Background: In chronic kidney disease (CKD), the accumulation of advanced oxidation protein products (AOPPs) is prevalent. Hypertrophy and epithelial-to-mesenchymal transition (EMT) of tubular cells are associated with the pathogenesis of CKD. However, whether AOPPs induce tubular-cell hypertrophy and EMT is unclear. In this study, we investigated the effect of AOPPs on human proximal tubular cells (HK-2 cells) and the mechanisms underlying tubular-cell hypertrophy and EMT in vitro. Methods: The mRNA and protein expression of CCAAT/enhancer-binding protein-homologous protein (CHOP), glucose-regulated protein (GRP) 78, p27, α-smooth muscle actin (α-SMA) and E-cadherin were evaluated by quantitative real-time PCR and western blot, respectively. Cell cycle was detected by flow cytometry. Bicinchoninic acid method was performed to measure total protein content. Results: AOPP treatment upregulated total protein expression, caused an increase in the percentage of G1-phase cells, and induced the overexpression of p27 and α-SMA, lowered the expression of E-cadherin. Furthermore, AOPP treatment induced the overexpression of GRP78 and CHOP. Moreover, the aforementioned effects were reversed following the treatment of cells with an NADPH oxidase inhibitor, a reactive oxygen species (ROS) scavenger, or salubrinal, which is an inhibitor of ER stress, whereas these effects were produced after exposure to thapsigargin, an inducer of ER stress. Conclusion: Our results suggest that AOPPs induced HK-2-cell hypertrophy and EMT by inducing ER stress, which was likely mediated by ROS. These findings could facilitate the development of novel therapeutic strategies for suppressing the progression of CKD.

Chronic kidney disease (CKD) often leads to end-stage renal disease and requires renal replacement therapy, and it is a global public health problem. CKD is known to progress to end-stage renal disease through the final common pathway of renal interstitial fibrosis [1]. Although the cellular mechanisms that facilitate tubulointerstitial fibrosis and the contribution of tubular epithelial-to-mesenchymal transition (EMT) to renal interstitial fibrosis are still controversial, tubular EMT is through to play a role in the pathogenesis of CKD [2,3,4,5,6,7,8]. Hypertrophy of tubular cells is the main cause of renal hypertrophy, because the tubulointerstitium makes up almost 90% of the kidney volume. Moreover, previous studies demonstrated that initial adaptive tubular hypertrophy will become maladaptive and lead to tubular atrophy and interstitial fibrosis, which is related to the pathogenesis of CKD [9]. Therefore, preventing the EMT and hypertrophy of renal tubular epithelial cells is critical for preventing and interrupting the accelerated progression of CKD.

Advanced oxidation protein products (AOPPs), which were first reported by Witko-Sarsat et al. in 1996, are dityrosine-containing and cross-linking protein products that are mainly formed as a result of the reaction of plasma albumin with chlorinated oxidants during oxidative stress [10,11]. Previous studies have suggested that AOPPs can be observed in the plasma of patients with early-stage CKD and that elevated levels of AOPPs are correlated positively with lesions of renal function [12]. Furthermore, growing evidence suggests that AOPPs play a crucial role in the progression of CKD [13,14,15]. AOPPs have been reported to induce podocyte apoptosis, renal tubular epithelial injury, and mesangial cell proliferation and differentiation [13,14,15,16]. However, although numerous factors might induce hypertrophy in renal tubular epithelial cells and initiate EMT in the kidney, whether AOPPs induce the hypertrophy and EMT of renal tubular epithelial cells remains poorly understood.

The endoplasmic reticulum (ER) functions as the major processing center where newly synthesized, secreted and membrane-associated proteins are correctly folded and assembled. Adverse environmental conditions such as hypoxia affect the function of the ER and cause an imbalance between the protein-folding capacity of the ER and its protein content, which results in the accumulation of unfolded or misfolded proteins in the ER lumen and leads to ER stress [17]. Unfolded protein response, which triggers both adaptive and apoptotic pathways, is initiated after ER stress [18]. ER stress has been increasingly recognized to participate in the development or pathology of several kidney diseases such as diabetic nephropathy, renal tubular interstitial lesions induced by toxins or drugs, renal ischemia-reperfusion injury, and CKD [19,20,21]. Moreover, AOPPs have been shown to induce inflammation in adipocytes by inducing ER stress in vitro [22]. However, whether AOPPs induce ER stress and how ER stress affects renal tubular epithelial cells are questions that remain poorly studied.

In this study, we investigated whether AOPPs induce hypertrophy and EMT in HK-2 cells (a human proximal tubular epithelial cell line), and whether the process is mediated by ER stress. Moreover, we examined whether oxidative stress is involved in this process.

AOPPs-BSA preparation

AOPPs-bovine serum albumin (BSA) was prepared as previously described. Briefly, a BSA solution was exposed to HOCl at a molar ratio of 1:140 for 30 min at room temperature and then dialyzed against PBS at 4ºC for 24 h to remove free HOCl. In the control incubation, native BSA was dissolved in PBS alone. All preparations were passed through a Detoxi-Gel column (Pierce, Rockford, IL, USA) in order to remove contaminating endotoxins. The endotoxin levels in the preparations were measured using the Amebocyte lysate assay kit (Sigma, St. Louis, MO, USA) and were determined to be <0.025 EU/mL. The AOPP content was determined by measuring the absorbance at 340 nm in an acidic condition and was calibrated using chloramine-T in the presence of potassium iodide.

