Nucleosome-binding protein HMGN2 exhibits antitumor activity in human SaO2 and U2-OS osteosarcoma cell lines

  • Authors:
    • Guojun Liang
    • Enjie Xu
    • Chaoqun Yang
    • Chenglin Zhang
    • Xiaolong Sheng
    • Xuhui Zhou
  • View Affiliations

  • Published online on: December 22, 2014     https://doi.org/10.3892/or.2014.3689
  • Pages: 1300-1306
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

High mobility group N (HMGNs) are members of the high mobility group protein family, and are involved in the development and progression of several tumors. HMGN1 and HMGN5 were previously shown to be associated with the bioactivities of osteosarcoma. However, the effects and molecular mechanisms of HMGN2 on osteosarcoma progression remain to be determined. In order to characterize the endogenous expression of HMGN2 in osteosarcoma cell lines, RT-PCR and western blot analysis were performed. Recombinant HMGN2 lentivirus was used to infect the osteosarcoma cell lines with relatively low HMGN2 expression to determine the functional relevance of HMGN2 overexpression in osteosarcoma cell growth and migration in vitro and in vivo, and to investigate the expression levels of Ki-67, PCNA, cyclin D1 and cyclin E. The results showed that osteosarcoma cell proliferation and migration were significantly reduced by HMGN2, as indicated by cell count and wound-healing assays. Cell apoptosis was markedly induced and HMGN2 increased the sensitivity to chemotherapy. When HMGN2 expression was enhanced, the expression of cyclin D1 and PCNA was downregulated in osteosarcoma cells. In addition, the tumor volumes in SaO2 and U2-OS subcutaneous nude mouse models treated with HMGN2 lentivirus were significantly decreased as compared to those of the GFP group. These results suggested that the enhanced expression of HMGN2 in osteosarcoma cells by HMGN2 lentivirus, exerts inhibitory effects on growth and migration of osteosarcoma cells.

Introduction

Osteosarcoma accounts for ~20% of all primary bone cancers, and is the second highest cause of cancer-associated mortality in the pediatric age group (1,2). Although modern surgery and neo-adjuvant chemotherapy have developed over the past decades, ~35% of patients are likely to succumb to the disease within 5 years of diagnosis (3). Originating from cells of the osteoblast lineage, osteosarcoma cells exhibit complex and unbalanced karyotypes, characterized by numerous recurrent DNA amplifications as well as chromosomal abnormalities (4).

The high mobility group protein family (HMG) consists of three subfamilies, including the high mobility group B (HMGB), high mobility group A (HMGA) and high mobility group N (HMGN) (5). Each subfamily appears to exert a single characteristic nuclear function, specifically binding to nucleosomes and contributing to the diversity of chromatin function (6). Among them, HMGN2 is an abundant, highly conserved cell protein, widely known as a nuclear DNA-binding protein, which stabilizes nucleosome formation and facilitates gene transcription.

Findings of previous studies demonstrated that HMGN2 was one of the most abundant non-histone nuclear proteins in vertebrates and invertebrates (6,7). Subsequently, it was found that HMGN2 was mainly involved in the growth of tumor vascular endothelia and played an important role in tumorigenesis (8). The aberrant expression of HMGN2 was correlated with several tumors (9,10). In vitro, HMGN2 was released by the stimulation of IL-2 in human peripheral blood mononuclear leukocytes (8). HMGN2 protein was demonstrated to inhibit growth and induce apoptosis in the Tca8113 oral squamous cell carcinoma cell line (11). In vivo, HMGN2 significantly inhibited growth of the transplanted tumor in nude mice (12).

Therefore, HMGN2 is a newly identified gene associated with cancer growth and metastasis, representing a new therapeutic target for the treatment of cancer. However, the effects and molecular mechanisms of HMGN2 on osteosarcoma progression have not yet been comprehensively explored. In the present study, we examined the endogenous expression of HMGN2 in osteosarcoma cell lines using RT-PCR and western blot analysis. HMGN2 lentivirus was used to infect the osteosarcoma SaO2 and U2-OS cell lines with a relatively low HMGN2 expression to determine the functional relevance of HMGN2 overexpression in osteosarcoma cell growth and migration in vitro, and to examine the underlying signaling pathway involved in the progression of osteosarcoma.

