RUNX2 promotes hepatocellular carcinoma cell migration and invasion by upregulating MMP9 expression

  • Authors:
    • Qian Wang
    • Wei Yu
    • Tao Huang
    • Yan Zhu
    • Changshan Huang
  • View Affiliations

  • Published online on: September 19, 2016     https://doi.org/10.3892/or.2016.5101
  • Pages: 2777-2784
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Runt-related transcription factor 2 (RUNX2) was first identified as a transcription factor to play an important role in different biological processes of osteoblast and chondrocyte, including differentiation and migration. Recently, RUNX2 has been implicated in promigratory/proinvasive behavior in different human malignancies. In the present study, we demonstrated that the RUNX2 mRNA and protein expression were both increased significantly in HCC tissues and cell lines. High RUNX2 expression was correlated obviously with poor clinicopathological characteristics including multiple tumor nodes, high histological grading, venous infiltration and advanced tumor-node-metastasis (TNM) stage. In addition, we demonstrated that RUNX2 was a prognostic indicator for predicting 5-year overall survival and disease-free survival of HCC patients. Our studies showed that RUXN2 overexpression promoted, while RUNX2 knockdown inhibited HCC cell migration and invasion in vitro. Notably, RUNX2 positively regulated matrix metalloproteinase 9 (MMP9) accumulation in HCC cells. Furthermore, we confirmed that RUNX2 was positively correlated with MMP9 expression in HCC tissues by Pearson correlation analysis. Mechanistically, we demonstrated that MMP9 overexpression increased HCC cell migration and invasion, while MMP9 knockdown reduced HCC cell migration and invasion in vitro. Alteration of MMP9 expression partially abrogated the effects of RUNX2 on HCC cell migration and invasion, which suggests that RUNX2 developed its pro-metastatic biological function by upregulating the expression of MMP9 in HCC cells. In conclusion, our results reveal that RUNX2 promotes HCC cell migration and invasion by MMP9-mediated pathway, and potentially serves as a new prognostic biomarker and in therapeutic strategies for HCC.

Introduction

Hepatocellular carcinoma (HCC) is one of the most common cancers worldwide and the third leading cause of cancer-related death globally (1). Despite considerable advances in treatment modalities, the long-term survival of HCC patients remains poor because of its high recurrence and early metastasis (2). However, the underlying mechanism responsible for the development and progression of HCC has not been fully elucidated (3). Therefore, it is urgent to develop a novel therapeutic target involved in HCC.

Runt-related transcription factor 2 (RUNX2), which belongs to RUNX family, are distinctive by a highly conserved 128 amino acid DNA binding/protein-protein interaction domain (4). It functions as a major regulator for osteoblast differentiation and regulates endochondral bone formation physiological progression (57). Recent studies suggested that RUNX2 are involved in many types of human cancer development, progression and metastasis (810). In vitro studies showed that RUNX2 promoted the migration and invasion capacity of prostate cancer cells (11,12). In breast cancer, RUNX2 promoted tumorsphere formation by regulating soluble E-cadherin expression associated with the TAZ transcriptional co-activator (13). Moreover, it was recently reported that RUNX2 played a critical role in the process of epithelial to mesenchymal transition (EMT) whose characteristics include increased migration, invasion and metastasis potential by upregulating the transcription factors, such as SOX9 and SMAD3 (12). In addition, RUNX2 was essential for cellular movement and cytoskeleton remodeling. In non-small cell lung cancer, the increased RUNX2 presents resistance to cisplatin chemosensitivity. Furthermore, RUNX2 promoted migration and invasion potential of thyroid tumor cells by activating the expression levels of MMPs (14). These studies suggest that RUNX2 probably functions as an oncogene for tumorigenesis and metastasis. However, the precise function of RUNX2 and the underlying mechanisms in HCC remain unclear.

In the present study, we demonstrated that RUNX2 expression is upregulated in HCC. Clinical analysis reveals that the increased RUNX2 expression was associated with poor prognostic features and was an independent prognostic marker for predicting survival of HCC patients. RUNX2 promotes cell migration and invasion by regulating MMP9 expression in HCC cells. Mechanistically, the pro-metastatic effect of RUNX2 could be abrogated by inhibiting MMP9 expression in vitro. Our data suggest that RUNX2 probably promotes MMP9 expression and thus, induces the metastasis of HCC cells.

