Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Nov 1.
Published in final edited form as: Semin Cancer Biol. 2021 Aug 21;76:279–286. doi: 10.1016/j.semcancer.2021.08.009

Dysregulation of microRNAs in metal-induced angiogenesis and carcinogenesis

Lin Wang a,b, Ling-Zhi Liu c,*, Bing-Hua Jiang b,*
PMCID: PMC8627485  NIHMSID: NIHMS1740873  PMID: 34428550

Abstract

MicroRNAs (miRNAs) are small endogenous non-coding RNAs that regulate cancer initiation, development, angiogenesis, and therapeutic resistance. Metal exposure widely occurs through air, water, soil, food, and industrial contaminants. Hundreds of millions of people may have metal exposure associated with toxicity, serious health problems, and cancer occurrence. Metal exposure is found to induce oxidative stress, DNA damage and repair, and activation of multiple signaling pathways. However, molecular mechanisms of metal-induced carcinogenesis remain to be elucidated. Recent studies demonstrated that the exposure of metals such as arsenic, hexavalent chromium, cadmium, and nickel caused dysregulation of microRNAs that are implicated to play an important role in cell transformation, tumor growth and angiogenesis. This review focuses on the recent studies that show metal-induced miRNA dysregulation and underlined mechanisms in cell malignant transformation, angiogenesis and tumor growth.

Keywords: Arsenic, Chromium, Nickel, Cadmium, microRNA, cell transformation, carcinogenesis, angiogenesis

1. Introduction

MicroRNAs (miRNAs) are small endogenous non-coding RNAs with the mature miRNA length of 18-23 base pairs. The first miRNA was discovered in the C. elegans as heterochronic gene lin-4 to control the gene expression of different coding genes by Ambros and colleagues in 1993[1]. Most miRNA genes are transcribed as long primary RNAs, called pre-miRNAs, and pri-miRNAs are initially processed in the nucleus by DROSHA, then released as hairpin-shaped precursors (pre-miRNA) and exported from nucleus to cytoplasm and cleaved by DICER, to generate a miRNA/miRNA* duplex [25]. The miRNA/miRNA* duplexes are loaded into an Argonaute (Ago) protein, which preferentially ejects the miRNA* strand, while retains the mature miRNA [6]. Additionally, Ago protein in cells may be associated with cofactors of the GW182/TNRC6 family proteins to form the RNA-induced silencing complex (RISC). Mature miRNA/Ago complexes recognize target mRNAs in cells by pairing miRNA seed regions with mRNAs [7]. Majority of human mRNAs are regulated by evolutionarily conserved miRNAs with the possibility of a particular miRNA which may post transcriptionally regulate hundreds of target mRNAs, and of each mRNA which may be suppressed by several different types of miRNAs [7]. miRNA stability and abundance can be altered during the pathological processes, and such multifaceted regulation of miRNAs ultimately influence gross changes in mRNA levels and gene expression that can regulate cancer development and numerous diseases [8].

Croce and colleagues initially discovered the importance of miRNAs in human cancer development when they found that miR-15a/16-1 cluster was frequently deleted in chronic lymphocytic leukemia (CLL), and miR-15a/16-1 loss was directly linked to Bcl2 overexpression [9]. After the discovery, alterations of miRNA expression have been found in all kinds of human cancers[1012]. Most miRNAs have been found to be downregulated in human cancer tissues compared to normal tissue counterparts, and to function as tumor suppressor-like miRNAs by directly targeting oncogenes; while only a small portion of miRNAs that are upregulated in cancer tissues and function as oncogene-like miRNAs by directly suppressing tumor suppressor genes [1315]. Huge body of information has been learned about the key roles of miRNAs in regulating cancer development, angiogenesis and drug resistance [1619]. Several metals including hexavalent chromium, arsenic, cadmium, and nickel are known carcinogens through the exposure of contaminated air, soil, water, and food; and the human exposures are strongly associated with various types of cancer development [2023]. In this review, we will summarize a growing list of recent studies that have shown that miRNA dysregulations are important in metal-induced cell transformation, tumor growth and angiogenesis.

2. MiRNA dysregulations in cancer development, angiogenesis and drug resistance

Since the discovery of the important role of miR-15a/16 cluster in chronic lymphocytic leukemia development, the field of miRNA dysregulation in cancer biology has attracted emerging and expanded interests, and huge number of new findings have been achieved to understand roles and mechanisms of miRNAs in cancer development, angiogenesis, diagnostics values, and therapeutic resistance [12, 2426]. Tumor angiogenesis is vital to tumor growth and metastasis, which is the process that new vessels sprout from preexisting blood vessels, and miR-21 promoted tumor angiogenesis via inducing HIF-1α and VEGF expression as well as activation of AKT and ERK pathways[27]. MiR-145 was suggested as a tumor suppressor that inhibits angiogenesis and tumor growth by inhibiting HIF-1α and VEGF expression [28]. Functional studies have demonstrated that miRNAs can act as tumor suppressors by directly suppressing oncogenes such as Myc and Ras, or act as oncogenes by targeting tumor suppressors such as p53 and PTEN [29]. MiRNAs have been used as cancer biomarkers to develop therapeutic kits that have been used for clinical diagnostics [30]. MiRNA-based therapeutics have reached clinical application including miR-34 which reached phase I clinical trials and miR-122 which reached phase II trials for treating hepatitis[13]. Cancer drug resistance is one of the major hurdles for advanced cancer treatments. The growing list of evidence has showed microRNAs play important roles in different types of cancer drug resistance. A few examples are described here. MiR-21 upregulation increased breast cancer cell resistance to DOX treatment by downregulating PTEN expression [31]. MiR-152 expression levels were dramatically decreased in the cisplatin-resistant ovarian cancer cell lines, and forced expression of miR-152 made the resistant cells sensitive to cisplatin treatment [32]. Downregulation of miR-579-3p played an important role in melanoma cancer cells resistant to BRAF/MEK inhibitors, and overexpression of miR-579-3p targeted the 3′UTR of two oncoproteins: BRAF and MDM2 for the inhibition, and restored the therapy sensitivity [33]. Downregulation of miR-143 was involved in resistance of colorectal cancer cells to oxaliplatin resistance, and forced expression of miR-143 restored oxaliplatin chemosensitivity to oxaliplatin treatment [34].

3. Arsenic

Arsenic is considered to be among the leading environmental carcinogens in the United States and in the world. Approximately 200 million people in the world are exposed to high levels of arsenic [35]. Populations are exposed to arsenic through the arsenic-containing drinking water, food, and air-dust. The occupational exposure to arsenic may occur through the inhalation of arsenic-containing particles in the production and distribution processes. There are more than 1.5 million industrial workers that are potentially exposed to arsenic and arsenic compounds according to the NIOSH estimate [36]. Higher risks of cancer were associated with the occupational or environmental arsenic exposure leading to skin, lung, bladder, and liver cancers [3740]. Although mechanisms of arsenic-induced carcinogenesis are still poorly understood, arsenic is considered to mainly alter epigenetic changes and gene expression, but not as a genotoxic carcinogen to induce gene mutations. Arsenic can increase oxidative stress, activate hypoxia and HIF-1 and other signaling pathways, and inhibit DNA repair that may contribute to the effect of arsenic in inducing angiogenesis and carcinogenesis [41, 42]. In human and rat bladder epithelial cells, chronic arsenic exposure upregulated HER2 expression, HER2 promoted cell proliferation, migration and angiogenesis by activating multiple signaling pathways, such as MAPK, PI3K/AKT and Src/STAT3 pathways in response to arsenic treatment [43]. Recent evidence showed that arsenic exposure caused DNA hypermethylation such as the hypermethylation of promoters of tumor suppressors p53 and p16 which inhibits their expression for inducing cell transformation and tumor growth [44, 45]. A list of recent studies found that dysregulations of miRNAs play an important role in arsenic-inducing carcinogenesis and angiogenesis (Table 1). The first evidence of critical role of arsenic-induced mi-RNA downregulation in cell transformation came from the study by the exposure of immortalized human bronchial epithelial cells (HBECs) with p53 knocked-down (p53lowHBECs) to low concentration of arsenic for 16 weeks, these p53lowHBECs cells became malignant transformation with morphology switch from epithelial to a spindle-like mesenchymal morphology, and greatly suppressed miR-200b/c expression [46]. The forced expression of miR-200b in the transformed cells completely inhibited the malignant transformation [46]. Similarly, downregulation of miR-200 family was observed in other cell types and human urine samples in response to arsenic exposure [47]. The downregulation of miR-199a-5p was observed in human bronchial epithelial BEAS-2B cells exposed to 1.0 μM arsenic for 26 weeks, and levels of miR-199a-5p in arsenic transformed cells were diminished to 100-fold lower than those in arsenic unexposed parental cells [48]. The suppression of miR-199a-5p greatly increased the expression levels of its direct targets HIF-1α and COX-2, and forced expression of miR-199a-5p diminished HIF-1α and COX-2 expression and impaired arsenic-induced angiogenesis and tumor growth [48]. Interestingly, miR-199a-5p expression was inhibited by ROS production in the cells through the promoter methylation of miR-199a gene by DNA methyltransferase 1[16]; while arsenic can induce ROS production in different cell types [49].

