Topic Highlight Open Access
Copyright ©2014 Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. May 21, 2014; 20(19): 5694-5699
Published online May 21, 2014. doi: 10.3748/wjg.v20.i19.5694
Role of microRNAs in gastric cancer
Hideyuki Ishiguro, Masahiro Kimura, Hiromitsu Takeyama, Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan
Author contributions: Ishiguro H and Kimura M gathered the papers and performed the research; Ishiguro H and Takeyama H analyzed the data and wrote the paper.
Correspondence to: Hideyuki Ishiguro, MD, PhD, Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi Mizuho-cho Mizuho-ku, Nagoya 467-8601, Japan. h-ishi@med.nagoya-cu.ac.jp
Telephone: +81-52-8538226 Fax: +81-52-8423906
Received: October 26, 2013
Revised: December 11, 2013
Accepted: January 19, 2014
Published online: May 21, 2014

Abstract

Although gastric cancer (GC) is one of the leading causes of cancer-related death, major therapeutic advances have not been made, and patients with GC still face poor outcomes. The prognosis of GC also remains poor because the molecular mechanisms of GC progression are incompletely understood. MicroRNAs (miRNAs) are noncoding RNAs that are associated with gastric carcinogenesis. Studies investigating the regulation of gene expression by miRNAs have made considerable progress in recent years, and abnormalities in miRNA expression have been shown to be associated with the occurrence and progression of GC. miRNAs contribute to gastric carcinogenesis by altering the expression of oncogenes and tumor suppressors, affecting cell proliferation, apoptosis, motility, and invasion. Moreover, a number of miRNAs have been shown to be associated with tumor type, tumor stage, and patient survival and therefore may be developed as novel diagnostic or prognostic markers. In this review, we discuss the involvement of miRNAs in GC and the mechanisms through which they regulate gene expression and biological functions. Then, we review recent research on the involvement of miRNAs in GC prognosis, their potential use in chemotherapy, and their effects on Helicobacter pylori infections in GC. A greater understanding of the roles of miRNAs in gastric carcinogenesis could provide insights into the mechanisms of tumor development and could help to identify novel therapeutic targets.

Key Words: MicroRNA, Gastric cancer, Reverse transcription-polymerase chain reaction, Chemosensitivity, Helicobacter pylori, Circulating MicroRNA

Core tip: In this review, we discuss the involvement of miRNAs in gastric cancer (GC) and the mechanisms through which they regulate gene expression and biological functions. Then, we review recent research on the involvement of miRNAs in GC prognosis, their potential use in chemotherapy, and their effects on Helicobacter pylori infections in GC. A greater understanding of the roles of miRNAs in gastric carcinogenesis could provide insights into the mechanisms of tumor development and could help to identify novel therapeutic targets.



INTRODUCTION

Despite the decreasing incidence of gastric cancer (GC) in developed countries, GC remains the second leading cause of cancer-related deaths worldwide[1,2], with 700000 deaths attributed to this malignancy annually[3]. Therefore, the development of novel therapies to improve the prognosis of patients with GC is critical.

MicroRNAs (miRNAs) are a subset of small noncoding RNA molecules, typically 21-23 nucleotides in length, that are believed to regulate the expression of several genes[4]. Mature miRNAs are cleaved from 70- to 100-nucleotide hairpin pre-miRNA precursors[4]. The precursor is cleaved by cytoplasmic RNase III Dicer into a miRNA duplex[5-7]. One strand of the short-lived duplex is degraded, whereas the other strand serves as the mature miRNA[8]. Mature miRNAs associate with a cellular complex that is similar to the RNA-induced silencing complex that participates in RNA interference[9,10]. Recent studies have reported miRNA-mediated regulation of cell growth and apoptosis[11,12]. Moreover, the measurement of miRNA expression has shown that certain miRNAs are specifically involved in cancer[12-15]. In the context of GC, the number of publications investigating the relationship between miRNAs and GC has been increasing each year. miRNAs have the unique ability to negatively regulate gene expression, thereby resulting in changes in cell development, proliferation, and apoptosis[16]. These biological properties of miRNAs may provide the ability to regulate a variety of human diseases, including cancer[17]. According to recent findings, miRNAs may play important roles in human cancer by acting as potential oncogenes or tumor-suppressor genes[18-20].

In this review, we introduce and discuss the newest knowledge on the relationship between GC and miRNAs.

Aberrant expression of miRNAs in GC

Although more than 1000 miRNAs are thought to exist, no comprehensive analysis of miRNA expression in GC has been performed to date. Each miRNA modulates the expression of hundreds of genes, and we speculate that miRNAs act in a network-type fashion to mediate the expression of genes.

While several reports have published comprehensive expression analysis of miRNAs[21,22], more data, including expression profile analysis by high-throughput real-time reverse transcription-polymerase chain reaction (RT-PCR) or miRNA microarrays, are required. A summary of reported miRNA expression abnormalities in GC is presented in Table 1.

