increase of TLR2 expression even at concentrations <0.5 μM, with further suppression seen in response to increasing concentrations of 15d-PGJ2 (Fig. 1⇓A). In contrast, no significant changes of TLR4 expression were observed under these conditions. The LPS-dependent decrease of TLR4 expression was not affected by exposure to 15d-PGJ2 at any of the tested concentrations. Moreover, we also observed that 15d-PGJ2 markedly altered the change of TLR2 expression, whereas it did not affect TLR4 expression in cells treated with ganglioside, an endogenous ligand for TLR2 and TLR4 (Refs. 37 and 38 ; Fig. 1⇓C). These results further support the suppressive effects of 15d-PGJ2 on the ligand-stimulated induction of TLR2 expression.

FIGURE 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 1.

15d-PGJ2 effectively attenuates the ligand-dependent enhancement of TLR2 cell surface expression in microglia. BV2 microglial cells were pretreated with 15d-PGJ2 or control medium for 1 h, and then left untreated (control) or treated with 100 ng/ml LPS (A and B), 25 μg/ml brain ganglioside mixture (Gmix, C), 1 μg/ml CpG (D), 10 μg/ml LTA (E), or 100 ng/ml Pam3CSK4 (E) for 12 h. TLR2, TLR4, or TLR9 expression was analyzed by flow cytometric analysis using FITC-conjugated anti-mouse TLR2, PE-conjugated anti-mouse TLR4, or FITC-conjugated anti-mouse TLR9 mAbs, respectively. Data shown are representative of at least four independent experiments.

The enhancement of TLR2 expression by other representative TLR2 agonists were also suppressed in the presence of 15d-PGJ2

To further explore the action of 15d-PGJ2 on TLR systems, we next examined whether 15d-PGJ2 could modulate TLR9 and TLR1 expressions. As shown in Fig. 1⇑D, TLR9 expression was increased after exposure to CpG, a ligand for TLR9. However, 15d-PGJ2 did not change the enhancement of TLR9 expression by CpG at any of the concentrations tested. Furthermore, we did not observe any effects of 15d-PGJ2 on TLR1 expression (data not shown). These results raised the possibility that 15d-PGJ2 may modulate the expression level of TLR2, but not TLR1, 4, and 9, in activated glia, thus regulating TLR2-mediated inflammatory responses.

To address this issue, we examined whether 15d-PGJ2 could also influence the enhancement of TLR2 expression induced by other representative TLR2 ligands such as LTA and Pam3CSK4. Consistent with the above results, the enhancement of TLR2 expression triggered by both LTA and Pam3CSK4 was significantly lower in 15d-PGJ2-treated cells compared with the untreated control (Fig. 1⇑E). In all cells treated with LTA, Pam3CSK4, or LPS, we observed the 15d-PGJ2-dependent reduction of TLR2 cell surface expression within 3 h after stimulator treatment (data not shown). These results provide strong evidence for the modulation of TLR2 cell surface expression by 15d-PGJ2 in TLR2 ligand-stimulated microglia, suggesting that 15d-PGJ2 may act to regulate TLR2-mediated inflammatory events through modulation of TLR2 expression level in brain microglia.

15d-PGJ2 also attenuates the expression of TLR2 at message and protein levels as well as cell surface level

TLR2 plays a critical role as a signal-transducing molecule for a large repertoire of exogenous and endogenous ligands, and it might contribute to the initiation and progression of CNS pathology (39, 40, 41, 42). To visualize the effect of 15d-PGJ2 on TLR2 expression, we performed immunofluorescence microscopy using TLR2 Ab. As compared with untreated control cells, TLR2 expression was considerably increased in cells within 3 h of LTA treatment, but this enhancement was apparently attenuated by treatment with 15d-PGJ2 (Fig. 2⇓A). Similar results were also observed in cells with Pam3CSK4 or LPS (data not shown).

FIGURE 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 2.

Message and protein levels of TLR2 are also affected by 15d-PGJ2. A, BV2 cells were left untreated (control) or treated with 10 μg/ml LTA in the absence or presence of 2.5 μM 15d-PGJ2, and then analyzed by immunofluorescence with anti-TLR2. DAPI, Nuclear staining. Original magnification, ×400. All images are representative of at least three independent experiments. B, Cells were left untreated or treated with 100 ng/ml LPS or 100 ng/ml Pam3CSK4 for 6 h following pretreatment with the indicated concentrations of 15d-PGJ2 for 1 h. Total RNA was then extracted for RT-PCR analysis. C, Cells were treated with 100 ng/ml LPS, 10 μg/ml LTA, or 100 ng/ml Pam3CSK4 for 12 h in the absence or presence of 2.5 μM 15d-PGJ2, followed by Western blot analysis using TLR2 and α-tubulin Abs. Data shown are representative of at least three independent experiments.

To further investigate the action of 15d-PGJ2 on TLR2 expression, we examined the effects of 15d-PGJ2 treatment on the message levels of TLR2 by RT-PCR analysis. As shown in Fig. 2⇑B, TLR2 message levels were markedly and concentration-dependently reduced by 15d-PGJ2. However, LPS-stimulated induction of TLR1 transcript was not influenced by 15d-PGJ2. TLR2 protein levels were also influenced by treatment with 15d-PGJ2. 15d-PGJ2 apparently suppressed the induction of TLR2 protein levels in all cells treated with LPS, LTA, or Pam3CSK4 (Fig. 2⇑C). Taken together, these results consistently indicated that 15d-PGJ2 modulates the expression of TLR2 at the message and protein levels as well as cell surface levels following treatment of cells with TLR2 ligands, further supporting the modulation of TLR2 expression by 15d-PGJ2.

