Establishment and genetic characterization of ANGM-CSS, a novel, immortal cell line derived from a human glioblastoma multiforme

  • Authors:
    • Angelantonio Notarangelo
    • Domenico Trombetta
    • Vincenzo D'Angelo
    • Paola Parrella
    • Orazio Palumbo
    • Clelia Tiziana Storlazzi
    • Luciana Impera
    • Lucia Anna Muscarella
    • Antonella La Torre
    • Andrea Affuso
    • Vito Michele Fazio
    • Massimo Carella
    • Leopoldo Zelante
  • View Affiliations

  • Published online on: December 23, 2013     https://doi.org/10.3892/ijo.2013.2224
  • Pages: 717-724
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Glioblastoma multiforme (World Health Organ­ization, grade IV astrocytoma) is the most common and most aggressive malignant primary brain tumor. We report a novel cell line, designated as ANGM-CSS, which was established from a 56-year-old male patient with a surgically removed glioblastoma multiforme. The ANGM-CSS cell line was established in vitro and characterized using histological and immunohistochemical staining, classical and molecular cytogenetic analyses, molecular studies and functional assays using a xenograft model in immunodeficient animals. ANGM-CSS was positive for CD133, nestin and vimentin proteins, whereas GFAP showed staining only in a fraction of the cells. Cytogenetic and molecular cytogenetic analysis revealed a near-tetraploid karyotype, with a modal chromosome number from 88 to 91, and additional cytogenetic abnormalities, such as the t(6;14)(p12;q11.2), t(8;10)(q24.2;q21.1) and t(5;9)(q34;p21) unbalanced translocations. Moreover, ANGM-CSS showed amplification of the MET and EGFR genes whose overexpression was observed at the mRNA level. Interestingly, ANGM-CSS is tumorigenic when implanted in immunodeficient mice, and the cells obtained from the xenografts showed the same morphology and karyotype in vitro as the original cell line. ANGM-CSS represents a biologically relevant cell line to be used to investigate the molecular pathology of glioblastoma multiforme, also to evaluate the efficacy of novel therapeutic drugs in vitro.

Introduction

Glioma is the most common primary brain tumor affecting yearly 3–5/100,000 and occurring mainly in adults >45 years old (13). The ability of glioma to invade and infiltrate diffusely contiguous brain tissue limits the complete surgical resection and the efficacy of standard therapies (4). Glioblastoma multiforme (GBM), a grade IV astrocytoma as currently defined by the World Health Organization (WHO) classification (5), is the most common and the most lethal form of brain tumor. The therapeutic approach against this tumor consists in surgical resection followed by radiation and chemotherapy with temozolomide (TMZ) (6,7). At present, such treatment can only slightly modify the patient’s outcome. In fact, the tumor typically recurs after an average of only 6.9 months, resulting in a median survival rate of <1 year following diagnosis (8). Permanent cell lines represent important tools to study the behaviour of human tumors, such as their growth and metabolism, drug sensitivity and resistance, as well as genomic and expression profiles (911). Furthermore, some cultivated cancer cells can originate a novel tumor when transplanted into nude mice, providing an experimental system to test potential novel therapeutic drugs (12). Here we report the establishment and the characterization, by cytogenetic and molecular approaches, of a novel cell line termed as ANGM-CSS, derived from a patient with GBM.

Materials and methods

Patient history

The patient was a 56-year-old male, surgically treated to remove a large primary tumor localized in the left temporo-occipital lobe with invasion of the ventricular cornus (Fig. 1A). Hematoxylin and eosin staining (Fig. 1B) and immunohistochemical examination for GFAP, α-SMA and HMB45 were performed as part of the routine assessment of tumor type/phenotype. Immunostaining demonstrated a weak positivity for GFAP (Fig. 1C) but was negative for α-SMA and HMB45 (data not shown). The final diagnosis was GBM with spindle, mitotically active cells showing a fascicular growth pattern. After surgery, the patient was treated with temozolomide at a daily dose of 75 mg/m2 of body surface in association with fractionated radiotherapy (60 Gy) for 6–7 weeks. The patient died 18 months after surgery.

