Skip to main content

Resolvin D1 promotes the interleukin-4-induced alternative activation in BV-2 microglial cells

Abstract

Background

Microglia play key roles in innate immunity, homeostasis, and neurotropic support in the central nervous system. Similar to macrophages, microglia adopt two different activation phenotypes, the classical and alternative activation. Resolvin D1 (RvD1) is considered to display potent anti-inflammatory and pro-resolving actions in inflammatory models. In this present study, we investigate the effect of RvD1 on IL-4-induced alternative activation in murine BV-2 microglial cells.

Methods

BV-2 cells were incubated with RvD1 alone, IL-4 alone, or the combination of RvD1 and IL-4. Western blot and immunofluorescence were performed to detect protein levels of alternative activation markers arginase 1 (Arg1), chitinase 3-like 3 (Ym1). Moreover, we investigated the effects of RvD1 on IL-4-induced activation of signal transducer and activators of transcription 6 (STAT6) and peroxisome proliferator-activated receptor gamma (PPARĪ³).

Results

RvD1 promoted IL-4-induced microglia alternative activation by increasing the expression of Arg1 and Ym1. RvD1 also enhanced phosphorylation of STAT6, nuclear translocation of PPARĪ³ and the DNA binding activity of STAT6 and PPARĪ³. These effects were reversed by butyloxycarbonyl-Phe-Leu-Phe-Leu-Phe (a formyl peptide receptor 2 antagonist). Further, the effects of RvD1 and IL-4 on Arg1 and Ym1 were blocked by the application of leflunomide (a STAT6 inhibitor) or GW9662 (a PPARĪ³ antagonist).

Conclusions

Our studies demonstrate that RvD1 promotes IL-4-induced alternative activation via STAT6 and PPARĪ³ signaling pathways in microglia. These findings suggest that RvD1 may have therapeutic potential for neuroinflammatory diseases.

Background

Microglia are considered to be the resident immune cells of the central nervous system (CNS). In the normal CNS, microglia survey their surrounding microenvironment [1]. However, in a pathological state, microglia respond to environmental changes with a variety of activation states, changing both morphology and phenotype [2].

It is becoming increasingly clear that microglia, like macrophages, have at least two polarized activation states: classical and alternative activation [3ā€“6]. Classical activation (M1) induced by lipopolysaccharide (LPS) or Th1 cytokines is associated with elevations of pro-inflammation cytokines such as IL-1, IL-6 and TNF-Ī±, nitric oxide (NO) and reactive oxygen species (ROS), which are involved in pathogen destruction and cytotoxicity [7, 8]. Alternative activation (M2) induced by Th2 cytokines IL-4, IL-13 or IL-10 is associated with elevation of arginase 1 (Arg1), chitinase 3-like 3 (Ym1), mannose receptor and found in inflammatory zone 1 (FIZZ1), which is primarily involved in tissue remodeling and healing [7, 8]. Therefore, modulating microglia activation states may be a potential therapeutic targeting of neuroinflammatory diseases.

Signal transducer and activators of transcription 6 (STAT6) is activated by IL-4 and IL-13 and plays an important role in alternative activation. STAT6 is able to regulate transcription of genes typical of M2 activation [9, 10]. Downstream of STAT6, peroxisome proliferator-activated receptor gamma (PPARĪ³) is activated in a STAT6-dependent manner [11]. PPARĪ³ is a member of the nuclear hormone receptor superfamily and is considered to associate with lipid metabolism, oxidative metabolism, macrophage and microglia M2 polarization [12ā€“14].

Resolvins are a family of bioactive metabolites derived from omega-3 fatty acids docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) and display potent anti-inflammatory and pro-resolving actions in inflammatory models [15]. Resolvin D1 (7S,8R,17S-trihydroxy-4Z,9E,11E,13Z,15E,19Z-DHA, RvD1) is produced physiologically from the sequential oxygenation of DHA by 15-lipoxygenase and 5-lipoxygenase [16]. RvD1 directly activates lipoxin A4 receptor/formyl peptide receptor 2 (ALX/FPR2) and orphan receptor G protein coupling receptor 32 (GPR32), limits excessive leukocyte infiltration, attenuates the production of pro-inflammatory cytokines [17], regulates macrophage phagocytosis [18] and up-regulates pro-resolving miRNAs [19]. RvD1 can also elicite macrophage polarization toward an M2-like phenotype [20]. Previous research has indicated that another ALX/FPR2 activator, aspirin-triggered lipoxin A4, can down-regulate the LPS-induced expression of M1 markers in microglia [21ā€“23]. However, the effect of RvD1 on microglia M2 polarization and molecular mechanisms is still unknown.

Here, we investigated the impact of RvD1 on IL-4-induced expression of M2 markers in BV-2 microglial cells and the signaling pathways involving in these processes. Our data show that RvD1 up-regulates expression of Arg1 and Ym1 in IL-4-treated microglial cells depending on STAT6 and PPARĪ³ signaling pathways.

Methods

Cell culture

The immortalized murine microglia cell line BV-2 was purchased from the Cell Resource Center of Peking Union Medical College (Beijing, China) and maintained in DMEM with F12 supplement (DMEM/F12, Gibco, Grand Island, NY, USA) supplemented with 10% FBS (Gibco, Grand Island, NY, USA), 100 U/ml penicillin and 100 Ī¼g/ml streptomycin at 37Ā°C in a humidified atmosphere of 95% air and 5% CO2. Before each experiment, cells were serum-starved for 12 hours. BV-2 cells were incubated with different concentrations (1 nM, 10 nM or 100 nM) of RvD1 (Cayman Chemical, Ann Arbor, MI, USA) or vehicle (0.038% ethanol) for 30 minutes before addition of 10 ng/ml murine IL-4 (PeproTech, Hamburg, Germany) under serum-free conditions. To investigate the involvement of ALX, STAT6 or PPARĪ³, the cells were treated with 10 Ī¼M butyloxycarbonyl-Phe-Leu-Phe-Leu-Phe (Boc-2) (GenScript Corporation, Piscataway, NJ, USA), 100 Ī¼M leflunomide (Sigma-Aldrich, St. Louis, MO, USA) or 1 Ī¼M GW9662 (Sigma-Aldrich, St. Louis, MO, USA) prior to the treatment with RvD1 for 30 minutes.

