Potassium bromate-induced cell model of age-related macular degeneration in vitro

  • Authors:
    • Shu-Chun Kuo
    • Yingxiao Li
    • Kai-Chun Cheng
    • Chia-Chen Hsu
    • Juei-Tang Cheng
    • Hui-Hsuan Lau
  • View Affiliations

  • Published online on: January 20, 2021     https://doi.org/10.3892/mmr.2021.11855
  • Article Number: 216
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Age‑related macular degeneration (AMD) progression occurs due to oxidative stress in retinal pigment epithelium (RPE) cells. To develop a new model of AMD, the present study investigated the effects of potassium bromate (KBrO3) on ARPE‑19 cells. Incubation with KBrO3 for 24 h significantly decreased ARPE‑19 cell viability in a concentration‑dependent manner compared with the control group. The MTT and lactate dehydrogenase assay results indicated that KBrO3 induced cell apoptosis. Compared with the control group, KBrO3 treatment significantly decreased the Bcl2/Bax ratio, as determined via western blotting, and caspase‑3 mRNA expression levels. Fluorescence microscopy indicated the increased ROS levels in cells treated with KBrO3. Endogenous antioxidant enzyme activities, including superoxide dismutase and glutathione peroxidase, were significantly inhibited by KBrO3 compared with the control group. Moreover, the antioxidants tiron and phloroglucinol inhibited KBrO3‑mediated effects on ARPE‑19 cells in a dose‑dependent manner. Additionally, GPR109A is the binding site of 4‑hydroxynonenal (4‑HNE). KBrO3 displayed cytotoxic effects in 293 cells, which naturally lack the GPR109A gene, but these effects were not observed in 4‑HNE‑treated 293 cells, suggesting that KBrO3 induced apoptosis without increasing endogenous 4‑HNE levels in cells. Moreover, the results suggested that KBrO3‑induced oxidative stress may activate STAT3 to increase VEGF expression in ARPE‑19 cells. Collectively, the results of the present study supported the potential use of KBrO3 to induce an in vitro model of AMD in ARPE‑19 cells.

Introduction

Aging and other factors, including increased oxidative stress and chronic inflammation, can diminish the function of the retinal pigment epithelium (RPE), which is located between the photoreceptors and vascular choroid in the eye, leading to eventual degeneration (1). Degenerated RPE cells may cause irreversible injury to photoreceptors and a disturbance in central vision, resulting in age-related macular degeneration (AMD) (2).

AMD is a progressive disease associated with retinal degeneration that can cause irreversible vision loss in elderly populations. A gradual increase in the prevalence of AMD has been observed among older individuals (3). AMD is a complex and multifaceted disease that is caused by a combination of genetic and environmental factors (4). Drusen accumulation in the macula between the RPE and Bruch's membrane has been observed in the early stages of AMD (4). Subsequently, AMD progresses to interconvertible ‘dry’ and ‘wet’ forms in the later stages (5). Dry AMD is characterized by the extensive loss of RPE cells, the overlying photoreceptors and the underlying choriocapillaris (6). Therefore, RPE cell degeneration by factors including increased oxidative stress and chronic inflammation may promote the irreversible injury of photoreceptors and loss of central vision, thus inducing AMD (7). However, at present, no effective treatment is available for AMD (8). Therefore, developing a new AMD model to investigate strategies for the prevention or delayed progression of the disease is important.

Oxidative stress is an important factor in the pathogenesis of AMD (9) that may initiate RPE cell damage, resulting in chronic inflammation (10). RPE cells can digest photoreceptor outer rod segments (POSs); however, the digestive rate of POSs decreases during oxidative stress, resulting in accumulated POS, which promotes lipofuscin formation, one of the major pathological alterations observed in AMD (11). Therefore, reducing oxidative stress may serve as a potential therapeutic strategy to prevent AMD.

Hydrogen peroxide (H2O2) is widely utilized to induce oxidative stress in human RPE cells (12), displaying dose- and time-dependent effects (12,13). Chemical oxidants, including tert-butyl hydroperoxide (t-BHP) (14) and bisphenol A (15), are also used to induce oxidative stress in vitro. Additionally, acrolein-induced reactive oxygen species (ROS) overproduction has been employed for the induction of oxidative stress (16). RPE cells are sensitive to oxidative stress as they contain photosensitizers that produce ROS in response to visible light (10). The antioxidative effects of RPE cells decrease with age, and aged RPE cells display an oxidative burden, which is a sign of AMD progression (12).

