MicroRNAs serve as a bridge between oxidative stress and gastric cancer (Review)

  • Authors:
    • Tianhe Huang
    • Feng Wang-Johanning
    • Fuling Zhou
    • Herbert Kallon
    • Yongchang Wei
  • View Affiliations

  • Published online on: September 9, 2016     https://doi.org/10.3892/ijo.2016.3686
  • Pages: 1791-1800
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Gastric cancer (GC) remains one of the most prevalent tumors worldwide and affects human health due to its high morbidity and mortality. Mechanisms underlying occurrence and development of GC have been widely studied. Studies have revealed reactive oxygen species (ROS) generated by cells under oxidative stress (OS) are involved in gastric tumorigenesis, and modulate expression of microRNAs (miRs). As such, miRs have been shown to be associated with OS-related GC. Given the association of OS and miRs in development of GC, this review aims to summarize the relationship between miRs and OS and their role in GC development. Serving as a link between OS and GC, miRs may offer new approaches for gaining a more in-depth understanding of mechanisms of GC and may lead to the identification of new therapeutic approaches against GC.

1. Introduction

Gastric cancer (GC) is one of the most common types of cancers worldwide. Each year, almost one million people are newly diagnosed with GC, and over 700,000 people die of GC (1). Due to high prevalence and death rate, GC remains a tremendous threat to word health. An increasing body of evidence shows that patients with GC experience high levels of OS, which contribute to progression of GC. Markers of OS, such as 8-hydroxy-2′-deoxyguanosine (8-OHdG), human oxoguanine glycosylase 1 (hOGG1) and xanthine oxidase (XOD) are aberrant in GC (2). Due to redox imbalance, OS participates in progression of GC by affecting expression of critical effectors, including miRs (3,4). miRs are small non-coding RNAs that modulate gene expression by either inhibiting mRNA translation or inducing mRNA degradation at post-transcriptional level (5). miRs have been relevant to promotion or suppression of tumorigenesis in GC and are implicated in cell growth, differentiation, invasion, metastasis, and apoptosis in GC (6). Additionally, OS gives rise to abnormal expression of miRs in various types of diseases, including cancers (79). Although function of miRs has been extensively investigated in GC, whether miRs may be associated with or serve as a link between OS and GC remains unknown. In this review, we focus on roles of OS in GC, OS-induced miRs in GC, and roles of dysfunctional miRs in GC to gain further understanding of the association of OS and miRs in gastric tumorigenesis.

2. OS is involved in the tumorigenesis of GC through ROS

OS, which refers to redox imbalance with excessive production of ROS exceeding the scavenging ability of human body (10), has been associated with pathophysiology of both solid tumors and hematological malignant diseases (11,12). ROS, including superoxide (O2•−), hydroxide (OH), and hydrogen peroxide (H2O2), lead to damage to cell membrane, protein, and DNA (13). Physiologically, glutathione peroxidase (GPX), superoxide dismutase (SOD), and catalase could scavenge ROS to preserve redox homeostasis. ROS also contribute to cell apoptosis by regulating p38α mitogen-activated protein kinases (MAPK) (14). More importantly, ROS stimulate the NF-E2-related factor (NRF2)-mediated activation of an antioxidant response element (ARE) to protect cells again OS (15). Nevertheless, under OS, excessive ROS in cells could damage tissues, leading to tumorigenesis, particularly in the gastrointestinal tract.

For GC, in both human and mouse, ROS are actuated factors in gastric carcinogenesis. In human GC, ROS are dysregulated in serum and tissue samples (16). In Helicobacter pylori (Hp) along with N-methyl-N′-nitro-N′-nitrosoguanidine (MNNG) triggered mouse GC models, ROS activated downstream targets including P53, Wnt, Ras, mTOR to initiate gastric carcinogenesis (17,18). Hypoxia leads to producing ROS, which prevent degradation of hypoxia-inducible factor 1α (HIF-1α) (19). As a transcription factor, accumulation of HIF-1α is able to modulate expression of various genes, which are important in gastric tumorigenesis. For example, HIF-1α attenuates expression of caveolin-1 (Cav-1), which can induce the epithelial-mesenchymal transition (EMT) in GC through regulation of E-cadherin (20). HIF-1α also activates vascular endothelial growth factor (VEGF) pathway to enhance angiogenesis in GC (21). In contrast, ROS act as a signaling molecule and trigger essential signaling pathways indispensible to promoting the occurrence and development of GC. As second messenger, ROS are well known to activate tyrosine kinases and MAPK, leading to cell proliferation (22), as well as trigger activation of protein kinase-B (Akt)/mammalian target of rapamycin (mTOR) signaling pathway to promote GC cell proliferation (23). Moreover, ROS have been found to stimulate nuclear factor-κB (NF-κB) to promote GC invasion (24,25).

In addition, OS exerts a crucial role in Hp-induced diseases. Hp infection is a recognized risk factor in pathogenesis of gastric disease, such as chronic atrophic gastritis, gastric ulcer, and GC. Chaturvedi et al showed that Hp activation of spermine oxidase (SMOX) in gastric epithelial cells results in OS (26). OS causes damage to biological structures such as proteins, carbohydrates, lipids, and nucleic acids (27). DNA damage promotes activation of the epidermal growth factor receptor (EFGR) signaling pathway and mutations of tumor-suppressor genes, such as p53 and calcium/calmodulin-dependent serine protein kinase (CASK) (28,29), which are critical initial events in gastric carcinogenesis. Additionally, Hp infection also induces changes in expression of miRs in GC. For example, miR-328 is a well-studied miR involved in Hp-induced GC. Ishimoto et al demonstrated that miR-328 is downregulated in Hp-infected gastritis (30). Analysis of expression of miR-328 in GC, yielded a result similar to that obtained in gastritis. A low level of miR-328 activates CD44, a target of miR-328, to promote GC stem cell differentiation (4). Effect of Hp infection on miR-328 expression could be mimicked by H2O2. Therefore, miR-328 may be a specific miR that triggers development of Hp-induced GC by ROS. However, these researchers did not provide an explanation for downregulation of miR-328 in Hp-infected GC. Indeed, previous studies have demonstrated that Hp induced expression miR-210 by regulating methylation of miR-210, thereby increasing proliferation of gastric epithelial cells through suppressing oncoprotein 18 or metablastin (STMN1) and dimethyladenosine transferase 1 (DIMT1) (31). Therefore, Hp infection may alter expression of miRs through ROS-mediated methylation of gene promoter region. Hp-induced OS, which interferes with methylation of miRs, may account for part of the mechanism underlying the initiation of GC. In studies of effect of radiation on GC cells, radiation, which acts similarly to OS, also leads to production of ROS (32).

In conclusion, hypoxia, Hp infection, and radiation are capable of triggering cellular OS, and excessive ROS lead to the inhibition of anti-oncogenes such as p53, and the activation of tumorigenesis-associated signaling pathways, which contribute to occurrence and development of GC (Fig. 1).

3. OS modulates expression of miRs

Since the first miR, lin-4 (3), was discovered, an increasing number of studies have focused on these small non-coding RNAs. miRs regulate gene expression by completely or incompletely binding to 3′ untranslated region (3′-UTR) of target messenger RNAs. miRs are demonstrated to modulate expression of target genes. However, the crucial factor affecting expression of miRs has not been identified. OS can influence gene expression by regulating their methylation status of genes (33,34). Whether OS can affect the expression of miRNAs remains an open question.

As mentioned previously, under OS, ROS are produced and play a key role under pathological conditions. Hydrogen peroxide is widely used in experiments to simulate the effect of OS in cell lines and results in the abnormal expression of miRs in numerous diseases, including vitiligo, idiopathic pulmonary fibrosis (IPF), and most cancers. ROS induced miR-25 has been shown to enhance degeneration of melanocytes in vitiligo (35). Elevated levels of miR-9-5p has been confirmed to be influenced by ROS both in vivo and in vitro. In specimens of patients with IPF, expression of miR-9-5p has been associated with ROS and is upregulated in human fetal lung fibroblasts treated with hydrogen peroxide (36). Independently, endoplasmic reticulum (ER) stress can be caused by hypoxia, failure of protein maturation, or degradation (37). Induced by ER stress, expression of miR-17, -34a, -96, and -125b and the consequent reduction in the translation of proapoptotic caspase-2 (38). Additionally, exposure of cells to arsenic triggers upregulation of miR-663, miR-222, and miR-638 (39).

