miRNA: The nemesis of gastric cancer (Review)

  • Authors:
    • Xiaohui Xu
    • Xiaodong Yang
    • Chungen Xing
    • Shuyu Zhang
    • Jianping Cao
  • View Affiliations

  • Published online on: June 26, 2013     https://doi.org/10.3892/ol.2013.1428
  • Pages: 631-641
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

microRNAs (miRNAs) are a group of small non‑coding RNAs that are ~22 (18 to 25) nucleotides (nt) long and have been associated with a variety of diseases, including cancer. Increasing evidence indicates that miRNAs are essential in the development, diagnosis, treatment and prognosis of a variety of tumors. The utility of miRNAs as biomarkers for diagnosis and of target molecules for the treatment of cancers is increasingly being recognized. With the discovery of circulating miRNAs, a non‑invasive approach for the diagnosis and treatment of cancer has been identified. This review summarizes the role of miRNAs in the development of different tumors, as well as a variety of other biological events. Moreover, this review focuses on analyzing the function and mechanism of gastric cancer‑related miRNAs and investigates the importance of circulating miRNAs in gastric cancer, as well as their origin. Finally, this review lists a number of the problems that must be solved prior to miRNAs being used as reliable non‑invasive tools for the diagnosis, treatment and prognosis of gastric cancer.

1. Introduction

The incidence of gastric cancer ranks fourth in males and fifth in females, while the mortality ranks third in males and fifth in females among all cancers worldwide (1). The incidence and mortality of gastric cancer in developing countries in males and females are much higher compared with developed countries (1). With improvements in diagnostic and therapeutic technology, the survival time of patients with early gastric cancer has been greatly extended. However, the prospects for the treatment of patients with advanced gastric cancer are not optimistic. At present, there are a number of non-surgical treatment approaches, although surgery remains the primary method for the radical treatment of gastric cancer.

microRNAs (miRNAs) are becoming increasingly recognized as important in cancers, including gastric cancer. To date, >1,400 miRNAs have been described in humans (2). Yu et al observed that in gastric cancer cell lines 17 miRNAs were upregulated and 146 miRNAs were downregulated compared with normal gastric mucosa (3). miRNAs specific to the tumorigenesis and development of tumors have become hot spots for cancer research. miRNAs are associated with the development of tumor biology, as well as the diagnosis, treatment and detection of prognosis of tumors. In addition, the reliability of miRNA detection in the circulatory system of tumor patients has also been demonstrated. One study showed that miRNA expression is closely associated with the tumorigenesis, progression and prognosis of gastric cancer (4), and the detection of miRNA in the circulating system may offer new biomarkers for gastric cancer (5). Consequently, detecting miRNAs in the serum/plasma may become a non-invasive pathway for providing a basis for the treatment, diagnosis and prognosis of gastric cancer.

2. Discovery of miRNAs

In 1993, Victor Ambros first identified miRNA in C. elegans(6). Simultaneously, Gary Ruvkun identified the gene Lin-14 as the first miRNA target (7). These two important findings demonstrated a new post-transcriptional gene regulation mechanism. Approximately seven years later, the importance of miRNAs was realized when Ruvkun and Horvitz identified another miRNA in C. elegans(8), and when interest was focused on another short-chain RNA, small interfering RNA (siRNA; involved in the process of RNA interference and related phenomena in plants and animals). In 2001, three research groups from different countries all identified 21–22 nucleotide (nt) non-coding small RNA molecules in C. elegans, Drosophila and the human body (9). These single-stranded small RNA molecules with spatial and temporal expression were different from the previously reported siRNA detected in the interference pathway (RNA interference; RNAi) and were subsequently named miRNA.

3. Biosynthesis and biological function of miRNAs

miRNAs are encoded by specific miRNA genes in the genomic DNA, which are first transcribed by RNA polymerase II into a stem-loop that is ~500 to 3,000 bp long (10). This initial transcript is known as pri-miRNA. These pri-miRNAs in the nucleus are further transformed into ~60 to 70-nt hairpin-shaped precursor miRNAs (pre-miRNAs) by the Drosha enzyme (a type of RNase III enzyme). Precision processing of the pre-miRNA is extremely important as this produces the ‘seed’ region of miRNA, which is the determining factor leading to gene silencing. This ‘seed’ region is the target area on the mRNA that is complementary to the 2–7 positions of the antisense miRNA oligonucleotide. The pre-miRNAs are exported into the cytoplasm by Exportin-5 (Exp-5) and cleaved by Dicer to generate 20–24 nt RNA duplexes, one strand of which is loaded into the Argonaute-containing RNA-induced silencing complex (RISC). miRNA-RISC complexes are able to silence target mRNAs via imperfect complementarity with sequences located in the 5′-UTR, coding sequences and, most commonly, the 3′-UTR (11). When pre-miRNA is transported to the cytoplasm by the transport protein Exp-5, the pre-miRNA hairpin in the cytoplasm is cleaved into two strands by Dicer (RNase III), with the release of two complementary 22-base nt chains. Subsequent to being processed by the enzyme Dicer, the antisense miRNA chain and mRNA target region complementary base pair together to form a complex with Argonaut proteins. Argonaut guides the miRNA chain to reach the target sequence on the mRNA and combines with RISC (12,13). RISC combines with target mRNA at the 3′-UTR and the miRNA base pairs with the target mRNA, causing target mRNA cleavage and inhibition of protein expression (1317).

miRNAs regulate gene expression at the post-transcriptional level (1820). Furthermore, miRNAs are members of the small RNA family, which includes small nuclear RNA (snRNA) involved in mRNA splicing (21), small nucleolar RNAs (snoRNAs), which directly modify ribosomal RNA (22), and siRNAs, which are generated by long double-stranded RNA precursors. siRNAs also functionally regulate gene expression, similar to miRNAs (23). miRNAs are highly-conserved endogenous RNA molecules that occur in the genome of animals, plants, fungi and viruses (24). It is currently estimated that miRNAs account for ~1% of the human genome (25) and they have been demonstrated to play crucial roles in the human body, including roles in growth (8), cell proliferation and cell apoptosis (26,27).

4. miRNAs and cancer

Tumor-specific miRNAs

miRNAs have been associated with a variety of diseases, including cancer. Increasing evidence has demonstrated the importance of miRNAs in regulating biological characteristics common to various tumors, including self-growth signals, insensitivity to anti-growth signals, abnormal apoptosis, unlimited replication potential, sustained induction of angiogenesis and invasion and metastasis organization (28). Numerous researchers have identified tumor-specific miRNA signatures that accurately distinguish malignant tumors from various types of benign tissues, and have demonstrated that certain miRNAs are carcinogenic depending on the other gene mutations in the tumors (29). miRNA regulation in tumor cell lines directly affects cell proliferation and apoptosis, and similarly, a number of studies have identified a link between abnormal miRNA expression and intracellular signal transduction pathway abnormalities, as well as tumorigenesis (3034). For example, miR-9 is activated by YC/MYCN, which induces cancer metastasis by regulating the metastasis suppressor protein E-cadherin (35), while miR-449a causes retinoblastoma (Rb)-dependent cell cycle arrest and cellular senescence in prostate cancer (36). Table I provides an overview of the expression of the same miRNAs in different tumor cells, the expression of different miRNAs in the same tumor cells and the associated target proteins and/or target genes of the miRNAs (3548).

Table I

Tumor-associated miRNAs and their targets.

