1. Introduction
Drug-induced liver injury (DILI) is a predictable event that occurs when an individual is exposed to toxic doses of certain compounds and is emerging as one of the leading causes of liver health in humans [
1]. Acetaminophen (APAP) is a widely used antipyretic analgesic drug worldwide that produces severe hepatotoxicity when overdosed or chronically used [
2]. The hepatotoxicity of APAP is mainly caused by its metabolite N-acetyl-p-benzoquinoimide (NAPQI) during the biotransformation process [
3]. NAPQI accumulation and glutathione (GSH) depletion after excess APAP enters the CYP450 enzyme (CYP2E1, CYP1A2, CYP3A4) pathway results in excessive reactive oxygen species production, thereby triggering oxidative stress. In addition, the accumulation of peroxides and GSH depletion ultimately lead to endoplasmic reticulum stress (ERS), hepatocyte apoptosis, and even hepatic dysfunction [
4].
The pregnane X receptor (
PXR) is a member of a subfamily of nuclear receptors, which are characterized by their ligand-dependency and their ability to modulate the expression of target genes. From a clinical perspective, approximately 50% of drugs undergo metabolism via the activation of
PXR-mediated drug-metabolizing enzymes, specifically cytochrome P450 3A11 (CYP3A11) and cytochrome P450 1A2 (CYP1A2). [
5]. CYP1A2, CYP2E1, and CYP3A11 (the mouse homologue of human CYP3A4) are the most active CYPs catalyzing the generation of toxic NAPQI from APAP in mice, and previous studies consistently show that induction of these enzymes exacerbates APAP hepatotoxicity. In addition, these CYPs can be regulated by the
PXR, an important transcriptional regulator of CYP3A11 and CYP1A2. However, mRNA expression of hepatic CYP3A11 and CYP1A2 in mice lacking
PXR was lower than in the controls [
6]. It indicated that
PXR-deficient mice are less susceptible to APAP-induced liver injury (AILI), which was confirmed by the study of Kristina KW et al. [
7].
ERS triggers complex adaptive or apoptosis-promoting signaling defined as the unfolded protein response (UPR) [
8]. ProteinkinaseR-likeERkinase (PERK) is a protein kinase distributed in the endoplasmic reticulum (ER) membrane [
9]. Released PERK is activated by oligomerization and reverse autophosphorylation, and activated PERK phosphorylates the alpha subunit of translation initiation factor 2 (eIF-2α). In the early stage of the stress response, phosphorylated eIF2α inhibits protein translation and synthesis, reduces the amount of protein folding load in the ER, and thus exerts a protective effect on the cell [
10]. Although UPR activation may maintain cell survival, under conditions of severe or persistent ERS, the adaptive response of the UPR fails to eliminate ERS, and the cell is unable to restore ER homeostasis, ultimately leading to the activation of apoptotic signaling pathways [
11,
12]. With the increase of the time and intensity of the stress response, phosphorylated eIF-2α induces the activation of transcriptional expression of the transcription factor 4 (ATF4), which promotes the apoptosis signaling molecule C/EBP homologous protein (CHOP) expression, which in turn promotes apoptosis, reduces liver function [
9,
13]. Apoptosis is a physiological and pathological stimulation signal of cells to the environment and a death process that responds to orderly changes caused by changes in environmental conditions or moderate damage [
14]. Apoptosis is a strictly controlled process of multiple genes. These genes are very conserved among species, such as the Bcl-2 family and the caspase family [
15]. In addition, oxidative stress mediates apoptosis through ERS [
16]. Studies have shown that excess APAP causes oxidative stress, ERS, apoptosis; whether FA attenuates these problems is a question to be addressed in this study [
17].
Forsythiae fructus, a shrub of the genus
Forsythia in the family
Oleaceae, is commonly used in the treatment of acute febrile colds and lymph node tuberculosis. The extract of
Forsythia suspensa has a protective effect against carbon tetrachloride-induced hepatotoxicity. [
18,
19].
