Open Access

Antitumorigenic effect of interferon-β by inhibition of undifferentiated glioblastoma cells

  • Authors:
    • Shun Yamamuro
    • Emiko Sano
    • Yutaka Okamoto
    • Yushi Ochiai
    • Takashi Ohta
    • Akiyoshi Ogino
    • Atsushi Natsume
    • Toshihiko Wakabayashi
    • Takuya Ueda
    • Hiroyuki Hara
    • Tomohiro Nakayama
    • Atsuo Yoshino
    • Yoichi Katayama
  • View Affiliations

  • Published online on: September 14, 2015     https://doi.org/10.3892/ijo.2015.3165
  • Pages: 1647-1654
  • Copyright: © Yamamuro et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

Glioma stem-like cells (GSCs) are undifferentiated cells that are considered to be an origin of glioblastomas. Furthermore, they may contribute to treatment resistance and recurrence in glioblastomas. GSCs differentiate into differentiated glioma cells (non-glioma stem-like cells: non‑GSCs), and interconversion might occur between GSCs and non-GSCs. We investigated whether interferon-beta (IFN-β) could exert any efficacy towards GSCs or such interconversion processes. The neural stem cell marker CD133 and pluripotency marker Nanog in GSCs were analyzed to evaluate their differentiation levels. GSCs were considered to undergo differentiation into non-GSCs upon serum exposure, since the expression of CD133 and Nanog in the GSCs was negatively affected. Furthermore, the cells regained their undifferentiated features upon removal of the serum. However, we verified that IFN-β reduced cell proliferation and tumor sphere formation in GSCs, and induced suppression of the restoration of such undifferentiated features. In addition, we also confirmed that IFN-β suppressed the acquisition process of undifferentiated features in human malignant glioma cell lines. Our data thus suggest that IFN-β could be an effective agent not only through its cell growth inhibitory effect on GSCs but also as a means of targeting the interconversion between GSCs and non-GSCs, indicating the possibility of IFN-β being used to prevent treatment resistance and recurrence in glioblastomas, via the inhibition of undifferentiated features.

Introduction

Malignant gliomas, especially glioblastomas, are the most frequently occurring primary tumors of the central nervous system, and represent one of the most lethal malignancies. Glioblastomas have been reported to be heterogeneous bulk tumors comprising differentiated and undifferentiated cells with a self-renewal ability, pluripotency, and tumorigenicity (1). Such heterogeneity may contribute to tumor expansion, invasion, metastasis, and drug resistance. The undifferentiated cells, a distinct subpopulation, within the tumors may derive from a limited source of glioblastoma cells and are termed glioblastoma stem-like cells (GSCs) (24). These cells are considered to be capable of aberrantly differentiating into diverse cell types, differentiated glioma cells (non-glioma stem-like cells: non-GSCs), in response to their microenvironment (57). Furthermore, there may be interconversion between GSCs and non-GSCs (7).

The efficacy of postoperative radiotherapy with concomitant and adjuvant temozolomide (TMZ) as the first-line treatment for glioblastomas was reported to be 9.8% in terms of the 5-year survival rate versus 1.9% with radiotherapy alone in a recent EORTC/NCIC randomized phase III trial (8,9). Concomitant radiotherapy with TMZ followed by adjuvant TMZ chemotherapy has thus become a current standard postoperative treatment for glioblastomas. Among the factors that may contribute to TMZ resistance, O6-methylguanine-DNA methyltransferase (MGMT, a protein that removes drug-induced alkylguanine adducts from DNA created by TMZ) is thought to be involved in its crucial mechanisms (9,10).

Human interferon-beta (IFN-β), a type I interferon, was first discovered on the basis of its antiviral activities. Subsequently, it was found to exhibit pleiotropic biological activities including immunomodulatory activity, anti-angiogenetic activity and direct antitumor effects: e.g., growth inhibition, and apoptosis (1113). Recently, a synergistic antitumor effect between TMZ and IFN-β was reported in malignant glioma cells in vitro (14,15). Natsume et al suggested that a sensitizing effect between IFN-β and TMZ in TMZ-resistant glioma cells was possibly due to attenuation of MGMT expression via induction of the protein p53 (14). More recently, the INTEGRA clinical study (integrated Japanese multicenter clinical trial: a phase II study on IFN-β and TMZ for glioma in combination with radiotherapy) was undertaken to evaluate the clinical effectiveness in glioblastomas (16,17).

