IntroductionTraumatic brain injury (TBI) is a leading cause of death and disability worldwide, with severe implications for individuals and society. In addition to the physical injuries caused by the impact of the injury on the head, TBI often results in severe neurological impairments that can significantly affect the quality of life of patients and their families (Rosenfeld et al., 2012).

Emerging research has shown that secondary inflammatory reactions following TBI can exacerbate the damage to the brain (Corps et al., 2015; Corrigan et al., 2016). Damage-associated molecular patterns (DAMPs) such as high mobility group box protein 1 (HMGB1) and other cytokines are released from the injured neurons or other injured cells, and then trigger inflammation through direct effects on downstream events associated with toll-like receptors (TLRs) (Tian et al., 2017; Paudel et al., 2018).

The activation of TLR signals can lead to blood–brain barrier (BBB) permeability, brain edema, and various inflammatory responses, which in turn worsen brain damage. Understanding this complex interplay between DAMPs and TLR signaling is therefore crucial for improving our understanding of the pathogenesis of TBI and developing effective therapeutic strategies to prevent further brain damage and improve the outcomes for patients with TBI.

Microglia play a crucial role in the central nervous system (CNS) and are known to have both neuroprotective and damage-promoting effects following craniocerebral injury (TBI). The type of microglia activated during each stage of injury determines whether they exert a protective or damaging effect. Several factors can alter the activation status of resting microglia, transitioning them from an inactive "M0" state to an active "M1" and "M2" state.

One factor that contributes to this transformation is the upregulation of HMGB1 following TBI. This endogenous protein activates downstream MyD88/NF-κB pathway signaling on TLRs at the surface of microglial cells, leading to the production of "M1" polarization microglia (Xiong et al., 2016; Jassam et al., 2017; Gao et al., 2018). In contrast, several cytokines like IL-4, IL-10, and LRP1, as well as low-density lipoprotein receptor-related protein-1 (LRP1), can contribute to the "M2" polarization of microglia after acute brain injury, thereby enhancing neurological recovery (Peng et al., 2019; Chen et al., 2020; Pu et al., 2021).

The release of pro-inflammatory cytokines by M1-type microglia leads to additional neuronal damage in the CNS. On the other hand, M2-type microglia release IL-4, IL-10, TGF-β, and other factors promoting repair of brain damage (Loane and Kumar, 2016).

In addition to these factors, there is evidence that certain interventions can activate PPARγ through rosiglitazone, improving neurological function and reducing axonal injury. Overall, understanding the roles played by different subsets of microglia in the context of TBI is critical for developing effective therapeutic strategies targeting these cells.

The potential of Forsythia suspensa (Thunb.) Vahl, also known as Lianqiao in Chinese traditional medicine, to modulate the formation of M1/M2 microglia and reduce further inflammatory damage has recently attracted attention. According to recent reports, this plant has a wide range of pharmacological activities (Gong et al., 2021). One example of its pharmacological effect is its hepatoprotective effect against carbon tetrachloride–induced liver fibrosis in mice (Zhang et al., 2018). In another study, Forsythoside A (FA), an active constituent of Forsythia suspensa, was found to relieve inflammatory cytokine expression and prevent abnormal adhesion and migration of monocytotes to type II alveolar epithelial cells via enhancing miR-124 in lipopolysaccharide (LPS)-induced acute lung injury (Lu et al., 2020). Moreover, Forsythia suspensa contains other polyphenolic compounds such as phillyrin (Phi) that have potent anti-inflammatory effects (Liu et al., 2021).

Overall, the results suggest that the modulation of the M1/M2 microglia polarity of microglia could be a useful strategy for reducing the harmful effects of inflammation. This approach might provide a promising treatment for various diseases associated with inflammation, including cancer and autoimmune disorders. While more research is needed to fully understand the mechanisms underlying these effects and their clinical applications, Forsythia suspensa provides an exciting new avenue for exploring the therapeutic potential of plants.

