Abstract
Hepatocellular carcinoma (HCC) is one of the most prevailing malignancies, and the molecular mechanisms underlying HCC tumorigenesis remain to be further clarified. The aim of our present study was to determine the biological functions and clinical significance of SATB2 in HCC. Quantitative RT-PCR was performed to detect SATB2 mRNA expression in HCC tissues and cells. Cell proliferation, migration and invasion were investigated by MTT assay and transwell assay. Western blotting was performed to evaluate the protein expression levels. Tumor xenografts were established to explore the effects of SATB2 on HCC tumor growth in vivo. Our results indicated that SATB2 was highly expressed in HCC tissues and cell lines, and its high expression was closely correlated with aggressive clinical phenotypes and poor prognosis of HCC patients. Through gain- and loss-of function experiments, we found that SATB2 significantly promoted HCC cell proliferation, migration, and invasion in vitro and HCC tumorigenicity in vivo. Furthermore, SATB2 was identified as an important regulator of stem-like properties and epithelial to mesenchymal transition (EMT) in HCC cells. In summary, our results suggested that SATB2 is a crucial upregulator of HCC, and SATB2 might be a potential prognostic marker and a therapeutic target for HCC.
Keywords: Hepatocellular carcinoma, SATB2, prognosis, tumorigenesis, stemness feature, epithelial-mesenchymal transition
Introduction
Hepatocellular carcinoma (HCC), accounting for about 90% of primary liver cancers, is the sixth most prevalent cancer and the third most frequent cause of cancer-associated death [1]. China alone accounts for nearly 55% of all global HCC cases [2]. Many HCC patients do not have any symptoms until an advanced stage [3]. Despite of recent advances in surgical resection and medical treatment, the overall survival for HCC patients remains dismal largely due to the high rates of recurrence and metastasis [4]. Therefore, it is imperative to investigate the mechanisms underlying HCC metastasis and develop effective individual therapeutic strategies for HCC patients.
SATB2 (special AT-rich binding protein-2), encoded on chromosome 2q32-33, is an evolutionarily conserved transcription factor, which regulates the expression of diverse sets of genes by binding to the nuclear matrix attachment regions [5,6]. Recent studies have indicated that SATB2 is associated with cancer progression. The expression of SATB2 was upregulated in HCC, and SATB2 rescued the miR-211-mediated inhibition of HCC cell invasion and proliferation [7]. However, the specific mechanisms by which SATB2 regulate the malignant progression of HCC are still largely unknown.
In our study, we detected the mRNA and protein expression levels of SATB2 in HCC tissues and cell lines, and explored the relationship of SATB2 with clinical characteristics and prognosis of HCC patients. Moreover, we further investigated the effects as well as the potential molecular mechanism of SATB2 involved in proliferation, migration and invasion of HCC cells in vitro. Our findings indicated the potential value of SATB2 in the clinical diagnosis and treatment of HCC.
Materials and methods
Human tissue samples
HCC tissue samples and their adjacent non-cancerous tissues (2 cm from the edge of the cancerous region) were obtained from 103 patients diagnosed with HCC who had undergone a routine hepatic resection at Sichuan Provincial People’s Hospital (Chengdu, China). The clinicopathological data of these patients are shown in Table 1. Patients who had preoperative radiotherapy or chemotherapy were excluded. Tissue samples were immediately snap-frozen in liquid nitrogen and stored at -80°C till use. This study was approved by the Ethics Committee of Sichuan Provincial People’s Hospital, and all patients provided written informed consent before enrollment in the study.
Table 1.
