Epigenetic modulation associated with carcinogenesis and prognosis of human gastric cancer (Review)

  • Authors:
    • Fuminori Sonohara
    • Yoshikuni Inokawa
    • Masamichi Hayashi
    • Yasuhiro Kodera
    • Shuji Nomoto
  • View Affiliations

  • Published online on: March 27, 2017     https://doi.org/10.3892/ol.2017.5912
  • Pages: 3363-3368
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Gastric cancer (GC) is a leading cause of cancer‑related death, particularly in Asia. Epidemiological and other clinical studies have identified an association between a number of risk factors, including Helicobacter pylori, and GC. A number of studies have also examined genetic changes associated with the development and progression of GC. When considering the clinical significance of the expression of a specific gene, its epigenetic modulation should be considered. Epigenetic modulation appears to be a primary driver of changes in gastric tissue that promotes carcinogenesis and progression of GC and other neoplasms. The role of epigenetic modulation in GC carcinogenesis and progression has been widely studied in recent years. In the present review, recent results of epigenetic modulation associated with GC and their effects on clinical outcome are examined, with particular respect to DNA methylation, histone modulation and non‑coding RNA. A number of studies indicate that epigenetic changes in the expression of specific genes critically affect their clinical significance and further study may reveal epigenetic changes as the basis for targeted molecular therapy or novel biomarkers that predict GC prognosis or extension of this often fatal disease.

Introduction

Gastric cancer/carcinoma (GC) is a relatively common cancer, particularly in Asia (1). Although its incidence is decreasing gradually in developed countries, high morbidity and mortality of GC remains among cancer types (2,3). Early GC may be treated, and even completely cured surgically, using endoscopic mucosal resection and endoscopic submucosal dissection (4,5). However, the prognosis of advanced or distantly metastasized GC is worse. Although a number of systemic chemotherapy regimens are available to treat unresectable or distantly metastatic GC, highly advanced GC is difficult to completely cure using chemotherapy. Therefore, prevention or early detection of this fatal disease is critical. The epigenetic aspects of GC are an important current frontier in understanding its development and progression, and identifying its earliest stages.

Epidemiology and other clinical research have identified an association between several risk factors and GC. Helicobacter pylori is an influential factor in the carcinogenesis of GC (6). Certain dietary characteristics, including high sodium intake or low produce consumption are recognized risk factors for GC (7,8). Many studies have also examined genetic changes related to GC development and progression. Whole human genome sequencing has been available since 2003, and has led to new insights into human diseases, including the genetic aspects of GC (9); both genetic and epigenetic changes have been demonstrated to orchestrate carcinogenesis and progression of neoplasms.

Notably, genetic and epigenetic changes have been identified to affect cancer development in a stepwise manner, although they may have limited effects separately. Epigenetic modulation, including DNA methylation status, histone modification and non-coding RNA modulation, greatly influences neoplastic development (10). Therefore, understanding how these epigenetic changes affect GC has importance in detecting, treating and preventing this fatal disease. In the present review, epigenetic modulation of GC in terms of its association with epidemiological factors and clinicopathological factors is outlined, and the clinical significance of epigenetic modulation in GC carcinogenesis and prognosis is summarized.

DNA methylation

Specific and genome-wide DNA methylation

Abnormal DNA methylation can be caused by internal and external factors, and is distributed in two general patterns, genome-wide hypomethylation and site-specific CpG island local hypermethylation (11,12), which affect the prognosis of GC and other types of cancer. The methylation status of a gene's promoter region may determine whether or not it acts as a tumor suppressor; promoter regions of various tumor suppressor genes have been independently associated with prognosis of GC (1320) (Table I).

Table I.

Genes identified to exhibit an association with promoter methylation and clinical outcome.

Table I.

Genes identified to exhibit an association with promoter methylation and clinical outcome.

Author, yearGeneFunctionClinical association(Refs.)
Graziano et al, 2004CDH1Regulates cell-cell adhesion, mobility and proliferation of epithelial cellsPoorer DFS, OS(13)
de Maat et al, 2007COX-2Catalyzes prostaglandin productionImproved survival, time to recurrence(14)
Ooki et al, 2010HOPX-βCardiac growth and developmentPoorer survival(15)
Xu et al, 2012BCL6BEarly B cell developmentPoorer survival(16)
Li et al, 2012PAX5B cell differentiation, neural development and spermatogenesisPoorer survival(17)
Du et al, 2012ADAMTS9Cleaves the proteoglycans aggrecan and versicanPoorer survival(18)
Wang et al, 2013ZNF545 (ZFP82)Transcriptional regulationPoorer survival(19)
Guo et al, 2013RKIP (PEBP1)Involved in the presynaptic cholinergic neurons of the central nervous systemPoorer survival(20)

[i] CDH1, cadherin 1; DFS, disease-free survival; OS, overall survival; COX-2, cyclo-oxygenase 2; HOPX-β, HOP homeobox β; BCL6B, B cell CLL/lymphoma 6B; PAX5, paired box 5; ADAMTS9, a disintegrin and metalloproteinase with thrombospondin motifs 9; ZNF545, zinc finger 545; ZFP82, zinc finger protein 82; RKIP, Raf kinase inhibitor protein; PEBP1, phosphatidylethanolamine-binding protein 1.