Potassium bromate (KBrO3) is an oxidant that is used as a food additive during the bread-baking process because it decomposes into a stable compound, potassium bromide (17). KBrO3-induced genotoxicity in HepG2 cells indicates that KBrO3 generates a variety of ROS, resulting in oxidative DNA damage (18). Additionally, KBrO3 induces an effect in cultured cardiac cells that was similar to the effect mediated by H2O2 (19). Superoxide anion radical generation by KBrO3 has also been observed (20). The aforementioned studies indicated that KBrO3 and H2O2 display the potential to induce AMD, thus it was hypothesized that KBrO3 may be used to induce AMD in cells.

In the present study, cultured ARPE-19 cells were employed to identify the effective dose of KBrO3 and explore the possibility of developing a new model of AMD.

Materials and methods

Cell culture

The human RPE cell line (ARPE-19) was obtained from The Culture Collection and Research Center, Food Industry Research and Development Institute [Bioresource Collection & Research Center (BCRC) no. 60383]. ARPE-19 cells were cultured in DMEM/F12 medium (Cytiva) with 10% fetal calf serum (Cytiva) and antibiotics (cat no. 15240062; Thermo Fisher Scientific, Inc.; solution contains 10,000 U/ml of penicillin, 10,000 µg/ml of streptomycin, and 25 µg/ml of Gibco Amphotericin B) at 37°C with 5% CO2 (12). 293 cells (BCRC no. 60019) were cultured in minimum essential medium (MEM) (Cytiva). After 24 h, the cell culture medium was refreshed. Following culture for 24 h, cells were used for subsequent experiments.

Cell viability assay

ARPE-19 and 293 cell viability was assessed by performing the MTT assay (19). The purple formazan was dissolved by DMSO. Briefly, ARPE-19 and 293 cells were seeded at 5,000 cells per well in 24-well plates and incubated with KBrO3 (25, 50 and 75 µM; Sigma-Aldrich, Merck KGaA) at 37°C for 24 h. 4-Hydroxynonenal (4-HNE; Cayman Chemical Company) and H2O2 (Sigma-Aldrich; Merck KGaA) was used as a reference to assess the effects of KBrO3. Subsequently, the culture medium was refreshed and 50 µl MTT (0.5 mg/ml) was added to each well for 4 h. The absorbance was measured at a wavelength of 595 nm using a Synergy HT Multi-Mode Microplate Reader (BioTek Instruments, Inc.).

Additionally, the cytotoxicity effect of KBrO3 in ARPE-19 and 293 cells was assessed using the lactate dehydrogenase (LDH) release assay kit (BioVision, Inc.) as previously described (12). The number of dead cells in the medium of cultured cells was measured to evaluate LDH activity. The absorbance was measured at a wavelength of 490 nm using a microplate spectrophotometer.

Determination of intracellular superoxide levels

According to the previously described protocol (19), ARPE-19 cells were seeded (7.5×103 cells/ml) into 24-well plates and incubated for 24 h. Following starvation in serum-free medium at 37°C for 4 h, cells were incubated with increasing amounts of KBrO3 (25, 50 and 75 µM) at 37°C for 72 h. Superoxide levels were measured by incubating cells with dihydroergotamine (DHE; Thermo Fisher Scientific, Inc.), which reacts with intracellular superoxide ions, for 30 min at 37°C. The entire field of vision was observed using an IX71 fluorescence microscope (Olympus Corporation; magnification, ×200) and a DP2-BSW imaging system (Olympus Corporation). The results were quantified using ImageJ software v2.0.0 (National Institutes of Health) (21).

Assay of intracellular ROS levels

ROS levels in ARPE-19 cells were assessed using the 2′,7′-dichlorodihydro-fluorescein (DCFH-DA) fluorescent probe (Sigma-Aldrich; Merck KGaA). ARPE-19 cells at 1×106 cells/ml were incubated with 25 µM DCFH-DA for 1 h at 37°C. Cells were pretreated with tiron (1 and 5 µM, Sigma-Aldrich; Merck KGaA) and phloroglucinol (1 and 5 µM, Sigma-Aldrich; Merck KGaA) for 30 min prior to KBrO3 at 37°C. The resulting fluorescence was measured at an excitation wavelength of 485 nm and an emission wavelength of 535 nm using a fluorescence plate reader as previously described (22).

Assay of antioxidant enzymes

Cells were harvested and sonicated at high frequency on ice. The lysates were incubated on ice for 30 min, to ensure protein solubilization. Cell debris was centrifuged at 10,000 × g for 20 min at 4°C. Total protein concentrations were measured by bicinchoninic acid protein assay (Thermo Fisher Scientific, Inc.).

Antioxidant enzyme activities, including superoxide dismutase (SOD) and glutathione peroxidase (GSHPx), were measured in total cell protein extracts using colorimetric assay kits (cat no. K555; BioVision, Inc.) according to the manufacturer's instructions. The absorbance was measured at a wavelength of 560 nm using a microplate spectrophotometer.

