Open Access

HIT000218960 promotes gastric cancer cell proliferation and migration through upregulation of HMGA2 expression

  • Authors:
    • Leitao Sun
    • Jieru Yu
    • Peipei Wang
    • Minhe Shen
    • Shanming Ruan
  • View Affiliations

  • Published online on: March 21, 2019     https://doi.org/10.3892/ol.2019.10176
  • Pages: 4957-4963
  • Copyright: © Sun et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The aim of the present study was to elucidate whether the long non‑coding RNA (lncRNA) HIT000218960 could accelerate the proliferative and migratory ability of gastric cancer (GC) cells by regulating high‑mobility group AT‑hook 2 (HMGA2) gene. The reverse transcription‑quantitative polymerase chain reaction was used to determine HIT000218960 and HMGA2 expression levels in GC tissues and cells. The HMGA2 protein level was detected by western blotting. A χ2 test was used to determine the association between the HIT000218960 expression level and the clinical characteristics of patients with GC. GC cells were transfected with small interfering (si)‑negative control, si‑HIT000218960 and si‑HIT000218960+pcDNA‑HMGA2, prior to assessing the cell proliferative and migratory ability using the Cell Counting Kit‑8 and Transwell assays, respectively. HIT000218960 and HMGA2 were highly expressed in GC tissues compared with in healthy tissues. In addition, HIT000218960 and HMGA2 were positively correlated in GC tissues. The HIT000218960 expression level was associated with tumor size, Tumor‑Node‑Metastasis staging and lymph node metastasis in patients with GC. HIT000218960 silencing decreased the proliferative and migratory ability of HGC27 and NCI‑N87 cells; however, HMGA2 overexpression partly reversed this inhibitory effect. The results of the present study indicated that HIT000218960 could promote HGC27 and NCI‑N87 cell proliferation and migration, which may be mediated by HMGA2.

Introduction

Gastric cancer (GC) is a malignancy that originates from the glandular epithelial cells of gastric mucosa. It is the second most common cause of cancer-associated mortality in China (1). According to the International Agency for Research on Cancer (https://www.iarc.fr/), GC was the fifth most common type of cancer worldwide in 2012 with 951,000 new cases identified, and represented the third most common causes of cancer-associated mortality, with 723,000 cases. In addition, >70% of new cases of GC occur in developing countries, and ~50% originate from Eastern Asia, mainly China (2). Treatment of GC essentially comprises surgery, chemotherapy and radiotherapy. However, the high rate of GC spreading to local lymph nodes, liver and peritoneal cavity contributes to a poor prognosis and a low 5-year survival rate (<30%) (3,4). Numerous studies suggest that the biological evolution of GC is complex. In China, GC is typically diagnosed at an advanced stage. It is therefore crucial to improve early diagnosis of GC and determine its underlying molecular mechanism in order to increase the 5-year survival rate and improve the quality of life of patients with GC (5,6).

Long non-coding RNA (lncRNA) is an RNA molecule of >200 nucleotides, which is not translated into protein (7). LncRNA was originally considered to be ‘noise’ in genome transcription. However, an increasing number of lncRNAs have been identified to serve a pivotal role in cancer development (8). A previous study (9) reported that lncRNA acts as a potential therapeutic target, which provides a novel direction for treatment development. Other studies (10,11) have revealed that lncRNAs are associated with stomach cancer development. A previous study identified that GACAT1 expression is correlated with tumor stage, metastasis, differentiation and invasion depth of GC (12). Yang et al (13) reported that H19 is overexpressed in GC and correlated with cancer cell proliferation and apoptosis. In addition, H19 interacts with p53 to suppress its activity, which leads to downregulation of the downstream Bax gene, and therefore results in increased cell proliferation and decreased cell apoptosis (13). Colon cancer-associated transcript 1 (CCAT1), which is overexpressed in GC, is associated with tumor size and lymphatic or distant metastasis (13,14). In vitro studies indicated that CCAT1 is associated with stomach carcinoma cell migration, which may result from c-Myc and E-box interaction (15). However, the expression profile and underlying mechanism of lncRNA in GC remain unclear. It is therefore crucial to investigate further the functions and underlying molecular mechanism of lncRNAs in GC.