Concerning the treatment of glioblastomas, it is important to elucidate the detailed features of GSCs as well as the underlying mechanisms of interconversion between GSCs and non-GSCs. To this end, we examined whether IFN-β could exert some effect on the interconversion between GSCs and non-GSCs, especially the conversion process of non-GSCs into GSCs.

Materials and methods

Cell culture

As GSCs, we employed 0222-GSC provided by Nagoya University School of Medicine (Nagoya, Japan) (7,8). The 0222-GSC satisfied the following criteria: i) the cell lines could be maintained in serum-free-media for 3 months (minimum) and ii) 103 cells formed tumors in the brain of nonobese diabetic mice with severe combined immunodeficiency disease (18). 0222-GSC culture was undertaken in serum-free neurobasal (NBE) media (Invitrogen, Carlsbad, CA, USA) comprising N2 and B27 supplements (Invitrogen), human recombinant basic fibroblast growth factor (bFGF; R&D Systems, Minneapolis, MN, USA), and epidermal growth factor (EGF; R&D Systems).

Human malignant glioma cell lines A-172, AM-38, T98G, U-251MG, YH-13 (purchased from Health Science Research Resources Bank, Sennan, Osaka, Japan), U-87MG, and U-138MG (purchased from American Type Culture Collection, Manassas, VA, USA) were also used in the present study. These human malignant glioma cell lines were cultured in Dulbecco's modified Eagle's medium (Nissui Pharmaceutical, Tokyo, Japan) containing 10% fetal bovine serum (FBS) (Life Technologies, Grand Island, NY, USA) (18,19).

Populations of serum-induced brain tumor cells (S-BTC) were established by culturing 0222-GSC in serum medium for 3 weeks. Moreover, populations of revertant-glioma stem-like cells (Rev-GSC) were established by additional culturing of S-BTC in serum-free medium for 2 weeks. On the other hand, populations of S-BTC+IFN were established by culturing 0222-GSC in serum medium with 10 IU/ml IFN-β (Toray Industries, Tokyo, Japan) twice a week for 3 weeks (the total number of administrations was 6). Populations of Rev-GSC+IFN were then established by additional culturing of S-BTC+IFN in serum-free medium for 2 weeks (Fig. 1). Additionally, populations of GSC+IFN were established by culturing 0222-GSC in serum-free medium with 10 IU/ml IFN-β for one week.

Rev-A-172, Rev-AM-38, Rev-T98G, Rev-U-87MG, Rev-U-138MG, Rev-U-251MG, and Rev-YH-13 were established by culturing the respective cells in serum-free medium for 2 weeks. Moreover, Rev-A-172+IFN, Rev-AM-38+IFN, Rev-T98G+IFN, Rev-U-87MG+IFN, Rev-U-138MG+IFN, Rev-U-251MG+IFN, and Rev-YH-13+IFN were established by culturing the respective cells in serum-free medium for 2 weeks after culture in serum supplemented medium with 10 IU/ml IFN-β twice a week for 3 weeks (the total number of administrations was 6) (Fig. 2).

Flow cytometric analysis

The neural stem cell marker CD133 was employed as a marker of GSCs. Furthermore, glial fibrillary acidic protein (GFAP) was used as a marker of astrocytes, and galactocerebroside C (GalC) was used as a marker of oligodendrocytes (2,18,2022). We employed the following fluorescence conjugated monoclonal antibodies: anti-CD133 (CD133-PE, 130-080-801; Miltenyi Biotec, Auburn, CA, USA), anti-GFAP (anti-GFAP-Alexa Fluor 488, 561449; Becton-Dickinson, NJ, USA), and anti-GalC (antiGalC-Alexa Fluor 488, MAB342A4; Millipore, Temecula, CA, USA).

The expressions of CD133, GFAP and GalC in 0222-GSC, S-BTC, Rev-GSC, Rev-GSC+IFN, and the 7 human malignant glioma cell lines were analyzed with a fluorescence-activated cell sorter (FACS). The fluorescence was measured using FACSCalibur flow cytometer (Becton-Dickinson, Franklin Lakes, NJ, USA), and the DNA histograms were analyzed with Flowjo software (BioLegend, San Diego, CA, USA).