Downstream events of TLR2 activation are significantly affected by treatment of glia with 15d-PGJ2

Activation of TLR2 cause triggering complex signaling cascades, thereby resulting in robust production of inflammation-associated molecules including cytokines and NO (40, 43). Having determined that 15d-PGJ2 regulates the expression level of TLR2, we next carefully investigated the functional importance of this effect by testing the action of 15d-PGJ2 on TLR2 ligand-stimulated phagocytosis. Phagocytosis is accompanied by secretion of inflammatory mediators, and TLR2 has been closely implicated in phagocytic events (5). The phagocytic response of BV2 cells was measured using FITC-conjugated fluorescent beads. Cells were treated with or without LPS, LTA, or Pam3CSK4 for 15 h, followed by incubation with FITC-conjugated beads for 1 h. The uptake of fluorescent beads by microglia was then determined by flow cytometry. Treatment of microglia with LPS, LTA, or Pam3CSK4 considerably increased the cellular uptake of fluorescent beads, and these enhancements were apparently decreased in the presence of 15d-PGJ2 (Fig. 3⇓, A and B, and data not shown). Using fluorescence microscopy, we also observed the effects of 15d-PGJ2 on TLR2 ligand-dependent phagocytosis. Compared with untreated control cells, LPS- or Pam3-treated cells were somewhat flattened and aggregated, where the uptake of fluorescent beads was increased. However, in the presence of 15d-PGJ2, the uptake of fluorescent beads by microglia was significantly attenuated (Fig. 3⇓C). Furthermore, we found that 15d-PGJ2 significantly suppressed the transcription of MCP-1 and TNF-α, iNOS expression, and NO release in cells treated with LTA or Pam3CSK4 (data not shown). Overall, these results show that not only TLR2 expression but also TLR2-mediated inflammatory events are suppressed by treatment of 15d-PGJ2, and convincingly suggest that 15d-PGJ2 may regulate the activation of microglia, at least in part, by down-regulating TLR2 expression.

FIGURE 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 3.

15d-PGJ2 does affect TLR2 ligand-stimulated phagocytosis. BV2 microglial cells were incubated with 100 ng/ml LPS (A), 10 μg/ml LTA (B), or 100 ng/ml Pam3CSK4 (C) for 15 h in the absence or presence of 2.5 μM 15d-PGJ2. The cells were then incubated with FITC-conjugated fluorescent beads for 1 h, and the cellular uptake of the fluorescent bead was determined by flow cytometric analysis and fluorescence microscopy as described in Materials and Methods. The mean fluorescence intensity (MFI) values are the mean ± SD of three independent experiments. ∗, p < 0.05; ∗∗, p < 0.01 when compared with ligand-treated cells, respectively.

15d-PGD2 and PGD2, but not PGE2 or arachidonic acid, modulate the expression of TLR2

PGs are inflammatory mediators that are produced via the metabolism of arachidonic acid by COX and a chain of PG synthase enzymes (18, 19). Because 15d-PGJ2 is derived from PGD2, a metabolite of PGH2, and PGD2 is now recognized to have not only proinflammatory but also anti-inflammatory properties during inflammation, we investigated whether PGD2 could also alter the ligand-dependent change of TLR2 expression. Like 15d-PGJ2, PGD2 significantly reduced the enhancement of TLR2 expression in cells treated with LPS, LTA, or Pam3CSK4 (Fig. 4⇓A and data not shown). In addition, 15d-PGD2, another metabolite of PGD2, also suppressed the ligand-triggered increase of TLR2 expression. Under these conditions, TLR2 ligand-stimulated induction of iNOS expression and phagocytosis was significantly attenuated (Fig. 4⇓, B and C).

FIGURE 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 4.

Both 15d-PGD2 and PGD2, but not PGE2 or arachidonic acid (AA), affect TLR2 agonist-stimulated enhancement of TLR2 expression, iNOS expression, and phagocytosis. A, BV2 microglial cells were pretreated with 20 μM 15d-PGD2, 5 μM PGD2, 20 μM PGE2, or 30 μM AA for 1 h, and stimulated with 100 ng/ml Pam3CSK4 for 12 h. TLR2 expression levels were then analyzed using flow cytometry. B, BV2 microglial cells were treated with or without 10 μg/ml LTA for 18 h in the absence or presence of 2.5 μM 15d-PGD2 or PGD2. Protein extracts were then prepared and subjected to Western blotting using Abs against iNOS and α-tubulin (loading control). C, The cellular uptake of fluorescent beads was analyzed as described in Materials and Methods. The MFI values are mean ± SD of three independent experiments. ∗, p < 0.05; ∗∗, p < 0.01 when compared with stimulator-treated cells. Data shown are representative of at least four independent experiments.

We then tested whether TLR2 expression could be altered by PGE2, another metabolite of PGH2. In contrast to the above results, no significant changes in the levels of TLR2 were detected in cells treated with PGE2. In addition, arachidonic acid, the precursor of PGD2, had no effect on ligand-dependent changes in TLR2 expression (Fig. 4⇑A). Taken together, these results indicate that some 15d-PGJ2-related PGs such as PGD2 and 15d-PGD2, but not PGE2 and arachidonic acid, markedly attenuate the ligand-dependent enhancement of TLR2 expression.

Similar effects are observed in primary glial cells and peritoneal macrophage

Because our results demonstrated that some PGs could modulate the ligand-dependent change of TLR2 expression in BV2 microglial cells, we next examined the contribution of these PGs to the regulation of TLR2 expression in mouse and rat primary glial cells. Consistent with the results from BV2 cells, 15d-PGJ2, 15d-PGD2, and PGD2 reduced the ligand-stimulated increase of TLR2 expression in primary microglia and astrocytes (Fig. 5⇓A and data not shown). Moreover, treatment with such PGs also apparently attenuated the response of TLR2 to its ligands in primary peritoneal macrophage (Fig. 5⇓B). Similar effects were observed in the mouse macrophage cell line, J774 (data not shown). Overall, our results indicate that PGs such as 15d-PGJ2, PGD2, and 15d-PGD2 influence TLR2 expression in both primary glia and macrophage, providing further evidence that these PGs control TLR activation by down-regulating TLR2 expression.

FIGURE 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 5.

Similar effects are observed in primary glial cells and peritoneal macrophage. A, Primary astrocytes were treated with 100 ng/ml LPS or 25 μg/ml brain gangliosides for 12 h in the absence or presence of 15d-PGJ2, then Western blot analysis was performed using TLR2 and α-tubulin Abs. B, Mouse primary peritoneal macrophage were stimulated with 100 ng/ml LPS for 12 h following pretreatment with 2.5 μM 15d-PGJ2 for 1 h. TLR2 expression was analyzed at the cell surface level by flow cytometric analysis. C, BV2 cells were untreated or treated with 100 ng/ml Pam3CSK4 for 6 h, after which cells were further incubated for 18 h with or without 2.5 μM 15d-PGJ2. TLR2 expression was analyzed at the cell surface levels by flow cytometry.

