Open Access

Effect of astragaloside IV and the role of nuclear receptor RXRα in human peritoneal mesothelial cells in high glucose‑based peritoneal dialysis fluids

  • Authors:
    • Weiwei Zhu
    • Xin Zhang
    • Kun Gao
    • Xufang Wang
  • View Affiliations

  • Published online on: August 22, 2019     https://doi.org/10.3892/mmr.2019.10604
  • Pages: 3829-3839
  • Copyright: © Zhu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Peritoneal fibrosis is a serious complication that can occur during peritoneal dialysis (PD), which is primarily caused by damage to peritoneal mesothelial cells (PMCs). The onset of peritoneal fibrosis is delayed or inhibited by promoting PMC survival and inhibiting PMC epithelial‑to‑mesenchymal transition (EMT). In the present study, the effect of astragaloside IV and the role of the nuclear receptor retinoid X receptor‑α (RXRα) in PMCs in high glucose‑based PD fluids was investigated. Human PMC HMrSV5 cells were transfected with RXRα short hairpin RNA (shRNA), or an empty vector, and then treated with PD fluids and astragaloside IV. Cell viability, apoptosis and EMT were examined using the Cell Counting Kit‑8 assay and flow cytometry, and by determining the levels of caspase‑3, E‑cadherin and α‑smooth muscle actin (α‑SMA) via western blot analysis. Cell viability and apoptosis were increased, as were the levels of E‑cadherin in HMrSV5 cells following treatment with PD fluid. The protein levels of α‑SMA and caspase‑3 were increased by treatment with PD fluid. Exposure to astragaloside IV inhibited these changes; however, astragaloside IV did not change cell viability, apoptosis, E‑cadherin or α‑SMA levels in HMrSV5 cells under normal conditions. Transfection of HMrSV5 cells with RXRα shRNA resulted in decreased viability and E‑cadherin expression, and increased apoptosis and α‑SMA levels, in HMrSV5 cells treated with PD fluids and co‑treated with astragaloside IV or vehicle. These results suggested that astragaloside IV increased cell viability, and inhibited apoptosis and EMT in PMCs in PD fluids, but did not affect these properties of PMCs under normal condition. Thus, the present study suggested that RXRα is involved in maintaining viability, inhibiting apoptosis and reducing EMT of PMCs in PD fluid.

Introduction

Peritoneal dialysis (PD) is an effective alternative treatment for end-stage renal disease (14). Peritoneal fibrosis is a serious complication during PD treatment that affects the survival and prognosis of patients undergoing PD. Peritoneal fibrosis is also one of the primary factors leading to withdrawal from treatment (57). The components and some bioincompatible properties of peritoneal dialysates, such as low pH, lactate buffer, high sugar, low calcium, plasticizer and glucose degradation products, cause loss of peritoneal mesothelial cells (PMCs), subcutaneous dense zone thickening, interstitial fibrosis, inflammation and neovascularization (812). Damage to PMCs is a key initiating factor that leads to peritoneal fibrosis (1315). After PMCs are damaged, extracellular matrix (ECM) components, including collagen, fibronectin, laminin, proteoglycan and various fibrogenic factors, such as transforming growth factor (TGF)-β1, fibroblast growth factor, connective tissue growth factor, platelet derived growth factor, toll-like receptors (TLRs), angiotensin II receptor and receptor tyrosine kinases, are highly expressed or secreted, which interferes with the normal metabolism of the ECM and promoting its overdeposition, ultimately leading to peritoneal fibrosis (10,1623). Peritoneal fibrosis can be delayed or inhibited by promoting PMC survival and inhibiting PMC epithelial-to-mesenchymal transition (EMT) (14,1619,21,22,24).

Previous studies have shown that Astragalus membranaceus inhibits peritoneal fibrosis in PD through monocyte chemoattractant protein-1 and the TGF-β1 pathway (25), and ameliorates renal interstitial fibrosis by inhibiting EMT, inflammation, TLR4/NF-κB and cyrillic B (2527). Astragalus inhibits PMC EMT by downregulating β-catenin (28). Astragaloside IV is a key compound extracted from Astragalus membranaceus (27,29,30). It has been shown that astragaloside IV inhibits TGF-β1-induced PMC EMT through the upregulation of Smad7 in the TGF-β1/Smad signaling pathway (31). However, the effect of astragaloside IV on viability and apoptosis of PMCs remains unclear.

Retinoid X receptor-α (RXRα) is a ligand-dependent nuclear receptor expressed in various tissues and cells (3234). RXRα can form heterodimers with other nuclear receptors, including peroxisome proliferator activated receptor (PPAR), vitamin D receptor (VDR) and thyroid hormone receptors, resulting in the involvement of RXRα in multiple signaling pathways (3540). Previous studies have shown that vitamin D/VDR can inhibit peritoneal fibrosis and functional deterioration induced by chlorhexidine gluconate by inhibiting PMC EMT (4143). Telmisartan inhibits peritoneal fibrosis through PPAR-γ activation (44). The PPAR-β/δ agonist GW501516 inhibits peritoneal inflammation in peritoneal fibrosis by inhibiting the TGF-β-activated kinase 1/NF-κB pathway (45). The PPAR-γ agonists rosiglitazone and pioglitazone protect rat PMCs against PD solution-induced damage (46,47). These previous studies indicated that the RXR signaling pathway is involved in regulating PMC EMT and peritoneal fibrosis. However, the role of RXRα in PMC activity, apoptosis and EMT in peritoneal fibrosis remains unclear.

In the present study, the human PMC HMrSV5 cell line and high glucose-based PD fluids were used as a model (31) to study the effects of astragaloside IV on PMC viability, apoptosis and EMT during PD. The role of RXRα in PMC viability, apoptosis and EMT during PD was also investigated. The findings of the present study may provide important information for the prevention and treatment of PD-induced fibrosis.

Materials and methods

Construction of RXRα short hairpin RNA (shRNA) plasmid

The synthetic DNA fragment targeting RXRα (GGATCCCGCACTATGGAGTGTACAGCTCAAGAGAGAGCTGTACACTCCAGTGCTTTTTTCCAAAAGCTT, synthesized by Western Biomedical Technology, Ltd.) and the vector SD1211 (Biovector Science Lab, Inc.) were modified with BamHI and HindIII (Takara Bio, Inc.) at 37°C for 30 min. After gel purification, the digested DNA fragment and the vector were ligated using T4 DNA ligase (Takara, Bio, Inc.) and then transfected into DH5α competent cells (Tiangen Biotech Co., Ltd.) for plasmid amplification. After selection and screening, plasmids were sequenced to confirm successful construction of the shRNA plasmid.