Forsythia has a variety of active ingredients, some of which have been proven to have significant pharmacological activity. Forsythiaside A (FA), a phenylethanoid glycoside isolated from
Forsythiae fructus, exhibits anti-oxidative, anti-inflammatory, and hepatoprotective pharmacological activities [
20]. FA has been demonstrated to be involved in regulating the development of a variety of diseases in the body, including inflammation, viral infections, neurodegeneration, oxidative stress, liver injury, and bacterial infections [
21,
22]. It has been shown that FA plays a protective role in AILI in zebrafish [
20]; however, the therapeutic effect of FA in mice with AILI and its mechanism have not been investigated yet.
Understanding the molecular mechanisms of APAP-induced acute liver injury is fundamental to designing new therapeutic strategies for treating this type of liver injury. We presently report that mice given FA exhibit much less liver injury than WT control mice after APAP overdose treatment, suggesting that FA is highly effective for the treatment of APAP-induced acute liver injury. We further demonstrated that FA ameliorates APAP hepatotoxicity metabolism by inhibiting PXR-mediated CYP1A2 and CYP3A11. Moreover, PXR interference attenuated APAP-induced oxidative stress, ERS, and apoptosis. Ultimately, our study emphasizes that previously undescribed FA alleviates APAP-induced liver dysfunction by inhibiting oxidative stress, ERS, and apoptosis in the liver.
3. Materials and Methods
3.1. Cell Experiments
AML12 cells obtained from Novobio Scientific (Shanghai, China) were incubated in DMEM/F-12 medium with 10% FBS at 37 °C and 5% CO
2 conditions. AML12 cells were inoculated overnight into 6-well plates at a density of 105 cells/well, treated with FA (0–80 μM) for 24 h, and the survival rate of FA was measured using the CCK8 method. To evaluate the therapeutic effects of FA on APAP damage, the cells were pretreated with APAP (5 mM) for 1 h and exposed to FA (20 μΜ) for 24 h. The cell supernatants were collected to detect the levels of oxidative stress and APAP metabolism. And then in order to detect whether FA alleviates APAP damage through PXR, the cells were divided into six groups: normal, APAP, FA, shPXR, FA + APAP, and shPXR + APAP. Short hairpin RNA (shRNA) was purchased from GenePharma (Shanghai, China). Cells were placed in six-well plates and incubated for 24 h before RNA interference. Diluted transfection reagent lip2000 was lightly mixed with shRNA suspension in DMEM (shRNA sequences are detailed in
Table 1). Cells were incubated at 37 °C for 6 h; the medium was changed and incubated for 48 h to confirm the infection rate; the medium was changed, and the infection rate was confirmed by incubation for 48 h. The supernatant and cells were collected for subsequent experimental analysis after treatment of the cells.
3.2. Animals and Treatment
Male C57BL/6 mice (18–22 g, 6 weeks old) were procured from the Lanzhou veterinary research institute, Chinese academy of agricultural sciences (certificate No. SCXK(J) 2011–0007, Lanzhou, China). The animals were raised in cages and provided free access to food and water under standard specific pathogen-free (SPF) conditions. Animal experiments, including sample collection, were performed in accordance with the guidelines of the Ethics Committee of Gansu Agricultural University. All experiments were approved by the Ethics Committee of Gansu Agricultural University, China (ethics approval file No. GSAU-Eth-VMC-2023-006). All mice were housed on a 12 h light and 12 h dark schedule and fed with deionized water and a standard mouse diet at a suitable temperature (22 ± 0.5 °C). In order to investigate the therapeutic effects of FA at different concentrations on APAP-induced liver injury and to compare the liver-protective effects of FA with those of the liver-protectant NAC (N-acetyl-cysteine), the mice were divided into six groups: the normal group, the APAP group, the NAC treatment group, the low-dose FA treatment group (50 mg/kg, LT), the medium-dose FA treatment group (100 mg/kg, MT), and the high-dose FA treatment group (200 mg/kg, HT). In order to investigate the role of PXR in the process of FA treating APAP-induced liver injury, The mice were divided into six groups: normal group, APAP group, FA group, shPXR group, FA + APAP group, and shPXR + APAP group. Liver and serum were collected for subsequent analysis. APAP solution was made fresh in PBS at 20 mg/mL, and C57BL/6 mice were administered a single dose of 400 mg/kg APAP by intraperitoneal injection [
24]. FA solution was prepared in physiological saline, and 100 mg/kg was given by single gavage. Plasmids were injected into mice via the tail vein, and Entranster TM in vivo transfection reagent (Engreen Biosystem Co., Ltd., Beijing, China) was used to deliver plasmids [
25].