ROS exhibit their characteristics in intervening in expression of miRs in cancers. Similarly, placing cells under hypoxic condition is a common way to test the effect of OS on cancer cell lines. In breast and prostate cancer cells, miR-196b could be restrained by hypoxia (40). In bladder cancer cells, hypoxia could induce expression of miR-145 (41). Additionally, in response to ionizing radiation, ROS were stated to upregulate miR-193a-3p and miR-30e (42,43). miR-193a-3p has been demonstrated to lead to apoptosis by targeting Mcl-1, and miR-30e promote glioma cell invasion through EGFR stabilization by directly targeting Casitas-B-lineage lymphoma (Cbl)-b.

In GC, miR-382 was upregulated by hypoxia and participate in hypoxia-induced angiogenesis of GC (44). As discussed previously, Hp infection alters expression of miRs, such as miR-328 and miR-210, through ROS. Moreover, our team and other researchers focused on activities of miR-21 in GC and concluded that ROS give rise to progression of GC through dysfunction of miR-21 (45,46). Both programmed cell death protein 4 (PDCD4) and phosphatase and tension homolog deleted on chromosome 10 (PTEN) have been confirmed to be targets of miR-21. The silencing of miR-21 with its inhibitor AC1 MMYR2 reverse the effect of EMT accompanied by suppression of proliferation and invasion of GC (47). We also sought to identify other miRs affected by ROS (O2•−, OH); but few studies have examined the relationship of O2•− and OH with miRs because H2O2 is easy to use and detect in experiments. However, we found that enzymes catalyzing O2•− and OH are positively correlated with the expression of some miRs, such as miR-200b (48). In fact, the role of OS in regulating the expression of miRs has been widely studied. Selected miRs responsive to OS are shown in Table I. In summary, under OS, product of OS in cells mediate expression of miRs by different mechanisms and participate in progression of various types of diseases, including GC. The detailed mechanism will be discussed in part 5.

Table I

OS regulates expression of miRs.

Table I

OS regulates expression of miRs.

ItemsmiRsExpressionInduced byRefs.
Gastric cancermiR-370DownregulatedH. pylori(49)
miR-203DownregulatedH. pylori(28)
miR-328DownregulatedH. pylori, H2O2(4,50)
Lung fibrosismiR-9-5pUpregulated H2O2(36)
VitiligomiR-25Upregulated H2O2(35)
Mesenchymal stem cellsmiR-181aUpregulated H2O2(51)
Endothelial innate immune responsemiR-92aUpregulated H2O2(52)
MELAS syndrome microRNA-9/9*Upregulated H2O2(53)
Cardiac muscle, vascular smooth muscle cellsmiR-30 familyDownregulated H2O2(54)
GliomamiR-193a-3pUpregulatedRadiation(42)
miR-30eUpregulatedRadiation(43)
Human bronchial epithelial cellsmiR-15b/16-2UpregulatedRadiation(55)
Nasopharyngeal carcinomamiR-504UpregulatedRadiation(56)
Bladder cancermiR-769-3pDownregulatedHypoxia(57)
miR-145UpregulatedHypoxia(41)
Breast and prostate cancermiRNA-196bDownregulatedHypoxia(40)

4. Alteration of miRs participates in tumorigenesis of GC

It is well known that miRs are differentially expressed in different types of cancers and are implicated in processes of carcinogenesis, such as cell proliferation, angiogenesis, invasion, and metastasis. miRs have been identified to predict prognosis of GC patients. For example, selective overexpression of miR-25 in advanced GC indicated poor survival (58) and downregulation of miR-451 was associated with worse prognosis of GC patients (59). miRs may regulate the cell cycle through direct interaction with key regulators, such as cyclin, cyclin-dependent kinase (CDK), and cyclin-dependent kinase inhibitor (CDI). Specifically, cyclins D1 and E1 are targets of miR-16, and low levels of miR-16 contribute to development of colon cancer (60). In leukemia, CDK2 has been found to be a target gene of miR-638, and depression of miR-638 was found to be crucial for differentiation and proliferation of leukemia cells (61). With regard to miR-related cancer invasion and metastasis, miR-367 has been shown to promote invasion by downregulating Smad7 and to stimulate the EMT through activation of the transforming growth factor-β (TGF-β) signaling pathway (62). Moreover, in chondrosarcoma, miR-200b directly repress VEGF to inhibit angiogenesis (63). In cancers, most decreased miRs show activities as tumor-suppressor gene, and miR mimics prevent development of cancer. Conversely, high levels of miRs mainly act as oncogene and promote tumorigenesis. Following this review of role of miRs in cancer, we will concentrate our discussion on roles of miRs in GC.

In GC, numerous miRNAs have been identified, and dysfunction of these miRs promotes progression of GC. Expression of miRs has been found to be either increased or decreased in both GC tissues and cell lines. For example, miR-133b/a-3p has been ascertained to be reduced in primary GC tissues and two GC cell lines, namely SGC7901 and MNK45 (64). Using TargetScan database and luciferase assay, myeloid cell leukemia 1 (Mcl-1) and BCL2-like 1 (Bcl-xL) were shown to be targets of miR-133b/a-3p. Furthermore, using mouse tumor-bearing model and cell lines, miR-133b/a-3p has been ascertained to inhibit tumor growth. Similarly, miR-940 has been shown to promote tumor cell invasion and metastasis by interacting with zinc finger transcription factor 24 (ZNF24) in GC (65); miR-130a and miR-495 have been confirmed to increase cell proliferation and angiogenesis by targeting runt-related transcription factor 3 (RUNX3) in GC (66); miR-544a could induce the EMT by sensitizing Wnt signaling pathway in GC (67); and a lower miR-100 level is associated with lymphatic metastasis by targeting zinc finger and BTB domain containing 7A (ZBTB7A) (68). In fact, miRs have been widely investigated in GC. Studies of miRs in GC almost follow the methods described above. A list of miRs in GC, including their expression types, targets, and functions in gastric cancer is presented in Table II. Similar to role of miRs in other types of cancers, increased levels of miRs in GC act as oncogenes and reduced levels of miRs act as tumor suppressor genes. In general, molecules downstream of these miR targets are key factors involved in the signaling pathways in GC. For example, miR-141 is capable of regulating EGFR2 signaling pathway to affect proliferation of GC cells (69), and miR-495 can influence migration and invasion by action of phosphatase on regenerating liver-3 (PRL-3) (70); and Kip1 (p27), acting as a CDK, is the direct target of miR-196a, and a lower level of Kip1 due to inhibition by miR-196a leads to the proliferation of GC cells (71).

Table II

Alteration, function, and target(s) of some miRs in gastric cancer.

