Articles Service
Research article
Schisandrin A in Schisandra chinensis Upregulates the LDL Receptor by Inhibiting PCSK9 Protein Stabilization in Steatotic Model
1Personalized Diet Research Group, Korea Food Research Institute, Wanju 55365, Republic of Korea
2Department of Food Biotechnology, University of Science & Technology, Daejeon 34113, Republic of Korea
3Department of Food and Nutrition, Gangseo University, Seoul 07661, Republic of Korea
J. Microbiol. Biotechnol. 2024; 34(2): 425-435
Published February 28, 2024 https://doi.org/10.4014/jmb.2306.06049
Copyright © The Korean Society for Microbiology and Biotechnology.
Abstract
Keywords
Graphical Abstract
Introduction
Cardiovascular disease (CVD) is one of the most significant causes of mortality and morbidity worldwide. Over the last ten years, the number of deaths due to CVD has increased by 12.5% globally [1, 2], with atherosclerosis being the leading cause of vascular disease and death. Atherosclerosis is caused by abnormal lipid metabolism, which leads to the accumulation of fatty streaks in the arterial layer, causing thickening and hardening of the arterial wall and resulting in narrowing of the vascular lumen [3]. Blood cholesterol, especially low-density lipoprotein (LDL), is a key driver of atherosclerosis [4]. Meta-analyses of large prospective observational studies have investigated the relationship between the risk of ischemic heart disease (IHD) and various lipid measures, including total cholesterol (TCHO), high-density lipoprotein (HDL) cholesterol, non-HDL cholesterol, triglycerides, and apolipoproteins [4-6]. These data suggest the existence of continuous positive log-linear relationships between normal blood levels of either total or LDL cholesterol (non-HDL cholesterol) and the risk of IHD. Meanwhile, a clear inverse correlation between HDL cholesterol and IHD risk has been found [5, 6].
Dietary intake of fat and cholesterol is an important environmental determinant of blood cholesterol levels, which can also be influenced by other factors, including exercise and genetics [7-10]. The consumption of dietary components that inhibit proprotein convertase subtilisin/kexin 9 (PCSK9) can positively influence lipid metabolism during the development and progression of atherosclerosis [11]. The role of PCSK9 in lipid metabolism has been extensively studied. PCSK9 binds to the LDL receptor, causing its degradation, thereby increasing circulating LDL levels in the blood. Cohort studies have demonstrated that circulating PCSK9 is a viable therapeutic target for reducing plasma LDL cholesterol levels to aid in the prevention of CVD [12, 13]. Observation of a large sample of healthy participants in the Brisighella Heart Cohort found that circulating PCSK9 levels were associated with increased arterial stiffness, and therefore potentially associated also with LDL cholesterol [13].
PCSK9 inhibitors bind to LDL receptors on the surface of liver cells, promoting receptor degradation and interfering with the clearance of LDL cholesterol from the bloodstream [14]. To date, specific natural substances that directly inhibit PCSK9 have not yet been reported. Widely known PCSK9 inhibitors are primarily synthetic compounds, such as evolocumab and alirocumab, but ongoing studies are exploring their safety [15]. The search for specific components of natural substances that directly inhibit PCSK9 remains an ongoing area of research and future findings may lead to new discoveries.
Materials and Methods
Dulbecco’s modified Eagle’s medium (DMEM) and antibiotic antimycotic solution (100X) were purchased from Welgene Inc. (Republic of Korea). Delipidated serum (DLPS) was prepared from fetal bovine serum (FBS; Thermo Fisher Scientific, USA) as described previously [19, 20]. Isopropanol and chloroform were obtained from EMD Millipore (USA). Cycloheximide (CHX), bafilomycin A1 (BA1), 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), and MG132 were purchased from Sigma-Aldrich (USA). Dimethyl sulfoxide (DMSO) was purchased from Duchefa Biochemie (Netherlands).
SCE Preparation
The fruit of
In Vivo Model
The animal study was approved by the Institutional Animal Care and Use Committee at Wonkwang University (WKU16-21). Male C57BL/6 mice (7-week-old) were purchased from SAMTAKO Bio Korea (Republic of Korea) and acclimated for 1 week in an environmentally controlled facility. At 8 weeks of age, mice were randomized to control diet (CD; #98121701, Research Diets Inc., USA), WD (#D12079B, Research Diets INC), or WD formulated with 1% SCE (w/w;
Serum Analysis
Blood samples were stored at 4°C for 2 h and centrifuged before serum was collected. Serum lipids (triglyceride, total cholesterol and LDL cholesterol) and serum hepatic injury markers (alanine transaminase (ALT) and aspartate aminotransferase (AST)) were analyzed using enzymatic colorimetric assay kits (
Cell Culture and Treatment
HepG2 cells (HB-8065) were purchased from the American Type Culture Collection (ATCC, USA), and maintained in DMEM containing 1% antibiotics and 10% (v/v) FBS at 37°C in a humidified atmosphere of 5% CO2 in an incubator. For sample (extract or chemicals) treatments, HepG2 cells were seeded (1.5 × 105 cells/ml) in DMEM containing 1% antibiotics and 10% FBS (day 0). On the following day, cells were washed with DPBS twice, and the medium was changed to DMEM containing 1% antibiotics and 10% DLPS (day 1). After 24 h incubation, the medium was replaced with fresh DMEM containing 1% antibiotics and 10% DLPS supplemented with appropriate levels of SCE or schisandrin A (SA) up to 100 μM (day 2). Finally, 18 h later, cells were harvested for further analysis following washing with DPBS.
Western Blotting
To evaluate hepatic protein expression, liver samples were homogenized with radioimmunoprecipitation assay (RIPA) buffer (Elpis Biotech, Inc., Republic of Korea) containing protease and phosphatase inhibitor (Roche), before being placed on ice for 30 min with intermittent vortexing. Following centrifugation (20,000 ×
Collected whole cell lysates were then used to analyze total protein content using a BCA kit (Pierce, USA). After quantification, 20 μg of protein was separated by 10% SDS-PAGE. Proteins were transferred to a nitrocellulose membrane (GE Healthcare BioScience, USA). The membrane was blocked in PBS containing 0.05% Tween 20 and 5% non-fat dried milk for 1 h at room temperature, and then incubated in blocking solution with primary antibodies against PCSK9 (Circulex, Medical & Biological Laboratories Co., Ltd., USA), LDLR (BioVision Inc., Milpitas, CA, USA), SREBP2 (Cayman Chemical, USA), hepatic nuclear factor (HNF)-1α (Cell Signaling Technology, USA) and β-actin (Bethyl Laboratories, USA) for 1 h at room temperature. After washing with PBS containing 0.05% Tween 20 and 5% non-fat dried milk, the membrane was incubated in horseradish peroxidase secondary antibody (Thermo Scientific, USA). Target proteins were visualized with a FUSION SOLO 6S imaging system (Vilber Lourmat, AZC de Lamirault, France) with an enhanced chemiluminescence detection reagent (Thermo Scientific). Target protein expression was normalized to β-actin.
Cell Viability
To examine viability of the HepG2 cells, a colorimetric assay using MTT was performed. To determine non-toxic concentrations for the cells, HepG2 cells were cultured with SCE (200, 400 μg/ml) or SA (10‒100 μM) in DMEM containing 10% DLPS. After an additional 18 h incubation, MTT solution (final concentration: 0.5 mg/mL) was added in each well. After a further 2 h incubation, a mixture of isopropanol and DMSO (1:1) was added to the wells before incubation for 5 min with gentle shaking. Sample absorbance was measured at 570 nm using a microplate reader (SpectraMax M2 microplate reader; Molecular Devices, USA).
Enzyme-Linked Immunosorbent Assay (ELISA)
HepG2 cells were cultured as described in Cell Culture and Treatment. Cells were treated with SCE or SA, and the plate was incubated for another 18 h. After incubation, both cell pellets and media were collected. Pellets were rinsed with DPBS and lysed with RIPA buffer (Elpis Biotech, Inc., Republic of Korea) containing protease and phosphatase inhibitor (Roche, Switzerland) in ice, before vortexing, centrifuging (15,000 ×
LDL Uptake Assay
To examine LDL uptake at the cellular level, HepG2 cells were seeded (3 × 104 cells/well) in 96-well plates and treated as previously described. We used an LDL uptake kit (Cayman Chemical) and performed the assay according to the manufacturer’s instructions. At the end of the treatment period, we added a fluorescent probe, DyLight 550, and incubated it for 12 h at 37°C in a humidified atmosphere of 5% CO2. Cells were rinsed and fixed for immunofluorescence staining. After washing with buffer, images were captured using a fluorescence microscope (IX71; Olympus, Japan).
Quantitative Real-Time Polymerase Chain Reaction (qRT-PCR)
HepG2 cells were maintained, seeded in 6-well plates, and cultured as described. On day 2, the cells were incubated with DMEM containing 1% antibiotics and 10% DLPS supplemented with SA (20, 40, and 80 μM), and incubated for an additional 18 h. Cells were washed with DPBS two times, and pellets were collected following centrifugation. Total RNA was extracted from pellets using TRIzol (Invitrogen, USA). cDNA was synthesized using a reverse transcription kit (Bio-Rad) and gene expression was quantified with SYBR Green PCR master mix (Thermo Fisher Scientific) on a Bio-Rad CFX384. Primers used for qRT-PCR are provided in Table 1. Target gene expression was quantified relative to GAPDH, using the 2-ΔΔCT method. All reactions were carried out in triplicate.
-
Table 1 . Sequence list of qRT-PCR primers.
