Int J Clin Exp Med 2020;13(12):9390-9397 www.ijcem.com /ISSN:1940-5901/IJCEM0119448

Original Article Inhibition of Bmi1 suppresses tumorigenicity in glioma stem cells via the YAP/Pax3 pathway

Ke Mao, Ding Lei, Yuan Fang

Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China Received August 3, 2020; Accepted September 28, 2020; Epub December 15, 2020; Published December 30, 2020

Abstract: Glioma stem cells (GSCs) are related to the recurrence of glioblastoma multiforme (GBM) due to their resistance to chemotherapy (CT) and radiotherapy (RT). Bmi1 is associated with self-renewal of malignant cancer stem cells (CSCs). The aim of this study was to evaluate the effect of Bmi1 downregulation in GSCs to target and eradicate CSCs in malignant tissues. A series of experiments involving Bmi1 knockdown, MTS assay, Pax3 and YAP overexpression, quantitative real-time PCR (qRT-PCR), western blotting, BrdU incorporation assay, and soft agar assay were performed in this study. Knockdown of Bmi1 notably suppressed GSC proliferation as shown in BrdU- positive, Bmi1-knockdown cells. Additionally, mRNA and expression levels of Pax3 were suppressed in Bmi1- knockdown cells, as shown by western blot and qRT-PCR results. Excessive Pax3 expression in Bmi1-knockdown cells attenuated the suppressive effect of Bmi1 knockdown on GSC proliferation. Pax3 expression was modulated by Bmi1 via the YAP pathway. Moreover, Pax3 modulated Bmi1 activity in GSCs. The above findings indicate that Bmi1 activity is involved in the proliferation and transformation of GSCs in GBM.

Keywords: Bmi1, GSC, Pax3, proliferation, tumorigenicity

Introduction that are essential for regulation, de- termination of the density of stem cells (SCs), Glioblastoma multiforme (GBM) is the most suppression of , and viabil- prevalent primary cancer of the adult brain [1, ity modulation of various malignancies [13, 14]. 2]. The clinical outcome of GBM is poor, with a Bmi1 regulates essential periods during the median survival rate of 14.6 months, despite development of malignancy such as metasta- therapeutic approaches such as surgery, che- sis, invasion by regulating epithelial-to-mesen- motherapy (CT), and radiotherapy (RT) [3-5]. chymal transition (EMT), and resistance to Unregulated proliferation, infiltration tendency, drugs [15, 16]. Bmi1 expression is remarkably unstable genomes, and hypervascularization associated with unfavorable clinical outcomes are the main contributors to the poor progn- and viability as well as increased aggression in osis [6]. The resistance of glioma stem cells patients with breast cancer (BC) [17-19]. (GSCs) to CT and RT results in recurrence and limited overall survival [7, 8]. Consequently, Pax3 participates in the development of malig- innovative strategies targeting GSCs are re- nancies [20]. Pax3 expression has been found quired to meet the challenge of resistance and to be positively associated with the develop- promote therapeutic efficiency. It is widely ac- ment of melanoma [21, 22]. Several reports cepted that GSCs contribute to the prolifera- have shown that Pax3 negatively regulates tion, recurrence, and resistance of GBM [9, 10]. glial fibrillary acidic protein (GFAP) expression Although investigation of GSC markers with high specificity and sensitivity is ongoing, sev- during differentiation of neural SCs (NSCs) to eral reports have shown that EGFRvIII+ cells dis- astrocytes [23, 24]. The aim of the present play the ability to promote malignancy [11, 12]. study was to elucidate the effects of Bmi1 on the role of GSCs, particularly in the regulation Bmi1 participates in the self-renewal of cancer of proliferation and the development of malig- stem cells (CSCs) via the modulation of nancy, using human primary GBM cells. Inhibition of Bmi1 suppresses tumorigenicity in GSCs

