Skip to main content

Sphingosine 1-phosphate receptor subtype 3 (S1P3) contributes to brain injury after transient focal cerebral ischemia via modulating microglial activation and their M1 polarization

Abstract

Background

The pathogenic roles of receptor-mediated sphingosine 1-phosphate (S1P) signaling in cerebral ischemia have been evidenced mainly through the efficacy of FTY720 that binds non-selectively to four of the five S1P receptors (S1P1,3,4,5). Recently, S1P1 and S1P2 were identified as specific receptor subtypes that contribute to brain injury in cerebral ischemia; however, the possible involvement of other S1P receptors remains unknown. S1P3 can be the candidate because of its upregulation in the ischemic brain, which was addressed in this study, along with underlying pathogenic mechanisms.

Methods

We used transient middle cerebral artery occlusion/reperfusion (tMCAO), a mouse model of transient focal cerebral ischemia. To identify S1P3 as a pathogenic factor in cerebral ischemia, we employed a specific S1P3 antagonist, CAY10444. Brain damages were assessed by brain infarction, neurological score, and neurodegeneration. Histological assessment was carried out to determine microglial activation, morphological transformation, and proliferation. M1/M2 polarization and relevant signaling pathways were determined by biochemical and immunohistochemical analysis.

Results

Inhibiting S1P3 immediately after reperfusion with CAY10444 significantly reduced tMCAO-induced brain infarction, neurological deficit, and neurodegeneration. When S1P3 activity was inhibited, the number of activated microglia was markedly decreased in both the periischemic and ischemic core regions in the ischemic brain 1 and 3 days following tMCAO. Moreover, inhibiting S1P3 significantly restored the microglial shape from amoeboid to ramified microglia in the ischemic core region 3 days after tMCAO, and it attenuated microglial proliferation in the ischemic brain. In addition to these changes, S1P3 signaling influenced the proinflammatory M1 polarization, but not M2. The S1P3-dependent regulation of M1 polarization was clearly shown in activated microglia, which was affirmed by determining the in vivo activation of microglial NF-κB signaling that is responsible for M1 and in vitro expression levels of proinflammatory cytokines in activated microglia. As downstream effector pathways in an ischemic brain, S1P3 influenced phosphorylation of ERK1/2, p38 MAPK, and Akt.

Conclusions

This study identified S1P3 as a pathogenic mediator in an ischemic brain along with underlying mechanisms, involving its modulation of microglial activation and M1 polarization, further suggesting that S1P3 can be a therapeutic target for cerebral ischemia.

Background

Sphingosine 1 phosphate (S1P), which is a bioactive sphingolipid, has been known to influence a variety of biological actions throughout the body [1]. These actions of S1P in various organs are mostly mediated by its five specific G-protein coupled receptors (S1P1–5) [1]. Based on the identified biological actions of S1P, a considerable effort has been made to develop a drug that targets S1P receptors, leading to the first successful output, FTY720 (fingolimod, Gilenya, Novartis), that binds non-selectively to 4 of the 5 S1P receptors after being phosphorylated [1] and is currently used for treatment of multiple sclerosis [2]. In addition to this success, FTY720 is now under clinical trials for the treatment of several disease types, including acute stroke, amyotrophic lateral sclerosis, schizophrenia, Rett syndrome, and glioblastoma [2], strongly suggesting that receptor-mediated S1P signaling can be a considerable drug target in different diseases. However, the S1P receptor subtypes involved in each disease type is still unclear. Even the efficacy of FTY720 has been assumed to be primarily mediated by S1P1, and no other subtypes targeted by FTY720 have been identified that mediate its efficacy.

Cerebral ischemia, which is caused by a sudden interruption of blood flow to the brain, is a disease type where S1P receptors become validated drug targets mainly due to the efficacy of FTY720. Numerous in vivo studies have been conducted to prove the neuroprotective effects of FTY720 in the brain against ischemic challenge [3,4,5,6,7,8,9]. FTY720 itself [10, 11] or combined with a thrombolytic agent [12] is under clinical trials for the treatment in acute stroke. Despite this validated efficacy, among the four S1P receptor subtypes targeted by FTY720, S1P1 is the only identified receptor subtype to be associated with cerebral ischemia [13], indicating the possible involvement of other subtypes of FTY720-relevant S1P receptors. Besides S1P1, S1P2 (which is not a target for FTY720) was also revealed to influence brain injury after ischemic challenge [14]. These two independent studies identified the importance of receptor-mediated S1P signaling in cerebral ischemia and further demonstrated the pathogenic roles of both receptor subtypes in this disease. Interestingly, the pathogenic roles of S1P1 in cerebral ischemia [13] demonstrated that FTY720’s efficacy in this disease is via its unique action as a functional antagonist for S1P1 [15, 16]. In addition to S1P1, FTY720-phosphate may also antagonize S1P3 because it reduced cellular responses through S1P-S1P3 signaling axis [17]. Furthermore, S1P3 was reported to be upregulated at mRNA levels in the brain after ischemic challenge [6]. This notion raised the possibility that S1P3 could be an additional pathogenic factor for cerebral ischemia, and FTY720’s efficacy in cerebral ischemia can also be mediated via suppressing S1P3. However, whether S1P3 influences brain injury in focal cerebral ischemia and the role of S1P3, pathogenic or neuroprotective, has not been identified.

In this study, we aimed to address the pathogenic role of S1P3 in transient focal cerebral ischemia with a mouse model of transient middle cerebral artery occlusion and reperfusion (tMCAO). To identify the role, we used a selective S1P3 antagonist, CAY10444, that was given to mice immediately after reperfusion. We then assessed brain damage such as brain infarction, neurological functional deficit, and neural cell death. We further assessed whether S1P3 influenced microglial activation and polarization, a core pathogenic event in cerebral ischemia, along with a clarification of S1P3-dependent effector pathways in the brain after tMCAO challenge.

Methods

Animals and surgical procedures

Male ICR mice (32 ± 2 g; 6 weeks old) were bought from the Orient Bio company (Korea) and housed under controlled environmental conditions of diurnal lighting (light on 07:00–19:00), temperature (22 ± 2 Â°C), and relative humidity (60 ± 10%). All animal handling and surgical procedures were carried out in accordance with the approved animal protocols specified by the Institutional Animal Care and Use Committee at Gachon University (Incheon, Republic of Korea) (no. of approved animal protocols: LCDI-2015–0048; LCDI-2014–0079). Following 1 week of laboratory acclimatization, the mice were challenged with tMCAO as described previously [18]. In brief, the mice were anesthetized with isoflurane (3% for induction and 1.5% for maintenance of anesthesia) in a N2O∶O2 (3∶1) mixture, and the right common carotid artery was isolated through a ventral neck incision. A silicone-coated 5–0 monofilament was introduced to the internal carotid artery from carotid bifurcation and advanced to occlude the middle cerebral artery (MCA). After 90 min of MCAO, the filament was withdrawn to allow complete reperfusion of the cerebral area. During surgery, rectal temperature was maintained at 37.0 ± 0.5 °C with a homoeothermic blanket. Sham-operated mice received similar surgical procedure except for the occlusion of MCA. After surgery, three mice were kept in a single cage; wet food and soft bedding were provided to minimize the suffering from the operation until they were sacrificed for brain sampling.

CAY10444 administration

CAY10444 (Cayman chemical, MI, USA) was dissolved in 1:1 mixture of chremophore EL and 100% ethanol, diluted in water, and injected intraperitoneally to mice at 0.1, 0.2, and 0.5 mg/kg at the time of reperfusion. For the tMCAO group, equal volumes of the vehicle were injected.

Neurological function assessment and brain infarction determination

Functional neurological deficit was assessed using modified neurological severity score (mNSS) scale to determine the motor, sensory, balance, and reflex disorder 24 h following MCAO, as described previously [18,19,20]. Following the neurological score assessment, the mice were sacrificed with CO2 exposure; their brains were quickly removed and sliced in the mice brain matrix at 2 mm thickness. The obtained brain slices were incubated with 2% 2,3,5-triphenyltetrazolium chloride (TTC) in physiological saline for 20 min at 37 Â°C. The TTC-stained brain slices were photographed, and the infarct area was calculated using ImageJ software (National Institute of Mental Health, Bethesda, MD).

