Downregulation of IDH2 exacerbates the malignant progression of osteosarcoma cells via increased NF-κB and MMP-9 activation

  • Authors:
    • Wan-Rong Yi
    • Zong-Huan Li
    • Bai-Wen Qi
    • Mendame Ehya Regis Ernest
    • Xiang Hu
    • Ai-Xi Yu
  • View Affiliations

  • Published online on: January 13, 2016     https://doi.org/10.3892/or.2016.4553
  • Pages: 2277-2285
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Isocitrate dehydrogenase 2 (IDH2) is a mitochondrial NADP-dependent isocitrate dehydrogenase. It is considered to be a novel tumor suppressor in several types of tumors. However, the role and related mechanism of IDH2 in osteosarcoma remain unknown. The expression and significance of IDH2 were investigated by immunohistochemistry in formalin-fixed paraffin sections from 44 osteosarcoma patients. IDH2 was downregulated via lentiviral vector‑mediated RNA interference (RNAi) in the Saos-2 and MG-63 human osteosarcoma cell lines. The effect of IDH2 downregulation on human osteosarcoma was studied in vitro by MTT, flow cytometry and invasion assays. Nuclear factor-κB (NF-κB) and matrix metalloproteinase-9 (MMP-9) assays were also used to study the likely molecular mechanism of IDH2 downregulation on the malignant progression of osteosarcoma cells. The results revealed that the expression of IDH2 was inversely correlated with pathological grade and metastasis in osteosarcoma. IDH2 downregulation promoted a pro-proliferative effect on the Saos-2 and MG-63 osteosarcoma cell lines. IDH2 downregulation accelerated cell cycle progression from S to G2/M phase. The pro-proliferative effect induced by IDH2 downregulation may be ascribed to increased NF-κB activity via IκBα phosphorylation. The invasive activity of osteosarcoma cells was also significantly promoted by IDH2 downregulation and may result from elevated MMP-9 activity. In conclusion, IDH2 downregulation may exacerbate malignant progression via increased NF-κB and MMP-9 activity and may implicate the potential biological importance of IDH2 targeting in osteosarcoma cells. Downregulation of IDH2 exacerbates the malignant progression of osteosarcoma cells via increased NF-κB and MMP-9 activation.

Introduction

Osteosarcoma is the most prevalent primary malignant bone tumor in children and young adults, and is characterized by aggressive invasion, early metastasis and resistance to existing chemotherapeutics (1). In recent years, the survival rate of osteosarcoma patients has improved due to advances in aggressive systemic chemotherapy. However, the survival rate of osteosarcoma patients is still low for those with primary metastases and relapse compared to patients with localized disease (2,3). Moreover, multidrug combination chemotherapy for osteosarcoma leads to ototoxicity, cardiac toxicity and secondary malignancies (2). Thus, it is vital to identify novel approaches for both diagnosis and treatment that are more efficient than the currently available methods to resolve these problems and improve the prognosis of osteosarcoma (4). An understanding of the molecular events that drive the progression and metastasis of osteosarcoma would facilitate better diagnosis and treatment strategies.

Recently, NADP+-dependent isocitrate dehydrogenases (IDHs), including IDH1 and IDH2, were found to be down-regulated in glioma, melanoma and bladder cancer (58). By providing NADPH, IDHs play an important role in controlling the mitochondrial redox balance and mitigating cellular oxidative damage (9,10). In our previous study, IDH1 was shown to be a tumor-suppressor gene in osteosarcoma and inhibited the malignant progression of osteosarcoma (11,12). However, the role of IDH2 in osteosarcoma remains unknown.

IDH2, a mitochondrial NADP-dependent enzyme, catalyzes oxidative decarboxylation and produces CO2, NADPH and α-ketoglutarate from isocitrate in the mitochondria (13). NADPH is a vital cofactor for many enzymatic reactions, including fat and cholesterol biosynthesis and glutathione metabolism (14). It was demonstrated that mitochondrial NADP+-dependent isocitrate dehydrogenase plays an important role in cellular defense against oxidative damage by providing NADPH, which is needed to regenerate the glutathione levels in mitochondria (1517). Reactive oxygen species (ROS), which are produced in mitochondria as a natural by-product of normal energy metabolism, are involved in over 150 human disorders (18). Increased ROS levels promote cellular oxidative stress that contributes to various processes in malignant tumors, including carcinogenesis, aberrant growth, angiogenesis and metastasis (19,20).

Tumor cell growth and invasion are important in malignant progression and are regulated by many biological regulators. One such regulator is NF-κB that regulates the expression of various genes involved in immunity, stress responses, inflammation and inhibition of apoptosis, thus providing appropriate conditions for tumor cell progression (21,22). NF-κB is constitutively activated in tumor cells, including osteosarcoma cells, and contributes to maintain the highly proliferative malignant phenotype as well as cellular invasion (23,24). NF-κB regulates several metastasis-related matrix metalloproteinases (MMPs), such as MMP1, MMP3 and MMP-9 (2529). MMP-9 is recognized as a classic invasion- or metastasis-related NF-κB target gene and is described as an important biomarker that is directly associated with the metastatic processes in osteosarcoma (11,30,31).

In the present study, we first investigated the expression and significance of IDH2 in osteosarcoma biopsies in vivo. Next, we studied the role of IDH2 downregulation in vitro in the Saos-2 and MG-63 human osteosarcoma cell lines. Furthermore, we studied the related biological mechanisms that were induced by IDH2 downregulation in osteosarcoma cells.

