1. Introduction
Arsenic is a well-known toxicant found ubiquitously in the environment. It has been used since ancient times both as a curative and poison. Typical uses of arsenic are in agricultural pesticides, pigmentation, metallurgy, medications, and pyrotechnics. Various epidemiological, case–control and experimental studies have linked arsenic exposure to the development of lung, skin, bladder, kidney, and liver cancers in humans. Hence, to protect human health, the International Agency for Research on Cancer (IARC) classified arsenic and its compounds into group IA carcinogens. However, despite the prohibition on the use of arsenic in commodities, approximately 200 million people worldwide are still being exposed to arsenic via drinking water, food, and air [
1,
2]. Drinking water contamination is the primary source of arsenic exposure worldwide. The current US Environmental Protection Agency (US EPA) and World Health Organization (WHO) arsenic standard in drinking water is 10 ppb (<0.1 μM, 10 μg/L). However, there are areas in the US, India, Bangladesh, China, Chile, Mongolia, Mexico, and Argentina where more than 50 ppb of arsenic is found in drinking water [
1,
2,
3]. Arsenic is known to induce cancer via various mechanisms, such as oxidative stress caused by the generation of reactive oxygen species (ROS), epigenetic alterations by regulating microRNAs, DNA and histone tail modifications, and several intracellular stress signaling pathways leading to the activation of JNK, STAT3, NF-κB, DNA damage response, and immune surveillance systems. Thus, interpreting the carcinogenic mechanisms of arsenic is the key to develop anti-cancer therapeutics.
Cancer-related mortality rates, especially of the lung, bronchus, and trachea cancers, hold the sixth position in the world’s top 10 list of leading causes of death, according to the World Health Organization (WHO) analysis of 2019. Lung cancer is the most common cancer worldwide and accounts for a significant portion of cancer-related deaths. Lung cancer can be categorized as non-small and small cell lung cancer. Non-small cell lung cancer (NSCLC) forms about 80% of the total lung cancer cases, which can be further categorized into adenocarcinoma (40%), squamous cell carcinoma (25%) and large cell carcinoma (10%). Despite advances in cancer therapy, relapse is still a major cause of cancer mortality [
4,
5]. This relapse has been partially attributed to a subpopulation of cells called the tumor-initiating cells or cancer stem-like cells (CSCs). The CSCs can develop from differentiated cells or bulk cancer cells. Like the normal stem cells, CSCs can self-renew and differentiate into new cell types [
6]. CSCs contribute to sustained tumor growth, heterogeneity, metastasis, and chemotherapy resistance [
7].
Previous studies from our lab have demonstrated that long-term treatment with an environmentally relevant concentration of inorganic arsenic (0.125–0.25 μM As
3+) leads to the malignant transformation of the human bronchial epithelial BEAS-2B cells. The transformed cells showed malignant characteristics such as asymmetric division and formation of tumorspheres in tumor formation media. These cells also showed higher expression of stemness factors such as Oct4, Sox2, Nanog, c-myc, etc., compared to the non-transformed cells. Henceforth, we refer to these cells as the arsenic-induced CSCs or CSCs. By subcutaneous inoculation of 10,000 control cells and the As
3+-induced CSCs, respectively, into athymic nude mice, only CSCs, but not the control cells, could form fast-growing tumors [
8,
9].
Meanwhile, a metabolic change was observed in the As
3+-induced CSCs compared to non-CSCs cells. Transcriptomics data revealed an upregulation of the glycolytic genes and metabolites. In contrast, a significant downregulation of mitochondrial oxidative phosphorylation (OXPHOS) genes was observed [
7]. Interestingly, a reduced expression of ER stress and autophagy genes in the As
3+-induced CSCs was also observed, which indicated a possible connection between ER stress signaling and energy metabolism during the generation of CSCs induced by As
3+.
