- Research
- Open access
- Published:
Punicalagin as a novel selective aryl hydrocarbon receptor (AhR) modulator upregulates AhR expression through the PDK1/p90RSK/AP-1 pathway to promote the anti-inflammatory response and bactericidal activity of macrophages
Cell Communication and Signaling volume 22, Article number: 473 (2024)
Abstract
Aryl hydrocarbon receptor (AhR) plays an important role in inflammation and immunity as a new therapeutic target for infectious disease and sepsis. Punicalagin (PUN) is a Chinese herbal monomer extract of pomegranate peel that has beneficial anti-inflammatory, antioxidant and anti-infective effects. However, whether PUN is a ligand of AhR, its effect on AhR expression, and its signaling pathway remain poorly understood. In this study, we found that PUN was a unique polyphenolic compound that upregulated AhR expression at the transcriptional level, and regulated the AhR nongenomic pathway. AhR expression in lipopolysaccharide-induced macrophages was upregulated by PUN in vitro and in vivo in a time- and dose-dependent manner. Using specific inhibitors and siRNA, induction of AhR by PUN depended on sequential phosphorylation of 90-kDa ribosomal S6 kinase (p90RSK), which was activated by the mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) and phosphoinositide-dependent protein kinase (PDK)1 pathways. PUN promoted p90RSK-mediated activator protein-1 (AP-1) activation. AhR knockout or inhibitors reversed suppression of interleukin (IL)-6 and IL-1β expression by PUN. PUN decreased Listeria load and increased macrophage survival via AhR upregulation. In conclusion, we identified PUN as a novel selective AhR modulator involved in AhR expression via the MEK/ERK and PDK1 pathways targeting p90RSK/AP-1 in inflammatory macrophages, which inhibited macrophage inflammation and promoted bactericidal activity.
Introduction
Infectious diseases, such as sepsis, are an important global health problem. As a syndrome caused by underlying infection, sepsis causes life-threatening organ dysfunction by dysregulation of the host response to infection [1]. The critical pathological mechanisms of sepsis are related to excessive inflammation caused by activation of the immune system and diminished ability to eliminate pathogenic bacteria. Within this context, macrophages perform a pivotal role and act as key target cells for therapeutic intervention [2]. The aryl hydrocarbon receptor (AhR) is a basic helix–loop–helix transcription factor. Accumulating evidence shows that AhR has a variety of physiological effects, and is related to the occurrence and development of various diseases [3]. The pivotal roles of AhR in human physiology and pathophysiology imply that it is a promising therapeutic target [4]. Recently, AhR has been shown to have a dual impact characterized by anti-inflammatory regulation and facilitation of bactericidal function in macrophages, which is a potential new interventional target in sepsis and infectious diseases.
Previous reports have demonstrated the pivotal role of AhR in inflammation and immunity. AhR activation contributes to the establishment of endotoxin tolerance, and septic mice characterized by AhR activation are resistant to lethal attack by Gram-positive and Gram-negative bacteria [5]. AhR senses distinct bacterial virulence factors and controls antibacterial responses, and acts as a major sensor of infection dynamics, coordinating host defense according to infection status [6, 7]. Recent studies have also shown that AhR is a proviral host factor and a candidate therapeutic target for SARS-CoV-2 [8].
AhR signaling pathways include genomic and nongenomic pathways [9]. Inactive AhR exists in complex form with Hsp90, p23, and Src in the cytoplasm, and activated AhR regulates downstream molecules through genomic and nongenomic signaling pathways [10]. Ligands bind to AhR and activate the genomic pathway by promoting AhR translocation to the nucleus. The formation of active heterodimers between AhR and AhR nuclear translocator (ARNT) in the nucleus promotes downstream target gene expression, including cytochrome P450 (CYP)1A1 [10]. In addition to the classical genomic pathway of AhR activation, several alternative nongenomic pathways are completely dependent on the presence of AhR, but do not require nuclear translocation of AhR and binding to dioxin response elements, and their regulatory targets are on the cytoplasm or cell membrane [11]. The genomic pathway tends to mediate the toxicological effects of AhR, while nongenomic pathways are mainly involved in inflammation and immunoregulation [11]. Therefore, it is a more accurate intervention strategy to search for regulators that selectively regulate AhR cytoplasmic expression and nongenomic pathways.
Many AhR ligands (synthetic and natural) have been found. Since its discovery, AhR has been characterized as a xenosensor, specifically activated by classical ligand 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), thereby causing harmful effects to host organisms [12]. Therefore, scientists have been hunting for nonclassical AhR ligands that are less harmful. Natural AhR ligands such as resveratrol, quercetin, and curcumin from polyphenolic compounds possess low toxicity while still maintaining efficacy via the AhR pathway [13]. Punicalagin (PUN) is a Chinese herbal monomer and the main polyphenol component in pomegranate peel extract, which exhibit diverse activities, including antimicrobial, antioxidant, anti-inflammatory, and antitumor activities [14,15,16]. Several studies have shown the roles of PUN in the modulation of inflammation-related disorders [17]. Despite growing interest in the biological and pharmacological activities of PUN, its pharmacological mechanisms and their effects on AhR signaling remain poorly understood.
The 90-kDa ribosomal S6 kinase (p90RSK) family is composed of a group of highly homologous serine/threonine kinases that regulate a wide range of cellular processes, such as cell growth, survival, proliferation, and motility. The p90RSK kinases are directly phosphorylated by extracellular signal-regulated kinase (ERK) and phosphoinositide-dependent protein kinase (PDK)1, resulting in their activation. p90RSK activation induces the downstream gene phosphorylation and transcription of c-fos, which is a part of activator protein-1 (AP-1) transcription factor family [18]. The AhR signaling pathway interconnects with various other pathways, such as ERK and AP-1 [19, 20]. The AhR ligands, including PM10 [21] and indoxyl sulfate [22], upregulate AP-1 activity in an AhR-dependent manner. Furthermore, latent membrane protein 2A inhibits the expression and pathway activation of AhR by inducing phosphorylation of ERK [23].
In this study, we screened PUN as a selective AhR modulator from polyphenolic compounds that upregulated AhR expression, which was different from resveratrol, curcumin, and quercetin. We also found that PUN promoted AhR expression at the transcriptional level, mainly regulating the AhR non-genomic pathway. Activation of p90RSK was a checkpoint for AhR upregulation by PUN and lipopolysaccharide (LPS), which upregulated AhR via the mitogen-activated protein kinase kinase (MEK)/ERK and PDK1 pathways targeting c-fos. Finally, AhR upregulation by PUN inhibited inflammation and promoted bactericidal activity in macrophages.
Materials and methods
Materials and reagents
LPS (Escherichia coli 0111: B4) (L4130) and thioglycollate medium (T9032) were procured from Sigma-Aldrich (St. Louis, USA). Punicalagin (A0226), resveratrol (A0051), quercetin (A0083) and curcumin (A0086) were provided by Chengdu MUST bio-technology co, LTD (Chengdu, China). Inhibitor PD98059 (S1177) and GSK2334470 (S7087) were purchased from Selleck (Houston, Texas, USA). Inhibitor SCH772984 (HY-50846), LJH685 (HY-19712) and T-5224 (HY-12270) were purchased from Med chem express (New Jersey, USA). Inhibitor CH-223191 (C8124), Benzopyrene (B1760) and β-Naphthoflavone (N3633) were purchased from Sigma-Aldrich. THP-1 were obtained from Procell Life Science& Technology Co. Ltd. (CL-0233, Wuhan, China). Inhibitor U0126 (S1901), nuclear and cytoplasmic protein extraction kit (P0028), BCA protein assay kit (ST2222), Cell lysis buffer for Western and IP (P0013) and Protease/Phosphatase Inhibitor Cocktail (P1051) were provided by Beyotime (Shanghai, China).
Animals
Adult C57BL/6 mice (average body weight of 20 ~ 22 g, aged 6 ~ 8 weeks) were acquired from the of Daping Hospital’s (Chongqing, China) Experimental Animal Center. The AhR-KO mice were derived from AhR heterozygote mice that were obtained from Jackson's laboratory. All animal experiments were conducted in the SPF animal room of Daping Hospital's Experimental Animal Center (SYXK20170002), whose ethics approval number is AMUWEC20211429.
Extract mice peritoneal macrophages and cells culture
Mice were injected with 4% thioglycolate broth three days ahead, the peritoneal macrophages obtained by washing the abdominal cavity with PBS and adhering to a Petri dish for 2 h. After the suspension cells were washed away by PBS, adherent macrophages were collected. Macrophage and THP-1 cells were cultured in RPMI-1640 with 10% FBS, penicillin, and streptomycin. Cells were maintained at 37 °C in a 5% CO2 humidified environment.
Western blotting
Cells lysed in buffer (20mM Tris, 150mM NaCl, 1% Triton X-100) for 15 min on ice, debris removed by centrifugation. Protein quantified by BCA assay. Equal protein per sample analyzed on 10% Tris-Glycine extended stain-free gels. Total protein stains used as loading controls for quantitative protein analysis. SDS gels transferred to PVDF membranes, blocked with 5% skim milk for 1 h, and incubated with primary antibodies overnight at 4 °C. Next day, membranes washed and probed with HRP-conjugated secondary antibody for 1 h. Immunoreactive bands were observed, identified, and quantitatively evaluated using enhanced chemiluminescence detection system. Detailed quantification was performed by Image J software for densitometric analysis. The antibodies information were listed in Table S1.