Primer name Direction Sequence (5'->3') PCSK9 Forward GGCAGGTTGGCAGCTGTTT Reverse CGTGTAGGCCCCGAGTGT LDLR Forward AGGAGACGTGCTTGTCTGTC Reverse CTGAGCCGTTGTCGCAGT
Protein Stability
To evaluate PCSK9 protein stability regulated by SA, cells were treated simultaneously with SA and cycloheximide (CHX). In brief, HepG2 cells were cultured and seeded as described previously, followed by incubation in fresh DMEM supplemented with DLPS for 24 h (day 1). On the following day (day 2), cells were treated with SA at 0 or 80 μM. On day 3, cells were treated with protein synthesis inhibitor CHX (C4895, Sigma-Aldrich) at 5 μg/ml. Cells were then harvested at 0, 1, 2, 4, and 8 h post incubation with CHX. Western blotting was performed as described above using all the collected pellets followed by protein lysis.
To evaluate the effect of SA against lysosomal and proteasomal degradation inhibition, HepG2 cells were cultured and seeded (day 0) as described previously. On the following day (day 1), the cells were incubated with DMEM supplemented with 10% DLPS, followed by treatment (day 2) with chemicals [bafilomycin A1 (BA1; Sigma-Aldrich) at 0 or 50 nM or MG132 (Sigma-Aldrich) at 0 or 1 μM] and SA at 0 or 80 μM. Following 24 h incubation on day 3, the cells were washed with DPBS, harvested, and centrifuged, with the upper layer removed to collect whole cell lysate.
Filipin Staining Assay
To measure for cholesterol, HepG2 cells were cultured, seeded, and treated as described in Cell Culture and Treatment section. After 18 h of further incubation at day 2, cells were washed with DPBS 3 times, and fixed with 3.7% paraformaldehyde for 1 h. Following washing with DPBS 3 times, glycine solution dissolved in DPBS (100 μl) was added to each well to remove residual paraformaldehyde. Filipin solution (50 μg/ml) was then added to each well and incubated for 2 h at room temperature, followed by rinsing with DPBS 3 times. Fluorescence intensity was measured at 360/480 nm (excitation/emission) using a SpectraMax M2 microplate reader (Molecular Devices).
Statistical Analysis
Data (means ± standard error of mean (SEM)) were analyzed using GraphPad Prism software (version 7.0; USA). Student’s
Results
SCE Supplementation Attenuates WD-Induced Increases in Body Weight, Serum Lipid Profiles, and Hepatic Injury Markers, as well as LDLR Protein Expression
SCE (1%, w/w) supplementation significantly attenuated the final body weight of WD-fed obese mice compared with that of the controls (Fig. 1A). Twelve weeks of SCE supplementation also significantly reduced total cholesterol, LDL cholesterol, and TG levels in the serum compared with WD feeding in obese mice (Fig. 1B). To examine the effect of WD and low-fat diet (LD) feeding for 12 weeks on liver injury in mice, serum ALT and AST activities were measured. We observed that the WD significantly increased both ALT and AST activity, which were significantly decreased by SCE supplementation (Fig. 1C). ALT and AST activities in the serum were increased approximately 5- and 2-fold by WD feeding, respectively, and were almost normalized in the LD group with SCE supplementation.
-
Fig. 1.
Schisandra chinensis extract (SCE) attenuates final body weight, serum lipid profiles, hepatic injury markers, and low-density lipoprotein receptor (LDLR) expression in liver tissues of Western diet (WD)-fed obese mice. Changes in body weight (A) serum lipid profiles (TG, TCHO, and LDL-C) (B) serum hepatic injury markers (ALT and AST) (C) and LDLR protein expression (D) in the liver tissue of WD-fed obese mice. The results are expressed as the mean ± SEM. # indicates a significant difference compared to the low-fat diet (LD) group (#p < 0.05, ##p < 0.01, ###p < 0.001). * indicates a significant difference compared to the WD group (*p < 0.05, **p < 0.01, ***p < 0.001). LD, low-fat diet; WD+SCE, WD plusSchisandra chinensis extract, TG; triglyceride, TCHO; total cholesterol, LDL-C; LDL-cholesterol, ALT; alanine transaminase, AST; aspartate aminotransferase.
LDLR is a key regulator of cholesterol metabolism. LDLR knockout (Ldlr-/-) Leiden mice are widely used to study dyslipidemia, obesity, and nonalcoholic steatohepatitis following high-fat diet feeding [21, 22]. Therefore, we examined whether the SCE-mediated reduction in LDL cholesterol levels were attributable to LDLR upregulation in the livers of these obese mice (Fig. 1D). We found that WD feeding markedly attenuated LDLR protein expression, which was reversed by SCE supplementation. LDLR protein expression in the WD+SCE group was similar to that observed in LD controls. Therefore, we deduced that SCE increased LDLR protein expression, contributing to reduced serum LDL cholesterol levels in obese mice.
SCE Increases LDLR Protein Expression and Reduces PCSK9 Secretion in DLPS-Treated HepG2 Cells
The regulation of hypercholesterolemia, attributed to LDLR upregulation by SCE treatment, was further examined in HepG2 cells. DLPS was used to simulate lipid-deprived conditions for the purpose of PCSK9 upregulation [19, 20]. SCE, at concentrations up to 400 μg/ml, did not affect the viability of DLPS-treated HepG2 cells (Fig. 2A). We then measured LDLR protein expression in DLPS-treated cells and found that SCE at a concentration of 200 ug/ml, markedly increased LDLR expression compared to the DLPS-treated cells (Fig. 2B). Next, we measured PCSK9 secretion using ELISA. DLPS significantly increased PCSK9 secretion, which was significantly reduced by SCE at concentrations of 100 and 200 μg/ml (Fig. 2C). These findings suggest that SCE inhibits PCSK9 secretion, thereby preventing LDLR degradation and likely contributing to the reduction of free cholesterol levels in HepG2 cells.
-
Fig. 2.
Schisandra chinensis extract increases low-density lipoprotein receptor (LDLR) expression and inhibits proprotein convertase subtilisin/kexin 9 (PCSK9) secretion in delipidated serum (DLPS)-induced lipid-deprived conditions. Cell viability (A) LDLR protein expression in HepG2 cells (B) and PCSK9 levels in the media (C). The results are expressed as the mean ± SEM. # indicates a significant difference compared to the normal control (#p < 0.05, ##p < 0.01, ###p < 0.001). * indicates a significant difference compared to DLPS-treated HepG2 cells (*p < 0.05, **p < 0.01, ***p < 0.001). FBS, fetal bovine serum; SCE,Schisandra chinensis extract.
Schisandrin A Induces LDLR Upregulation and PCSK9 Inhibition in DLPS-Treated HepG2 Cells
We examined the components of SCE to determine which of them contribute to the observed hypocholesterolemic activity of PCSK9 inhibition. SA is a major bioactive component of
-
Fig. 3. Schisandrin A (SA) increases low-density lipoprotein (LDL) uptake in delipidated serum (DLPS)- treated HepG2 cells.
Chemical structure of SA (A) cell viability (B) and representative immunofluorescence images and fluorescence intensity of the LDL-cholesterol uptake assay (C) in HepG2 cells. The results are expressed as the mean ± SEM. # indicates a significant difference compared to the normal control (#
p < 0.05, ##p < 0.01, ###p < 0.001). * indicates a significant difference compared to DLPS-treated HepG2 cells (*p < 0.05, **p < 0.01, ***p < 0.001).
SA Significantly Regulates LDLR/PCSK9 Protein Expression but Not PCSK9 Gene Expression
LDLR and PCSK9 protein expression levels were confirmed using western blot analysis. SA (10, 20, 40, and 80 μM) increased LDLR protein expression, and 80 μM SA inhibited PCSK9 expression in DLPS-treated HepG2 cells (Fig. 4A).
-
Fig. 4. Schisandrin A (SA) regulates low-density lipoprotein receptor (LDLR)- proprotein convertase subtilisin/kexin 9 (PCSK9) protein expression but is not associated with their gene expression or the upregulation of hepatic nuclear factor (HNF)-1α and SREBP2 protein expression in delipidated serum (DLPS)-treated HepG2 cells.
LDLR and PCSK9 protein expression (A) LDLR and PCSK9 gene expression (B) and SREBP2 and HNF-1α protein expression (C) in HepG2 cells. The results are expressed as the mean ± SEM. # indicates a significant difference compared to the normal control (#
p < 0.05, ##p < 0.01, ###p < 0.001). * indicates a significant difference compared to DLPS-treated HepG2 cells (*p < 0.05, **p < 0.01, ***p < 0.001). n.s., no significant difference.
In HepG2 cells, LDLR gene expression was significantly increased by DLPS treatment, which was in turn significantly increased by treatment with SA (80 μM) (Fig. 4B). DLPS-mediated increases in PCSK9 gene expression were not affected by any level of SA treatment (Fig. 4B).
LDLR-PCSK9 Protein Modulation by SA Is Not associated with HNF-1α or SREBP2 Regulation in HepG2 Cells
The expression levels of SREBP2 and HNF-1α were examined by western blotting. We found that DLPS treatment downregulated SREBP2 protein expression, while SA slightly downregulated SREBP2 (Fig. 4C). Hepatic nuclear factor HNF-1α is a transcription factor that regulates PCSK9 independently of LDLR [23]. We found that HNF-1α protein expression was unaffected by DLPS or SA treatments (Fig. 4C). These findings suggested that the hypocholesterolemic effect of SA was not associated with the regulation of transcription factors that mediate LDLR and PCSK9 protein expression.
SA Induces PCSK9 Destabilization, thereby Preventing LDLR Lysosomal Degradation in HepG2 Cells
To examine PCSK9 protein stabilization by SA treatment, cells were simultaneously treated with CHX, an inhibitor of new protein synthesis, and SA. When PCSK9 translation was blocked, PCSK9 protein expression reduced in a time-dependent manner up to 8 h, which was also consistently observed with 80 μM of SA treatment (Fig. 5A), suggesting that SA does not contribute to stabilization of the PCSK9 protein.
-
Fig. 5. Schisandrin A (SA) effectively blocks proprotein convertase subtilisin/kexin 9 (PCSK9) secretion in delipidated serum (DLPS)-treated HepG2 cells.