Table 1 Aberrant expression of miRNAs in gastric cancer.
Up-regulated miRNAs
let-7a, miR-9, -10a, -10b, -17, -17-5p, -18a, -18b, -19a, -19b, -20a, -20b, -21, -23a, -23b
miR-25, -26b, -27, -29b-1, -30b, -31, -34a, -34b, -34c, -92, -98, -99a, -100, -103, -106a
miR-106b, -107, -125b, -126, -128a, -130b, -138, -142-3p, -146a, -147, -150, -151-5p
miR-155, 181a, -181a-2, -181b, -181c, -185, -191, -192, -194, -196a, -196b, -199a
miR-199a-3p, -200b, -210, -214, -215, -221, -222, -223, -296-5p
miR-301a, -302f, -337-3p, -340, -370, -421, -520c-3p, -575, -601, -616, -658, -1259
Down-regulated miRNAs
let-7a, -7f, miR-7, -9, -22, -29c, -30a-5p, -31, -34a, -34b, -34c
miR-101, -126, -128b, -129, -129-2, -129-3p, -130b, -133b, -135a, -137, -141, -145
miR-146a, -148, -148b, -149, -152, -155, -181b, -181c, -182, -193b, -195, -195-5p, -197
miR-200, -204, -206, -210, -212, -218, -219-2-3p, -302b, -331-3p, -375
miR-378, -408-3p, -429, -433, -486, -495, -551a, -574-3p, -610, -622, -638, -663, -874

Upregulated miRNAs (oncomirs): Aberrant expression of miRNAs has been observed in many cancers[19,23]. Oncomirs are oncogenic miRNAs that are up-regulated in cancer cells and have been shown to act as oncogenes in GC. One such oncomir is miR-21, which has been widely reported as an oncomir in GC[24-26].

Overexpression of miR-21 has been reported in various cancers, such as esophageal cancer[27], breast cancer[28], and glioblastoma[29]. In GC, the expression of miR-21 is upregulated compared with normal tissues[22,30,31]. Motoyama et al[31] and Cao et al[32] reported an inverse correlation between miR-21 and PDCD4 expression in GC. PDCD4, a direct target gene of miR-21, encodes a protein that inhibits cell growth and invasion[33,34]. Moreover, Zhang et al[26] reported that PTEN, a well-known tumor-suppressor gene, is a target of miR-21. Thus, these data support the idea that miR-21 acts as a key oncomir in GC by inhibiting the tumor-suppressor genes PDCD4 and PTEN.

In addition to miR-21, miR-106a expression is also upregulated in GC (Table 1), as well as in several other human tumors, compared with adjacent normal tissues[35]. miR-106a mimics the function of positive regulators of the G1-to-S transition[35]. In several human hematopoietic cell lines, miR-106a has been shown to target interleukin (IL) 10[36], downregulating the expression of this critical cytokine by binding to the 3′ untranslated region[36]. As a further regulatory element, SP1 and EGR1 indirectly downregulate IL10 expression by inducing miR-106a expression[36].

Downregulated miRNAs (tumor-suppressor miRNAs): Down-regulated miRNAs in cancer tissue are referred to as tumor-suppressor miRNAs[37-39]. The important target gene of a tumor-suppressor miRNA is usually an oncogene. Therefore, decreased expression of the tumor-suppressive miRNA induces the expression of the oncogene. The genomic loss of miR-101 in cancer leads to overexpression of EZH2 and concomitant dysregulation of epigenetic pathways, resulting in cancer progression[40,41]. Because miR-101 targets cyclooxygenase (COX) 2 in GC, downregulation of miR-101 induces COX2 expression[42]. COX2 activates the arachidonic acid/prostaglandin E2 pathway following cell proliferation[42]. Oncogenic targets of miR-101 induce cell proliferation in GC. Therefore, miR-101 may be useful for gene therapy in GC.

Another miRNA that has been suggested to have a role in cancer is let-7. Expression of let-7 reduces the expression of 3 human RAS genes, HRAS, KRAS, and NRAS. Moreover, let-7 expression is lower in lung tumors than in normal lung tissue, whereas expression of the RAS proteins is significantly higher in lung tumors, suggesting a possible role of let-7 in cancer[43]. The expression of let-7 miRNA is also reduced in human lung cancer[44], breast cancer[45], and hepatocellular carcinomas[46]. In addition, overexpression of let-7 inhibits the growth of lung cancer cells in vitro[44]. In GC, RAB40C, a target of let-7a, has been reported to play an essential role in gastric tumorigenesis[47].

MiR-148a has been shown to act as a tumor suppressor in prostate cancer, and its expression is lower in prostate cancer cells compared with normal prostate epithelial cells[48]. In GC, miR-148a is inactivated by hypermethylation of the promoter region[49]. This may result in the upregulation of DNA methyltransferase, which is a target of miR-148a[49]. Moreover, miR-148a suppresses tumor cell invasion by downregulating ROCK1[50]. In a report from our laboratory, we found that miR-148a expression is downregulated in undifferentiated GC[51].