Characteristics | Total number (n=103) | SATB2 expression | P value | |
---|---|---|---|---|
| ||||
Low (n=59) | High (n=44) | |||
Age (years) | 0.182 | |||
≤50 | 38 | 25 | 13 | |
>50 | 65 | 34 | 31 | |
Gender | 0.371 | |||
Male | 70 | 38 | 32 | |
Female | 33 | 21 | 12 | |
HBsAg status | 0.847 | |||
Positive | 81 | 46 | 35 | |
Negative | 22 | 13 | 9 | |
Serum AFP level (ng/ml) | 0.251 | |||
≥400 | 76 | 41 | 35 | |
<400 | 27 | 18 | 9 | |
Liver cirrhosis | 0.357 | |||
Yes | 65 | 35 | 30 | |
No | 38 | 24 | 14 | |
Tumor size (cm) | 0.049 | |||
<5 | 56 | 37 | 19 | |
≥5 | 47 | 22 | 25 | |
Number of tumor nodules | 0.082 | |||
1 | 83 | 51 | 32 | |
≥2 | 20 | 8 | 12 | |
TNM stage | 0.017 | |||
I+II | 65 | 43 | 22 | |
III+IV | 38 | 16 | 22 | |
Edmondson-Steiner grading | 0.215 | |||
I+II | 61 | 38 | 23 | |
III+IV | 42 | 21 | 21 | |
Vascular invasion | 0.023 | |||
Yes | 43 | 19 | 24 | |
No | 60 | 40 | 20 |
Cell culture and transfection
Five HCC cell lines (SMMC-7721, Huh-7, HepG2, HCCLM3 and Hep3B) and one normal liver cell line L02, obtained from the Cell Bank of Chinese Academy of Sciences (Shanghai, China), were cultured in Dulbecco’s modified Eagle’s medium (DMEM; Hyclone, Logan, UT, USA) supplemented with 10% fetal bovine serum (FBS; Biowest, Loire, France) in a humidified incubator with 5% CO2 at 37°C.
The full-length SATB2 cDNA was amplified and cloned into the pcDNA3.1 (+) vector (Invitrogen, Carlsbad, CA, USA). The oligonucleotides to inhibit SATB2 expression were synthesized and inserted to the shRNA expression vector pGPH1/Neo (GenePharma, Shanghai, China). HepG2 cells were transfected with pcDNA3.1-SATB2 for overexpressing SATB2, and SMMC-7721 cells were transfected with sh-SATB2 for suppression of SATB2 using Lipofectamine 3000 (Invitrogen) following the manufacturer’s protocol.
Quantitative RT-PCR
Total RNA was extracted from using TRIzol reagent (Invitrogen). 1 μg of total RNA was used for complementary DNA synthesis using the Primer Script RT reagent Kit (Takara, Dalian, China). Quantitative RT-PCR was conducted using SYBR Green Real-time PCR Master Mix (TOYOBO, Shanghai, China) on the LightCycler 480 II System (Roche, Basel, Switzerland). Comparative quantification was determined using the 2-ΔΔCt method [8]. GAPDH was used as an internal control. The following primers were used: SATB2 forward: 5’-GGAGAACGACAGCGAGGAA-3’, reverse: 5’-CCGATGTATTGCTTTGCCTAGT-3’; GAPDH forward: 5’-ACCGCAATTGGGAGTCAGGATT-3’, reverse: 5’-CCAATGTGTCCAGGAGGATT-3’.
Western blotting
Total protein was isolated using RIPA lysis buffer (Beyotime, Shanghai, China) with PMSF (Roche, Basel, Switzerland). Equal amounts of total protein were separated by SDS-PAGE and then transferred onto PVDF membrane (Millipore, Bedford, MA, USA). The membrane was blocked in 5% nonfat dry milk in TBST for 2 h and then incubated with appropriate primary antibodies in 4°C for one night. Next the membranes were incubated with HRP-conjugated secondary antibodies for 1 h at room temperature. The proteins of interest were detected using Image-Pro Plus software 6.0 (Media Cybernetics Inc., Rockville, MD, USA), and GAPDH was used as an internal control.