High-mobility group AT-hook 2 (HMGA2) is a crucial protein, which is highly expressed during embryonic development and responsible for cell differentiation (16). The gene coding for HMGA2 is located on loci 13–15 of the long arm of chromosome 12 (17). Previous studies have reported that HMGA2 is overexpressed in various types of cancer, including thyroid (17), colon (18), non-small cell lung (19), squamous cell (20) and gastric (21) cancer, and is associated with invasion and metastasis of certain types of malignancy. Furthermore, HMGA2 is an independent factor that can predict the prognosis of certain malignant tumors (20). As a structural transcriptional regulator, HMGA2 is involved in numerous biological processes. For example, HMGA2 mediates glioblastoma cancer stem cell proliferation via forkhead box M1 and plasminogen activator urokinase regulation (22). In addition, HMGA2 serves a role in DNA damage repair (23,24). In addition, previous studies identified that HMGA2 promotes the epithelial-mesenchymal transition (EMT) by targeting Twist family basic helix-loop-helix transcription factor 1 (Twist1), Snail family transcriptional repressor 2 and Wnt/β signaling pathways in tumors (2528). In summary, HMGA2 serves important roles in the biological evolution of cancer. HIT000218960 is an lncRNA which has not been extensively investigated. A recent study identified that HMGA2 regulates HIT000218960 expression in thyroid carcinoma tissues (29); however, the association between HMGA2 and HIT000218960 in GC remains unclear.

To the best of our knowledge, the present study is the first to investigate the role of HIT000218960 and HMGA2 in GC. The present study clarified the regulatory effects of HIT000218960 and HMGA2 on cell proliferation and migration in gastric tumor, which may provide a novel strategy and potential target for the treatment of GC.

Materials and methods

Tissue samples and clinical characteristics of patients with GC

In total, 60 tumor samples were obtained from patients with GC who were admitted to The First Affiliated Hospital of Zhejiang Chinese Medical University (Hangzhou, China) between March 2016 and March 2017. A total of 25 normal tissues adjacent to cancer were selected as controls. All fresh tissues were quickly frozen in liquid nitrogen. The clinical characteristics of patients with GC were collected, including age, sex, tumor size, Tumor-Node-Metastasis (TNM) staging and lymph node metastasis. The present study was approved by the Ethics Committee of The First Affiliated Hospital of Zhejiang Chinese Medical University. All subjects provided written informed consent.

Cell culture and transfection

The HGC27 and NCI-N87 cell lines (purchased from Shanghai Huiying Biotechnology Co., Ltd., Shanghai, China), which are commonly used as GC cell lines, were selected for the present study (30,31). HGC27 and NCI-N87 cells were cultured in RPMI-1640 (HyClone; GE Healthcare Life Sciences, Logan, UT, USA) containing 10% fetal bovine serum (FBS) (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) and placed at 37°C in a humidified incubator containing 5% CO2. Culture medium was changed after 2 days and replaced according to the cell culture conditions. Cell lines were transfected with small interfering (si)-negative control (NC) (5′-UCCGGCGACCGGGUAACCUTT-3′), si-HIT000218960 (5′-CCUAACCCAGAGCUCUGAUUATT-3′) and si-HIT000218960+pcDNA-HMGA2, which were designed and synthesized by GenePharma Biotech Corp., Taipei, Taiwan. A total of 5 µl si-NC, si-HIT000218960 and pcDNA-HMGA2 were dissolved in 250 µl transfection dilution solution, containing LipoHigh transfection reagent (catalog no. E607403; Sangon Biotech Co., Ltd., Shangai China). The mixture was incubated at room temperature for 20 min. The cells were washed three times with PBS prior to transfection and the PBS was then replaced with fresh serum-free culture medium. The cells were incubated with the transfection mixture at 37°C in a 5% CO2 incubator and the medium was changed 6 h post-transfection.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA was isolated from tissues samples and cancer cells using TRIzol® (Invitrogen; Thermo Fisher Scientific, Inc.). RNA purity and concentration were detected using a Nanodrop 2000c spectrophotometer (Thermo Fisher Scientific, Inc.). The reverse transcription was operated using a PrimeScript™ RT kit (Takara Bio, Inc., Otsu, Japan) according to the manufacturer's protocol. Following reverse transcription, cDNA was diluted with diethy; pyrocarbonate at 1:2 and premixed. Subsequently, 1 µl cDNA, primers, premixed liquid and double-distilled water were mixed into a 20 µl volume in PCR tubes. Following centrifugation at 800 × g for 5 min at 4°C, PCR amplification was performed under the recommended parameters: 40 cycles of 50°C for 5 min, 95°C for 5 min, 95°C for 5 sec, 60°C for 15 sec and 72°C for 15 sec, followed by 94°C for 15 sec. The fluorescence signals were collected and analyzed. The primer sequences were as follows: HIT000218960 forward, 5-CCACCTACCCATCTGACTTTG-3′ and reverse, 5-CCACTATTTCCCACTGCCTT-3; HMGA2 forward, 5-AGAATCTGGTGCAGGAATGG-3 and reverse, 5-TCGTATTTAGTGTCTCCAGCC-3; and β-actin forward, 5′-CCTCGCCTTTGCCGATCC-3′ and reverse, 5′-GGATCTTCATGAGGTAGTCAGTC-3′. The relative RNA expression levels were analyzed using the 2−ΔΔCq method (32).