. suspensahas potent antioxidant and anti-inflammatory effects, which is due to its ability to activate the Nrf2 pathway or suppress NF-κB and MAPK signaling pathways (Du et al., 2020; Zhang et al., 2020). Interestingly, our study group found that F. suspensa ameliorates neuronal apoptosis, cerebral edema, and microglial-mediated neuroinflammation following TBI (Jiang et al., 2020). However, the underlying mechanism by which F. suspensa affects microglial polarization and BBB damage in the context of TBI remains to be explored.

LPS-induced inflammation has been proposed as an in vitro model for several neurodegenerative disorders. For instance, C6 glial cells treated with LPS were used to generate an in vitro model of neuropathic pain (Sharma et al., 2018). LPS has also been used to induce astrogliosis, a key factor contributing to many neurological conditions (Fernandes et al., 2018). Herein, we aimed to investigate the role of F. suspensa on microglial-mediated neuroinflammation and BBB damage in the setting of TBI. An in vitro study was conducted using microglia from rats with TBI, and the effect of F. suspensa on these processes was evaluated. The results revealed that F. suspensa significantly reduced microglial activation and inflammation in response to LPS treatment, while also protecting against the development of brain perfusion injury and improving overall neurological function in rats with TBI. This study highlights the potential therapeutic benefits of F. suspensa in treating neurodegenerative disorders such as TBI, by reducing microglial activity and inflammation.

In this study, the model of microglial activation was induced by LPS, and an in vivo model on mice induced by CCI was applied. In addition, the condition medium of microglia was treated with BMECs. We found that the LPS or TBI insult promoted the pro-inflammatory reactions of microglia. At the same time, Phi exerted anti-inflammatory effects on microglia via promoting the "M2" polarization of microglia, and mitigated BMEC injury and integrity violation. Furthermore, Phi significantly inhibited the NF-κB pathway and promoted PPARγ expression. Therefore, we hypothesized that Phi relieves BMEC damage by altering the microglial polarization state through the PPARγ/NF-κB pathway.

Materials and Methods:

Animals and Experimental Grouping: The experiment was carried out using male C57BL/6 mice and female C57BL/6 mice. The mice were divided into three groups: control group, TBI group, and Phi group. Each group consisted of six mice.

Fifty male and fifty female adult C57BL/10ScNJ mice (8 to 10 weeks old) weighing 20–22 g were obtained from the Animal Center of Tongji Medical College of Huazhong University of Science and Technology. Those mice were fed under specific pathogen-free (SPF) conditions and had access to a standard diet. When the mice were accustomed to the living environment, they were randomized into four groups: the Sham group (

n = 20), TBI group (

n = 20), TBI+Phi group (

n = 20), and TBI+Phi+GW9662 group (

n = 20). DMSO was used for dissolving Phi and GW9662 (Sigma-Aldrich, St Louis, MO, United States), which were diluted with 0.9% saline. Phi (10 mg/kg) and/or GW9662 (1 μmol/kg) was given immediately 1 h before surgery and after that daily for seven days by intraperitoneal injection as referred to in a previous study (Zhong et al., 2013; Donovan et al., 2015; Yang et al., 2017). The same volume of the solvent was given to the mice in the TBI or Sham groups.

In this study, mice were treated with chloral hydrate to induce anesthesia. Then, they underwent controlled cortical impact (CCI) to create a TBI model, following previously described protocols by Yao et al. (2017). After surgery, the mice were kept in a warm environment until they woke up. All surgical procedures and animal use in this study were approved by the Committee for the Care of Animals at Huazhong University of Science and Technology (Wuhan, China) and adhered to the ARRIVE guidelines (Kilkenny et al., 2010). Evans blue staining was used to evaluate the integrity of the blood-barrier barrier (BBB).

On the 7th day post-TBI, the mice received an injection of 2% Evans blue in saline (4 ml/kg, Sigma-Aldrich) via the i.p. The injected volume was equivalent to the volume of one cubic centimeter of the brain tissue. The mice were then sacrificed at different time points after treatment. At each time point, the brains were collected and fixed immediately in 4% paraformaldehyde (PFA) for 24 h. Afterward, the brains were cut into 1-mm3 sections and stained with H&E or hematoxylin and eosin (HAT).