3.3. Drugs and Reagents
Forsythoside A (purity > 99.0%) was purchased from Shanghai yuanye Bio-Technology Co., Ltd. (Shanghai, China). NAC was purchased from Beijing Solarbio Science & Technology Co., Ltd. (Beijing, China). APAP was obtained from Shanghai Aladdin Biochemical Technology Co., Ltd (Shanghai, China). DMEM/F-12 and fetal bovine serum (FBS) were obtained from Gibco Life Technologies (Grand Island, NE, USA), fetal bovine serum (FBS). NAPQI enzyme-linked immunosorbent assay (ELISA) kits were obtained from Shanghai huabang Biochemical Technology Co., Ltd. (Shanghai, China). Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) apoptosis detection kit and Reactive oxygen species assay kit were obtained from Beyotime Biotechnology (Shanghai, China). The serum enzyme activities of aspartate aminotransferase (AST), alanine aminotransferase (ALT), superoxide dismutase (SOD), malondialdehyde (MDA), hydrogen peroxide (H2O2) and glutathione (GSH) were measured by the commercial quantitative kits (Nanjing Jiancheng Bioengineering Institute, Nanjing, China).
3.4. Molecular Docking
Molecular docking technology is a method that predicts the interaction mechanism between small molecule ligands and biological macromolecular receptors (such as proteins and nucleic acids) through computational simulation. It is achieved through the following steps: Conformational Search: Generate possible binding conformations of the ligand and the receptor; Energy Evaluation: Calculate the binding free energy of different conformations; Scoring and Ranking: Rank the candidate conformations according to their affinities. Proteins were semiflexibly docked to the receptor, and the molecular docking was visualized using PyMOL 2.5. The docking was performed using the software AutoDock 4.2. The 2D structure of FA was downloaded from PubChem. The three-dimensional protein data bank (PDB) coordinates for PXR (PDB ID: 6TFI) were retrieved from the RCSB Protein Data Bank.
3.5. PXR Promoter Reporter Gene
According to the principles of homologous recombination primer design and the promoter sequence of the mouse PXR gene, primers were designed using the homologous recombination method. The purified PCR products and pGL3-basic were digested with two restriction enzymes and then ligated. The ligated PXR products were transformed into competent cells, which were then inoculated into LB medium for cultivation. After that, positive clone colonies were picked out, and plasmids were extracted for sequencing. After cell culture and transfection, the dual-luciferase activity assay can be used to detect the activation effect of drugs on PXR.ROS detection
The medium was removed, and the cells were washed three times with PBS, followed by incubation with 10 mmol/L DCFH-DA for 20 min at 37 °C. We set the excitation wavelength at 488 nm and the emission wavelength at 525 nm, and directly observed and took photos using a laser confocal microscope.
3.6. Histopathological Examination of Liver
Liver tissue samples were fixed in 4% paraformaldehyde for 72 h. For H&E staining, tissue samples were dehydrated, washed, paraffin-embedded, and sectioned at a thickness of 5 μm. H&E staining was completed according to the manufacturer’s instructions.
3.7. TUNEL Fluorescence Staining
The TdT-UTP barbed end labeling (TUNEL) kit was purchased from Beyotime Biotechnology (Shanghai) Co., Ltd., (Shanghai, China). The TUNEL assay is performed using a one-step TUNEL kit according to the manufacturer’s instructions. In brief, cells were permeabilized with 0.1% Triton X-100 for 5 min and then incubated with TUNEL assay solution for 1 h at 37 °C, protected from light. FITC-labeled TUNEL-positive cells were imaged under a fluorescence microscope using 488 nm excitation and 530 nm emission. Cells with green fluorescence were defined as apoptotic cells.
