Skip to main content
Food Science and Biotechnology logoLink to Food Science and Biotechnology
. 2023 Nov 18;33(8):1939–1946. doi: 10.1007/s10068-023-01440-9

Hepatoprotective effects of paeonol by suppressing hepatic stellate cell activation via inhibition of SMAD2/3 and STAT3 pathways

Hye-Jin Jeong 1, Sooyeon Koo 1, Yeon-Ho Kang 1, Tae Won Kim 1,2, Hye Kyung Kim 1,2,✉,#, Yong Joo Park 1,✉,#
PMCID: PMC11091017  PMID: 38752108

Abstract

Hepatic stellate cell (HSC) activation is a key event in extracellular matrix accumulation, causing hepatic fibrosis. Therefore, identifying chemicals that inhibit HSC activation is an important therapeutic strategy for hepatic fibrosis. The aim of this study was to investigate the therapeutic effects of paeonol on HSC activation. In LX-2 cells, paeonol inhibited the expression of collagen and decreased the expression of HSC activation markers. In mice with thioacetamide-induced liver fibrosis, paeonol treatment decreased the serum levels of aspartate aminotransferase and alanine transaminase and mRNA expression of α-smooth muscle actin, platelet-derived growth factor-β, and connective-tissue growth factor. Investigation of the underlying molecular mechanism of paeonol showed that paeonol inhibits the SMAD2/3 and STAT3 signaling pathways that are important for HSC activation. On the basis of these results, paeonol should be investigated and developed further for hepatic fibrosis treatment.

Supplementary Information

The online version contains supplementary material available at 10.1007/s10068-023-01440-9.

Keywords: Hepatic stellate cell, Collagen accumulation, Liver fibrosis, Paeonol

Introduction

The liver plays a pivotal role in metabolic activities, nutrient storage, and hazardous substance clearance. It is a primary target organ of various foreign toxic chemicals. Liver fibrosis (LF) is a chronic liver disease characterized by the accumulation of extracellular matrix (ECM) proteins including fibronectin and collagen (Bataller and Brenner, 2005). Although the global burden of LF has increased, there are no US Food and Drug Administration-approved drugs for LF treatment (Ginès et al., 2022; Zhang et al., 2021).

The inhibition of hepatic stellate cell (HSC) activation is considered an important therapeutic target for LF. HSCs are vitamin A- and fat-storing cells that exist in the space between hepatocytes and the liver sinusoidal endothelium (Senoo et al., 2010). When the liver is injured, quiescent HSCs transform into activated HSCs and transdifferentiate into myofibroblast-like cells, stimulating excessive ECM accumulation (Tsuchida and Friedman, 2017).

Natural compounds are a valuable source for new drug discovery. As synthetic chemical compounds have the limitation of structural diversity, studies have focused on discovering new medicine from natural products. Paeonia suffruticosa Andrews belongs to the family Ranunculaceae and is an edible, ornamental, and medicinal plant widely cultivated in Asia, Europe, and America (Han and Bhat, 2014; Zhu et al., 2018). Several pharmacological studies have demonstrated that P. suffruticosa has anti-inflammatory, anti-invasive, antidiabetic, anti-obesity, and anti-oxidant activities (Chun et al., 2007; Han and Bhat, 2014; Lee et al., 2003; Nagasawa et al., 1991; Pisani et al., 1999; Zhu et al., 2018). P. suffruticosa contains different types of active compounds such as phenols, monoterpene glycosides, flavonoids, galloylglucoses, triterpenoids, and gallic acid derivatives (Pan et al., 2020). Paeonol, which is a phenolic compound, is a predominant compound in P. suffruticosa. Paeonol has anti-oxidant and anti-inflammatory activities (Chou, 2003; Dai et al., 1999; Hsieh et al., 2006). Furthermore, it has been shown to alleviate alcoholic liver injury by inhibiting hepatic steatosis and inflammation (Hu et al., 2010). Paeonol-containing herbal medicines have traditionally been used to alleviate various liver diseases, including hepatitis; however, scientific evidence in this regard is limited.