Materials and methods

Tissue specimens and clinical data

Fifty-one formalin-fixed, paraffin-embedded osteosarcoma specimens (before the administration of neo-adjuvant chemotherapy) were collected according to the Chinese National Ethical Guidelines ('Code for Proper Secondary Use of Human Tissue', Chinese Federation of Medical Scientific Societies). Due to the limited available tumor material and follow-up information, only 44 of these osteosarcoma tumor samples were eligible for the present study. The mean age of the patients (mean ± SD) was 25.25±13.61 years (range 9–61; male/female ratio 32:12). Of the 44 eligible patients, 23 had non-metastatic tumors (53.3%). Osteoblastic osteosarcoma was the most common histopathological subtype and occurred in 29 patients (65.9%). The lower end of the femur was the most common site and was observed in 13 patients (29.5%). The patient distribution according to Rosen's histological grade (19,20) included stage I in 5 (11.3%), II in 16 (36.4%), III in 16 (36.4%) and stage IV in 7 patients (15.9%). Stages I and II were defined as low Rosen grade osteosarcoma specimens and stages III and IV were defined as high grade specimens. The patients were followed-up until death from the disease, or until their most recent clinical therapy at the end of the present study. The mean follow-up time (mean ± SD) was 4.26±1.99 years (range 0.5–9.0). All patients were diagnosed according to the osteosarcoma criteria defined by the World Health Organization. Written informed consent was obtained from each patient before he/she entered into the present study, and all study protocols were approved by the Ethics Committee for Clinical Research of Wuhan University, China.

Immunohistochemistry and specimen evaluation

The sections were cut from the formalin-fixed, paraffin-embedded osteosarcoma tissue and hydrated through graded alcohol solutions. For antigen unmasking, the sections were treated in a trypsin solution at 37°C for 10 min. The sections were then washed with deionized water and incubated with 3% H2O2 for 5 min. They were then incubated with the anti-IDH2 mAb at room temperature for 1 h, followed by a secondary antibody and the peroxidase-conjugated streptavidin-biotin complex (both from Santa Cruz Biotechnology, Santa Cruz, CA, USA) at 37°C for 30 min. The immunoreactivity was visualized with 3,3′-diaminobenzidine (DAB) (Zymed, South San Francisco, CA, USA). The negative controls were obtained by omitting the primary antibody.

IDH2 staining was detected in the mitochondria and was scored by adding the number of cells displaying clear tumor cell labeling; the intensity of staining was scored between 0 and 6 (32,33). The proportion score was as follows: 0 indicates negative staining; +1 indicates ≤25% positive labeling in tumor cells; +2 indicates 25–50% positive tumor cells; and +3 indicates >50% positive tumor cells. The intensity score was as follows: 0 indicates no staining; +1 indicates weak staining; +2 indicates intermediate staining; and +3 indicates strong staining (32,33). For statistical analysis, the osteosarcoma patients were grouped as either the low expression group (scored 1–4) or the high expression group (scored 5–6). At least 5 separated neoplastic infiltration foci were analyzed in each specimen, followed by the evaluation of 10 slides/patient and 6 sections/slide. The immunostaining was assessed by three independent observers. The slides were scanned using a microscope (Carl Zeiss AG, Germany) by reviewing the entire spot and the images were recorded using a digital camera (DC500; Leica) and Leica FW4000 software. The images were processed using Adobe Photoshop.

Cell lines, culture and lentiviral infection

The Saos-2 and MG63 tumor cells (ATCC, LGC Promochem, Germany) were grown in Roswell Park Memorial Institute (RPMI)-1640 medium (Sigma-Aldrich, USA) with 10% fetal bovine serum (FBS) (Amresco, USA) and 0.1% penicillin/streptomycin and maintained in an atmosphere with 5% CO2 at 37°C.

To downregulate IDH2, small interfering RNA (siRNA) sequences: LV-KD, 5′-GTGGACATCCAGCTAAAGTAT-3′, were inserted into the pLL3.7 shRNA lentiviral vector (Genesil, Wuhan, China). The pLL3.7 lentiviral vector LV-mIκB (Genesil), which was aimed at overexpressing the mutant IκB (mIκB), was constructed as previously described to suppress NF-κB activity (13). siRNA sequences for MMP-9 downregulation were: 5′-ACCACAACAUCACCUAUUGTT-3′ (34). The empty lentiviral vector (Genesil), LV-EV was used as a control. In some experiments, non-treated cells, named the NT cells, were used as another control. The lentiviral stocks were added to the Saos-2 and MG63 osteosarcoma cell lines.

The cells were infected with the lentivirus and selected with an 800 μg/ml G418 solution for the Saos-2 cells and a 500 μg/ml solution for the MG63 cells. The efficiency of the highest infection, as determined by G418 selection, was obtained at a multiplicity of infection (MOI) of 10 for the Saos-2 cells and 50 for the MG63 cells. The cells transfected with LV-KD or LV-EV were named the KD or EV cells, respectively. After selection, the efficiency of infection was verified by western blotting. Polyclonal populations and clones displaying higher levels of IDH2 downregulation were chosen for the subsequent studies. After IDH2 downregulation, LV-mIκB or LV-siMMP-9 was transfected into the cells for the NF-κB and MMP-9 assays, respectively.