The FACS analyses were repeated at least 3 times in each experiment, and we confirmed that similar tendencies were obtained.

mRNA expressions of Nanog

We analyzed the mRNA expression of pluripotency markers, Nanog, in 0222-GSC, S-BTC, Rev-GSC, Rev-GSC+IFN, Rev-U-87MG, and Rev-U-87MG+IFN by the real-time polymerase chain reaction (real-time PCR) (23,24). An RNeasy Mini kit (Qiagen Inc., Valencia, CA, USA) was employed for the extraction of mRNA. A SepOne Real-time PCR System (Applied Biosystems, Foster City, CA, USA) was used for the RT-PCR reaction. The housekeeping gene glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was employed for the control. The following primers, synthesized by Opero (Tokyo, Japan), were used in the real-time PCR as described previously (7): Nanog (forward, 5′-GTC CCG GTC AAG AAA CAG AA; reverse, 5′-TGC GTC ACA CCA TTG CTA TT) and GAPDH (forward, 5′-TCG GTG CGT GCC CAG TTG AAC C; reverse: 5′-ATG CGG CTG ACT GTC GAA CAG GAG). The real-time PCR was carried out with a final volume of 50 μl containing 10 pmol of each sense and antisense primer, 2.5 μl of 50 mM Mn(OAc)2, 25 μl of RNA-direct™ SYBR Green Real-time PCR Master Mix (Toyobo, Osaka, Japan), 2 μg of extracted mRNA, and RNA-free water. Amplification was carried out by initial denaturing at 90°C for 30 sec, reverse transcription at 61°C for 20 min, second denaturing at 95°C for 1 min, followed by 40 cycles of extension at 95°C for 15 sec, 55°C for 15 sec, and 74°C for 45 sec. The expression levels were calculated using the following equations by comparing the threshold cycles (CT): ΔCT = CT of Nanog - CT of GAPDH, ΔΔCT = ΔCT (target cell line) - ΔCT (reference cell line), and ratio = 2−ΔΔCT (25).

Growth inhibitory effect of IFN-β on 0222-GSC

The growth inhibitory effect of IFN-β was evaluated by counting the number of cells using a Coulter Counter (Coulter Counter ZI, Beckman coulter, Fullerton, CA, USA). Briefly, cells were plated at 2×104 cells per well in 24-well, flat-bottomed plates (Iwaki, Chiba, Japan) and incubated in the medium with or without 1.0–100 IU/ml of IFN-β. After 5 days of exposure to various concentrations of IFN-β, the cells were counted with the cell counter.

Sphere formation assay

0222-GSC cells were placed into 96-well plates (50 cells/well) in serum-free medium. IFN-β (10 IU/ml) was administered on one side (48-wells). At day 7 after seeding, the spheres containing >10 cells were counted.

Statistical evaluations

Statistical analyses were performed using the unpaired, Mann-Whitney U test. If the samples comprised more than three groups, the significance of the overall samples was evaluated by the Kruskal-Wallis test before evaluating the significant differences between pairs of groups by the Mann-Whitney U test. All quantitative data are presented as the means ± SE from at least six samples per data point. Statistical software IBM SPSS Statistics version 21.0 (International Business Machines Corp., Armonk, NY, USA) was employed for the data analysis.

Results

Characteristics of GSCs and effects of IFN-β

0222-GSC formed tumor spheres in serum-free medium. However, the cells did not show tumor spheres in response to a change to serum medium for 3 weeks (S-BTC). In addition, S-BTC reformed tumor spheres again after culturing in serum-free medium for 2 weeks (Rev-GSC) (Fig. 3A).

In the FACS analysis, 0222-GSC expressed a positive reaction for CD133, but the expressions became negatively converted in S-BTC. Further, the expression of CD133 again positively converted in Rev-GSC (expression was not observed when S-BTC were cultured in serum-free medium for only a week: data not shown). On the other hand, the expression of CD133 was suppressed in Rev-GSC+IFN (Fig. 3B).

The mRNA expression of Nanog in 0222-GSC, S-BTC, Rev-GSC, and Rev-GSC+IFN were analyzed by the real-time PCR. The expression in S-BTC was reduced to 0.06±0.03-fold as compared to the expressions in 0222-GSC. However, the expression in Rev-GSC increased again to 0.79±0.09-fold as compared to the expressions in 0222-GSC. On the other hand, the expression of mRNA Nanog in Rev-GSC+IFN was also increased to 0.42±0.07-fold as compared to the expressions in 0222-GSC, although the level was significantly lower than that for Rev-GSC (p<0.01; Fig. 3C).

Furthermore, Rev-GSC and Rev-GSC+IFN were cultured continuously for an additional 2 weeks in serum-free media (total of 4 weeks culture in serum-free media). Although the expression of mRNA Nanog in Rev-GSC+IFN was reduced to 0.86±0.08-fold as compared to those in Rev-GSC, there was no significant difference between Rev-GSC+IFN and Rev-GSC (p=0.09; Fig. 3D).

