Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

The Downregulation of MiR-182 Is Associated with the Growth and Invasion of Osteosarcoma Cells through the Regulation of TIAM1 Expression

  • Jun Hu,

    Affiliations Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China, Department of Orthopedics, The First Hospital of Kunming, Kunming, Yunnan, China

  • Guohua Lv ,

    gfq4e1@yeah.net

    Affiliation Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China

  • Shuguang Zhou,

    Affiliations Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China, Department of Orthopedics, The Affiliated Hospital of Hunan University of Medicine and The Third People’s Hospital of Huaihua, Huaihua, Hunan, China

  • Yucheng Zhou,

    Affiliations Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China, Department of Orthopedics, The People’s Hospital of Qingyuan, Qingyuan, Guangdong, China

  • Bangxu Nie,

    Affiliation Department of Orthopedics, The First Hospital of Kunming, Kunming, Yunnan, China

  • Hong Duan,

    Affiliation Department of Orthopedics, The First Hospital of Kunming, Kunming, Yunnan, China

  • Yunfeng Zhang,

    Affiliation Department of Orthopedics, The First Hospital of Kunming, Kunming, Yunnan, China

  • Xiaofeng Yuan

    Affiliation Department of Orthopedics, The First Hospital of Kunming, Kunming, Yunnan, China

Retraction

The PLOS ONE Editors retract this article [1] because it was identified as one of a series of submissions for which we have concerns about peer review, authorship, and similarities across articles. Image similarities were noted between panels in Fig 3B in [1], between Fig 4D in [1] and Fig 5D in [2, 3], between Fig 4E in [1] and Fig 3D in [4, 5], and between Fig 5B in [1], Fig 5D in [6, 7], Fig 4A in [8, 9] and Fig 7E in [10]. These concerns call into question the validity and provenance of the reported results. We regret that the issues were not addressed prior to the article’s publication.

All authors either did not respond directly or could not be reached.

21 Apr 2023: The PLOS ONE Editors (2023) Retraction: The Downregulation of MiR-182 Is Associated with the Growth and Invasion of Osteosarcoma Cells through the Regulation of TIAM1 Expression. PLOS ONE 18(4): e0283115. https://doi.org/10.1371/journal.pone.0283115 View retraction

Abstract

Background

Osteosarcoma is the most common primary bone malignancy in children and young adults. Increasing results suggest that discovery of microRNAs (miRNAs) might provide a novel therapeutical target for osteosarcoma.

Methods

MiR-182 expression level in osteosarcoma cell lines and tissues were assayed by qRT-PCR. MiRNA mimics or inhibitor were transfected for up-regulation or down-regulation of miR-182 expression. Cell function was assayed by CCK8, migration assay and invasion assay. The target genes of miR-182 were predicated by bioinformatics algorithm (TargetScan Human).

Results

MiR-182 was down-regulated in osteosarcoma tissues and cell lines. Overexpression of miR-182 inhibited tumor growth, migration and invasion. Subsequent investigation revealed that TIAM1 was a direct and functional target of miR-182 in osteosarcoma cells. Overexpression of miR-182 impaired TIAM1-induced inhibition of proliferation and invasion in osteosarcoma cells.

Conclusions

Down-expression of miR-182 in osteosarcoma promoted tumor growth, migration and invasion by targeting TIAM1. MiR-182 might act as a tumor suppressor gene whose down-regulation contributes to the progression and metastasis of osteosarcoma, providing a potential therapy target for osteosarcoma patients.

Introduction

Osteosarcoma is a primary malignant bone tumor with high morbidity in children and young adults, accounting for approximately 60% of malignant bone tumors in the first 2 decades of life[13]. The 5-year survival rate of the patients with osteosarcoma has significantly improved over the past decades to approximately 60–70% since the introduction of wide tumor excision, adjuvant chemotherapy and radiotherapy[4, 5]. However, a high proportion of osteosarcoma patients still respond poorly to chemotherapy, and they have a great risk of distant metastasis or local relapse even after curative resection of the primary tumor and adjuvant chemotherapy or radiotherapy[6, 7]. Therefore, discovery of new therapeutic targets and approaches is one of the most important challenges in the management of osteosarcoma.

MicroRNA (miRNA) belongs to small non-coding RNA molecules that contain about 22 nucleotides[8, 9]. MiRNAs regulate gene expression by binding to complementary sites in their 3’-UTR, resulting in translational repression or gene silencing[10, 11]. At present, approximately 450 miRNAs have been cloned in mammalian cells, and it is believed that up to 1,000 miRNAs genes exist[12]. Aberrant expression of miRNAs occurs in many types of cancers and plays significant roles by regulating the expression of various tumor suppressors and oncogenes[1315]. MiRNAs have been suggested to be associated with the proliferation, differentiation, apoptosis and invasion of tumor cells [1619]. However, the role of miRNAs in osteosarcoma development has only recently been investigated and still remains largely ambiguous.

