Functional analysis of HOXA10 and HOXB4 in human medulloblastoma cell lines

  • Authors:
    • Ricardo Bonfim-Silva
    • Fernanda Ursoli Ferreira Melo
    • Carolina Hassibe Thomé
    • Kuruvilla Joseph Abraham
    • Fábio Augusto Labre De Souza
    • Fernando Silva Ramalho
    • Hélio Rubens Machado
    • Ricardo Santos De Oliveira
    • Angelo A. Cardoso
    • Dimas Tadeu Covas
    • Aparecida Maria Fontes
  • View Affiliations

  • Published online on: October 10, 2017     https://doi.org/10.3892/ijo.2017.4151
  • Pages: 1929-1940
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Medulloblastoma (MB) is a malignant childhood brain tumor which at molecular level is classified into at least four major subtypes: WNT, SHH, group C and group D differing in response to treatment. Previous studies have associated changes in expression levels and activation of certain HOX genes with MB development. In the present study, we investigate the role of HOX genes in two attributes acquired by tumor cells: migration and proliferation potential, as well as, in vivo tumorigenic potential. We analyzed UW402, UW473, DAOY and ONS-76 human pediatric MB cell lines and cerebellum primary cultures. Two-color microarray-based gene expression analysis was used to identify differentially expressed HOX genes. Among the various HOX genes significantly overexpressed in DAOY and ONS-76 cell lines compared to UW402 and UW473 cell lines, HOXA10 and HOXB4 were selected for further analysis. The expression levels of these HOX genes were validated by real-time PCR. A mouse model was used to study the effect of the HOXA10 and HOXB4 genes on the in vivo tumorigenic potential and the in vitro proliferative and migration potential of MB cell lines. Our results show that the inhibition of HOXA10 in DAOY cell line led to increased in vitro cell migration while in vitro cell proliferation or in vivo tumorigenic potential were unaffected. We also observed that induced expression of HOXB4 in the UW473 cell line significantly reduced in vitro cell proliferation and migration capability of UW473 cells with no effect on the in vivo tumorigenicity. This suggests that HOXA10 plays a role in migration events and the HOXB4 gene is involved in proliferation and migration processes of medulloblastoma cells, however, it appears that these genes are not essential for the tumorigenic process of these cells.

Introduction

Medulloblastoma (MB) represents a genetically and epigenetically heterogeneous group of neuroepithelial primary tumors arising from the cerebellum (1). The World Health Organization (WHO) has classified four histological variants of MB which include classic, desmoplastic nodular (MB/N), MB with extensive nodularity (MBEN) and large cell/anaplastic type (LC/A) (2). Later, Garré et al proposed the classification of MB into two groups, standard-risk (SR) and high-risk (HR) groups (3,4) according to the risk-adapted treatments. In 2012, an international collaborative microarray study established four molecular subgroups in MB (WNT, SHH, group 3 and 4) (5,6). It is noteworthy that Gibson et al (7) found that WNT- and SHH-subtype medulloblastoma have different anatomical locations. For example, the WNT-Subtype is located within the IV ventricle and arises at the dorsal surface of the brain-stem, while the SHH-subtype arises within the cerebellar hemispheres. Current treatment protocols are dependent on the age of patients; for patients older than 36 months of age options include maximal safe tumor resection followed by craniospinal radiotherapy and chemotherapy. Infants younger than 36 months do not receive radiotherapy due the risk of severe side-effects (8). Cure rates are dependent on the MB molecular subtype. Patients with MB-WNT have a cure rate of over 90%, while in patients with MB-group 3 40–60% face chemo or radio-resistance or relapse (8,9). This variation in treatment response is a motivation to better understand the biology of these tumors, including gene expression patterns, in order to increase cure rates and/or to reduce treatment-related sequelae.

An earlier comprehensive sequence analysis study in 88 pediatric MB samples led to the discovery of a gene, histone-lysine N-methyltransferase MLL2, not previously known to be altered in MBs (10). In 14% of MB patients in the same study somatic mutations were observed in MLL2, which is known to be a member of the trithorax complex that induces epigenetic marks that correlate with accessible euchromatin (11). The connection between MLL2 alterations and tumorigenesis has not been investigated in medulloblastoma. One possibility is that, the MLL2 gene (new approved symbol KMT2D), which codifies the MLL2 protein and which plays a role in maintaining HOX gene expression (10,12) might deregulate HOX gene expression in cancer. HOX genes encode evolutionarily conserved homeodomain-containing transcription factors and in humans there are four clusters named HOXA, HOXB, HOXC and HOXD, which are located on chromosomes 7p14, 17a21, 12q13 and 2q31, respectively.

An additional motivation for studying HOX gene expression in MB arises from the role of HOX genes in cerebellum development. As described in Wang and Zoghbi (13), cerebellum development commences from the dorsal region of the posterior neural tube. Two of the regions of the posterior neural tube, the mesencephalon and the metencephalon depend on molecular signals received from the Isthmus Organizer (IO). The Otx-2 (MB oncogene) plays a role in IO development while various genes in HOX family affect metencephalon development (13).

Moreover, the expression of HOX genes is not limited to the embryonic phase of development (14). Takahashi et al (15) studied the expression profiles of 39 HOX genes in 20 different human adult normal organs by real-time PCR. They showed that in the cerebellum six HOX genes are expressed, among them: HOXB2, HOXB3, HOXB4, HOXC4, HOXD1 and HOXD3. Other studies have investigated the expression of HOX genes in cerebellum suggesting that the number of HOX genes transcribed in the cerebellum can be even higher. For example, recently, Hutlet et al (16) demonstrated low expression of 21 specific HOX genes in adult mice cerebella, all belonging to paralogy groups (PG) 1–8 (16). These data suggest complex patterns of HOX expression in the cerebellum, which in turn suggest the relevance of multiple HOX gene regulation mechanisms. These mechanisms include the use of multiple promoters, alternative splicing, antisense transcripts, long non-coding RNA, miRNA, as well as some translation controls (16,17).

In some cancers, certain HOX genes may function as oncogene or tumor suppressor genes (18). For example, in prostate, breast and colorectal cancers loss-of-function of HOXB13 mutations suggests that HOXB13 may function as a tumor suppressor gene (19). On the other hand, in breast and colorectal cancer, upregulation of HOXB7 promotes cell proliferation, which implies that it might function as an oncogene (20,21). Apart from loss of function, additional progress has been made in understanding changes in expression levels and the activation of certain HOX genes and cancer development (22,23). In brain tumors, the deregulation of HOX genes has been described in glioblastoma (2427) neuroblastoma (28,29), tumor atypical teratoid rhabdoid, juvenile pilocytic astrocytoma and ependymomas (30). In medulloblastoma, Bodey et al (31) observed by immunocytochemistry techniques that HOXB3, HOXB4 and HOXC6 proteins were expressed in medulloblastoma tissues. In 2014, Chakravadhanula et al (30) reported that HOXC4-6, HOXC8-13, HOXD3-4, HOXD8-11 and HOXD13 genes were upregulated and HOXD1 gene was downregulated in medulloblastoma tissues when compared to control tissues.