Materials and methods

Materials

MG63, HOS, SaO2 and U2-OS osteosarcoma cell lines were purchased from the American Type Culture Collection (ATCC, Vanassas, MA, USA). HMGN2 lentivirus vector, negative control vector GFP and virion-packaging elements were purchased from Genechem (Shanghai, China). All the antibodies were purchased from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA, USA).

Pharmaceuticals and reagents

TRIzol reagent, Lipofectamine 2000, Double stain apoptosis detection kit (Annexin V-FITC/PI), Dulbecco’s modified Eagle’s medium (DMEM) and fetal bovine serum (FBS) were purchased from Gibco Invitrogen Corporation (Grand Island, NY, USA). 2-(2-methoxy-4-nitrophenyl)-3-(4-nitrophenyl)-5-(2,4-disulfo-phenyl)-2H-tetrazolium (WST-8) was purchased from Beyotime (Beyotime, Haimen, China) and SYBR-Green master mixture was purchased from Takara (Otsu, Japan).

Overexpression of HMGN2

Human-HMGN2 cDNA was prepared by RT-PCR (Fig. 1A) and inserted into the BamHI and XhoI restriction sites of the pGC-FU-3FLAG vector plasmid (Addgene, Cambridge, MA, USA). For the stable overexpression of HMGN2, HEK293T cells were plated in 75-cm2 culture flasks and transfected with 10 μg HMGN2 or GFP (control) lentivirus vectors. The medium was changed and the viral supernatant was harvested 48 h later. Viral-containing medium was collected and passed through 0.45-μm syringe filters. SaO2 and U2-OS cells were incubated with the lentivirus supernatant for 24 h and selected with 2 μg/ml puromycin (Sigma, St. Louis, MO, USA) according to the manufacturer’s instructions. The clone in which the HMGN2 lentivirus vectors transfected was designated as the HMGN2 group, the negative control GFP transfected vectors was designated as the GFP group and SaO2 and U2-OS osteosarcoma cells were designated as the CON group.

RT-qPCR

To quantitatively determine the expression level of HMGN2 in osteosarcoma cell lines, HMGN2 mRNA expression was determined by RT-qPCR using SYBR-Premix Ex Taq (Takara, Japan) and an ABI Prism 7500 sequence detection system (Applied Biosystems, USA). Total RNA of each clone was extracted with TRIzol according to the manufacturer’s instructions. The genes were amplified using specific oligonucleotide primers and human β-actin gene was used as an endogenous control. The PCR primer sequences used were: HMGN2, forward: 5′-CCAGCCATCAGCCATGAGGGT-3′ and reverse: 5′-GGAGCCCTTTCTGAATCCGCA-3′); β-actin, forward: 5′-GCGGGAAATCGTGCGTGACATT-3′ and reverse: 5′-GGCAGATGGTCGTTTGGCTGAATA-3′. Data were analyzed using the comparative Ct method (2−ΔΔCt). Three separate experiments were performed for each clone.

Western blot assay

Treated cells were collected and extracted using an assay kit (Beyotime, China) and the concentration was determined by using a Bio-Rad protein assay (Hercules, CA, USA). Equal amounts of cell extracts were separated on SDS-PAGE gels according to the molecular weight of the tested proteins. Separated protein bands were transferred onto polyvinylidene fluoride membranes and blocked in 5% skimmed milk powder. The primary antibodies against HMGN2, cyclin E, cyclin D1, PCNA, and Ki-67 (all from Santa Cruz Biotechnology) were diluted according to the instructions and incubated overnight at 4°C. Horseradish peroxidase-linked secondary antibodies were added at a dilution ratio of 1:1,000, and incubated at room temperature for 2 h. The membranes were washed with phosphate-buffered saline three times and the immunoreactive bands were visualized using an ECL-Plus kit according to the manufacturer’s instructions [Multi Sciences (Lianke) Biotech Co., Ltd., Hangzhou, China]. The relative protein level in different cell lines was normalized to β-actin concentration. Three separate experiments were performed for each clone.