Histological analysis

Tissue preparation

Brain tissue samples for histological analysis were obtained 1 or 3 days after tMCAO. Mice were anesthetized with a mixture of Zoletil 50® (10 mg/kg, i.m.) and Rompun® (3 mg/kg, i.m.), and their brains were perfused with ice-cold phosphate-buffered saline (PBS; pH 7.4) followed by 4% paraformaldehyde. The brains were incubated in the same fixative solution overnight, cryoprotected with 30% sucrose, and cut into 20-μm sections using a microtome cryostat. To ensure anatomical similarity of brain regions, two coronal brain sections obtained from the rostral to middle portion of the striatum and the cortex of each mouse brain were used for histological evaluation. In a different set of experiments, the mice brains were transcardially washed with ice-cold PBS and the ipsilateral brain hemisphere was used for RNA and protein extraction.

Fluoro Jade B staining

In order to identify any degenerating neurons following the tMCAO challenge, Fluoro Jade B (FJB) histochemical staining was performed 1 day after tMCAO induction. Brain sections were sequentially immersed in ethanol series (100% for 3 min, and 70% and 30% for 1 min each), rinsed in deionized water, and oxidized in 0.06% w/v KMnO4. Then, sections were stained with 0.001% (w/v) FJB in 0.1% (v/v) acetic acid solution for 30 min, rinsed in deionized water, dried in a slide warmer, cleared in xylene, and then cover-slipped.

Iba1 or glial fibrillary acidic protein (GFAP) immunohistochemistry

To evaluate the effect of S1P3 activity on microglia or astrocyte activation, Iba1 or GFAP immunohistochemistry was performed 1 or 3 days after tMCAO. Brain sections were oxidized with 1% H2O2 in PBS for 15 min and blocked with 1% fetal bovine serum (FBS) in 0.3% Triton-X100 in PBS for 1 h to block non-specific protein binding. Then, the brain sections were incubated with primary antibody against Iba1 (1:500, Wako) or GFAP (1: 500, Invitrogen) overnight at 4 Â°C followed by anti-rabbit secondary antibody (1:200). Sections were exposed to avidin and biotinylated horse-radish peroxidase macromolecular complex (ABC) kit (1:100, Vector Labs) and visualized with 3, 3′-diaminobenzidine tetrahydrochloride (DAB) exposure (0.02% DAB and 0.01% H2O2 in 0.05 M TRIS solution), dehydrated with ethanol, cleared in xylene, and mounted using mounting media.

Iba1/NF-κB double-immunohistochemistry

In order to determine whether the NF-κB pathway is triggered in activated microglia after the tMCAO challenge, cryostat brain sections were processed for double immunolabeling using antibodies against NF-κB (p65) and Iba1. The sections were incubated with TRIS-EDTA solution at 100 Â°C for 30 min for antigen retrieval, blocked with 1% FBS in 0.3% Triton X-100, and labeled with rabbit NF-κB (p65) (1:100) antibody overnight at 4 Â°C. The sections were labeled with a biotinylated secondary antibody (1:200) followed by incubation with an ABC kit. The signals were visualized with DAB staining (0.02% DAB and 0.01% H2O2 for 2 min). The stained sections were then washed with PBS (3 × 5 min), blocked, and incubated with primary antibodies against Iba1 (1:500) overnight at 4 Â°C. Sections were then labeled with appropriate secondary antibodies conjugated with Cy3 (1:1000) and mounted with VECTA SHIELD mounting medium.

Bromodeoxyuridine (BrdU)/Iba1 immunofluorescence

The role of S1P3 activity on tMCAO-induced microglia proliferation was determined using Iba1/BrdU double immunofluorescence. BrdU (50 mg/kg in PBS, i.p.) was administered twice a day at 12-h intervals on the second and third day after tMCAO challenge. Brain sections were prepared for Iba1/BrdU immunofluorescence as described previously [18, 21].

Image preparation and quantification

The brain sections after staining or immunolabeling were photographed using bright-field and fluorescence microscopy (BX53T, Olympus, Japan) equipped with a DP72 camera. Representative images were prepared using Adobe Photoshop CS3. For quantification, three photographs were taken from different area of each region and the number of immunopositive cells was counted. Then, the average number of immunopositive cells from each region was expressed in per unit area (mm2).

Western blot analysis

Ipsilateral brain hemispheres were obtained 24 h following tMCAO induction and triturated with neuronal protein extraction reagent (NPER); the obtained proteins was thus separated in a 10% SDS-PAGE system and transferred to the polyvinylidene difluoride membrane. The membrane was blocked with 5% skim milk to avoid non-specific protein bindings and incubated with primary antibodies against rabbit pAkt, Akt, pERK1/2, ERK1/2, pp38, p38 (Cell signaling, all at 1:1000 dilution), and mouse β-actin (Sigma Aldrich, 1:5000) overnight at 4 Â°C followed by incubation with respective secondary antibodies (Jackson ImmunoResearch, 1:10000) for 2 h at room temperature and visualized with enhanced chemiluminescence (ECL) solution. The band intensity of each protein was analyzed using ImageQuant (TM) TL software, normalized with β-actin, and then expressed as fold changes of the sham-operated group.

Mouse primary microglia culture, CAY10444 treatment, and transfection with S1P3 shRNA

Primary microglial cells were obtained from the brain cortices of 1–2-day-old mouse pups as described previously [13]. The microglial cells were seeded on 6-well plates at a density of 1 × 105 cells/well. Microglial cells were starved overnight and treated with CAY10444 (1 Î¼M) or vehicle (0.1% DMSO in DMEM). Thirty minutes later, microglia were stimulated with lipopolysaccharides (LPS) (100 ng/ml) for additional 24 h. Alternatively, the shRNA targeted with S1P3 (shS1P3) receptor or non-targeted control shRNA was transfected into the cells in serum and antibiotic-free medium. After 6 h of incubation, the media were replaced with serum and antibiotic containing media for an additional 42 h. S1P3-infected microglial cells were then challenged with serum starvation for 12 h and stimulated with LPS, and then harvested for qRT-PCR analysis.

Quantitative real-time polymerase chain reaction (qRT-PCR)

Total RNA was extracted from the ipsilateral hemisphere of mice brain and cultured microglia cells using TRIzol Reagent (Invitrogen). One microgram of total RNA was reverse transcribed (RT) to synthesize cDNA. The gene expression levels of the different markers of M1- and M2-polarized microglia were determined using the StepOnePlus™ qRT-PCR system (Applied Biosystems) with the FG Power SYBR Green PCR master mix (Life Technologies) and primer sets (Additional file 1: Table S1). β-actin was used as the housekeeping gene.

Statistical analysis

All statistical tests were performed using Graph Pad Prism 5 (Graph Pad Software Inc., La Jolla, CA, USA), and the data are presented as mean ± S.E.M. One-way ANOVA followed by the Newman-Keuls post hoc test was used to compare the data among the multiple experimental groups, while comparisons between the two groups were performed using the Student’s t test. p < 0.05 was set as statistically significant.

