Skip to main content
Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
Toxicol Sci. 2018 Oct; 165(2): 284–290.
Published online 2018 May 28. doi: 10.1093/toxsci/kfy128
PMCID: PMC6154275
PMID: 29846715

Arsenic-Induced Carcinogenesis: The Impact of miRNA Dysregulation

Abstract

Arsenic is a toxic metalloid widely present in the earth’s crust, and is a proven human carcinogen. Chronic arsenic exposure mainly through drinking water causes skin, lung, and urinary bladder cancers, and is associated with liver, prostate, and kidney cancers, cardiovascular and neurological disorders, and diabetes. Several modes of action have been suggested in arsenic carcinogenesis. However, the molecular etiology of arsenic-induced cancer remains unclear. Recent evidence clearly indicates that gene expression modifications induced by arsenic may involve epigenetic alterations, including miRNA dysregulation. Many miRNAs have been implicated in different human cancers as a consequence of losses and or gains of miRNA function that contribute to cancer development. Progress in identifying miRNA dysregulation induced by arsenic has been made using different approaches and models. The present review discusses the recent data regarding dysregulated expression of miRNA in arsenic-induced malignant transformation in vitro, gaps in current understanding and deficiencies in current models for arsenic-induced carcinogenesis, and future directions of research that would improve our knowledge regarding the mechanisms involved in arsenic-induced carcinogenesis.

Keywords: arsenic, carcinogenesis, miRNA, metals non-genotoxic agents

Carcinogenesis is a complicated process that, despite much progress in delineating the role of mutation, is still incompletely understood. Recent studies have provided evidence that microRNAs (miRNAs or miRs) dysregulation plays a role in carcinogenesis of many tissue types. Focused studies in arsenic-induced cancers suggest that mutations may play a lesser role in arsenic-induced carcinogenesis (States, 2015). Instead, the findings from many studies have shown that miRNAs are critically involved in arsenic-induced malignant transformation. Here, we explore the recent data on dysregulation of miRNA expression in arsenic-exposed cells in vitro and discuss gaps in current understanding and deficiencies in current models for arsenic-induced carcinogenesis.

ARSENIC

Arsenic is a toxic metalloid present in the earth’s crust, and is one of the most well-known human carcinogens (IARC Working Group on the Evaluation of Carcinogenic Risks to Humans., 2012). Chronic arsenic exposure in drinking water is a global issue affecting at least 70 countries and >140 million people (Mukherjee et al., 2006). The U.S. Environmental Protection Agency (USEPA) and the World Health Organization (WHO) have set the Maximum Contaminant Level (MCL) for arsenic in water at 10 ppb (10 μg/l). However, a recent study showed that 2.1 million people in the U.S. use water from domestic wells with predicted arsenic concentration >10 μg/l (Ayotte et al., 2017). Epidemiological evidence has shown that long-term arsenic exposure is associated with human diseases such as skin lesions, cardiovascular disease, neurological disorders, and diabetes, and increased risk of developing skin, lung, and urinary bladder cancers (Chen et al., 2017; States, 2015). Several modes of action have been suggested in arsenic carcinogenesis including DNA repair inhibition, comutagenicity, oxidative stress, cell proliferation, aneuploidy, interaction with zinc finger proteins and epigenetic alterations including changes in DNA methylation, histone modification, and miRNA expression (Bailey and Fry, 2014; Reichard and Puga, 2010; States, 2015; Zhou et al., 2014). Recent in vitro studies support an association of exposure to arsenic with altered miRNA expression and alternative mRNA splicing (Al-Eryani et al., 2018; Riedmann et al., 2015).

miRNAs

Biogenesis and Biological Function

miRNAs are part of the epigenome and compose a dominating class of small RNAs in most somatic tissues. They are 21–22 nucleotide long RNAs that mediate posttranscriptional gene silencing by guiding Argonaute (AGO) proteins to mRNA targets (Ha and Kim, 2014; Hausser and Zavolan, 2014). miRNA biogenesis occurs in both the nucleus and the cytoplasm. The formation of mature miRNA is a 2-step process (Figure 1). In the first step, the primary transcript (pri-miRNA) is cleaved in the nucleus by the microprocessor complex into pre-miRNA (∼85-nucleotide pre-miRNA hairpin) (Peng and Croce, 2016). Then, the trimmed pre-miRNA is transported from the nucleus to the cytoplasm where the pre-miRNA is processed to a ∼22-nucleotide single-stranded mature miRNA (Peng and Croce, 2016). Mature miRNA is incorporated into an RNA-induced silencing complex (RISC) and guides the RISC to the target mRNA to regulate translation (Peng and Croce, 2016).