Establishment of primary culture

After surgical removal, upon institutional Ethics Committee approval, the tumor specimen was placed immediately in DMEM-F12 medium without serum, repeatedly washed with phosphate-buffered saline (PBS, Invitrogen, Carlsbad, CA, USA) and then placed in a 30-mm Petri dish. The specimen was cut in 1–2 mm or tinier fragments and transferred in a poly-D-lysine treated flask with a small amount of Dulbecco’s modified Eagle’s medium/F12 medium (D-MEM/F12, Invitrogen) (1:1, v/v) supplemented with 10% fetal bovine serum (FBS, Invitrogen), 100 U/ml penicillin and 100 μg/ml streptomycin (PenStrep, Invitrogen) in order to allow the fragments to adhere to the surface of the flask. Primary culture was incubated at 37°C in 5% CO2 humidified atmosphere, and 5 ml of complete medium was added 24 h later. After one week, the primary culture was washed with PBS to remove non-adherent fragments and fresh medium prewarmed at 37°C was added. These procedures were repeated every 3 days until primary culture reached local confluence. Then cells were treated with 0.05% trypsin (Invitrogen) and 0.02% EDTA (Invitrogen), washed with PBS and transferred into a T-75 flask without biocoat and containing complete medium DMEM/F12. Next, the culture was serially transferred by using the same procedures once or twice a week. Every 10 passages, one amount of cells was counted with Z1-Coulter (IL-Laboratories). Cells (50×106) were frozen in medium with 10% dimethyl sulfoxide (DMSO) and stored in liquid nitrogen. Cells were propagated by serial passages (split ratio 1:3) along 2 years, until 105th passage. Growth curves were established at the 32nd passages by seeding 1×105 cells into three 35-mm culture dishes. Triplicate dishes were harvested and counted daily with Z1-Coulter (IL-Laboratories). The cell number was determined as the average number of cells ± SD in each time interval.

Immunophenotypical characterization of ANGM-CSS cell line

Immunofluorescence analysis was performed at different passages to establish GFAP, nestin, CD133 and vimentin localization in the ANGM cell cultures. After trypsin treatment, 105 cells were seeded on 20×20 mm coverslips, rinsed twice in PBS, and fixed in PBS containing 4% formaldehyde (pH 7.2–7.4) for 10 min at room temperature (RT), washed for 10 min three times with PBS, permeabilized with PBS/0.2% Triton X-100 (MP Biomedical) and blocked for 30 min with PBS containing bovine serum albumin (BSA). After three washings with PBS, the cells were incubated with a primary antibody against CD133 (AP2010b, purified rabbit, ABGENT), nestin (sc-23927, mouse monoclonal IgG1, Santa Cruz Biotechnology), vimentin (sc-6260, mouse monoclonal IgG1, Santa Cruz Biotechnology), GFAP (sc-58766, mouse monoclonal IgG1, Santa Cruz Biotechnology) diluted 1:300 in BSA for 1 h at RT. After extensive washing in PBS, the coverslips were incubated with the secondary antibodies for 30 min at room temperature. A goat anti-mouse IgG-FITC conjugated (sc-2010, 1:200, Santa Cruz Biotechnology) for vimentin, nestin and GFAP, and a goat anti-rabbit Ig-G FITC conjugated (sc-2012, 1:200, Santa Cruz Biotechnology) for CD-133 were used as secondary antibodies. Nuclei were then washed only once with PBS and stained with DAPI (1 mg/ml). The fluorescent staining was visualized using Nikon E1000 microscopy.

Cytogenetic analysis

ANGM-CSS cells were subcultured at passages 5, 32, 67, 86 and incubated overnight with Colcemid (0.05 mg/ml, Invitrogen). Cells were dispersed with 0.5% trypsin (Invitrogen) and 0.02 EDTA (Invitrogen), then washed with 1X PBS and treated with 0.075 M KCl hypotonic solution and fetal bovine serum (1:1, v/v) for 20 min at 37°C. The cells were then fixed with methanol:acetic acid (3:1, v/v) solution and stored for 1 h at −20°C. Cell suspension were dropped on ice glass slides and stained in Giemsa stain after banding with GAG-acid solution at 55°C. Karyotyping was performed by use of the Genikon software (Nikon Italia, Firenze, Italy) and described in accordance to the International System for Human Cytogenetic Nomenclature (ISCN 2009).

SNP array analysis

SNP array experiments and data analysis were performed using the Genome-Wide Human SNP 6.0 array (Affymetrix, Santa Clara, CA, USA) as previously described (13).

Multicolor fluorescence in situ hybridization (M-FISH)

M-FISH analysis of ANGM-CSS cells was performed using the commercially available 24-colour SpectraVysion probe (Abbott), according to the manufacturer’s instructions. Metaphase images were captured using a Leica DM-RXA2 epifluorescence microscope equipped with an 8-position automated filter wheel and a cooled CCD camera (Princeton Instruments). Six fluorescent images per metaphase were captured using filter combinations specific for SpectrumGold, SpectrumAqua, SpectrumGreen, FRed, Red, and DAPI. Images were processed using the Leica CW4000 M-FISH software.

