Skip to main content
Proceedings of the National Academy of Sciences of the United States of America logoLink to Proceedings of the National Academy of Sciences of the United States of America
. 2009 Dec 7;106(51):21807–21812. doi: 10.1073/pnas.0906891106

Targeted lipidomics reveals mPGES-1-PGE2 as a therapeutic target for multiple sclerosis

Yasuyuki Kihara a, Takuya Matsushita b, Yoshihiro Kita a, Satoshi Uematsu c, Shizuo Akira c, Jun-ichi Kira b, Satoshi Ishii a,d,1, Takao Shimizu a,1
PMCID: PMC2789753  PMID: 19995978

Abstract

The arachidonic acid (AA) cascade produces eicosanoids, such as prostaglandins (PGs), that regulate physiological and pathological functions. Although various nonsteroidal anti-inflammatory drugs have been developed, blocking upstream components (cyclooxygenase-1 and -2) of the AA cascade leads to severe side effects, including gastrointestinal ulcers and cardiovascular events, respectively, due to the complexity of the AA cascade. Here, using an AA cascade-targeted lipidomics approach, we report that microsomal PGE synthase 1 (mPGES-1) plays a key role in experimental autoimmune encephalomyelitis (EAE). Eicosanoids (mainly PGD2) are produced constitutively in the spinal cord of naive mice. However, in EAE lesions, the PGE2 pathway is favored and the PGD2, PGI2, and 5-lipoxygenase pathways are attenuated. Furthermore, mPGES-1−/− mice showed less severe symptoms of EAE and lower production of IL-17 and IFN-γ than mPGES-1+/+ mice. Expression of PGE2 receptors (EP1, EP2, and EP4) was elevated in EAE lesions and correlated with clinical symptoms. Immunohistochemistry on central nervous systems of EAE mice and multiple sclerosis (MS) patients revealed overt expression of mPGES-1 protein in microglia/macrophages. Thus, the mPGES-1-PGE2-EPs axis of the AA cascade may exacerbate EAE pathology. Our findings have important implications for the design of therapies for MS.

Keywords: autoimmunity, demyelination, lipid mediator, mass spectrometry, Th17


Multiple sclerosis (MS) is the most prevalent autoimmune disorder of the central nervous system (CNS), with neurological symptoms caused by inflammation and demyelination (1). Studies of experimental autoimmune encephalomyelitis (EAE), an animal model for MS, have shown that autoreactive T cells secreting IL-17 (TH17 cells) and IFN-γ (TH1 cells) are involved in EAE/MS pathogenesis (24).

In an effort to understand and treat this complex disease, lipids have emerged as one of the targets for developing drugs. Membrane lipids have been identified as autoantigens in MS and EAE pathologies (5). In addition to membrane components, lipids play roles in cell–cell interaction as autacoids. Arachidonic acid (AA) is released from membrane glycerophospholipids by the action of cytosolic phospholipase A2α (cPLA2α) (6, 7). Released AA is further converted into prostaglandins (PGs), leukotrienes (LTs), lipoxins, and hydroxy-eicosatetraenoic acids (HETEs), collectively termed eicosanoids, by cyclooxygenases (COXs), lipoxygenases (LOs), and terminal enzymes (8). Involvement of eicosanoids and related lipid mediators has been reported in EAE, collagen-induced arthritis, and other immunological disorders (6, 7, 9). Previously, we and others clearly demonstrated the importance of cPLA2α in EAE pathology by genetically and pharmacologically ablated mouse studies (1012). Furthermore, cPLA2α expression and activities were up-regulated in the spinal cords (SCs) of EAE mice (13). However, it remains totally elusive which eicosanoids downstream of cPLA2α are involved in the EAE induction and exacerbation. Miyamoto et al. (14) demonstrated that COX-2−/− mice develop EAE with comparable severity to wild-type control mice, while indomethacin, a nonsteroidal anti-inflammatory drug (NSAID), prevents EAE. However, all mice treated with effective doses of indomethacin died of gastrointestinal bleeding (14). On the other hand, 5-LO−/− or 12/15-LO−/− mice developed more severe EAE than control wild-type mice (15), while pharmacological studies showed different results (12). Previous sketchy studies to focus on individual eicosanoids in EAE or MS patients have obvious limitations, as >20 different eicosanoids are produced by the concerted actions of cPLA2α and downstream enzymes (Fig. S1) (1618). To overcome such complexity and some controversy, we evaluated the cascade in the SCs of naive and EAE mice by the nonbiased top-down approach (AA cascade-targeted lipidomics and transcriptomics) and confirmed the results from knockout studies.

Results

AA Cascade-Targeted Transcriptomics Imply That AA Cascade Plays Important Roles in EAE Pathology.

C57BL/6 mice were immunized with a myelin oligodendrocyte glycoprotein 35–55 (MOG35–55) peptide to induce EAE and monitored daily (Fig. 1A). As described in refs. 13 and 19, the course of the disease is divided into induction, acute, and chronic phases in accordance with the clinical symptoms (Fig. 1A). We collected the SCs, spleen, and plasma of naive mice and EAE mice in these three phases.

Fig. 1.

Fig. 1.

AA cascade-targeted transcriptomics analysis. (A) C57BL/6 female mice were immunized with the MOG35–55 peptide. Mice were monitored and weighed daily. Data are the mean clinical score and body weight ± SEM of eight animals. (B) Expression of AA cascade related transcripts was estimated using the comparative CT method with the SCs of naive mice and EAE mice in the induction, acute, and chronic phases (n = 6, 5, 6, and 5 animals, respectively). The relative abundance of mRNA levels in EAE mice compared with naive mice is shown. Data represent means ± SEM. #, P < 0.001, **, P < 0.01, *, P < 0.05 compared with naive mice using the Kruskal-Wallis test with Dunn's post-hoc test.

