Silibinin A decreases statin‑induced PCSK9 expression in human hepatoblastoma HepG2 cells

  • Authors:
    • Zhewen Dong
    • Wenxiang Zhang
    • Siyu Chen
    • Chang Liu
  • View Affiliations

  • Published online on: June 5, 2019     https://doi.org/10.3892/mmr.2019.10344
  • Pages: 1383-1392
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hypercholesterolemia is one of the major risk factors for the occurrence and development of atherosclerosis. The most common drugs used to treat hypercholesterolemia are 3‑hydroxy‑3‑methyl‑glutaryl‑CoA reductase inhibitors, known as statins. Statins induce a beneficial increase in the levels of the low density lipoprotein receptor (LDLR) and additionally upregulate proprotein convertase subtilisin/kexin type 9 (PCSK9), which leads to LDLR degradation. This process causes a negative feedback response that attenuates the lipid lowering effects of statins. Therefore, the development of PCSK9 inhibitors may increase the lipid‑lowering functions of statins. In the present study, a drug‑screening assay was developed using the human PCSK9 promoter, based on data from a dual‑luciferase reporter assay, and the efficacies of various compounds from Traditional Chinese Medicine were examined. Among the compounds examined, SIL was demonstrated to function by targeting PCSK9. It was identified that SIL treatment decreased the expression levels of PCSK9 in HepG2 cells by decreasing the activity of the PCSK9 promoter in a dose‑and time‑dependent manner. Notably, SIL antagonized the statin‑induced phosphorylation of the p38 MAPK signaling pathway. The present study suggested that SIL may be developed as a novel PCSK9 inhibitor that may increase the efficiency of statin treatment.

Introduction

Hypercholesterolemia is one of the major risk factors responsible for the occurrence and development of atherosclerosis. This condition has become the primary therapeutic focus of atherosclerosis treatment (1). The most common cholesterol-lowering drugs are statins. These compounds decrease intracellular cholesterol levels by selectively inhibiting the enzyme 3-hydroxy-3-methylglutaryl-coenzyme A reductase (2). This in turn inhibits cholesterol biosynthesis and decreases hepatic cholesterol concentration (2). Statins increase the expression levels of the low density lipoprotein receptor (LDLR) in liver cell membranes by the sterol regulatory element binding protein (SREBP) pathway, leading to an increase in the clearance of LDL particles from the blood circulation (2). However, the use of statins causes several side effects. High doses of statins may increase the incidence and severity of multiple adverse events, including hepatotoxicity and myopathy, which are accompanied by muscle cramps, stiffness and weakness (3). In addition, statins increase the levels of the proprotein convertase subtilisin/kexin type 9 (PCSK9), which leads to LDLR degradation, thereby causing a negative feedback response that attenuates their lipid lowering effect (4). Therefore, the development of PCSK9 inhibitors may, in theory, enhance the lipid-lowering functions of statins.

PCSK9 is a newly identified serine protease that has emerged as a critical regulator in the pathogenesis of hypercholesterolemia and atherosclerosis (5). PCSK9 consists of 692 amino acids and is synthesized in the cytoplasm of hepatocytes. This protein enters the endoplasmic reticulum and the signal peptide of PCSK9 is cleaved. Pro-PCSK9 undergoes autocatalytic cleavage at residue Gln152 and binds to the catalytic domain of the protein to form a mature protein by the secretory pathway (6,7). The mature PCSK9 protein binds directly to the epidermal growth factor repeat A of the LDLR, and the PCSK9: LDLR complex is transferred to the lysosomes for degradation. Therefore, LDLR is no longer recycled back to the cell membrane surface, which leads to elevated LDL levels in the plasma resulting from decreasing binding of the LDL to its receptor (810). Previous studies have indicated that functional mutations of PCSK9 are associated with human hypercholesterolemia (gain-of-function mutation) or hypocholesterolemia (loss-of-function mutation), which are associated with increased and decreased cardiovascular risk, respectively (1114). In human studies, PCSK9 has been demonstrated to be an important target for the decrease of plasma cholesterol concentration and the decrease in cardiovascular risk (5).

The United States of America Food and Drug Administration (FDA) has approved the marketing of 2 monoclonal antibodies against PCSK9, namely alirocumab and evolocumab, which are effective in decreasing levels of atherogenic lipoproteins and are well tolerated (15,16). However, these 2 inhibitors are expensive; their estimated cost ranges from $12,000-$15,000 for each patient per year (17). Therefore, the development of low-cost PCSK9 inhibitors will have significant commercial interest. In contrast to these observations, traditional Chinese medicine (TCM) has been used in clinical practice for >2,000 years in China and has exhibited marked beneficial effects on human health (18). Recently, several studies demonstrated a favorable effect of TCM for the treatment of dyslipidemia by the regulation of PCSK9, for example; berberine is a compound isolated from a Chinese herb that has exhibited cholesterol-lowering activity. Berberine inhibits PCSK9 transcription by downregulating hepatic hepatocyte nuclear factor 1 (HNF1) protein expression via the ubiquitin-proteasome degradation pathway (19,20). Therefore, TCM may serve as a promising candidate to screen functional PCSK9 inhibitors.

In the present study, a novel drug-screening assay was developed based on the human PCSK9 promoter. The assay used data from a dual-luciferase reporter assay and the compound silibinin A (SIL), a TCM compound, was identified to repress PCSK9 promoter activity. SIL has been demonstrated to have a broad range of pharmacological activities, including anti-inflammatory, anti-oxidant, anti-cancer, neuroprotective and cardioprotective effects (2126). Therefore, SIL has been used as a popular dietary supplement due to its optimal tolerability and low toxicity, and its diverse biological functions. The present study suggested that SIL may be developed as a novel PCSK9 inhibitor used in combination with statins, in order to retain their lipid lowering activity.

Materials and methods

Cell culture

The 293 and human hepatoblastoma HepG2 cells were obtained from the American Type Culture Collection and cultured in Dulbecco's modified Eagle's medium (DMEM; Gibco; Thermo Fisher Scientific, Inc.) supplemented with 10% fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc.) and 1% penicillin/streptomycin solution. All cells were incubated in a cell culture chamber at 37°C under a humidified atmosphere with 5% CO2.