Materials and methods Poly-Transfecter Cell Reagent was used for transient transfection. Derivation and culture of glioma cell lines Quantitative Real-Time PCR (qRT-PCR) We harvested primary glioblastoma multifor- me cells from the corresponding cases. The The purified RNA was dissolved in 30 μL of Papain Dissociation System (Worthington, NJ, RNase-free water. cDNA was synthesized using USA) was used to digest the excised glioma 2 μg of total RNA with the PrimeScript™ RT specimens. Malignant cells were cultured in reagent kit and gDNA Eraser (Takara, Dalian, Dulbecco’s Modified Eagle Medium/Nutrient Liaoning, Japan), in preparation for the subse- Mixture F-12 (DMEM/F-12; Invitrogen, Carls- quent qRT-PCR of Bmi1. The SYBR Premix Ex bad, CA, USA), containing B27 minus vitamin A Taq II kit and an Applied Biosystems 7500 (1/50; Invitrogen, Carlsbad, CA, USA), 20 ng/ Fluorescent Quantitative PCR system was us- mL endothelial growth factor (EGF; eBioscienc- ed for qRT-PCR. Expression of β-actin was us- es, CA, USA), penicillin/streptomycin (1/100; ed for the normalization of the expression of Fisher, Waltham, MA, USA), 20 ng/mL basic the target genes. The relative mRNA concen- fibroblast growth factor (bFGF; eBiosciences, tration was determined according to Ct values. CA, USA), and glutamine (1/100; Corning, VA, All specimens were evaluated in triplicate. USA). Growth factors, such as EGF and bFGF, were supplemented twice a week. Cells were Western blotting (WB) separated and plated in DMEM/F12 containing 20% (v/v) fetal bovine serum (FBS) in prepara- Lysates containing total protein were acquired tion for culture. Accutase (Fisher, Waltham, MA, using the modified RIPA buffer (0.5% deoxycho- USA) was applied during cell separation for pas- late, 0.1% sodium dodecyl sulfate (SDS), 1% saging. This study has been approved by the Igepal CA-630, and Tris-HCl, pH 8.0), and SDS Ethics Committee of the West China Hospital, polyacrylamide gel electrophoresis was sub- Sichuan University. sequently performed. Overnight incubation wi- Bmi1 knockdown th primary antibodies was performed at 4°C. Thereafter, incubation was carried out with the Three lentivirus vector plasmids specific for tar- secondary antibodies for 1 h at room tem- get sequences contained in the Bmi1 shRNA perature. plasmid (Santa Cruz Biotechnology, Dallas, TX, USA) were used to knockdown Bmi1 using pu- BrdU incorporation assay romycin selection genes. The cells were trans- fected with the Bmi1 shRNA plasmid using After seven-days in culture, cells were incubat- media devoid of antibiotics or serum supple- ed for 4-6 h with 10 μmol of BrdU. Cells were ments. fixed with 4% paraformaldehyde (PFA; Sigma, St. Louis, MI, USA) and stained with an anti- MTS assay BrdU antibody (BD, Franklin Lakes, NJ, USA). Counterstaining was performed with Hoechst Cells from each distinct group were cultured in 33258 (Sigma, St. Louis, MI, USA). The number 96-well plates (2000 cells/well). MTS assay kit of cells positive for BrdU was recorded as the (Promega, Madison, WI, USA) was used to de- proportion of the total number of cells coun- termined cell viability. The assay was perfor- terstained with Hoechst 33258. A Nikon DS- med in triplicate. For Pax3 overexpression, Bmi1 knockdown cells were seeded in 12-well Ri1 microscope was used for observation. plates, and 24 h later, cells were transfected Procedures were carried out in triplicate. with vector or Pax3 plasmid. After 24 h, cells Soft agar assay were split into a 96-well plate. Thereafter, cell viability was determined using the MTS assay Ten thousand cells were separated and resus- kit according to the manufacturer’s instru- pended in SeaPlaque agar (0.7%). Cells were ctions. added to culture medium containing the gr- Pax3 and YAP overexpression owth factors twice a week for 3 weeks. Cells were stained with nitro blue tetrazolium (NBT; Plasmid vectors that overexpressed YAP and Sigma, St. Louis, MI, USA) and fixed with 4% Pax3 were obtained from Addgene. The Micro- PFA. Procedures were carried out in triplicate.

9391 Int J Clin Exp Med 2020;13(12):9390-9397 Inhibition of Bmi1 suppresses tumorigenicity in GSCs

Figure 1. Bmi1 is preferentially expressed in glioma stem cells (GSCs). The level of Bmi1 was determined by western blot (A) and qRT-PCR (B) in glioblastoma multiforme (GBM) cells treated with GSCs or 20% FBS media. (C) The mRNA expression levels of CD133, CD44, Sox2, and OCT4 was analyzed by qRT-PCR. *P<0.05; **P<0.01; ***P<0.001.

evaluate the differences bet- ween the groups. The differ- ence was regarded as signifi- cant with P<0.05.