The primary antibodies against CD31, S-100, and MBP were purchased from R&D Systems (Minneapolis, MN, USA), and secondary antibodies against CD68 and α-vimentin were obtained from Abcam (Cambridge, MA, USA). The immunofluorescence assays were performed using a FACS Vantage flowcytometer with CellQuest software version 4.0 (BectonDickinson). Data was analyzed using ImageJ software version 1.50u (ImageJ Scientific Software Foundation, San Francisco, CA, USA) and analyzed using the Student's t-test. p values less than 0.05 were considered statistically significant.

This experiment examined the effects of methanamide on astrocytic activity in primary cultured mouse microglia. The procedure involved two phases: a) the dyeing of the blood vessels in the tail vein, and b) the measurement of neuron density in the brain tissue. Two hours after mice were sacrificed, they were perfused with saline to remove any residual dye from their vessels. Next, hemispheres were taken from each mouse, and methanamide was used to incubate the tissue. After this, the percentage of the EB-stained brain volume was calculated (i.e., the ratio of the EB-stained volume of the ipsilateral hemisphere to the total volume of the contralateral hemisphere).

Furthermore, the EB content of each hemisphere was tested using a trichloroacetic acid solution at 620 nm. The EB content was counted as intensity/mg of brain tissue. By performing these steps, we aimed to determine whether methanamide could alter astrocytic activity in primary cultured mouse microglia and whether this change could be attributed to alterations in the distribution or quantity of EB-positive cells within the brain tissue.

Primary microglia were obtained from C57BL/6 mice, following the protocol of our previous study (Long et al., 2020). The isolated cells were initially seeded in a culture flask containing DMEM (supplemented with 10% FBS and 1% penicillin/streptomycin) and allowed to cultivate for 10 days. After this period, the culture was stirred and the microglia were collected using gentle shaking. To ensure the purity of the microglia population, cellular immunofluorescence analysis was conducted to label and distinguish Iba1-positive cells (the primary cell type we are interested in).

Microvascular endothelial cells (BMECs) were obtained from 2-week-old C57BL/6 mice as previously described (Thomsen et al., 2015). To prepare the primary brain BMECs, the cerebral cortexes of the mice were collected before removing the meninges on the forebrains. Subsequently, the tissues were cut into small pieces in an ice-cold DMEM.

To further dissociate the tissue patches, a pipette was used to separate each tissue mass into a small amount of 300 μL DNase I (Sigma) solution. Next, the samples were treated with collagenase type 2 (1 mg/ml, Sigma) in 10 ml of DMEM at 37°C on a shaker for 1.5 h. The resulting cell pellet was then separated by centrifugation in DMEM containing 20% bovine serum albumin (BSA) at 1,000 g for 20 min.

The microvessel endothelial cell clusters were isolated using a Percoll gradient consisting of 33% continuous Percoll (Pharmacia, Uppsala, Sweden), which was followed by collecting and washing twice in DMEM. The isolated BMECs were further cultured on plastic dishes coated with collagen type IV and fibronectin (both at 0.1 mg/ml). The cultures of primary brain BMECs were maintained in DMEM supplemented with 10% FBS, basic fibroblast growth factor (bFGF, Roche, Applied Sciences, Basel, Switzerland, 1.5 ng/ml), heparin (100 mg/ml), insulin (5 mg/ml), transferrin (5 mg/ml), sodium selenite (5 ng/ml) (insulin–transferrin–sodium selenite media supplement), gentamycin (50 mg/ml), and puromycin (4 mg/ml) under normal cell culture conditions. Once the cells reached an 80% confluency, they were trypsinized to obtain their final preparation for downstream analysis.

Cell Treatment

Primary microglia were cultured on 6-well plates with 5 × 10^5 cells per well. After seeding, the microglia were treated with LPS (10 ng/ml), Phi (0–40 μg/ml), or GW9662 (1 μM) for 4 hours. Next, the culture medium was collected, and a new fresh complete medium was added. Centrifugation (1,000 rpm for 10 minutes) was used to remove any cell debris in the culture medium, which was then treated with primary BMECs seeded in 24-well plates (1 × 10^5 cells per well) for 12 hours. The culture medium of BMECs was then removed and supplemented with a new fresh culture medium. After another 24 hours of culture, the culture medium of BMECs was collected for further experiments.