Table II

Alteration, function, and target(s) of some miRs in gastric cancer.

miRsExpressionTarget (s)FunctionRefs.
miR-7DownregulatedRELA, FOSInhibition of proliferation(72)
miR-100DownregulatedZBTB7AInhibition of proliferation and metastasis(68)
miR-141DownregulatedEGFR2Inhibition of proliferation(69)
miR-182DownregulatedCREB1Inhibition of proliferation(73,74)
miR-100DownregulatedHS3ST2Inhibition of proliferation(75)
miR-409-3pDownregulatedRDX, PHF10Inhibition of proliferation, invasion, and metastasis(76,77)
miR-124DownregulatedSPHK1Inhibition of proliferation(78)
miR-495/551aDownregulatedPRL-3Inhibition of migration and invasion(70)
miR-625DownregulatedILKInhibition of invasion and metastasis(79)
miR-145DownregulatedCDH2Inhibition of invasion, metastasis, and angiogenesis(80)
miR-874DownregulatedSTAT3Inhibition of angiogenesis(81)
miR-21UpregulatedRECK, PTEN, PDCD4, TPM1, MaspinPromotion of proliferation and metastasis(8284)
miR-301aUpregulatedRUNX3Promotion of proliferation and metastasis(85)
miR-196aUpregulatedKIP1Promotion of proliferation(71)
miR-296-5pUpregulatedCDX1Promotion of proliferation(86)
miR-223UpregulatedEPB41L3Promotion of invasion and metastasis(87)

Few studies have evaluated screening or diagnostic value of miRs in gastric juice and serum as a noninvasive method for GC treatment. The miR-21 and miR-106a levels in gastric juice were lower in GC than in benign gastric diseases, and showed a specificity of 0.969 and 0.871, respectively, for the identification of GC compared with benign gastric diseases (88). In GC, serum level of miR-203 is significantly lower compared with the control. Similar to its value in GC tissue, lower serum levels of miR-203 reflect an advanced clinical stage and poor overall survival, which is consistent with well-studied activity of miR-203 in inhibiting the EMT (89,90).

Despite the roles of these miRs, few studies have compared levels of miR in both tissue and serum from time of occurrence of morbidity to time of death of GC patients. This question is challenging for several reasons, such as the fact that we may not know when GC begins and cannot obtain adequate tissue samples once tumor progresses. By studying changes in the levels of miRs in body fluids, it may be possible for us to identify specific markers for GC.

5. miRs serve as a link between OS and GC

As discussed above, product of OS is involved in development of GC, and production of OS thus leads to an abnormal expression profile of miRs in diseases including cancers. Importantly, miRs can either promote or suppress tumorigenesis in GC. Whether miRs may serve as a link between OS and GC remains unclear.

To the best of our knowledge, epigenetic factors play a crucial role in carcinogenesis during OS. ROS contribute to methylation of miR genes, subsequently resulting in development of cancer by influencing expression of miRs, such as miR-145-5p, miR-362-3p, miR-329, miR-199, and miR-125 (9193). Frequently, ROS increase methylation of miRs, which play roles of tumor suppressor genes. For example, the hypermethylation of miR-199a and miR-125b induced by ROS is modulated by DNA methyltransferase (DNMT) 1, which downregulated these two miRs in an ovarian cancer cell line (93).

In Hp-induced GC, a decreased expression of miR-328 is mediated by ROS (4). Expression of miR-210 and miR-149 is thought to be modulated by methylation in Hp-positive human gastric biopsies (31,94). Hypoxia also leads to alteration of miR expression in GC. For example, miR-495 is upregulated under hypoxic conditions in two gastric cell lines, SNU5 and SNU484 (66), and methylation of corresponding promoter region contributes to dysregulation of miR-495 in GC. Following inhibition by DNMT1 and Zeste homolog 2 (EZH2), a repressed level of miR-200b/a/429 leads to progression of GC (95). The methylation-mediated silencing of miR-9 family in a GC cell line and the patient specimens was confirmed by combined bisulfate restriction analysis (COBRA) (96). Importantly, treating cells with 5-aza-2′-deoxycytidine (5-aza-CdR) reverses this action. Similarly, methylation of miR-137 gradually increases in normal gastric tissue, chronic gastritis, and GC tissue (97), and treatment of GC cell lines with 5-aza-CdR and 4-phenylbutyric acid (PBA) activates previously silenced miR-512-5p, causing its raised levels to induce cell apoptosis (98). Collectively, miRs are frequently dysregulated in GC (Table III). Interestingly, levels of most miRs are decreased by hypomethylation, but only two miRs, namely miR-196b and miR-127, have been identified to be elevated by hypomethylation in GC (99,100).

Table III

Expression of miRs is modulated by epigenetic factors in GC.

Table III

Expression of miRs is modulated by epigenetic factors in GC.

miRsExpressionMethylation statusModulated byRefs.
miR-200b/a/429Downregulated HypermethylationDNMT1/EZH2(95)
let-7DownregulatedHypermethylation, H3K27 trimethylationDNMT3B/EZH2(101)
miR-99a/449aDownregulatedH3K27 trimethylationEZH2(102)
miR-124a-1/2/3Downregulated HypermethylationNAa(100)
miR-139Downregulated HypermethylationNA(103)
miR-490-3pDownregulatedHypermethylation, H3K4 trimethylationNA(104)
miR-335Downregulated HypermethylationNA(105)
miR-34b/cDownregulated HypermethylationNA(106)
miR-200bDownregulated HypermethylationNA(107)
miR-149Downregulated HypermethylationNA(94)
miR-196bUpregulated HypomethylationNA(99)
miR-127UpregulatedDemethylationNA(100)

a NA, not available.

Actually, ROS-mediated deregulation of DNA methylation is quite an important aspect of biological role of ROS. ROS-mediated elevation of 8-OHdG in CpG island is one of the major mechanism of ROS-mediated hypomethylation, however, some form of ROS induced DNA damage also stimulate DNA methylation of specific loci. DNA hypermethylation of promoter region of genes is known to result in gene silencing, whereas hypomethylation contributes to increased gene expression. In GC, ROS induce expression of HIF-1α (19), which increases expression of DNMT enzymes DNMT1 and DNMT3B, resulting in gene silencing through gene hypermethylation (108). Interestingly, HIF-1α can also result in hypomethylation of genes by regulating MAT1A/MAT2A switch (109). By contrast, expression of miRs could also be modulated by histone deacetylases (HDACs). For example, miR-466 h-5p is elevated when histone deacetylation is inhibited by glucose deprivation-induced OS (110).

In conclusion, this review infers that in GC, ROS lead to changes in DNMT, which results in methylation changes in promoter region of miR genes that play roles in development of GC through inhibition of their targets and related signaling pathways (Fig. 2).

6. Conclusion

We summarized changes of OS as well as miRs and analyzed their function in gastric carcinogenesis. We conclude that ROS and parts of miRs exhibit overlapped characters in gastric carcinogenesis. Given that ROS mediate expression of miRs through epigenetic mechanism, we predict miRs serve as a bridge between ROS and gastric cancer. ROS are the driving force for gastric carcinogenesis. In both human and mouse gastric cancer models, ROS triggered crucial signal pathways and vital molecules such as Wnt, ERK, P53, and miRs to induce initiation and development of GC. In Hp-positive GC patients as well as Hp evoked mouse model of GC, ROS are also confirmed dysfunctional. While, we must accept that in treatment of GC, most drugs function through ROS-activated apoptosis of tumor cells, and some researchers found antioxidant can promote progress of cancer cells. Activities of ROS levels were dependent on cancer cells. How to define the threshold of ROS may be an important approach to identify their function in cancer cells. miRs are hot topics in cancer research. Diverse miRs change in GC making them hard to be specific targets in treatment of GC. However, detection of miRs in body fluid and tissues will be a treatment reaction or prognostic factor. miR expression profiling studies make it possible for us to probe miR change in different tissues or body fluid. Despite the encouraging prospect, we are still faced with many difficulties in the fields of OS-induced miRs in GC, tumor-related miRs, signaling pathway network between OS and gastric cancer and interaction between them. Expression of miRs presents tissue, time, and space specificity and can be affected by a variety of factors, which makes it hard to identify a consistent miRNA signature with high specificity for diagnosis and prognosis purpose. Furthermore, miRs exert a limited role in complex gene regulation network and modulate expression of multiple genes, suggesting complexity of miR function and both the potentially limited and undeliberate effect of therapy targeted to a limited number of miRs.

Acknowledgements

This study was supported by Natural Science Foundation of Shaanxi Province (no. 99SM50), National Natural Science Foundation of China (no. 81171288) for the research of oxidative stress and depression, and the Fundamental Research Funds for the Central Universities (no. 0601-08143036).