Cell culture and grouping

The human PMC cell line HMrSV5 was obtained from the Type Culture Collection of the Cell Bank of Chinese Academy of Sciences. This cell line was established by Professor Pierre Ronco, Hospital Tenon (Paris, France) (48) and had been used in a number of previous studies (4952). HMrSV5 cells were cultured in DMEM containing 10% FBS (Gibco; Thermo Fisher Scientific, Inc.) and 1% penicillin-streptomycin in an incubator at 37°C supplemented with 5% CO2 and a saturated humidity. To investigate the effects of astragaloside IV (Yuanye Bio-Technology Co., Ltd.) on PMCs in high glucose-based PD fluids, HMrSV5 cells were divided into four groups: i) Normal + vehicle control group, cells were cultured in regular media and treated with DMSO; ii) PD model + vehicle group, cells were cultured in PD fluids and treated with DMSO; iii) normal + astragaloside IV group, cells were cultured in regular media and treated with astragaloside IV; and iv) PD model regular astragaloside IV group, cells were cultured in PD fluids and treated with astragaloside IV. To investigate the role of RXRα in maintaining PMCs in PD fluids, HMrSV5 cells were divided into four groups: i) Blank + vehicle control group, cells were transfected with the SD1211 empty plasmid, cultured in PD fluids and treated with DMSO; ii) RXRα shRNA plasmid + vehicle group, cells were transfected with the RXRα shRNA plasmid, cultured in PD fluids and treated with DMSO; iii) blank + astragaloside IV group, cells were transfected with the SD1211 empty plasmid, cultured in PD fluids and treated with astragaloside IV; and iv) RXRα shRNA + astragaloside IV group, cells were transfected with the SD1211 RXRα shRNA plasmid, cultured in PD fluids and treated with astragaloside IV. Cells in each group were first transfected with the appropriate plasmid, for 6 h and then cultured in fresh media for 24 h. These cells were then cultured in PD fluids and astragaloside IV or DMSO was added. The PD fluids used for cell culture were made from original PD fluids with the addition of 10% FBS. The original PD fluids (Lactate-G4.25%; cat. no. 6AB9896) were purchased from Guangzhou Baxter Medical Products Co., Ltd. Its components include 4.25 g glucose, 538 mg sodium chloride, 26 mg calcium chloride, 5.1 mg magnesium chloride and 448 mg sodium lactate/100 ml. The final concentration of astragaloside IV in the normal media or PD fluids was 40 µg/ml. Astragaloside IV was dissolved in DMSO to make a 40 mg/ml stock solution. The same volume of DMSO and astragaloside IV stock solution was used to treat cells. All cells were cultured and treated with pertinent chemicals at 37°C.

Cell Counting Kit-8 (CCK-8) assay

The CCK-8 assay was used to determine the viability of HMrSV5 cells. To investigate the effects of astragaloside IV on the viability of PMCs in PD fluids, HMrSV5 cells in the log phase growth from each group were seeded in triplicate into 96-well plates at a density 1×105 cells/cm2 and cultured overnight. Cells were treated with astragaloside IV, PD and their respective controls for 24, 48 or 72 h. To investigate the role of RXRα in maintaining the viability of PMCs in PD fluids, HMrSV5 cells from each group were seeded in triplicate into 96-well plates at a density 1×105 cells/cm2 and cultured overnight. Following overnight culture, cells were transfected with SD1211 empty vector (0.4 µg/cm2) or RXRα shRNA plasmid (0.4 µg/cm2) for 6 h and then cultured in fresh media for 24 h. These cells were then treated with astragaloside IV, PD and DMSO for 24, 48 or 72 h. Cell viability was determined using a CCK-8 kit (Sigma-Aldrich; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. The absorbance (A) at 450 nm was measured using a microplate reader (Thermo Fisher Scientific, Inc.). A 96-well plate with medium and CCK reagent was used as a blank control. Cell viability (%)=[A (experiment)-A (blank plate)]/A (normal + vehicle control group) ×100; or cell viability (%)=[A (experiment)-A (blank plate)]/A (blank + vehicle control group) ×100.

Flow cytometry

Flow cytometry was used to examine the level of apoptosis in HMrSV5 cells. To investigate the effects of astragaloside IV on the level of apoptosis of PMCs in PD fluids, HMrSV5 cells were seeded into 6-well plates at a density 1×105 cells/cm2, cultured overnight and treated with astragaloside IV, PD and their respective controls for 48 h. To investigate the role of RXRα in apoptosis of PMCs in PD fluids, HMrSV5 cells were seeded into 6-well plates at a density 1×105 cells/cm2. Following overnight culture, cells were transfected with SD1211 empty vector (0.4 µg/cm2) or RXRα shRNA plasmid (0.4 µg/cm2) for 6 h and then cultured in fresh media for 24 h. These cells were then treated with PD and astragaloside IV or DMSO for 48 h. Cells were collected and the rate of apoptosis was determined using a EPICS XL flow cytometer (Beckman Coulter, USA) and Annexin V-PE Apoptosis Detection Kit I (BD Biosciences, 559763) according to the manufacturer's instructions. Briefly, cells were washed twice with cold PBS and then resuspended in 1X Binding Buffer (BD Biosciences, 51–66121E) at a concentration of 1×106 cells/ml. Then, 100 µl of the solution (1×105 cells) was transferred to a 5 ml culture tube and 5 µl of Annexin V-PE (BD Biosciences, 51-65875X) and 5 µl of 7-Amino-actinomycin D (7-AAD; BD Biosciences, 51-68981E) added. The cells were gently vortexed and incubated for 15 min at room temperature (25°C) in the dark. 1X binding buffer (400 µl) was added to each tube. Flow cytometry analysis was performed within one hour. The results were analyzed using CytExpert 1.2 software (Beckman Coulter, Inc.).

Western blotting

To investigate the effects of astragaloside IV on caspase-3 levels and EMT of PMCs in PD fluids, HMrSV5 cells were seeded in 6-well plates at a density 1×105 cells/cm2. After overnight culture, cells were treated with astragaloside IV, PD and their respective controls for 48 h. To investigate the role of RXRα on the caspase-3 levels and EMT of PMCs in PD fluids, HMrSV5 cells were seeded in 6-well plates at a density 1×105 cells/cm2. After overnight culture, cells were transfected with SD1211 empty vector (0.4 µg/cm2) or RXRα shRNA plasmid (0.4 µg/cm2) for 6 h and then cultured in fresh media for 24 h. These cells were then treated with astragaloside IV, PD and DMSO for 48 h. Cells were collected and lysed using RIPA buffer [50 mM Tris-HCl (pH 7.4), 150 mM NaCl, 1% (v/v) NP40, 0.1% (w/v) SDS, 0.5% (w/v) sodium deoxycholate] with protease inhibitor PMSF (100 mM; Beyotime Institute of Biotechnology). Equal amounts of proteins (50 µg) were separated by SDS-PAGE on 10% gels and then transferred onto PVDF membranes. After blocking with 5% non-fat milk at room temperature for 2 h, membranes were incubated overnight at 4°C with the following primary antibodies: E-cadherin (1:500; Abcam, ab1416), α-smooth muscle actin (α-SMA; 1:500; Abcam, ab32575), caspase-3 (1:500; Abcam, ab32351), β-actin (1:500; Abcam, ab179467) or GAPDH (1:1,000; Cell Signaling Technologies, Inc., 2118). This was followed by incubation with horseradish peroxidase-coupled secondary antibodies (1:1,000; Cell Signaling Technologies, Inc.; 7074 and 7076). Bands were visualized using the Enhanced Chemiluminescence Reagent kit (EMD Millipore) and analyzed using the GDS8000 system GelDoc-It310 and software VisionWorks LS v6.5.2 (UVP, LLC).