Culture of HK-2 cells

HK-2 cells were purchased from American Type Culture Collection (Rockville, MD, USA) and cultured Dulbecco's modified Eagle's medium (DMEM/F12) supplemented with 10% heat-inactivated fetal bovine serum (FBS) and maintained at 37°C in humidified air/5% CO2. Cells were trypsinized using 0.25% trypsin-EDTA and then incubated in 6-well plates until they reached approximately 80% confluence. All experiments were performed using the differentiated HK-2 cells that were 70%-80% confluent. In the blocking experiments, cells were pretreated with the indicated inhibitors or inducer and then incubated with or without AOPPs until the end of the experiments. The inhibitors include diphenyleneiodonium (DPI) (Santa Cruz Biotechnology, Santa Cruz, CA, USA), salubrinal (Santa Cruz Biotechnology, Santa Cruz, CA, USA), and superoxide dismutase (SOD) (Cayman, USA) and the inducer is thapsigargin (Santa Cruz Biotechnology, Santa Cruz, CA, USA).

Total RNA isolation, Reverse Transcription (RT), and Real-Time RT-Polymerase Chain Reaction (RT-PCR)

Total RNA was extracted from control and treated HK-2 cells by using TRIzol reagent (Invitrogen, USA) according to the manufacturer's protocol. Total RNA was reverse transcribed by using the M-MLV reverse transcriptase kit according to the manufacturer's instructions, and the cDNA was synthesized using TRIzol and Oligo dT (Invitrogen). Quantitative real-time PCR was performed using appropriate primers in a PCR-system-9700 and then the products were examined using the SYBR Green assay (TaKaRa, Japan). The amplification conditions were the following: 95ºC for 2 min, followed by 40 cycles of 95ºC for 30 s and 60°C for 35 s. The following sets of primers were used: β-actin: 5'-TGGCACCCAGCACAATGAA-3' (forward) and 5'-CTAAGTCATAGTCCGCCTAGAAGCA-3' (reverse); CCAAT/enhancer-binding protein-homologous protein (CHOP): 5'-CCTGGAAATGAAGAGGAAGAATCAA-3' (forward) and 5'-GGAGGTGCTTGTGACCTCTG-3' (reverse); glucose-regulated protein (GRP) 78: 5'-ACCTACTCCTGCGTCGGCGTGTT-3' (forward) and 5'-CGG CATCGCCAATCAGACGTTCC-3' (reverse); p27: 5'-CACAGTGAGGGGAAGCCC CTGAA-3' (forward) and 5'-CAAGCCTCTTCCAGATCCTTATGC-3' (reverse); α-smooth muscle actin (α-SMA): 5'-TACTACTGCTGAGCGTGAGA-3' (forward) and 5'-CATCAGGCAACTCGTAACTC-3' (reverse); and E-cadherin: 5'- CTGCTGCAGG TCTCCTCTTG-3' (forward) and 5'-TGTCGACCGGTGCAATCTTC-3' (reverse). All data were normalized using the internal control β-actin, and expression levels were analyzed using the 2-DDCt method.

Western-blot analysis

Western blotting was performed as described previously. Briefly, cells were lysed in radioimmunoprecipitation assay (RIPA) buffer, and the lysates were separated using SDS-PAGE. The separated proteins were transferred to polyvinylidene fluoride (PVDF) membranes, and the blots were probed with primary antibodies (overnight, 4ºC). After incubating the blots with appropriate secondary antibodies conjugated to horseradish peroxidase, immunoreactive proteins were visualized using an enhanced chemiluminescence system and quantified densitometrically using the software LabWorks 4.5 (SensiAnsys, China). The following primary antibodies (1:1000) were used: anti-CHOP, anti-GRP78, anti-p27, anti-α-SMA, and anti-E-cadherin (Santa Cruz Biotechnology, Santa Cruz, CA, USA). The internal control was β-actin.

Flow cytometry

We used flow cytometry to analyze the cell cycle as described previously [23]. Briefly, adherent cells was collected from control and experimental groups and fixed using ice-cold 70% ethanol for 30 min. The cells were washed with PBS and then resuspended in 1 mL of cell-cycle buffer (0.05 mg/mL propidium iodide in PBS containing 1 mg/mL RNase) at 37ºC for 30 min. After staining, the distributions of cell-cycle phases were analyzed using flow cytometry (BD FACSCalibur™, USA) and the percentages of cells were measured using Multicycle software.

Determination of total protein content in HK-2 cells

Cells were treated with AOPPs and indicated reagents, after which the cells were trypsinized, washed with PBS, and counted using a hemocytometer. Equal numbers of cells were lysed in RIPA buffer containing (in PBS) 0.1% (w/v) SDS, 0.5% (w/v) sodium deoxycholate, and 1.0% (w/v) Nonidet P-40. The total protein content was measured using the bicinchoninic acid method.

Statistical analysis

All experiments were conducted in triplicate. Results are expressed as means ± SEM. Multiple groups were compared using one-way ANOVA. For comparing 2 groups, the LSD method was used, or when the assumption of equal variance did not hold, the Dunnett's T3 method was used. Differences were considered statistically significant at P < 0.05. Statistical analyses were conducted using SPSS 20.0 software.

