Abstract

Arsenic trioxide (As2O3) is a promising effective chemotherapeutic agent for cancer treatment; however, how and through what molecular mechanisms the oxidative damage of As2O3 is controlled remains poorly understood. Recently, the involvement of dysregulated long noncoding RNA ovarian tumor domain containing 6B antisense RNA1 (lncRNA OTUD6B-AS1) in tumorigenesis is established. Here, for the first time, we characterize the regulation of As2O3 in the oxidative damage against bladder cancer via lncRNA OTUD6B-AS1. As2O3 could activate lncRNA OTUD6B-AS1 transcription in bladder cancer cells, and these findings were validated in a xenograft tumor model. Functional assays showed that lncRNA OTUD6B-AS1 dramatically exacerbated As2O3-mediated oxidative damage by inducing oxidative stress. Mechanistically, As2O3 increased levels of metal-regulatory transcription factor 1 (MTF1), which regulates lncRNA OTUD6B-AS1, in response to oxidative stress. Further, lncRNA OTUD6B-AS1 inhibited mitochondrial NADP+-dependent isocitrate dehydrogenase 2 (IDH2) expression by stabilizing miR-6734-5p, which contributed to cytotoxicity by enhancing oxidative stress. Together, our findings offer new insights into the mechanism of As2O3-induced oxidative damage and identify important factors in the pathway, As2O3/lncRNA OTUD6B-AS1/miR-6734-5p/IDH2, expanding the knowledge of activity of As2O3 as cancer treatment.

1. Introduction

Bladder cancer is associated with high morbidity and mortality rates and is the ninth most common cancer worldwide [1, 2]. The International Agency for Research on Cancer website reported that more than 120 000 people are diagnosed with bladder cancer annually in the European Union, with upwards of 40 000 people dying from the disease each year [3]. Environmental exposure to carcinogens, particularly toxic heavy metals, is a major risk factor for bladder cancer [4, 5]. Bladder cancer develops as either nonpapillary muscle-invasive tumors or non-muscle-invasive papillary tumors. Complete resection is the mainstay of treatment for non-muscle-invasive bladder cancer, while multimodal treatment, involving neoadjuvant chemotherapy and radical cystectomy, offers the best chance for cure of muscle-invasive bladder cancer [68]; however, several factors have resulted in limited uptake of clinical treatment. Until recently, there were no therapeutic options for metastatic bladder cancer beyond cisplatin-based therapy available in the clinic [9]. Hence, there has been considerable research interest in searching for new treatment strategies.

Arsenic trioxide (As2O3), a traditional remedy in China, is proven to induce complete remission in acute promyelocytic leukemia [10]. Further, evidence reported by Mathews et al. [11] showed that, for newly diagnosed cases of acute promyelocytic leukemia, 86.1% treated with single-agent As2O3 achieved complete hematologic remission. The antitumor properties of As2O3 for solid tumors have been under intense investigation; however, whether the success of this treatment for blood tumors can be repeated in solid tumors remains to be determined. Liu et al. [12] previously showed that intravenous administration and transarterial chemoembolization of As2O3 were safe and effective for treatment of unresectable hepatocellular carcinoma with lung metastasis, whereas Bajorin et al. [13] reported that As2O3 did not have any obvious activity against previously treated urothelial cancer and was associated with significant toxicity in the patient population tested. Several attempts have since been made to increase the antitumor effects of As2O3 toward solid cancers [14]. Further, significant insight has been obtained into the molecular mechanisms underlying the function of As2O3, including induction of oxidative stress, cancer stem cell inhibition, and regulation of noncoding RNAs [1517]. Gu et al. [18] reported that endoplasmic reticulum stress and mitochondrial dysfunction, mediated by reactive oxygen species (ROS), were involved in apoptosis induction by As2O3. Nonetheless, knowledge gaps remain in understanding of the molecular mechanisms involved in As2O3 cytotoxicity to solid tumors. Hence, it is of great interest to investigate the molecular mechanism underlying As2O3 induction of ROS production.

There is ample evidence supporting a direct role for long noncoding RNAs (lncRNAs) in modulation of cancer cell proliferation, apoptosis, and metastasis [19, 20]. For example, lncRNA SNHG is upregulated in bladder cancer tissue and involved in tumor proliferation [21]. Further, lncRNAs are involved in the oxidative stress system that regulates cancer progression [22, 23]. In addition, lncRNA is regulated in response to oxidative stress. Wang et al. [24] reported that the lncRNAs, H19 and HULC, are activated by oxidative stress and promote cholangiocarcinoma metastasis through regulation of miRNA. Interestingly, dysregulation of lncRNAs can be corrected by toxic heavy metals, plant extracts, and chemotherapeutic drugs [2528].

Here, we aimed to evaluate the endogenous cellular mechanisms involved in As2O3-mediated oxidative damage through regulation of lncRNA ovarian tumor domain containing 6B antisense RNA1 (lncRNA OTUD6B-AS1). Collectively, our results reveal that As2O3 provokes oxidative stress by upregulating lncRNA OTUD6B-AS1, which downregulates mitochondrial NADP+-dependent isocitrate dehydrogenase 2 (IDH2) by stabilizing its negative regulator, miR-6734-5p.