FISH

BAC clones for FISH analysis were selected according to the March 2006 release of the UCSC Human Genome Browser (http://genome.ucsc.edu) (data not shown). FISH experiments were carried out as previously described (14).

Quantitative reverse transcription polymerase chain reaction (RTq-PCR)

Expression analysis of EGFR and MET genes was performed comparing the mRNA levels in the ANGM-CSS cell line and normal human astrocytes (NHA) (Lonza Walkersville, MD, USA) by QRT-PCR on 7700 sequence detection system (Applied Biosystems, Foster City, CA, USA) using MGB TaqMan chemistry and the 2−ΔCt relative method for relative quantification (15). For the analysis were used the following TaqMan® Gene Expression Assays (Applied Biosystems): MET (Hs01565580_m1) and EGFR (Hs01076092_m1). The human large ribosomal protein transcript (Human RPLP09, Applied Biosystems) was used as endogenous control.

Quantitative methylation-specific PCR (QMSP) for the MGMT gene

DNA extracted from ANGM-CSS underwent bisulfite treatment and subsequent DNA purification using the Epitect Bisulfate kit (Qiagen Sci, MD, USA) according to the manufacturer’s instructions. Bisulphite converted DNA was used as template for fluorescence-based real-time QMSP. Real-time PCR experiments for MGMT were performed as previously described (16).

TP53 and KRAS mutation analysis

PCR amplifications of KRAS gene exon 2 and TP53 gene exons 4–8 were performed as previously described (17). Amplification reactions were performed in a GeneAmp PCR System 9700 (Perkin-Elmer, Foster City, CA, USA) in a final reaction volume of 25 μl containing 100 ng of genomic DNA template, 0.25 nM dNTPs, 20 pmol of each primers, 1 U HotMaster Taq polymerase (Eppendorf), in 1X PCR reaction buffer. All PCR products were purified using GFX PCR DNA and Gel Band Purification kit (GE Healthcare, Buckinghamshire, UK) and sequenced. Sequencing reactions were performed in 10 μl of final volume using 3 pmol of primer, 4–6 ng of DNA template and 1 μl of Big Dye Terminator Ready Reaction mix v.1.1 (Applied Biosystems). Sequencing reactions were loaded on an ABI 3100 capillary sequencer (Applied Biosystems) and by the Sequencing Analysis software v.3.7 (PE Applied Biosystems).

FIG-ROS1 and FGFR3-TACC3 fusion gene detection

Total RNA was isolated from ANGM-CSS cell line using the TRIzol reagent (InvitrogenTM Life Technologies, Carlsbad, CA, USA) according to the manufacturer’s instructions. The Agilent 2100 Bioanalyzer was used to measure the quantity, integrity and purity of total RNA. RNA (1 μg) was reverse transcribed by High Capacity cDNA Reverse Transcription kit (Life Technologies) according to the manufacturer’s instructions. To detect the possible presence of FIG-ROS1 fusion transcript, reverse transcriptase (RT)-PCR experiments were performed using primers previously reported (18). The detection of the FGFR3-TACC3 fusion gene was performed on the basis of the results previously described (19) and using the primers TACC3_Ex5_f (CTTGAACTCTGCCAGCACCT) and FGFR3_Ex16_r GTGGGCAAACACGGAGTC. Briefly, for both chimeric genes, 2 μl cDNA was used as template in a final volume of 50 μl containing 10X PCR buffer, 0.25 mM of each dNTP, 0.5 μM of each forward and reverse primer, 0.5 U HotMaster Taq DNA polymerase (5Prime). The PCRs were run on a GeneAmp PCR System 9700 (Applied Biosystem) with the cycling profile of initial denaturation for 2 min at 94°C followed by 35 cycles of 1 min at 94°C, 1 min at 56°C and 2 min at 72°C, with a final extension for 10 min at 72°C. For the experiments for FIG-ROS1 detection, the cDNA of U118MG cell line was used as positive control (1820). The PCR product (20 μl) was analyzed by electrophoresis through a 1% agarose gel containing ethidium bromide for staining.

Murine xenograft model

Six female, 6-week old athymic nude mice (Crl:CD1-NU-Foxn1nu from Charles River Laboratories, Italy) were used for transplantation studies in accordance with national and institutional guidelines and were kept under specific pathogen-free (SPF) conditions. The mice were inoculated subcutaneously with 200 μl of cell suspension at four different concentrations (from a minimum concentration of 5×105 cells in 200 μl of mixture PBS/Matrigel 1:1, to a maximum concentration of 10×106 cells in 200 μl of mixture PBS/Matrigel 1:1). Mice were bilaterally inoculated on the flank (three injections for each concentration), while a control mouse was inoculated with Matrigel. The tumor diameter was measured weekly with a digital caliper and the tumor volume (mm3) was calculated as previously described (21). All mice used in the experiment were monitored daily for signs of suffering and to identify cases of spontaneous death.