AOPPs induced hypertrophy in HK-2 cells

Cell hypertrophy is characterized by an enlargement of cells that occurs because of an increase in protein and RNA content in the absence of DNA replication [24,25]. Normal renal tubular epithelial cells are quiescent in G0 phase, and when the cells prepare to proliferate, they enter the cell cycle at G1 phase and protein synthesis increases. G1 arrest is widely recognized to relate to antiproliferation or hypertrophy [26], and cell-cycle transit is known to be blocked by the cyclin-dependent kinase (CDK)-interacting protein/kinase-inhibitor protein(CIP/KIP) family of CDK inhibitors (CKIs:p21,p27, and p57). Tubular-cell hypertrophy is associated with the expression of p27 and p21 [27]. To investigate whether AOPPs induce HK-2-cell hypertrophy, we measured he total protein content in HK-2 cells. AOPP stimulation significantly increased the total protein content in HK-2 cells (Fig. 1). Additionally, we also measured the protein and mRNA expression of p27 and cell-cycle distribution of HK-2 cells. Treatment of the cells with AOPPs induced the overexpression of p27 at the protein (Fig. 2, A & B) and gene (Fig. 2, C & D) levels and increased the percentage of HK-2 cells arrested in G1 phase (Fig. 2 E & F) in a dose- and time-dependent manner. Although the maximal expression of p27, the total protein content, and the maximal number of cell arrested in G1 phase were not detected in the 24-h AOPP-treatment group, their levels in this group were significantly higher than that in the control group. Moreover, this effect was not observed in either the control group or the unmodified-BSA group. These results indicated that the overexpression of p27, the accumulation of total proteins, and the increased percentage of HK-2 cells arrested in G1 phase were all associated with the advanced oxidation of BSA. Taken together, these data suggested that AOPPs induced HK-2-cell hypertrophy through induction of the overexpression of p27 and G1 phase arrest.

Fig. 1

AOPPs induced hypertrophy in HK-2 cells. HK-2 cells were treated with control medium, native BSA (50 µg/ml), or indicated concentration of AOPPs for 24 h or 200 µg/mL of AOPPs for indicated times. A, B: AOPP treatment increased the total protein content in HK-2 cells, which was measured using the bicinchoninic acid method. Data are expressed as means ± SEM of 3 independent experiments; *P < 0.05 vs. control.

Fig. 1

AOPPs induced hypertrophy in HK-2 cells. HK-2 cells were treated with control medium, native BSA (50 µg/ml), or indicated concentration of AOPPs for 24 h or 200 µg/mL of AOPPs for indicated times. A, B: AOPP treatment increased the total protein content in HK-2 cells, which was measured using the bicinchoninic acid method. Data are expressed as means ± SEM of 3 independent experiments; *P < 0.05 vs. control.

Close modal
Fig. 2

AOPPs induced hypertrophy in HK-2 cells via induction of overexpression of p27 and G1 phase arrest. HK-2 cells were treated with control medium, native BSA (50 µg/ml), or indicated concentration of AOPPs for 24 h or 200 µg/mL of AOPPs for indicated times. A, B: Western-blotting results showing that AOPP treatment induced the overexpression of p27 at the protein level. C, D: RT-PCR results showing that AOPP treatment induced the overexpression of p27 at the gene level. E, F: Flow-cytometry results showing that AOPP challenge increased the percentage of cells arrested in G1 phase in a dose- and time- dependent manner. Data are expressed as means ± SEM of 3 independent experiments; *P < 0.05 vs. control.

Fig. 2

AOPPs induced hypertrophy in HK-2 cells via induction of overexpression of p27 and G1 phase arrest. HK-2 cells were treated with control medium, native BSA (50 µg/ml), or indicated concentration of AOPPs for 24 h or 200 µg/mL of AOPPs for indicated times. A, B: Western-blotting results showing that AOPP treatment induced the overexpression of p27 at the protein level. C, D: RT-PCR results showing that AOPP treatment induced the overexpression of p27 at the gene level. E, F: Flow-cytometry results showing that AOPP challenge increased the percentage of cells arrested in G1 phase in a dose- and time- dependent manner. Data are expressed as means ± SEM of 3 independent experiments; *P < 0.05 vs. control.

Close modal

AOPPs induced EMT in HK-2 cells

During EMT, renal tubular cells lose their epithelial phenotype and acquire the characteristics of mesenchymal cells. The first phase of EMT is characterized by a loss of epithelial cell adhesion, including the loss of proteins associated with epithelial membrane junctions and tight junctions, such as E-cadherin. In the intermediate phase, the cells express a combination of epithelial and mesenchymal markers such as α-smooth muscle actin (α-SMA). E-cadherin and α-SMA are the most critical proteins involved in EMT [28]. Thus, to determine whether AOPPs induce tubular EMT, we measured the expression of E-cadherin and α-SMA. Treatment with AOPPs downregulated the mRNA and protein levels of E-cadherin but upregulated those of α-SMA in a concentration- and time-dependent manner (Fig. 3). The AOPP-induced increase in the mRNA levels of α-SMA peaked at 12 h and then decreased by 24 h, but the level was still significantly more than that in the control (P < 0.05). The expression of E-cadherin and α-SMA was not changed in control cells and in cells treated with unmodified BSA, which indicated that the loss of E-cadherin and the overexpression of α-SMA were associated with the advanced oxidation of BSA. Collectively, these results indicated that AOPPs induced EMT in HK-2 cells.

Fig. 3

AOPPs induced EMT in HK-2 cells. HK-2 cells were cultured as control cells, were cultured with unmodified BSA (50 µg/ml), or were treated with the indicated concentrations of AOPPs for 24 h or with 200 µg/mL of AOPPs for indicated times. A-D: RT-PCR results showing that AOPP treatment lowered the expression of E-cadherin and induced the overexpression of α-SMA at the gene level in a concentration-and time-dependent manner. E, F: Western-blotting results showing that AOPP treatment suppressed the expression of E-cadherin and induced α-SMA overexpression at the protein level in a concentration- and time-dependent manner. Data are presented as means ± SEM of 3 independent experiments; *P < 0.05 vs. control.