Here, we wondered whether miR-182 was involved in the pathogenesis of osteosarcoma. Our aim is to identify the role of miR-182 in osteosarcoma cells growth, migration and invasion, and the target gene of miR-182.

Materials and Methods

Ethics statement

All of the patients (or patients’ parents on behalf of the children) agreed to participate in the study and gave written informed consent. Both this study and consent were approved by the Ethics Committee of The Second Xiangya Hospital of Central South University and complied with the Declaration of Helsinki.

Osteosarcoma tissues

Fifty paired tissue specimens of osteosarcoma and matched normal tissues were obtained from Department of Orthopedics in The Second Xiangya Hospital of Central South University. The matched normal tissues were further confirmed by pathologist for their normal origin that they do not have tumor cells. All the tissues were obtained at the time of surgery and immediately stored in liquid nitrogen until use. Overall survival was measured from the time at which the tissue was obtained to the date of death from any cause or date of last follow-up. All of the patients were anonymised. The Institute Research Medical Ethics Committee of The Second Xiangya Hospital of Central South University granted approval for this study.

Cell lines and cell culture

The following human cell lines were used in this study: MG-63 (14 years old, male), U2OS (15 years old, female), SOSP-9607(17 years old, male), SAOS-2 (11 years old, female) and hFOB. These cell lines were purchased from the Cell Resource Center of the Institute of Basic Medical Sciences at the Chinese Academy of Medical Sciences and the Peking Union Medical College (Beijing, China) and were propagated in Dulbecco’s modified Eagle’s medium (Gibco; Invitrogen; Life Technologies, Germany) that was supplemented with 10% fetal bovine serum (GIBCO, NY, USA), streptomycin (100 μg/ml), and penicillin (100 U/ml).

Cell transfection

The miR-182 mimics, inhibitor and their controls were purchased from GenePharma (Shanghai, China). MG63 cells were infected into the cells with a final oligonucleotide concentration of 20 nmol/L. All of the cell transfections were performed with DharmaFECT1 reagent (Dharmacon, TX, USA), according to the manufacturer’s instructions.

Cell proliferation assay

Cells were plated in 96-well plates at 1× 104 per well in a final volume of 100 μL, and cultured for 1–4 days after transfection. Cells were incubated in 10% CCK-8 (Dojindo; Kumamoto, Japan) that was diluted in normal culture medium at 37°C until the visual color conversion occurred. After incubating, the absorbance at 450 nm was measured by an enzyme-linked immunosorbent assay plate reader.

Cell migration and invasion assays

A wound-healing assay was done to assess cell migration. An artificial wound was created 24 h after transfection using a 200-ll pipette tip on the confluent cell monolayer, and mitomyclin C was added to the culture wells. To visualize migrated cells and wound healing, images were taken at 0h and 48h. For invasion assays, 1×105 cells were plated in the top chamber of the insert precoated with Matrigel (BD Bioscience). Cells were plated in medium without serum, and medium supplemented with 10% FBS was used as a chemoattractant in the lower chamber. The cells were incubated for 48 h and cells that migrated to the underside of the membrane. The cells that were located on the lower surface of the chamber were stained with 0.1% crystal violet (Sigma) and were then counted.

Luciferase assay

Mutant (Mut) constructs were generated by mutating the seed region of the miR-182 binding site. miR-182 expressing or control cells were cultured in 24-well plates, and transfected with 100 ng luciferase reporter plasmid and 5 ng pRL-TK vector expressing the Renilla luciferase (Promega, Madison, WI, USA) using Lipofectamine 2000 reagent (Invitrogen). After 48 h, cells were harvested, lysed and luciferase activity was measured using the Dual-Luciferase Reporter Assay System (Promega) according to the manufacturer’s protocol. Renilla luciferase was used for normalization.

RNA extraction and RT-PCR

We extracted total RNA with the miRNeasy RNA isolation kit (Qiagen). Purified RNA was reverse transcribed with the iScript cDNA Synthesis Kit (Bio-Rad). RT-PCR was performed with TaqMan probes for both genes and miRNAs (Applied Biosystems). PCR was performed at 95°C for 10 min, followed by 40 cycles of 95°C for 10s and 60°C for 1 min. The relative expression ratio of miR-182 in tissues and cells was quantified by the 2-ΔΔCT method. All miRNA data were normalized to the internal control small RNAs U6. For the mRNA samples, GAPDH was used as an internal control.

Western blot analysis

Total cell lysates were prepared using RIPA buffer (150 mM NaCl, 1 mM EDTA, 50 mM Tris—HCl pH 7.4, 1% Triton X-100, 0.5% deoxycholic acid, 0.1% SDS) and the protein concentration was measured by BCA method. Equal amounts of proteins were separated by 10% SDS-PAGE and blotted to PVDF membranes (Millipore, Billerica, MA, USA). The membranes were blocked with 5% non-fat milk powder at room temperature for 2 h, then incubated with specific antibody for TIMA1 or GAPHD (Abcam, England) at 4°C followed by incubation with secondary antibody (zsgb-bio, Beijing, China) for 1 h at room temperature. The membranes were developed using ECL kit (Pierce, Rockford, IL, USA) and exposed to X-ray film to visualize the images.