Effect of IFN-β on GSC

As shown in Fig. 4A, cell growth inhibitory effects of IFN-β on 0222-GSC were observed in a dose-dependent manner.

We next examined the expression of CD133, GFAP and GalC in 0222-GSC and GSC+IFN by FACS analysis. The expression of CD133 and GFAP was suppressed, but the expression of GalC was enhanced in GSC+IFN as compared to those in 0222-GSC (Fig. 4B). The data obtained indicated IFN-β induced oligodendrogenesis in 0222-GSCs, as reported previously (18).

The numbers of tumor spheres were counted after 7 days of culture in serum-free medium and compared between IFN-β treatment, but not in 0222-GSC. As shown in Fig. 4C, the number of tumor spheres in the IFN-β-treated cells were significantly lower than the number of tumor spheres in the non-treated cells (p<0.01). We found that the sphere formation ability was attenuated by IFN-β treatment, although both groups could form tumor spheres.

Effect of IFN-β on malignant glioma cell lines

The human malignant glioma cell lines did not form tumor spheres. On FACS analysis, U-251MG expressed CD133, even though the other 6 cell lines did not (Fig. 5A). Subsequently, each of the cell lines was able to form tumor spheres when cultured in serum-free medium for 2 weeks (viz., Rev-A-172, Rev-AM-38, Rev-T98G, Rev-U-87MG, Rev-U-138MG, Rev-U-251MG and Rev-YH-13). Expressions of CD133 was newly observed in U-87MG, U-138MG and YH-13 on FACS analysis (Fig. 5B). Each of the cell lines, which had been administered IFN-β previously under serum medium, also formed tumor spheres when cultured in serum-free medium for 2 weeks (viz., Rev-A-172+IFN, Rev-AM-38+IFN, Rev-T98G+IFN, Rev-U-87MG+IFN, Rev-U-138MG+IFN, Rev-U-251MG+IFN and Rev-YH-13+IFN), but none of these cell lines expressed CD133 (Fig. 5C).

Moreover, we analyzed the mRNA expressions of Nanog in Rev-U-87MG and Rev-U-87MG+IFN by the real-time PCR. The expressions in Rev-U-87MG+IFN was significantly reduced to 0.61±0.07-fold as compared to the expression in Rev-U-87MG (p<0.01; Fig. 5D).

Discussion

GSCs share many properties with normal stem cells including self-renewal and pluripotency, and exhibit tumorigenetic ability. Furthermore, GSCs may contribute to tumor development, invasion, recurrence and chemo/radiation resistance in glioblastomas (7,18,26,27). It is important therefore to take GSCs fully into accound when deciding treatment strategies for glioblastomas.

The undifferentiated state of GSCs displays characteristics such as tumor sphere formation, CD133 expression, and mRNA Nanog expression (7,18,2022). The GSC cell line, 0222-GSC, demonstrated tumor sphere formation, CD133 expression, and a high expression of mRNA Nanog. However, it lost such characteristics on exposure to serum medium (S-BTC). This may indicate that GSCs can change/differentiate to non-GSCs in response to signals from their microenvironment (7,18). On the other hand, S-BTC re-exhibited the characteristics of tumor sphere formation, CD133 expression, and a high expression of mRNA Nanog upon removal of the serum from the medium. Such changes suggested that the non-GSCs had regained an undifferentiated state in response to signals from their microenvironment. These findings are in keeping with those described in previous reports (7).

Human malignant glioma cell lines cultured in conventional medium did not form tumor spheres, nor did they express CD133 (except for U-251MG) in the present study. All cell lines formed tumor spheres when cultured in serum-free medium, and furthermore U-87MG, U-138MG, and YH-13 expressed CD133. Qiang et al, investigated the percentage of CD133-positive cells in A-172, U-87MG, and U-251MG by FACS analysis, and found that the existence of CD133-positive cells was common in U-251MG (28). They also reported that human malignant glioma cell lines formed tumor spheres and showed increased expression of CD133 when cultured in serum-free medium (28). We obtained similar results, and confirmed that human malignant glioma cells (non-GSCs) could acquire an undifferentiated state (return to GSCs) in response to signals from their microenvironment.