Table I

Tumor-associated miRNAs and their targets.

miRNAUp- or down-regulationTarget gene(s)Target proteinsCancerReferences
miR-9UpYC/MYCNE-cadherinBreast cancer cells(35)
miR-449aDownRb-Prostate cancer cells(36)
miR-223DownE2F1C/EBPαAcute myeloid leukemia(37)
miR-34a and miR-34b/cDownP53CDK4/6, Cyclin E2, MET and Bcl-2Neuroblastoma, colorectal, lung, pancreatic, breast and ovarian cancer, bladder and renal cell carcinoma, liver carcinoma, oral squamous cell carcinoma,(38,39)
miR-21UpPdcd4u-PARColon cancer(40,41)
miR-17–92UpK-Ras and c-MycTsp1 and CTGFColon cancer(42)
miR-145DownTP53ER-αBreast cancer cells(43)
miR- 483-3 and miR-421UpDPC4/Smad4DPC4/Smad4Pancreatic cancer(44,45)
miR-224UpCDC42, CDH1, PAK2, BCL-2 and MAPK1CDC42, CDH1, PAK2, BCL-2 and MAPK1Hepatocellular carcinoma(46)
miR-106b-25UpCaspase-7Focal adhesionProstate cancer(47)
miR-34DownMet and Bcl-2p53Lung adenocarcinoma(48)

[i] miRNA, microRNA; Rb, retinoblastoma.

miRNAs constitute a new class of molecules that are able to promote the formation of cancer through interactions with oncogenes and/or tumor suppressor genes (49). However, different miRNAs have different signaling pathways and the target proteins/genes through which they affect the biological changes of cancer.

Circulating miRNAs in cancer

The aforementioned examples show that miRNAs have the potential to function as biomarkers for the diagnosis, treatment and prognosis of cancer, although miRNA expression profiling may not be used without a tumor biopsy. Therefore, the identification of non-invasive methods for the diagnosis and treatment of cancer has become a goal for cancer research. A number of studies have proposed that the characteristics of the cancer genome, genetic and epigenetic, may be detected in the serum and plasma of cancer patients (50). Therefore, the detection of plasma miRNA is a useful approach for the diagnosis, treatment and monitoring of the prognosis for cancer patients. miRNAs are involved in the development and differentiation of tumors and the lineage-dependent mechanism. Consequently, this becomes the basis for tumor development. Since miRNA detection by PCR is highly sensitive and of low complexity compared with protein detection, modifications of miRNAs are possible and synthetic high-affinity miRNA “capture” reagents are available, miRNAs may have a high value as biomarkers. In addition, circulating miRNAs in human blood and other body fluids are plentiful, have remarkable stability and may be measured in routine clinical diagnosis. Therefore, circulating miRNAs are considered to be extremely promising cancer biomarkers (5153). At present, an increasing number of scholars are attempting to identify more specific and highly sensitive miRNAs from the plasma of various tumor patients for use as non-invasive biomarkers for the diagnosis, treatment and prognostic evaluation of tumors. Such a simple, feasible and acceptable method is likely to optimize the therapeutic effect for patients. In recent years, there are have been a number of studies on plasma miRNAs as biomarkers. Lawrie et al identified the first circulating miRNA in 2008 (54). The study observed that miR-21 expression was associated with the relapse-free survival of patients with diffuse large B-cell lymphoma (DLBCL) and suggested that miRNAs have potential as non-invasive diagnostic markers for DLBCL and possibly other cancers (54). Subsequently, the plasma-based detection of miRNA has become a research hot spot. Table II provides an overview of the functions of plasma or serum miRNAs in various types of cancer (5564).

Table II

Functions of plasma or serum miRNAs in different cancer.

Table II

Functions of plasma or serum miRNAs in different cancer.

miRNASerum/plasmaAUCSensitivity (%)Specificity (%)FunctionsCancerReferences
miR-21Plasma0.636489Discriminate cancer or normalPancreatic ductal adenocarcinoma(55)
miR-2100.62
miR-1550.60
miR-196a0.66
miR-141Serum0.8860100Discriminate cancer or normalProstate cancer(56)
miR-141Plasma0.887790Relevant to tumor stageColorectal cancer stage I–III(57)
9177Colorectal cancer stage IV
miR-92Plasma0.888970Discriminate cancer or normalColorectal cancer(58)
miR-29a and miR-92aPlasma0.888385Discriminate cancer or normalAdvanced colorectal cancer(59)
microRNA-29aSerum0.887575Relevant to liver metastasisColorectal cancer(60,61)
miR-221Plasma0.888641Relevant to plasma level of miR-221 and p53Colorectal cancer(62)
miR-15b and miR-27bSerum0.9810084Discriminate cancer or normalNon-small cell lung cancer(63)
miR-125bSerum0.669638Relevant to poor prognosisNon-small-cell lung cancer stage I–II(64)
0.849366Non-small-cell lung cancer stage III
0.909567Non-small-cell lung cancer stage IV

[i] AUC, area under the curve; miRNA, microRNA.

All current research results show that miRNAs have promise as novel non-invasive biomarkers for the elimination of false-positive and -negative results in routine testing. However, the source of the circulating miRNAs and how these circulating miRNAs function must be identified for the proper use of circulating miRNA biomarkers in evidence-based medicine (65).

5. miRNAs and gastric cancer

Since miRNAs have been shown to be significant factors during the progression of gastric cancer, global miRNA expression profiles have been performed using microarrays, real-time PCR or next-generation sequencing approaches. A significant number of the functions and mechanisms of miRNAs in the process of the development of gastric cancer have been identified.

Biological effects of miRNAs in gastric cancer

The incidence of gastric cancer is the result of interactions between Helicobacter pylori, genetic predisposition, eating habits and environmental factors, although oncogenes and tumor suppressor genes eventually determine gastric carcinogenesis. However, miRNAs are not only involved in oncogene-induced tumors, but also in tumors caused by tumor suppressor genes; their role in gastric carcinogenesis complements and enriches the mechanism of tumorigenesis. Table III provides an overview of the roles of various miRNAs in the proliferation of gastric cancer (6682). Considerable progress has also been made in research into the mechanism of action of miRNAs in gastric cancer cell invasion and metastasis. Table IV provides an overview of the role of various miRNAs in the invasion and metastasis of gastric cancer (8390).

Table III

Role of different miRNAs in the proliferation of gastric cancer.

Table III

Role of different miRNAs in the proliferation of gastric cancer.

miRNAUp- or down-regulationRegulatory pathwayInhibit or promote biological functionRelated biological functionDetection methodCell linesReferences
miR-449UpActivating p53 pathwayInhibitProliferationTaqman miRNA assaysSNU638 and MKN74(66)
miR-21UpPTEN expressionPromoteProliferation and invasionReal-time PCRBGC-823(67,68)
miR-9 and miR-433DownGRB2 and RAB34 expressionPromoteProliferationqRT-PCR PCRSGC7901(69)
miR-222Upp27 and p57PromoteProliferationReal-timeSNU-638, AGS and MKN-28(70)
miR-375UpPDK1/Akt signalling pathwayInhibitProliferationmiRNA MicroarrayNUGC3, AZ521 and MKN74(71)
miR-375UpJAK2 oncogene PDK1InhibitProliferationqRT-PCRMGC-803, BGC-823, SGC-7901, et al(72,73)
miR-9UpNF-κB1geneInhibitProliferationStem-loop RT-PCRMGC803(74)
miR-141DownFGFR2 signalling pathwayPromoteProliferationqRT-PCRMGC-803, HGC-27, SGC-7901, et al(75)
miR-199aUpSmad4TGF-β signalling pathwayPromoteGrowth and survivalReal-time RT-PCRSNU-16, AGS, BGC-823(76)
miR-181aUpKLF6 genePromoteProliferation, colony formation, migration, and invasionqRT-PCRSGC-7901(77)
miR-181bUpCREB1InhibitProliferation and colony formation--(78)
miR-124UpSPHK1InhibitProliferation and tumourigenicityqRT-PCRMGC-803 and SGC-7901(79)
miR-409-3pUpPHF10InhibitProliferationqRT-PCRSGC-7901(80)
miR-182UpCREB1InhibitProliferation and colony formationqRT-PCRMGC-803, BGC-823 and SGC-7901(81)
miR-223UpFBXW7/hCdc4 genePromoteProliferationReal-time RT-PCRSGC7901(82)

[i] miRNA, microRNA; CREB1, cAMP responsive element binding protein 1; SPHK1, sphingosine kinase 1.