Cell proliferation assay

Cell proliferation was analyzed with the cell count assay using WST-8 kits. Briefly, cells infected with HMGN2 were incubated in 96-well plates at a density of 1×105 cells/well with DMEM with 10% FBS. The cells were treated with 10 μl WST at 6, 12 and 24 h. The color reaction was measured at 570 nm with enzyme immunoassay analyzer (Bio-Rad Laboratories, Hercules, CA, USA). The proliferative activities were calculated for each clone.

Cell cycle analysis

For the cell cycle analysis, harvested cells were centrifuged at 1,000 × g for 5 min, washed with phosphate buffered saline and fixed in 70% ethanol overnight. The cells were then treated with 100 μg/ml propidium iodide plus RNase (10 μg/ml) for 30 min. The cell-cycle phase distribution was determined by analytical DNA flow cytometry (FACSCalibur; BD Biosciences, Becton-Dickinson, San Jose, CA, USA) as described by Evans et al (13).

Wound-healing assay

SaO2 and U2-OS cells were plated in each well of a 6-well culture plate and allowed to grow to 90% confluence. Treatment with HMGN2 lentivirus was then performed. The following day, a wound was created using a 10 μl micropipette tip. The migration of cells towards the wound was monitored daily, and images were captured at time intervals of 24 h.

In vivo tumor xenograft studies

Three mice were injected subcutaneously with 1×108 SaO2 and U2-OS cells in 50 ml of PBS pre-mixed with an equal volume of Matrigel matrix (Becton Dickinson). When the tumor size reached ~5 mm in length, sections were surgically removed, cut to 1-mm3, and re-seeded individually into other mice. When the tumor size reached ~5 mm in length, the mice were randomly assigned to the GFP and HMGN2 groups, in which 15 ml of lentivirus was injected into subcutaneous tumors using a multi-site injection format. The injections were repeated on the third day after initial treatment. The tumor volume was measured every three days with a caliper, using the formula volume = (length × width)2/2.

Statistical analysis

The results obtained were expressed as the mean ± standard error values from at least three independent experiments. One-way analysis of variance (ANOVA) was used to analyze the differences between groups. The LSD method of multiple comparisons was used when the probability for ANOVA was statistically significant. P<0.05 was considered to indicate statistical significance.

Results

Endogenous expression of HMGN2 in osteosarcoma cell lines

The endogenous expression of HMGN2 in human osteosarcoma SaO2, MG-63, U2-OS and HOS cell lines was evaluated using RT-qPCR and western blot analysis. As shown in Fig. 1A and B, there were different levels of mRNA and protein expression of HMGN2 in SaO2, MG-63, U-2 OS, and HOS cell lines, while the expression levels of HMGN2 were significantly reduced in the SaO2 and U-2 OS cell lines than those in the MG-63 and HOS cell lines.

Since HMGN2 exhibited a low expression in SaO2 and U-2 OS cell lines, the cell lines were selected as the infective objects of HMGN2 lentivirus. The infection efficiencies of HMGN2 (at a multiplicity of infection =50) in SaO2 and U-2 OS cell lines were >80% under fluorescence microscopy (Fig. 2C). To confirm that HMGN2 successfully transfected into SaO2 and U-2 OS cells, western blot analysis was applied. After 48 h following HMGN2 or GFP lentivirus infection and the expression of HMGN2 protein was significantly increased in the HMGN2 group (Fig. 1D).

HMGN2 inhibits osteosarcoma cell growth

Deregulated cell proliferation is a hallmark of cancer (16). To determine the effect of HMGN2 overexpression on SaO2 and U-2 OS cell growth, we investigated the proliferative activities by WST assay. Overexpression of HMGN2 significantly reduced the proliferative activities of SaO2 and U-2 OS cells compared with the GFP and CON groups (Fig. 2A). In addition, PCNA and Ki-67, which were indicators for cell proliferation, were examined by western blot assay to determine whether HMGN2 overexpression suppressed cell growth through translational repression. The expression of PCNA protein was significantly decreased in Lenti-HMGN2 group compared with the CON and GFP groups (P<0.01) while Ki-67 did not alter among these groups (Fig. 2B). These data suggested that the overexpression of HMGN2 may inhibit osteosarcoma cell proliferation through the downregulation of PCNA expression.