15d-PGJ2 and PGD2 are also capable of suppressing a pre-existing increase in TLR2 expression

The above findings prompted us to consider whether these PGs could modulate a pre-existing activation of glia. To address this more clinically relevant question, we examined the expression level of TLR2 in cells treated with 15d-PGJ2 at various times after treatment with TLR2 ligands. BV2 cells were left untreated or stimulated with 100 ng/ml Pam3CSK4 for 6 h, whereupon the cells were further incubated for 18 h with or without 2.5 μM 15d-PGJ2. Interestingly, the suppressive effects of 15d-PGJ2 on cell surface expression of TLR2 were consistently observed in cells receiving ligand 6 h before the addition of 15d-PGJ2 (Fig. 5⇑C). Similar results were observed in cells treated with PGD2 and 15d-PGD2 (data not shown). These results indicate that—in addition to preventing ligand-dependent increases of TLR2—15d-PGJ2 and PGD2 are capable of attenuating an ongoing ligand-induced increase in TLR2 expression.

Thiazolidinediones, representative PPARγ agonists, do not mimic the actions of 15d-PGD2 and PGD2 on TLR2 expression

PPARγ is a ligand-dependent nuclear receptor whose ligands include several PGs and antidiabetic thiazolidinediones such as rosiglitazone (44). 15d-PGJ2 is shown to be a natural endogenous ligand for PPARγ and to exert some of its anti-inflammatory effects by binding to PPARγ. Therefore, to gain insight into the action mechanism by which these PGs suppress TLR2 expression, we tested the possibility that their inhibitory effects on TLR2 expression could be mediated by PPARγ. For this, we examined whether potent synthetic PPARγ agonists such as rosiglitazone and pioglitazone could affect the cell surface expression of TLR2 in cells stimulated with LPS, LTA, or Pam3CSK4. Neither rosiglitazone nor pioglitazone had any effects on the cell surface expression of TLR2 at any of tested concentrations and time points (Fig. 6⇓). In addition, we did not detect any changes of TLR2 message and protein levels following treatment with thiazolidinediones (data not shown). Under the same conditions, we observed the significant activation of a PPARγ-responsive element-luciferase reporter construct and the inhibitory effects of synthetic PPARγ agonists on expression of inflammatory mediators (data not shown). These results indicate that the inhibitory actions of 15d-PGJ2 and PGD2 on the ligand-dependent enhancement of TLR2 expression do not occur through PPARγ.

FIGURE 6.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 6.

Synthetic PPARγ agonists do not affect the ligand-dependent changes in TLR2 expression. BV2 microglial cells were treated with 100 ng/ml LPS, 10 μg/ml LTA, or 100 ng/ml Pam3CSK4 for 12 h in the presence or absence of 20 μM rosiglitazone (A) or pioglitazone (B), after which TLR2 expression was analyzed at the cell surface protein level. Data shown are representative of at least four independent experiments.

Attenuation of ligand-induced TLR2 expression is not dependent of DP1 or DP2

PGD2 binds and activates two distinct G protein-coupled receptors, DP1 and chemoattractant receptor-like molecule expressed on Th2 (CRTH2; also named DP2). It has been shown that the physiological effects of PGD2 and 15d-PGD2 are mediated in part by DP1 or DP2 (21, 28, 30). Thus, we questioned whether 15d-PGJ2, PGD2, and 15d-PGD2 could exert their effects on TLR2 expression through DP1 or DP2. To address it, we examined the effects of the selective DP1 agonist BW245C or the selective DP2 agonist DK-PGD2 on ligand-dependent change in TLR2 expression. As shown in Fig. 7⇓A, treatment with BW245C or DK-PGD2 did not reduce the increased levels of TLR2 message in activated primary microglia. Moreover, neither BW245C nor DK-PGD2 had any effect on the LTA-stimulated increase of TLR2 protein expression at all tested concentrations and time points, whereas BW245C, but not DK-PGD2, suppressed the increase of COX2 under the same conditions (Fig. 7⇓B). Similar results were observed in primary microglia and astrocytes treated with LPS or Pam3CSK4 (data not shown). Taken together, these results suggest that neither DP1 nor DP2 mediate the regulatory effects of PGD2, 15d-PGJ2, and 15d-PGD2 on ligand-dependent increase of TLR2 expression.

FIGURE 7.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 7.

Neither the DP1 agonist BW245C nor the DP2 agonist DK-PGD2 attenuates the ligand-stimulated increase of TLR2 expression. A, Rat primary microglia was treated with 10 μg/ml LTA or 100 ng/ml LPS for 6 h following pretreatment with 50 nM BW245C or 0.5 μM DK-PGD2 for 1 h. Total RNA was then extracted for RT-PCR analysis, and cell extract was prepared for Western blot analysis. B, Primary astrocytes were treated with 10 μg/ml LTA for 12 h in the indicated concentrations of BW245C or DK-PGD2, then Western blot analysis was performed using TLR2, COX2, and α-tubulin Abs. Data shown are representative of at least three independent experiments.

Discussion

The TLR system has been extensively characterized over the past several years, and considerable progress has been made in identifying the involved ligand specificities and downstream signaling cascades. However, relatively little is known regarding how the TLR system is regulated to maintain the balance between defense functions and harmful effects. Recent studies have revealed that dysregulation of TLR signaling may contribute significantly to a variety of diseases (4, 45), and many efforts are now being made to understand the regulation of the TLR system and develop therapeutic strategies to stimulate or inhibit TLR signaling (46, 47, 48). Here, we provide important new results showing that some PGs may contribute to the regulation of the TLR2-associated inflammatory responses through down-regulation of TLR2 expression in the brain.

Many CNS diseases including Alzheimer disease, Parkinson’s disease, multiple sclerosis, ischemia, and cancer are accompanied by robust activation of immune and inflammatory responses; these effects are closely associated with glial cells, which are the main immune cells of the brain (17, 18, 49). Recently, we and other researchers have shown that microglia and astrocytes express TLRs, and TLR-mediated inflammatory signaling may play a crucial role in glial activation and inflammation-associated neuronal diseases (37, 38, 50, 51). In particular, several reports have shown that TLR2 and TLR4 might contribute to the initiation and progression of CNS pathologies (39, 40, 41, 42, 52, 53). We have therefore focused on investigating the regulatory mechanisms that limit excessive TLR-mediated responses and bring about the resolution of brain inflammation. Interestingly, we found that 15d-PGJ2, a cyclopentenone PG, significantly modulates TLR2 message, protein, and cell surface levels in microglia and astrocytes activated by Pam3CSK4, LTA, as well as LPS and gangliosides. Notably, 15d-PGJ2 appears to have differential effects on the various TLR types in glia; whereas TLR2 expression was regulated by 15d-PGJ2, expression of TLR1, 4, and TLR9 was not affected by 15d-PGJ2 treatment of ligand-stimulated cells (Fig. 1⇑). TLR expression has been shown to be strictly regulated in a stimulus-dependent, cell type-, and state-specific manner (54, 55, 56, 57). Our results suggest the possibility that 15d-PGJ2 may exert a role in the modulation of TLR2 expression, thus contributing to the resolution of TLR2-stimulated inflammation in the brain.