Results

The primary culture grew initially slowly, reaching cell confluence three weeks after surgical removal. Analysis by phase contrast microscopy showed a mixed population with dendritic-like and spindle cells (Fig. 1D). From the 26th passage, the cells showed a spindle shape with large nuclei. Cells were propagated until the 105th passage, growing continuously for >2 years, without morphological changes. The doubling time was calculated at the 32nd passage. One day after subculturing, the cells entered an exponential growth phase, where the doubling time was ∼60 h. Immunofluorescence analysis showed a decrease in GFAP immunoreactivity, starting from the 17th passage to completely disappear during further serial passages in vitro. The cells were persistently positive for vimentin and nestin, while only a small fraction of the ANGM-CSS cell population showed a CD133 staining (data not shown). Cytogenetic and molecular analyses were performed at the 32nd, at the 70th and at the last passages. Metaphase spreads from long-term cultured cells were treated by conventional cytogenetic methods for karyotype analysis and thirty-two metaphases were analyzed. The cytogenetic analysis showed a strong karyotypic complexity and heterogeneity; the chromosome number was near-tetraploid and in addition to whole chromosome losses (chromosomes 10, 14 and 21) and gains (chromosomes 7, 20 and 19), two clonal chromosomal translocations were observed: a t(6;14) (p12;q11.2), and a t(8;10) (q24.2;q21.1). Moreover, the recurrent deletion of the long arm of chromosome 6 was detected and one marker chromosome apparently composed of chromosome 12 material [mar (12)] was found to be recurrent in 100% of the cell population (Fig. 2). According to the 2009 recommendations of the International System for Human Cytogenetic Nomenclature (22), the karyotype was described as follows: 88∼91,XXYY,−6,+7,del(9)(p21.1)×2,−10,−12,+der(14)t(6;14)(p12;q11.2)×2,−17,+19,+20,−21,+marx2[p32].

To better define genomic gains and losses, SNP array analysis was performed on DNA extracted at the same passages. The SNP analysis confirmed the high genomic complexity, showing, in addition to the losses and gains of whole chromosomes detected by chromosome banding, the deletion of the 6q, copy number alterations at 5q (gain of a 14.6 Mb region from 5q34 to 5qter) and 9p (loss of a 28.7-Mb segment from 9p24.3 to 9p21.1); in addition, chromosome 12 showed copy number changes from 10 to 1 along its long arm (Fig. 3A), similarly to regions of chromosomes 7, 16 and 17. The overall SNP array results are listed in Table II.

Table II.

SNP array results.

Table II.

SNP array results.

ChromosomeGain (chromosome/cytoband)Loss (chromosome/cytoband)
22p11.2
33q13.31–3q13.32, 3q13.31
44q34.3
55q34–5q35.3,5q34, 5q34
66q16.3–6q27, 6q11.1–6q16.2
77, 7p22.3–7q36.3
99p24.2, 9p24.3-9p24.2, 9p24.2-9p21.1
1010, 10q23.1, 10p15.3–10q23.1, 10q23.1–10q26.3
1212q24.23–12q24.31, 12q24.32, 12q22–12q23.1, 12q14.1–12q21.1, 12q24.12–12q24.13, 12q23.1, 12q23.1–12q23.2, 12q13.2–12q13.3, 12q24.31, 12q24.13, 12q23.3, 12q13.3–12q14.112q12, 12q23.1, 12q21.1, 12q23.1, 12q23.1, 12q23.2–12q23.3, 12q21.1–12q22, 12q23.3–12q24.12, 12q24.13–12q24.23, 12q14.1, 12q24.32–12q24.33
1414q11.214
1717p13.1, 17p13.2, 17p13.3 17q11.1–17q21.31
1919, 19p13.3–19q13.43
2020, 20p13–20q13.33
2121
XXp22.33–Xq28