The effect of whole-genome hypomethylation on GC was first described in 1996 (21). In that study, the authors identified that hypomethylation of DNA was increased in a stepwise manner in normal gastric mucosae, superficial gastritis and atrophic gastritis, but did not significantly increase between atrophic gastritis and GC. The European Prospective Investigation into Cancer and Nutrition cohort study identified that global demethylation of tumor cell genomes occurred in GC, which is consistent with the idea that abnormal hypermethylation of specific genes occurs concomitantly with genome-wide hypomethylation (22). Long interspersed element 1 (LINE-1), a retrotransposon, occupies ~17% of human DNA; its hypomethylation is hypothesized to be a surrogate for whole-DNA hypomethylation. Bae et al (23) demonstrated that LINE-1 hypomethylation was an independent prognostic factor following curative resection for advanced GC. Shigaki et al (24) also identified an association between LINE-1 hypomethylation and shorter survival time in GC, which suggests that it is a potent prognostic biomarker.

DNA methylation associated with etiological factors

A strong etiological factor in GC is H. pylori. Maekita et al (25) identified an association between H. pylori infection and hypermethylation of certain specific CpG islands. Furthermore, Chan et al (26) demonstrated that aberrant methylation of CpG islands in the epithelial cadherin promotor was an early event in H. pylori-induced GC carcinogenesis. Aberrant methylation has also been associated with Epstein-Barr virus, another virus associated with development of GC (27). Although associations between DNA methylation and habits including smoking or high sodium intake remain unclear, Xu et al (28) recently demonstrated that methylation of the gene encoding transmembrane protein 106A in primary GC was significantly associated with smoking and tumor metastasis. Furthermore, methylation of the genes encoding human MutL homolog 1 and human MutS homolog 3 were identified to be age-related and may therefore serve an important role in carcinogenesis of GC in the older population (29).

Histone modulation

Histone octamers package and order the DNA of eukaryotic cells into nucleosomes as part of the chromatin structure. Histones possess a tail-like structure that extends beyond the nucleosome and may be modulated; the majority of well-investigated histone modulations affect transcription. Histone modification assists with the regulation of pre-mRNA splicing (30). Ubiquitin-like containing PHD and RING finger domains 1-dependent histone H3 ubiquitination has been demonstrated to be critical in maintaining methylation during DNA duplication (31). Histone modulation, including methylation, acetylation, phosphorylation and ubiquitination, affect oncogene expression (3236), although their clinical significance in patients with GC remains unclear. Cai et al (37) recently revealed that expression of histone-lysine N-methyltransferase Suv39H1 and trimethylated histone H3 methylated lysine 9 (H3K9) was increased in GC, and trimethylated H3K9 was identified to be positively associated with tumor stage and metastatic status (37).

Non-coding RNA

The Human Genome Project identified that although ~80% of the human genome is transcribed into RNA, only ~2% of those transcripts are translated into proteins. Numerous RNAs do not code for proteins in human cells, and are therefore called non-coding RNA. The functions of non-coding RNA have been partly elucidated, particularly for smaller transcripts. However, larger sized non-coding RNA is becoming more widely studied, particularly with regard to its roles in carcinogenesis and tumor progression.

MicroRNA (miRNA/miR)

miRNA is a relatively short non-coding RNA of between 20 and 28 base pairs, and is derived from host DNA. miRNA has emerged as an important modulator of post-transcriptional regulation. miRNA regulates gene expression primarily by interfering with the transcription and cleavage of mRNA (38,39). This interference may explain some discrepancies between mRNA production and corresponding protein expression. miRNA targets are thought to include <30% of the human genome (40), and misregulation of miRNA expression affects development and progression of various diseases, including neoplasms. miRNA expression levels are widely reported to be associated with clinical effects of GC (4168) (Table II). miRNA polymorphisms are also associated with GC outcomes. Stenholm et al (69) identified that polymorphisms of miR-26a, pre-miRNA of miR-27-a1 and pre-miRNA of miR-196-a2 were significantly associated with overall survival rates. As miRNA in formalin-fixed tissues is highly stable, and detection of circulating serum miRNA from tumors is widely studied, miRNA may be the basis of a novel system of detecting early-stage cancers (70). miRNA in serum is encapsulated in an ‘exosome’ that makes miRNA stable against various stresses and allows miRNA to be transferred between cells (71). Certain studies have investigated the possibility of using circulating miRNA as less invasive markers to detect and monitor GC (70,72,73).

Table II.

miRNAs identified to be associated with clinical outcome of gastric cancer.

Table II.

miRNAs identified to be associated with clinical outcome of gastric cancer.