Roles of miRNAs as prognostic factors

Because many factors affect the prognosis of cancer patients, it is difficult to clarify how miRNAs are involved in the prognosis of patients with GC. However, advances in research on the potential role of miRNAs in patient prognosis may lead to the use of miRNAs as tools in medical treatment or diagnosis in the future. The detection of miRNAs involved in the prognosis of GC patients will not only be useful for predicting prognosis but helpful for developing therapeutic targets in the future. In Table 2, we have summarized the current knowledge on the relationship between miRNAs and prognosis. In particular, miR-21, -93, and -125 have been well studied in this context. While we mentioned the usefulness of miR-21 previously, overexpression of miR-93 in GC cells has been shown to reduce the cellular response to transforming growth factor (TGF)-β (TGFB1) by interfering with the synthesis of p21 (CDKN1A) and BIM (BCL2L11), the 2 most important downstream effectors of TGF-β-dependent cell cycle arrest and apoptosis, respectively[52]. High expression of miR-98 was found to predict poor survival[53,54]. Interestingly, low expression of miR-125 in GC has been shown to be an independent prognostic factor for survival[55,56]. One target gene of miR-125a-5p is ERBB2 (HER2), which is an important molecular target in chemotherapy[56].

Table 2 miRNAs associated with prognosis in gastric cancer patients.
let-7a ,-7i
miR-10b, -20a, -20b, -21, -22, -25, -27a, -30a-5p, -34a, -93
miR-103, -106a, -106b, -107, -125a-5p, -126, -130, -142-5p, -144, -146a, -150
miR-155, -181c, -195, -196a, -199a-3p, -200c, -206, -221, -222, -223
miR-335, -338, -372, -375, -451

Notably, many genes act as prognostic factors for patients with GC; miRNAs may regulate these genes, thereby affecting prognosis. Thus, further analysis of candidate miRNAs is necessary.

MiRNAs involved in chemosensitivity

Chemotherapy is an important tool for the treatment of GC. However, with currently available tools, it is impossible to predict whether GC patients will respond to chemotherapeutic approaches. The ability to predict the effects of chemotherapy may help reduce the unnecessary use of chemotherapeutics in GC. Current chemotherapeutic agents used in the treatment of GC include 5FU, CDDP, taxan, and irinotecan. Many studies have reported that miRNAs may affect the efficacy of chemotherapy. Wang et al[57] identified 9 upregulated miRNAs and 18 downregulated miRNAs involved in 5FU sensitivity by microarray and RT-PCR (Table 3). Moreover, miR-143, miR-145, and miR-144 have been reported to be involved in 5FU sensitivity[58,59]. In Table 3, we summarize the involvement of miRNAs in CPT, CDDP, and CF sensitivity. In the near future, we will be able to use miRNA expression as a predictor of chemotherapeutic efficacy. Additionally, gene therapy with miRNAs may be able to induce chemosensitivity in patients with GC.

Table 3 MiRNAs involved in chemosensitivity.
5FU sensitivity
let-7g
miR-10b, -22, -30c, -31, -32,
miR-133b, -143, -144, -145, -181b, -190, -197, -200c, -204, -210
miR-335, -501, -501-5p, -532, -615, -615-5p, -766, -877
miR-1224-3p, -1229, -3131, -3149, -3162-3p, -4763-3p
CPT sensitivity
let-7g
miR-7, -31, -98, -126, -196a, -200, -338
CDDP, CF sensitivity
let-7g
miR-1, -16, -21, -34, -181, -181b, -342, -497
MiRNAs involved in Helicobacter pylori infection

Helicobacter pylori (H. pylori) selectively colonize the gastric epithelium and typically persist for the lifetime of the host. Among colonized individuals, however, only a fraction develop gastric adenocarcinoma, emphasizing the importance of understanding the pathogenic mechanisms through which H. pylori promote chronic inflammation and the progression to GC[60]. miRNAs involved in H. pylori infections have been reported in several papers. Let-7 expression has been shown to be downregulated by Cag A after H. pylori infection, and Ras, a target of let-7, is overexpressed in GC[61]. Additionally, miR-17/92, the miR-106b-93-25 cluster, miR-21, miR-194, miR-196, miR155, miR-222, and miR-223 are upregulated in gastric mucosa infected by H. pylori[62-64]. Among these miRNAs, Li et al[64] revealed that miR-222 targets RECK, which inhibits the tumorigenicity of GC[64-66]. Further analysis of miRNAs and target genes may clarify the complicated mechanism of GC that occurs in the context of H. pylori infections.