Quantitative reverse transcription PCR analysis
Total RNA was extracted from macrophages using the RNA rapid extraction kit (BioTeke, Beijing, China). The cDNA was synthesized from 1 μg RNA, and qRT-PCR was performed with SYBR Premix (TaKaRa). All the primer sequences used for each targeted gene were provided in Supporting Information Table S2.
Flow cytometry
Cells were stained with FITC-F4/80 and BV605-CD11b antibodies, and incubated 15 min in the dark. Fixed and permeabilized with Fixation/Permeabilization Solution Kit (BD, Biosciences) for 2 h. Stained with APC-AhR or APC-isotype antibodies for 30 min. Washed and centrifuged, cells resuspended in PBS and analyzed by flow cytometry. The antibodies information were listed in Table S3.
Immunofluorescence and confocal analysis
Cells on Confocal Petri Dish were fixed for 30 min with 4% Paraformaldehyde, permeabilized for 10 min with 0.5% Triton X-100, and blocked for 60 min with QuickBlock™ Blocking Buffer. Cells were incubated with AhR (GTX22770, GeneTex) or Phospho-ERK (4370, CST) antibodies overnight at 4 °C, then incubated with Alexa Fluor 647-conjugated or 480-conjugated secondary antibody for 1 h. DAPI Staining Solution was added for 10 min. Imaging was performed using a laser-scanning confocal microscope. After staining the nucleus with DAPI, the images were then collected by laser scanning confocal microscope for analysis (Olympus).
RNA interference
Cells were transfected with 100 nM of scrambled or p90RSK-specific siRNAs (Ribobio) using riboFECTTM CP Reagent in six-well plates. After 48 h, siRNA and oligofectamine mixtures were discarded. The mRNA and protein expression of p90RSK were detected by qPCR or Western blotting to confirm the efficiency of gene interference. The siRNA sequences information were listed in Table S4.
EMSA
The DNA–nuclear protein binding activity was evaluated by electrophoresis on a native gel. The expression of the complex on the gel was then measured using the Odyssey Scanning system. The DNA-binding sequences of AhR and AP-1 on the promoter were synthesized into infrared ray 700 labeled probes, and their information is listed in Table S5.
Preparation of Listeria cells
Listeria monocytogenes (ATCC19115) was grown in LB medium for 16 h at 37 °C on an orbital shaker. After washing with 0.9% NaCl and centrifugation, bacterial sediments were resuspended in sterile glycerine to form a 1 × 108 CFU/mL bacterial solution.
In vitro bactericidal assay
Macrophages from wild-type and AhR-KO mice were collected and seeded to the 12-well plates with 1 × 106 cells per well, and were pretreated with PUN for 12 h. Discard the culture medium and wash three times with sterile PBS. Peritoneal macrophages were infected with Listeria for 1 h (MOI = 1). The medium was discarded and washed, then the extracellular bacteria were killed with the antibiotic culture medium. The cells were dissolved in 200µL 0.5% TritonX-100 for 5 min, the supernatant was centrifuged, and the precipitate was re-suspended in 100µL sterile PBS. After 10 times dilution, 30µL bacterial solution was applied to LB medium and incubate at 37 ℃ for 18 h. The quantitative enumeration of bacterial colonies was performed using the colony-forming unit (CFU) method.
Analysis of cytotoxicity
Lactate dehydrogenase cytotoxicity test kit purchased from Beyotime (C0016). Prepare macrophages into 1 × 105 cell/mL dilution solution. Seeded to the 96-well plates with 100 μL cell dilution solution per wells. Cells were pretreated with PUN for 12 h and subsequently challenged with Listeria for additional 24 h (MOI = 1). The LDH assay was performed in accordance with the product manual.
Transcriptome sequencing
Macrophages RNA was extracted using the TRIzol extraction method. After the concentration and purity of RNA were qualified, sequencing was performed on the second-generation sequencing platform. According to the read count of each gene, the relative quantitative analysis of gene expression and enrichment analysis of differential genes were carry out. When comparing between groups, the genes with a P value less than 0.05 and a fold change greater than 2 were identified as significantly differentially expressed genes. The analysis and illustration of results were performed using Majorbio Cloud Software.
Molecular docking and affinity prediction
AhR consisted of PASA and PASB two key domains, the latter of which was a target for conventional drugs. Adopting AlphaFold2 forecasted crystal structure. The PASB crystal structure was downloaded in the https:/ww.alphafold.ebi.ac.uk/entry/P30561. The two-dimensional structure of each compound was obtained from pubchem, and Gaussian 09 was used to optimize the molecule, B3LYP density functional was used to optimize the molecule, 6-31 g (d) was selected as the base group, and aqueous solution was selected as the solution. AutoDock Vina was used to dock each compound with the crystal structure separately. The molecular size of each compound was calculated using VEGA ZZ3.2.2.
Statistical analyses
Data were presented as mean ± standard error (SEM). The number of independent experiments is indicated by N. Statistical analyses were performed using appropriate tests for multiple comparisons (one-way ANOVA) or for pairwise comparisons (t-test). All statistical analyses were conducted using Prism version 8.02 (GraphPad Software). of P values < 0.05 were considered statistically significant.
Results
PUN screened from polyphenolic compounds was a selective AhR modulator
Previous studies have reported that the plant polyphenol flavonoids resveratrol (RES), quercetin (QUE) and curcumin (CUR) act as ligands of AhR and induce expression of CYP1A1 [24]. We conducted a comparative examination of the effects of PUN and the above compounds on AhR expression in LPS-induced mouse macrophages. As reported previously [9, 25], AhR expression was increased by LPS in peritoneal macrophages. We found that PUN alone or combined with LPS upregulated expression of AhR, and their combination was more potent (Fig. 1A, B). However, treatment with resveratrol, quercetin, and curcumin, alone or in combination with LPS, did not cause any significant change in AhR expression (Fig. 1A, B). To evaluate the binding of PUN to AhR, we used molecular docking techniques to evaluate the affinity of polyphenol compounds to AhR. Compared with TCDD, resveratrol, quercetin, and curcumin had a significantly lower affinity for AhR, decreasing by a factor of 4–10 times. The affinity of PUN for AhR was reduced by ~ 40 times compared with TCDD (Fig. 1C and S1A). We discovered that TCDD penetrated the hydrophobic pocket of AhR. In contrast, PUN was too large to enter the hydrophobic pocket, thus, it did not activate AhR (Fig. 1D). Benzo (a) pyrene (Bap) and β-naphthoflavone (βNAF) are agonists of AhR, which can promote the nuclear translocation of AhR. However, PUN did not induce AhR nuclear translocation but rather promoted its cytoplasmic expression (Fig. 1E). The downstream factors of AhR, including CYP-related genes, were not affected by PUN, regardless of CYP1B1 (Fig. 1F, G, and S1B, C). Additionally, PUN did not affect the DNA binding activity between nucleoproteins and dioxin response elements on the AhR promoter (Fig. 1H). Taken together, PUN screened from polyphenolic compounds was a selective AhR modulator that upregulated AhR expression, but did not induce nuclear translocation and downstream gene transcription of AhR.
Punicalagin screened from polyphenolic compounds was selective AhR modulator. A Western blot analysis of AhR in peritoneal macrophages were induced with LPS (10 μg/mL) for 12 h after pre-treated with Punicalagin (50 μM), Resveratrol (25 μM), Quercetin (10 μM) and Curcumin (10 μM) for 1 h. B The relative abundance of protein AhR was calculated by loading control normalization (n = 3 bands per group). C Predicted affinities of TCDD, Punicalagin, Resveratrol, Quercetin, and Curcumin compounds to AhR proteins. D Evaluated the affinity of TCCD/PUN to AhR with molecular docking techniques. E AhR expression in the cytoplasm and nucleus was analyzed by Western blot. Peritoneal macrophages were pretreated with PUN for 1 h and then induced with LPS for 8 h. Alternatively, macrophages were induced with LPS for 8 h and then treated with Benzo (a) pyrene (Bap) (10 μM) and β-Naphthoflavone (βNAF) (1 μM) for 0.5 h. F-G Expression of AhR downstream related genes in peritoneal macrophages. F Transcriptome analysis; (G) qRT-PCR analysis (n = 3 groups per group). H Macrophages were stimulated with or without LPS for 12 h and PUN pretreated 1 h, nuclear-extract proteins were then prepared and incubated with AhR-binding site probe. Binding activity was measured using EMSA. The experiment was repeated three times, the data are shown as mean ± SEM. Combined results are compared using an one-way ANOVA test. N.S P > 0.05, ∗ P < 0.05, ∗ ∗ P < 0.01
PUN upregulated AhR expression in LPS-induced macrophages in vivo and in vitro
Several studies have demonstrated that AhR inhibited expression of interleukin (IL)-6 and IL-1β [26], and stimulated expression of IL-10 in inflammatory macrophages [9]. Therefore, we focused on the effect of PUN on AhR in inflammatory macrophages. In vivo, intraperitoneal injection of PUN increased the percentage of AhR-positive peritoneal macrophages in LPS-induced mice (Fig. 2A). In vitro, we confirmed that PUN alone or combined with LPS upregulated AhR protein expression (Fig. 2B) and mRNA expression (Fig. 2C). Following PUN pretreatment, AhR level were significantly increased at both 8 h and 12 h (Fig. 2D). AhR expression was initially triggered at a PUN concentration of 12.5 μM, with a maximum level observed at 50 μM (Fig. 2E). Therefore, AhR expression in macrophages treated with LPS was significantly augmented by PUN in a time- and dose-dependent manner (Fig. 2D, E). Compared with LPS-stimulated macrophages, the mean fluorescence intensity (MFI) of AhR was significantly enhanced in PUN-stimulated macrophages (Fig. 2F). Furthermore, the majority of the increased AhR was localized to the cytoplasm (Fig. 2F). In human THP-1 cells, we also demonstrated that PUN has the effect of up-regulating AhR expression (Fig. 2G). In summary, PUN exhibited the ability to augment AhR expression in inflammatory macrophages.