Recently, in order to elucidate epigenetic changes that contribute to medulloblastoma progression, Vo et al (32) demonstrated through the use of engineered deletions of Ezh2 by gene editing nucleases, that inactivation of Ezh2 drives aggressive medulloblastoma. Ezh2 is a member of the polycomb complex known to induce marks that correlate with heterochromatinization and mediate repression of HOX genes (33). In addition to repression, the authors showed that loss of Ezh2 is also accompanied by upregulation of several HOX genes (32).

These studies suggest that several epigenetic mechanisms of HOX regulation might play a role in medulloblastoma. However, the role of HOX gene dysregulation in medulloblastoma has not been investigated. In 2011, a seminal study (34) suggested ten attributes which are acquired by tumor cells and which also sustain the malignant behavior in various types of cancer. In this study, we investigate the role of two specific HOX genes, HOXA10 and HOXB4, in two of these attributes in meduloblastoma: migration and proliferation potential.

Materials and methods

Cell lines

Four independent medulloblastoma cell lines (UW402, UW473, DAOY and ONS-76) were used in this study. UW402 and UW473 cell lines, have been previously characterized regarding chromosomal heterogeneity, instability profiles, cell morphology, cell population doubling time, colony-forming efficiency as well as chemo-sensitivity heterogeneity (35). Both DAOY and ONS-76 cell lines are long-established MB cell lines (3638). The ONS-76, UW402 and UW473 cell lines were kindly provided by the Laboratory of Pediatric Oncology of the Clinical Hospital of the Medical School of Ribeirão Preto (CHMSRP), while the DAOY cell line was purchased from the American Type Culture Collection (ATCC: HTB-186; ATTC, Manassas, VA, USA).

The three cerebellum primary cultures (CP4, CP5 and CP6) were generated from cerebellum specimens that were harvested from infants at autopsy in the Pathology Service of the CHMSRP. CP4 was obtained from cerebellum of a full term newborn who died at two months old. CP5 and CP6 were collected from the cerebella of two infants born at 22 and 23 weeks of gestation, respectively, who died soon after birth. The causes of the babies' deaths were not brain related.

The primary cells were cultured in Dulbecco's modified Eagle's medium (DMEM)-F12 medium (Gibco, Grand Island, NY, USA) with 10% fetal bovine serum (FBS; HyClone Laboratories, Inc., Logan, UT, USA), the same medium in which UW402 and UW473 cell lines were cultured. The DAOY cell line was cultured in a-MEM (Gibco) 10% FBS and ONS-76 cell line was cultured in RPMI-1640 medium (Gibco) 10% FBS in an incubator at 37°C and 5% CO2. All culture media contained penicillin at 100 units/ml and streptomycin 100 μg/ml (Gibco). The study was approved by the Ethics Research Committee of the Clinical Hospital of Ribeirão Preto and Medical School of Ribeirão Preto of the University of São Paulo under protocol number 14920/2011. All the animal procedures of the present study were conducted in agreement with the ethical principles in animal research adopted by the Brazilian College of Animal Experimentation (COBEA) and this study, under protocol number 137/2011, was also approved by the Medical School of Ribeirão Preto of the University of São Paulo, Ethics Commission of Ethics in Animal Research (CETEA).

Preparation of total RNA and microarray experiments

Total RNA was isolated from cerebellum primary cultures and medulloblastoma cell lines using RNeasy Mini kit according to the manufacturer's instructions (Qiagen, Hilden, Germany). The quantity and purity of the total RNA was determined by spectrophotometer (NanoDrop; Thermo Fisher Scientific, Waltham, MA, USA) and samples with OD 260/289 between 1.8 and 2.0 were used. In order to check the integrity of the RNA 500 ng of RNA with loading-buffer (xylene cyanol containing gelred fluorescente dye from Biotium) was loaded in a 1% miniagarose gel in Tris-Acetate-EDTA buffer. After electrophoresis, the gel was analyzed using the ImageQuant 350 transilluminator (GE Healthcare) with short-wave-length UV illumination to visualize 28S and 18S bands. The integrity of the total RNA was also evaluated by microfluidic-based electrophoresis (Agilent 2100 Bioanalyzer; Agilent Technologies, Santa Clara, CA, USA) and samples with RIN >7.0 were used for microarray experiments.

A total of 200 ng of RNA of samples and reference were reverse transcribed by the Low RNA Input linear amplification kit and then transcribed to Cy3-labelled (samples) or Cy5-labelled (reference) according to the manufacturer's instructions (Agilent Technologies). The protocols used were described in de Oliveira et al (39). Briefly, the reverse transcription reaction was incubated at 40°C for 2 h and then incubated at 70°C for 15 min to degrade the excessive RNA. Next, in vitro transcription was carried out using the T7 RNA polymerase and the dyes Cy3 and Cy5. This reaction was incubated at 40°C for 2 h. After labeling and purification (Mini Spin kit; GE Healthcare), the efficiency of Cy3 and Cy5 labeling was quantified using the NanoDrop 1000 spectrophotometer (Thermo Fisher Scientific). The numerical values for the labeling efficiency were determined by the base-to-dye ratios of Cy3-dCTP and Cy5-dCTP per μg of cRNA. The values obtained were consistent with those recommended for hybridization onto cDNA microaarrays. Next, the labeled samples and reference were fragmented according to the manufacturer's instructions and the hybridization buffer added. After the arrays were loaded, the arrays were incubated at 65°C for 17 h for hybridization.

Dye normalization was performed by the linear lowess method. For the spike in controls, the expected log ratio was plotted against the observed log ratio as a quality control check. For each lamina, the log ratio was plotted against the log processed signal to ensure that the signal strength distribution for upregulated and downregulated features were comparable.

Microarray data analysis

The raw data were obtained from a two-color Agilent platform (SurePrint G3 Human Gene Expression 8×60K Microarray kit; G4851A) using DAOY1 as a common reference. Quantile normalization was performed on the raw data after hybridization followed by normalization between arrays to obtain the gene expression levels. Expression levels for the various HOX genes were obtained by mapping probe names to the HOX genes using the NCBI GenBank. Whenever there were multiple probes present for the same HOX gene, the average over all probes was taken to determine the expression level for that particular gene.