Western blotting

Total protein was extracted from ARPE-19 and 293 cell lysates using ice-cold RIPA buffer (Thermo Fisher Scientific, Inc.) (19). Total protein concentrations were measured by bicinchoninic acid protein assay (Thermo Fisher Scientific, Inc.). Proteins (10 µg) were separated by 10% or 15% SDS-PAGE and electro-transferred onto polyvinylidene fluoride membrane (EMD Millipore). After blocking with 5% (v/v) bovine serum albumin (MDBio, Inc.) for 1 h at room temperature, membranes were incubated with primary antibodies, Bcl2 (cat. no. 2870, 1:1,000; Cell Signaling Technology, Inc.), Bax (cat. no. 2772, 1:1,000; Cell Signaling Technology, Inc.), phosphorylated (p-)STAT3 (cat. no. ab76315, 1:1,000; Abcam), STAT3 (cat. no. ab68153, 1:1,000; Abcam) or β-actin (cat. no. sc-1615, 1:5,000; Santa Cruz Biotechnology, Inc.) at 4°C overnight. The membranes were then incubated with the corresponding secondary antibodies, goat anti-rabbit IgG (cat. no. 12-348, 1:5,000; Sigma-Aldrich; Merck KGaA) or goat anti-mouse IgG (cat. no. 12-349, 1:5,000; Sigma-Aldrich; Merck KGaA) at room temperature for 1 h. Protein bands were visualized using a chemiluminescence kit (PerkinElmer, Inc.). The optical densities of the bands for Bax (18 kDa), Bcl2 (100 kDa) and β-actin (43 kDa) were quantified using software (Gel-Pro Analyzer v4.0 software; Media Cybernetics Inc.) (23).

Reverse transcription-quantitative PCR (qPCR)

VEGF mRNA expression levels in ARPE-19 cells were measured. Total RNA was extracted from ARPE-19 and 293 cells using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.). Reverse-transcriptase reactions contained 5 µg of total RNA extract, 4 µl 5X PCR buffer, 1 µl 0.1 M DTT, 1 µl SuperScript™ III RT (cat. no. 18080093; Invitrogen; Thermo Fisher Scientific, Inc.), which were performed at room temperature for 10 min, then at 42°C for 30 min and at 95°C for 5 min. Subsequently, qPCR was performed using a LightCycler (Roche Diagnostics GmbH). The qPCR reactions contained LightCycler DNA Master Hybridization Probes reaction mix (Roche Diagnostics GmbH) and the dual-labeled fluorescent probes. PCR cycles were: Pretreatment at 95°C for 10 sec, 96°C for 10 sec, 60°C for 30 sec, 72°C for 1 sec, 40°C for 30 sec (45 cycles) and a final extension at 72°C for 10 min. Analysis of relative gene expression data was performed using the 2−ΔΔCq method (24). The following primers were used for qPCR: VEGF forward, 5′-AGGAGGGCAGAATCATCACG-3′ and reverse, 5′-CAAGGCCCACAGGGATTTTCT-3′; caspase-3 forward, 5′-CCGACTTCCTGTATGCTTACTCTA-3′ and reverse, 5′-CATGACCCGTCCCTTGAA-3′; and β-actin forward, 5′-CTAAGGCCAACCGTGAAAAG-3′ and reverse, 5′-GCCTGGATGGCTACGTACA-3′. VEGF mRNA, caspase-3 mRNA expression levels were normalized to the internal reference gene β-actin.

Statistical analysis

Data are presented as the mean ± SEM (n=6). All statistical analyses were carried out by SPSS v21 (IBM Corp.). Comparisons among multiple groups were analyzed using one-way ANOVA followed by Tukey's post hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

Effects of KBrO3 on ARPE-19 cell viability

Compared with the control group, incubation with KBrO3 for 24 h significantly inhibited ARPE-19 cell viability in a concentration-dependent manner KBrO3 decreased cell viability, and increased cell apoptosis and cell cytotoxicity at all three tested concentrations (Fig. 1A). The Bcl2/Bax ratio is a useful index for evaluating KBrO3-induced apoptosis (25). KBrO3 significantly decreased the Bcl2/Bax ratio in a concentration-dependent manner compared with the control group (Fig. 1B). Caspase-3 activity also serves as a biomarker of apoptosis (26). KBrO3 significantly decreased Caspase-3 expression levels in a concentration-dependent manner compared with the control group (Fig. 1C). The results were further supported by the results of the LDH assay that demonstrated that KBrO3 significantly increased cell cytotoxicity in a concentration-dependent manner compared with the control group (Fig. 1D). In contrast to the other 2 pro-oxidants (4-NHE, H2O2), KBrO3 showed lower cell toxicity at 25 µM. Moreover, 75 µM KBrO3 and 30 µM 4-HNE, which has been widely used to induce oxidative stress in RPE cells, displayed similar effects on ARPE-19 cells (23).