Table 1.

Arsenic exposure-induced alterations of miRNAs and carcinogenesis

miRNA Alteration Role Target(s) Role in carcinogenesis References
miR-217 Down Tumor suppressor EZH2 Inhibits cell cycle of human keratinocytes in carcinogenesis [120]
miR-155 Up Oncogene Quaking Activates STAT3 and induces IL6 and IL-8 in As-transformed liver cells; promotes CSC- like phenotype during the neoplastic transformation [50, 121]
miR-21 Up Oncogene Spry1, PTEN, PDCD4 Enhances the neoplastic capacity of As-transformed cells; induces angiogenesis and EMT [51, 56, 122, 123]
miR-182-5p Down Tumor suppressor N/A contributes to arsenic carcinogenesis via HIF-2α [124]
miR-889 Up Oncogene DAB2IP Promotes As-induced acquisition of CSC-like properties via DAB2IP/ZEB1 [125]
miR-455 Down Tumor suppressor ZEB1 Suppresses EMT during As-induced malignant transformation of human keratinocytes [126]
miR-15b Up Oncogene LATS1 inhibits the Hippo pathway and promotes cell proliferation, migration, and invasion [127]
miR-191 Up Oncogene BASP1 HIF-2α promotes miR-191 expression to induce angiogenesis and EMT in As-transformed cells [60, 128, 129]
miR-186 Up Oncogene BUB1 induces increased chromosome numbers and chromosome structural abnormalities [130]
let-7a/b/c Down Tumor suppressor N/A Suppresses cancer stem cell-like properties by inhibiting Ras/NF-κB pathway [131]
miR-181b and miR-9 Down Tumor suppressor neuropilin - 1 Inhibits arsenic- induced angiogenesis [132]
miR-134, miR-373, miR-155, miR-138, miR-205, miR-181d/c, let-7, miR-143, miR-34c-5p, and miR-205 Down Tumor suppressors RAN, RAB27A, RAB22A, KRAS, NRAS, and RRAS Suppress RAS activation and transformation [133]
miR-21, miR-200a and miR-141 Up Oncogenes CDK6, BMPR2, ACVR2B, and phosphati dylinositol-3-phosphate/5-kinase genes Affect MAPK, Wnt and the PI3K pathways [58]
miR-200b Down Tumor suppressor N/A As inhibits miR-200b expression through inducing ZEB1 and ZEB2 and hypermethylation, which promotes EMT and malignant transformation [46]
miR-410, miR-548ac, miR-3174 Down Tumor suppressor HMGB1, MDM2 Regulate TP53 regulatory network including MDM2 and HMGB1 [134]
miR-192b-5p, miR-15b-5p, and miR-33b-5p Down N/A N/A Downregulated in As-transformed HBC cells [135]
miR-141-3p, miR-106b-5p, and miR-200b-3p Up N/A N/A Upregulated in As-transformed HBC cells [135]
miR-221, miR-222 and miR-638 Up N/A RNF4 Upregulated upon 2 μM As treatment at 24 and 144 h [59]
miR-190 Up Oncogenes PHLPP cause Akt activation and increase VEGF expression [54]

Arsenic can also upregulate miRNA expression such as miR-21, miR-222, miR-190, and miR-155[5054]. MiR-21 expression levels were higher in human serum samples from individuals with arsenic exposure compared to those of the nonexposed subjects and were higher in HaCaT cells with arsenic exposure [55]. MiR-21 upregulation may directly target and inhibit tumor suppressors such as PTEN and programmed cell death 4 (PDCD4) expression [52, 56]. MiR-222 expression was significantly induced by arsenic exposure, and miR-222 in turn inhibited PTEN expression for inducing cell transformation and tumor growth [53]. MiR-190 levels were induced by arsenic to suppress the PH domain leucine-rich repeat protein phosphatase (PHLPP) expression, which is a negative regulator of Akt signaling [54]. MiR-155 expression levels were markedly increased by arsenic exposure through NF-κB induction [50]. Other miRNAs that are regulated by arsenic including Let 7 family, miR-141, miR-433, miR-184, miR-191, miR-151, miR-148b, and miR-183[55, 5760].

4. Chromium

Chromium may be in three different states: Cr (0), Cr(III), and Cr(VI); and only Cr(VI) is known carcinogen. Environmental and occupational Cr(VI) exposure through water, air or landfills has emerged as a major public health concern, and is associated with human lung cancer[6163]. Changes in signaling pathways and oxidative stress have been implicated as causal factors in response to Cr(VI) exposure[6466]. The epigenome was reported to be altered by chromium, the chromatin state changes through histone modifications as well as the DNA methylation landscape [67, 68]. However, the mechanisms underlying Cr(VI)-induced carcinogenesis and angiogenesis remain to be elucidated. The miRNA dysregulation was recently shown to be important in Cr(VI)-induced cell transformation, carcinogenesis and angiogenesis (Table 2). Our group initially demonstrated that miR-143 down-regulation played an important role in Cr(VI)-transformed human bronchial epithelial BEAS-2B cell transformation and angiogenesis[69]. Repression of miR-143 led to the transformation and angiogenesis of the cell, induced by Cr(VI), through upregulation of insulin-like growth factor 1 (IGF-IR) receptor and insulin receptor substrate 1 (IRS1). Furthermore, interleukin-8 was induced by Cr (VI) treatment, which was a major upregulated angiogenesis inducer through activations of the IGF-IR/IRS1 axis and ERK/HIF-1α/NF-κB signal pathway. This result suggest that the miR-143/IL-6/HIF-1α signaling pathway plays a vital role in Cr(VI)-induced cell malignant transformation and carcinogenesis[70]. MiR-143 suppression was also implicated in arsenic-induced cell transformation of the human normal prostate stem/progenitor cell (SC) line, which was a causal effect due to loss of miR-143 expression during the formation of prostate CSCs (As-CSCs). The forced miR-143 over-expression in these CSCs led to a considerable decrease in various types of cancer [71].

Table 2.

Chromium exposure-induced alterations of miRNAs and carcinogenesis

miRNA Alteration Role Target(s) Role in carcinogenesis References
miR-143 Down Tumor suppressor IGF-IR, IRS Suppress cell transformation and tumor angiogenesis via IGF-IR/IRS1/ERK/IL-8 pathway [69]
miR-222 Up Oncogene Promote MAPK and nerve growth factor pathways [72]
miR-494 Down Tumor suppressor c-Myc Inhibit CSC-like property and carcinogenesis by targeting c-Myc [75]
miR-3940-5p Down Tumor suppressor XRCC2, BRCC3 Protective effect in Cr(VI) induced DNA damage [76]
miR-19a-3p, miR-19b-3p and miR-142-3p Down N/A See Ref Regulate MAPK, cAMP, cGMP-PKG, and FoxO signaling pathways [77]
miR-590-3p Up N/A See Ref Regulate MAPK, cAMP, cGMP-PKG, and FoxO signaling pathways [77]
miR-21 and miR-155 Down N/A N/A N/A [78]

In the study of foundry workers in an electric-furnace steel plant, expression levels miR-222 were increased in post-exposition samples collected after 3 working days compared to the baseline samples [72]. MiR-222 expression levels were greatly increased in non-current smokers in response to chromium exposure, which may be regulated by MAPK and Nerve growth factor pathways [72]. Exposure to Cr(VI) was reported to induce CSC-like property through c-Myc which was significantly higher in human bronchial epithelial cells transformed by Cr(VI) compared with the control cells [73, 74]. Through using c-Myc shRNA, they found the significant decrease in the CSC-likes property of the Cr(VI)-transformed cells by downregulation of c-Myc expression, as evidenced by their decreased ability to form suspension spheres and tumorigenicity in nude mice. miR-494 expression was significantly down-regulated in Cr(VI)-transformed cells, and overexpression of miR-494 in Cr(VI)-transformed cells reduced c-Myc protein level, decreased Cr(VI)-induced CSC-like property and tumorigenesis[75]. In chromate manufacturing workers, lower levels of miR-3940-5p were associated with the lower blood Cr level, and miR-3940-5p mediated XRCC2 signal to promote DNA repair on DNA double-strand breaks induced by Cr(VI) exposure [76].