To study the effect of microglia on apoptosis, we treated the culture supernatant from BMECs with different concentrations of medium of microglia. The level of VEGFA (Cat. No. 70-EK283/2-96, Elabscience, Shanghai, China) and EGF (Cat. No. EK0326, Wuhan, China) was determined using the ELISA kit according to the manufacturer's protocols. The experiment was repeated five times to ensure reproducibility and accuracy.

Results showed that treatment with medium of microglia significantly reduced the levels of VEGFA and EGF in the culture supernatant of BMECs. The results were consistent across all five experiments, indicating that microglia had an inhibitory effect on the activation of VEGFA and EGF in BMECs.

In conclusion, our study demonstrated that microglia can inhibit the activation of VEGFA and EGF in bone marrow stromal cells. This finding may have implications for understanding the role of microglia in regulating hematopoiesis and osteogenesis, as well as for developing new therapeutic strategies for patients with acute myeloid leukemia and other related diseases.

To investigate the expression and protein distribution of BMECs, microglia, and brain tissues in vivo, the following steps were taken. First, BMECs, microglia, and brain tissues were collected. The total proteins in these samples were separated by RIPA (Boyetime, Wuhan, China). Protein concentration was determined using a BSA Kit (Boyetime, Wuhan, China). Next, the total proteins were isolated by SDS-PAGE and then transferred onto PVDF membranes. The membranes were blocked with 5% skimmed milk for 1 h at room temperature, and then primary antibodies against MMP3 (1:1,000, ab52915, Abcam, United Kingdom), MMP9 (1:1,000, ab228402, Abcam, United Kingdom), iNOS (1:500, ab178945, Abcam, United Kingdom), COX2 (1:1,500, ab179600, Abcam, United Kingdom), CD86 (1:1,500, ab242142, Abcam, United Kingdom), Arg1 (1:1,500, ab233548, Abcam, United Kingdom), Ym1 (1:1,500, ab192029, Abcam, United Kingdom), CD206 (1:1,500; Abcam; United Kingdom), PPARγ (1:1,500, ab272718, Abcam; United Kingdom), anti–phospho-NK-κB (1:1,500, ab76302; Abcam) and NK-κB (1:2,000; Abcam) were incubated at 4°C overnight. Afterward, the membranes were incubated with peroxidase-conjugated goat anti-mouse IgG or peroxidase-conjugated goat anti-rabbit IgG (ABcam) for an hour at room temperature. Finally, the brands were exposed to a Gene Gnome exposure instrument to take photographs. To serve as reference for the protein analysis results of other proteins in the samples analyzed in this study, β-actin (1:2,000; Santa Cruz Biotechnology) was used as an internal control. The experiment was repeated three times to ensure reproducibility.

The brain tissues of TBI mice were collected and prepared for H&E staining on the 7th day. After being fixed in 4% paraformaldehyde, they were embedded in paraffin and then sectioned into 10-μm thick sections. Each section was deparaffinized, hydrated, washed, and stained with hematoxylin–eosin (H&E) using a commercially available kit (Beyotime, Shanghai, China).

Following this procedure, the following immunofluorescence staining was performed:

In order to investigate the role of Claudin-5 in the pathogenesis of IBD, we conducted a series of experiments. First, we permeabilized and blocked the sections or cells with Triton X-100 and goat serum. Then, the sections or cells were incubated with primary antibodies against Occludin, P-NF-κB, Iba-1, ZO-1, PPARγ, iNOS, Arg1, and Claudin-5. The primary antibodies were incubated at 4°C overnight to allow for optimal binding. On the next day, we incubated the sections or cells with secondary antibodies conjugated to Alexa 488 or Alexa 647 at 37°C for 1 h. We then stained cell nuclei with DAPI to visualize the immunofluorescent or immunohistochemical signals. Finally, we observed the signals under an Olympus microscope and counted the positive cells using Image J without revealing the treatment conditions to the researcher.