Results

Expression of Bmi1 was pro- moted in GSCs

To examine the influence of Bmi1 on the preservation of the GSC state, we compared the expression of Bmi1 in SCs to that of non-SCs in malignant tissues. Cultures from freshly excised tumors were digested and plated in medium contain- ing growth factors or serum. This resulted in the derivation Figure 2. Bmi1 knockdown in GSCs. A. Cells were infected with lentiviruses and separation of matched gli- expressing shcon or shRNAs. The mRNA level was detected by qRT-PCR. B. Protein levels were determined by western blot. **P<0.01. oma cells of typical popula- tions of CSCs or cells after differentiation. WB was sub- Statistical analysis sequently performed to evaluate Bmi1 expres- sion in distinct cellular compartments. The pro- The results were reported as the mean ± SD. tein and mRNA levels of Bmi1 were evaluated Student’s t-test or ANOVA was performed to by GSC/differentiated cultivation with distinct

9392 Int J Clin Exp Med 2020;13(12):9390-9397 Inhibition of Bmi1 suppresses tumorigenicity in GSCs

Figure 3. Bmi1 knockdown confers proliferation defects in GSCs. A. MTS analysis of sh-con and sh-Bmi1 at the indicated time points. B. Representative images and quantified values from the BrdU analysis of the GSCs. Scale bar: 50 μm. *P<0.05; **P<0.01. concentrations of protein (Figure 1A and 1B). noticeable suppression of Bmi1 expression Bmi1 expression increased remarkably in GS- was shown via WB (Figure 2B). Cs compared to that in the differentiated cells (Figure 1A and 1B). In addition, the mRNA lev- Bmi1 knockdown in GSCs inhibited prolifera- tion els of CD133, CD44, Sox2, and OCT4 increased in the GSCs (Figure 1C). The MTS assay was used to quantify prolifera- tion. Bmi1 knockdown notably suppressed pro- Stable shRNA transfection reduced expression liferation (Figure 3A). We also carried out BrdU of Bmi1 incorporation assays to investigate the influ- ence of Bmi1 on the proliferation of GSCs. GSC We subsequently conducted qRT-PCR of GSCs proliferation was notably suppressed in BrdU- transfected with a shRNA plasmid specific for positive, Bmi1-knockdown cells (Figure 3B). Bmi1. Transfection with the shRNAs notably These findings indicated that the expression suppressed Bmi1 transcription (Figure 2A). A of Bmi1 is related for the proliferation of GSCs.

9393 Int J Clin Exp Med 2020;13(12):9390-9397 Inhibition of Bmi1 suppresses tumorigenicity in GSCs

knockdown cells as shown by the WB and RT-PCR results (Figure 4A and 4B). Further- more, excessive Pax3 expres- sion in Bmi1 knockdown cells attenuated the suppressive ef- fect of Bmi1 knockdown on proliferation (Figure 4C).

YAP modulated Bmi1-stimulat- ed Pax3 expression

We examined the participation of YAP in Pax3 regulation via Bmi1 in GSCs. In Bmi1 knock- down cells, suppression of Pa- x3 expression in GSCs that excessively expressed YAP was inhibited, as presented in Fi- gure 5.

Bmi1 knockdown suppressed tumorigenicity in GSCs

Colony formation was utilized to evaluate the anchorage-in- dependent growth of GSCs in which Bmi1 expression was knocked down. Infected GSCs, Figure 4. Bmi1 regulates Pax3 expression. A. The mRNA levels of Pax3 in suppressed colony growth (Fi- Bmi1 knockdown cells were analyzed by qRT-PCR. B. The protein level of gure 6A and 6B). The above Pax3 in Bmi1 knockdown cells was analyzed by western blot. C. The cell via- findings indicated that Bmi1 is bility of GSCs with Bmi1 knockdown and/or Pax3 overexpression. *P<0.05; **P<0.01. correlated to the development of tumorigenicity in GSCs.

Discussion

GSCs were first isolated from leukemic cells [25]. Reya et al. observed the expression of the same biomarkers in SCs and malignant cells. It was hypoth- esized that malignancies co- uld arise from a subgroup of precursor SCs [26]. SCs were isolated from various malig- Figure 5. YAP is involved in Bmi1-regulated Pax3 expression. Pax3 expres- nancies, such as prostate and sion in shcon and sh-Bmi1 GSCs with or without YAP transfection was ana- breast cancers [27]. Despite lyzed by western blot. the isolation of GSCs from glio- mas, our understanding of the Bmi1 knockdown inhibited Pax3 expression participation of GSCs in the development of malignancy is insufficient [28, 29]. Our resear- Our research subsequently examined the influ- ch aimed to explore the influence of GSCs on ence of Bmi1 knockdown on the expression of the hallmarks of malignancies. In this study, Pax3 in GSCs. Pax3 was suppressed in Bmi1 our findings demonstrate that Bmi1 is essenti-