Table IV

Role of different miRNAs in the invasion and metastasis of gastric cancer.

Table IV

Role of different miRNAs in the invasion and metastasis of gastric cancer.

miRNAUp- or down- regulationRegulatory pathwayInhibit or promote biological functionRelated biological functionDetection methodCell linesReferences
miR-409-3pUpRDXInhibitInvasion and metastasisRT-PCRSGC-7901, MKN-45 and GES-1(83)
miR-27UpEMTPromoteMetastasisRT-PCRAGS(84)
miR-625Up LIMS1-ILK-parvinInhibitMigration and invasionqRT-PCRSGC-7901 and MKN-45(85)
miR-625Down LIMS1-ILK-parvinPromoteMigration and invasionqRT-PCRGES-1 and NCI-N87(85)
miR-495 and miR-551aUpPRL-3InhibitMigration and invasionqRT-PCRSGC7901, MKN45, MKN28, and BGC823(86)
miR-10bUpHOXD10RhoC-AKT signaling pathwayPromoteInvasionqRT-PCRGES-1(87)
miR-196bUpETS2PromoteMigration and invasionqRT-PCRAGS, HR and the embryonic kidney cell, 293T(88)
miR-145UpN-cadherin proteinInhibitInvasion and metastasisRT-PCRGES-1 and NCI-N87(89)
miR-610UpVASPInhibitMigration and invasionRT-PCRBGC-823 and MKN-28(90)

[i] miRNA, microRNA.

miRNAs are pivotal in the development of gastric cancer. It is of note that certain miRNAs do not function alone and various clusters of miRNAs share functional associations. Therefore, it is extremely important to clarify whether their role is independent or synergistic with other miRNAs when the biological functions of certain clusters of miRNAs are studied.

miRNAs and gastric cancer pathology

The detection of specific miRNAs may become a potential field in marker development (91). Studies have shown that different tumors have specific miRNA expression patterns and that a variety of tumors have specific miRNA expression profiles (92). A comparative analysis of miRNA expression profiles between tumors and normal tissue suggests a new approach for the genetic diagnosis of tumors. At present, the role of miRNAs in the diagnosis and treatment of gastric cancer has increased significantly worldwide. miR-21 appears to be important in tumorigenesis due to its upregulation in almost all types of human cancer. Furthermore miR-21 is overexpressed in 92% (34/37) of gastric cancer samples and is thus considered to be a promising novel biomarker for gastric cancer, as well as lymph node metastasis (93,94). In gastric cancer, upregulated miRNA-106a and miR-143, as well as downregulated miR-203, are associated with gastric tumor size, stage, lymph nodes and distant metastasis. Consequently, they may become a new type of potential biological diagnostic molecule for gastric cancer (9597). A study has shown that the reduced expression of miR-574–3p occurs mainly in the early stages of gastric cancer or in cancers with a high level of differentiation, suggesting that it may be used as a marker for mild cases of gastric cancer (98).

Therefore, miRNAs are important in pathological staging, lymph node metastasis and distant metastasis of gastric cancer. If miRNA changes specific to the development of gastric cancer are identified and the stage of gastric cancer is determined, this approach may become a pathway for aiding in the diagnosis of gastric cancer by detecting this type of miRNA. However, further important information for the clinical diagnosis of gastric cancer should be provided and more clinical data must be collected to determine the feasibility.

miRNAs and gastric cancer diagnosis and treatment

At present, the main methods of treatment for gastric cancer are surgery, chemotherapy, radiotherapy and biologically targeted therapy, of which surgery is the most important. In numerous cases, a tumor diagnostic tool also provides a therapeutic approach. There are a number of studies on gastric cancer treatment through the targeting of miRNA. For example, cell proliferation, migration and invasion in gastric cancer cells have been demonstrated to be significantly increased following miR-181b transfection, so miR-181b may be a potential molecular target for anticancer therapeutics in gastric cancer (99). Zhao et al revealed that miR-7 functions as an anti-metastatic microRNA in gastric cancer by targeting the insulin-like growth factor-1 receptor (100). Targeting the miR-7/IGF1R/Snail axis may be useful as a therapeutic approach for blocking gastric cancer metastasis. miRNAs produce a negative regulation of proteins by combining with the target mRNA, and it is possible to take advantage of their features to up- or downregulate the expression of key tumor-associated genes for therapeutic purposes. miRNAs, either as oncogenes or tumor suppressor genes, regulate the biological characteristics of gastric cancer. Inhibiting miRNAs as oncogenes or increasing miRNAs as tumor suppressor genes using antisense oligonucleotides and siRNA technology presents a new approach to biological therapy for gastric cancer. The identification of specific tumor-associated miRNA coding genes provides new targets for cancer gene therapy. Studies to identify drug targets for miRNAs, as well as small molecule compounds that are able to inhibit miRNA activity, are being conducted by predicting three-dimensional structures of the miRNA (101). Certain studies have investigated the use of anti-miRNA antisense oligonucleotides (AMOs) to reduce or knock out overexpressed tumor-associated miRNAs, producing tumor suppressor effects. The knockout of miR-21 by AMOS causes gastric cancer cell proliferation to be reduced significantly and apoptosis to increase significantly (67). The downregulation of prohibitin by miR-27a may explain why the suppression of miR-27a inhibits gastric cancer cell growth, further supporting the hypothesis that miR-27a functions as an oncogene (102). The inhibition of miR-421 expression has been shown to decrease the growth of MGC-803 and SGC-7901 gastric cancer cells in vitro, upregulating the expression of its cancer-associated target genes, CBX7 and RBMXL1 (103). By contrast, the expression of tumor suppressor miRNA, induced by miRNA mimics and delivered by virus or liposome, gradually reduced the growth of gastric tumors (104). miR-34 is has been demonstrated to be involved in the downstream p53 pathway and is a potential tumor suppressor of the target genes Notch, HMGA2 and Bcl-2, which are involved in cancer stem cell self-renewal and survival (105). The miR-34 mimics or infection with the lentivirus function as miR-34. A study has shown that miR-34 is able to impair cell growth, cause the accumulation of the cells in the G1 phase, increase caspase-3 activation and more significantly, inhibit tumorsphere formation and growth, indicating that the restoration of the tumor-suppressor, miR-34, may provide a novel molecular therapy for p53-mutant gastric cancer (106). Resistance to chemotherapy remains one of the main obstacles for improving the overall survival and quality of life of patients with gastric cancer. Treatment with miRNA may be used as a regulatory tool for tumor cells in the chemotherapy response. Xia et al suggested that miRNAs may be involved in the development of multidrug resistance (MDR) in gastric cancer cells (107). miR-15b and miR-16 are able to modulate the sensitivity of gastric cancer cells to certain anticancer drugs, at least in part, by regulating BCL2 expression (108). So far, the potential of miRNA-based treatment for malignant disease remains largely to be developed. In addition to the previously mentioned research areas, miRNA remains to be investigated in tumor metastasis, angiogenesis and radiation resistance.