In this study, we aimed to evaluate the inhibitory effects of P. suffruticosa extract (PSE) and paeonol on collagen synthesis via HSC activation. The hepatoprotective effects of paeonol were examined in a thioacetamide (TAA)-induced LF model. We also investigated the effects of paeonol on the SMAD2/3 and signal transducer and activator of transcription 3 (STAT3) signaling pathways to elucidate the molecular mechanisms involved in HSC activation.

Materials and methods

Preparation of PSE

The root cortex of P. suffruticosa was purchased from an herbal medicine market (Seoul, South Korea). Shade-dried root cortex of P. suffruticosa (300 g) was pulverized with a blender (SMX-M41KP, Shinil Industrial, Seoul, South Korea). The pulverized sample (300 g) was extracted two times with 1.5 L of 100% ethanol (Duksan Pure Chemicals, Ansan, South Korea) for 3 h under reflux at 80 °C. The PSE was passed through a filter paper (5-μm pore size; Toyo Roshi Kaishi, Tokyo, Japan). To obtain 100% ethanol extract of P. suffruticosa, the filtered PSE was concentrated using a rotary vacuum evaporator (Rotavapor R-100; Buchi, Flawil, Switzerland) and lyophilized using a freeze dryer (FD5512; Ilshin, Seoul, South Korea). The yield of PSE was 25.9%. The pulverized PSE was stored at 4 °C until use. Before use, the stored PSE was dissolved in dimethyl sulfoxide (Daejung, Seoul, South Korea) to the required concentration.

High-performance liquid chromatography analysis

The paeonol standard (H35803) was purchased from Sigma-Aldrich (St. Louis, MO, USA). Analytical and gradient grade acetonitrile and water were purchased from Duksan Pure Chemicals. The paeonol standard and PSE were dissolved in 100% acetonitrile to make 1 mg/mL paeonol. High-performance liquid chromatography (HPLC) analysis was then performed on a Zorbax Eclipse Plus C18 (250 mm × 4.6 mm, 5 µm) analytical column equipped with the Hitachi LaChrom Elite HPLC system (Hitachi High-Tech Corp., Tokyo, Japan). Acetonitrile in solvent (A) and distilled water in solvent (B) were used as mobile phases in the gradient elution system (Supplementary Table 1).

Chemicals

Transforming growth factor (TGF)-β1 was obtained from R&D Systems (Minneapolis, MN, USA), and TAA, 2′-hydroxy-4′-methoxyacetophenone (paeonol), and silymarin were purchased from Sigma-Aldrich.

Cell culture

The human HSC line LX-2 was obtained from the American Type Culture Collection (Manassas, VA, USA). The cells were cultured with Dulbecco’s modified Eagle medium (Sigma-Aldrich) containing 10% fetal bovine serum (Thermo Scientific, Waltham, MA, USA), penicillin (100 units/mL), and streptomycin (100 μg/mL). All cell cultures were maintained at 37 °C in a humidified atmosphere containing 5% CO2.

Cell viability

Cell viability was analyzed using the EZ-Cytox cell viability assay kit (DoGenBio, Seoul, South Korea). LX-2 cells (1 × 104 cells/well) were seeded in transparent 96-well plates in a growth medium and incubated for 24 h. The cells were then treated with PSE (10.5–100 μg/mL) or paeonol (5–100 μM) with or without TGF-β1 (10 ng/mL) for 48 h. According to the manufacturer’s protocol, a 10% diluted water-soluble tetrazolium salt reagent was added to each well (100 µL per well), and the plates were incubated at 37 °C. Absorbance of the samples was measured at 450 nm using a spectrophotometer (Allsheng, Zhejiang, China). Cell viability was calculated based on the absorbance ratio between cells treated with P. suffruticosa and paeonol and untreated cells.

Animal experiments

Male C57BL/6 J (7 weeks old) mice weighing between 21 and 22 g were purchased from Daehan BioLink (Chungbuk, South Korea). After 1 week of acclimatization, the mice were randomly assigned to five groups: saline and saline group (n = 5), TAA (100 mg/kg) and saline group (n = 6), TAA and paeonol (2.5 and 40 mg/kg) group (n = 6), and TAA and silymarin (40 mg/kg) group (n = 6). The mice were administered TAA through intraperitoneal injection (I.P.) three times per week for 4 weeks. To evaluate the hepatoprotective effect of paeonol and silymarin, paeonol and silymarin were administered through I.P. to the mice three times per week for 2 weeks on day 15 after 2 weeks of TAA administration. The mice were euthanized on day 29, and blood and liver tissue were collected for further analysis under isoflurane anesthesia. Blood was collected by puncture of the retro-orbital sinus and cervical dislocation was performed according to Institutional Animal Care and Use Committee (IACUC) guideline after blood and tissue collection under anesthesia. Serum levels of aspartate aminotransferase and alanine transaminase were quantitatively evaluated as biochemical indicators of hepatic function in mice (Southeast Medi-Chem Institute, Busan, South Korea).