Punicalagin upregulated AhR expression in LPS-induced macrophages in vivo and in vitro. A Flow cytometry analysis of AhR+ peritoneal macrophages in mice (n = 12). The mice were pretreated with 12.5 mg/kg PUN or equal volume PBS for 12 h and then stimulated with 10 mg/kg LPS for 8 h. The results are compared using t test. B Western blot analysis of AhR in the cell lysates. The cells were pretreated with PUN (50 μM) for 1 h, and then induced with LPS (10 µg/mL) for 12 h. Gray value analysis of relative abundance of AhR normalized to loading control. C The AhR mRNA levels were detected with qRT-PCR. The cells were pretreated with PUN (50 μM) for 1 h, and then induced with LPS (10 µg/mL) for 4 h or 8 h. D Western blot analysis of AhR in the cell lysates. Peritoneal macrophages were pretreated with PUN (50 μM) for 1 h, and then induced with LPS (10 µg/mL) for for different times (8 h, 12 h). E Western blot analysis of AhR in the cell lysates. Peritoneal macrophages were stimulated either with different doses of PUN (12.5 μM, 25 μM, 50 μM) for 1 h, and then induced with LPS (10 µg/mL) for 12 h. F AhR protein expression was measured with immunofluorescence. Cells were pretreted with 50 μM PUN for 1 h and induced by 1mg/mL LPS for 8 h, then immunostained with AhR specific antibody (Purple), cell nucleus stained with DAPI (Blue). Bar = 20 µm. The fluorescence intensity of AhR was measured (n = 6 sheets per group). G The THP-1 cells were pretreated with PUN (50 μM) for 1 h, and then induced with 10 µg/mL LPS for 12 h. Western blotting analysis with AhR antibodies. Gray value analysis of relative abundance of AhR normalized to loading control. The experiment was repeated three times. The data are shown as mean ± SEM. Combined results are compared using one-way ANOVA test or t test, ∗ P < 0.05
AP-1 was involved in AhR induction by PUN
Activated nuclear factor (NF)-κB is a key mediator of AhR expression [9, 27]. We found that the increase in AhR expression induced by PUN was not associated with NF-κB (Fig. S2A), prompting us to investigate additional transcription factors involved in AhR regulation. The AhR gene promoter contains an AP-1 binding site [28], and inhibition of AP-1 activity reduces AhR expression [29], suggesting the correlation between AP-1 and AhR expression. AP-1 is a transcription factor, usually composed of a heterodimer of the proteins c-fos and c-jun. Compared with untreated cells, serine phosphorylation of c-fos was increased in PUN-treated cells following LPS stimulation, while PUN had a tendency to increase the serine phosphorylation of c-jun (Fig. 3A). Within 0.5–2 h after LPS stimulation, phosphorylation of c-fos in macrophages pretreated with PUN increased significantly (Fig. 3B). Our analysis of the JASPAR database [30] indicated that the sequence spanning -697 to -687 bp on the AhR promoter would have the highest c-fos binding score (Fig. 3C). Next, we confirmed by electrophoresis mobility shift assay that PUN significantly enhanced the ability of nuclear protein to bind to AhR promoter in LPS-stimulated macrophages (Fig. 3D). We explored whether the suppression of c-fos affected AhR expression, using an AP-1 inhibitor (T-5224), which significantly inhibited phosphorylation of c-fos (Fig. 3E and S2B). Compared with control cells, AhR mRNA (Fig. 3F) and protein (Fig. 3G) expression levels were significantly lower in LPS- and PUN-stimulated cells exposed to T-5224. Above all, AP-1 was thought to play a crucial role in the triggering of AhR induction by PUN in macrophages exposed to LPS.
AP-1 was involved in AhR induction by PUN. A The cells were pretreated with PUN (50 μM) for 1 h, and induced with LPS (10 µg/mL) for 30 min. cell lysates subjected to western blotting analysis with total c-fos, phospho-c-fos (Ser32), total c-jun, phospho-c-jun (Ser63). Gray value analysis of relative abundance of phospho-c-fos (Ser32) and phospho-c-jun (Ser63). B The cells were pretreated with PUN (50 μM) for 1 h, and induced with LPS (10 µg/mL) for 30 min, 1 h, 2 h. Cell lysates subjected to western blotting analysis with phospho-c-fos (Ser32). C ~ G PUN promoted AhR expression through transcription factor of c-fos. C The AhR promoter contains a binding site for the transcription factor c-fos. D Macrophages were stimulated with PUN pretreated 1 h and LPS for 0.5 h, nuclear-extract proteins were then prepared and incubated with AP-1-binding site probe. Binding activity was measured using EMSA. E Macrophages were pretreated with transcription factor AP-1 inhibitor T-5224 (10 μM) for 2 h, and then pretreated with PUN (50 μM) for 1 h, and induced with LPS (10 µg/mL) for 12 h. Cell lysates subjected to western blotting analysis with total c-fos, phospho-c-fos (Ser32). F The mRNA expression of AhR was analyzed by qRT-PCR after the application of transcription factor AP-1 inhibitor T-5224. G The protein expression of AhR was analyzed by western blotting after the application of T-5224. Gray value analysis of relative abundance of AhR normalized to loading control. The experiment was repeated three times. The data are shown as mean ± SEM. Combined results are compared using one-way ANOVA test. N.S P > 0.05, ∗ P < 0.05
MEK/ERK pathway, but not ERK phosphorylation and nuclear translocation, mediated PUN induction of AhR expression in LPS-induced macrophages
To explore the molecular mechanism of upregulation of AhR by PUN, we performed transcriptome sequencing on macrophages treated with LPS, PUN, or both. As shown in the Venn diagram, among the 1291 differentially expressed genes affected by LPS stimulation, 197 were detected in response to PUN treatment (Fig. 4A). Enrichment analysis of the KEGG signaling pathway suggested possible correlation with phosphoinositide 3-kinase (PI3K)/AKT, NF-κB, and mitogen-activated protein kinase (MAPK) signaling pathways (Fig. 4B). However, we revealed that pretreatment of macrophages with the NF-κB inhibitor pyrrolidine dithiocarbamate (PDTC) did not suppress PUN-induced AhR expression (Fig. S2A). After pretreatment with MEK (U0126 and PD98059) and ERK (SCH772984) inhibitors, the expression of AhR coactivated by LPS and PUN was significantly decreased (Figs. 4C–E and S2C, D). MEK (U0126) and ERK (SCH772984) inhibitors also downregulated phosphorylation of c-fos activated by LPS and PUN (Fig. S3C–F). In comparison to LPS, expression of phosphorylated MEK and total MEK was significantly augmented by PUN (Fig. 4F). Remarkably, PUN decreased the extent of ERK phosphorylation in LPS-stimulated macrophages (Fig. 4G, H) and reduced the translocation of phosphorylated ERK into the nucleus (Fig. 4I). Similarly, PUN also reduced phosphorylation of mitogen- and stress-activated kinase 1, a molecule downstream of ERK (Fig. S2E). These results supported the hypothesis that the MEK/ERK pathway, but not ERK phosphorylation and nuclear translocation, mediated PUN induction of AhR expression in LPS-induced macrophages. This suggests that additional transcription factors mediate the effect of PUN on upregulation of AhR expression.