Protein isolation and western blot analysis

The cell lysates were prepared using lysis buffer from the Dual-Luciferase assay kit (Promega, Madison, WI, USA) according to the manufacturer's instructions. The lysates were separated on 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) gels and transferred onto polyvinylidene difluoride (PVDF) membranes (Millipore, Billerica, MA, USA). The membranes were then blocked with 5% skim milk in Tris-buffered saline with 0.05% Tween (TBST) and washed 6 times with TBST. The IDH2 and MMP-9 proteins were detected using rabbit polyclonal primary antibodies (Protein Technology Group, USA). NF-κB, Bcl-2, JNK, p-JNK, ERK, p-ERK, IκBα and p-IκBα were detected by mouse monoclonal primary antibodies (Santa Cruz Biotechnology). The β-actin proteins were recognized by a monoclonal β-actin-specific mouse IgG (Santa Cruz Biotechnology) and used as the internal loading control. The antibodies were diluted according to the manufacturer's instructions and incubated overnight at 4°C, followed by incubations with the peroxidase-conjugated goat anti-rabbit or anti-mouse immunoglobulins (1:2,000; Santa Cruz Biotechnology) in TBST for 1 h. The signals were developed using an enhanced chemiluminescent reagent (Pierce Biotechnology, Rockford, IL, USA).

MTT assay

A total of 3.5×103 cells were seeded in each test well in a 96-well plate to detect cell growth. After 1–6 days of culture, the cells were washed with phosphate-buffered saline (PBS). MTT (5 mg/ml) was then added to each well (including the control) and the mixture was incubated at 37°C for 4 h. The culture medium was then replaced with an equal volume of dimethylsulfoxide (DMSO). After shaking the plate at room temperature for 10 min, the absorbance of each well was determined at 570 nm using a VersaMax microplate reader (Molecular Devices, Sunnyvale, CA, USA).

Cell cycle analysis with flow cytometry

The cells were harvested by trypsinization, fixed with 70% pre-chilled alcohol and stored at 4°C overnight. The alcohol was then removed by centrifugation at 1,000 rpm for 5 min, and the cells were treated with 0.1% Triton-X and DNase-free RNase (10 mg/ml) for 30 min (35). The cell DNA was stained with 1 mg/ml propidium iodide (PI) for 15 min in the dark and analyzed using a flow cytometer (FACScan; Becton-Dickinson, New York, NY, USA). The relative proportions of cells in the G0–G1, S and G2-M phases of the cell cycle were determined from the flow cytometry data.

Cell invasion assay

Cell invasion was determined using a two-chamber Transwell (Corning, New York, NY, USA). The upper surface of a polycarbonate membrane with 8-μm pores was coated with 1 mg/ml Matrigel (36). The cells (~10×105) were suspended in RPMI-1640 serum-free medium (Gibco, USA) and placed in the upper chamber; RPMI-1640 medium containing 10% FBS was placed in the lower chamber. The cells were incubated at 37°C for 48 h with 5% CO2 in an incubator. At the end of the incubation, the cells on the upper surface of the membrane were completely removed by wiping with a cotton swab. Then, the membrane was fixed with methanol and stained with 0.1% crystal violet. The cells that invaded the Matrigel and reached the lower surface of the membrane were counted and photographed under a microscope.

NF-κB activity assay

An NF-κB reporter plasmid, NF-κB-Luc, was constructed by cloning five repeats of the NF-κB regulatory elements into the pLuc plasmid (Stratagene, La Jolla, CA, USA) to drive luciferase expression. The empty plasmid, Luc, containing a minimal TATA box, was used as a negative control. Approximately 2.0×106 cells were transiently transfected with 10 μg of the NF-κB reporter plasmid by electroporation (37). Then, the cells were seeded and incubated in 24-well plates at 37°C. After 24 h, the luciferase activity was analyzed using the Luciferase assay system (Promega) according to the manufacturer's instructions.

Intracellular ROS assay

To obtain dissociated Saos2 and MG63 cells for the ROS assay, the culture medium was first removed and the cells were washed two times with RPMI-1640 serum-free medium (Sigma-Aldrich). DCFH-DA, diluted to a final concentration of 10 μM with RPMI-1640 medium, was added to the medium and incubated for 20 min at 37°C. The fluorescence was read at 488 nm for excitation and 530 nm for emission by flow cytometry (FACScan). The increase in the value compared to the control was viewed as the increase in intracellular ROS levels.

MMP-9 activity assay

The IDH2 and/or MMP-9 down-regulated osteosarcoma cells were seeded in 6-well plates and incubated at 37°C. After 24 h, the medium was removed. Then, the cells were washed and incubated in serum-free medium for 48 h (37). The MMP-9 activity in the medium and cell lysate was detected using the Fluorokine E Human MMP-9 Activity assay kit (R&D Systems) according to the manufacturer's protocol.

Statistical analysis

The statistical analyses were performed using the SPSS 13.0 software package for Windows (SPSS, Inc., Chicago, IL, USA). Comparisons between groups were analyzed by the t-test or Mann-Whitney U test. Associations were assessed by Pearson's or Spearman's correlation coefficients. Event-free survival was calculated from the start of treatment to relapse or metastasis. Overall survival was calculated from the beginning of treatment to the last follow-up or death of the patient. The Kaplan-Meier method was used for the survival analysis. P<0.05 was considered to indicate a statistically significant result. P<0.01 was considered to indicate a highly statistically significant result.