2. Materials and Methods

2.1. Cell Culture

T24 cells (a human bladder cancer cell line) were purchased from Shanghai Academy of Life Sciences (Shanghai, China) and cultured in RPMI-1640 (Thermo Fisher Scientific, Inc., Waltham, MA, USA), supplemented with sodium pyruvate (0.11 g/L), D-glucose (2.5 g/L), NaHCO3 (1.5 g/L), and 10% fetal bovine serum (FBS; Thermo Fisher Scientific). To assess the effects of As2O3 on T24 cells, they were maintained in an incubator with 5% CO2 and humidity of 70%–80% at 37°C. T24 cells were seeded into 96-well plates ( cells/well), then cultured for 12 h, followed by incubation with As2O3 (Sigma-Aldrich, Merck KGaA, Darmstadt, Germany) at 0, 10, and 20 μmol/L for 6 h, then collected for use in subsequent experiments.

2.2. RNA Extraction and Quantitative Real-Time PCR (qRT-PCR)

Total RNA was extracted from T24 cells using a Total RNA Extraction Kit (Thermo Fisher Scientific), according to the manufacturer’s instructions. A Maxima First Strand cDNA Synthesis Kit (Thermo Fisher Scientific) was used to synthesize complementary DNA (cDNA) and a One-Step SYBR Real-Time RT-PCR Kit (Bibaoman Biotechnology Co., Ltd., Shanghai, China) for qRT-PCR. Reaction conditions were as follows: 95°C for 600 s, then 45 cycles of 95°C for 10 s, 60°C for 20 s, and 72°C for 30 s. cDNA levels were calculated using the 2ΔΔCt method [29]. GAPDH served as an internal reference gene for evaluation of mRNA levels, and U6 was used for miRNAs. Each reaction was performed in triplicate. Primer sequences included the following: miR-6734-5p, forward 5-GGT CAC AGT GAA CCG GTC-3 and reverse 5-GTG CAG GGT CCG AGG T-3; U6, forward 5-CTC GCT TCG GCA GCA CA-3 and reverse 5-AAC GCT TCA CGA ATT TGC GT-3; lncRNA OTUD6B-AS1, forward 5-AGC ACA CCC AGT CAG AAA CCA G-3 and reverse 5-TCT ACA AAC GGG AAT GTC G-3; and GAPDH, forward 5-GGT CAC AGT GAA CCG GTC-3 and reverse 5-GTG CAG GGTC CGA GGT-3.

2.3. Cell Transfection

Lentiviral lncRNA OTUD6B-AS1 plasmids (lncRNA) were constructed by Hanheng Biotechnology Co., Ltd. (Shanghai, China). For lncRNA OTUD6B-AS1 overexpression, T24 cells were transfected with lentiviral lncRNA OTUD6B-AS1 plasmids or an empty vector. Then, transfected cells were harvested for further analysis. For ectopic expression of miR-6734-5p, T24 cells were transfected with negative control miRNA 5-CAG UAC UUU UGU GUA GUA CAA A-3 (NC miRNA) or miRNA mimics 5-AAA GAG ACC GGU UCAC UGU GA-3 (Sangon Biotech (Shanghai) Co., Ltd., Shanghai, China) using Lipofectamine 2000 (Thermo Fisher Scientific), according to the manufacturer’s instructions. Then, T24 cells were incubated for an additional 48 h, then harvested for further analysis. Lentiviral plasmids expressing IDH2-specific shRNA were constructed by Hanheng Biotechnology. For IDH2 knockdown, T24 cells were transfected with lentiviral IDH2 plasmids or an empty vector; then, transfected cells were harvested for further analysis.

2.4. Cell Counting Kit-8 (CCK-8) and Flow Cytometry Assays

T24 cells were seeded into 96-well plates ( cells/well) and cultured for 12 h. Next, T24 cells were incubated with As2O3 (Sigma-Aldrich, Merck KGaA, Darmstadt, Germany) at 0, 10, and 20 μmol/L for 6 h. After washing, T24 cells were incubated with 10% CCK-8 (Dojindo Molecular Technologies, Inc., Minato-ku, Tokyo, Japan) and optical density measured using a xMark Microporous Plate Absorption Spectrophotometer (Bio-Rad Laboratories, Inc., Hercules, CA, USA).

The apoptotic rate of T24 cells was detected using an Annexin V, 633 Apoptosis Detection Kit (Dojindo Molecular Technologies), following the kit instructions. T24 cells were seeded into 6-well plates ( cells/well) and cultured for 12 h. Next, cells were incubated with As2O3 (Sigma-Aldrich) at 20 μmol/L for 6 h, then incubated with Annexin V, followed by propidium iodide (PI) buffer for 15 min at 25°C in a dark room. Subsequently, apoptotic cells were quantified using a NovoCyte 1040 flow cytometer (ACEA Biosciences, Inc., Zhejiang, China).

2.5. TUNEL Staining

TUNEL assays for cells were conducted using a One Step TUNEL Apoptosis Assay Kit (Beyotime Institute of Biotechnology, Haimen, China) and for tumor tissues with a Colorimetric TUNEL Apoptosis Assay Kit (Beyotime), according to the kit instructions. Harvested tumor tissues were cut into 2.5 mm blocks and fixed in paraformaldehyde overnight. Then, blocks were embedded in paraffin and cut into 4 μm coronal sections. Next, apoptotic images were acquired using the Xmatrx Infinity Automatic Section Dyeing System (BioGenex Life Sciences Pvt. Ltd., San Ramon, California). For quantification of apoptosis, six sections were selected from each sample and five fields examined from each section.