Ethics statement

All animal experiments were approved by the Kyungsung University IACUC (2022-006A) and conducted in accordance with the guidelines of the National Institute of Health. Surgical tissue isolation was performed as a terminal procedure with the mice under anesthesia as described below, and all precautions were taken to minimize animal suffering.

RNA extraction and quantitative real-time polymerase chain reaction

LX-2 cells were seeded in six-well plates at 8 × 104 cells/well and incubated for 24 h. The cells were treated with paeonol (5–50 μM) for 48 h after TGF-β1 (10 ng/mL) treatment for 48 h for HSC activation. The total RNA was isolated using TRIzol reagent (Takara Bio Inc, Shiga, Japan). The extracted RNA was measured using NanoDrop-400A (Allsheng). RNA concentration was 2 μg. The total RNA was reverse transcribed to cDNA using a cDNA synthesis kit (Takara Bio Inc.). The quantitative polymerase chain reaction (qPCR) was performed in duplicate for each sample using the SYBR® Green qPCR Master Mix (Toyobo, Osaka, Japan). cDNA was amplified using PCR with the primers listed in Table 1. The expression levels of mRNA were normalized to the level of GAPDH.

Table 1.

List of qPCR primers

Gene Species Forward Reverse
α-SMA Human 5′-CTGGCATCGTGCTGGACTCT-3′ 5′-GATCTCGGCCAGCCAGATC-3′
Col 1A1 Human 5′-GGCAACAGCCGCTTCACCTAC-3′ 5′-GCGGGAGGACTTGGTGGTTTT-3′
Fibronectin Human 5′-CAGTGGGAGACCTCGAGAAG-3′ 5′-TCCCTCGGAACATCAGAAAC-3′
PDGFR_β1 Human 5′-CAGGAGAGACAGCAACAGCA-3′ 5′-AACTGTGCCCACACCAGAAG-3′
GAPDH Human 5′-AATCCCATCACCATCTTCCA-3′ 5′-TGGACTCCACGACGTACTCA-3′
IL-1β Human 5′-AAGCTGATGGCCCTAAACAG-3′ 5′-AGGTGCATCGTGCACATAAG-3′
INOS Human 5′-TGTGCTCTTGCCTGTATGC-3′ 5′-TTGCCAAACGTACTGGTCAC-3′
MMP-2 Human 5′-GAGAACCAAAGTCTGAAGAG-3′ 5′-GGAGTGAGAATGCTGATTAG-3′
MMP-9 Human 5′-TTTGACAGCGACAAGAAGTGG-3′ 5′-GGGCGAGGACCATAGAGG-3′
α-SMA Mouse 5′-GTTCAGTGGTGCCTCTGTCA-3′ 5′-GTTCAGTGGTGCCTCTGTCA-3′
Col 1A1 Mouse 5′-TTCGGACTAGACATTGG-3′ 5′-GGGTTGTTCGTCTGTTTC-3′
Col 3A1 Mouse 5′-ACGTAGATGAATTGGGATGCAG-3′ 5′-GGGTTGGGGCAGTCTAGTG-3′
IL-6 Mouse 5′-GGTGACAACCACGGCCTTCCC-3′ 5′-TTAAGCCTCCGACTTGTGAAGTGGT-3′
GAPDH Mouse 5′-TTGATGGCAACAATCTCCAC-3′ 5′-CGTCCCGTAGACAAAATGGT-3′

Western blot analysis

LX-2 cells were seeded in a six-well plate at 8 × 104 cells/well and incubated for 24 h. The cells were treated with paeonol for 48 h after TGF-β1 treatment for 48 h. The cells were washed twice with PBS and lysed with radio immunoprecipitation assay buffer (Thermo Scientific) with protease inhibitor cocktails (GenDEPOT, Barker, TX, USA). The cell lysates were incubated on ice for 30 min and centrifuged at 21,130×g for 15 min at 4 °C. The protein content of the lysates was determined using a BCA protein assay kit (Pierce, Rockford, IL, USA). Fifteen micrograms of each protein was resolved in a 6%–10% gradient SDS-PAGE gel and transferred on to a polyvinylidene difluoride membrane (Bio-Rad, Hercules, CA, USA). The membrane was blocked with 3% bovine serum albumin in TBS-T at 20 °C for 1 h and incubated with primary antibodies overnight at 4 °C.