Results

In vivo tissues
IDH2 correlates with Rosen's histological grade and metastasis in osteosarcoma

IDH2 was mainly found in the mitochondria (Fig. 1A). Of the 44 osteosarcoma specimens, 41 cases were IDH2-positive using immunohistochemistry (93.2%) and 19 exhibited high levels of staining (43.1%). The average IDH2 immunostaining score was 3.15 (SD, 1.46; range from 0 to 6). Lower IDH2 expression was observed in high Rosen's histological grade (38,39) osteosarcoma compared to the low grade specimens (P=0.005; r=−0.505) (Fig. 1A). IDH2 expression was inversely correlated with metastasis (P=0.026; r=−0.334). There was no significant correlation between IDH2 expression and overall survival (P=0.063) or event-free (relapse and metastasis included) survival (P=0.074) (Fig. 1B). IDH2 expression did not correlate with other clinical features, such as age, location of the primary tumor and histological type (P>0.05).

In vitro cell lines
IDH2 downregulation promotes cell proliferation

IDH2 downregulation was verified by western blotting. The IDH2 protein was significantly decreased in the Saos-2 KD cells compared to the EV or NT cells (P<0.01) (Fig. 2A), respectively; similar results were obtained in the MG63 cells (Fig. 2A). There was no difference in IDH2 expression between the NT and EV cells, either in the Saos-2 or MG63 cells (P>0.05) (Fig. 2A).

IDH2 downregulation increased the cell growth rate in the Saos-2 KD cells by 1.7-fold and in the MG63 KD cells by 1.5-fold on day 6 compared to the Saos-2 EV or MG-63 EV cells (P<0.01) (Fig. 2B). IDH2 downregulation promoted the growth of osteosarcoma cells.

IDH2 downregulation decreases the distribution of cells in the S phase and increases the distribution in the G2/M phase

The DNA of the cell populations was analyzed after stable transfection of IDH2 siRNAs into the Saos-2 and MG63 cells. IDH2 downregulation induced an increase in the G2/M population in the Saos-2 and MG-63 cell lines by 180.4±3.5 and 60.3±2.2% (P<0.05), respectively, whereas the S phase population was reduced by 53.2±5.8 and 69.6±2.7% (P<0.05), respectively, compared to the empty vector control (Fig. 2C and D). The population in the G0/G1 phase was not significantly changed in the present study (P>0.05) (Fig. 2C and D). The EV cells did not show significant changes in the cell cycle distribution in the Saos-2 and MG63 cells compared to the NT cells (P>0.05) (Fig. 2C and D). IDH2 downregulation in the osteosarcoma cells induced cell cycle progression from the S to the G2/M phase.

IDH2 downregulation exacerbates cell invasion

Next, the effect of IDH2 on cell invasion was investigated. As shown in Fig. 3A and B, IDH2 downregulation promoted the cell invasive activity of the Saos-2 KD cells by 2.8-fold and the MG63 KD cells by 2.2-fold compared to the Saos-2 EV or MG-63 EV cells (P<0.01). The EV cells did not show significant changes in invasion activity in the Saos-2 and MG63 cells compared to the NT cells (P>0.05) (Fig. 3A and B). IDH2 downregulation promoted the invasion of osteosarcoma cells.

IDH2 downregulation does not change the intracellular ROS levels

The intracellular ROS levels in the Saos-2 and MG63 cell lines were also investigated. IDH2 downregulation did not induce a significant difference in the ROS levels in the Saos-2 KD or MG63 KD cells (P>0.05), respectively, compared to the EV and NT cells (Fig. 3C and D).

IDH2 downregulation increases NF-κB activation and IkBα phosphorylation

In both the Saos-2 and MG63 cells, the degradation of inactivated NF-κB was increased by IDH2 downregulation compared to the EV cells (Fig. 4A). However, there was no significant difference in the protein expression of other regulators, such as Bcl-2, JNK and ERK (Fig. 4A). NF-κB transcriptional activity was analyzed by comparing the luciferase activity of the pNF-κB-Luc transfected EV cells to the basal activity. We investigated the overexpression of mIκB (a constitutive NF-κB inhibitor) on the cells with IDH2 downregulation. NF-κB activity was promoted in the Saos-2 and MG63 KD cells with stable IDH2 downregulation compared to the respective EV and/or NT cells (P<0.01). The increased NF-κB activity induced by IDH2 downregulation was inhibited by mIκB overexpression (P<0.01) (Fig. 4B). In addition, Fig. 4C shows that IDH2 downregulation increased the expression of p-IκBα (P<0.01). The significant p-IκBα upregulation induced by IDH2 downregulation was inhibited by mIκB overexpression (Fig. 4C).

Figure 4

IDH2 downregulation elevates NF-κB transcriptional activity and IκB phosphorylation. (A) In both the Saos-2 and MG63 cells, the degradation of inactivated NF-κB was promoted by IDH2 downregulation compared to the EV cells (P<0.05). However, there was no significant difference in other proteins, such as Bcl-2, phosphorylated and total ERK, and phosphorylated and total JNK (P>0.05). A representative western blot result is shown. (B) Increased NF-κB transcriptional activity was detected in the siIDH2-transfected Saos-2 or MG63 cells (Saos-2 pNF-κB-Luc siIDH2 or MG63 pNF-κB-Luc siIDH2 cells) (P<0.05) compared to the EV cells (pNF-κB-Luc EV cells) using the NF-κB-Luc reporter. The NF-κB activity was significantly reduced by mIκB overexpression (pNF-κB-Luc mIκB cells). When the siIDH2-transfected Saos-2 and MG63 cells were transfected with mIκB (pNF-κB-Luc siIDH2 + mIκB cells), the NF-κB activity was markedly decreased (n=3; P<0.01). Transfection of the empty luc reporter in the NT cells (p-Luc NT cells) and EV cells (p-Luc EV cells) showed minimal background luciferase activity. The NT cells transfected with the NF-κB-Luc reporter (pNF-κB-Luc NT cells) did not show a significant change in luciferase activity; *P<0.05 and **P<0.01. Columns, means; bars, SE (standard deviation). (C) The Saos-2 and MG63 cells transfected with siIDH2 (siIDH2 cells) exhibited suppression of the expression of the NF-κB inhibitor IκBα and promoted the activation of a subset of p-IκBα compared to the EV cells (P<0.05). mIκB suppressed the IDH2 downregulation-induced IκBα phosphorylation (P<0.05). Representative results are shown.