Effects of KBrO3 on oxidative stress biomarkers in ARPE-19 cells

In ARPE-19 cells, KBrO3 significantly inhibited the activities of antioxidant enzymes, including SOD or GSHPx, in a concentration-dependent manner compared with the control group (Fig. 2A and B). Therefore, the results indicated that KBrO3 induced oxidative stress in ARPE-19 cells.

To determine intracellular superoxide levels in ARPE-19 cells, DCFH-DA was used to detect intracellular ROS levels and DHE was used to detect intracellular superoxide levels. Compared with the control group, KBrO3 treatment significantly elevated ROS and superoxide levels (Fig. 2C and D). KBrO3-mediated effects were significantly inhibited by the antioxidants tiron and phloroglucinol in a concentration-dependent manner (Fig. 2C and D).

Inhibitory effects of antioxidants on KBrO3-mediated effects in ARPE-19 cells

Intracellular ROS levels were measured using the DCFH-DA fluorescence probe. Compared with the control group, KBrO3 significantly increased intracellular ROS levels in a concentration-dependent manner (Fig. 3A). Additionally, the effects of KBrO3 on intracellular ROS levels were significantly inhibited by the antioxidants tiron and phloroglucinol (Fig. 3B and C).

Moreover, compared with the control group, KBrO3 notably increased Bax protein expression levels in ARPE-19 cells. The antioxidant phloroglucinol markedly inhibited KBrO3-induced Bax protein expression levels in a concentration-dependent manner (Fig. 3E). Additionally, phloroglucinol reversed KBrO3-mediated downregulation of Bcl2 protein expression levels in ARPE-19 cells (Fig. 3E). Therefore, phloroglucinol significantly attenuated KBrO3-mediated downregulation of the Bcl2/Bax ratio in ARPE-19 cells (Fig. 3E). The antioxidant tiron displayed similar effects on the Bcl2/Bax ratio (Fig. 3D). The results indicated that antioxidants inhibited KBrO3-mediated effects on apoptotic factors in ARPE-19 cells.

Role of 4-HNE in KBrO3-mediated effects

KBrO3-induced apoptosis might be mediated via endogenous 4-HNE in ARPE-19 cells, as 4-HNE is produced in RPE cells during high oxygen consumption (27). Recently, 4-HNE was reported to serve as an agonist of the nicotinic receptor GPR109A (28). Therefore, 293 cells lacking the GPR109A gene (29) were used to investigate the effects of KBrO3.

Compared with the control group, the three tested concentrations of KBrO3 significantly inhibited 293 cell viability in a concentration-dependent manner (Fig. 4A), although the effective dose was higher and the incubation time was two-fold higher in 293 cells compared with ARPE-19 cells. However, compared with the control group, 4-HNE failed to significantly inhibit 293 cell viability at a concentration sufficient to inhibit ARPE-19 cell viability. Additionally, compared with the control group, KBrO3 significantly induced 293 cell apoptosis, but 4-HNE did not significantly alter 293 cell apoptosis as demonstrated by significantly increased caspase-3 expression levels in KBrO3-treated cells compared with control cells, but no significant alteration in caspase-3 expression levels in 4-HNE-treated cells compared with control cells (Fig. 4B). KBrO3 showed higher apoptosis induction in all fractionated doses in contrast to 4-HNE. Similarly, KBrO3 significantly increased cytotoxicity in 293 cells compared with the control group, but 4-HNE did not significantly increase cytotoxicity in 293 cells (Fig. 4C). Moreover, the antioxidant tiron significantly increased the Bcl2/Bax ratio in KBrO3-treated 293 cells in a concentration-dependent manner (Fig. 4D). Therefore, the results suggested that 4-HNE did not serve a role in KBrO3-induced apoptosis.

Effects of KBrO3 on VEGF expression in ARPE-19 cells

Choroidal neovascularization (CNV) occurs during AMD, and VEGF is a critical signal in AMD-associated CNV (30). ROS inhibition may reduce VEGF mRNA expression levels (31). Therefore, alterations in VEGF mRNA expression levels in ARPE-19 cells treated with KBrO3 for 48 h were investigated.