Author, yearmiRNA(Refs.)
Highly expressed
  Wang et al, 2013miR-10b(43)
  Katada et al, 2009miR-20b(53)
  Katada et al, 2009miR-27a(53)
  Osawa et al, 2011miR-34a(65)
  Inoue et al, 2012miR-107(42)
  Liu et al, 2014miR-132(46)
  Naito et al, 2014miR-143(49)
  Katada et al, 2009miR-150(53)
  Chen et al, 2013miR-181a-5p(54)
  Brenner et al, 2011miR-199a-3p(56)
  Yang et al, 2013miR-214(58)
  Liu et al, 2012miR-221(61)
  Wang et al, 2013miR-301a(62)
  Yan et al, 2013miR-335(63)
Poorly expressed
  Liu et al, 2012Let-7i(41)
  Wang et al, 2013miR-22(60)
  Hashiguchi et al, 2012miR-125a-3p(44)
  Guo et al, 2013miR-127(45)
  Shin et al, 2013miR-135a(47)
  Bao et al, 2011miR-139(48)
  Akiyoshi et al, 2012miR-144(50)
  Zheng et al, 2011;miR-148a(51,52)
  Sakamoto et al, 2014
  Tan et al, 2014miR-185(55)
  Tang et al, 2013miR-200b(57)
  Tang et al, 2013miR-200c(57)
  Wang et al, 2014miR-214(59)
  Zheng et al, 2012miR-409-3p(64)
  Guo et al, 2013miR-433(45)
  Bandres et al, 2009miR-451(66)
  Iwaya et al, 2013miR-494(67)
  He et al, 2014miR-760(68)

[i] miRNA/miR, microRNA.

Long non-coding RNA (lncRNA)

Non-coding RNA transcripts of >200 base pairs are designated lncRNA. Previously, lncRNA was thought to be ‘junk nucleotides’, but recent evidence indicates that lncRNA is as important in epigenetic modulation as miRNA (74). Aberrant expression of certain types of lncRNA has been associated with poorer prognosis in breast cancer (75), hepatocellular carcinoma (76) and lung carcinoma (77); and it has recently been demonstrated to possess clinical significance in GC (7885) (Table III). lncRNA in plasma is reportedly detectable in patients with GC, as with miRNA (86).

Table III.

Long non-coding RNA identified to be associated with promoter methylation and clinical outcomes.

Table III.

Long non-coding RNA identified to be associated with promoter methylation and clinical outcomes.

Author, yearGeneStatusFunction(Refs.)
Yang et al, 2012H19IncreasedCellular proliferation, p53 inactivation(78)
Xu et al, 2013HOTAIRIncreasedPoor OS, invasiveness(79)
Yang et al, 2014GHET1IncreasedTumor size, tumor invasion and poor survival(80)
Lee et al, 2014nc886DecreasedPoor survival(81)
Xu et al, 2014FENDRRDecreasedTumor invasion, stage, lymph node metastasis, poor prognosis(82)
Han et al, 2014LEIGCDecreasedTumor growth, cell proliferation, 5-FU sensitivity(83)
Okugawa et al, 2014MALAT1IncreasedPeritoneal dissemination(84)
Xu et al, 2014LSINCT5IncreasedTumor size, depth, stage, worse DFS and DSS(85)

[i] H19, H19 imprinted maternally expressed transcript; HOTAIR, HOX transcript antisense RNA; OS, overall survival; GHET1, gastric carcinoma proliferation enhancing transcript 1; nc886, vault RNA 2; FENDRR, FOXF1 adjacent non-coding developmental regulatory RNA; LEIGC, lower expression in gastric cancer; 5-FU, 5-fluorouracil; MALAT1, metastasis-associated lung adenocarcinoma transcript 1; LSINCT5, long stress-induced non-coding transcript 5; DFS, disease-free survival; DSS, disease-specific survival.

Conclusions

In the present review, recent studies that have identified associations between epigenetic modulation and clinical outcomes in GC have been summarized and discussed. Epigenetic changes in the expression of specific genes critically affect their clinical significance. Further study may reveal epigenetic changes as the basis for targeted molecular therapy or novel biomarkers that predict GC prognosis or extension of this often fatal disease.

References

1 

Bray F, Ren JS, Masuyer E and Ferlay J: Global estimates of cancer prevalence for 27 sites in the adult population in 2008. Int J Cancer. 132:1133–1145. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Ferlay J, Soerjomataram I, Ervik M, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D and Bray F: GLOBOCAN 2012 v1.0, Cancer Incidence and Mortality Worldwide, IARC CancerBase No. 11. International Agency for Research on Cancer Lyon (France). 2013 http://globocan.iarc.frAccessed. September 15–2015.