2.6. In Vivo Tumorigenesis in Nude Mice

BALB/c nude mice (4 weeks old; 50% female and 50% male) were purchased from the Laboratory Animal Center of China Medical University (Production License: SCXK (Liaoning) 2018-0004). Mice were allowed free access to food and water and maintained at a controlled temperature range (). All animal studies were performed based on the principles and procedures outlined in the National Institutes of Health Guide for the Care and Use of Animals under assurance number A3873-1. T24 cell suspensions (100 μL; cells) were subcutaneously injected into the right flanks of mice, and xenograft tumors were measured every 2 days and tumor volumes calculated using the equation: . BALB/c nude mice were sacrificed at 14 days of measurement, and tumors were excised and weighed.

2.7. Detection of Oxidative Stress

Total ROS levels in T24 cells were measured using a reactive oxygen species assay kit (Beijing Solarbio Science & Technology Co., Ltd., Beijing, China). T24 cells were collected and suspended in diluted 2,7-dichlorofluorescin diacetate ( cells/mL). After incubation for 20 min and washing with culture medium (serum-free), fluorescence intensity was detected at 525 nm emission and 490 nm excitation. Levels of malondialdehyde (MDA), superoxide dismutase (SOD), and glutathione peroxidase (GSH-Px) in T24 cell lysates were measured using standard assay kits, including the Lipid Peroxidation MDA Assay Kit (Beyotime), the Total Superoxide Dismutase Assay Kit (Beyotime), and the Total Glutathione Peroxidase Assay Kit (Beyotime), according to their respective instructions. Enzyme levels and activity were measured using an xMark Microporous Plate Absorption Spectrophotometer (Bio-Rad).

2.8. Chromatin Immunoprecipitation (ChIP) Assays

ChIP assays were performed using EpiQuik Chromatin Immunoprecipitation Kits (AmyJet Scientific Inc., Wuhan, Hubei, China), according to the kit instructions. An MTF1 antibody was obtained from Abcam (ab236401). ChIP primer sequences were synthesized by Sangon Biotech, and immunoprecipitated DNA was quantified by qRT-PCR using the 2ΔΔCt method.

2.9. Pathological Examination

Hematoxylin-eosin (H&E) staining was performed using a Hematoxylin and Eosin Staining Kit (Beyotime). Cancer tissues were fixed in formaldehyde (10%) for 24 h, followed by dehydration, permeabilization, wax dipping, paraffin embedding, and cutting into 3 μm sections. After staining with hematoxylin (300 sec), sections were stained with eosin solution for 30 sec. Next, sections were dehydrated and mounted using neutral gum. Images were captured by microscopy (Olympus CX23; Olympus Corporation, Tokyo, Japan); all specimens were assessed by three observers.

For immunohistochemical analyses, cancer tissue specimens were fixed in formaldehyde (10%) for 1 day, followed by dehydration, permeabilization, wax dipping, paraffin embedding, and cutting into small pieces. Then, tissues were digested with proteinase K (20 mM; 5 min), followed by incubation in methanol (30 min), then 3% skim milk in 0.3% Triton X-100. Subsequently, rat monoclonal Ki-67 (2 μg/mL; Abcam, ab15580) and mouse monoclonal p57 (0.5 μg/mL; Abcam, ab220207) antibodies were added, followed by incubation with goat anti-rabbit IgG-HRP (1 : 100; HuaBio) and goat anti-mouse IgG-HRP (1 : 100; HuaBio) and image capture by microscopy (Olympus). Immunohistochemical staining intensity was visually assessed at 200x magnification by three observers. Intensity ratings were as follows: strongly positive (+++), moderately positive (++), weakly positive (+), and negative (-) expression.

2.10. lncRNA Target Prediction and Verification

Target genes were predicted using miRDB (http://mirdb.org/), TargetScan (http://www.targetscan.org/vert_72/), and EVmiRNA (http://bioinfo.life.hust.edu.cn/EVmiRNA#!/). Transcription factors and lncRNA binding sites were predicted using JASPAR (http://jaspar.genereg.net/). For RNA-RNA pull-down assays, biotinylated wild-type miR-6734-5p (miR-Wt Bio), biotinylated mutant miR-6734-5p (miR-Mut Bio), biotinylated NC (NC Bio), and lncRNA OTUD6B-AS1 were synthesized by Sangon Biotech. After 12 h of incubation, miRNAs and lncRNA or miRNAs and mRNA were cotransfected in T24 cells into 6-well plates. Forty-eight hours after transfection, T24 cells were lysed and incubated with Pierce Nucleic Acid-Compatible Streptavidin Magnetic Beads (Thermo Fisher) to bind to biotinylated probes. Bound lncRNAs or mRNA were purified and analyzed by qRT-PCR. For dual-luciferase reporter assay, the 3-untranslated region (3UTR) of IDH2 (WT) was amplified by Sangon, and the sequences were inserted into a pMIR-reporter luciferase vector (Ambion, Rockville, MD, USA). A mutated 3UTR (MUT) was also synthesized and inserted into the luciferase vector. MUT or WT vectors were cotransfected into HEK-293 cells with NC miRNA or miRNA mimics and luciferase intensity measured 2 days later.