MTT assay
The proliferation of HCC cells was measured using 3-(4,5-dimethylthiazal-2-yl)-2,5-diphenyl-tetrazolium bromide (MTT) assay. Briefly, approximately 2,000 cells were seeded in a 96-well culture plate incubated for 24, 48, 72 and 96 h. At various time points, 0.5 mg/ml MTT solution was added to each well, and then the plate was incubated at 37°C for 4 h. Next, the supernatants were removed and 100 μL of DMSO was added to solubilize the formed crystals. Absorbance levels were measured at the wavelength of 490 nm.
Transwell assay
HCC cell migration and invasion were evaluated using Matrigel-uncoated and -coated transwell inserts (8 μm pore size; BD Biosciences, Erembodegem, Belgium). Briefly, 2 × 104 cells suspended in 200 μL serum-free medium were into the upper chamber, and the lower chamber was filled with 500 µl medium with 10% FBS. After 48 h of incubation at 37°C, the cells that had traversed the membrane were fixed with using 4% paraformaldehyde and stained with 0.1% crystal violet dye. The images were obtained and cells were counted using a light microscope.
Tumorsphere formation assay
About 1 × 103 single HCC cells were plated onto 6-well ultralow attachment plates (Corning Inc., Corning, NY, USA) in DMEM/F12 supplemented with 2% B27 (Life Technologies, Invitrogen, USA), 20 ng/ml human EGF, 10 ng/ml human bFGF and 5 μg/ml insulin. After 14 days at 37°C, plates were observed for tumorsphere formation.
Tumor xenografts
The 6-week-old BALB/c-nu mice, purchased from Shanghai Laboratory Animals Center (Shanghai, China), were randomized into two groups (n=8/group). SMMC-7721 cells (1 × 106 cells in 200 μl PBS) transfected with sh-SATB2 or sh-NC were inoculated subcutaneously into the right flank of the nude mice. Tumor volume was measured by an external caliper and calculated using the formula: Volume =0.5 × width2 × length. 28 days after inoculation, the mice were killed and the tumors were excised and weighted. All experimental procedures were carried out with the approval of the Animal Care and Use Committee of Sichuan Provincial People’s Hospital.
Statistical analysis
Statistical analyses were performed by using GraphPad Prism 6.0 software (GraphPad Software Inc., San Diego, CA, USA) and SPSS 20.0 software (IBM, Chicago, IL, USA). Data are expressed as the mean ± standard deviation (SD) from at least three independent experiments, and inter-group differences were assessed for significance using two-tailed unpaired Student’s t-test. Chi-square test was applied to analyze the correlation of SATB2 expression and clinicopathological features of HCC patients. Association between SATB2 expression and overall survival was estimated using Kaplan-Meier method and evaluated using the log-rank test. A P value <0.05 was considered to indicate statistical significance.
Results
SATB2 is highly expressed in human HCC tissues and cell lines
Firstly, the expression levels of SATB2 in HCC tissues and cell lines were measured by quantitative RT-PCR and western blot analysis. The results showed that SATB2 mRNA was expressed at low levels in the adjacent normal liver tissues, while it was significantly upregulated in the HCC tissues (Figure 1A). Then, we detected the expression levels of SATB2 in a panel of HCC cell lines, and found that the expression levels of SATB2 was significantly increased in HCC cells when compared to normal human hepatocyte cell line L02 (Figure 1B, 1C). The expression of SATB2 was most highly in SMMC-7721 cells and was lowest in HepG2 cells. Thus, these two cell lines were chosen for subsequent analysis.
Clinical significance of SATB2 expression in HCC patients
In addition, to understand the clinical significance of SATB2 upregulation in HCC, HCC patients were divided into two groups according to the SATB2 expression in HCC tissues, including the high-expression group (n=44) and the low-expression group (n=59). Then we analyzed the relationship between SATB2 expression and clinicopathological characteristics of 103 HCC patients. As shown in Table 1, increased SATB2 expression was significantly associated with tumor size (P=0.049), TNM stage (P=0.017) and vascular invasion (P=0.023). Additionally, the overall survival was higher in HCC patients with lower SATB2 expression than in those with higher SATB2 expression (P=0.015; Figure 1D).