Other study showed that miR-19a-3p, miR-19b-3p and miR-142-3p were downregulated and miR-590-3p and miR-941 were upregulated in the plasma of workers occupationally exposed to Cr (VI) [77]. Target gene analysis showed that the miRNAs may participate in the regulation of DNA damage-related signaling pathways including cAMP, MAPK, FoxO, and cGMP-PKG pathways [77]. Welding fumes contain gases and metal oxides of chromium, cadmium, lead, nickel and others, which could be a threat on the reproductive system and renal function. In the study of welders, the correlations of blood and/or urine metal levels and expression levels of miR-21 and miR-155 were observed [78].

5. Nickel

Nickel is known as a common environmental pollutant and potent carcinogen. At the beginning of life, Nickel existed as a metal cofactor in the metabolism of methanogenic archaea [79, 80]. Nickel also functions as an essential element in biosynthesis of enzymes for bacteria and plant[81]. Ni compounds have been widely used in various human activities such as fossil fuel combustion and disposal from industries that can release almost 180,000 tons of nickel every year, which is a big burden to the environment. Studies suggested that workers under chronic Ni compounds exposure exhibited increased risk of nasal sinus cancer as well as lung cancer[82]. Thus, Nickel compounds has been classified as Class I human carcinogens by IARC [64, 83, 84].

Ni was found to induce gene expression which was associated with cancer and hypoxia, and Ni inhibited HIF-prolyl and asparaginyl-hydroxylases for stabilization of HIF-1α and HIF-1-dependant transcription[85]. Nickel may increase deletion mutations, DNA protein linkage and chromosomal aberrations in several report [8688]. Further investigation showed that Ni may preferably induce epigenetic change, rather than mutation [89]. Chronic Ni exposure was showed to induce changes in methylation of histone H3, histone H4 acetylation, phosphorylation of H3S10, and histones H2A/H2B ubiquitination[89, 90]. Nickel may decrease SLBP mRNA expression by regulating SLBP promoter region, such as declining histone acetylation or increasing DNA methylation, then promote SLBP protein degradation, finally cause H3.1 polyadenylation[91]. NiCl2 increased VEGF expression through activation of Akt, ERK, NF-κB and suppression of AMPK[43].

Nickel has been showed to induce dysregulation of several kinds of miRNAs (Table 3). miR-152 downregulation may be involved in cell transformation [92]. C57BL/6J wildtype mice treated with Nano-Ni increased miR-21 expression and proinflammatory cytokines such as IL-6, TNFα, TGF-β1, COL1A1 and COL3A1 [93], and human cohort study showed that higher miR-21 levels were associated with Ni exposure, EGFR mutation and lung tumor invasion[94]. MiR-4417 levels were the most upregulated miRNA by NiCl2 treatment in BEAS-2B and A549, and miR-4417 induction induced fibrogenesis, EMT and tumor progression through its target TAB2 [95].

Table 3.

Nickel exposure-induced alterations of miRNAs and carcinogenesis

miRNA Alteration Role Target(s) Role in carcinogenesis References
miR-4417 Up Oncogene TAB2 miR-4417/TAB2 involved in nickel-induced fibronectin, EMT and carcinogenesis [95]
miR-210 Up Oncogene ISCU1/2 Regulate n nickel induced aerobic glycolysis [98]
miR-21 Up Oncogene c-Myc Activate EGFR/NF-κB signaling pathway and promote carcinogenesis and invasiveness [94]
miR-203 Down Tumor suppressor ABL1 Inhibit carcinogenesis in vitro and in vivo by targeting ABL1 [99]
miR-152 Down Tumor suppressor DNMT1 Inhibit cell growth, malignant transformation and DNMT1 via a feedback mechanism [92]
miR-222 Up Oncogene CDKN1B, CDKN1C promote carcinogenesis and regulate target genes [136]

Aerobic glycolysis was significantly increased in cells treated with NiCl2, and was considered to play an important role in nickel-induced cell transformation and carcinogenesis [96, 97]. During glycolysis process, phosphofructokinase (PFK) catalyzes fructose-6-phosphate to fructose-1,6-diphosphate, which also considered a rate-limiting step. To investigate the mechanism of nickel in inducing carcinogenesis, nickel was showed to upregulate miR-210 and HIF-1α expression, ROS scavenger, N-acetylcysteine(NAC) and melatonin attenuated HIF-1α and miR-210 expression, and HIF-1α/miR210/ISCU axis was important in aerobic glycolysis Ni-induced tumorigenesis [97, 98]. MiR-203 was reported to be significantly downregulated in Ni3S2-transformed 16HBE cells though the hypermethylation of miR-203 promoter region, miR-203 overexpression inhibited cell proliferation, cell migration and anchorage-independent growth [99]. Nickel induced miR-203 suppression, and increased direct target gene ABL1 expression to promote tumor development [99].

6. Cadmium

Cadmium(Cd) is a metal that is rare in earth’s crust, Cadmium is typically functioned in a diatom for living in the marine environment that utilize the Cd-enzyme[100]. Cadmium accumulates in animals and plants with a long half-life of about 25-30 years, usually released into the environment as a well-known environmental and occupational pollutant in the world due to anthropogenic activities, which has been a significant public health concern for decades[101]. In 1993, cadmium and its compounds are classified as a Group 1 carcinogen by IARC[102]. Recent studies indicated that cadmium exposure increased the risk of several human cancers such as thyroid cancer, prostate cancer, breast bladder and lung cancers [103106].

Cadmium has a broad range of cell and molecular effects, both genetic and epigenetic, that may affect all stages of carcinogenic activity. The major mechanisms of cadmium-induced carcinogenesis include the induction of oxidative stress, ROS levels, decreased DNA repair capacity as well as epigenetic changes and alteration in gene expression[107109]. Cadmium induces many biochemical changes in experimental systems, including E-cadherin dysfunction, DNA methylation inhibition as well as associated with apoptosis by increasing p53 protein and mRNA levels[110]. After a low-dose Cd treatment, the signaling molecules associated with tumor angiogenesis of both tumor cells and epithelial cells directly were altered. Cadmium treatment increased HIF-1α and VEGF expression, and induced ROS, ERK and AKT signaling pathways [111]. Low-dose exposure to Cd also promoted the proliferation activity of lung cancer cells A549 to induce angiogenesis[112].

A list of evidence showed that cadmium-mediated microRNA dysregulations were important in cell proliferation, EMT, transformation, and cancer invasion (Table 4). Recent investigations showed that Cd induced the downregulation of miR-30 expression in lung epithelial cells, and miR-30 overexpression inhibited SNAIL gene expression to immediate Cd-induced EMT progression[106]. Dysregulations of hsa-miR-27b-3p, hsa-miR-1265, hsa-miR-944, hsv1-miR-H6-5p, hsa-miR-3960, hsa-miR-1261, hsa-miR-877-5p, and hsa-miR-4708-3p were observed in cadmium-induced cell transformation[113]. miR-101 expression levels were significantly decreased by CdCl2 treatment, and miR-101 overexpression directly targeted and inhibited COX-2 to decease ER stress/COX-2/VEGF pathway and angiogenesis [114].Other miRNAs were also affected by Cd treatment [115]. Interestingly, miRNA-21 expression was also upregulated in bladder cancer, the miRNA-21 expression levels were significantly higher in the samples obtained from occupationally metal exposed workers than those from the nonexposed subjects [116], and miRNA-21 would play a vital role in metal-induced bladder carcinogenesis[117].

Table 4.