M-FISH analysis confirmed the presence of the unbalanced translocation der(14)t(6;14)(p12;q11.2) and detected two additional unbalanced translocations: der(8)t(8;10)(q24.2;q21.1) and der(9)t(5;9)(q34;p21), in agreement with the SNP array data and clarified that the additional material on der(12), already detected by G-banding and SNP-array CGH, originated from amplified material from chromosomes 7, 12 and 17 (Fig. 3B–E). Chromosome aberrations detected by both SNP array and M-FISH analyses, were further validated by FISH experiments with locus-specific BAC probes. Interestingly, the long arm of the der(12) was shown to be almost entirely composed by amplified chromosome 12 sequences, together with chromosomes 7, 16 and 17 material (Fig. 4). Moreover, we finely mapped the breakpoints of all unbalanced translocations (data not shown). Then, combining classical cytogenetic, molecular cytogenetic and SNP array data for ANGM-CCS cell line, it was possible to define the karyotype as: 88∼91,XXYY,−6,+7,+add(9)(p21.1)ishdup(5)(q34)×2,+der(8)t(8;10)(q24.2;q21.1),−10,+der(12)ishins(7;17;16)(12 qter→12q24::7?::17?::16?::12q24.3)×2,+der(14)t(6;14)(p12;q11.2)×2,−17,+19,+20[cp32]. ANGM-CSS showed an increased expression of EGFR and MET as compared to a normal human astrocytes (NHA) cell line (data not shown). The relative mRNA expression value for the MET and EGFR in the tumor cell line was, respectively 4.65 and 1.45. ANGM-CSS did not show methylation of the MGMT promoter or pathogenic mutations in the hotspot regions (exons 5–8) of the TP53 gene, neither were mutations detected in codons 12 and 13 of the KRAS gene. No FIG-ROS1 or FGFR3-TACC3 chimeric gene was detected. Six weeks after the subcutaneous injection of ANGM-CSS cells in athymic nude mice, all animals showed growth of a macroscopically visible tumor. Mice were sacrified eight weeks after injections and the tumors were immediately excised. One fragment was used for serial transplantation in other mice whereas the other section was used to start a new culture. The newly injected mice developed tumors two weeks after inoculation. The morphology of the cells grown after heterotransplantation did not differ from the initial culture (Fig. 1E). The human origin of the tumor cells was confirmed by chromosome analysis that revealed the same karyotype (data not shown).

Discussion

We successfully established a novel cell line, named ANGM-CSS, from a patient with GBM. Cell line immunoreactivity for GFAP showed a decrease after serial passages in culture, as previously described (2327); whereas, the immunohistochemical positivity for vimentin and nestin was persistent. The immunohistochemical results recapitulate the phenotype of the GMB cell lines, according to data from literature (2628). We provided a detailed characterization of ANGM-CSS by use of cytogenetic and molecular approaches. As expected, the cytogenetic analysis of ANGM-CSS showed a very complex karyotype characterized by several chromo-some aberrations, in line with the data described in previous studies concerning human glioma cell lines (2931). Gliomas have been extensively analyzed by genetic techniques. They typically show highly complex karyotypes (31). The most common numerical chromosomal changes include losses of 9p, chr10, chr17 and chr22 and gain of chromosomes chr7 and chr20 (3135). Other structural abnormalities were also reported in chromosomal arms 1p, 6q, 9p, 9q and 13q (31). Similary to the latter data, G-banding, molecular cytogenetic and SNP array analysis performed on ANGM-CSS disclosed a near-tetraploid karyotype harbouring numerous copy number changes, including gain of whole chromosomes 7, 20 and 19 and loss of whole chromosomes 10, 14 and 21, in addition to gains of sub-regions in 5q, 7 and 17 and loss of 6q and 9p. Moreover, three structural chromosome rearrangements were identified in ANGM-CSS: a t(8;10)(q24.2;q21.1), a t(6;14)(p12;q11.2) and an add(9)(p21.1) ish dup(5)(q34). In summary, the genetic analyses indicate that the ANGM-CSS cell line recapitulates the key properties of GBM. The Cancer Genome Atlas Research Network reported in GBM frequent genetic alterations in three critical pathways: the RTK/RAS/PI3K, the p53 and the RB signalling pathways (36). ANGM-CSS presents a profound deregulation of proliferation and survival due to disruption of the RTK/RAS/PI3K pathway due to the amplification of the EGFR and MET genes and the homozygous deletion of NF1 and PTEN genes, as detected by SNP array analysis (Table I). Quantitative RT-PCR analysis confirmed an increased expression at the mRNA level of the EGFR and MET genes. The cell line under study does not show the presence of FIG-ROS1 and FGFR3-TACC3 chimeric transcripts, already described for U118MG glioblastoma cell line and for primary tumors, respectively (19,20).

Table I.

Comparison between the genetic abnormalities reported by the Cancer Genome Atlas (TCGC) research network at gene loci critically involved in gliomas and gains and losses detected in ANGM-CSS cell line by SNP array analysis.a

Table I.