ER stress-activated unfolded protein response (UPR) and autophagy are essential in maintaining mitochondrial dynamics, such as mitochondrial fusion, fission, and metabolism, and recycling misfolded proteins and organelles to provide alternative fuel during stress/nutrient deprivation conditions. There are three master regulatory arms of the UPR signaling: protein kinase RNA-like ER kinase (PERK), inositol-requiring enzyme 1 (IRE1) and activating transcription factor 6 (ATF6). The ATF6 has two genes at different genomic locations. The gene encodes ATF6A, also named ATF6, is at the gene locus of chromosome 1 (q23.3), whereas the gene of ATF6B is at a locus of chromosome 6 (p21.32). Under certain stress conditions, an intimate connection between the ER, mitochondrion and autophagy has been well-established. Impairment in either one of them directly affects the functioning of the other. Thus, it becomes essential to investigate whether As
3+ affects the interaction among ER, mitochondrion, and autophagy during the process of As
3+-induced generation of the CSCs [
10,
11,
12]. Meanwhile, induction of the CSCs by As
3+ is made by a relatively long-term treatment scheme, in which cells undergo positive and negative selection, adaptation and repopulation. Thus, recognizing the initiation cellular response, or short-term treatment of the cells with As
3+, will be critical to determine how ER stress signaling contributes to the As
3+-induced carcinogenesis. Answering these questions, accordingly, may also help us to understand if UPR, metabolism and autophagy play a role in the malignant transformation of the BEAS2B cells or maintenance of the stemness features of the As
3+-induced CSCs.
2. Materials and Methods
2.1. Cell Culture
The human bronchial epithelial cell line BEAS-2B was purchased from American Type Culture Collection (ATCC, Manassas, VA, USA). The BEAS-2B cells were cultured in Dulbecco’s Modified Eagle’s Medium-High Glucose (Sigma-Aldrich #D5796) with added 5% Fetal Bovine Serum, 1% L-Glutamine and 1% Penicillin/Streptomycin in a humidified incubator at 37° C and 5% CO2. The cells were starved with serum-free medium (overnight), followed by treatment of the cells with various concentrations of As3+ [arsenic (III) chloride, Sigma-Aldrich, St. Louis, MO, USA] for the indicated times. For long-term treatment, BEAS-2B cells were exposed to 0.125–0.25 μM As3+ for six months. These cells are referred to as transformed BEAS-2B cells.
2.2. Human Unfolded Protein Response Qiagen RT2 Profiler PCR Array
Total RNA was isolated from control BEAS-2B cells, or the cells were treated with As3+ using Qiagen RNeasy plus mini kit (Qiagen, Germantown, MD, USA). The RNA quality was determined by agarose gel electrophoresis, and concentration was calculated using the Thermofisher nanodrop spectrophotometer. RNA was reverse transcribed to cDNA using a High-Capacity RNA-to-cDNA kit (Applied Biosystems, Waltham, MA, USA) according to the manufacturer’s instructions. The cDNA and SYBR-Green qPCR Master mix (Qiagen Cat. no. 330529) were added to the Qiagen Human UPR array plate (Qiagen, Cat. no. PAHS-089Z) to detect changes in gene expression in the UPR with or without As3+ treatment.
2.3. Regular Reverse Transcription Polymerase Chain Reaction (PCR)
Total RNA was isolated from As3+-treated and control BEAS-2B cells prepared using Qiagen RNeasy plus mini kit according to the manufacturer’s protocol. Reverse transcription was performed to convert RNA to cDNA using High-Capacity RNA-to-cDNA kit (Applied Biosystems) according to the manufacturer’s instructions. ATF6 and UBE2G2 primers were designed using the NCBI primer blast. GAPDH was used as a loading control. RT-PCR primer sequences for ATF6α (exon 16) are as follows: forward primer, 5′-GAAGCTTATGGCAGAGATGCAC-3′; reverse primer, 5′-CAGTGCTTTCCAAATAGATGGGTA-3′. For UBE2G2 (exon 4), they are: forward primer, 5′-CATTTGTCAATTGTGGTCGACGTT-3′; reverse primer, 5′-AATACCACCATGCTTACTTGGCT-3′. Primer sequences for GAPDH are as follows: sense primer, 5′-CTGAACGGGAAGCTGGCATGGCCTTCC-3′; anti-sense primer, 5′-CATGAGGTCCACCTGTTGCTGTAGCC-3′. PCR products were run on 1% agarose gels with DNA ladders. Samples without cDNA or RT templates served as negative controls.