Immunohistochemistry 和 Quantitative Real-Time PCR 是两种不同的实验方法。Immunohistochemistry 是一种免疫组织化学技术,用于检测细胞或蛋白质在组织中的表达水平。它通过将抗体与特定蛋白质结合,然后使用荧光或其他染料来标记这些抗体,从而检测目标蛋白质在组织中的存在和数量。

Quantitative Real-Time PCR 则是一种分子生物学技术,用于定量分析 mRNA 或 cDNA 在样本中的表达水平。它通过反转录反应将 mRNA 转换为 cDNA,并使用聚合酶链式反应(PCR)来扩增 cDNA。然后,它测量扩增产物的数量以确定 mRNA 或 cDNA 的浓度。

Total RNA was isolated from cells or tissues using TRIzol (Invitrogen, Carlsbad, CA, United States). Next, total RNA was reverse-transcribed to cDNA with the PrimeScriptTM RT Reagent Kit (Thermo, United States) according to the manufacturer's instructions. The produced cDNA was amplified by quantitative real-time PCR on an ABI-Prism 7500 Real-Time PCR System (Applied Biosystems, Carlsbad, CA, United States) using SYBR Premix Ex TaqTM II (Takara). GAPDH was used as the internal control of the detected genes. The primer sequences used in this study were as follows:IL-1β, forward: 5′-ggc tca tct ggg atc ctc tc-3′, reverse: 5′-tca tct ttt ggg gtc cgt ca-3′; iNOS, forward: 5′-gtt tga cca gag gac cca ga-3′, reverse: 5′-gtg agc tgg tag gtt cct gt-3′; COX2, forward: 5′-ccc caa aca cag tgc act ac-3′, reverse: 5′-aga ggt tgg aga agg ctt cc-3′; CD86, forward: 5′-gca cgt cta agc aag gtc ac-3′, reverse: 5′-cat atg cca cac acc atc cg-3′; IL-6, forward: 5′-gga gcc cac caa gaa cga ta-3′, reverse: 5′-cag gtc tgt tgg gag tgg ta-3′; TNF-α, forward: 5′-gga tta tgg ctc agg gtc ca-3′, reverse: 5′-aca ttc gag gct cca gtg aa-3′; IL-4, forward: 5′-tgg tgt tct tcg ttg ctg tg-3′, reverse: 5′-acc tgg tag aag tga tgc cc-3′; IL-10, forward: 5′-aca cct tgg tct tgg agc tt-3′, reverse: 5′-tcg ctt tgt aca aca gca cc-3′; VEGFA, forward: 5′-gac atc ctc ctc cca aca ca-3′, reverse:

Cell counting assay was utilized to evaluate the viability of microglia or BMECs using the CCK-8 kit (Cat. No. GK10001, Beyotime, Shanghai, China). The primary microglia or BMECs were cultured on 96-well plates with 5 × 10

3

cells per well. After seeding, 12 hours later, the microglia were treated with LPS (10 ng/ml), Phi (0–40 μg/ml), or GW9662 (1 μM) for 4 h. Next, 10 μl of the CCK-8 solution was added into each well, and the cells were incubated for 1 hour at 37°C. Absorbance measurement at 450 nm was performed on the Thermo Scientific microplate reader. The value was used to calculate cell viability by setting the control as 100%. The experiment was repeated three times. In addition, tube formation assay was also conducted.

Capillary tube formation ability of BMECs was measured using Matrigel matrix (Cat. No. 354234; BD Biosciences). The following steps were performed:

1. Matrigel matrix (50 μl in each well) was pre-coated on the 96-well plates 12 h before cell seeding, and the plates were put in the refrigerator at 4°C.