Promoter construction

The promoter of human PCSK9 (−1,833- +100 bp) was retrieved from the National Centre for Biotechnology Information. As demonstrated previously, PCSK9 transcription is controlled through cis regulatory elements located in the proximal promoter region of the PCSK9 gene where the transcription factor Sp1, HNF1 and sterol regulatory element-binding protein 1 (SRE-1) sites are located (−430- −345) (27). Notably, the SRE-1 motif is responsible for the statins-induced PCSK9 transcription (28). Therefore, the region containing all these functional sites was selected for the drug screening protocol. Genomic DNA was extracted from human liver HL7702 cells using a Wizard® Genomic DNA Purification kit (Promega Corporation). The primers were designed using Primer Premier 5 software (Premier Biosoft International), and MluI and XhoI restriction sites were added upstream and downstream of the promoter sequence. The specific sequences were as follows: Forward (F), 5′-TGCAAAAAAGAGTATGCCCGT-3′; reverse (R), 5′-TCCCCAAACAGCGTCAGATTA-3′. The PCSK9 promoter fragment was amplified by polymerase chain reaction (PCR). PCR was conducted by activating the DNA polymerase at 94°C for 5 min, followed by 30 cycles of three-step PCR (94°C for 30 sec, 66.8°C for 20 sec and 72°C for 2 min), a final extension at 72°C for 10 min and holding at 4°C (PrimeSTAR® HS DNA Polymerase; Takara Bio, Inc.), and the band size was detected by 1% agarose gel electrophoresis. The recovered product and the pGL3-basic vector (MiaoLingPlasmid) were double-digested with MluI (Takara Bio, Inc.) and XhoI (Takara Bio, Inc.) at 37°C for 4 h, and annealed together using T4 DNA ligase (Takara Bio, Inc.) at 16°C overnight. The purified product was ligated (the size of the PCSK9-luc promoter plasmid was 6,751 bp), and finally the sequence of the pGL3-basic PCSK9-luc promoter plasmid was verified by GenScript Biotech Corp.

Drug screening and dual-luciferase reporter assay

TCM compounds (detailed drug information were presented in Table I) were used to construct a library in Jiangsu Key Laboratory for Molecular Medical Biotechnology. These compounds were purchased from Nanjing Cebai Biotechnology Co., Ltd. and Shanghai Aladdin Biochemical Technology Co., Ltd. The treatment doses for these compounds were selected based on previous studies (Table I) (2966), which confirmed their functional effects for the treatment of various diseases. To analyze the activity of the PCSK9 promoter, pGL3-PCSK9-luc (60 ng) and internal control plasmid pRL-TK (10 ng; MiaoLingPlasmid) was co-transfected into cells using Lipofectamine® 3000 (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. Following 36 h of culture, 293 or HepG2 cells were incubated with serum-free DMEM containing 1% dimethyl sulfoxide (DMSO) as a negative control, or with the indicated drugs (Table I) for 12 h at 37°C. The cell lysates were extracted and luciferase signal intensities (ratios of firefly luciferase signal normalized to Renilla luciferase) were measured using a Microplate Luminometer (Promega Corporation).

Table I.

Potential drugs that decrease proprotein convertase subtilisin/kexin type 9 expression.

Table I.

Potential drugs that decrease proprotein convertase subtilisin/kexin type 9 expression.

No.DrugsConcentration(Refs.)
1Isoorientin30 µM(29)
2 Dihydrokaempferol690 µM(30)
3Hyperin100 µM(31)
4Glycitein100 µM(32)
5Genistein60 µM(33)
6Calycosin100 µM(34)
7Formononetin200 µM(35)
8Irisflorentin40 µM(36)
9Dichotomitin3 µM(37)
10Iristectorigenin A20 µM(38)
11 6″-O-xylosyl-glycitein350 µM(32)
12Corylin30 µM(39)
13Kaempferide50 µM(40)
14Isovitexin20 µM(41)
15Naringenin150 µM(42)
16Isofraxidin70 µM(43)
17Oleanolic acid80 µM(44)
18Berberine50 µM(45)
19Resveratrol20 µM(46)
20Rosiglitazone300 µM(47)
21Oridonin160 µM(48)
22Lithium chloride100 mM(49)
23Chlorogenic acid450 µM(50)
24Baicalin220 µM(51)
25Nicotinic acid10 mM(52)
26 Cycloastragenol20 µM(53)
27 Prim-O-glucosylcimifugin210 µM(54)
28Epimedin C500 µM(55)
29Linarin10 µM(56)
30Vitexin20 µM(57)
31Myricetin75 µM(58)
32Herbacetin100 µM(59)
33Typhaneoside10 µM(60)
34Afzelin460 µM(61)
35Taxifolin50 µM(62)
36Silibinin A100 µM(63)
37Daidzein10 µM(64)
38 6″-O-Xylosyltectoridin100 µM(65)
39Schaftoside2 µM(66)
Cytotoxicity assay

Cell viability was examined by the Cell Counting Kit-8 assay (CCK-8; Nanjing Jiancheng Bioengineering Institute Co., Ltd.). To determine the non-toxic concentration for cells, SIL (1, 5, 10, 25, 50, 100 and 200 µM) was added to each well (1×104 cells/well). The plates were subsequently incubated for 12 h. CCK-8 solution (10 µl/well) was added to each well and the cells were cultured for an additional 2 h. Finally, a microplate reader was used to measure the absorbance at 450 nm.

In situ terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling (TUNEL) assay

TUNEL assays were performed to detect DNA strand breaks using a commercial kit provided by the Vazyme. In brief, HepG2 cells were treated as aforementioned and fixed with 4% paraformaldehyde for 15 min at room temperature. Following washing, the cells were permeabilized with 0.1% Triton X-100, and finally incubated with TUNEL labeling reagent for 60 min at 37°C. The cells were double-stained with 10 µg/ml DAPI for 10 min at 37°C (Sigma-Aldrich; Merck KGaA). The sections were captured in three different fields per sample with a DP70 digital camera connected to an ECLIPSE Ts2R-FL microscope (Nikon Corporation). Representative images from at least three separate experiments were presented.

