Snail homolog 1 is involved in epithelial-mesenchymal transition-like processes in human glioblastoma cells

  • Authors:
    • Caspar D. Kühnöl
    • Carina Würfel
    • Martin S. Staege
    • Christof Kramm
  • View Affiliations

  • Published online on: March 17, 2017     https://doi.org/10.3892/ol.2017.5875
  • Pages: 3882-3888
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Despite advancements in neurosurgery, chemotherapy and radiation therapy, the outcome of patients with glioblastoma remains poor. The migration of tumor cells from the primary tumor site with subsequent invasion of these cells into the surrounding normal brain tissue is frequently responsible for relapse and treatment failure. The present study hypothesized that snail homolog 1 (SNAI1), a factor critically involved in the epithelial‑mesenchymal transition (EMT) of human carcinoma cells, may also contribute to an invasive EMT‑like phenotype of glioblastoma cells. The majority of glioblastoma cell lines investigated in the present study expressed SNAI1 at basal levels. The present study overexpressed SNAI1 in glioblastoma cell lines by lentiviral transfer of human SNAI1 complementary DNA. In addition, the inhibition of SNAI1 expression was achieved by lentiviral transfer of a short hairpin RNA specific for SNAI1. SNAI1 overexpression increased proliferation of one of the cell lines, U251MG, but exhibited only a weak effect on the migration and invasion of glioblastoma cells. However, downregulation of SNAI1 significantly decreased the invasive capacity of all investigated cell lines. In parallel, regained expression of E‑cadherin, a marker that is usually lost during EMT, was observed subsequent to SNAI1 knockdown in the glioblastoma cell lines U87MG and U251MG. The data of the present study suggest that certain key genes of the EMT in carcinoma are also involved in the migration and invasion of human glioblastoma cells.

Introduction

Approximately 1–2% of neoplastic diseases are located in the central nervous system (1). The majority of these tumors, ~50–60%, are gliomas that develop more often in Caucasian patients compared with those of Asian or African origin (2). The incidence of gliomas is 5–11/100,000 in Western Europe, Australia and Northern America (3). The majority of patients are older than 50 years, and males are more often affected than females (4). In Germany, every year ~65 children are diagnosed with high-grade glioma (HGG) (5). Despite advancements in neurosurgery, chemotherapy and radiation therapy, the outcome remains poor, with a median survival subsequent to diagnosis of only 14 months (6). Glioblastoma multiforme is the most common type of HGG, accounting for ~50% of cases (7). The tumors may be located in all parts of the brain, but are mostly located supratentorially within the cerebral lobes (8).

The histogenetic origin of glioma cells is not clear yet. Since the beginning of the 20th century, it has been assumed that the tumor cells develop from precursor glial cells (9,10). An association with neuroectodermal stem cells appears to be possible (1).

For glioblastoma, an association between clinical characteristics, histology and genetics may be observed (11,12). The malignant progression is associated with an abnormal expression of proto-oncogenes and tumor-suppressor genes (7,1315).

The poor outcome following treatment of glioblastoma is likely associated with the invasive capability of the tumor cells. Even subsequent to gross total tumor resection, vital tumor cells may have already migrated into the surrounding brain tissue, unnoticed during surgical intervention. The infiltrative growth of glioblastomas is mediated by the interplay of cell invasion, cell motility and the interaction between matrix proteins and growth factors (16,17). However, models for glioma cell migration and invasion are missing. Thus, the adaptation of models from other types of tumor disease may be a promising method. In carcinoma, the change from a local carcinoma into an infiltrative and metastasizing tumor is described by the model of epithelial-mesenchymal transition (EMT) (18). Under certain circumstances, epithelial cells may adopt a mesenchymal phenotype (1922). This change is marked by the replacement of E-cadherin (CDH1) by N-cadherin. Another important protein in this process is vimentin (VIM), which allows the shape of tumor cells to change, thus enabling these cells to move into the extracellular matrix (23). Notably, not all cells undergoing EMT exhibit all the EMT-specific features. Some tumors exhibit only certain EMT-typical changes or opposed phenotypical changes in terms of mesenchymal-epithelial transition (2426).

Several EMT inducers have been described, including transforming growth factor-β (TGF-β), snail homologs 1 and 2 (SNAI1/SNAI2), twist homolog 1 (TWIST1) and wingless-type mouse mammary tumor virus integration site family members (WNT) (1933). The progression of tumors may also be part of EMT programs (34). The effect of TWIST1, a gene involved in EMT programs in carcinoma, on the invasion level of glioblastoma was previously described (35). A central role of the nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) pathway in the induction of EMT has been revealed in different tumor models (36).

The present study focused on the effect of the EMT-inducer SNAI1 on the proliferation, migration and invasion of human glioblastoma cells. SNAI1 belongs to the family of snail genes. The members of the snail superfamily encode for zinc finger transcriptions factors and serve an important role during the development of the mesoderm and transcriptional-repressor functions (3740). Another function of SNAI1 is its role in cell death and cell survival programs (41,42). SNAI1, as a key EMT-associated gene, is involved in several pathways, including the TGF-β, WNT, NOTCH and endothelin receptor A pathways (39,42). In addition, the direct inhibition of expression of CDH1, the hallmark of EMT, is mediated by members of the snail superfamily (43).