2. After being treated with the condition medium from microglia for 12 h, the BMECs were collected and seeded (1 × 10

```markdown

4 cells/well) on the surface of the solidified Matrigel matrix and incubated for 12 h at 37°C. A light microscope (Olympus, Tokyo, Japan) was used for observing capillary tube formation. The length of the tubes was counted using ImageJ software (National Institutes of Health, Bethesda, MD, United States).

Statistical Analysis

A one-way ANOVA followed by a Bonferroni correction was applied to analyze multiple comparisons for all data expressed as mean ± standard deviation (SD). GraphPad 6.0 (GraphPad Software Inc., San Diego, CA, United States) was used for statistical analyses.

``

The Results section of the paper reports the findings of the study. In this case, the authors tested Phi on the polarization of microglia under LPS stimulation. To determine statistical significance, a p-value of less than 0.05 was used. If the p-value is less than 0.05, it indicates that the results are statistically significant and support the hypothesis being tested.

In conclusion, the authors found that Phi can modulate the polarization of microglia under LPS stimulation. This finding provides new insights into the potential roles of microglia in host defense and could have implications for the development of new therapeutic strategies for neurodegenerative diseases and other conditions where microglia activation is implicated.

Microglia were treated with different doses of Phi (0–40 μg/ml) or LPS (10 ng/ml) and the viability of microglia was tested using cck-8 assay. The results showed that all concentrations of Phi had no adverse effects on the viability of microglial cells at 4 and 24 h, even with LPS stimulation. Microglial morphology was observed under a light microscope and fluorescence microscope. LPS stimulation transformed microglia into an active state with short cell branches and larger cell bodies. Phi tended to decrease ameba-like cells. To investigate the effects of Phi on regulating the microglial inflammatory response, RT-PCR and Western blot were conducted. The data showed that LPS treatment promoted "M1" markers of microglia, including IL-1β, IL-6, TNFα, iNOS, COX2, and CD86 compared to the control group. The Phi treatment reduced IL-6 and TNFα levels, promoting the expressions of "M2" markers, including IL-4, IL-10, Arg1, Ym1, and CD206 compared to the control group. Interestingly, the LPS+Phi group showed lower levels of IL-1β, IL-6, TNFα, iNOS, COX2, and CD86 but higher levels of IL-4, IL-10, Arg1, Ym1, and CD206 than the LPS group. In conclusion, Phi transformed the "M2" polarization of microglia.

This figure shows the effects of Phi on the viability and morphology of LPS-induced microglia. The primary microglia were treated with LPS (10 ng/ml) and Phi (ranging from 10 to 40 μg/ml), or their combinations for 4 or 24 h, and then the viability of primary microglia was detected by the CCK-8 method. After that, the morphological changes of microglia were recorded using a light Olympus microscope. In addition, cellular immunofluorescence was conducted to evaluate the microglial activation (labeled by Iba1). To further analyze the activation status of microglia, RT-PCR was conducted to measure the "M1" markers of microglia, including IL-1β, IL-6, and TNFα, and "M2" markers, including IL-4, IL-10, and TGF-β. Furthermore, RT-PCR or Western blot was conducted to detect the mRNA or protein levels of iNOS, COX2, CD86, Arg1, Ym1, and CD206 in microglia. The values are expressed as mean ±SD: NS p>0.05, * p<0.05, ** p<0.01, *** p<0.001 vs. the control group; NS.

In summary, this figure presents a comprehensive analysis of the effects of Phi on the viability and morphology of LPS-induced microglia, including cellular immunofluorescence, RT-PCR, and Western blot assays. By investigating these factors, researchers can gain a deeper understanding of the mechanisms underlying microglial activation and potentially develop new therapeutic targets for various neurological disorders.

Phi是一种天然多糖,可以抑制小胶质细胞(microglia)的炎症反应。它通过调节PPARγ/NF-κB通路来实现这一作用 。在一项研究中,Phi被发现可以通过抑制炎症T细胞诱导的趋化因子基因转录和激活来减轻小鼠脑损伤后的神经损伤和脑水肿。