PCSK9 protein expression in the presence or absence of cycloheximide (CHX) (A) bafilomycin A1 (BA1) (B) and MG132 (C) in HepG2 cells. Released PCSK9 levels into the medium (D) and free low-density lipoprotein-cholesterol levels in the medium (E). The results are expressed as the mean ± SEM. # indicates a significant difference compared to the normal control (#
p < 0.05, ##p < 0.01, ###p < 0.001). * indicates a significant difference compared to DLPS-treated HepG2 cells (*p < 0.05, **p < 0.01, ***p < 0.001).
Circulating PCSK9 binds to LDLR and is transported to lysosomes for degradation [24]. As expected, treatment with the lysosomal degradation inhibitor, BA1, increased PCSK9 protein expression in DLPS-treated cells. However, SA enhanced the BA1-mediated increase in PCSK9 protein expression in DLPS-treated HepG2 cells (Fig. 5B). These results indicated that SA contributes to the stabilization of PCSK9 protein binding to LDLR in lysosomes.
PCSK9 stabilization by SA was further evaluated using the proteasome degradation inhibitor MG132 in DLPS-treated HepG2 cells (Fig. 5C). SA co-treated with MG132 markedly decreased PCSK9 protein levels, suggesting that SA destabilizes free PCSK9 in HepG2 cells treated with DLPS.
Next, we sought to confirm whether SA caused the destabilization of free PCSK9, thereby blocking PCSK9 secretion. Levels of PCSK9 secreted into the medium were measured using ELISA. DLPS was found to significantly increase PCSK9 secretion, which was in turn significantly reduced by SA (80 μM) treatment in HepG2 cells (Fig. 5D). SA also reduced free LDL-cholesterol (LDL-C) levels in DLPS-treated HepG2 cells (Fig. 5E). Taken together, these observations support the previous finding that SA destabilizes free PCSK9, leading to the attenuation of circulating PCSK9, thereby protecting LDLR from protein degradation and subsequently reducing cholesterol levels in HepG2 cells.
Discussion
Cholesterol homeostasis is an essential, yet complex, aspect of metabolic regulation [25, 26]. Dietary cholesterol is absorbed into the intestine, whereas endogenous cholesterol is synthesized in a number of tissues, primarily in the liver. Lipoprotein particles play an important role in maintaining cholesterol homeostasis in the liver. Lipoproteins belong to several categories, including chylomicrons, very low-density lipoproteins (VLDL), LDL, and HDL.
Chylomicrons transport esterified dietary cholesterol from the intestine to the liver. VLDL is produced in the liver by combining triglycerides (TG) and cholesterol ester (CE) with apolipoproteins and other lipoproteins, and secreted into the bloodstream to be transported to peripheral tissues, such as muscle and adipose tissues. During this process, TG was removed from VLDL, converting to LDL containing more CE than TG [25, 27]. LDL is taken up by tissue expressing the apo B-containing receptor, and the CE transported by LDL becomes available for use by cells. Excess LDL (or oxidized LDL) often accumulates in cells via macrophages to form foam cells, which are a primary component of plaques during the development of atherosclerosis. Oxidized LDL is taken up by lesional macrophages before delivery to lysosomes, where oxidized LDL is hydrolyzed to free cholesterol by lysosomal acid lipase [28]. Free cholesterol can cause inflammation, is associated with various chronic diseases, and is transported from the lysosome to the endoplasmic reticulum, where it is esterified and stored as lipid droplets under normal conditions, whereas both CE and free cholesterol accumulate in cells where cholesterol trafficking occurs [29, 30]. Accumulated CE (as lipid droplets) causes the foamy morphology of macrophages, contributing to the production of atherosclerotic plaques. Together with the buildup of free cholesterol, which causes inflammation, these factors increase the risk of atherosclerosis [31].
Several strategies mediated by food compounds or diet plans can reduce circulating LDL cholesterol levels. In addition to enhancing LDLR expression, cholesterol excretion or absorption can be mediated when LDL cholesterol reaches critical levels. A meta-analysis of 14 randomized controlled trials with 1,136 individuals indicated that the consumption of green tea and its extracts significantly reduced fasting serum total cholesterol and LDL cholesterol levels (but not that of HDL cholesterol) in adults [32]. There are several intermediates through which flavonoids attenuate LDL cholesterol levels, which are often LDLR/PCSK9-independent. The underlying mechanism of PCSK9 inhibition may involve the upregulation of LDLR recycling and LDL uptake, providing a scientific basis for the prevention and treatment of steatosis. Therefore, our study focused on evaluating the potential of SCE and its constituents, which are natural substances, to regulate LDL levels via the LDLR-PCSK9 pathway.
Several food extracts with PCSK9 inhibitory activity have been identified. For instance, extracts of
In our study, SA reduced PCSK9 stabilization in the proteasome, a likely mechanism by which it contributes to the protection of LDLR against protein degradation by lysosomes (Fig. 5). Destabilization of PCSK9 aims to increase the availability of functional LDLRs and promote the clearance of LDL-C from the bloodstream. This can be beneficial for the management of high LDL-C levels, reducing the risk of CVDs and improving lipid profiles.
Taken together, these observations suggest that SA effectively blocks the free (not bound to LDLR) PCSK9 form, thereby recycling it back to the cell membrane of LDLR. Sun
Conclusively, SA, a natural bioactive compound found in
Acknowledgments
This study was supported by the Main Research Program of the Korea Food Research Institute (KFRI) and was funded by the Ministry of Science and ICT (E0210601-03).
Author Contributions
Conceptualization; S.K.P. and M.-Y.C., Methodology; M.-Y.C., Validation; J.-H.P. and S.H.P., Formal Analysis; H.-J.K., Investigation; H.-J.K. and S.K.P., Data Curation; S.H.P. and Y.G.L. Writing – Original Draft Preparation; H.-J.K. and S.K.P., Writing – Review & Editing; J.-T.H. and M.-Y.C., Visualization; Y.G.L, Supervision; J.-T.H. and M.-Y.C., Project Administration; J.-H.P. and J.-T.H.
Conflict of Interest
The authors have no financial conflicts of interest to declare.
References
- Joseph P, Leong D, McKee M, Anand SS, Schwalm JD, Teo K,
et al . 2017. Reducing the global burden of cardiovascular disease, part 1: the epidemiology and risk factors.Circ. Res. 121 : 677-694. - Roth GA, Johnson C, Abajobir A, Abd-Allah F, Abera SF, Abyu G,
et al . 2017. Global, regional, and national burden of cardiovascular diseases for 10 causes, 1990 to 2015.J. Am. Coll. Cardiol. 70 : 1-25. - Herrington W, Lacey B, Sherliker P, Armitage J, Lewington S. 2016. Epidemiology of atherosclerosis and the potential to reduce the global burden of atherothrombotic disease.
Circ. Res. 118 : 535-546. - Baigent C, Blackwell L, Emberson J, Holland LE, Reith C,
et al . 2010. Efficacy and safety of more intensive lowering of LDL cholesterol: a meta-analysis of data from 170,000 participants in 26 randomised trials.Lancet 376 : 1670-1681. - Lewington S, Whitlock G, Clarke R, Sherliker P, Emberson J,
et al . 2007. Blood cholesterol and vascular mortality by age, sex, and blood pressure: a meta-analysis of individual data from 61 prospective studies with 55,000 vascular deaths.Lancet 370 : 1829-1839. - Di Angelantonio E, Sarwar N, Perry P, Kaptoge S, Ray KK,
et al . 2009. Major lipids, apolipoproteins, and risk of vascular disease.JAMA 302 : 1993-2000. - Hegsted DM, Ausman LM, Johnson JA, Dallal GE. 1993. Dietary fat and serum lipids: an evaluation of the experimental data.
Am. J. Clin. Nutr. 57 : 875-883. - Keys A, Anderson JT, Grande F. 1957. Prediction of serum-cholesterol responses of man to changes in fats in the diet.
Lancet 273 : 959-966. - Clarke R, Frost C, Collins R, Appleby P, Peto R. 1997. Dietary lipids and blood cholesterol: quantitative meta-analysis of metabolic ward studies.
BMJ 314 : 112-117. - Dattilo AM, Kris-Etherton PM. 1992. Effects of weight reduction on blood lipids and lipoproteins: a meta-analysis.
Am. J. Clin. Nutr. 56 : 320-328. - Patriki D, Saravi SSS, Camici GG, Liberale L, Beer JH. 2021. PCSK 9: a link between inflammation and atherosclerosis.
Curr. Med. Chem. 29 : 251-267. - Leander K, Malarstig A, Van't Hooft FM, Hyde C, Hellenius ML, Troutt JS,
et al . 2016. Circulating proprotein convertase subtilisin/kexin type 9 (PCSK9) predicts future risk of cardiovascular events independently of established risk factors.Circulation 133 : 1230-1239. - Ruscica M, Ferri N, Fogacci F, Rosticci M, Botta M, Marchiano S,
et al . 2017. Circulating levels of proprotein convertase subtilisin/kexin type 9 and arterial stiffness in a large population sample: data from the brisighella heart study.J. Am. Heart Assoc. 6 : e005764. - Horton JD, Cohen JC, Hobbs HH. 2007. Molecular biology of PCSK9: Its role in LDL metabolism.
Trends Biochem. Sci. 32 : 71-77. - Xu S, Luo S, Zhu Z, Xu J. 2019. Small molecules as inhibitors of PCSK9: current status and future challenges.