Results

Suppression of S1P3 activity attenuates brain infarction and neurological deficit in tMCAO-challenged mice

The vehicle-administered mice developed severe brain infarction in both the ischemic cortex and striatum 24 h after the tMCAO challenge (Fig. 1a, b). However, the mice administered with S1P3 antagonist (CAY10444) showed significantly decreased brain infarction in a dose-dependent manner (Fig. 1a, b). The brain infarction volume of the vehicle-administered tMCAO group was 31.20 ± 1.65%, whereas that in the different dosages of CAY10444-administered mice were 28.63 ± 0.97%, 25.20 ± 1.15%, and 21.96 ± 1.68% at 0.1, 0.2, and 0.5 mg/kg, respectively (Fig. 1a, b). The lowest dose of CAY10444 (0.1 mg/kg) was not effective, but 0.2 and 0.5 mg/kg were effective to attenuate the brain infarction. Similarly, the neurological deficit parameters reflecting motor, sensory, reflex, and balance disorders, as evidenced by mNSS analysis, were significantly improved in the CAY10444-administered mice compared to the vehicle-administered group (Fig. 1c). Among the tested doses, 0.5 mg/kg was found to be the most effective to attenuate brain infarction and neurological deficit; this dose was therefore chosen for the remaining experiments. The neuroprotective potential of CAY10444 in tMCAO-induced brain damage was further affirmed by analyzing the extent of neurodegeneration 24 h following ischemic challenge using FJB staining. CAY10444 reduced the extent of neuronal damage compared with vehicle exposure (Additional file 1: Figure S1). These results clearly demonstrated that the suppression of S1P3 activity attenuated tMCAO-induced brain damage, indicating the pathogenic role of S1P3 in cerebral ischemia.

Fig. 1
figure 1

CAY10444 (CAY) administration attenuates tMCAO-induced brain infarction and neurological deficit. Mice were challenged with tMCAO, and CAY (0.1. 0.2, and 0.5 mg/kg) was administered intraperitoneally immediately after reperfusion. Brain damage was ascertained 24 h after tMCAO challenge (a–c). Effects of different dosage of CAY on infarct volume (a, b) and neurological function (c) were determined. Representative images of TTC-stained brain tissue (a), quantification of brain infarction (b), and neurological deficit (c) are shown. n = 10 ~ 12 mice per group. **p < 0.01 and ***p < 0.001 versus vehicle-administered tMCAO group

Suppression of S1P3 activity attenuates microglial activation and proliferation in the brain of tMCAO-challenged mouse

Focal cerebral ischemia-induced microglial activation was analyzed in the brain through Iba1 immunohistochemistry 1 and 3 days following tMCAO challenge. The vehicle-administered tMCAO group showed the robust activation of microglia, as demonstrated by an increased number of Iba1-immunopositive cells in the ischemic hemisphere at both time points. CAY10444 administration significantly reduced the number of Iba1-immunopositive cells in a time- and region-dependent manner compared with the vehicle administration (Figs. 2 and 3). The number of activated microglia was significantly reduced in both the periischemic and ischemic core regions of the CAY10444-administered mice compared with the vehicle-administered mice at both time points (Figs. 2 and 3). Moreover, the number of amoeboid microglia in the ischemic core region was significantly reduced in the CAY10444-administered mice, as depicted by the reduced ratio of amoeboid/ramified microglia (Fig. 3c). These data demonstrated that suppressing S1P3 activity in an ischemic brain not only attenuated the activation of microglia, but also reduced the morphological transformation of ramified microglia to amoeboid microglia.

Fig. 2
figure 2

CAY10444 (CAY) administration attenuates tMCAO-induced microglia activation in 1 day post-ischemic brain. Mice were challenged with tMCAO, and CAY (0.5 mg/kg) was administered intraperitoneally immediately after reperfusion. The effect of CAY on microglial activation was determined by Iba1 immunohistochemistry in 1 day post-ischemic brain. a Representative images of Iba1-immunopositive cells in periischemic (P) and ischemic core (C) regions. Scale bars, 200 Î¼m (top panels) and 50 Î¼m (middle and bottom panels). b Quantification of the number of Iba1-immunopositive cells in both regions. n = 4 ~ 5 mice per group. ***p < 0.001 versus sham. ##p < 0.01 and ###p < 0.001 versus vehicle-administered tMCAO group (tMCAO+veh)

Fig. 3
figure 3

CAY10444 (CAY) administration attenuates tMCAO-induced microglia activation in 3 days post-ischemic brain. Mice were challenged with tMCAO, and CAY (0.5 mg/kg) was administered intraperitoneally immediately after reperfusion. The effect of CAY on microglial activation was determined by Iba1 immunohistochemistry in 3 days post-ischemic brain. a Representative images of Iba1-immunopositive cells in periischemic (P) and ischemic core (C) regions. Scale bars, 200 Î¼m (top panels) and 50 Î¼m (middle and bottom panels). b Quantification of the number of Iba1-immunopositive cells in both regions. c Quantification of morphological changes of Iba1-positive cells in ischemic core regions (ratio of amoeboid to ramified microglia). n = 5 mice per group. **p < 0.01 and ***p < 0.001 versus sham group. #p < 0.05 and ###p < 0.001 versus vehicle-administered tMCAO group (tMCAO+veh)

The brain resident microglia proliferated during the first week following the ischemic challenge, and these newly born microglia may participate in inflammatory responses [22]. To analyze the regulatory roles of S1P3 on microglial proliferation in the ischemic brain, we performed double immunofluorescence for BrdU and Iba1 in the brain 3 days after the tMCAO challenge. Microglial proliferation was obviously observed in the ischemic penumbra region of the vehicle-administered tMCAO group as evidenced by the increased number of BrdU/Iba1 double-immunopositive cells. The administration of CAY10444 significantly decreased the number of BrdU/Iba1 double-immunopositive cells compared with the vehicle administration (Fig. 4a, b), demonstrating that S1P3 is involved in microglial proliferation following ischemic challenge.

Fig. 4
figure 4

CAY10444 (CAY) administration attenuates tMCAO-induced microglia proliferation in 3 days post-ischemic brain. Mice were challenged with tMCAO, and CAY (0.5 mg/kg) was administered intraperitoneally immediately after reperfusion. The effect of CAY on microglial proliferation was determined by BrdU/Iba1 double immunofluorescence analysis in 3 days post-ischemic brain. a Representative images of BrdU/Iba1 double-immunopositive cells in marginal zone. Scale bars, 50 Î¼m. b Quantification of the number of BrdU/Iba1-immunopositive cells. n = 5 mice per group. ***p < 0.001 versus sham group. ##p < 0.01 versus vehicle-administered tMCAO group (tMCAO+veh)

Besides microglial activation, astrogliosis is another core pathogenesis in cerebral ischemia [23], and S1P3 regulates inflammatory responses in activated astrocytes [24]. In this study, we also determined whether suppressing S1P3 activity reduced astrogliosis following ischemic challenge through GFAP immunohistochemistry. The vehicle-administered mice developed a significant astrogliosis in the corpus callosum as evidenced by the increased number of GFAP-immunopositive cells 1 and 3 days after the tMCAO challenge. CAY10444 administration significantly reduced the number of GFAP-immunopositive cells at both time points. In addition, the morphology of astrocytes was transformed towards reactive phenotype, particularly, 3 days after the tMCAO challenge, which was markedly attenuated by CAY10444 administration (Additional file 1: Figure S3). These results demonstrated that S1P3 signaling also regulated astrogliosis in the ischemic brain.

S1P3 regulates microglial M1 polarization in the brain of tMCAO-challenged mouse

Following ischemic injury, activated microglia become polarized into two distinct phenotypes, broadly known as proinflammatory M1 and anti-inflammatory M2 phenotypes [25]. In order to identify the association between S1P3 activity and M1/M2 polarization in the ischemic brain, the mRNA expression levels of different markers, both surface and soluble, of M1 and M2 polarization were determined. The mRNA expression levels of M1 surface markers (CD11b, CD16, CD32, and CD86) were significantly upregulated 1 and 3 days following the tMCAO challenge (Fig. 5). The upregulated surface markers of M1 polarization in the ischemic brain, such as CD16 and CD32, were significantly downregulated in the CAY10444-administered mice 1 day after the ischemic challenge (Fig. 5a–d). Similarly, CAY10444 administration significantly downregulated the mRNA expression levels of M1 surface markers (CD11b, CD16, and CD32) 3 days after the tMCAO challenge (Fig. 5e–h). We then determined whether S1P3 also regulated the expression of soluble markers that are functionally more important M1 markers. The administration of CAY10444 significantly reduced the mRNA expression levels of the proinflammatory cytokines, such as TNF-α and IL-1β, but not IL-6 (Fig. 6a–c) in the 1-day post-ischemic brain, which were reproduced in the 3-day post-ischemic brain (Fig. 6d–f). These data demonstrated that S1P3 triggered the proinflammatory responses of M1-polarized cells in the ischemic brain. We further determined whether S1P3, in the ischemic brain, had a role in the anti-inflammatory M2 polarization. However, the administration of CAY10444 did not alter the gene expression levels of the M2 markers (Arg1, CCL-22, CD206, TGF-β, and Ym-1) at both day 1 (Additional file 1: Figure S2a–e) and day 3 (Additional file 1: Figure S2f–j) following the ischemic challenge, suggesting that S1P3 in an ischemic brain is mainly associated with M1 polarization rather than M2 polarization.