Western blotting

Cells were washed with PBS, and lysed using radioimmunoprecipitation assay buffer (Beyotime Institute of Biotechnology, Haimen, China) for 10 min on ice. The BCA method was used for protein quantification. Following centrifugation at 14,000 × g for 15 min at 4°C, the supernatant was collected. Proteins (30 µg) were separated by SDS-PAGE (12% gel), and transferred onto a polyvinylidene fluoride membrane (EMD Millipore, Billerica, MA, USA). Membranes were washed with Tris-buffered saline containing Tween-20 (TBST), and blocked with 5% skimmed milk with TBST at 25°C for 1 h. Membranes were then incubated with monoclonal primary antibody at 4°C overnight. The following primary antibodies were used: Anti-HMGA2 (1:500; catalog no. ab52039; Abcam, Cambridge, UK) and anti-GAPDH (1:500; catalog no. ab9485; Abcam). Membranes were washed with TBST for 30 min and incubated at 25°C for 1 h with goat anti-rabbit IgG H&L secondary antibody (1:1,000; catalog no. ab6940; Abcam). Enhanced chemiluminescence reagent (Thermo Fisher Scientific, Inc.) was used to detect the signal on the membrane.

Cell Counting Kit-8 (CCK-8) assay

Cells (2,000 cells/well) were seeded in 96-well plates. CCK-8 reagent (10 µl; Dojindo Molecular Technologies, Inc., Kumamoto, Japan) was added to each well at 4, 24, 48, 72, 96 and 120 h after seeding for 1.5 h. Each group comprised 6 wells and contained a blank control. The absorbance value of each well was determined using a microplate reader at a wavelength of 490 nm.

Migration assay

A Transwell assay was performed to determine the cell migratory ability. HCG2 and NCI-N87 cells (25×104) were digested, suspended in serum-free RPMI-1640 medium and seeded onto the upper non-Matrigel-coated chamber with a membrane pore size of 8.0-µm (Corning, Inc., Corning, NY, USA). The lower chamber was filled with 500 µl RPMI-1640 medium containing 10% FBS. After 2 h, cells that had invaded the lower chamber were fixed for 20 min with 95% ethanol at room temperature, stained for 20 min with 0.5% crystal violet at room temperature and washed with water. Subsequently, images were captured with a light microscope (magnification, ×400) and cells were counted.

Statistical analysis

SPSS software (version 22.0; IBM Corp., Armonk, NY, USA) was used to analyze data. Images were edited with GraphPad Prism 6.0 software (GraphPad Software, Inc., La Jolla, CA, USA). Clinical characteristics were evaluated using a χ2 test. Student's t-test was applied for comparing data between two groups. Comparison between multiple groups was performed using one-way analysis of variance followed a least significant difference multiple-range post hoc test. Pearson correlation analysis was used to determine the correlation between one dependent variable and single independent variable. P<0.05 was considered to indicate a statistically significant difference.

Results

Expression levels of HIT000218960 and HMGA2 in GC tissue samples

A total of 60 GC tissue samples and 25 normal control samples were collected. Expression levels of HIT000218960 and HMGA2 in the two groups were detected using RT-qPCR. The results indicated that HIT000218960 and HMGA2 expression levels were significantly higher in GC samples compared with in control samples (P<0.001; Fig. 1A and B, respectively). In addition, western blotting revealed that HMGA2 protein expression was higher in GC samples compared with in control samples (Fig. 1C). Further analysis indicated a positive correlation between HIT000218960 and HMGA2 expression levels in GC tissues (P<0.001; R2=0.6020; Fig. 1D). However, there was no difference in HIT000218960 and HMGA2 expression levels between HGC27 and NCI-N87 cell lines (Fig. 1E).