Circulating miRNAs as biomarkers

Presently, circulating miRNAs found in the blood of patients constitute the most promising type of miRNA for clinical use, because these are concise for blood collecting. We summarized the circulating miRNAs in the blood of GC patients in Table 4.

Table 4 Circulating miRNAs as biomarkers.
Up-regulated miRNAs
miR-1, -17, -17-5p, -20a, -21, -27a, -31, -34, -103, -106a, -106b, -107,-194, -200c
miR-210, -221, -223, -370, -376a, -378, 421, 423-5p, 451, -486, 744
Down-regulated miRNAs
miR-218, -375

There are a number of reports describing the use of miR-106[67-69], miR-17[67,70], miR-21[67,71], and miR-221 as potential biomarkers[72]. The detection of miRNA in peripheral blood may be a novel tool for monitoring circulating tumor cells in patients with gastric cancers. Moreover, circulating miRNA may be a promising, non-invasive molecular marker for tracking pathological progression, predicting prognosis and monitoring chemotherapeutic effects in gastric cancer.

Finally, the precise mechanism of each miRNA is not well known. Further reports about miRNA are expected to help us better understand cancer mechanisms. This research will be useful for clinical diagnosis or treatment for GC patients.

CONCLUSION

In this review, we presented and discussed the newest knowledge on miRNAs in gastric cancer and their potential usefulness as future medical treatments and diagnostic tools. Although the molecular biology of GC has been well characterized, research on miRNAs in GC is still in its infancy. Thus, in the near future, we anticipate that advances in miRNA research in GC may help to develop novel medical treatments or diagnostic tools, thereby improving the prognosis of GC patients.

Footnotes

P- Reviewers: de Franciscis V, Poltronieri P, Ren T S- Editor: Ma YJ L- Editor: A E- Editor: Wang CH