All heatmaps were generated using the gplots package of Bioconductor. The identification of differentially expressed genes was performed using the Limma package in Bioconductor. A significance threshold of 0.05, along with P-values adjusted according to the Benjamini-Hochberg method, was used to determine which genes were differentially expressed. Anova contrasts were used to identify differentially expressed genes in the various comparisons shown in Fig. 1C and D. All P-values were converted to their log to the base 10 equivalents before use in Fig. 1C and D. The log fold change values used in the figures were those obtained from the Limma output.

The raw data from high-throughput microarray experiments are available on the Gene Expression Omnibus (GEO) website under accession number GSE95684.

Real-time PCR

The cDNA was directly synthesized from 1 μg total RNA from cerebellum primary cultures and medulloblastoma cell lines using the High Capacity cDNA Reverse Transcription kit (Life Technologies, Carlsbad, CA, USA) according to the manufacturer's protocol. Each 25 μl cDNA synthesis reaction solution contained 1 μg of total RNA, 1X buffer, 1X random primers, dNTP (8 mM), RNase inhibitor (2 U) and 62 U of MultiScribe Reverse Transcriptase (Life Technologies). The cDNA synthesis was carried out with 1 cycle at 25°C for 10 min, 1 cycle at 37°C for 120 min followed by heating to 85°C for 5 min to terminate the reaction. Gene expression of the two HOX genes were quantified using the TaqMan Universal Master Mix as previously reported by Covas et al (40) and Fontes et al (41). Briefly, cDNA (2 μl of cDNA diluted 1:10) was applied in the real-time PCR assay with TaqMan probes (Applied Biosystems, Waltham, MA, USA) HOXA10 (Hs00172012_m1), HOXB4 (Hs00256884_m1) and the TaqMan Universal Master Mix according to the manufacturer's protocol. Beta actin (ACTB) gene (4310881E) was used as reference gene. Cycling was done for 2 min at 50°C, followed by denaturation at 95°C for 10 min. The amplification was carried out with 40 cycles of 15 sec at 95°C and 60 sec at 60°C. We obtained the number of amplification cycles needed for reaching the threshold fluorescence within the log-linear phase of the amplification curves after setting the appropriate baseline in each color channel of the GeneAmp® 7500 software using the manual baseline method. The relative expression level of target gene was shown as expression relative units (ERU) and was quantified according to the formula ERU = 10,000/2dCT following Albesiano et al (42), where ΔCT = CT target sample - CT reference sample. As in the study by Albesiano, the reference gene level (in our case ACTB level) is set equal 10,000. For each sample in Fig. 2A and B, cDNA was independently synthesized three times, and two technical replicates were carried out on each batch of independently synthesized cDNA. The same procedure was followed for the samples in Fig. 3C and E.

Gene silencing

The lentiviral particles (Santa Cruz Biotechnology, Santa Cruz, CA, USA) used contained the sequences of short hairpin for human HOXA10 gene (shHOXA10), the green fluorescent protein (GFP) reporter gene and the resistance gene to the puromycin antibiotic (PUR), described as shHOXA10-GFP-PUR, and the empty vector control containing only the GFP and PUR gene sequences (sh-GFP-PUR). The DAOY cell line was transduced using hexadimethrine bromide (6 μg/ml; Sigma-Aldrich, St. Louis, MO, USA) at a multiplicity of infection (MOI) of 5. After 24 h, the cells were treated with puromycin antibiotic (1 μg/ml; Sigma-Aldrich, Hamburg, Germany) for 6 days. Cell lines were identified as DAOY/shHOXA10+ and DAOY/control. The cell transduction efficiency was evaluated by fluorescence microscopy and flow cytometry for GFP, and gene silencing efficiency was evaluated by real-time PCR and western blot analysis.

Gene overexpression

The human HOXB4 gene was over-expressed in UW473 the cell line by the cell transduction method. The vector used contained the sequences of human HOXB4 gene, the GFP reporter gene and the resistance gene to the antibiotic puromycin, described as HOXB4-PUR-GFP (VB150311-10017; Cyagen Biosciences, Santa Clara, CA, USA). As a control, we used an empty vector containing only the GFP and PUR gene sequences (VB150311-10018; Cyagen Biosciences). Vectors related to the viral capsid (8.91) and viral envelope (VSV-G) were used as accessory vectors for the production of lentiviruses by transfection of the 293FT cell line using Lipofectamine™ 2000 (Invitrogen, Carlsbad, CA, USA) according to the manufacturer's protocol. The UW473 cell line was transduced with HOXB4-GFP-PUR and control lentiviral particles with a MOI of 2. After 24 h, the cells were treated with puromycin antibiotic (1 μg/ml; Sigma-Aldrich) for 6 days. Cell lines were identified as UW473/HOXB4+ and UW473/control. The cell transduction efficiency was evaluated by fluorescence microscopy and flow cytometry for GFP, and HOXB4 overexpression efficiency was evaluated by real-time PCR and western blot analysis.

Western blot analysis

Total protein was extracted using RIPA buffer (Sigma-Aldrich, St. Louis, MO, USA) supplemented with protease inhibitor (Complete Mini Protease Inhibitor Cocktail; Roche Diagnostics GmbH, Mannheim, Germany) and phosphatase (PhosSTOP phosphatase inhibitor cocktail; Roche Diagnostics), and quantified using the BCA protein assay kit (Pierce, Rockford, IL, USA) according to the manufacturer's protocol. Proteins were separated by SDS-PAGE using 10% polyacrylamide gels (Mini-PROTEAN TGX; Bio-Rad Laboratories, Hercules, CA, USA), transferred from the gel to a nitrocellulose membrane (40 μm, Hybond-C extra; Amersham Biosciences, Little Chalfont, UK) and incubated in a 5% skim milk blocking solution. The following primary antibodies were used: mouse anti-human HOXA10 (E-12) (1:250, sc-271139; Santa Cruz Biotechnology), mouse anti-human HOXB4 (D-1) (1:500, sc-365927; Santa Cruz Biotechnology), rabbit anti-human β-actin (1:4,000, #4967; Cell Signaling Technology, Danvers, MA, USA) and rabbit anti-human GAPDH (14C10) (1:4,000, #2118; Cell Signaling Technology). Mouse anti-sheep IgG (1:4,000, NA931V; GE Healthcare, Aurora, OH, USA) and goat anti-mouse IgG rabbit (1:4000, #7074; Cell Signaling Technology) HRP-conjugated secondary antibodies were used. The proteins bands on the membrane were developed using ECL Western Blotting Detection Reagent Prime kit (GE Healthcare) according to the manufacturer's instructions. As mentioned by Ferguson et al (43) β-actin and β-tubulin proteins (both being part of the cytoskeleton of eukaryotic cells), and GAPDH enzyme (glyceraldehyde-3-phosphate dehydrogenase), are classically used as internal standards for normalization of signals in western blot analysis in order to compare differences in the expression of the target proteins among samples and eliminating variations arising from technical reasons, such as differences in amount of total load protein (43). The protocols have been previously used by various co-authors of this study, as described in Thomé et al (44) (β-actin) and Palma et al (45) who have used both these control antibodies. As in this study, in Palma et al (45) both control antibodies, β-tubulin and GAPDH, were used.