Effect of HMGN2 on osteosarcoma cycle distribution

The cycle distribution of SaO2 and U-2 OS cells was also analyzed. As shown in Fig. 3A, the percentage of untransfected SaO2 and U-2 OS cells in S phase was 52.1 and 58%, respectively, whereas the percentage of SaO2 and U-2 OS cells transfected with HMGN2 in S phase was 35.5 and 33%, respectively. In addition, 10 and 15% of untreated SaO2 and U-2 OS cells were in the G2/M phase compared with 5 and 8% of cells transfected with HMGN2 lentivirus. These results indicated that cell cycle was arrested in G0/G1 phase in HMGN2 group compared with the CON and GFP groups. Subsequently, cyclin D1 and cyclin E, two regulators of cell cycle progression from G1 to S phase, were assessed by western blotting (17). The results showed that only HMGN2 led to a decreased level of cyclin D1, whereas no significant changes were identified in the levels of cyclin E (Fig. 3B), suggesting that HMGN2 modulates the cell cycle through the regulation of cyclin D1.

Effect of HMGN2 on osteosarcoma cell invasion and metastasis

To determine the effect of HMGN2 on osteosarcoma cell invasion and metastasis, Transwell assay and wound-healing assay were carried out. As was shown in Fig. 4A, the migrative ability of SaO2 and U-2 OS cells in the HMGN2 group was lower than that in the CON and GFP groups. However, there were no significant differences between the CON and GFP groups. Furthermore, a Transwell assay was performed to determine the ability of cells to invade a matrix barrier. The representative micrographs of Transwell filters are shown in Fig. 4B. The invasive cell count demonstrated that invasive potential was significantly reduced in the HMGN2 group relative to the CON and GFP groups.

HMGN2 increases apoptosis and sensitivity to chemotherapy

The action mechanism of many anticancer factors is based on their ability to induce apoptosis. Consequently, SaO2 and U-2 OS osteosarcoma cells treated with HMGN2 lentivirus underwent apoptosis as their mode of cell death. At the end of the incubation period for 24, 48 and 72 h, the osteosarcoma cells were stained using an Annexin V-FITC/PI detection kit. As shown in Fig. 4C, the number of SaO2 and U-2 OS apoptotic cells in the HMGN2 group significantly increased compared with that in the GFP and CON groups at various time points. To further investigate whether HMGN2 affected the sensitivity to chemotherapy, the apoptosis in HMGN2-overexpressed osteosarcoma cells was induced by chemotherapy. A significant increase of apoptosis incidence was detected in cells treated with Dox, Cis, and Mtx for 24 h (Fig. 4D). Overexpression of HMGN2 resulted in a further increase of apoptosis induced by these anticancer agents, suggesting that HMGN2 increased the sensitivity to chemotherapy.

Antitumor effect of lenti-HMGN2 in the osteosarcoma xenograft model

The in vitro experiments confirmed that HMGN2 efficiently inhibited the growth and migration of U2-OS and SaO2 cells. However, whether HMGN2 has the same inhibitory effect on in vivo osteosarcoma remains to be determined. We investigated the antitumor effect of HMGN2 in vivo using SaO2 and U2-OS xenograft models. The mean volumes of SaO2 and U2-OS xenograft tumors were 52.3±10.5 and 35.5±8.6 mm3 in the experimental mice prior to treatment. On day 14, the average volumes of SaO2 and U2-OS xenograft tumors were significantly smaller in the HMGN2 group than those in the CON and GFP groups (Fig. 5A). During the whole tumor growth period, the tumor growth activity was measured. Tumors treated with HMGN2 lentivirus grew substantially slower than the GFP group (Fig. 5B). When the tumors were harvested, the average weights of SaO2 and U-2 OS xenograft tumors in HMGN2 group were significantly lighter than those in the GFP group (**P<0.01; Fig. 5C). These results in vivo indicated that overexpression of HMGN2 was able to inhibit SaO2 and U-2 OS cell growth.