IDH2 downregulation elevates MMP-9 activation

Western blot analysis and an MMP-9 activity assay were conducted. We found that the MMP-9 protein levels were markedly increased in the IDH2 downregulated Saos-2 or MG63 cells (Fig. 5A and B). In addition, there was a 3.5- and 2.7-fold increase in the MMP-9 activity (Fig. 5A and B) in these cells, respectively. Furthermore, the increased protein expression and activity of MMP-9 induced by IDH2 downregulation were significantly inhibited by MMP-9 downregulation (Fig. 5A and B).

Discussion

To date, the alteration of IDH2 expression levels has been identified in several types of carcinoma (5,4144). In melanoma, IDH2 was found to be frequently downregulated, and an increase in tumor-free survival resulted from the overexpression of IDH2 (9). IDH2 was also reported to be downregulated in early phase colon carcinoma compared to peritumoral tissues (45). However, the expression and significance of IDH2 in osteosarcoma remain unknown. In our study, it was observed that IDH2 was expressed at a lower level in high-grade osteosarcoma, and 3 of the 44 osteosarcoma patients did not express IDH2. There was a similar trend toward increased metastasis in patients with low IDH2 expression, although no significant correlation was observed between IDH2 expression and survival. Our previous study indicated that patients with low IDH1 expression tended to have higher pathological grade tumors with increased metastatic potential (12). A higher 5-year survival rate was also found in the IDH1 high expression group vs. the IDH1 low expression group, although there was no significant correlation between IDH1 expression and overall survival (12). The significant similarity of IDH1 and IDH2 in osteosarcoma suggests that IDH1 and IDH2 could both be potential biomarkers for assessing malignant progression and predicting the risk of metastasis in osteosarcoma.

Next, cell proliferation and metastasis studies were performed to explore the biological significant of IDH2 downregulation in osteosarcoma. In our study, IDH2 downregulation increased cell growth in osteosarcoma. Furthermore, it decreased the proportion of cells in the S phase and increased the proportion in the G2/M phase, suggesting a pro-progression effect. We also found that IDH2 downregulation exacerbated cell invasion in the Saos-2 and MG63 osteosarcoma cell lines. In colon cancer, IDH2 downregulation has been reported to inhibit the growth of colon carcinoma cells (45). Kim et al demonstrated that the reduction in IDH2 levels in malignant melanoma cells has antitumorigenic effects (13). However, our previous study showed that IDH1 upregulation inhibited cell proliferation in the 143B and MG63 osteosarcoma cell lines, whereas IDH1 downregulation exacerbated cell proliferation (12). In the present study, we found a similar biological significance of IDH1 and IDH2 downregulation in osteosarcoma cells. Moreover, this suggested that, similar to IDH1, IDH2 downregulation exacerbated cell proliferation and metastasis in osteosarcoma cells.

The levels of reactive oxygen species (ROS) were analyzed in the osteosarcoma cell lines to explore the mechanism of IDH2 downregulation in cell proliferation and metastasis. Mitochondrial NADP+-dependent isocitrate dehydrogenase was reported to have an important role in cellular defense against oxidative damage by supplying NADPH, which is needed to produce glutathione (1517). Cellular oxidative stress arising from high levels of ROS contributes to the development and progression of malignant tumors, including carcinogenesis, aberrant growth, angiogenesis and metastasis (19). Thus, mitochondrial NADP+-dependent isocitrate dehydrogenase is fundamentally important for the defense against ROS, which was detected in our study. The result showed that there was no significant difference in the intracellular ROS levels between the downregulated osteosarcoma cells and non-treated cells. ROS, which were involved in the effects of IDH2 downregulation in melanoma cells, were significantly elevated (19). In addition, this surprising difference may suggest different mechanisms of IDH2 in different tumor cells.

NF-κB is constitutively activated and is implicated in cellular proliferation and invasion in osteosarcoma cells (21,23). NF-κB activation occurs via phosphorylation of IκBα and IκBβ and IκBα phosphorylation is essential for the release of active NF-κB (46). Therefore, we examined whether and how IDH2 affected NF-κB activity. Notably, IDH2 downregulation increased NF-κB degradation in osteosarcoma cells, suggesting that NF-κB may be involved in the IDH2 downregulation-induced pro-proliferation effect. IDH2 downregulation increased NF-κB activity as well as IκBα phosphorylation in the Saos-2 and MG63 cells compared to the control cells. This result is supported by the finding that increased NF-κB activity and IκBα phosphorylation can be inhibited by overexpression of mIκB (a constitutive NF-κB inhibitor). IDH2 downregulation increased the activity of NF-κB, which was likely due to IκBα phosphorylation, and, therefore, contributed to the IDH2 downregulation-induced pro-growth function in the osteosarcoma cell lines.