In vitro proliferative and migration potential assay

The proliferative potential was assessed by cell counting in a Neubauer chamber. Initially, 8×104 viable cells were cultured in 75-cm2 flask for a period of 120 h. Next, the cells were trypsinized and viable cells were counted in the Neubauer Chamber with trypan blue in quadruplicate. The proliferative potential (PP) was calculated as PP = N - No, in which N is the final number of cells and No is the initial number of cells.

Scratch assay was used to evaluate the migration potential. Cells were cultured in 6-well plates (in triplicate for each cell line), and when they reached ~95% confluence a scratch was made. The wells were photodocumented by phase contrast microscopy (Olympus IX71 and PD controller software, USA) in two different points along the scratch in each well, and identified as time 0. After 16 or 20 h, the wells were again photodocumented at the same positions using the TScratch software (software developed by Tobias Gebäck and Schulz, ETH Zurich, Switzerland). The percentage of scratch area was measured in the images of initial and final time. The migration potential was calculated as percentage of migrated area (%MA), %MA = 1- (N/No), in which N is the final area (%) and No is the initial area (%).

In vivo tumorigenic potential assay

Male nude mice weighing 20–25 g were obtained from the Central Animal House of the University of São Paulo (Ribeirão Preto, Brazil), and were housed at 23±2C with 12-h light/dark cycle. This experimental protocol was approved by the Animal Research Ethical Committee of the Medical School of Ribeirão Preto, USP (protocol number 137/2011).

For the evaluation of the tumorigenic potential, 3×106 cells of medulloblastoma cell lines in 100 μl of Matrigel (Matrigel® Matrix; Corning, Inc., Corning, NY, USA) were infused subcutaneously into the backs of nude mice, which were previously anesthetized with a mixture of 2% isoflurane and oxygen. The mice were monitored in the periods 7, 15, 30 and 60 days after cell infusion, for the detection and quantification of GFP fluorescence using the in vivo imaging system IVIS Lumina and Living Image software (Perkin-Elmer, Waltham, MA, USA); and for measurement of the nodular Volume using a digital caliper. The nodular volume (NV) was calculated as: NV= L × W × H × 4/3 π, where L is the length, W is the width and H is the nodule height.

Statistical analysis

The Student's t-test and the one-way ANOVA test were used to evaluate the HOX expression level differences, as well as migration and proliferation potential, before and after genetic modification of the MB cell lines. For these assays, statistical tests were performed using the GraphPad Prism 5 for Mac OS X. All results were expressed as mean ± standard deviation. R was used for all other statistical analyses. Tests were declared statistically significant for P<0.05.

Results

Higher numbers of HOX genes are significantly deregulated in DAOY and ONS-76 cell lines compared to UW473 and UW402 cell lines

In Fig. 1 we show the results of hierarchical clustering of 39 HOX genes with tumor cell lines which are not characterized (UW473 and UW402 cell lines) and primary cell cultures from the cerebellum (CP4, CP5 and CP6), and also 39 HOX genes with tumor cell lines which are characterized as being from the SHH subgroup (ON-S76 and DAOY cell lines) (Fig. 1A and B) (35,46,47).

In both dendograms tumor cell lines and primary cell cultures fall into two distinct groups. For the DAOY and ONS-76 cell lines this distinction is even clearer producing two distinct clusters (Fig. 1B). We observed that in DAOY and ONS-76 cell lines several HOX genes are expressed, while these genes are not expressed in primary cell cultures. As regards the other two cell lines, we observed a distinction between HOX gene profiles in UW473 and UW402 cell lines. HOX genes are more expressed in UW402 cell line compared with UW473 cell line (Fig. 1A).

To visualize the significantly dysregulated HOX genes we constructed the volcano plot. We observed that in the UW402 and UW473 cell lines only 6 HOX genes are significantly upregulated (HOXB9, HOXB13, HOXC6, HOXC13, HOXD10 and HOXD11) compared with the primary cell lines from cerebellum (Fig. 1C). On the other hand, in the DAOY and ONS-76 cell lines 24 HOX genes are significantly upregulated: nine from HOXA group (HOXA2, HOXA3, HOXA4, HOXA5, HOXA6, HOXA7, HOXA9, HOXA10 and HOXA13), six from HOXB group (HOXB2, HOXB4, HOXB5, HOXB6, HOXB7 and HOXB9), six from HOXC group (HOXC4, HOXC5, HOXC6, HOXC9, HOXC10 and HOXC13) and three from HOXD group (HOXD3, HOXD10 and HOXD11) (Fig. 1D). Out of these 24 HOX genes, HOXA3 was the most dysregulated gene, however, upregulation of this gene has not been demonstrated in medulloblastoma or other tumor from the central nervous systems (CNS). These results are summarized in Table I, which contains all log fold change values. Gene names in bold face are those mentioned above as being statistically significantly upregulated.

Table I

Comparison of fold change using microarray data set from HOX genes in UW402 and UW473 vs. cerebellum primary cells and DAOY and ONS-76 vs. cerebellum primary cells.

Table I

Comparison of fold change using microarray data set from HOX genes in UW402 and UW473 vs. cerebellum primary cells and DAOY and ONS-76 vs. cerebellum primary cells.

UW473 and UW402
DAOY and ONS-76
GenesLog fold changeGenesLog fold change
HOXA1−0.569958863HOXA1−0.597828379
HOXA20.095698182HOXA22.656257452
HOXA32.500799762HOXA35.535882266
HOXA40.803518199HOXA43.429616377
HOXA51.091665053HOXA52.184209412
HOXA61.611731106HOXA62.779732951
HOXA71.371254281HOXA72.825761681
HOXA9−0.262664551HOXA91.664946587
HOXA100.131334357HOXA101.866723269
HOXA110.143180662HOXA111.846451081
HOXA132.670226837HOXA133.755042843
HOXB10.946528446HOXB10.324546332
HOXB2−0.182254131HOXB23.150953293
HOXB32.293320983HOXB31.472715878
HOXB40.483773574HOXB44.027400665
HOXB50.726902915HOXB54.049698398
HOXB62.769873572HOXB63.992872898
HOXB71.744203467HOXB72.169081951
HOXB80.213373435HOXB82.286976211
HOXB93.155648057HOXB94.359008666
HOXB134.43490889HOXB132.78047806
HOXC42.657559554HOXC44.752524627
HOXC50.994376614HOXC51.697798782
HOXC64.164237557HOXC63.346096371
HOXC81.042329918HOXC81.991863755
HOXC92.852715702HOXC94.663234774
HOXC102.095372335HOXC102.858686398
HOXC11−0.378263228HOXC11−0.496091564
HOXC120.317817366HOXC121.876619216
HOXC134.232848464HOXC133.531212456
HOXD10.307750142HOXD11.833684289
HOXD30.779984978HOXD31.743693697
HOXD40.924886063HOXD41.133382434
HOXD81.49312174HOXD82.089211292
HOXD91.620166724HOXD91.288963756
HOXD103.44561321HOXD104.045820474
HOXD112.529309801HOXD112.805492204
HOXD120.064926757HOXD120.579149569
HOXD131.71540358HOXD133.112325861