Before the application of miRNA-based therapy may occur, there are several major obstacles to overcome. First, miRNAs have multiple targets, producing the risk of accidental off-target effects, and therefore miRNA-based therapy requires careful assessment. Second, the expression of target genes is controlled by several different miRNAs, which may reduce the effect of miRNA-based therapy. Finally, there is a lack of a sufficiently specific and effective transport system for miRNA (109).

Although miRNA-targeted treatment is likely to provide a new approach for the treatment of gastric cancer, effective treatment may require a combinatorial approach to target multiple oncogenic miRNA clusters. If good use is made of multi-targeted miRNA in treatment, particularly when adaptive resistance occurs, it may produce therapeutic effects via a variety of alternative pathways in tumor cells. However, there is a long way to go before miRNAs are suitable as a therapeutic target for clinical application.

miRNAs and gastric cancer prognosis

The prognosis of patients subsequent to treatment is important for each patient and their families. In recent years, attempts have been made to identify prompt detection methods for the prognosis of gastric cancer using the fields of biochemistry, immunohistochemistry and genetic testing (110,111), although a reliable, specific, sensitive and practical detection method has yet to be detected. However, it has been possible to identify a new method for detecting the prognosis of gastric cancer patients through the study of miRNAs. Yu et al and Chen et al detected high expression levels of miR-93 in gastric cancer, particularly in advanced and metastatic cases (3,103). The authors suggested that miR-93 may be critical in the carcinogenesis of gastric cancer. Notably, miR-93, as well as miR-1, may predict a poor survival outcome in gastric cancer patients (3,112,113). Furthermore, Tsai et al demonstrated that miR-196a may be a novel biomarker for detecting gastric cancer and monitoring disease recurrence (114). Multivariate analyses have indicated that low miR-125a-3p expression is an independent prognostic factor for survival, while in vitro assays have demonstrated that miR-125a-3p suppresses the proliferation of gastric cancer cells (115).

Therefore, the continuous investigation of gastric cancer-associated miRNAs may lead them to become new assessment biomarkers following treatment for gastric cancer. These biomarkers may not only increase the effectiveness of the evaluation but may also become the main basis of this evaluation.

6. Serum/plasma miRNAs as biomarkers/signaling molecules for gastric cancer

At present, the majority of the discoveries of gastric cancer-associated miRNAs have been based on gastric carcinoma and adjacent normal tissues. Since this is an invasive approach, it is a significant request to make of patients and not one that is readily accepted. However, great progress has been made in investigating whether serum and plasma miRNAs may be used as non-invasive biomarkers for the diagnosis, treatment and detection of prognosis in gastric cancer patients. Tsujiura et al published a study demonstrating that plasma miRNA assays have several potential clinical uses, including for screening patients at a high risk of gastric cancer and monitoring disease recurrence during the follow-up period after gastrectomy (5). These miRNA biomarkers may be powerful and useful for confirming the completeness of tumor resection and evaluating the efficacy of adjuvant therapies if the clearance of plasma miRNAs may be elucidated (5). In addition, Konishi et al observed that plasma miR-451 and miR-486 may be useful blood-based biomarkers for the screening of gastric cancer. The authors noted that the levels of these two miRNAs decrease in post-operative plasma in 90 and 93% of patients, respectively. However, in comparison with healthy controls, the levels of the two miRNAs were demonstrated to be significantly higher in the plasma of post-operative patients (116). The results of further studies have shown that the levels of circulating miR-17–5p/20a may be a promising non-invasive molecular marker for pathological progression, predicting prognosis and monitoring chemotherapeutic effects for gastric cancer (117). Studies have also shown that serum miR-221, miR-376c, miR-744 and miR-378 have clear potential as novel non-invasive biomarkers for the early detection of gastric cancer, and that miR-378 may be used as a valuable biomarker alone (118,119). These findings have pioneered a new approach for the diagnosis and treatment of gastric cancer and demonstrated that miRNAs are likely to become a routine examination indicator for the development and prognosis of gastric cancer, similar to CA199, p53 or other tumor markers.

As mentioned previously, miRNAs are extremely stable in blood plasma and serum (56). Consequently, this makes miRNA levels as suitable for testing for clinical diagnosis as tumor markers and biomarkers. The source of the miRNAs circulating in blood samples remains unclear. Chim et al suggested two hypotheses for this: The presence of circulating miRNAs may be due to tumor cell death and lysis or they may be released by tumor cells into the extracellular microenvironment of blood vessels (120). Ohshima et al demonstrated that the amount of total RNA in culture media was relatively higher in the NCI-H69, Lu-135 and Colo205 cell lines compared with in the exosomes, which was consistent with the ‘tumor cell death’ hypothesis. However, the authors also revealed that the let-7 miRNA family is selectively secreted into serum and plasma via exosomes in metastatic gastric cancer cell lines (121), while Tanaka et al demonstrated that the level of miR-92a was decreased in the blood plasma of acute leukemia patients while remaining high in the tissue samples (121). These results demonstrated that miRNAs may serve as signaling molecules transferred by exosomes between tumor cells and serum/plasma.

Most significantly, circulating miRNAs are not only tumor biomarkers for gastric cancer, but also signaling molecules involved in the proliferation, tumorigenesis and metastasis of gastric cancer.

7. Conclusions

In summary, increasing attention is being paid to miRNAs in gastric cancer. To identify the best approach for the diagnosis, treatment and evaluation of the prognosis of gastric cancer via miRNAs, the detection of serum/plasma miRNAs is likely to be the first choice. However, to make this a reality, breakthroughs are required in the following areas: i) Determining the source of circulating miRNAs; ii) validating a classical pathway of circulating miRNA transmittance; iii) clarifying the mechanism of the action of various circulating miRNAs in gastric cancer; iv) determining how the circulating miRNAs in different stages of gastric cancer development are expressed and how miRNAs develop in this development process; v) highly specific and sensitive detection of miRNA pre- and post-operatively, as well as at various stages of gastric cancer; vi) clarifying the mechanism of synergy/antagonism of various circulating miRNAs in the occurrence and development of gastric cancer; and vii) determining the feasibility of certain miRNA target therapies at different stages of gastric cancer according to associated miRNA specificity.

Acknowledgements

The present study was partially supported by the National Natural Science Foundation of China (81172348, 81102078), the Jiangsu Province Health Department Research Project (H201313) and the Suzhou Social Development Fund Project (200823).

References

1 

Jemal A, Bray F, Center MM, Ferlay J, Ward E and Forman D: Global cancer statistics. CA Cancer J Clin. 61:69–90. 2011. View Article : Google Scholar

2 

Schoof CR, Botelho EL, Izzotti A and dos Vasques LR: MicroRNAs in cancer treatment and prognosis. Am J Cancer Res. 2:414–433. 2012.PubMed/NCBI

3 

Yu BQ, Su LP, Li JF, Cai Q, Yan M, Chen XH, et al: microRNA expression signature of gastric cancer cells relative to normal gastric mucosa. Mol Med Rep. 6:821–826. 2012.PubMed/NCBI

4 

Ueda T, Volinia S, Okumura H, Shimizu M, Taccioli C, Rossi S, et al: Relation between microRNA expression and progression and prognosis of gastric cancer: a microRNA expression analysis. Lancet Oncol. 11:136–146. 2010. View Article : Google Scholar : PubMed/NCBI

5 

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

6 

Lee RC, Feinbaum RL and Ambros V: The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell. 75:843–854. 1993.