Materials and methods

Clinical tissues and data

Ninety-six HCC tissues and matched tumor-adjacent tissues were obtained from patients including 82 males and 14 females, who underwent curative resection surgery in the Department of Hepato-Biliary-Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University from January 2006 to December 2009. All tissues were used after obtaining informed consent. Patients did not receive preoperative chemotherapy or embolization. The demographic and clinicopathological data were obtained through medical records. The experimental protocols were approved by the Zhengzhou University Ethics Committee according to the Declaration of Helsinki.

Cell lines and transfection

The human HCC cell lines (HepG2, MHCC-97L, Hep3B, SMMC-7721, MHCC-97H and HCCLM3) and immortalized normal hepatic cell line LO2 were purchased from the Institute of Biochemistry and Cell Biology (Chinese Academy of Sciences, Shanghai, China). The cells were cultured in complete Dulbecco's modified Eagle's medium (DMEM; Invitrogen, Carlsbad, CA, USA) supplemented with 10% fetal bovine serum (FBS; Gibco, Grand Island, NY, USA) at 37°C in a humidified incubator in 5% CO2.

Retroviral vectors pMMP-RUNX2 and pMMP-MMP9 were generated by inserting the cDNA into pMMP. The specific siRNA against RUNX2 (5′-UAACAGCAGAGGCAUUUCGUAGCUC-3′), MMP9 (5′-CUAUGGUCCUCGCCCUGAA-3′) and scramble siRNA (5′-UUCUCCGAACGUGUCACGUUUGUGC-3′) were synthesized by Shanghai GenePharma Co., Ltd. (Shanghai, China). Cells were transfected with the siRNAs mentioned above using Lipofectamine 2000 (Invitrogen) according to the manufacturer's instructions.

Real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR)

Total RNA was extracted from respective HCC cells and clinical samples using TRIzol reagent (Invitrogen). The first strand cDNA was reverse transcribed with miRNA assay kit (Applied Biosystems, Foster City, CA, USA) and quantified by a SYBR® Premix Ex Taq™ II (Perfect Real-Time) kit (Takara Bio, Inc., Shiga, Japan) and performed on the ABI PRISM 7300 Sequence Detection System (Applied Biosystems). qPCR primer against RUNX2 (HQP016478) and GAPDH (HQP006940) were purchased from Genecopoeia (Guangzhou, China).

Western blot analysis

The HCC cells and clinical tissues were collected and lysed, then the protein concentration was quantified using the BCA reagent (Pierce, Rockford, IL, USA). A total of 30 µg protein was separated by 10% SDS-PAGE and transferred onto a PVDF membrane (Bio-Rad Laboratories, Hercules, CA, USA). The membranes were probed with the following primary antibodies: RUNX2 (1:1,000; Cell Signaling Technology, Inc., Danvers, MA, USA), MMP9 (1:1,000; Santa Cruz Biotechnology, Santa Cruz, CA, USA), GAPDH (G8140; US Biological, Salem, MA, USA) overnight. The membranes were incubated with appropriate HRP-conjugated secondary antibody (ZSGB-BIO, Beijing, China). Protein bands were visualized using an enhanced chemiluminescence kit (Amersham, Little Chalfont, UK).

Immunohistochemical staining

Samples were fixed in formalin and embedded in paraffin and sections were cut at 4 µm thickness. RUNX2 and MMP9 (1:400; Cell Signaling Technology) antibody was performed in immunohistochemistry according to a standard streptavidin-peroxidase-conjugated (SP-IHC) procedures. The staining results for the RUNX2 and MMP9 protein were semi-quantitatively evaluated by the staining intensity and the percentage of positive staining cells. The percentage of positive cells was divided into four grades: 0 for <5%; 1 for 6–25%; 2 for 26–50%; 3 for 51–75% and 4 for >75%. Staining intensity was assessed by four degrees: 0, negative; 1, weak; 2, moderate; and 3, strong. Each section was assayed for ten independent high magnification (×400) fields to get the average scores.