Reverse transcription quantitative PCR (RT-qPCR) analysis

Total RNA was isolated using the TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.) and reverse transcribed with the PrimeScript™ RT reagent kit (Takara Bio, Inc.). The resulting cDNA was amplified by qPCR using SYBR Green (Takara Bio, Inc.) and the LightCycler® 480 System (Roche Diagnostics) under the following thermocycling conditions: Denaturation at 95°C for 5 min, followed by 95°C for 10 sec and 60°C for 30 sec for 40 cycles. The relative expression of each targeted gene was determined using the 2−∆∆Cq comparative method (67). The primers for human β-actin were included for normalization. The primer sequences were as follows: PCSK9 F, 5′-AGGGGAGGACATCATTGGTG-3′; PCSK9 R, 5′-CAGGTTGGGGGTCAGTACC-3′; β-actin F, 5′-CACCCACACTGTGCCCATCTACGA-3′; β-actin R, 5′-CAGCGGAACCGCTCATTGCCAATGG-3′.

Western blot analysis

Total cellular proteins were isolated from HepG2 cells using radioimmunoprecipitation assay lysis buffer (Beyotime Institute of Biotechnology). The protein concentration was determined using the BCA protein assay reagent (Beyotime Institute of Biotechnology). Equal amounts of protein (20 µg/lane) were loaded and separated via 10% SDS-PAGE, and then transferred onto PVDF membranes (EMD Millipore). Subsequently, the membranes were blocked with 5% fat-free dry milk at room temperature for 1 h. Membranes were incubated with rabbit anti-PCSK9 (1:1,000; cat. no. BS71876; Bioworld Technology, Inc.), mouse anti-β-actin (1:1,000; cat. no. AP0060; Bioworld Technology, Inc.), rabbit anti-p38 MAPK (1:1,000; cat. no. 9212S; Cell Signaling Technology, Inc.) or rabbit anti-phosphorylated (p)-p38 MAPK (p-Tyr182; (1:1,000; cat. no. 11253; Signalway Antibody LLC) antibodies in 5% fat-free dry milk overnight on a shaker at 4°C. Membranes were subsequently washed three times in PBST and incubated with horseradish peroxidase-conjugated goat anti-mouse IgG secondary antibodies (1:2,000; cat. no. sc-2005; Santa Cruz Biotechnology, Inc.) or goat anti-rabbit IgG secondary antibodies (1:2,000; cat. no. sc-2004; Santa Cruz Biotechnology, Inc.) at room temperature for 1 h. The immunoreactive bands were visualized using electrochemiluminescence (Tanon-5200 Multi; Tanon Science and Technology Co., Ltd.) and quantified with the AlphaEase FC version 3.1.2 software (Alpha Innotech; Cell Biosciences).

Statistical analysis

Groups of data were presented as mean ± standard deviation. The data were analyzed using one-way analysis of variance followed by Fisher's Least Significant Difference post hoc test. The calculations were performed using Origin 8 software (v8.6, OriginLab Corporation). P<0.05 was considered to indicate a statistically significant difference.

Results

SIL decreases the activity of PCSK9-luc in 293 cells

The 293 cells transfected with PCSK9-luc were used to screen the active ingredients present in the TCM compounds (specific compounds were presented in Table I), which have been suggested to inhibit the transcriptional activity of the PCSK9 promoter. The transfected cells were incubated with various drug concentrations for 12 h. Among all the tested drugs, chlorogenic acid, nicotinic acid, epmedin C, linarin, herbacetin, afzelin and SIL decreased the activity of PCSK9-luc (Fig. 1). In addition, SIL was the most effective drug, which decreased PCSK9 promoter activity by 46.6% compared with that measured in the control samples.

Detection of HepG2 cell cytotoxicity and HepG2 apoptosis induction by SIL

The CCK-8 assay was used to assess the potential cytotoxicity of SIL on human HepG2 cells. The data indicated that treatment of SIL at the concentrations of 1, 5, 10 and 25 µM was not cytotoxic to HepG2 cells. SIL at the doses of 50 and 100 µM caused a slight decrease in cell viability, although the differences were not significant (Fig. 2B). To exclude the possibility that the SIL-induced decrease in PCSK9 would induce apoptosis in HepG2 cells, a TUNEL assay was performed and it was identified that 100 µM of SIL did not induce DNA fragmentation in these cells (Fig. 2C). This result indicated that the decrease of PCSK9 expression is not relevant to the apoptosis or cell growth. Therefore, 100 µM SIL was selected as the maximum safe dose for subsequent experiments.

SIL decreases PCSK9 promoter activity and the expression of PCSK9 in a dose-dependent manner

To confirm the results of the drug screening experiments, HepG2 cells were transfected with a firefly luciferase vector containing PCSK9-luc and subsequently treated with SIL for 12 h. When compared with the cells treated with DMSO alone, SIL decreases the activity of luciferase in a dose-dependent manner (Fig. 3A). The maximum decrease in the PCSK9 promoter activity was estimated to be 45.6% for the cells treated with 100 µM SIL.

To examine whether SIL affected the expression levels of PCSK9 mRNA, HepG2 cells were incubated with increasing concentrations of SIL, and the mRNA levels of PCSK9 were detected by RT-qPCR. The results indicated that SIL decreased PCSK9 mRNA levels in a dose-dependent manner (Fig. 3B). At the concentration of 100 µM, SIL inhibited PCSK9 mRNA levels by 65.8% (P<0.01). The suppressive effects of SIL on the PCSK9 protein expression levels were verified by western blot analysis (Fig. 3C).

SIL decreases PCSK9 mRNA and protein expression levels in a time-dependent manner

Subsequently, the effects of 100 µM SIL on PCSK9 expression in HepG2 cells at different time periods were assessed. PCSK9 mRNA and protein expression levels were significantly decreased following treatment of HepG2 cells with SIL in a time-dependent manner (Fig. 4). Notably, the maximum level of inhibition (62.3% decrease compared with control; P<0.05) was observed at 12 h. A similar trend was observed with regard to the expression of the PCSK9 protein.

SIL antagonizes atorvastatin (ATV)-induced upregulation of PCSK9 expression

It was previously reported that ATV may increase PCSK9 expression in HepG2 cells (68). In the present study, PCSK9 mRNA expression levels were 2.3-fold increased following treatment of HepG2 cells with 10 µM ATV, which was consistent with previous studies (68,69). Notably, co-incubation of HepG2 cells with ATV and SIL for 12 h resulted in an inhibition of the ATV-induced increase of PCSK9 mRNA levels by 78.1% (Fig. 5A). Similarly, western blot analysis in HepG2 cell lysates revealed that ATV increased PCSK9 protein expression levels by 51.0%, and SIL attenuated this effect by 43.2% (Fig. 5B).