MEK-ERK pathway, but not ERK phosphorylation and nuclear translocation, mediated PUN induction of AhR expression in LPS-induced macrophages. A Venn diagram was used to analyze the differential gene sets between the two groups, LPS vs Control and LPS vs LPS + PUN. B KEGG enrichment analysis signaling pathway enriched by differential expression genes. C ~ E Peritoneal macrophages were pretreated with MEK inhibitors U0126 (20 μM) or PD98059 (10 μM), ERK inhibitors SCH772984 (1 μM) for 1 h and then pretreated with PUN (50 μM) for 1 h, and then treated with LPS (10 µg/mL) for 12 h, cells lysates subjected to western blotting analysis with AhR antibodies. Gray value analysis of relative abundance of AhR normalized to loading control. F ~ I Peritoneal macrophages were pretreated with PUN (50 μM) for 1 h and then treated with LPS (10 µg/mL) for 30 min. Cells lysates subjected to western blotting analysis the protein levels of MEK / p-MEK (F), ERK / p-ERK (G), and ERK / p-ERK in the nucleus and cytoplasm respectively(I). Gray value analysis of relative abundance of p-ERK in nucleus normalized to loading control. H Cells were immunostained with p-ERK antibody (Green) and co-stained with DAPI (Blue) to detect nucleus. Bar = 20 µm. The experiment was repeated three times. The data are shown as mean ± SEM. Combined results are compared using one-way ANOVA test or t test. ∗ P < 0.05, ∗ ∗ P < 0.01
p90RSK activation was critical for AhR expression in macrophages increased by PUN
Previous studies have reported that ERK activated p90RSK, and phosphorylated p90RSK translocated into the nucleus to stimulate activation of the downstream transcription factor c-fos [18, 31, 32]. p90RSK contains four essential phosphorylation sites (Ser221, Thr359, Ser380, and Thr573) (Fig. 5A) [18]. In this study, PUN promoted LPS-induced phosphorylation of Ser221, Thr359, Ser380, and Thr573 of p90RSK in macrophages (Fig. 5B). PUN significantly increased the translocation of phosphorylated p90RSK (Ser221) into the nucleus, while exerting no influence on the nuclear translocation of phosphorylated p90RSK (Thr573) (Fig. 5C). Additionally, we explored the potential impact of p90RSK inhibitor (LJH685) on the regulation of AhR by detecting the phosphorylation sites (Ser221, Thr359, Ser380, and Thr573) of p90RSK activated by LPS + PUN (Fig. 5D). Consistent with our hypothesis, we observed a decrease in the expression of AhR and phosphorylation of c-fos in PUN-stimulated cells following treatment with a p90RSK inhibitor (Figs. 5E and S3G, H). Subsequently, we utilized siRNA knockdown of p90RSK to identify the individual with the highest knockdown efficiency for subsequent experimentation (Fig. 5F). AhR expression induced by LPS and PUN was decreased in macrophages treated with p90RSK siRNA compared with macrophages treated with scrambled siRNA (Fig. 5G). In addition, MEK (U0126) and ERK (SCH772984) inhibitors also downregulated phosphorylation of p90RSK activated by LPS and PUN (Fig. S3A, B), suggesting that p90RSK was regulated by upstream MEK/ERK in promoting AhR expression. These results supported the hypothesis that p90RSK activation was critical for AhR expression in macrophages increased by LPS and PUN.
p90RSK activation was critical for AhR expression in macrophages increased by PUN. A Structure diagram of the RSK, containing the S221, T359, S380 and T573 phosphorylation sites. B ~ C Peritoneal macrophages cells were treated with LPS (10 µg/mL) for 30 min after pretreated with PUN (50 μM) for 1 h. Cells lysates used for western blotting analysis. B The protein level of phospho-p90RSK (Thr573), phospho-p90RSK (Ser380), phospho-p90RSK (Thr359), phospho-p90RSK (Ser221) and RSK. C The protein level of p-p90RSK (Thr753) and p-p90RSK (Ser221) in cytoplasm and nucleus. D ~ E Peritoneal macrophages were pretreated with p90RSK inhibitors LJH685 (10 μM) for 1 h and then pretreated with PUN (50 μM) for 1 h, and then treated with LPS (10 µg/mL) for 0.5 h or 12 h, cells lysates subjected to western blotting analysis. D The protein level of phospho-p90RSK (Thr573), phospho-p90RSK (Ser380), phospho-p90RSK (Thr359), phospho-p90RSK (Ser221). E The protein level of AhR. F Expression of RSK protein after RSK silencing in macrophages by Western blotting analysis. G Western blotting detected the AhR protein level after RSK silencing and pretreated with (50 μM) PUN for 1 h and then induced by 10 µg/mL LPS for 12 h. All the bands gray value analysis of relative abundance of protein normalized to loading control. The experiment was repeated three times. The data are shown as mean ± SEM. Combined results are compared using one-way ANOVA test. ∗ P < 0.05
PUN promoted p90RSK mediated AhR expression through the PDK1 pathway in LPS-induced macrophages
Complete activation of p90RSK necessitated the cooperative effect of the MEK/ERK and PDK1 signaling pathways [18, 33]. The former has been demonstrated to mediate PUN-induced AhR expression. Consequently, we examined the effect of PUN on the PDK1 signaling pathway. PUN promoted PDK1 phosphorylation and total PKD1 protein expression (Fig. 6A). We further investigated the impact of PDK1 inhibition on AhR expression using PDK1 inhibitor (GSK2334470). This significantly counteracted the phosphorylation of PDK1 triggered by LPS + PUN (Fig. 6B). When PUN-activated cells were treated with GSK2334470, AhR expression was significantly reduced compared with control cells (Fig. 6C, D). GSK2334470 also decreased the phosphorylation of p90RSK, c-fos, and c-jun activated by LPS + PUN (Fig. 6E–G). These results identified that PDK1 was involved in the induction of AhR by PUN in LPS-induced macrophages.
PUN promoted p90RSK mediated AhR expression through PDK1 pathway in LPS-induced macrophages. A, B Peritoneal macrophages cells were pretreated with PUN (50 μM) for 1 h, and then treated with LPS (10 µg/mL) for 30 min. Cells lysates subjected to western blotting analysis with total PDK1, phospho-PDK1. B ~ G Before cells were treated with LPS and PUN at the same dose and time, cells were pretreated with PDK1 inhibitor GSK2334470 (5 nM) for 1 h. Cells lysates subjected to western blotting analysis. B PDK1 and phospho-PDK1. C AhR. D Gray value analysis of relative abundance of AhR normalized to loading control. E phospho-p90RSK (Thr573), phospho-p90RSK (Ser380), phospho-p90RSK (Thr359) and phospho-p90RSK (Ser221). F c-fos and phospho-c-fos(s32). G c-jun and phospho-c-jun(s63). The experiment was repeated three times. The data are shown as mean ± SEM. Combined results are compared using one-way ANOVA test. ∗ P < 0.05
AhR upregulated by PUN inhibited macrophage inflammation and promoted bactericidal activity
Recently, it has been found that AhR negatively regulates the inflammatory response of macrophages, and contributes to the bactericidal capacity of macrophages [6, 34, 35]. In LPS-induced macrophages, PUN inhibited mRNA expression levels of inflammatory cytokines IL-6, IL-1β, and IL-27, but after pretreatment with AhR inhibitor CH223191, the fold change by PUN on reducing these inflammatory factors was partially reversed (Fig. 7A–C). The above results were also confirmed in AhR knockout macrophages (Fig. 7D). More significantly, in AhR knockout macrophages, the effect of PUN in reducing IL-6 and IL-1β was almost completely eliminated (Fig. 7E, F). In Listeria-infected macrophages, AhR promoted bacterial clearance by enhancing reactive oxygen species (ROS) production and inhibited apoptosis by inducing apoptosis inhibitor of macrophage expression [34]. Therefore, we investigated whether PUN enhanced macrophage clearance of Listeria monocytogenes and inhibited macrophage death in an AhR-dependent manner. PUN promoted Listeria clearance in wild-type macrophages, but this effect was reversed in AhR knockout macrophages (Fig. 7G, H). Similarly, the effect of PUN on reducing the cell death rate of Listeria-induced macrophages was partially reversed in AhR knockout macrophages (Fig. 7I). These results suggested that AhR upregulated by PUN inhibited macrophage inflammation and promoted bactericidal activity.