7 

Wightman B, Ha I and Ruvkun G: Posttranscriptional regulation of the heterochronic gene lin-14 by lin-4 mediates temporal pattern formation in C. elegans. Cell. 75:855–862. 1993. View Article : Google Scholar : PubMed/NCBI

8 

Reinhart BJ, Slack FJ, Basson M, Pasquinelli AE, Bettinger JC, Rougvie AE, et al: The 21-nucleotide let-7 RNA regulates developmental timing in Caenorhabditis elegans. Nature. 403:901–906. 2000. View Article : Google Scholar : PubMed/NCBI

9 

Hutvagner G, McLachlan J, Pasquinelli AE, et al: A cellular function for the RNA-interference enzyme Dicer in the maturation of the let-7 small temporal RNA. Science. 293:834–838. 2001. View Article : Google Scholar : PubMed/NCBI

10 

Han J, Lee Y, Yeom KH, Nam JW, Heo I, Rhee JK, et al: Molecular basis for the recognition of primary microRNAs by the Drosha-DGCR8 complex. Cell. 125:887–901. 2006. View Article : Google Scholar : PubMed/NCBI

11 

Klase Z, Houzet L and Jeang KT: Replication competent HIV-1 viruses that express intragenomic microRNA reveal discrete RNA-interference mechanisms that affect viral replication. Cell Biosci. 1:382011. View Article : Google Scholar : PubMed/NCBI

12 

Bernstein E, Caudy AA, Hammond SM and Hannon GJ: Role for a bidentate ribonuclease in the initiation step of RNA interference. Nature. 409:363–366. 2001. View Article : Google Scholar : PubMed/NCBI

13 

Han J, Lee Y, Yeom KH, Kim YK, Jin H and Kim VN: The Drosha-DGCR8 complex in primary microRNA processing. Genes Dev. 18:3016–3027. 2004. View Article : Google Scholar : PubMed/NCBI

14 

Lee Y, Kim M, Han J, Yeom KH, Lee S, Baek SH and Kim VN: MicroRNA genes are transcribed by RNA polymerase II. EMBO J. 23:4051–4060. 2004. View Article : Google Scholar : PubMed/NCBI

15 

Cai X, Hagedorn CH and Cullen BR: Human microRNAs are processed from capped, polyadenylated transcripts that can also function as mRNAs. RNA. 10:1957–1966. 2004. View Article : Google Scholar : PubMed/NCBI

16 

Kim YK and Kim VN: Processing of intronic microRNAs. EMBO J. 26:775–783. 2007. View Article : Google Scholar : PubMed/NCBI

17 

Kim VN: MicroRNA biogenesis: coordinated cropping and dicing. Nat Rev Mol Cell Biol. 6:376–385. 2005. View Article : Google Scholar : PubMed/NCBI

18 

Kusenda B, Mraz M, Mayer J and Pospisilova S: MicroRNA biogenesis, functionality and cancer relevance. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub. 150:205–215. 2006. View Article : Google Scholar : PubMed/NCBI

19 

Place RF, Li LC, Pookot D, Noonan EJ and Dahiya R: MicroRNA-373 induces expression of genes with complementary promoter sequences. Proc Natl Acad Sci USA. 105:1608–1613. 2008. View Article : Google Scholar : PubMed/NCBI

20 

Vasudevan S, Tong Y and Steitz JA: Switching from repression to activation: microRNAs can up-regulate translation. Science. 318:1931–1934. 2007. View Article : Google Scholar : PubMed/NCBI

21 

Mattaj IW, Tollervey D and Séraphin B: Small nuclear RNAs in messenger RNA and ribosomal RNA processing. FASEB J. 7:47–53. 1993.PubMed/NCBI

22 

Bachellerie JP, Cavaillé J and Hüttenhofer A: The expanding snoRNA world. Biochimie. 84:775–790. 2002. View Article : Google Scholar : PubMed/NCBI

23 

Ambros V, Lee RC, Lavanway A, Williams PT and Jewell D: MicroRNAs and other tiny endogenous RNAs in C. elegans. Curr Biol. 13:807–818. 2003. View Article : Google Scholar : PubMed/NCBI

24 

Du T and Zamore PD: microPrimer: the biogenesis and function of microRNA. Development. 132:4645–4652. 2005. View Article : Google Scholar : PubMed/NCBI

25 

Lim LP, Glasner ME, Yekta S, Burge CB and Bartel DP: Vertebrate microRNA genes. Science. 299:15402003. View Article : Google Scholar : PubMed/NCBI

26 

Chan JA, Krichevsky AM and Kosik KS: MicroRNA-21 is an antiapoptotic factor in human glioblastoma cells. Cancer Res. 65:6029–6033. 2005. View Article : Google Scholar : PubMed/NCBI

27 

Cheng AM, Byrom MW, Shelton J and Ford LP: Antisense inhibition of human miRNAs and indications for an involvement of miRNA in cell growth and apoptosis. Nucleic Acids Res. 33:1290–1297. 2005. View Article : Google Scholar : PubMed/NCBI

28 

Santarpia L, Nicoloso M and Calin GA: MicroRNAs: a complex regulatory network drives the acquisition of malignant cell phenotype. Endocr Relat Cancer. 17:F51–F75. 2010. View Article : Google Scholar : PubMed/NCBI

29 

Mattie MD, Benz CC, Bowers J, et al: Optimized high-throughput microRNA expression profiling provides novel biomarker assessment of clinical prostate and breast cancer biopsies. Mol Cancer. 5:242006. View Article : Google Scholar

30 

Volinia S, Calin GA, Liu CG, Ambs S, Cimmino A, Petrocca F, et al: A microRNA expression signature of human solid tumors defines cancer gene targets. Proc Natl Acad Sci USA. 103:2257–2261. 2006. View Article : Google Scholar : PubMed/NCBI

31 

Grady WM, Parkin RK, Mitchell PS, Lee JH, Kim YH, Tsuchiya KD, et al: Epigenetic silencing of the intronic microRNA hsa-miR-342 and its host gene EVL in colorectal cancer. Oncogene. 27:3880–3888. 2008. View Article : Google Scholar : PubMed/NCBI

32 

Lanza G, Ferracin M, Gafà R, Veronese A, Spizzo R, Pichiorri F, et al: mRNA/microRNA gene expression profile in microsatellite unstable colorectal cancer. Mol Cancer. 6:542007. View Article : Google Scholar : PubMed/NCBI

33 

Gaur A, Jewell DA, Liang Y, Ridzon D, Moore JH, Chen C, et al: Characterization of microRNA expression levels and their biological correlates in human cancer cell lines. Cancer Res. 67:2456–2468. 2007. View Article : Google Scholar : PubMed/NCBI

34 

Saito Y, Suzuki H and Hibi T: The role of microRNAs in gastrointestinal cancers. J Gastroenterol. 44(Suppl 19): 18–22. 2009. View Article : Google Scholar

35 

Ma L, Young J, Prabhala H, Pan E, Mestdagh P, Muth D, et al: miR-9, a MYC/MYCN-activated microRNA, regulates E-cadherin and cancer metastasis. Nat Cell Biol. 12:247–256. 2010.PubMed/NCBI