From the results of these analyses, we continued the present study with focus on two genes HOXA10 and HOXB4, which are significantly upregulated in DAOY and ONS-76 cell lines but not in UW402 and UW473. In mice MB-derived cancer stem cell (CSC), HOXA10 has been found to be one of the top-ranking genes highly expressed in highly tumorigenic (HT)-CSCs. Also, HOXA10 was found to have a high protein expression level in glioblastoma-derived neurospheres, and is associated with poor outcomes and treatment resistance in glioblastoma multiforme (GBM) (48). Moreover, the HOXA10 locus was associated with expression of a stem cell related HOX-signature in glioblastoma and with temozolomide resistance (49). HOXB4 was found to be expressed in medulloblastoma tissues (31) and recently, loss of Ezh2, an epigenetic regulator of HOX genes, drives aggressive medulloblastoma and was accompanied by upregulation of HOXB4 (32).

We validated the microarray data of HOXA10 and HOXB4 genes and compared gene expression levels by RT-qPCR in these four MB cell lines and control cells. As seen from Fig. 2A and B, HOXA10 and HOXB4 are upregulated in ONS-76 and DAOY, consistent with the results from the microarray analysis. The analysis showed that HOXA10 and HOXB4 transcripts are not expressed in primary cerebellum cells. Previous studies by Takahashi et al (15) and Hutlet et al (16) have demonstrated the absence of HOXA10 expression in human and mouse cerebellum tissues. These authors also showed the expression of HOX genes belonging to the 8 paralogy groups (PG1-8), which includes HOXB4, but not HOXA10. Therefore, one possible explanation for the absence of the HOXB4 expression in our primary cerebellum culture could be due to the differences between tissues and isolated cells. In cell therapy studies differences in gene expression between tissues and isolated cells have been previously observed (40,50). In our analysis of cell lines, HOXA10 transcripts are not expressed in two of the MB cell lines, UW402 and UW473. In the ONS-76 and DAOY cell lines, the observed expression levels of HOXA10 are 502.4±16.07 and 1305±22.42 URE, respectively (Fig. 2A). As regards the HOXB4 transcripts, they are expressed at low level in the UW402 and UW473 cell lines 1.05±0.16 URE and 1.072±0.33 URE, respectively. In the ONS-76 and DAOY cell lines, the expression levels of HOXB4 transcripts are 178.4±15.85 URE and 192.5±1.97 URE, respectively (Fig. 2B).

To better understand the classification of these four MB cell lines, we examine the expression levels of 22 signature genes proposed by Northcott et al (6) for molecular characterization of MB subgroups (Shh, Wnt, group C and group D). As shown Fig. 2C, we observed that under our culture conditions, in the DAOY cell line two Shh markers (EYA1 and PDL) but also the Wnt marker (WIF1) and the group C marker (GABRA5) are expressed. In the ONS-76 cell line the Shh marker (HHIP) and the group C markers (MAB21L2, EYS and NPR3) are expressed. On the other hand, in the UW473 cell line, genes from the four subgroups [GAD1 (from WNT), EYA1 [from Shh], EYS and NPR3 (from group C)] and OAS1 and RBM24 (from group D) are expressed and in the UW402 cell line genes from four subgroups [TNC and GAD1 (from Wnt), SFRP1 (from Shh), IMPG2 (from group C) and EOMES (from group D)] (Fig. 2C) are expressed.

Next, we investigated whether or not the HOXA10 gene has a direct effect on the in vivo tumorigenic potential and on the in vitro proliferative and migration potential of DAOY cell line by knockdown of this transcript. We also investigated whether or not HOXB4 transcripts have a direct effect on the in vivo and the in vitro properties in UW473 cell line after overexpression of this gene.

HOXA10 knockdown increases the migration potential but does not affect the proliferative potential of the DAOY medulloblastoma cell line

The HOXA10 gene was silenced in the DAOY cell line by shHOXA10 and an empty vector was used as control. The transduction efficiency was evaluated by fluorescence microscopy and flow cytometry, which showed that >95% of DAOY/shHOXA10+ cells were GFP+ (Fig. 3A). The HOXA10 knockdown was confirmed; the HOXA10 mRNA levels were reduced by 78% in comparison with the DAOY-control cells (268.1±16.25 vs. 59.20±2.1 URE) (P=0.0002) (Fig. 3C). The HOXA10 protein levels were also reduced in DAOY/shHOXA10+ cells (Fig. 3D). Analyzing the migration potential, the DAOY/shHOXA10+ cells displayed showed a migrated area 37±8% bigger when compared to DAOY-control cells (P=0.0046) (Fig. 4A and B). However, no difference was observed in the proliferative potential in which DAOY/shHOXA10+ and DAOY/control cells showed a similar amount of viable cells after 120 h of cell culture (P=0.868) (Fig. 4C). This result suggests that the low expression of HOXA10 gene is associated with an increase in the in vitro migration potential; however, there is no relationship with the in vitro proliferative potential of the DAOY medulloblastoma cell line.

HOXB4 overexpression decreases the proliferative and migration potential of the UW473 medulloblastoma cell line

The HOXB4 gene was overexpressed in the UW473 cell line and the transduction efficiency was evaluated by fluorescence microscopy and flow cytometry, which showed that >93% of cells were GFP+ in UW473/HOXB4+ and UW473-control cells (Fig. 3B). We also compared gene expression levels of HOXB4 by qPCR in UW473-control cells and in UW473/HOXB4+ cells. We observed that in UW473-control cells HOXB4 mRNA is present at a very low level (1.05±0.69 URE). However, in UW493/HOXB4+ cells the expression level of HOXB4 is 33.735±2.971 URE (P=0.0077) (Fig. 3E and F). Assessing the migration potential, the UW473/HOXB4+ cell line showed a migrated area 32±15% smaller when compared to the UW473-control cells (P=0.033) (Fig. 5A and B). The UW473/HOXB4+ cells line also showed a proliferative potential 34±6% lower compared to the UW473-control cells (P=0.032; Fig. 5C). These results suggest that the high expression of the HOXB4 gene is associated with a decrease in the in vitro proliferative and migration potential of the UW473 medulloblastoma cell line.