Fig. 3

AOPPs induced EMT in HK-2 cells. HK-2 cells were cultured as control cells, were cultured with unmodified BSA (50 µg/ml), or were treated with the indicated concentrations of AOPPs for 24 h or with 200 µg/mL of AOPPs for indicated times. A-D: RT-PCR results showing that AOPP treatment lowered the expression of E-cadherin and induced the overexpression of α-SMA at the gene level in a concentration-and time-dependent manner. E, F: Western-blotting results showing that AOPP treatment suppressed the expression of E-cadherin and induced α-SMA overexpression at the protein level in a concentration- and time-dependent manner. Data are presented as means ± SEM of 3 independent experiments; *P < 0.05 vs. control.

Close modal

AOPPs induced ER stress in HK-2 cells

ER stress is a self-protection mechanism of cells that can restore homoeostasis within the ER. However, prolonged or severe ER stress can lead to apoptosis. GRP78 is involved in the unfolded protein response and the protection mechanism that functions during ER stress. Conversely, CHOP is associated with the injury mechanism induced by excessive ER stress. These represent 2 distinct mechanisms that are induced during ER stress and are regarded as markers of ER stress [29,30]. In this study, to examine whether AOPPs triggered ER stress, we examined the expression of GRP78 and CHOP by performing RT-PCR and western-blot assays. Compared with the levels in control cells and in cells treated with unmodified BSA, the protein and mRNA levels of GRP78 and CHOP were significantly increased in AOPP-treated HK-2 cells (Fig. 4). Although the maximal expression of CHOP and GRP78 at both gene and protein levels was not in the 24-h AOPP-treatment group, the expression levels were still at significantly higher than those in the control group. Collectively, these data indicated that AOPPs induced ER stress in HK-2 cells.

Fig. 4

AOPPs induced ER stress in cultured HK-2 cells. HK-2 cells were incubated with control medium, unmodified BSA (50 µg/ml), or indicated concentrations of AOPPs for 24 h or 200 µg/mL of AOPPs for indicated times. A-D: RT-PCR results showing that AOPP treatment increased the expression of GRP78 and CHOP at the gene level in a concentration- and time-dependent manner. E, F: Western-blotting results showing that AOPP challenge increased the expression of GRP78 and CHOP at the protein level in a concentration- and time-dependent manner. Data are presented as means ± SEM of 3 independent experiments; *P < 0.05 vs. control.

Fig. 4

AOPPs induced ER stress in cultured HK-2 cells. HK-2 cells were incubated with control medium, unmodified BSA (50 µg/ml), or indicated concentrations of AOPPs for 24 h or 200 µg/mL of AOPPs for indicated times. A-D: RT-PCR results showing that AOPP treatment increased the expression of GRP78 and CHOP at the gene level in a concentration- and time-dependent manner. E, F: Western-blotting results showing that AOPP challenge increased the expression of GRP78 and CHOP at the protein level in a concentration- and time-dependent manner. Data are presented as means ± SEM of 3 independent experiments; *P < 0.05 vs. control.

Close modal

AOPPs induced hypertrophy and EMT in HK-2 cells through induction of ER stress

To investigate the role of ER stress in AOPP-induced hypertrophy and EMT in HK-2 cells, we measured the expression of CHOP, GRP78, p27, E-cadherin, and α-SMA in cells treated without or with AOPPs and an ER stress inhibitor (salubrinal), or in cells treated with an ER stress inducer (thapsigargin) alone. AOPP again induced the overexpression of CHOP, GRP78, p27, and α-SMA and suppressed the expression of E-cadherin at the transcriptional and translational levels, and these effects of AOPPs were partly reversed when the cells were treated with both AOPPs and the ER stress inhibitor. By contrast, the aforementioned effects of AOPPs were reproduced when cells were treated with only the ER stress inducer (Fig. 5). Furthermore, the AOPP-induced increases in the total protein content in HK-2 cells and the percentage of HK-2 cells arrested in G1 phase were also partly reversed following treatment with the ER stress inhibitor, whereas both of these increases were produced in cells after treatment with the ER stress inducer alone (Fig. 6). Collectively, these results indicated that AOPP-induced hypertrophy and EMT in HK-2 cells was mediated by the activation of ER stress.

Fig. 5

AOPPs induced HK-2-cell hypertrophy and EMT through ER stress response, which was mediated by ROS. HK-2 cells were treated with 200 µg/mL of AOPPs or native BSA (50 µg/ml) for 24 h in the presence or absence of DPI (10 µmol/L), SOD (200 U/mL), or salubrinal (50 µmol/L), or they were treated with thapsigargin (0.25 µmol/L) alone. The results of RT-PCR assays (A-C) and western blotting (D) showed that AOPP-induced overexpression of α-SMA (A. b), GRP78 (B. a), p27 (C), and CHOP (B. b) and suppression of E-cadherin expression (A. a) at both gene and protein levels were partly inhibited by DPI, SOD, and salubrinal, whereas the effects of AOPPs were reproduced in cells treated with only thapsigargin. Data are expressed as means ± SEM of 3 independent experiments; *P < 0.05 vs. control, #P < 0.05 vs. AOPP-treatment group.