36 

Noonan EJ, Place RF, Basak S, Pookot D and Li LC: miR-449a causes Rb-dependent cell cycle arrest and senescence in prostate cancer cells. Oncotarget. 1:349–358. 2010.PubMed/NCBI

37 

Pulikkan JA, Dengler V, Peramangalam PS, Peer Zada AA, Müller-Tidow C, Bohlander SK, et al: Cell-cycle regulator E2F1 and microRNA-223 comprise an autoregulatory negative feedback loop in acute myeloid leukemia. Blood. 115:1768–1778. 2010. View Article : Google Scholar : PubMed/NCBI

38 

Hermeking H: The miR-34 family in cancer and apoptosis. Cell Death Differ. 17:193–199. 2010. View Article : Google Scholar : PubMed/NCBI

39 

Vogt M, Munding J, Grüner M, Liffers ST, Verdoodt B, Hauk J, et al: Frequent concomitant inactivation of miR-34a and miR-34b/c by CpG methylation in colorectal, pancreatic, mammary, ovarian, urothelial, and renal cell carcinomas and soft tissue sarcomas. Virchows Arch. 458:313–322. 2011. View Article : Google Scholar : PubMed/NCBI

40 

Allgayer H: Pdcd4, a colon cancer prognostic that is regulated by a microRNA. Crit Rev Oncol Hematol. 73:185–191. 2010. View Article : Google Scholar : PubMed/NCBI

41 

Lu Z, Liu M, Stribinskis V, Klinge CM, Ramos KS, Colburn NH and Li Y: MicroRNA-21 promotes cell transformation by targeting the programmed cell death 4 gene. Oncogene. 27:4373–4379. 2008. View Article : Google Scholar : PubMed/NCBI

42 

Dews M, Homayouni A, Yu D, Murphy D, Sevignani C, Wentzel E, et al: Augmentation of tumor angiogenesis by a Myc-activated microRNA cluster. Nat Genet. 38:1060–1065. 2006. View Article : Google Scholar : PubMed/NCBI

43 

Spizzo R, Nicoloso MS, Lupini L, Lu Y, Fogarty J, Rossi S, et al: miR-145 participates with TP53 in a death-promoting regulatory loop and targets estrogen receptor-alpha in human breast cancer cells. Cell Death Differ. 17:246–254. 2010. View Article : Google Scholar : PubMed/NCBI

44 

Hao J, Zhang S, Zhou Y, Hu X and Shao C: MicroRNA 483–3p suppresses the expression of DPC4/Smad4 in pancreatic cancer. FEBS Lett. 585:207–213. 2011.

45 

Hao J, Zhang S, Zhou Y, Liu C, Hu X and Shao C: MicroRNA 421 suppresses DPC4/Smad4 in pancreatic cancer. Biochem Biophys Res Commun. 406:552–557. 2011. View Article : Google Scholar : PubMed/NCBI

46 

Zhang Y, Takahashi S, Tasaka A, Yoshima T, Ochi H and Chayama K: Involvement of microRNA-224 in cell proliferation, migration, invasion and anti-apoptosis in hepatocellular carcinoma. J Gastroenterol Hepatol. 28:565–575. 2013. View Article : Google Scholar : PubMed/NCBI

47 

Hudson RS, Yi M, Esposito D, Glynn SA, Starks AM, Yang Y, et al: MicroRNA-106b-25 cluster expression is associated with early disease recurrence and targets caspase-7 and focal adhesion in human prostate cancer. Oncogene. Sep 17–2012.(Epub ahead of print).

48 

Kasinski AL and Slack FJ: miRNA-34 prevents cancer initiation and progression in a therapeutically resistant K-ras and p53-induced mouse model of lung adenocarcinoma. Cancer Res. 72:5576–5587. 2012. View Article : Google Scholar : PubMed/NCBI

49 

Panarelli NC and Yantiss RK: MicroRNA Expression in Selected Carcinomas of the Gastrointestinal Tract. Patholog Res Int. 2011:1246082011.PubMed/NCBI

50 

Tokumaru Y, Yamashita K, Osada M, et al: Inverse correlation between cyclin A1 hypermethylation and p53 mutation in head and neck cancer identified by reversal of epigenetic silencing. Cancer Res. 64:5982–5987. 2004. View Article : Google Scholar : PubMed/NCBI

51 

Wittmann J and Jäck HM: Serum microRNAs as powerful cancer biomarkers. Biochim Biophys Acta. 1806:200–207. 2010.PubMed/NCBI

52 

Krutovskikh VA and Herceg Z: Oncogenic microRNAs (OncomiRs) as a new class of cancer biomarkers. Bioessays. 32:894–904. 2010. View Article : Google Scholar : PubMed/NCBI

53 

Brase JC, Wuttig D, Kuner R and Sültmann H: Serum microRNAs as non-invasive biomarkers for cancer. Mol Cancer. 9:3062010. View Article : Google Scholar : PubMed/NCBI

54 

Lawrie CH, Gal S, Dunlop HM, Pushkaran B, Liggins AP, Pulford K, et al: Detection of elevated levels of tumour-associated microRNAs in serum of patients with diffuse large B-cell lymphoma. Br J Haematol. 141:672–675. 2008. View Article : Google Scholar : PubMed/NCBI

55 

Wang J, Chen J, Chang P, LeBlanc A, Li D, Abbruzzesse JL, et al: MicroRNAs in plasma of pancreatic ductal adenocarcinoma patients as novel blood-based biomarkers of disease. Cancer Prev Res (Phila). 2:807–813. 2009. View Article : Google Scholar : PubMed/NCBI

56 

Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, Pogosova-Agadjanyan EL, et al: Circulating microRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci USA. 105:10513–10518. 2008. View Article : Google Scholar : PubMed/NCBI

57 

Ng EK, Chong WW, Jin H, Lam EK, Shin VY, Yu J, et al: Differential expression of microRNAs in plasma of patients with colorectal cancer: a potential marker for colorectal cancer screening. Gut. 58:1375–1381. 2009. View Article : Google Scholar : PubMed/NCBI

58 

Cheng H, Zhang L, Cogdell DE, Zheng H, Schetter AJ, Nykter M, et al: Circulating plasma MiR-141 is a novel biomarker for metastatic colon cancer and predicts poor prognosis. PLoS One. 6:e177452011. View Article : Google Scholar : PubMed/NCBI

59 

Huang Z, Huang D, Ni S, Peng Z, Sheng W and Du X: Plasma microRNAs are promising novel biomarkers for early detection of colorectal cancer. Int J Cancer. 127:118–126. 2010. View Article : Google Scholar : PubMed/NCBI

60 

Wang LG and Gu J: Serum microRNA-29a is a promising novel marker for early detection of colorectal liver metastasis. Cancer Epidemiol. 36:e61–e67. 2012. View Article : Google Scholar : PubMed/NCBI

61 

Luo X, Burwinkel B, Tao S and Brenner H: MicroRNA signatures: novel biomarker for colorectal cancer? Cancer Epidemiol Biomarkers Prev. 20:1272–1286. 2011. View Article : Google Scholar

62 

Pu XX, Huang GL, Guo HQ, Guo CC, Li H, Ye S, et al: Circulating miR-221 directly amplified from plasma is a potential diagnostic and prognostic marker of colorectal cancer and is correlated with p53 expression. J Gastroenterol Hepatol. 25:1674–1680. 2010. View Article : Google Scholar : PubMed/NCBI