Association between HIT000218960 and clinical characteristics of patients with GC

To clarify the function of HIT000218960 in GC tissue, the association between the clinical characteristics and HIT000218960 expression levels of the 60 patients with GC was analyzed. Patients were divided into lower-expression and higher-expression groups according to the median level of HIT000218960. Patients with an expression level ≥2.1 were placed in the higher-expression group and patients with an expression level <2.1 were placed in the lower-expression group, and each group consisted of 30 patients (Table I). Analysis using a χ2 test indicated that the tumor size of patients in the higher-expression group was significantly larger compared with those in the lower-expression group (P=0.0019). In addition, compared with the lower-expression group, the rate of advanced TNM staging (P=0.0008) and the incidence of lymph node metastasis (P=0.0195) were higher in the higher-expression group (P=0.0008 and P=0.0195, respectively). These data suggested that the level of HIT000218960 expression was associated with tumor size, TNM stage and lymph node metastasis in patients with GC.

Table I.

Association between HIT000218960 expression level and clinical characteristics of patients with gastric cancer (n=60).

Table I.

Association between HIT000218960 expression level and clinical characteristics of patients with gastric cancer (n=60).

HIT000218960 expression level

Clinical characteristicnLow (n=30)High (n=30)P-value
Age, years
  ≤603114170.4383
  >60291613
Sex
  Male2812160.3006
  Female321814
Tumor size, cm
  ≤32820  80.0019a
  >3321022
TNM stage
  I–II3122  90.0019a
  III–IV29  821
Lymph node metastasis
  Negative2718  90.0195a
  Positive331221

a P<0.05. TNM, Tumor-Node-Metastasis.

HIT000218960 silencing inhibits HGC27 and NCI-N87 cell proliferation and migration

RT-qPCR analysis indicated that si-HIT000218960 transfection significantly decreased HIT000218960 expression levels in HGC27 and NCI-N87 cell lines (P<0.001; Fig. 2A). CCK-8 assay demonstrated that HGC27 and NCI-N87 cell proliferation was significantly decreased following si-HIT000218960 transfection compared with si-NC (P<0.001; Fig. 2B). The results of the Transwell assay indicated that HIT000218960 silencing decreased HGC27 and NCI-N87 cell migratory ability (Fig. 2C). These results suggested that decreasing HIT000218960 expression decreased GC cell proliferative and migratory ability.

HMGA2 mediates the regulatory effect of HIT000218960 on GC cell proliferation and migration

Western blotting indicated that HIT000218960 knockdown induced HMGA2 downregulation in HGC27 and NCI-N87 cells, whereas pcDNA-HMGA2 reversed this phenomenon (Fig. 3A). In order to further clarify the association between HIT000218960 and HMGA2, HGC27 and NCI-N87 cells were transfected with si-NC, si-HIT000218960 or si-HIT000218960+pcDNA-HMGA2. A CCK-8 assay revealed that HMGA2 overexpression reversed the si-HIT000218960-induced decrease in cell proliferation (Fig. 3B) and migration (Fig. 3C) of HGC27 and NCI-N87 cell lines. These results suggested that HIT000218960 may promote the proliferative and migratory ability of HGC27 and NCI-N87 cells through HMGA2 upregulation.

Discussion

GC is a common gastrointestinal cancer in China, which leads to high mortality and low 5-year survival rates. The treatment of GC is essentially based on conventional surgery; however, there is currently a lack of research on the mechanisms of occurrence and development of GC, compared with other gastrointestinal malignancies. It has been reported that lncRNAs are involved in numerous biological processes, and that dysregulation of RNA expression is associated with various types of disease, including cancer. Subsequently, studies have identified the underlying mechanisms of lncRNA in cancer. For example, the antisense non-coding RNA in the INK4 locus and HOX transcript antisense RNA can determine chromatin localization by recruiting chromatin-modifying complexes and can regulate histone modifications (33,34). To the best of our knowledge, only a few studies on HIT000218960 have been performed. A recent study reported that HIT000218960 is highly expressed in thyroid carcinoma tissues, and that HIT000218960 inhibition induces a decrease in the papillary thyroid carcinoma cell proliferative, migratory ability and invasive capacity (29). They also reported for the first time that HMGA2 regulates HIT00021896 expression. However, the effect of HIT000218960 in GC has not been yet reported. Further investigation on the mechanism of action of HIT000218960 in GC is therefore required. The aim of the present study was to investigate the effect of HIT000218960 in GC. The results demonstrated for the first time that HIT000218960 expression level was significantly higher in GC tissues compared with normal tissues, and that HIT000218960 knockdown inhibited HGC27 and NCI-N87 cell proliferative and migratory ability.