Cell migration and invasion assays

Transwell cell migration and invasion assays were carried out by the 8 µM pore-sized Transwell inserts (Nalge Nunc International Corp., Naperville, IL, USA). Transfected cells were seeded at 2.5×105/ml in 200 µl serum-free DMEM medium into the upper chamber, and 750 µl DMEM medium containing 10% FBS was placed in the lower chamber. After 24-h incubation, cells were fixed in 4% paraformaldehyde for 20 min and stained with 0.1% crystal violet dye for 15 min. The cells on the inner layer were softly removed with a cotton swab; 1:6 dilution Matrigel invasion chamber (BD Biosciences, San Jose, CA, USA) was performed for invasion assays and the following assays were same as before.

Statistical analysis

Results are shown as mean ± standard deviation. The SPSS 13 (SPSS, Inc., Chicago, IL, USA) and GraphPad Prism 5 software (GraphPad Software, Inc., San Diego, CA, USA) were used for Pearson Chi-squared test and the multivariate Cox regression analysis. Two-tailed Student's t-test, a Kaplan-Meier plot, a log-rank test, a Spearman's rank correlation coefficient or an ANOVA were used to evaluate the statistical significance. Difference was defined as P<0.05.

Results

The expression level of RUNX2 in HCC tissues and cells

We investigated the expression level of RUNX2 in 96 pairs of HCC tissues and corresponding adjacent non-tumor tissues by qRT-PCR and western blot analysis. We found that RUNX2 mRNA and protein expression in HCC tissues were both obviously higher than those in corresponding adjacent non-tumor tissues (P<0.05; Fig. 1A and B). Moreover, RUNX2 mRNA levels were increased in HCC cell lines (HepG2, Hep3B, SMMC-7721, MHCC-97L, HCCLM3 and MHCC-97H) compared to the normal hepatocyte cell line, LO2 (P<0.05; Fig. 1C). In addition, the levels of RUNX2 protein showed the same result with mRNA levels in all cell lines by western blot analysis (P<0.05; Fig. 1D). Furthermore, RUNX2 expression in MHCC-97H and HCCLM3, which was considered as highly metastasis were prominently higher than those in the low metastasis HCC cell lines (HepG2, Hep3B, SMMC-7721 and MHCC-97L) (P<0.05; Fig. 1D). Thus, these results suggest that elevated RUNX2 expression probably plays a critical role in the development of HCC.

Correlation between RUNX2 expression and clinicopathological features

We defined the mean level of RUNX2 protein as a cut-off value to distinguish the RUNX2 expression level. As shown in Table I, the high expression of RUNX2 protein was significantly associated with multiple tumor nodes (P=0.006), venous infiltration (P=0.009), high Edmondson-Steiner grading (P=0.001) and advanced tumor-node-metastasis (TNM) stage (P= 0.004). Hence, these data suggest that the increased expression of RUNX2 is correlated with adverse prognostic features of HCC. In addition, Kaplan-Meier analysis revealed that high RUNX2 expression was correlated with worse overall survival (P=0.0005; Fig. 2A) and disease-free survival (P=0.0001, Fig. 2B) of HCC patients. In addition, RUNX2 expression was a novel independent factor for predicting both 5-year overall and disease-free survival in HCC patients (P=0.021 and 0.039, respectively, Table II). Taken together, these results highlight the potential value of RUNX2 for the outcome of HCC.

Table I

Clinical correlation of RUNX2 expression in HCC (n=96).

Table I

Clinical correlation of RUNX2 expression in HCC (n=96).

Clinical parametersCases
(n)
Expression level
P-value (aP<0.05)
RUNX2high
(n=61)
RUNX2low
(n=35)
Age (years)
 <652818100.923
 ≥65684325
Gender
 Male8252300.950
 Female1495
Tumor size (cm)0.128
 <5372710
 ≥5593425
Tumor number 0.006a
 Solitary764333
 Multiple20182
Edmondson 0.001a
 I+II321319
 III+IV644816
TNM stage 0.004a
 I+II653530
 III+IV31265
Venous infiltration 0.009a
 Present26224
 Absent703931
AFP (ng/ml)0.619
 <400382315
 ≥400583820
HBsAg0.810
 Positive845331
 Negative1284

Table II

Multivariate Cox regression analysis of 5-year overall and disease-free survival of 96 HCC patients.