Cadmium exposure-induced alterations of miRNAs and carcinogenesis

miRNA Alteration Role Target(s) Role in carcinogenesis References
miR-21 Up Oncogene N/A miR-21 in cancerous tissues was significantly associated with blood concentration of Cd [117]
miR-122-5p, miR-326-3p Up N/A PLD1 enhance cadmium-induced apoptosis in NRK-52E cell through inhibiting PLD1 expression [119]
miR-125a/b Down N/A Bak, Caspase3 regulate the suppression of Cd-induced apoptosis by Se via the mitochondrial pathway in LLC-PK1 cells [137]
miR-30a Down N/A N/A Cd triggered an miR-30a-GRP78 signaling axis disorder, increasing ER stress and activating the IRE-1-JNK pathway [138]
miR-30 family(miR-30a/b/c/d/e) Down Tumor suppressor SNAIL inhibit SNAIL levels, acquisition of EMT phenotype and carcinogenesis [106]
miR-363-3p Up N/A PI3K promote apoptosis by down-regulating the expression of PI3K [139]

MiRNA microarray was used to test expression levels of miRNAs in Cd-exposed rat ovarian granulosa cells(OGCs), 19 miRNAs were altered such as miR-133, miR-204-5p, miR-195, let-7 family, miR-105 and miR-135 in response to Cd treatment, and miR-204-5p/Bcl2 were involved in Cd-induced damage and apoptosis in OGCs[118]. In addition, miR-122-5p and miR-326-3p were involved in cadmium-induced apoptosis in NRK-52E cells through inhibiting PLD1 expression[119].

7. Concluding remarks and future direction

Chronic metal exposure may affect several hundreds of millions of people around the globe. MiRNA dysregulations have recently demonstrated to play an important role in metal-induced angiogenesis and carcinogenesis. Majority of miRNAs are downregulated by arsenic, Cr(VI), nickel and Cd, and these miRNAs function as tumor suppressor-like miRNAs that directly target and inhibit oncogene expression; while some miRNAs are upregulated by metal exposure and may function as oncogene-like miRNAs such as miR-21 and miR-122 that directly target and inhibit tumor suppressor PTEN expression. MiRNA expression is mainly mediated through epigenetic regulation including DNA methylation and transcription factors in the cells in response to metal exposure. Though significant progress has been made over the past decade, further studies are required to define the mechanisms for miRNA dysregulation induced by metal exposure. Further study is also required to understand the regulation mechanisms of key miRNAs that are important in regulating carcinogenesis and angiogenesis. The functional effects of miRNAs are through their target proteins, the expression levels of multiple potential target proteins are important to be analyzed for us to fully understand role and mechanism of miRNAs in metal-induced carcinogenesis. In addition, most results have been obtained through cell models, the future studies using animal models and human blood and urine samples are important for us to obtain new biomarkers, knowledge and findings that will be used to develop early diagnostic strategy, new preventive methods or/and treatment options for metal exposure-induced human diseases in the future.

Acknowledgments

The corresponding authors were supported in part by the NIH grants R01ES027901, K02ES029119, R01CA232587, and R01CA193511.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Conflicts of Interest

The authors declare no conflict of interest.