2.11. Western Blot Analysis

Cells were lysed using lysis buffer solution (Beyotime); then, proteins were quantified using the BCA method. Protein extracts (50 μg) were separated by 8% SDS-PAGE, then transferred onto polyvinylidene difluoride membranes. Next, proteins were probed with primary antibodies against mitochondrial NADP+-dependent isocitrate dehydrogenase 2 (IDH2, 1 μg/mL; Abcam, ab55271), Bax (1 : 500; Abcam, ab53154), Bcl-2 (1 μg/mL; Abcam, ab185002), Caspase3 (1 : 5000; Abcam, ab90437), MTF1 (1 : 1000; Abcam, ab236401), nuclear factor E2-related factor 2 (Nrf2, 1 : 1000; Abcam, ab89443), Lamin B (1 : 4000; Abcam, ab151735), and β-actin (1 : 500; Abcam, ab179467). After washing, membranes were incubated with secondary goat anti-mouse IgG (1 : 1000, EMD Millipore, Billerica, MA, USA) antibody. Band intensity was quantified using ImageJ 1.52 software (National Institutes of Health, Bethesda, MD, USA).

2.12. Statistical Analysis

Each experiment was repeated three times independently. Data are presented as the . Statistical analyses were conducted using SPSS 21.0 (IBM Corp., Armonk, NY, USA) and GraphPad software 7 (GraphPad Software, Inc., La Jolla, CA, USA). Student’s -test was used to analyze the significance of differences between groups, while differences among more than two groups were evaluated by one-way analysis of variance (ANOVA), followed by post hoc correction for multiple comparisons using the Tukey test. Two-sided values < 0.05 were considered to indicate a statistically significant difference.

3. Results

3.1. As2O3 Increases lncRNA OTUD6B-AS1 Expression

First, we investigated the effect of As2O3 on lncRNA OTUD6B-AS1 expression. The results of qRT-PCR analyses showed that lncRNA OTUD6B-AS1 levels in T24 bladder cancer cells were significantly and dose-dependently increased in response to As2O3; lncRNA OTUD6B-AS1 levels were increased by more than 8.65-fold by As2O3 treatment at 20 μmol/L for 6 h, relative to untreated T24 cells () (Figure 1(a)). Furthermore, significant increases in lncRNA OTUD6B-AS1 levels were also observed in xenograft tumor model nude mice treated with As2O3 at 1 mg/kg and 5 mg/kg (Figure 1(b)). These data suggest that As2O3 activates lncRNA OTUD6B-AS1 transcription in bladder cancer.

3.2. lncRNA OTUD6B-AS1 Overexpression Inhibits Bladder Cancer Progression

Subsequently, we overexpressed lncRNA OTUD6B-AS1 8.79-fold by transfecting T24 cells with a lentiviral vector (Figure 1(c)), to quantitatively evaluate the biological function of lncRNA OTUD6B-AS1 in As2O3-induced cytotoxicity in vitro. The results of the CCK-8 assay (Figure 1(d)) showed that both lncRNA OTUD6B-AS1 overexpression and As2O3 treatment (20 μmol/L) significantly decreased T24 cell viability to 40% (). As shown in Figures 1(e) and 1(f), lncRNA OTUD6B-AS1 overexpression markedly increased apoptosis of T24 cells (). Interestingly, the elevated T24 cell apoptosis rate induced by As2O3 was further significantly enhanced by lncRNA OTUD6B-AS1 overexpression (). Further, a similar pattern was observed in TUNEL staining assays (Figures 1(g) and 1(h)). Western blotting showed that, under lncRNA OTUD6B-AS1 overexpression and As2O3 treatment (20 μmol/L), the expression of Bax and Caspase3 was significantly increased, while levels of Bcl-2 were significantly decreased relative to lncRNA OTUD6B-AS1 overexpression or As2O3 treatment (; Figures 1(i)1(l)).

The effect of lncRNA OTUD6B-AS1 in enhancing As2O3-induced cytotoxicity was further explored in vivo. The tumor volumes and weights of xenografts treated with lncRNA OTUD6B-AS1 or As2O3 (5 mg/kg) were clearly reduced and much lower in those treated with both lncRNA OTUD6B-AS1 and As2O3 (all ; Figures 2(a)2(c)). In addition, levels of lncRNA OTUD6B-AS1 in xenograft tumors were confirmed to increase with lncRNA OTUD6B-AS1 overexpression, As2O3 treatment, or both (all ; Figure 2(d)). The effects of lncRNA OTUD6B-AS1 were further assessed by pathological examination of harvested tumors. The results of H&E staining indicated that, compared with xenograft tumors with lncRNA OTUD6B-AS1 overexpression or As2O3 treatment (5 mg/kg), xenograft tumors subjected to both treatments exhibited more severe edema and had increased numbers of vacuolated cells (Figure 2(e)). As shown in Figures 2(f) and 2(g), compared with untreated xenograft tumors, more obvious apoptosis was detected in xenograft tumors in which lncRNA OTUD6B-AS1 was overexpressed or those treated with As2O3 (). Similar to the in vitro results, elevated apoptosis rates induced by As2O3 were found to undergo a further significant increase in response to lncRNA OTUD6B-AS1 overexpression (). Next, levels of apoptosis-related proteins were determined by immunohistochemistry. Xenograft tumors with both lncRNA OTUD6B-AS1 overexpression and As2O3 treatment showed significantly weaker Ki-67 expression (Figure 2(h)) and significantly stronger p57 expression (Figure 2(i)), compared with xenograft tumors with lncRNA OTUD6B-AS1 overexpression or As2O3 treatment alone. Together, these results further indicate that lncRNA OTUD6B-AS1 exacerbates As2O3-induced cytotoxicity.