Table II

Multivariate Cox regression analysis of 5-year overall and disease-free survival of 96 HCC patients.

VariablesOverall survival
Disease-free survival
HR95% CIP-valueHR95% CIP-value
RUNX22.1791.107–4.6580.021a3.6481.084–5.1030.039a
Edmondson grade2.1480.846–5.0820.0681.9810.834–4.5960.135
TNM stage1.1341.013–1.9480.023a1.0231.004–1.7680.003a
No. of tumor nodule1.7120.684–4.2490.2141.1320.954–3.8210.873
Venous infiltration1.0780.987–1.2470.0571.0230.658–1.7840.856

{ label (or @symbol) needed for fn[@id='tfn1-or-36-05-2777'] } HR, hazard ratio; CI, confidence interval;

a statistically significant.

RUNX2 promotes HCC cell migration and invasion

To explore the biological function of RUNX2 in HCC, we transfected HCC cell line MHCC-97L with empty vector (EV) or RUNX2 retroviruses (P<0.01; Fig. 3A). We demonstrated that RUNX2 overexpression prominently promoted HCC cell migration and invasion in MHCC-97L (P<0.05; Fig. 3B). Moreover, RUNX2 was knocked down using a specific siRNA in HCCLM3 cells (P<0.05; Fig. 3C). As expected, downregulated RUNX2 led to a significant reduction of cell migration and invasion (P<0.05; Fig. 3D). These data suggest that RUNX2 regulates the migration and invasion of HCC cells.

RUNX2 positively regulates MMP9 in HCC

The positive correlation between RUNX2 and MMP9 expression was reported previously in breast cancer (1517), thus, we evaluated the effect of RUNX2 on MMP9. Amazingly, our results showed that RUNX2 overexpression obviously increased the expression level of MMP9 in MHCC-97L cells (P<0.05; Fig. 4A). Moreover, RUNX2 knockdown significantly decreased the level of MMP9 in HCCLM3 cells (P<0.05; Fig. 4B). We then analysed RUNX2 and MMP9 expression by immunostaining in HCC samples. Both RUNX2 and MMP9 expression in cancer tissues were significantly higher than those in paired non-cancerous tissues (P<0.05; Fig. 4C). Moreover, IHC scores were evaluated for semi-quantitative analysis, we found a strong positive correlation between RUNX2 and MMP9 (r=0.534, P<0.001; Fig. 4D). These data revealed that RUNX2 exerts its biological promotive function through elevating expression of MMP9.

RUNX2 promotes HCC cell migration and invasion by increasing MMP9

To explore whether MMP9 participates in RUNX2 mediated promotion of HCC cell migration and invasion, MMP9 expression was significantly increased by MMP9 retroviruses in MHCC-97L cells (P<0.05; Fig. 5A) and significantly knocked down using a specific siRNA in HCCLM3 cells (P<0.05; Fig. 5C). As shown in Fig. 5B and 5D, MMP9 overexpression obviously promoted, while MMP9 knockdown obviously blocked HCC cell migration and invasion (P<0.05; respectively). In addition, RUNX2 overexpressing MHCC-97L cells were subsequently transfected with MMP9 siRNA (P<0.05; Fig. 6A). MMP9 knockdown markedly abrogated the function of exogenous RUNX2 overexpression, causing a significant decrease in the number of migrated and invaded cells (P<0.05; Fig. 6B). Similarly, MMP9 overexpression promoted HCC cell migration and invasion in RUNX2-knockdown HCCLM3 cells (P<0.05, respectively; Fig. 6C and D). In conclusion, these results suggest that MMP9 may function as a downstream factor in RUNX2 mediated promotion of HCC cell migration and invasion.

Discussion

In the present study, we initially checked RUNX2 expression level in 96 specimens of HCC tissues. Our results show that the RUNX2 expression level in HCC was obviously higher than that in non-tumor tissues. Increased expression levels of RUNX2 mRNA and protein were also confirmed in different HCC cell lines, especially in the highly metastatic cell lines. Moreover, elevated expression of RUNX2 was significantly correlated with multiple tumor nodes, high histological grade, TNM stage and venous infiltration. These data suggest that the increased RUNX2 expression is correlated with adverse prognostic characters in HCC. Furthermore, patients with higher RUNX2 had a worse prognosis of HCC patients, which was consistent with that in acute lymphoblastic leukemia patients (18). Multivariate Cox repression analysis demonstrated that RUNX2 was a novel independent prognostic factor for predicting survival of HCC patients. Taken together, these results indicate that RUNX2 expression is critical for prognosis of HCC patients.