Statistical analysis

Statistical analysis was performed using SPSS 17.0 software (SPSS, USA). Data were expressed as the mean ± SD from at least three separate experiments. Differences between groups were analyzed using Student’s t test and the χ2 test. P < 0.05 was considered statistically significant. Multivariate survival analyses were performed using Cox regression models. We estimated hazard ratios (HRs), 95% CI and p values.

Result

MiR-182 was down-regulated in osteosarcoma cell lines and tissues

MiR-182 expression in osteosarcoma cell lines was assayed by qRT-PCR. A lower expression of miR-182 was observed in osteosarcoma cell lines (U2OS, MG-63, SOSP-9607, and SAOS-2), as comparing with human osteoblast cell lines (hFOB) (Fig 1A). MiR-182 level in osteosarcoma tissues and corresponding adjacent normal tissues were also assayed by qRT-PCR. Our results demonstrated that miR-182 was significantly down-regulated in osteosarcoma tissues (Fig 1B). The level of miR-182 was lower in osteosarcoma tissue than in normal tissues in the 40 pairs (Fig 1C). In addition, loss of miR-182 levels in patients with osteosarcoma was associated with considerably shortened disease-free survival (hazards ratio = 3.22, Fig 1D).

thumbnail
Fig 1. miR-182 was down-regulated in osteosarcoma cell lines and tissues.

(A) The expression levels of miR-182 were measured by qRT-PCR in osteosarcoma cell lines (U2OS, MG-63, SOSP-9607, and SAOS-2) and human normal osteoblastic cell line (hFOB). The expression of miR-182 was down-regulated in osteosarcoma cell lines. (B) Relative expression of miR-182 in 40 primary osteosarcoma tissues compared with their pair-matched nontumor tissues. Data are shown as log2 of relative ratio change of osteosarcoma tissues relative to normal tissues. (C) The expression of miR-182 in the human osteosarcoma tissues was lower than that in non-tumor adjacent tissues. (D) Loss of miR-182 levels in patients with osteosarcoma was associated with associated with considerably shortened disease-free survival. All data were normalized by U6 snRNA. ***p<0.001.

https://doi.org/10.1371/journal.pone.0121175.g001

Overexpression of miR-182 inhibited tumor growth

After 48h transfection, MG-63 that transfected with miR-182 mimic showed higher expression of miR-182 (Fig 2A), while MG-63 that transfected with miR-182 inhibitor showed lower miR-182 expression (Fig 2B). Then, we assayed cell proliferation of these cell line using CCK8. We found that overexpression of miR-182 inhibited MG-63 cells proliferation (Fig 2C) and inhibition of miR-182 promoted MG-63 cells proliferation (Fig 2D). Thus our data indicated that overexpression of miR-182 inhibited osteosarcoma cells growth.

thumbnail
Fig 2. Overexpression of miR-182 inhibited tumor growth.

(A) The miR-182 expression level of the indicated cells was measured by qRT-PCR. The expression of miR-182 was normalized to U6 snRNA. (B) The expression level of miR-182 in the indicated cells was measured by qRT-PCR. The expression of miR-182 was normalized to U6 snRNA. (C) Cell proliferation of the indicated cells was analyzed using CCK-8 assay. (D) Inhibition of miR-182 promote the osteosarcoma cell proliferation. *p<0.05, ** p<0.01, and ***p<0.001.

https://doi.org/10.1371/journal.pone.0121175.g002

Overexpression of miR-182 inhibited tumor migration and invasion

A wound-healing assay and invasion assay were performed to investigate the effects of miR-182 on the migratory and invasive behaviors of osteosarcoma cells in vitro. Our results demonstrated that cells in miR-182 mimics group exhibited significant declines in migration and invasion capacities compared with cells in the control group and scramble group respectively (Fig 3A and 3B). Conversely, miR-182 inhibitor significantly promoted the cell migration and invasion of the MG-63 cells (Fig 3A and 3B).

thumbnail
Fig 3. Overexpression of miR-182 inhibited tumor invasion.

(A) Wound-healing assay was employed to evaluate the migratory ability of MG-63 cells after treatment with miR-182 mimics, inhibitors or scramble or no transfection. Relative ratio of wound closure is shown on the right. (B) Invasion analysis were employed to evaluate the invasive ability of the MG-63 cells after treatment with miR-182 mimics, inhibitors or scramble or no transfection; the relative ratio of invasive cells per field is shown on the right, *p<0.05, ** p<0.01, and ***p<0.001.

https://doi.org/10.1371/journal.pone.0121175.g003

MiR-182 directly targets the TIAM1 in osteosarcoma cells

We found that the TIAM1 harbored a potential miR-182 binding site (Fig 4A). Luciferase reporter assays using 3’-untranslated region (3’-UTR) of TIAM1 gene further demonstrated that miR-182 mimics significantly reduced the activity of TIAM1 3’-UTR, but not the binding motif mutant one (Fig 3B). In agreement, miR-182 mimics significantly reduced the protein and mRNA abundance of TIAM1 in MG-63 cells (Fig 4C and 4D). Moreover, miR-182 inhibitor increased both the protein and mRNA levels of TIAM1 (Fig 4C and 4F), further indicating that TIAM1 was a target of miR-182 in osteosarcoma cells.

thumbnail
Fig 4. miR-182 directly targets the TIAM1 in osteosarcoma cells.