Fig. 5

AOPPs induced HK-2-cell hypertrophy and EMT through ER stress response, which was mediated by ROS. HK-2 cells were treated with 200 µg/mL of AOPPs or native BSA (50 µg/ml) for 24 h in the presence or absence of DPI (10 µmol/L), SOD (200 U/mL), or salubrinal (50 µmol/L), or they were treated with thapsigargin (0.25 µmol/L) alone. The results of RT-PCR assays (A-C) and western blotting (D) showed that AOPP-induced overexpression of α-SMA (A. b), GRP78 (B. a), p27 (C), and CHOP (B. b) and suppression of E-cadherin expression (A. a) at both gene and protein levels were partly inhibited by DPI, SOD, and salubrinal, whereas the effects of AOPPs were reproduced in cells treated with only thapsigargin. Data are expressed as means ± SEM of 3 independent experiments; *P < 0.05 vs. control, #P < 0.05 vs. AOPP-treatment group.

Close modal
Fig. 6

AOPPs induced HK-2-cell hypertrophy through ER stress response, which was mediated by ROS. HK-2 cells were treated with 200 µg/mL of AOPPs or native BSA for 24 h in the presence or absence of DPI (10 µmol/L), SOD (200 U/mL), or salubrinal (50 µmol/L), or they were treated with thapsigargin (0.25 µmol/L) alone. AOPP-induced increases in total protein content (A) and the percentage of cells arrested in G1 phase (B) were partly reversed by DPI, SOD, and salubrinal, but these effects of AOPPs were reproduced in cells treated with thapsigargin alone. Data are expressed as means ± SEM of 3 independent experiments; *P < 0.05 vs. control, #P < 0.05 vs. AOPP-treatment group.

Fig. 6

AOPPs induced HK-2-cell hypertrophy through ER stress response, which was mediated by ROS. HK-2 cells were treated with 200 µg/mL of AOPPs or native BSA for 24 h in the presence or absence of DPI (10 µmol/L), SOD (200 U/mL), or salubrinal (50 µmol/L), or they were treated with thapsigargin (0.25 µmol/L) alone. AOPP-induced increases in total protein content (A) and the percentage of cells arrested in G1 phase (B) were partly reversed by DPI, SOD, and salubrinal, but these effects of AOPPs were reproduced in cells treated with thapsigargin alone. Data are expressed as means ± SEM of 3 independent experiments; *P < 0.05 vs. control, #P < 0.05 vs. AOPP-treatment group.

Close modal

ER stress is associated with oxidative stress

Oxidative stress has been reported to activate ER stress [31,32,33]. Here, to determine the role of oxidative stress in ER stress in HK-2 cells, we measured the expression of CHOP, GRP78, p27, α-SMA, and E-cadherin in the presence or absence of AOPPs and either an NADPH oxidase inhibitor (diphenyleneiodonium, DPI) or superoxide dismutase (SOD), a cell-permeable scavenger of reactive oxygen species (ROS). Our results showed that AOPP-induced loss of E-cadherin and overexpression of α-SMA, CHOP, and GRP78 were partly suppressed after treatment with the NADPH oxidase inhibitor and the ROS scavenger (Fig. 5). Moreover, the increases in the total protein content in HK-2 cells and the percentage of HK-2 cells arrested in G1 phase after AOPP treatment were also partly reversed by DPI and SOD (Fig. 6). Taken together, these data suggested that oxidative stress played a role upstream or downstream of ER stress and participated in the process of AOPP-induced HK-2-cell hypertrophy and EMT.

In this study, we firstly demonstrated that AOPPs induced tubular hypertrophy and confirmed that AOPPs induced EMT in cultured human proximal renal tubular cells. Furthermore, our data also firstly revealed that AOPP-induced hypertrophy and EMT in human renal tubular cells were associated with ER stress, which was mediated by oxidative stress.

The first key finding of this study is that AOPPs induced hypertrophy in HK-2 cells. Cellular hypertrophy, an increase in cell size, is attributed to an increase in protein and RNA content in the absence of DNA replication. Previously, angiotensin II was shown to induce the overexpression of the CDK inhibitor p27 and cause cell-cycle arrest in G1 phase and lead to hypertrophy [27,34,35]. In accord with previous studies, we showed that AOPP treatment increased the total protein content in HK-2 cells, the expression of p27 at the gene level and protein levels, and the percentage of cells arrested in G1 phase. Based on these lines of evidence, we can conclude that AOPPs induce HK-2-cell hypertrophy via induction of overexpression of p27 and G1 phase arrest, which is the main cause of renal hypertrophy and which is associated with the pathogenesis of CKD.

Our results suggested that AOPPs induced not only hypertrophy but also EMT in HK-2 cells. Accumulating evidence indicates that renal tubular EMT is associated with tubulointerstitial fibrosis [2,3,36]. However, the contribution of tubular EMT to renal interstitial fibrosis is still controversial. In a previous study, it was shown that the number of myofibroblasts derived from EMT is only 5% [7]. In addition, Wilhelm et al. demonstrated that EMT might not be an in vivo process in renal fibrosis [6]. The discrepancy about the effect of EMT in kidney fibrosis may be attributed to the experimental conditions such as the mouse strain and type of disease model [37]. It is worth noting that, many studies revealed that tubular EMT plays an important role in the pathogenesis of CKD and is regarded as a potential therapeutic target for CKD [8]. The process of tubular EMT is characterized by the loss of an epithelial phenotype and the gain of profibrotic features that are characteristic of mesenchymal cells. E-cadherin and α-SMA are the most critical proteins known to be involved in EMT, during which the expression of E-cadherin is decreased and that of α-SMA is increased [4,28]. Our results showed that AOPP treatment induced tubular EMT, which was revealed by a loss of E-cadherin and the overexpression of α-SMA. By contrast, HK-2 cells treated with unmodified BSA did not exhibit marked changes in E-cadherin and α-SMA expression. These results showed that AOPPs, but not BSA, caused HK-2 cells to undergo EMT. Consistent with our finding, AOPPs were recently reported to induce EMT in human proximal renal tubular cells through oxidative stress [38]. Thus, we confirmed that AOPPs induce human proximal renal tubular cells to undergo EMT, which is related to the pathogenesis of CKD.