9394 Int J Clin Exp Med 2020;13(12):9390-9397 Inhibition of Bmi1 suppresses tumorigenicity in GSCs

Figure 6. Bmi1 regulates the tumorigenicity of GSCs. The soft agar assay for GBM #1 (A) and GBM #2 (B). Bmi1 knockdown resulted in a significant decrease in colony formation. **P<0.01. al for CSC modulation in gliomas. GSCs were crest, and somites of the vertebrate embryos. transfected with shRNAs targeting Bmi1 to As differentiation progresses, expression decr- downregulate its expression. It was observed eases. In the early stages of embryogenesis, that Bmi1 transcription and translation were Pax3 is transiently expressed in the posterior noticeably suppressed in transfected GSCs. and dorsal neural tube. Pax3 is essential for Bmi1 knockdown inhibited the proliferation of the correct specification of these development GSCs. Moreover, Bmi1 knockdown resulted in tissues and cell survival, as homozygous dele- reduced Pax3 expression mediated by the YAP tion of Pax3 in mice results in severe neural pathway. As Pax3 induces proliferation, the crest and neural tube defects due to -de- above findings indicate that Bmi1 participates pendent cell death. Our results indicate Pax3 in the proliferation of GSCs through the YAP/ mediates the function of Bmi1 in CSCs. Pax3 axis. Various studies have examined the expression The polycomb group protein Bmi1 is overex- and activities of Bmi1 in CSCs [17, 30]. They pressed and acts as an in a variety have reported that Bmi1 deficiency results in of human tumors. The Hippo/YAP signaling the shortage of features related to CSCs [31]. pathway is a major integrator of cell surface- Our study is the first to directly evaluate the mediated signaling and regulates key process- influence of Bmi1 suppression on GSCs. Bmi1 es in development and tumorigenesis. Previ- stimulation is regarded as a trigger for the ous studies have reported Bmi1-mediated YAP development of malignancy [32, 33]. Studies stabilization and the active hippo/YAP signal- show that Bmi1 is a downstream modulator of ing pathway. In the current study, we elucidated in BC, acute myeloid leukemia, gas- the involvement of YAP in Bmi1-induced Pax3 tric carcinoma, and GBM [32, 34, 35]. upregulation. Pax3 is a developmental tran- scription factor that displays a regionalized The findings of our research indicate that Bmi1 expression pattern in the neural tube, neural plays a role in the functions of GSCs by enhanc-