Materials and methods

Cell culture

The human glioblastoma U87MG (44), T98G (45), LN-18 (46) and U251MG (47) cell lines were donated by Professor M. Hegi (Department of Clinical Neuroscience, University of Lausanne, Lausanne, Switzerland) and Dr W. Maes (Laboratory for Thrombosis Research, Interdisciplinary Research Facility Life Sciences Kulak, Kortrijk, Belgium), and the human embryonal kidney (HEK) 293T cell line were obtained from the American Type Culture Collection (Manassas, VA. Cells were grown in a humidified, 5% CO2 atmosphere at 37°C in Dulbecco's modified Eagle's medium (DMEM; PAA Laboratories; GE Healthcare Life Sciences, Chalfont, UK) with 10% fetal calf serum (FCS; Biochrom GmbH, Berlin, Germany), 100 U/ml penicillin and 100 µg/ml streptomycin (PAA Laboratories; GE Healthcare Life Sciences) in cell culture flasks. Twice a week, the cells were washed twice with PBS and detached with a trypsin/EDTA solution (0.05% trypsin and 0.02% EDTA in PBS; PAA Laboratories; GE Healthcare Life Sciences). Subsequent to centrifugation at 350 × g for 7 min, the cells were suspended in DMEM at a ratio of 1:10 in new cell culture flasks.

Overexpression of SNAI1 in tumor cell lines

RNA was isolated using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA). The complementary DNA (cDNA) of SNAI1 from the cell line T98G was amplified by reverse transcription-polymerase chain reaction (RT-PCR) using the following PCR primers: 5′-GTTCTTCTGCGCTACTGCTG-3′ (forward) and 5′-GCAGGTATGGAGAGGAAGAGG-3′ (reverse). A total of 2 µl cDNA was mixed with 2.5 µl 10X buffer (Promega Corporation, Madison, WI, USA), 1.5 µl MgCl2 (25 mM), 0.2 µl Taq polymerase (Promega Corporation), 0.5 µl dNTP mix (10 mM; Fermentas; Thermo Fisher Scientific, Inc., Pittsburgh, PA, USA), 0.25 µl sequence-specific primers (Invitrogen; Thermo Fisher Scientific, Inc.), 2.5 µl dimethylsulfoxide and 15.3 µl H2O. The thermocycling conditions were as follows: 95°C for 5 min; 35 cycles of 94°C for 30 sec, 60°C for 35 sec and 72°C for 45 sec; followed by 72°C for 5 min and an infinite holding temperature of 4°C. The PCR product was cloned into the vector pGEM-T Easy (Promega Corporation). From this vector, SNAI1 was cloned into the lentiviral vector puc2CL6IN, donated by Professor H. Hanenberg (Clinic of Pediatrics and Adolescent Medicine III, Essen University Hospital, Essen, Germany), containing a neomycin resistance cassette. Sequencing of the vector was performed as previously described (48). For lentiviral vector production, the envelope plasmid pczVSV-G and helper plasmid pCD/NL-BH, both donated by Professor H. Hanenberg, were co-transfected with puc2CL6IN-SNAI1 or the control vector puc2CL6IN into HEK293T cells. The resulting lentivirus-containing supernatants were used to infect U87MG, T98G, LN-18 and U251MG glioblastoma cells. Three independent transductions were performed. Selection of transduced cells was performed with 1 mg/ml G418 (PAA Laboratories; GE Healthcare Life Sciences).

Knockdown of SNAI1 in tumor cell lines

Potential short hairpin RNA sequences for knockdown of SNAI1 in tumor cells were identified using the BLOCK-iT™ RNAi Designer (Invitrogen; Thermo Fisher Scientific, Inc.). The two oligonucleotides 5′-CGCGTCCCCGCTGCAGGACTCTAATCCATTCAAGAGATGGATTAGAGTCCTGCAGCTTTTTGGAAATT-3′ and 5′-AGGGGCGACGTCCTGAGATTAGGTAAGTTCTCTACCTAATCTCAGGACGTCGAAAAACCTTTAGC-3′ were annealed and cloned in the enhanced green fluorescent protein (EGFP)-containing lentiviral vector pCL2.THPC, donated by Professor H. Hanenberg. The production of the lentiviral vectors, infection and selection of lentivirally transduced U87MG, T98G, LN-18 and U251MG cell lines was performed as aforementioned. A total of three independent transductions were performed.