Eur. J. Med. Chem. 162 : 212-233. - Szopa A, Ekiert R, Ekiert H. 2017. Current knowledge of
Schisandra chinensis (Turcz.) Baill.(Chinese magnolia vine) as a medicinal plant species: a review on the bioactive components, pharmacological properties, analytical and biotechnological studies.Phytochem. Rev. 16 : 195-218. - Kopustinskiene DM, Bernatoniene J. 2021. Antioxidant effects of
Schisandra chinensis fruits and their active constituents.Antioxidants 10 : 620. - Seo HJ, Ji SB, Kim SE, Lee GM, Moon SH, Jang DS,
et al . 2020. Analysis of dibenzocyclooctadiene lignans in Omija Wine and Cheong by liquid chromatography-tandem mass spectrometry.Mass. Spectrom. Lett. 11 : 30-35. - Jeong HJ, Lee HS, Kim KS, Kim YK, Yoon D, Park SW. 2008. Sterol-dependent regulation of proprotein convertase subtilisin/kexin type 9 expression by sterol-regulatory element binding protein-2.
J. Lipid. Res. 49 : 399-409. - Hwang JT, Choi E, Choi HK, Park JH, Chung MY. 2021. The cholesterol-lowering effect of
Capsella Bursa-Pastoris is mediated via SREBP2 and HNF-1alpha-regulated PCSK9 inhibition in obese mice and HepG2 cells.Foods 10 : 408. - Morrison MC, Verschuren L, Salic K, Verheij J, Menke A, Wielinga PY,
et al . 2018. Obeticholic acid modulates serum metabolites and gene signatures characteristic of human NASH and attenuates inflammation and fibrosis progression in Ldlr-/-. leiden mice.Hepatol. Commun. 2 : 1513-1532. - Tengeler AC, Gart E, Wiesmann M, Arnoldussen IAC, van Duyvenvoorde W, Hoogstad M,
et al . 2020. Propionic acid and not caproic acid, attenuates nonalcoholic steatohepatitis and improves (cerebro) vascular functions in obese Ldlr(-/-) .Leiden mice.FASEB J. 34 : 9575-9593. - Li H, Dong B, Park SW, Lee HS, Chen W, Liu J. 2009. 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. - Lambert G, Sjouke B, Choque B, Kastelein JJ, Hovingh GK. 2012. The PCSK9 decade.
J. Lipid. Res. 53 : 2515-2524. - Maxfield FR, Tabas I. 2005. Role of cholesterol and lipid organization in disease.
Nature 438 : 612-621. - Estronca LM, Filipe HA, Salvador A, Moreno MJ, Vaz WL. 2014. Homeostasis of free cholesterol in the blood: a preliminary evaluation and modeling of its passive transport.
J. Lipid. Res. 55 : 1033-1043. - Rothblat GH, Phillips MC. 2010. High-density lipoprotein heterogeneity and function in reverse cholesterol transport.
Curr. Opin. Lipidol. 21 : 229-238. - Xu X, Zhang A, Halquist MS, Yuan X, Henderson SC, Dewey WL,
et al . 2017. Simvastatin promotes NPC1-mediated free cholesterol efflux from lysosomes through CYP7A1/LXRalpha signalling pathway in oxLDL-loaded macrophages.J. Cell Mol. Med. 21 : 364-374. - Pfisterer SG, Peranen J, Ikonen E. 2016. LDL-cholesterol transport to the endoplasmic reticulum: current concepts.
Curr. Opin. Lipidol. 27 : 282-287. - Vanier MT. 2015. Complex lipid trafficking in Niemann-Pick disease type C.
J. Inherit. Metab. Dis. 38 : 187-199. - Tall AR, Yvan-Charvet L. 2015. Cholesterol, inflammation and innate immunity.
Nat. Rev. Immunol. 15 : 104-116. - Zheng XX, Xu YL, Li SH, Liu XX, Hui R, Huang XH. 2011. Green tea intake lowers fasting serum total and LDL cholesterol in adults: a meta-analysis of 14 randomized controlled trials.
Am. J. Clin. Nutr. 94 : 601-610. - Park JY, Shin HK, Lee YJ, Choi YW, Bae SS, Kim CD. 2009. The mechanism of vasorelaxation induced by
Schisandra chinensis extract in rat thoracic aorta.J. Ethnopharmacol. 121 : 69-73. - Brown MS, Goldstein JL. 1984. How LDL receptors influence cholesterol and atherosclerosis.
Sci. Am. 251 : 58-66. - Sirinian MI, Belleudi F, Campagna F, Ceridono M, Garofalo T, Quagliarini F,
et al . 2005. Adaptor protein ARH is recruited to the plasma membrane by low density lipoprotein (LDL) binding and modulates endocytosis of the LDL/LDL receptor complex in hepatocytes.J. Biol. Chem. 280 : 38416-38423. - Ragusa R, Basta G, Neglia D, De Caterina R, Del Turco S, Caselli C. 2021. PCSK9 and atherosclerosis: looking beyond LDL regulation.
Eur. J. Clin. Invest. 51 : e13459. - Macchi C, Sirtori C, Corsini A, Santos R, Watts G, Ruscica M. 2019. A new dawn for managing dyslipidemias: the era of RNA-based therapies.
Pharmacol. Res. 150 : 104413. - Choi HK, Hwang JT, Nam TG, Kim SH, Min DK, Park SW,
et al . 2017. Welsh onion extract inhibits PCSK9 expression contributing to the maintenance of the LDLR level under lipid depletion conditions of HepG2 cells.Food Funct. 8 : 4582-4591. - Choi HK, Kim HJ, Hwang JT, Chung MY. 2017.
Allium tuberosum reverses PCSK9-mediated LDLR degradation by inhibition of HNF1α.J. Korean Soc. Food Sci. Nutr. 46 : 1278-1285. - Wang F, Zhao C, Tian G, Wei X, Ma Z, Cui J,
et al . 2020. Naringin alleviates atherosclerosis in ApoE(-/-) mice by regulating cholesterol metabolism involved in gut microbiota remodeling.J. Agric. Food Chem. 68 : 12651-12660. - Hwang JT, Kim HJ, Choi HK, Park JH, Chung S, Chung MY. 2020. Butein synergizes with statin to upregulate low-density lipoprotein receptor through HNF1alpha-mediated PCSK9 inhibition in HepG2 cells.
J. Med. Food 23 : 1102-1108. - Cui CJ, Jin JL, Guo LN, Sun J, Wu NQ, Guo YL,
et al . 2020. Beneficial impact of epigallocatechingallate on LDL-C through PCSK9/LDLR pathway by blocking HNF1alpha and activating FoxO3a.J. Transl. Med. 18 : 1-13. - Jia Q, Cao H, Shen D, Li S, Yan L, Chen C,
et al . 2019. Quercetin protects against atherosclerosis by regulating the expression of PCSK9, CD36, PPARgamma, LXRalpha and ABCA1.Int. J. Mol. Med. 44 : 893-902. - Dong Z, Zhang W, Chen S, Liu C. 2019. Silibinin A decreases statininduced PCSK9 expression in human hepatoblastoma HepG2 cells.
Mol. Med. Rep. 20 : 1383-1392. - Gao WY, Chen PY, Chen SF, Wu MJ, Chang HY, Yen JH. 2018. Pinostrobin inhibits proprotein convertase subtilisin/kexin-type 9 (PCSK9) gene expression through the modulation of FoxO3a protein in HepG2 cells.
J. Agric. Food Chem. 66 : 6083-6093. - Razgonova M, Zakharenko A, Pikula K, Kim E, Chernyshev V, Ercisli S,
et al . 2020. Rapid mass spectrometric study of a supercritical CO2-extract from woody lianaSchisandra chinensis by HPLC-SPD-ESI-MS/MS.Molecules 25 : 2689. - Kim Y, Kim E, Jeong G. 2019. Isolation and quantitative analysis of schisandrin, gomisin A and gomisin M2 from
Schisandra chinensis .Korean J. Pharmacogn. 50 : 148-153. - Choi YH. 2018. Schisandrin A prevents oxidative stress-induced DNA damage and apoptosis by attenuating ROS generation in C2C12 cells.
Biomed. Pharmacother. 106 : 902-909. - Fu K, Zhou H, Wang C, Gong L, Ma C, Zhang Y,
et al . 2022. A review: pharmacology and pharmacokinetics of schisandrin A.Phytother. Res. 36 : 2375-2393. - Kwon DH, Cha HJ, Choi EO, Leem SH, Kim GY, Moon SK,
et al . 2018. Schisandrin A suppresses lipopolysaccharide-induced inflammation and oxidative stress in RAW 264.7 macrophages by suppressing the NF-κB, MAPKs and PI3K/Akt pathways and activating Nrf2/HO-1 signaling.Int. J. Mol. Med. 41 : 264-274. - Jeong MJ, Kim SR, Jung UJ. 2019. Schizandrin A supplementation improves nonalcoholic fatty liver disease in mice fed a high-fat and high-cholesterol diet.
Nutr. Res. 64 : 64-71. - Sun JH, Liu X, Cong LX, Li H, Zhang CY, Chen JG,
et al . 2017. Metabolomics study of the therapeutic mechanism ofSchisandra chinensis lignans in diet-induced hyperlipidemia mice.Lipids Health Dis. 16 : 145.
Related articles in JMB
Article
Research article
J. Microbiol. Biotechnol. 2024; 34(2): 425-435
Published online February 28, 2024 https://doi.org/10.4014/jmb.2306.06049
Copyright © The Korean Society for Microbiology and Biotechnology.
Schisandrin A in Schisandra chinensis Upregulates the LDL Receptor by Inhibiting PCSK9 Protein Stabilization in Steatotic Model
Hyo-Jin Kim1,2†, Seon Kyeong Park1†, Soo Hyun Park1, Yu Geon Lee1, Jae-Ho Park1, Jin-Taek Hwang1*, and Min-Yu Chung3*
1Personalized Diet Research Group, Korea Food Research Institute, Wanju 55365, Republic of Korea
2Department of Food Biotechnology, University of Science & Technology, Daejeon 34113, Republic of Korea
3Department of Food and Nutrition, Gangseo University, Seoul 07661, Republic of Korea
Correspondence to:Jin-Taek Hwang, jthwang@kfri.re.kr
Min-Yu Chung, chungmy@gangseo.ac.kr
†These authors contributed equally to this work.