AhR upregulated by PUN inhibits macrophage inflammation and promotes bactericidal activity. A ~ C Peritoneal macrophages cells were pretreated with AhR inhibitor CH-223191 and then treated with LPS (10 µg/mL) for 4 h after pretreated with PUN (50 μM) for 1 h. qRT-PCR analysis IL-6, IL-β and IL-27 mRNA expression level. D ~ F WT and AhR-KO peritoneal macrophages treated with LPS (10 µg/mL) for 4 h after pretreated with PUN (50 μM) for 1 h. qRT-PCR analysis AhR, IL-6 and IL-β mRNA expression level. G ~ I WT and AhR-KO peritoneal macrophages treated with Listeria monocytogenes for 1 h after pretreated with PUN (50 μM) for 1 h. G Results of intracellular bacterial plating. H Statistical results of bacterial loads. I Amount of LDH released from macrophages infected with Listeria. The experiment was repeated three times. The data are shown as mean ± SEM. Combined results are compared using one-way ANOVA test or t test. N.S P> 0.05, P < 0.05, ∗P < 0.01
Discussion
At present, AhR ligands can be broken down into endogenous and exogenous forms [4]. Endogenous ligands include those produced by the human body or gut microbiota, while exogenous ligands come from dietary compounds, environmental contaminants, drugs, and synthetic compounds. AhR activation by varying ligand classes results in context-specific outcomes. Natural plant compounds are preferred to synthetic pharmaceuticals due to their lower toxicity, higher bioavailability, and low cost. Polyphenols, abundant in plants, exert potent antioxidant and anti-inflammatory effects for disease prevention and health maintenance [36]. Polyphenolic compounds are AhR ligands belonging to a class of dietary compounds, including the currently known resveratrol, quercetin, and curcumin [24]. Resveratrol, a class of phytochemicals found particularly in certain plants when exposed to stress or injury, has been shown to be an antagonist of AhR [37]. Quercetin, a natural flavonoid, acts as an AhR agonist [37] and regulates CYP1A1 gene expression [24, 38]. Curcumin is derived from the plant Curcuma longa, and is a yellow spice and coloring agent that activates AhR [37]. In this study, we found that PUN, a polyphenol compound derived from pomegranate peel extract, was not an agonist or inhibitor of AhR like other polyphenol compounds, but rather a selective modulator. Previous studies have classified AhR ligands as agonists or antagonists based primarily on their ability to promote or inhibit AhR nuclear translocation. Recently, a third class of AhR ligands has been discovered [39, 40]. These compounds are known as selective AhR modulators (SAhRMs), displaying anti-inflammatory activity that is mainly mediated by nongenomic signaling pathways [12, 13]. The selected synthetic compound SGA360, as an SAhRM, significantly suppresses ear swelling and multiple inflammatory gene expression in mice [41], and inhibits the acute inflammatory response of macrophages, thus alleviating arthritis and reducing the mortality of endotoxic shock in mice [42]. Our results demonstrated that PUN enhanced AhR expression but did not promote AhR nuclear transport or gene transcription in macrophages. In addition, PUN inhibited macrophage inflammation and promoted bactericidal activity in an AhR-dependent manner. Therefore, given the regulation of AhR and its signaling pathway by PUN, we concluded that PUN could be defined as an SAhRM like SGA360.
Although ligand binding is crucial for the activation of AhR, the amount of AhR expression determines ligand activation and subsequent nongenomic effects. AhR knockout mice have increased sensitivity to septic shock and higher levels of inflammatory cytokines in AhR-deficient macrophages after LPS stimulation [43]. AhR is compensatively elevated in LPS-stimulated macrophages through the NF-κB pathway, resulting in inhibition of macrophage inflammatory response [9]. Expression of AhR in microglia is upregulated during ischemia, triggering neuroinflammation that subsequently causes vasogenic edema [44]. The metabolites of 1-methoxypyrene [45] and L-kynurenine [46] dose-dependently activate the AhR pathway, thus mediating different biological effects. However, the effects of polyphenolic compounds on AhR expression remain poorly understood. This study found that treatment with resveratrol, quercetin, and curcumin alone or in combination with LPS did not show any significant changes in AhR expression. We found for the first time that PUN, which is a polyphenol compound, upregulated the expression of AhR either alone or in combination with LPS, and combination treatment was more effective. Therefore, the discovery of PUN as a selective modulator that upregulated AhR expression has implications for the treatment of diseases targeted by AhR.
To investigate the molecular mechanism of upregulation of AhR by PUN, the results of transcriptome sequencing suggested that it is related to NF-κB, MAPK, and PI3K/AKT signaling pathways. Our team previously reported that LPS increased AhR expression in macrophages through the NF-κB signaling pathway [9], but Xu et al. demonstrated that PUN was capable of inhibition of NF-κB activation [16]. We also found that the NF-κB inhibitor PDTC did not reverse the upregulatory effect of PUN on AhR. These data indicate that the NF-κB signaling pathway is not involved in PUN-mediated AhR expression. Regarding the MAPK pathway, our results were consistent with previous studies [16], in which PUN inhibited the phosphorylation of EKR1/2 in LPS-induced macrophages. Surprisingly, MEK (U0126 and PD98059) and ERK (SCH772984) inhibitors reversed upregulation of AhR by PUN, suggesting that additional transcription factors might be involved in PUN-induced upregulation of AhR. Upon screening, we found that PUN promoted LPS-induced phosphorylation of Ser221, Thr359, Ser380, and Thr573 of p90RSK in macrophages. The p90RSK inhibitor LJH685, which was designed to bind to the N-terminal ATP domain of p90RSK kinase, has been shown to inhibit PUN-induced upregulation of AhR expression in macrophages but not LPS-induced AhR expression. The above effect was confirmed in macrophages treated with p90RSK siRNA. Activation of this key molecule, p90RSK, is essential for PUN-induced AhR expression in macrophages. As far as we know, this is the first study to demonstrate the involvement of p90RSK in the regulation of AhR expression. The p90RSK protein kinases, which include p90RSK1–4, belong to the highly conserved AGC kinase family. These kinases regulate various cellular processes and are activated by the Ras/ERK signaling cascade [18, 47]. Interestingly, in Fig. 5D and E, we found that LPS-induced levels of p-p90RSK(Thr573) and p-p90RSK(Ser380) increased more after treatment with the p90RSK inhibitor LJH685, as well as the expression level of AhR. p90RSK contains C-terminal and N-terminal kinase domains [48], and all isoforms have the four crucial phosphorylation sites at Ser221, Ser363, Ser380, and Thr573 [18]. The activation process of p90RSK is achieved through a sequential phosphorylation cascade at Thr573 and Ser221, respectively, by ERK and PDK1 [49]. After complete activation of p90RSK, Ser737 located on the KIM motifs exhibits self phosphorylation, resulted in ERK separation from Thr573 site of p90RSK. Therefore, a negative feedback loop is formed to prevent the continued activation of p90RSK. LJH685 is an N-terminal inhibitor of p90RSK kinase. It is possible that LJH685 prevented the negative feedback loop of p90RSK, which resulted in the sustained Thr573 and Ser380 activation of p90RSK and the subsequent elevation of AhR. Purified p90RSK protein can be fully activated by phosphorylated ERK and phosphorylated PDK1 in vitro [50]. Our research demonstrated that PDK1 (GSK2334470), MEK (U0126), and ERK (SCH772984) inhibitors effectively suppressed PUN-mediated AhR expression, by blocking phosphorylation of p90RSK induced by LPS and PUN. In alignment with earlier studies [47, 51], we confirmed that PDK1 and MEK/ERK synergistically fully activated full-length p90RSK, thereby contributing to the enhancement effect of AhR.
Previous studies focused on the regulation of the AP-1 signaling pathway by AhR, influencing the expression of various members of the AP-1 family in keratinocytes [20]. On the one hand, AhR ligand indolyl sulfate activated AhR and then upregulated E-selectin expression in vascular endothelium by promoting AP-1 transcriptional activity [22]. On the other hand, TCDD-activated AhR inhibited AP-1 protein expression in B cells [52]. However, there is evidence that AP-1 might also be associated with AhR expression. AP-1 upregulates TH17/ILC3 genes, including AhR, in lymphoma cells, so AP-1 inhibition leads to AhR reduction [29]. As indicated in the JASPAR database [30], we proposed that the binding checkpoint of the AP-1 family member c-fos on the AhR promoter was a crucial regulatory mechanism. Our hypothesis was verified through experimental evidence, which indicated that AP-1 played a key role in regulating AhR expression by PUN in LPS-induced macrophages. Finally, we proposed for the first time a new synergistic model to upregulate AhR expression. The partial activation of p90RSK by the LPS-activated MEK/ERK signaling pathway was the basis for its full activation. PUN promoted the full activation of p90RSK through the PDK1 pathway, and then synergistically enhanced c-fos-mediated AhR expression with LPS (Fig. 8).
A new synergistic model of AhR up-regulated by PUN against inflammation and infection in inflammatory macrophages. The activation of p90RSK served as a critical checkpoint for AhR upregulation by PUN and LPS, which induced AhR upregulation by activating MEK/ERK and PDK1 pathways that ultimately targeted c-fos. The partial activation of p90RSK by LPS-activated MEK-ERK signaling pathway was the basis for its full activation. PUN promoted the full activation of p90RSK through PDK1 pathway, and then synergistically enhanced c-fos mediated AhR expression with LPS. Furthermore, AhR up-regulated by PUN could inhibit macrophage inflammation and promote its bactericidal activity
AhR serves as a critical regulator of both inflammatory responses and microbial defense [6, 8, 53]. AhR is intricately connected to the functional regulation of macrophages. In particular, activation of AhR influences the M1/M2 polarization of macrophages [54]. In inflammatory macrophages, AhR decreases the expression of IL-6, tumor necrosis factor-α, IL-12p40, and IL-1β [25, 43, 55], and promotes IL-10 expression in a ligand-dependent or nondependent manner [9, 56]. Our previous study also showed that the increased level of AhR in post-traumatic macrophages substantially decreased the production of inflammatory cytokines [57]. The present study showed that the AhR was a vital regulator in controlling macrophage response and its expression within macrophages was critical for maintaining homeostasis and during inflammatory responses. Nevertheless, although there are reports of anti-inflammatory drugs that target AhR, additional research is needed to discover more potent natural regulators.