Immunocytochemistry

BV-2 cells were cultured on sterile glass cover slips and treated according to the experimental design. Afterward, cells were fixed with 4% paraformaldehyde in PBS and permeabilized with 0.1% Triton X-100 in PBS. After rinsing, cells were blocked with 3% BSA in PBS for one hour and incubated with primary antibodies overnight at 4Ā°C. The primary antibodies used were as follows: rabbit anti-arginase-1 (1:100, Santa Cruz, Heidelberg, Germany), Ym1 (1:50, Stem Cell Technologies, Vancouver, Canada), PPARĪ³ (1:100, Santa Cruz, Heidelberg, Germany). After washing, cells were incubated with FITC-conjugated goat anti-rabbit IgG (1:400, Jackson Immuno Research Laboratories, West Grove, PA, USA) for one hour and counterstained with 4, 6-diamidino-2-phenylindole (DAPI, Roche, Shanghai, China) for the identification of nuclei. After washing with PBS, the cover slips were mounted with antifade mounting medium (Beyotime, Shanghai, China) on slides, and the cells were observed with an Olympus immunofluorescence microscope (Olympus, Tokyo, Japan).

Protein extraction

For making whole cell lysates, the cells were lysed in radioimmune precipitation assay (RIPA) buffer supplemented with protease inhibitor cocktail (Roche, Shanghai, China). Nuclear and cytoplasmic fractionations were performed with NE-PER Nuclear and Cytoplasmic Extraction Reagents (Thermo Scientific, Rockford, IL, USA) according to manufacturerā€™s protocol.

Western blot analysis

Equal amounts of nuclear or whole cell extracts were electrophoresed on sodium dodecyl sulfate-polyacrylamide gels, and then transferred onto a polyvinylidene difluoride membrane (Millipore, Schwalbach, Germany). The transformed membrane was blocked with 5% non-fat dry milk in Tris-buffered saline containing 0.05% Tween-20 (TBST) for one hour and incubated with primary antibodies overnight at 4Ā°C. The primary antibodies used were as follows: rabbit anti-arginase-1 (1:500), Ym1 (1:1,000), PPARĪ³ (1:500), phospho-Stat6 (1:500, Santa Cruz, Heidelberg, Germany), Ī²-actin (1:1,000, Santa Cruz, Heidelberg, Germany), lamin B1 (1:1,000, Santa Cruz, Heidelberg, Germany). The membrane was washed three times with TBST for ten minutes and incubated with anti-rabbit IgG-horseradish peroxidase (1:5,000, Jackson ImmunoResearch Laboratories, West Grove, PA, USA) at room temperature for one hour. The Supersignal West Pico chemiluminescent substrate system (Millipore, Schwalbach, Germany) was used to detect immunoreactive bands. The intensity of protein bands after Western blot were quantitated by using Quantity One Version 4.6.3 Image software (Bio-Rad, Hercules, CA, USA) and normalized against proper loading controls.

Electrophoretic mobility shift assay (EMSA)

Nuclear extracts were prepared as described above. Oligonucleotides corresponding to the STAT6 (5ā€²-TGCCTTAGTCAACTTCCCAAGAACAGA-3ā€²) and PPARĪ³ (5ā€²- GGAACTAGGTCAAAGGTCATCCCCT-3ā€²) binding site consensus sequences were synthesized and end-labeled with biotin by Invitrogen (Invitrogen, Shanghai, China). EMSAs were performed using the LightShift chemiluminescent EMSA kit (Thermo Scientific, Rockford, IL, USA). Briefly, 10 fmol of biotin-labeled, double strand probe were incubated for 20 minutes at room temperature in 20 Ī¼l of EMSA binding buffer containing 2.5% glycerol, 5 mM MgCl2, 50 ng/Ī¼l poly (dI-dC), 0.05% Nonidet P-40, and 6 Ī¼g of nuclear proteins. For competition EMSA, 200-fold (2 pmol) excess unlabeled, double strand probe was added to the binding reaction. The DNA-nuclear protein complexes were resolved by electrophoresis in 6% nondenaturing polyacrylamide gel in 0.5ā€‰Ć—ā€‰Tris-borate-EDTA (TBE) buffer at 100 V. Gels were then electroblotted onto Hybond nylon membranes (GE Healthcare, Freiburg, Germany) at 380 mA for 50 minutes. The membranes were then cross-linked for 15 minutes with the membrane face down on a transilluminator at 312 nm, and the biotinylated protein-DNA bands were detected with HRP-conjugated streptavidin using the chemiluminescent nucleic acid detection system.

Statistical analysis

Data are expressed as meansā€‰Ā±ā€‰SEM of the indicated number of independent experiments. Statistical significance between multiple groups was analyzed by one-way ANOVA. Least significant difference (LSD) post hoc test was used for multiple comparisons. Statistical analysis was performed using the SPSS software version 13.0 (SPSS Inc., Chicago, IL, USA). Pā€‰<ā€‰0.05 was considered statistically significant.