HOXA10 gene silencing and HOXB4 gene overexpression do not affect the tumorigenic potential of medulloblastoma cell lines

The effect of HOXA10 gene inhibition and HOXB4 gene overexpression on the tumorigenic potential of medulloblastoma cell lines was evaluated. DAOY/shHOXA10+, UW473/HOXB4+, DAOY-control and UW473-control cells were injected subcutaneously in mice and the GFP fluorescence intensity and tumor volume were monitored for a period of 7, 15, 30 and 60 days. After 60 days, the GFP fluorescence intensity (Fig. 6A and B) and tumor volume (Fig. 6C) were similar in DAOY-control and DAOY/shHOXA10+ cell lines with both cell groups giving rise to tumor nodules. No formation of tumor nodules was observed in either UW473-control or UW473/HOXA10+ cell lines, GFP fluorescence intensity (Fig. 6D and E) and tumor volume (Fig. 6F) were similar in both cell groups. These results demonstrate that the modulation of HOXA10 and HOXB4 gene expression did not change the tumorigenic potential of these medulloblastoma cell lines in nude mice.

Discussion

This study evaluated the differences in aberrant upregulated HOX gene expression among the UW402, UW473, DAOY and ONS-76 human medulloblastoma cell lines and investigated the role of two HOX genes (HOXA10 and HOXB4) in the in vitro proliferative and migration potential and in vivo tumorigenic potential of these cell lines. Moreover, medulloblastoma is a heterogeneous disease and 22 transcripts have been used to stratify MB subgroups (6). Therefore, in order to understand these differences according to MB subtypes we compared the 22 transcript signatures in these MB-derived cell lines.

This study shows that different groups of HOX genes are differentially expressed according to MB-derived cell line. We identify 6 HOX genes (HOXB9, HOXB13, HOXC6, HOXC13, HOXD10 and HOXD11) whose expression levels are significant upregulated between UW402 and UW473 cell lines and normal cerebellum primary cells; and 24 HOX genes (HOXA2, HOXA3, HOXA4, HOXA5, HOXA6, HOXA7, HOXA9, HOXA10, HOXA13, HOXB2, HOXB4, HOXB5, HOXB6, HOXB7, HOXB9, HOXC4, HOXC5, HOXC6, HOXC9, HOXC10, HOXC13, HOXD3, HOXD10 and HOXD11) whose expression levels are significantly different between DAOY and ONS-76 cell lines and normal cerebellum primary cells.

To the best of our knowledge this is the first study that analyzes the expression pattern of all 39 HOX genes in MB-derived cell lines. HOX mRNA expression has been detected in MB tissues, Chakravadhanula et al (30) examined 10 MB tissues and studied the expression levels of 21 HOX genes. They showed that 10 HOX genes (HOXC4-6, HOXC8-10, HOXD3-4, HOXD8 and HOXD10) were significantly upregulated in medulloblastoma tissues when compared to control tissues. Also, Bodey et al (31) observed by immunocytochemistry techniques that HOXB3, HOXB4 and HOXC6 proteins were expressed in medulloblastoma tissues. In this study some of these HOX genes are also significantly upregulated. Two of them (HOXC6 and HOXD10) are significantly upregulated in these four MB cell lines (UW473, UW402, DAOY and ONS-76) and another five (HOXC4-5, HOXC9-10 and HOXD3) were identified as significantly upregulated in DAOY and ONS-76 cells.

The noticeable change in expression levels of most HOX genes in DAOY and ONS-76 cell lines compared with UW402 and UW473 merits attention. In order to determine whether differences in HOX deregulation are related to MB subtype we used the 22-transcript predictor in these 4 MB cell lines. We observed that UW473 and UW402 cell lines express genes from all four MB groups, and DAOY and ONS-76 previously classified as SHH subtype (38) also express genes from the other MB subtypes. DAOY expresses SHH, WNT and group C markers and ONS-76 expresses SHH and group C markers. These results suggest that under our cell culture conditions these cell lines cannot be classified as belonging to a unique molecular subtype and therefore the interesting differences in HOX pattern expression among these four MB cell lines cannot be directly associated with MB subtype. A similar effect has been observed in the D238-MB cell line, which has been classified as group 4 or group 3 (51,52).

Among the HOX genes significantly overexpressed in DAOY and ONS-76 cell lines compared to the UW402 and UW473 cell lines we selected HOXA10 and HOXB4 as mentioned earlier. In mouse medulloblastoma (MB)-derived cancer stem cell (CSC) the role of miR-135a, which binds to HOXA10 3'UTR in other cancers, was investigated (53,54). Hemmesi et al (54) showed that induced overexpression of miR-135a in mouse MB-derived CSC was responsible for a significant decrease in the expression of HOXA10 at mRNA level, but not at the protein level. Moreover, considering that miR-135a restoration inhibits tumor progression in CSC-derived MBs with no change in HOXA10 protein level, which suggests that in MB-CSCs HOXA10 can contribute to the malignant properties of MB cells depending on genetic background. HOXA10 is also one of the HOX genes, which shows a markedly higher expression in glioblastoma-derived neurospheres and was associated with worse outcome in patients assigned to TMZ/RT-TMZ therapy (48). From our initial analysis which showed that HOXA10 is differentially overexpressed in DAOY and ONS-76 cell lines but not in UW402 and UW473 cell lines, we decided to investigate whether knockdown of HOXA10 in DAOY cell line will result in changes in vitro migration and proliferation potential changes and in vivo tumor growth. We observe that knockdown of HOXA10 does in fact lead to an increase in the in vitro migration potential of the DAOY medulloblastoma cell line, however, there was no relationship between the HOXA10 expression level and the in vitro proliferative and the in vivo tumorigenic potential of that cell line. Altogether, although the exact mechanism that accounts for these effects in MB is unknown, these data might indicate that HOXA10 could contain different epigenetic regulators and has a role in several physiological processes.

Regarding the HOXB4 gene, we investigated whether or not overexpression of HOXB4 in the UW473 cell line promotes proliferation, migration and in vivo MB tumorigenesis. In the present study, we showed that the HOXB4 expression level is associated with a decrease in the in vitro proliferative and migration potential of the UW473/HOXB4+ cell line, however, it did not influence the in vivo tumorigenic potential of that cell line. It was previously known that HOXB4 is expressed in MB tissues (31) but the effect of this gene in medulloblastogenesis was not investigated. A recent study by Vo et al (32) measured RNA levels of HOXB4 gene in MB cells after gene-editing systems to induce loss-of-function mutation in Ezh2. In these Ezh2-mutated cells, HOXB4 was expressed more highly and the tumor became more aggressive. Ezh2 is an epigenetic regulator of HOX expression as a member of polycomb-repressive complex 2 (PRC2) together with EED, SUZ12 and other accessory proteins. Altogether, these results suggest that in UW473/HOXB4+ cells additional changes at the genetic or epigenetic levels are required to inactivate essential genes necessary for normal cellular development and function and to induce tumorigenesis.