An external file that holds a picture, illustration, etc.
Object name is kfy128f1.jpg

miRNA biogenesis. The multiple steps of miRNAs biogenesis in both the nucleus and cytoplasm and their role in regulating their target mRNAs.

miRNAs target cytosolic mRNA by hybridization to complementary sequences, typically in 3′-untranslated regions (UTRs) called the “seed region” leading to decreased translation, deadenylation, or degradation of the mRNA (Bushati and Cohen, 2007; Hayes et al., 2014). Extensive pairing of miRNAs with their target mRNAs, eg, the seed region of mRNA matches perfectly with the targeting miRNA, leads to the degradation of the mRNA. However, imperfect base pairing leads to repression of mRNA translation and the possibility of restoring translation once the repressor miRNA is degraded (Macfarlane and Murphy, 2010).

miRNAs play a crucial role during developmental processes, apoptosis, and cell proliferation, and in regulating translation of most mammalian protein-coding genes (Friedman et al., 2009). Some miRNAs can also regulate a large number of zinc finger (ZNF) genes by directly targeting their coding regions (Huang et al., 2010). Dysregulation of miRNA is often associated with development, progression, and response to therapy of viral, immune-related and neurodegenerative diseases, and cancer (Li and Kowdley, 2012). The involved mechanisms of miRNA dysregulation in cancer include transcriptional dysregulation, epigenetic alterations, defects in the miRNA biogenesis pathway, and chromosomal instability (Peng and Croce, 2016).

Role in Carcinogenesis

miRNAs regulate molecular pathways by targeting various oncogenes and tumor suppressors having a function in cancer and stem cell biology, angiogenesis, the epithelial-mesenchymal transition, metastasis, and drug resistance (Humphries et al., 2016). Dysregulated miRNAs have a fundamental activity in cancer development and progression and may act as a novel class of oncogenes (by suppressing tumor suppressor genes) or of tumor suppressor genes (by suppressing oncogenes) (Figure 2) (Lotterman et al., 2008; Zhou et al., 2017). For example, miRNA let-7a down regulated MYC, a proto oncogene, in Burkitt lymphoma cells inhibiting cell growth induced by MYC (Sampson et al., 2007). On the other hand, overexpression of miR-504 negatively regulated p53, decreasing the p53-mediated apoptosis, and cell cycle arrest in response to stress (Hu et al., 2010). These 2 examples highlight the importance of microRNAs in tumorigenesis. Aberrant miRNA expression has been found in different cancers, including breast, colon, gastric, lung, prostate, and thyroid (Reddy, 2015). Thus, identification of miRNAs differentially expressed between normal and tumor tissues may help to define the contribution of miRNA in carcinogenesis initiation and progression and additionally may function as biomarkers for cancer diagnosis and prognosis (Humphries et al., 2016). A further role of miRNA may be implicated with chemical carcinogenesis since many studies have linked dysregulated miRNA expression and function after chemical exposure. Moreover, recent data have shown that arsenic can alter miRNA expression patterns in in vitro and in vivo models of arsenic-induced carcinogenesis (Table 1) (Humphries et al., 2016; Ren et al., 2015).

Table 1.

miRNA Dysregulation in Models of Arsenic-Induced Carcinogenesis

Type of StudyTarget OrganCell Type or Animal StrainArsenic ExposuremiRNAmiRNA ChangeReferences
In vitroSkinHaCaT500 nM for 4 weeks21, 200a, 141Upregulation(32)
HaCaT0.05 ppm for 8 weeks21Upregulation(33)
HaCaT1 μM for 15 weekslet-7a, let-7b, let7-cDownregulation(34)
Lung16-HBE2.5 μM for 13 weeks155Upregulation(5)
HBE1 μM for 30 passages21Upregulation(36)
BEAS-2B0.5 μM for 24 weeks21Upregulation(38)
BEAS-2B1 μM for 26 weeks199-aDownregulation(40)
Urinary bladderHUC11 μM for 48 and 60 weeks200a, 200b, 200cDownregulation(41)
LiverL-021, 2, 4, or 8 μM for 24 h21Upregulation(42)
L-022 μM for 15 weeks191Upregulation(43)
In vivoLiverSprague Dawley rats0.1, 1, 10, and 100 mg/l for 60 days151, 183Upregulation(24)
423, 26a, 148bDownregulation
An external file that holds a picture, illustration, etc.
Object name is kfy128f2.jpg

miRNAs as tumor suppressors and oncogenes. miRNA may act as transducer/mediator among oncogene and tumor suppressor genes leading to tumor formation (adapted from Paranjape et al., 2009).

miRNA DYSREGULATION IN ARSENIC-INDUCED CARCINOGENESIS

After the identification of miRNAs, several studies were performed exploring environmental carcinogens such as arsenic, altered miRNA expression profile, and carcinogenesis. The first work evaluating the impact of arsenic exposure on miRNA expression was published in 2006 (Marsit et al., 2006). Until 2010, relatively few studies in this field had been published. Some of these studies have focused on the therapeutic potential of arsenic in leukemia by modulating miRNAs, resulting in apoptosis inhibition (Gao et al., 2010; Li et al., 2010). Since then, a variety of studies have assessed the involvement of miRNAs mediating arsenic-induced carcinogenesis. As for example, miR-200 (Sahu, 2012), miR-190, (Ren et al., 2010), and miR-21 (Gonzalez et al., 2015; Sherr et al., 2016; Ye et al., 2017). However, as indicated by some authors, the majority of these studies have been performed in vitro using different cell lines, but the functional consequences of miRNA altered profile still need to be addressed (Bailey and Fry, 2014; Ren et al., 2010; Sahu, 2012). In the past 5 years, progress in identifying miRNA changes after arsenic exposure has been made using different approaches and models.