Results

RvD1 promotes IL-4-induced alternative activation of BV-2 microglial cells

Initially, we evaluated the effects of RvD1 on IL-4-induced microglia alternative activation; BV-2 cells were treated with vehicle or different concentrations of RvD1 (1 nM, 10 nM and 100 nM) for 30 minutes and stimulated with IL-4 (10 ng/ml) for 24 hours. As the assessment of the alternative activation relied on the molecule markers such as Arg1 and Ym1, the expression of Arg1 and Ym1 were analyzed. We tested the protein levels of Arg1 and Ym1 using Western blot analysis. As shown in FigureĀ 1A, IL-4 stimulation significantly enhanced Arg1 and Ym1 protein levels in BV-2 cells (P <0.01). Treatment with RvD1 alone did not affect Arg1 and Ym1 protein levels (Pā€‰>ā€‰0.05); while combination of RvD1 and IL-4 markedly increased expression of Arg1 and Ym1 in BV-2 cells compared to IL-4 treatment alone, the effects of RvD1 were concentration dependent. Immunofluorescence staining demonstrated that Arg1 and Ym1 staining intensity increased after IL-4 treatment, pretreatment with RvD1 further increased the staining intensity (FigureĀ 1C and D).

Figure 1
figure 1

RvD1 promotes the IL-4-induced alternative activation of BV-2 cells. BV-2 cells were treated with vehicle (0.038% ethanol) or different concentrations of RvD1 (1 nM, 10 nM and 100 nM) for 30 minutes in the absence or presence of Boc-2 (10 Ī¼M), and then stimulated by IL-4 (10 ng/ml) for 24 hours. (A, B) The expression of Arg1 and Ym1 proteins was assessed by Western blot. A representative result from three independent experiments is shown. Quantification for Arg1 and Ym1 was normalized by Ī²-actin. Data are presented as meanā€‰Ā±ā€‰SEM for three independent experiments. Asterisks indicate statistically significant difference (*Pā€‰<ā€‰0.05, **Pā€‰<ā€‰0.01). (C, D) Representative images of BV-2 cells with positive immunofluorescence staining for Arg1 (C) and Ym1 (D). Scale bars indicate 20 Ī¼m.

To evaluate the role of ALX/FPR2 in the effects of RvD1, an ALX antagonist Boc-2 (10 Ī¼M) was administrated 30 minutes prior to treatment with RvD1. The effects in response to RvD1 were reversed by Boc-2 (FigureĀ 1B-D).

RvD1 promotes IL-4-induced STAT6 phosphorylation and activation

STAT6 plays a critical role in IL-4-dependent induction of Arg1 and Ym1 [24, 25]. As RvD1 promotes IL-4-induced expression of Arg1 and Ym1 (FigureĀ 1), we investigated the involvement of STAT6 activation in these effects. STAT6 phosphorylated at 60 minutes after IL-4 treated (FigureĀ 2A), the 60 minute treatment of IL-4 was determined to examine the effect of RvD1 on STAT6 phosphorylation. RvD1 markedly enhanced IL-4-induced phosphorylation of STAT6 (Pā€‰<ā€‰0.01), the effect of RvD1 was reversed by Boc-2 (P <0.05, FigureĀ 2B). Further, we performed EMSA to determine the effects RvD1 and IL-4 on STAT6 DNA binding activity in BV-2 cells. IL-4 treatment increased STAT6 binding activity, combination of RvD1 and IL-4 further increased the activity. Moreover, treatment with Boc-2 reversed the effect of RvD1 (FigureĀ 2C).

Figure 2
figure 2

RvD1 enhances IL-4-induced STAT6 activation. (A) BV-2 cells were treated with IL-4 (10 ng/ml) for the indicated times, expression of phosphorylated STAT6 was assessed by Western blot. Quantification of phospo-STAT6 from three independent experiments indicated that the effect of IL-4 was obvious at 60 minutes. (B) BV-2 cells were treated with RvD1 (100 nM), IL-4 (10 ng/ml), Boc-2 (10 Ī¼M) or combination of them. Levels of phosphorylated STAT6 were detected by Western blot 60 minutes after IL-4 stimulation. A representative result from three independent experiments is shown. Quantification of phosphorylated STAT6 was normalized by Ī²-actin. (C) BV-2 cells were treated as in (B). Nuclear extracts were prepared 120 minutes after IL-4 treatment and used to analyze STAT6 DNA binding activity by Electrophoretic Mobility Shift Assay (EMSA). Binding specificity was confirmed by unlabelled probe (200-fold in excess) to compete with labeled oligonucleotide. Results were confirmed by three independent experiments. Data are presented as meanā€‰Ā±ā€‰SEM for three independent experiments. Asterisks indicate statistically significant difference (*Pā€‰<ā€‰0.05, **Pā€‰<ā€‰0.01).

RvD1 promotes IL-4-induced PPARĪ³ nuclear translocation and activation

Along with STAT6, PPARĪ³ is known to control genes associated with alternatively activation [12, 26]. Therefore, we examined the effects of RvD1 on IL-4-stimulated PPARĪ³ nuclear translocation and DNA binding activity in BV-2 cells. Immunofluorescence staining indicated that PPARĪ³ translocated from the cytoplasm into the nucleus obviously 120 minutes after IL-4 stimulation (FigureĀ 3A). Then we tested whether IL-4 and RvD1 affected PPARĪ³ protein levels in nucleus and DNA binding activity. As shown in FigureĀ 3B and C, IL-4 increased PPARĪ³ protein levels in nucleus and DNA binding activity. RvD1 alone failed to activate PPARĪ³ (Pā€‰>ā€‰0.05). However, combination of RvD1 and IL-4 further increased PPARĪ³ protein levels in nucleus and DNA binding activity compared to treatment with IL-4 alone, the effects of RvD1 were abolished by pretreatment with Boc-2.