To determine which enzymes and receptors in the AA cascade are involved in EAE pathology, AA cascade-targeted transcriptomics of the SCs was performed by quantitative RT-PCR (Fig. 1B). Components immediately downstream of cPLA2α are COX-1/2, 5-LO/FLAP, and 12/15-LO, whose expression levels were highly up-regulated (Fig. 1B). Large part of terminal enzymes and receptors, such as microsomal PGE synthase 1 (mPGES-1), were substantially up-regulated in the SCs of EAE mice (Fig. 1B). However, expression levels of PGI synthase (PGIS) and lipocalin-type PGDS (L-PGDS) were down-regulated in the induction and acute phases of EAE, respectively, and then returned to the basal levels in the chronic phase (Fig. 1B). The PGI2, PGE2, and LT receptor (IP, EP1/2/4, BLT1, and CysLT1) gene expression was up-regulated in the acute phase of EAE (Fig. 1B). A correlation between the gene expression and the clinical score was observed for COX-1, H-PGDS, EP1/2/4, BLT1, CysLT1, etc. (Fig. S2). These results imply that AA cascade profoundly affects the pathogenesis of EAE.

AA Cascade-Targeted Lipidomics Reveals That the PGE2 Pathway Is Favored in EAE Lesions.

The lipidomics approaches disclosed the constitutive production of eicosanoids in the SCs of naive mice and the AA cascade dynamics during EAE (Fig. 2). Approximately 90% of the eicosanoids in the SCs of naive mice were derived from the COX pathway (Fig. S3). The rank order of eicosanoids in the SCs of naive mice was the following: PGD2 and metabolites > PGE2 and metabolites >6-keto-PGF > PGF and metabolites > cysteinyl LTs >11-HETE >12-HETE >15-HETE > thromboxane B2 (TXB2) > 5-HETE >8-HETE > LTB4 (Fig. 2A). Cluster analysis demonstrated pathway-dependent fluctuations in the AA cascade, such as COX (clusters I and III) and LO (cluster II), during the course of EAE (Fig. 2B). Eicosanoids belonging to cluster I, such as PGD2 and 6-keto-PGF, were markedly suppressed in the acute phase and returned to the basal levels in the chronic phase (Fig. 2 A and B). Suppression of these metabolites was probably caused by the fluctuation of L-PGDS and PGIS, respectively (Fig. 1B). 5-LO metabolites (LTB4, LTC4, LTD4, and 5-HETE) belonging to cluster II considerably diminished during the disease course (Fig. 2 A and B), although the up-regulation of 5-LO pathway components were remarkable (Fig. 1B). In the cluster III, the levels of PGE2 and 13, 14-dihydro-15-keto-PGE2 (DHK-PGE2, a major tissue metabolite of PGE2) were increased, thereby making them the major eicosanoids in acute phase SCs instead of PGD2 and its metabolites (Fig. 2A). In the chronic phase of EAE, the level of PGE2 remained higher than in naive mice (Fig. 2A). Among PGESs, only mPGES-1 was up-regulated in the SCs of EAE mice (Fig. 1B), suggesting that this is the key enzyme for the PGE2 accumulation in EAE lesions. Cluster IV consists of only platelet-activating factor (PAF), which is structurally distinct from the eicosanoids. We previously reported that PAF exacerbates inflammation in the chronic phase of EAE by the enhancement of phagocytosis in microglia/macrophages and subsequent production of TNF-α (19). Furthermore, there was a strong correlation between PAF levels and clinical scores of EAE (13, 19). However, in the current study, no eicosanoid was correlated with the clinical scores of EAE. Although we also measured eicosanoid levels in the spleen and plasma, there were only a few fluctuations in these samples (Fig. S4). These results suggest that the AA cascade was strictly regulated in the SCs of naive mice and this regulation was disrupted by EAE pathology.

Fig. 2.

Fig. 2.

AA cascade-targeted metabolomics analysis. (A) Eicosanoid levels in the SCs of naive mice and EAE mice in the induction, acute, and chronic phases (n = 8–10 animals) were determined. Data represent means ± SEM. #, P < 0.001, **, P < 0.01, *, P < 0.05 compared with naive mice using the Kruskal-Wallis test with Dunn's post-hoc test. (B) The means of eicosanoid levels were normalized with those of naive mice for each eicosanoid. Then, a cluster analysis was performed by the Ward method. The normalized eicosanoid levels were divided into seven levels according to the indicated color scale. (C) Correlations of COX/5-LO in naive mice (light blue) and EAE mice in the induction (pink), acute (red), and chronic (orange) phases are shown (n = 8–10 animals). Each data point represents the results from a single animal. Data were analyzed statistically by Pearson's correlation.

Next, correlations between the pathways were analyzed to understand the selectivity of the downstream pathways in the AA cascade (Fig. 2C, Fig. S5, and Table S1), because the cluster analysis revealed the pathway-dependent fluctuations. The COX pathway (PGs, TXs, and 11-HETE) correlated with the 5-LO pathway (LTs and 5-HETE) in naive mice (Fig. 2C). Of note, AA metabolism was shifted into the COX pathway, rather than the 5-LO pathway, in the acute phase of EAE (Fig. 2C). Within the COX pathway, the PGE2 pathway was correlated with both the PGD2 and PGI2 pathways in naive mice (Fig. S5 and Table S1). After the induction phase, the PGE2 pathway was correlated with both the PGD2 and PGI2 pathways, while PGE2 production was facilitated, as shown by the small slope (Fig. S5). Correlations between other pathways (Table S1), such as the PGD2 and PGI2 pathways (Fig. S5), were largely conserved throughout the disease course, as indicated by the stable slopes. Taken together, the COX-mPGES-1-PGE2-EP axis may aggravate EAE pathology.

mPGES-1 Exacerbates EAE Pathology Through TH1 and TH17 Cytokine Production.