Knockdown of RXRα expression in HMrSV5 cells and determination of relative RXRα mRNA levels using reverse transcription-quantitative PCR (RT-qPCR)

HMrSV5 cells were transfected with SD1211 empty vector (0.4 µg/cm2) or RXRα shRNA plasmid (0.4 µg/cm2) using Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) and Opti-MEM (Gibco; Thermo Fisher Scientific, Inc.), which was added to cells, incubated for 4–6 h and then cultured with regular media. After 24 h of culture, total RNA was extracted from cells using a MiniBEST Universal RNA Extraction kit (Takara Bio, Inc., 9767), according to the manufacturer's instructions, and reverse transcribed using oligo dT primers and a PrimeScript II 1st Strand cDNA Synthesis kit (Takara Bio, Inc., 6210A) according to the manufacturer's instructions. The thermocycling conditions used for the reverse transcription was as follows: 30°C 10 min, 42°C 30–60 min, 95°C 5 min, and then chilled in ice. Relative mRNA levels were analyzed by RT-qPCR using a PowerUp SYBR Green Master Mix (Applied Biosystems) and an ABI 7500 fast cycler (Applied Biosystems; Thermo Fisher Scientific, Inc.). GAPDH was used for normalization. The relative RXRα mRNA levels were calculated using the 2−ΔΔCq method after normalization (53). All experiments were repeated three times. The following primers were used: GAPDH forwards, AGATCCCTCCAAAATCAAGTGG and reverse, GGCAGAGATGATGACCCTTTT; RXRα forward, CGGGAAGGTTCGCTAAGCT, and reverse, TGTTCCAGGCATTTGAGCC. Primers were designed using Primer 5 according to reference sequences in NCBI and synthesized by Invitrogen (Thermo Fisher Scientific, Inc.).

Statistical analysis

Quantitative data are expressed as the mean ± standard deviation for three experimental repeats. All the data were analyzed using SPSS 17.0 software (SPSS, Inc.). Graphics for quantitative data were processed using Prism 5 (GraphPad). Statistical analysis of the difference among multiple groups was performed by one-way ANOVA followed by the post hoc Student-Newman-Keuls test. Statistical analysis of the difference in RXRα levels in HMrSV5 cells transfected with SD1211 empty vector and those transfected with RXRα shRNA plasmid was performed using Student's t-test. P<0.05 was considered to indicate a statistically significant difference.

Results

Astragaloside IV enhances the viability of PMCs in PD fluid

To study the effect of astragaloside IV on the viability of PMCs during high-glucose PD, HMrSV5 cells were treated with PD fluid and astragaloside IV, and cell viability was determined using a CCK-8 assay. The viability of HMrSV5 cells was decreased in the PD model + vehicle groups compared with the normal + vehicle controls (Fig. 1). Viability in the PD model group was increased significantly after treatment with astragaloside IV compared with the PD model + vehicle groups (Fig. 1). Under normal conditions, the viability of HMrSV5 cells was not affected by astragaloside IV (Fig. 1). These results suggested that astragaloside IV increased viability of PMCs cultured in PD fluid, but did not affect cell viability under normal conditions.

Astragaloside IV reduces apoptosis of PMCs cultured in PD fluid

To examine the effect of astragaloside IV on the rate of apoptosis in PMCs during high glucose-based PD, PD fluid-treated HMrSV5 cells were used as a PD model. These cells were treated with astragaloside IV or vehicle control, and the rate of apoptosis was determined using flow cytometry. Additionally, the protein levels of caspase-3, a hallmark of apoptosis, were determined using western blotting. The number of apoptotic HMrSV5 cells was significantly increased in PD fluids compared with the normal control (Fig. 2A and B). Apoptosis was significantly reduced in PD model cells after treatment with astragaloside IV, while the HMrSV5 cells apoptosis was not affected by astragaloside IV under normal conditions (Fig. 2A and B). Caspase-3 protein levels in HMrSV5 cells were significantly increased in the PD model group compared with the normal control. Caspase-3 levels were significantly decreased in the PD model cells following treatment with astragaloside IV (Fig. 2C). Caspase-3 protein levels were not affected by astragaloside IV under normal conditions (Fig. 2C). These results suggested that astragaloside IV inhibited apoptosis of PMCs in the PD model, but that astragaloside IV did not affect apoptosis under normal conditions.

Astragaloside IV inhibits EMT of PMCs cultured in PD fluid

To investigate the effect of astragaloside IV on EMT of PMCs in PD, HMrSV5 were treated with high-glucose PD fluid and astragaloside IV, and the levels of E-cadherin and α-SMA were determined using western blot analysis. The level of E-cadherin was decreased in HMrSV5 cells cultured in PD fluid compared with the normal control (Fig. 3). The level of E-cadherin in the PD model was significantly increased by treatment with astragaloside IV (Fig. 3). The level of α-SMA was significantly increased in HMrSV5 cells cultured in PD fluid compared with the normal control (Fig. 3). The level of α-SMA was significantly decreased in the PD model after treatment with astragaloside IV (Fig. 3). The levels of both E-cadherin and α-SMA were not affected by astragaloside IV under normal conditions (Fig. 3). These results suggested that astragaloside IV inhibited PMC EMT induced by PD fluid, but did not affect PMC EMT under normal conditions.

Knockdown of RXRα expression in HMrSV5 cells

To knockdown the expression of RXRα in HMrSV5 cells, the RXRα shRNA plasmid was constructed using the SD1211 vector and transfected into HMrSV5 cells. The relative expression level of RXRα mRNA was determined using RT-qPCR to examine the effect and efficiency of RXRα shRNA on RXRα expression in HMrSV5 cells. The level of RXRα mRNA was decreased by ~80% in the HMrSV5 cells after transfection with SD1211 RXRα shRNA compared with the empty vector control (Fig. 4). These data suggested that RXRα expression was significantly and efficiently reduced in HMrSV5 cells by the SD1211 RXRα shRNA plasmid.

RXRα is required to maintain the viability of PMCs in PD fluids

To examine the role of RXRα in maintaining the viability of PMCs during high glucose-based PD, HMrSV5 cells were transfected with SD1211 RXRα shRNA or empty SD1211, these cells were treated with PD fluids and astragaloside IV or vehicle control, and the cell viability was determined using the CCK-8 assay. The results showed that the viability of HMrSV5 cells treated with vehicle or astragaloside IV was decreased after RXRα shRNA transfection compared with the empty vector transfections (Fig. 5). Treatment with astragaloside IV resulted in increases in the viability of HMrSV5 cells after RXRα shRNA or blank vector transfection compared with the vehicle control treatment (Fig. 5). These results suggested that a decrease in the level of RXRα results in reduced viability of PMCs in PD fluid. Therefore, RXRα is required to maintain the viability of PMCs in PD fluid.