3.3. As2O3 Upregulates lncRNA OTUD6B-AS1 Expression via MTF1

To gain insights into the molecular mechanism underlying lncRNA OTUD6B-AS1 exacerbation of As2O3-induced cytotoxicity, levels of oxidative stress were evaluated in T24 cells. As illustrated in Figures 3(a) and 3(b), ROS and MDA production was clearly elevated following As2O3 treatment (). Furthermore, ROS and MDA production also increased significantly in response to lncRNA OTUD6B-AS1 overexpression (). Interestingly, following lncRNA OTUD6B-AS1 overexpression, levels of ROS and MDA in T24 cells treated with As2O3 were significantly enhanced (); however, the activities of the antioxidant enzymes SOD and GSH-Px showed the opposite trend, suggesting an impaired antioxidant defense system (Figures 3(c) and 3(d)). In addition, we explored the expression of Nrf2 and found that Nrf2 levels in cytoplasm were clearly elevated following lncRNA OTUD6B-AS1 overexpression or treatment with As2O3; however, Nrf2 in the nucleus showed the opposite trend (Figures 3(e)3(g)). Together, these results suggest that lncRNA OTUD6B-AS1 exacerbation of As2O3-induced cytotoxicity is achieved by enhancement of oxidative stress.

Next, we investigated the molecular process by which As2O3 upregulates lncRNA OTUD6B-AS1 expression. According to the JASPAR database, the lncRNA OTUD6B-AS1 promoter has a potential binding site for the transcription factor, MTF1 (score, 5.75) (Figure 3(h)). Consequently, we evaluated MTF1 levels in bladder cancer cells treated with different concentrations of As2O3 for 6 h. As2O3 induced a significant and dose-dependent increase of MTF1 expression (; Figures 3(f) and 3(g)). Pretreatment with the NADPH oxidase inhibitor, apocynin, at 20 μm for 1 h significantly attenuated the elevation in MTF1 levels (; Figures 3(i) and 3(j)), indicating that As2O3-induced oxidative stress contributes to MTF1 expression. To further verify whether MTF1 is responsible for As2O3 regulation of lncRNA OTUD6B-AS1 transcription, endogenous MTF1 was knocked down using shRNA (Figure 3(k)). Significantly reduced lncRNA OTUD6B-AS1 levels were observed in MTF1 knockdown cells (), and the elevated lncRNA OTUD6B-AS1 levels in As2O3-treated T24 cells were significantly attenuated by MTF1 knockdown () (Figure 3(l)). These data suggest that lncRNA OTUD6B-AS1 transcription is regulated by MTF1. To corroborate these findings, we conducted ChIP assays and found that the pull-down with the MTF1 antibody greatly enriched the lncRNA OTUD6B-AS1 promoter region containing putative metal response elements (nucleotides 754 to 854 in the promoter) by almost 5.20-fold relative to control IgG (), and a reference region (nucleotides 100 to 200 in the promoter) without putative MRE was used as a negative control (Figure 3(m). In addition, we assessed whether MTF1 could activate transcription from the lncRNA OTUD6B-AS1 promoter using a dual-luciferase reporter assay. The luciferase reporter plasmid promoter region was modified to contain WT or MUT MTF1 binding sites. As shown in Figure 3(n), the luciferase activity driven by the WT promoter sequence was markedly reduced by MTF1 knockdown, indicating that the WT sites are required for binding of MTF1 to the lncRNA OTUD6B-AS1 promoter. Collectively, these findings indicate that lncRNA OTUD6B-AS1 is directly regulated by MTF1 at the transcriptional level upon As2O3 treatment.

3.4. lncRNA OTUD6B-AS1 Downregulates IDH2 Expression by Stabilizing miR-6734-5p

To further interrogate the molecular mechanism by which lncRNA OTUD6B-AS1 exacerbates As2O3-induced oxidative damage, we investigated the downstream targets of lncRNA OTUD6B-AS1 by bioinformatic analysis. First, the subcellular localization of lncRNA OTUD6B-AS1 in T24 cells was determined using a nuclear mass separation assay. As shown in Figure 4(a), 86.53% of lncRNA OTUD6B-AS1 expression was detected in the cytoplasm fraction in T24 bladder cancer cells, suggesting that the oxidative stress induced by lncRNA OTUD6B-AS1 may be achieved through regulation of miRNA. Therefore, we predicted potential lncRNA binding sites for miRNAs using miRDB. As shown in Figure 4(b), miR-6734-5p was predicted to have three binding sites in lncRNA OTUD6B-AS1. Interestingly, miR-6734-5p was also found to bind to the 3UTR of IDH2 mRNA by miRDB, TargetScanHuman, and EVmiRNA (Figure 4(c)). Therefore, miR-6734-5p was selected as a potential downstream target of lncRNA and further analyses were conducted.