To further elucidate the roles of PGE2 pathway in EAE, we particularly focused on mPGES-1, a key enzyme responsible for PGE2 production in inflammation (2022). In EAE lesions, mPGES-1 was colocalized with F4/80, a marker for macrophages, but not with CD4 (Fig. 3A). PGE2 production in SCs of EAE mice was completely blocked by disruption of the mPGES-1 gene (Fig. 3B), suggesting that PGE2 production in EAE lesions depends on mPGES-1 expressed in macrophages/microglia. The clinical course of EAE in mPGES-1−/− mice was less severe than in mPGES-1+/+ mice (Fig. 3C), consistent with the lower cumulative scores in mPGES-1−/− mice (Table S2). To understand the mechanisms underlying the attenuated symptoms of EAE in mPGES-1−/− mice, proliferation and cytokine production of T cells in response to the MOG35–55 peptide were investigated (Fig. 3 D–F). When stimulated with the MOG35–55 peptide, cells isolated from lymph nodes (LNs) of immunized mPGES-1+/+ and mPGES-1−/− mice proliferated similarly (Fig. 3D). However, LN cells from mPGES-1−/− mice produced significantly lower levels of cytokines (TNF-α, IFN-γ, IL-6, and IL-17) than mPGES-1+/+ mice (Fig. 3E). Flow cytometry of MOG35–55-treated CD4+ T cells also revealed that IFN-γ and IL-17 production in mPGES-1−/− mice was reduced (Fig. 3F). Taken together, PGE2 derived from mPGES-1 appears to support TH1 and TH17 cytokine production in EAE lesions.

Fig. 3.

Fig. 3.

Suppression of EAE pathology and TH1/TH17 responses in the absence of mPGES-1. (A) SCs of EAE mice in the acute phase were stained with anti-mPGES-1 (green), F4/80 (red), and CD4 (red) Abs. (Scale bar, 50 μm.) These pictures are representative of two different experiments. (B) PGE2 levels were measured in the SCs of mPGES-1+/+, +/− and mPGES-1−/− mice on day 26 (n = 13 and 7 animals, respectively). *, P < 0.05 versus mPGES-1+/+, +/− mice by Mann–Whitney U test. Data represent means ± SEM. (C) mPGES-1+/+ (filled circles) and mPGES-1−/− (open circles) mice were immunized with the MOG35–55 peptide and clinical scores were assessed daily for 26 days. Data represent means ± SEM from two independent experiments with a total of 14 and 9 animals for mPGES-1+/+ and mPGES-1−/− mice, respectively. *, P < 0.0001 versus mPGES-1+/+ mice determined by two-way repeated measures ANOVA. (D) LN cells from EAE-induced mPGES-1+/+ and mPGES-1−/− mice on day 11 were stimulated in vitro with various concentrations of MOG35–55 peptide, and the proliferative responses were measured (n = 5 animals). Data represent means ± SEM. (E) Supernatants of LN cell cultures were used to measure the concentrations of IFN-γ, IL-17, TNF-α, and IL-6 (n = 5 animals). Data represent means ± SEM. *, P < 0.05, **, P < 0.01 versus mPGES-1+/+ mice by two-tailed Student's t test. (F) Two days after activation, cells were restimulated with PMA/ionomycin and subjected to intracellular cytokine staining for IFN-γ and IL-17 (n = 5 animals). Each data point represents the results from a single animal and the horizontal bars designate the mean values of individual groups. *, P < 0.05, by two-tailed Student's t test.

mPGES-1 Is Expressed in Human MS Lesions.

To investigate whether the mPGES-1 is a therapeutic target in MS patients, we examined autopsy brain tissues obtained from MS patients (Fig. 4). In accordance with murine EAE (Fig. 3A), immunohistochemistry on MS lesions revealed the overt expression of mPGES-1 protein (Fig. 4 A, B, D, and E) in CD68+ macrophages (Fig. 4 G–Q). Immunoreactivity of mPGES-1 was not detected with antigen-absorbed Ab (Fig. 4 C and F). These data suggest that not only murine EAE, but also human MS pathology seems to be influenced by the mPGES-1-PGE2 axis of the AA cascade.

Fig. 4.

Fig. 4.

mPGES-1 expression in infiltrated macrophages of MS lesions. Brain tissues of MS patients' autopsied materials displaying periventricular demyelinating lesions were stained with anti-mPGES-1. (Scale bar, 50 μm A, B, D, and E.) The high magnification images of A and D are shown in B and E, respectively. (Scale bar, 10 μm.) The antigen-peptide blocks the signals of anti-mPGES-1 Ab staining. (Scale bar, 10 μm C and F.) Both MS #1 (subacute stage, A–C) and MS #2 (acute stage, D–F) showed extensive infiltration of macrophages characterized by foamy appearance. The mPGES-1 is colocalized with CD68-immunoreactive macrophage in the MS lesions. (Scale bar, 10 μm G–Q.)