RXRα is required to reduce apoptosis and the level of caspase-3 in PMCs cultured in PD fluid

To examine the role of RXRα in the apoptosis of PMCs during high glucose-based PD, HMrSV5 cells were transfected with SD1211 RXRα shRNA or SD1211 empty vector, these cells were treated with PD fluid and astragaloside IV or vehicle control. The rate of apoptosis was determined using flow cytometry and the level of caspase-3 was determined using western blotting. The results showed that the rate of apoptosis in HMrSV5 cells treated with vehicle or astragaloside IV were increased after RXRα shRNA transfection compared with the blank shRNA transfection (Fig. 6A and B). Treatment with astragaloside IV resulted in decreased apoptosis of HMrSV5 cells transfected with RXRα shRNA or blank vector compared with the vehicle control treatments (Fig. 6A and B). Similar changes in the levels of caspase-3 were observed (Fig. 6C). These results suggested that a decrease in the level of RXRα resulted in an increase in apoptosis and the level of caspase-3 in PMCs cultured in PD fluid. Therefore, RXRα may be involved in reducing the apoptosis of PMCs in PD fluids.

RXRα is silencing increased EMT of PMCs cultured in PD fluid

To examine the role of RXRα in EMT of PMCs during high glucose-based PD, HMrSV5 cells were transfected with SD1211 RXRα shRNA or SD1211 empty vector, and cultured in PD fluid with astragaloside IV or vehicle control. The levels of E-cadherin and α-SMA were determined using western blot analysis. The level of E-cadherin in HMrSV5 cells treated with vehicle or astragaloside IV were decreased after RXRα shRNA transfection compared with the blank shRNA transfections (Fig. 7). Treatment with astragaloside IV resulted in an increase in the level of E-cadherin in HMrSV5 cells after RXRα shRNA or blank vector transfections compared the vehicle control treatments (Fig. 7). By contrast, the level of α-SMA in HMrSV5 cells treated with vehicle or astragaloside IV were increased after RXRα shRNA transfection compared with the empty shRNA transfections (Fig. 7). Treatment with astragaloside IV resulted in a decrease in the level of α-SMA in HMrSV5 cells after RXRα shRNA or empty vector transfections compared with the vehicle control treatments (Fig. 7). These results suggested that a decrease in the level of RXRα resulted in an increase in EMT in PMCs in PD fluid. Therefore, RXRα may be required inhibit EMT in PMCs in PD fluid.

Discussion

Damage to PMCs is an initiating and important factor in peritoneal fibrosis. A number of preclinical animal and in vitro studies have revealed that the onset of peritoneal fibrosis is delayed or inhibited by promoting PMC survival and inhibiting PMC EMT (812,14,1619,21,22,24). Previous studies have revealed that several drugs can inhibit PMC EMT and inhibit peritoneal fibrosis. Melatonin can reverse lipopolysaccharide-induced EMT (54). Fluvastatin inhibits high glucose-based PD-induced fibronectin expression in human PMCs via the serum- and glucocorticoid-inducible kinase 1 pathway (55). The histone acetyltransferase inhibitor C646 reverses EMT in human PMCs via the TGF-β/Smad3 signaling pathway (56). The adenosine 5′-monophosphate (AMP)-activated protein kinase activator HL156A protects against peritoneal fibrosis (57). Suramin inhibits the occurrence and deterioration of peritoneal fibrosis (58). Selenium inhibits EMT by regulating reactive oxygen species (ROS) and the ROS/matrix metalloproteinase-9 signaling pathways and the PI3K/AKT pathways in PMCs (59). Hydrogen sulfide can improve peritoneal fibrosis by inhibiting inflammation and TGF-β synthesis (60). Metformin ameliorates the transition phenotype of PMCs and peritoneal fibrosis via the modulation of oxidative stress (61). The data in the present study showed that astragaloside IV increases cell viability and inhibits apoptosis and EMT in PMCs cultured in high-glucose PD fluid, without affecting PMCs under normal conditions. This is consistent with a previous report by Zhang et al (31). Astragaloside IV may be a potential drug that could be used for the inhibition of peritoneal fibrosis.

Previous studies have shown that several signaling pathways are involved in the protective effects of astragaloside IV in different cell types during fibrosis and under high glucose challenge. Astragaloside IV inhibits TGF-β1/PI3K/AKT-induced forehead box O3a hyper-phosphorylation and downregulation to reverse EMT during the progression of bleomycin-induced pulmonary fibrosis (62). Astragaloside IV has been reported to inhibit renal fibrosis and promote renal function in diabetic KK-Ay mice through inhibition of glucose-induced EMT in glomerular podocytes by activating autophagy and Sirtuin-1 expression, which results in decreased acetylation of NF-κB subunit p65 (63). Astragaloside IV also downregulates the calcineurin/nuclear factor of activated T cells/transient receptor potential channel 6 pathway to prevent high glucose-induced podocyte apoptosis (64). Astragaloside IV prevents high glucose-induced apoptosis and inflammatory reactions by inhibiting the JNK pathway in human umbilical vein endothelial cells (65). Astragaloside IV ameliorates high glucose-induced apoptosis and oxidative stress in the human proximal tubular HK-2 cell line by regulating the nuclear factor erythroid 2-related factor 2/antioxidant responsive element (NFE2L2/ARE) signaling pathway (66). Astragaloside IV protects primary cerebral cortical neurons from oxygen and glucose deprivation/reoxygenation by activating the cyclic AMP (cAMP)-dependent protein kinase/cAMP response element-binding protein pathway (67). Astragaloside IV inhibits cell viability, invasion, migration and TGF-β1-induced EMT in gastric cancer cells through inhibition of the PI3K/AKT/NF-κB pathway (68). Astragaloside IV inhibits the invasion and migration of hepatocellular carcinoma cells by reducing EMT via effect on the AKT/glycogen synthase kinase-3β/β-catenin pathway (69), and suppressing long noncoding RNA activated by TGF-β/interleukin-11/STAT3 signaling (70). Therefore, it is speculated that similar signaling pathways may also be involved in the protective effects of astragaloside IV in PMCs to prevent damage from high glucose-based PD fluids, in addition to the upregulation of Smad7 in the TGF-β1/Smad signaling pathway during the inhibition of TGF-β1-induced PMC EMT by astragaloside IV (31).