Subsequently, we investigated the regulation of miR-6734-5p by lncRNA OTUD6B-AS1. The results revealed that miR-6734-5p levels were significantly increased, by more than 5.18-fold, in T24 cells overexpressing lncRNA OTUD6B-AS1 relative to untreated control cells () (Figure 4(d)). Levels of miR-6734-5p increased significantly in T24 cells after treatment with miR-6734-5p mimics (Figure 4(e)). Nevertheless, we found that miR-6734-5p mimics did not significantly downregulate lncRNA OTUD6B-AS1 levels (; Figure 4(f)). To further elucidate whether lncRNA OTUD6B-AS1 could directly bind to miR-6734-5p, RNA pull-down assays were performed and showed that lncRNA OTUD6B-AS1 was more highly enriched in miR-6734-5p pull-down cell lysates, relative to control cells (Figures 4(g) and 4(h)). These results demonstrate that lncRNA OTUD6B-AS1 functions to increase the miR-6734-5p level by enhancing miR-6734-5p stability.

In addition, to validate the prediction that miR-6734-5p can regulate IDH2 expression, dual-luciferase reporter assays were performed and the results confirmed that, compared with the miR-6734-5p control, luciferase activity was markedly lower (approximately 80%) following transfection of IDH2 WT and miR-6734-5p mimics (), whereas the luciferase activity was not significantly altered after transfection of IDH2 MUT and miR-6734-5p mimics () (Figure 4(i)). The results of RNA pull-down assays showed that IDH2 mRNA was more highly enriched in miR-6734-5p pull-down cell lysates relative to control cells (Figures 4(j) and 4(k)). We then examined IDH2 levels, to further confirm the interaction between miR-6734-5p and the IDH2 3UTR, by Western blot analysis. As demonstrated in Figures 4(l) and 4(m), transfection of miR-6734-5p mimics resulted in significantly reduced IDH2 expression (). Further, lncRNA OTUD6B-AS1 significantly reduced IDH2 levels by 50% () (Figures 4(n) and 4(o)). Together, these results demonstrate that miR-6734-5p serves as an intermediate in the regulation of IDH2 by lncRNA OTUD6B-AS1.

3.5. The lncRNA OTUD6B-AS1/miR-6734-5p/IDH2 Pathway Is Correlated with Oxidative Damage

We next characterized the interactions between lncRNA OTUD6B-AS1 and miR-6734-5p in evoking oxidative damage. As shown in Figures 5(a)5(d), lncRNA OTUD6B-AS1 upregulated ROS and MDA and this activity could be significantly enhanced by miR-6734-5p. Upregulation of SOD and GSH-Px was reduced in T24 cells in response to miR-6734-5p mimics (). Furthermore, the elevation of Nrf2 in cytoplasm induced by lncRNA OTUD6B-AS1 was further significantly enhanced by miR-6734-5p; however, Nrf2 in the nucleus showed the opposite trend (Figures 5(e)5(g)). These data indicate that miR-6734-5p contributes to oxidative stress as a downstream target of lncRNA OTUD6B-AS1. Additionally, flow cytometry experiments revealed that miR-6734-5p mimics could significantly increase apoptosis relative to controls (). Further, upregulation of miR-6734-5p also significantly elevated the T24 cell apoptosis rate in response to lncRNA OTUD6B-AS1 overexpression, relative to lncRNA OTUD6B-AS1 overexpression alone () (Figures 5(h) and 5(i)). Importantly, a similar pattern was observed in TUNEL staining assays (Figures 5(j) and 5(k)). The results of CCK-8 assays also revealed significantly lower cell viability of bladder cells overexpressing lncRNA OTUD6B-AS1 and miR-6734-5p mimics () (Figure 5(l)). Together, these results offer new insights into the biological function of miR-6734-5p in lncRNA OTUD6B-AS1-induced oxidative damage.

Next, we sought to delineate the interactions between miR-6734-5p and IDH2 in evoking oxidative damage. In the context of oxidative stress, we found that the increases in ROS and MDA induced by miR-6734-5p mimics were further significantly enhanced by IDH2 knockdown, while the decreases in SOD and GSH-Px were significantly reduced by IDH2 knockdown (Figures 6(a)6(d)).

In addition, Nrf2 levels in cytoplasm were clearly increased by IDH2 knockdown; however, Nrf2 in the nucleus showed the opposite trend (Figures 6(e)6(g)). The results presented in Figures 6(h) and 6(i) indicate that the apoptosis rate was significantly elevated in response to IDH2 knockdown (). In addition, the elevated rate of T24 cell apoptosis on miR-6734-5p mimics was significantly enhanced by IDH2 knockdown (). The TUNEL staining assay revealed a similar pattern, where introduction of miR-6734-5p significantly enhanced apoptosis in cells with IDH2 knockdown (Figures 6(j) and 6(k)). Further, the results of CCK-8 assays also indicated that T24 cells with miR-6734-5p mimics treated with IDH2 knockdown exhibited significantly lower cell viability () (Figure 6(l)). Taken together, these data highlight the important roles of miR-6734-5p and IDH2 in lncRNA OTUD6B-AS1-induced oxidative damage.

4. Discussion

It is not established whether As2O3 exerts effective antitumor activity against bladder cancer [13, 30], while it is well known that the oxidative stress triggered by As2O3 is key to its antitumor effects [31, 32]. Nevertheless, knowledge gaps remain regarding the endogenous cellular machinery through which As2O3 exacerbates oxidative damage. This study was prompted by reports of the dysregulation of lncRNA expression in tumors and the regulation of lncRNAs by toxic heavy metals [33, 34]. lncRNA OTUD6B-AS1 can inhibit cancer cell proliferation [35]; however, whether lncRNA OTUD6B-AS1 is also involved in oxidative damage triggered by As2O3 in bladder cancer was previously unknown. Here, for the first time, we demonstrate that lncRNA OTUD6B-AS1 expression is upregulated by As2O3 and that lncRNA OTUD6B-AS1 in regulated in response to oxidative stress. Further, we show that lncRNA OTUD6B-AS1 induction by As2O3 is dependent on MTF1. Importantly, lncRNA OTUD6B-AS1 dramatically exacerbated As2O3-induced cytotoxicity by inducing oxidative stress. More interestingly, another major finding was that the molecular mechanism involved lncRNA OTUD6B-AS1 inhibition of IDH2 expression through stabilization of miR-6734-5p (Figure 7).