9395 Int J Clin Exp Med 2020;13(12):9390-9397 Inhibition of Bmi1 suppresses tumorigenicity in GSCs ing the development and proliferation of malig- transcriptional activity in glioblastoma multi- nant cells. Targeting GSCs is important due forme. PLoS One 2014; 9: e86102. to its role in recurrence and the debilitating [7] Auffinger B, Spencer D, Pytel P, Ahmed AU and effects of GBM. The clear distinction between Lesniak MS. The role of glioma stem cells in chemotherapy resistance and glioblastoma SCs and non-SCs is necessary for the develop- multiforme recurrence. Expert Rev Neurother ment of GBM treatments. 2015; 15: 741-752. [8] Safari M and Khoshnevisan A. Cancer stem The findings of our research indicate that Bmi1 cells and chemoresistance in glioblastoma participates in the proliferation of glioma SCs multiform: a review article. J Stem Cells 2015; and potentially contributes to the selective tar- 10: 271-285. geting of the Bmi1 axis stimulation in GBM. [9] Safa AR, Saadatzadeh MR, Cohen-Gadol AA, Pollok KE and Bijangi-Vishehsaraei K. Glioblas- Acknowledgements toma stem cells (GSCs) epigenetic plasticity and interconversion between differentiated This work was supported by the Key R&D non-GSCs and GSCs. Genes Dis 2015; 2: 152- Project of Sichuan Provincial Science and Te- 163. chnology Department (grant number 2018FZ- [10] Yi Y, Hsieh IY, Huang X, Li J and Zhao W. Glio- 0055). blastoma stem-like cells: characteristics, mi- croenvironment, and therapy. Front Pharmacol Disclosure of conflict of interest 2016; 7: 477. [11] Kwon SJ, Kwon OS, Kim KT, Go YH, Yu SI, Lee None. BH, Miyoshi H, Oh E, Cho SJ and Cha HJ. Role of MEK partner-1 in cancer stemness through Address correspondence to: Ding Lei, Department MEK/ERK pathway in cancerous neural stem cells, expressing EGFRviii. Mol Cancer 2017; of Neurosurgery, West China Hospital, Sichuan 16: 140. University, No. 37, Guoxue Lane, Wuhou District, [12] Eskilsson E, Rosland GV, Talasila KM, Knappsk- Chengdu, Sichuan, China. Tel: +86-18980767530; og S, Keunen O, Sottoriva A, Foerster S, Solecki Fax: +86-028-85582944; E-mail: babyface1243@ G, Taxt T, Jirik R, Fritah S, Harter PN, Valk K, Al 126.com Hossain J, Joseph JV, Jahedi R, Saed HS, Pic- cirillo SG, Spiteri I, Leiss L, Euskirchen P, Gra- References ziani G, Daubon T, Lund-Johansen M, Enger PO, Winkler F, Ritter CA, Niclou SP, Watts C, [1] Hanif F, Muzaffar K, Perveen K, Malhi SM and Bjerkvig R and Miletic H. EGFRvIII mutations Simjee ShU. Glioblastoma multiforme: a review can emerge as late and heterogenous events of its epidemiology and pathogenesis through in glioblastoma development and promote an- clinical presentation and treatment. Asian Pac giogenesis through Src activation. Neuro Oncol J Cancer Prev 2017; 18: 3-9. 2016; 18: 1644-1655. [2] Noroxe DS, Poulsen HS and Lassen U. Hall- [13] Siddique HR and Saleem M. Role of BMI1, a marks of glioblastoma: a systematic review. stem cell factor, in cancer recurrence and che- ESMO Open 2016; 1: e000144. moresistance: preclinical and clinical evidenc- [3] Cannon GM, Tome WA, Robins HI and Howard es. Stem Cells 2012; 30: 372-378. SP. Pulsed reduced dose-rate radiotherapy: [14] Chen D, Wu M, Li Y, Chang I, Yuan Q, Ekimyan- case report: a novel re-treatment strategy in Salvo M, Deng P, Yu B, Yu Y, Dong J, Szymanski the management of recurrent glioblastoma JM, Ramadoss S, Li J and Wang CY. Targeting multiforme. J Neurooncol 2007; 83: 307-311. BMI1(+) cancer stem cells overcomes chemo- [4] Reitman ZJ, Winkler F and Elia AEH. New direc- resistance and inhibits metastases in squa- tions in the treatment of glioblastoma. Semin mous cell carcinoma. Cell Stem Cell 2017; 20: Neurol 2018; 38: 50-61. 621-634, e626. [5] Stoyanov GS, Dzhenkov D, Ghenev P, Iliev B, [15] Paranjape AN, Balaji SA, Mandal T, Krushik EV, Enchev Y and Tonchev AB. Cell biology of glio- Nagaraj P, Mukherjee G and Rangarajan A. blastoma multiforme: from basic science to Bmi1 regulates self-renewal and epithelial to diagnosis and treatment. Med Oncol 2018; mesenchymal transition in breast cancer cells 35: 27. through Nanog. BMC Cancer 2014; 14: 785. [6] Wang E, Zhang C, Polavaram N, Liu F, Wu G, [16] Wang MC, Jiao M, Wu T, Jing L, Cui J, Guo H, Schroeder MA, Lau JS, Mukhopadhyay D, Jiang Tian T, Ruan ZP, Wei YC, Jiang LL, Sun HF, SW, O’Neill BP, Datta K and Li J. The role of fac- Huang LX, Nan KJ and Li CL. Polycomb com- tor inhibiting HIF (FIH-1) in inhibiting HIF-1 plex protein BMI-1 promotes invasion and me-