It might be possible to enhance the effects of radiotherapy/chemotherapy for glioblastomas, if we could promote the differentiation of GSCs and/or suppress the return process of non-GSCs to GSCs. Not only GSCs themselves but also the interconversion between GSCs and non-GSCs could be new targets in glioblastoma treatment. In the present study, IFN-β revealed a cell growth inhibitory effect and a cell differentiation effect with suppression of tumor sphere formation and CD133 expression in 0222-GSC. In addition, IFN-β suppressed the acquisition process of undifferentiated features in S-BTC and some of the human malignant glioma cell lines investigated. Thus, IFN-β might represent an effective agent not only through its cell growth inhibitory effect on GSCs but also as a means of targeting the interconversion between GSCs and non-GSCs.

0222-GSC cells were induced to differentiate into oligodendrogenetic cells in the present study, as reported previously (8). The oligodendrogliomas display a better prognosis than the astrocytomas, through their high sensitivity to adjuvant therapy including radiotherapy and chemotherapy (29). Such a differentiation effect is therefore considered to offer a new treatment strategy for glioblastomas, and IFN-β may represent an effective drug for the treatment of glioblastomas, not only when used alone but also in combination with TMZ. Further, in the present study, IFN-β also induced suppression of acquisition processes involved in the undifferentiation of non-GSCs at 2 weeks, although the suppression of mRNA Nanog was not significant at 4 weeks after IFN-β treatment. The interval of IFN-β a dministration employed during maintenance therapy in the INTEGRA study was once every 4 weeks (17), so that a higher effect could be expected if the intervals were shorter.

Finally, the effects of IFN-β on the tumor cells were considered as not only temporary, but also genetic or epigenetic changes, since the effects of IFN-β lasted >2 weeks. DNA methylation, and histone modification (acetylation, methylation, and phosphorylation) are known to represent epigenetic changes of the cells. Among them, histone methylation has been reported to play an important role in the control of normal stem cell differentiation. In particular, histone H3 lysine 27 trimethylation (H3K27me3) in the promoter region of the differentiation-associated genes is known to inhibit the transcription of these genes (30,31). H3K27me3 is catalyzed by enhancer of zeste homolog 2 (EZH2; histone methyltransferase). EZH2 has been reported to contribute to undifferentiated features in normal stem cells (3236). Enhanced gene expressions of EZH2 have been observed in various cancers (32,3739), and inhibitors of EZH2 have been found to exhibit antitumor effects (40,41). While EZH2 has been described as an oncogene, it has also been reported as a tumor-suppressor gene since mutations of EZH2 have been observed extinguished in several cancers (42). Although the detailed relationships between EZH2 and cancer remain to be elucidated, there is a report suggesting that EZH2 may contribute to the interconversion between GSCs and non-GSCs (7). Further research is clearly necessary, but an association could exist between EZH2 and the effects of IFN-β.

Acknowledgements

This study was supported in part by a grant from the Health Sciences Research Institute, Inc., Yokohama, Japan, to the Division of Companion Diagnostics, Department of Pathology of Microbiology, Nihon University School of Medicine. The authors are grateful to Hiroyuki Satake and Nobuo Miyazaki, Toray Industries Inc. (Tokyo, Japan), for their invaluable discussions and supply of natural type IFN-β. Some parts of this study have been submitted for the Japanese-language thesis of Shun Yamamuro's Ph.D. degree at Nihon University School of Medicine. Toshihiko Wakabayashi received funds for other research projects not related to this study from Eisai Co., Ltd, Chugai Pharmaceutical Co., Ltd., MSD K.K., Mizuho Co., Ltd., Otsuka Pharmaceutical Co., Ltd., Takeda Pharmaceutical Co., Ltd., and Toray Industries Inc. Takuya Ueda received funds for other research projects not related to this study from the Ministry of Education, Culture, Sports, Science and Technology, Japan, Minister of Economy, Trade and Industry, Japan and Human Frontier Science Program. Yoichi Katayama received research funds for another research project from Medtronic Japan Co., Ltd.

References

1 

Lee J, Kotliarova S, Kotliarov Y, Li A, Su Q, Donin NM, Pastorino S, Purow BW, Christopher N, Zhang W, et al: Tumor stem cells derived from glioblastomas cultured in bFGF and EGF more closely mirror the phenotype and genotype of primary tumors than do serum-cultured cell lines. Cancer Cell. 9:391–403. 2006. View Article : Google Scholar : PubMed/NCBI

2 

Wilson RJ, Thomas CD, Fox R, Roy DB and Kunin WE: Spatial patterns in species distributions reveal biodiversity change. Nature. 432:393–396. 2004. View Article : Google Scholar : PubMed/NCBI