Previous studies suggest that RUNX2 functions as an oncogene in breast cancer development through Wnt and TGF-β signaling pathways (1921). Moreover, downregulated RUNX2 inhibits the invasion of osteosarcoma (22). In the present study, we found that RUNX2 overexpression prominently promoted MHCC-97L cell migration and invasion and RUNX2 knockdown significantly reduced the migrated and invaded HCCLM3 cells. These data suggest that RUNX2 exerts its biological function on HCC cell migration and invasion. Peng et al (23) reported that RUNX2 promoted trophoblast invasion via MMP9 expression. Our data demonstrated that RUNX2 overexpression upregulated MMP9 in MHCC-97L cells and RUNX2 knockdown led to MMP9 reduction in HCCLM3 cells. In addition, we demonstrated that the MMP9 expression level in high RUNX2-expressed HCC tissues was prominently higher than that from low RUNX2-expressed group. Spearman correlation analysis suggest a positive correlation between RUNX2 and MMP9 expression in HCC tissues. Taken together, these data indicate that RUNX2 positively regulate MMP9 accumulation in HCC. Furthermore, we confirmed that MMP9 overexpression obviously promoted HCC cell migration and invasion in MHCC-97L cells, while MMP9 knockdown obviously inhibited migration and invasion in HCCLM3 cells, which was consistent with previous reports. Moreover, the promotion in HCC migration and invasion by RUNX2 overexpression could be abolished by MMP9 knockdown, and the suppression in HCC cell migration and invasion by RUNX2 knockdown could be reverted by restoring MMP9 expression. Taken together, these data indicate that RUNX2 promotes HCC cell migration and invasion by increasing MMP9 expression. In osteoblast, RUNX2 exerts as a regulator in metastasis by interaction with PI3K/AKT signaling (24,25). Similarly, in breast cancer, RUNX2 directly regulates the expression of MMP9 and MMP13. Furthermore, RUNX2 overexpression upregulates transcription factors (SOX9, SMAD3 and SNAI2) implicated in the process of epithelial to mesenchymal transition (12,2629). Therefore, we need further investigation to explore the molecular mechanisms between RUNX2 and MMP9 in HCC.

In conclusion, the data show that the expression of RUNX2 is elevated in HCC tissues and cell lines and its high expression is associated with malignant clinicopathological features. We confirm that RUNX2 is an independent prognostic marker for predicting 5-year survival of HCC patients. We demonstrate that RUNX2 promotes HCC cell migration and invasion in vitro. Mechanistically, we suggest that RUNX2 may promote HCC invasion and metastasis by increasing MMP9. Taken together, we consider that RUNX2 may potentially act as a clinical biomarker, and may also be a therapeutic target in HCC.

Acknowledgments

The present study was supported by a grant from the Scientific Research Foundation of Henan (no. 092102310090).

References

1 

El-Serag HB and Rudolph KL: Hepatocellular carcinoma: Epidemiology and molecular carcinogenesis. Gastroenterology. 132:2557–2576. 2007. View Article : Google Scholar : PubMed/NCBI

2 

Talwalkar JA and Gores GJ: Diagnosis and staging of hepatocellular carcinoma. Gastroenterology. 127(Suppl 1): S126–S132. 2004. View Article : Google Scholar : PubMed/NCBI

3 

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

4 

Ito Y: Oncogenic potential of the RUNX gene family: 'overview'. Oncogene. 23:4198–4208. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Li J, Hao L, Wu J, Zhang J and Su J: Linarin promotes osteogenic differentiation by activating the BMP-2/RUNX2 pathway via protein kinase A signaling. Int J Mol Med. 37:901–910. 2016.PubMed/NCBI