Abbreviations:

GC

gastric cancer

ROS

reactive oxygen species

OS

oxidative stress

miRs

microRNAs

8-OHdG

8-hydroxy-2′-deoxyguanosine

hOGG1

human oxoguanine glycosylase 1

XOD

xanthine oxidase

O2•-

superoxide

OH

hydroxide

H2O2

hydrogen peroxide

MAPK

mitogen-activated protein kinases

HIF-1α

hypoxia-inducible factor 1α

Cav-1

caveolin-1

EMT

epithelial-mesenchymal transition

VEGF

vascular endothelial growth factor

Akt

protein kinase-B

mTOR

mammalian target of rapamycin

NRF2

NF-E2-related factor

ARE

antioxidant response element

NF-κB

nuclear factor-κB

Hp

Helicobacter pylori

SMOX

spermine oxidase

EFGR

epidermal growth factor receptor

CASK

calmodulin-dependent serine protein kinase

STMN1

oncoprotein 18 or metablastin

DIMT1

demethyladenosine transferease 1

3′-UTR

3′ untranslated region

IPF

idiopathic pulmonary fibrosis

ER

endoplasmic reticulum

Cbl

Casitas-B-lineage lymphoma

PDCD4

programmed cell death protein 4

PTEN

phosphatase and tension homolog deleted on chromosome 10

CDK

cyclin-dependent kinase

CDI

cyclin-dependent kinase inhibitor

TGF-β

transforming growth factor-β

Mcl-1

myeloid cell leukemia 1

Bcl-xL

BCL2-like 1

ZNF24

zinc finger transcription factor 24

RUNX3

runt-related transcription factor 3

ZBTB7A

zinc finger and BTB domain containing 7A

PRL-3

phosphatase of regenerating liver-3

p27

Kip1

RELA

v-rel avian reticuloendotheliosis viral oncogene homolog A

FOS

FBJ osteosarcoma oncogene

CREB1

cAMP responsive element binding protein 1

HS3ST2

heparan sulfate (glucosamine) 3-O-sulfotransferase 2

RDX

radixin

PHF10

PHD finger protein 10

SPHK1

sphingosine kinase 1

ILK

integrin-linked kinase

CDH2

cadherin 2

STAT3

signal transducer and activator of transcription 3

RECK

reversion-inducing-cysteine-rich protein with kazal motifs

TPM1

tropomyosin 1 (α)

CDX1

caudal type homeobox 1

EPB41 L3

erythrocyte membrane protein band 4.1-like 3

DNMT

DNA methyltransferase

EZH2

zeste homolog 2

COBRA

combined bisulfiterestriction analysis

H3K27

histone H3 lysine 27

H3K4

histone 3 lysine 4

HDACs

histone deacetylases

References

1 

Riquelme I, Saavedra K, Espinoza JA, Weber H, García P, Nervi B, Garrido M, Corvalán AH, Roa JC and Bizama C: Molecular classification of gastric cancer: Towards a pathway-driven targeted therapy. Oncotarget. 6:24750–24779. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Wei YC, Zhou FL, He DL, Bai JR, Ding H, Wang XY and Nan KJ: Oxidative stress in depressive patients with gastric adenocarcinoma. Int J Neuropsychopharmacol. 12:1089–1096. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Chaturvedi R, de Sablet T, Asim M, Piazuelo MB, Barry DP, Verriere TG, Sierra JC, Hardbower DM, Delgado AG, Schneider BG, et al: Increased Helicobacter pylori-associated gastric cancer risk in the Andean region of Colombia is mediated by spermine oxidase. Oncogene. 34:3429–3440. 2015. View Article : Google Scholar :

4 

Ishimoto T, Sugihara H, Watanabe M, Sawayama H, Iwatsuki M, Baba Y, Okabe H, Hidaka K, Yokoyama N, Miyake K, et al: Macrophage-derived reactive oxygen species suppress miR-328 targeting CD44 in cancer cells and promote redox adaptation. Carcinogenesis. 35:1003–1011. 2014. View Article : Google Scholar

5 

Benfey PN: Molecular biology: microRNA is here to stay. Nature. 425:244–245. 2003. View Article : Google Scholar : PubMed/NCBI

6 

Song JH and Meltzer SJ: MicroRNAs in pathogenesis, diagnosis, and treatment of gastroesophageal cancers. Gastroenterology. 143:35–47.e2. 2012. View Article : Google Scholar : PubMed/NCBI

7 

Wang L, Tang ZP, Zhao W, Cong BH, Lu JQ, Tang XL, Li XH, Zhu XY and Ni X: MiR-22/Sp-1 links estrogens with the up-regulation of cystathionine γ-lyase in myocardium, which contributes to estrogenic cardioprotection against oxidative stress. Endocrinology. 156:2124–2137. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Xiao Y, Yan W, Lu L, Wang Y, Lu W, Cao Y and Cai W: p38/p53/miR-200a-3p feedback loop promotes oxidative stress-mediated liver cell death. Cell Cycle. 14:1548–1558. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Xu L, Ziegelbauer J, Wang R, Wu WW, Shen RF, Juhl H, Zhang Y and Rosenberg A: Distinct profiles for mitochondrial t-RNAs and small nucleolar RNAs in locally invasive and metastatic colorectal cancer. Clin Cancer Res. 22:773–784. 2016. View Article : Google Scholar

10 

Maya-Mendoza A, Ostrakova J, Kosar M, Hall A, Duskova P, Mistrik M, Merchut-Maya JM, Hodny Z, Bartkova J, Christensen C, et al: Myc and Ras oncogenes engage different energy metabolism programs and evoke distinct patterns of oxidative and DNA replication stress. Mol Oncol. 9:601–616. 2015. View Article : Google Scholar

11 

Zhou F, Shen Q and Claret FX: Novel roles of reactive oxygen species in the pathogenesis of acute myeloid leukemia. J Leukoc Biol. 94:423–429. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Jeon SM, Chandel NS and Hay N: AMPK regulates NADPH homeostasis to promote tumour cell survival during energy stress. Nature. 485:661–665. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Farías JG, Herrera EA, Carrasco-Pozo C, Sotomayor-Zárate R, Cruz G, Morales P and Castillo RL: Pharmacological models and approaches for pathophysiological conditions associated with hypoxia and oxidative stress. Pharmacol Ther. 158:1–23. 2016. View Article : Google Scholar

14 

Dolado I, Swat A, Ajenjo N, De Vita G, Cuadrado A and Nebreda AR: p38alpha MAP kinase as a sensor of reactive oxygen species in tumorigenesis. Cancer Cell. 11:191–205. 2007. View Article : Google Scholar : PubMed/NCBI

15 

Abu-Alainin W, Gana T, Liloglou T, Olayanju A, Barrera LN, Ferguson R, Campbell F, Andrews T, Goldring C, Kitteringham N, et al: UHRF1 regulation of the Keap1-Nrf2 pathway in pancreatic cancer contributes to oncogenesis. J Pathol. 238:423–433. 2016. View Article : Google Scholar

16 

Mashimo M, Nishikawa M, Higuchi K, Hirose M, Wei Q, Haque A, Sasaki E, Shiba M, Tominaga K, Watanabe T, et al: Production of reactive oxygen species in peripheral blood is increased in individuals with Helicobacter pylori infection and decreased after its eradication. Helicobacter. 11:266–271. 2006. View Article : Google Scholar : PubMed/NCBI

17 

Mikuni T and Tatsuta M: Production of hydroxyl free radical in the xanthine oxidase system with addition of 1-methyl-3-nitro-1-nitrosoguanidine. Free Radic Res. 36:641–647. 2002. View Article : Google Scholar : PubMed/NCBI

18 

Tatsuta M, Iishi H, Baba M, Mikuni T, Narahara H, Uedo N and Yano H: Suppression by iron chelator phenanthroline of sodium chloride-enhanced gastric carcinogenesis induced by N-methyl-N′-nitro-N-nitrosoguanidine in Wistar rats. Cancer Lett. 191:9–16. 2003. View Article : Google Scholar : PubMed/NCBI