(A) The potential miR-182 binding site at the 3'-UTR of TIAM1 mRNA was computationally predicted by Targetscan. (B) Cells were transfected with wild type 3'-UTR reporter or mutant constructs together with 20 nM of miR-182 mimics or controls. Luciferase activity was normalized by the ratio of firefly and Renilla luciferase signals. (C) Relative expression of TIAM1 in the MG-63 cells transfected with miR-182 mimics or negative control or inhibitor. The expression of TIAM1 was normalized to GAPDH. (D) Western blot analysis of TIMA1 expression in MG-63 cells transfected with miR-182 mimics or negative control or untreat group. GAPDH was also detected as a loading control. (E) Western blot analysis of TIAM1 expression in MG-63 cells transfected with miR-182 inhibitor or negative control or untreat group. GAPDH was also detected as a loading control. ***p<0.001.

https://doi.org/10.1371/journal.pone.0121175.g004

Restoration of miR-182 inhibited TIAM1-induce osteosarcoma cell proliferation and invasion

The TIAM1 expression in osteosarcoma cell lines was assayed by qRT-PCR and western blot. The osteosarcoma cell lines (U2OS, MG-63, SOSP-9607, and SAOS-2) showed a higher TIAM1 mRNA and protein expression as compared with human osteoblast cell lines (hFOB) (Fig 5A and 5B). PcDNA-TIAM1 can enhance the TIAM1 protein expression using western blot (Fig 5C). TIAM1 significantly inhibited osteosarcoma cell proliferation and invasion (Fig 5D and 5E). When miR-182 mimics and pcDNA-TIAM1 were cotransfected into MG-63 cells, miR-182 expression significantly enhanced the TIAM1-induced osteosarcoma cell proliferation and invasion (Fig 5D and 5E).

thumbnail
Fig 5. Restoration of miR-182 inhibits TIAM1-inducing osteosarcoma cell proliferation and invasion.

(A) qRT-PCR analysis of TIAM1 expression in osteosarcoma cell lines (U2OS, MG-63, SOSP-9607, and SAOS-2) and human normal osteoblastic cell line (hFOB). The mRNA expression of TIAM1 was up-regulated in osteosarcoma cell lines. The expression of TIAM1 was normalized to GAPDH. (B) Western blot analysis of TIAM1 expression in osteosarcoma cell lines (U2OS, MG-63, SOSP-9607, and SAOS-2) and human normal osteoblastic cell line (hFOB). The protein expression of TIAM1 was up-regulated in osteosarcoma cell lines. GAPDH was also detected as a loading control. (C) The effects of pCDNA-TIAM1 on the expression of TIAM1 was detected by Western blotting. GAPDH was also detected as a loading control. (B) The cell growth in MG-63 co-transfected with either miR-182 mimic or scramble and 2.0 μg pCDNA-TIAM1 or pCDNA empty vector using CCK-8 proliferation assay. (C) The cell invasive in MG-63 co-transfected with either miR-182 mimic or scramble and 2.0 μg pCDNA-TIAM1 or pCDNA empty vector using invasion assay. *p<0.05, ** p<0.01, and ***p<0.001. Figs 5B and 5D are excluded from this article's CC-BY license. See the accompanying retraction notice for more information.

https://doi.org/10.1371/journal.pone.0121175.g005

Discussion

Osteosarcoma is the most common malignant bone tumor in childhood and adolescent and always results in an aggressive clinical course[4, 20, 21]. However, the molecular mechanism under osteosarcoma carcinogenesis and progression still remains to be explored[22]. Increasing evidence has indicated that deregulation of miRNAs was involved in the development of tumors, such as gastric cancer, breast cancer, hepatocellular carcinoma and osteosarcoma[2328]. In this study, we showed that miR-182 expression was downregulated in osteosarcoma tissues and cell lines. Forced overexpression of miR-182 inhibited cell proliferation, migration, and invasion in MG-63 cells. We further revealed that TIAM1 was a target of miR-182 in osteosarcoma cells, and overexpression of miR-182 impaired TIAM1-induced promotion of proliferation and invasion in osteosarcoma cells. Moreover, TIAM1 was inversely correlated with miR-182 in osteosarcoma tissues. Therefore, our study identifies that miR-182 might be a tumor suppressor in the progression of osteosarcoma via targeting TIAM1.