63 

Hennessey PT, Sanford T, Choudhary A, Mydlarz WW, Brown D, Adai AT, et al: Serum microRNA biomarkers for detection of non-small cell lung cancer. PLoS One. 7:e323072012. View Article : Google Scholar : PubMed/NCBI

64 

Yuxia M, Zhennan T and Wei Z: Circulating miR-125b is a novel biomarker for screening non-small-cell lung cancer and predicts poor prognosis. J Cancer Res Clin Oncol. 138:2045–2050. 2012. View Article : Google Scholar : PubMed/NCBI

65 

Kosaka N, Iguchi H and Ochiya T: Circulating microRNA in body fluid: a new potential biomarker for cancer diagnosis and prognosis. Cancer Sci. 101:2087–2092. 2010. View Article : Google Scholar : PubMed/NCBI

66 

Bou Kheir T, Futoma-Kazmierczak E, Jacobsen A, Krogh A, Bardram L, Hother C, et al: miR-449 inhibits cell proliferation and is down-regulated in gastric cancer. Mol Cancer. 10:292011.PubMed/NCBI

67 

Zhang Z, Li Z, Gao C, Chen P, Chen J, Liu W, et al: miR-21 plays a pivotal role in gastric cancer pathogenesis and progression. Lab Invest. 88:1358–1366. 2008. View Article : Google Scholar : PubMed/NCBI

68 

Zhang BG, Li JF, Yu BQ, Zhu ZG, Liu BY and Yan M: microRNA-21 promotes tumor proliferation and invasion in gastric cancer by targeting PTEN. Oncol Rep. 27:1019–1026. 2012.PubMed/NCBI

69 

Luo H, Zhang H, Zhang Z, Zhang X, Ning B, Guo J, et al: Down-regulated miR-9 and miR-433 in human gastric carcinoma. J Exp Clin Cancer Res. 28:822009. View Article : Google Scholar : PubMed/NCBI

70 

Kim YK, Yu J, Han TS, Park SY, Namkoong B, Kim DH, et al: Functional links between clustered microRNAs: suppression of cell-cycle inhibitors by microRNA clusters in gastric cancer. Nucleic Acids Res. 37:1672–1681. 2009. View Article : Google Scholar : PubMed/NCBI

71 

Tsukamoto Y, Nakada C, Noguchi T, Tanigawa M, Nguyen LT, Uchida T, et al: MicroRNA-375 is downregulated in gastric carcinomas and regulates cell survival by targeting PDK1 and 14-3-3zeta. Cancer Res. 70:2339–2349. 2010. View Article : Google Scholar : PubMed/NCBI

72 

Ding L, Xu Y, Zhang W, Deng Y, Si M, Du Y, et al: MiR-375 frequently downregulated in gastric cancer inhibits cell proliferation by targeting JAK2. Cell Res. 20:784–793. 2010. View Article : Google Scholar : PubMed/NCBI

73 

Xu Y, Deng Y, Yan X and Zhou T: Targeting miR-375 in gastric cancer. Expert Opin Ther Targets. 15:961–972. 2011. View Article : Google Scholar : PubMed/NCBI

74 

Wan HY, Guo LM, Liu T, Liu M, Li X and Tang H: Regulation of the transcription factor NF-kappaB1 by microRNA-9 in human gastric adenocarcinoma. Mol Cancer. 9:162010. View Article : Google Scholar : PubMed/NCBI

75 

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

76 

Zhang Y, Fan KJ, Sun Q, Chen AZ, Shen WL, Zhao ZH, et al: Functional screening for miRNAs targeting Smad4 identified miR-199a as a negative regulator of TGF-β signalling pathway. Nucleic Acids Res. 40:9286–9297. 2012.PubMed/NCBI

77 

Zhang X, Nie Y, Du Y, Cao J, Shen B and Li Y: MicroRNA-181a promotes gastric cancer by negatively regulating tumor suppressor KLF6. Tumour Biol. 33:1589–1597. 2012. View Article : Google Scholar : PubMed/NCBI

78 

Chen L, Yang Q, Kong WQ, Liu T, Liu M, Li X and Tang H: MicroRNA-181b targets cAMP responsive element binding protein 1 in gastric adenocarcinomas. IUBMB Life. 64:628–635. 2012. View Article : Google Scholar : PubMed/NCBI

79 

Xia J, Wu Z, Yu C, He W, Zheng H, He Y, et al: 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

80 

Li C, Nie H, Wang M, Su L, Li J, Yu B, 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

81 

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

82 

Li J, Guo Y, Liang X, Sun M, Wang G, De W and Wu W: MicroRNA-223 functions as an oncogene in human gastric cancer by targeting FBXW7/hCdc4. J Cancer Res Clin Oncol. 138:763–774. 2012. View Article : Google Scholar : PubMed/NCBI

83 

Zheng B, Liang L, Huang S, Zha R, Liu L, Jia D, 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

84 

Zhang Z, Liu S, Shi R and Zhao G: miR-27 promotes human gastric cancer cell metastasis by inducing epithelial-to-mesenchymal transition. Cancer Genet. 204:486–491. 2011. View Article : Google Scholar : PubMed/NCBI

85 

Wang M, Li C, Nie H, Lv X, Qu Y, Yu B, 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

86 

Li Z, Cao Y, Jie Z, Liu Y, Li Y, Li J, 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

87 

Liu Z, Zhu J, Cao H, Ren H and Fang X: miR-10b promotes cell invasion through RhoC-AKT signaling pathway by targeting HOXD10 in gastric cancer. Int J Oncol. 40:1553–1560. 2012.PubMed/NCBI

88 

Liao YL, Hu LY, Tsai KW, Wu CW, Chan WC, Li SC, et al: Transcriptional regulation of miR-196b by ETS2 in gastric cancer cells. Carcinogenesis. 33:760–769. 2012. View Article : Google Scholar : PubMed/NCBI

89 

Gao P, Xing AY, Zhou GY, Zhang TG, Zhang JP, Gao C, et al: The molecular mechanism of microRNA-145 to suppress invasion-metastasis cascade in gastric cancer. Oncogene. 32:491–501. 2013. View Article : Google Scholar : PubMed/NCBI

90 

Wang J, Zhang J, Wu J, Luo D, Su K, Shi W, et al: MicroRNA-610 inhibits the migration and invasion of gastric cancer cells by suppressing the expression of vasodilator-stimulated phosphoprotein. Eur J Cancer. 48:1904–1913. 2012. View Article : Google Scholar : PubMed/NCBI

91 

Leja M, Wex T and Malfertheiner P: Markers for gastric cancer premalignant lesions: where do we go? Dig Dis. 30:268–276. 2012. View Article : Google Scholar : PubMed/NCBI

92 

Calin GA and Croce CM: MicroRNA signatures in human cancers. Nat Rev Cancer. 6:857–866. 2006. View Article : Google Scholar : PubMed/NCBI

93 

Chan SH, Wu CW, Li AF, Chi CW and Lin WC: miR-21 microRNA expression in human gastric carcinomas and its clinical association. Anticancer Res. 28:907–911. 2008.PubMed/NCBI

94 

Xu Y, Sun J, Xu J, Li Q, Guo Y and Zhang Q: miR-21 Is a Promising Novel Biomarker for Lymph Node Metastasis in Patients with Gastric Cancer. Gastroenterol Res Pract. 2012:6401682012.PubMed/NCBI