To date, the clinical success of As2O3 in treating hematological cancers has not been translated to application in solid tumors of the urinary system [13, 36]. Researchers have made efforts to improve the antitumor activity of As2O3 toward solid tumors. The novel lncRNA, OTUD6B-AS1, is an important target of gene expression [35]. Data from The Cancer Genome Atlas database reveal that lncRNA OTUD6B-AS1 levels are reduced and that lncRNA OTUD6B-AS1 acts as an antioncogene in various tumors [35]. Here, we found that lncRNA OTUD6B-AS1 transcription was activated by As2O3 in bladder cancer cells, and these results were validated using a xenograft tumor model, suggesting that the antitumor effects of As2O3 may be via upregulation of lncRNA OTUD6B-AS1; however, the roles of lncRNA OTUD6B-AS1 in exacerbated As2O3-induced oxidative damage have yet to be elucidated. Here, we investigated the function of lncRNA OTUD6B-AS1 and found that its overexpression clearly suppressed proliferation and elevated apoptosis of bladder cancer cells. Additionally, overexpression of lncRNA OTUD6B-AS1 significantly reduced tumor size and tumor weight in a xenograft model. These findings indicate that increased lncRNA OTUD6B-AS1 may be able to inhibit bladder cancer development, which is similar to the findings of Wang et al. [35] that lncRNA OTUD6B-AS1 can suppress progression of clear cell renal cell carcinoma. Further, functional assays showed that overexpression of lncRNA OTUD6B-AS1 could exacerbate As2O3-induced cell damage both in vitro and in vivo. Cumulatively, we believe that As2O3-induced cell damage is achieved by upregulation of lncRNA OTUD6B-AS1. Numerous studies have shown that ROS underlies the induction of cancer cell apoptosis by As2O3, and following lncRNA OTUD6B-AS1 overexpression, the elevation of ROS and MDA in T24 cells induced by As2O3 was further significantly enhanced. Interestingly, activities of the antioxidant enzymes, SOD and GSH-Px, were also significantly reduced. Furthermore, the elevation of Nrf2 in cytoplasm induced by As2O3 was further significantly enhanced by lncRNA OTUD6B-AS1; however, Nrf2 in the nucleus showed the opposite trend. This discovery was consistent with Li et al.’s report [37] and Zhang et al.’s study [38]. A marked increase in the generation of ROS exceeds the physiological capacity of SOD and GSH-Px, and the exhaustion of these enzymes reduces their activity, ultimately leading to oxidative damage. Furthermore, several previous studies have demonstrated that As2O3 inhibits Nrf2 transposition through inhibition of the PI3K/Akt pathway, ultimately leading to the reduction of SOD and GSH-Px generation [39, 40]. These results further indicate that exacerbation of oxidative stress is key for enhancement of As2O3-induced cytotoxicity by lncRNA OTUD6B-AS1.

Accumulated evidence supports the regulation of lncRNA transcription by multiple factors, including toxic heavy metals, plant extracts, and chemotherapeutic drugs [41]. MTF1, a transcription factor, drives mRNA expression in response to toxic heavy metals [42]. van Loo et al. previously demonstrated that zinc regulates a key transcriptional pathway involved in epileptogenesis via MTF1 [43]. Accordingly, we sought out to determine the mechanism through which MTF1 regulates lncRNA OTUD6B-AS1 expression. First, we explored MTF1 expression in response to As2O3 and oxidative stress. Interestingly, treatment with As2O3 clearly increased MTF1 expression. In addition, apocynin, a NADPH oxidase inhibitor, eliminated this increase of MTF1 expression, consistent with the reports of Tavera-Montanez et al. that MTF1 levels were increased following exposure to toxic heavy metals [44]. Furthermore, we found that MTF1 knockdown eliminated the As2O3-induced increase of lncRNA OTUD6B-AS1 expression. Collectively, these results offer new insights into the mechanisms involved in As2O3 anticancer activity and demonstrate that lncRNA OTUD6B-AS1 expression can be activated by As2O3 through ROS-mediated upregulation of MTF1. Furthermore, binding of MTF1 to the lncRNA OTUD6B-AS1 promoter sequence was directly proven by both ChIP and dual-luciferase reporter assays. Previous reports demonstrated that MTF-1 can effectively protect cells from oxidative stresses by binding toxic metal ions to activate the expression of metallothioneins [4547]. We speculate that higher doses of As2O3 may exceed the tolerance of metallothioneins and that lncRNA OTUD6B-AS1 induces MTF1 in response to uncleared As2O3, promoting ROS production and oxidative damage.