The membrane was incubated with the following antibodies: anti-alpha smooth muscle actin (AB5694; Abcam, MA, USA), anti-collagen IA1 (AB138492; Abcam), anti-SMAD2/3 (3102S; Cell Signaling Technology, MA, USA), anti-phospho-SMAD2/3 (8828S; Cell Signaling Technology), anti-STAT3 (9139S; Cell Signaling Technology), anti-phospho-STAT3 (9145S; Cell Signaling Technology), and anti-GAPDH (015-25473; FUJIFILM Wako Pure Chemical Corporation, Osaka, Japan).

Statistical analysis

The results were analyzed using GraphPad Prism version 7.00 (GraphPad software Inc., San Diego, CA, USA) and Excel (Microsoft, Redmond, WA, USA). Each assay was performed at least three times. The results of each assay are expressed as mean ± standard deviation (SD). The differences between the groups were assessed using Duncan’s post-hoc test after a one-way analysis of variance. Statistical significance was accepted at p < 0.05.

Results and discussion

As a part of a research project to discover bioactive products from natural resources, we found that PSE has hepatoprotective effects. It inhibits the accumulation of collagen in LX-2 human HSCs without cytotoxicity (Fig. 1).

Fig. 1.

Fig. 1

Inhibitory effects of methanol extract of Paeonia suffruticosa on hepatic stellate cell (HSC) activation. (A) Cytotoxicity of PSE in LX-2 cells was measured using the cell viability assay after treatment with different concentrations of PSE for 48 h. (B) Protein expression level of COL1A1 in LX-2 cells was measured using western blotting. GAPDH was used as the loading control. Each experiment was repeated three times and the values represent mean ± standard deviation (SD). ##p < 0.01 compared with non-treated cells. *p < 0.05 compared with TGF-β1-treated cells. n.s not significant compared to TGF-β1-treated cells, TGF-β1, transforming growth factor-β1, COL1A1, collagen 1A1, PSE, P. suffruticosa extract

While investigating the active compounds in PSE, we discovered that paeonol inhibits the activation of HSCs (Fig. 2A). Paeonol was detected to be the major active compound in PSE (Sun et al., 2018). In this study, paeonol inhibited the accumulation of collagen and expression of alpha smooth muscle (α-SMA) without cytotoxicity (Fig. 2B, C). α-SMA is an important marker of HSC activation and helps diagnose hepatic fibrosis in the initial stages (Carpino et al., 2005). To the best of our knowledge, this is the first study to report the inhibitory effect of paeonol on HSC activation. In a previous study, paeonol alleviated alcoholic liver injury by inhibiting the inflammation and apoptosis of hepatocytes; however, the mechanism was not associated with HSC activation (Hu et al., 2010). Before paeonol treatment, LX-2 cells were activated by pre-treatment with TGF-β for 48 h. TGF-β is a potent profibrogenic cytokine, and its signaling promotes HSC activation and ECM production (Dewidar et al., 2015; Fabregat and Caballero-Díaz, 2018; Park et al., 2021). In LX-2 cells, TGF-β (10 ng/mL) increased the expression of α-SMA and collagen at the mRNA and protein levels (Fig. 2C, D). Our study indicates that paeonol inhibits TGF-β-induced HSC activation and suppresses mRNA expression of fibrogenic markers, such as fibronectin and platelet-derived growth factor-β (PDGFR-β).

Fig. 2.