Compared with the control group, KBrO3 significantly increased VEGF mRNA expression levels in a concentration-dependent manner at concentrations sufficient to induce ROS production (Fig. 5A). The upstream STAT3 may enhance VEGF gene expression (32). Compared with the control group, KBrO3 treatment also significantly increased STAT3 phosphorylation (Fig. 5B), which was inhibited by tiron. Cells were pretreated with stattic (0.5 and 1 µM; Sigma-Aldrich; Merck KGaA) for 30 min prior to addition of KBrO3. Stattic significantly attenuated KBrO3-mediated upregulation of VEGF mRNA expression levels (Fig. 5C). However, stattic failed to inhibit KBrO3-induced ROS production (Fig. 5D). Therefore, the results suggested that KBrO3 activated STAT3 via ROS to promote VEGF expression in ARPE-19 cells.

Discussion

The present study identified a novel AMD-like model using KBrO3 to induce oxidative stress in ARPE-19 cells. In the present study, KBrO3-induced cell injury was reversed by antioxidants. Moreover, the results suggested that KBrO3 induced apoptosis without mediating endogenous 4-HNE in ARPE-19 cells.

KBrO3 is an established oxidant (18), although to the best of our knowledge, its effects on ARPE-19 cells have not been previously reported. Oxidants, including H2O2 and t-BHP, have been reported to produce ROS in cells (12,13,16). In the present study, KBrO3 significantly induced ROS production and apoptosis in ARPE-19 cells in a concentration-depended manner. RPE cells may digest photoreceptor POSs (10); however, oxidative stress reduces the digestive rate of POS (10), and higher POS results in lipofuscin formation, one of the major alterations associated with AMD (11). Therefore, the present study indicated that KBrO3 may be useful to establish a cell model of AMD in ARPE-19 cells.

Similar to other oxidants, in the present study, KBrO3 significantly increased cytotoxicity in ARPE-19 cells compared with the control group. Moreover, compared with the control group, KBrO3 inhibited ARPE-19 cell viability in a dose-dependent manner at the tested concentrations (25, 50 and 75 µM). At 75 µM, KBrO3 induced ARPE-19 cell apoptosis to a similar level to ARPE-19 cell apoptosis induced by 30 µM 4-HNE. Also, 4-HNE produced in the RPE during high oxygen consumption is associated with AMD (33). Therefore, 4-HNE was used as a reference in the present study. Treatment with at higher concentrations (30 µM) causes cytotoxicity and ROS formation effect in cells compared with that in KBrO3 (25 µM) (34). In addition, t-BHP has been reported to induce cell apoptosis at doses >250 µM (14). In RPE cells, exposure to 1 mM H2O2 for 24 h markedly inhibited viability compared with the control group (12,13). Therefore, the results of the present study suggested that KBrO3 might be more potent than the aforementioned oxidants in ARPE-19 cells.

It has been reported that KBrO3-induced apoptosis is mediated via endogenous 4-HNE in ARPE-19 cells (33). 4-HNE is effective as an agonist for the nicotinic receptor GPR109A (28), thus 293 cells, which lack GPR109A expression (29), were used in the present study. In 293 cells, compared with the control group, KBrO3 significantly induced apoptosis and cytotoxicity, but 4-HNE did not display these effects. Therefore, the results suggested that endogenous 4-HNE was not involved in KBrO3-induced apoptosis.

The present study demonstrated that KBrO3 induced oxidative stress, which promoted cytotoxicity and apoptosis in ARPE-19 cells, but KBrO3-mediated effects were reversed by treatment with the antioxidants tiron and phloroglucinol. Tiron is a mitochondria-targeted antioxidant (35) and phloroglucinol protects RPE (36). The mediation of ROS in KBrO3-treated ARPE cells was also increased in a concentration-dependent manner. Moreover, KBrO3 treatment significantly inhibited antioxidant enzyme activities, including SOD, GSHPx in ARPE-19 cells, suggesting that KBrO3-induced oxidative stress in ARPE-19 cells. This was consistent with previous studies using hepatic (18) and cardiac (19) cells.

CNV occurs during the ‘wet’ phase of AMD, which occurs during the late stages of the disease (37). Hyperglycemia-induced oxidative stress may induce activation of STAT3-regulated VEGF expression in RPE cells (38). In the present study, the results suggested that KBrO3 activated STAT3 to promote VEGF expression in ARPE-19 cells. The results indicated that oxidative stress mediated this effect as KBrO3-induced effects were reversed by antioxidants. Moreover, the STAT3 inhibitor stattic significantly attenuated KBrO3-induced upregulation of VEGF mRNA expression levels at a dose that failed to inhibit KBrO3-induced ROS production. Stattic is an established STAT3 inhibitor that is pharmacologically applied in vitro and in vivo (39). Collectively, the results indicated that KBrO3-induced oxidative stress may activate STAT3 to promote VEGF expression in ARPE-19 cells.