SIL decreases PCSK9 expression by suppressing the p38 mitogen-activated protein kinases (MAPK) pathway

In the liver, the p38 MAPK signaling pathway serves a central role in lipid and cholesterol metabolism, and is induced by statins to cause hepatic oxidative stress and apoptosis (70). To investigate the cellular and molecular mechanism underlying SIL-induced alleviation of ATV-based PCSK9 accumulation, HepG2 cells were treated with ATV (10 µM) and SIL (100 µM) for 0.5 h and 1 h, respectively. As presented in Fig. 6, SIL treatment decreased the phosphorylation of p38 MAPK protein following ATV treatment for 0.5 h by ~34% in HepG2 cells. This effect occurred prior to the induction of p38 phosphorylation triggered by 1 h of ATV treatment. SIL consistently inhibited p38 phosphorylation induced by 1 h of ATV treatment by ~46%. These data indicated that SIL may enhance the lipid-lowering functions of statins.

Discussion

Hypercholesterolemia is a major risk factor for the development of atherosclerosis. Decrease in plasma cholesterol levels is beneficial for the treatment of hypercholesterolemia-associated diseases. In a clinical setting, statins are the most frequently used drugs for lowering blood lipid levels (71). Statins increase LDLR expression by the inhibition of cholesterol synthesis, leading to the clearance of cholesterol in the blood. In addition, they increase PCSK9 expression at the transcriptional level and cause LDLR degradation within the cell membrane, therefore attenuating their lipid-lowering effects (72). The present study aimed to examine the ability of a novel compound to inhibit PCSK9 expression. Therefore, 39 monomers isolated from TCM studies were screened, and it was identified that SIL inhibited PCSK9 expression in a time- and dose-dependent manner. In addition, SIL inhibited ATV-induced upregulation of PCSK9. These results suggested that SIL may be a promising compound for the attenuation of the negative feedback response of statins with regard to PCSK9 expression.

The monomers isolated from TCM have several advantages, including low cost, low toxicity and optimal isolation procedures. Therefore, the present study focused on the screening of those monomers that targeted PCSK9. Previous studies have indicated that berberine decreases PCSK9 expression and improves the LDL-C uptake from the hepatocytes (19,20,73). However, berberine may also cause detrimental effects, which occur primarily in the digestive system, including nausea, diarrhea, constipation and abdominal pain (74,75). Furthermore, the oral bioavailability of berberine is <1%, due to its poor solubility. In contrast to berberine, the biological half-life time and oral bioavailability levels of SIL are considerably increased, which renders it a good candidate for the development of novel PSCK9 inhibitors. Indeed, it has been demonstrated that when administrated orally with 120 mg SIL, the peak concentration in the plasma (Cmax) in patients was 2.7 µM (76). In addition, the recommended oral dosage of SIL in phase I clinical trials (ClinicalTrials.gov Identifier: NCT00487721) for the treatment of prostate cancer is 13 g and Cmax is 100 µM (77). Taken together, the plasma levels of SIL administrated in the clinical setting were comparable to the concentration used in the present study.

SIL is a polyphenolic that belongs to the flavonoid family of compounds (78). SIL has been used clinically to treat acute and chronic hepatitis, early cirrhosis and poisonous liver injury (22). SIL is a drug with multiple biological targets, including PCSK9, and therefore has potential use in the therapy of lipid metabolism disorders. Notably, the plasma half-life of ATV and SIL is 7 and 6.21 h, respectively. The corresponding time interval to reach Cmax levels is 1.5 h for ATV, and between 1–2 h for SIL, respectively (79,80). These two important parameters have similar values, suggesting that ATV and SIL may be orally administered simultaneously in order to achieve an improved therapeutic effect of ATV.

The liver serves an important role in the physiological process of lipid synthesis, gluconeogenesis and cholesterol metabolism (81). It has been suggested that ATV induces oxidative stress and apoptotic injury in hepatocytes by increasing the phosphorylation of p38, JNK and ERK MAPK enzymes (70). In HepG2 cells, C-reactive protein increases PCSK9 expression by the activation of the p38 MAPK-hepatocyte nuclear factor 1 homeobox A signaling pathway (82). Similarly, leptin additionally induced PCSK9 expression by the activation of p38 MAPK (83). Therefore, p38 MAPK is potentially involved in the regulation of PCSK9 expression. In the present study, it was demonstrated that ATV activated p38 MAPK, while SIL suppressed the ATV-induced phosphorylation of p38 MAPK. The activation of the p38 MAPK signaling pathway downregulated peroxisome proliferator-activated receptor α and its transcriptional target genes carnitine palmitoyltransferase 1A and Acyl-coenzyme A oxidase 1, leading to the inhibition of fatty acid β-oxidation (84). Whether other MAPK enzymes are also involved in the inhibition of PCSK9 by SIL remains unknown.

In conclusion, the present study demonstrated that SIL inhibited the ATV-induced PCSK9 upregulation in HepG2 cells, and that it may be used to decrease the adverse effects of statins following simultaneous administration of these 2 drugs to patients with hypercholesterolemia. Future studies are required to confirm the beneficial effects of SIL in vivo by the use of hyperlipidemic animal models.

Acknowledgements

Not applicable.

Funding

The present study was financially supported by grants from the National Natural Science Foundation of China (grant nos. 31800992, 31771298 and 81800512), the Natural Science Foundation of the Jiangsu Higher Education Institutions of China (grant nos. BK20180554 and BK20180577), the Project of State Key Laboratory of Natural Medicines, China Pharmaceutical University (grant no. SKLNMZZRC201803), the ‘Double First-Class’ University Project (grant no. CPU2018GY17) and the Open Fund of State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, China (grant no. KF-GN-201901).

Availability of data and materials

The analyzed data sets generated during the present study are available from the corresponding author on reasonable request.

Authors' contributions

ZD designed and performed the study, analyzed the data and wrote the manuscript. WZ and SC performed the experiments and analyzed the data. CL designed the experiments, analyzed the data and wrote the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

All authors declare that they have no competing interests.