Fig. 5
figure 5

CAY10444 (CAY) administration attenuates expression level of surface markers of M1 polarization in post-ischemic brain. Mice were challenged with tMCAO, and CAY (0.5 mg/kg) was administered intraperitoneally immediately after reperfusion. The effect of CAY on mRNA expression of M1 surface markers in 1 day (a–d) and 3 days (e–h) post-ischemic brain was determined by qRT-PCR analysis. n = 5 mice per group. *p < 0.05 and **p < 0.01 versus sham group. #p < 0.05 versus vehicle-administered tMCAO group (tMCAO+veh)

Fig. 6
figure 6

CAY10444 (CAY) administration attenuates expression level of soluble markers of M1 polarization in post-ischemic brain. Mice were challenged with tMCAO, and CAY (0.5 mg/kg) was administered intraperitoneally immediately after reperfusion. The effect of CAY on mRNA expression of M1 soluble markers in 1 day (a–d) and 3 days (e–h) post-ischemic brain was determined by qRT-PCR analysis. n = 5 mice per group. *p < 0.05, **p < 0.01, and ***p < 0.001 versus sham group. #p < 0.05, ##p < 0.01, and ###p < 0.001versus vehicle-administered tMCAO group (tMCAO+veh)

The M1 polarization is closely related to NF-κB signaling as the expression of most of the soluble M1 markers are dependent on a transcriptional activation of NF-κB. S1P3 was also found to regulate microglial activation and M1 polarization following ischemic injury in this study. Therefore, we tried to correlate the roles of S1P3 with NF-κB activation, especially in activated microglia, which was addressed by double immunolabeling for NF-κB(p65) and Iba1 1 day after the ischemic challenge. The vehicle-administered tMCAO group showed an enhanced expression of NF-κB(p65) which are easily identified in Iba1-immunopositive cells in the ischemic core region (Fig. 7a, b). CAY10444 administration significantly decreased the number of NF-κB(p65)-immunopositive cells or NF-κB(p65)/Iba1 double-immunopositive cells (Fig. 7a, b). These data further demonstrated that S1P3 in the ischemic brain mediated the M1 polarization through the activation of NF-κB signaling, in particular, in activated microglia. The regulatory role of S1P3 on M1 microglial polarization was reaffirmed using LPS-stimulated mouse primary microglia. For this purpose, we used LPS because LPS is a well-known stimulus to induce M1 polarization of microglia [26, 27]. The mRNA expression levels of M1-soluble markers (TNF-α, IL-6, and IL-1β) were significantly upregulated in LPS-treated cells. Suppressing S1P3 activity either pharmacologically, using CAY10444 (Fig. 8a–c), or genetically, using S1P3-specific shRNA lentivirus (Fig. 8d–g), attenuated the expression of these M1 markers. These data ensured that S1P3 in the ischemic brain might be associated with the inflammatory M1 polarization of activated microglia.

Fig. 7
figure 7

CAY10444 (CAY) administration attenuates tMCAO-induced microglial NF-κB expression in post-ischemic brain. Mice were challenged with tMCAO, and CAY (0.5 mg/kg) was administered intraperitoneally immediately after reperfusion. The effect of CAY on NF-κB expression in activated microglia was determined by NF-κB(p65)/Iba1 double immunohistochemical analysis in 1 day post-ischemic brain. a Representative images of NF-κB(p65)/Iba1-immunopositive cells in ischemic core regions. Scale bars, 50 Î¼m. b Quantification of the number of NF-κB(p65)- and NF-κB(p65)/Iba1-immunopositive cells. n = 4~ 5 mice per group. ***p < 0.001 versus sham group and ###p < 0.001 versus vehicle-administered tMCAO group (tMCAO+veh)

Fig. 8
figure 8

Suppression of S1P3 activity attenuates proinflammatory cytokine expression in LPS-stimulated mouse primary microglial cells. Mouse primary microglial cells were stimulated with LPS (100 ng/ml) 30 min after CAY (1 Î¼M) treatment. Alternatively, cells were stimulated with LPS 2 days after transfection with S1P3 shRNA (shS1P3) or non-target control shRNA (shNC). a–c The mRNA expression levels of TNF-α, IL-6, and IL-1β were determined 24 h after LPS treatment using qRT-PCR analysis. n = 4 per group. **p < 0.01 versus vehicle-treated cells. #p < 0.05, ##p < 0.01, and ###p < 0.001 versus LPS-stimulated cells. d–g Effects of S1P3 knockdown (d) on mRNA expression levels of TNF-α, IL-6, and IL-1β (e–g) were determined 24 h after LPS treatment in S1P3 shRNA and shNC transfected cells through qRT-PCR. n = 3 per group. **p < 0.01 versus shNC in d. *p < 0.05, **p < 0.01, and ***p < 0.001 versus vehicle-treated cells (e–g). #p < 0.05, ##p < 0.01, and ###p < 0.001 versus LPS-stimulated cells

S1P3 activity in ischemic brain was linked with activation of ERK1/2, p38 MAPK, and Akt effector pathways

Microglial activation and their phenotype shift towards M1 polarization are linked to several signaling molecules, including ERK1/2, p38 MAPK, and PI3K/Akt [28,29,30,31]. Additionally, these signaling pathways function as Gi protein-associated effector systems under S1P3 activation [1]. Therefore, we determined whether S1P3 influenced the activation of these signaling components in an ischemic brain 24 h after tMCAO. In the ischemic brain, ERK1/2 and p38 MAPKs were significantly activated, as assessed by Western blotting for their phosphorylated forms (Fig. 9a, b). When S1P3 activity was blocked by CAY10444 administration, the increased phosphorylation of ERK1/2 and p38 MAPKs was significantly attenuated (Fig. 9a, b). Akt phosphorylation was reduced in the ischemic brain, and this reduction was significantly reversed by S1P3 antagonism (Fig. 9a, b), further implying the neurotoxic roles of S1P3 following the ischemic challenge because Akt phosphorylation is a well-known survival factor [32]. These data demonstrated that S1P3 influenced the activation of ERK1/2 and p38 MAPKs as well as the inactivation of Akt as downstream signaling cascades in cerebral ischemia.

Fig. 9
figure 9

CAY10444 (CAY) administration alters tMCAO-induced Akt and MAPK expression in post-ischemic brain. Mice were challenged with tMCAO, and CAY (0.5 mg/kg) was administered intraperitoneally immediately after reperfusion. The effect of CAY on expression in MAPKs and Akt was determined by Western blot analysis. a Representative Western blots. b Quantification. n = 4 mice per group. *p < 0.05, **p < 0.01, and ***p < 0.001 versus sham group, and #p < 0.05 vehicle-treated tMCAO (tMCAO+veh)

Discussion

In the current study, we identified S1P3 as another S1P receptor subtype that triggers pathogenesis in transient focal cerebral ischemia along with mechanistic features, particularly in terms of microglial biology, and the effector signaling pathways after S1P3 activation. Suppression of S1P3 activity after tMCAO by its specific antagonist results in attenuation of brain damages. The pathogenic roles of S1P3 in the ischemic brain are closely associated with microglial activation, involving an increased number of activated microglia, morphological transformation into amoeboid shape, and microglial proliferation. In addition, S1P3 regulates M1 microglial polarization, but not M2 polarization, in the ischemic brain because inhibiting S1P3 after tMCAO weakened the characteristics of M1 polarization without any influence on the M2 markers. These biological roles were further supported in vitro using LPS-stimulated primary microglia. Finally, PI3K/Akt, ERK1/2 MAPK, and p38 MAPK pathways were identified as effector pathways after S1P3 activation in the ischemic brain.