2.4. Western Blotting
BEAS-2B cells were seeded in a 6-well plate (3.0 × 105 cells per well) or 10 cm dish (1.0 × 106 cells per plate) and treated with various concentrations of As3+ for the indicated times. Cells were lysed by 1 × RIPA lysis buffer (Cell signaling) supplemented with protease and phosphatase inhibitors cocktail (Roche, Indianapolis, IN, USA) and 1 mM PMSF. Cell lysates were homogenized by sonication, and insoluble debris was removed through centrifugation of 12,500× g for 15 min at 4 °C. The protein concentrations were then determined using Pierce BCA Protein Assay Kit (Thermo Scientific, Rockford, IL, USA). The protein samples were prepared using 4 × LDS sample buffer (Invitrogen, Carlsbad, CA, USA) with dithiothreitol (final concentration of 200 mM) and were denatured by boiling at 95 °C for 5 min before separation by SDS-PAGE gel. Separated samples were then transferred onto PVDF membrane (Invitrogen) and blocked with 5% non-fat milk diluted in 1 × TBST for 1 h at room temperature followed by a quick TBST rinse. The membranes were then incubated with the indicated primary antibodies overnight at 4 °C. The membranes were further washed thrice, each wash of TBST for a period of 10 min. The membranes were further incubated at room temperature with the corresponding HRP-linked secondary antibodies for 1 h, followed by TBST wash thrice. ECL substrates (Thermo Scientific, Millipore and Westpico Plus, Waltham, MA, USA) were used to visualize the signals. Primary antibodies against p-CEBPβ Threonine 235 (3084S), SCAP (13102s), GAPDH and HRP-linked rat, mouse and rabbit IgG were purchased from Cell Signaling technology. Antibodies against ATF6 and ATF6B were purchased from Biolegend for Western blot. UBE2G2 (ab235790), HSPA1B/Hsp70 (ab231637), anti-htrA4 antibody-catalytic domain (ab65915), anti-ERN2 antibody (ab135795) and CEBPβ primary antibodies (ab32358) were purchased from Abcam.
2.5. Seahorse Bioanalyzer for Cell Energy Phenotype Test
The Seahorse XF24 Extracellular Flux Analyzer was used to obtain real-time measurements of extracellular acidification rates (ECAR) and oxygen consumption rate (OCR) in BEAS-2B cells. Cells (1.0 × 104) were plated in a 24-well Agilent Seahorse XF24 plate in DMEM media. The plate was coated with 0.1% gelatin prior to cell seeding to improve cell adhesion. The extracellular flux assay kit cartridge was hydrated with XF calibrant/well. The hydrated cartridge was incubated overnight in the CO2-free incubator prior to use. After the cells reached 70–80% confluency, the media was changed to serum-free DMEM (overnight). The cells were treated with the indicated doses of arsenic. The media was changed to Seahorse media with appropriate supplements and arsenic 45 min–1 h prior to ECAR and OCR measurements.
2.6. Immunohistochemistry
Lung adenocarcinoma tissue microarray slide LC10013c (lung cancer and matched adjacent normal lung tissue array) was purchased from US Biomax, Inc. (Rockville, MD, USA). It was processed for immunohistochemical staining using ATF6 and ATF6B antibodies (Novus biologicals, Littleton, CO, USA). Paraffin-embedded tissue sections were deparaffinized with xylene and hydrated in a series of alcohol gradients. To quench endogenous peroxidase activity, slides were incubated with 1.5 to 3% H2O2 in PBS for 20 min at room temperature. Heat-mediated antigen retrieval was performed by boiling tissue sections in citrate buffer with pH 6.0 for 20 min in a microwave. To block nonspecific binding of immunoglobulin, slides were incubated with a solution containing 5% goat serum, 0.2% Triton X-100 in PBS for 2 h at room temperature, followed by incubation with primary antibodies against ATF6 and ATF6B (1:50) overnight at 4 °C. Goat anti-mouse and goat anti-rabbit biotinylated secondary antibodies were subsequently applied at 1:100 dilution and incubated for 2 h at room temperature. Slides were then incubated with ABC reagent (Vectastatin Elite ABC kit, Vector Laboratories Inc., Burlingame, CA, USA) for 45 minutes at room temperature. The chromogen was developed with diaminobenzidine (DAB). Slides were counterstained with hematoxylin (Sigma-Aldrich, St. Louis, MO, USA) and mounted with Entellan® (Electron Microscopy Sciences, Hatfield, PA, USA). All incubation steps were carried out in a humidified chamber, and all washing steps were performed with 1 × PBS. Images were captured under a bright field of a Nikon Eclipse Ti-S Inverted microscope (Mager Scientific, Dexter, MI, USA).