RT-quantitative (q)PCR

Total RNA was isolated from glioblastoma cell lines using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.). First strand cDNA synthesis was performed with 2 µg total RNA using oligo-(dT)12–18 primers (Fermentas; Thermo Fisher Scientific, Inc.). RT was carried out with Moloney Murine Leukemia Virus Reverse Transcriptase (Fermentas; Thermo Fisher Scientific, Inc.), for 1 h at 37°C. A total of 2 µl cDNA was amplified by PCR [25 (β-actin) or 35 (other targets) cycles of 94°C for 30 sec, 60°C for 35 sec and 72°C for 45 sec] using Taq polymerase (Promega Corporation) in a final volume of 25 µl in a Mastercycler personal (Eppendorf, Hamburg, Germany). All primers were purchased from Invitrogen; Thermo Fisher Scientific, Inc. The following primers were used: β-actin (ACTB) forward, 5′-GGCATCGTGATGGACTCCG-3′ and reverse, 5′-GCTGGAAGGTGGACAGCGA-3′; CDH1 forward, 5′-GCTGGAGATTAATCCGGACA-3′ and reverse, 5′-ACCCACCTCTAAGGCCATCT-3′; EGFP forward, 5′-ACGTAAACGCCCACAAGTTC-3′ and reverse, 5′-AAGTCGTGCTGCTTCATGTG-3′; lymphoid enhancer-binding factor 1 (LEF1) forward, 5′-AGCACTTTTCTCCAGGGTCA-3′ and reverse, 5′-CCCGTGATGGGATATACAGG-3′; neomycin phosphotransferase II forward, 5′-AGACAATCGGCTGCTCTGAT-3′ and reverse, 5′-AGTGACAACGTCGAGCACAG-3′; NF-κB1 forward, 5′-CACCTAGCTGCCAAAGAAGG-3′ and reverse, 5′-TCAGCCAGCTGTTTCATGTC-3′; SNAI1 forward, 5′-ACCCCACATCCTTCTCACTG-3′ and reverse, 5′-CCGACAAGTGACAGCCAT-3′; and TWIST2 forward, 5′-CGAGGAGGAGCTCGAGAGG-3′ and reverse, 5′-CTAGTGGGAGGCGGACAT-3′. RT-qPCR was performed using the Maxima SYBR-Green qPCR Master Mix kit (Fermentas; Thermo Fisher Scientific, Inc.) using the following conditions: Denaturation, 94°C for 45 sec; annealing, 62°C for 45 sec; and elongation 72°C for 60 sec. Each reaction was subjected to melting temperature analysis to confirm the presence of the expected products. Specific gene amplification was normalized to ACTB. Target genes and ACTB were amplified with 40 cycles using a Rotor Gene RG-3000 (Corbett Life Science; Qiagen GmbH, Hilden, Germany) and Rotor Gene 6 software version 6.1 (build 93; Corbett Life Science; Qiagen GmbH). Relative expression values were calculated using the 2−ΔΔCq method (49).

Cell proliferation assay

Analysis of cell proliferation was performed with the MTT Cell Proliferation kit (Roche Applied Science, Mannheim, Germany) according to the protocol of the manufacturer. The cells were seeded at a density of 1×103 cells/well in 96-well plates for 72 h in a humidified, 5% CO2 atmosphere at 37°C. Turnover of MTT was detected using an ELx808 microplate reader (BioTek Instruments, Inc., Winooski, VT, USA).

Cell migration assay

To analyze tumor cell migration, ThinCert cell culture inserts with a pore size of 8.0 µm (Greiner Bio-One GmbH, Frickenhausen, Germany) were used. The tumor cells were washed twice with PBS, suspended in serum-free medium containing 0.2% bovine serum albumin (BSA; SERVA Electrophoresis GmbH, Heidelberg, Germany) and cultured in cell culture flasks overnight in a humidified, 5% CO2 atmosphere at 37°C. On the next day, the wells of a 24-well plate were filled with 600 µl medium (DMEM with 10% FCS, 100 U/ml penicillin and 100 µg/ml streptomycin), and a ThinCert insert was placed in every well. Subsequently, 2×105 tumor cells in 200 µl serum-free medium (DMEM with 0.2% BSA) were placed in the inserts. After 24 h, the ThinCert inserts containing the cells were removed and placed in new wells of a 24-well plate containing 500 µl trypsin/EDTA solution. The corresponding cell numbers were subsequently determined using a Neubauer chamber following staining with trypan blue (Invitrogen; Thermo Fisher Scientific, Inc.).

Matrigel invasion assay

To investigate the invasion capacity of the different glioblastoma cell lines, a Matrigel invasion chamber (BD Biosciences, Franklin Lakes, NJ, USA) was used. The cell lines were harvested and resuspended in DMEM containing 0.1% BSA at a concentration of 5×104 cells/ml. Matrigel invasion and BD BioCoat Control inserts (BD Biosciences) were filled with 0.5 ml DMEM pre-warmed to 37°C containing 0.1% BSA. After 2 h the medium was removed and the inserts were transferred into 24-well plates containing DMEM with 10% FCS as a chemoattractant. Immediately thereafter, 0.5 ml cell suspension was added to the inside of the insert. The Matrigel invasion and control chambers were incubated for 24 h in a humidified, 5% CO2 atmosphere at 37°C. Inserts with invaded cells were stained with Diff-Quick (Medion Grifols Diagnostics AG, Düdingen, Switzerland). To determine the number of invaded cells, membranes were cut and placed on a microscope slide with a drop of immersion oil, and the cells were counted using an Axiolab light microscope (Zeiss GmbH, Jena, Germany). To calculate the percentage of invading cells, the mean number of cells invading into the Matrigel insert membrane was divided by the mean number of cells migrating through the control inserts and multiplied by 100.