Figure 3
figure 3

RvD1 enhances IL-4-induced PPARĪ³ activation. (A) BV-2 cells were treated with IL-4 (10 ng/ml) for the indicated times, subcellular localization of PPARĪ³ was evaluated using immunofluorescence staining. DNA was stained using 6-diamidino-2-phenylindole (DAPI) to visualize nuclei. The nuclear translocation of PPARĪ³ was obvious 120 minutes after stimulation. Scale bars indicate 20 Ī¼m. (B, C) BV-2 cells were treated with RvD1 (100 nM), IL-4 (10 ng/ml), Boc-2 (10 Ī¼M) or a combination of them. Nuclear extracts were prepared 120 minutes after IL-4 treatment, Western blot or electrophoretic mobility shift assay (EMSA) were performed to detect PPARĪ³ protein level in nucleus (B) or DNA binding activity (C). A representative result from three independent experiments is shown. Quantification of PPARĪ³ was normalized by lamin B1. Data are presented as meanā€‰Ā±ā€‰SEM for three independent experiments. Asterisks indicate statistically significant difference (*Pā€‰<ā€‰0.05, **Pā€‰<ā€‰0.01).

STAT6 is required for PPARĪ³ activation induced by IL-4 and RvD1

STAT6 is a regulator of PPARĪ³ response; IL-4 induces augmented PPARĪ³ activity through a STAT6-dependent mechanism [11, 27]. To identify the role of STAT6 on PPARĪ³ activation induced by IL-4 and RvD1, we used leflunomide (an inhibitor suppressing tyrosine phosphorylation of STAT6 and preventing subsequent DNA binding) and GW9662 (a PPARĪ³ antagonist) [26, 28]. One hundred micromoles of leflunomide or 1 Ī¼M GW9662 was administrated 30 minutes prior to treatment with RvD1, PPARĪ³ expression and DNA binding activity were analyzed. Consistent with GW9662, leflunomide decreased PPARĪ³ protein levels in nucleus and DNA binding activity in IL-4 and RvD1 treated BV-2 cells (FigureĀ 4).

Figure 4
figure 4

STAT6 is required for PPARĪ³ activation. BV-2 cells were treated with leflunomide (10 Ī¼M) or GW9662 (1 Ī¼M) 30 minutes prior to administration of RvD1 (100 nM), followed by incubation with IL-4 (10 ng/ml) for 120 minutes. PPARĪ³ protein level in nucleus (A) and DNA binding activity (B) were analyzed using Western blot or electrophoretic mobility shift assay (EMSA). A representative result from three independent experiments is shown. Quantification of PPARĪ³ was normalized by lamin B1. Data are presented as meanā€‰Ā±ā€‰SEM for three independent experiments. Asterisks indicate statistically significant difference (*Pā€‰<ā€‰0.05, **Pā€‰<ā€‰0.01).

STAT6 and PPARĪ³ are essential for induction of Arg1 and Ym1 by IL-4 and RvD1

RvD1 promotes IL-4-induced STAT6 and PPARĪ³ activation, these facts suggest, but do not prove, that STAT6 and PPARĪ³ are required for the effects of RvD1 and IL-4 on Arg1 and Ym1 induction. To directly test the effects of STAT6 and PPARĪ³, BV-2 cells were pretreated with 100 Ī¼M leflunomide or 1 Ī¼M GW9662 for 30 minutes to inhibit STAT6 or PPARĪ³ signaling pathway and stimulated with RvD1 and IL-4 for 24 hours. Immunofluorescence and Western blot were performed to determine Arg1 and Ym1 expression. As expected, Arg1 and Ym1 induction by RvD1 and Ym1 was markedly blunted by inhibition of STAT6 and PPARĪ³ (FigureĀ 5).

Figure 5
figure 5

Inhibition of STAT6 and PPARĪ³ impacts microglia alternative activation induced by IL-4 and RvD1. BV-2 cells were treated with leflunomide (10 Ī¼M) or GW9662 (1 Ī¼M) 30 minutes prior to administration of RvD1 (100 nM) and then stimulated with IL-4 (10 ng/ml) for 24 hours. Immunofluorescence assay for Arg1 (A) and Ym1 (B) indicated that the induction of Arg1 and Ym1 by RvD1 and IL-4 was disappeared when blocking STAT6 or PPARĪ³ signaling pathway. Scale bars indicate 20 Ī¼m. (C) The protein levels of Arg1 and Ym1 were measured by Western blot. A representative result from three independent experiments is shown. Quantification for Arg1 and Ym1 was normalized by Ī²-actin. Data are presented as meanā€‰Ā±ā€‰SEM for three independent experiments. Asterisks indicate statistically significant difference (*Pā€‰<ā€‰0.05, **Pā€‰<ā€‰0.01).

Discussion

In this study, we provide the first evidence that RvD1 enhances the IL-4-induced M2 polarization in BV-2 microglial cells. Pretreatment with RvD1 was able to increase IL-4-induced expression of two major alternative activation markers, Arg1 and Ym1. In addition, RvD1 potentiated the effects of IL-4 on STAT6 and PPARĪ³ signaling pathways, administering leflunomide or GW9662 before RvD1 and IL-4 blunted the induction of Arg1 and Ym1. These results suggest that RvD1 can promote IL-4-induced M2 polarization in microglial cells via STAT6 and PPARĪ³ signaling pathways.