KBrO3 is easily prepared in solution for both in vitro and in vivo investigations (40). As an oxidative stress inducer in cells, KBrO3 displays improved stability compared with H2O2 (41). However, the application of KBrO3 in animals may result in damage to the kidney when injury is initiated (42), whereas the ROS scavenger rutin may inhibit KBrO3 (43). Therefore, systemic application of KBrO3 to induce AMD may be difficult to achieve in animals. However, local application of KBrO3 in the eye may serve as an alternative method to prepare AMD model animals, but further investigation is required.

In conclusion, the present study identified a novel method to establish an AMD cell model using KBrO3 as an oxidant in ARPE-19 cells. KBrO3 treatment may serve as a simple and useful method to induce AMD cell models in the future.

Acknowledgements

The authors would like to thank laboratory assistants of Chi-Mei Medical Center, Ms. Y.L. Yen and Ms. Y.P. Lin, for their kind help with the experiments.

Funding

No funding was received.

Availability of data and materials

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

Authors' contributions

SCK and JTC designed the study and analyzed the data. YL assisted with performing the experiments and prepared the manuscript. JTC supervised the project. CCH analyzed the data and prepared the manuscript. KCC analyzed the data and revised the manuscript. HHL contributed to the study design and performed the VEGF assay. All authors read and approved the final 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 

Datta S, Cano M, Ebrahimi K, Wang L and Handa JT: The impact of oxidative stress and inflammation on RPE degeneration in non-neovascular AMD. Prog Retin Eye Res. 60:201–218. 2017. View Article : Google Scholar : PubMed/NCBI

2 

van Lookeren Campagne M, LeCouter J, Yaspan BL and Ye W: Mechanisms of age-related macular degeneration and therapeutic opportunities. J Pathol. 232:151–164. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Jager RD, Mieler WF and Miller JW: Age-related macular degeneration. N Engl J Med. 358:2606–2617. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Chen Y, Bedell M and Zhang K: Age-related macular degeneration: Genetic and environmental factors of disease. Mol Interv. 10:271–281. 2010. View Article : Google Scholar : PubMed/NCBI

5 

Coleman HR, Chan CC, Ferris FL III and Chew EY: Age-related macular degeneration. Lancet. 372:1835–1845. 2008. View Article : Google Scholar : PubMed/NCBI

6 

Bhutto I and Lutty G: Understanding age-related macular degeneration (AMD): Relationships between the photoreceptor/retinal pigment epithelium/Bruch's membrane/choriocapillaris complex. Mol Aspects Med. 33:295–317. 2012. View Article : Google Scholar : PubMed/NCBI

7 

Ding X, Patel M and Chan CC: Molecular pathology of age-related macular degeneration. Prog Retin Eye Res. 28:1–18. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Lorés-Motta L, de Jong EK and den Hollander AI: Exploring the use of molecular biomarkers for precision medicine in age-related macular degeneration. Mol Diagn Ther. 22:315–343. 2018. View Article : Google Scholar : PubMed/NCBI

9 

Bonilha VL: Oxidative Stress Regulation and DJ-1 Function in the Retinal Pigment Epithelium: Implications for AMD. Adv Exp Med Biol. 1074:3–9. 2018. View Article : Google Scholar : PubMed/NCBI

10 

Liang FQ and Godley BF: Oxidative stress-induced mitochondrial DNA damage in human retinal pigment epithelial cells: A possible mechanism for RPE aging and age-related macular degeneration. Exp Eye Res. 76:397–403. 2003. View Article : Google Scholar : PubMed/NCBI

11 

Wihlmark U, Wrigstad A, Roberg K, Brunk UT and Nilsson SE: Lipofuscin formation in cultured retinal pigment epithelial cells exposed to photoreceptor outer segment material under different oxygen concentrations. APMIS. 104:265–271. 1996. View Article : Google Scholar : PubMed/NCBI

12 

Hu X, Liang Y, Zhao B and Wang Y: Thymoquinone protects human retinal pigment epithelial cells against hydrogen peroxide induced oxidative stress and apoptosis. J Cell Biochem. 120:4514–4522. 2019. View Article : Google Scholar : PubMed/NCBI

13 

Lu L, Hackett SF, Mincey A, Lai H and Campochiaro PA: Effects of different types of oxidative stress in RPE cells. J Cell Physiol. 206:119–125. 2006. View Article : Google Scholar : PubMed/NCBI

14 

Shen C, Ma W, Zheng W, Huang H, Xia R, Li C and Zhu X: The antioxidant effects of riluzole on the APRE-19 celll model injury-induced by t-BHP. BMC Ophthalmol. 17:2102017. View Article : Google Scholar : PubMed/NCBI

15 

Abdel-Wahab WM: Thymoquinone attenuates toxicity and oxidative stress induced by bisphenol A in liver of male rats. Pak J Biol Sci. 17:1152–1160. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Sheu SJ, Chao YM, Liu NC and Chan JY: Differential effects of bevacizumab, ranibizumab and aflibercept on cell viability, phagocytosis and mitochondrial bioenergetics of retinal pigment epithelial cell. Acta Ophthalmol. 93:e631–e643. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Cunningham DK and Anderson JA: Decomposition of bromate in fermenting and nonfermenting doughs. Cereal Chem. 33:290–299. 1956.