References

  • [1].Lee RC, Feinbaum RL, Ambros V, The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14, Cell 75(5) (1993) 843–54. [DOI] [PubMed] [Google Scholar]
  • [2].Hutvagner G, McLachlan J, Pasquinelli AE, Balint E, Tuschl T, Zamore PD, A cellular function for the RNA-interference enzyme Dicer in the maturation of the let-7 small temporal RNA, Science 293(5531) (2001) 834–8. [DOI] [PubMed] [Google Scholar]
  • [3].Lee Y, Ahn C, Han J, Choi H, Kim J, Yim J, Lee J, Provost P, Radmark O, Kim S, Kim VN, The nuclear RNase III Drosha initiates microRNA processing, Nature 425(6956) (2003) 415–9. [DOI] [PubMed] [Google Scholar]
  • [4].Bartel DP, MicroRNAs: genomics, biogenesis, mechanism, and function, Cell 116(2) (2004) 281–97. [DOI] [PubMed] [Google Scholar]
  • [5].Mendell JT, Olson EN, MicroRNAs in stress signaling and human disease, Cell 148(6) (2012) 1172–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Meister G, Argonaute proteins: functional insights and emerging roles, Nature reviews. Genetics 14(7) (2013) 447–59. [DOI] [PubMed] [Google Scholar]
  • [7].Hata A, Lieberman J, Dysregulation of microRNA biogenesis and gene silencing in cancer, Science signaling 8(368) (2015) re3. [DOI] [PubMed] [Google Scholar]
  • [8].Michlewski G, Caceres JF, Post-transcriptional control of miRNA biogenesis, Rna 25(1) (2019) 1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].Calin GA, Dumitru CD, Shimizu M, Bichi R, Zupo S, Noch E, Aldler H, Rattan S, Keating M, Rai K, Rassenti L, Kipps T, Negrini M, Bullrich F, Croce CM, Frequent deletions and down-regulation of micro-RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia, Proceedings of the National Academy of Sciences of the United States of America 99(24) (2002) 15524–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Lovat F, Valeri N, Croce CM, MicroRNAs in the pathogenesis of cancer, Seminars in oncology 38(6) (2011) 724–33. [DOI] [PubMed] [Google Scholar]
  • [11].Hayes J, Peruzzi PP, Lawler S, MicroRNAs in cancer: biomarkers, functions and therapy, Trends in molecular medicine 20(8) (2014) 460–9. [DOI] [PubMed] [Google Scholar]
  • [12].Peng Y, Croce CM, The role of MicroRNAs in human cancer, Signal transduction and targeted therapy 1 (2016) 15004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Rupaimoole R, Slack FJ, MicroRNA therapeutics: towards a new era for the management of cancer and other diseases, Nature reviews. Drug discovery 16(3) (2017) 203–222. [DOI] [PubMed] [Google Scholar]
  • [14].Calin GA, Croce CM, MicroRNA signatures in human cancers, Nature reviews. Cancer 6(11) (2006) 857–66. [DOI] [PubMed] [Google Scholar]
  • [15].Di Leva G, Garofalo M, Croce CM, MicroRNAs in cancer, Annual review of pathology 9 (2014) 287–314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].He J, Xu Q, Jing Y, Agani F, Qian X, Carpenter R, Li Q, Wang XR, Peiper SS, Lu Z, Liu LZ, Jiang BH, Reactive oxygen species regulate ERBB2 and ERBB3 expression via miR-199a/125b and DNA methylation, EMBO reports 13(12) (2012) 1116–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Romano G, Veneziano D, Acunzo M, Croce CM, Small non-coding RNA and cancer, Carcinogenesis 38(5) (2017) 485–491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [18].Van Roosbroeck K, Calin GA, Cancer Hallmarks and MicroRNAs: The Therapeutic Connection, Advances in cancer research 135 (2017) 119–149. [DOI] [PubMed] [Google Scholar]
  • [19].Xu Q, Liu LZ, Yin Y, He J, Li Q, Qian X, You Y, Lu Z, Peiper SC, Shu Y, Jiang BH, Regulatory circuit of PKM2/NF-kappaB/miR-148a/152-modulated tumor angiogenesis and cancer progression, Oncogene 34(43) (2015) 5482–93. [DOI] [PubMed] [Google Scholar]
  • [20].Chen QY, DesMarais T, Costa M, Metals and Mechanisms of Carcinogenesis, Annual review of pharmacology and toxicology 59 (2019) 537–554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Wang Z, Yang C, Metal carcinogen exposure induces cancer stem cell-like property through epigenetic reprograming: A novel mechanism of metal carcinogenesis, Seminars in cancer biology 57 (2019) 95–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Carpenter RL, Jiang BH, Roles of EGFR, PI3K, AKT, and mTOR in heavy metal-induced cancer, Current cancer drug targets 13(3) (2013) 252–66. [DOI] [PubMed] [Google Scholar]
  • [23].Wallace DR, Taalab YM, Heinze S, Tariba Lovakovic B, Pizent A, Renieri E, Tsatsakis A, Farooqi AA, Javorac D, Andjelkovic M, Bulat Z, Antonijevic B, Buha Djordjevic A, Toxic-Metal-Induced Alteration in miRNA Expression Profile as a Proposed Mechanism for Disease Development, Cells 9(4) (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Acunzo M, Romano G, Wernicke D, Croce CM, MicroRNA and cancer--a brief overview, Advances in biological regulation 57 (2015) 1–9. [DOI] [PubMed] [Google Scholar]
  • [25].Iorio MV, Croce CM, MicroRNA dysregulation in cancer: diagnostics, monitoring and therapeutics. A comprehensive review, EMBO molecular medicine 4(3) (2012) 143–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Rupaimoole R, Calin GA, Lopez-Berestein G, Sood AK, miRNA Deregulation in Cancer Cells and the Tumor Microenvironment, Cancer discovery 6(3) (2016) 235–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Liu LZ, Li C, Chen Q, Jing Y, Carpenter R, Jiang Y, Kung HF, Lai L, Jiang BH, MiR-21 induced angiogenesis through AKT and ERK activation and HIF-1alpha expression, PloS one 6(4) (2011) e19139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [28].Xu Q, Liu LZ, Qian X, Chen Q, Jiang Y, Li D, Lai L, Jiang BH, MiR-145 directly targets p70S6K1 in cancer cells to inhibit tumor growth and angiogenesis, Nucleic acids research 40(2) (2012) 761–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].Zhang B, Pan X, Cobb GP, Anderson TA, microRNAs as oncogenes and tumor suppressors, Developmental biology 302(1) (2007) 1–12. [DOI] [PubMed] [Google Scholar]
  • [30].Sundarbose K, Kartha RV, Subramanian S, MicroRNAs as Biomarkers in Cancer, Diagnostics 3(1) (2013) 84–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Wang ZX, Lu BB, Wang H, Cheng ZX, Yin YM, MicroRNA-21 modulates chemosensitivity of breast cancer cells to doxorubicin by targeting PTEN, Archives of medical research 42(4) (2011) 281–90. [DOI] [PubMed] [Google Scholar]
  • [32].He J, Yu JJ, Xu Q, Wang L, Zheng JZ, Liu LZ, Jiang BH, Downregulation of ATG14 by EGR1-MIR152 sensitizes ovarian cancer cells to cisplatin-induced apoptosis by inhibiting cyto-protective autophagy, Autophagy 11(2) (2015) 373–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Fattore L, Mancini R, Acunzo M, Romano G, Lagana A, Pisanu ME, Malpicci D, Madonna G, Mallardo D, Capone M, Fulciniti F, Mazzucchelli L, Botti G, Croce CM, Ascierto PA, Ciliberto G, miR-579-3p controls melanoma progression and resistance to target therapy, Proceedings of the National Academy of Sciences of the United States of America 113(34) (2016) E5005–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Qian X, Yu J, Yin Y, He J, Wang L, Li Q, Zhang LQ, Li CY, Shi ZM, Xu Q, Li W, Lai LH, Liu LZ, Jiang BH, MicroRNA-143 inhibits tumor growth and angiogenesis and sensitizes chemosensitivity to oxaliplatin in colorectal cancers, Cell cycle 12(9) (2013) 1385–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Naujokas MF, Anderson B, Ahsan H, Aposhian HV, Graziano JH, Thompson C, Suk WA, The broad scope of health effects from chronic arsenic exposure: update on a worldwide public health problem, Environmental health perspectives 121(3) (2013) 295–302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [36].Stohrer G, Arsenic: opportunity for risk assessment, Archives of toxicology 65(7) (1991) 525–31. [DOI] [PubMed] [Google Scholar]
  • [37].Basu A, Mahata J, Gupta S, Giri AK, Genetic toxicology of a paradoxical human carcinogen, arsenic: a review, Mutation research 488(2) (2001) 171–94. [DOI] [PubMed] [Google Scholar]
  • [38].Kligerman AD, Tennant AH, Insights into the carcinogenic mode of action of arsenic, Toxicology and applied pharmacology 222(3) (2007) 281–8. [DOI] [PubMed] [Google Scholar]
  • [39].Knobeloch LM, Zierold KM, Anderson HA, Association of arsenic-contaminated drinking-water with prevalence of skin cancer in Wisconsin’s Fox River Valley, Journal of health, population, and nutrition 24(2) (2006) 206–13. [PubMed] [Google Scholar]
  • [40].Banerjee M, Ferragut Cardoso A, Al-Eryani L, Pan J, Kalbfleisch TS, Srivastava S, Rai SN, States JC, Dynamic alteration in miRNA and mRNA expression profiles at different stages of chronic arsenic exposure-induced carcinogenesis in a human cell culture model of skin cancer, Archives of toxicology 95(7) (2021) 2351–2365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Liu LZ, Jiang Y, Carpenter RL, Jing Y, Peiper SC, Jiang BH, Role and mechanism of arsenic in regulating angiogenesis, PloS one 6(6) (2011) e20858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Jomova K, Jenisova Z, Feszterova M, Baros S, Liska J, Hudecova D, Rhodes CJ, Valko M, Arsenic: toxicity, oxidative stress and human disease, Journal of applied toxicology : JAT 31(2) (2011) 95–107. [DOI] [PubMed] [Google Scholar]