In our study, we also examined the effects of IDH2 downregulation on the invasion of osteosarcoma cells. We found that IDH2 downregulation promoted the invasion of osteosarcoma cells into Matrigel. MMPs are involved in the processes of tumor cell invasion and metastasis (25) and are directly associated with the metastatic processes in osteosarcoma (30,31). We found, for the first time, that IDH2 downregulation increased MMP-9 expression at the protein level and also activated MMP-9. This is supported by the finding that increased MMP-9 expression and activation can be inhibited by siMMP-9. These findings suggest that the potential pro-metastatic activities following IDH2 downregulation could be partially interpreted by the elevated MMP-9 activity. In addition, NF-κB activity was increased following IDH2 downregulation in our study. Based on our results, IDH2 downregulation likely promoted cell invasion, at least in part, through increased activation of NF-κB and its target genes: MMPs.

The limitation of the present study is that it is a retrospective study with limited samples. In addition, the lower IDH2 expression in the higher grade osteosarcoma samples may not indicate a mutation in this gene. Furthermore, IDH2 siRNA was used only in a small number of cell lines. Further studies are needed to confirm the precise molecular regulation of IDH2 and NF-κB and their interaction to elucidate the role of IDH2 in cell growth and invasion in animal models. However, it may still be valuable to study the role of IDH2 in osteosarcoma. In addition, the pro-proliferation and pro-invasion activities as well as the potential effects of IDH2 on cell immortalization and the inflammatory response in osteosarcoma remain to be elucidated in relevant models.

In conclusion, IDH2 downregulation may indicate high pathological grade and metastasis. IDH2 downregulation induced malignant progression via increased NF-κB and MMP-9 activity in osteosarcoma in vitro. IDH2 may be an effective target by which to develop new therapeutic strategies against osteosarcoma.

Acknowledgments

We thank Guorong Yu, Shengxiang Tao, Zhenyu Pan and Weidong Xiao for technical assistance. The present study was supported by Hubei Province's Outstanding Medical Academic Leader Program.

Abbreviations:

IDH2

isocitrate dehydrogenase 2

NF-κB

nuclear factor-κB

MMPs

matrix metalloproteinases

MMP-9

matrix metalloproteinase-9

JNK

Jun N-terminal kinase

ERK

extracellular signal-regulated kinase

MAPKs

mitogen-activated protein kinases

ROS

reactive oxygen species

References

1 

Raymond AK and Jaffe N: Osteosarcoma multidisciplinary approach to the management from the pathologist's perspective. Cancer Treat Res. 152:63–84. 2009. View Article : Google Scholar

2 

Gorlick R, Anderson P, Andrulis I, Arndt C, Beardsley GP, Bernstein M, Bridge J, Cheung NK, Dome JS, Ebb D, et al: Biology of childhood osteogenic sarcoma and potential targets for therapeutic development: Meeting summary. Clin Cancer Res. 9:5442–5453. 2003.PubMed/NCBI

3 

Kager L, Zoubek A, Pötschger U, Kastner U, Flege S, Kempf-Bielack B, Branscheid D, Kotz R, Salzer-Kuntschik M, Winkelmann W, et al: Primary metastatic osteosarcoma: Presentation and outcome of patients treated on neoadjuvant Cooperative Osteosarcoma Study Group protocols. J Clin Oncol. 21:2011–2018. 2003. View Article : Google Scholar : PubMed/NCBI

4 

Bacci G, Longhi A, Versari M, Mercuri M, Briccoli A and Picci P: Prognostic factors for osteosarcoma of the extremity treated with neoadjuvant chemotherapy: 15-Year experience in 789 patients treated at a single institution. Cancer. 106:1154–1161. 2006. View Article : Google Scholar : PubMed/NCBI

5 

Memon AA, Chang JW, Oh BR and Yoo YJ: Identification of differentially expressed proteins during human urinary bladder cancer progression. Cancer Detect Prev. 29:249–255. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Cancer Genome Atlas Research Network; Brat DJ, Verhaak RG, Aldape KD, Yung WK, Salama SR, Cooper LA, Rheinbay E, Miller CR, Vitucci M, Morozova O, et al: Comprehensive, integrative genomic analysis of diffuse lower-grade gliomas. N Engl J Med. 372:2481–2498. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Ikota H, Nobusawa S, Arai H, Kato Y, Ishizawa K, Hirose T and Yokoo H: Evaluation of IDH1 status in diffusely infiltrating gliomas by immunohistochemistry using anti-mutant and wild type IDH1 antibodies. Brain Tumor Pathol. 32:237–244. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Lian CG, Xu Y, Ceol C, Wu F, Larson A, Dresser K, Xu W, Tan L, Hu Y, Zhan Q, et al: Loss of 5-hydroxymethylcytosine is an epigenetic hallmark of melanoma. Cell. 150:1135–1146. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Park SY, Lee SM, Shin SW and Park JW: Inactivation of mitochondrial NADP+-dependent isocitrate dehydrogenase by hypochlorous acid. Free Radic Res. 42:467–473. 2008. View Article : Google Scholar : PubMed/NCBI

10 

Jo SH, Son MK, Koh HJ, Lee SM, Song IH, Kim YO, Lee YS, Jeong KS, Kim WB, Park JW, et al: Control of mitochondrial redox balance and cellular defense against oxidative damage by mitochondrial NADP+-dependent isocitrate dehydrogenase. J Biol Chem. 276:16168–16176. 2001. View Article : Google Scholar : PubMed/NCBI

11 

Hu X, Liu Y, Qin C, Pan Z, Luo J, Yu A and Cheng Z: Up-regulated isocitrate dehydrogenase 1 suppresses proliferation, migration and invasion in osteosarcoma: In vitro and in vivo. Cancer Lett. 346:114–121. 2014. View Article : Google Scholar