Previous studies have indicated that miR-182 is frequently aberrantly expressed in various human malignancies, including gastric cancer, malignant pleural mesothelioma, ovarian carcinoma, prostate, bladder, lung, endometrial, colon and breast cancers, polycythemia vera, pediatric acute leukemia and melanoma[2939]. MiR-182 functions as tumor-suppressor or oncogene in different human malignancies. The tumor suppressor function of miR-182 was shown in lung adenocarcinoma cells, where it inhibits the proliferation and invasion by targeting CTTN[40]. MiR-182 suppressed lung tumorigenesis by targeting RGS17 [39, 41]. In addition, the expression of miR-182 is up-regulated and functions as an oncogene in several human cancers such as melanoma, it promotes anchorage independent growth, invasion and metastasis[29]. In breast cancer, it targets FOXO1 together with miR-27a and miR-96[42]. In ovarian carcinoma, miR-182 promotes cell proliferation, invasion and chemoresistance by targeting programmed cell death 4 [43]. The overexpression of miR-182 is also up-regulated in colorectal cancer and is associated with adverse clinical characteristics and poor prognosis[44], and in hepatocellular carcinoma, it is correlated with metastatic behavior[36]. In the present study, we demonstrated that miR-182 was down-regulated in three four osteosarcoma cell lines and tissues. Moreover, restoration of miR-182 can reduce osteosarcoma cell proliferation and invasion. These results suggest that miR-182 might act as a tumor suppressor gene whose down-regulation contributes to the progression and metastasis of osteosarcoma.

In this study, target validation experiments showed that miR-182 efficiently bound to TIAM1 mRNA and inhibited TIAM1 expression. Overexpression of miR-182 inhibited TIAM1 mRNA and protein expression. Thus, our results suggest that reduced miR-182 expression may be a pivotal component in regulation of TIAM1 and osteosarcoma cell proliferation. Our findings reveal a previous unknown mechanism for miR-182-mediated regulation of cancer cell proliferation. However, the comprehensive understanding of the roles of miR-182 in regulating TIAM1 in various cancers needs further exploration.

Tiam1 gene, located on human chromosome 21, is a member of the guanine nucleotide exchange factor (GNEF) family. Tiam1 gene is found to be an important gene in various cellular functions, such as cell proliferation, adhesion, migration, invasion and metastasis[45, 46]. It has been reported that Tiam1 is up-regulated in T lymphoma, B lymphoma, pancreatic carcinoma, gastric cancer, breast and lung carcinoma[45, 4751]. Overexpression of TIAM1 is considered to be a new potential or even an independent predictor of poor prognosis for clinical patients. Qi et al has shown a connection between Tiam1 overexpression and nasopharyngeal carcinoma[52]. Tiam1 has also been found to be significantly up-regulated in prostate carcinoma and to be a negative predictor[53]. Moreover, Jin et al demonstrated miR-218 acted as a tumor suppressor in osteosarcomas by down-regulating TIAM1, MMP2 and MMP9 expression[54]. However, the detailed mechanisms are still unclear. In this study, we identified TIAM1 as a direct and functional target of miR-182. We confirmed the overexpression of TIAM1 can promote osteosarcomas cell proliferation and invasion in vitro. Moreover, miR-182-mediated suppression of TIAM1 depended on the 3’-UTR. Finally, TIAM1-induced cell proliferation and invasion were reversed by miR-182. Taken together, these results establish a functional connection between miR-182 and TIAM1, and confirm that miR-182 can function as an anti-metastatic miRNA in osteosarcomas cells by targeting TIAM1. The ability of miR-182 to target TIAM1 may be one mechanism underlying the post-transcriptional control of TIAM1.

In conclusion, this study demonstrates that miR-182 functions as a tumor suppressor miRNA in osteosarcoma by suppressing TIAM1 expression. Importantly, the miR-182-TIAM1 axis identified in our study worth further investigation for therapeutic interventio for osteosarcoma.

Supporting Information

S1 Table. Summary of clinic pathological parameters of patients with osteosarcoma.

https://doi.org/10.1371/journal.pone.0121175.s001

(DOCX)

Author Contributions

Conceived and designed the experiments: JH GL SZ YCZ BN HD YFZ XY. Performed the experiments: JH GL SZ YCZ BN HD YFZ XY. Analyzed the data: JH GL SZ YCZ BN HD YFZ XY. Contributed reagents/materials/analysis tools: JH GL SZ YCZ BN HD YFZ XY. Wrote the paper: JH GL SZ YCZ BN HD YFZ XY.