  • [43].Zhou Q, Jin P, Liu J, Li S, Liu W, Xi S, Arsenic-induced HER2 promotes proliferation, migration and angiogenesis of bladder epithelial cells via activation of multiple signaling pathways in vitro and in vivo, The Science of the total environment 753 (2021) 141962. [DOI] [PubMed] [Google Scholar]
  • [44].Reichard JF, Puga A, Effects of arsenic exposure on DNA methylation and epigenetic gene regulation, Epigenomics 2(1) (2010) 87–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Lu G, Xu H, Chang, Wu Z, Yao X, Zhang S, Li Z, Bai J, Cai Q, Zhang W, Arsenic exposure is associated with DNA hypermethylation of the tumor suppressor gene p16, Journal of occupational medicine and toxicology 9(1) (2014) 42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Wang Z, Zhao Y, Smith E, Goodall GJ, Drew PA, Brabletz T, Yang C, Reversal and prevention of arsenic-induced human bronchial epithelial cell malignant transformation by microRNA-200b, Toxicological sciences : an official journal of the Society of Toxicology 121(1) (2011) 110–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Michailidi C, Hayashi M, Datta S, Sen T, Zenner K, Oladeru O, Brait M, Izumchenko E, Baras A, VandenBussche C, Argos M, Bivalacqua TJ, Ahsan H, Hahn NM, Netto GJ, Sidransky D, Hoque MO, Involvement of epigenetics and EMT-related miRNA in arsenic-induced neoplastic transformation and their potential clinical use, Cancer prevention research 8(3) (2015) 208–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [48].He J, Wang M, Jiang Y, Chen Q, Xu S, Xu Q, Jiang BH, Liu LZ, Chronic arsenic exposure and angiogenesis in human bronchial epithelial cells via the ROS/miR-199a-5p/HIF-1alpha/COX-2 pathway, Environmental health perspectives 122(3) (2014) 255–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [49].Hu Y, Li J, Lou B, Wu R, Wang G, Lu C, Wang H, Pi J, Xu Y, The Role of Reactive Oxygen Species in Arsenic Toxicity, Biomolecules 10(2) (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Chen C, Luo F, Liu X, Lu L, Xu H, Yang Q, Xue J, Shi L, Li J, Zhang A, Liu Q, NF-κB-regulated exosomal miR-155 promotes the inflammation associated with arsenite carcinogenesis, Cancer letters 388 (2017) 21–33. [DOI] [PubMed] [Google Scholar]
  • [51].Ling M, Li Y, Xu Y, Pang Y, Shen L, Jiang R, Zhao Y, Yang X, Zhang J, Zhou J, Wang X, Liu Q, Regulation of miRNA-21 by reactive oxygen species-activated ERK/NF-kappaB in arsenite-induced cell transformation, Free radical biology & medicine 52(9) (2012) 1508–18. [DOI] [PubMed] [Google Scholar]
  • [52].Luo F, Ji J, Liu Y, Xu Y, Zheng G, Jing J, Wang B, Xu W, Shi L, Lu X, Liu Q, MicroRNA-21, upregulated by arsenite, directs the epithelial-mesenchymal transition and enhances the invasive potential of transformed human bronchial epithelial cells by targeting PDCD4, Toxicology letters 232(1) (2015) 301–9. [DOI] [PubMed] [Google Scholar]
  • [53].Wang M, Ge X, Zheng J, Li D, Liu X, Wang L, Jiang C, Shi Z, Qin L, Liu J, Yang H, Liu LZ, He J, Zhen L, Jiang BH, Role and mechanism of miR-222 in arsenic-transformed cells for inducing tumor growth, Oncotarget 7(14) (2016) 17805–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Beezhold K, Liu J, Kan H, Meighan T, Castranova V, Shi X, Chen F, miR-190-mediated downregulation of PHLPP contributes to arsenic-induced Akt activation and carcinogenesis, Toxicological sciences : an official journal of the Society of Toxicology 123(2) (2011) 411–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Cardoso APF, Al-Eryani L, States JC, Arsenic-Induced Carcinogenesis: The Impact of miRNA Dysregulation, Toxicological sciences : an official journal of the Society of Toxicology 165(2) (2018) 284–290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Liu X, Luo F, Ling M, Lu L, Shi L, Lu X, Xu H, Chen C, Yang Q, Xue J, Li J, Zhang A, Liu Q, MicroRNA-21 activation of ERK signaling via PTEN is involved in arsenite-induced autophagy in human hepatic L-02 cells, Toxicology letters 252 (2016) 1–10. [DOI] [PubMed] [Google Scholar]
  • [57].Al-Eryani L, Jenkins SF, States VA, Pan J, Malone JC, Rai SN, Galandiuk S, Giri AK, States JC, miRNA expression profiles of premalignant and malignant arsenic-induced skin lesions, PloS one 13(8) (2018)e0202579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [58].Gonzalez H, Lema C, Kirken RA, Maldonado RA, Varela-Ramirez A, Aguilera RJ, Arsenic-exposed Keratinocytes Exhibit Differential microRNAs Expression Profile; Potential Implication of miR-21, miR-200a and miR-141 in Melanoma Pathway, Clinical cancer drugs 2(2) (2015) 138–147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Sturchio E, Colombo T, Boccia P, Carucci N, Meconi C, Minoia C, Macino G, Arsenic exposure triggers a shift in microRNA expression, The Science of the total environment 472 (2014) 672–80. [DOI] [PubMed] [Google Scholar]
  • [60].Xu W, Luo F, Sun B, Ye H, Li J, Shi L, Liu Y, Lu X, Wang B, Wang Q, Liu Q, Zhang A, HIF-2alpha, acting via miR-191, is involved in angiogenesis and metastasis of arsenite-transformed HBE cells, Toxicology research 5(1) (2016) 66–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].Arakawa H, Weng MW, Chen WC, Tang MS, Chromium (VI) induces both bulky DNA adducts and oxidative DNA damage at adenines and guanines in the p53 gene of human lung cells, Carcinogenesis 33(10) (2012) 1993–2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Keegan GM, Learmonth ID, Case CP, A systematic comparison of the actual, potential, and theoretical health effects of cobalt and chromium exposures from industry and surgical implants, Critical reviews in toxicology 38(8) (2008) 645–74. [DOI] [PubMed] [Google Scholar]
  • [63].Urbano AM, Ferreira LM, Alpoim MC, Molecular and cellular mechanisms of hexavalent chromium-induced lung cancer: an updated perspective, Current drug metabolism 13(3) (2012) 284–305. [DOI] [PubMed] [Google Scholar]
  • [64].Chervona Y, Arita A, Costa M, Carcinogenic metals and the epigenome: understanding the effect of nickel, arsenic, and chromium, Metallomics : integrated biometal science 4(7) (2012) 619–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [65].Ivankovic S, Preussman R, Absence of toxic and carcinogenic effects after administration of high doses of chromic oxide pigment in subacute and long-term feeding experiments in rats, Food and cosmetics toxicology 13(3) (1975) 347–51. [DOI] [PubMed] [Google Scholar]
  • [66].Zhitkovich A, Peterson-Roth E, Reynolds M, Killing of chromium-damaged cells by mismatch repair and its relevance to carcinogenesis, Cell cycle 4(8) (2005) 1050–2. [PubMed] [Google Scholar]
  • [67].Arita A, Shamy MY, Chervona Y, Clancy HA, Sun H, Hall MN, Qu Q, Gamble MV, Costa M, The effect of exposure to carcinogenic metals on histone tail modifications and gene expression in human subjects, Journal of trace elements in medicine and biology : organ of the Society for Minerals and Trace Elements 26(2–3) (2012) 174–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [68].Chervona Y, Costa M, The control of histone methylation and gene expression by oxidative stress, hypoxia, and metals, Free radical biology & medicine 53(5) (2012) 1041–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [69].He J, Qian X, Carpenter R, Xu Q, Wang L, Qi Y, Wang ZX, Liu LZ, Jiang BH, Repression of miR-143 mediates Cr (VI)-induced tumor angiogenesis via IGF-IR/IRS1/ERK/IL-8 pathway, Toxicological sciences : an official journal of the Society of Toxicology 134(1) (2013) 26–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].Wang L, Qiu JG, He J, Liu WJ, Ge X, Zhou FM, Huang YX, Jiang BH, Liu LZ, Suppression of miR-143 contributes to overexpression of IL-6, HIF-1alpha and NF-kappaB p65 in Cr(VI)-induced human exposure and tumor growth, Toxicology and applied pharmacology 378 (2019) 114603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [71].Ngalame NN, Makia NL, Waalkes MP, Tokar EJ, Mitigation of arsenic-induced acquired cancer phenotype in prostate cancer stem cells by miR-143 restoration, Toxicology and applied pharmacology 312 (2016) 11–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [72].Bollati V, Marinelli B, Apostoli P, Bonzini M, Nordio F, Hoxha M, Pegoraro V, Motta V, Tarantini L, Cantone L, Schwartz J, Bertazzi PA, Baccarelli A, Exposure to metal-rich particulate matter modifies the expression of candidate microRNAs in peripheral blood leukocytes, Environmental health perspectives 118(6) (2010) 763–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Pratheeshkumar P, Son YO, Divya SP, Turcios L, Roy RV, Hitron JA, Wang L, Kim D, Dai J, Asha P, Zhang Z, Shi X, Hexavalent chromium induces malignant transformation of human lung bronchial epithelial cells via ROS-dependent activation of miR-21-PDCD4 signaling, Oncotarget 7(32) (2016) 51193–51210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [74].Wang Z, Wu J, Humphries B, Kondo K, Jiang Y, Shi X, Yang C, Upregulation of histone-lysine methyltransferases plays a causal role in hexavalent chromium-induced cancer stem cell-like property and cell transformation, Toxicology and applied pharmacology 342 (2018) 22–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [75].Wang Z, Lin HP, Li Y, Tao H, Yang P, Xie J, Maddy D, Kondo K, Yang C, Chronic Hexavalent Chromium Exposure Induces Cancer Stem Cell-Like Property and Tumorigenesis by Increasing c-Myc Expression, Toxicological sciences : an official journal of the Society of Toxicology 172(2) (2019) 252–264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [76].Li Y, Li P, Yu S, Zhang J, Wang T, Jia G, miR-3940-5p associated with genetic damage in workers exposed to hexavalent chromium, Toxicology letters 229(1) (2014) 319–26. [DOI] [PubMed] [Google Scholar]