Various adverse conditions such as viral infection and glucose deprivation can result in the accumulation of unfolded proteins in the ER and then lead to ER stress [39]. Zhou et al. showed that AOPPs induced adipocyte perturbation through ER stress in vitro [22]. Considerable evidence suggests that ER stress is involved in a wide range of kidney diseases [19,40,41,42,43]. ER stress has been reported to play a crucial role in cardiac hypertrophy [44]. Moreover, the results of Wu et al. indicate that ER stress is closely correlated with renal tubular interstitial fibrosis in patients with diabetic nephropathy [45]. In this study, our results provided several lines of evidence suggesting that AOPP-induced hypertrophy and EMT in tubular cells was mediated by ER stress. First, exposure of HK-2 cells to AOPPs increased the expression of GRP78 and CHOP at both mRNA and protein levels, which indicated that AOPPs induced ER stress. Second, AOPP-induced hypertrophy and EMT of HK-2 cells were potently blocked by salubrinal, an inhibitor of eukaryotic initiation factor-2α dephosphorylation, which can inhibit ER stress. Lastly, thapsigargin, an inducer of ER stress, stimulated HK-2 cell hypertrophy and EMT. These results support the previous finding that ER stress might lead to EMT in HK-2 cells [46] and are consistent with the evidence suggesting that ER stress is involved in the pathological process in the progression of kidney diseases [20]. Collectively, these data indicated that AOPPs induced HK-2-cell hypertrophy and EMT by inducing ER stress.

ROS accumulation has been reported to activate ER stress [15,16,17]. However, although studies have suggested cross-talk between ER stress and oxidative stress, the mechanistic link between them is incompletely understood, and the potential effect of oxidative stress on AOPP-induced ER stress in HK-2 cells has not been previously investigated. Our results showed that an NADPH oxidase inhibitor and an ROS scavenger could partly prevent ER stress and also alleviate hypertrophy and EMT in HK-2 cells, suggesting that oxidative stress is involved in the pathway through which AOPPs induce ER stress. These results agree with the evidence indicating that oxidative stress can function upstream or downstream of ER stress [47]. Furthermore, oxidative stress can induce ER stress in neural and tumor cells [32,33], as well as in adipocytes [22]. Therefore, we conclude that oxidative stress and ER stress are associated.

Although we confirmed that AOPPs induced HK-2-cell hypertrophy and EMT through the induction of ER stress, which was mediated by ROS, whether ROS is upstream or downstream of ER stress has not been fully elucidated. Moreover, the signal transduction pathways that are activated by AOPP-induced ER stress in HK-2 cells remain to be fully elucidated. Three pathways are widely recognized to be involved in the unfolded protein response: the activating transcription factor 6 pathway, the inositol-requiring enzyme 1 pathway, and the double-stranded RNA-activated protein kinase-like ER kinase pathway [18]. Further investigation must be conducted to determine whether all of these pathways are involved in AOPP-induced hypertrophy and EMT in HK-2 cells and to identify the pathway that is most critical.

In conclusion, our results demonstrate that AOPPs induce hypertrophy and EMT in HK-2 cells. Importantly, AOPPs triggered hypertrophy and EMT in cultured HK-2 cells probably through the induction of ER stress. The induction of oxidative stress might also be involved, at least partially, in AOPP-induced tubular hypertrophy and EMT. AOPPs commonly accumulate in CKD patients and play a crucial role in the development and progression of CKD. Thus, our results provide a framework for developing novel nephroprotective therapies by targeting ER stress.

The authors declare no competing financial interests.

This work was supported by the Natural Science Foundation of Guangdong Province (No. 10151051501000030, No. S2011010004053, No. S2011040003566), the Science and Technical Plan of Guangzhou, Guangdong, China (No. 11C22120703), the Science and Technical Plan of Guangdong, China (No. 2013B021800149), and the National Natural Science Foundation of China (No. 81202842).