Comparison between the genetic abnormalities reported by the Cancer Genome Atlas (TCGC) research network at gene loci critically involved in gliomas and gains and losses detected in ANGM-CSS cell line by SNP array analysis.a

GeneLocusTCGAbANGM-CSS
Genes involved in RTK/RAS/PI-3K signaling
EGFR7p12.3-p12.1Amplified45% amplified
ERBB217q21.1Mutated8% homozygous deletion
PDGFRA4q12Amplified13% no change
MET7q31Amplified4% amplified
NF117q11.2Homozygous deletion18% homozygous deletion
RAS6p21.3Mutated2% no change
PI3K3q26.3Mutated15% no change
PTEN10q23.31Homozygous deletion36% homozygous deletion
AKT14q32.3Amplified2% no change
FOXO6q21Mutated1% homozygous deletion
TP53 regulation
CDKN2A9p21Homozygous deletion49% deleted Hom
MDM212q14.3–q15Amplified14% amplified
MDM41q32Amplified7% no change
TP5317p13.1Homozygous deletion35% no change
RB signaling
CDKN2A9p21Homozygous deletion52% homozygous deletion
CDKN2B9p21Homozygous deletion47% homozygous deletion
CDKN2C1p32Homozygous deletion2% no change
CDK412q14Amplified18% amplified
CCND212p13Amplified2% amplified
CDK67q21–22Amplified1% amplified
RB113q14.1–q14.2Homozygous deletion11% homozygous deletion

a The ANGM-CSS cell line is characterized by amplification of the EGFR, MET, MDM2, CDK4 and CDK6 genes and homozygous deletion of NF1, CDKN2A and CDKN2B genes.

b TCGA, The Cancer Genome Atlas.

While no mutation or deletions were detected in the TP53 gene, ANGM-CSS was characterized by amplification of the MDM2 gene and homozygous deletion of the CDKN2A gene (Table I). The MDM2 gene encodes for a protein involved in the degradation of the p53 protein and its expression is negatively regulated by the ARF protein, encoded by CDKN2A, leading to an abnormal regulation of apoptosis and senescence mediated by p53 (37). The Rb pathway was affected by the amplification of CDK4 and CDK6 and the homozygous deletion of the CDKN2A/CDKN2B genes. All these alterations affect the G1/S phase progression. Another feature of the ANGM-CSS is the absence of methylation in the promoter region of the MGMT gene. Alkylating agents induce cell death by forming cross-links between adjacent DNA strands through the alkylation of the O6 position of guanine. MGMT promoter hypermethylation with consequent loss of MGMT protein expression reduces the DNA repair activity of glioma cells overcoming resistance to alkylating agents. The absence of MGMT promoter hypermethylation in ANGM-CSS leads to a transcriptionally active MGMT which rapidly removes the alkyl adducts preventing the formation of cross-links thereby causing resistance to alkylating drugs (38,39). ANGM-CSS cell also gave rise to tumors in vivo. All six SCID mice that were injected with tumor cells developed solid tumors, demonstrating that this cell line is capable of being propagated in animal models, which may aid in the development of test systems for new therapies.

Acknowledgements

This study was supported by Italian Health Ministry.

References

1. 

Furnari FB, Fenton T, Bachoo RM, et al: Malignant astrocytic glioma: genetics, biology, and paths to treatment. Genes Dev. 21:2683–26710. 2007. View Article : Google Scholar : PubMed/NCBI

2. 

Porter K, McCarthy B, Berbaum M, et al: Conditional survival of all primary brain tumor patients by age, behavior, and histology. Neuroepidemiology. 36:230–239. 2011. View Article : Google Scholar : PubMed/NCBI

3. 

Preusser M, de Ribaupierre S, Wöhrer A, et al: Current concepts and management of glioblastoma. Ann Neurol. 70:9–21. 2011. View Article : Google Scholar

4. 

Cheng L, Wu Q, Guryanova O, et al: Elevated invasive potential of glioblastoma stem cells. Biochem Biophys Res Commun. 406:643–648. 2011. View Article : Google Scholar : PubMed/NCBI

5. 

Louis DN, Ohgaki H, Wiestler OD, et al: The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol. 114:97–109. 2007. View Article : Google Scholar : PubMed/NCBI

6. 

Stupp R, Mason WP, van den Bent MJ, et al: Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 352:987–996. 2005. View Article : Google Scholar : PubMed/NCBI

7. 

Stupp R, Hegi ME, Mason WP, et al: 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.

8. 

Wen PY and Kesari S: Malignant gliomas in adults. N Engl J Med. 359:492–507. 2008. View Article : Google Scholar : PubMed/NCBI

9. 