6 

Ozaki T, Nakamura M and Shimozato O: Novel implications of DNA damage response in drug resistance of malignant cancers obtained from the functional interaction between p53 family and RUNX2. Biomolecules. 5:2854–2876. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Sugimoto H, Nakamura M, Yoda H, Hiraoka K, Shinohara K, Sang M, Fujiwara K, Shimozato O, Nagase H and Ozaki T: Silencing of RUNX2 enhances gemcitabine sensitivity of p53-deficient human pancreatic cancer AsPC-1 cells through the stimulation of TAp63-mediated cell death. Cell Death Dis. 6:e19142015. View Article : Google Scholar : PubMed/NCBI

8 

Wysokinski D, Blasiak J and Pawlowska E: Role of RUNX2 in breast carcinogenesis. Int J Mol Sci. 16:20969–20993. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Shrivats AR, McDermott MC, Klimak M, Averick SE, Pan H, Matyjaszewski K, Mishina Y and Hollinger JO: Nanogel-mediated RNAi against Runx2 and Osx inhibits osteogenic differentiation in constitutively active BMPR1A osteoblasts. ACS Biomater Sci Eng. 1:1139–1150. 2015. View Article : Google Scholar

10 

Fujita T, Azuma Y, Fukuyama R, Hattori Y, Yoshida C, Koida M, Ogita K and Komori T: Runx2 induces osteoblast and chondrocyte differentiation and enhances their migration by coupling with PI3K-Akt signaling. J Cell Biol. 166:85–95. 2004. View Article : Google Scholar : PubMed/NCBI

11 

Akech J, Wixted JJ, Bedard K, van der Deen M, Hussain S, Guise TA, van Wijnen AJ, Stein JL, Languino LR, Altieri DC, et al: Runx2 association with progression of prostate cancer in patients: Mechanisms mediating bone osteolysis and osteoblastic metastatic lesions. Oncogene. 29:811–821. 2010. View Article : Google Scholar :

12 

Baniwal SK, Khalid O, Gabet Y, Shah RR, Purcell DJ, Mav D, Kohn-Gabet AE, Shi Y, Coetzee GA and Frenkel B: Runx2 transcriptome of prostate cancer cells: Insights into invasiveness and bone metastasis. Mol Cancer. 9:2582010. View Article : Google Scholar : PubMed/NCBI

13 

Brusgard JL, Choe M, Chumsri S, Renoud K, MacKerell AD Jr, Sudol M and Passaniti A: RUNX2 and TAZ-dependent signaling pathways regulate soluble E-cadherin levels and tumorsphere formation in breast cancer cells. Oncotarget. 6:28132–28150. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Sancisi V, Borettini G, Maramotti S, Ragazzi M, Tamagnini I, Nicoli D, Piana S and Ciarrocchi A: Runx2 isoform I controls a panel of proinvasive genes driving aggressiveness of papillary thyroid carcinomas. J Clin Endocrinol Metab. 97:E2006–E2015. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Mendoza-Villanueva D, Deng W, Lopez-Camacho C and Shore P: The Runx transcriptional co-activator, CBFbeta, is essential for invasion of breast cancer cells. Mol Cancer. 9:1712010. View Article : Google Scholar : PubMed/NCBI

16 

Pratap J, Javed A, Languino LR, van Wijnen AJ, Stein JL, Stein GS and Lian JB: The Runx2 osteogenic transcription factor regulates matrix metalloproteinase 9 in bone metastatic cancer cells and controls cell invasion. Mol Cell Biol. 25:8581–8591. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Selvamurugan N, Kwok S and Partridge NC: Smad3 interacts with JunB and Cbfa1/Runx2 for transforming growth factor-beta1-stimulated collagenase-3 expression in human breast cancer cells. J Biol Chem. 279:27764–27773. 2004. View Article : Google Scholar : PubMed/NCBI

18 

Edwards H, Xie C, LaFiura KM, Dombkowski AA, Buck SA, Boerner JL, Taub JW, Matherly LH and Ge Y: RUNX1 regulates phosphoinositide 3-kinase/AKT pathway: Role in chemotherapy sensitivity in acute megakaryocytic leukemia. Blood. 114:2744–2752. 2009. View Article : Google Scholar : PubMed/NCBI