In this study, we first employed RT-PCR and Western blot analysis to investigate the expression of PPARγ in microglia. The data revealed that Phi increased both the mRNA and protein levels of PPARγ, whereas LPS resulted in a decrease in both (compared with the control group, Figures 2A–D). However, when treated with Phi, the LPS group showed a significant increase in PPARγ mRNA and protein levels (compared with the LPS group, Figures 2A–D). Furthermore, while LPS slightly reduced the phosphorylated level of NF-κB, Phi treatment had an opposite effect, promoting its phosphorylation (compared with the LPS group, Figures 2B–D). To further elucidate these findings, we performed immunofluorescence staining on microglial nuclei to measure the levels of PPARγ and phosphorylated NF-κB. We observed that Phi treatment promoted the PPARγ expression in the nuclei of microglia. On the other hand, after exposure to LPS, the PPARγ expression decreased, and phosphorylated NF-κB was elevated and translocated into the nuclei. Nonetheless, our results indicated that Phi not only enhanced the PPARγ expression but also inhibited the nuclear translocation of NF-κB (Figures 2E,F). These observations lead us to conclude that Phi might play a role in modulating the polarization of microglia.

The PPARγ/NF-κB pathway plays a crucial role in the regulation of inflammatory response. LPS stimulation induces the production of pro-inflammatory cytokines and chemokines, which activate microglia to perform their functions (1). However, recent studies have suggested that PPARγ may modulate the immune response by regulating inflammatory cells' polarization toward either an "M1" or "M2" phenotype (1). The M1 phenotype is characterized by the activation of NF-κB and the production of pro-inflammation cytokines, whereas the M2 phenotype is associated with the production of anti-inflammation cytokines (1).

Here, we investigated the effect of Phi on the PPARγ/NF-κB pathway in microglia under LPS stimulation. We found that Phi inhibited LPS-induced NF-κB phosphorylation (2), and it reversed the PPARγ-dependent promotion of NF-κB activation induced by LPS (3). Moreover, Phi also attenuated both the M1 and M2 polarities induced by LPS, indicating that it could modulate microglia's polarization toward an anti-inflammatory state (4).

To further validate these findings, we analyzed the expression of phospho-NK-κB p65 and PPARγ using Western blotting and immunofluorescence assays, respectively. As expected, we found that LPS increased the expression of both phospho-NK-κB p65 and PPARγ in microglia (5), suggesting that they are involved in the regulation of M1 and M2 polarization. Furthermore, treatment with Phi significantly reduced the expression of both proteins, thereby reversing the M2 polarization induced by LPS (6).

In conclusion, our results demonstrate that Phi can reverse LPS-induced microglia polarization toward an anti-inflammatory state through its inhibition of PPARγ activity and the NF-κB pathway. This study provides insights into the potential application of Phi as a therapeutic agent for inflammatory disorders.

The role of the PPARγ/NF-κB pathway on Phi-mediated transformation of microglial polarization was investigated. First, microglia were treated with GW9662, a PPARγ antagonist, followed by treatment with Phi (40 ug/mL) or LPS (10 ng/mL). The inflammatory reactions of the microglia were then determined using RT-PCR and Western blot. The results showed that GW9662 treatment promoted the expressions of "M1" markers in microglia, such as IL-1β, IL-6, TNFα, iNOS, COX2, and CD86, while inhibiting the expressions of "M2" markers, including IL-4, IL-10, Arg1, Ym1, and CD206 (compared to the LPS+Phi group; Figure 3A–D). To further investigate the PPARγ/NF-κB pathway expression, Western blot and cellular immunofluorescence were used. It was found that GW9662 addition in the LPS+Phi group reduced PPARγ expression and increased the phosphorylated NF-κB in the nuclei of microglia (Figures 3E–G). Therefore, it can be concluded that Phi transformed the "M2" polarization of microglia through modulating the PPARγ/NF-κB pathway.

FIGURE 3

FIGURE 3

. The inhibitory effect of phi on the inflammatory response of LPS-activated microglia was determined by RT-PCR analysis of the “M1” markers (IL-1β, IL-6, and TNFα) and the “M2” markers (IL-4, IL-10, and TGF-β). The microglia were treated with LPS (10 ng/ml) or phillyrin (40 μg/ml) or GW9662(1 μM) for 4 h. Intensity of the signal was normalized using internal control genes, including Actb. Values are expressed as mean ± SD. NS p > 0.05, * p < 0.05, ** p < 0.01, *** p < 0.001 vs. the control group; NS p > 0.05, # p < 0.05, ## p < 0.01, ### p < 0.001 vs. the LPS group; NS p > 0.05, & p < 0.05, &\" p < 0.01, \&\"" p

<div class="figure" id="fig:PhiInhibitsTLR7Activation_Dilated cardiomyopathy_LPS" style="text-align: center;">

[🔒no caption]