SATB2 promotes HCC cell proliferation, migration and invasion in vitro
Next, we explored the regulatory functions of SATB2 in HCC cells in vitro. We knocked down SATB2 in SMMC-7721 cells and overexpressed SATB2 in HepG2 cells. As expected, the expression of SATB2 was significantly modulated by pcDNA3.1-SATB2 and sh-SATB2 in HepG2 and SMMC-7721 cells (Data not shown). MTT assay revealed that cell proliferation was significantly promoted by overexpression of SATB2 in HepG2 cells; conversely, proliferation of SMMC-7721 cells was suppressed after knockdown of SATB2 (Figure 2A). Additionally, transwell assay demonstrated that HepG2 cells with SATB2 overexpression displayed more invasive and migratory properties, while knockdown of SATB2 significantly reduced the number of SMMC-7721 cells crossing the membrane (Figure 2B).
Downregulation of SATB2 inhibits tumorigenesis of HCC cells in vivo
The effects of SATB2 on tumorigenesis of HCC cells were further determined in vivo. SMMC-7721 cells transfected with sh-SATB2 or sh-NC were injected into the flanks of nude mice. As shown in Figure 3A, 3B, downregulation of SATB2 markedly inhibited the growth of HCC xenografts in nude mice. Moreover, the average tumor weight was also remarkably decreased in the sh-SATB2 group compared with that of sh-NC group (Figure 3C).
SATB2 induces stem-like properties in HCC cells
Considering stem-like cells as an important factor for tumor metastasis, we also examined assessed whether SATB2 modulated the stem-like properties in HCC cells. The result showed that overexpression of SATB2 increased the numbers and sizes of spheres formed by HepG2 cells, whereas knockdown of SATB2 reduced the numbers and sizes of spheres made by SMMC-7721 cells (Figure 4A). Stem cell markers, including Nanog, Oct4 and Sox2, were upregulated in SATB2-overexpressing HepG2 cells and downregulated when SATB2 was silenced in SMMC-7721 cells (Figure 4B).
SATB2 induces epithelial to mesenchymal transition in HCC cells
EMT is of critical importance for migration and invasion of HCC cells. Accordingly we determined to find whether SATB2 could exert effects on EMT of HCC cells. The results demonstrated that overexpression of SATB2 in HepG2 cells led to a remarkable increase in the mesenchymal makers, including N-cadherin and Vimentin, and a significant decrease in the epithelial markers (E-cadherin) (Figure 5A). On the contrary, as shown in Figure 5B, inhibition of SATB2 in SMMC-7721 cells decreased the mesenchymal markers whereas increased epithelial markers.
Discussion
Surgery is still the best therapeutic strategy for HCC. Metastasis is a key cause of poor long-term survival after surgical resection in HCC patients [9]. It is widely accepted that cancer metastasis is a complex multistep process that involves a myriad of genetic alterations. SATB2 is universally expressed in a variety of tissues, and serves important roles in multiple biological processes. However, the impact of SATB2 in cancerogenesis is still controversial. Overexpression of SATB2 suppressed cell proliferation and tumor progression ability in laryngeal squamous cell carcinoma [10], gastric cancer [11] and non-small cell lung cancer [12]. But other study also reported that SATB2 was upregulated in breast cancer cell lines [13]. Therefore SATB2 has been shown to serve as either an oncogenic factor or a tumor suppressor, with their functions depending on specific tissue samples. In the present study we found that SATB2 was significantly upregulated in HCC tissues and cell lines, and SATB2 expression was positively associated with unfavorable prognosis of HCC patients. Besides, gain- and loss-of-function experiment validated that SATB2 overexpression promoted the migratory and invasive capacities of HCC cells whereas SATB2 knockdown reduced these metastatic behaviors in vitro.