Discussion

HMGNs were initially regarded as transcription coregulators, however, their roles in DNA repair and cancer progression were determined using HMGN1 knockout mice (14). In addition to HMGN1, the expression of HMGN5 (formerly NSBP1) was found to be one of the significant factors in the prognosis of bone tumor (15), breast cancer (16) and prostate cancer (17). Collectively, results of those studies suggested that HMGNs were involved in osetosarcoma cell progression and exhibited characteristics of a tumor-suppressor gene. However, whether HMGN2 was expressed in osteosarcoma and the role of HMGN2 was not previously reported. The present study investigated the activity of HMGN2 in the MG-63, HOS, SaO2 and U2-OS osteosarcoma cell lines. To the best of our knowledge, the present study documented for the first time the endogenous expression of HMGN2 in osteosarcoma cell lines, and demonstrated that the expression levels of HMGN2 were significantly lower in SaO2 and U2-OS cell lines than those in MG-63 and HOS cell lines, which were selected as the infective objects of HMGN2 lentivirus. In pilot studies, the infectious efficiency of HMGN2 in SaO2 and U2-OS cell lines was extremely high, and a marked increase of HMGN2 expression was observed in the HMGN2 group compared with the GFP and CON groups. HMGN2 is mainly expressed in vertebrates and invertebrates (6,7) and functions as a modifier of the nucleosomal organization. In addition to the above function, the findings in our study demonstrated that overexpression of HMGN2 significantly reduced the proliferative activities of SaO2 and U2-OS cell lines in a time dependent manner. As shown in our findings, the possible underlying mechanism is that HMGN2, via downregulation of PCNA and cyclin D1 expression, inhibits osteosarcoma cell proliferation.

More importantly, HMGN2 has been confirmed to prevent migration and invasiveness and induce apoptosis, which suggesting that HMGN2 is a tumor suppressor and apoptosis regulator in osteosarcoma. However, the response to HMGN2 is not concordant among all types of cancer. The biological response of cancer cells to HMGN2 may depend, not only on the particular cell type, but also on the presence of other factors that remain to be defined. Our gain-of-function studies in vitro and in vivo using HMGN2 lentivirus revealed a significant decrease in growth and migration and an increase in apoptosis in SaO2 and U2-OS cells, suggesting that HMGN2 may function as a tumor suppressor in osteosarcoma.

To the best of our knowledge, this is the first study to provide data demonstrating that HMGN2 has an inhibitory effect on growth and migration of osteosarcoma cells. However, limited evidence was obtained regarding the use of osteosarcoma cells in the two cell lines. Investigations using more cell lines and primary tumor are therefore crucial to confirm the findings of this study. In conclusion, the present results have shown that the enhanced expression of HMGN2 in osteosarcoma cells by HMGN2 lentivirus exerts inhibitory effects on growth and migration of osteosarcoma cells. HMGN2 as a tumor suppressor may provide a novel approach to human osteosarcoma treatment.

References

1 

Huang J, Ni J, Liu K, et al: HMGB1 promotes drug resistance in osteosarcoma. Cancer Res. 72:230–238. 2012. View Article : Google Scholar

2 

Clark JC, Dass CR and Choong PF: A review of clinical and molecular prognostic factors in osteosarcoma. J Cancer Res Clin Oncol. 134:281–297. 2008. View Article : Google Scholar

3 

Meyers PA, Schwartz CL, Krailo MD, et al: Osteosarcoma: the addition of muramyl tripeptide to chemotherapy improves overall survival - a report from the Children’s Oncology Group. J Clin Oncol. 26:633–638. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Lau CC, Harris CP, Lu XY, et al: Frequent amplification and rearrangement of chromosomal bands 6p12-p21 and 17p11.2 in osteosarcoma. Genes Chromosomes Cancer. 39:11–21. 2004. View Article : Google Scholar

5 

Musselman CA and Kutateladze TG: Methyl fingerprinting of the nucleosome reveals the molecular mechanism of high-mobility group nucleosomal-2 (HMGN2) association. Proc Natl Acad Sci USA. 108:12189–12190. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Bustin M: Regulation of DNA-dependent activities by the functional motifs of the high-mobility-group chromosomal proteins. Mol Cell Biol. 19:5237–5246. 1999.PubMed/NCBI