19 

Park JH, Kim TY, Jong HS, Kim TY, Chun YS, Park JW, Lee CT, Jung HC, Kim NK and Bang YJ: Gastric epithelial reactive oxygen species prevent normoxic degradation of hypoxia-inducible factor-1alpha in gastric cancer cells. Clin Cancer Res. 9:433–440. 2003.PubMed/NCBI

20 

Kannan A, Krishnan A, Ali M, Subramaniam S, Halagowder D and Sivasithamparam ND: Caveolin-1 promotes gastric cancer progression by up-regulating epithelial to mesenchymal transition by crosstalk of signalling mechanisms under hypoxic condition. Eur J Cancer. 50:204–215. 2014. View Article : Google Scholar

21 

Rath S, Das L, Kokate SB, Pratheek BM, Chattopadhyay S, Goswami C, Chattopadhyay R, Crowe SE and Bhattacharyya A: Regulation of Noxa-mediated apoptosis in Helicobacter pylori-infected gastric epithelial cells. FASEB J. 29:796–806. 2015. View Article : Google Scholar

22 

Hao W, Yuan X, Yu L, Gao C, Sun X, Wang D and Zheng Q: Licochalcone A-induced human gastric cancer BGC-823 cells apoptosis by regulating ROS-mediated MAPKs and PI3K/AKT signaling pathways. Sci Rep. 5:103362015. View Article : Google Scholar : PubMed/NCBI

23 

Chen JJ, Huang WC and Chen CC: Transcriptional regulation of cyclooxygenase-2 in response to proteasome inhibitors involves reactive oxygen species-mediated signaling pathway and recruitment of CCAAT/enhancer-binding protein delta and CREB-binding protein. Mol Biol Cell. 16:5579–5591. 2005. View Article : Google Scholar : PubMed/NCBI

24 

Leone A, Roca MS, Ciardiello C, Terranova-Barberio M, Vitagliano C, Ciliberto G, Mancini R, Di Gennaro E, Bruzzese F and Budillon A: Vorinostat synergizes with EGFR inhibitors in NSCLC cells by increasing ROS via up-regulation of the major mitochondrial porin VDAC1 and modulation of the c-Myc-NRF2-KEAP1 pathway. Free Radic Biol Med. 89:287–299. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Yuan X, Zhou Y, Wang W, Li J, Xie G, Zhao Y, Xu D and Shen L: Activation of TLR4 signaling promotes gastric cancer progression by inducing mitochondrial ROS production. Cell Death Dis. 4:e7942013. View Article : Google Scholar : PubMed/NCBI

26 

Chaturvedi R, Asim M, Piazuelo MB, Yan F, Barry DP, Sierra JC, Delgado AG, Hill S, Casero RA Jr, Bravo LE, et al: Activation of EGFR and ERBB2 by Helicobacter pylori results in survival of gastric epithelial cells with DNA damage. Gastroenterology. 146:1739–1751. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Ferreira AG, Scherer EB, da Cunha AA, Manfredini V, Biancini GB, Vanzin CS, Vargas CR and Wyse AT: Hyper-prolinemia induces DNA, protein and lipid damage in blood of rats: Antioxidant protection. Int J Biochem Cell Biol. 54:20–25. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Zhou X, Xu G, Yin C, Jin W and Zhang G: Down-regulation of miR-203 induced by Helicobacter pylori infection promotes the proliferation and invasion of gastric cancer by targeting CASK. Oncotarget. 5:11631–11640. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Morgan C, Jenkins GJ, Ashton T, Griffiths AP, Baxter JN, Parry EM and Parry JM: Detection of p53 mutations in precancerous gastric tissue. Br J Cancer. 89:1314–1319. 2003. View Article : Google Scholar : PubMed/NCBI

30 

Ishimoto T, Izumi D, Watanabe M, Yoshida N, Hidaka K, Miyake K, Sugihara H, Sawayama H, Imamura Y, Iwatsuki M, et al: Chronic inflammation with Helicobacter pylori infection is implicated in CD44 overexpression through miR-328 suppression in the gastric mucosa. J Gastroenterol. 50:751–757. 2015. View Article : Google Scholar

31 

Kiga K, Mimuro H, Suzuki M, Shinozaki-Ushiku A, Kobayashi T, Sanada T, Kim M, Ogawa M, Iwasaki YW, Kayo H, et al: Epigenetic silencing of miR-210 increases the proliferation of gastric epithelium during chronic Helicobacter pylori infection. Nat Commun. 5:44972014. View Article : Google Scholar : PubMed/NCBI

32 

Bhattacharyya A, Chattopadhyay R, Mitra S and Crowe SE: Oxidative stress: An essential factor in the pathogenesis of gastrointestinal mucosal diseases. Physiol Rev. 94:329–354. 2014. View Article : Google Scholar : PubMed/NCBI

33 

Niu Y, DesMarais TL, Tong Z, Yao Y and Costa M: Oxidative stress alters global histone modification and DNA methylation. Free Radic Biol Med. 82:22–28. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Salminen A, Haapasalo A, Kauppinen A, Kaarniranta K, Soininen H and Hiltunen M: Impaired mitochondrial energy metabolism in Alzheimer's disease: Impact on pathogenesis via disturbed epigenetic regulation of chromatin landscape. Prog Neurobiol. 131:1–20. 2015. View Article : Google Scholar : PubMed/NCBI

35 

Shi Q, Zhang W, Guo S, Jian Z, Li S, Li K, Ge R, Dai W, Wang G, Gao T, et al: Oxidative stress-induced overexpression of miR-25: The mechanism underlying the degeneration of melanocytes in vitiligo. Cell Death Differ. 23:496–508. 2016. View Article : Google Scholar

36 

Fierro-Fernández M, Busnadiego Ó, Sandoval P, Espinosa-Díez C, Blanco-Ruiz E, Rodríguez M, Pian H, Ramos R, López-Cabrera M, García-Bermejo ML, et al: miR-9-5p suppresses pro-fibrogenic transformation of fibroblasts and prevents organ fibrosis by targeting NOX4 and TGFBR2. EMBO Rep. 16:1358–1377. 2015. View Article : Google Scholar : PubMed/NCBI

37 

Ando Y and Leung AK: Does an emergency visit to the ER make microRNAs stronger during stress? Mol Cell. 52:1–3. 2013. View Article : Google Scholar : PubMed/NCBI

38 

Upton JP, Wang L, Han D, Wang ES, Huskey NE, Lim L, Truitt M, McManus MT, Ruggero D, Goga A, et al: IRE1α cleaves select microRNAs during ER stress to derepress translation of proapoptotic Caspase-2. Science. 338:818–822. 2012. View Article : Google Scholar : PubMed/NCBI

39 

Sturchio E, Colombo T, Carucci N, Meconi C, Boccia P and Macino G: Molecular biomarkers in workers and population exposed to inorganic arsenic: Preliminary study in vitro. G Ital Med Lav Ergon. 34(Suppl): 678–681. 2012.(In Italian).