Discussion

In the present study, we provided a comprehensive overview of the AA cascade dynamics in EAE lesions and determined the roles of mPGES-1 and PGE2 in EAE. By the AA cascade-targeted lipidomics approach, we found that the PGE2 pathway is favored and the PGD2, PGI2, and 5-LO pathways are attenuated. Correlation analysis imply that the AA producing enzyme cPLA2α possibly passes more AA to the COX than the 5-LO pathway in EAE, because the sequential actions of eicosanoid-synthesizing enzymes are regulated spatially and temporally in the cells, the so-called functional coupling (1618). Likewise, PGH2, a common precursor of PGs, appears to be selectively consumed by the PGE2 pathway rather than the PGI2 and PGD2 pathways, thereby producing PGE2 in the SCs of EAE mice.

The symptoms of EAE in COX-2−/− mice were comparable to those of wild-type controls and administration of indomethacin, a nonselective COX inhibitor, prevented development of the disease (14). We found that COX-2 expression was substantially up-regulated in the SCs of EAE mice from the induction phase (Fig. 1B). The absolute number of microglia/macrophages increased before the onset and throughout the disease course (23), and COX-2 is generally induced by the inflammatory mediators in these cells (24), implying that the up-regulation of COX-2 depended on these cells. In human MS, Rose et al. reported the COX-2 expression in microglia/macrophages of MS lesions (25). We demonstrated here that COX-1 mRNA expression is up-regulated in the SCs of EAE mice (Fig. 1B) and is correlated with clinical symptoms (Fig. S2). Deininger and Schluesener showed the constitutive expression of COX-1 in the microglia/macrophages of rat brain and an elevation of its expression levels in the cells with the progression of EAE (26). Further studies are needed to disclose the functions of COX-1 and COX-2 in EAE pathology.

Among the eicosanoids examined, only the PGE2 level was significantly elevated in the SCs of EAE mice (Fig. 2). This up-regulation seems to depended on the mPGES-1 expressed in the microglia/macrophages of EAE lesions, because the PGE2 production was almost completely suppressed in mPGES-1−/− mice (Fig. 3B). The expression of EP1, EP2, and EP4 was altered depending on the disease severity (Fig. 1B and Fig. S2). Therefore, these EPs are candidates for the downstream effectors of PGE2. Indeed, the exogenous and endogenous PGE2 activates EP2 and EP4 on antigen presenting cells to stimulate expression of IL-23 and IL-6, resulting in a shift toward TH17 responses (2730). PGE2 synergized with IL-23 in expanding human TH17 cell (31) and directly promoted differentiation of human and murine TH17 cells through EP2/EP4 signaling (32). In addition, Yao et al. (30) recently reported that PGE2 acting on EP4 on T cells amplifies IL-23-mediated TH17 expansion in vitro and an EP4-selective antagonist suppressed the EAE pathology. In the case of TH1 immune responses, differentiation of TH1 cells from naive T cells in vitro was facilitated by EP1 (33), EP2, or EP4 agonists (30). T cells appear to encounter the autoantigens mainly in the peripheral lymphoid organs in the induction phase, and then in inflammatory foci of CNS in the acute and chronic phases. The levels of PGE2 in the spleen were unchanged during the disease course (Fig. S4), whereas those in the SCs were drastically elevated in wild-type mice (Fig. 2). Furthermore, the EAE pathology of mPGES-1−/− mice was attenuated in the chronic phase. These results imply that EP4-expressing T cells are affected by PGE2 in the CNS rather than in peripheral lymphoid organs. We showed that the mPGES-1 deficiency abrogated the TH1 and TH17 cytokine production in EAE (Fig. 3 E and F), suggesting that PGE2 produced by microglia/macrophages in inflammatory foci may aggravate EAE by promoting the differentiation of TH1 and the expansion of TH17 cells through these EPs (Fig. 5).

Fig. 5.

Fig. 5.

Conceptual model for the role of AA cascade in EAE pathology. PGE2 is produced by mPGES-1 in activated microglia (MG), infiltrating macrophages (MΦ) and dendritic cells (DC), and then activates these cells in an autocrine/paracrine manner through EP2/EP4. T cells differentiate into TH17 cells by stimulation with IL-6 and IL-23 secreted from activated MG/MΦ/DC. TH1 differentiation and expansion of TH17 cells are accelerated by the direct effects of PGE2 on EP1 and EP2/EP4.

We found that the PGIS expression was essentially diminished in EAE-induced C57BL/6 mice, resulting in a significant decrease of 6-keto-PGF levels in the acute phase of EAE (Fig. 2). Because PGI2 enhances endothelial barrier function (34), PGI2 seems to protect mice from EAE by suppressing the inflammatory cell infiltration. Several reports showed that loss of mPGES-1 results in the rediversion of a substrate PGH2 to other prostanoids, like PGI2, which may affect the physiological and pathophysiological conditions of mPGES-1−/− mice (3537). Therefore, it is possible that the amelioration of EAE in mPGES-1−/− mice is due to the up-regulation of PGI2 levels (Fig. S6). On the other hand, the collagen-induced arthritis was aggravated by the PGI2-IP axis (38), raising another possibility that the PGI2 is an exacerbating factor in TH1 and TH17 immune responses and EAE pathology. The roles of PGI2 in EAE pathology remain controversial, and further studies are needed.