Some PGs have been reported to be produced during inflammatory episodes and have an anti-inflammatory role in the resolution of inflammation, in addition to well-known proinflammatory abilities in the pathogenic state (21, 22, 23, 24, 25, 58, 59). Therefore, we questioned whether not only 15d-PGJ2 but also its related PG metabolites could influence the ligand-stimulated increase of TLR2 expression. Interestingly, we found that both PGD2 and 15d-PGD2 significantly reduced the ligand-dependent enhancement of TLR2 expression and the events downstream of TLR2 activation, but neither PGE2 nor arachidonic acid had any effect on TLR2 expression (Fig. 4⇑). Consistent with this notion, we observed similar effects in primary microglia and astrocytes, and obtained the concordant data from experiments in primary macrophage and macrophage cell lines (Fig. 5⇑).

Our above results raised the question of how these PGs could influence the ligand-stimulated enhancement of TLR2 expression in glial cells. 15d-PGJ2 and PGD2 can activate PPARγ, which has been shown to exert roles in suppression of inflammation as well as adipocyte differentiation (21). In vivo and in vitro studies have provided that 15d-PGJ2 effectively suppresses inflammation, which is either dependent on or independent of PPARγ (60, 61, 62, 63, 64). Based on these reports, we examined whether PPARγ could mediate the inhibitory action of 15d-PGJ2 on TLR2 expression using synthetic PPARγ agonists such as rosiglitazone and pioglitazone. However, even though rosiglitazone and pioglitazone are more potent PPARγ agonists than 15d-PGJ2, they did not have any detectable effects on the ligand-stimulated increase of TLR2 cell surface expression (Fig. 6⇑). In addition, the TLR2 message and protein levels in our system were also unaffected by treatment with rosiglitazone or pioglitazone, whereas PPARγ-responsive element-luciferase reporter activity was significantly increased and the expression levels of proinflammatory mediators were attenuated under the same conditions. Thus, we conclude that the inhibitory actions of 15d-PGJ2 and PGD2 on the ligand-dependent enhancement of TLR2 expression occur independently of PPARγ activation in the brain.

How, then, might they regulate the ligand-stimulated enhancement of TLR2 expression independent of PPARγ? We next considered the possibility that the DP1 or DP2 receptors could mediate these effects because numerous physiological activities of PGD2 and its metabolites have been demonstrated to be dependent on these two G protein-coupled receptors (27, 28, 29, 30). Activation of the DP1 by a selective agonist has been shown to suppress asthma by modulation of lung dendritic cell function and induction of regulatory T cells (27, 28). In the CNS, Liang et al. (29) showed that PGD2 and the DP1 selective agonist mediated neuronal protection via the DP1 receptor in hippocampal neurons. Taniguchi et al. (30) also reported that PGD2 protected the neonatal brain from hypoxic ischemic injury mainly via DP1 receptors. However, we did not detect any effect of the DP1 selective agonist BW245C or the DP2 agonist DK-PGD2 in ligand-stimulated increase of TLR2 expression.

We have previously reported that JAK-STAT signaling contributes to activation of glia in response to several stimuli including LPS and gangliosides, and that inhibition of JAK-STAT signaling by SOCS, Src homology domain-containing tyrosine kinase, or pharmacological inhibitors significantly reduces the abnormal activation of glia (60, 65, 66). Thus, we next examined the possibility that the JAK-STAT-signaling pathway could be linked with the action of some PGs on TLR2 expression using pharmacological inhibitors. However, we did not observe any meaningful effects of JSI-124, an inhibitor for STAT, or AG490, an inhibitor for JAK, on the suppression of TLR2 up-regulation by PG (data not shown). In addition, we did not obtain results supporting an association between induction of SOCS 1–3 and regulation of TLR2 expression by PGs (data not shown). Although further research will be required to definitely elucidate it, it seems unlikely that STAT signaling may be directly linked with the regulation of TLR2 expression by PGs.

Another possibility is that they may act by modification of specific cellular molecules. Some PGD2 metabolites have been shown to interact directly with intracellular proteins and regulate signaling pathway through modifying protein function. For instance, 15d-PGJ2 inhibited NF-κB signaling through binding to IκB kinase and NF-κB (67, 68), and also inhibited AP-1 activity by interaction with c-Jun (69). In addition to inflammation-associated transcription factors, there are reports that 15d-PGJ2 also regulates the activities of several proteins, including H-Ras and estrogen receptor-α (70, 71). Based on these reports and our current findings, it is possible that such PGs may functionally associate with certain signaling molecules in the TLR2-specific signaling pathway. In this regard, we are now investigating this possibility.

In the CNS, the levels of PGs are low under normal conditions but increase during inflammatory processes. In vivo and in vitro studies have shown that some PGs including PGD2 and its related cyclopentenone PGs play a crucial role in the resolution of inflammation by acting as endogenous anti-inflammatory mediators although other reports dispute these properties (22, 23, 24, 25, 26, 27, 28, 64, 72, 73). Our present data indicate that some PGs can attenuate TLR2 expression even when administered following exposure to TLR2 ligands. This observation suggests that they may participate in balancing appropriate inflammatory responses by acting as feedback inhibitors of TLR2-dependent inflammatory responses. The capacity of such PGs to reduce ongoing inflammatory responses is notable in the development of novel therapeutic approaches for TLR2-associated diseases. Further study on the precise molecular mechanism underlying these effects should contribute to a better understanding of the inflammation system, and may lead to the development of new therapeutic interventions for existing and emerging inflammation-associated neuronal diseases.

Acknowledgments

We thank all the anonymous reviewers for their thoughtful comments and all the members of the Division of Fusion Technology at the National Cancer Center for their helpful discussions and suggestions.

Disclosures

The authors have no financial conflict of interest.

Footnotes

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

  • ↵1 This work was supported by the National Cancer Center (0710320-1) and by KRF grant funded by the Korean Government (KRF-2005-041-E00103).