40 

Rebucci M, Sermeus A, Leonard E, Delaive E, Dieu M, Fransolet M, Arnould T and Michiels C: miRNA-196b inhibits cell proliferation and induces apoptosis in HepG2 cells by targeting IGF2BP1. Mol Cancer. 14:792015. View Article : Google Scholar : PubMed/NCBI

41 

Blick C, Ramachandran A, McCormick R, Wigfield S, Cranston D, Catto J and Harris AL: Identification of a hypoxia-regulated miRNA signature in bladder cancer and a role for miR-145 in hypoxia-dependent apoptosis. Br J Cancer. 113:634–644. 2015. View Article : Google Scholar : PubMed/NCBI

42 

Kwon JE, Kim BY, Kwak SY, Bae IH and Han YH: Ionizing radiation-inducible microRNA miR-193a-3p induces apoptosis by directly targeting Mcl-1. Apoptosis. 18:896–909. 2013. View Article : Google Scholar : PubMed/NCBI

43 

Kwak SY, Kim BY, Ahn HJ, Yoo JO, Kim J, Bae IH and Han YH: Ionizing radiation-inducible miR-30e promotes glioma cell invasion through EGFR stabilization by directly targeting CBL-B. FEBS J. 282:1512–1525. 2015. View Article : Google Scholar : PubMed/NCBI

44 

Seok JK, Lee SH, Kim MJ and Lee YM: MicroRNA-382 induced by HIF-1α is an angiogenic miR targeting the tumor suppressor phosphatase and tensin homolog. Nucleic Acids Res. 42:8062–8072. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Tu H, Sun H, Lin Y, Ding J, Nan K, Li Z, Shen Q and Wei Y: Oxidative stress upregulates PDCD4 expression in patients with gastric cancer via miR-21. Curr Pharm Des. 20:1917–1923. 2014. View Article : Google Scholar

46 

Polytarchou C, Iliopoulos D, Hatziapostolou M, Kottakis F, Maroulakou I, Struhl K and Tsichlis PN: Akt2 regulates all Akt isoforms and promotes resistance to hypoxia through induction of miR-21 upon oxygen deprivation. Cancer Res. 71:4720–4731. 2011. View Article : Google Scholar : PubMed/NCBI

47 

Shi Z, Zhang J, Qian X, Han L, Zhang K, Chen L, Liu J, Ren Y, Yang M, Zhang A, et al: AC1MMYR2, an inhibitor of dicer-mediated biogenesis of Oncomir miR-21, reverses epithelial-mesenchymal transition and suppresses tumor growth and progression. Cancer Res. 73:5519–5531. 2013. View Article : Google Scholar : PubMed/NCBI

48 

Gu H, Yu J, Dong D, Zhou Q, Wang J-Y, Fang S and Yang P: High glucose-repressed CITED2 expression through miR-200b triggers the unfolded protein response and endoplasmic reticulum stress. Diabetes. 65:149–163. 2016.

49 

Feng Y, Wang L, Zeng J, Shen L, Liang X, Yu H, Liu S, Liu Z, Sun Y, Li W, et al: FoxM1 is overexpressed in Helicobacter pylori-induced gastric carcinogenesis and is negatively regulated by miR-370. Mol Cancer Res. 11:834–844. 2013. View Article : Google Scholar : PubMed/NCBI

50 

Serguienko A, Grad I, Wennerstrøm AB, Meza-Zepeda LA, Thiede B, Stratford EW, Myklebost O and Munthe E: Metabolic reprogramming of metastatic breast cancer and melanoma by let-7a microRNA. Oncotarget. 6:2451–2465. 2015. View Article : Google Scholar : PubMed/NCBI

51 

Lee S, Yun I, Ham O, Lee SY, Lee CY, Park JH, Lee J, Seo HH, Choi E and Hwang KC: Suppression of miR-181a attenuates H2O2-induced death of mesenchymal stem cells by maintaining hexokinase II expression. Biol Res. 48:452015. View Article : Google Scholar

52 

Chen Z, Wen L, Martin M, Hsu CY, Fang L, Lin FM, Lin TY, Geary MJ, Geary GG, Zhao Y, et al: Oxidative stress activates endothelial innate immunity via sterol regulatory element binding protein 2 (SREBP2) transactivation of microRNA-92a. Circulation. 131:805–814. 2015. View Article : Google Scholar : PubMed/NCBI

53 

Meseguer S, Martínez-Zamora A, García-Arumí E, Andreu AL and Armengod ME: The ROS-sensitive microRNA-9/9* controls the expression of mitochondrial tRNA-modifying enzymes and is involved in the molecular mechanism of MELAS syndrome. Hum Mol Genet. 24:167–184. 2015. View Article : Google Scholar

54 

Chen M, Ma G, Yue Y, Wei Y, Li Q, Tong Z, Zhang L, Miao G and Zhang J: Downregulation of the miR-30 family microRNAs contributes to endoplasmic reticulum stress in cardiac muscle and vascular smooth muscle cells. Int J Cardiol. 173:65–73. 2014. View Article : Google Scholar : PubMed/NCBI

55 

Rahman M, Lovat F, Romano G, Calore F, Acunzo M, Bell EH and Nana-Sinkam P: miR-15b/16-2 regulates factors that promote p53 phosphorylation and augments the DNA damage response following radiation in the lung. J Biol Chem. 289:26406–26416. 2014. View Article : Google Scholar : PubMed/NCBI

56 

Zhao L, Tang M, Hu Z, Yan B, Pi W, Li Z, Zhang J, Zhang L, Jiang W, Li G, et al: miR-504 mediated down-regulation of nuclear respiratory factor 1 leads to radio-resistance in nasopharyngeal carcinoma. Oncotarget. 6:15995–16018. 2015. View Article : Google Scholar : PubMed/NCBI

57 

Luo EC, Chang YC, Sher YP, Huang WY, Chuang LL, Chiu YC, Tsai MH, Chuang EY and Lai LC: MicroRNA-769-3p downregulates NDRG1 and enhances apoptosis in MCF-7 cells during reoxygenation. Sci Rep. 4:59082014.

58 

Li BS, Zuo QF, Zhao YL, Xiao B, Zhuang Y, Mao XH, Wu C, Yang SM, Zeng H, Zou QM, et al: MicroRNA-25 promotes gastric cancer migration, invasion and proliferation by directly targeting transducer of ERBB2, 1 and correlates with poor survival. Oncogene. 34:2556–2565. 2015. View Article : Google Scholar

59 

Bandres E, Bitarte N, Arias F, Agorreta J, Fortes P, Agirre X, Zarate R, Diaz-Gonzalez JA, Ramirez N, Sola JJ, et al: microRNA-451 regulates macrophage migration inhibitory factor production and proliferation of gastrointestinal cancer cells. Clin Cancer Res. 15:2281–2290. 2009. View Article : Google Scholar : PubMed/NCBI

60 

Witalison EE, Cui X, Causey CP, Thompson PR and Hofseth LJ: Molecular targeting of protein arginine deiminases to suppress colitis and prevent colon cancer. Oncotarget. 6:36053–36062. 2015.PubMed/NCBI

61 

Lin Y, Li D, Liang Q, Liu S, Zuo X, Li L, Sun X, Li W, Guo M and Huang Z: miR-638 regulates differentiation and proliferation in leukemic cells by targeting cyclin-dependent kinase 2. J Biol Chem. 290:1818–1828. 2015. View Article : Google Scholar :

62 

Zhu Z, Xu Y, Zhao J, Liu Q, Feng W, Fan J and Wang P: miR-367 promotes epithelial-to-mesenchymal transition and invasion of pancreatic ductal adenocarcinoma cells by targeting the Smad7-TGF-β signalling pathway. Br J Cancer. 112:1367–1375. 2015. View Article : Google Scholar : PubMed/NCBI

63 

Liu GT, Chen HT, Tsou HK, Tan TW, Fong YC, Chen PC, Yang WH, Wang SW, Chen JC and Tang CH: CCL5 promotes VEGF-dependent angiogenesis by down-regulating miR-200b through PI3K/Akt signaling pathway in human chondrosarcoma cells. Oncotarget. 5:10718–10731. 2014. View Article : Google Scholar : PubMed/NCBI

64 

Liu Y, Zhang X, Zhang Y, Hu Z, Yang D, Wang C, Guo M and Cai Q: Identification of miRNomes in human stomach and gastric carcinoma reveals miR-133b/a-3p as therapeutic target for gastric cancer. Cancer Lett. 369:58–66. 2015. View Article : Google Scholar : PubMed/NCBI

65 

Liu X, Ge X, Zhang Z, Zhang X, Chang J, Wu Z, Tang W, Gan L, Sun M and Li J: MicroRNA-940 promotes tumor cell invasion and metastasis by downregulating ZNF24 in gastric cancer. Oncotarget. 6:25418–25428. 2015. View Article : Google Scholar : PubMed/NCBI

66 

Lee SH, Jung YD, Choi YS and Lee YM: Targeting of RUNX3 by miR-130a and miR-495 cooperatively increases cell proliferation and tumor angiogenesis in gastric cancer cells. Oncotarget. 6:33269–33278. 2015.PubMed/NCBI