18 

Zhang Y, Jiang L, Jiang L, Geng C, Li L, Shao J and Zhong L: Possible involvement of oxidative stress in potassium bromate-induced genotoxicity in human HepG2 cells. Chem Biol Interact. 189:186–191. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Kuo SC, Li Y, Cheng YZ, Lee WJ, Cheng JT and Cheng KC: Molecular mechanisms regarding potassium bromate induced cardiac hypertrophy without apoptosis in H9c2 cells. Mol Med Rep. 18:4700–4708. 2018.PubMed/NCBI

20 

Watanabe S, Togashi S and Fukui T: Contribution of nitric oxide to potassium bromate-induced elevation of methaemoglobin concentration in mouse blood. Biol Pharm Bull. 25:1315–1319. 2002. View Article : Google Scholar : PubMed/NCBI

21 

Li CJ, Lv L, Li H and Yu DM: Cardiac fibrosis and dysfunction in experimental diabetic cardiomyopathy are ameliorated by alpha-lipoic acid. Cardiovasc Diabetol. 11:732012. View Article : Google Scholar : PubMed/NCBI

22 

Liu WY, Liou SS, Hong TY and Liu IM: The benefits of the citrus flavonoid diosmin on human retinal pigment epithelial cells under high-glucose conditions. Molecules. 22:22512017. View Article : Google Scholar

23 

Hytti M, Piippo N, Salminen A, Honkakoski P, Kaarniranta K and Kauppinen A: Quercetin alleviates 4-hydroxynonenal-induced cytotoxicity and inflammation in ARPE-19 cells. Exp Eye Res. 132:208–215. 2015. View Article : Google Scholar : PubMed/NCBI

24 

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

25 

Salakou S, Kardamakis D, Tsamandas AC, Zolota V, Apostolakis E, Tzelepi V, Papathanasopoulos P, Bonikos DS, Papapetropoulos T, Petsas T, et al: Increased Bax/Bcl-2 ratio up-regulates caspase-3 and increases apoptosis in the thymus of patients with myasthenia gravis. In Vivo. 21:123–132. 2007.PubMed/NCBI

26 

Wang Y, Shen D, Wang VM, Yu CR, Wang RX, Tuo J and Chan CC: Enhanced apoptosis in retinal pigment epithelium under inflammatory stimuli and oxidative stress. Apoptosis. 17:1144–1155. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Sharma A, Sharma R, Chaudhary P, Vatsyayan R, Pearce V, Jeyabal PV, Zimniak P, Awasthi S and Awasthi YC: 4-Hydroxynonenal induces p53-mediated apoptosis in retinal pigment epithelial cells. Arch Biochem Biophys. 480:85–94. 2008. View Article : Google Scholar : PubMed/NCBI

28 

Gautam J, Banskota S, Shah S, Jee JG, Kwon E, Wang Y, Kim DY, Chang HW and Kim JA: 4-Hydroxynonenal-induced GPR109A (HCA2 receptor) activation elicits bipolar responses, Gαi-mediated anti-inflammatory effects and Gβγ-mediated cell death. Br J Pharmacol. 175:2581–2598. 2018. View Article : Google Scholar : PubMed/NCBI

29 

Li G, Shi Y, Huang H, Zhang Y, Wu K, Luo J, Sun Y, Lu J, Benovic JL and Zhou N: Internalization of the human nicotinic acid receptor GPR109A is regulated by G(i), GRK2, and arrestin3. J Biol Chem. 285:22605–22618. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Wang X, Ma W, Han S, Meng Z, Zhao L, Yin Y, Wang Y and Li J: TGF-β participates choroid neovascularization through Smad2/3-VEGF/TNF-α signaling in mice with Laser-induced wet age-related macular degeneration. Sci Rep. 7:96722017. View Article : Google Scholar : PubMed/NCBI

31 

Ushio-Fukai M and Nakamura Y: Reactive oxygen species and angiogenesis: NADPH oxidase as target for cancer therapy. Cancer Lett. 266:37–52. 2008. View Article : Google Scholar : PubMed/NCBI