1.
Li Y, Sun Y, Liu F, Sun L, Li J, Duan S, Liu H, Peng Y, Xiao L, Liu Y, Xi Y, You Y, Li H, Wang M, Wang S, Hou T: Correction: Norcantharidin Inhibits Renal Interstitial Fibrosis by Blocking the Tubular Epithelial-Mesenchymal Transition. PLoS One 2013;8:e66356.
2.
Kalluri R, Neilson EG: Epithelial-mesenchymal transition and its implications for fibrosis. J Clin Invest 2003;112:1776-1784.
3.
Liu Y: Epithelial to mesenchymal transition in renal fibrogenesis: pathologic significance, molecular mechanism, and therapeutic intervention. J Am Soc Nephrol 2004;15:1-12.
4.
Burns WC, Kantharidis P, Thomas MC: The role of tubular epithelial-mesenchymal transition in progressive kidney disease. Cells Tissues Organs 2007;185:222-231.
5.
Carew RM, Wang B, Kantharidis P: The role of EMT in renal fibrosis. Cell Tissue Res 2012;347:103-116.
6.
Kriz W, Kaissling B, Le Hir M: Epithelial-mesenchymal transition (EMT) in kidney fibrosis: fact or fantasy? J Clin Invest 2011;121:468-474.
7.
LeBleu VS, Taduri G, O'Connell J, Teng Y, Cooke VG, Woda C, Sugimoto H, Kalluri R: Origin and function of myofibroblasts in kidney fibrosis. Nat Med 2013;19:1047-1053.
8.
Liu Y: New insights into epithelial-mesenchymal transition in kidney fibrosis. J Am Soc Nephrol 2010;21:212-222.
9.
Wolf G, Jablonski K, Schroeder R, Reinking R, Shankland SJ, Stahl RA: Angiotensin II-induced hypertrophy of proximal tubular cells requires p27Kip1. Kidney Int 2003;64:71-81.
10.
Witko-Sarsat V, Friedlander M, Capeillere-Blandin C, Nguyen-Khoa T, Nguyen AT, Zingraff J, Jungers P, Descamps-Latscha B: Advanced oxidation protein products as a novel marker of oxidative stress in uremia. Kidney Int 1996;49:1304-1313.
11.
Witko-Sarsat V, Friedlander M, Nguyen Khoa T, Capeillere-Blandin C, Nguyen AT, Canteloup S, Dayer JM, Jungers P, Drueke T, Descamps-Latscha B: Advanced oxidation protein products as novel mediators of inflammation and monocyte activation in chronic renal failure. J Immunol 1998;161:2524-2532.
12.
Liu B, Hou X, Zhou Q, Tian J, Zhu P, Xu J, Hou F, Fu N: Detection of advanced oxidation protein products in patients with chronic kidney disease by a novel monoclonal antibody. Free Radic Res 2011;45:662-671.
13.
Wei XF, Zhou QG, Hou FF, Liu BY, Liang M: Advanced oxidation protein products induce mesangial cell perturbation through PKC-dependent activation of NADPH oxidase. Am J Physiol Renal Physiol 2009;296:F427-437.
14.
Li HY, Hou FF, Zhang X, Chen PY, Liu SX, Feng JX, Liu ZQ, Shan YX, Wang GB, Zhou ZM, Tian JW, Xie D: Advanced oxidation protein products accelerate renal fibrosis in a remnant kidney model. J Am Soc Nephrol 2007;18:528-538.
15.
Iwao Y, Nakajou K, Nagai R, Kitamura K, Anraku M, Maruyama T, Otagiri M: CD36 is one of important receptors promoting renal tubular injury by advanced oxidation protein products. Am J Physiol Renal Physiol 2008;295:F1871-1880.
16.
Liu Y: Advanced oxidation protein products: a causative link between oxidative stress and podocyte depletion. Kidney Int 2009;76:1125-1127.
17.
Cnop M, Foufelle F, Velloso LA: Endoplasmic reticulum stress, obesity and diabetes. Trends Mol Med 2012;18:59-68.
18.
Wu J, Kaufman RJ: From acute ER stress to physiological roles of the Unfolded Protein Response. Cell Death Differ 2006;13:374-384.
19.
Kimura K, Jin H, Ogawa M, Aoe T: Dysfunction of the ER chaperone BiP accelerates the renal tubular injury. Biochem Biophys Res Commun 2008;366:1048-1053.
20.
Pallet N, Bouvier N, Beaune P, Legendre C, Thervet E, Anglicheau D: [Endoplasmic reticulum stress in kidney diseases: a question of life and death?]. Nephrol Ther 2009;5:173-180.
21.
Zhang J, Guo TT, Yang L, Du QS, Hua J, Liu RZ, Tang X: [Effect of arctiin on mouse podocyte epithelial-mesenchymal transition induced by advanced oxidation protein products]. Nan Fang Yi Ke Da Xue Xue Bao 2012;32:379-382.
22.
Zhou QG, Zhou M, Lou AJ, Xie D, Hou FF: Advanced oxidation protein products induce inflammatory response and insulin resistance in cultured adipocytes via induction of endoplasmic reticulum stress. Cell Physiol Biochem 2010;26:775-786.
23.
Lin HP, Kulp SK, Tseng PH, Yang YT, Yang CC, Chen CS, Chen CS: Growth inhibitory effects of celecoxib in human umbilical vein endothelial cells are mediated through G1 arrest via multiple signaling mechanisms. Mol Cancer Ther 2004;3:1671-1680.
24.
Fine L: The biology of renal hypertrophy. Kidney Int 1986;29:619-634.
25.
Fine LG, Norman J: Cellular events in renal hypertrophy. Annu Rev Physiol 1989;51:19-32.
26.
Preisig PA, Franch HA: Renal epithelial cell hyperplasia and hypertrophy. Semin Nephrol 1995;15:327-340.
27.
Shankland SJ, Wolf G: Cell cycle regulatory proteins in renal disease: role in hypertrophy, proliferation, and apoptosis. Am J Physiol Renal Physiol 2000;278:F515-529.
28.
Veerasamy M, Nguyen TQ, Motazed R, Pearson AL, Goldschmeding R, Dockrell ME: Differential regulation of E-cadherin and alpha-smooth muscle actin by BMP 7 in human renal proximal tubule epithelial cells and its implication in renal fibrosis. Am J Physiol Renal Physiol 2009;297:F1238-1248.
29.