PMC homeostasis is important for resistance against peritoneal fibrosis (1315). Previous studies have identified several molecular components that are essential for PMC homeostasis. Heat shock protein 70 has been reported to protect PMCs from late glycation end products-induced EMT through the mitogen-activated protein kinase/ERK and TGF-β/Smad3 pathways (71,72). NF-κB mediates the inhibition of high glucose induced PMC extracellular matrix synthesis by pioglitazone (46) and the effects of chondroitin sulfate on peritoneal fibrosis (73). Twist promotes cell proliferation and EMT-induced fibrosis by regulating Y-box binding protein 1 in PMCs (74). Acidic organelles mediate TGF-β1-induced cellular fibrosis via (pro)renin receptor and vacuolar ATPase trafficking in human PMCs (75). MicroRNA-15a-5p suppressed PMC EMT (76) and microRNA-21 promoted PMC EMT (77). VDR, PPARγ and PPARβ/δ are also involved in the regulation of PMC activity and homeostasis during peritoneal dialysis (4147). As RXRα is a dimerization partner for VDR and PPAR, the role of RXRα in PMC homeostasis during PD was investigated. The data from the present study indicated that RXRα was required to maintain viability, inhibit apoptosis and reduce EMT of PMCs in high glucose-based PD fluid. Therefore, RXRα is an important factor for PMC viability and the ability to resist apoptosis and EMT induction.

It has previously been established that Astragalus membranaceus inhibits peritoneal fibrosis during PD (1820,22). The data from the present study, and the study by Zhang et al (31), showed that astragaloside IV, a component of Astragalus membranaceus, increased cell viability and inhibited apoptosis and EMT in PMCs in high glucose-based PD fluids without affecting PMCs under normal conditions, suggesting that astragaloside IV is an active, key component of Astragalus membranaceus that contributes to its anti-fibrosis function. It was also shown that RXRα was required to maintain viability, inhibit apoptosis and reduce EMT in PMCs cultured in high-glucose PD fluids. A limitation of the present study was that the cause-effect relationship between astragaloside IV and RXRα was not investigated. It has been previously shown that astragaloside IV can bind to glucocorticoid receptor (GR) with a low affinity, modulating the GR-mediated signaling pathway, including dephosphorylation of PI3K, AKT, inhibitor of κB and NF-κB in microglia (78). Astragaloside IV is a natural PPARγ agonist that suppresses the activity of β-secretase 1 and amyloid β (Aβ) levels in SH-SY5Y cells, and reduces neuritic plaque formation and Aβ levels in the brains of APP/PS1 mice, a model of Alzheimer's disease (79). Whether and how the effect of astragaloside IV on PMCs is mediated by RXRα remains to be determined; this question requires further investigation in future studies.

In conclusion, astragaloside IV increased cell viability, and inhibited apoptosis and EMT of PMCs in high-glucose PD fluid, but did not affect PMCs under normal condition. RXRα silencing reduced viability, inhibited apoptosis and reduced EMT of PMCs in high-glucose PD fluid. Astragaloside IV may be a potential drug that could be used for the inhibition of peritoneal fibrosis. RXRα was found to be an important factor involved in maintaining the viability of PMCs, and in their ability to resist apoptosis and EMT induction. The findings of the present study may provide important information for the prevention and treatment of PD-induced fibrosis.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (NSFC; grant nos. 81673912 and 81873259).

Availability of data and materials

The datasets used and/or analyzed during the present study are available from the corresponding author on reasonable request.

Authors' contributions

WZ, XZ and KG performed experiments, and collected and analyzed data. XW conceived the study and wrote the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Khan S and Rosner MH: Peritoneal dialysis for patients with end-stage renal disease and liver cirrhosis. Perit Dial Int. 38:397–401. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Vareldzis R, Naljayan M and Reisin E: The incidence and pathophysiology of the obesity paradox: Should peritoneal dialysis and kidney transplant be offered to patients with obesity and end-stage renal disease? Curr Hypertens Rep. 20:842018. View Article : Google Scholar : PubMed/NCBI

3 

Javaid MM, Khan BA and Subramanian S: Peritoneal dialysis as initial dialysis modality: A viable option for late-presenting end-stage renal disease. J Nephrol. 32:51–56. 2019. View Article : Google Scholar : PubMed/NCBI

4 

Wang WN, Zhang WL, Sun T, Ma FZ, Su S and Xu ZG: Effect of peritoneal dialysis versus hemodialysis on renal anemia in renal in end-stage disease patients: A meta-analysis. Ren Fail. 39:59–66. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Krediet RT, Abrahams AC, de Fijter CWH, Betjes MGH, Boer WH, van Jaarsveld BC, Konings CJAM and Dekker FW: The truth on current peritoneal dialysis: State of the art. Neth J Med. 75:179–189. 2017.PubMed/NCBI

6 

Krediet RT and Struijk DG: Peritoneal changes in patients on long-term peritoneal dialysis. Nat Rev Nephrol. 9:419–429. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Bargman JM: Advances in peritoneal dialysis: A review. Semin Dial. 25:545–549. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Davies SJ: Unraveling the mechanisms of progressive peritoneal membrane fibrosis. Kidney Int. 89:1185–1187. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Witowski J, Kawka E, Rudolf A and Jörres A: New developments in peritoneal fibroblast biology: Implications for inflammation and fibrosis in peritoneal dialysis. Biomed Res Int. 2015:1347082015. View Article : Google Scholar : PubMed/NCBI

10 

Raby AC and Labéta MO: Preventing peritoneal dialysis-associated fibrosis by therapeutic blunting of peritoneal Toll-like receptor activity. Front Physiol. 9:16922018. View Article : Google Scholar : PubMed/NCBI

11 

Zhang Z, Jiang N and Ni Z: Strategies for preventing peritoneal fibrosis in peritoneal dialysis patients: New insights based on peritoneal inflammation and angiogenesis. Front Med. 11:349–358. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Zhou Q, Bajo MA, Del Peso G, Yu X and Selgas R: Preventing peritoneal membrane fibrosis in peritoneal dialysis patients. Kidney Int. 90:515–524. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Lee HB and Ha H: Mechanisms of epithelial-mesenchymal transition of peritoneal mesothelial cells during peritoneal dialysis. J Korean Med Sci. 22:943–945. 2007. View Article : Google Scholar : PubMed/NCBI

14 

De Vriese AS, Tilton RG, Mortier S and Lameire NH: Myofibroblast transdifferentiation of mesothelial cells is mediated by RAGE and contributes to peritoneal fibrosis in uraemia. Nephrol Dial Transplant. 21:2549–2555. 2006. View Article : Google Scholar : PubMed/NCBI

15 

Wu J, Xing C, Zhang L, Mao H, Chen X, Liang M, Wang F, Ren H, Cui H, Jiang A, et al: Autophagy promotes fibrosis and apoptosis in the peritoneum during long-term peritoneal dialysis. J Cell Mol Med. 22:1190–1201. 2018.PubMed/NCBI

16 

Dobbie JW: Pathogenesis of peritoneal fibrosing syndromes (sclerosing peritonitis) in peritoneal dialysis. Perit Dial Int. 12:14–27. 1992.PubMed/NCBI

17 

Strippoli R, Moreno-Vicente R, Battistelli C, Cicchini C, Noce V, Amicone L, Marchetti A, Del Pozo MA and Tripodi M: Molecular mechanisms underlying peritoneal EMT and fibrosis. Stem Cells Int. 2016:35436782016. View Article : Google Scholar : PubMed/NCBI