The use of receptor-mediated S1P signaling has been assumed as a possible therapeutic strategy to overcome cerebral ischemia because FTY720, which is a non-selective modulator of 4 of 5 S1P receptors after being phosphorylated, exerts neuroprotective effects in rodent models [3,4,5,6,7,8]. Currently, FTY720 is under clinical trial for the treatment of acute stroke [10, 11], and another trial for acute ischemic stroke is underway to determine its clinical efficacy in combination with a thrombolytic therapy, alteplase [12]. Despite these successful efforts, until recently, which S1P receptor subtypes are actual mediators for FTY720’s efficacy has remained uncertain. Our previous report proposed the first possibility for this, demonstrating S1P1 as a pathogenic factor in focal cerebral ischemia using a mouse model for transient focal cerebral ischemia [13]. The current study identified S1P3 as an additional S1P receptor subtype to mediate brain injury in cerebral ischemia. Notably, it has been discovered that FTY720-phosphate acts as a functional antagonist for S1P1 [15, 16] and possibly for S1P3. Even with no direct evidence for the latter, a few findings indicate that FTY720-phosphate antagonizes S1P3 signaling. Either FTY720-phosphate or TY-52156 (a selective S1P3 antagonist) reduced p-selectin production and leucocyte rolling via S1P-S1P3 signaling axis, which was reaffirmed in S1P3 knockouts [17]. FTY720-phosphate was also reported to antagonize Gq-mediated signaling pathway under S1P3 activation [33]. Considering the inhibitory roles of FTY720-phosphate for S1P1 and S1P3, our previous and current in vivo findings strongly indicate that the reported FTY720’s efficacy in cerebral ischemia may be through suppressing at least the S1P1 and S1P3 activities. Besides FTY720-relevant target receptors, S1P2 was also identified to mediate brain injury in cerebral ischemia through the disruption of vascular integrity in the ischemic brain [14], even though it is not a target for FTY720-phosphate. Therefore, three subtypes of S1P receptors have been identified as pathogenic factors for cerebral ischemia. However, it is still unclear whether the mediation of the brain injury in the cerebral ischemia differs among the receptor subtypes and whether additional S1P receptor subtypes participate, such as S1P4 or S1P5.

Despite the clear pathogenic role of S1P3 in the brain, its roles in ischemic conditions seem to be tissue-specific. In fact, earlier studies reported controversial roles of S1P3 in non-neural ischemic models: protective or harmful. In the heart, the deletion of both S1P2 and S1P3 was shown to aggravate myocardial infarction in mice, which supported the cardioprotective role of S1P3 [34]. In the kidneys, however, S1P3 was shown to be associated with tissue injury after ischemic challenge. Deletion of bone marrow S1P3 attenuated tissue damage following renal ischemia/reperfusion, in which its deletion reduced the expression levels of proinflammatory cytokines and increased the expression levels of anti-inflammatory cytokines [35, 36]. These disparate roles for S1P receptors were similarly observed in the case of S1P1. Renal injury after ischemic challenge was reduced or exacerbated by exposure to an S1P1 agonist [37] or endothelial S1P1 deletion [38]. However, in the brain, S1P1 knockdown reduced brain injury after ischemic challenge [13]. Regardless of the different roles of receptor-mediated S1P signaling in non-neural tissues, it should be noted that all three identified S1P receptors (S1P1, S1P2, and S1P3) mediate pathogenesis in ischemic brain.

In this study, we have used CAY10444 to address the role of S1P3 in cerebral ischemia because CAY10444 has been widely used as a specific antagonist for S1P3 [39,40,41]. But, additional possible modes of actions of CAY10444 were suggested, which included S1P2, P2 receptor, or α1A-adrenoceptor [42]. CAY10444 at 10 Î¼M blocked the S1P2- and S1P3-mediated increase in the intracellular calcium levels in Chinese hamster ovary cells. This inhibitory effect of CAY10444 was also mediated through the stimulation of P2 receptor or α1A-adrenoceptor [42]. These findings indicate that CAY10444 could also act as an antagonist for S1P2 and an agonist for P2 receptor or α1A-adrenoceptor. The latter agonistic property could be excluded in the protective effects of CAY10444 against cerebral ischemia: the association of α1A-adrenoceptor with cerebral ischemia is unclear and suppressing P2 receptor is neuroprotective in this disease [43, 44]. Unlikely, S1P2 could mediate the neuroprotective effects of CAY10444 in cerebral ischemia because S1P2 was reported as a pathogenic factor in this disease [14]. However, it is also possible that CAY10444’s efficacy is solely mediated through S1P3 in cerebral ischemia. In renal ischemic injury, blocking S1P2 activity by JTE013 resulted in renoprotection, whereas CAY10444 did not [45]. The latter indicates that CAY10444 does not act as S1P2 antagonist. It would be tempting to address these opposite notions using genetic tools such as knockout mice for S1P3 in future studies.

The neuroharmful role of S1P3 in the ischemic brain appears to be associated with the activation of brain residence microglia, which is a common pathogenic event in several central nervous system (CNS) disorders, including stroke [46, 47]. Previously, receptor-mediated S1P signaling was reported to be involved in microglial activation through both in vitro and in vivo studies [6, 48]. Recently, we identified that S1P1-mediated brain damage after focal cerebral ischemia was mainly mediated through microglial activation [13]. In this study, we identified that S1P3 was also associated with microglial activation: inhibiting S1P3 using its specific antagonist reduced the number of activated microglia in the ischemic brain, in both a time- and region-dependent manner. Furthermore, the suppression of S1P3 activity in the ischemic brain attenuated microglial proliferation. In addition to the increase in the population, S1P3 in the ischemic brain was closely associated with the morphological transformation of activated microglia. In the ischemic core regions 3 days or more after ischemic challenge, most of the activated microglia were amoeboid shaped and were mainly responsible for neuronal damage in ischemic brain by releasing several proinflammatory mediators [49, 50]. We demonstrated that inhibiting S1P3 resulted in a significant attenuation of the transformation of activated microglia into an amoeboid shape.

S1P3 in the ischemic brain may also link into astrogliosis, a core pathogenesis associated with inflammatory responses in cerebral ischemia [23]. In fact, S1P microinjection into the brain has been reported to cause astrogliosis [6, 51]. Recently, S1P3 was identified as the receptor subtype to regulate astrogliosis, in which a pharmacological antagonism or genetic deletion of S1P3 reduced S1P-triggered inflammatory responses in astrocytes [24]. These previous findings indicate that S1P3-triggered astrogliosis may occur in an ischemic brain. Indeed, we demonstrated that inhibiting S1P3 after tMCAO challenge resulted in a significant attenuation of astrogliosis.

The phenotypical shift of activated microglia has also been extensively considered to understand the pathogenesis of cerebral ischemia [25]. Activated microglia in the ischemic brain become polarized to different phenotypes: classically activated M1- or alternatively activated M2-polarized microglia [25]. M1 microglia are considered as toxic and proinflammatory cells in diverse CNS disorders including cerebral ischemia [52], and the prevention of toxic transformation towards M1 phenotypes has been considered as a possible therapeutic strategy for cerebral ischemia [53, 54]. In contrast, M2-polarized microglia are involved in the repair and resolution phase of ischemic recovery, leading to the neuroprotection [53]. In this study, the suppression of S1P3 activity in the ischemic brain attenuated M1 polarization, as evidenced by the attenuated gene expression of relative markers following tMCAO. However, S1P3 suppression did not alter the expression levels of M2 polarization-relevant markers following tMCAO. These data demonstrate that S1P3 in the ischemic brain is selectively associated with the M1 polarization. This unique role of S1P3 in M1 polarization was obvious in activated microglia, which was confirmed by determining the expression levels of microglial NF-κB, a characteristic marker for M1 polarization [54]. Inhibiting S1P3 significantly reduced the number of Iba1/p65 NF-κB double-immunopositive cells. These in vivo findings of the link between S1P3 and M1 microglial polarization were further affirmed in LPS-stimulated mouse primary microglia, in which inhibiting S1P3 by both genetic and pharmacological tools ensured the attenuation of proinflammatory cytokines. Therefore, S1P3 may mediate brain injury following tMCAO by altering the microglial polarization states to M1, further suggesting that S1P3 is a novel and selective player in regulating M1 microglial polarization.