References

  1. 1. Tang J, Shen L, Yang Q, Zhang C. Overexpression of metadherin mediates metastasis of osteosarcoma by regulating epithelial-mesenchymal transition. Cell proliferation. 2014;47(5):427–34. pmid:25174891
  2. 2. Montanaro L, Mazzini G, Barbieri S, Vici M, Nardi-Pantoli A, Govoni M, et al. Different effects of ribosome biogenesis inhibition on cell proliferation in retinoblastoma protein- and p53-deficient and proficient human osteosarcoma cell lines. Cell proliferation. 2007;40(4):532–49. pmid:17635520
  3. 3. Li G, Cai M, Fu D, Chen K, Sun M, Cai Z, et al. Heat shock protein 90B1 plays an oncogenic role and is a target of microRNA-223 in human osteosarcoma. Cellular physiology and biochemistry: international journal of experimental cellular physiology, biochemistry, and pharmacology. 2012;30(6):1481–90.
  4. 4. Zhu J, Feng Y, Ke Z, Yang Z, Zhou J, Huang X, et al. Down-regulation of miR-183 promotes migration and invasion of osteosarcoma by targeting Ezrin. The American journal of pathology. 2012;180(6):2440–51. pmid:22525461
  5. 5. Hasei J, Sasaki T, Tazawa H, Osaki S, Yamakawa Y, Kunisada T, et al. Dual programmed cell death pathways induced by p53 transactivation overcome resistance to oncolytic adenovirus in human osteosarcoma cells. Molecular cancer therapeutics. 2013;12(3):314–25. pmid:23315976
  6. 6. Wang XH, Cai P, Wang MH, Wang Z. microRNA25 promotes osteosarcoma cell proliferation by targeting the cellcycle inhibitor p27. Molecular medicine reports. 2014;10(2):855–9. pmid:24859599
  7. 7. Zhou Y, Huang Z, Wu S, Zang X, Liu M, Shi J. miR-33a is up-regulated in chemoresistant osteosarcoma and promotes osteosarcoma cell resistance to cisplatin by down-regulating TWIST. Journal of experimental & clinical cancer research: CR. 2014;33:12.
  8. 8. Ohdaira H, Sekiguchi M, Miyata K, Yoshida K. MicroRNA-494 suppresses cell proliferation and induces senescence in A549 lung cancer cells. Cell proliferation. 2012;45(1):32–8. pmid:22151897
  9. 9. Lee JE, Hong EJ, Nam HY, Kim JW, Han BG, Jeon JP. MicroRNA signatures associated with immortalization of EBV-transformed lymphoblastoid cell lines and their clinical traits. Cell proliferation. 2011;44(1):59–66. pmid:21199010
  10. 10. Li M, Yu M, Liu C, Zhu H, He X, Peng S, et al. miR-34c works downstream of p53 leading to dairy goat male germline stem-cell (mGSCs) apoptosis. Cell proliferation. 2013;46(2):223–31. pmid:23510477
  11. 11. Li J, You T, Jing J. MiR-125b inhibits cell biological progression of Ewing's sarcoma by suppressing the PI3K/Akt signalling pathway. Cell proliferation. 2014;47(2):152–60. pmid:24517182
  12. 12. Du B, Wang Z, Zhang X, Feng S, Wang G, He J, et al. MicroRNA-545 Suppresses Cell Proliferation by Targeting Cyclin D1 and CDK4 in Lung Cancer Cells. PloS one. 2014;9(2):e88022. pmid:24505359
  13. 13. Khatri R, Subramanian S. MicroRNA-135b and Its Circuitry Networks as Potential Therapeutic Targets in Colon Cancer. Frontiers in oncology. 2013;3:268. pmid:24156094
  14. 14. Lin CW, Chang YL, Chang YC, Lin JC, Chen CC, Pan SH, et al. MicroRNA-135b promotes lung cancer metastasis by regulating multiple targets in the Hippo pathway and LZTS1. Nature communications. 2013;4:1877. pmid:23695671
  15. 15. Satzger I, Mattern A, Kuettler U, Weinspach D, Voelker B, Kapp A, et al. MicroRNA-15b represents an independent prognostic parameter and is correlated with tumor cell proliferation and apoptosis in malignant melanoma. International journal of cancer Journal international du cancer. 2010;126(11):2553–62. pmid:19830692
  16. 16. Chang SJ, Weng SL, Hsieh JY, Wang TY, Chang MD, Wang HW. MicroRNA-34a modulates genes involved in cellular motility and oxidative phosphorylation in neural precursors derived from human umbilical cord mesenchymal stem cells. BMC medical genomics. 2011;4:65. pmid:21923954
  17. 17. Bier A, Giladi N, Kronfeld N, Lee HK, Cazacu S, Finniss S, et al. MicroRNA-137 is downregulated in glioblastoma and inhibits the stemness of glioma stem cells by targeting RTVP-1. Oncotarget. 2013;4(5):665–76. pmid:23714687