3.8. Western Blotting (WB)
First, we used RIPA buffer (Solarbio, China) to extract protein from mice liver and cultured cells. Then, protein was separated by SDS-PAGE and transferred onto PVDF membranes (Merck Life Science, Darmstadt, Germany). After that, specific primary antibodies were used to incubate with membranes at 4 °C overnight. The next morning, the membranes were washed for 30 min and incubated with the secondary antibodies at room temperature for 1 h. Protein was visualized by chemiluminescence reagents (YEASRN, Shanghai, China). Band intensities were quantified using Image J 1.54f (National Institutes of Health, Bethesda, MD, USA). In this research, we used GAPDH as the normalization. The primary antibodies and secondary antibodies of Western blot: PXR (Proteintech group, Wuhan, China), GAPDH (Affinity Biosciences, Cincinnati, OH, USA), CYP1A2 (Affinity Biosciences), CYP3A11 (Bioss, Beijing, China), CYP2E1 (Bioss), PERK (Affinity Biosciences), p-PERK (Affinity Biosciences), eIF-2α (Bioss), p-eIF-2α (Affinity Biosciences), CHOP (Proteintech group), Bax (Bioss), Bcl-2 (Affinity Biosciences), Caspase 3 (Abmart, Shanghai, China), Caspase 7 (Bioss), Goat Anti-Mouse IgG H&L/HRP antibody (Bioss), Goat Anti-Rabbit IgG H&L/HRP antibody (Bioss).
3.9. Quantitative Polymerase Chain Reaction (qPCR)
Cells and mouse livers were collected, and total RNA was extracted with TRIzol reagent and reverse transcribed into cDNA using Hiscript II QRT SuperMix for qPCR (Nanjing Vazyme Biotech Co., Ltd., Nanjing, China). PCR amplification was performed in LightCycler 96 (Roche Diagnostics Products (Shanghai) Co., Ltd., Basel, Switzerland) using SYBR qPCR Master Mix. Primers (
Table 2) were synthesized by Sangon Biotech (Shanghai, China) Co., Ltd. and used for PCR. The relative expression levels of
PXR, CYP1A2, CYP3A11, and CYP2E1 mRNA were calculated by the 2
−ΔΔCt method.
3.10. Statistical Analysis
The values were expressed as mean ± standard deviation (SD). One-way analysis of variance (ANOVA) was performed using SPSS Statistics 29. p-values less than 0.05 were considered statistically significant, and values were expressed as mean ± standard deviation (SD).
4. Discussion
PXR, a nuclear receptor and major regulator of drug metabolism, is highly expressed in the liver and plays an integral role in the control of exogenous and endogenous metabolism. As a potentially important therapeutic target, it is important to fully understand the biological and physiological properties of
PXR and its functions. Recently, it has been found that excessive intake of APAP induces aberrant activation of
PXR, which produces pathogenic effects ultimately resulting in liver injury, whereas this phenomenon is significantly attenuated in
PXR knockout mice [
7,
26,
27]. These studies demonstrate that
PXR is an important target for the treatment of APAP liver injury, and
PXR antagonists may be helpful in reducing APAP-induced hepatotoxicity.
Lately, we have observed that a drug monomer, FA, appears to significantly attenuate overdose AILI; however, it is not clear what mechanistic changes within the liver are occurring that lead to recovery from hepatotoxicity, and the molecular signatures involved remain poorly characterized. FA is the main bioactive index constituent of Forsythia, which has significant hepatoprotective, antioxidant, and other pharmacological effects [
28]. In the present research, we confirmed that FA significantly attenuated APAP hepatotoxicity in mice by detecting liver function indexes, oxidative stress indexes, and observing pathological sections. In addition, to explore the optimal therapeutic dose of FA, we conducted investigations using low, medium, and high concentrations of FA, with the NAC treatment group serving as a control. The results demonstrated that 100 mg/kg of FA was the most effective in treating APAP-induced liver injury and outperformed the traditional liver-protectant NAC. This suggests that FA could potentially serve as a new alternative drug for clinically treating liver injury. However, before its clinical application, we need to elucidate the mechanism by which FA treats APAP-induced liver injury.