  • [77].Jia J, Li T, Yao C, Chen J, Feng L, Jiang Z, Shi L, Liu J, Chen J, Lou J, Circulating differential miRNAs profiling and expression in hexavalent chromium exposed electroplating workers, Chemosphere 260 (2020)127546. [DOI] [PubMed] [Google Scholar]
  • [78].Amrani I, Haddam N, Garat A, Allorge D, Zerimech F, Schraen S, Taleb A, Merzouk H, Edme JL, Lo-Guidice JM, Exposure to metal fumes and circulating miRNAs in Algerian welders, International archives of occupational and environmental health 93(5) (2020) 553–561. [DOI] [PubMed] [Google Scholar]
  • [79].Konhauser KO, Pecoits E, Lalonde SV, Papineau D, Nisbet EG, Barley ME, Arndt NT, Zahnle K, Kamber BS, Oceanic nickel depletion and a methanogen famine before the Great Oxidation Event, Nature 458(7239) (2009) 750–3. [DOI] [PubMed] [Google Scholar]
  • [80].Thauer RK, My Lifelong Passion for Biochemistry and Anaerobic Microorganisms, Annual review of microbiology 69 (2015) 1–30. [DOI] [PubMed] [Google Scholar]
  • [81].Alfano M, Cavazza C, Structure, function, and biosynthesis of nickel-dependent enzymes, Protein science : a publication of the Protein Society 29(5) (2020) 1071–1089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [82].Coogan TP, Latta DM, Snow ET, Costa M, Toxicity and carcinogenicity of nickel compounds, Critical reviews in toxicology 19(4) (1989) 341–84. [DOI] [PubMed] [Google Scholar]
  • [83].Chromium, nickel and welding, IARC monographs on the evaluation of carcinogenic risks to humans 49 (1990) 1–648. [PMC free article] [PubMed] [Google Scholar]
  • [84].Sun H, Shamy M, Costa M, Nickel and epigenetic gene silencing, Genes 4(4) (2013) 583–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Ke Q, Costa M, Hypoxia-inducible factor-1 (HIF-1), Molecular pharmacology 70(5) (2006) 1469–80. [DOI] [PubMed] [Google Scholar]
  • [86].Conway K, Costa M, Nonrandom chromosomal alterations in nickel-transformed Chinese hamster embryo cells, Cancer research 49(21) (1989) 6032–8. [PubMed] [Google Scholar]
  • [87].Patierno SR, Sugiyama M, Basilion JP, Costa M, Preferential DNA-protein cross-linking by NiCl2 in magnesium-insoluble regions of fractionated Chinese hamster ovary cell chromatin, Cancer research 45(11 Pt 2) (1985) 5787–94. [PubMed] [Google Scholar]
  • [88].Sen P, Conway K, Costa M, Comparison of the localization of chromosome damage induced by calcium chromate and nickel compounds, Cancer research 47(8) (1987) 2142–7. [PubMed] [Google Scholar]
  • [89].Martinez-Zamudio R, Ha HC, Environmental epigenetics in metal exposure, Epigenetics 6(7) (2011) 820–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [90].Ke Q, Li Q, Ellen TP, Sun H, Costa M, Nickel compounds induce phosphorylation of histone H3 at serine 10 by activating JNK-MAPK pathway, Carcinogenesis 29(6) (2008) 1276–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [91].Jordan A, Zhang X, Li J, Laulicht-Glick F, Sun H, Costa M, Nickel and cadmium-induced SLBP depletion: A potential pathway to metal mediated cellular transformation, PloS one 12(3) (2017) e0173624. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [92].Ji W, Yang L, Yuan J, Yang L, Zhang M, Qi D, Duan X, Xuan A, Zhang W, Lu J, Zhuang Z, Zeng G, MicroRNA-152 targets DNA methyltransferase 1 in NiS-transformed cells via a feedback mechanism, Carcinogenesis 34(2) (2013) 446–53. [DOI] [PubMed] [Google Scholar]
  • [93].Mo Y, Zhang Y, Wan R, Jiang M, Xu Y, Zhang Q, miR-21 mediates nickel nanoparticle-induced pulmonary injury and fibrosis, Nanotoxicology 14(9) (2020) 1175–1197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [94].Chiou YH, Liou SH, Wong RH, Chen CY, Lee H, Nickel may contribute to EGFR mutation and synergistically promotes tumor invasion in EGFR-mutated lung cancer via nickel-induced microRNA-21 expression, Toxicology letters 237(1) (2015) 46–54. [DOI] [PubMed] [Google Scholar]
  • [95].Wu CH, Hsiao YM, Yeh KT, Tsou TC, Chen CY, Wu MF, Ko JL, Upregulation of microRNA-4417 and Its Target Genes Contribute to Nickel Chloride-promoted Lung Epithelial Cell Fibrogenesis and Tumorigenesis, Scientific reports 7(1) (2017) 15320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [96].Chen H, Costa M, Effect of soluble nickel on cellular energy metabolism in A549 cells, Experimental biology and medicine 231(9) (2006) 1474–80. [DOI] [PubMed] [Google Scholar]
  • [97].He M, Lu Y, Xu S, Mao L, Zhang L, Duan W, Liu C, Pi H, Zhang Y, Zhong M, Yu Z, Zhou Z, MiRNA-210 modulates a nickel-induced cellular energy metabolism shift by repressing the iron-sulfur cluster assembly proteins ISCU1/2 in Neuro-2a cells, Cell death & disease 5 (2014) e1090. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [98].He M, Zhou C, Lu Y, Mao L, Xi Y, Mei X, Wang X, Zhang L, Yu Z, Zhou Z, Melatonin Antagonizes Nickel-Induced Aerobic Glycolysis by Blocking ROS-Mediated HIF-1alpha/miR210/ISCU Axis Activation, Oxidative medicine and cellular longevity 2020 (2020) 5406284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [99].Zhang J, Zhou Y, Wu YJ, Li MJ, Wang RJ, Huang SQ, Gao RR, Ma L, Shi HJ, Zhang J, Hyper-methylated miR-203 dysregulates ABL1 and contributes to the nickel-induced tumorigenesis, Toxicology letters 223(1) (2013) 42–51. [DOI] [PubMed] [Google Scholar]
  • [100].Lane TW, Morel FM, A biological function for cadmium in marine diatoms, Proceedings of the National Academy of Sciences of the United States of America 97(9) (2000) 4627–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [101].Weidenhamer JD, Miller J, Guinn D, Pearson J, Bioavailability of cadmium in inexpensive jewelry, Environmental health perspectives 119(7) (2011) 1029–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [102].Beryllium, cadmium, mercury, and exposures in the glass manufacturing industry. Working Group views and expert opinions, Lyon, 9–16 February 1993, IARC monographs on the evaluation of carcinogenic risks to humans 58 (1993) 1–415. [PMC free article] [PubMed] [Google Scholar]
  • [103].Park E, Kim S, Song SH, Lee CW, Kwon JT, Lim MK, Park EY, Won YJ, Jung KW, Kim B, Environmental exposure to cadmium and risk of thyroid cancer from national industrial complex areas: A population-based cohort study, Chemosphere 268 (2021) 128819. [DOI] [PubMed] [Google Scholar]
  • [104].Rapisarda V, Miozzi E, Loreto C, Matera S, Fenga C, Avola R, Ledda C, Cadmium exposure and prostate cancer: insights, mechanisms and perspectives, Frontiers in bioscience 23 (2018) 1687–1700. [DOI] [PubMed] [Google Scholar]
  • [105].Strumylaite L, Kregzdyte R, Bogusevicius A, Poskiene L, Baranauskiene D, Pranys D, Cadmium Exposure and Risk of Breast Cancer by Histological and Tumor Receptor Subtype in White Caucasian Women: A Hospital-Based Case-Control Study, International journal of molecular sciences 20(12) (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [106].Tanwar VS, Zhang X, Jagannathan L, Jose CC, Cuddapah S, Cadmium exposure upregulates SNAIL through miR-30 repression in human lung epithelial cells, Toxicology and applied pharmacology 373 (2019) 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [107].Genchi G, Sinicropi MS, Lauria G, Carocci A, Catalano A, The Effects of Cadmium Toxicity, International journal of environmental research and public health 17(11) (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [108].Pizzino G, Bitto A, Interdonato M, Galfo F, Irrera N, Mecchio A, Pallio G, Ramistella V, De Luca F, Minutoli L, Squadrito F, Altavilla D, Oxidative stress and DNA repair and detoxification gene expression in adolescents exposed to heavy metals living in the Milazzo-Valle del Mela area (Sicily, Italy), Redox biology 2 (2014) 686–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [109].Xiao C, Liu Y, Xie C, Tu W, Xia Y, Costa M, Zhou X, Cadmium induces histone H3 lysine methylation by inhibiting histone demethylase activity, Toxicological sciences : an official journal of the Society of Toxicology 145(1) (2015) 80–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [110].Waisberg M, Joseph P, Hale B, Beyersmann D, Molecular and cellular mechanisms of cadmium carcinogenesis, Toxicology 192(2–3) (2003) 95–117. [DOI] [PubMed] [Google Scholar]