9396 Int J Clin Exp Med 2020;13(12):9390-9397 Inhibition of Bmi1 suppresses tumorigenicity in GSCs

tastasis of stem cells by ac- [26] Reya T, Morrison SJ, Clarke MF and Weissman tivating PI3K/AKT signaling, an ex vivo, in vitro, IL. Stem cells, cancer, and cancer stem cells. and in vivo study. Oncotarget 2016; 7: 9586- Nature 2001; 414: 105-111. 9599. [27] Mastelic A, Cikes Culic V, Rezic Muzinic N, Vui- [17] Srinivasan M, Bharali DJ, Sudha T, Khedr M, ca-Ross M, Barker D, Leung EY, Reynisson J Guest I, Sell S, Glinsky GV and Mousa SA. and Markotic A. Glycophenotype of breast and Downregulation of Bmi1 in breast cancer stem stem cells treated with cells suppresses tumor growth and prolifera- thieno[2,3-b]pyridine anticancer compound. tion. Oncotarget 2017; 8: 38731-38742. Drug Des Devel Ther 2017; 11: 759-769. [18] Wang Y, Zhe H, Ding Z, Gao P, Zhang N and Li [28] Ahmed AU, Auffinger B and Lesniak MS. Under- G. marker Bmi-1 expression standing glioma stem cells: rationale, clinical is associated with basal-like phenotype and relevance and therapeutic strategies. Expert poor survival in breast cancer. World J Surg Rev Neurother 2013; 13: 545-555. 2012; 36: 1189-1194. [29] Gersey ZC, Rodriguez GA, Barbarite E, Sanchez [19] Arnes JB, Collett K and Akslen LA. Independent A, Walters WM, Ohaeto KC, Komotar RJ and prognostic value of the basal-like phenotype of Graham RM. Curcumin decreases malignant breast cancer and associations with EGFR and characteristics of glioblastoma stem cells via candidate stem cell marker BMI-1. Histopa- induction of reactive oxygen species. BMC thology 2008; 52: 370-380. Cancer 2017; 17: 99. [20] Navet S, Buresi A, Baratte S, Andouche A, Bon- [30] Proctor E, Waghray M, Lee CJ, Heidt DG, Yala- naud-Ponticelli L and Bassaglia Y. The Pax manchili M, Li C, Bednar F and Simeone DM. family: highlights from cephalopods. Bmi1 enhances tumorigenicity and cancer PLoS One 2017; 12: e0172719. stem cell function in pancreatic adenocarcino- [21] Autilio C, Paolillo C, Lavieri MM, Pocino K, De ma. PLoS One 2013; 8: e55820. Paolis E, Di Stasio E, Marchetti P, Gian Carlo CA [31] Wang MC, Li CL, Cui J, Jiao M, Wu T, Jing LI and and Capoluongo E. PAX3d mRNA over 2.76 Nan KJ. BMI-1, a promising therapeutic target copies/microL in the bloodstream predicts cu- for human cancer. Oncol Lett 2015; 10: 583- taneous malignant melanoma relapse. Onco- 588. target 2017; 8: 85479-85491. [32] Zhang R, Wu WR, Shi XD, Xu LB, Zhu MS, Zeng [22] Kubic JD, Little EC, Kaiser RS, Young KP and H and Liu C. Dysregulation of Bmi1 promotes Lang D. FOXD3 promotes PAX3 expression in malignant transformation of hepatic progeni- melanoma cells. J Cell Biochem 2016; 117: tor cells. Oncogenesis 2016; 5: e203. 533-541. [33] Sahasrabuddhe AA. BMI1: a biomarker of he- [23] Su X, Liu X, Ni L, Shi W, Zhu H, Shi J, Chen J, Gu matologic malignancies. Biomark Cancer Z, Gao Y, Lan Q and Huang Q. GFAP expression 2016; 8: 65-75. is regulated by Pax3 in brain glioma stem cells. [34] Peng HX, Liu XD, Luo ZY, Zhang XH, Luo XQ, Oncol Rep 2016; 36: 1277-1284. Chen X, Jiang H and Xu L. Upregulation of the [24] Liu Y, Zhu H, Liu M, Du J, Qian Y, Wang Y, Ding proto-oncogene Bmi-1 predicts a poor progno- F and Gu X. Downregulation of Pax3 expres- sis in pediatric acute lymphoblastic leukemia. sion correlates with acquired GFAP expression BMC Cancer 2017; 17: 76. during NSC differentiation towards astrocytes. [35] Jacobs JJ, Kieboom K, Marino S, DePinho RA FEBS Lett 2011; 585: 1014-1020. and van Lohuizen M. The oncogene and Poly- [25] Liebelt BD, Shingu T, Zhou X, Ren J, Shin SA comb-group gene bmi-1 regulates cell prolifer- and Hu J. Glioma stem cells: signaling, micro- ation and senescence through the ink4a locus. environment, and therapy. Stem Cells Int Nature 1999; 397: 164-168. 2016; 2016: 7849890.

9397 Int J Clin Exp Med 2020;13(12):9390-9397