Bakir A, Gezen F, Yildiz O, et al: Establishment and characterization of a human glioblastoma multiforme cell line. Cancer Genet Cytogenet. 103:46–51. 1998. View Article : Google Scholar : PubMed/NCBI

10. 

Nuki Y, Uchinokura S, Miyata S, et al: Establishment and characterization of a new human glioblastoma cell line, NYGM. Hum Cell. 17:145–150. 2004. View Article : Google Scholar : PubMed/NCBI

11. 

Wang J, Wang X, Jiang S, et al: Establishment of a new human glioblastoma multiforme cell line (WJ1) and its partial characterization. Cell Mol Neurobiol. 27:831–843. 2007. View Article : Google Scholar : PubMed/NCBI

12. 

Fomchenko EI and Holland EC: Mouse models of brain tumors and their applications in preclinical trials. Clin Cancer Res. 12:5288–5297. 2006. View Article : Google Scholar : PubMed/NCBI

13. 

Palumbo O, Palumbo P, Palladino T, et al: A novel deletion in 2q24.1q24.2 in a girl with mental retardation and generalized hypotonia: a case report. Mol Cytogenet. 5:12012. View Article : Google Scholar : PubMed/NCBI

14. 

Macchia G, Trombetta D, Möller E, et al: FOSL1 as a candidate target gene for 11q12 rearrangements in desmoplastic fibroblastoma. Lab Invest. 92:735–743. 2012. View Article : Google Scholar : PubMed/NCBI

15. 

Scintu M, Vitale R, Prencipe M, et al: Genomic instability and increased expression of BUB1B and MAD2L1 genes in ductal breast carcinoma. Cancer Lett. 254:298–307. 2012. View Article : Google Scholar : PubMed/NCBI

16. 

Parrella P, la Torre A, Copetti M, et al: High specificity of quantitative methylation-specific PCR analysis for MGMT promoter hypermethylation detection in gliomas. J Biomed Biotechnol. 2009:5316922009. View Article : Google Scholar : PubMed/NCBI

17. 

Hibi K, Robinson CR, Booker S, et al: Molecular detection of genetic alterations in the serum of colorectal cancer patients. Cancer Res. 58:1405–1407. 1998.PubMed/NCBI

18. 

Gu TL, Deng X, Huang F, et al: Survey of tyrosine kinase signaling reveals ROS kinase fusions in human cholangiocarcinoma. PLoS One. 6:e156402011. View Article : Google Scholar : PubMed/NCBI

19. 

Singh D, Chan JM, Zoppoli P, et al: Transforming fusions of FGFR and TACC genes in human glioblastoma. Science. 337:1231–1235. 2012. View Article : Google Scholar : PubMed/NCBI

20. 

Charest A, Lane K, McMahon K, et al: Fusion of FIG to the receptor tyrosine kinase ROS in a glioblastoma with an interstitial del(6) (q21q21). Genes Chromosomes Cancer. 37:58–71. 2003. View Article : Google Scholar : PubMed/NCBI

21. 

Burfeind P, Chernicky CL, Rininsland F, et al: Antisense RNA to the type I insulin-like growth factor receptor suppresses tumor growth and prevents invasion by rat prostate cancer cells in vivo. Proc Natl Acad Sci USA. 93:7263–7268. 1996. View Article : Google Scholar : PubMed/NCBI

22. 

Shaffer LG, Slovak ML and Campbell LJ: ISCN 2009: An International System for Human Cytogenetic Nomenclature. S. Karger; Basel: 2009

23. 

Lolait SJ, Harmer JH, Auteri G, et al: Expression of glial fibrillary acidic protein, actin, fibronectin and factor VIII antigen in human astrocytomas. Pathology. 15:373–378. 1983. View Article : Google Scholar : PubMed/NCBI

24. 

Lipsky RH and Silverman SJ: Effects of mycophenolic acid on detection of glial filaments in human and rat astrocytoma cultures. Cancer Res. 47:4900–4904. 1987.PubMed/NCBI

25. 

Bocchini V, Casalone R, Collini P, et al: Changes in glial fibrillary acidic protein and karyotype during culturing of two cell lines established from human glioblastoma multiforme. Cell Tissue Res. 265:73–81. 1991. View Article : Google Scholar

26. 

Veselska R, Kuglik P, Cejpek P, et al: Nestin expression in the cell lines derived from glioblastoma multiforme. BMC Cancer. 6:322003. View Article : Google Scholar : PubMed/NCBI

27. 

Jiang Y and Uhrbom L: On the origin of glioma. Ups J Med Sci. 117:113–121. 2012. View Article : Google Scholar

28. 