Abstract
Schisandra chinensis extract (SCE) protects against hypocholesterolemia by inhibiting proprotein convertase subtilisin/kexin 9 (PCSK9) protein stabilization. We hypothesized that the hypocholesterolemic activity of SCE can be attributable to upregulation of the PCSK9 inhibition-associated low-density lipoprotein receptor (LDLR). Male mice were fed a low-fat diet or a Western diet (WD) containing SCE at 1% for 12 weeks. WD increased final body weight and blood LDL cholesterol levels as well as alanine transaminase and aspartate aminotransferase expression. However, SCE supplementation significantly attenuated the increase in blood markers caused by WD. SCE also attenuated WD-mediated increases in hepatic LDLR protein expression in the obese mice. In addition, SCE increased LDLR protein expression and attenuated cellular PCSK9 levels in HepG2 cells supplemented with delipidated serum (DLPS). Non-toxic concentrations of schisandrin A (SA), one of the active components of SCE, significantly increased LDLR expression and tended to decrease PCSK9 protein levels in DLPS-treated HepG2 cells. High levels of SA-mediated PCSK9 attenuation was not attributable to reduced PCSK9 gene expression, but was associated with free PCSK9 protein degradation in this cell model. Our findings show that PCSK9 secretion can be significantly reduced by SA treatment, contributing to reductions in free cholesterol levels.
Keywords: Schisandra chinensis, low-density lipoprotein receptor, proprotein convertase subtilisin/kexin 9, schisandrin A, HepG2 cells
Introduction
Cardiovascular disease (CVD) is one of the most significant causes of mortality and morbidity worldwide. Over the last ten years, the number of deaths due to CVD has increased by 12.5% globally [1, 2], with atherosclerosis being the leading cause of vascular disease and death. Atherosclerosis is caused by abnormal lipid metabolism, which leads to the accumulation of fatty streaks in the arterial layer, causing thickening and hardening of the arterial wall and resulting in narrowing of the vascular lumen [3]. Blood cholesterol, especially low-density lipoprotein (LDL), is a key driver of atherosclerosis [4]. Meta-analyses of large prospective observational studies have investigated the relationship between the risk of ischemic heart disease (IHD) and various lipid measures, including total cholesterol (TCHO), high-density lipoprotein (HDL) cholesterol, non-HDL cholesterol, triglycerides, and apolipoproteins [4-6]. These data suggest the existence of continuous positive log-linear relationships between normal blood levels of either total or LDL cholesterol (non-HDL cholesterol) and the risk of IHD. Meanwhile, a clear inverse correlation between HDL cholesterol and IHD risk has been found [5, 6].
Dietary intake of fat and cholesterol is an important environmental determinant of blood cholesterol levels, which can also be influenced by other factors, including exercise and genetics [7-10]. The consumption of dietary components that inhibit proprotein convertase subtilisin/kexin 9 (PCSK9) can positively influence lipid metabolism during the development and progression of atherosclerosis [11]. The role of PCSK9 in lipid metabolism has been extensively studied. PCSK9 binds to the LDL receptor, causing its degradation, thereby increasing circulating LDL levels in the blood. Cohort studies have demonstrated that circulating PCSK9 is a viable therapeutic target for reducing plasma LDL cholesterol levels to aid in the prevention of CVD [12, 13]. Observation of a large sample of healthy participants in the Brisighella Heart Cohort found that circulating PCSK9 levels were associated with increased arterial stiffness, and therefore potentially associated also with LDL cholesterol [13].
PCSK9 inhibitors bind to LDL receptors on the surface of liver cells, promoting receptor degradation and interfering with the clearance of LDL cholesterol from the bloodstream [14]. To date, specific natural substances that directly inhibit PCSK9 have not yet been reported. Widely known PCSK9 inhibitors are primarily synthetic compounds, such as evolocumab and alirocumab, but ongoing studies are exploring their safety [15]. The search for specific components of natural substances that directly inhibit PCSK9 remains an ongoing area of research and future findings may lead to new discoveries.
Materials and Methods
Dulbecco’s modified Eagle’s medium (DMEM) and antibiotic antimycotic solution (100X) were purchased from Welgene Inc. (Republic of Korea). Delipidated serum (DLPS) was prepared from fetal bovine serum (FBS; Thermo Fisher Scientific, USA) as described previously [19, 20]. Isopropanol and chloroform were obtained from EMD Millipore (USA). Cycloheximide (CHX), bafilomycin A1 (BA1), 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), and MG132 were purchased from Sigma-Aldrich (USA). Dimethyl sulfoxide (DMSO) was purchased from Duchefa Biochemie (Netherlands).
SCE Preparation
The fruit of
In Vivo Model
The animal study was approved by the Institutional Animal Care and Use Committee at Wonkwang University (WKU16-21). Male C57BL/6 mice (7-week-old) were purchased from SAMTAKO Bio Korea (Republic of Korea) and acclimated for 1 week in an environmentally controlled facility. At 8 weeks of age, mice were randomized to control diet (CD; #98121701, Research Diets Inc., USA), WD (#D12079B, Research Diets INC), or WD formulated with 1% SCE (w/w;
Serum Analysis
Blood samples were stored at 4°C for 2 h and centrifuged before serum was collected. Serum lipids (triglyceride, total cholesterol and LDL cholesterol) and serum hepatic injury markers (alanine transaminase (ALT) and aspartate aminotransferase (AST)) were analyzed using enzymatic colorimetric assay kits (
Cell Culture and Treatment
HepG2 cells (HB-8065) were purchased from the American Type Culture Collection (ATCC, USA), and maintained in DMEM containing 1% antibiotics and 10% (v/v) FBS at 37°C in a humidified atmosphere of 5% CO2 in an incubator. For sample (extract or chemicals) treatments, HepG2 cells were seeded (1.5 × 105 cells/ml) in DMEM containing 1% antibiotics and 10% FBS (day 0). On the following day, cells were washed with DPBS twice, and the medium was changed to DMEM containing 1% antibiotics and 10% DLPS (day 1). After 24 h incubation, the medium was replaced with fresh DMEM containing 1% antibiotics and 10% DLPS supplemented with appropriate levels of SCE or schisandrin A (SA) up to 100 μM (day 2). Finally, 18 h later, cells were harvested for further analysis following washing with DPBS.
Western Blotting
To evaluate hepatic protein expression, liver samples were homogenized with radioimmunoprecipitation assay (RIPA) buffer (Elpis Biotech, Inc., Republic of Korea) containing protease and phosphatase inhibitor (Roche), before being placed on ice for 30 min with intermittent vortexing. Following centrifugation (20,000 ×
Collected whole cell lysates were then used to analyze total protein content using a BCA kit (Pierce, USA). After quantification, 20 μg of protein was separated by 10% SDS-PAGE. Proteins were transferred to a nitrocellulose membrane (GE Healthcare BioScience, USA). The membrane was blocked in PBS containing 0.05% Tween 20 and 5% non-fat dried milk for 1 h at room temperature, and then incubated in blocking solution with primary antibodies against PCSK9 (Circulex, Medical & Biological Laboratories Co., Ltd., USA), LDLR (BioVision Inc., Milpitas, CA, USA), SREBP2 (Cayman Chemical, USA), hepatic nuclear factor (HNF)-1α (Cell Signaling Technology, USA) and β-actin (Bethyl Laboratories, USA) for 1 h at room temperature. After washing with PBS containing 0.05% Tween 20 and 5% non-fat dried milk, the membrane was incubated in horseradish peroxidase secondary antibody (Thermo Scientific, USA). Target proteins were visualized with a FUSION SOLO 6S imaging system (Vilber Lourmat, AZC de Lamirault, France) with an enhanced chemiluminescence detection reagent (Thermo Scientific). Target protein expression was normalized to β-actin.
Cell Viability
To examine viability of the HepG2 cells, a colorimetric assay using MTT was performed. To determine non-toxic concentrations for the cells, HepG2 cells were cultured with SCE (200, 400 μg/ml) or SA (10‒100 μM) in DMEM containing 10% DLPS. After an additional 18 h incubation, MTT solution (final concentration: 0.5 mg/mL) was added in each well. After a further 2 h incubation, a mixture of isopropanol and DMSO (1:1) was added to the wells before incubation for 5 min with gentle shaking. Sample absorbance was measured at 570 nm using a microplate reader (SpectraMax M2 microplate reader; Molecular Devices, USA).
Enzyme-Linked Immunosorbent Assay (ELISA)
HepG2 cells were cultured as described in Cell Culture and Treatment. Cells were treated with SCE or SA, and the plate was incubated for another 18 h. After incubation, both cell pellets and media were collected. Pellets were rinsed with DPBS and lysed with RIPA buffer (Elpis Biotech, Inc., Republic of Korea) containing protease and phosphatase inhibitor (Roche, Switzerland) in ice, before vortexing, centrifuging (15,000 ×
LDL Uptake Assay
To examine LDL uptake at the cellular level, HepG2 cells were seeded (3 × 104 cells/well) in 96-well plates and treated as previously described. We used an LDL uptake kit (Cayman Chemical) and performed the assay according to the manufacturer’s instructions. At the end of the treatment period, we added a fluorescent probe, DyLight 550, and incubated it for 12 h at 37°C in a humidified atmosphere of 5% CO2. Cells were rinsed and fixed for immunofluorescence staining. After washing with buffer, images were captured using a fluorescence microscope (IX71; Olympus, Japan).
Quantitative Real-Time Polymerase Chain Reaction (qRT-PCR)
HepG2 cells were maintained, seeded in 6-well plates, and cultured as described. On day 2, the cells were incubated with DMEM containing 1% antibiotics and 10% DLPS supplemented with SA (20, 40, and 80 μM), and incubated for an additional 18 h. Cells were washed with DPBS two times, and pellets were collected following centrifugation. Total RNA was extracted from pellets using TRIzol (Invitrogen, USA). cDNA was synthesized using a reverse transcription kit (Bio-Rad) and gene expression was quantified with SYBR Green PCR master mix (Thermo Fisher Scientific) on a Bio-Rad CFX384. Primers used for qRT-PCR are provided in Table 1. Target gene expression was quantified relative to GAPDH, using the 2-ΔΔCT method. All reactions were carried out in triplicate.