3 

Lee J, Son MJ, Woolard K, Donin NM, Li A, Cheng CH, Kotliarova S, Kotliarov Y, Walling J, Ahn S, et al: Epigenetic-mediated dysfunction of the bone morphogenetic protein pathway inhibits differentiation of glioblastoma-initiating cells. Cancer Cell. 13:69–80. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Peñuelas S, Anido J, Prieto-Sánchez RM, Folch G, Barba I, Cuartas I, García-Dorado D, Poca MA, Sahuquillo J, Baselga J, et al: TGF-beta increases glioma-initiating cell self-renewal through the induction of LIF in human glioblastoma. Cancer Cell. 15:315–327. 2009. View Article : Google Scholar : PubMed/NCBI

5 

Gupta PB, Chaffer CL and Weinberg RA: Cancer stem cells: Mirage or reality? Nat Med. 15:1010–1012. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Gupta PB, Fillmore CM, Jiang G, Shapira SD, Tao K, Kuperwasser C and Lander ES: Stochastic state transitions give rise to phenotypic equilibrium in populations of cancer cells. Cell. 146:633–644. 2011. View Article : Google Scholar : PubMed/NCBI

7 

Natsume A, Ito M, Katsushima K, Ohka F, Hatanaka A, Shinjo K, Sato S, Takahashi S, Ishikawa Y, Takeuchi I, et al: Chromatin regulator PRC2 is a key regulator of epigenetic plasticity in glioblastoma. Cancer Res. 73:4559–4570. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, Belanger K, Brandes AA, Marosi C, Bogdahn U, et al; European Organisation for Research and Treatment of Cancer Brain Tumor and Radiotherapy Groups; National Cancer Institute of Canada Clinical Trials Group. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 352:987–996. 2005. View Article : Google Scholar : PubMed/NCBI

9 

Stupp R, Hegi ME, Mason WP, van den Bent MJ, Taphoorn MJ, Janzer RC, Ludwin SK, Allgeier A, Fisher B, Belanger K, et al; European Organisation for Research and Treatment of Cancer Brain Tumour and Radiation Oncology Groups; National Cancer Institute of Canada Clinical Trials Group. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol. 10:459–466. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Pegg AE: Mammalian O6-alkylguanine-DNA alkyltransferase: Regulation and importance in response to alkylating carcinogenic and therapeutic agents. Cancer Res. 50:6119–6129. 1990.PubMed/NCBI

11 

Saito R, Mizuno M, Hatano M, Kumabe T, Yoshimoto T and Yoshida J: Two different mechanisms of apoptosis resistance observed in interferon-beta induced apoptosis of human glioma cells. J Neurooncol. 67:273–280. 2004. View Article : Google Scholar : PubMed/NCBI

12 

Vannucchi S, Chiantore MV, Mangino G, Percario ZA, Affabris E, Fiorucci G and Romeo G: Perspectives in biomolecular therapeutic intervention in cancer: From the early to the new strategies with type I interferons. Curr Med Chem. 14:667–679. 2007. View Article : Google Scholar : PubMed/NCBI

13 

Yoshino A, Katayama Y, Yokoyama T, Watanabe T, Ogino A, Ota T, Komine C, Fukushima T and Kusama K: Therapeutic implications of interferon regulatory factor (IRF)-1 and IRF-2 in diffusely infiltrating astrocytomas (DIA): Response to interferon (IFN)-beta in glioblastoma cells and prognostic value for DIA. J Neurooncol. 74:249–260. 2005. View Article : Google Scholar : PubMed/NCBI

14 

Natsume A, Ishii D, Wakabayashi T, Tsuno T, Hatano H, Mizuno M and Yoshida J: IFN-beta down-regulates the expression of DNA repair gene MGMT and sensitizes resistant glioma cells to temozolomide. Cancer Res. 65:7573–7579. 2005.PubMed/NCBI

15 

Park JA, Joe YA, Kim TG and Hong YK: Potentiation of anti-glioma effect with combined temozolomide and interferon-beta. Oncol Rep. 16:1253–1260. 2006.PubMed/NCBI

16 

Wakabayashi T, Kayama T, Nishikawa R, Takahashi H, Yoshimine T, Hashimoto N, Aoki T, Kurisu K, Natsume A, Ogura M, et al: A multicenter phase I trial of interferon-beta and temozolomide combination therapy for high-grade gliomas (INTEGRA Study). Jpn J Clin Oncol. 38:715–718. 2008. View Article : Google Scholar : PubMed/NCBI