Krupkova O, Loja T, Zambo I, et al: Nestin expression in human tumors and tumor cell lines. Neoplasma. 57:291–298. 2010. View Article : Google Scholar : PubMed/NCBI

29. 

Bigner DD, Bigner SH, Ponten J, et al: Heterogeneity of Genotypic and phenotypic characteristics of fifteen permanent cell lines derived from human gliomas. J Neuropathol Exp Neurol. 40:201–229. 1981. View Article : Google Scholar : PubMed/NCBI

30. 

Roversi G, Pfundt R, Moroni RF, et al: Identification of novel genomic markers related to progression to glioblastoma through genomic profiling of 25 primary glioma cell lines. Oncogene. 25:1571–1583. 2006. View Article : Google Scholar : PubMed/NCBI

31. 

Dahlback HS, Brandal P, Meling TR, et al: Genomic aberrations in 80 cases of primary glioblastoma multiforme: Pathogenetic heterogeneity and putative cytogenetic pathways. Genes Chromosomes Cancer. 48:908–924. 2009. View Article : Google Scholar

32. 

Rey JA, Bello MJ, de Campos JM, et al: Chromosomal patterns in human malignant astrocytomas. Cancer Genet Cytogenet. 29:201–221. 1987. View Article : Google Scholar : PubMed/NCBI

33. 

Bigner SH, Mark J and Bigner DD: Cytogenetics of human brain tumors. Cancer Genet Cytogenet. 47:141–154. 1990. View Article : Google Scholar : PubMed/NCBI

34. 

Kruse CA, Mitchell DH, Kleinschmidt-DeMasters BK, et al: Characterization of a continuous human glioma cell line DBTRG-05MG: growth kinetics, karyotype, receptor expression, and tumor suppressor gene analyses. In Vitro Cell Dev Biol. 28A:609–614. 1992. View Article : Google Scholar : PubMed/NCBI

35. 

Gjerset RA, Fakhrai H, Shawler DL, et al: Characterization of a new human glioblastoma cell line that expresses mutant p53 and lacks activation of the PDGF pathway. In Vitro Cell Dev Biol Anim. 31:207–214. 1995. View Article : Google Scholar : PubMed/NCBI

36. 

Cancer Genome Atlas Research Network: Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature. 455:1061–1068. 2008. View Article : Google Scholar : PubMed/NCBI

37. 

Uhrinova S, Uhrin D, Powers H, et al: Structure of free MDM2 N-terminal domain reveals conformational adjustments that accompany p53-binding. J Mol Biol. 350:587–598. 2005. View Article : Google Scholar : PubMed/NCBI

38. 

Esteller M, Garcia-Foncillas J, Andion E, et al: Inactivation of the DNA-repair gene MGMT and the clinical response of gliomas to alkylating agents. N Engl J Med. 343:1350–1354. 2000. View Article : Google Scholar

39. 

Hegi ME, Diserens AC, Gorlia T, et al: MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med. 352:997–1003. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

2014-March
Volume 44 Issue 3

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
Notarangelo A, Trombetta D, D'Angelo V, Parrella P, Palumbo O, Storlazzi CT, Impera L, Muscarella LA, La Torre A, Affuso A, Affuso A, et al: Establishment and genetic characterization of ANGM-CSS, a novel, immortal cell line derived from a human glioblastoma multiforme. Int J Oncol 44: 717-724, 2014
APA
Notarangelo, A., Trombetta, D., D'Angelo, V., Parrella, P., Palumbo, O., Storlazzi, C.T. ... Zelante, L. (2014). Establishment and genetic characterization of ANGM-CSS, a novel, immortal cell line derived from a human glioblastoma multiforme. International Journal of Oncology, 44, 717-724. https://doi.org/10.3892/ijo.2013.2224
MLA
Notarangelo, A., Trombetta, D., D'Angelo, V., Parrella, P., Palumbo, O., Storlazzi, C. T., Impera, L., Muscarella, L. A., La Torre, A., Affuso, A., Fazio, V. M., Carella, M., Zelante, L."Establishment and genetic characterization of ANGM-CSS, a novel, immortal cell line derived from a human glioblastoma multiforme". International Journal of Oncology 44.3 (2014): 717-724.
Chicago
Notarangelo, A., Trombetta, D., D'Angelo, V., Parrella, P., Palumbo, O., Storlazzi, C. T., Impera, L., Muscarella, L. A., La Torre, A., Affuso, A., Fazio, V. M., Carella, M., Zelante, L."Establishment and genetic characterization of ANGM-CSS, a novel, immortal cell line derived from a human glioblastoma multiforme". International Journal of Oncology 44, no. 3 (2014): 717-724. https://doi.org/10.3892/ijo.2013.2224