67 

Yanaka Y, Muramatsu T, Uetake H, Kozaki K and Inazawa J: miR-544a induces epithelial-mesenchymal transition through the activation of WNT signaling pathway in gastric cancer. Carcinogenesis. 36:1363–1371. 2015. View Article : Google Scholar : PubMed/NCBI

68 

Shi DB, Wang YW, Xing AY, Gao JW, Zhang H, Guo XY and Gao P: C/EBPα-induced miR-100 expression suppresses tumor metastasis and growth by targeting ZBTB7A in gastric cancer. Cancer Lett. 369:376–385. 2015. View Article : Google Scholar : PubMed/NCBI

69 

Du Y, Xu Y, Ding L, Yao H, Yu H, Zhou T and Si J: Downregulation of miR-141 in gastric cancer and its involvement in cell growth. J Gastroenterol. 44:556–561. 2009. View Article : Google Scholar

70 

Li Z, Cao Y, Jie Z, Liu Y, Li Y, Li J, Zhu G, Liu Z, Tu Y, Peng G, et al: miR-495 and miR-551a inhibit the migration and invasion of human gastric cancer cells by directly interacting with PRL-3. Cancer Lett. 323:41–47. 2012. View Article : Google Scholar : PubMed/NCBI

71 

Sun M, Liu XH, Li JH, Yang JS, Zhang EB, Yin DD, Liu ZL, Zhou J, Ding Y, Li SQ, et al: MiR-196a is upregulated in gastric cancer and promotes cell proliferation by downregulating p27 (kip1). Mol Cancer Ther. 11:842–852. 2012. View Article : Google Scholar : PubMed/NCBI

72 

Zhao XD, Lu YY, Guo H, Xie HH, He LJ, Shen GF, Zhou JF, Li T, Hu SJ, Zhou L, et al: MicroRNA-7/NF-κB signaling regulatory feedback circuit regulates gastric carcinogenesis. J Cell Biol. 210:613–627. 2015. View Article : Google Scholar : PubMed/NCBI

73 

Kong WQ, Bai R, Liu T, Cai CL, Liu M, Li X and Tang H: MicroRNA-182 targets cAMP-responsive element-binding protein 1 and suppresses cell growth in human gastric adenocarcinoma. FEBS J. 279:1252–1260. 2012. View Article : Google Scholar : PubMed/NCBI

74 

Zheng L, Pu J, Qi T, Qi M, Li D, Xiang X, Huang K and Tong Q: miRNA-145 targets v-ets erythroblastosis virus E26 oncogene homolog 1 to suppress the invasion, metastasis, and angiogenesis of gastric cancer cells. Mol Cancer Res. 11:182–193. 2013. View Article : Google Scholar

75 

Yang G, Gong Y, Wang Q, Wang Y and Zhang X: The role of miR-100-mediated Notch pathway in apoptosis of gastric tumor cells. Cell Signal. 27:1087–1101. 2015. View Article : Google Scholar : PubMed/NCBI

76 

Zheng B, Liang L, Huang S, Zha R, Liu L, Jia D, Tian Q, Wang Q, Wang C, Long Z, et al: MicroRNA-409 suppresses tumour cell invasion and metastasis by directly targeting radixin in gastric cancers. Oncogene. 31:4509–4516. 2012. View Article : Google Scholar

77 

Li C, Nie H, Wang M, Su L, Li J, Yu B, Wei M, Ju J, Yu Y, Yan M, et al: MicroRNA-409-3p regulates cell proliferation and apoptosis by targeting PHF10 in gastric cancer. Cancer Lett. 320:189–197. 2012. View Article : Google Scholar : PubMed/NCBI

78 

Xia J, Wu Z, Yu C, He W, Zheng H, He Y, Jian W, Chen L, Zhang L and Li W: miR-124 inhibits cell proliferation in gastric cancer through down-regulation of SPHK1. J Pathol. 227:470–480. 2012. View Article : Google Scholar : PubMed/NCBI

79 

Wang M, Li C, Nie H, Lv X, Qu Y, Yu B, Su L, Li J, Chen X, Ju J, et al: Down-regulated miR-625 suppresses invasion and metastasis of gastric cancer by targeting ILK. FEBS Lett. 586:2382–2388. 2012. View Article : Google Scholar : PubMed/NCBI

80 

Gao P, Xing AY, Zhou GY, Zhang TG, Zhang JP, Gao C, Li H and Shi DB: The molecular mechanism of microRNA-145 to suppress invasion-metastasis cascade in gastric cancer. Oncogene. 32:491–501. 2013. View Article : Google Scholar

81 

Zhang X, Tang J, Zhi X, Xie K, Wang W, Li Z, Zhu Y, Yang L, Xu H and Xu Z: miR-874 functions as a tumor suppressor by inhibiting angiogenesis through STAT3/VEGF-A pathway in gastric cancer. Oncotarget. 6:1605–1617. 2015. View Article : Google Scholar : PubMed/NCBI

82 

Huang TH, Wu F, Loeb GB, Hsu R, Heidersbach A, Brincat A, Horiuchi D, Lebbink RJ, Mo YY, Goga A, et al: Up-regulation of miR-21 by HER2/neu signaling promotes cell invasion. J Biol Chem. 284:18515–18524. 2009. View Article : Google Scholar : PubMed/NCBI

83 

Si ML, Zhu S, Wu H, Lu Z, Wu F and Mo YY: miR-21-mediated tumor growth. Oncogene. 26:2799–2803. 2007. View Article : Google Scholar

84 

Zhu S, Wu H, Wu F, Nie D, Sheng S and Mo YY: MicroRNA-21 targets tumor suppressor genes in invasion and metastasis. Cell Res. 18:350–359. 2008. View Article : Google Scholar : PubMed/NCBI

85 

Wang M, Li C, Yu B, Su L, Li J, Ju J, Yu Y, Gu Q, Zhu Z and Liu B: Overexpressed miR-301a promotes cell proliferation and invasion by targeting RUNX3 in gastric cancer. J Gastroenterol. 48:1023–1033. 2013. View Article : Google Scholar : PubMed/NCBI

86 

Li T, Lu YY, Zhao XD, Guo HQ, Liu CH, Li H, Zhou L, Han YN, Wu KC, Nie YZ, et al: MicroRNA-296-5p increases proliferation in gastric cancer through repression of Caudal-related homeobox 1. Oncogene. 33:783–793. 2014. View Article : Google Scholar

87 

Li X, Zhang Y, Zhang H, Liu X, Gong T, Li M, Sun L, Ji G, Shi Y, Han Z, et al: miRNA-223 promotes gastric cancer invasion and metastasis by targeting tumor suppressor EPB41L3. Mol Cancer Res. 9:824–833. 2011. View Article : Google Scholar : PubMed/NCBI

88 

Cui L, Zhang X, Ye G, Zheng T, Song H, Deng H, Xiao B, Xia T, Yu X, Le Y, et al: Gastric juice microRNAs as potential biomarkers for the screening of gastric cancer. Cancer. 119:1618–1626. 2013. View Article : Google Scholar : PubMed/NCBI

89 

Imaoka H, Toiyama Y, Okigami M, Yasuda H, Saigusa S, Ohi M, Tanaka K, Inoue Y, Mohri Y and Kusunoki M: Circulating microRNA-203 predicts metastases, early recurrence, and poor prognosis in human gastric cancer. Gastric Cancer. 19:744–753. 2016. View Article : Google Scholar

90 

Taube JH, Malouf GG, Lu E, Sphyris N, Vijay V, Ramachandran PP, Ueno KR, Gaur S, Nicoloso MS, Rossi S, et al: Epigenetic silencing of microRNA-203 is required for EMT and cancer stem cell properties. Sci Rep. 3:26872013. View Article : Google Scholar : PubMed/NCBI

91 

Donzelli S, Mori F, Bellissimo T, Sacconi A, Casini B, Frixa T, Roscilli G, Aurisicchio L, Facciolo F, Pompili A, et al: Epigenetic silencing of miR-145-5p contributes to brain metastasis. Oncotarget. 6:35183–35201. 2015.PubMed/NCBI