Reduction in PGD2 levels in the acute phase and its recovery in the chronic phase could be explained by the expression changes of L-PGDS (Figs. 1B and 2). One of the features of EAE/MS is demyelination, which coincides with oligodendrocyte cell death. L-PGDS is thought to be an anti-apoptotic molecule that protects oligodendrocytes, because the oligodendrocyte apoptosis was enhanced in the brain of L-PGDS-deficient twitcher mice (39). In human MS, L-PGDS expression was increased especially in the remyelinated lesions (40). Thus, the decrease and the subsequent recovery of L-PGDS may be associated with the demyelination and remyelination in EAE/MS, respectively. Another PGD synthase, H-PGDS, showed the pattern opposite to L-PGDS expression (Fig. 1B). Although H-PGDS is highly expressed in hematopoietic cells and microglia/macrophages (41), PGD2 levels were not elevated in the SCs of EAE mice in the acute phase. Because these cells also express mPGES-1, COX-1/2 seem to prefer to functionally couple with mPGES-1 rather than H-PGDS in the cells of EAE lesions.

Contrary to the up-regulation of 5-LO/FLAP transcripts (Fig. 1B), we demonstrated here the significant suppression of 5-LO metabolites during EAE pathology (Fig. 2). The discrepancy between transcripts and metabolites is explained by posttranscriptional/translational (such as phosphorylation) regulation of enzymes (4244) and/or functional coupling with cPLA2α. Furthermore, COX enzymes were up-regulated in the SC around the same time (Fig. 1B), which results in the reduction of the substrate availability for 5-LO. Emerson and LeVine reported that 5-LO−/− mice developed more severe EAE than control wild-type mice (15), suggesting a possibility of increased PGE2 production in the SCs of 5-LO−/− mice. Meanwhile, the report raises another possibility that lipid mediators downstream of 5-LO may play protective roles in EAE pathology. Because Gladue et al. (45) showed that a BLT1 antagonist protects mice from EAE, LTB4 is not such a candidate for the protective lipid mediators. 5-LO generates not only LTs but also lipoxins and resolvins (46, 47), which are anti-inflammatory/proresolving lipid mediators derived from AA and eicosapentaenoic acid, respectively, although we did not measure these lipid mediators in the current study. 12/15-LO deficiency conferred more severe EAE pathology (15), possibly because 12/15-LO also mediates lipoxin production. Resolvin E1 reduces dendritic cell migration and IL-12 production, suggesting a protective role in EAE pathology (48).

As a therapeutic strategy, blocking the hub of the AA cascade, like COX-1/2, seems to be highly effective in preventing EAE pathology. Although treatment of indomethacin actually ameliorates EAE, all mice died of gastrointestinal ulcers and bleeding (14), a well-known side effect of NSAIDs (49). In 2004, rofecoxib, a selective COX-2 inhibitor, was withdrawn because of an increased cardiovascular risk in patients taking the drug for >18 months (50, 51). A possible reason why NSAIDs are currently only used for treatment of flu-like symptoms associated with IFN therapy for MS is that inhibition of COX-1/2 may put the patients at high risk for adverse effects. Therefore, it seems that targeting AA cascade downstream of COX-1/2 is highly effective in treating MS without side effects. As discussed above, components of AA cascade contribute to EAE pathology via intricate mechanisms (Fig. 5), while our AA cascade-targeted lipidomics approach and knockout study identified the mPGES-1-PGE2-EPs axis as the critical pathway of AA cascade in EAE pathology. Given the function of EPs in TH1 and TH17 immune responses (2729, 38) and our findings of mPGES-1 expression in MS lesions (Fig. 4), inhibition of mPGES-1 has a much better chance of blocking individual EPs. Although mPGES-1 inhibitors which effectively suppress PGE2 production in rodents (22) have not been published yet, MF63, a selective human mPGES-1 inhibitor, suppressed PGE2 production, pyresis and inflammatory pain in knock-in mouse expressing human mPGES-1 (52). We believe that mPGES-1 inhibitors provide a significant treatment option for MS.

Materials and Methods

Mice.

Female C57BL/6 mice were purchased from Charles River for lipidomics analysis. The mPGES-1−/− mice were generated as described in ref. 53. See SI Materials and Methods for more information.

Induction of EAE.

Mice were s.c. immunized with MOG35–55 peptide. Pertussis toxin was i.p. injected. See SI Materials and Methods for more information.

Quantification of Eicosanoids and mRNA Levels in SCs.

Eicosanoid levels were estimated simultaneously as described in refs. 13, 19, 54, and 55. See SI Materials and Methods for more information.

Ex Vivo Experiments.

The details of recall responses to the MOG35–55 peptide antigen, ELISA, and flow cytometry are provided in SI Materials and Methods.

Immunohistochemistry.

For murine EAE, SCs obtained from EAE-induced mice were stained with either anti-F4/80 (Serotec) or CD4 (BD Biosciences) Ab and mPGES-1 (Affinity BioReagents) Ab as primary Abs. For human MS, cerebral specimens were stained with rabbit anti-human mPGES-1 polyclonal Ab (Cayman) or mouse anti-human CD68 monoclonal Ab (DAKO, clone KP1) as a first Ab. See SI Materials and Methods for more information.

Statistical Analysis.

As appropriate, data were analyzed statistically by means of a Student's t test, Mann–Whitney U test, Fisher's exact test, two-way repeated measures ANOVA, or Kruskal-Wallis test with Dunn's post-hoc test. Statistical, clustering, and correlation analyses were performed using JMP6 (SAS Institute) or Prism 4 (GraphPad). P values <0.05 were considered to be statistically significant.

Supplementary Material

Supporting Information

Acknowledgments.