So far, two receptors for RvD1 have been indentified, ALX/FPR2 and an orphan, GPR32. RvD1 activates ALX and GPR32 with high affinity (EC50 approximately 1.2 pM for ALX/FPR2 and 8.8 pM for GPR32) [18]. Interestingly, on human polymorphonuclear leukocytes, low concentration of RvD1 (1 nM) appear sensitive to GPR32 blockade, while responses to high concentration (10 nM) are ALX-dependent [29]. Our findings demonstrated that RvD1 promoted IL-4-induced microglia alternative activation with high concentrations (10 nM and 100 nM). Moreover, in murine physiology, we did not identify the murine orthologs of human GPR32, while mouse ALX has been confirmed to express in neuron, astrocyte, microglia, neutrophils, macrophage and monocyte, as well as BV-2 cells [21, 30]. Boc-2 is an ALX/FPR2 antagonist, and can block the ability of RvD1 [31]. Thus, we investigated the receptor dependency of RvD1 in microglia M2 polarization by using Boc-2. Previous studies have shown that activation of ALX by lipoxin A4, RvD1, BML-111 or other agonists have potential anti-inflammatory and pro-resolving effects in a rat brain ischemia-reperfusion injury model [32, 33] and other inflammatory disease models [34ā€“37]. These findings suggest that RvD1 could modulate the inflammatory reaction in CNS diseases.

Microglia exhibit various functions at different stages in life or in response to different pathological situations [6]. The activation states of microglia are plastic and flexible, both blunting classical activation and promoting alternative activation are involving in anti-inflammatory and pro-resolving process. Here, we used Arg1 and Ym1 as markers for microglia alternative activation. Expression of Arg1 in microglia is induced by Th2 cytokines [38]. Arg1 can reduce NO production, and it is also involved in wound healing, fibrosis and supporting neuron survival [39ā€“41]. Apart from Arg1, Ym1 is another marker for alternative activation [42]. Ym1 is considered to be associated with tissue remodeling and regulating inflammation [43]. First, we examined the effect of IL-4 and RvD1 on Arg1 and Ym1 expression. According with previous studies, our results showed that expression of Arg1 and Ym1 was low in unstimulated BV-2 microglial cells, which was enhanced by stimulation with 10ng/ml IL-4 [7, 44]. Titos et al. reported that RvD1 and its precursor DHA promoted resolution by eliciting macrophage M2 polarization [20]. In our study, treatment with RvD1 alone had no effect on Arg1 and Ym1 expression in microglia, while pretreatment with RvD1 enhanced the effect of IL-4 on Arg1 and Ym1 expression. This indicated that RvD1 may be possible to promote resolution of neuroinflammation by regulating microglia polarization. The effect of RvD1 was receptor-dependent as it was reversed by the use of ALX antagonist Boc-2.

The synergistic effect of RvD1 and IL-4 on Arg1 and Ym1 expression might be explained by RvD1 potentiating IL-4-induced STAT6 and PPARĪ³ activation. IL-4-induced expression of Arg1 and Ym1 depends on activation of STAT6. In response to IL-4, STAT6 is phosphorylated, translates into the nucleus and binds to consensus sites at the Jmjd3 promoter, then regulates transcriptional activation of M2 marker genes [45]. STAT6 can also modulate the activity of nuclear receptor PPARĪ³ [11]. PPARĪ³ plays an important role in macrophage/microglia alternative activation; the addition of a PPARĪ³ agonist directly regulates the Arg1 and Ym1 expression [12, 14, 46, 47]. In this study, RvD1 was able to enhance the phosphorylation and DNA binding activity of STAT6 induced by IL-4. Inhibiting STAT6 by leflunomide abolished the induction of Arg1 and Ym1. Moreover, leflunomide inhibited PPARĪ³ translocating into the nucleus and binding to DNA sites, which suggested that STAT6 was required for PPARĪ³ activation induced by RvD1 and IL-4. Consistent with STAT6, the activity of PPARĪ³ induced by RvD1 and IL-4 was greater than stimulating with IL-4 alone. A previous study reported that activation of PPARĪ³ by pioglitazone promoted microglia M2 polarization by increasing Arg1 and Ym1 expression [14]. In our research, the effects of RvD1 and IL-4 on Arg1 and Ym1 were impaired by using PPARĪ³ antagonist GW9662, which proved PPARĪ³ was essential for RvD1 and IL-4-induced microglia alternative activation. These findings suggested that RvD1 enhanced IL-4-induced microglia alternative activation depending on the STAT6/PPARĪ³ signaling pathway (FigureĀ 6).

Figure 6
figure 6

Schematic representation of the RvD1 contribution to microglia alternative activation. RvD1 promotes IL-4-induced microglia alternative activation by increasing Arg1 and Ym1 expression via STAT6/PPARĪ³ signaling pathway.

Conclusions

Our results show that RvD1 promotes IL-4-induced microglia alternative activation. A possible mechanism relates to activation of STAT6 and PPARĪ³ signaling pathways. Given the anti-inflammatory and pro-resolving effects of RvD1, it may be a potential treatment for CNS diseases involving neuroinflammation.

Abbreviations

ALX:

lipoxin A4 receptor

Arg1:

arginase 1

Boc-2:

butyloxycarbonyl-Phe-Leu-Phe-Leu-Phe

BSA:

bovine serum albumin

CNS:

central nervous system

DAPI:

6-diamidino-2-phenylindole

DHA:

docosahexaenoic acid

DMEM:

Dulbeccoā€™s modified Eagleā€™s medium

EMSA:

electrophoretic Mobility Shift Assay

EPA:

eicosapentaenoic acid

Fizz1:

found in inflammatory zone 1

FPR2:

formyl peptide receptor 2

GPR 32:

G protein coupling receptor 32

IL:

interleukin

LPS:

lipopolysaccharide

NO:

nitric oxide

PBS:

phosphate-buffered saline

RvD1:

resolvin D1

PPARĪ³:

peroxisome proliferator-activated receptor gamma

RIPA:

radioimmune precipitation assay buffer

ROS:

reactive oxygen species

STAT6:

signal transducer and activators of transcription 6

TBE:

Tris-borate-EDTA

TBST:

Tris-buffered saline containing 0.05% Tween-20

TNF:

tumour necrosis factor

Ym1:

chitinase 3-like 3.