Skin

miR-21, miR-200a, and miR-141 were found to be overexpressed in HaCaT cells (immortalized human keratinocytes) after 4-week treatment with 500 nM sodium arsenite (Gonzalez et al., 2015). miR-21 and miR-141 have been reported to be associated with most human tumors (Gonzalez et al., 2015; Ye et al., 2017) and miR-200 family has been shown to have a role in the epithelial-mesenchymal transition (EMT), a manifestation of transformed cells, and cancer progression (Gonzalez et al., 2015). Arsenic may affect several signaling pathways through the modulation of these 3 dysregulated miRNAs, including mitogen-activated protein kinase (MAPK), Jak-STAT, and Wnt pathways. Genes simultaneously regulated by miR-21, miR-200a, and miR-141 through these pathways that may be considered for future work include cyclin-dependent kinase 6 (CKD6), which has important function in cell cycle progression and regulates the activity of the tumor suppressor retinoblastoma (RB) (Sherr et al., 2016). Recently, miR-21 expression was evaluated in human serum samples from individuals exposed to arsenic in West Bengal, India (Banerjee et al., 2017). The levels of miR-21 were upregulated in individuals exposed to arsenic compared with nonexposed. Furthermore, within the exposed group, miR-21 expression levels were higher in the individuals with skin lesions when compared with the individuals without skin lesions. miR-21 levels were also upregulated in HaCaT cells exposed to 0.05 ppm sodium arsenite for 8 weeks (Banerjee et al., 2017). This study also evaluated the levels of phosphatase and tensin homolog (PTEN) and programmed cell death 4 (PDCD4), downstream targets of miR-21, and survival proteins phosphorylated protein kinase B (pAKT) and phosphatidyl inositol 3 kinase (PI3K) in vivo and in vitro. PTEN and PCDC4 levels changed inversely with miR-21 expression, and pAKT and PI3K protein levels were found increased as miR-21 levels decreased (Banerjee et al., 2017). The relevant findings from this study show that miR-21 likely contributes to arsenic-induced skin lesions in an exposed population, and results from the in vitro work validate the in vivo data. However, it is noteworthy to mention the poor quality of the Western-blot images presented in the paper that could lead to a misinterpretation of the results. The let-7 family members have been identified as tumor-suppressing miRNAs (Jiang et al., 2014). Jiang et al. (2014) have induced neoplastic transformation in HaCaT cells after exposure to 1 µM arsenite for 15 weeks. Transformed cells showed decreased levels of let-7a, let-7b, and let-7c. These levels were also reduced in a time-dependent manner when HaCaT cells were acutely exposed to 1 µM arsenite. Further investigation of the role of let-7c suggested that arsenite activates the RAS/nuclear factor kappa-light-chain-enhancer of activated B cells (RAS/NF-κB) signaling pathway by decreasing the levels of let-7c through hypermethylation (Jiang et al., 2014).

Lung

Malignant transformation induced by arsenic has been observed in several types of lung cells. Overexpression of miR-155 in normal cells can lead to lung cancer development (Chen et al., 2017). To determine the role of miR-155 in arsenic-induced transformation of human bronchial epithelial cells (16-HBE), the expression levels of miR-155, nuclear factor (erythroid-derived 2)-like 2 (NRF2), and NF-kB were analyzed (Chen et al., 2017). 16-HBE cells underwent malignant transformation after 2.5 µM arsenite exposure for 13 weeks. miR-155 expression was increased in arsenic transformed cells, the levels of NRF2 were decreased and no change was observed in NF-kB levels. Inhibiting miR-155 expression enhanced NRF2 expression and decreased colony formation suggesting that miR-155 may play a role in the NRF2 signaling pathway regulation during arsenite-induced malignant transformation (Chen et al., 2017). In addition to inducing human skin cell malignant transformation, miR-21 up-regulation was reported in studies evaluating human lung cell transformation (Luo et al., 2013). Luo et al. (2015) investigated the effects of miR-21 on the EMT. Chronic 1.0 µM arsenite exposure induced human bronchial epithelial (HBE) cell transformation and EMT transition, characterized by loss of apical-basal polarity, redefining cell shape, and acquiring cancer stem cell (CSC) phenotype (Lamouille et al., 2014). Mechanistic studies reported that the levels of PDCD4, a tumor suppressor, and miR-21 target, decreased in transformed HBE cells. In addition, after inhibiting miR-21 and silencing PDCD4 RNA the EMT phenotype was still observed, indicating that miR-21 regulates EMT in arsenic-transformed HBE cells through PDCD4 (Luo et al., 2015). Interestingly, chronic exposure of human bronchial epithelial cell line (BEAS-2B) to arsenic-induced changes similar to those observed in HBE cells, such as overexpression of miR-21 with an associated decrease of PDCD4 levels leading to malignant transformation and tumorigenesis (Pratheeshkumar et al., 2016). Furthermore, arsenic-induced reactive oxygen species (ROS) and Neutrophil Cytosolic Factor 1 (NCF1, aka p47phox) expression; NCF1 is a neutrophil NADPH oxidase subunit required for ROS generation. Increased levels of interleukin 6 (IL-6), transcriptional activation of signal transducer and activator of transcription 3 (STAT3), and STAT3 phosphorylation were reported in BEAS-2B cells after arsenic exposure. Together, these results suggest that a ROS-STAT3-miR-21-PDCD4 signaling pathway is involved in arsenic-induced BEAS-2B malignant transformation (Pratheeshkumar et al., 2016). miR-199a is dysregulated and represses tumor progression in some types of cancer, including chondrosarcoma, hepatocellular, and ovarian cancer (Zhang et al., 2017). He et al. (2014) performed miRNA microarray analysis comparing the miRNA profiles between transformed BEAS-2B cells, chronically exposed to 1 μM arsenic for 26 weeks, and untreated cells. miR-199a was found down-regulated in arsenic transformed cells, and the levels of proangiogenic hypoxia-inducible factor (HIF)-1α and cyclooxygenase-2 (COX-2) were up-regulated, which could lead to enhanced angiogenesis and tumor growth, as suggested by the tube formation assay. Oxidative stress can be related to arsenic-mediated carcinogenesis (Zhang et al., 2015) and ROS can activate HIF-1α and COX-2 (Chen et al., 2012) and inhibit miR-199a expression (He et al., 2012). To evaluate the interaction between ROS and HIF-1α and COX-2, arsenic treated BEAS-2B cells overexpressing miR-199a were treated with H2O2. The cells did not express COX-2, demonstrating that downregulation of miR-199a by arsenic-induced ROS activates HIF-1α and COX-2 (He et al., 2014).