18 

Wang L, Liu N, Xiong C, Xu L, Shi Y, Qiu A, Zang X, Mao H and Zhuang S: Inhibition of EGF receptor blocks the development and progression of peritoneal fibrosis. J Am Soc Nephrol. 27:2631–2644. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Morinelli TA, Luttrell LM, Strungs EG and Ullian ME: Angiotensin II receptors and peritoneal dialysis-induced peritoneal fibrosis. Int J Biochem Cell Biol. 77:240–250. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Bartosova M, Schaefer B, Vondrak K, Sallay P, Taylan C, Cerkauskiene R, Dzierzega M, Milosevski-Lomic G, Büscher R, Zaloszyc A, et al: Peritoneal dialysis vintage and glucose exposure but not peritonitis episodes drive peritoneal membrane transformation during the first years of PD. Front Physiol. 10:3562019. View Article : Google Scholar : PubMed/NCBI

21 

Choi SY, Ryu HM, Choi JY, Cho JH, Kim CD, Kim YL and Park SH: The role of Toll-like receptor 4 in high-glucose-induced inflammatory and fibrosis markers in human peritoneal mesothelial cells. Int Urol Nephrol. 49:171–181. 2017. View Article : Google Scholar : PubMed/NCBI

22 

Wang L and Zhuang S: The role of tyrosine kinase receptors in peritoneal fibrosis. Perit Dial Int. 35:497–505. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Tomino Y: Mechanisms and interventions in peritoneal fibrosis. Clin Exp Nephrol. 16:109–114. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Duan S, Yu J, Liu Q, Wang Y, Pan P, Xiao L, Ling G and Liu F: Epithelial-to-mesenchymal transdifferentiation of peritoneal mesothelial cells mediated by oxidative stress in peritoneal fibrosis rats. Zhong Nan Da Xue Xue Bao Yi Xue Ban. 36:34–43. 2011.PubMed/NCBI

25 

Li Z, Zhang L, He W, Zhu C, Yang J and Sheng M: Astragalus membranaceus inhibits peritoneal fibrosis via monocyte chemoattractant protein (MCP)-1 and the transforming growth factor-β1 (TGF-β1) pathway in rats submitted to peritoneal dialysis. Int J Mol Sci. 15:12959–12971. 2014. View Article : Google Scholar : PubMed/NCBI

26 

Shan G, Zhou XJ, Xia Y and Qian HJ: Astragalus membranaceus ameliorates renal interstitial fibrosis by inhibiting tubular epithelial-mesenchymal transition in vivo and in vitro. Exp Ther Med. 11:1611–1616. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Zhou X, Sun X, Gong X, Yang Y, Chen C, Shan G and Yao Q: Astragaloside IV from Astragalus membranaceus ameliorates renal interstitial fibrosis by inhibiting inflammation via TLR4/NF-кB in vivo and in vitro. Int Immunopharmacol. 42:18–24. 2017. View Article : Google Scholar : PubMed/NCBI

28 

Yu M, Shi J, Sheng M, Gao K, Zhang L, Liu L and Zhu Y: Astragalus inhibits Epithelial-to-Mesenchymal transition of peritoneal mesothelial cells by Down-regulating β-catenin. Cell Physiol Biochem. 51:2794–2813. 2018. View Article : Google Scholar : PubMed/NCBI

29 

Zhang WD, Chen H, Zhang C, Liu RH, Li HL and Chen HZ: Astragaloside IV from Astragalus membranaceus shows cardioprotection during myocardial ischemia in vivo and in vitro. Planta Med. 72:4–8. 2006. View Article : Google Scholar : PubMed/NCBI

30 

Li W and Fitzloff JF: Determination of astragaloside IV in Radix astragali (Astragalus membranaceus var. monghulicus) using high-performance liquid chromatography with evaporative light-scattering detection. J Chromatogr Sci. 39:459–462. 2001. View Article : Google Scholar : PubMed/NCBI

31 

Zhang L, Li Z, He W, Xu L, Wang J, Shi J and Sheng M: Effects of Astragaloside IV Against the TGF-β1-induced Epithelial-to-Mesenchymal transition in peritoneal mesothelial cells by promoting Smad 7 expression. Cell Physiol Biochem. 37:43–54. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Fagerberg L, Hallström BM, Oksvold P, Kampf C, Djureinovic D, Odeberg J, Habuka M, Tahmasebpoor S, Danielsson A, Edlund K, et al: Analysis of the human tissue-specific expression by genome-wide integration of transcriptomics and antibody-based proteomics. Mol Cell Proteomics. 13:397–406. 2014. View Article : Google Scholar : PubMed/NCBI

33 

Yue F, Cheng Y, Breschi A, Vierstra J, Wu W, Ryba T, Sandstrom R, Ma Z, Davis C, Pope BD, et al: A comparative encyclopedia of DNA elements in the mouse genome. Nature. 515:355–364. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Bourguet W, Ruff M, Chambon P, Gronemeyer H and Moras D: Crystal structure of the ligand-binding domain of the human nuclear receptor RXR-alpha. Nature. 375:377–382. 1995. View Article : Google Scholar : PubMed/NCBI

35 

Shulman AI and Mangelsdorf DJ: Retinoid × receptor heterodimers in the metabolic syndrome. N Engl J Med. 353:604–615. 2005. View Article : Google Scholar : PubMed/NCBI

36 

Rastinejad F: Retinoid X receptor and its partners in the nuclear receptor family. Curr Opin Struct Biol. 11:33–38. 2001. View Article : Google Scholar : PubMed/NCBI

37 

Watanabe M and Kakuta H: Retinoid X receptor antagonists. Int J Mol Sci. 19(pii): E23542018. View Article : Google Scholar : PubMed/NCBI

38 

Reitzel AM, Macrander J, Mane-Padros D, Fang B, Sladek FM and Tarrant AM: Conservation of DNA and ligand binding properties of retinoid X receptor from the placozoan Trichoplax adhaerens to human. J Steroid Biochem Mol Biol. 184:3–10. 2018. View Article : Google Scholar : PubMed/NCBI

39 

Morishita KI and Kakuta H: Retinoid X receptor ligands with anti-type 2 diabetic activity. Curr Top Med Chem. 17:696–707. 2017. View Article : Google Scholar : PubMed/NCBI

40 

Menéndez-Gutiérrez MP and Ricote M: The multi-faceted role of retinoid X receptor in bone remodeling. Cell Mol Life Sci. 74:2135–2149. 2017. View Article : Google Scholar : PubMed/NCBI

41 

Lee YC, Hung SY, Liou HH, Lin TM, Tsai CH, Lin SH, Tsai YS, Chang MY, Wang HH, Ho LC, et al: Vitamin D can ameliorate chlorhexidine gluconate-induced peritoneal fibrosis and functional deterioration through the inhibition of Epithelial-to-mesenchymal transition of mesothelial cells. Biomed Res Int. 2015:5950302015. View Article : Google Scholar : PubMed/NCBI