Liu H, Qian J, Wang F, Sun X, Xu X, Xu W, Zhang X, Zhang X: Expression of two endoplasmic reticulum stress markers, GRP78 and GADD153, in rat retinal detachment model and its implication. Eye (Lond) 2010;24:137-144.
30.
Woo KJ, Lee TJ, Lee SH, Lee JM, Seo JH, Jeong YJ, Park JW, Kwon TK: Elevated gadd153/chop expression during resveratrol-induced apoptosis in human colon cancer cells. Biochem Pharmacol 2007;73:68-76.
31.
Gregor MF, Hotamisligil GS: Thematic review series: Adipocyte Biology. Adipocyte stress: the endoplasmic reticulum and metabolic disease. J Lipid Res 2007;48:1905-1914.
32.
Xue X, Piao JH, Nakajima A, Sakon-Komazawa S, Kojima Y, Mori K, Yagita H, Okumura K, Harding H, Nakano H: Tumor necrosis factor alpha (TNFalpha) induces the unfolded protein response (UPR) in a reactive oxygen species (ROS)-dependent fashion, and the UPR counteracts ROS accumulation by TNFalpha. J Biol Chem 2005;280:33917-33925.
33.
Holtz WA, Turetzky JM, Jong YJ, O'Malley KL: Oxidative stress-triggered unfolded protein response is upstream of intrinsic cell death evoked by parkinsonian mimetics. J Neurochem 2006;99:54-69.
34.
Wolf G, Neilson EG: Angiotensin II induces cellular hypertrophy in cultured murine proximal tubular cells. Am J Physiol 1990;259:F768-777.
35.
Wolf G, Stahl RA: Angiotensin II-stimulated hypertrophy of LLC-PK1 cells depends on the induction of the cyclin-dependent kinase inhibitor p27Kip1. Kidney Int 1996;50:2112-2119.
36.
Ng YY, Huang TP, Yang WC, Chen ZP, Yang AH, Mu W, Nikolic-Paterson DJ, Atkins RC, Lan HY: Tubular epithelial-myofibroblast transdifferentiation in progressive tubulointerstitial fibrosis in 5/6 nephrectomized rats. Kidney Int 1998;54:864-876.
37.
Inoue T, Umezawa A, Takenaka T, Suzuki H, Okada H: The contribution of epithelial-mesenchymal transition to renal fibrosis differs among kidney disease models. Kidney Int 2014;10.1038/ki.2014.235.
38.
Zhang J, Qiu M, Ma Y, Bu Y, Yang L, Tang X: [Advanced oxidation protein products induce epithelial-to-mesenchymal transition in cultured human proximal tubular epithelial cells via oxidative stress]. Nan Fang Yi Ke Da Xue Xue Bao 2014;34:659-663.
39.
Rutkowski DT, Kaufman RJ: A trip to the ER: coping with stress. Trends Cell Biol 2004;14:20-28.
40.
Kitamura M: Endoplasmic reticulum stress and unfolded protein response in renal pathophysiology: Janus faces. Am J Physiol Renal Physiol 2008;295:F323-334.
41.
Dickhout JG, Krepinsky JC: Endoplasmic reticulum stress and renal disease. Antioxid Redox Signal 2009;11:2341-2352.
42.
Inagi R: Endoplasmic reticulum stress as a progression factor for kidney injury. Curr Opin Pharmacol 2010;10:156-165.
43.
Inagi R: Endoplasmic reticulum stress in the kidney as a novel mediator of kidney injury. Nephron Exp Nephrol 2009;112:e1-9.
44.
Dickhout JG, Carlisle RE, Austin RC: Interrelationship between cardiac hypertrophy, heart failure, and chronic kidney disease: endoplasmic reticulum stress as a mediator of pathogenesis. Circ Res 2011;108:629-642.
45.
Wu J, Zhang R, Torreggiani M, Ting A, Xiong H, Striker GE, Vlassara H, Zheng F: Induction of diabetes in aged C57B6 mice results in severe nephropathy: an association with oxidative stress, endoplasmic reticulum stress, and inflammation. Am J Pathol 2010;176:2163-2176.
46.
Pallet N, Bouvier N, Bendjallabah A, Rabant M, Flinois JP, Hertig A, Legendre C, Beaune P, Thervet E, Anglicheau D: Cyclosporine-induced endoplasmic reticulum stress triggers tubular phenotypic changes and death. Am J Transplant 2008;8:2283-2296.
47.
Santos CX, Tanaka LY, Wosniak J, Laurindo FR: Mechanisms and implications of reactive oxygen species generation during the unfolded protein response: roles of endoplasmic reticulum oxidoreductases, mitochondrial electron transport, and NADPH oxidase. Antioxid Redox Signal 2009;11:2409-2427.

X. Tang, G. Rong contributed equally to this work.

Open Access License / Drug Dosage / Disclaimer
Open Access License: This is an Open Access article licensed under the terms of the Creative Commons Attribution-NonCommercial 3.0 Unported license (CC BY-NC) (www.karger.com/OA-license), applicable to the online version of the article only. Distribution permitted for non-commercial purposes only.
Drug Dosage: The authors and the publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accord with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government regulations, and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for any changes in indications and dosage and for added warnings and precautions. This is particularly important when the recommended agent is a new and/or infrequently employed drug.
Disclaimer: The statements, opinions and data contained in this publication are solely those of the individual authors and contributors and not of the publishers and the editor(s). The appearance of advertisements or/and product references in the publication is not a warranty, endorsement, or approval of the products or services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property resulting from any ideas, methods, instructions or products referred to in the content or advertisements.