17 

Wakabayashi T, Kayama T, Nishikawa R, Takahashi H, Hashimoto N, Takahashi J, Aoki T, Sugiyama K, Ogura M, Natsume A, et al: A multicenter phase I trial of combination therapy with interferon-β and temozolomide for high-grade gliomas (INTEGRA study): The final report. J Neurooncol. 104:573–577. 2011. View Article : Google Scholar : PubMed/NCBI

18 

Yuki K, Natsume A, Yokoyama H, Kondo Y, Ohno M, Kato T, Chansakul P, Ito M, Kim SU and Wakabayashi T: Induction of oligodendrogenesis in glioblastoma-initiating cells by IFN-mediated activation of STAT3 signaling. Cancer Lett. 284:71–79. 2009. View Article : Google Scholar : PubMed/NCBI

19 

Yoshino A, Tashiro S, Ogino A, Yachi K, Ohta T, Fukushima T, Watanabe T, Katayama Y, Okamoto Y, Sano E, et al: Gene expression profiles predicting the response to IFN-β and a combination of temozolomide and IFN-β in malignant gliomas. Int J Oncol. 39:529–542. 2011.PubMed/NCBI

20 

Griguer CE, Oliva CR, Gobin E, Marcorelles P, Benos DJ, Lancaster JR Jr and Gillespie GY: CD133 is a marker of bioenergetic stress in human glioma. PLoS One. 3:e36552008. View Article : Google Scholar : PubMed/NCBI

21 

Poltavtseva RA, Marey MV, Aleksandrova MA, Revishchin AV, Korochkin LI and Sukhikh GT: Evaluation of progenitor cell cultures from human embryos for neurotransplantation. Brain Res Dev Brain Res. 134:149–154. 2002. View Article : Google Scholar : PubMed/NCBI

22 

Singh SK, Clarke ID, Terasaki M, Bonn VE, Hawkins C, Squire J and Dirks PB: Identification of a cancer stem cell in human brain tumors. Cancer Res. 63:5821–5828. 2003.PubMed/NCBI

23 

Hatano SY, Tada M, Kimura H, Yamaguchi S, Kono T, Nakano T, Suemori H, Nakatsuji N and Tada T: Pluripotential competence of cells associated with Nanog activity. Mech Dev. 122:67–79. 2005. View Article : Google Scholar

24 

Mitsui K, Tokuzawa Y, Itoh H, Segawa K, Murakami M, Takahashi K, Maruyama M, Maeda M and Yamanaka S: The homeoprotein Nanog is required for maintenance of pluripotency in mouse epiblast and ES cells. Cell. 113:631–642. 2003. View Article : Google Scholar : PubMed/NCBI

25 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar

26 

Clevers H: The cancer stem cell: Premises, promises and challenges. Nat Med. 17:313–319. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, Hjelmeland AB, Dewhirst MW, Bigner DD and Rich JN: Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature. 444:756–760. 2006. View Article : Google Scholar : PubMed/NCBI

28 

Qiang L, Yang Y, Ma YJ, Chen FH, Zhang LB, Liu W, Qi Q, Lu N, Tao L, Wang XT, et al: Isolation and characterization of cancer stem like cells in human glioblastoma cell lines. Cancer Lett. 279:13–21. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Smith TJ: The art of oncology: When the tumor is not the target. Tell it like it is. J Clin Oncol. 18:3441–3445. 2000.PubMed/NCBI

30 

Kondo Y, Shen L, Cheng AS, Ahmed S, Boumber Y, Charo C, Yamochi T, Urano T, Furukawa K, Kwabi-Addo B, et al: Gene silencing in cancer by histone H3 lysine 27 trimethylation independent of promoter DNA methylation. Nat Genet. 40:741–750. 2008. View Article : Google Scholar : PubMed/NCBI

31 

Sparmann A and van Lohuizen M: Polycomb silencers control cell fate, development and cancer. Nat Rev Cancer. 6:846–856. 2006. View Article : Google Scholar : PubMed/NCBI

32 

Chi P, Allis CD and Wang GG: Covalent histone modifications--miswritten, misinterpreted and mis-erased in human cancers. Nat Rev Cancer. 10:457–469. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Ougolkov AV, Bilim VN and Billadeau DD: Regulation of pancreatic tumor cell proliferation and chemoresistance by the histone methyltransferase enhancer of zeste homologue 2. Clin Cancer Res. 14:6790–6796. 2008. View Article : Google Scholar : PubMed/NCBI