3 

Ferro A, Peleteiro B, Malvezzi M, Bosetti C, Bertuccio P, Levi F, Negri E, La Vecchia C and Lunet N: Worldwide trends in gastric cancer mortality (1980–2011), with predictions to 2015, and incidence by subtype. Eur J Cancer. 50:1330–1344. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Ono H, Kondo H, Gotoda T, Shirao K, Yamaguchi H, Saito D, Hosokawa K, Shimoda T and Yoshida S: Endoscopic mucosal resection for treatment of early gastric cancer. Gut. 48:225–229. 2001. View Article : Google Scholar : PubMed/NCBI

5 

Gotoda T, Yamamoto H and Soetikno RM: Endoscopic submucosal dissection of early gastric cancer. J Gastroenterol. 41:929–942. 2006. View Article : Google Scholar : PubMed/NCBI

6 

An international association between Helicobacter-pylori infection and gastric-cancer. The EUROGAST Study Group. Lancet. 341:1359–1362. 1993. View Article : Google Scholar : PubMed/NCBI

7 

Dias-Neto M, Pintalhao M, Ferreira M and Lunet N: Salt intake and risk of gastric intestinal metaplasia: Systematic review and meta-analysis. Nutr Cancer. 62:133–147. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Tsugane S: Salt, salted food intake, and risk of gastric cancer: Epidemiologic evidence. Cancer Sci. 96:1–6. 2005. View Article : Google Scholar : PubMed/NCBI

9 

International Human Genome Sequencing Consortium: Finishing the euchromatic sequence of the human genome. Nature. 431:931–945. 2004. View Article : Google Scholar : PubMed/NCBI

10 

Jones PA and Baylin SB: The epigenomics of cancer. Cell. 128:683–692. 2007. View Article : Google Scholar : PubMed/NCBI

11 

Gaudet F, Hodgson JG, Eden A, Jackson-Grusby L, Dausman J, Gray JW, Leonhardt H and Jaenisch R: Induction of tumors in mice by genomic hypomethylation. Science. 300:489–492. 2003. View Article : Google Scholar : PubMed/NCBI

12 

Merlo A, Herman JG, Mao L, Lee DJ, Gabrielson E, Burger PC, Baylin SB and Sidransky D: 5′ CpG island methylation is associated with transcriptional silencing of the tumor-suppressor p16/CDKN2/MTS1 in human cancers. Nat Med. 1:686–692. 1995. View Article : Google Scholar : PubMed/NCBI

13 

Graziano F, Arduini F, Ruzzo A, Bearzi I, Humar B, More H, Silva R, Muretto P, Guilford P, Testa E, et al: Prognostic analysis of E-cadherin gene promoter hypermethylation in patients with surgically resected, node-positive, diffuse gastric cancer. Clin Cancer Res. 10:2784–2789. 2004. View Article : Google Scholar : PubMed/NCBI

14 

de Maat MF, van de Velde CJ, Umetani N, de Heer P, Putter H, van Hoesel AQ, Meijer GA, van Grieken NC, Kuppen PJ, Bilchik AJ, et al: Epigenetic silencing of cyclooxygenase-2 affects clinical outcome in gastric cancer. J Clin Oncol. 25:4887–4894. 2007. View Article : Google Scholar : PubMed/NCBI

15 

Ooki A, Yamashita K, Kikuchi S, Sakuramoto S, Katada N, Kokubo K, Kobayashi H, Kim MS, Sidransky D and Watanabe M: Potential utility of HOP homeobox gene promoter methylation as a marker of tumor aggressiveness in gastric cancer. Oncogene. 29:3263–3275. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Xu L, Li X, Chu ES, Zhao G, Go MY, Tao Q, Jin H, Zeng Z, Sung JJ and Yu J: Epigenetic inactivation of BCL6B, a novel functional tumour suppressor for gastric cancer, is associated with poor survival. Gut. 61:977–985. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Li X, Cheung KF, Ma X, Tian L, Zhao J, Go MY, Shen B, Cheng AS, Ying J, Tao Q, et al: Epigenetic inactivation of paired box gene 5, a novel tumor suppressor gene, through direct upregulation of p53 is associated with prognosis in gastric cancer patients. Oncogene. 31:3419–3430. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Du W, Wang S, Zhou Q, Li X, Chu J, Chang Z, Tao Q, Ng EK, Fang J, Sung JJ and Yu J: ADAMTS9 is a functional tumor suppressor through inhibiting AKT/mTOR pathway and associated with poor survival in gastric cancer. Oncogene. 32:3319–3328. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Wang S, Cheng Y, Du W, Lu L, Zhou L, Wang H, Kang W, Li X, Tao Q, Sung JJ and Yu J: Zinc-finger protein 545 is a novel tumour suppressor that acts by inhibiting ribosomal RNA transcription in gastric cancer. Gut. 62:833–841. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Guo W, Dong Z, Guo Y, Lin X, Chen Z, Kuang G and Yang Z: Aberrant methylation and loss expression of RKIP is associated with tumor progression and poor prognosis in gastric cardia adenocarcinoma. Clin Exp Metastasis. 30:265–275. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Cravo M, Pinto R, Fidalgo P, Chaves P, Glória L, Nobre-Leitão C and Costa Mira F: Global DNA hypomethylation occurs in the early stages of intestinal type gastric carcinoma. Gut. 39:434–438. 1996. View Article : Google Scholar : PubMed/NCBI