Statistical analysis

Statistical analysis was performed using Microsoft Excel 2010 (version 14.0.7128.5000; Microsoft Corporation, Redmont, WA, USA), Student's t-test was used to analyze differences. P<0.05 was considered to indicate a statistically significant difference.

Results

Modulation of EMT-associated genes by overexpression of SNAI1 in glioblastoma cells

To investigate the effect of SNAI1 on glioblastoma cells, glioblastoma U87MG, T98G, LN-18 and U251MG cell lines were transduced with lentiviral vectors allowing the overexpression or inhibition of SNAI1. Neither SNAI1 overexpression nor inhibition changed the microscopic appearance (size and shape) of the cells. SNAI1 overexpression was confirmed by RT-qPCR analysis (Fig. 1). In all used glioma cell lines, the expression of SNAI increased following transduction. A significantly (P<0.05) increased proliferation rate of SNAI1-overexpressing U251MG cells compared with the vehicle control (Fig. 2) was observed. The three other cell lines exhibited no significant proliferative changes upon SNAI1 overexpression. Furthermore, none of the cell lines exhibited significant changes on their migration rate, although there was a tendency towards a relative higher migration rate in the U87MG, T98G and LN-18 cell lines (Fig. 3A). Similarly, no significant differences in the invasion rate were detectable when comparing each cell type to the control (Fig. 3B).

The present study investigated whether EMT target genes may be affected by the overexpression of SNAI1 in glioblastoma cells. The regulation of TWIST2 in three of four glioma cell lines, U87MG, T98G and U251MG, upon SNAI1 overexpression (Fig. 4) was observed. In addition, RT-PCR revealed an increased expression of NF-κB1 in SNAI1-overexpressing U87MG and T98G cells. On the contrary, the overexpression of SNAI1 resulted in the decreased expression of NF-κB1 in LN-18 cells and U251MG cells. LEF1 exhibited low expression in untreated U87MG, LN-18 and T98G cells. An increased LEF1 expression was also detected in T98G and U251MG cells upon SNAI1 overexpression (Fig. 4). No differences were observed for VIM, which was stably detectable in U251MG, T98G and U87MG cells. CDH1 was absent from all cell lines independent from SNAI1 transduction (data not shown; see also below; Fig. 5A).

Re-induction of CDH1 subsequent to knockdown of SNAI1 in glioblastoma cells

Upon SNAI1 inhibition, as confirmed by RT-PCR analysis (Fig. 5A), a significantly higher proliferation rate in the cell line T98G was observed. In all the other cell lines, no significant effect on cellular proliferation was observed (Fig. 5B). Loss of CDH1 is a central step during EMT, accompanying the establishment of a highly infiltrative and metastasizing tumor phenotype (50). Thus, it was notable to observe that two out of four cell lines (U87MG and U251MG; Fig. 5A) regained CDH1 expression upon inhibition of SNAI1. No changes in the expression of VIM were observed (data not shown). The migration rate appeared relatively decreased compared with the mock transfected cells in the cell lines T98G, LN-18 and U251MG, but the differences did not reach significance (Fig. 6A). In U87MG cells, the migration rate was enhanced. However, the invasion capability significantly decreased in all four human glioma cell lines subsequent to SNAI1 inhibition (Fig. 6B).

Discussion

EMT represents a well-established model explaining the switch from a local into an infiltrating and metastasizing carcinoma phenotype on a signal transduction level (50). The present study investigated if this model may also be adaptable to human glioblastoma cells, which are characterized by extensively infiltrating tumor cells (16,17).

A key gene of EMT is SNAI, which is involved in different signaling pathways that downregulate CDH1 expression as a hallmark of EMT (50). A baseline SNAI1 expression in all investigated four human glioma cell lines, U87MG, T98G, U251MG and LN-18, was observed in the present study. High SNAI1 baseline expression may contribute to the infiltrative and migratory phenotype of glioblastoma cells, similar to EMT-associated functions of SNAI in carcinoma cells (51). Growth inhibition of U87MG cells subsequent to knockdown of SNAI1 has been described (37). The present study did not observe inhibition of proliferation subsequent to SNAI1 knockdown in any of the investigated cell lines. This divergence may be a consequence of experimental differences. For instance, viral transduction was used for genetic engineering, whereas in the study by Han et al (37), transfection of small interfering RNA was performed. In addition, proliferation was not investigated directly in the present study. Han et al (37) used a viability assay that depends on an intact respiratory chain, whereas the present study used an assay that depends on intact glycolysis. The effect of SNAI1 on the proliferation of glioma cells requires additional investigation.

The SNAI baseline expression may also be responsible for the observation of the present study that the additional increase in SNAI expression subsequent to lentiviral gene transfer did not result in significantly increased cell migration or invasion in any investigated cell line, although there was at least a tendency towards increased cell migration subsequent to SNAI overexpression in the majority of the cell lines.