Urinary Bladder

Few available studies have linked miRNA dysregulation to arsenic-induced human urothelial cell transformation. Immortalized human urothelial cells (HUC1) were chronically exposed to 1 μM sodium arsenite resulting in malignant cell transformation and EMT acquisition (Michailidi et al., 2015). Because the miR-200 family members can reverse the EMT process, their levels were analyzed. Expression of miR-200a, miR-200b, and miR-200c was down-regulated in arsenic-exposed HUC1 cells compared with nonexposed HUC1 cells. Similarly, the levels of miR-200a, miR-200b, and miR-200c in the urine of arsenic-exposed individuals were decreased (Michailidi et al., 2015). These results suggest that miR-200 family members could have an important role in the development of urothelial human cancer induced by arsenic.

Liver

Acute exposure to sodium arsenite at 1, 2, 4, or 8 μM induced autophagy in human hepatic epithelial (L-02) cells (Liu et al., 2016). In addition, L-02 cells showed decreased expression of miR-21 target proteins, PTEN, PDCD4, and sprouty RTK signaling antagonist 1 (SPRY1) with upregulation of miR-21 levels in a concentration-dependent manner. Further investigation associated extracellular-signal-regulated kinase (ERK) activation via PTEN inhibition through up-regulation of miR-21 in arsenite-induced autophagy. Autophagy may influence cancer initiation and progression. Therefore, the proposed mechanism of arsenite-induced autophagy thorough miR-21 may be involved in arsenic-induced carcinogenesis (Liu et al., 2016). Chen et al. (2017, 2018) induced malignant transformation in L-02 cells by chronic exposure to 2 μM sodium arsenite. The expression of miR-191, an oncogenic miRNA (Nagpal et al., 2013), was found higher in transformed L-02 cells than in untransformed L-02 cells, as well as the markers for EMT induction (N-cadherin and α-smooth muscle actin [α-SMA]) and for liver CSCs (Cluster of Differentiation 90 [CD90] and Epithelial cell adhesion molecule [EpCAM]) (Chen et al., 2018). HIF-2α has been reported to exert a function in the arsenite-induced malignant transformation of HBE cells (Xu et al., 2012). Thus, the authors suggested that HIF-2α upregulates miR-191 levels, which are involved in the acquisition of a stem cell-like phenotype and in the epithelial-mesenchymal transition (Chen et al., 2018). An in vivo study was carried out by Ren et al. (2015) to analyze miRNA expression profiles in liver samples from Sprague Dawley (SD) rats exposed for 60 days to sodium arsenite at 0.1, 1, 10, and 100 mg/l in drinking water. The RNA-Seq and qPCR profile data demonstrated that miR-151 and miR-183 were up-regulated and miR-423, miR-26a, and miR-148b were down-regulated by arsenic exposure (Ren et al., 2015). These dysregulated miRNAs might be subjects for further investigations regarding mechanisms of arsenic-induced carcinogenesis in vivo and in vitro.

CURRENT DATA GAPS IN ARSENIC-INDUCED CARCINOGENESIS

Oral administration of arsenic to rodent models has provided significant information on human arsenic carcinogenesis although there is still a general lack of animal models, particularly an in vivo model for arsenic-induced skin cancer. The available studies evaluating arsenic and skin in rodents did not provide evidence of arsenic carcinogenicity by itself, rather, they demonstrated that arsenic is not a complete carcinogen for rodent skin (Burns et al., 2004; Motiwale et al., 2005; Rossman et al., 2001; Uddin et al., 2005), indicating that the biological response of humans to arsenic is different from that of rodents. Thus, the in vitro studies are the major source of information regarding arsenic-induced skin carcinogenesis. However, the in vitro models used to evaluate arsenic toxicity often use immortalized cell lines instead of primary cells. Although results provided by immortalized cell systems are generally consistent, not only may they not adequately represent primary cells but also may provide different results due to differences between immortalized and primary cells. Immortalized cell lines provide unlimited supply of material and should maintain functional features of the primary cells, although serial passages of cells could lead to genotypic and phenotypic variation over time and genetic drift could also cause heterogeneity in cultures. For these reasons, immortalized cells may not adequately mimic primary cells and may provide different results (Kaur and Dufour, 2012). A better understanding of how arsenic changes primary cells is essential to integrate epidemiological evidence and experimental data on arsenic and cancer risk.