42 

Yang L, Wu L, Zhang X, Hu Y, Fan Y and Ma J: 1,25(OH)2D3/VDR attenuates high glucoseinduced epithelialmesenchymal transition in human peritoneal mesothelial cells via the TGFβ/Smad3 pathway. Mol Med Rep. 15:2273–2279. 2017. View Article : Google Scholar : PubMed/NCBI

43 

Yang L, Wu L, Du S, Hu Y, Fan Y and Ma J: 1,25(OH)2D3 inhibits high glucose-induced apoptosis and ROS production in human peritoneal mesothelial cells via the MAPK/P38 pathway. Mol Med Rep. 14:839–844. 2016. View Article : Google Scholar : PubMed/NCBI

44 

Su X, Yu R, Yang X, Zhou G, Wang Y, Li L and Li D: Telmisartan attenuates peritoneal fibrosis via peroxisome proliferator-activated receptor-gamma activation in rats. Clin Exp Pharmacol Physiol. 42:671–679. 2015. View Article : Google Scholar : PubMed/NCBI

45 

Su X, Zhou G, Wang Y, Yang X, Li L, Yu R and Li D: The PPARβ/δ agonist GW501516 attenuates peritonitis in peritoneal fibrosis via inhibition of TAK1-NFκB pathway in rats. Inflammation. 37:729–737. 2014. View Article : Google Scholar : PubMed/NCBI

46 

Zhou G, Su X, Ma J, Wang L and Li D: Pioglitazone inhibits high glucose-induced synthesis of extracellular matrix by NF-κB and AP-1 pathways in rat peritoneal mesothelial cells. Mol Med Rep. 7:1336–1342. 2013. View Article : Google Scholar : PubMed/NCBI

47 

Zhang YF, Wang Q, Su YY, Wang JL, Hua BJ, Yang S, Feng JX and Li HY: PPAR-γ agonist rosiglitazone protects rat peritoneal mesothelial cells against peritoneal dialysis solution-induced damage. Mol Med Rep. 15:1786–1792. 2017. View Article : Google Scholar : PubMed/NCBI

48 

Rougier JP, Moullier P, Piedagnel R and Ronco PM: Hyperosmolality suppresses but TGF beta 1 increases MMP9 in human peritoneal mesothelial cells. Kidney Int. 51:337–347. 1997. View Article : Google Scholar : PubMed/NCBI

49 

Zhao JL, Guo MZ, Zhu JJ, Zhang T and Min DY: Curcumin suppresses epithelial-to-mesenchymal transition of peritoneal mesothelial cells (HMrSV5) through regulation of transforming growth factor-activated kinase 1 (TAK1). Cell Mol Biol Lett. 24:322019. View Article : Google Scholar : PubMed/NCBI

50 

Zhang P, Dai H and Peng L: Involvement of STAT3 signaling in high glucose-induced epithelial mesenchymal transition in human peritoneal mesothelial cell line HMrSV5. Kidney Blood Press Res. 44:179–187. 2019. View Article : Google Scholar : PubMed/NCBI

51 

Chu Y, Wang Y, Zheng Z, Lin Y, He R, Liu J and Yang X: Proinflammatory effect of high glucose concentrations on HMrSV5 cells via the autocrine effect of HMGB1. Front Physiol. 8:7622017. View Article : Google Scholar : PubMed/NCBI

52 

Tani H, Sato Y, Ueda M, Miyazaki Y, Suginami K, Horie A, Konishi I and Shinomura T: Role of versican in the pathogenesis of peritoneal endometriosis. J Clin Endocrinol Metab. 101:4349–4356. 2016. View Article : Google Scholar : PubMed/NCBI

53 

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

54 

Shi S, Zhang Y, Wen W, Zhao Y and Sun L: Molecular mechanisms of melatonin in the reversal of LPS-induced EMT in peritoneal mesothelial cells. Mol Med Rep. 14:4342–4348. 2016. View Article : Google Scholar : PubMed/NCBI

55 

Zhang L, Liu J, Liu Y, Xu Y, Zhao X, Qian J, Sun B and Xing C: Fluvastatin inhibits the expression of fibronectin in human peritoneal mesothelial cells induced by high-glucose peritoneal dialysis solution via SGK1 pathway. Clin Exp Nephrol. 19:336–342. 2015. View Article : Google Scholar : PubMed/NCBI

56 

Yang Y, Liu K, Liang Y, Chen Y, Chen Y and Gong Y: Histone acetyltransferase inhibitor C646 reverses epithelial to mesenchymal transition of human peritoneal mesothelial cells via blocking TGF-β1/Smad3 signaling pathway in vitro. Int J Clin Exp Pathol. 8:2746–2754. 2015.PubMed/NCBI

57 

Ju KD, Kim HJ, Tsogbadrakh B, Lee J, Ryu H, Cho EJ, Hwang YH, Kim K, Yang J, Ahn C and Oh KH: HL156A, a novel AMP-activated protein kinase activator, is protective against peritoneal fibrosis in an in vivo and in vitro model of peritoneal fibrosis. Am J Physiol Renal Physiol. 310:F342–F350. 2016. View Article : Google Scholar : PubMed/NCBI

58 

Xiong C, Liu N, Fang L, Zhuang S and Yan H: Suramin inhibits the development and progression of peritoneal fibrosis. J Pharmacol Exp Ther. 351:373–382. 2014. View Article : Google Scholar : PubMed/NCBI

59 

Liu J, Zeng L, Zhao Y, Zhu B, Ren W and Wu C: Selenium suppresses lipopolysaccharide-induced fibrosis in peritoneal mesothelial cells through inhibition of epithelial-to-mesenchymal transition. Biol Trace Elem Res. 161:202–209. 2014. View Article : Google Scholar : PubMed/NCBI

60 

Lu Y, Gao L, Li L, Zhu Y, Wang Z, Shen H and Song K: Hydrogen sulfide alleviates peritoneal fibrosis via attenuating inflammation and TGF-β1 synthesis. Nephron. 131:210–219. 2015. View Article : Google Scholar : PubMed/NCBI

61 

Shin HS, Ko J, Kim DA, Ryu ES, Ryu HM, Park SH, Kim YL, Oh ES and Kang DH: Metformin ameliorates the phenotype transition of peritoneal mesothelial cells and peritoneal fibrosis via a modulation of oxidative stress. Sci Rep. 7:56902017. View Article : Google Scholar : PubMed/NCBI

62 

Qian W, Cai X, Qian Q, Zhang W and Wang D: Astragaloside IV modulates TGF-β1-dependent epithelial-mesenchymal transition in bleomycin-induced pulmonary fibrosis. J Cell Mol Med. 22:4354–4365. 2018. View Article : Google Scholar : PubMed/NCBI

63 

Wang X, Gao Y, Tian N, Wang T, Shi Y, Xu J and Wu B: Astragaloside IV inhibits glucose-induced epithelial-mesenchymal transition of podocytes through autophagy enhancement via the SIRT-NF-κB p65 axis. Sci Rep. 9:3232019. View Article : Google Scholar : PubMed/NCBI