References

  1. Nimmerjahn A, Kirchhoff F, Helmchen F: Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo. Science. 2005, 308: 1314-1318. 10.1126/science.1110647.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  2. Nelson PT, Soma LA, Lavi E: Microglia in diseases of the central nervous system. Ann Med. 2002, 34: 491-500. 10.1080/078538902321117698.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  3. Mosser DM: The many faces of macrophage activation. J Leukoc Biol. 2003, 73: 209-212. 10.1189/jlb.0602325.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  4. Hu X, Li P, Guo Y, Wang H, Leak RK, Chen S, Gao Y, Chen J: Microglia/macrophage polarization dynamics reveal novel mechanism of injury expansion after focal cerebral ischemia. Stroke. 2012, 43: 3063-3070. 10.1161/STROKEAHA.112.659656.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  5. Lee DC, Ruiz CR, Lebson L, Selenica ML, Rizer J, Hunt JB, Rojiani R, Reid P, Kammath S, Nash K, Dickey CA, Gordon M, Morgan D: Aging enhances classical activation but mitigates alternative activation in the central nervous system. Neurobiol Aging. 2013, 34: 1610-1620. 10.1016/j.neurobiolaging.2012.12.014.

    ArticleĀ  PubMed CentralĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  6. Boche D, Perry VH, Nicoll JA: Review: activation patterns of microglia and their identification in the human brain. Neuropathol Appl Neurobiol. 2013, 39: 3-18. 10.1111/nan.12011.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  7. Varnum MM, Ikezu T: The classification of microglial activation phenotypes on neurodegeneration and regeneration in Alzheimerā€™s disease brain. Arch Immunol Ther Exp (Warsz). 2012, 60: 251-266. 10.1007/s00005-012-0181-2.

    ArticleĀ  CASĀ  Google ScholarĀ 

  8. Gordon S, Martinez FO: Alternative activation of macrophages: mechanism and functions. Immunity. 2010, 32: 593-604. 10.1016/j.immuni.2010.05.007.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  9. Goenka S, Kaplan MH: Transcriptional regulation by STAT6. Immunol Res. 2011, 50: 87-96. 10.1007/s12026-011-8205-2.

    ArticleĀ  PubMed CentralĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  10. Mandal P, Pratt BT, Barnes M, McMullen MR, Nagy LE: Molecular mechanism for adiponectin-dependent M2 macrophage polarization: link between the metabolic and innate immune activity of full-length adiponectin. J Biol Chem. 2011, 286: 13460-13469. 10.1074/jbc.M110.204644.

    ArticleĀ  PubMed CentralĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  11. Szanto A, Balint BL, Nagy ZS, Barta E, Dezso B, Pap A, Szeles L, Poliska S, Oros M, Evans RM, Barak Y, Schwabe J, Nagy L: STAT6 transcription factor is a facilitator of the nuclear receptor PPARgamma-regulated gene expression in macrophages and dendritic cells. Immunity. 2010, 33: 699-712. 10.1016/j.immuni.2010.11.009.

    ArticleĀ  PubMed CentralĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  12. Odegaard JI, Ricardo-Gonzalez RR, Goforth MH, Morel CR, Subramanian V, Mukundan L, Red Eagle A, Vats D, Brombacher F, Ferrante AW, Chawla A: Macrophage-specific PPARgamma controls alternative activation and improves insulin resistance. Nature. 2007, 447: 1116-1120. 10.1038/nature05894.

    ArticleĀ  PubMed CentralĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  13. Jiang Q, Heneka M, Landreth GE: The role of peroxisome proliferator-activated receptor-gamma (PPARgamma) in Alzheimerā€™s disease: therapeutic implications. CNS Drugs. 2008, 22: 1-14. 10.2165/00023210-200822010-00001.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  14. Mandrekar-Colucci S, Karlo JC, Landreth GE: Mechanisms underlying the rapid peroxisome proliferator-activated receptor-gamma-mediated amyloid clearance and reversal of cognitive deficits in a murine model of Alzheimerā€™s disease. J Neurosci. 2012, 32: 10117-10128. 10.1523/JNEUROSCI.5268-11.2012.

    ArticleĀ  PubMed CentralĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  15. Seki H, Sasaki T, Ueda T, Arita M: Resolvins as regulators of the immune system. ScientificWorldJournal. 2010, 10: 818-831.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  16. Hong S, Gronert K, Devchand PR, Moussignac RL, Serhan CN: Novel docosatrienes and 17S-resolvins generated from docosahexaenoic acid in murine brain, human blood, and glial cells. Autacoids in anti-inflammation. J Biol Chem. 2003, 278: 14677-14687. 10.1074/jbc.M300218200.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  17. Rogerio AP, Haworth O, Croze R, Oh SF, Uddin M, Carlo T, Pfeffer MA, Priluck R, Serhan CN, Levy BD: Resolvin D1 and aspirin-triggered resolvin D1 promote resolution of allergic airways responses. J Immunol. 2012, 189: 1983-1991. 10.4049/jimmunol.1101665.

    ArticleĀ  PubMed CentralĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  18. Krishnamoorthy S, Recchiuti A, Chiang N, Yacoubian S, Lee CH, Yang R, Petasis NA, Serhan CN: Resolvin D1 binds human phagocytes with evidence for proresolving receptors. Proc Natl Acad Sci U S A. 2010, 107: 1660-1665. 10.1073/pnas.0907342107.