References

1 

Benito-Vicente A, Uribe KB, Jebari S, Galicia-Garcia U, Ostolaza H and Martin C: Familial hypercholesterolemia: The most frequent cholesterol metabolism disorder caused disease. Int J Mol Sci. 19(pii): E34262018. View Article : Google Scholar : PubMed/NCBI

2 

Randomised trial of cholesterol lowering in 4444 patients with coronary heart disease, . The Scandinavian Simvastatin Survival Study (4S). Lancet. 344:1383–1389. 1994.PubMed/NCBI

3 

Golomb BA and Evans MA: Statin adverse effects: A review of the literature and evidence for a mitochondrial mechanism. Am J Cardiovasc Drugs. 8:373–418. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Dubuc G, Chamberland A, Wassef H, Davignon J, Seidah NG, Bernier L and Prat A: Statins upregulate PCSK9, the gene encoding the proprotein convertase neural apoptosis-regulated convertase-1 implicated in familial hypercholesterolemia. Arterioscler Thromb Vasc Biol. 24:1454–1459. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Gouni-Berthold I: PCSK9 antibodies: A new class of lipid-lowering drugs. Atheroscler Suppl. 18:21–27. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Horton JD, Cohen JC and Hobbs HH: Molecular biology of PCSK9: Its role in LDL metabolism. Trends Biochem Sci. 32:71–77. 2007. View Article : Google Scholar : PubMed/NCBI

7 

Goldstein JL and Brown MS: A century of cholesterol and coronaries: From plaques to genes to statins. Cell. 161:161–172. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Cunningham D, Danley DE, Geoghegan KF, Griffor MC, Hawkins JL, Subashi TA, Varghese AH, Ammirati MJ, Culp JS, Hoth LR, et al: Structural and biophysical studies of PCSK9 and its mutants linked to familial hypercholesterolemia. Nat Struct Mol Biol. 14:413–419. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Fisher TS, Lo Surdo P, Pandit S, Mattu M, Santoro JC, Wisniewski D, Cummings RT, Calzetta A, Cubbon RM, Fischer PA, et al: Effects of pH and low density lipoprotein (LDL) on PCSK9-dependent LDL receptor regulation. J Biol Chem. 282:20502–20512. 2007. View Article : Google Scholar : PubMed/NCBI

10 

Zhang DW, Lagace TA, Garuti R, Zhao Z, McDonald M, Horton JD, Cohen JC and Hobbs HH: Binding of proprotein convertase subtilisin/kexin type 9 to epidermal growth factor-like repeat A of low density lipoprotein receptor decreases receptor recycling and increases degradation. J Biol Chem. 282:18602–18612. 2007. View Article : Google Scholar : PubMed/NCBI

11 

Abifadel M, Varret M, Rabès JP, Allard D, Ouguerram K, Devillers M, Cruaud C, Benjannet S, Wickham L, Erlich D, et al: Mutations in PCSK9 cause autosomal dominant hypercholesterolemia. Nat Genet. 34:154–156. 2003. View Article : Google Scholar : PubMed/NCBI

12 

Maxwell KN and Breslow JL: Proprotein convertase subtilisin kexin 9: The third locus implicated in autosomal dominant hypercholesterolemia. Curr Opin Lipidol. 16:167–172. 2005. View Article : Google Scholar : PubMed/NCBI

13 

Allard D, Amsellem S, Abifadel M, Trillard M, Devillers M, Luc G, Krempf M, Reznik Y, Girardet JP, Fredenrich A, et al: Novel mutations of the PCSK9 gene cause variable phenotype of autosomal dominant hypercholesterolemia. Hum Mutat. 26:4972005. View Article : Google Scholar : PubMed/NCBI

14 

Hallman DM, Srinivasan SR, Chen W, Boerwinkle E and Berenson GS: Relation of PCSK9 mutations to serum low-density lipoprotein cholesterol in childhood and adulthood (from The Bogalusa Heart Study). Am J Cardiol. 100:69–72. 2007. View Article : Google Scholar : PubMed/NCBI

15 

Fala L: Repatha (Evolocumab): Second PCSK9 inhibitor approved by the FDA for patients with familial hypercholesterolemia. Am Health Drug Benefits 9 (Spec Feature). 136–139. 2016.

16 

Raedler LA: Praluent (Alirocumab): First PCSK9 inhibitor approved by the FDA for hypercholesterolemia. Am Health Drug Benefits 9 (Spec Feature). 123–126. 2016.

17 

White CM: Therapeutic potential and critical analysis of the PCSK9 monoclonal antibodies evolocumab and alirocumab. Ann Pharmacother. 49:1327–1335. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Dai L, Lu A, Zhong LLD, Zheng G and Bian Z: Chinese herbal medicine for hyperlipidaemia: A review based on data mining from 1990 to 2016. Curr Vasc Pharmacol. 15:520–531. 2017. View Article : Google Scholar : PubMed/NCBI

19 

Kong W, Wei J, Abidi P, Lin M, Inaba S, Li C, Wang Y, Wang Z, Si S, Pan H, et al: Berberine is a novel cholesterol-lowering drug working through a unique mechanism distinct from statins. Nat Med. 10:1344–1351. 2004. View Article : Google Scholar : PubMed/NCBI

20 

Dong B, Li H, Singh AB, Cao A and Liu J: Inhibition of PCSK9 transcription by berberine involves down-regulation of hepatic HNF1α protein expression through the ubiquitin-proteasome degradation pathway. J Biol Chem. 290:4047–4058. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Flora K, Hahn M, Rosen H and Benner K: Milk thistle (Silybum marianum) for the therapy of liver disease. Am J Gastroenterol. 93:139–143. 1998. View Article : Google Scholar : PubMed/NCBI

22 

Wellington K and Jarvis B: Silymarin: A review of its clinical properties in the management of hepatic disorders. BioDrugs. 15:465–489. 2001. View Article : Google Scholar : PubMed/NCBI

23 

Saller R, Meier R and Brignoli R: The use of silymarin in the treatment of liver diseases. Drugs. 61:2035–2063. 2001. View Article : Google Scholar : PubMed/NCBI

24 

Zi X and Agarwal R: Silibinin decreases prostate-specific antigen with cell growth inhibition via G1 arrest, leading to differentiation of prostate carcinoma cells: Implications for prostate cancer intervention. Proc Natl Acad Sci USA. 96:7490–7495. 1999. View Article : Google Scholar : PubMed/NCBI

25 

Gallo D, Giacomelli S, Ferlini C, Raspaglio G, Apollonio P, Prislei S, Riva A, Morazzoni P, Bombardelli E and Scambia G: Antitumour activity of the silybin-phosphatidylcholine complex, IdB 1016, against human ovarian cancer. Eur J Cancer. 39:2403–2410. 2003. View Article : Google Scholar : PubMed/NCBI