Another issue concerning in vitro data consists of the arsenic concentration used in most of the peer-reviewed literature, which do not represent internal arsenic exposures at physiological levels. Epidemiological studies had measured arsenic physiological levels as ∼100 nM (Pi, 2000), a lower level than those that have been used in many arsenic-induced transformation studies. The high concentrations of arsenic might induce molecular responses and biological effects in vitro that might not reflect the chronic health outcomes experienced by individuals exposed to low to moderate arsenic concentrations. A large number of pathways and proteins have been analyzed in order to investigate arsenic effects. The levels of tumor protein P53 (TP53) expression remain conflicting, having been reported to be enhanced or decreased after arsenic treatment, depending on the cell line, dosing, and timing of exposure (Huang et al., 2008). Because TP53 plays an important role in the induction of cell cycle arrest, DNA repair signaling and/or apoptosis, further investigation is needed to explain the differences found in TP53 levels after arsenic exposure (Fischer, 2017). These results could clarify early signaling events caused by arsenic, delineating some of the details involved in cancer induction.

FUTURE DIRECTIONS

The mechanisms of arsenic-induced carcinogenesis are not yet clear. Several mechanisms, including epigenetic alterations are proposed. miRNAs are part of the epigenome and play an important role in gene regulation and cellular development (Leichter et al., 2017). The studies discussed above strongly suggest that dysregulated miRNA expression plays a critical role in arsenic-induced tumorigenesis and carcinogenesis although there are still some important aspects to consider in further investigations. First, the use of primary cells and physiologically relevant arsenic exposure could provide more relevant details about mechanisms involved in arsenic-induced cancer in humans. Second, findings from epidemiological and in vivo arsenic-related studies must be considered for choosing potential miRNAs for future research. Current studies analyzed selected miRNAs based on previous data showing differential expression in human tumors in general but not related to arsenic exposure. An unpublished pioneering work identified differential miRNA expression between human arsenic-induced premalignant and malignant skin lesions (Al-Eryani et al., manuscript submitted). Thus, further investigation could consider the identified miRNAs as targets for understanding their role in carcinogenesis induced by chronic exposure to arsenic. Furthermore, longitudinal studies following the miRNA differential expression throughout the transformation process are necessary to evaluate the critical changes during arsenic-associated transformation and carcinogenesis. Such information would improve our knowledge regarding the mechanisms involved in arsenic-induced carcinogenesis and direct the next steps of research.

FUNDING

The authors were supported in part by NIH-NIEHS grants 5R21ES023627-02 and 1R01ES027778-01A1.