The underlying signaling mechanisms for the pathogenic roles of S1P3 in cerebral ischemia were linked to PI3K/Akt and MAPK pathways, including ERK1/2 and p38 MAPK. Inhibiting S1P3 following tMCAO increased the Akt phosphorylation in the ischemic brain, whereas it attenuated the phosphorylation of ERK1/2 and p38 MAPK. These signaling molecules are, in particular, considered to regulate the phenotype shift between M1 and M2 polarization. Akt activation in microglia is a signaling molecule that drives activated microglia towards M2 polarization. Additionally, the activation of PI3K/Akt signaling is critical for restricting inflammatory activation of microglia/macrophages and negatively regulates NF-κB signaling, whereas its inhibition drives activated microglia/macrophages towards their M1 polarization [31, 55]. In this context, Akt activation is crucial for cell phenotype shift by inhibiting M1 and activating M2 polarization. In this study, the suppression of S1P3 activity in ischemic brain increased Akt phosphorylation without altering the expression markers of M2 polarization, indicating that the increased Akt phosphorylation by S1P3 inhibition may be linked to the restriction of M1 polarization rather than to the enhancing of M2 polarization. Persistent activation of ERK1/2 signaling has been reported to trigger NF-κB transcriptional activity [28, 29] similar to the activation of p38 [30], both of which eventually lead to the secretion of proinflammatory mediators that are associated with the M1 polarization of activated microglia [56,57,58]. This further ensured that S1P3 activation is closely associated with the M1 polarization of activated microglia in the ischemic brain because the suppression of S1P3 activity attenuated ERK1/2 and p38 MAPK phosphorylation in the ischemic brain.

Conclusions

This study identified S1P3 as a novel pathogenic factor in cerebral ischemia and provided underlying mechanisms, particularly in view of microglial activation. The medically relevant roles of the S1P receptor subtypes in cerebral ischemia have emerged through translational studies. Now, at least three subtypes have been identified to mediate brain injury in cerebral ischemia, including S1P1 [13], S1P2 [14], and S1P3 (the current study). Even though S1P3 may be limited as a therapeutic target because of its negative effects on the heart, it would be a good therapeutic strategy for cerebral ischemia if S1P3-specific antagonist can act inside the CNS. In addition to the identification of novel roles of S1P3, our findings also implicate that the neuroprotective effects exerted by FTY720 in cerebral ischemia in previous studies occur additionally via suppressing S1P3 activity [17], similar to the case of S1P1 [13, 59, 60].

Abbreviations

ABC:

Avidin and biotinylated horse-radish peroxidase macromolecular complex

BrdU:

Bromodeoxyuridine

CNS:

Central nervous system

DAB:

3, 3′-diaminobenzidine tetrahydrochloride

ECL:

Enhanced chemiluminescence

FBS:

Fetal bovine serum

FJB:

Fluoro Jade B

GFAP:

Glial fibrillary acidic protein

LPS:

Lipopolysaccharides

mNSS:

Modified neurological severity score

NPER:

Neuronal protein extraction reagent

PBS:

Phosphate-buffered saline

S1P:

Sphingosine 1-phosphate

tMCAO:

Transient middle cerebral artery occlusion

TTC:

2,3,5-Triphenyltetrazolium chloride

References

  1. Choi JW, Chun J. Lysophospholipids and their receptors in the central nervous system. Biochim Biophys Acta. 1831;2013:20–32.

    Google Scholar 

  2. O'Sullivan S, Dev KK. Sphingosine-1-phosphate receptor therapies: advances in clinical trials for CNS-related diseases. Neuropharmacology. 2017;113:597–607.

    Article  CAS  PubMed Central  Google Scholar 

  3. Czech B, Pfeilschifter W, Mazaheri-Omrani N, Strobel MA, Kahles T, Neumann-Haefelin T, Rami A, Huwiler A, Pfeilschifter J. The immunomodulatory sphingosine 1-phosphate analog FTY720 reduces lesion size and improves neurological outcome in a mouse model of cerebral ischemia. Biochem Biophys Res Commun. 2009;389:251–6.

    Article  CAS  PubMed Central  Google Scholar 

  4. Hasegawa Y, Suzuki H, Sozen T, Rolland W, Zhang JH. Activation of sphingosine 1-phosphate receptor-1 by FTY720 is neuroprotective after ischemic stroke in rats. Stroke. 2010;41:368–74.

    Article  CAS  PubMed Central  Google Scholar 

  5. Kraft P, Gob E, Schuhmann MK, Gobel K, Deppermann C, Thielmann I, Herrmann AM, Lorenz K, Brede M, Stoll G, et al. FTY720 ameliorates acute ischemic stroke in mice by reducing thrombo-inflammation but not by direct neuroprotection. Stroke. 2013;44:3202–10.

    Article  CAS  PubMed Central  Google Scholar 

  6. Moon E, Han JE, Jeon S, Ryu JH, Choi JW, Chun J. Exogenous S1P exposure potentiates ischemic stroke damage that is reduced possibly by inhibiting S1P receptor signaling. Mediat Inflamm. 2015;2015:492659.

    Article  Google Scholar 

  7. Nazari M, Keshavarz S, Rafati A, Namavar MR, Haghani M. Fingolimod (FTY720) improves hippocampal synaptic plasticity and memory deficit in rats following focal cerebral ischemia. Brain Res Bull. 2016;124:95–102.

    Article  CAS  PubMed Central  Google Scholar 

  8. Shichita T, Sugiyama Y, Ooboshi H, Sugimori H, Nakagawa R, Takada I, Iwaki T, Okada Y, Iida M, Cua DJ, et al. Pivotal role of cerebral interleukin-17-producing gammadeltaT cells in the delayed phase of ischemic brain injury. Nat Med. 2009;15:946–50.

    Article  CAS  PubMed Central  Google Scholar 

  9. Wei Y, Yemisci M, Kim HH, Yung LM, Shin HK, Hwang SK, Guo S, Qin T, Alsharif N, Brinkmann V, et al. Fingolimod provides long-term protection in rodent models of cerebral ischemia. Ann Neurol. 2011;69:119–29.

    Article  CAS  Google Scholar 

  10. Fu Y, Hao J, Zhang N, Ren L, Sun N, Li YJ, Yan Y, Huang D, Yu C, Shi FD. Fingolimod for the treatment of intracerebral hemorrhage: a 2-arm proof-of-concept study. JAMA Neurol. 2014;71:1092–101.

    Article  PubMed Central  Google Scholar 

  11. Fu Y, Zhang N, Ren L, Yan Y, Sun N, Li YJ, Han W, Xue R, Liu Q, Hao J, et al. Impact of an immune modulator fingolimod on acute ischemic stroke. Proc Natl Acad Sci U S A. 2014;111:18315–20.

    Article  CAS  PubMed Central  Google Scholar 

  12. Zhu Z, Fu Y, Tian D, Sun N, Han W, Chang G, Dong Y, Xu X, Liu Q, Huang D, Shi FD. Combination of the immune modulator fingolimod with alteplase in acute ischemic stroke: a pilot trial. Circulation. 2015;132:1104–12.

    Article  CAS  PubMed Central  Google Scholar 

  13. Gaire BP, Lee CH, Sapkota A, Lee SY, Chun J, Cho HJ, Nam TG, Choi JW. Identification of sphingosine 1-phosphate receptor subtype 1 (S1P1) as a pathogenic factor in transient focal cerebral ischemia. Mol Neurobiol. 2018;55:2320–32.