Fig. 2

Paeonol inhibited hepatic stellate cell (HSC) activation. (A) Chemical structure of paeonol. (B) Cytotoxicity of PAE in LX-2 cells was measured using the cell viability assay after treatment with different concentrations of PAE for 48 h. (C) Protein expression levels of α-SMA and COL1A1 in LX-2 cells were measured using western blotting. GAPDH was used as the loading control. (D) Gene expression of α-SMA, COL1A1, fibronectin, and PDGFR-β was measured using qPCR analysis. Each experiment was repeated three times, and the values represent mean ± standard deviation (SD). ##p < 0.01 compared with non-treated cells. *p < 0.05 **p < 0.01 compared with TGF-β1-treated cells. n.s, not significant compared to TGF-β1-treated cells. TGF-β1, transforming growth factor-β1, α-SMA, α-smooth muscle actin, COL1A1, collagen 1A1, PAE paeonol

To determine the content of paeonol in PSE, the retention time of PSE was compared with that of the paeonol standard. The paeonol standard peak and PSE peak were clearly separated without any analytical interference (Supplementary Fig. 1). The calibration curve of the paeonol standard was calculated as a regression equation y = ax + b and the correlation coefficient R2 was 0.999. The content of paeonol in the PSE calculated with a linear calibration curve was approximately 0.49% (Supplementary Table 2).

To evaluate the effects of paeonol in an animal model, paeonol (2.5 and 40 mg/kg) was administered six times for 2 weeks after the induction of liver injury by repeated treatment with TAA (100 mg/kg) for 2 weeks (Fig. 3A). Thioacetamide is a well-known hepatotoxic chemical bioactivated in the liver via oxidation processes (Hu et al., 2010). Hepatotoxic damage induced by TAA treatment induces collagen deposition, similar to that observed in the carbon tetrachloride mouse model or bile duct ligation model (Liu et al., 2017). Our results showed that paeonol has protective effects against TAA, similar to silymarin, which is the most well-known natural product with the potential to treat hepatic diseases such as fibrosis and cirrhosis. Paeonol reduced liver injury and aspartate transaminase and alanine transaminase levels, which were increased by TAA injection (Fig. 3B, C). Moreover, paeonol treatment substantially suppressed the expression of inflammation and fibrosis-related genes, such as α-SMA, COL1A2, TGF-β, PDGFR-β, CTGF, TIMP-1, and MCP-1, which was increased by TAA injection (Fig. 3D).

Fig. 3.

Fig. 3

Paeonol attenuated thioacetamide (TAA)-induced liver fibrosis. (A) Mice were intraperitoneally injected with saline or PAE for 2 weeks after 2 weeks of saline or TAA (100 mg/kg) treatment. (B) Serum aspartate aminotransferase (AST) and (C) alanine transaminase (ALT) levels were analyzed. (D) Gene expression of α-SMA, COL1A2, TGF-β, PDGFR-β, CTGF, TIMP-1, and MCP-1 was measured using qPCR analysis. ##p < 0.01 compared with the non-treated group. *p < 0.05, **p < 0.01 compared with the TAA-treated group

To explore the hepatoprotective mechanisms of paeonol, we tested its effects on the SMAD and STAT3 pathways. SMAD transfers signal in the TGF-β canonical pathway. Hetero-dimerization of TGF-β type I and II receptors promotes SMAD2 and SMAD3 phosphorylation. SMAD2/3 then forms a heterocomplex with SMAD4 and translocates to the nucleus to regulate target gene transcription (Hata and Chen, 2016; Zhang, 2009). In this study, paeonol inhibited SMAD 2/3 phosphorylation induced by TGF-β1 at different time points (15, 30, 60, 120, and 240 min) (Fig. 4). The inhibitory effects of paeonol on the TGF-β1/SMAD pathway was previously reported in pancreatic cancer cells (Cheng et al., 2020). This inhibition is considered an important therapeutic target for cancer or fibrotic diseases (Chung et al., 2021; Hu et al., 2018; Peng et al., 2022). The STAT3 pathway also plays an important role in fibrotic diseases via inflammatory responses and fibroblast activation (Chakraborty et al., 2017; Kasembeli et al., 2018). In LF, the STAT3 pathway is a key regulator of development of non-alcoholic fatty liver disease, schistosomiasis, chemical liver injury, and hepatitis B virus infection (Park et al., 2020, 2019; Wang et al., 2011; Zhao et al., 2021). STAT3 signals represent the non-canonical TGF-β pathway, and it has been reported that the JAK1/STAT pathway in association with the SMAD pathway is essential for HSC activation (Tang et al., 2017).