We thank Drs. T. Yokomizo (Kyushu University), N. Uozumi, K. Masago, and K. Yanagida (University of Tokyo) for valuable suggestions and Drs. T. Iwaki and S. Suzuki (Kyushu University) for providing autopsied MS samples. This work was supported, in part, by Grants-in-Aid for Scientific Research from the Ministry of Education, Science, Culture, Sports, and Technology of Japan (T.S., S.I., Y. Kita, and J.-i.K.), Health and Labor Sciences Research Grants for the Comprehensive Research on Aging and Health (S.I.) and the Research on Allergic disease and Immunology (S.I.) from the Ministry of Health, Labor and Welfare of Japan, grants to the Respiratory Failure Research Group (S.I.) and the Research Committee of Neuroimmunological Diseases (J.-i.K.) from the Ministry of Health, Labor and Welfare of Japan, a grant from the Kato Memorial Trust for Nambyo Research (S.I.), and the Research Fellowships of Japanese Society for the Promotion of Science (Y. Kihara).

Footnotes

The authors declare no conflict of interest.

This article is a PNAS Direct Submission.

This article contains supporting information online at www.pnas.org/cgi/content/full/0906891106/DCSupplemental.

References

  • 1.McFarland HF, Martin R. Multiple sclerosis: A complicated picture of autoimmunity. Nat Immunol. 2007;8:913–919. doi: 10.1038/ni1507. [DOI] [PubMed] [Google Scholar]
  • 2.Bettelli E, Korn T, Oukka M, Kuchroo VK. Induction and effector functions of T(H)17 cells. Nature. 2008;453:1051–1057. doi: 10.1038/nature07036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Bettelli E, Oukka M, Kuchroo VK. T(H)-17 cells in the circle of immunity and autoimmunity. Nat Immunol. 2007;8:345–350. doi: 10.1038/ni0407-345. [DOI] [PubMed] [Google Scholar]
  • 4.Kroenke MA, Carlson TJ, Andjelkovic AV, Segal BM. IL-12- and IL-23-modulated T cells induce distinct types of EAE based on histology, CNS chemokine profile, and response to cytokine inhibition. J Exp Med. 2008;205:1535–1541. doi: 10.1084/jem.20080159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Kanter JL, et al. Lipid microarrays identify key mediators of autoimmune brain inflammation. Nat Med. 2006;12:138–143. doi: 10.1038/nm1344. [DOI] [PubMed] [Google Scholar]
  • 6.Shimizu T. Lipid mediators in health and disease: Enzymes and receptors as therapeutic targets for the regulation of immunity and inflammation. Annu Rev Pharmacol Toxicol. 2009;49:123–150. doi: 10.1146/annurev.pharmtox.011008.145616. [DOI] [PubMed] [Google Scholar]
  • 7.Shimizu T, Ohto T, Kita Y. Cytosolic phospholipase A2: Biochemical properties and physiological roles. IUBMB Life. 2006;58:328–333. doi: 10.1080/15216540600702289. [DOI] [PubMed] [Google Scholar]
  • 8.Funk CD. Prostaglandins and leukotrienes: Advances in eicosanoid biology. Science. 2001;294:1871–1875. doi: 10.1126/science.294.5548.1871. [DOI] [PubMed] [Google Scholar]
  • 9.Narumiya S, FitzGerald GA. Genetic and pharmacological analysis of prostanoid receptor function. J Clin Invest. 2001;108:25–30. doi: 10.1172/JCI13455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Kalyvas A, David S. Cytosolic phospholipase A2 plays a key role in the pathogenesis of multiple sclerosis-like disease. Neuron. 2004;41:323–335. doi: 10.1016/s0896-6273(04)00003-0. [DOI] [PubMed] [Google Scholar]
  • 11.Marusic S, et al. Cytosolic phospholipase A2 alpha-deficient mice are resistant to experimental autoimmune encephalomyelitis. J Exp Med. 2005;202:841–851. doi: 10.1084/jem.20050665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Marusic S, et al. Blockade of cytosolic phospholipase A(2)alpha prevents experimental autoimmune encephalomyelitis and diminishes development of Th1 and Th17 responses. J Neuroimmunol. 2008;204:29–37. doi: 10.1016/j.jneuroim.2008.08.012. [DOI] [PubMed] [Google Scholar]
  • 13.Kihara Y, et al. Platelet-activating factor production in the spinal cord of experimental allergic encephalomyelitis mice via the group IVA cytosolic phospholipase A2-Lyso-PAFAT axis. J Immunol. 2008;181:5008–5014. doi: 10.4049/jimmunol.181.7.5008. [DOI] [PubMed] [Google Scholar]
  • 14.Miyamoto K, et al. Selective COX-2 inhibitor celecoxib prevents experimental autoimmune encephalomyelitis through COX-2-independent pathway. Brain. 2006;129:1984–1992. doi: 10.1093/brain/awl170. [DOI] [PubMed] [Google Scholar]
  • 15.Emerson MR, LeVine SM. Experimental allergic encephalomyelitis is exacerbated in mice deficient for 12/15-lipoxygenase or 5-lipoxygenase. Brain Res. 2004;1021:140–145. doi: 10.1016/j.brainres.2004.06.045. [DOI] [PubMed] [Google Scholar]
  • 16.Murakami M, Kambe T, Shimbara S, Kudo I. Functional coupling between various phospholipase A2s and cyclooxygenases in immediate and delayed prostanoid biosynthetic pathways. J Biol Chem. 1999;274:3103–3115. doi: 10.1074/jbc.274.5.3103. [DOI] [PubMed] [Google Scholar]