The dissemination of cancerous cells from the primary tumor must acquire specific characteristics, including migratory and invasive abilities, which allow them to reestablish in a secondary site [14]. Epithelial-to-mesenchymal transition (EMT) plays an important role in regulating metastasis and invasion in malignant tumors, including HCC [15]. During EMT, epithelial cells lose their apical-basal polarity and transition to a mesenchymal phenotype [16]. Downregulation of SATB2 is sufficient to induce EMT and promote migration and invasion in non-small cell lung cancer cells [17]. We found that overexpression of SATB2 significantly up-regulated the expression of N-Cadherin and Vimentin, and down-regulated the expression of E-Cadherin, further promoting the process of EMT in HCC cells. EMT has emerged as an important regulator of cancer cells exhibiting stem cell-like properties [18]. Stemness characteristics in cancer cells are also responsible for cancer relapse and metastasis [19]. SATB2 has been reported to induce cellular transformation and stemness in pancreatic cancer [20]. But SATB2 might serve as a negative regulator of stemness in colorectal cancer cells [21]. Here, we revealed that overexpression of SATB2 promotes stem-like characteristics in HCC cells, as evidenced by an increase in cell spheres and the expression of stem cell markers.
In summary, our data demonstrated that the aberrant high expression of SATB2 in HCC promotes the process of EMT and stemness. In view of our present results, we hypothesize that SATB2 might be an attractive target for prognostic and therapeutic interventions in HCC. Future studies should be performed to determine the precise mechanisms of the aberrant expression of SATB2 in HCC.
Disclosure of conflict of interest
None.
References
- 1.Forner A, Llovet JM, Bruix J. Hepatocellular carcinoma. Lancet. 2012;379:1245–1255. doi: 10.1016/S0140-6736(11)61347-0. [DOI] [PubMed] [Google Scholar]
- 2.Yuen MF, Hou JL, Chutaputti A. Hepatocellular carcinoma in the Asia pacific region. J Gastroenterol Hepatol. 2009;24:346–353. doi: 10.1111/j.1440-1746.2009.05784.x. [DOI] [PubMed] [Google Scholar]
- 3.Bruix J, Gores GJ, Mazzaferro V. Hepatocellular carcinoma: clinical frontiers and perspectives. Gut. 2014;63:844–855. doi: 10.1136/gutjnl-2013-306627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Schwartz M, Roayaie S, Konstadoulakis M. Strategies for the management of hepatocellular carcinoma. Nat Clin Pract Oncol. 2007;4:424–432. doi: 10.1038/ncponc0844. [DOI] [PubMed] [Google Scholar]
- 5.FitzPatrick DR, Carr IM, McLaren L, Leek JP, Wightman P, Williamson K, Gautier P, McGill N, Hayward C, Firth H, Markham AF, Fantes JA, Bonthron DT. Identification of SATB2 as the cleft palate gene on 2q32-q33. Hum Mol Genet. 2003;12:2491–2501. doi: 10.1093/hmg/ddg248. [DOI] [PubMed] [Google Scholar]
- 6.Szemes M, Gyorgy A, Paweletz C, Dobi A, Agoston DV. Isolation and characterization of SATB2, a novel AT-rich DNA binding protein expressed in development- and cell-specific manner in the rat brain. Neurochem Res. 2006;31:237–246. doi: 10.1007/s11064-005-9012-8. [DOI] [PubMed] [Google Scholar]
- 7.Jiang G, Cui Y, Yu X, Wu Z, Ding G, Cao L. miR-211 suppresses hepatocellular carcinoma by downregulating SATB2. Oncotarget. 2015;6:9457–9466. doi: 10.18632/oncotarget.3265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Thomsen R, Solvsten CA, Linnet TE, Blechingberg J, Nielsen AL. Analysis of qPCR data by converting exponentially related Ct values into linearly related X0 values. J Bioinform Comput Biol. 2010;8:885–900. doi: 10.1142/s0219720010004963. [DOI] [PubMed] [Google Scholar]