    ArticleĀ  PubMed CentralĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  19. Krishnamoorthy S, Recchiuti A, Chiang N, Fredman G, Serhan CN: Resolvin D1 receptor stereoselectivity and regulation of inflammation and proresolving microRNAs. Am J Pathol. 2018ā€“2027, 2012: 180.

    Google ScholarĀ 

  20. Titos E, Rius B, Gonzalez-Periz A, Lopez-Vicario C, Moran-Salvador E, Martinez-Clemente M, Arroyo V, Claria J: Resolvin D1 and its precursor docosahexaenoic acid promote resolution of adipose tissue inflammation by eliciting macrophage polarization toward an M2-like phenotype. J Immunol. 2011, 187: 5408-5418. 10.4049/jimmunol.1100225.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  21. Wang YP, Wu Y, Li LY, Zheng J, Liu RG, Zhou JP, Yuan SY, Shang Y, Yao SL: Aspirin-triggered lipoxin A4 attenuates LPS-induced pro-inflammatory responses by inhibiting activation of NF-kappaB and MAPKs in BV-2 microglial cells. J Neuroinflammation. 2011, 8: 95-10.1186/1742-2094-8-95.

    ArticleĀ  PubMed CentralĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  22. Wu Y, Zhai H, Wang Y, Li L, Wu J, Wang F, Sun S, Yao S, Shang Y: Aspirin-triggered lipoxin A(4) attenuates lipopolysaccharide-induced intracellular ROS in BV2 microglia cells by inhibiting the function of NADPH oxidase. Neurochem Res. 2012, 37: 1690-1696. 10.1007/s11064-012-0776-3.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  23. Ye XH, Wu Y, Guo PP, Wang J, Yuan SY, Shang Y, Yao SL: Lipoxin A4 analogue protects brain and reduces inflammation in a rat model of focal cerebral ischemia reperfusion. Brain Res. 2010, 1323: 174-183.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  24. Pourcet B, Pineda-Torra I: Transcriptional regulation of macrophage arginase 1 expression and its role in atherosclerosis. Trends Cardiovasc Med. 2013, 23: 143-152. 10.1016/j.tcm.2012.10.003.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  25. Welch JS, Escoubet-Lozach L, Sykes DB, Liddiard K, Greaves DR, Glass CK: TH2 cytokines and allergic challenge induce Ym1 expression in macrophages by a STAT6-dependent mechanism. J Biol Chem. 2002, 277: 42821-42829. 10.1074/jbc.M205873200.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  26. Gallardo-Soler A, Gomez-Nieto C, Campo ML, Marathe C, Tontonoz P, Castrillo A, Corraliza I: Arginase I induction by modified lipoproteins in macrophages: a peroxisome proliferator-activated receptor-gamma/delta-mediated effect that links lipid metabolism and immunity. Mol Endocrinol. 2008, 22: 1394-1402. 10.1210/me.2007-0525.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  27. Vats D, Mukundan L, Odegaard JI, Zhang L, Smith KL, Morel CR, Wagner RA, Greaves DR, Murray PJ, Chawla A: Oxidative metabolism and PGC-1beta attenuate macrophage-mediated inflammation. Cell Metab. 2006, 4: 13-24. 10.1016/j.cmet.2006.05.011.

    ArticleĀ  PubMed CentralĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  28. Siemasko K, Chong AS, Jack HM, Gong H, Williams JW, Finnegan A: Inhibition of JAK3 and STAT6 tyrosine phosphorylation by the immunosuppressive drug leflunomide leads to a block in IgG1 production. J Immunol. 1998, 160: 1581-1588.

    CASĀ  PubMedĀ  Google ScholarĀ 

  29. Norling LV, Dalli J, Flower RJ, Serhan CN, Perretti M: Resolvin D1 limits polymorphonuclear leukocyte recruitment to inflammatory loci: receptor-dependent actions. Arterioscler Thromb Vasc Biol. 1970ā€“1978, 2012: 32.

    Google ScholarĀ 

  30. Ye RD, Boulay F, Wang JM, Dahlgren C, Gerard C, Parmentier M, Serhan CN, Murphy PM: International Union of Basic and Clinical Pharmacology. LXXIII. Nomenclature for the formyl peptide receptor (FPR) family. Pharmacol Rev. 2009, 61: 119-161. 10.1124/pr.109.001578.

    ArticleĀ  PubMed CentralĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  31. Odusanwo O, Chinthamani S, McCall A, Duffey ME, Baker OJ: Resolvin D1 prevents TNF-alpha-mediated disruption of salivary epithelial formation. Am J Physiol Cell Physiol. 2012, 302: C1331-C1345. 10.1152/ajpcell.00207.2011.

    ArticleĀ  PubMed CentralĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  32. Wu Y, Wang YP, Guo P, Ye XH, Wang J, Yuan SY, Yao SL, Shang Y: A lipoxin A4 analog ameliorates bloodā€“brain barrier dysfunction and reduces MMP-9 expression in a rat model of focal cerebral ischemia-reperfusion injury. J Mol Neurosci. 2012, 46: 483-491. 10.1007/s12031-011-9620-5.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  33. Wu Y, Ye XH, Guo PP, Xu SP, Wang J, Yuan SY, Yao SL, Shang Y: Neuroprotective effect of lipoxin A4 methyl ester in a rat model of permanent focal cerebral ischemia. J Mol Neurosci. 2010, 42: 226-234. 10.1007/s12031-010-9355-8.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  34. Cattaneo F, Parisi M, Ammendola R: Distinct signaling cascades elicited by different formyl Peptide receptor 2 (FPR2) agonists. Int J Mol Sci. 2013, 14: 7193-7230. 10.3390/ijms14047193.