  • ↵2 Address correspondence and reprint requests to Dr. Eun Jung Park, Immune and Cell Therapy Branch, National Cancer Center, Goyang, 410-351, Korea. E-mail address: ejpark{at}ncc.re.kr

  • ↵3 Abbreviations used in this paper: SOCS, suppressor of cytokine signaling; DP, D prostanoid receptor; COX, cyclooxygenase; 15d-PGJ2, 15-deoxy-Δ12,14-PG J2; PPAR, peroxisome proliferator-activated receptor; LTA, lipoteichoic acid; iNOS, inducible NO synthase; DAPI, 4′,6′-diamidino-2-phenylindole hydrochloride.

  • Received August 24, 2007.
  • Accepted April 10, 2008.
  • Copyright © 2008 by The American Association of Immunologists

References

  1. ↵
    Medzhitov, R., P. Preston-Hurlburt, C. A. Janeway, Jr. 1997. A human homologue of the Drosophila Toll protein signals activation of adaptive immunity. Nature 388: 394-397.
    OpenUrlCrossRefPubMed
  2. ↵
    Aderem, A., R. J. Ulevitch. 2000. Toll-like receptors in the induction of the innate immune response. Nature 406: 782-787.
    OpenUrlCrossRefPubMed
  3. ↵
    Underhill, D. M., A. Ozinsky, A. M. Hajjar, A. Stevens, C. B. Wilson, M. Bassetti, A. Aderem. 1999. The Toll-like receptor 2 is recruited to macrophage phagosomes and discriminates between pathogens. Nature 401: 811-815.
    OpenUrlCrossRefPubMed
  4. ↵
    Sabroe, I., R. C. Read, M. K. Whyte, D. H. Dockrell, S. N. Vogel, S. K. Dower. 2003. Toll-like receptors in health and disease: complex questions remain. J. Immunol. 171: 1630-1635.
    OpenUrlFREE Full Text
  5. ↵
    Kawai, T., S. Akira. 2007. TLR signaling. Semin. Immunol. 19: 24-32.
    OpenUrlCrossRefPubMed
  6. ↵
    O'Neill, L. A.. 2006. How Toll-like receptors signal: what we know and what we don’t know. Curr. Opin. Immunol. 18: 3-9.
    OpenUrlCrossRefPubMed
  7. ↵
    Trinchieri, G., A. Sher. 2007. Cooperation of Toll-like receptor signals in innate immune defense. Nat. Rev. Immunol. 7: 179-190.
    OpenUrlCrossRefPubMed
  8. ↵
    Kaisho, T., S. Akira. 2006. Toll-like receptor function and signaling. J. Allergy Clin. Immunol. 117: 979-987.
    OpenUrlCrossRefPubMed
  9. ↵
    Kobayashi, K., L. D. Hernandez, J. E. Galan, C. A. Janeway, Jr, R. Medzhitov, R. A. Flavell. 2002. IRAK-M is a negative regulator of Toll-like receptor signaling. Cell 110: 191-202.
    OpenUrlCrossRefPubMed
  10. ↵
    Nakagawa, R., T. Naka, H. Tsutsui, M. Fujimoto, A. Kimura, T. Abe, E. Seki, S. Sato, O. Takeuchi, K. Takeda, et al 2002. SOCS-1 participates in negative regulation of LPS responses. Immunity 17: 677-687.
    OpenUrlCrossRefPubMed
  11. ↵
    Zhang, G., S. Ghosh. 2002. Negative regulation of Toll-like receptor-mediated signaling by Tollip. J. Biol. Chem. 277: 7059-7065.
    OpenUrlAbstract/FREE Full Text
  12. ↵
    Kobayashi, K. S., R. A. Flavell. 2004. Shielding the double-edged sword: negative regulation of the innate immune system. J. Leukocyte Biol. 75: 428-433.
    OpenUrlAbstract/FREE Full Text
  13. ↵
    Saitoh, T., M. Yamamoto, M. Miyagishi, K. Taira, M. Nakanishi, T. Fujita, S. Akira, N. Yamamoto, S. Yamaoka. 2005. A20 is a negative regulator of IFN regulatory factor 3 signaling. J. Immunol. 174: 1507-1512.
    OpenUrlAbstract/FREE Full Text
  14. ↵
    Tanaka, T., M. J. Grusby, T. Kaisho. 2007. PDLIM2-mediated termination of transcription factor NF-κB activation by intranuclear sequestration and degradation of the p65 subunit. Nat. Immunol. 8: 584-591.
    OpenUrlCrossRefPubMed
  15. ↵
    Ridet, J. L., S. K. Malhotra, A. Privat, F. H. Gage. 1997. Reactive astrocytes: cellular and molecular cues to biological function. Trends Neurosci. 20: 570-577.
    OpenUrlCrossRefPubMed
  16. ↵
    Kigerl, K. A., W. Lai, S. Rivest, R. P. Hart, A. R. Satoskar, P. G. Popovich. 2007. Toll-like receptor (TLR)-2 and TLR-4 regulate inflammation, gliosis, and myelin sparing after spinal cord injury. J. Neurochem. 102: 37-50.
    OpenUrlCrossRefPubMed
  17. ↵
    Matyszak, M. K.. 1998. Inflammation in the CNS: balance between immunological privilege and immune responses. Prog. Neurobiol. 56: 19-35.
    OpenUrlCrossRefPubMed
  18. ↵
    Minghetti, L., G. Levi. 1998. Microglia as effector cells in brain damage and repair: focus on prostanoids and nitric oxide. Prog. Neurobiol. 54: 99-125.
    OpenUrlCrossRefPubMed
  19. ↵
    Straus, D. S., C. K. Glass. 2001. Cyclopentenone prostaglandins: new insights on biological activities and cellular targets. Med. Res. Rev. 21: 185-210.
    OpenUrlCrossRefPubMed
  20. ↵
    Hata, A. N., R. M. Breyer. 2004. Pharmacology and signaling of prostaglandin receptors: multiple roles in inflammation and immune modulation. Pharmacol. Ther. 103: 147-166.
    OpenUrlCrossRefPubMed
  21. ↵
    Sandig, H., J. E. Pease, I. Sabroe. 2007. Contrary prostaglandins: the opposing roles of PGD2 and its metabolites in leukocyte function. J. Leukocyte Biol. 81: 372-382.
    OpenUrlAbstract/FREE Full Text
  22. ↵
    Gilroy, D. W., P. R. Colville-Nash, D. Willis, J. Chivers, M. J. Paul-Clark, D. A. Willoughby. 1999. Inducible cyclooxygenase may have anti-inflammatory properties. Nat. Med. 5: 698-701.
    OpenUrlCrossRefPubMed
  23. ↵
    Shibata, T., M. Kondo, T. Osawa, N. Shibata, M. Kobayashi, K. Uchida. 2002. 15-Deoxy-δ 12,14-prostaglandin J2: a prostaglandin D2 metabolite generated during inflammatory processes. J. Biol. Chem. 277: 10459-10466.
    OpenUrlAbstract/FREE Full Text
  24. ↵
    Trivedi, S. G., J. Newson, R. Rajakariar, T. S. Jacques, R. Hannon, Y. Kanaoka, N. Eguchi, P. Colville-Nash, D. W. Gilroy. 2006. Essential role for hematopoietic prostaglandin D2 synthase in the control of delayed type hypersensitivity. Proc. Natl. Acad. Sci. USA 103: 5179-5184.
    OpenUrlAbstract/FREE Full Text
  25. ↵
    Gavett, S. H., S. L. Madison, P. C. Chulada, P. E. Scarborough, W. Qu, J. E. Boyle, H. F. Tiano, C. A. Lee, R. Langenbach, V. L. Roggli, D. C. Zeldin. 1999. Allergic lung responses are increased in prostaglandin H synthase-deficient mice. J. Clin. Invest. 104: 721-732.