2.7. Kaplan–Meier Survival Analysis
A Kaplan–Meier survival analysis was carried out using the Kaplan–Meier plotter (lung cancer) from mRNA database [
13], which contains overall survival (OS) of 1925 lung cancer patients, using the option of the best perform threshold as a cutoff. The probe IDs are indicated on the top of each panel in the figures. Survival curves resulting in
p values of <0.05 were considered statistically significantly. The gene expression of ATF6 and ATF6B in human lung cancer was determined by using TCGA platforms that contain genomic and transcriptiomic data of the lung cancer patient databases. The differential expression of ATF6 and ATF6B in normal lungs and lung tumors was calculated by TNMplot [
14].
2.8. Chromatin Immunoprecipitation with Global Parallel DNA Sequencing (ChIP-seq)
ChIP-seq for Nrf2 and HIF1a was performed as reported previously [
15]. Briefly, BEAS-2B cells were seeded in 10 cm dishes and treated with 1 mM As
3+ or without treatment for 6 h. At the end of culture, approximately 10 million control cells and the As
3+-treated cells were fixed using formaldehyde solution. The fixation was then stopped by adding glycine solution. The cell pellet was washed twice with 1× PBS-Igepal and snap-frozen in dry ice. The cells were then subjected to immunoprecipitation using ChIP-grade antibodies against Nrf2 and HIF1α from Active Motif (Carlsbad, CA, USA). The procedures of ChIP, preparation of input and control DNA, DNA sequencing, and data analysis were performed as what we had recently reported [
16]. The sequene tags were aligned to the reference genome hg19 using the Burrows–Wheeler Aligner (BWA) algorithm with default settings. The enrichment data of Nrf2 and HIF1a were visualized using University of California Santa Cruz (UCSC) genome browser. All ChIP-seq data can be accessed at
https://www.ncbi.nih.gov/geo/query/acc.cgi?acc=GSE145834 (accessed on 25 February 2020).
2.9. Statistical Analysis
All cell culture experiments were performed independently in triplicate at a minimum (unless otherwise indicated). Western blot images were analyzed using NIH ImageJ software. One-way Anova with 95% confidence interval followed by Tukey’s post hoc test was performed using IBM SPSS statistical software for Western blots and Seahorse cell phenotype data. Wilcoxon signed-rank tests and Kruskal–Wallis H tests were used for immunohistochemistry data. Figures were prepared using GraphPad Prism 5 and plotted as mean values with SEM. A p-value of < 0.05 was considered statistically significant.
4. Discussion
Arsenic, derived from the Greek word “arsenikos,” meaning potent, is a metalloid naturally present in the earth’s crust. However, natural and predominantly anthropogenic activities such as mining and agriculture have disturbed arsenic’s geochemical cycles, leading to its exposure in food, water, and air [
23]. Various case–control, cohort and ecological studies have demonstrated a positive correlation between high arsenic exposure and lung cancer development [
24]. To protect human health, the WHO International Agency for Research on Cancer (IARC) and US Environmental Protection Agency (EPA) have set an arsenic limit of 10 ppb in drinking water. Despite the regulations, arsenic exposure, especially the inorganic trivalent arsenic (As
3+) via contaminated drinking water is still a global health issue affecting more than 200 million people worldwide [
12].