References
1.  Jemal A, Siegel R, Ward E, Hao Y, Xu J, Murray T, Thun MJ. Cancer statistics, 2008. CA Cancer J Clin. 2008;58:71-96.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8283]  [Cited by in F6Publishing: 8159]  [Article Influence: 509.9]  [Reference Citation Analysis (0)]
2.  Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J Clin. 2011;61:69-90.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23762]  [Cited by in F6Publishing: 25182]  [Article Influence: 1937.1]  [Reference Citation Analysis (3)]
3.  Parkin DM, Bray F, Ferlay J, Pisani P. Global cancer statistics, 2002. CA Cancer J Clin. 2005;55:74-108.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13286]  [Cited by in F6Publishing: 13416]  [Article Influence: 706.1]  [Reference Citation Analysis (1)]
4.  Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116:281-297.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25833]  [Cited by in F6Publishing: 26782]  [Article Influence: 1339.1]  [Reference Citation Analysis (0)]
5.  Rossbach M. Small non-coding RNAs as novel therapeutics. Curr Mol Med. 2010;10:361-368.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 50]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
6.  Nelson KM, Weiss GJ. MicroRNAs and cancer: past, present, and potential future. Mol Cancer Ther. 2008;7:3655-3660.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 205]  [Cited by in F6Publishing: 222]  [Article Influence: 14.8]  [Reference Citation Analysis (0)]
7.  Wiemer EA. The role of microRNAs in cancer: no small matter. Eur J Cancer. 2007;43:1529-1544.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 248]  [Cited by in F6Publishing: 271]  [Article Influence: 15.9]  [Reference Citation Analysis (0)]
8.  Liu W, Mao SY, Zhu WY. Impact of tiny miRNAs on cancers. World J Gastroenterol. 2007;13:497-502.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Hutvágner G, Zamore PD. A microRNA in a multiple-turnover RNAi enzyme complex. Science. 2002;297:2056-2060.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1474]  [Cited by in F6Publishing: 1429]  [Article Influence: 65.0]  [Reference Citation Analysis (0)]
10.  Lin SL, Kim H, Ying SY. Intron-mediated RNA interference and microRNA (miRNA). Front Biosci. 2008;13:2216-2230.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 48]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
11.  Cheng AM, Byrom MW, Shelton J, Ford LP. Antisense inhibition of human miRNAs and indications for an involvement of miRNA in cell growth and apoptosis. Nucleic Acids Res. 2005;33:1290-1297.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1089]  [Cited by in F6Publishing: 1129]  [Article Influence: 59.4]  [Reference Citation Analysis (0)]
12.  Matsubara H, Takeuchi T, Nishikawa E, Yanagisawa K, Hayashita Y, Ebi H, Yamada H, Suzuki M, Nagino M, Nimura Y. Apoptosis induction by antisense oligonucleotides against miR-17-5p and miR-20a in lung cancers overexpressing miR-17-92. Oncogene. 2007;26:6099-6105.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 261]  [Cited by in F6Publishing: 267]  [Article Influence: 15.7]  [Reference Citation Analysis (0)]
13.  Akao Y, Nakagawa Y, Naoe T. let-7 microRNA functions as a potential growth suppressor in human colon cancer cells. Biol Pharm Bull. 2006;29:903-906.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 442]  [Cited by in F6Publishing: 467]  [Article Influence: 25.9]  [Reference Citation Analysis (0)]
14.  Cahill S, Smyth P, Finn SP, Denning K, Flavin R, O’Regan EM, Li J, Potratz A, Guenther SM, Henfrey R. Effect of ret/PTC 1 rearrangement on transcription and post-transcriptional regulation in a papillary thyroid carcinoma model. Mol Cancer. 2006;5:70.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 51]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
15.  Iorio MV, Ferracin M, Liu CG, Veronese A, Spizzo R, Sabbioni S, Magri E, Pedriali M, Fabbri M, Campiglio M. MicroRNA gene expression deregulation in human breast cancer. Cancer Res. 2005;65:7065-7070.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2930]  [Cited by in F6Publishing: 2983]  [Article Influence: 157.0]  [Reference Citation Analysis (0)]
16.  Ambros V. The functions of animal microRNAs. Nature. 2004;431:350-355.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7919]  [Cited by in F6Publishing: 8284]  [Article Influence: 414.2]  [Reference Citation Analysis (0)]
17.  Alvarez-Garcia I, Miska EA. MicroRNA functions in animal development and human disease. Development. 2005;132:4653-4662.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1018]  [Cited by in F6Publishing: 1007]  [Article Influence: 53.0]  [Reference Citation Analysis (0)]
18.  Zhang B, Pan X, Cobb GP, Anderson TA. microRNAs as oncogenes and tumor suppressors. Dev Biol. 2007;302:1-12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1765]  [Cited by in F6Publishing: 1899]  [Article Influence: 105.5]  [Reference Citation Analysis (0)]
19.  Volinia S, Calin GA, Liu CG, Ambs S, Cimmino A, Petrocca F, Visone R, Iorio M, Roldo C, Ferracin M. A microRNA expression signature of human solid tumors defines cancer gene targets. Proc Natl Acad Sci USA. 2006;103:2257-2261.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4162]  [Cited by in F6Publishing: 4422]  [Article Influence: 245.7]  [Reference Citation Analysis (0)]
20.  He L, Thomson JM, Hemann MT, Hernando-Monge E, Mu D, Goodson S, Powers S, Cordon-Cardo C, Lowe SW, Hannon GJ. A microRNA polycistron as a potential human oncogene. Nature. 2005;435:828-833.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2729]  [Cited by in F6Publishing: 2776]  [Article Influence: 146.1]  [Reference Citation Analysis (0)]