    OpenUrlCrossRefPubMed
  26. ↵
    Peebles, R. S., Jr, K. Hashimoto, J. D. Morrow, R. Dworski, R. D. Collins, Y. Hashimoto, J. W. Christman, K. H. Kang, K. Jarzecka, J. Furlong, et al 2002. Selective cyclooxygenase-1 and -2 inhibitors each increase allergic inflammation and airway hyperresponsiveness in mice. Am. J. Respir. Crit. Care Med. 165: 1154-1160.
    OpenUrlCrossRefPubMed
  27. ↵
    Hammad, H., H. J. de Heer, T. Soullie, H. C. Hoogsteden, F. Trottein, B. N. Lambrecht. 2003. Prostaglandin D2 inhibits airway dendritic cell migration and function in steady state conditions by selective activation of the D prostanoid receptor 1. J. Immunol. 171: 3936-3940.
    OpenUrlAbstract/FREE Full Text
  28. ↵
    Hammad, H., M. Kool, T. Soullie, S. Narumiya, F. Trottein, H. C. Hoogsteden, B. N. Lambrecht. 2007. Activation of the D prostanoid 1 receptor suppresses asthma by modulation of lung dendritic cell function and induction of regulatory T cells. J. Exp. Med. 204: 357-367.
    OpenUrlAbstract/FREE Full Text
  29. ↵
    Liang, X., L. Wu, T. Hand, K. Andreasson. 2005. Prostaglandin D2 mediates neuronal protection via the DP1 receptor. J. Neurochem. 92: 477-486.
    OpenUrlCrossRefPubMed
  30. ↵
    Taniguchi, H., I. Mohri, H. Okabe-Arahori, K. Aritake, K. Wada, T. Kanekiyo, S. Narumiya, M. Nakayama, K. Ozono, Y. Urade, M. Taniike. 2007. Prostaglandin D2 protects neonatal mouse brain from hypoxic ischemic injury. J. Neurosci. 27: 4303-4312.
    OpenUrlAbstract/FREE Full Text
  31. ↵
    Petrova, T. V., K. T. Akama, L. J. Van Eldik. 1999. Cyclopentenone prostaglandins suppress activation of microglia: down-regulation of inducible nitric-oxide synthase by 15-deoxy-δ12,14-prostaglandin J2. Proc. Natl. Acad. Sci. USA 96: 4668-4673.
    OpenUrlAbstract/FREE Full Text
  32. ↵
    Combs, C. K., D. E. Johnson, J. C. Karlo, S. B. Cannady, G. E. Landreth. 2000. Inflammatory mechanisms in Alzheimer’s disease: inhibition of β-amyloid-stimulated proinflammatory responses and neurotoxicity by PPARγ agonists. J. Neurosci. 20: 558-567.
    OpenUrlAbstract/FREE Full Text
  33. ↵
    Heneka, M. T., T. Klockgether, D. L. Feinstein. 2000. Peroxisome proliferator-activated receptor-γ ligands reduce neuronal inducible nitric oxide synthase expression and cell death in vivo. J. Neurosci. 20: 6862-6867.
    OpenUrlAbstract/FREE Full Text
  34. ↵
    Scher, J. U., M. H. Pillinger. 2005. 15d-PGJ2: the anti-inflammatory prostaglandin?. Clin. Immunol. 114: 100-109.
    OpenUrlCrossRefPubMed
  35. ↵
    Rehli, M.. 2002. Of mice and men: species variations of Toll-like receptor expression. Trends Immunol. 23: 375-378.
    OpenUrlCrossRefPubMed
  36. ↵
    Weiss, D. S., B. Raupach, K. Takeda, S. Akira, A. Zychlinsky. 2004. Toll-like receptors are temporally involved in host defense. J. Immunol. 172: 4463-4469.
    OpenUrlAbstract/FREE Full Text
  37. ↵
    Jou, I., J. H. Lee, S. Y. Park, H. J. Yoon, E. H. Joe, E. J. Park. 2006. Gangliosides trigger inflammatory responses via TLR4 in brain glia. Am. J. Pathol. 168: 1619-1630.
    OpenUrlCrossRefPubMed
  38. ↵
    Yoon, H. J., S. B. Jeon, K. H. Seok, D. K. Choi, Y. J. Hong, E. J. Park. 2008. Contribution of TLR2 in the initiation of gangliosides-triggered inflammatory signaling. Mol. Cells 25: 99-104.
    OpenUrlPubMed
  39. ↵
    Kielian, T., A. Haney, P. M. Mayes, S. Garg, N. Esen. 2005. Toll-like receptor 2 modulates the proinflammatory milieu in Staphylococcus aureus-induced brain abscess. Infect. Immun. 73: 7428-7435.
    OpenUrlAbstract/FREE Full Text
  40. ↵
    Chen, K., P. Iribarren, J. Hu, J. Chen, W. Gong, E. H. Cho, S. Lockett, N. M. Dunlop, J. M. Wang. 2006. Activation of Toll-like receptor 2 on microglia promotes cell uptake of Alzheimer disease-associated amyloid β peptide. J. Biol. Chem. 281: 3651-3659.
    OpenUrlAbstract/FREE Full Text
  41. ↵
    Lehnardt, S., P. Henneke, E. Lien, D. L. Kasper, J. J. Volpe, I. Bechmann, R. Nitsch, J. R. Weber, D. T. Golenbock, T. Vartanian. 2006. A mechanism for neurodegeneration induced by group B streptococci through activation of the TLR2/MyD88 pathway in microglia. J. Immunol. 177: 583-592.
    OpenUrlAbstract/FREE Full Text
  42. ↵
    Hoffmann, O., J. S. Braun, D. Becker, A. Halle, D. Freyer, E. Dagand, S. Lehnardt, J. R. Weber. 2007. TLR2 mediates neuroinflammation and neuronal damage. J. Immunol. 178: 6476-6481.
    OpenUrlAbstract/FREE Full Text
  43. ↵
    Jiang, D., J. Liang, J. Fan, S. Yu, S. Chen, Y. Luo, G. D. Prestwich, M. M. Mascarenhas, H. G. Garg, D. A. Quinn, et al 2005. Regulation of lung injury and repair by Toll-like receptors and hyaluronan. Nat. Med. 11: 1173-1179.
    OpenUrlCrossRefPubMed
  44. ↵
    Lehmann, J. M., L. B. Moore, T. A. Smith-Oliver, W. O. Wilkison, T. M. Willson, S. A. Kliewer. 1995. An antidiabetic thiazolidinedione is a high affinity ligand for peroxisome proliferator-activated receptor γ (PPAR γ). J. Biol. Chem. 270: 12953-12956.
    OpenUrlAbstract/FREE Full Text
  45. ↵
    Miyake, K.. 2007. Innate immune sensing of pathogens and danger signals by cell surface Toll-like receptors. Semin. Immunol. 19: 3-10.
    OpenUrlCrossRefPubMed
  46. ↵
    Han, J., R. J. Ulevitch. 2005. Limiting inflammatory responses during activation of innate immunity. Nat. Immunol. 6: 1198-1205.
    OpenUrlCrossRefPubMed
  47. ↵
    Miggin, S. M., L. A. O'Neill. 2006. New insights into the regulation of TLR signaling. J. Leukocyte Biol. 80: 220-226.
    OpenUrlAbstract/FREE Full Text
  48. ↵
    Kanzler, H., F. J. Barrat, E. M. Hessel, R. L. Coffman. 2007. Therapeutic targeting of innate immunity with Toll-like receptor agonists and antagonists. Nat. Med. 13: 552-559.
    OpenUrlCrossRefPubMed
  49. ↵
    Streit, W. J., S. A. Walter, N. A. Pennell. 1999. Reactive microgliosis. Prog. Neurobiol. 57: 563-581.
    OpenUrlCrossRefPubMed
  50. ↵