REFERENCES

  • Al-Eryani L., Waigel S., Tyagi A., Peremarti J., Jenkins S. F., Damodaran C., States J. C. (2018). Differentially expressed mRNA targets of differentially expressed miRNAs predict changes in the TP53 axis and carcinogenesis related pathways in human keratinocytes chronically exposed to arsenic. Toxicol. Sci. 162, 645–654. [PMC free article] [PubMed] [Google Scholar]
  • Ayotte J. D., Medalie L., Qi S. L., Backer L. C., Nolan B. T. (2017). Estimating the high-arsenic domestic-well population in the conterminous United States. Environ. Sci. Technol. 51, 12443–12454. [PMC free article] [PubMed] [Google Scholar]
  • Bailey K. A., Fry R. C. (2014). Arsenic-associated changes to the epigenome: What are the functional consequences? Curr. Environ. Health Rep. 1, 22–34. [PMC free article] [PubMed] [Google Scholar]
  • Banerjee N., Bandyopadhyay A. K., Dutta S., Das J. K., Roy Chowdhury T., Bandyopadhyay A., Giri A. K. (2017). Increased microRNA 21 expression contributes to arsenic induced skin lesions, skin cancers and respiratory distress in chronically exposed individuals. Toxicology 378, 10–16. [PubMed] [Google Scholar]
  • Burns F. J., Uddin A. N., Wu F., Nádas A., Rossman T. G. (2004). Arsenic-induced enhancement of ultraviolet radiation carcinogenesis in mouse skin: A dose-response study. Environ. Health Perspect. 112, 599–603. [PMC free article] [PubMed] [Google Scholar]
  • Bushati N., Cohen S. M. (2007). microRNA functions. Annu. Rev. Cell Dev. Biol. 23, 175–205. [PubMed] [Google Scholar]
  • Chen C., Jiang X., Gu S., Zhang Z. (2017). MicroRNA-155 regulates arsenite-induced malignant transformation by targeting Nrf2-mediated oxidative damage in human bronchial epithelial cells. Toxicol. Lett. 278, 38–47. [PubMed] [Google Scholar]
  • Chen C., Yang Q., Wang D., Luo F., Liu X., Xue J., Yang P., Xu H., Lu J., Zhang A., et al. (2018). MicroRNA-191, regulated by HIF-2alpha, is involved in EMT and acquisition of a stem cell-like phenotype in arsenite-transformed human liver epithelial cells. Toxicol. In Vitro 48, 128–136. [PubMed] [Google Scholar]
  • Chen C.-C., Cheng Y.-Y., Chen S.-C., Tuan Y.-F., Chen Y.-J., Chen C.-Y., Chen L.-C. (2012). Cyclooxygenase-2 expression is up-regulated by 2-aminobiphenyl in a ROS and MAPK-dependent signaling pathway in a bladder cancer cell line. Chem. Res. Toxicol. 25, 695–705. [PubMed] [Google Scholar]
  • Fischer M. (2017). Census and evaluation of p53 target genes. Oncogene 36, 3943–3956. [PMC free article] [PubMed] [Google Scholar]
  • Friedman R. C., Farh K. K.-H., Burge C. B., Bartel D. P. (2009). Most mammalian mRNAs are conserved targets of microRNAs. Genome Res. 19, 92–105. [PMC free article] [PubMed] [Google Scholar]
  • Gao S-m., Chen C., Wu J., Tan Y., Yu K., Xing C.-Y., Ye A., Yin L., Jiang L. (2010). Synergistic apoptosis induction in leukemic cells by miR-15a/16-1 and arsenic trioxide. Biochem. Biophys. Res. Commun. 403, 203–208. [PubMed] [Google Scholar]
  • Gonzalez H., Lema C., Kirken R., A. Maldonado R., Varela-Ramirez A., J. Aguilera R. (2015). Arsenic-exposed keratinocytes exhibit differential microRNAs expression profile; potential implication of miR-21, miR-200a and miR-141 in melanoma pathway. Clin. Cancer Drugs 2, 138–147. [PMC free article] [PubMed] [Google Scholar]
  • Ha M., Kim V. N. (2014). Regulation of microRNA biogenesis. Nat. Rev. Mol. Cell Biol. 15, 509–524. [PubMed] [Google Scholar]
  • Hausser J., Zavolan M. (2014). Identification and consequences of miRNA-target interactions – Beyond repression of gene expression. Nat. Rev. Genet. 15, 599–612. [PubMed] [Google Scholar]
  • Hayes J., Peruzzi P. P., Lawler S. (2014). MicroRNAs in cancer: Biomarkers, functions and therapy. Trends Mol. Med. 20, 460–469. [PubMed] [Google Scholar]
  • He J., Wang M., Jiang Y., Chen Q., Xu S., Xu Q., Jiang B. H., Liu L. Z. (2014). Chronic arsenic exposure and angiogenesis in human bronchial epithelial cells via the ROS/miR-199a-5p/HIF-1alpha/COX-2 pathway. Environ. Health Perspect. 122, 255–261. [PMC free article] [PubMed] [Google Scholar]
  • He J., Xu Q., Jing Y., Agani F., Qian X., Carpenter R., Li Q., Wang X.-R., Peiper S. S., Lu Z., et al. (2012). Reactive oxygen species regulate ERBB2 and ERBB3 expression via miR-199a/125b and DNA methylation. EMBO Rep. 13, 1116–1122. [PMC free article] [PubMed] [Google Scholar]
  • Hu W., Chan C. S., Wu R., Zhang C., Sun Y., Song J. S., Tang L. H., Levine A. J., Feng Z. (2010). Negative regulation of tumor suppressor p53 by microRNA miR-504. Mol. Cell 38, 689–699. [PMC free article] [PubMed] [Google Scholar]
  • Huang S., Wu S., Ding J., Lin J., Wei L., Gu J., He X. (2010). MicroRNA-181a modulates gene expression of zinc finger family members by directly targeting their coding regions. Nucleic Acids Res. 38, 7211–7218. [PMC free article] [PubMed] [Google Scholar]