21.  Yao Y, Suo AL, Li ZF, Liu LY, Tian T, Ni L, Zhang WG, Nan KJ, Song TS, Huang C. MicroRNA profiling of human gastric cancer. Mol Med Rep. 2009;2:963-970.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 149]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
22.  Li X, Zhang Y, Zhang H, Liu X, Gong T, Li M, Sun L, Ji G, Shi Y, Han Z. miRNA-223 promotes gastric cancer invasion and metastasis by targeting tumor suppressor EPB41L3. Mol Cancer Res. 2011;9:824-833.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 256]  [Cited by in F6Publishing: 288]  [Article Influence: 22.2]  [Reference Citation Analysis (0)]
23.  Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, Sweet-Cordero A, Ebert BL, Mak RH, Ferrando AA. MicroRNA expression profiles classify human cancers. Nature. 2005;435:834-838.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7124]  [Cited by in F6Publishing: 7179]  [Article Influence: 377.8]  [Reference Citation Analysis (0)]
24.  Zeng Z, Wang J, Zhao L, Hu P, Zhang H, Tang X, He D, Tang S, Zeng Z. Potential role of microRNA-21 in the diagnosis of gastric cancer: a meta-analysis. PLoS One. 2013;8:e73278.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 51]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
25.  Osawa S, Shimada Y, Sekine S, Okumura T, Nagata T, Fukuoka J, Tsukada K. MicroRNA profiling of gastric cancer patients from formalin-fixed paraffin-embedded samples. Oncol Lett. 2011;2:613-619.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 34]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
26.  Zhang BG, Li JF, Yu BQ, Zhu ZG, Liu BY, Yan M. microRNA-21 promotes tumor proliferation and invasion in gastric cancer by targeting PTEN. Oncol Rep. 2012;27:1019-1026.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 193]  [Cited by in F6Publishing: 218]  [Article Influence: 18.2]  [Reference Citation Analysis (0)]
27.  Mori Y, Ishiguro H, Kuwabara Y, Kimura M, Mitsui A, Ogawa R, Katada T, Harata K, Tanaka T, Shiozaki M. MicroRNA-21 induces cell proliferation and invasion in esophageal squamous cell carcinoma. Mol Med Rep. 2009;2:235-239.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 16]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
28.  Si ML, Zhu S, Wu H, Lu Z, Wu F, Mo YY. miR-21-mediated tumor growth. Oncogene. 2007;26:2799-2803.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1136]  [Cited by in F6Publishing: 1170]  [Article Influence: 65.0]  [Reference Citation Analysis (0)]
29.  Chan JA, Krichevsky AM, Kosik KS. MicroRNA-21 is an antiapoptotic factor in human glioblastoma cells. Cancer Res. 2005;65:6029-6033.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1865]  [Cited by in F6Publishing: 1856]  [Article Influence: 97.7]  [Reference Citation Analysis (0)]
30.  Inoue T, Iinuma H, Ogawa E, Inaba T, Fukushima R. Clinicopathological and prognostic significance of microRNA-107 and its relationship to DICER1 mRNA expression in gastric cancer. Oncol Rep. 2012;27:1759-1764.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 47]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
31.  Motoyama K, Inoue H, Mimori K, Tanaka F, Kojima K, Uetake H, Sugihara K, Mori M. Clinicopathological and prognostic significance of PDCD4 and microRNA-21 in human gastric cancer. Int J Oncol. 2010;36:1089-1095.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Cao Z, Yoon JH, Nam SW, Lee JY, Park WS. PDCD4 expression inversely correlated with miR-21 levels in gastric cancers. J Cancer Res Clin Oncol. 2012;138:611-619.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 39]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
33.  Hiyoshi Y, Kamohara H, Karashima R, Sato N, Imamura Y, Nagai Y, Yoshida N, Toyama E, Hayashi N, Watanabe M. MicroRNA-21 regulates the proliferation and invasion in esophageal squamous cell carcinoma. Clin Cancer Res. 2009;15:1915-1922.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 207]  [Cited by in F6Publishing: 220]  [Article Influence: 14.7]  [Reference Citation Analysis (0)]
34.  Lu Z, Liu M, Stribinskis V, Klinge CM, Ramos KS, Colburn NH, Li Y. MicroRNA-21 promotes cell transformation by targeting the programmed cell death 4 gene. Oncogene. 2008;27:4373-4379.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 526]  [Cited by in F6Publishing: 541]  [Article Influence: 33.8]  [Reference Citation Analysis (0)]
35.  Ivanovska I, Ball AS, Diaz RL, Magnus JF, Kibukawa M, Schelter JM, Kobayashi SV, Lim L, Burchard J, Jackson AL. MicroRNAs in the miR-106b family regulate p21/CDKN1A and promote cell cycle progression. Mol Cell Biol. 2008;28:2167-2174.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 422]  [Cited by in F6Publishing: 442]  [Article Influence: 27.6]  [Reference Citation Analysis (0)]
36.  Sharma A, Kumar M, Aich J, Hariharan M, Brahmachari SK, Agrawal A, Ghosh B. Posttranscriptional regulation of interleukin-10 expression by hsa-miR-106a. Proc Natl Acad Sci USA. 2009;106:5761-5766.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 150]  [Cited by in F6Publishing: 146]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
37.  Grammatikakis I, Gorospe M, Abdelmohsen K. Modulation of Cancer Traits by Tumor Suppressor microRNAs. Int J Mol Sci. 2013;14:1822-1842.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 23]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
38.  Yim RL, Kwong YL, Wong KY, Chim CS. DNA Methylation of Tumor Suppressive miRNAs in Non-Hodgkin’s Lymphomas. Front Genet. 2012;3:233.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 12]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