To conclude, the present study was motivated by an earlier study of the importance of MLL2 in MB and the observation that MLL2 is an epigenetic regulator of the HOX gene. From the results of the microarray analysis presented (which were validated by RT-qPCR), we selected two HOX genes for functional analysis in vitro and in a mouse model. The in vitro analysis showed that these genes could be involved in migration and proliferative potential. The in vivo study with mice was not conclusive, which may be due to a variety of different causes. Other members of the HOX gene family could be involved. Another possibility is that MLL2 deletions have an effect on MB, but via other genes for example the MB oncogene named OTX2 or β-catenin known to regulate Wnt pathways, and which is implicated in one of the medulloblastoma subtype. Studies similar to the one we have undertaken should be extended to other genes and could help identify potential drug targets.

Acknowledgments

We thank the Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq) (Process no. 310619/2012-2) and Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP) (Process nos. 2011/20829-4 and 2011/18664-7) for the financial support for the present study. We also thank Sandra Navarro Bresciani of the Regional Blood Center of Ribeirão Preto-FMRP-USP, Brazil for help with the figures.

References

1 

Gilbertson RJ and Ellison DW: The origins of medulloblastoma subtypes. Annu Rev Pathol. 3:341–365. 2008. View Article : Google Scholar

2 

Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A, Scheithauer BW and Kleihues P: The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol. 114:97–109. 2007. View Article : Google Scholar : PubMed/NCBI

3 

Garrè ML, Cama A, Bagnasco F, Morana G, Giangaspero F, Brisigotti M, Gambini C, Forni M, Rossi A, Haupt R, et al: Medulloblastoma variants: Age-dependent occurrence and relation to Gorlin syndrome - a new clinical perspective. Clin Cancer Res. 15:2463–2471. 2009. View Article : Google Scholar

4 

Ramaswamy V, Remke M, Bouffet E, Bailey S, Clifford SC, Doz F, Kool M, Dufour C, Vassal G, Milde T, et al: Risk stratifi-cation of childhood medulloblastoma in the molecular era: The current consensus. Acta Neuropathol. 131:821–831. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Northcott PA, Dubuc AM, Pfister S and Taylor MD: Molecular subgroups of medulloblastoma. Expert Rev Neurother. 12:871–884. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Northcott PA, Shih DJ, Remke M, Cho YJ, Kool M, Hawkins C, Eberhart CG, Dubuc A, Guettouche T, Cardentey Y, et al: Rapid, reliable, and reproducible molecular sub-grouping of clinical medulloblastoma samples. Acta Neuropathol. 123:615–626. 2012. View Article : Google Scholar :

7 

Gibson P, Tong Y, Robinson G, Thompson MC, Currle DS, Eden C, Kranenburg TA, Hogg T, Poppleton H, Martin J, et al: Subtypes of medulloblastoma have distinct developmental origins. Nature. 468:1095–1099. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Coluccia D, Figuereido C, Isik S, Smith C and Rutka JT: Medulloblastoma: Tumor biology and relevance to treatment and prognosis paradigm. Curr Neurol Neurosci Rep. 16:432016. View Article : Google Scholar : PubMed/NCBI

9 

Khatua S: Evolving molecular era of childhood medulloblastoma: Time to revisit therapy. Future Oncol. 12:107–117. 2016. View Article : Google Scholar

10 

Parsons DW, Li M, Zhang X, Jones S, Leary RJ, Lin JC, Boca SM, Carter H, Samayoa J, Bettegowda C, et al: The genetic landscape of the childhood cancer medulloblastoma. Science. 331:435–439. 2011. View Article : Google Scholar :

11 

Roy DM, Walsh LA and Chan TA: Driver mutations of cancer epigenomes. Protein Cell. 5:265–296. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Schuettengruber B, Martinez AM, Iovino N and Cavalli G: Trithorax group proteins: Switching genes on and keeping them active. Nat Rev Mol Cell Biol. 12:799–814. 2011. View Article : Google Scholar : PubMed/NCBI

13 

Wang VY and Zoghbi HY: Genetic regulation of cerebellar development. Nat Rev Neurosci. 2:484–491. 2001. View Article : Google Scholar : PubMed/NCBI

14 

Ruddle FH: The role of Hox and Dlx gene cluster in evolution and development. Inborn Errors of Development. Epstein CJ, Erickson RP and Wynshaw-Boris A: Oxford University Press; New York: pp. 653–658. 2008

15 

Takahashi Y, Hamada J, Murakawa K, Takada M, Tada M, Nogami I, Hayashi N, Nakamori S, Monden M, Miyamoto M, et al: Expression profiles of 39 HOX genes in normal human adult organs and anaplastic thyroid cancer cell lines by quantitative real-time RT-PCR system. Exp Cell Res. 293:144–153. 2004. View Article : Google Scholar : PubMed/NCBI

16 

Hutlet B, Theys N, Coste C, Ahn MT, Doshishti-Agolli K, Lizen B and Gofflot F: Systematic expression analysis of Hox genes at adulthood reveals novel patterns in the central nervous system. Brain Struct Funct. 221:1223–1243. 2016. View Article : Google Scholar

17 

Mallo M and Alonso CR: The regulation of Hox gene expression during animal development. Development. 140:3951–3963. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Shah N and Sukumar S: The Hox genes and their roles in oncogenesis. Nat Rev Cancer. 10:361–371. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Quinonez SC and Innis JW: Human HOX gene disorders. Mol Genet Metab. 111:4–15. 2014. View Article : Google Scholar

20 

Wu X, Chen H, Parker B, Rubin E, Zhu T, Lee JS, Argani P and Sukumar S: HOXB7, a homeodomain protein, is overexpressed in breast cancer and confers epithelial-mesenchymal transition. Cancer Res. 66:9527–9534. 2006. View Article : Google Scholar : PubMed/NCBI

21 

Liao WT, Jiang D, Yuan J, Cui YM, Shi XW, Chen CM, Bian XW, Deng YJ and Ding YQ: HOXB7 as a prognostic factor and mediator of colorectal cancer progression. Clin Cancer Res. 17:3569–3578. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Wang Y, Dang Y, Liu J and Ouyang X: The function of homeobox genes and lncRNAs in cancer. Oncol Lett. 12:1635–1641. 2016.PubMed/NCBI