12 

Hu X, Yu AX, Qi BW, Fu T, Wu G, Zhou M, Luo J and Xu JH: The expression and significance of IDH1 and p53 in osteosarcoma. J Exp Clin Cancer Res. 29:432010. View Article : Google Scholar : PubMed/NCBI

13 

Kim SH, Yoo YH, Lee JH and Park JW: Mitochondrial NADP+-dependent isocitrate dehydrogenase knockdown inhibits tumorigenicity of melanoma cells. Biochem Biophys Res Commun. 451:246–251. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Koh HJ, Lee SM, Son BG, Lee SH, Ryoo ZY, Chang KT, Park JW, Park DC, Song BJ, Veech RL, et al: Cytosolic NADP+-dependent isocitrate dehydrogenase plays a key role in lipid metabolism. J Biol Chem. 279:39968–39974. 2004. View Article : Google Scholar : PubMed/NCBI

15 

Nakamura H: Thioredoxin and its related molecules: Update 2005. Antioxid Redox Signal. 7:823–828. 2005. View Article : Google Scholar : PubMed/NCBI

16 

Kirsch M and De Groot H: NAD(P)H, a directly operating antioxidant? FASEB J. 15:1569–1574. 2001. View Article : Google Scholar : PubMed/NCBI

17 

Avery AM, Willetts SA and Avery SV: Genetic dissection of the phospholipid hydroperoxidase activity of yeast gpx3 reveals its functional importance. J Biol Chem. 279:46652–46658. 2004. View Article : Google Scholar : PubMed/NCBI

18 

Nishikawa M: Reactive oxygen species in tumor metastasis. Cancer Lett. 266:53–59. 2008. View Article : Google Scholar : PubMed/NCBI

19 

Kim S, Kim SY, Ku HJ, Jeon YH, Lee HW, Lee J, Kwon TK, Park KM and Park JW: Suppression of tumorigenesis in mitochondrial NADP+-dependent isocitrate dehydrogenase knock-out mice. Biochim Biophys Acta. 1842:135–143. 2014. View Article : Google Scholar

20 

Kim ES and Moon A: Ursolic acid inhibits the invasive phenotype of SNU-484 human gastric cancer cells. Oncol Lett. 9:897–902. 2015.PubMed/NCBI

21 

Inoue R, Matsuki NA, Jing G, Kanematsu T, Abe K and Hirata M: The inhibitory effect of alendronate, a nitrogen-containing bisphosphonate on the PI3K-Akt-NFkappaB pathway in osteosarcoma cells. Br J Pharmacol. 146:633–641. 2005. View Article : Google Scholar : PubMed/NCBI

22 

Ghosh S and Karin M: Missing pieces in the NF-kappaB puzzle. Cell. 109(Suppl 1): S81–S96. 2002. View Article : Google Scholar : PubMed/NCBI

23 

Eliseev RA, Schwarz EM, Zuscik MJ, O'Keefe RJ, Drissi H and Rosier RN: Smad7 mediates inhibition of Saos2 osteosarcoma cell differentiation by NFkappaB. Exp Cell Res. 312:40–50. 2006. View Article : Google Scholar

24 

Andela VB, Sheu TJ, Puzas EJ, Schwarz EM, O'Keefe RJ and Rosier RN: Malignant reversion of a human osteosarcoma cell line, Saos-2, by inhibition of NFkappaB. Biochem Biophys Res Commun. 297:237–241. 2002. View Article : Google Scholar : PubMed/NCBI

25 

Han YP, Tuan TL, Wu H, Hughes M and Garner WL: TNF-alpha stimulates activation of pro-MMP2 in human skin through NF-(kappa)B mediated induction of MT1-MMP. J Cell Sci. 114:131–139. 2001.

26 

Sun P, Mu Y and Zhang S: A novel NF-κB/MMP-3 signal pathway involves in the aggressivity of glioma promoted by Bmi-1. Tumour Biol. 35:12721–12727. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Park SL, Kim WJ and Moon SK: p21WAF1 mediates the IL-15-induced migration and invasion of human bladder cancer 5637 cells via the ERK1/2/NF-κB/MMP-9 pathway. Int Immunopharmacol. 22:59–65. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Li Z, Guo Y, Jiang H, Zhang T, Jin C, Young CY and Yuan H: Differential regulation of MMPs by E2F1, Sp1 and NF-kappa B controls the small cell lung cancer invasive phenotype. BMC Cancer. 14:2762014. View Article : Google Scholar : PubMed/NCBI

29 

Poudel B and Kim DK, Ki HH, Kwon YB, Lee YM and Kim DK: Downregulation of ERK signaling impairs U2OS osteosarcoma cell migration in collagen matrix by suppressing MMP9 production. Oncol Lett. 7:215–218. 2014.

30 

Wang IC, Chen YJ, Hughes DE, Ackerson T, Major ML, Kalinichenko VV, Costa RH, Raychaudhuri P, Tyner AL and Lau LF: FoxM1 regulates transcription of JNK1 to promote the G1/S transition and tumor cell invasiveness. J Biol Chem. 283:20770–20778. 2008. View Article : Google Scholar : PubMed/NCBI

31 

Egeblad M and Werb Z: New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer. 2:161–174. 2002. View Article : Google Scholar : PubMed/NCBI

32 

Xu J, Wu S and Shi X: Expression of matrix metalloproteinase regulator, RECK, and its clinical significance in osteosarcoma. J Orthop Res. 28:1621–1625. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Hirotsu M, Setoguchi T, Sasaki H, Matsunoshita Y, Gao H, Nagao H, Kunigou O and Komiya S: Smoothened as a new therapeutic target for human osteosarcoma. Mol Cancer. 9:52010. View Article : Google Scholar : PubMed/NCBI