-
Table 1 . Sequence list of qRT-PCR primers..
Primer name Direction Sequence (5'->3') PCSK9 Forward GGCAGGTTGGCAGCTGTTT Reverse CGTGTAGGCCCCGAGTGT LDLR Forward AGGAGACGTGCTTGTCTGTC Reverse CTGAGCCGTTGTCGCAGT
Protein Stability
To evaluate PCSK9 protein stability regulated by SA, cells were treated simultaneously with SA and cycloheximide (CHX). In brief, HepG2 cells were cultured and seeded as described previously, followed by incubation in fresh DMEM supplemented with DLPS for 24 h (day 1). On the following day (day 2), cells were treated with SA at 0 or 80 μM. On day 3, cells were treated with protein synthesis inhibitor CHX (C4895, Sigma-Aldrich) at 5 μg/ml. Cells were then harvested at 0, 1, 2, 4, and 8 h post incubation with CHX. Western blotting was performed as described above using all the collected pellets followed by protein lysis.
To evaluate the effect of SA against lysosomal and proteasomal degradation inhibition, HepG2 cells were cultured and seeded (day 0) as described previously. On the following day (day 1), the cells were incubated with DMEM supplemented with 10% DLPS, followed by treatment (day 2) with chemicals [bafilomycin A1 (BA1; Sigma-Aldrich) at 0 or 50 nM or MG132 (Sigma-Aldrich) at 0 or 1 μM] and SA at 0 or 80 μM. Following 24 h incubation on day 3, the cells were washed with DPBS, harvested, and centrifuged, with the upper layer removed to collect whole cell lysate.
Filipin Staining Assay
To measure for cholesterol, HepG2 cells were cultured, seeded, and treated as described in Cell Culture and Treatment section. After 18 h of further incubation at day 2, cells were washed with DPBS 3 times, and fixed with 3.7% paraformaldehyde for 1 h. Following washing with DPBS 3 times, glycine solution dissolved in DPBS (100 μl) was added to each well to remove residual paraformaldehyde. Filipin solution (50 μg/ml) was then added to each well and incubated for 2 h at room temperature, followed by rinsing with DPBS 3 times. Fluorescence intensity was measured at 360/480 nm (excitation/emission) using a SpectraMax M2 microplate reader (Molecular Devices).
Statistical Analysis
Data (means ± standard error of mean (SEM)) were analyzed using GraphPad Prism software (version 7.0; USA). Student’s
Results
SCE Supplementation Attenuates WD-Induced Increases in Body Weight, Serum Lipid Profiles, and Hepatic Injury Markers, as well as LDLR Protein Expression
SCE (1%, w/w) supplementation significantly attenuated the final body weight of WD-fed obese mice compared with that of the controls (Fig. 1A). Twelve weeks of SCE supplementation also significantly reduced total cholesterol, LDL cholesterol, and TG levels in the serum compared with WD feeding in obese mice (Fig. 1B). To examine the effect of WD and low-fat diet (LD) feeding for 12 weeks on liver injury in mice, serum ALT and AST activities were measured. We observed that the WD significantly increased both ALT and AST activity, which were significantly decreased by SCE supplementation (Fig. 1C). ALT and AST activities in the serum were increased approximately 5- and 2-fold by WD feeding, respectively, and were almost normalized in the LD group with SCE supplementation.
-
Figure 1.
Schisandra chinensis extract (SCE) attenuates final body weight, serum lipid profiles, hepatic injury markers, and low-density lipoprotein receptor (LDLR) expression in liver tissues of Western diet (WD)-fed obese mice. Changes in body weight (A) serum lipid profiles (TG, TCHO, and LDL-C) (B) serum hepatic injury markers (ALT and AST) (C) and LDLR protein expression (D) in the liver tissue of WD-fed obese mice. The results are expressed as the mean ± SEM. # indicates a significant difference compared to the low-fat diet (LD) group (#p < 0.05, ##p < 0.01, ###p < 0.001). * indicates a significant difference compared to the WD group (*p < 0.05, **p < 0.01, ***p < 0.001). LD, low-fat diet; WD+SCE, WD plusSchisandra chinensis extract, TG; triglyceride, TCHO; total cholesterol, LDL-C; LDL-cholesterol, ALT; alanine transaminase, AST; aspartate aminotransferase.
LDLR is a key regulator of cholesterol metabolism. LDLR knockout (Ldlr-/-) Leiden mice are widely used to study dyslipidemia, obesity, and nonalcoholic steatohepatitis following high-fat diet feeding [21, 22]. Therefore, we examined whether the SCE-mediated reduction in LDL cholesterol levels were attributable to LDLR upregulation in the livers of these obese mice (Fig. 1D). We found that WD feeding markedly attenuated LDLR protein expression, which was reversed by SCE supplementation. LDLR protein expression in the WD+SCE group was similar to that observed in LD controls. Therefore, we deduced that SCE increased LDLR protein expression, contributing to reduced serum LDL cholesterol levels in obese mice.
SCE Increases LDLR Protein Expression and Reduces PCSK9 Secretion in DLPS-Treated HepG2 Cells
The regulation of hypercholesterolemia, attributed to LDLR upregulation by SCE treatment, was further examined in HepG2 cells. DLPS was used to simulate lipid-deprived conditions for the purpose of PCSK9 upregulation [19, 20]. SCE, at concentrations up to 400 μg/ml, did not affect the viability of DLPS-treated HepG2 cells (Fig. 2A). We then measured LDLR protein expression in DLPS-treated cells and found that SCE at a concentration of 200 ug/ml, markedly increased LDLR expression compared to the DLPS-treated cells (Fig. 2B). Next, we measured PCSK9 secretion using ELISA. DLPS significantly increased PCSK9 secretion, which was significantly reduced by SCE at concentrations of 100 and 200 μg/ml (Fig. 2C). These findings suggest that SCE inhibits PCSK9 secretion, thereby preventing LDLR degradation and likely contributing to the reduction of free cholesterol levels in HepG2 cells.
-
Figure 2.
Schisandra chinensis extract increases low-density lipoprotein receptor (LDLR) expression and inhibits proprotein convertase subtilisin/kexin 9 (PCSK9) secretion in delipidated serum (DLPS)-induced lipid-deprived conditions. Cell viability (A) LDLR protein expression in HepG2 cells (B) and PCSK9 levels in the media (C). The results are expressed as the mean ± SEM. # indicates a significant difference compared to the normal control (#p < 0.05, ##p < 0.01, ###p < 0.001). * indicates a significant difference compared to DLPS-treated HepG2 cells (*p < 0.05, **p < 0.01, ***p < 0.001). FBS, fetal bovine serum; SCE,Schisandra chinensis extract.
Schisandrin A Induces LDLR Upregulation and PCSK9 Inhibition in DLPS-Treated HepG2 Cells
We examined the components of SCE to determine which of them contribute to the observed hypocholesterolemic activity of PCSK9 inhibition. SA is a major bioactive component of
-
Figure 3. Schisandrin A (SA) increases low-density lipoprotein (LDL) uptake in delipidated serum (DLPS)- treated HepG2 cells.
Chemical structure of SA (A) cell viability (B) and representative immunofluorescence images and fluorescence intensity of the LDL-cholesterol uptake assay (C) in HepG2 cells. The results are expressed as the mean ± SEM. # indicates a significant difference compared to the normal control (#
p < 0.05, ##p < 0.01, ###p < 0.001). * indicates a significant difference compared to DLPS-treated HepG2 cells (*p < 0.05, **p < 0.01, ***p < 0.001).
SA Significantly Regulates LDLR/PCSK9 Protein Expression but Not PCSK9 Gene Expression
LDLR and PCSK9 protein expression levels were confirmed using western blot analysis. SA (10, 20, 40, and 80 μM) increased LDLR protein expression, and 80 μM SA inhibited PCSK9 expression in DLPS-treated HepG2 cells (Fig. 4A).
-
Figure 4. Schisandrin A (SA) regulates low-density lipoprotein receptor (LDLR)- proprotein convertase subtilisin/kexin 9 (PCSK9) protein expression but is not associated with their gene expression or the upregulation of hepatic nuclear factor (HNF)-1α and SREBP2 protein expression in delipidated serum (DLPS)-treated HepG2 cells.
LDLR and PCSK9 protein expression (A) LDLR and PCSK9 gene expression (B) and SREBP2 and HNF-1α protein expression (C) in HepG2 cells. The results are expressed as the mean ± SEM. # indicates a significant difference compared to the normal control (#
p < 0.05, ##p < 0.01, ###p < 0.001). * indicates a significant difference compared to DLPS-treated HepG2 cells (*p < 0.05, **p < 0.01, ***p < 0.001). n.s., no significant difference.
In HepG2 cells, LDLR gene expression was significantly increased by DLPS treatment, which was in turn significantly increased by treatment with SA (80 μM) (Fig. 4B). DLPS-mediated increases in PCSK9 gene expression were not affected by any level of SA treatment (Fig. 4B).
LDLR-PCSK9 Protein Modulation by SA Is Not associated with HNF-1α or SREBP2 Regulation in HepG2 Cells
The expression levels of SREBP2 and HNF-1α were examined by western blotting. We found that DLPS treatment downregulated SREBP2 protein expression, while SA slightly downregulated SREBP2 (Fig. 4C). Hepatic nuclear factor HNF-1α is a transcription factor that regulates PCSK9 independently of LDLR [23]. We found that HNF-1α protein expression was unaffected by DLPS or SA treatments (Fig. 4C). These findings suggested that the hypocholesterolemic effect of SA was not associated with the regulation of transcription factors that mediate LDLR and PCSK9 protein expression.