  • 17.Murakami M, et al. Regulation of prostaglandin E2 biosynthesis by inducible membrane-associated prostaglandin E2 synthase that acts in concert with cyclooxygenase-2. J Biol Chem. 2000;275:32783–32792. doi: 10.1074/jbc.M003505200. [DOI] [PubMed] [Google Scholar]
  • 18.Ueno N, et al. Coupling between cyclooxygenase, terminal prostanoid synthase, and phospholipase A2. J Biol Chem. 2001;276:34918–34927. doi: 10.1074/jbc.M100429200. [DOI] [PubMed] [Google Scholar]
  • 19.Kihara Y, et al. Dual phase regulation of experimental allergic encephalomyelitis by platelet-activating factor. J Exp Med. 2005;202:853–863. doi: 10.1084/jem.20050660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Hofstetter AO, et al. The induced prostaglandin E2 pathway is a key regulator of the respiratory response to infection and hypoxia in neonates. Proc Natl Acad Sci USA. 2007;104:9894–9899. doi: 10.1073/pnas.0611468104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Ikeda-Matsuo Y, et al. Microsomal prostaglandin E synthase-1 is a critical factor of stroke-reperfusion injury. Proc Natl Acad Sci USA. 2006;103:11790–11795. doi: 10.1073/pnas.0604400103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Samuelsson B, Morgenstern R, Jakobsson PJ. Membrane prostaglandin E synthase-1: A novel therapeutic target. Pharmacol Rev. 2007;59:207–224. doi: 10.1124/pr.59.3.1. [DOI] [PubMed] [Google Scholar]
  • 23.Ponomarev ED, Shriver LP, Maresz K, Dittel BN. Microglial cell activation and proliferation precedes the onset of CNS autoimmunity. J Neurosci Res. 2005;81:374–389. doi: 10.1002/jnr.20488. [DOI] [PubMed] [Google Scholar]
  • 24.Smith WL, DeWitt DL, Garavito RM. Cyclooxygenases: Structural, cellular, and molecular biology. Annu Rev Biochem. 2000;69:145–182. doi: 10.1146/annurev.biochem.69.1.145. [DOI] [PubMed] [Google Scholar]
  • 25.Rose JW, Hill KE, Watt HE, Carlson NG. Inflammatory cell expression of cyclooxygenase-2 in the multiple sclerosis lesion. J Neuroimmunol. 2004;149:40–49. doi: 10.1016/j.jneuroim.2003.12.021. [DOI] [PubMed] [Google Scholar]
  • 26.Deininger MH, Schluesener HJ. Cyclooxygenases-1 and -2 are differentially localized to microglia and endothelium in rat EAE and glioma. J Neuroimmunol. 1999;95:202–208. doi: 10.1016/s0165-5728(98)00257-4. [DOI] [PubMed] [Google Scholar]
  • 27.Sheibanie AF, Khayrullina T, Safadi FF, Ganea D. Prostaglandin E2 exacerbates collagen-induced arthritis in mice through the inflammatory interleukin-23/interleukin-17 axis. Arthritis Rheum. 2007;56:2608–2619. doi: 10.1002/art.22794. [DOI] [PubMed] [Google Scholar]
  • 28.Sheibanie AF, et al. Prostaglandin E2 induces IL-23 production in bone marrow-derived dendritic cells. FASEB J. 2004;18:1318–1320. doi: 10.1096/fj.03-1367fje. [DOI] [PubMed] [Google Scholar]
  • 29.Sheibanie AF, et al. The proinflammatory effect of prostaglandin E2 in experimental inflammatory bowel disease is mediated through the IL-23–>IL-17 axis. J Immunol. 2007;178:8138–8147. doi: 10.4049/jimmunol.178.12.8138. [DOI] [PubMed] [Google Scholar]
  • 30.Yao C, et al. Prostaglandin E2-EP4 signaling promotes immune inflammation through TH1 cell differentiation and TH17 cell expansion. Nat Med. 2009;15:633–640. doi: 10.1038/nm.1968. [DOI] [PubMed] [Google Scholar]
  • 31.Chizzolini C, et al. Prostaglandin E2 (PGE2) synergistically with interleukin-23 (IL-23) favors human Th17 expansion. Blood. 2008;112:3696–3703. doi: 10.1182/blood-2008-05-155408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Boniface K, et al. Prostaglandin E2 regulates Th17 cell differentiation and function through cyclic AMP and EP2/EP4 receptor signaling. J Exp Med. 2009;206:535–548. doi: 10.1084/jem.20082293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Nagamachi M, et al. Facilitation of Th1-mediated immune response by prostaglandin E receptor EP1. J Exp Med. 2007;204:2865–2874. doi: 10.1084/jem.20070773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Fukuhara S, et al. Cyclic AMP potentiates vascular endothelial cadherin-mediated cell–cell contact to enhance endothelial barrier function through an Epac-Rap1 signaling pathway. Mol Cell Biol. 2005;25:136–146. doi: 10.1128/MCB.25.1.136-146.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Boulet L, et al. Deletion of microsomal prostaglandin E2 (PGE2) synthase-1 reduces inducible and basal PGE2 production and alters the gastric prostanoid profile. J Biol Chem. 2004;279:23229–23237. doi: 10.1074/jbc.M400443200. [DOI] [PubMed] [Google Scholar]
  • 36.Trebino CE, et al. Redirection of eicosanoid metabolism in mPGES-1-deficient macrophages. J Biol Chem. 2005;280:16579–16585. doi: 10.1074/jbc.M412075200. [DOI] [PubMed] [Google Scholar]