A previous study reported that HMGA2 overexpression is closely associated with the occurrence and development of GC. Motoyama et al (33) observed that HMGA2 expression level in cancerous tissue was high in 110 patients with gastric cancer and was positively correlated with infiltration depth and lymph node metastasis. In addition, follow-up analysis revealed that patients with a high HMGA2 expression level had a shorter survival time compared with that of healthy subjects. This was consistent with the results from the present study, where HMGA2 expression was higher in GC tissues compared with in normal tissues. In addition, HIT000218960 expression level was positively correlated with HMGA2 expression level in GC tissues. HMGA2, which is a non-histone protein present in the nucleus, can change chromosome structure and regulate the expression of various target genes (15). HMGA2 is now considered to be a oncogene (27). However, the underlying molecular mechanism of HMGA2 in tumors is not fully understood and may be associated with activation or translocation of certain oncogenes. Motoyama et al (33) reported that in the transforming growth factor β signaling pathway, Smad upregulates its downstream target genes, including Snail, Twist1 and Snail family transcriptional repressor 2 through HMGA2 and functions as an epithelial (E-)cadherin transcription inhibitor. This results in downregulation of E-cadherin expression and loss of intercellular polarity, which induces EMT. Currently, the association between HMGA2 and the clinical characteristics of patients is being investigated to determine whether HMGA2 can be used to distinguish benign from malignant tumors. For example, a high HMGA2 expression level is associated with N staging, 2-year metastasis and poor prognosis in patients with nasopharyngeal carcinoma (35,36). In the present study, the expression levels of HIT000218960 and HMGA2 were associated with the clinical characteristics of patients with GC. These results suggested that HIT000218960 may serve a tumorigenic role by upregulating HMGA2 expression.

In conclusion, the results of the present study indicated that HIT000218960 and HMGA2 were significantly overexpressed in GC tissues and HGC27 an NCI-N87 cell lines. HIT000218960 and HMGA2 expression levels were positively correlated in GC tissues, and the HIT000218960 expression level in patients with GC was associated with tumor size, TNM stage and lymph node metastasis. In addition, HIT000218960 promoted the HGC27 and NCI-N87 cell proliferative and migratory capacity by upregulating HMGA2 expression.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contributions

LS and SR designed the study and performed the experiments. JY, PW and MS collected the data. JY and PW analyzed the data. LS and SR prepared the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

This study was approved by the Ethics Committee of The First Affiliated Hospital of Zhejiang Chinese Medical University (Hangzhou, China). All subjects and/or guardians provided written informed consent.

Patients consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ and He J: Cancer statistics in China, 2015. CA Cancer J Clin. 66:115–132. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D and Bray F: Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 136:E359–E386. 2015. View Article : Google Scholar : PubMed/NCBI

3 

Ilson DH: Adjuvant treatment for gastric cancer: Too much is not enough. Lancet Oncol. 15:788–789. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Bagcchi S: Radiotherapy for testicular cancer increases gastric cancer risk. Lancet Oncol. 15:e5932014. View Article : Google Scholar : PubMed/NCBI

5 

Konishi H, Ichikawa D, Komatsu S, Shiozaki A, Tsujiura M, Takeshita H, Morimura R, Nagata H, Arita T, Kawaguchi T, 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

6 

Zirtiloglu A, Cil I, Mehmet VF and Tural D: What are the optimal treatment modalities according to age group in gastric cancer patients? J Buon. 22:312–319. 2017.PubMed/NCBI

7 

Spizzo R, Almeida MI, Colombatti A and Calin GA: Long non-coding RNAs and cancer: A new frontier of translational research? Oncogene. 31:4577–4587. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Batista PJ and Chang HY: Long noncoding RNAs: Cellular address codes in development and disease. Cell. 152:1298–1307. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Xia Y, Yan Z, Wan Y, Wei S, Bi Y, Zhao J, Liu J, Liao DJ and Huang H: Knockdown of long noncoding RNA GHET1 inhibits cellcycle progression and invasion of gastric cancer cells. Mol Med Rep. 18:3375–3381. 2018.PubMed/NCBI