34 

Luo Y, Liang F and Zhang ZY: PRL1 promotes cell migration and invasion by increasing MMP2 and MMP9 expression through Src and ERK1/2 pathways. Biochemistry. 48:1838–1846. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Wang R, Dong K, Lin F, Wang X, Gao P, Wei SH, Cheng SY and Zhang HZ: Inhibiting proliferation and enhancing chemosensitivity to taxanes in osteosarcoma cells by RNA interference-mediated downregulation of stathmin expression. Mol Med. 13:567–575. 2007. View Article : Google Scholar : PubMed/NCBI

36 

Koppikar P, Lui VW, Man D, Xi S, Chai RL, Nelson E, Tobey AB and Grandis JR: Constitutive activation of signal transducer and activator of transcription 5 contributes to tumor growth, epithelial-mesenchymal transition, and resistance to epidermal growth factor receptor targeting. Clin Cancer Res. 14:7682–7690. 2008. View Article : Google Scholar : PubMed/NCBI

37 

Boye K, Grotterød I, Aasheim HC, Hovig E and Maelandsmo GM: Activation of NF-kappaB by extracellular S100A4: Analysis of signal transduction mechanisms and identification of target genes. Int J Cancer. 123:1301–1310. 2008. View Article : Google Scholar : PubMed/NCBI

38 

Huvos AG, Rosen G and Marcove RC: Primary osteogenic sarcoma: Pathologic aspects in 20 patients after treatment with chemotherapy en bloc resection, and prosthetic bone replacement. Arch Pathol Lab Med. 101:14–18. 1977.PubMed/NCBI

39 

Rosen G, Marcove RC, Caparros B, Nirenberg A, Kosloff C and Huvos AG: Primary osteogenic sarcoma: The rationale for preoperative chemotherapy and delayed surgery. Cancer. 43:2163–2177. 1979. View Article : Google Scholar : PubMed/NCBI

40 

Pessôa IA, Sagica FE, Anselmo NP, Brito JR and de Oliveira EH: IDH1 and IDH2 mutations in different histologic subtypes and WHO grading gliomas in a sample from Northern Brazil. Genet Mol Res. 14:6533–6542. 2015. View Article : Google Scholar : PubMed/NCBI

41 

Waitkus MS, Diplas BH and Yan H: Isocitrate dehydrogenase mutations in gliomas. Neuro Oncol. Jul 16–2015.(Epub ahead of print). pii: nov136. PubMed/NCBI

42 

Chan SM, Thomas D, Corces-Zimmerman MR, Xavy S, Rastogi S, Hong WJ, Zhao F, Medeiros BC, Tyvoll DA and Majeti R: Isocitrate dehydrogenase 1 and 2 mutations induce BCL-2 dependence in acute myeloid leukemia. Nat Med. 21:178–184. 2015. View Article : Google Scholar : PubMed/NCBI

43 

Aref S, Kamel Areida el S, Abdel Aaal MF, Adam OM, El-Ghonemy MS, El-Baiomy MA and Zeid TA: Prevalence and clinical effect of IDH1 and IDH2 mutations among cytogenetically normal acute myeloid leukemia patients. Clin Lymphoma Myeloma Leuk. 15:550–555. 2015. View Article : Google Scholar : PubMed/NCBI

44 

Liu WR, Tian MX, Jin L, Yang LX, Ding ZB, Shen YH, Peng YF, Zhou J, Qiu SJ, Dai Z, et al: High expression of 5-hydroxymethylcytosine and isocitrate dehydrogenase 2 is associated with favorable prognosis after curative resection of hepatocellular carcinoma. J Exp Clin Cancer Res. 33:322014. View Article : Google Scholar : PubMed/NCBI

45 

Lv Q, Xing S, Li Z, Li J, Gong P, Xu X, Chang L, Jin X, Gao F, Li W, et al: Altered expression levels of IDH2 are involved in the development of colon cancer. Exp Ther Med. 4:801–806. 2012.PubMed/NCBI

46 

Simeonidis S, Stauber D, Chen G, Hendrickson WA and Thanos D: Mechanisms by which IkappaB proteins control NF-kappaB activity. Proc Natl Acad Sci USA. 96:49–54. 1999. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2016
Volume 35 Issue 4

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
Yi W, Li Z, Qi B, Ernest ME, Hu X and Yu A: Downregulation of IDH2 exacerbates the malignant progression of osteosarcoma cells via increased NF-κB and MMP-9 activation. Oncol Rep 35: 2277-2285, 2016
APA
Yi, W., Li, Z., Qi, B., Ernest, M.E., Hu, X., & Yu, A. (2016). Downregulation of IDH2 exacerbates the malignant progression of osteosarcoma cells via increased NF-κB and MMP-9 activation. Oncology Reports, 35, 2277-2285. https://doi.org/10.3892/or.2016.4553
MLA
Yi, W., Li, Z., Qi, B., Ernest, M. E., Hu, X., Yu, A."Downregulation of IDH2 exacerbates the malignant progression of osteosarcoma cells via increased NF-κB and MMP-9 activation". Oncology Reports 35.4 (2016): 2277-2285.
Chicago
Yi, W., Li, Z., Qi, B., Ernest, M. E., Hu, X., Yu, A."Downregulation of IDH2 exacerbates the malignant progression of osteosarcoma cells via increased NF-κB and MMP-9 activation". Oncology Reports 35, no. 4 (2016): 2277-2285. https://doi.org/10.3892/or.2016.4553