Since PXR is an important target for the treatment of AILI, we considered whether the mechanism of FA treatment of AILI is related to PXR. We examined PXR levels in APAP liver-injured mice given FA and found that PXR expression was significantly correlated with whether FA was given or not. Next, we examined PXR expression levels in mice after FA administration alone and showed that FA significantly reduced PXR expression. In order to exclude the influence of other factors, we set the shPXR treatment group and FA treatment group as a control, and the results showed that these two groups had the same treatment effect. Combined with the previous studies, we conclude that FA is able to alleviate AILI by inhibiting PXR.
In addition, our results demonstrated that FA was able to affect APAP metabolism through inhibition of PXR, and after PXR was inhibited, CYP1A2 and CYP3A11, two APAP metabolizing enzymes, were inhibited, whereas CYP2E1 was unaltered, which proved that FA did not affect CYP2E1 through PXR. Moreover, the metabolite of APAP, NAPQI, was efficiently removed by FA, which explains that FA affects APAP metabolism via PXR, thereby ameliorating APAP hepatotoxicity.
Some reports have demonstrated that FA can attenuate ischemic brain injury through the ERS signaling pathway, but the mechanism of treating liver injury is not clear, and the present study fills this gap [
29]. After the occurrence of APAP liver injury, the organism will have a series of stress responses, such as ER stress, in order to maintain homeostasis [
30]. Preliminary studies in our laboratory have found that drugs for APAP liver injury can significantly reduce the level of ERS, and in conjunction with the study of
PXR in this experiment, we need to consider whether the alleviation of APAP liver injury caused by FA by inhibiting
PXR is related to ERS. Our studies at the in vitro and in vivo levels in mice by means of WB and immunofluorescence double staining revealed a positive correlation between
PXR and PERK expression in all groups and corresponding changes in eIF2α, ATF4, and CHOP. To exclude the influence of other factors, we set up the shPXR + APAP group as a way to demonstrate that the reduction of
PXR in the APAP model of liver injury can alleviate ER stress, which was confirmed by our data. Previously, we have demonstrated that FA acts as an antagonist of
PXR in the treatment of APAP hepatotoxicity, and the level of ER stress was significantly decreased in the FA-treated model group, and combined with the above results, we conclude that FA regulates ER stress by affecting the level of
PXR, which subsequently attenuates liver injury.
Furthermore, ERS can mediate apoptosis, so does it alter apoptosis in a model group treated with FA? We performed the next step of the study. Regarding ERS, we examined the expression of the PERK-eIF2α-ATF4-CHOP pathway and showed that AILI-triggered ERS induced CHOP. The study demonstrated that CHOP is able to cause apoptosis, but it does not mean that all apoptosis that occurs is caused by the CHOP pathway, so we supplementally examined other apoptotic hallmark factors, such as Bax, Bcl-2, Caspase 3, Caspase 7, and flow cytometry with TUNEL staining, which confirmed that excess APAP did trigger apoptosis. The apoptosis rate was significantly decreased after treatment with FA or shPXR, and we finally concluded that FA alleviated ERS-induced apoptosis by inhibiting
PXR. Previous studies have found that there is a possibility of false positives in gene-level regulation. Therefore, to establish a reliable causal logic, this study designed a rescue experiment. In this experiment, using APAP +
PXR-overexpressing mice as a control, FA could significantly down-regulate the level of oxidative stress. On this basis, the
PXR gene was interfered with to investigate whether it could rescue the damage caused by
PXR overexpression. The results showed that down-regulating
PXR could rescue the liver injury caused by up-regulating
PXR (see
Figure S1 in the
Supplementary Materials), clarifying that the mechanism of FA in treating APAP-induced liver injury is through inhibiting
PXR-mediated oxidative stress and pathological damage.
However, this study still has certain limitations. In the current paper, the mechanistic depth (PXR pathway) has not been fully explored. Although we have preliminarily confirmed that FA can alleviate AILI by inhibiting PXR, the more detailed signaling network downstream of PXR and its interactions with other intracellular signaling pathways remain unclear. Future research will focus on using cutting-edge technologies such as transcriptomics and proteomics to comprehensively analyze the detailed molecular mechanisms of the PXR pathway in AILI. Through transcriptomic analysis, we can gain in-depth insights into the overall changes in gene expression and identify the key genes and signaling pathways regulated by PXR; proteomics will help reveal the changes at the protein level and further clarify the interaction relationships between PXR and other proteins.