Fig. 4.

Fig. 4

Paeonol suppressed SMAD2/3 and STAT3 phosphorylation in TGF-β1-activated LX-2 cells. LX-2 cells were treated with TGF-β1 or TGF-β1 + PAE for different periods. GAPDH was used as the loading control. TGF-β1, transforming growth factor-β1, PAE, paeonol

Systematic screening for bioactive compounds that inhibit HSC activation led to the identification and isolation of paeonol, the primary phenolic compound from PSE. Our study indicates that paeonol has inhibitory effects on HSC activation by suppressing SMAD2/3 and STAT3 phosphorylation and thus has potential to be developed further for hepatic fibrosis treatment.

Supplementary Information

Below is the link to the electronic supplementary material.

Acknowledgements

This research was supported by Kyungsung University Research Grants in 2021.

Declarations

Conflict of interest

The authors declare no conflicts of interest.

Footnotes

Publisher's Note

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

Hye Kyung Kim and Yong Joo Park contributed equally to this work.

Contributor Information

Hye Kyung Kim, Email: fiona30@ks.ac.kr.

Yong Joo Park, Email: yjpark@ks.ac.kr.

References

  1. Bataller R, Brenner DA. Liver fibrosis. Journal of Clinical Investigation. 2005;115:209–218. doi: 10.1172/JCI24282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Carpino G, Morini S, Ginanni Corradini S, Franchitto A, Merli M, Siciliano M, Gentili F, Onetti Muda A, Berloco P, Rossi M, Attili AF, Gaudio E. Alpha-SMA expression in hepatic stellate cells and quantitative analysis of hepatic fibrosis in cirrhosis and in recurrent chronic hepatitis after liver transplantation. Digestive and Liver Disease. 2005;37:349–356. doi: 10.1016/j.dld.2004.11.009. [DOI] [PubMed] [Google Scholar]
  3. Chakraborty D, Šumová B, Mallano T, Chen C-W, Distler A, Bergmann C, Ludolph I, Horch RE, Gelse K, Ramming A, Distler O, Schett G, Šenolt L, Distler JHW. Activation of STAT3 integrates common profibrotic pathways to promote fibroblast activation and tissue fibrosis. Nature Communications. 2017;8:1130. doi: 10.1038/s41467-017-01236-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Cheng CS, Chen JX, Tang J, Geng YW, Zheng L, Lv LL, Chen LY, Chen Z. Paeonol inhibits pancreatic cancer cell migration and invasion through the inhibition of TGF-β1/Smad signaling and epithelial-mesenchymal-transition. Cancer Management and Research. 2020;12:641–651. doi: 10.2147/CMAR.S224416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Chou TC. Anti-inflammatory and analgesic effects of paeonol in carrageenan-evoked thermal hyperalgesia. British Journal of Pharmacology. 2003;139:1146–1152. doi: 10.1038/sj.bjp.0705360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Chun SC, Jee SY, Lee SG, Park SJ, Lee JR, Kim SC. Anti-inflammatory activity of the methanol extract of moutan cortex in LPS-activated Raw264.7 cells. Evid. Complementary and Alternative Medicine. 4: 327–333 (2007) [DOI] [PMC free article] [PubMed]
  7. Chung JY, Chan MK, Li JS, Chan AS, Tang PC, Leung KT, To KF, Lan HY, Tang PM. TGF-β signaling: from tissue fibrosis to tumor microenvironment. International Journal of Molecular Sciences. 2021;22:7575. doi: 10.3390/ijms22147575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Dai M, Zhi X, Peng D, Liu Q. Inhibitory effect of paeonol on experimental atherosclerosis in quails. Zhongguo Zhong Yao Za Zhi. 24: 488–490, 512 (1999) [PubMed]
  9. Dewidar B, Soukupova J, Fabregat I, Dooley S. TGF-β in hepatic stellate cell activation and liver fibrogenesis: updated. Current Pathobiology Reports. 2015;3:291–305. doi: 10.1007/s40139-015-0089-8. [DOI] [Google Scholar]