RT-PCR analyses of EMT target genes in the four glioblastoma cell lines also exhibited heterogeneous results. Mock transfected cell lines displayed no baseline expression of TWIST2, but the overexpression of SNAI1 induced a high expression of TWIST2 in three of four cell lines. TWIST2 is known as a direct inducer of EMT (52,53). In addition, there is evidence that TWIST1 may act as a potential oncogene in gliomas exhibiting an increased expression of TWIST1 during the transformation from low-grade glioma to HGG (54). There appear to be different signaling pathways involved in TWIST activation upon the induction of SNAI expression in the various glioblastoma cell models used in the present study. In U87MG and T98G cells, the activation of TGF-β-dependent signaling pathways appeared to be involved, as these cells exhibited increased NF-κB1 expression upon induction of SNAI expression; NF-κB1 is one of the key genes of the TGF-β pathway, but may also be involved in other pathways (55). However, in LN-18 and U251MG cells, NF-κB1-associated signaling did not appear to be driving TWIST expression subsequent to SNAI gene transfer. In these cell lines, NF-κB1 expression decreased subsequent to the overexpression of SNAI1.

In T98G and U251MG cells, the WNT signaling pathway appears to be activated subsequent to SNAI gene transfer. In the present study, increased expression of TWIST and LEF1 as target genes of the canonical WNT pathway was observed. However, the present study did not additionally corroborate if the activation of WNT signaling led to TWIST expression or vice versa. The involvement of WNT and TWIST in EMT is well known, and it has been shown that TWIST may activate canonical WNT signaling in carcinoma (56).

However, the involvement of SNAI1 in EMT-like pathways appeared much clearer following inhibition of SNAI expression compared with overexpression of SNAI1. Subsequent to the inhibition of SNAI1 by lentiviral knockdown, re-expression of CDH1 in U87MG and U251MG cells was observed. Since in carcinoma cells the downregulation of CDH1 expression represents the hallmark of EMT induction, the finding of the re-induction of CDH1 expression subsequent to SNAI inhibition may provide indirect evidence that there may be an EMT-like phenotype in human glioblastoma. CDH1 was not expressed in wild-type or SNAI1-overexpressing glioblastoma cells, but was induced in half of the cell lines subsequent to the knockdown of SNAI1. This observation is in agreement with the significantly lower cellular invasion capabilities observed subsequent to the inhibition of SNAI expression.

Cell lines are suitable models for glioblastoma research in vitro (57,58). However, cell culture-dependent effects may also be observed with respect to the behavior of tumor cells, and certain widely used tumor cell lines do not exhibit the typical glioblastoma growth pattern in xenotransplantation models (59). Therefore, the analysis of SNAI1 and interacting factors in patient-derived material is required.

Taken together, the data of the present study may indicate that EMT-like processes similar to those in carcinoma may also occur in human glioblastoma, thereby potentially contributing to their aggressive and invasive phenotype. However, systematic investigation of the key EMT genes and associated signaling pathways is required to obtain a more comprehensive overview, as the signaling network may be the same as that in carcinoma or there may be differences. In addition, an analysis of the involved factors at the protein level will increase the level of information about the relevant pathways.

Acknowledgements

The authors would like to thank Professor Monika Hegi (Department of Clinical Neuroscience, University of Lausanne, Lausanne, Switzerland) and Dr Wim Maes (Laboratory for Thrombosis Research, Interdisciplinary Research Facility Life Sciences Kulak, Kortrijk, Belgium) for providing the glioblastoma cell lines, as well as Professor Helmut Hanenberg (Clinic of Pediatrics and Adolescent Medicine III, Essen University Hospital, Essen, Germany) for providing the vectors pczVSV-G, pCD/NL-BH, pCL2.THPC, pcl2.THPC and puc2CL6IN.

References

1 

Kleihues P, Louis DN, Scheithauer BW, Rorke LB, Reifenberger G, Burger PC and Cavenee WK: The WHO classification of tumours of the nervous system. J Neuropathol Exp Neurol. 61:215–225. 2002. View Article : Google Scholar : PubMed/NCBI

2 

Ohgaki H and Kleihues P: Epidemiology and etiology of gliomas. Acta Neuropathol. 109:93–108. 2005. View Article : Google Scholar : PubMed/NCBI

3 

Legler JM, Ries LA, Smith MA, Warren JL, Heineman EF, Kaplan RS and Linet MS: Cancer surveillance series (corrected): Brain and other central nervous system cancers: Recent trends in incidence and mortality. J Natl Cancer Inst. 91:1382–1390. 1999. View Article : Google Scholar : PubMed/NCBI

4 

Schwartzbaum JA, Fisher JL, Aldape KD and Wrensch M: Epidemiology and molecular pathology of glioma. Nat Clin Pract Neurol. 2:494–503. 2006. View Article : Google Scholar : PubMed/NCBI

5 

Kramm C, Rausche U, Butenhoff S, Kühnöl C, Kunze C, Kortmann R, Wolff J and van Gool S: Hochmaligne Gliome im Kindes-und Jugendalter. Monatschr Kinderheilkd. 156:1201–1207. 2008. View Article : Google Scholar