    Bowman, C. C., A. Rasley, S. L. Tranguch, I. Marriott. 2003. Cultured astrocytes express Toll-like receptors for bacterial products. Glia 43: 281-291.
    OpenUrlCrossRefPubMed
  51. ↵
    Olson, J. K., S. D. Miller. 2004. Microglia initiate central nervous system innate and adaptive immune responses through multiple TLRs. J. Immunol. 173: 3916-3924.
    OpenUrlAbstract/FREE Full Text
  52. ↵
    Lehnardt, S., C. Lachance, S. Patrizi, S. Lefebvre, P. L. Follett, F. E. Jensen, P. A. Rosenberg, J. J. Volpe, T. Vartanian. 2002. The Toll-like receptor TLR4 is necessary for lipopolysaccharide-induced oligodendrocyte injury in the CNS. J. Neurosci. 22: 2478-2486.
    OpenUrlAbstract/FREE Full Text
  53. ↵
    Tanga, F. Y., N. Nutile-McMenemy, J. A. DeLeo. 2005. The CNS role of Toll-like receptor 4 in innate neuroimmunity and painful neuropathy. Proc. Natl. Acad. Sci. USA 102: 5856-5861.
    OpenUrlAbstract/FREE Full Text
  54. ↵
    Visintin, A., A. Mazzoni, J. H. Spitzer, D. H. Wyllie, S. K. Dower, D. M. Segal. 2001. Regulation of Toll-like receptors in human monocytes and dendritic cells. J. Immunol. 166: 249-255.
    OpenUrlAbstract/FREE Full Text
  55. ↵
    Muzio, M., D. Bosisio, N. Polentarutti, G. D'Amico, A. Stoppacciaro, R. Mancinelli, C. van't Veer, G. Penton-Rol, L. P. Ruco, P. Allavena, A. Mantovani. 2000. Differential expression and regulation of Toll-like receptors (TLR) in human leukocytes: selective expression of TLR3 in dendritic cells. J. Immunol. 164: 5998-6004.
    OpenUrlAbstract/FREE Full Text
  56. ↵
    Reis e Sousa, C.. 2004. Toll-like receptors and dendritic cells: for whom the bug tolls. Semin. Immunol. 16: 27-34.
    OpenUrlCrossRefPubMed
  57. ↵
    Iwasaki, A., R. Medzhitov. 2004. Toll-like receptor control of the adaptive immune responses. Nat. Immunol. 5: 987-995.
    OpenUrlCrossRefPubMed
  58. ↵
    Wayman, N. S., Y. Hattori, M. C. McDonald, H. Mota-Filipe, S. Cuzzocrea, B. Pisano, P. K. Chatterjee, C. Thiemermann. 2002. Ligands of the peroxisome proliferator-activated receptors (PPAR-γ and PPAR-α) reduce myocardial infarct size. FASEB J. 16: 1027-1040.
    OpenUrlAbstract/FREE Full Text
  59. ↵
    Murphy, G. J., J. C. Holder. 2000. PPAR-γ agonists: therapeutic role in diabetes, inflammation and cancer. Trends Pharmacol. Sci. 21: 469-474.
    OpenUrlCrossRefPubMed
  60. ↵
    Park, E. J., S. Y. Park, E. H. Joe, I. Jou. 2003. 15d-PGJ2 and rosiglitazone suppress Janus kinase-STAT inflammatory signaling through induction of suppressor of cytokine signaling 1 (SOCS1) and SOCS3 in glia. J. Biol. Chem. 278: 14747-14752.
    OpenUrlAbstract/FREE Full Text
  61. ↵
    Jiang, C., A. T. Ting, B. Seed. 1998. PPAR-γ agonists inhibit production of monocyte inflammatory cytokines. Nature 391: 82-86.
    OpenUrlCrossRefPubMed
  62. ↵
    Ricote, M., A. C. Li, T. M. Willson, C. J. Kelly, C. K. Glass. 1998. The peroxisome proliferator-activated receptor-γ is a negative regulator of macrophage activation. Nature 391: 79-82.
    OpenUrlCrossRefPubMed
  63. ↵
    Chawla, A., Y. Barak, L. Nagy, D. Liao, P. Tontonoz, R. M. Evans. 2001. PPAR-γ dependent and independent effects on macrophage-gene expression in lipid metabolism and inflammation. Nat. Med. 7: 48-52.
    OpenUrlCrossRefPubMed
  64. ↵
    Diab, A., C. Deng, J. D. Smith, R. Z. Hussain, B. Phanavanh, A. E. Lovett-Racke, P. D. Drew, M. K. Racke. 2002. Peroxisome proliferator-activated receptor-γ agonist 15-deoxy-δ(12,14)-prostaglandin J2 ameliorates experimental autoimmune encephalomyelitis. J. Immunol. 168: 2508-2515.
    OpenUrlAbstract/FREE Full Text
  65. ↵
    Kim, O. S., E. J. Park, E. H. Joe, I. Jou. 2002. JAK-STAT signaling mediates gangliosides-induced inflammatory responses in brain microglial cells. J. Biol. Chem. 277: 40594-40601.
    OpenUrlAbstract/FREE Full Text
  66. ↵
    Kim, H. Y., E. J. Park, E. H. Joe, I. Jou. 2003. Curcumin suppresses Janus kinase-STAT inflammatory signaling through activation of Src homology 2 domain-containing tyrosine phosphatase 2 in brain microglia. J. Immunol. 171: 6072-6079.
    OpenUrlAbstract/FREE Full Text
  67. ↵
    Rossi, A., P. Kapahi, G. Natoli, T. Takahashi, Y. Chen, M. Karin, M. G. Santoro. 2000. Anti-inflammatory cyclopentenone prostaglandins are direct inhibitors of IκB kinase. Nature 403: 103-108.
    OpenUrlCrossRefPubMed
  68. ↵
    Straus, D. S., G. Pascual, M. Li, J. S. Welch, M. Ricote, C. H. Hsiang, L. L. Sengchanthalangsy, G. Ghosh, C. K. Glass. 2000. 15-deoxy-δ 12, 14-prostaglandin J2 inhibits multiple steps in the NF-κB signaling pathway. Proc. Natl. Acad. Sci. USA 97: 4844-4849.
    OpenUrlAbstract/FREE Full Text
  69. ↵
    Perez-Sala, D., E. Cernuda-Morollon, F. J. Canada. 2003. Molecular basis for the direct inhibition of AP-1 DNA binding by 15-deoxy-δ 12,14-prostaglandin J2. J. Biol. Chem. 278: 51251-51260.
    OpenUrlAbstract/FREE Full Text
  70. ↵
    Oliva, J. L., D. Perez-Sala, A. Castrillo, N. Martinez, F. J. Canada, L. Bosca, J. M. Rojas. 2003. The cyclopentenone 15-deoxy-δ 12,14-prostaglandin J2 binds to and activates H-Ras. Proc. Natl. Acad. Sci. USA 100: 4772-4777.
    OpenUrlAbstract/FREE Full Text
  71. ↵
    Kim, H. J., J. Y. Kim, Z. Meng, L. H. Wang, F. Liu, T. P. Conrads, T. R. Burke, T. D. Veenstra, W. L. Farrar. 2007. 15-deoxy-δ12,14-prostaglandin J2 inhibits transcriptional activity of estrogen receptor-α via covalent modification of DNA-binding domain. Cancer Res. 67: 2595-2602.
    OpenUrlAbstract/FREE Full Text
  72. ↵
    Lawrence, T., D. A. Willoughby, D. W. Gilroy. 2002. Anti-inflammatory lipid mediators and insights into the resolution of inflammation. Nat. Rev. 2: 787-795.
    OpenUrlCrossRef
  73. ↵
    Reilly, C. M., J. C. Oates, J. A. Cook, J. D. Morrow, P. V. Halushka, G. S. Gilkeson. 2000. Inhibition of mesangial cell nitric oxide in MRL/lpr mice by prostaglandin J2 and proliferator activation receptor-γ agonists. J. Immunol. 164: 1498-1504.
    OpenUrlAbstract/FREE Full Text
PreviousNext
Back to top