10 

Lv Z, Zhang Y, Yu X, Lin Y and Ge Y: The function of long non-coding RNA MT1JP in the development and progression of gastric cancer. Pathol Res Pract. 214:1218–1223. 2018. View Article : Google Scholar : PubMed/NCBI

11 

Zhu C, Ma J, Li Y, Zhang Y and Da M: Low expression of long noncoding RNA MT1JP is associated with poor overall survival in gastric cancer patients: Protocol for meta-analysis. Medicine (Baltimore). 97:e103942018. View Article : Google Scholar : PubMed/NCBI

12 

Shi X, Wang X and Hua Y: LncRNA GACAT1 promotes gastric cancer cell growth, invasion and migration by regulating miR-149-mediated of ZBTB2 and SP1. J Cancer. 9:3715–3722. 2018. View Article : Google Scholar : PubMed/NCBI

13 

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

14 

Liu G, Xiang T, Wu QF and Wang WX: Curcumin suppresses the proliferation of gastric cancer cells by downregulating H19. Oncol Lett. 12:5156–5162. 2016. View Article : Google Scholar : PubMed/NCBI

15 

Yang F, Xue X, Bi J, Zheng L, Zhi K, Gu Y and Fang G: Long noncoding RNA CCAT1, which could be activated by c-Myc, promotes the progression of gastric carcinoma. J Cancer Res Clin Oncol. 139:437–445. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Henriksen J, Stabell M, Meza-Zepeda LA, Lauvrak SA, Kassem M and Myklebost O: Identification of target genes for wild type and truncated HMGA2 in mesenchymal stem-like cells. BMC Cancer. 10:3292010. View Article : Google Scholar : PubMed/NCBI

17 

Sun T, Zhao N, Zhao XL, Gu Q, Zhang SW, Che N, Wang XH, Du J, Liu YX and Sun BC: Expression and functional significance of Twist1 in hepatocellular carcinoma: Its role in vasculogenic mimicry. Hepatology. 51:545–556. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Wagenblast E, Soto M, Gutierrez-Angel S, Hartl CA, Gable AL, Maceli AR, Erard N, Williams AM, Kim SY, Dickopf S, et al: A model of breast cancer heterogeneity reveals vascular mimicry as a driver of metastasis. Nature. 520:358–362. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Williamson SC, Metcalf RL, Trapani F, Mohan S, Antonello J, Abbott B, Leong HS, Chester CP, Simms N, Polanski R, et al: Vasculogenic mimicry in small cell lung cancer. Nat Commun. 7:133222016. View Article : Google Scholar : PubMed/NCBI

20 

Wang JY, Sun T, Zhao XL, Zhang SW, Zhang DF, Gu Q, Wang XH, Zhao N, Qie S and Sun BC: Functional significance of VEGF-a in human ovarian carcinoma: Role in vasculogenic mimicry. Cancer Biol Ther. 7:758–766. 2008. View Article : Google Scholar : PubMed/NCBI

21 

Baeten CI, Hillen F, Pauwels P, de Bruine AP and Baeten CG: Prognostic role of vasculogenic mimicry in colorectal cancer. Dis Colon Rectum. 52:2028–2035. 2009. View Article : Google Scholar : PubMed/NCBI

22 

Zhong X, Liu X, Li Y, Cheng M, Wang W, Tian K, Mu L, Zeng T, Liu Y, Jiang X, et al: HMGA2 sustains self-renewal and invasiveness of glioma-initiating cells. Oncotarget. 7:44365–44380. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Fujikane R, Komori K, Sekiguchi M and Hidaka M: Function of high-mobility group A proteins in the DNA damage signaling for the induction of apoptosis. Sci Rep. 6:317142016. View Article : Google Scholar : PubMed/NCBI