Previous research showed that PUN treatment effectively combated inflammation in RAW264.7 cells exposed to LPS by reducing Toll-like-receptor-4-related MAPK and NF-κB activation and interfering with the FoxO3a/autophagy signaling pathway [16, 58]. PUN is also beneficial in minimizing pathological inflammation by suppressing M1 phenotype polarization and pyroptosis [59]. We demonstrated that PUN inhibited the production of proinflammatory cytokines IL-6, IL-1β, and IL-27 in a partially AhR-dependent manner. In AhR-inhibitor-pretreated or AhR knockout macrophages, the inhibitory effect of PUN on inflammatory cytokines was partially reversed. Our results demonstrated the anti-inflammatory mechanism of PUN, suggesting that PUN also regulates expression of AhR, which might further inhibit the inflammatory response of macrophages through a nongenomic mechanism of AhR. In macrophages, AhR upregulation by PUN played a crucial role in modulating the secretory function of these cells.
Recent evidence suggests that AhR and its pathway play a crucial role in combating common bacterial infections. TCDD improves survival and reduces lung bacterial load in mice infected with Streptococcus pneumoniae by activating AhR [60, 61]. In the Salmonella Typhimurium infection model in mice, sublethal doses of LPS enhanced resistance to infection by activating AhR [5]. MOTS-c [62], benzothiazole [63], coal tar [64], and other substances exerted an AhR-dependent killing or inhibitory effect on Staphylococcus aureus. AhR ligands tryptophan and Lactobacillus plantarum increased intestinal AhR level and ROS generation, and improved alveolar macrophage phagocytosis of drug-resistant Pseudomonas aeruginosa [65]. Intestinal microbiota-produced indole-3-propionic acid activated AhR and promoted macrophage phagocytosis, reducing bacterial burden in mice [66]. The above results highlight the critical function of AhR in combating extracellular bacteria, while it is also key in combatting intracellular bacteria. For instance, AhR also participated in macrophage infection by Mycobacterium tuberculosis, triggering innate immune response against the bacteria, and upregulating IL-23 and IL-22 production [67]. L. monocytogenes is a Gram-positive, facultative intracellular bacterium that causes severe illness and is commonly studied as a model intracellular pathogen. It is typically engulfed by macrophages, which are essential for L. monocytogenes killing [34]. Kimura et al. showed that AhR protected against macrophage cell death and promoted ROS production for L. monocytogenes confinement and clearance. Selective AhR-ligand activation may be ideal for treatment of listeriosis. Our group found that AhR expression in macrophages correlated with L. monocytogenes clearance, and that PUN enhanced macrophage clearance of L. monocytogenes and inhibited macrophage death in an AhR-dependent manner. Our findings confirmed previous studies and underscored the unique role of SAhRMs in regulating biological functions such as AhR bactericidal activity. The aforementioned research suggested that AhR served as a significant factor in the treatment of pathogenic infections and the regulation of inflammatory reactions.
The current study had several limitations that need to be considered. The impact of PUN on AhR was exclusively observed in macrophages. Future research will explore the potential of PUN in regulating AhR activity in other cells, including T cells and dendritic cells. Our investigation concentrated on understanding the transcriptional mechanisms used by PUN to augment AhR expression, but the question of whether PUN also influences AhR protein degradation remains to be answered. Moreover, the upstream mechanism of upregulation of AhR by PUN, particularly which receptor mediates this effect, remains to be studied.
Conclusions
We found for the first time that PUN, screened from polyphenols, upregulated AhR expression and mainly affected its non-genomic pathway, providing a new natural regulator for regulating AhR expression in cells. In addition, c-fos mediated the transcriptional regulation of AhR in macrophages. It was confirmed that activation of p90RSK served as a critical checkpoint for AhR upregulation by PUN and LPS, which induced AhR upregulation by activating the MEK/ERK and PDK1 pathways that ultimately targeted c-fos. Furthermore, AhR upregulated by PUN inhibited macrophage inflammation and promoted bactericidal activity (Fig. 8). PUN as a selective regulator of AhR has potential application in the treatment of various diseases targeting AhR.
Availability of data and materials
No datasets were generated or analysed during the current study.
References
Rudd KE, Johnson SC, Agesa KM, Shackelford KA, Tsoi D, Kievlan DR, Colombara DV, Ikuta KS, Kissoon N, Finfer S, et al. Global, regional, and national sepsis incidence and mortality, 1990–2017: analysis for the Global Burden of Disease Study. Lancet. 2020;395:200–11.
Steinhagen F, Schmidt SV, Schewe JC, Peukert K, Klinman DM, Bode C. Immunotherapy in sepsis - brake or accelerate? Pharmacol Ther. 2020;208:107476.
Stockinger B. Shah KA-O, Wincent E: AHR in the intestinal microenvironment: safeguarding barrier function. Nat Rev Gastroenterol Hepatol. 2021;18(8):559–70.
Sládeková L, Mani S, Dvořák Z. Ligands and agonists of the aryl hydrocarbon receptor AhR: Facts and myths. Biochem Pharmacol. 2023;213:115626.
Bessede A, Gargaro M, Pallotta MT, Matino D, Servillo G, Brunacci C, Bicciato S, Mazza EM, Macchiarulo A, Vacca C, et al. Aryl hydrocarbon receptor control of a disease tolerance defence pathway. Nature. 2014;511:184–90.
Moura-Alves PA-O, Puyskens AA-O, Stinn AA-O, Klemm M, Guhlich-Bornhof UA-O, Dorhoi A, Furkert J, Kreuchwig A, Protze JA-O, Lozza LA-O, et al. Host monitoring of quorum sensing during Pseudomonas aeruginosa infection. Science. 2019;366:eaaw1629.
Moura-Alves P, Faé K, Houthuys E, Dorhoi A, Kreuchwig A, Furkert J, Barison N, Diehl A, Munder A, Constant P, et al. AhR sensing of bacterial pigments regulates antibacterial defence. Nature. 2014;512:387–92.
Shi JA-O, Du TA-O, Wang JA-O, Tang CA-O, Lei M, Yu W, Yang Y, Ma Y, Huang P, Chen H, et al. Aryl hydrocarbon receptor is a proviral host factor and a candidate pan-SARS-CoV-2 therapeutic target. Sci Adv. 2023;9:eadf0211.
Zhu J, Luo L, Tian L, Yin S, Ma X, Cheng S, Tang W, Yu J, Ma W, Zhou X, et al. Aryl hydrocarbon receptor promotes IL-10 expression in inflammatory macrophages through Src-STAT3 signaling pathway. Front Immunol. 2018;9:2033.
Larigot L, Juricek L, Dairou J, Coumoul X. AhR signaling pathways and regulatory functions. Biochim Open. 2018;7:1–9.
Bock KW. Aryl hydrocarbon receptor (AHR)-mediated inflammation and resolution: non-genomic and genomic signaling. Biochem Pharmacol. 2020;182:114220.
Safe S, Jin UA-O, Park H, Chapkin RA-O, Jayaraman A. Aryl Hydrocarbon Receptor (AHR) Ligands as Selective AHR Modulators (SAhRMs). Int J Mol Sci. 2020;21:6654.
Dolciami D, Ballarotto M, Gargaro M, López-Cara LC, Fallarino F, Macchiarulo A. Targeting Aryl hydrocarbon receptor for next-generation immunotherapies: Selective modulators (SAhRMs) versus rapidly metabolized ligands (RMAhRLs). Eur J Med Chem. 2020;185:111842.
Venusova E, Kolesarova A. Horky PA-OX, Slama PA-OX: physiological and immune functions of punicalagin. Nutrients. 2021;13:2150.
Pirzadeh M, Caporaso N, Rauf A, Shariati MA, Yessimbekov Z, Khan MU, Imran M, Mubarak MS. Pomegranate as a source of bioactive constituents: a review on their characterization, properties and applications. Crit Rev Food Sci Nutr. 2020;61:982–99.
Xu X, Yin P, Wan C, Chong X, Liu M, Cheng P, Chen J, Liu F, Xu J. Punicalagin inhibits inflammation in LPS-induced RAW264.7 macrophages via the suppression of TLR4-mediated MAPKs and NF-κB activation. Inflammation. 2014;37:956–65.
Xu J, Cao K, Liu X, Zhao L, Feng ZA-O, Liu J. Punicalagin regulates signaling pathways in inflammation-associated chronic diseases. Antioxidants (Basel). 2021;11:29.
Romeo Y, Zhang X, Roux PP. Regulation and function of the RSK family of protein kinases. Biochem J. 2012;441:553–69.
Vázquez-Gómez G, Rocha-Zavaleta L, Rodríguez-Sosa M, Petrosyan P, Rubio-Lightbourn J. Benzo[a]pyrene activates an AhR/Src/ERK axis that contributes to CYP1A1 induction and stable DNA adducts formation in lung cells. Toxicol Lett. 2018;289:54–62.
Di Meglio P, Duarte JH, Ahlfors H, Owens ND, Li Y, Villanova F, Tosi I, Hirota K, Nestle FO, Mrowietz U, et al. Activation of the aryl hydrocarbon receptor dampens the severity of inflammatory skin conditions. Immunity. 2014;40:989–1001.
Morales-Bárcenas R, Sánchez-Pérez Y, Santibáñez-Andrade M, Chirino YI, Soto-Reyes E, García-Cuellar CM. Airborne particulate matter (PM(10)) induces cell invasion through Aryl Hydrocarbon Receptor and Activator Protein 1 (AP-1) pathway deregulation in A549 lung epithelial cells. Mol Biol Rep. 2023;50:107–19.