In this issue

The Journal of Immunology: 180 (12)
The Journal of Immunology
Vol. 180, Issue 12
15 Jun 2008
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Advertising (PDF)
  • Back Matter (PDF)
  • Editorial Board (PDF)
  • Front Matter (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word about The Journal of Immunology.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Regulation of TLR2 Expression by Prostaglandins in Brain Glia
(Your Name) has forwarded a page to you from The Journal of Immunology
(Your Name) thought you would like to see this page from the The Journal of Immunology web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Regulation of TLR2 Expression by Prostaglandins in Brain Glia
Hee Jung Yoon, Sae-Bom Jeon, In-Hoo Kim, Eun Jung Park
The Journal of Immunology June 15, 2008, 180 (12) 8400-8409; DOI: 10.4049/jimmunol.180.12.8400

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Regulation of TLR2 Expression by Prostaglandins in Brain Glia
Hee Jung Yoon, Sae-Bom Jeon, In-Hoo Kim, Eun Jung Park
The Journal of Immunology June 15, 2008, 180 (12) 8400-8409; DOI: 10.4049/jimmunol.180.12.8400
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like

Jump to section

  • Article
    • Abstract
    • Materials and Methods
    • Results
    • Discussion
    • Acknowledgments
    • Disclosures
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

Cited By...

More in this TOC Section

  • Anti-inflammatory and anti-bacterial effect of polyacetylene compound from Cirsium japonicum var. ussuriense (54.19)
  • CD11b+/Gr-1int monocytic myeloid derived suppressor cells contribute to high-fat induced inflammation and delayed tolerance in mouse liver (54.15)
  • Antigen presentation by dendritic cells in the aortic wall triggers T helper immune responses in atherosclerosis (54.16)
Show more Inflammation

Similar Articles

Navigate

  • Home
  • Current Issue
  • Next in The JI
  • Archive
  • Brief Reviews
  • Pillars of Immunology
  • Translating Immunology

For Authors

  • Submit a Manuscript
  • Instructions for Authors
  • About the Journal
  • Journal Policies
  • Editors

General Information

  • Advertisers
  • Subscribers
  • Rights and Permissions
  • Accessibility Statement
  • Public Access
  • Privacy Policy
  • Disclaimer

Journal Services

  • Email Alerts
  • RSS Feeds
  • ImmunoCasts
  • Twitter

Copyright © 2021 by The American Association of Immunologists, Inc.

Print ISSN 0022-1767        Online ISSN 1550-6606