  10. Fabregat I, Caballero-Díaz D. Transforming growth factor-β-induced cell plasticity in liver fibrosis and hepatocarcinogenesis. Frontiers in Oncology. 2018;8:357. doi: 10.3389/fonc.2018.00357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Ginès P, Castera L, Lammert F, Graupera I, Serra-Burriel M, Allen AM, Wong VWS, Hartmann P, Thiele M, Caballeria L, De Knegt RJ, Grgurevic I, Augustin S, Tsochatzis EA, Schattenberg JM, Guha IN, Martini A, Morillas RM, Garcia-Retortillo M, De Koning HJ, Fabrellas N, Pich J, Ma AT, Diaz MA, Roulot D, Newsome PN, Manns M, Kamath PS, Krag A. Population screening for liver fibrosis: toward early diagnosis and intervention for chronic liver diseases. Hepatology. 2022;75:219–228. doi: 10.1002/hep.32163. [DOI] [PubMed] [Google Scholar]
  12. Han CV, Bhat R. In vitro control of food-borne pathogenic bacteria by essential oils and solvent extracts of underutilized flower buds of Paeonia suffruticosa (Andr.). Industrial Crops and Products. 54: 203–208 (2014)
  13. Hata A, Chen YG. TGF-β signaling from receptors to Smads. Cold Spring Harbor Perspectives in Biology. 2016;8:a022061. doi: 10.1101/cshperspect.a022061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Hsieh CL, Cheng CY, Tsai TH, Lin IH, Liu CH, Chiang SY, Lin JG, Lao CJ, Tang NY. Paeonol reduced cerebral infarction involving the superoxide anion and microglia activation in ischemia-reperfusion injured rats. Journal of Ethnopharmacology. 2006;106:208–215. doi: 10.1016/j.jep.2005.12.027. [DOI] [PubMed] [Google Scholar]
  15. Hu HH, Chen DQ, Wang YN, Feng YL, Cao G, Vaziri ND, Zhao YY. New insights into TGF-β/Smad signaling in tissue fibrosis. Chemico-Biological Interactions. 2018;292:76–83. doi: 10.1016/j.cbi.2018.07.008. [DOI] [PubMed] [Google Scholar]
  16. Hu S, Shen G, Zhao W, Wang F, Jiang X, Huang D. Paeonol, the main active principles of Paeonia moutan, ameliorates alcoholic steatohepatitis in mice. Journal of Ethnopharmacology. 2010;128:100–106. doi: 10.1016/j.jep.2009.12.034. [DOI] [PubMed] [Google Scholar]
  17. Kasembeli MM, Bharadwaj U, Robinson P, Tweardy DJ. Contribution of STAT3 to inflammatory and fibrotic diseases and prospects for its targeting for treatment. International Journal of Molecular Sciences. 2018;19:2299. doi: 10.3390/ijms19082299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Lee SJ, Lee IS, Mar W. Inhibition of inducible nitric oxide synthase and cyclooxygenase-2 activity by 1,2,3,4,6-penta-O-galloyl-beta-D-glucose in murine macrophage cells. Archives of Pharmacal Research. 2003;26:832–839. doi: 10.1007/BF02980029. [DOI] [PubMed] [Google Scholar]
  19. Liu F, Chen L, Rao H-Y, Teng X, Ren Y-Y, Lu Y-Q, Zhang W, Wu N, Liu F-F, Wei L. Automated evaluation of liver fibrosis in thioacetamide, carbon tetrachloride, and bile duct ligation rodent models using second-harmonic generation/two-photon excited fluorescence microscopy. Laboratory Investigation. 2017;97:84–92. doi: 10.1038/labinvest.2016.128. [DOI] [PubMed] [Google Scholar]
  20. Nagasawa H, Iwabuchi T, Inatomi H. Protection by tree-peony (Paeonia suffruticosa Andr) of obesity in (SLN x C3H/He) F1 obese mice. In Vivo. 1991;5:115–118. [PubMed] [Google Scholar]
  21. Pan Y, Gao Z, Huang X-Y, Chen J-J, Geng C-A. Chemical and biological comparison of different parts of Paeonia suffruticosa (Mudan) based on LCMS-IT-TOF and multi-evaluation in vitro. Industrial Crops and Products. 2020;144:112028. doi: 10.1016/j.indcrop.2019.112028. [DOI] [Google Scholar]