Ito S, Osaka M, Edamatsu T, Itoh Y, Yoshida M. Crucial Role of the Aryl Hydrocarbon Receptor (AhR) in Indoxyl Sulfate-Induced Vascular Inflammation. J Atheroscler Thromb. 2016;23:960–75.
Jiang Y, Xiao H, Sun L, Zhang Y, Liu S, Luo B. LMP2A suppresses the role of AHR pathway through ERK signal pathway in EBV-associated gastric cancer. Virus Res. 2021;297:198399.
Mohammadi-Bardbori A, Bengtsson J, Rannug U, Rannug A, Wincent E. Quercetin, resveratrol, and curcumin are indirect activators of the aryl hydrocarbon receptor (AHR). Chem Res Toxicol. 2012;25:1878–84.
Kimura A, Naka T, Nakahama T, Chinen I, Masuda K, Nohara K, Fujii-Kuriyama Y, Kishimoto T. Aryl hydrocarbon receptor in combination with Stat1 regulates LPS-induced inflammatory responses. J Exp Med. 2009;206:2027–35.
Schmidt JR, Haupt J, Riemschneider S, Kämpf C, Löffler D, Blumert C, Reiche K, Koehl U, Kalkhof S, Lehmann J. Transcriptomic signatures reveal a shift towards an anti-inflammatory gene expression profile but also the induction of type I and type II interferon signaling networks through aryl hydrocarbon receptor activation in murine macrophages. Front Immunol. 2023;14:1156493.
Vogel CF, Khan EM, Leung PS, Gershwin ME, Chang WW, Wu D, Haarmann-Stemmann T, Hoffmann A, Denison MS. Cross-talk between aryl hydrocarbon receptor and the inflammatory response: a role for nuclear factor-κB. J Biol Chem. 2014;289:1866–75.
Schmidt JV, Carver LA, Bradfield CA. Molecular characterization of the murine Ahr gene. Organization, promoter analysis, and chromosomal assignment. J Biol Chem. 1993;268:22203–9.
Schleussner N, Merkel O, Costanza MA-O, Liang HC, Hummel F, Romagnani C, Durek P, Anagnostopoulos I, Hummel M, Jöhrens K, et al. The AP-1-BATF and -BATF3 module is essential for growth, survival and TH17/ILC3 skewing of anaplastic large cell lymphoma. Leukemia. 2018;32:994–2007.
Castro-Mondragon JA-OX, Riudavets-Puig RA-O, Rauluseviciute IA-O, Lemma RA-O, Turchi LA-O, Blanc-Mathieu RA-O, Lucas JA-O, Boddie P, Khan AA-O, Manosalva Pérez NA-O, et al. JASPAR 2022: the 9th release of the open-access database of transcription factor binding profiles. Nucleic Acids Res. 2022;2022(50):D165–73.
Yoo SM, Lee CJ, An HJ, Lee JY, Lee HA-O, Kang HA-O, Cho SJ, Kim SM, Park J, Kim DA-O, Cho YY. RSK2-mediated ELK3 activation enhances cell transformation and breast cancer cell growth by regulation of c-fos promoter activity. Int J Mol Sci. 1994;2019:20.
Cho YY. Molecular targeting of ERKs/RSK2 signaling in cancers. Curr Pharm Des. 2017;23:4247–58.
Richards SA, Fu J, Romanelli A, Shimamura A, Blenis J. Ribosomal S6 kinase 1 (RSK1) activation requires signals dependent on and independent of the MAP kinase ERK. Curr Biol. 1999;9:810–20.
Kimura A, Abe H, Tsuruta S, Chiba S, Fujii-Kuriyama Y, Sekiya T, Morita R, Yoshimura A. Aryl hydrocarbon receptor protects against bacterial infection by promoting macrophage survival and reactive oxygen species production. Int Immunol. 2014;26:209–20.
Lamorte S, Shinde R, McGaha TL. Nuclear receptors, the aryl hydrocarbon receptor, and macrophage function. Mol Aspects Med. 2021;78:100942.
Iqbal IA-O, Wilairatana PA-O, Saqib FA-O, Nasir B, Wahid MA-O, Latif MF, Iqbal A, Naz R, Mubarak MA-O. Plant polyphenols and their potential benefits on cardiovascular health: a review. Molecules. 2023;28:6403.
Abudahab S, Price ET, Dozmorov MG, Deshpande LS, McClay JL. The aryl hydrocarbon receptor, epigenetics and the aging process. J Nutr Health Aging. 2023;27:291–300.
Satake K, Ishii T, Morikawa T, Sakamoto T, Nishii YA-O. Quercetin reduces the development of 2,3,7,8-tetrachlorodibenzo-p-dioxin-induced cleft palate in mice by suppressing CYP1A1 via the Aryl hydrocarbon receptor. Nutrients. 2022;14:2448.
Murray IA, Flaveny CA, Chiaro CR, Sharma AK, Tanos RS, Schroeder JC, Amin SG, Bisson WH, Kolluri SK, Perdew GH. Suppression of cytokine-mediated complement factor gene expression through selective activation of the Ah receptor with 3’,4’-dimethoxy-α-naphthoflavone. Mol Pharmacol. 2011;79:508–19.
Murray IA, Morales JI, Flaveny CA, Dinatale BC, Chiaro C, Gowdahalli K, Gowdahalli K, Amin S, Perdew GH. Evidence for ligand-mediated selective modulation of aryl hydrocarbon receptor activity. Mol Pharmacol. 2010;77:247–54.
Murray IA, Krishnegowda G, DiNatale BC, Flaveny C, Chiaro C, Lin JM, Sharma AK, Amin S, Perdew GH. Development of a selective modulator of aryl hydrocarbon (Ah) receptor activity that exhibits anti-inflammatory properties. Chem Res Toxicol. 2010;23:955–66.
Muku GE, Lahoti TS, Murray IA, Podolsky MA, Smith KJ, Hubbard TD, Kuzu G, Gowda K, Amin SG, Perdew GH. Ligand-mediated cytoplasmic retention of the Ah receptor inhibits macrophage-mediated acute inflammatory responses. Lab Invest. 2017;97:1471–87.
Sekine H, Mimura J, Oshima M, Okawa H, Kanno J, Igarashi K, Gonzalez FJ, Ikuta T, Kawajiri K, Fujii-Kuriyama Y. Hypersensitivity of aryl hydrocarbon receptor-deficient mice to lipopolysaccharide-induced septic shock. Mol Cell Biol. 2009;29:6391–400.
Tanaka M, Fujikawa M, Oguro A, Itoh K, Vogel CFA, Ishihara Y. Involvement of the microglial Aryl hydrocarbon receptor in neuroinflammation and vasogenic edema after ischemic stroke. Cells. 2021;10:718.
Cao G, Miao H, Wang YN, Chen DQ, Wu XQ, Chen L, Guo Y, Zou L, Vaziri ND, Li P, Zhao YY. Intrarenal 1-methoxypyrene, an aryl hydrocarbon receptor agonist, mediates progressive tubulointerstitial fibrosis in mice. Acta Pharmacol Sin. 2022;43:2929–45.
Jia Y, Guo J, Zhao Y, Zhang Z, Shi L, Fang Y, Wu D, Wu L, Chang C. AHR signaling pathway reshapes the metabolism of AML/MDS cells and potentially leads to cytarabine resistance. Acta Biochim Biophys Sin (Shanghai). 2021;53:492–500.
Koutsougianni F, Alexopoulou D, Uvez A, Lamprianidou A, Sereti E, Tsimplouli C, Ilkay Armutak E, Dimas K. P90 ribosomal S6 kinases: A bona fide target for novel targeted anticancer therapies? Biochem Pharmacol. 2023;210:115488.
Fruergaard MU, Nielsen CJF, Kjeldsen CAO, Iversen LA-O, Andersen JL, Nissen PA-O. Activation and inhibition of the C-terminal kinase domain of p90 ribosomal S6 kinases. Life Sci Alliance. 2023;20:e202201425.
Takata TA-O, Araki S, Tsuchiya YA-O, Watanabe YA-O. Oxidative stress orchestrates MAPK and nitric-oxide synthase signal. Int J Mol Sci. 2020;21:8750.
Utepbergenov D, Hennig PA-OX, Derewenda U, Artamonov MV, Somlyo AV, Derewenda ZS. Bacterial Expression, Purification and In Vitro Phosphorylation of Full-Length Ribosomal S6 Kinase 2 (RSK2). PLoS One. 2016;11:e0164343.
Watanabe D, Nogami A, Okada K, Akiyama H, Umezawa Y, Miura O. FLT3-ITD Activates RSK1 to Enhance Proliferation and Survival of AML Cells by Activating mTORC1 and eIF4B Cooperatively with PIM or PI3K and by Inhibiting Bad and BIM. Cancers (Basel). 2019;11:1827.
Zhang Q, Kline DE, Bhattacharya S, Crawford RB, Conolly RB, Thomas RS, Andersen ME, Kaminski NE. All-or-none suppression of B cell terminal differentiation by environmental contaminant 2,3,7,8-tetrachlorodibenzo-p-dioxin. Toxicol Appl Pharmacol. 2013;268:17–26.