26 

Saliou C, Rihn B, Cillard J, Okamoto T and Packer L: Selective inhibition of NF-kappaB activation by the flavonoid hepatoprotector silymarin in HepG2. Evidence for different activating pathways. FEBS Lett. 440:8–12. 1998. View Article : Google Scholar : PubMed/NCBI

27 

Li H, Dong B, Park SW, Lee HS, Chen W and Liu J: Hepatocyte nuclear factor 1alpha plays a critical role in PCSK9 gene transcription and regulation by the natural hypocholesterolemic compound berberine. J Biol Chem. 284:28885–28895. 2009. View Article : Google Scholar : PubMed/NCBI

28 

Costet P, Cariou B, Lambert G, Lalanne F, Lardeux B, Jarnoux AL, Grefhorst A, Staels B and Krempf M: Hepatic PCSK9 expression is regulated by nutritional status via insulin and sterol regulatory element-binding protein 1c. J Biol Chem. 281:6211–6218. 2006. View Article : Google Scholar : PubMed/NCBI

29 

Ko FN, Chu CC, Lin CN, Chang CC and Teng CM: Isoorientin-6′-O-glucoside, a water-soluble antioxidant isolated from Gentiana arisanensis. Biochim Biophys Acta. 1389:81–90. 1998. View Article : Google Scholar : PubMed/NCBI

30 

Lu CL, Zhu W, Wang DM, Chen WL, Hu MM, Wang M, Xu XJ and Lu CJ: Inhibitory effects of chemical compounds isolated from the rhizome of Smilax glabra on nitric oxide and tumor necrosis factor-α production in lipopolysaccharide-induced RAW264.7 cell. Evid Based Complement Alternat Med. 2015:6024252015. View Article : Google Scholar : PubMed/NCBI

31 

Lee S, Jung SH, Lee YS, Yamada M, Kim BK, Ohuchi K and Shin KH: Antiinflammatory activity of hyperin from Acanthopanax chiisanensis roots. Arch Pharm Res. 27:628–632. 2004. View Article : Google Scholar : PubMed/NCBI

32 

Zhang B, Su JP, Bai Y, Li J and Liu YH: Inhibitory effects of O-methylated isoflavone glycitein on human breast cancer SKBR-3 cells. Int J Clin Exp Pathol. 8:7809–7817. 2015.PubMed/NCBI

33 

Matsukawa Y, Marui N, Sakai T, Satomi Y, Yoshida M, Matsumoto K, Nishino H and Aoike A: Genistein arrests cell cycle progression at G2-M. Cancer Res. 53:1328–1331. 1993.PubMed/NCBI

34 

Chen J, Zhao X, Ye Y, Wang Y and Tian J: Estrogen receptor beta-mediated proliferative inhibition and apoptosis in human breast cancer by Calycosin and Formononetin. Cell Physiol Biochem. 32:1790–1797. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Auyeung KK, Law PC and Ko JK: Novel anti-angiogenic effects of formononetin in human colon cancer cells and tumor xenograft. Oncol Rep. 28:2188–2194. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Gao Y, Fang L, Liu F, Zong C, Cai R, Chen X and Qi Y: Suppressive effects of irisflorentin on LPS-induced inflammatory responses in RAW 264.7 macrophages. Exp Biol Med (Maywood). 239:1018–1024. 2014. View Article : Google Scholar : PubMed/NCBI

37 

Effect of Dichotomitin on relieving cough induced by cigarette and infection and serum cytokines of model guinea pigs. Zhonghua Zhongyiyao Xuekan. 34:2902–2904. 2016.

38 

Jun HJ, Hoang MH, Lee JW, Yaoyao J, Lee JH, Lee DH, Lee HJ, Seo WD, Hwang BY and Lee SJ: Iristectorigenin B isolated from Belamcanda chinensis is a liver X receptor modulator that increases ABCA1 and ABCG1 expression in macrophage RAW 264.7 cells. Biotechnol Lett. 34:2213–2221. 2012. View Article : Google Scholar : PubMed/NCBI

39 

Chen CC, Chen CY, Ueng SH, Hsueh C, Yeh CT, Ho JY, Chou LF and Wang TH: Corylin increases the sensitivity of hepatocellular carcinoma cells to chemotherapy through long noncoding RNA RAD51-AS1-mediated inhibition of DNA repair. Cell Death Dis. 9:5432018. View Article : Google Scholar : PubMed/NCBI

40 

Martineti V, Tognarini I, Azzari C, Carbonell Sala S, Clematis F, Dolci M, Lanzotti V, Tonelli F, Brandi ML and Curir P: Inhibition of in vitro growth and arrest in the G0/G1 phase of HCT8 line human colon cancer cells by kaempferide triglycoside from Dianthus caryophyllus. Phytother Res. 24:1302–1308. 2010. View Article : Google Scholar : PubMed/NCBI

41 

Lin CM, Huang ST, Liang YC, Lin MS, Shih CM, Chang YC, Chen TY and Chen CT: Isovitexin suppresses lipopolysaccharide-mediated inducible nitric oxide synthase. Planta Med. 71:748–753. 2005. View Article : Google Scholar : PubMed/NCBI

42 

Zygmunt K, Faubert B, MacNeil J and Tsiani E: Naringenin, a citrus flavonoid, increases muscle cell glucose uptake via AMPK. Biochem Biophys Res Commun. 398:178–183. 2010. View Article : Google Scholar : PubMed/NCBI

43 

Niu X, Xing W, Li W, Fan T, Hu H and Li Y: Isofraxidin exhibited anti-inflammatory effects in vivo and inhibited TNF-α production in LPS-induced mouse peritoneal macrophages in vitro via the MAPK pathway. Int Immunopharmacol. 14:164–171. 2012. View Article : Google Scholar : PubMed/NCBI

44 

Shyu MH, Kao TC and Yen GC: Oleanolic acid and ursolic acid induce apoptosis in HuH7 human hepatocellular carcinoma cells through a mitochondrial-dependent pathway and downregulation of XIAP. J Agric Food Chem. 58:6110–6118. 2010. View Article : Google Scholar : PubMed/NCBI

45 

Ko BS, Choi SB, Park SK, Jang JS, Kim YE and Park S: Insulin densitizing and insulinotropic sction of Berberine from Cortidis Rhizoma. Biol Pharm Bull. 28:1431–1437. 2005. View Article : Google Scholar : PubMed/NCBI