6 

Stupp R and Weber DC: The role of radio- and chemotherapy in glioblastoma. Onkologie. 28:315–317. 2005.PubMed/NCBI

7 

Ohgaki H and Kleihues P: Population-based studies on incidence, survival rates and genetic alterations in astrocytic and oligodendroglial gliomas. J Neuropathol Exp Neurol. 64:479–489. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Larjavaara S, Mäntylä R, Salminen T, Haapasalo H, Raitanen J, Jääskeläinen J and Auvinen A: Incidence of gliomas by anatomic location. Neuro Oncol. 9:319–325. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Smith MT, Ludwig CL, Godfrey AD and Armbrustmacher VW: Grading of oligodendrogliomas. Cancer. 52:2107–2114. 1983. View Article : Google Scholar : PubMed/NCBI

10 

Bailey P and Cushing H: Microchemical color reactions as an Aid to the Identification and classification of brain tumours. Proc Natl Acad Sci USA. 11:82–84. 1925. View Article : Google Scholar : PubMed/NCBI

11 

Collins VP: Gene amplification in human gliomas. Glia. 15:289–296. 1995. View Article : Google Scholar : PubMed/NCBI

12 

Dirks PB and Rutka JT: Current concepts in neuro-oncology: The cell cycle - a review. Neurosurgery. 40:1000–1015. 1997. View Article : Google Scholar

13 

Ohgaki H, Dessen P, Jourde B, Horstmann S, Nishikawa T, Di Patre PL, Burkhard C, Schüler D, Probst-Hensch NM, Maiorka PC, et al: Genetic pathways to glioblastoma: A population-based study. Cancer Res. 64:6892–6899. 2004. View Article : Google Scholar : PubMed/NCBI

14 

Schmidt MC, Antweiler S, Urban N, Mueller W, Kuklik A, Meyer-Puttlitz B, Wiestler OD, Louis DN, Fimmers R and von Deimling A: Impact of genotype and morphology on the prognosis of glioblastoma. J Neuropathol Exp Neurol. 61:321–328. 2002. View Article : Google Scholar : PubMed/NCBI

15 

Nozaki M, Tada M, Kobayashi H, Zhang CL, Sawamura Y, Abe H, Ishii N and Van Meir EG: Roles of the functional loss of p53 and other genes in astrocytoma tumorigenesis and progression. Neuro Oncol. 1:124–137. 1999. View Article : Google Scholar : PubMed/NCBI

16 

Rao JS: Molecular mechanisms of glioma invasiveness: The role of proteases. Nat Rev Cancer. 3:489–501. 2003. View Article : Google Scholar : PubMed/NCBI

17 

Maria BL, Eskin TA and Quisling RG: Brainstem and other malignant gliomas: II. Possible mechanisms of brain infiltration by tumour cells. J Child Neurol. 8:292–305. 1993. View Article : Google Scholar : PubMed/NCBI

18 

Micalizzi DS, Farabaugh SM and Ford HL: Epithelial-mesenchymal transition in cancer: Parallels between normal development and tumour progression. J Mammary Gland Biol Neoplasia. 15:117–134. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Townsend TA, Wrana JL, Davis GE and Barnett JV: Transforming growth factor-beta-stimulated endocardial cell transformation is dependent on Par6c regulation of RhoA. J Biol Chem. 283:13834–13841. 2008. View Article : Google Scholar : PubMed/NCBI

20 

Greenburg G and Hay ED: Epithelia suspended in collagen gels can lose polarity and express characteristics of migrating mesenchymal cells. J Cell Biol. 95:333–339. 1982. View Article : Google Scholar : PubMed/NCBI

21 

Yang J and Weinberg RA: Epithelial-mesenchymal transition: At the crossroads of development and tumour metastasis. Dev Cell. 14:818–829. 2008. View Article : Google Scholar : PubMed/NCBI

22 

Savanger P: Leaving the neighbourhood: Molecular mechanisms involved during epithelial-mesenchymal transition. Bioessays. 23:912–923. 2001. View Article : Google Scholar : PubMed/NCBI

23 

Mendez MG, Kojima S and Goldman RD: Vimentin induces changes in cell shape, motility and adhesion during the epithelial to mesenchymal transition. FASEB J. 24:1838–1851. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Yilmaz M and Christofori G: EMT, the cytoskeleton, and cancer cell invasion. Cancer Metastasis Rev. 28:15–33. 2009. View Article : Google Scholar : PubMed/NCBI

25 

Boyer B, Vallès AM and Edme N: Induction and regulation of epithelial-mesenchymal transition. Biochem Pharmacol. 60:1091–1099. 2000. View Article : Google Scholar : PubMed/NCBI

26 

Chaffer CL, Thompson EW and Williams ED: Mesenchymal to epithelial transition in development and disease. Cells Tissues Organs. 185:7–19. 2007. View Article : Google Scholar : PubMed/NCBI