39.  Wang LQ, Liang R, Chim CS. Methylation of tumor suppressor microRNAs: lessons from lymphoid malignancies. Expert Rev Mol Diagn. 2012;12:755-765.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 13]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
40.  Varambally S, Cao Q, Mani RS, Shankar S, Wang X, Ateeq B, Laxman B, Cao X, Jing X, Ramnarayanan K. Genomic loss of microRNA-101 leads to overexpression of histone methyltransferase EZH2 in cancer. Science. 2008;322:1695-1699.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 808]  [Cited by in F6Publishing: 812]  [Article Influence: 50.8]  [Reference Citation Analysis (0)]
41.  Carvalho J, van Grieken NC, Pereira PM, Sousa S, Tijssen M, Buffart TE, Diosdado B, Grabsch H, Santos MA, Meijer G. Lack of microRNA-101 causes E-cadherin functional deregulation through EZH2 up-regulation in intestinal gastric cancer. J Pathol. 2012;228:31-44.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 79]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
42.  He XP, Shao Y, Li XL, Xu W, Chen GS, Sun HH, Xu HC, Xu X, Tang D, Zheng XF. Downregulation of miR-101 in gastric cancer correlates with cyclooxygenase-2 overexpression and tumor growth. FEBS J. 2012;279:4201-4212.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 78]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
43.  Johnson SM, Grosshans H, Shingara J, Byrom M, Jarvis R, Cheng A, Labourier E, Reinert KL, Brown D, Slack FJ. RAS is regulated by the let-7 microRNA family. Cell. 2005;120:635-647.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2664]  [Cited by in F6Publishing: 2651]  [Article Influence: 139.5]  [Reference Citation Analysis (0)]
44.  Takamizawa J, Konishi H, Yanagisawa K, Tomida S, Osada H, Endoh H, Harano T, Yatabe Y, Nagino M, Nimura Y. Reduced expression of the let-7 microRNAs in human lung cancers in association with shortened postoperative survival. Cancer Res. 2004;64:3753-3756.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1813]  [Cited by in F6Publishing: 1812]  [Article Influence: 90.6]  [Reference Citation Analysis (0)]
45.  Yu F, Yao H, Zhu P, Zhang X, Pan Q, Gong C, Huang Y, Hu X, Su F, Lieberman J. let-7 regulates self renewal and tumorigenicity of breast cancer cells. Cell. 2007;131:1109-1123.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1416]  [Cited by in F6Publishing: 1439]  [Article Influence: 89.9]  [Reference Citation Analysis (0)]
46.  Gramantieri L, Ferracin M, Fornari F, Veronese A, Sabbioni S, Liu CG, Calin GA, Giovannini C, Ferrazzi E, Grazi GL. Cyclin G1 is a target of miR-122a, a microRNA frequently down-regulated in human hepatocellular carcinoma. Cancer Res. 2007;67:6092-6099.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 614]  [Cited by in F6Publishing: 621]  [Article Influence: 36.5]  [Reference Citation Analysis (0)]
47.  Yang Q, Jie Z, Cao H, Greenlee AR, Yang C, Zou F, Jiang Y. Low-level expression of let-7a in gastric cancer and its involvement in tumorigenesis by targeting RAB40C. Carcinogenesis. 2011;32:713-722.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 87]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
48.  Fujita Y, Kojima K, Ohhashi R, Hamada N, Nozawa Y, Kitamoto A, Sato A, Kondo S, Kojima T, Deguchi T. MiR-148a attenuates paclitaxel resistance of hormone-refractory, drug-resistant prostate cancer PC3 cells by regulating MSK1 expression. J Biol Chem. 2010;285:19076-19084.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 147]  [Cited by in F6Publishing: 156]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
49.  Zhu A, Xia J, Zuo J, Jin S, Zhou H, Yao L, Huang H, Han Z. MicroRNA-148a is silenced by hypermethylation and interacts with DNA methyltransferase 1 in gastric cancer. Med Oncol. 2012;29:2701-2709.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 78]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
50.  Zheng B, Liang L, Wang C, Huang S, Cao X, Zha R, Liu L, Jia D, Tian Q, Wu J. MicroRNA-148a suppresses tumor cell invasion and metastasis by downregulating ROCK1 in gastric cancer. Clin Cancer Res. 2011;17:7574-7583.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 201]  [Cited by in F6Publishing: 226]  [Article Influence: 17.4]  [Reference Citation Analysis (0)]
51.  Katada T, Ishiguro H, Kuwabara Y, Kimura M, Mitui A, Mori Y, Ogawa R, Harata K, Fujii Y. microRNA expression profile in undifferentiated gastric cancer. Int J Oncol. 2009;34:537-542.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Petrocca F, Visone R, Onelli MR, Shah MH, Nicoloso MS, de Martino I, Iliopoulos D, Pilozzi E, Liu CG, Negrini M. E2F1-regulated microRNAs impair TGFbeta-dependent cell-cycle arrest and apoptosis in gastric cancer. Cancer Cell. 2008;13:272-286.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 654]  [Cited by in F6Publishing: 682]  [Article Influence: 42.6]  [Reference Citation Analysis (0)]
53.  Yu BQ, Su LP, Li JF, Cai Q, Yan M, Chen XH, Yu YY, Gu QL, Zhu ZG, Liu BY. microrna expression signature of gastric cancer cells relative to normal gastric mucosa. Mol Med Rep. 2012;6:821-826.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 21]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
54.  Chen L, Jiang M, Yuan W, Tang H. Prognostic value of miR-93 overexpression in resectable gastric adenocarcinomas. Acta Gastroenterol Belg. 2012;75:22-27.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Hashiguchi Y, Nishida N, Mimori K, Sudo T, Tanaka F, Shibata K, Ishii H, Mochizuki H, Hase K, Doki Y. Down-regulation of miR-125a-3p in human gastric cancer and its clinicopathological significance. Int J Oncol. 2012;40:1477-1482.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 45]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