  18. 18. Althoff K, Beckers A, Odersky A, Mestdagh P, Koster J, Bray IM, et al. MiR-137 functions as a tumor suppressor in neuroblastoma by downregulating KDM1A. International journal of cancer Journal international du cancer. 2013;133(5):1064–73. pmid:23400681
  19. 19. Luo C, Tetteh PW, Merz PR, Dickes E, Abukiwan A, Hotz-Wagenblatt A, et al. miR-137 inhibits the invasion of melanoma cells through downregulation of multiple oncogenic target genes. The Journal of investigative dermatology. 2013;133(3):768–75. pmid:23151846
  20. 20. Ziyan W, Shuhua Y, Xiufang W, Xiaoyun L. MicroRNA-21 is involved in osteosarcoma cell invasion and migration. Med Oncol. 2011;28(4):1469–74. pmid:20480266
  21. 21. Pan W, Wang H, Jianwei R, Ye Z. MicroRNA-27a promotes proliferation, migration and invasion by targeting MAP2K4 in human osteosarcoma cells. Cellular physiology and biochemistry: international journal of experimental cellular physiology, biochemistry, and pharmacology. 2014;33(2):402–12.
  22. 22. Yan K, Gao J, Yang T, Ma Q, Qiu X, Fan Q, et al. MicroRNA-34a inhibits the proliferation and metastasis of osteosarcoma cells both in vitro and in vivo. PloS one. 2012;7(3):e33778. pmid:22457788
  23. 23. Fu Z, Qian F, Yang X, Jiang H, Chen Y, Liu S. Circulating miR-222 in plasma and its potential diagnostic and prognostic value in gastric cancer. Med Oncol. 2014;31(9):164. pmid:25129310
  24. 24. Inoue T, Iinuma H, Ogawa E, Inaba T, Fukushima R. Clinicopathological and prognostic significance of microRNA-107 and its relationship to DICER1 mRNA expression in gastric cancer. Oncology reports. 2012;27(6):1759–64. pmid:22407237
  25. 25. Li T, Yin J, Yuan L, Wang S, Yang L, Du X, et al. Downregulation of microRNA-139 is associated with hepatocellular carcinoma risk and short-term survival. Oncology reports. 2014;31(4):1699–706. pmid:24549282
  26. 26. Liang YJ, Wang QY, Zhou CX, Yin QQ, He M, Yu XT, et al. MiR-124 targets Slug to regulate epithelial-mesenchymal transition and metastasis of breast cancer. Carcinogenesis. 2013;34(3):713–22. pmid:23250910
  27. 27. Namlos HM, Meza-Zepeda LA, Baroy T, Ostensen IH, Kresse SH, Kuijjer ML, et al. Modulation of the osteosarcoma expression phenotype by microRNAs. PloS one. 2012;7(10):e48086. pmid:23133552
  28. 28. Geng S, Zhang X, Chen J, Liu X, Zhang H, Xu X, et al. The tumor suppressor role of miR-124 in osteosarcoma. PloS one. 2014;9(6):e91566. pmid:24971902
  29. 29. Segura MF, Hanniford D, Menendez S, Reavie L, Zou X, Alvarez-Diaz S, et al. Aberrant miR-182 expression promotes melanoma metastasis by repressing FOXO3 and microphthalmia-associated transcription factor. Proceedings of the National Academy of Sciences of the United States of America. 2009;106(6):1814–9. pmid:19188590
  30. 30. Perilli L, Vicentini C, Agostini M, Pizzini S, Pizzi M, D'Angelo E, et al. Circulating miR-182 is a biomarker of colorectal adenocarcinoma progression. Oncotarget. 2014;5(16):6611–9. pmid:25115394
  31. 31. Wang M, Wang Y, Zang W, Wang H, Chu H, Li P, et al. Downregulation of microRNA-182 inhibits cell growth and invasion by targeting programmed cell death 4 in human lung adenocarcinoma cells. Tumour biology: the journal of the International Society for Oncodevelopmental Biology and Medicine. 2014;35(1):39–46.
  32. 32. Tsuchiyama K, Ito H, Taga M, Naganuma S, Oshinoya Y, Nagano K, et al. Expression of microRNAs associated with Gleason grading system in prostate cancer: miR-182-5p is a useful marker for high grade prostate cancer. The Prostate. 2013;73(8):827–34. pmid:23184537
  33. 33. Li C, Du X, Tai S, Zhong X, Wang Z, Hu Z, et al. GPC1 regulated by miR-96-5p, rather than miR-182-5p, in inhibition of pancreatic carcinoma cell proliferation. International journal of molecular sciences. 2014;15(4):6314–27. pmid:24736782
  34. 34. Wang PY, Gong HT, Li BF, Lv CL, Wang HT, Zhou HH, et al. Higher expression of circulating miR-182 as a novel biomarker for breast cancer. Oncology letters. 2013;6(6):1681–6. pmid:24260062
  35. 35. Zhu YJ, Xu B, Xia W. Hsa-mir-182 downregulates RASA1 and suppresses lung squamous cell carcinoma cell proliferation. Clin Lab. 2014;60(1):155–9. pmid:24600991
  36. 36. Wang J, Li J, Shen J, Wang C, Yang L, Zhang X. MicroRNA-182 downregulates metastasis suppressor 1 and contributes to metastasis of hepatocellular carcinoma. BMC cancer. 2012;12:227. pmid:22681717