64 

Yao XM, Liu YJ, Wang YM, Wang H, Zhu BB, Liang YP, Yao WG, Yu H, Wang NS, Zhang XM and Peng W: Astragaloside IV prevents high glucose-induced podocyte apoptosis via downregulation of TRPC6. Mol Med Rep. 13:5149–5156. 2016. View Article : Google Scholar : PubMed/NCBI

65 

You L, Fang Z, Shen G, Wang Q, He Y, Ye S, Wang L, Hu M, Lin Y, Liu M and Jiang A: Astragaloside IV prevents high glucoseinduced cell apoptosis and inflammatory reactions through inhibition of the JNK pathway in human umbilical vein endothelial cells. Mol Med Rep. 19:1603–1612. 2019.PubMed/NCBI

66 

Wang J and Guo HM: Astragaloside IV ameliorates high glucose-induced HK-2 cell apoptosis and oxidative stress by regulating the Nrf2/ARE signaling pathway. Exp Ther Med. 17:4409–4416. 2019.PubMed/NCBI

67 

Xue B, Huang J, Ma B, Yang B, Chang D and Liu J: Astragaloside IV protects primary cerebral cortical neurons from oxygen and glucose deprivation/reoxygenation by activating the PKA/CREB pathway. Neuroscience. 404:326–337. 2019. View Article : Google Scholar : PubMed/NCBI

68 

Zhu J and Wen K: Astragaloside IV inhibits TGF-β1-induced epithelial-mesenchymal transition through inhibition of the PI3K/Akt/NF-κB pathway in gastric cancer cells. Phytother Res. 32:1289–1296. 2018. View Article : Google Scholar : PubMed/NCBI

69 

Qin CD, Ma DN, Ren ZG, Zhu XD, Wang CH, Wang YC, Ye BG, Cao MQ, Gao DM and Tang ZY: Astragaloside IV inhibits metastasis in hepatoma cells through the suppression of epithelial-mesenchymal transition via the Akt/GSK-3β/β-catenin pathway. Oncol Rep. 37:1725–1735. 2017. View Article : Google Scholar : PubMed/NCBI

70 

Li Y, Ye Y and Chen H: Astragaloside IV inhibits cell migration and viability of hepatocellular carcinoma cells via suppressing long noncoding RNA ATB. Biomed Pharmacother. 99:134–141. 2018. View Article : Google Scholar : PubMed/NCBI

71 

Yang J, Zhu T, Liu X, Zhang L, Yang Y, Zhang J and Guο M: Heat shock protein 70 protects rat peritoneal mesothelial cells from advanced glycation end-products-induced Epithelial-to-mesenchymal transition through mitogen-activated protein Kinases/extracellular signal-regulated kinases and transforming growth factor-β/Smad pathways. Mol Med Rep. 11:4473–4481. 2015. View Article : Google Scholar : PubMed/NCBI

72 

Liu J, Bao J, Hao J, Peng Y and Hong F: HSP70 inhibits high glucose-induced Smad3 activation and attenuates epithelial-to-mesenchymal transition of peritoneal mesothelial cells. Mol Med Rep. 10:1089–1095. 2014. View Article : Google Scholar : PubMed/NCBI

73 

Abe S, Obata Y, Oka S, Koji T, Nishino T and Izumikawa K: Chondroitin sulfate prevents peritoneal fibrosis in mice by suppressing NF-κB activation. Med Mol Morphol. 49:144–153. 2016. View Article : Google Scholar : PubMed/NCBI

74 

He L, Che M, Hu J, Li S, Jia Z, Lou W, Li C, Yang J, Sun S, Wang H and Chen X: Twist contributes to proliferation and epithelial-to-mesenchymal transition-induced fibrosis by regulating YB-1 in human peritoneal mesothelial cells. Am J Pathol. 185:2181–2193. 2015. View Article : Google Scholar : PubMed/NCBI

75 

Oba-Yabana I, Mori T, Takahashi C, Hirose T, Ohsaki Y, Kinugasa S, Muroya Y, Sato E, Nguyen G, Piedagnel R, et al: Acidic organelles mediate TGF-β1-induced cellular fibrosis via (pro)renin receptor and vacuolar ATPase trafficking in human peritoneal mesothelial cells. Sci Rep. 8:26482018. View Article : Google Scholar : PubMed/NCBI

76 

Shang J, He Q, Chen Y, Yu D, Sun L, Cheng G, Liu D, Xiao J and Zhao Z: miR-15a-5p suppresses inflammation and fibrosis of peritoneal mesothelial cells induced by peritoneal dialysis via targeting VEGFA. J Cell Physiol. 234:9746–9755. 2019. View Article : Google Scholar : PubMed/NCBI

77 

Gao Q, Xu L, Yang Q and Guan TJ: MicroRNA-21 contributes to high glucose-induced fibrosis in peritoneal mesothelial cells in rat models by activation of the Ras-MAPK signaling pathway via Sprouty-1. J Cell Physiol. 234:5915–5925. 2019. View Article : Google Scholar : PubMed/NCBI

78 

Liu HS, Shi HL, Huang F, Peterson KE, Wu H, Lan YY, Zhang BB, He YX, Woods T, Du M, et al: Astragaloside IV inhibits microglia activation via glucocorticoid receptor mediated signaling pathway. Sci Rep. 6:191372016. View Article : Google Scholar : PubMed/NCBI

79 

Wang X, Wang Y, Hu JP, Yu S, Li BK, Cui Y, Ren L and Zhang LD: Astragaloside IV, a natural PPARgamma agonist, reduces Aβ production in Alzheimer's disease through inhibition of BACE1. Mol Neurobiol. 54:2939–2949. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2019
Volume 20 Issue 4

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
Zhu W, Zhang X, Gao K and Wang X: Effect of astragaloside IV and the role of nuclear receptor RXRα in human peritoneal mesothelial cells in high glucose‑based peritoneal dialysis fluids. Mol Med Rep 20: 3829-3839, 2019
APA
Zhu, W., Zhang, X., Gao, K., & Wang, X. (2019). Effect of astragaloside IV and the role of nuclear receptor RXRα in human peritoneal mesothelial cells in high glucose‑based peritoneal dialysis fluids. Molecular Medicine Reports, 20, 3829-3839. https://doi.org/10.3892/mmr.2019.10604
MLA
Zhu, W., Zhang, X., Gao, K., Wang, X."Effect of astragaloside IV and the role of nuclear receptor RXRα in human peritoneal mesothelial cells in high glucose‑based peritoneal dialysis fluids". Molecular Medicine Reports 20.4 (2019): 3829-3839.
Chicago
Zhu, W., Zhang, X., Gao, K., Wang, X."Effect of astragaloside IV and the role of nuclear receptor RXRα in human peritoneal mesothelial cells in high glucose‑based peritoneal dialysis fluids". Molecular Medicine Reports 20, no. 4 (2019): 3829-3839. https://doi.org/10.3892/mmr.2019.10604