27 

Ozdamar B, Bose R, Barrios-Rodiles M, Wang HR, Zhang Y and Wrana JL: Regulation of the polarity protein Par6 by TGFbeta receptors controls epithelial cell plasticity. Science. 307:1603–1609. 2005. View Article : Google Scholar : PubMed/NCBI

28 

Ikenouchi J, Matsuda M, Furuse M and Tsukita S: Regulation of tight junctions during the epithelium-mesenchyme transition: Direct repression of the gene expression of claudins/occludin by Snail. J Cell Sci. 116:1959–1967. 2003. View Article : Google Scholar : PubMed/NCBI

29 

Zavadil J and Böttinger EP: TGF-beta and epithelial-to-mesenchymal transitions. Oncogene. 24:5764–5774. 2005. View Article : Google Scholar : PubMed/NCBI

30 

Huber MA, Kraut N and Beug H: Molecular requirements for epithelial-mesenchymal transition during tumour progression. Curr Opin Cell Biol. 17:548–558. 2005. View Article : Google Scholar : PubMed/NCBI

31 

Savagner P, Yamada KM and Thiery JP: The zinc-finger protein slug causes desmosome dissociation, an initial and necessary step for growth factor-induced epithelial-mesenchymal transition. J Cell Biol. 137:1403–1419. 1997. View Article : Google Scholar : PubMed/NCBI

32 

Yang J, Mani SA, Donaher JL, Ramaswamy S, Itzykson RA, Come C, Savagner P, Gitelman I, Richardson A and Weinberg RA: Twist, a master regulator of morphogenesis, plays an essential role in tumour metastasis. Cell. 117:927–939. 2004. View Article : Google Scholar : PubMed/NCBI

33 

Kim K, Lu Z and Hay ED: Direct evidence for a role of beta-catenin/LEF-1 signaling pathway in induction of EMT. Cell Biol Int. 26:463–476. 2002. View Article : Google Scholar : PubMed/NCBI

34 

Logullo AF, Nonogaki S, Pasini FS, Osório CA, Soares FA and Brentani MM: Concomitant expression of epithelial-mesenchymal transition biomarkers in breast ductal carcinoma: Association with progression. Oncol Rep. 23:313–320. 2010.PubMed/NCBI

35 

Mikheeva SA, Mikheev AM, Petit A, Beyer R, Oxford RG, Khorasani L, Maxwell JP, Glackin CA, Wakimoto H, González-Herrero I, et al: TWIST1 promotes invasion through mesenchymal change in human glioblastoma. Mol Cancer. 9:1942010. View Article : Google Scholar : PubMed/NCBI

36 

Zhu QC, Gao RY, Wu W and Qin HL: Epithelial-mesenchymal transition and its role in the pathogenesis of colorectal cancer. Asian Pac J Cancer Prev. 14:2689–2698. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Han SP, Kim JH, Han ME, Sim HE, Kim KS, Yoon S, Baek SY, Kim BS and Oh SO: SNAI1 is involved in the proliferation and migration of glioblastoma cells. Cell Mol Neurobiol. 31:489–496. 2011. View Article : Google Scholar : PubMed/NCBI

38 

Nieto MA: The snail superfamily of zinc-finger transcription factors. Nat Rev Mol Cell Biol. 3:155–166. 2002. View Article : Google Scholar : PubMed/NCBI

39 

Barrallo-Gimeno A and Nieto MA: The Snail genes as inducers of cell movement and survival: implications in development and cancer. Development. 132:3151–3161. 2005. View Article : Google Scholar : PubMed/NCBI

40 

Paznekas WA, Okajima K, Schertzer M, Wood S and Jabs EW: Genomic organization, expression and chromosome location of the human SNAIL gene (SNAI1) and a related processed pseudogene (SNAI1P). Genomics. 62:42–49. 1999. View Article : Google Scholar : PubMed/NCBI

41 

Inukai T, Inoue A, Kurosawa H, Goi K, Shinjyo T, Ozawa K, Mao M, Inaba T and Look AT: SLUG, a ces-1-related zinc finger transcription factor gene with antiapoptotic activity, is a downstream target of the E2A-HLF oncoprotein. Mol Cell. 4:343–352. 1999. View Article : Google Scholar : PubMed/NCBI

42 

Rosanò L, Cianfrocca R, Spinella F, Di Castro V, Nicotra MR, Lucidi A, Ferrandina G, Natali PG and Bagnato A: Acquisition of chemoresistance and EMT phenotype is linked with activation of the endothelin A receptor pathway in ovarian carcinoma cells. Clin Cancer Res. 17:2350–2360. 2011. View Article : Google Scholar : PubMed/NCBI

43 

Thiery JP and Sleeman JP: Complex networks orchestrate epithelial-mesenchymal transition. Nat Rev Mol Cell Biol. 7:131–142. 2006. View Article : Google Scholar : PubMed/NCBI

44 

Beckman G, Beckman L, Pontén J and Westermark B: G-6-PD and PGM phenotypes of 16 continuous human tumor cell lines. Evidence against cross-contamination and contamination by HeLa cells. Hum Hered. 21:238–241. 1971. View Article : Google Scholar : PubMed/NCBI