  37. 37. Tang T, Wong HK, Gu W, Yu MY, To KF, Wang CC, et al. MicroRNA-182 plays an onco-miRNA role in cervical cancer. Gynecologic oncology. 2013;129(1):199–208. pmid:23313739
  38. 38. Wang YQ, Guo RD, Guo RM, Sheng W, Yin LR. MicroRNA-182 promotes cell growth, invasion, and chemoresistance by targeting programmed cell death 4 (PDCD4) in human ovarian carcinomas. Journal of cellular biochemistry. 2013;114(7):1464–73. pmid:23296900
  39. 39. Kong WQ, Bai R, Liu T, Cai CL, Liu M, Li X, et al. MicroRNA-182 targets cAMP-responsive element-binding protein 1 and suppresses cell growth in human gastric adenocarcinoma. The FEBS journal. 2012;279(7):1252–60. pmid:22325466
  40. 40. Zhang L, Liu T, Huang Y, Liu J. microRNA-182 inhibits the proliferation and invasion of human lung adenocarcinoma cells through its effect on human cortical actin-associated protein. International journal of molecular medicine. 2011;28(3):381–8. pmid:21503569
  41. 41. Sun Y, Fang R, Li C, Li L, Li F, Ye X, et al. Hsa-mir-182 suppresses lung tumorigenesis through down regulation of RGS17 expression in vitro. Biochemical and biophysical research communications. 2010;396(2):501–7. pmid:20420807
  42. 42. Guttilla IK, White BA. Coordinate regulation of FOXO1 by miR-27a, miR-96, and miR-182 in breast cancer cells. The Journal of biological chemistry. 2009;284(35):23204–16. pmid:19574223
  43. 43. Liu Z, Liu J, Segura MF, Shao C, Lee P, Gong Y, et al. MiR-182 overexpression in tumourigenesis of high-grade serous ovarian carcinoma. The Journal of pathology. 2012;228(2):204–15. pmid:22322863
  44. 44. Liu H, Du L, Wen Z, Yang Y, Li J, Wang L, et al. Up-regulation of miR-182 expression in colorectal cancer tissues and its prognostic value. International journal of colorectal disease. 2013;28(5):697–703. pmid:23474644
  45. 45. Chen B, Ding Y, Liu F, Ruan J, Guan J, Huang J, et al. Tiam1, overexpressed in most malignancies, is a novel tumor biomarker. Molecular medicine reports. 2012;5(1):48–53. pmid:21971868
  46. 46. Huang J, Ye X, Guan J, Chen B, Li Q, Zheng X, et al. Tiam1 is associated with hepatocellular carcinoma metastasis. International journal of cancer Journal international du cancer. 2013;132(1):90–100. pmid:22573407
  47. 47. Zhu JM, Yu PW. Downregulation of Tcell lymphoma invasion and metastasisinducing factor 1 induces cytoskeletal rearrangement and inhibits the invasive capacity of gastric cancer cells. Molecular medicine reports. 2013;8(2):425–33. pmid:23754572
  48. 48. Chen JS, Su IJ, Leu YW, Young KC, Sun HS. Expression of T-cell lymphoma invasion and metastasis 2 (TIAM2) promotes proliferation and invasion of liver cancer. International journal of cancer Journal international du cancer. 2012;130(6):1302–13. pmid:21469146
  49. 49. Li Z, Lei H, Luo M, Wang Y, Dong L, Ma Y, et al. DNA methylation downregulated mir-10b acts as a tumor suppressor in gastric cancer. Gastric cancer: official journal of the International Gastric Cancer Association and the Japanese Gastric Cancer Association. 2014.
  50. 50. Liu S, Li Y, Qi W, Zhao Y, Huang A, Sheng W, et al. Expression of Tiam1 predicts lymph node metastasis and poor survival of lung adenocarcinoma patients. Diagnostic pathology. 2014;9:69. pmid:24661909
  51. 51. Zhao L, Liu Y, Sun X, He M, Ding Y. Overexpression of T lymphoma invasion and metastasis 1 predict renal cell carcinoma metastasis and overall patient survival. Journal of cancer research and clinical oncology. 2011;137(3):393–8. pmid:20429013
  52. 52. Qi Y, Huang B, Yu L, Wang Q, Lan G, Zhang Q. Prognostic value of Tiam1 and Rac1 overexpression in nasopharyngeal carcinoma. ORL J Otorhinolaryngol Relat Spec. 2009;71(3):163–71. pmid:19506399
  53. 53. Engers R, Mueller M, Walter A, Collard JG, Willers R, Gabbert HE. Prognostic relevance of Tiam1 protein expression in prostate carcinomas. British journal of cancer. 2006;95(8):1081–6. pmid:17003780
  54. 54. Jin J, Cai L, Liu ZM, Zhou XS. miRNA-218 Inhibits Osteosarcoma Cell Migration and Invasion by Down-regulating of TIAM1, MMP2 and MMP9. Asian Pacific journal of cancer prevention: APJCP. 2013;14(6):3681–4. pmid:23886165