Previous studies from our group have shown that long-term treatment of the BEAS-2B cells with the environmentally relevant concentration of As
3+ leads to the malignant transformation of these cells, and some of these cells acquire cancer stem-like (CSC) properties. Gene profiling and metabolomics analysis suggest impairment of the ER stress signaling, autophagy and mitochondrial functions [
7]. It is well documented that the UPR, mitochondria and autophagy work in conjunction under internal and external cellular stresses. It is also well known that the tri-components of the global ER stress response can restore cellular homeostasis or activate apoptosis under unresolved stress conditions [
12,
25]. However, since the long-term treatment procedure very likely causes adaptation and repopulation of the cells, it is important to determine whether the altered ER stress signaling is an initial event or a result of cellular adaptation. In a real-time metabolic phenotype assay in BEAS-2B cells treated with As
3+ for 6 h compared to control cells, we indeed noted a significant upregulation in glycolysis at a relatively high concentration of As
3+ (
Figure 6A), which is consistent with our results in As
3+-induced CSCs [
7]. This finding is also in agreement with the report by Zhao et al., who showed that As
3+ exposure in BEAS-2B cells induced an increased rate of extracellular acidification, which was inhibited by non-metabolized glucose analog 2-deoxy-D-glucose [
26]. Interestingly, unlike what was observed in the CSCs, the short-term As
3+ treatment also upregulated the mitochondrial respiration rates (
Figure 6B). Such a difference in mitochondrial metabolism between short- and long-term As
3+ treatment is very likely due to the accumulative damages of the functional proteins in mitochondria during a long-term As
3+ exposure.
Treatment of the BEAS-2B cells with As
3+ also caused a more than two-fold upregulation of five UPR genes, including ATF6, CEBPB, HSPA1B, MAPK10 and UBE2G2. ATF6 is an ER-resident UPR protein and a member of the basic leucine zipper family of transcription factors. It is a 90 kDa protein that is cleaved to a 50 kDa protein in the Golgi apparatus under ER stress. The cleaved form of N-ATF6 translocates to the nucleus to activate ER stress response genes [
27]. Mammals express two isoforms of ATF6 proteins, the bona fide ATF6, ATF6A (670 aa), and ATF6B (703 aa). The ATF6 and ATF6B proteins have similar highly conserved b-Zip domains that allow them to bind to ER stress response element (ERSE) as homo or heterodimers. However, they have a divergent N-terminal domain and vary in their ability to induce an ER stress response. The ATF6 protein is more studied than ATF6B protein [
20]. In the present study, we found that both ATF6 and ATF6B protein levels can be induced by As
3+, and both proteins showed proteolytic products in the As
3+-treated cells (
Figure 2 and
Figure 3) in the As
3+-treated cells. It has been implicated that only the cleaved forms of ATF6 and ATF6B can function as transcription factors for the transcription of the ER stress response genes in human bronchial epithelial cells, such as Grp78, Grp94, Xbp1, VEGF, etc. [
28,
29].
Mutual regulation of ER stress and Nrf2 activation had been studies in
C. elegans and certain neurodegenerative diseases [
30]. In this report, we provided the first evidence showing that Nrf2 is also a master regulator for the expression of the ER stress-associated UPR in cellular response to As
3+ through ChIP-seq and RNA-seq. On the genomic level, all of the As
3+-induced UPR genes contain conserved Nrf2-binding elements in either the promoter region or upstream of the transcription start site. Furthermore, some of them also showed significant enrichment of HIF1a binding in the promoter or gene body, such as CEBPB, HSPA1B and UBE2G2. HIF1a is a well-established oncogenic transcription factor during tumorigenesis. We had shown that the non-hypoxic induction of HIF1a by As
3+ is also Nrf2 dependent [
22]. Accordingly, there is a self-amplification and forward feedback loop among ER stress, Nrf2 and HIF1a induced by As
3+, which may serve as one of the key mechanisms of As
3+-induced carcinogenesis.
Although IHC evaluation of the ATF6 and ATF6B in lung cancer tissues vs. case-matched non-cancerous tissues is somewhat inconclusive, the overall survival analysis for the lung cancer patients unraveled an opposite prognostic value between ATF6 and ATF6B. Higher expression of ATF6 and ATF6B predicts a better and poorer OS of the lung cancer patients, respectively, indicating a possible tumor suppressor-like activity of ATF6 and an oncogenic property of the ATF6B for lung carcinogenesis. These findings also suggest that ATF6 and ATF6B may have different target genes for transcriptional regulation under physiological and carcinogenic conditions. Future studies are needed to define what sets of genes are regulated by these two transcription factors. Taken together, the data presented in this report demonstrated that As3+ is capable of activating the ER stress response signaling that may be actively involved in mediating the carcinogenic and/or tumorigenic processes associated with environmental As3+ exposure.