22 

Balassiano K, Lima S, Jenab M, Overvad K, Tjonneland A, Boutron-Ruault MC, Clavel-Chapelon F, Canzian F, Kaaks R, Boeing H, et al: Aberrant DNA methylation of cancer-associated genes in gastric cancer in the European Prospective Investigation into Cancer and Nutrition (EPIC-EURGAST). Cancer Lett. 311:85–95. 2011. View Article : Google Scholar : PubMed/NCBI

23 

Bae JM, Shin SH, Kwon HJ, Park SY, Kook MC, Kim YW, Cho NY, Kim N, Kim TY, Kim D and Kang GH: ALU and LINE-1 hypomethylations in multistep gastric carcinogenesis and their prognostic implications. Int J Cancer. 131:1323–1331. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Shigaki H, Baba Y, Watanabe M, Murata A, Iwagami S, Miyake K, Ishimoto T, Iwatsuki M and Baba H: LINE-1 hypomethylation in gastric cancer, detected by bisulfite pyrosequencing, is associated with poor prognosis. Gastric Cancer. 16:480–487. 2013. View Article : Google Scholar : PubMed/NCBI

25 

Maekita T, Nakazawa K, Mihara M, Nakajima T, Yanaoka K, Iguchi M, Arii K, Kaneda A, Tsukamoto T, Tatematsu M, et al: High levels of aberrant DNA methylation in Helicobacter pylori-infected gastric mucosae and its possible association with gastric cancer risk. Clin Cancer Res. 12:989–995. 2006. View Article : Google Scholar : PubMed/NCBI

26 

Chan AO, Lam SK, Wong BC, Wong WM, Yuen MF, Yeung YH, Hui WM, Rashid A and Kwong YL: Promoter methylation of E-cadherin gene in gastric mucosa associated with Helicobacter pylori infection and in gastric cancer. Gut. 52:502–506. 2003. View Article : Google Scholar : PubMed/NCBI

27 

Kang GH, Lee S, Kim WH, Lee HW, Kim JC, Rhyu MG and Ro JY: Epstein-barr virus-positive gastric carcinoma demonstrates frequent aberrant methylation of multiple genes and constitutes CpG island methylator phenotype-positive gastric carcinoma. Am J Pathol. 160:787–794. 2002. View Article : Google Scholar : PubMed/NCBI

28 

Xu D, Qu L, Hu J, Li G, Lv P, Ma D, Guo M and Chen Y: Transmembrane protein 106A is silenced by promoter region hypermethylation and suppresses gastric cancer growth by inducing apoptosis. J Cell Mol Med. 18:1655–1666. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Kim HG, Lee S, Kim DY, Ryu SY, Joo JK, Kim JC, Lee KH and Lee JH: Aberrant methylation of DNA mismatch repair genes in elderly patients with sporadic gastric carcinoma: A comparison with younger patients. J Surg Oncol. 101:28–35. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Luco RF, Pan Q, Tominaga K, Blencowe BJ, Pereira-Smith OM and Misteli T: Regulation of alternative splicing by histone modifications. Science. 327:996–1000. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Nishiyama A, Yamaguchi L, Sharif J, Johmura Y, Kawamura T, Nakanishi K, Shimamura S, Arita K, Kodama T, Ishikawa F, et al: Uhrf1-dependent H3K23 ubiquitylation couples maintenance DNA methylation and replication. Nature. 502:249–253. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Barski A, Cuddapah S, Cui K, Roh TY, Schones DE, Wang Z, Wei G, Chepelev I and Zhao K: High-resolution profiling of histone methylations in the human genome. Cell. 129:823–837. 2007. View Article : Google Scholar : PubMed/NCBI

33 

Archer SY and Hodin RA: Histone acetylation and cancer. Curr Opin Genet Dev. 9:171–174. 1999. View Article : Google Scholar : PubMed/NCBI

34 

Ota T, Suto S, Katayama H, Han ZB, Suzuki F, Maeda M, Tanino M, Terada Y and Tatsuka M: Increased mitotic phosphorylation of histone H3 attributable to AIM-1/Aurora-B overexpression contributes to chromosome number instability. Cancer Res. 62:5168–5177. 2002.PubMed/NCBI

35 

Ke Q, Davidson T, Chen H, Kluz T and Costa M: Alterations of histone modifications and transgene silencing by nickel chloride. Carcinogenesis. 27:1481–1488. 2006. View Article : Google Scholar : PubMed/NCBI

36 

Dawson MA and Kouzarides T: Cancer epigenetics: From mechanism to therapy. Cell. 150:12–27. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Cai L, Ma X, Huang Y, Zou Y and Chen X: Aberrant histone methylation and the effect of Suv39H1 siRNA on gastric carcinoma. Oncol Rep. 31:2593–2600. 2014.PubMed/NCBI

38 

Yekta S, Shih IH and Bartel DP: MicroRNA-directed cleavage of HOXB8 mRNA. Science. 304:594–596. 2004. View Article : Google Scholar : PubMed/NCBI

39 

Hutvagner G and Zamore PD: A microRNA in a multiple-turnover RNAi enzyme complex. Science. 297:2056–2060. 2002. View Article : Google Scholar : PubMed/NCBI