19 

Tandon M, Chen Z, Othman AH and Pratap J: Role of Runx2 in IGF-1Rβ/Akt- and AMPK/Erk-dependent growth, survival and sensitivity towards metformin in breast cancer bone metastasis. Oncogene. Jan 25–2016.Epub ahead of print. View Article : Google Scholar

20 

Sancisi V, Gandolfi G, Ragazzi M, Nicoli D, Tamagnini I, Piana S and Ciarrocchi A: Cadherin 6 is a new RUNX2 target in TgF-β signalling pathway. PLoS One. 8:e754892013. View Article : Google Scholar

21 

van der Deen M, Akech J, Wang T, Fitzgerald TJ, Altieri DC, Languino LR, Lian JB, van Wijnen AJ, Stein JL and Stein GS: The cancer-related Runx2 protein enhances cell growth and responses to androgen and TGFbeta in prostate cancer cells. J Cell Biochem. 109:828–837. 2010.PubMed/NCBI

22 

Zeng H and Xu X: RUNX2 RNA interference inhibits the invasion of osteosarcoma. Oncol Lett. 9:2455–2458. 2015.PubMed/NCBI

23 

Peng B, Zhu H, Klausen C, Ma L, Wang YL and Leung PC: GnRH regulates trophoblast invasion via RUNX2-mediated MMP2/9 expression. Mol Hum Reprod. 22:119–129. 2016. View Article : Google Scholar

24 

Cohen-Solal KA, Boregowda RK and Lasfar A: RUNX2 and the PI3K/AKT axis reciprocal activation as a driving force for tumor progression. Mol Cancer. 14:1372015. View Article : Google Scholar : PubMed/NCBI

25 

Tandon M, Chen Z and Pratap J: Runx2 activates PI3K/Akt signaling via mTORC2 regulation in invasive breast cancer cells. Breast Cancer Res. 16:R162014. View Article : Google Scholar : PubMed/NCBI

26 

Niu DF, Kondo T, Nakazawa T, Oishi N, Kawasaki T, Mochizuki K, Yamane T and Katoh R: Transcription factor Runx2 is a regulator of epithelial-mesenchymal transition and invasion in thyroid carcinomas. Lab Invest. 92:1181–1190. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Zhang X, Akech J, Browne G, Russell S, Wixted JJ, Stein JL, Stein GS and Lian JB: Runx2-Smad signaling impacts the progression of tumor-induced bone disease. Int J Cancer. 136:1321–1332. 2015. View Article : Google Scholar :

28 

Chimge NO, Baniwal SK, Little GH, Chen YB, Kahn M, Tripathy D, Borok Z and Frenkel B: Regulation of breast cancer metastasis by Runx2 and estrogen signaling: The role of SNAI2. Breast Cancer Res. 13:R1272011. View Article : Google Scholar : PubMed/NCBI

29 

Zaidi SK, Sullivan AJ, van Wijnen AJ, Stein JL, Stein GS and Lian JB: Integration of Runx and Smad regulatory signals at transcriptionally active subnuclear sites. Proc Natl Acad Sci USA. 99:8048–8053. 2002. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2016
Volume 36 Issue 5

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang Q, Yu W, Huang T, Zhu Y and Huang C: RUNX2 promotes hepatocellular carcinoma cell migration and invasion by upregulating MMP9 expression. Oncol Rep 36: 2777-2784, 2016
APA
Wang, Q., Yu, W., Huang, T., Zhu, Y., & Huang, C. (2016). RUNX2 promotes hepatocellular carcinoma cell migration and invasion by upregulating MMP9 expression. Oncology Reports, 36, 2777-2784. https://doi.org/10.3892/or.2016.5101
MLA
Wang, Q., Yu, W., Huang, T., Zhu, Y., Huang, C."RUNX2 promotes hepatocellular carcinoma cell migration and invasion by upregulating MMP9 expression". Oncology Reports 36.5 (2016): 2777-2784.
Chicago
Wang, Q., Yu, W., Huang, T., Zhu, Y., Huang, C."RUNX2 promotes hepatocellular carcinoma cell migration and invasion by upregulating MMP9 expression". Oncology Reports 36, no. 5 (2016): 2777-2784. https://doi.org/10.3892/or.2016.5101