45 

Stein GH: T98G: An anchorage-independent human tumor cell line that exhibits stationary phase G1 arrest in vitro. J Cell Physiol. 99:43–54. 1979. View Article : Google Scholar : PubMed/NCBI

46 

Diserens AC, de Tribolet N, Martin-Achard A, Gaide AC, Schnegg JF and Carrel S: Characterization of an established human malignant glioma cell line: LN-18. Acta Neuropathol. 53:21–28. 1981. View Article : Google Scholar : PubMed/NCBI

47 

Pontén J and Macintyre EH: Long term culture of normal and neoplastic human glia. Acta Pathol Microbiol Scand. 74:465–486. 1968. View Article : Google Scholar : PubMed/NCBI

48 

Neumann I, Foell JL, Bremer M, Volkmer I, Korholz D, Burdach S and Staege MS: Retinoic acid enhances sensitivity of neuroblastoma cells for imatinib mesylate. Pediatr Blood Cancer. 55:464–470. 2010. View Article : Google Scholar : PubMed/NCBI

49 

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 : PubMed/NCBI

50 

Heerboth S, Housman G, Leary M, Longacre M, Byler S, Lapinska K, Willbanks A and Sarkar S: EMT and tumor metastasis. Clin Transl Med. 4:62015. View Article : Google Scholar : PubMed/NCBI

51 

Casas E, Kim J, Bendesky A, Ohno-Machado L, Wolfe CJ and Yang J: Snail2 is an essential mediator of Twist1-induced epithelial mesenchymal transition and metastasis. Cancer Res. 71:245–254. 2011. View Article : Google Scholar : PubMed/NCBI

52 

Li Y, Wang W, Wang W, Yang R, Wang T, Su T, Weng D, Tao T, Li W, Ma D, et al: Correlation of TWIST2 up-regulation and epithelial-mesenchymal transition during tumorigenesis and progression of cervical carcinoma. Gynecol Oncol. 124:112–118. 2012. View Article : Google Scholar : PubMed/NCBI

53 

Fang X, Cai Y, Liu J, Wang Z, Wu Q, Zhang Z, Yang CJ, Yuan L and Ouyang G: Twist2 contributes to breast cancer progression by promoting an epithelial-mesenchymal transition and cancer stem-like cell self-renewal. Oncogene. 30:4707–4720. 2011. View Article : Google Scholar : PubMed/NCBI

54 

Elias MC, Tozer KR, Silber JR, Mikheeva S, Deng M, Morrison RS, Manning TC, Silbergeld DL, Glackin CA, Reh TA, et al: TWIST is expressed in human gliomas and promotes invasion. Neoplasia. 7:824–837. 2005. View Article : Google Scholar : PubMed/NCBI

55 

Maier HJ, Schmidt-Strassburger U, Huber MA, Wiedemann EM, Beug H and Wirth T: NF-kappaB promotes epithelial-mesenchymal transition, migration and invasion of pancreatic carcinoma cells. Cancer Lett. 295:214–228. 2010. View Article : Google Scholar : PubMed/NCBI

56 

Luo GQ, Li JH, Wen JF, Zhou YH, Hu YB and Zhou JH: Effect and mechanism of the Twist gene on invasion and metastasis of gastric carcinoma cells. World J Gastroenterol. 14:2487–2493. 2008. View Article : Google Scholar : PubMed/NCBI

57 

Wick W, Weller M, Weiler M, Batchelor T, Yung AW and Platten M: Pathway inhibition: Emerging molecular targets for treating glioblastoma. Neuro Oncol. 13:566–579. 2011. View Article : Google Scholar : PubMed/NCBI

58 

Niclou SP, Fack F and Rajcevic U: Glioma proteomics: Status and perspectives. J Proteomics. 73:1823–1838. 2010. View Article : Google Scholar : PubMed/NCBI

59 

Frosina G: Development of therapeutics for high grade gliomas using orthotopic rodent models. Curr Med Chem. 20:3272–3299. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2017
Volume 13 Issue 5

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Kühnöl CD, Würfel C, Staege MS and Kramm C: Snail homolog 1 is involved in epithelial-mesenchymal transition-like processes in human glioblastoma cells. Oncol Lett 13: 3882-3888, 2017
APA
Kühnöl, C.D., Würfel, C., Staege, M.S., & Kramm, C. (2017). Snail homolog 1 is involved in epithelial-mesenchymal transition-like processes in human glioblastoma cells. Oncology Letters, 13, 3882-3888. https://doi.org/10.3892/ol.2017.5875
MLA
Kühnöl, C. D., Würfel, C., Staege, M. S., Kramm, C."Snail homolog 1 is involved in epithelial-mesenchymal transition-like processes in human glioblastoma cells". Oncology Letters 13.5 (2017): 3882-3888.
Chicago
Kühnöl, C. D., Würfel, C., Staege, M. S., Kramm, C."Snail homolog 1 is involved in epithelial-mesenchymal transition-like processes in human glioblastoma cells". Oncology Letters 13, no. 5 (2017): 3882-3888. https://doi.org/10.3892/ol.2017.5875