- 9.Tang ZY, Ye SL, Liu YK, Qin LX, Sun HC, Ye QH, Wang L, Zhou J, Qiu SJ, Li Y, Ji XN, Liu H, Xia JL, Wu ZQ, Fan J, Ma ZC, Zhou XD, Lin ZY, Liu KD. A decade’s studies on metastasis of hepatocellular carcinoma. J Cancer Res Clin Oncol. 2004;130:187–196. doi: 10.1007/s00432-003-0511-1. [DOI] [PubMed] [Google Scholar]
- 10.Liu TR, Xu LH, Yang AK, Zhong Q, Song M, Li MZ, Hu LJ, Chen FJ, Hu ZD, Han P, Zeng MS. Decreased expression of SATB2: a novel independent prognostic marker of worse outcome in laryngeal carcinoma patients. PLoS One. 2012;7:e40704. doi: 10.1371/journal.pone.0040704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Wu L, Chen J, Qin Y, Mo X, Huang M, Ru H, Yang Y, Liu J, Lin Y. SATB2 suppresses gastric cancer cell proliferation and migration. Tumour Biol. 2016;37:4597–4602. doi: 10.1007/s13277-015-4282-5. [DOI] [PubMed] [Google Scholar]
- 12.Ma YN, Zhang HY, Fei LR, Zhang MY, Wang CC, Luo Y, Han YC. SATB2 suppresses nonsmall cell lung cancer invasiveness by G9a. Clin Exp Med. 2017 doi: 10.1007/s10238-017-0464-3. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
- 13.Luo LJ, Yang F, Ding JJ, Yan DL, Wang DD, Yang SJ, Ding L, Li J, Chen D, Ma R, Wu JZ, Tang JH. MiR-31 inhibits migration and invasion by targeting SATB2 in triple negative breast cancer. Gene. 2016;594:47–58. doi: 10.1016/j.gene.2016.08.057. [DOI] [PubMed] [Google Scholar]
- 14.Tam WL, Weinberg RA. The epigenetics of epithelial-mesenchymal plasticity in cancer. Nat Med. 2013;19:1438–1449. doi: 10.1038/nm.3336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.van Zijl F, Zulehner G, Petz M, Schneller D, Kornauth C, Hau M, Machat G, Grubinger M, Huber H, Mikulits W. Epithelial-mesenchymal transition in hepatocellular carcinoma. Future Oncol. 2009;5:1169–1179. doi: 10.2217/fon.09.91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Thompson EW, Newgreen DF, Tarin D. Carcinoma invasion and metastasis: a role for epithelial-mesenchymal transition? Cancer Res. 2005;65:5991–5995. doi: 10.1158/0008-5472.CAN-05-0616. discussion 5995. [DOI] [PubMed] [Google Scholar]
- 17.Kucuksayan H, Ozes ON, Akca H. Downregulation of SATB2 is critical for induction of epithelial-to-mesenchymal transition and invasion of NSCLC cells. Lung Cancer. 2016;98:122–129. doi: 10.1016/j.lungcan.2016.05.032. [DOI] [PubMed] [Google Scholar]
- 18.Scheel C, Weinberg RA. Phenotypic plasticity and epithelial-mesenchymal transitions in cancer and normal stem cells? Int J Cancer. 2011;129:2310–2314. doi: 10.1002/ijc.26311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Mishra L, Banker T, Murray J, Byers S, Thenappan A, He AR, Shetty K, Johnson L, Reddy EP. Liver stem cells and hepatocellular carcinoma. Hepatology. 2009;49:318–329. doi: 10.1002/hep.22704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Yu W, Ma Y, Shankar S, Srivastava RK. Role of SATB2 in human pancreatic cancer: Implications in transformation and a promising biomarker. Oncotarget. 2016;7:57783–57797. doi: 10.18632/oncotarget.10860. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Li Y, Liu YH, Hu YY, Chen L, Li JM. Special AT-rich sequence-binding protein 2 acts as a negative regulator of stemness in colorectal cancer cells. World J Gastroenterol. 2016;22:8528–8539. doi: 10.3748/wjg.v22.i38.8528. [DOI] [PMC free article] [PubMed] [Google Scholar]