SA Induces PCSK9 Destabilization, thereby Preventing LDLR Lysosomal Degradation in HepG2 Cells
To examine PCSK9 protein stabilization by SA treatment, cells were simultaneously treated with CHX, an inhibitor of new protein synthesis, and SA. When PCSK9 translation was blocked, PCSK9 protein expression reduced in a time-dependent manner up to 8 h, which was also consistently observed with 80 μM of SA treatment (Fig. 5A), suggesting that SA does not contribute to stabilization of the PCSK9 protein.
-
Figure 5. Schisandrin A (SA) effectively blocks proprotein convertase subtilisin/kexin 9 (PCSK9) secretion in delipidated serum (DLPS)-treated HepG2 cells.
PCSK9 protein expression in the presence or absence of cycloheximide (CHX) (A) bafilomycin A1 (BA1) (B) and MG132 (C) in HepG2 cells. Released PCSK9 levels into the medium (D) and free low-density lipoprotein-cholesterol levels in the medium (E). The results are expressed as the mean ± SEM. # indicates a significant difference compared to the normal control (#
p < 0.05, ##p < 0.01, ###p < 0.001). * indicates a significant difference compared to DLPS-treated HepG2 cells (*p < 0.05, **p < 0.01, ***p < 0.001).
Circulating PCSK9 binds to LDLR and is transported to lysosomes for degradation [24]. As expected, treatment with the lysosomal degradation inhibitor, BA1, increased PCSK9 protein expression in DLPS-treated cells. However, SA enhanced the BA1-mediated increase in PCSK9 protein expression in DLPS-treated HepG2 cells (Fig. 5B). These results indicated that SA contributes to the stabilization of PCSK9 protein binding to LDLR in lysosomes.
PCSK9 stabilization by SA was further evaluated using the proteasome degradation inhibitor MG132 in DLPS-treated HepG2 cells (Fig. 5C). SA co-treated with MG132 markedly decreased PCSK9 protein levels, suggesting that SA destabilizes free PCSK9 in HepG2 cells treated with DLPS.
Next, we sought to confirm whether SA caused the destabilization of free PCSK9, thereby blocking PCSK9 secretion. Levels of PCSK9 secreted into the medium were measured using ELISA. DLPS was found to significantly increase PCSK9 secretion, which was in turn significantly reduced by SA (80 μM) treatment in HepG2 cells (Fig. 5D). SA also reduced free LDL-cholesterol (LDL-C) levels in DLPS-treated HepG2 cells (Fig. 5E). Taken together, these observations support the previous finding that SA destabilizes free PCSK9, leading to the attenuation of circulating PCSK9, thereby protecting LDLR from protein degradation and subsequently reducing cholesterol levels in HepG2 cells.
Discussion
Cholesterol homeostasis is an essential, yet complex, aspect of metabolic regulation [25, 26]. Dietary cholesterol is absorbed into the intestine, whereas endogenous cholesterol is synthesized in a number of tissues, primarily in the liver. Lipoprotein particles play an important role in maintaining cholesterol homeostasis in the liver. Lipoproteins belong to several categories, including chylomicrons, very low-density lipoproteins (VLDL), LDL, and HDL.
Chylomicrons transport esterified dietary cholesterol from the intestine to the liver. VLDL is produced in the liver by combining triglycerides (TG) and cholesterol ester (CE) with apolipoproteins and other lipoproteins, and secreted into the bloodstream to be transported to peripheral tissues, such as muscle and adipose tissues. During this process, TG was removed from VLDL, converting to LDL containing more CE than TG [25, 27]. LDL is taken up by tissue expressing the apo B-containing receptor, and the CE transported by LDL becomes available for use by cells. Excess LDL (or oxidized LDL) often accumulates in cells via macrophages to form foam cells, which are a primary component of plaques during the development of atherosclerosis. Oxidized LDL is taken up by lesional macrophages before delivery to lysosomes, where oxidized LDL is hydrolyzed to free cholesterol by lysosomal acid lipase [28]. Free cholesterol can cause inflammation, is associated with various chronic diseases, and is transported from the lysosome to the endoplasmic reticulum, where it is esterified and stored as lipid droplets under normal conditions, whereas both CE and free cholesterol accumulate in cells where cholesterol trafficking occurs [29, 30]. Accumulated CE (as lipid droplets) causes the foamy morphology of macrophages, contributing to the production of atherosclerotic plaques. Together with the buildup of free cholesterol, which causes inflammation, these factors increase the risk of atherosclerosis [31].
Several strategies mediated by food compounds or diet plans can reduce circulating LDL cholesterol levels. In addition to enhancing LDLR expression, cholesterol excretion or absorption can be mediated when LDL cholesterol reaches critical levels. A meta-analysis of 14 randomized controlled trials with 1,136 individuals indicated that the consumption of green tea and its extracts significantly reduced fasting serum total cholesterol and LDL cholesterol levels (but not that of HDL cholesterol) in adults [32]. There are several intermediates through which flavonoids attenuate LDL cholesterol levels, which are often LDLR/PCSK9-independent. The underlying mechanism of PCSK9 inhibition may involve the upregulation of LDLR recycling and LDL uptake, providing a scientific basis for the prevention and treatment of steatosis. Therefore, our study focused on evaluating the potential of SCE and its constituents, which are natural substances, to regulate LDL levels via the LDLR-PCSK9 pathway.
Several food extracts with PCSK9 inhibitory activity have been identified. For instance, extracts of
In our study, SA reduced PCSK9 stabilization in the proteasome, a likely mechanism by which it contributes to the protection of LDLR against protein degradation by lysosomes (Fig. 5). Destabilization of PCSK9 aims to increase the availability of functional LDLRs and promote the clearance of LDL-C from the bloodstream. This can be beneficial for the management of high LDL-C levels, reducing the risk of CVDs and improving lipid profiles.
Taken together, these observations suggest that SA effectively blocks the free (not bound to LDLR) PCSK9 form, thereby recycling it back to the cell membrane of LDLR. Sun
Conclusively, SA, a natural bioactive compound found in
Acknowledgments
This study was supported by the Main Research Program of the Korea Food Research Institute (KFRI) and was funded by the Ministry of Science and ICT (E0210601-03).
Author Contributions
Conceptualization; S.K.P. and M.-Y.C., Methodology; M.-Y.C., Validation; J.-H.P. and S.H.P., Formal Analysis; H.-J.K., Investigation; H.-J.K. and S.K.P., Data Curation; S.H.P. and Y.G.L. Writing – Original Draft Preparation; H.-J.K. and S.K.P., Writing – Review & Editing; J.-T.H. and M.-Y.C., Visualization; Y.G.L, Supervision; J.-T.H. and M.-Y.C., Project Administration; J.-H.P. and J.-T.H.
Conflict of Interest
The authors have no financial conflicts of interest to declare.
Fig 1.
Fig 2.
Fig 3.
Fig 4.
Fig 5.
-
Table 1 . Sequence list of qRT-PCR primers..
Primer name Direction Sequence (5'->3') PCSK9 Forward GGCAGGTTGGCAGCTGTTT Reverse CGTGTAGGCCCCGAGTGT LDLR Forward AGGAGACGTGCTTGTCTGTC Reverse CTGAGCCGTTGTCGCAGT
References
- Joseph P, Leong D, McKee M, Anand SS, Schwalm JD, Teo K,
et al . 2017. Reducing the global burden of cardiovascular disease, part 1: the epidemiology and risk factors.Circ. Res. 121 : 677-694. - Roth GA, Johnson C, Abajobir A, Abd-Allah F, Abera SF, Abyu G,
et al . 2017. Global, regional, and national burden of cardiovascular diseases for 10 causes, 1990 to 2015.J. Am. Coll. Cardiol. 70 : 1-25. - Herrington W, Lacey B, Sherliker P, Armitage J, Lewington S. 2016. Epidemiology of atherosclerosis and the potential to reduce the global burden of atherothrombotic disease.
Circ. Res. 118 : 535-546. - Baigent C, Blackwell L, Emberson J, Holland LE, Reith C,
et al . 2010. Efficacy and safety of more intensive lowering of LDL cholesterol: a meta-analysis of data from 170,000 participants in 26 randomised trials.Lancet 376 : 1670-1681. - Lewington S, Whitlock G, Clarke R, Sherliker P, Emberson J,
et al . 2007. Blood cholesterol and vascular mortality by age, sex, and blood pressure: a meta-analysis of individual data from 61 prospective studies with 55,000 vascular deaths.Lancet 370 : 1829-1839. - Di Angelantonio E, Sarwar N, Perry P, Kaptoge S, Ray KK,
et al . 2009. Major lipids, apolipoproteins, and risk of vascular disease.JAMA 302 : 1993-2000. - Hegsted DM, Ausman LM, Johnson JA, Dallal GE. 1993. Dietary fat and serum lipids: an evaluation of the experimental data.
Am. J. Clin. Nutr. 57 : 875-883. - Keys A, Anderson JT, Grande F. 1957. Prediction of serum-cholesterol responses of man to changes in fats in the diet.
Lancet 273 : 959-966. - Clarke R, Frost C, Collins R, Appleby P, Peto R. 1997. Dietary lipids and blood cholesterol: quantitative meta-analysis of metabolic ward studies.
BMJ 314 : 112-117. - Dattilo AM, Kris-Etherton PM. 1992. Effects of weight reduction on blood lipids and lipoproteins: a meta-analysis.
Am. J. Clin. Nutr. 56 : 320-328. - Patriki D, Saravi SSS, Camici GG, Liberale L, Beer JH. 2021. PCSK 9: a link between inflammation and atherosclerosis.