  • Huang Y., Zhang J., McHenry K. T., Kim M. M., Zeng W., Lopez-Pajares V., Dibble C. C., Mizgerd J. P., Yuan Z.-M. (2008). Induction of cytoplasmic accumulation of p53: A mechanism for low levels of arsenic exposure to predispose cells for malignant transformation. Cancer Res. 68, 9131–9136. [PMC free article] [PubMed] [Google Scholar]
  • Humphries B., Wang Z., Yang C. (2016). The role of microRNAs in metal carcinogen-induced cell malignant transformation and tumorigenesis. Food Chem. Toxicol. 98, 58–65. [PMC free article] [PubMed] [Google Scholar]
  • IARC Working Group on the Evaluation of Carcinogenic Risks to Humans. (2012). Arsenic, metals, fibres, and dusts. IARC Monogr. Eval. Carcinog. Risks Hum. 100(Pt C), 11–465. [PMC free article] [PubMed] [Google Scholar]
  • Jiang R., Li Y., Zhang A., Wang B., Xu Y., Xu W., Zhao Y., Luo F., Liu Q. (2014). The acquisition of cancer stem cell-like properties and neoplastic transformation of human keratinocytes induced by arsenite involves epigenetic silencing of let-7c via Ras/NF-kappaB. Toxicol. Lett. 227, 91–98. [PubMed] [Google Scholar]
  • Kaur G., Dufour J. M. (2012). Cell lines: Valuable tools or useless artifacts. Spermatogenesis 2, 1–5. [PMC free article] [PubMed] [Google Scholar]
  • Lamouille S., Xu J., Derynck R. (2014). Molecular mechanisms of epithelial-mesenchymal transition. Nat. Rev. Mol. Cell. Biol. 15, 178–196. [PMC free article] [PubMed] [Google Scholar]
  • Leichter A. L., Sullivan M. J., Eccles M. R., Chatterjee A. (2017). MicroRNA expression patterns and signalling pathways in the development and progression of childhood solid tumours. Mol. Cancer 16, 15.. [PMC free article] [PubMed] [Google Scholar]
  • Li Y., Kowdley K. V. (2012). MicroRNAs in common human diseases. Genomics Proteomics Bioinformatics 10, 246–253. [PMC free article] [PubMed] [Google Scholar]
  • Li Y., Zhu X., Gu J., Dong D., Yao J., Lin C., Huang K., Fei J. (2010). Anti-miR-21 oligonucleotide sensitizes leukemic K562 cells to arsenic trioxide by inducing apoptosis. Cancer Sci. 101, 948–954. [PMC free article] [PubMed] [Google Scholar]
  • Liu X., Luo F., Ling M., Lu L., Shi L., Lu X., Xu H., Chen C., Yang Q., Xue J., et al. (2016). MicroRNA-21 activation of ERK signaling via PTEN is involved in arsenite-induced autophagy in human hepatic L-02 cells. Toxicol. Lett. 252, 1–10. [PubMed] [Google Scholar]
  • Lotterman C. D., Kent O. A., Mendell J. T. (2008). Functional integration of microRNAs into oncogenic and tumor suppressor pathways. Cell Cycle 7, 2493–2499. [PMC free article] [PubMed] [Google Scholar]
  • Luo F., Ji J., Liu Y., Xu Y., Zheng G., Jing J., Wang B., Xu W., Shi L., Lu X., et al. (2015). MicroRNA-21, up-regulated by arsenite, directs the epithelial-mesenchymal transition and enhances the invasive potential of transformed human bronchial epithelial cells by targeting PDCD4. Toxicol. Lett. 232, 301–309. [PubMed] [Google Scholar]
  • Luo F., Xu Y., Ling M., Zhao Y., Xu W., Liang X., Jiang R., Wang B., Bian Q., Liu Q., et al. (2013). Arsenite evokes IL-6 secretion, autocrine regulation of STAT3 signaling, and miR-21 expression, processes involved in the EMT and malignant transformation of human bronchial epithelial cells. Toxicol. Appl. Pharmacol. 273, 27–34. [PubMed] [Google Scholar]
  • Macfarlane L. A., Murphy P. R. (2010). MicroRNA: Biogenesis, function and role in cancer. Curr. Genomics 11, 537–561. [PMC free article] [PubMed] [Google Scholar]
  • Marsit C. J., Eddy K., Kelsey K. T. (2006). MicroRNA responses to cellular stress. Cancer Res. 66, 10843–10848. [PubMed] [Google Scholar]
  • Michailidi C., Hayashi M., Datta S., Sen T., Zenner K., Oladeru O., Brait M., Izumchenko E., Baras A., VandenBussche C., et al. (2015). Involvement of epigenetics and EMT-related miRNA in arsenic-induced neoplastic transformation and their potential clinical use. Cancer Prev. Res. (Phila) 8, 208–221. [PMC free article] [PubMed] [Google Scholar]
  • Motiwale L., Ingle A. D., Rao K. V. (2005). Mouse skin tumor promotion by sodium arsenate is associated with enhanced PCNA expression. Cancer Lett. 223, 27–35. [PubMed] [Google Scholar]
  • Mukherjee A., Sengupta M. K., Hossain M. A., Ahamed S., Das B., Nayak B., Lodh D., Rahman M. M., Chakraborti D. (2006). Arsenic contamination in groundwater: A global perspective with emphasis on the Asian scenario. J. Health Popul. Nutr. 24, 142–163. [PubMed] [Google Scholar]
  • Nagpal N., Ahmad H. M., Molparia B., Kulshreshtha R. (2013). MicroRNA-191, an estrogen-responsive microRNA, functions as an oncogenic regulator in human breast cancer. Carcinogenesis 34, 1889–1899. [PubMed] [Google Scholar]
  • Paranjape T., Slack F. J., Weidhaas J. B. (2009). MicroRNAs: Tools for cancer diagnostics. Gut 58, 1546–1554. [PMC free article] [PubMed] [Google Scholar]