An antioxidant system that relies on NADPH is needed for maintenance of proper redox status [48]. Mitochondrial NADP+-dependent IDH2 is confirmed as an essential enzyme for mitochondria to maintain their antioxidant system by generating NADPH [49]. Recent studies have demonstrated that IDH2 deficiency exacerbates acetaminophen hepatotoxicity in mice via increasing susceptibility to ROS generation and oxidative stress [50]. Therefore, we speculated that lncRNA OTUD6B-AS1 might enhance oxidative stress by downregulating IDH2. To date, many lncRNAs have been characterized and shown to regulate gene expression by regulating miRNA [51, 52]. To our knowledge, no mechanism underlying the regulation of miRNA by lncRNA OTUD6B-AS1 has previously been reported. We found that lncRNA OTUD6B-AS1 expression was significantly higher in the cytoplasm fraction of T24 bladder cancer cells, supporting a role for lncRNA OTUD6B-AS1 function in regulation of miRNA. Intriguingly, we established that miR-6734-5p acts as a bridge molecule between lncRNA OTUD6B-AS1 and IDH2. Consequently, levels of miR-6734-5p were elevated in response to increased lncRNA OTUD6B-AS1 levels. Further, RNA-RNA pull-down assays revealed tight binding between lncRNA OTUD6B-AS1 and miR-6734-5p, indicating that lncRNA OTUD6B-AS1 may stabilize miR-6734-5p, preventing its degradation. Previously, miR-6734-5p was reported to target a complementary p21 promoter sequence to inhibit cancer development [53]; however, to our knowledge, the role of miR-6734-5p in regulation of oxidative stress-related proteins remains largely unknown. Dual-luciferase reporter and RNA-RNA pull-down assays indicated that miR-6734-5p could bind to the IDH2 mRNA 3UTR. Additionally, gain of miR-6734-5p inhibited IDH2 expression. These data indicate that lncRNA OTUD6B-AS1 can suppress IDH2 expression through stabilization of miR-6734-5p.

Previously, lncRNA OTUD6B-AS1 was reported to have an antioncogenic role in clear cell renal cell carcinoma, associated with the Wnt/β-catenin signaling pathway [35]. Furthermore, miR-6734-5p can also inhibit development of colon cancer and acute myeloid leukemia [53, 54]; however, the oxidative damage effects of lncRNA OTUD6B-AS1 and miR-6734-5p against bladder cancer have not previously been reported. Therefore, we sought to determine the interactions between lncRNA OTUD6B-AS1 and miR-6734-5p in evoking oxidative damage against bladder cancer. Intriguingly, gain-of-function experiments demonstrated the effects of the combination of lncRNA OTUD6B-AS1 and miR-6734-5p in promoting oxidative stress, and knockdown of IDH2 also had a similar effect. These results represent new data indicating how lncRNA OTUD6B-AS1 exacerbates As2O3-induced oxidative damage via miR-6734-5p-mediated functional inhibition of IDH2. Cytotoxicity analyses also indicated that both lncRNA OTUD6B-AS1 and miR-6734-5p, or both miR-6734-5p and knockdown of IDH2, clearly increased apoptosis rates relative to lncRNA OTUD6B-AS1, miR-6734-5p, or IDH2 knockdown alone. In contrast, cell viability was evidently decreased. Overall, our results prove, for the first time, that the function of lncRNA OTUD6B-AS1 in enhancing As2O3-mediated oxidative damage is achieved by downregulation of IDH2 through stabilization of its negative regulator, miR-6734-5p.

Some limitations in this study will need to be addressed in future investigations. The regulation of lncRNA OTUD6B-AS1 expression by As2O3 requires further exploration in a clinical context, to corroborate our findings. Additionally, further studies are needed to determine whether other As2O3-induced cytotoxic effects, including inhibition of cancer stem-like cells, are also mediated by lncRNA OTUD6B-AS1.

5. Conclusions

In conclusion, the current study has elucidated a novel molecular mechanism of As2O3 in evoking oxidative stress in bladder cancer, where MTF1 was increased by As2O3, thereby activating transcription of lncRNA OTUD6B-AS1. Subsequently, lncRNA OTUD6B-AS1 inhibited IDH2 expression by stabilizing miR-6734-5p, exacerbating As2O3-induced oxidative stress. These data reveal a new mechanism underlying lncRNA OTUD6B-AS1-mediated signaling that promotes As2O3 cytotoxicity against bladder cancer, suggesting a potential new strategy to facilitate the development of As2O3 for use in treatment of bladder cancer.

Data Availability

All data generated or analyzed during this study are included in this published article. The data used to support the findings of this study are available from the corresponding author upon request.

Additional Points

Highlights. As2O3 activates transcription of lncRNA OTUD6B-AS1 by upregulating MTF1expression. lncRNA OTUD6B-AS1 exacerbates As2O3-induced oxidative damage by evoking oxidative stress. lncRNA OTUD6B-AS1 inhibits IDH2 expression by stabilizing miR-6734-5p.

Conflicts of Interest

The authors declared that they do not have anything to disclose regarding conflict of interest with respect to this manuscript.

Authors’ Contributions

The contributions of the authors involved in this study are as follows: conceptualization, Y.W. and S.X.; methodology, Y.W. and J.L.; software, T.Y. and Y.H.; investigation, Z.R. and J.Z.; formal analysis, T.Y. and Y.H.; resources, Z.R. and S.X.; writing—original draft preparation, Y.W.; writing—review and editing, S.X.; visualization, Y.H. and J.L.; supervision, S.X.; project administration, S.X.; and funding acquisition, S.X.

Acknowledgments

The work was supported by grants from the National Nature Science Foundation of China (Grant No. 81673207).