56.  Nishida N, Mimori K, Fabbri M, Yokobori T, Sudo T, Tanaka F, Shibata K, Ishii H, Doki Y, Mori M. MicroRNA-125a-5p is an independent prognostic factor in gastric cancer and inhibits the proliferation of human gastric cancer cells in combination with trastuzumab. Clin Cancer Res. 2011;17:2725-2733.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 167]  [Cited by in F6Publishing: 199]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
57.  Wang Y, Gu X, Li Z, Xiang J, Jiang J, Chen Z. microRNA expression profiling in multidrug resistance of the 5-Fu-induced SGC-7901 human gastric cancer cell line. Mol Med Rep. 2013;7:1506-1510.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 22]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
58.  Takagi T, Iio A, Nakagawa Y, Naoe T, Tanigawa N, Akao Y. Decreased expression of microRNA-143 and -145 in human gastric cancers. Oncology. 2009;77:12-21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 207]  [Cited by in F6Publishing: 238]  [Article Influence: 15.9]  [Reference Citation Analysis (0)]
59.  Akiyoshi S, Fukagawa T, Ueo H, Ishibashi M, Takahashi Y, Fabbri M, Sasako M, Maehara Y, Mimori K, Mori M. Clinical significance of miR-144-ZFX axis in disseminated tumour cells in bone marrow in gastric cancer cases. Br J Cancer. 2012;107:1345-1353.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 66]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
60.  Noto JM, Peek RM. The role of microRNAs in Helicobacter pylori pathogenesis and gastric carcinogenesis. Front Cell Infect Microbiol. 2011;1:21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 32]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
61.  Hayashi Y, Tsujii M, Wang J, Kondo J, Akasaka T, Jin Y, Li W, Nakamura T, Nishida T, Iijima H. CagA mediates epigenetic regulation to attenuate let-7 expression in Helicobacter pylori-related carcinogenesis. Gut. 2013;62:1536-1546.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 96]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
62.  Shiotani A, Uedo N, Iishi H, Murao T, Kanzaki T, Kimura Y, Kamada T, Kusunoki H, Inoue K, Haruma K. H. pylori eradication did not improve dysregulation of specific oncogenic miRNAs in intestinal metaplastic glands. J Gastroenterol. 2012;47:988-998.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 41]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
63.  Oertli M, Engler DB, Kohler E, Koch M, Meyer TF, Müller A. MicroRNA-155 is essential for the T cell-mediated control of Helicobacter pylori infection and for the induction of chronic Gastritis and Colitis. J Immunol. 2011;187:3578-3586.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 128]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
64.  Li N, Tang B, Zhu ED, Li BS, Zhuang Y, Yu S, Lu DS, Zou QM, Xiao B, Mao XH. Increased miR-222 in H. pylori-associated gastric cancer correlated with tumor progression by promoting cancer cell proliferation and targeting RECK. FEBS Lett. 2012;586:722-728.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 75]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
65.  Hong KJ, Wu DC, Cheng KH, Chen LT, Hung WC. RECK inhibits stemness gene expression and tumorigenicity of gastric cancer cells by suppressing ADAM-mediated Notch1 activation. J Cell Physiol. 2014;229:191-201.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 50]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
66.  Du YY, Dai DQ, Yang Z. Role of RECK methylation in gastric cancer and its clinical significance. World J Gastroenterol. 2010;16:904-908.  [PubMed]  [DOI]  [Cited in This Article: ]
67.  Komatsu S, Ichikawa D, Tsujiura M, Konishi H, Takeshita H, Nagata H, Kawaguchi T, Hirajima S, Arita T, Shiozaki A. Prognostic impact of circulating miR-21 in the plasma of patients with gastric carcinoma. Anticancer Res. 2013;33:271-276.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Zhou H, Guo JM, Lou YR, Zhang XJ, Zhong FD, Jiang Z, Cheng J, Xiao BX. Detection of circulating tumor cells in peripheral blood from patients with gastric cancer using microRNA as a marker. J Mol Med (Berl). 2010;88:709-717.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 124]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
69.  Tsujiura M, Ichikawa D, Komatsu S, Shiozaki A, Takeshita H, Kosuga T, Konishi H, Morimura R, Deguchi K, Fujiwara H. Circulating microRNAs in plasma of patients with gastric cancers. Br J Cancer. 2010;102:1174-1179.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 449]  [Cited by in F6Publishing: 487]  [Article Influence: 34.8]  [Reference Citation Analysis (0)]
70.  Wang M, Gu H, Wang S, Qian H, Zhu W, Zhang L, Zhao C, Tao Y, Xu W. Circulating miR-17-5p and miR-20a: molecular markers for gastric cancer. Mol Med Rep. 2012;5:1514-1520.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 74]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
71.  Zheng Y, Cui L, Sun W, Zhou H, Yuan X, Huo M, Chen J, Lou Y, Guo J. MicroRNA-21 is a new marker of circulating tumor cells in gastric cancer patients. Cancer Biomark. 2001;10:71-77.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 60]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
72.  Cai H, Yuan Y, Hao YF, Guo TK, Wei X, Zhang YM. Plasma microRNAs serve as novel potential biomarkers for early detection of gastric cancer. Med Oncol. 2013;30:452.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 76]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]