40 

Lewis BP, Burge CB and Bartel DP: Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell. 120:15–20. 2005. View Article : Google Scholar : PubMed/NCBI

41 

Liu K, Qian T, Tang L, Wang J, Yang H and Ren J: Decreased expression of microRNA let-7i and its association with chemotherapeutic response in human gastric cancer. World J Surg Oncol. 10:2252012. View Article : Google Scholar : PubMed/NCBI

42 

Inoue T, Iinuma H, Ogawa E, Inaba T and Fukushima R: Clinicopathological and prognostic significance of microRNA-107 and its relationship to DICER1 mRNA expression in gastric cancer. Oncol Rep. 27:1759–1764. 2012.PubMed/NCBI

43 

Wang YY, Ye ZY, Zhao ZS, Li L, Wang YX, Tao HQ, Wang HJ and He XJ: Clinicopathologic significance of miR-10b expression in gastric carcinoma. Hum Pathol. 44:1278–1285. 2013. View Article : Google Scholar : PubMed/NCBI

44 

Hashiguchi Y, Nishida N, Mimori K, Sudo T, Tanaka F, Shibata K, Ishii H, Mochizuki H, Hase K, Doki Y and Mori M: Down-regulation of miR-125a-3p in human gastric cancer and its clinicopathological significance. Int J Oncol. 40:1477–1482. 2012.PubMed/NCBI

45 

Guo LH, Li H, Wang F, Yu J and He JS: The tumor suppressor roles of miR-433 and miR-127 in gastric cancer. Int J Mol Sci. 14:14171–14184. 2013. View Article : Google Scholar : PubMed/NCBI

46 

Liu X, Yu H, Cai H and Wang Y: The expression and clinical significance of miR-132 in gastric cancer patients. Diagn Pathol. 9:572014. View Article : Google Scholar : PubMed/NCBI

47 

Shin JY, Kim YI, Cho SJ, Lee MK, Kook MC, Lee JH, Lee SS, Ashktorab H, Smoot DT, Ryu KW, et al: MicroRNA 135a suppresses lymph node metastasis through down-regulation of ROCK1 in early gastric cancer. PLoS One. 9:e852052014. View Article : Google Scholar : PubMed/NCBI

48 

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

49 

Naito Y, Sakamoto N, Oue N, Yashiro M, Sentani K, Yanagihara K, Hirakawa K and Yasui W: MicroRNA-143 regulates collagen type III expression in stromal fibroblasts of scirrhous type gastric cancer. Cancer Sci. 105:228–235. 2014. View Article : Google Scholar : PubMed/NCBI

50 

Akiyoshi S, Fukagawa T, Ueo H, Ishibashi M, Takahashi Y, Fabbri M, Sasako M, Maehara Y, Mimori K and Mori M: Clinical significance of miR-144-ZFX axis in disseminated tumour cells in bone marrow in gastric cancer cases. Br J Cancer. 107:1345–1353. 2012. View Article : Google Scholar : PubMed/NCBI

51 

Zheng B, Liang L, Wang C, Huang S, Cao X, Zha R, Liu L, Jia D, Tian Q, Wu J, et al: MicroRNA-148a suppresses tumor cell invasion and metastasis by downregulating ROCK1 in gastric cancer. Clin Cancer Res. 17:7574–7583. 2011. View Article : Google Scholar : PubMed/NCBI

52 

Sakamoto N, Naito Y, Oue N, Sentani K, Uraoka N, Zarni Oo H, Yanagihara K, Aoyagi K, Sasaki H and Yasui W: MicroRNA-148a is downregulated in gastric cancer, targets MMP7, and indicates tumor invasiveness and poor prognosis. Cancer Sci. 105:236–243. 2014. View Article : Google Scholar : PubMed/NCBI

53 

Katada T, Ishiguro H, Kuwabara Y, Kimura M, Mitui A, Mori Y, Ogawa R, Harata K and Fujii Y: microRNA expression profile in undifferentiated gastric cancer. Int J Oncol. 34:537–542. 2009.PubMed/NCBI

54 

Chen G, Shen ZL, Wang L, Lv CY, Huang XE and Zhou RP: Hsa-miR-181a-5p expression and effects on cell proliferation in gastric cancer. Asian Pac J Cancer Prev. 14:3871–3875. 2013. View Article : Google Scholar : PubMed/NCBI

55 

Tan Z, Jiang H, Wu Y, Xie L, Dai W, Tang H and Tang S: miR-185 is an independent prognosis factor and suppresses tumor metastasis in gastric cancer. Mol Cell Biochem. 386:223–231. 2014. View Article : Google Scholar : PubMed/NCBI

56 

Brenner B, Hoshen MB, Purim O, David MB, Ashkenazi K, Marshak G, Kundel Y, Brenner R, Morgenstern S, Halpern M, et al: MicroRNAs as a potential prognostic factor in gastric cancer. World J Gastroenterol. 17:3976–3985. 2011. View Article : Google Scholar : PubMed/NCBI