24 

Yu H, Lim HH, Tjokro NO, Sathiyanathan P, Natarajan S, Chew TW, Klonisch T, Goodman SD, Surana U and Droge P: Chaperoning HMGA2 protein protects stalled replication forks in stem and cancer cells. Cell Rep. 6:684–697. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Tan EJ, Thuault S, Caja L, Carletti T, Heldin CH and Moustakas A: Regulation of transcription factor twist expression by the DNA architectural protein high mobility group A2 during epithelial-to-mesenchymal transition. J Biol Chem. 287:7134–7145. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Tan EJ, Kahata K, Idas O, Thuault S, Heldin CH and Moustakas A: The high mobility group A2 protein epigenetically silences the Cdh1 gene during epithelial-to-mesenchymal transition. Nucleic Acids Res. 43:162–178. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Li Y, Zhao Z, Xu C, Zhou Z, Zhu Z and You T: HMGA2 induces transcription factor Slug expression to promote epithelial-to-mesenchymal transition and contributes to colon cancer progression. Cancer Lett. 355:130–140. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Singh I, Mehta A, Contreras A, Boettger T, Carraro G, Wheeler M, Cabrera-Fuentes HA, Bellusci S, Seeger W, Braun T and Barreto G: Hmga2 is required for canonical WNT signaling during lung development. BMC Biol. 12:212014. View Article : Google Scholar : PubMed/NCBI

29 

Li T, Yang XD, Ye CX, Shen ZL, Yang Y, Wang B, Guo P, Gao ZD, Ye YJ, Jiang KW and Wang S: Long noncoding RNA HIT000218960 promotes papillary thyroid cancer oncogenesis and tumor progression by upregulating the expression of high mobility group AT-hook 2 (HMGA2) gene. Cell Cycle. 16:224–231. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Liu C, Ren L, Wang Y, Liu Y and Xiao J: The interaction between RACK1 and WEE1 regulates the growth of gastric cancer cell line HGC27. Oncol Lett. 14:4784–4792. 2017. View Article : Google Scholar : PubMed/NCBI

31 

Kawai S, Kato S, Imai H, Okada Y and Ishioka C: Suppression of FUT1 attenuates cell proliferation in the HER2-overexpressing cancer cell line NCI-N87. Oncol Rep. 29:13–20. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2−ΔΔCt method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

33 

Motoyama K, Inoue H, Nakamura Y, Uetake H, Sugihara K and Mori M: Clinical significance of high mobility group A2 in human gastric cancer and its relationship to let-7 microRNA family. Clin Cancer Res. 14:2334–2340. 2008. View Article : Google Scholar : PubMed/NCBI

34 

Tsai MC, Manor O, Wan Y, Mosammaparast N, Wang JK, Lan F, Shi Y, Segal E and Chang HY: Long noncoding RNA as modular scaffold of histone modification complexes. Science. 329:689–693. 2010. View Article : Google Scholar : PubMed/NCBI

35 

Xia YY, Yin L, Tian H, Guo WJ, Jiang N, Jiang XS, Wu J, Chen M, Wu JZ and He X: HMGA2 is associated with epithelial-mesenchymal transition and can predict poor prognosis in nasopharyngeal carcinoma. Onco Targets Ther. 8:169–176. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Xia YY, Yin L, Jiang N, Guo WJ, Tian H, Jiang XS, Wu J, Chen M, Wu JZ and He X: Downregulating HMGA2 attenuates epithelial-mesenchymal transition-induced invasion and migration in nasopharyngeal cancer cells. Biochem Biophys Res Commun. 463:357–363. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2019
Volume 17 Issue 6

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
Sun L, Yu J, Wang P, Shen M and Ruan S: HIT000218960 promotes gastric cancer cell proliferation and migration through upregulation of HMGA2 expression. Oncol Lett 17: 4957-4963, 2019
APA
Sun, L., Yu, J., Wang, P., Shen, M., & Ruan, S. (2019). HIT000218960 promotes gastric cancer cell proliferation and migration through upregulation of HMGA2 expression. Oncology Letters, 17, 4957-4963. https://doi.org/10.3892/ol.2019.10176
MLA
Sun, L., Yu, J., Wang, P., Shen, M., Ruan, S."HIT000218960 promotes gastric cancer cell proliferation and migration through upregulation of HMGA2 expression". Oncology Letters 17.6 (2019): 4957-4963.
Chicago
Sun, L., Yu, J., Wang, P., Shen, M., Ruan, S."HIT000218960 promotes gastric cancer cell proliferation and migration through upregulation of HMGA2 expression". Oncology Letters 17, no. 6 (2019): 4957-4963. https://doi.org/10.3892/ol.2019.10176