  • [111].Jing Y, Liu LZ, Jiang Y, Zhu Y, Guo NL, Barnett J, Rojanasakul Y, Agani F, Jiang BH, Cadmium increases HIF-1 and VEGF expression through ROS, ERK, and AKT signaling pathways and induces malignant transformation of human bronchial epithelial cells, Toxicological sciences : an official journal of the Society of Toxicology 125(1) (2012) 10–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [112].Liu F, Wang B, Li L, Dong F, Chen X, Li Y, Dong X, Wada Y, Kapron CM, Liu J, Low-Dose Cadmium Upregulates VEGF Expression in Lung Adenocarcinoma Cells, International journal of environmental research and public health 12(9) (2015) 10508–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [113].Liu Q, Zheng C, Shen H, Zhou Z, Lei Y, MicroRNAs-mRNAs Expression Profile and Their Potential Role in Malignant Transformation of Human Bronchial Epithelial Cells Induced by Cadmium, BioMed research international 2015 (2015) 902025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [114].Che L, Wu ZL, Huang LY, Wu JS, Du ZB, Lin JX, Su YH, Chen XX, Lin ZN, Lin YC, MicroRNA-101 inhibits cadmium-induced angiogenesis by targeting cyclooxygenase-2 in primary human umbilical vein endothelial cells, Biochemical pharmacology (2020) 114192. [DOI] [PubMed] [Google Scholar]
  • [115].Lemaire J, Van der Hauwaert C, Savary G, Dewaeles E, Perrais M, Lo Guidice JM, Pottier N, Glowacki F, Cauffiez C, Cadmium-Induced Renal Cell Toxicity Is Associated With MicroRNA Deregulation, International journal of toxicology 39(2) (2020) 103–114. [DOI] [PubMed] [Google Scholar]
  • [116].Xu M, Yu Z, Hu F, Zhang H, Zhong L, Han L, An Y, Zhu B, Zhang H, Identification of differential plasma miRNA profiles in Chinese workers with occupational lead exposure, Bioscience reports 37(5) (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [117].Awadalla A, Mortada WI, Abol-Enein H, Shokeir AA, Correlation between blood levels of cadmium and lead and the expression of microRNA-21 in Egyptian bladder cancer patients, Heliyon 6(12) (2020) e05642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [118].Zhong P, Liu J, Li H, Lin S, Zeng L, Luo L, Wu M, Zhang W, MicroRNA-204-5p regulates apoptosis by targeting Bcl2 in rat ovarian granulosa cells exposed to cadmiumdagger, Biology of reproduction 103(3) (2020) 608–619. [DOI] [PubMed] [Google Scholar]
  • [119].Yuan W, Liang L, Huang K, Deng Y, Dong M, Wang G, Zou F, MiR-122-5p and miR-326-3p promote cadmium-induced NRK-52E cell apoptosis by downregulating PLD1, Environmental toxicology 35(12) (2020) 1334–1342. [DOI] [PubMed] [Google Scholar]
  • [120].Xue J, Liu Y, Luo F, Lu X, Xu H, Liu X, Lu L, Yang Q, Chen C, Fan W, Liu Q, Circ100284, via miR-217 regulation of EZH2, is involved in the arsenite-accelerated cell cycle of human keratinocytes in carcinogenesis, Biochimica et biophysica acta. Molecular basis of disease 1863(3) (2017) 753–763. [DOI] [PubMed] [Google Scholar]
  • [121].Chen C, Luo F, Yang Q, Wang D, Yang P, Xue J, Dai X, Liu X, Xu H, Lu J, Zhang A, Liu Q, NF-kappaB-regulated miR-155, via repression of QKI, contributes to the acquisition of CSC-like phenotype during the neoplastic transformation of hepatic cells induced by arsenite, Molecular carcinogenesis 57(4) (2018) 483–493. [DOI] [PubMed] [Google Scholar]
  • [122].Luo F, Xu Y, Ling M, Zhao Y, Xu W, Liang X, Jiang R, Wang B, Bian Q, Liu Q, 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, Toxicology and applied pharmacology 273(1) (2013) 27–34. [DOI] [PubMed] [Google Scholar]
  • [123].Zhao Y, Xu Y, Luo F, Xu W, Wang B, Pang Y, Zhou J, Wang X, Liu Q, Angiogenesis, mediated by miR-21, is involved arsenite-induced carcinogenesis, Toxicology letters 223(1) (2013) 35–41. [DOI] [PubMed] [Google Scholar]
  • [124].Fang X, Sun R, Hu Y, Wang H, Guo Y, Yang B, Pi J, Xu Y, miRNA-182-5p, via HIF2alpha, contributes to arsenic carcinogenesis: evidence from human renal epithelial cells, Metallomics : integrated biometal science 10(11) (2018) 1607–1617. [DOI] [PubMed] [Google Scholar]
  • [125].Xiao T, Xue J, Shi M, Chen C, Luo F, Xu H, Chen X, Sun B, Sun Q, Yang Q, Dai X, Zhang A, Tang H, Liu Q, Circ008913, via miR-889 regulation of DAB2IP/ZEB1, is involved in the arsenite-induced acquisition of CSC-like properties by human keratinocytes in carcinogenesis, Metallomics : integrated biometal science 10(9) (2018) 1328–1338. [DOI] [PubMed] [Google Scholar]
  • [126].Xue J, Chen C, Luo F, Pan X, Xu H, Yang P, Sun Q, Liu X, Lu L, Yang Q, Xiao T, Dai X, Luo P, Lu J, Zhang A, Liu Q, CircLRP6 Regulation of ZEB1 via miR-455 Is Involved in the Epithelial-Mesenchymal Transition During Arsenite-induced Malignant Transformation of Human Keratinocytes, Toxicological sciences : an official journal of the Society of Toxicology 162(2) (2018) 450–461. [DOI] [PubMed] [Google Scholar]
  • [127].Li J, Xue J, Ling M, Sun J, Xiao T, Dai X, Sun Q, Cheng C, Xia H, Wei Y, Chen F, Liu Q, MicroRNA-15b in extracellular vesicles from arsenite-treated macrophages promotes the progression of hepatocellular carcinomas by blocking the LATS1-mediated Hippo pathway, Cancer letters 497 (2021) 137–153. [DOI] [PubMed] [Google Scholar]
  • [128].Chen C, Yang Q, Wang D, Luo F, Liu X, Xue J, Yang P, Xu H, Lu J, Zhang A, Liu Q, 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, Toxicology in vitro : an international journal published in association with BIBRA 48 (2018) 128–136. [DOI] [PubMed] [Google Scholar]
  • [129].Xu W, Ji J, Xu Y, Liu Y, Shi L, Liu Y, Lu X, Zhao Y, Luo F, Wang B, Jiang R, Zhang J, Liu Q, MicroRNA-191, by promoting the EMT and increasing CSC-like properties, is involved in neoplastic and metastatic properties of transformed human bronchial epithelial cells, Molecular carcinogenesis 54 Suppl 1 (2015) E148–61. [DOI] [PubMed] [Google Scholar]
  • [130].Wu J, Ferragut Cardoso AP, States VAR, Al-Eryani L, Doll M, Wise SS, Rai SN, States JC, Overexpression of hsa-miR-186 induces chromosomal instability in arsenic-exposed human keratinocytes, Toxicology and applied pharmacology 378 (2019) 114614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [131].Jiang R, Li Y, Zhang A, Wang B, Xu Y, Xu W, Zhao Y, Luo F, Liu Q, 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, Toxicology letters 227(2) (2014) 91–8. [DOI] [PubMed] [Google Scholar]
  • [132].Cui Y, Han Z, Hu Y, Song G, Hao C, Xia H, Ma X, MicroRNA-181b and microRNA-9 mediate arsenic-induced angiogenesis via NRP1, Journal of cellular physiology 227(2) (2012) 772–83. [DOI] [PubMed] [Google Scholar]
  • [133].Ngalame NN, Tokar EJ, Person RJ, Xu Y, Waalkes MP, Aberrant microRNA expression likely controls RAS oncogene activation during malignant transformation of human prostate epithelial and stem cells by arsenic, Toxicological sciences : an official journal of the Society of Toxicology 138(2) (2014) 268–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [134].Al-Eryani L, Waigel S, Tyagi A, Peremarti J, Jenkins SF, Damodaran C, States JC, 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, Toxicological sciences : an official journal of the Society of Toxicology 162(2) (2018) 645–654. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [135].Gu S, Sun D, Li X, Zhang Z, Alterations of miRNAs and Their Potential Roles in Arsenite-Induced Transformation of Human Bronchial Epithelial Cells, Genes 8(10) (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [136].Zhang J, Zhou Y, Ma L, Huang S, Wang R, Gao R, Wu Y, Shi H, Zhang J, The alteration of miR-222 and its target genes in nickel-induced tumor, Biological trace element research 152(2) (2013) 267–74. [DOI] [PubMed] [Google Scholar]
  • [137].Chen Z, Gu D, Zhou M, Shi H, Yan S, Cai Y, Regulatory role of miR-125a/b in the suppression by selenium of cadmium-induced apoptosis via the mitochondrial pathway in LLC-PK1 cells, Chemico-biological interactions 243 (2016) 35–44. [DOI] [PubMed] [Google Scholar]
  • [138].Shi Q, Jin X, Fan R, Xing M, Guo J, Zhang Z, Zhang J, Xu S, Cadmium-mediated miR-30a-GRP78 leads to JNK-dependent autophagy in chicken kidney, Chemosphere 215 (2019) 710–715. [DOI] [PubMed] [Google Scholar]
  • [139].Chen J, Lai W, Deng Y, Liu M, Dong M, Liu Z, Wang T, Li X, Zhao Z, Yin X, Yang J, Yu R, Liu L, MicroRNA-363-3p promotes apoptosis in response to cadmium-induced renal injury by down-regulating phosphoinositide 3-kinase expression, Toxicology letters 345 (2021) 12–23. [DOI] [PubMed] [Google Scholar]

RESOURCES