57 

Tang H, Deng M, Tang Y and Xie X, Guo J, Kong Y, Ye F, Su Q and Xie X: miR-200b and miR-200c as prognostic factors and mediators of gastric cancer cell progression. Clin Cancer Res. 19:5602–5612. 2013. View Article : Google Scholar : PubMed/NCBI

58 

Yang TS, Yang XH, Wang XD, Wang YL, Zhou B and Song ZS: MiR-214 regulate gastric cancer cell proliferation, migration and invasion by targeting PTEN. Cancer Cell Int. 13:682013. View Article : Google Scholar : PubMed/NCBI

59 

Wang M, Zhao C, Shi H, Zhang B, Zhang L, Zhang X, Wang S, Wu X, Yang T, Huang F, et al: Deregulated microRNAs in gastric cancer tissue-derived mesenchymal stem cells: Novel biomarkers and a mechanism for gastric cancer. Br J Cancer. 110:1199–1210. 2014. View Article : Google Scholar : PubMed/NCBI

60 

Wang W, Li F, Zhang Y, Tu Y, Yang Q and Gao X: Reduced expression of miR-22 in gastric cancer is related to clinicopathologic characteristics or patient prognosis. Diagn Pathol. 8:1022013. View Article : Google Scholar : PubMed/NCBI

61 

Liu K, Li G, Fan C, Diao Y, Wu B and Li J: Increased expression of MicroRNA-221 in gastric cancer and its clinical significance. J Int Med Res. 40:467–474. 2012. View Article : Google Scholar : PubMed/NCBI

62 

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

63 

Yan Z, Xiong Y, Xu W, Gao J, Cheng Y, Wang Z, Chen F and Zheng G: Identification hsa-miR-335 as a prognostic signature in gastric cancer. PLoS One. 7:e400372012. View Article : Google Scholar : PubMed/NCBI

64 

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 : PubMed/NCBI

65 

Osawa S, Shimada Y, Sekine S, Okumura T, Nagata T, Fukuoka J and Tsukada K: MicroRNA profiling of gastric cancer patients from formalin-fixed paraffin-embedded samples. Oncol Lett. 2:613–619. 2011.PubMed/NCBI

66 

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

67 

Iwaya T, Fukagawa T, Suzuki Y, Takahashi Y, Sawada G, Ishibashi M, Kurashige J, Sudo T, Tanaka F, Shibata K, et al: Contrasting expression patterns of histone mRNA and microRNA 760 in patients with gastric cancer. Clin Cancer Res. 19:6438–6449. 2013. View Article : Google Scholar : PubMed/NCBI

68 

He W, Li Y, Chen X, Lu L, Tang B, Wang Z, Pan Y, Cai S, He Y and Ke Z: miR-494 acts as an anti-oncogene in gastric carcinoma by targeting c-myc. J Gastroenterol Hepatol. 29:1427–1434. 2014. View Article : Google Scholar : PubMed/NCBI

69 

Stenholm L, Stoehlmacher-Williams J, Al-Batran SE, Heussen N, Akin S, Pauligk C, Lehmann S, Senff T, Hofheinz RD, Ehninger G, et al: Prognostic role of microRNA polymorphisms in advanced gastric cancer: A translational study of the Arbeitsgemeinschaft Internistische Onkologie (AIO). Ann Oncol. 24:2581–2588. 2013. View Article : Google Scholar : PubMed/NCBI

70 

Tsujiura M, Ichikawa D, Komatsu S, Shiozaki A, Takeshita H, Kosuga T, Konishi H, Morimura R, Deguchi K, Fujiwara H, et al: Circulating microRNAs in plasma of patients with gastric cancers. Br J Cancer. 102:1174–1179. 2010. View Article : Google Scholar : PubMed/NCBI

71 

Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ and Lötvall JO: Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 9:654–659. 2007. View Article : Google Scholar : PubMed/NCBI

72 

Liu R, Zhang C, Hu Z, Li G, Wang C, Yang C, Huang D, Chen X, Zhang H, Zhuang R, et al: A five-microRNA signature identified from genome-wide serum microRNA expression profiling serves as a fingerprint for gastric cancer diagnosis. Eur J Cancer. 47:784–791. 2011. View Article : Google Scholar : PubMed/NCBI

73 

Liu H, Zhu L, Liu B, Yang L, Meng X, Zhang W, Ma Y and Xiao H: Genome-wide microRNA profiles identify miR-378 as a serum biomarker for early detection of gastric cancer. Cancer Lett. 316:196–203. 2012. View Article : Google Scholar : PubMed/NCBI

74 

Ponting CP, Oliver PL and Reik W: Evolution and functions of long noncoding RNAs. Cell. 136:629–641. 2009. View Article : Google Scholar : PubMed/NCBI

75 

Gupta RA, Shah N, Wang KC, Kim J, Horlings HM, Wong DJ, Tsai MC, Hung T, Argani P, Rinn JL, et al: Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature. 464:1071–1076. 2010. View Article : Google Scholar : PubMed/NCBI