95 

Xiao B, Guo J, Miao Y, Jiang Z, Huan R, Zhang Y, et al: Detection of miR-106a in gastric carcinoma and its clinical significance. Clin Chim Acta. 400:97–102. 2009. View Article : Google Scholar : PubMed/NCBI

96 

Chiang Y, Song Y, Wang Z, Chen Y, Yue Z, Xu H, et al: Aberrant expression of miR-203 and its clinical significance in gastric and colorectal cancers. J Gastrointest Surg. 15:63–70. 2011. View Article : Google Scholar : PubMed/NCBI

97 

Takagi T, Iio A, Nakagawa Y, Naoe T, Tanigawa N and Akao Y: Decreased expression of microRNA-143 and -145 in human gastric cancers. Oncology. 77:12–21. 2009. View Article : Google Scholar : PubMed/NCBI

98 

Su Y, Ni Z, Wang G, Cui J, Wei C, Wang J, et al: Aberrant expression of microRNAs in gastric cancer and biological significance of miR-574-3p. Int Immunopharmacol. 13:468–475. 2012. View Article : Google Scholar : PubMed/NCBI

99 

Guo JX, Tao QS, Lou PR, Chen XC, Chen J and Yuan GB: miR-181b as a potential molecular target for anticancer therapy of gastric neoplasms. Asian Pac J Cancer Prev. 13:2263–2267. 2012. View Article : Google Scholar : PubMed/NCBI

100 

Zhao X, Dou W, He L, Liang S, Tie J, Liu C, et al: MicroRNA-7 functions as an anti-metastatic microRNA in gastric cancer by targeting insulin-like growth factor-1 receptor. Oncogene. 32:1363–1372. 2013. View Article : Google Scholar : PubMed/NCBI

101 

Jiang W, Chen X, Liao M, et al: Identification of links between small molecules and miRNAs in human cancers based on transcriptional responses. Sci Rep. 2:2822012. View Article : Google Scholar : PubMed/NCBI

102 

Liu T, Tang H, Lang Y, Liu M and Li X: MicroRNA-27a functions as an oncogene in gastric adenocarcinoma by targeting prohibitin. Cancer Lett. 273:233–242. 2009. View Article : Google Scholar : PubMed/NCBI

103 

Jiang Z, Guo J, Xiao B, Miao Y, Huang R, Li D and Zhang Y: Increased expression of miR-421 in human gastric carcinoma and its clinical association. J Gastroenterol. 45:17–23. 2010. View Article : Google Scholar : PubMed/NCBI

104 

Link A, Kupcinskas J, Wex T, et al: Macro-role of microRNA in gastric cancer. Dig Dis. 30:255–267. 2012. View Article : Google Scholar : PubMed/NCBI

105 

Tarasov V, Jung P, Verdoodt B, Lodygin D, Epanchintsev A, Menssen A, et al: Differential regulation of microRNAs by p53 revealed by massively parallel sequencing: miR-34a is a p53 target that induces apoptosis and G1-arrest. Cell Cycle. 6:1586–1593. 2007. View Article : Google Scholar : PubMed/NCBI

106 

Ji Q, Hao X, Meng Y, Zhang M, Desano J, Fan D and Xu L: Restoration of tumor suppressor miR-34 inhibits human p53-mutant gastric cancer tumorspheres. BMC Cancer. 8:2662008. View Article : Google Scholar : PubMed/NCBI

107 

Xia L, Zhang D, Du R, Pan Y, Zhao L, Sun S, et al: miR-15b and miR-16 modulate multidrug resistance by targeting BCL2 in human gastric cancer cells. Int J Cancer. 123:372–379. 2008. View Article : Google Scholar : PubMed/NCBI

108 

Shen J, Wan R, Hu G, et al: miR-15b and miR-16 induce the apoptosis of rat activated pancreatic stellate cells by targeting Bcl-2 in vitro. Pancreatology. 12:91–99. 2012. View Article : Google Scholar : PubMed/NCBI

109 

Wang J, Wang Q, Liu H, Hu B, Zhou W and Cheng Y: MicroRNA expression and its implication for the diagnosis and therapeutic strategies of gastric cancer. Cancer Lett. 297:137–143. 2010. View Article : Google Scholar : PubMed/NCBI

110 

Lastraioli E, Romoli MR and Arcangeli A: Immunohistochemical biomarkers in gastric cancer research and management. Int J Surg Oncol. 2012:8686452012.PubMed/NCBI

111 

Grady WM and Tewari M: The next thing in prognostic molecular markers: microRNA signatures of cancer. Gut. 59:706–708. 2010. View Article : Google Scholar : PubMed/NCBI

112 

Chen L, Jiang M, Yuan W and Tang H: Prognostic value of miR-93 overexpression in resectable gastric adenocarcinomas. Acta Gastroenterol Belg. 75:22–27. 2012.PubMed/NCBI

113 

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

114 

Tsai KW, Liao YL, Wu CW, Hu LY, Li SC, Chan WC, et al: Aberrant expression of miR-196a in gastric cancers and correlation with recurrence. Genes Chromosomes Cancer. 51:394–401. 2012. View Article : Google Scholar : PubMed/NCBI

115 

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

116 

Konishi H, Ichikawa D, Komatsu S, Shiozaki A, Tsujiura M, Takeshita H, et al: Detection of gastric cancer-associated microRNAs on microRNA microarray comparing pre- and post-operative plasma. Br J Cancer. 106:740–747. 2012. View Article : Google Scholar : PubMed/NCBI

117 

Wang M, Gu H, Wang S, Qian H, Zhu W, Zhang L, et al: Circulating miR-17-5p and miR-20a: molecular markers for gastric cancer. Mol Med Report. 5:1514–1520. 2012.PubMed/NCBI

118 

Song MY, Pan KF, Su HJ, Zhang L, Ma JL, Li JY, et al: Identification of serum microRNAs as novel non-invasive biomarkers for early detection of gastric cancer. PLoS One. 7:e336082012. View Article : Google Scholar : PubMed/NCBI

119 

Liu H, Zhu L, Liu B, Yang L, Meng X, Zhang W, et al: 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

120 

Chim SS, Shing TK, Hung EC, Leung TY, Lau TK, Chiu RW and Lo YM: Detection and characterization of placental microRNAs in maternal plasma. Clin Chem. 54:482–490. 2008. View Article : Google Scholar : PubMed/NCBI

121 

Ohshima K, Inoue K, Fujiwara A, Hatakeyama K, Kanto K, Watanabe Y, et al: Let-7 microRNA family is selectively secreted into the extracellular environment via exosomes in a metastatic gastric cancer cell line. PLoS One. 5:e132472010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September 2013
Volume 6 Issue 3

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
Xu X, Yang X, Xing C, Zhang S and Cao J: miRNA: The nemesis of gastric cancer (Review). Oncol Lett 6: 631-641, 2013
APA
Xu, X., Yang, X., Xing, C., Zhang, S., & Cao, J. (2013). miRNA: The nemesis of gastric cancer (Review). Oncology Letters, 6, 631-641. https://doi.org/10.3892/ol.2013.1428
MLA
Xu, X., Yang, X., Xing, C., Zhang, S., Cao, J."miRNA: The nemesis of gastric cancer (Review)". Oncology Letters 6.3 (2013): 631-641.
Chicago
Xu, X., Yang, X., Xing, C., Zhang, S., Cao, J."miRNA: The nemesis of gastric cancer (Review)". Oncology Letters 6, no. 3 (2013): 631-641. https://doi.org/10.3892/ol.2013.1428