    Article  CAS  PubMed Central  Google Scholar 

  14. Kim GS, Yang L, Zhang G, Zhao H, Selim M, McCullough LD, Kluk MJ, Sanchez T. Critical role of sphingosine-1-phosphate receptor-2 in the disruption of cerebrovascular integrity in experimental stroke. Nat Commun. 2015;6:7893.

    Article  CAS  PubMed Central  Google Scholar 

  15. LaMontagne K, Littlewood-Evans A, Schnell C, O'Reilly T, Wyder L, Sanchez T, Probst B, Butler J, Wood A, Liau G, et al. Antagonism of sphingosine-1-phosphate receptors by FTY720 inhibits angiogenesis and tumor vascularization. Cancer Res. 2006;66:221–31.

    Article  CAS  PubMed Central  Google Scholar 

  16. Quancard J, Bollbuck B, Janser P, Angst D, Berst F, Buehlmayer P, Streiff M, Beerli C, Brinkmann V, Guerini D, et al. A potent and selective S1P(1) antagonist with efficacy in experimental autoimmune encephalomyelitis. Chem Biol. 2012;19:1142–51.

    Article  CAS  PubMed Central  Google Scholar 

  17. Nussbaum C, Bannenberg S, Keul P, Graler MH, Goncalves-de-Albuquerque CF, Korhonen H, von Wnuck Lipinski K, Heusch G, de Castro Faria Neto HC, Rohwedder I, et al. Sphingosine-1-phosphate receptor 3 promotes leukocyte rolling by mobilizing endothelial P-selectin. Nat Commun. 2015;6:6416.

    Article  CAS  PubMed Central  Google Scholar 

  18. Gaire BP, Kwon OW, Park SH, Chun KH, Kim SY, Shin DY, Choi JW. Neuroprotective effect of 6-paradol in focal cerebral ischemia involves the attenuation of neuroinflammatory responses in activated microglia. PLoS One. 2015;10:e0120203.

    Article  PubMed Central  Google Scholar 

  19. Chen J, Sanberg PR, Li Y, Wang L, Lu M, Willing AE, Sanchez-Ramos J, Chopp M. Intravenous administration of human umbilical cord blood reduces behavioral deficits after stroke in rats. Stroke. 2001;32:2682–8.

    Article  CAS  Google Scholar 

  20. Han JE, Lee EJ, Moon E, Ryu JH, Choi JW, Kim HS. Matrix metalloproteinase-8 is a novel pathogenetic factor in focal cerebral ischemia. Mol Neurobiol. 2016;53:231–9.

    Article  CAS  PubMed Central  Google Scholar 

  21. Sapkota A, Gaire BP, Cho KS, Jeon SJ, Kwon OW, Jang DS, Kim SY, Ryu JH, Choi JW. Eupatilin exerts neuroprotective effects in mice with transient focal cerebral ischemia by reducing microglial activation. PLoS One. 2017;12:e0171479.

    Article  PubMed Central  Google Scholar 

  22. Li T, Pang S, Yu Y, Wu X, Guo J, Zhang S. Proliferation of parenchymal microglia is the main source of microgliosis after ischaemic stroke. Brain. 2013;136:3578–88.

    Article  PubMed Central  Google Scholar 

  23. Takano T, Oberheim N, Cotrina ML, Nedergaard M. Astrocytes and ischemic injury. Stroke. 2009;40:S8–12.

    Article  PubMed Central  Google Scholar 

  24. Dusaban SS, Chun J, Rosen H, Purcell NH, Brown JH. Sphingosine 1-phosphate receptor 3 and RhoA signaling mediate inflammatory gene expression in astrocytes. J Neuroinflammation. 2017;14:111.

    Article  PubMed Central  Google Scholar 

  25. 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;43:3063–70.

    Article  CAS  PubMed Central  Google Scholar 

  26. Chhor V, Le Charpentier T, Lebon S, Ore MV, Celador IL, Josserand J, Degos V, Jacotot E, Hagberg H, Savman K, et al. Characterization of phenotype markers and neuronotoxic potential of polarised primary microglia in vitro. Brain Behav Immun. 2013;32:70–85.

    Article  CAS  PubMed Central  Google Scholar 

  27. Orihuela R, McPherson CA, Harry GJ. Microglial M1/M2 polarization and metabolic states. Br J Pharmacol. 2016;173:649–65.

    Article  CAS  PubMed Central  Google Scholar 

  28. Jiang B, Brecher P, Cohen RA. Persistent activation of nuclear factor-kappaB by interleukin-1beta and subsequent inducible NO synthase expression requires extracellular signal-regulated kinase. Arterioscler Thromb Vasc Biol. 2001;21:1915–20.

    Article  CAS  PubMed Central  Google Scholar 

  29. Jiang B, Xu S, Hou X, Pimentel DR, Brecher P, Cohen RA. Temporal control of NF-kappaB activation by ERK differentially regulates interleukin-1beta-induced gene expression. J Biol Chem. 2004;279:1323–9.

    Article  CAS  PubMed Central  Google Scholar 

  30. Olson CM, Hedrick MN, Izadi H, Bates TC, Olivera ER, Anguita J. p38 mitogen-activated protein kinase controls NF-kappaB transcriptional activation and tumor necrosis factor alpha production through RelA phosphorylation mediated by mitogen- and stress-activated protein kinase 1 in response to Borrelia burgdorferi antigens. Infect Immun. 2007;75:270–7.

    Article  CAS  PubMed Central  Google Scholar 

  31. Vergadi E, Ieronymaki E, Lyroni K, Vaporidi K, Tsatsanis C. Akt signaling pathway in macrophage activation and M1/M2 polarization. J Immunol. 2017;198:1006–14.

    Article  CAS  PubMed Central  Google Scholar 

  32. Zhao H, Sapolsky RM, Steinberg GK. Phosphoinositide-3-kinase/akt survival signal pathways are implicated in neuronal survival after stroke. Mol Neurobiol. 2006;34:249–70.

    Article  CAS  Google Scholar 

  33. Sensken SC, Staubert C, Keul P, Levkau B, Schoneberg T, Graler MH. Selective activation of G alpha i mediated signalling of S1P3 by FTY720-phosphate. Cell Signal. 2008;20:1125–33.

    Article  CAS  PubMed Central  Google Scholar 

  34. Means CK, Xiao CY, Li Z, Zhang T, Omens JH, Ishii I, Chun J, Brown JH. Sphingosine 1-phosphate S1P2 and S1P3 receptor-mediated Akt activation protects against in vivo myocardial ischemia-reperfusion injury. Am J Physiol Heart Circ Physiol. 2007;292:H2944–51.

    Article  CAS  PubMed Central  Google Scholar 

  35. Bajwa A, Huang L, Ye H, Dondeti K, Song S, Rosin DL, Lynch KR, Lobo PI, Li L, Okusa MD. Dendritic cell sphingosine 1-phosphate receptor-3 regulates Th1-Th2 polarity in kidney ischemia-reperfusion injury. J Immunol. 2012;189:2584–96.

    Article  CAS  PubMed Central  Google Scholar 

  36. Bajwa A, Huang L, Kurmaeva E, Gigliotti JC, Ye H, Miller J, Rosin DL, Lobo PI, Okusa MD. Sphingosine 1-phosphate receptor 3-deficient dendritic cells modulate splenic responses to ischemia-reperfusion injury. J Am Soc Nephrol. 2016;27:1076–90.

    Article  CAS  PubMed Central  Google Scholar 

  37. Awad AS. Selective sphingosine 1-phosphate 1 receptor activation reduces ischemia-reperfusion injury in mouse kidney. AJP: Renal Physiol. 2006;290:F1516–F24.

    CAS  Google Scholar 

  38. Ham A, Kim M, Kim JY, Brown KM, Fruttiger M, D'Agati VD, Thomas LH. Selective deletion of the endothelial sphingosine-1-phosphate 1 receptor exacerbates kidney ischemia–reperfusion injury. Kidney Int. 2013;85:807–23.