76 

Yang Z, Zhou L, Wu LM, Lai MC, Xie HY, Zhang F and Zheng SS: Overexpression of long non-coding RNA HOTAIR predicts tumor recurrence in hepatocellular carcinoma patients following liver transplantation. Ann Surg Oncol. 18:1243–1250. 2011. View Article : Google Scholar : PubMed/NCBI

77 

Gutschner T, Hammerle M, Eissmann M, Hsu J, Kim Y, Hung G, Revenko A, Arun G, Stentrup M, Gross M, et al: The noncoding RNA MALAT1 is a critical regulator of the metastasis phenotype of lung cancer cells. Cancer Res. 73:1180–1189. 2013. View Article : Google Scholar : PubMed/NCBI

78 

Yang F, Bi J, Xue X, Zheng L, Zhi K, Hua J and Fang G: Up-regulated long non-coding RNA H19 contributes to proliferation of gastric cancer cells. FEBS J. 279:3159–3165. 2012. View Article : Google Scholar : PubMed/NCBI

79 

Xu ZY, Yu QM, Du YA, Yang LT, Dong RZ, Huang L, Yu PF and Cheng XD: Knockdown of long non-coding RNA HOTAIR suppresses tumor invasion and reverses epithelial-mesenchymal transition in gastric cancer. Int J Biol Sci. 9:587–597. 2013. View Article : Google Scholar : PubMed/NCBI

80 

Yang F, Xue X, Zheng L, Bi J, Zhou Y, Zhi K, Gu Y and Fang G: Long non-coding RNA GHET1 promotes gastric carcinoma cell proliferation by increasing c-Myc mRNA stability. FEBS J. 281:802–813. 2014. View Article : Google Scholar : PubMed/NCBI

81 

Lee KS, Park JL, Lee K, Richardson LE, Johnson BH, Lee HS, Lee JS, Kim SB, Kwon OH, Song KS, et al: nc886, a non-coding RNA of anti-proliferative role, is suppressed by CpG DNA methylation in human gastric cancer. Oncotarget. 5:3944–3955. 2014. View Article : Google Scholar : PubMed/NCBI

82 

Xu TP, Huang MD, Xia R, Liu XX, Sun M, Yin L, Chen WM, Han L, Zhang EB, Kong R, et al: Decreased expression of the long non-coding RNA FENDRR is associated with poor prognosis in gastric cancer and FENDRR regulates gastric cancer cell metastasis by affecting fibronectin1 expression. J Hematol Oncol. 7:632014. View Article : Google Scholar : PubMed/NCBI

83 

Han Y, Ye J, Wu D, Wu P, Chen Z, Chen J, Gao S and Huang J: LEIGC long non-coding RNA acts as a tumor suppressor in gastric carcinoma by inhibiting the epithelial-to-mesenchymal transition. BMC Cancer. 14:9322014. View Article : Google Scholar : PubMed/NCBI

84 

Okugawa Y, Toiyama Y, Hur K, Toden S, Saigusa S, Tanaka K, Inoue Y, Mohri Y, Kusunoki M, Boland CR and Goel A: Metastasis-associated long non-coding RNA drives gastric cancer development and promotes peritoneal metastasis. Carcinogenesis. 35:2731–2739. 2014. View Article : Google Scholar : PubMed/NCBI

85 

Xu MD, Qi P, Weng WW, Shen XH, Ni SJ, Dong L, Huang D, Tan C, Sheng WQ, Zhou XY and Du X: Long non-coding RNA LSINCT5 predicts negative prognosis and exhibits oncogenic activity in gastric cancer. Medicine (Baltimore). 93:e3032014. View Article : Google Scholar : PubMed/NCBI

86 

Arita T, Ichikawa D, Konishi H, Komatsu S, Shiozaki A, Shoda K, Kawaguchi T, Hirajima S, Nagata H, Kubota T, et al: Circulating long non-coding RNAs in plasma of patients with gastric cancer. Anticancer Res. 33:3185–3193. 2013.PubMed/NCBI

Related Articles

Journal Cover

May-2017
Volume 13 Issue 5

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Sonohara F, Inokawa Y, Hayashi M, Kodera Y and Nomoto S: Epigenetic modulation associated with carcinogenesis and prognosis of human gastric cancer (Review). Oncol Lett 13: 3363-3368, 2017
APA
Sonohara, F., Inokawa, Y., Hayashi, M., Kodera, Y., & Nomoto, S. (2017). Epigenetic modulation associated with carcinogenesis and prognosis of human gastric cancer (Review). Oncology Letters, 13, 3363-3368. https://doi.org/10.3892/ol.2017.5912
MLA
Sonohara, F., Inokawa, Y., Hayashi, M., Kodera, Y., Nomoto, S."Epigenetic modulation associated with carcinogenesis and prognosis of human gastric cancer (Review)". Oncology Letters 13.5 (2017): 3363-3368.
Chicago
Sonohara, F., Inokawa, Y., Hayashi, M., Kodera, Y., Nomoto, S."Epigenetic modulation associated with carcinogenesis and prognosis of human gastric cancer (Review)". Oncology Letters 13, no. 5 (2017): 3363-3368. https://doi.org/10.3892/ol.2017.5912