  22. Park YJ, An H-T, Park J-S, Park O, Duh AJ, Kim K, Chung KH, Lee KC, Oh Y, Lee S. Tyrosine kinase inhibitor neratinib attenuates liver fibrosis by targeting activated hepatic stellate cells. Scientific Reports. 2020;10:14756. doi: 10.1038/s41598-020-71688-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Park YJ, Jeon MS, Lee S, Kim JK, Jang TS, Chung KH, Kim KH. Anti-fibrotic effects of brevilin A in hepatic fibrosis via inhibiting the STAT3 signaling pathway. Bioorganic & Medicinal Chemistry Letters. 2021;41:127989. doi: 10.1016/j.bmcl.2021.127989. [DOI] [PubMed] [Google Scholar]
  24. Park YJ, Kim DM, Jeong MH, Yu JS, So HM, Bang IJ, Kim HR, Kwon SH, Kim KH, Chung KH. (-)-Catechin-7-O-β-d-apiofuranoside inhibits hepatic stellate cell activation by suppressing the STAT3 signaling pathway. Cells. 2019;9:30. doi: 10.3390/cells9010030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Peng D, Fu M, Wang M, Wei Y, Wei X. Targeting TGF-β signal transduction for fibrosis and cancer therapy. Molecular Cancer. 2022;21:104. doi: 10.1186/s12943-022-01569-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Pisani P, Parkin DM, Bray F, Ferlay J. Estimates of the worldwide mortality from 25 cancers in 1990. International Journal of Cancer. 1999;83:18–29. doi: 10.1002/(SICI)1097-0215(19990924)83:1&#x0003c;18::AID-IJC5&#x0003e;3.0.CO;2-M. [DOI] [PubMed] [Google Scholar]
  27. Senoo H, Yoshikawa K, Morii M, Miura M, Imai K, Mezaki Y. Hepatic stellate cell (vitamin A-storing cell) and its relative–past, present and future. Cell Biology International. 2010;34:1247–1272. doi: 10.1042/CBI20100321. [DOI] [PubMed] [Google Scholar]
  28. Sun SW, Du J, Hwang E, Yi TH. Paernol extracted from Paeonia suffruticosa Andr. amealiorated UVB-induced skin photoaging via DLD/Nrf2/ARE and MAPK/AP-1 pathway. Phytotherapy Research. 32(9): 1741-1749 (2018) [DOI] [PubMed]
  29. Tang LY, Heller M, Meng Z, Yu LR, Tang Y, Zhou M, Zhang YE. Transforming growth factor-β (TGF-β) directly activates the JAK1-STAT3 axis to induce hepatic fibrosis in coordination with the SMAD pathway. Journal of Biological Chemistry. 2017;292:4302–4312. doi: 10.1074/jbc.M116.773085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Tsuchida T, Friedman SL. Mechanisms of hepatic stellate cell activation. Nature Reviews Gastroenterology & Hepatology. 2017;14:397–411. doi: 10.1038/nrgastro.2017.38. [DOI] [PubMed] [Google Scholar]
  31. Wang H, Lafdil F, Kong X, Gao B. Signal transducer and activator of transcription 3 in liver diseases: a novel therapeutic target. International Journal of Biological Sciences. 2011;7:536–550. doi: 10.7150/ijbs.7.536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Zhang J, Liu Q, He J, Li Y. Novel therapeutic targets in liver fibrosis. Frontiers in Molecular Biosciences. 2021;8:766855. doi: 10.3389/fmolb.2021.766855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Zhang YE. Non-Smad pathways in TGF-β signaling. Cell Research. 2009;19:128–139. doi: 10.1038/cr.2008.328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Zhao J, Qi YF, Yu YR. STAT3: a key regulator in liver fibrosis. Annals of Hepatology. 2021;21:100224. doi: 10.1016/j.aohep.2020.06.010. [DOI] [PubMed] [Google Scholar]
  35. Zhu S, Shirakawa A, Shi Y, Yu X, Tamura T, Shibahara N, Yoshimatsu K, Komatsu K. Impact of different post-harvest processing methods on the chemical compositions of peony root. Journal of Natural Medicines. 2018;72:757–767. doi: 10.1007/s11418-018-1214-x. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials


Articles from Food Science and Biotechnology are provided here courtesy of Springer

RESOURCES