Curr. Med. Chem. 29 : 251-267. - Leander K, Malarstig A, Van't Hooft FM, Hyde C, Hellenius ML, Troutt JS,
et al . 2016. Circulating proprotein convertase subtilisin/kexin type 9 (PCSK9) predicts future risk of cardiovascular events independently of established risk factors.Circulation 133 : 1230-1239. - Ruscica M, Ferri N, Fogacci F, Rosticci M, Botta M, Marchiano S,
et al . 2017. Circulating levels of proprotein convertase subtilisin/kexin type 9 and arterial stiffness in a large population sample: data from the brisighella heart study.J. Am. Heart Assoc. 6 : e005764. - Horton JD, Cohen JC, Hobbs HH. 2007. Molecular biology of PCSK9: Its role in LDL metabolism.
Trends Biochem. Sci. 32 : 71-77. - Xu S, Luo S, Zhu Z, Xu J. 2019. Small molecules as inhibitors of PCSK9: current status and future challenges.
Eur. J. Med. Chem. 162 : 212-233. - Szopa A, Ekiert R, Ekiert H. 2017. Current knowledge of
Schisandra chinensis (Turcz.) Baill.(Chinese magnolia vine) as a medicinal plant species: a review on the bioactive components, pharmacological properties, analytical and biotechnological studies.Phytochem. Rev. 16 : 195-218. - Kopustinskiene DM, Bernatoniene J. 2021. Antioxidant effects of
Schisandra chinensis fruits and their active constituents.Antioxidants 10 : 620. - Seo HJ, Ji SB, Kim SE, Lee GM, Moon SH, Jang DS,
et al . 2020. Analysis of dibenzocyclooctadiene lignans in Omija Wine and Cheong by liquid chromatography-tandem mass spectrometry.Mass. Spectrom. Lett. 11 : 30-35. - Jeong HJ, Lee HS, Kim KS, Kim YK, Yoon D, Park SW. 2008. Sterol-dependent regulation of proprotein convertase subtilisin/kexin type 9 expression by sterol-regulatory element binding protein-2.
J. Lipid. Res. 49 : 399-409. - Hwang JT, Choi E, Choi HK, Park JH, Chung MY. 2021. The cholesterol-lowering effect of
Capsella Bursa-Pastoris is mediated via SREBP2 and HNF-1alpha-regulated PCSK9 inhibition in obese mice and HepG2 cells.Foods 10 : 408. - Morrison MC, Verschuren L, Salic K, Verheij J, Menke A, Wielinga PY,
et al . 2018. Obeticholic acid modulates serum metabolites and gene signatures characteristic of human NASH and attenuates inflammation and fibrosis progression in Ldlr-/-. leiden mice.Hepatol. Commun. 2 : 1513-1532. - Tengeler AC, Gart E, Wiesmann M, Arnoldussen IAC, van Duyvenvoorde W, Hoogstad M,
et al . 2020. Propionic acid and not caproic acid, attenuates nonalcoholic steatohepatitis and improves (cerebro) vascular functions in obese Ldlr(-/-) .Leiden mice.FASEB J. 34 : 9575-9593. - Li H, Dong B, Park SW, Lee HS, Chen W, Liu J. 2009. 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. - Lambert G, Sjouke B, Choque B, Kastelein JJ, Hovingh GK. 2012. The PCSK9 decade.
J. Lipid. Res. 53 : 2515-2524. - Maxfield FR, Tabas I. 2005. Role of cholesterol and lipid organization in disease.
Nature 438 : 612-621. - Estronca LM, Filipe HA, Salvador A, Moreno MJ, Vaz WL. 2014. Homeostasis of free cholesterol in the blood: a preliminary evaluation and modeling of its passive transport.
J. Lipid. Res. 55 : 1033-1043. - Rothblat GH, Phillips MC. 2010. High-density lipoprotein heterogeneity and function in reverse cholesterol transport.
Curr. Opin. Lipidol. 21 : 229-238. - Xu X, Zhang A, Halquist MS, Yuan X, Henderson SC, Dewey WL,
et al . 2017. Simvastatin promotes NPC1-mediated free cholesterol efflux from lysosomes through CYP7A1/LXRalpha signalling pathway in oxLDL-loaded macrophages.J. Cell Mol. Med. 21 : 364-374. - Pfisterer SG, Peranen J, Ikonen E. 2016. LDL-cholesterol transport to the endoplasmic reticulum: current concepts.
Curr. Opin. Lipidol. 27 : 282-287. - Vanier MT. 2015. Complex lipid trafficking in Niemann-Pick disease type C.
J. Inherit. Metab. Dis. 38 : 187-199. - Tall AR, Yvan-Charvet L. 2015. Cholesterol, inflammation and innate immunity.
Nat. Rev. Immunol. 15 : 104-116. - Zheng XX, Xu YL, Li SH, Liu XX, Hui R, Huang XH. 2011. Green tea intake lowers fasting serum total and LDL cholesterol in adults: a meta-analysis of 14 randomized controlled trials.
Am. J. Clin. Nutr. 94 : 601-610. - Park JY, Shin HK, Lee YJ, Choi YW, Bae SS, Kim CD. 2009. The mechanism of vasorelaxation induced by
Schisandra chinensis extract in rat thoracic aorta.J. Ethnopharmacol. 121 : 69-73. - Brown MS, Goldstein JL. 1984. How LDL receptors influence cholesterol and atherosclerosis.
Sci. Am. 251 : 58-66. - Sirinian MI, Belleudi F, Campagna F, Ceridono M, Garofalo T, Quagliarini F,
et al . 2005. Adaptor protein ARH is recruited to the plasma membrane by low density lipoprotein (LDL) binding and modulates endocytosis of the LDL/LDL receptor complex in hepatocytes.J. Biol. Chem. 280 : 38416-38423. - Ragusa R, Basta G, Neglia D, De Caterina R, Del Turco S, Caselli C. 2021. PCSK9 and atherosclerosis: looking beyond LDL regulation.
Eur. J. Clin. Invest. 51 : e13459. - Macchi C, Sirtori C, Corsini A, Santos R, Watts G, Ruscica M. 2019. A new dawn for managing dyslipidemias: the era of RNA-based therapies.
Pharmacol. Res. 150 : 104413. - Choi HK, Hwang JT, Nam TG, Kim SH, Min DK, Park SW,
et al . 2017. Welsh onion extract inhibits PCSK9 expression contributing to the maintenance of the LDLR level under lipid depletion conditions of HepG2 cells.Food Funct. 8 : 4582-4591. - Choi HK, Kim HJ, Hwang JT, Chung MY. 2017.
Allium tuberosum reverses PCSK9-mediated LDLR degradation by inhibition of HNF1α.J. Korean Soc. Food Sci. Nutr. 46 : 1278-1285. - Wang F, Zhao C, Tian G, Wei X, Ma Z, Cui J,
et al . 2020. Naringin alleviates atherosclerosis in ApoE(-/-) mice by regulating cholesterol metabolism involved in gut microbiota remodeling.J. Agric. Food Chem. 68 : 12651-12660. - Hwang JT, Kim HJ, Choi HK, Park JH, Chung S, Chung MY. 2020. Butein synergizes with statin to upregulate low-density lipoprotein receptor through HNF1alpha-mediated PCSK9 inhibition in HepG2 cells.
J. Med. Food 23 : 1102-1108. - Cui CJ, Jin JL, Guo LN, Sun J, Wu NQ, Guo YL,
et al . 2020. Beneficial impact of epigallocatechingallate on LDL-C through PCSK9/LDLR pathway by blocking HNF1alpha and activating FoxO3a.J. Transl. Med. 18 : 1-13. - Jia Q, Cao H, Shen D, Li S, Yan L, Chen C,
et al . 2019. Quercetin protects against atherosclerosis by regulating the expression of PCSK9, CD36, PPARgamma, LXRalpha and ABCA1.Int. J. Mol. Med. 44 : 893-902. - Dong Z, Zhang W, Chen S, Liu C. 2019. Silibinin A decreases statininduced PCSK9 expression in human hepatoblastoma HepG2 cells.
Mol. Med. Rep. 20 : 1383-1392. - Gao WY, Chen PY, Chen SF, Wu MJ, Chang HY, Yen JH. 2018. Pinostrobin inhibits proprotein convertase subtilisin/kexin-type 9 (PCSK9) gene expression through the modulation of FoxO3a protein in HepG2 cells.
J. Agric. Food Chem. 66 : 6083-6093. - Razgonova M, Zakharenko A, Pikula K, Kim E, Chernyshev V, Ercisli S,
et al . 2020. Rapid mass spectrometric study of a supercritical CO2-extract from woody lianaSchisandra chinensis by HPLC-SPD-ESI-MS/MS.Molecules 25 : 2689. - Kim Y, Kim E, Jeong G. 2019. Isolation and quantitative analysis of schisandrin, gomisin A and gomisin M2 from
Schisandra chinensis .Korean J. Pharmacogn. 50 : 148-153. - Choi YH. 2018. Schisandrin A prevents oxidative stress-induced DNA damage and apoptosis by attenuating ROS generation in C2C12 cells.
Biomed. Pharmacother. 106 : 902-909. - Fu K, Zhou H, Wang C, Gong L, Ma C, Zhang Y,
et al . 2022. A review: pharmacology and pharmacokinetics of schisandrin A.Phytother. Res. 36 : 2375-2393. - Kwon DH, Cha HJ, Choi EO, Leem SH, Kim GY, Moon SK,
et al . 2018. Schisandrin A suppresses lipopolysaccharide-induced inflammation and oxidative stress in RAW 264.7 macrophages by suppressing the NF-κB, MAPKs and PI3K/Akt pathways and activating Nrf2/HO-1 signaling.Int. J. Mol. Med. 41 : 264-274. - Jeong MJ, Kim SR, Jung UJ. 2019. Schizandrin A supplementation improves nonalcoholic fatty liver disease in mice fed a high-fat and high-cholesterol diet.
Nutr. Res. 64 : 64-71. - Sun JH, Liu X, Cong LX, Li H, Zhang CY, Chen JG,
et al . 2017. Metabolomics study of the therapeutic mechanism ofSchisandra chinensis lignans in diet-induced hyperlipidemia mice.Lipids Health Dis. 16 : 145.