    Article  PubMed Central  Google Scholar 

  39. Shirakawa H, Katsumoto R, Iida S, Miyake T, Higuchi T, Nagashima T, Nagayasu K, Nakagawa T, Kaneko S. Sphingosine-1-phosphate induces Ca(2+) signaling and CXCL1 release via TRPC6 channel in astrocytes. Glia. 2017;65:1005–16.

    Article  PubMed Central  Google Scholar 

  40. Li C, Li JN, Kays J, Guerrero M, Nicol GD. Sphingosine 1-phosphate enhances the excitability of rat sensory neurons through activation of sphingosine 1-phosphate receptors 1 and/or 3. J Neuroinflammation. 2015;12:70.

    Article  PubMed Central  Google Scholar 

  41. Tang HB, Jiang XJ, Wang C, Liu SC. S1P/S1PR3 signaling mediated proliferation of pericytes via Ras/pERK pathway and CAY10444 had beneficial effects on spinal cord injury. Biochem Biophys Res Commun. 2018;498:830–6.

    Article  CAS  PubMed Central  Google Scholar 

  42. Jongsma M, Hendriks-Balk MC, Michel MC, Peters SL, Alewijnse AE. BML-241 fails to display selective antagonism at the sphingosine-1-phosphate receptor, S1P(3). Br J Pharmacol. 2006;149:277–82.

    Article  CAS  PubMed Central  Google Scholar 

  43. Pedata F, Dettori I, Coppi E, Melani A, Fusco I, Corradetti R, Pugliese AM. Purinergic signalling in brain ischemia. Neuropharmacology. 2016;104:105–30.

    Article  CAS  PubMed Central  Google Scholar 

  44. Webster CM, Hokari M, McManus A, Tang XN, Ma H, Kacimi R, Yenari MA. Microglial P2Y12 deficiency/inhibition protects against brain ischemia. PLoS One. 2013;8:e70927.

    Article  CAS  PubMed Central  Google Scholar 

  45. Park SW, Kim M, Brown KM, D'Agati VD, Lee HT. Inhibition of sphingosine 1-phosphate receptor 2 protects against renal ischemia-reperfusion injury. J Am Soc Nephrol. 2012;23:266–80.

    Article  CAS  PubMed Central  Google Scholar 

  46. Block ML, Zecca L, Hong JS. Microglia-mediated neurotoxicity: uncovering the molecular mechanisms. Nat Rev Neurosci. 2007;8:57–69.

    Article  CAS  Google Scholar 

  47. Gerhard A, Schwarz J, Myers R, Wise R, Banati RB. Evolution of microglial activation in patients after ischemic stroke: a [11C](R)-PK11195 PET study. NeuroImage. 2005;24:591–5.

    Article  PubMed Central  Google Scholar 

  48. Nayak D, Huo Y, Kwang WX, Pushparaj PN, Kumar SD, Ling EA, Dheen ST. Sphingosine kinase 1 regulates the expression of proinflammatory cytokines and nitric oxide in activated microglia. Neuroscience. 2010;166:132–44.

    Article  CAS  PubMed Central  Google Scholar 

  49. Taylor RA, Sansing LH. Microglial responses after ischemic stroke and intracerebral hemorrhage. Clin Dev Immunol. 2013;2013:746068.

    Article  PubMed Central  Google Scholar 

  50. Sawano T, Watanabe F, Ishiguchi M, Doe N, Furuyama T, Inagaki S. Effect of Sema4D on microglial function in middle cerebral artery occlusion mice. Glia. 2015;63:2249–59.

    Article  PubMed Central  Google Scholar 

  51. Sorensen SD, Nicole O, Peavy RD, Montoya LM, Lee CJ, Murphy TJ, Traynelis SF, Hepler JR. Common signaling pathways link activation of murine PAR-1, LPA, and S1P receptors to proliferation of astrocytes. Mol Pharmacol. 2003;64:1199–209.

    Article  CAS  PubMed Central  Google Scholar 

  52. London A, Cohen M, Schwartz M. Microglia and monocyte-derived macrophages: functionally distinct populations that act in concert in CNS plasticity and repair. Front Cell Neurosci. 2013;7:34.

    Article  CAS  PubMed Central  Google Scholar 

  53. Hu X, Leak RK, Shi Y, Suenaga J, Gao Y, Zheng P, Chen J. Microglial and macrophage polarization-new prospects for brain repair. Nat Rev Neurol. 2015;11:56–64.

    Article  Google Scholar 

  54. Xia CY, Zhang S, Gao Y, Wang ZZ, Chen NH. Selective modulation of microglia polarization to M2 phenotype for stroke treatment. Int Immunopharmacol. 2015;25:377–82.

    Article  CAS  Google Scholar 

  55. Byles V, Covarrubias AJ, Ben-Sahra I, Lamming DW, Sabatini DM, Manning BD, Horng T. The TSC-mTOR pathway regulates macrophage polarization. Nat Commun. 2013;4:2834.

    Article  PubMed Central  Google Scholar 

  56. Harari OA, Liao JK. NF-kappaB and innate immunity in ischemic stroke. Ann N Y Acad Sci. 2010;1207:32–40.

    Article  CAS  PubMed Central  Google Scholar 

  57. Gabriel C, Justicia C, Camins A, Planas AM. Activation of nuclear factor-kappaB in the rat brain after transient focal ischemia. Brain Res Mol Brain Res. 1999;65:61–9.

    Article  CAS  PubMed Central  Google Scholar 

  58. Mattson MP, Camandola S. NF-kappaB in neuronal plasticity and neurodegenerative disorders. J Clin Invest. 2001;107:247–54.

    Article  CAS  PubMed Central  Google Scholar 

  59. Choi JW, Gardell SE, Herr DR, Rivera R, Lee CW, Noguchi K, Teo ST, Yung YC, Lu M, Kennedy G, Chun J. FTY720 (fingolimod) efficacy in an animal model of multiple sclerosis requires astrocyte sphingosine 1-phosphate receptor 1 (S1P1) modulation. Proc Natl Acad Sci U S A. 2011;108:751–6.

    Article  CAS  PubMed Central  Google Scholar 

  60. Graler MH, Goetzl EJ. The immunosuppressant FTY720 down-regulates sphingosine 1-phosphate G-protein-coupled receptors. FASEB J. 2004;18:551–3.

    Article  CAS  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank YJ Bae for the assistance with primary microglia culture and Western blot analysis.

Funding

This work was supported by grants from the National Research Foundation (NRF) to JWC (NRF-2014M3A9B6069339 and NRF-2017R1A2B4002818).

Availability of data and materials

The data generated and analyzed as a part of this study are included within this article (as well as supplementary additional files).

Author information

Authors and Affiliations

Authors

Contributions

BPG, MRS, and JWC designed the research. BPG carried out the in vivo and in vitro experiments. BPG, MRS, and JWC analyzed the data and wrote the manuscript. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Mi-Ryoung Song or Ji Woong Choi.

Ethics declarations

Ethics approval

There are no human participants in this study. All animal handling and surgical procedures were carried out in accordance with the approved animal protocols specified by the Institutional Animal Care and Use Committee at Gachon University.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Additional file

Additional file 1:

Figure S1. CAY10444 (CAY) administration attenuates tMCAO-induced neurodegeneration in post-ischemic brain. Figure S2. CAY10444 (CAY) administration does not alter tMCAO-induced microglial M2 polarization in post-ischemic brain. Figure S3. CAY10444 (CAY) administration attenuates tMCAO-induced astrocytes activation in post-ischemic brain. Table S1. Primer sets used for qRT-PCR analysis. (DOCX 1425 kb)

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gaire, B.P., Song, MR. & Choi, J.W. Sphingosine 1-phosphate receptor subtype 3 (S1P3) contributes to brain injury after transient focal cerebral ischemia via modulating microglial activation and their M1 polarization. J Neuroinflammation 15, 284 (2018). https://doi.org/10.1186/s12974-018-1323-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12974-018-1323-1

Keywords