34 

Raaphorst FM, Meijer CJ, Fieret E, Blokzijl T, Mommers E, Buerger H, Packeisen J, Sewalt RA, Otte AP and van Diest PJ: Poorly differentiated breast carcinoma is associated with increased expression of the human polycomb group EZH2 gene. Neoplasia. 5:481–488. 2003. View Article : Google Scholar

35 

Sauvageau M and Sauvageau G: Polycomb group proteins: Multifaceted regulators of somatic stem cells and cancer. Cell Stem Cell. 7:299–313. 2010. View Article : Google Scholar : PubMed/NCBI

36 

Schwartz YB and Pirrotta V: Polycomb silencing mechanisms and the management of genomic programmes. Nat Rev Genet. 8:9–22. 2007. View Article : Google Scholar

37 

Bracken AP and Helin K: Polycomb group proteins: Navigators of lineage pathways led astray in cancer. Nat Rev Cancer. 9:773–784. 2009. View Article : Google Scholar : PubMed/NCBI

38 

Kleer CG, Cao Q, Varambally S, Shen R, Ota I, Tomlins SA, Ghosh D, Sewalt RG, Otte AP, Hayes DF, et al: EZH2 is a marker of aggressive breast cancer and promotes neoplastic transformation of breast epithelial cells. Proc Natl Acad Sci USA. 100:11606–11611. 2003. View Article : Google Scholar : PubMed/NCBI

39 

Cebrià F, Kobayashi C, Umesono Y, Nakazawa M, Mineta K, Ikeo K, Gojobori T, Itoh M, Taira M, Sánchez Alvarado A, et al: FGFR-related gene nou-darake restricts brain tissues to the head region of planarians. Nature. 419:620–624. 2002. View Article : Google Scholar : PubMed/NCBI

40 

Fiskus W, Wang Y, Sreekumar A, Buckley KM, Shi H, Jillella A, Ustun C, Rao R, Fernandez P, Chen J, et al: Combined epigenetic therapy with the histone methyltransferase EZH2 inhibitor 3-deazaneplanocin A and the histone deacetylase inhibitor panobinostat against human AML cells. Blood. 114:2733–2743. 2009. View Article : Google Scholar : PubMed/NCBI

41 

Tan J, Yang X, Zhuang L, Jiang X, Chen W, Lee PL, Karuturi RK, Tan PB, Liu ET and Yu Q: Pharmacologic disruption of Polycomb-repressive complex 2-mediated gene repression selectively induces apoptosis in cancer cells. Genes Dev. 21:1050–1063. 2007. View Article : Google Scholar : PubMed/NCBI

42 

Ernst T, Chase AJ, Score J, Hidalgo-Curtis CE, Bryant C, Jones AV, Waghorn K, Zoi K, Ross FM, Reiter A, et al: Inactivating mutations of the histone methyltransferase gene EZH2 in myeloid disorders. Nat Genet. 42:722–726. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2015
Volume 47 Issue 5

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yamamuro S, Sano E, Okamoto Y, Ochiai Y, Ohta T, Ogino A, Natsume A, Wakabayashi T, Ueda T, Hara H, Hara H, et al: Antitumorigenic effect of interferon-β by inhibition of undifferentiated glioblastoma cells. Int J Oncol 47: 1647-1654, 2015
APA
Yamamuro, S., Sano, E., Okamoto, Y., Ochiai, Y., Ohta, T., Ogino, A. ... Katayama, Y. (2015). Antitumorigenic effect of interferon-β by inhibition of undifferentiated glioblastoma cells. International Journal of Oncology, 47, 1647-1654. https://doi.org/10.3892/ijo.2015.3165
MLA
Yamamuro, S., Sano, E., Okamoto, Y., Ochiai, Y., Ohta, T., Ogino, A., Natsume, A., Wakabayashi, T., Ueda, T., Hara, H., Nakayama, T., Yoshino, A., Katayama, Y."Antitumorigenic effect of interferon-β by inhibition of undifferentiated glioblastoma cells". International Journal of Oncology 47.5 (2015): 1647-1654.
Chicago
Yamamuro, S., Sano, E., Okamoto, Y., Ochiai, Y., Ohta, T., Ogino, A., Natsume, A., Wakabayashi, T., Ueda, T., Hara, H., Nakayama, T., Yoshino, A., Katayama, Y."Antitumorigenic effect of interferon-β by inhibition of undifferentiated glioblastoma cells". International Journal of Oncology 47, no. 5 (2015): 1647-1654. https://doi.org/10.3892/ijo.2015.3165