92 

Kang H, Kim C, Lee H, Rho JG, Seo JW, Nam JW, Song WK, Nam SW, Kim W and Lee EK: Downregulation of microRNA-362-3p and microRNA-329 promotes tumor progression in human breast cancer. Cell Death Differ. 23:484–495. 2016. View Article : Google Scholar

93 

He J, Xu Q, Jing Y, Agani F, Qian X, Carpenter R, Li Q, Wang XR, Peiper SS, Lu Z, et al: Reactive oxygen species regulate ERBB2 and ERBB3 expression via miR-199a/125b and DNA methylation. EMBO Rep. 13:1116–1122. 2012. View Article : Google Scholar : PubMed/NCBI

94 

Li P, Shan JX, Chen XH, Zhang D, Su LP, Huang XY, Yu BQ, Zhi QM, Li CL, Wang YQ, et al: Epigenetic silencing of microRNA-149 in cancer-associated fibroblasts mediates prostaglandin E2/interleukin-6 signaling in the tumor microenvironment. Cell Res. 25:588–603. 2015. View Article : Google Scholar : PubMed/NCBI

95 

Ning X, Shi Z, Liu X, Zhang A, Han L, Jiang K, Kang C and Zhang Q: DNMT1 and EZH2 mediated methylation silences the microRNA-200b/a/429 gene and promotes tumor progression. Cancer Lett. 359:198–205. 2015. View Article : Google Scholar : PubMed/NCBI

96 

Tsai KW, Liao YL, Wu CW, Hu LY, Li SC, Chan WC, Ho MR, Lai CH, Kao HW, Fang WL, et al: Aberrant hypermethylation of miR-9 genes in gastric cancer. Epigenetics. 6:1189–1197. 2011. View Article : Google Scholar : PubMed/NCBI

97 

Steponaitiene R, Kupcinskas J, Langner C, Balaguer F, Venclauskas L, Pauzas H, Tamelis A, Skieceviciene J, Kupcinskas L, Malfertheiner P, et al: Epigenetic silencing of miR-137 is a frequent event in gastric carcinogenesis. Mol Carcinog. 55:376–386. 2016. View Article : Google Scholar

98 

Saito Y, Suzuki H, Tsugawa H, Nakagawa I, Matsuzaki J, Kanai Y and Hibi T: Chromatin remodeling at Alu repeats by epigenetic treatment activates silenced microRNA-512-5p with downregulation of Mcl-1 in human gastric cancer cells. Oncogene. 28:2738–2744. 2009. View Article : Google Scholar : PubMed/NCBI

99 

Tsai KW, Hu LY, Wu CW, Li SC, Lai CH, Kao HW, Fang WL and Lin WC: Epigenetic regulation of miR-196b expression in gastric cancer. Genes Chromosomes Cancer. 49:969–980. 2010. View Article : Google Scholar : PubMed/NCBI

100 

Ando T, Yoshida T, Enomoto S, Asada K, Tatematsu M, Ichinose M, Sugiyama T and Ushijima T: DNA methylation of microRNA genes in gastric mucosae of gastric cancer patients: Its possible involvement in the formation of epigenetic field defect. Int J Cancer. 124:2367–2374. 2009. View Article : Google Scholar : PubMed/NCBI

101 

Hayashi Y, Tsujii M, Wang J, Kondo J, Akasaka T, Jin Y, Li W, Nakamura T, Nishida T, Iijima H, et al: CagA mediates epigenetic regulation to attenuate let-7 expression in Helicobacter pylori-related carcinogenesis. Gut. 62:1536–1546. 2013. View Article : Google Scholar

102 

Zhang EB, Kong R, Yin DD, You LH, Sun M, Han L, Xu TP, Xia R, Yang JS, De W, et al: Long noncoding RNA ANRIL indicates a poor prognosis of gastric cancer and promotes tumor growth by epigenetically silencing of miR-99a/miR-449a. Oncotarget. 5:2276–2292. 2014. View Article : Google Scholar : PubMed/NCBI

103 

Bao W, Fu HJ, Xie QS, Wang L, Zhang R, Guo ZY, Zhao J, Meng YL, Ren XL, Wang T, et al: HER2 interacts with CD44 to up-regulate CXCR4 via epigenetic silencing of microRNA-139 in gastric cancer cells. Gastroenterology. 141:2076–2087.e6. 2011. View Article : Google Scholar : PubMed/NCBI

104 

Shen J, Xiao Z, Wu WK, Wang MH, To KF, Chen Y, Yang W, Li MS, Shin VY, Tong JH, et al: Epigenetic silencing of miR-490-3p reactivates the chromatin remodeler SMARCD1 to promote Helicobacter pylori-induced gastric carcinogenesis. Cancer Res. 75:754–765. 2015. View Article : Google Scholar

105 

Li Z, Li D, Zhang G, Xiong J, Jie Z, Cheng H, Cao Y, Jiang M, Lin L, Le Z, et al: Methylation-associated silencing of MicroRNA-335 contributes tumor cell invasion and migration by interacting with RASA1 in gastric cancer. Am J Cancer Res. 4:648–662. 2014.PubMed/NCBI

106 

Suzuki H, Yamamoto E, Nojima M, Kai M, Yamano HO, Yoshikawa K, Kimura T, Kudo T, Harada E, Sugai T, et al: Methylation-associated silencing of microRNA-34b/c in gastric cancer and its involvement in an epigenetic field defect. Carcinogenesis. 31:2066–2073. 2010. View Article : Google Scholar : PubMed/NCBI

107 

Kurashige J, Mima K, Sawada G, Takahashi Y, Eguchi H, Sugimachi K, Mori M, Yanagihara K, Yashiro M, Hirakawa K, et al: Epigenetic modulation and repression of miR-200b by cancer-associated fibroblasts contribute to cancer invasion and peritoneal dissemination in gastric cancer. Carcinogenesis. 36:133–141. 2015. View Article : Google Scholar

108 

Watson CJ, Collier P, Tea I, Neary R, Watson JA, Robinson C, Phelan D, Ledwidge MT, McDonald KM, McCann A, et al: Hypoxia-induced epigenetic modifications are associated with cardiac tissue fibrosis and the development of a myofibroblast-like phenotype. Hum Mol Genet. 23:2176–2188. 2014. View Article : Google Scholar

109 

Frau M, Feo F and Pascale RM: Pleiotropic effects of methionine adenosyltransferases deregulation as determinants of liver cancer progression and prognosis. J Hepatol. 59:830–841. 2013. View Article : Google Scholar : PubMed/NCBI

110 

Druz A, Betenbaugh M and Shiloach J: Glucose depletion activates mmu-miR-466h-5p expression through oxidative stress and inhibition of histone deacetylation. Nucleic Acids Res. 40:7291–7302. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2016
Volume 49 Issue 5

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Huang T, Wang-Johanning F, Zhou F, Kallon H and Wei Y: MicroRNAs serve as a bridge between oxidative stress and gastric cancer (Review). Int J Oncol 49: 1791-1800, 2016
APA
Huang, T., Wang-Johanning, F., Zhou, F., Kallon, H., & Wei, Y. (2016). MicroRNAs serve as a bridge between oxidative stress and gastric cancer (Review). International Journal of Oncology, 49, 1791-1800. https://doi.org/10.3892/ijo.2016.3686
MLA
Huang, T., Wang-Johanning, F., Zhou, F., Kallon, H., Wei, Y."MicroRNAs serve as a bridge between oxidative stress and gastric cancer (Review)". International Journal of Oncology 49.5 (2016): 1791-1800.
Chicago
Huang, T., Wang-Johanning, F., Zhou, F., Kallon, H., Wei, Y."MicroRNAs serve as a bridge between oxidative stress and gastric cancer (Review)". International Journal of Oncology 49, no. 5 (2016): 1791-1800. https://doi.org/10.3892/ijo.2016.3686