7 

Postnikov YV, Herrera JE, Hock R, Scheer U and Bustin M: Clusters of nucleosomes containing chromosomal protein HMG-17 in chromatin. J Mol Biol. 274:454–465. 1997. View Article : Google Scholar

8 

Porkka K, Laakkonen P, Hoffman JA, Bernasconi M and Ruoslahti E: A fragment of the HMGN2 protein homes to the nuclei of tumor cells and tumor endothelial cells in vivo. Proc Natl Acad Sci USA. 99:7444–7449. 2002. View Article : Google Scholar : PubMed/NCBI

9 

Srikantha T, Landsman D and Bustin M: Retropseudogenes for human chromosomal protein HMG-17. J Mol Biol. 197:405–413. 1987. View Article : Google Scholar : PubMed/NCBI

10 

Spieker N, Beitsma M, van Sluis P, et al: An integrated 5-Mb physical, genetic, and radiation hybrid map of a 1p36.1 region implicated in neuroblastoma pathogenesis. Genes Chromosomes Cancer. 27:143–152. 2000. View Article : Google Scholar

11 

Hu A, Dong X, Liu X, et al: Nucleosome-binding protein HMGN2 exhibits antitumor activity in oral squamous cell carcinoma. Oncol Lett. 7:115–120. 2014.

12 

Liu X, Dong X, Zhang Y, et al: Inhibitory effects of high mobility group chromosomal protein N2 on human tongue carcinoma transplanted in nude mice. Hua Xi Kou Qiang Yi Xue Za Zhi. 32:5–8. 2014.(In Chinese). PubMed/NCBI

13 

Evans DL, Bishop GR and Jaso-Friedmann L: Methods for cell cycle analysis and detection of apoptosis of teleost cells. Methods Cell Sci. 22:225–231. 2000. View Article : Google Scholar

14 

Birger Y, Catez F, Furusawa T, et al: Increased tumorigenicity and sensitivity to ionizing radiation upon loss of chromosomal protein HMGN1. Cancer Res. 65:6711–6718. 2005. View Article : Google Scholar : PubMed/NCBI

15 

Zhou X, Yuan B, Yuan W, Wang C, Gao R and Wang J: The expression and clinical significance of high mobility group nucleosome binding domain 5 in human osteosarcoma. Tumour Biol. 35:6539–6547. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Li DQ, Hou YF, Wu J, et al: Gene expression profile analysis of an isogenic tumour metastasis model reveals a functional role for oncogene AF1Q in breast cancer metastasis. Eur J Cancer. 42:3274–3286. 2006. View Article : Google Scholar : PubMed/NCBI

17 

Huang C, Zhou LQ and Song G: Effect of nucleosomal binding protein 1 in androgen-independent prostatic carcinoma. Zhonghua Yi Xue Za Zhi. 88:657–660. 2008.(In Chinese). PubMed/NCBI

Related Articles

Journal Cover

March-2015
Volume 33 Issue 3

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
Liang G, Xu E, Yang C, Zhang C, Sheng X and Zhou X: Nucleosome-binding protein HMGN2 exhibits antitumor activity in human SaO2 and U2-OS osteosarcoma cell lines. Oncol Rep 33: 1300-1306, 2015
APA
Liang, G., Xu, E., Yang, C., Zhang, C., Sheng, X., & Zhou, X. (2015). Nucleosome-binding protein HMGN2 exhibits antitumor activity in human SaO2 and U2-OS osteosarcoma cell lines. Oncology Reports, 33, 1300-1306. https://doi.org/10.3892/or.2014.3689
MLA
Liang, G., Xu, E., Yang, C., Zhang, C., Sheng, X., Zhou, X."Nucleosome-binding protein HMGN2 exhibits antitumor activity in human SaO2 and U2-OS osteosarcoma cell lines". Oncology Reports 33.3 (2015): 1300-1306.
Chicago
Liang, G., Xu, E., Yang, C., Zhang, C., Sheng, X., Zhou, X."Nucleosome-binding protein HMGN2 exhibits antitumor activity in human SaO2 and U2-OS osteosarcoma cell lines". Oncology Reports 33, no. 3 (2015): 1300-1306. https://doi.org/10.3892/or.2014.3689