46 

Tang FY, Su YC, Chen NC, Hsieh HS and Chen KS: Resveratrol inhibits migration and invasion of human breast-cancer cells. Mol Nutr Food Res. 52:683–691. 2008. View Article : Google Scholar : PubMed/NCBI

47 

Fryer LG, Parbu-Patel A and Carling D: The Anti-diabetic drugs rosiglitazone and metformin stimulate AMP-activated protein kinase through distinct signaling pathways. J Biol Chem. 277:25226–25232. 2002. View Article : Google Scholar : PubMed/NCBI

48 

Cui Q, Tashiro S, Onodera S, Minami M and Ikejima T: Autophagy preceded apoptosis in oridonin-treated human breast cancer MCF-7 cells. Biol Pharm Bull. 30:859–864. 2007. View Article : Google Scholar : PubMed/NCBI

49 

Miyoshi K, Kasahara K, Miyazaki I and Asanuma M: Factors that influence primary cilium length. Acta Med Okayama. 65:279–185. 2011.PubMed/NCBI

50 

Li X, Liu Y, Hou X, Peng H, Zhang L, Jiang Q, Shi M, Ji Y, Wang Y and Shi W: Chlorogenic acid inhibits the replication and viability of enterovirus 71 in vitro. PLoS One. 8:e760072013. View Article : Google Scholar : PubMed/NCBI

51 

Zhang CJ and Yu HT: The signal pathways of immune inflammation mediated By The Tlr3/Nf-Kappab and activator protein-1 in cells infected with influenza A virus antagonized by Baicalin. Adv Mat Res. 345:201–209. 2012.

52 

Ida C, Ogata S, Okumura K and Taguchi H: Changes in the gene expression of C-myc and CD38 in HL-60 cells during differentiation induced by nicotinic acid-related compounds. Biosci Biotechnol Biochem. 72:868–871. 2008. View Article : Google Scholar : PubMed/NCBI

53 

Sun C, Jiang M, Zhang L, Jian Y, Zhang G, Du B, Ren Y, Li X and Yao J: Cycloastragenol mediates activation and proliferation suppression in concanavalin A-induced mouse lymphocyte pan-activation model. Immunopharmacol Immunotoxicol. 39:131–139. 2017. View Article : Google Scholar : PubMed/NCBI

54 

Zhou J, Sun YY, Sun MY, Mao WA, Wang L, Zhang J and Zhang H: Prim-O-glucosylcimifugin attenuates lipopolysaccharide induced inflammatory response in RAW 264.7 macrophages. Pharmacogn Mag. 13:378–384. 2017. View Article : Google Scholar : PubMed/NCBI

55 

Liu TZ, Chen CY, Yiin SJ, Chen CH, Cheng JT, Shih MK, Wang YS and Chern CL: Molecular mechanism of cell cycle blockage of hepatoma SK-Hep-1 cells by Epimedin C through suppression of mitogen-activated protein kinase activation and increased expression of CDK inhibitors p21(Cip1) and p27(Kip1). Food Chem Toxicol. 44:227–235. 2006. View Article : Google Scholar : PubMed/NCBI

56 

Li J, Hao L, Wu J, Zhang J and Su J: Linarin promotes osteogenic differentiation by activating the BMP-2/RUNX2 pathway via protein kinase A signaling. Int J Mol Med. 37:901–910. 2016. View Article : Google Scholar : PubMed/NCBI

57 

Zhou J, Hu H, Long J, Wan F, Li L, Zhang S, Shi YE and Chen Y: Vitexin 6, a novel lignan, induces autophagy and apoptosis by activating the Jun N-terminal kinase pathway. Anticancer Drugs. 24:928–936. 2013. View Article : Google Scholar : PubMed/NCBI

58 

Lu J, Papp LV, Fang J, Rodriguez-Nieto S, Zhivotovsky B and Holmgren A: Inhibition of mammalian thioredoxin reductase by some flavonoids: Implications for myricetin and quercetin anticancer activity. Cancer Res. 66:4410–4418. 2006. View Article : Google Scholar : PubMed/NCBI

59 

Li L, Sapkota M, Kim SW and Soh Y: Herbacetin inhibits inducible nitric oxide synthase via JNK and nuclear factor-κB in LPS-stimulated RAW264.7 cells. Eur J Pharmacol. 765:115–123. 2015. View Article : Google Scholar : PubMed/NCBI

60 

Cao S, Ni B, Feng L, Yin X, Dou H, Fu J, Lin L and Ni J: Simultaneous determination of typhaneoside and isorhamnetin-3-O-neohesperidoside in rats after oral administration of pollen Typhae extract by UPLC-MS/MS. J Chromatogr Sci. 53:866–871. 2015. View Article : Google Scholar : PubMed/NCBI

61 

Shin SW, Jung E, Kim S, Kim JH, Kim EG, Lee J and Park D: Antagonizing effects and mechanisms of Afzelin against UVB-induced cell damage. PLoS One. 8:e619712013. View Article : Google Scholar : PubMed/NCBI

62 

Guo H, Zhang X, Cui Y, Zhou H, Xu D, Shan T, Zhang F, Guo Y, Chen Y and Wu D: Taxifolin protects against cardiac hypertrophy and fibrosis during biomechanical stress of pressure overload. Toxicol Appl Pharmacol. 287:168–177. 2015. View Article : Google Scholar : PubMed/NCBI

63 

Chu SC, Chiou HL, Chen PN, Yang SF and Hsieh YS: Silibinin inhibits the invasion of human lung cancer cells via decreased productions of urokinase-plasminogen activator and matrix metalloproteinase-2. Mol Carcinog. 40:143–149. 2004. View Article : Google Scholar : PubMed/NCBI

64 

Sugimoto E and Yamaguchi M: Stimulatory effect of Daidzein in osteoblastic MC3T3-E1 cells. Biochem Pharmacol. 59:471–475. 2000. View Article : Google Scholar : PubMed/NCBI

65 

Lee KT, Sohn IC, Kim YK, Choi JH, Choi JW, Park HJ, Itoh Y and Miyamoto K: Tectorigenin, an isoflavone of Pueraria thunbergiana Benth., induces differentiation and apoptosis in human promyelocytic leukemia HL-60 cells. Biol Pharm Bull. 24:1117–1121. 2001. View Article : Google Scholar : PubMed/NCBI