32 

Wei D, Le X, Zheng L, Wang L, Frey JA, Gao AC, Peng Z, Huang S, Xiong HQ, Abbruzzese JL, et al: Stat3 activation regulates the expression of vascular endothelial growth factor and human pancreatic cancer angiogenesis and metastasis. Oncogene. 22:319–329. 2003. View Article : Google Scholar : PubMed/NCBI

33 

Ethen CM, Reilly C, Feng X, Olsen TW and Ferrington DA: Age-related macular degeneration and retinal protein modification by 4-hydroxy-2-nonenal. Invest Ophthalmol Vis Sci. 48:3469–3479. 2007. View Article : Google Scholar : PubMed/NCBI

34 

Raza H and John A: 4-hydroxynonenal induces mitochondrial oxidative stress, apoptosis and expression of glutathione S -transferase A4-4 and cytochrome P450 2E1 in PC12 cells. Toxicol Appl Pharmacol. 216:309–318. 2006. View Article : Google Scholar : PubMed/NCBI

35 

Oyewole AO and Birch-Machin MA: Mitochondria-targeted antioxidants. FASEB J. 29:4766–4771. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Cia D, Cubizolle A, Crauste C, Jacquemot N, Guillou L, Vigor C, Angebault C, Hamel CP, Vercauteren J and Brabet P: Phloroglucinol protects retinal pigment epithelium and photoreceptor against all-trans-retinal-induced toxicity and inhibits A2E formation. J Cell Mol Med. 20:1651–1663. 2016. View Article : Google Scholar : PubMed/NCBI

37 

Calabrèse A, Bernard JB, Hoffart L, Faure G, Barouch F, Conrath J and Castet E: Wet versus dry age-related macular degeneration in patients with central field loss: Different effects on maximum reading speed. Invest Ophthalmol Vis Sci. 52:2417–2424. 2011. View Article : Google Scholar : PubMed/NCBI

38 

Li X, Cai Y, Wang YS, Shi YY, Hou W, Xu CS, Wang HY, Ye Z, Yao LB and Zhang J: Hyperglycaemia exacerbates choroidal neovascularisation in mice via the oxidative stress-induced activation of STAT3 signalling in RPE cells. PLoS One. 7:e476002012. View Article : Google Scholar : PubMed/NCBI

39 

Dural E, Shah UK, Pritchard D, Chapman KE, Doak SH and Jenkins GJ: The effect of chronic dosing and p53 status on the genotoxicity of pro-oxidant chemicals in vitro. Mutagenesis geaa024. 2020. View Article : Google Scholar

40 

Kurokawa Y, Maekawa A, Takahashi M and Hayashi Y: Toxicity and carcinogenicity of potassium bromate--a new renal carcinogen. Environ Health Perspect. 87:309–335. 1990. View Article : Google Scholar : PubMed/NCBI

41 

Parsons JL and Chipman JK: The role of glutathione in DNA damage by potassium bromate in vitro. Mutagenesis. 15:311–316. 2000. View Article : Google Scholar : PubMed/NCBI

42 

Giri U, Iqbal M and Athar M: Potassium bromate (KBrO3) induces renal proliferative response and damage by elaborating oxidative stress. Cancer Lett. 135:181–188. 1999. View Article : Google Scholar : PubMed/NCBI

43 

Khan RA, Khan MR and Sahreen S: Protective effects of rutin against potassium bromate induced nephrotoxicity in rats. BMC Complement Altern Med. 12:2042012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2021
Volume 23 Issue 3

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
Kuo S, Li Y, Cheng K, Hsu C, Cheng J and Lau H: Potassium bromate-induced cell model of age-related macular degeneration <em>in vitro</em>. Mol Med Rep 23: 216, 2021
APA
Kuo, S., Li, Y., Cheng, K., Hsu, C., Cheng, J., & Lau, H. (2021). Potassium bromate-induced cell model of age-related macular degeneration <em>in vitro</em>. Molecular Medicine Reports, 23, 216. https://doi.org/10.3892/mmr.2021.11855
MLA
Kuo, S., Li, Y., Cheng, K., Hsu, C., Cheng, J., Lau, H."Potassium bromate-induced cell model of age-related macular degeneration <em>in vitro</em>". Molecular Medicine Reports 23.3 (2021): 216.
Chicago
Kuo, S., Li, Y., Cheng, K., Hsu, C., Cheng, J., Lau, H."Potassium bromate-induced cell model of age-related macular degeneration <em>in vitro</em>". Molecular Medicine Reports 23, no. 3 (2021): 216. https://doi.org/10.3892/mmr.2021.11855