  • 37.Wang M, et al. Deletion of microsomal prostaglandin E synthase-1 augments prostacyclin and retards atherogenesis. Proc Natl Acad Sci USA. 2006;103:14507–14512. doi: 10.1073/pnas.0606586103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Honda T, Segi-Nishida E, Miyachi Y, Narumiya S. Prostacyclin-IP signaling and prostaglandin E2-EP2/EP4 signaling both mediate joint inflammation in mouse collagen-induced arthritis. J Exp Med. 2006;203:325–335. doi: 10.1084/jem.20051310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Taniike M, et al. Perineuronal oligodendrocytes protect against neuronal apoptosis through the production of lipocalin-type prostaglandin D synthase in a genetic demyelinating model. J Neurosci. 2002;22:4885–4896. doi: 10.1523/JNEUROSCI.22-12-04885.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Kagitani-Shimono K, et al. Lipocalin-type prostaglandin D synthase (beta-trace) is upregulated in the alphaB-crystallin-positive oligodendrocytes and astrocytes in the chronic multiple sclerosis. Neuropathol Appl Neurobiol. 2006;32:64–73. doi: 10.1111/j.1365-2990.2005.00690.x. [DOI] [PubMed] [Google Scholar]
  • 41.Kanaoka Y, Urade Y. Hematopoietic prostaglandin D synthase. Prostaglandins Leukot Essent Fatty Acids. 2003;69:163–167. doi: 10.1016/s0952-3278(03)00077-2. [DOI] [PubMed] [Google Scholar]
  • 42.Haeggstrom JZ. Leukotriene A4 hydrolase/aminopeptidase, the gatekeeper of chemotactic leukotriene B4 biosynthesis. J Biol Chem. 2004;279:50639–50642. doi: 10.1074/jbc.R400027200. [DOI] [PubMed] [Google Scholar]
  • 43.Radmark O, Werz O, Steinhilber D, Samuelsson B. 5-Lipoxygenase: Regulation of expression and enzyme activity. Trends Biochem Sci. 2007;32:332–341. doi: 10.1016/j.tibs.2007.06.002. [DOI] [PubMed] [Google Scholar]
  • 44.Welsch DJ, et al. Molecular cloning and expression of human leukotriene-C4 synthase. Proc Natl Acad Sci USA. 1994;91:9745–9749. doi: 10.1073/pnas.91.21.9745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Gladue RP, et al. Inhibition of leukotriene B4-receptor interaction suppresses eosinophil infiltration and disease pathology in a murine model of experimental allergic encephalomyelitis. J Exp Med. 1996;183:1893–1898. doi: 10.1084/jem.183.4.1893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Serhan CN, Chiang N, Van Dyke TE. Resolving inflammation: Dual anti-inflammatory and pro-resolution lipid mediators. Nat Rev Immunol. 2008;8:349–361. doi: 10.1038/nri2294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Serhan CN, Savill J. Resolution of inflammation: The beginning programs the end. Nat Immunol. 2005;6:1191–1197. doi: 10.1038/ni1276. [DOI] [PubMed] [Google Scholar]
  • 48.Arita M, et al. Stereochemical assignment, antiinflammatory properties, and receptor for the omega-3 lipid mediator resolvin E1. J Exp Med. 2005;201:713–722. doi: 10.1084/jem.20042031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Wolfe MM, Lichtenstein DR, Singh G. Gastrointestinal toxicity of nonsteroidal anti-inflammatory drugs. N Engl J Med. 1999;340:1888–1899. doi: 10.1056/NEJM199906173402407. [DOI] [PubMed] [Google Scholar]
  • 50.James MJ, Cook-Johnson RJ, Cleland LG. Selective COX-2 inhibitors, eicosanoid synthesis and clinical outcomes: A case study of system failure. Lipids. 2007;42:779–785. doi: 10.1007/s11745-007-3069-x. [DOI] [PubMed] [Google Scholar]
  • 51.Juni P, et al. Risk of cardiovascular events and rofecoxib: Cumulative meta-analysis. Lancet. 2004;364:2021–2029. doi: 10.1016/S0140-6736(04)17514-4. [DOI] [PubMed] [Google Scholar]
  • 52.Xu D, et al. MF63 [2-(6-chloro-1H-phenanthro[9,10-d]imidazol-2-yl)-isophthalonitrile], a selective microsomal prostaglandin E synthase-1 inhibitor, relieves pyresis and pain in preclinical models of inflammation. J Pharmacol Exp Ther. 2008;326:754–763. doi: 10.1124/jpet.108.138776. [DOI] [PubMed] [Google Scholar]
  • 53.Uematsu S, Matsumoto M, Takeda K, Akira S. Lipopolysaccharide-dependent prostaglandin E(2) production is regulated by the glutathione-dependent prostaglandin E(2) synthase gene induced by the Toll-like receptor 4/MyD88/NF-IL6 pathway. J Immunol. 2002;168:5811–5816. doi: 10.4049/jimmunol.168.11.5811. [DOI] [PubMed] [Google Scholar]
  • 54.Kita Y, et al. Pathway-oriented profiling of lipid mediators in macrophages. Biochem Biophys Res Commun. 2005;330:898–906. doi: 10.1016/j.bbrc.2005.03.055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Kita Y, Takahashi T, Uozumi N, Shimizu T. A multiplex quantitation method for eicosanoids and platelet-activating factor using column-switching reversed-phase liquid chromatography-tandem mass spectrometry. Anal Biochem. 2005;342:134–143. doi: 10.1016/j.ab.2005.03.048. [DOI] [PubMed] [Google Scholar]

Associated Data

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

Supplementary Materials

Supporting Information

Articles from Proceedings of the National Academy of Sciences of the United States of America are provided here courtesy of National Academy of Sciences

RESOURCES