    ArticleĀ  PubMed CentralĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  35. Gong J, Guo S, Li HB, Yuan SY, Shang Y, Yao SL: BML-111, a lipoxin receptor agonist, protects haemorrhagic shock-induced acute lung injury in rats. Resuscitation. 2012, 83: 907-912. 10.1016/j.resuscitation.2011.12.035.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  36. Li HB, Wang GZ, Gong J, Wu ZY, Guo S, Li B, Liu M, Ji YD, Tang M, Yuan SY, Yao SL: BML-111 attenuates hemorrhagic shock-induced acute lung injury through inhibiting activation of mitogen-activated protein kinase pathway in rats. J Surg Res. 2013, 183: 710-719. 10.1016/j.jss.2013.03.007.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  37. Shang Y, Jiang YX, Ding ZJ, Shen AL, Xu SP, Yuan SY, Yao SL: Valproic acid attenuates the multiple-organ dysfunction in a rat model of septic shock. Chin Med J (Engl). 2010, 123: 2682-2687.

    CASĀ  Google ScholarĀ 

  38. Pauleau AL, Rutschman R, Lang R, Pernis A, Watowich SS, Murray PJ: Enhancer-mediated control of macrophage-specific arginase I expression. J Immunol. 2004, 172: 7565-7573.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  39. Morris SM: Enzymes of arginine metabolism. J Nutr. 2004, 134: 2743S-2747S. discussion 2765S-2767S

    CASĀ  PubMedĀ  Google ScholarĀ 

  40. Iniesta V, Gomez-Nieto LC, Molano I, Mohedano A, Carcelen J, Miron C, Alonso C, Corraliza I: Arginase I induction in macrophages, triggered by Th2-type cytokines, supports the growth of intracellular Leishmania parasites. Parasite Immunol. 2002, 24: 113-118. 10.1046/j.1365-3024.2002.00444.x.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  41. Ma TC, Campana A, Lange PS, Lee HH, Banerjee K, Bryson JB, Mahishi L, Alam S, Giger RJ, Barnes S, Morris SM, Willis DE, Twiss JL, Filbin MT, Ratan RR: A large-scale chemical screen for regulators of the arginase 1 promoter identifies the soy isoflavone daidzeinas a clinically approved small molecule that can promote neuronal protection or regeneration via a cAMP-independent pathway. J Neurosci. 2010, 30: 739-748. 10.1523/JNEUROSCI.5266-09.2010.

    ArticleĀ  PubMed CentralĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  42. Colton CA, Mott RT, Sharpe H, Xu Q, Van Nostrand WE, Vitek MP: Expression profiles for macrophage alternative activation genes in AD and in mouse models of AD. J Neuroinflammation. 2006, 3: 27-10.1186/1742-2094-3-27.

    ArticleĀ  PubMed CentralĀ  PubMedĀ  Google ScholarĀ 

  43. Giannetti N, Moyse E, Ducray A, Bondier JR, Jourdan F, Propper A, Kastner A: Accumulation of Ym1/2 protein in the mouse olfactory epithelium during regeneration and aging. Neuroscience. 2004, 123: 907-917. 10.1016/j.neuroscience.2003.09.024.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  44. Zhou X, Spittau B, Krieglstein K: TGFbeta signalling plays an important role in IL4-induced alternative activation of microglia. J Neuroinflammation. 2012, 9: 210-10.1186/1742-2094-9-210.

    ArticleĀ  PubMed CentralĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  45. Ishii M, Wen H, Corsa CA, Liu T, Coelho AL, Allen RM, Carson WF, Cavassani KA, Li X, Lukacs NW, Hogaboam CM, Dou Y, Kunkel SL: Epigenetic regulation of the alternatively activated macrophage phenotype. Blood. 2009, 114: 3244-3254. 10.1182/blood-2009-04-217620.

    ArticleĀ  PubMed CentralĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  46. Cuartero MI, Ballesteros I, Moraga A, Nombela F, Vivancos J, Hamilton JA, Corbi AL, Lizasoain I, Moro MA: N2 neutrophils, novel players in brain inflammation after stroke: modulation by the PPARgamma agonist rosiglitazone. Stroke. 2013, 44: 3498-3508. 10.1161/STROKEAHA.113.002470.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  47. Bouhlel MA, Derudas B, Rigamonti E, Dievart R, Brozek J, Haulon S, Zawadzki C, Jude B, Torpier G, Marx N, Staels B, Chinetti-Gbaguidi G: PPARgamma activation primes human monocytes into alternative M2 macrophages with anti-inflammatory properties. Cell Metab. 2007, 6: 137-143. 10.1016/j.cmet.2007.06.010.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

Download references

Acknowledgements

This study was supported by the grants from the National Natural Science Foundation of China (81271270, 81270018 and 81200909) and Science and Technology Bureau of Wuhan (2013060501010155).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to You Shang.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authorsā€™ contributions

LYL and YW designed the experiments. LYL, YW, WJX and LMS performed the experiments. LYL and YW analyzed the data. YPW, JW and LP provided useful advice and reviewed the manuscript. SYY and YS supervised the experimental work. YS conceived the study, participated in its design and coordination, and wrote the manuscript. All authors of this paper have read and approved the final version the manuscript.

Longyan Li, Yan Wu contributed equally to this work.

Authorsā€™ original submitted files for images

Rights and permissions

Open Access Ā This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made.

The images or other third party material in this article are included in the articleā€™s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the articleā€™s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.

To view a copy of this licence, visit https://creativecommons.org/licenses/by/4.0/.

The Creative Commons Public Domain Dedication waiver (https://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Cite this article

Li, L., Wu, Y., Wang, Y. et al. Resolvin D1 promotes the interleukin-4-induced alternative activation in BV-2 microglial cells. J Neuroinflammation 11, 72 (2014). https://doi.org/10.1186/1742-2094-11-72

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1742-2094-11-72

Keywords