  • Peng Y., Croce C. M. (2016). The role of MicroRNAs in human cancer. Signal. Transduct. Target Ther. 1, 15004.. [PMC free article] [PubMed] [Google Scholar]
  • Pi J. (2000). Decreased serum concentrations of nitric oxide metabolites among Chinese in an endemic area of chronic arsenic poisoning in inner Mongolia. Free Radic. Biol. Med. 28, 1137–1142. [PubMed] [Google Scholar]
  • Pratheeshkumar P., Son Y.-O., Divya S. P., Wang L., Zhang Z., Shi X. (2016). Oncogenic transformation of human lung bronchial epithelial cells induced by arsenic involves ROS-dependent activation of STAT3-miR-21-PDCD4 mechanism. Sci. Rep. 6, 37227.. [PMC free article] [PubMed] [Google Scholar] Retracted
  • Reddy K. B. (2015). MicroRNA (miRNA) in cancer. Cancer Cell Int. 15, 38.. [PMC free article] [PubMed] [Google Scholar]
  • Reichard J. F., Puga A. (2010). Effects of arsenic exposure on DNA methylation and epigenetic gene regulation. Epigenomics 2, 87–104. [PMC free article] [PubMed] [Google Scholar]
  • Ren X., Gaile D. P., Gong Z., Qiu W., Ge Y., Zhang C., Huang C., Yan H., Olson J. R., Kavanagh T. J., et al. (2015). Arsenic responsive microRNAs in vivo and their potential involvement in arsenic-induced oxidative stress. Toxicol. Appl. Pharmacol. 283, 198–209. [PMC free article] [PubMed] [Google Scholar]
  • Ren X., McHale C. M., Skibola C. F., Smith A. H., Smith M. T., Zhang L. (2010). An emerging role for epigenetic dysregulation in arsenic toxicity and carcinogenesis. Environ. Health Perspect. 119, 11–19. [PMC free article] [PubMed] [Google Scholar]
  • Riedmann C., Ma Y., Melikishvili M., Godfrey S. G., Zhang Z., Chen K. C., Rouchka E. C., Fondufe-Mittendorf Y. N. (2015). Inorganic arsenic-induced cellular transformation is coupled with genome wide changes in chromatin structure, transcriptome and splicing patterns. BMC Genomics 16, 212.. [PMC free article] [PubMed] [Google Scholar]
  • Rossman T. G., Uddin A. N., Burns F. J., Bosland M. C. (2001). Arsenite is a cocarcinogen with solar ultraviolet radiation for mouse skin: An animal model for arsenic carcinogenesis. Toxicol. Appl. Pharmacol. 176, 64–71. [PubMed] [Google Scholar]
  • Sahu S. C. (2012). Toxicology and Epigenetics. Chichester, West Sussex, U.K.; Hoboken, N.J: John Wiley & Sons. xxix, 658 p. [Google Scholar]
  • Sampson V. B., Rong N. H., Han J., Yang Q., Aris V., Soteropoulos P., Petrelli N. J., Dunn S. P., Krueger L. J. (2007). MicroRNA let-7a down-regulates MYC and reverts MYC-induced growth in Burkitt lymphoma cells. Cancer Res. 67, 9762–9770. [PubMed] [Google Scholar]
  • Sherr C. J., Beach D., Shapiro G. I. (2016). Targeting CDK4 and CDK6: From discovery to therapy. Cancer Discov. 6, 353–367. [PMC free article] [PubMed] [Google Scholar]
  • States J. C. (2015). Disruption of mitotic progression by arsenic. Biol. Trace Elem. Res. 166, 34–40. [PMC free article] [PubMed] [Google Scholar]
  • Uddin A. N., Burns F. J., Rossman T. G. (2005). Vitamin E and organoselenium prevent the cocarcinogenic activity of arsenite with solar UVR in mouse skin. Carcinogenesis 26, 2179–2186. [PubMed] [Google Scholar]
  • Xu Y., Li Y., Pang Y., Ling M., Shen L., Jiang R., Zhao Y., Zhou J., Wang X., Liu Q., et al. (2012). Blockade of p53 by HIF-2alpha, but not HIF-1alpha, is involved in arsenite-induced malignant transformation of human bronchial epithelial cells. Arch. Toxicol. 86, 947–959. [PubMed] [Google Scholar]
  • Ye Q., Lei L., Shao L., Shi J., Jia J., Tong X. (2017). MicroRNA141 inhibits epithelialmesenchymal transition, and ovarian cancer cell migration and invasion. Mol. Med. Rep. 16, 6743–6749. [PMC free article] [PubMed] [Google Scholar]
  • Zhang X., Li P., Ding Z., Wang H., Wang J., Han L., Ding S. (2017). The putative tumor suppressor, miR-199a, regulated by Snail, modulates clear cell renal cell carcinoma aggressiveness by repressing ROCK1. Oncol. Targets Ther. 11, 103–112. [PMC free article] [PubMed] [Google Scholar]
  • Zhang Z., Pratheeshkumar P., Budhraja A., Son Y.-O., Kim D., Shi X. (2015). Role of reactive oxygen species in arsenic-induced transformation of human lung bronchial epithelial (BEAS-2B) cells. Biochem. Biophys. Res. Commun. 456, 643–648. [PMC free article] [PubMed] [Google Scholar]
  • Zhou K., Liu M., Cao Y. (2017). New insight into microRNA functions in cancer: Oncogene-microRNA-tumor suppressor gene network. Front. Mol. Biosci. 4, 46.. [PMC free article] [PubMed] [Google Scholar]
  • Zhou X., Sun X., Mobarak C., Gandolfi A. J., Burchiel S. W., Hudson L. G., Liu K. J. (2014). Differential binding of monomethylarsonous acid compared to arsenite and arsenic trioxide with zinc finger peptides and proteins. Chem. Res. Toxicol. 27, 690–698. [PMC free article] [PubMed] [Google Scholar]

Articles from Toxicological Sciences are provided here courtesy of Oxford University Press

-