66 

Kim PS, Shin JH, Jo DS, Shin DW, Choi DH, Kim WJ, Park K, Kim JK, Joo CG, Lee JS, et al: Anti-melanogenic activity of schaftoside in Rhizoma Arisaematis by increasing autophagy in B16F1 cells. Biochem Biophys Res Commun. 503:309–315. 2018. View Article : Google Scholar : PubMed/NCBI

67 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

68 

Davignon J and Dubuc G: Statins and ezetimibe modulate plasma proprotein convertase subtilisin kexin-9 (PCSK9) levels. Trans Am Clin Climatol Assoc. 120:163–173. 2009.PubMed/NCBI

69 

Welder G, Zineh I, Pacanowski MA, Troutt JS, Cao GQ and Konrad RJ: High-dose atorvastatin causes a rapid sustained increase in human serum PCSK9 and disrupts its correlation with LDL cholesterol. J Lipid Res. 51:2714–2721. 2010. View Article : Google Scholar : PubMed/NCBI

70 

Pal S, Ghosh M, Ghosh S, Bhattacharyya S and Sil PC: Atorvastatin induced hepatic oxidative stress and apoptotic damage via MAPKs, mitochondria, calpain and caspase12 dependent pathways. Food Chem Toxicol. 83:36–47. 2015. View Article : Google Scholar : PubMed/NCBI

71 

Puccetti L, Acampa M and Auteri A: Pharmacogenetics of statins therapy. Recent Pat Cardiovasc Drug Discov. 2:228–236. 2007. View Article : Google Scholar : PubMed/NCBI

72 

Taylor BA and Thompson PD: Statins and their effect on PCSK9-impact and clinical relevance. Curr Atheroscler Rep. 18:462016. View Article : Google Scholar : PubMed/NCBI

73 

Cameron J, Ranheim T, Kulseth MA, Leren TP and Berge KE: Berberine decreases PCSK9 expression in HepG2 cells. Atherosclerosis. 201:266–273. 2008. View Article : Google Scholar : PubMed/NCBI

74 

Kumar A, Ekavali, Chopra K, Mukherjee M, Pottabathini R and Dhull DK: Current knowledge and pharmacological profile of berberine: An update. Eur J Pharmacol. 761:288–297. 2015. View Article : Google Scholar : PubMed/NCBI

75 

Lan J, Zhao Y, Dong F, Yan Z, Zheng W, Fan J and Sun G: Meta-analysis of the effect and safety of berberine in the treatment of type 2 diabetes mellitus, hyperlipemia and hypertension. J Ethnopharmacol. 161:69–81. 2015. View Article : Google Scholar : PubMed/NCBI

76 

Wu JW, Lin LC and Tsai TH: Drug-drug interactions of silymarin on the perspective of pharmacokinetics. J Ethnopharmacol. 121:185–193. 2009. View Article : Google Scholar : PubMed/NCBI

77 

Flaig TW, Gustafson DL, Su LJ, Zirrolli JA, Crighton F, Harrison GS, Pierson AS, Agarwal R and Glodé LM: A phase I and pharmacokinetic study of silybin-phytosome in prostate cancer patients. Invest New Drugs. 25:139–146. 2007. View Article : Google Scholar : PubMed/NCBI

78 

Stolf AM, Cardoso CC and Acco A: Effects of silymarin on diabetes mellitus complications: A review. Phytother Res. 31:366–374. 2017. View Article : Google Scholar : PubMed/NCBI

79 

Lennernäs H: Clinical pharmacokinetics of atorvastatin. Clin Pharmacokinet. 42:1141–1160. 2003. View Article : Google Scholar : PubMed/NCBI

80 

Wu JW, Lin LC and Tsai TH: Drug-drug interactions of silymarin on the perspective of pharmacokinetics. J Ethnopharmacol. 121:185–193. 2009. View Article : Google Scholar : PubMed/NCBI

81 

Pang Y, Zhu H, Xu J, Yang L, Liu L and Li J: β-arrestin-2 is involved in irisin-induced glucose metabolism in type 2 diabetes via p38 MAPK signaling. Exp Cell Res. 360:199–204. 2017. View Article : Google Scholar : PubMed/NCBI

82 

Cui CJ, Li S, Zhu CG, Sun J, Du Y, Zhang Y, Wu NQ, Guo YL, Xu RX, Gao Y and Li JJ: Enhanced pro-protein convertase subtilisin/kexin type 9 expression by C-reactive protein through p38MAPK-HNF1α pathway in HepG2 cells. J Cell Mol Med. 20:2374–2383. 2016. View Article : Google Scholar : PubMed/NCBI

83 

Du Y, Li S, Cui CJ, Zhang Y, Yang SH and Li JJ: Leptin decreases the expression of low-density lipoprotein receptor via PCSK9 pathway: Linking dyslipidemia with obesity. J Transl Med. 14:2762016. View Article : Google Scholar : PubMed/NCBI

84 

Li J, Huang Q, Long X, Zhang J, Huang X, Aa J, Yang H, Chen Z and Xing J: CD147 reprograms fatty acid metabolism in hepatocellular carcinoma cells through Akt/mTOR/SREBP1c and P38/PPARα pathways. J Hepatol. 63:1378–1389. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2019
Volume 20 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Dong Z, Zhang W, Chen S and Liu C: Silibinin A decreases statin‑induced PCSK9 expression in human hepatoblastoma HepG2 cells. Mol Med Rep 20: 1383-1392, 2019
APA
Dong, Z., Zhang, W., Chen, S., & Liu, C. (2019). Silibinin A decreases statin‑induced PCSK9 expression in human hepatoblastoma HepG2 cells. Molecular Medicine Reports, 20, 1383-1392. https://doi.org/10.3892/mmr.2019.10344
MLA
Dong, Z., Zhang, W., Chen, S., Liu, C."Silibinin A decreases statin‑induced PCSK9 expression in human hepatoblastoma HepG2 cells". Molecular Medicine Reports 20.2 (2019): 1383-1392.
Chicago
Dong, Z., Zhang, W., Chen, S., Liu, C."Silibinin A decreases statin‑induced PCSK9 expression in human hepatoblastoma HepG2 cells". Molecular Medicine Reports 20, no. 2 (2019): 1383-1392. https://doi.org/10.3892/mmr.2019.10344