Pinto CJG, Ávila-Gálvez M, Lian Y, Moura-Alves P, Nunes Dos Santos C. Targeting the aryl hydrocarbon receptor by gut phenolic metabolites: a strategy towards gut inflammation. Redox Biol. 2023;61:102622.
Yang X, Liu H, Ye T, Duan C, Lv P, Wu X, Liu J, Jiang K, Lu H, Yang H, et al. AhR activation attenuates calcium oxalate nephrocalcinosis by diminishing M1 macrophage polarization and promoting M2 macrophage polarization. Theranostics. 2020;10:12011–25.
Wakamatsu T, Yamamoto S, Ito T, Sato Y, Matsuo K, Takahashi Y, Kaneko Y, Goto S, Kazama JJ, Gejyo F, Narita I. Indoxyl sulfate promotes macrophage IL-1β production by activating Aryl hydrocarbon receptor/NF-κ/MAPK cascades, but the NLRP3 inflammasome was not activated. Toxins (Basel). 2018;10:124.
Fueldner C, Kohlschmidt J, Riemschneider S, Schulze F, Zoldan K, Esser C, Hauschildt S, Lehmann J. Benzo(a)pyrene attenuates the pattern-recognition-receptor induced proinflammatory phenotype of murine macrophages by inducing IL-10 expression in an aryl hydrocarbon receptor-dependent manner. Toxicology. 2018;409:80–90.
Kuang TY, Yin SQ, Dai WH, Luo L, Chen T, Liang XH, Wang RX, Liang HP, Zhu JY. Effects of enhancing the expression of aryl hydrocarbon receptor in post-traumatic mice macrophages on the inflammatory cytokine level and bactericidal ability. Zhonghua Shao Shang Za Zhi. 2023;39:633–40.
Cao Y, Chen J, Ren G, Zhang Y, Tan X, Yang L. Punicalagin prevents inflammation in LPS-induced RAW264.7 macrophages by inhibiting FoxO3a/Autophagy signaling pathway. Nutrients. 2019;11:2794.
Ge G, Bai J, Wang Q, Liang X, Tao H, Chen H, Wei M, Niu J, Yang H, Xu Y, et al. Punicalagin ameliorates collagen-induced arthritis by downregulating M1 macrophage and pyroptosis via NF-κB signaling pathway. Sci China Life Sci. 2022;65:588–603.
Vorderstrasse BA, Lawrence BP. Protection against lethal challenge with Streptococcus pneumoniae is conferred by aryl hydrocarbon receptor activation but is not associated with an enhanced inflammatory response. Infect Immun. 2006;74:5679–86.
Wang T, Wyrick KL, Pecka MR, Wills TB, Vorderstrasse BA. Mechanistic exploration of AhR-mediated host protection against Streptococcus pneumoniae infection. Int Immunopharmacol. 2012;13:490–8.
Zhai D, Ye Z, Jiang Y, Xu C, Ruan B, Yang Y, Lei X, Xiang A, Lu H, Zhu Z, et al. MOTS-c peptide increases survival and decreases bacterial load in mice infected with MRSA. Mol Immunol. 2017;92:151–60.
Goya-Jorge E, Abdmouleh F, Carpio LE, Giner RM, Sylla-Iyarreta Veitía M. Discovery of 2-aryl and 2-pyridinylbenzothiazoles endowed with antimicrobial and aryl hydrocarbon receptor agonistic activities. Eur J Pharm Sci. 2020;51:105386.
Smits JPH, Ederveen THA, Rikken G, van den Brink NJM, van Vlijmen-Willems I, Boekhorst J, Kamsteeg M, Schalkwijk J, van Hijum S, Zeeuwen P, van den Bogaard EH. Targeting the cutaneous microbiota in atopic dermatitis by coal tar via AHR-dependent induction of antimicrobial peptides. J Invest Dermatol. 2020;140:415–24.
Tsay TB, Chen PH. Chen LA-O: Aryl hydrocarbon receptor ligands enhance lung immunity through intestinal IKKβ pathways. J Transl Med. 2019;17:304.
Huang ZB, Hu Z, Lu CX, Luo SD, Chen Y, Zhou ZP, Hu JJ, Zhang FL, Deng F, Liu KX. Gut microbiota-derived indole 3-propionic acid partially activates aryl hydrocarbon receptor to promote macrophage phagocytosis and attenuate septic injury. Front Cell Infect Microbiol. 2022;12:1015386.
Memari BA-O, Bouttier M, Dimitrov V, Ouellette M, Behr MA, Fritz JH, White JH. Engagement of the Aryl hydrocarbon receptor in mycobacterium tuberculosis-infected macrophages has pleiotropic effects on innate immune signaling. J Immunol. 2015;195:4479–91.
Acknowledgements
This work was supported by grants from the Basic research project (2022-JCJQ, 2021-JCJQ), National Natural Science Foundation of China (82202422), General Program of Chongqing Natural Science Foundation (cstc2021jcyj-msxmX0234), Special Project of Science and Technology Innovation Capacity Promotion of Army Medical University (2023XJS47), General Program of Hainan Natural Science Foundation (821MS41), Hainan Clinical Medical Research Center Project (LCYX202205), Hainan Medical College Research and Cultivation Fund Project (XPY200032). We thank International Science Editing (http://www.internationalscienceediting.com) for editing this manuscript.
Funding
Basic research project (2022-JCJQ, 2021-JCJQ), National Natural Science Foundation of China (82202422), General Program of Chongqing Natural Science Foundation (cstc2021jcyj-msxmX0234), Special Project of Science and Technology Innovation Capacity Promotion of Army Medical University (2023XJS47), General Program of Hainan Natural Science Foundation (821MS41), Hainan Clinical Medical Research Center Project (LCYX202205), Hainan Medical College Research and Cultivation Fund Project (XPY200032).
Author information
Authors and Affiliations
Contributions
Weihong Dai and Junyu Zhu performed most of the experiments, analyzed the research data and wrote the manuscript. Shuangqin Yin helped with transcriptome and immunofluorescence analysis. Fangjie Wang, Dongmei He and Xue Yang assisted in mice feeding and flow cytometry analysis. Tianyin Kuang, Wei Li and Chao Fan performed bactericidal experiment. Wenyuan Kang and He Jin performed affinity prediction and molecular docking. Caihong Yun, Wanqi Tang and Lizhu Liu assisted with qRT-PCR assay. Fei Wang, Li Luo and Shengxiang Ao helped with western blotting analysis. Hongyan Xiao and Jing Zhou assisted in siRNA and inhibitor experiments. Junyu Zhu, Huaping Liang and Rixing Wang designed experiments, interpreted data and supervised the work.
Corresponding authors
Ethics declarations
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Supplementary Information
12964_2024_1847_MOESM1_ESM.pdf
Additional file 1: Figure S1. (A) Molecular structural formula of Punicalagin (PUN), Resveratrol (RES), Quercetin (QUE) and Curcumin (CUR). (B, C) Peritoneal macrophages cells were treated with LPS (10 µg/mL) for 8 h after pretreated with PUN (50 μM) for 1 h. Cells lysates used for western blotting analysis with ARNT and CYP1A1 protein expression. The experiment was repeated three times. Figure S2. (A~E) Peritoneal macrophages were pretreated with MEK inhibitors U0126 (20 μM) or PD98059 (10 μM) , ERK inhibitors SCH772984 (1 μM) for 1 h and then pretreated with PUN (50 μM) for 1 h, and then treated with LPS (10 µg/mL) for 0.5 h, 1 h or 12 h, cells lysates subjected to western blotting analysis with AhR, ERK, p-ERK, c-jun, p-c-jun, p-MSK1 protein expressions. The experiment was repeated three times. Figure S3. (A, B)Peritoneal macrophages were pretreated with MEK inhibitors U0126 (20 μM), ERK inhibitors SCH772984 (1 μM), p90RSK inhibitors LJH685(10 μM) for 1 h and then pretreated with PUN (50 μM) for 1 h, and then treated with LPS (10 µg/mL) for 0.5 h, 1 h or 12 h, cells lysates subjected to western blotting analysis with p90RSK (Thr573, Ser380, Thr359, Ser221), c-fos, p-c-fos(Ser32), c-jun, p-c-jun(Ser63). The experiment was repeated three times. Table S1. Antibodies information of western blotting and Immunofluorescence. Table S2. Gene-specific primer sequences for qRT-PCR. Table S3. Antibodies information of flow cytometry. Table S4. siRNA sequences information of p90RSK. Table S5. AhR and AP-1 sequence information of the consensus recognition sit.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Dai, W., Yin, S., Wang, F. et al. Punicalagin as a novel selective aryl hydrocarbon receptor (AhR) modulator upregulates AhR expression through the PDK1/p90RSK/AP-1 pathway to promote the anti-inflammatory response and bactericidal activity of macrophages. Cell Commun Signal 22, 473 (2024). https://doi.org/10.1186/s12964-024-01847-9
Received:
Accepted:
Published:
DOI: https://doi.org/10.1186/s12964-024-01847-9