23 

Rodrigues MF, Esteves CM, Xavier FC and Nunes FD: Methylation status of homeobox genes in common human cancers. Genomics. 108:185–193. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Costa BM, Smith JS, Chen Y, Chen J, Phillips HS, Aldape KD, Zardo G, Nigro J, James CD, Fridlyand J, et al: Reversing HOXA9 oncogene activation by PI3K inhibition: Epigenetic mechanism and prognostic significance in human glioblastoma. Cancer Res. 70:453–462. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Tabuse M, Ohta S, Ohashi Y, Fukaya R, Misawa A, Yoshida K, Kawase T, Saya H, Thirant C, Chneiweiss H, et al: Functional analysis of HOXD9 in human gliomas and glioma cancer stem cells. Mol Cancer. 10:602011. View Article : Google Scholar : PubMed/NCBI

26 

Se YB, Kim SH, Kim JY, Kim JE, Dho YS, Kim JW, Kim YH, Woo HG, Kim SH, Kang SH, et al: Underexpression of HOXA11 is associated with treatment resistance and poor prognosis in glioblastoma. Cancer Res Treat. 49:387–398. 2017. View Article : Google Scholar :

27 

Munthe S, Sørensen MD, Thomassen M, Burton M, Kruse TA, Lathia JD, Poulsen FR and Kristensen BW: Migrating glioma cells express stem cell markers and give rise to new tumors upon xenografting. J Neurooncol. 130:53–62. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Manohar CF, Salwen HR, Furtado MR and Cohn SL: Up-regulation of HOXC6, HOXD1, and HOXD8 homeobox gene expression in human neuroblastoma cells following chemical induction of differentiation. Tumour Biol. 17:34–47. 1996. View Article : Google Scholar : PubMed/NCBI

29 

Zhang X, Hamada J, Nishimoto A, Takahashi Y, Murai T, Tada M and Moriuchi T: HOXC6 and HOXC11 increase transcription of S100beta gene in BrdU-induced in vitro differentiation of GOTO neuroblastoma cells into Schwannian cells. J Cell Mol Med. 11:299–306. 2007. View Article : Google Scholar : PubMed/NCBI

30 

Chakravadhanula M, Ozols VV, Hampton CN, Zhou L, Catchpoole D and Bhardwaj RD: Expression of the HOX genes and HOTAIR in atypical teratoid rhabdoid tumors and other pediatric brain tumors. Cancer Genet. 207:425–428. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Bodey B, Bodey B Jr, Siegel SE and Kaiser HE: Immunocytochemical detection of the homeobox B3, B4, and C6 gene products in childhood medulloblastomas/primitive neuroectodermal tumors. Anticancer Res. 20:1769–1780. 2000.PubMed/NCBI

32 

Vo BT, Li C, Morgan MA, Theurillat I, Finkelstein D, Wright S, Hyle J, Smith SMC, Fan Y, Wang YD, et al: Inactivation of Ezh2 upregulates Gfi1 and drives aggressive Myc-driven group 3 medulloblastoma. Cell Rep. 18:2907–2917. 2017. View Article : Google Scholar : PubMed/NCBI

33 

Margueron R and Reinberg D: The Polycomb complex PRC2 and its mark in life. Nature. 469:343–349. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

35 

Castro-Gamero AM, Borges KS, Lira RC, Andrade AF, Fedatto PF, Cruzeiro GA, Silva RB, Fontes AM, Valera ET, Bobola M, et al: Chromosomal heterogeneity and instability characterize pediatric medulloblastoma cell lines and affect neoplastic phenotype. Cytotechnology. 65:871–885. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Jacobsen PF, Jenkyn DJ and Papadimitriou JM: Establishment of a human medulloblastoma cell line and its heterotransplantation into nude mice. J Neuropathol Exp Neurol. 44:472–485. 1985. View Article : Google Scholar : PubMed/NCBI

37 

Yamada M, Shimizu K, Tamura K, Okamoto Y, Matsui Y, Moriuchi S, Park K, Mabuchi E, Yamamoto K, Hayakawa T, et al: Establishment and biological characterization of human medulloblastoma cell lines. No To Shinkei. 41:695–702. 1989.In Japanese. PubMed/NCBI

38 

Ivanov DP, Coyle B, Walker DA and Grabowska AM: In vitro models of medulloblastoma: Choosing the right tool for the job. J Biotechnol. 236:10–25. 2016. View Article : Google Scholar : PubMed/NCBI

39 

de Oliveira GP, Alves CJ and Chadi G: Early gene expression changes in spinal cord from SOD1G93A Amyotrophic Lateral Sclerosis animal model. Front Cell Neurosci. 7:2162013.

40 

Covas DT, Panepucci RA, Fontes AM, Silva WA Jr, Orellana MD, Freitas MC, Neder L, Santos AR, Peres LC, Jamur MC, et al: Multipotent mesenchymal stromal cells obtained from diverse human tissues share functional properties and gene-expression profile with CD146+ perivascular cells and fibroblasts. Exp Hematol. 36:642–654. 2008. View Article : Google Scholar : PubMed/NCBI

41 

Fontes AM, Melo FU, Greene LJ, Faça VM, Lin Y, Gerson SL and Covas DT: Production of human factor VIII-FL in 293T cells using the bicistronic MGMT(P140K)-retroviral vector. Genet Mol Res. 11:775–789. 2012. View Article : Google Scholar : PubMed/NCBI

42 

Albesiano E, Messmer BT, Damle RN, Allen SL, Rai KR and Chiorazzi N: Activation-induced cytidine deaminase in chronic lymphocytic leukemia B cells: Expression as multiple forms in a dynamic, variably sized fraction of the clone. Blood. 102:3333–3339. 2003. View Article : Google Scholar : PubMed/NCBI

43 

Ferguson RE, Carroll HP, Harris A, Maher ER, Selby PJ and Banks RE: Housekeeping proteins: A preliminary study illustrating some limitations as useful references in protein expression studies. Proteomics. 5:566–571. 2005. View Article : Google Scholar : PubMed/NCBI

44 

Thomé CH, dos Santos GA, Ferreira GA, Scheucher PS, Izumi C, Leopoldino AM, Simão AM, Ciancaglini P, de Oliveira KT, Chin A, et al: Linker for activation of T-cell family member2 (LAT2) a lipid raft adaptor protein for AKT signaling, is an early mediator of alkylphospholipid anti-leukemic activity. Mol Cell Proteomics. 11:1898–1912. 2012. View Article : Google Scholar : PubMed/NCBI

45 

Palma CS, Grassi ML, Thomé CH, Ferreira GA, Albuquerque D, Pinto MT, Ferreira Melo FU, Kashima S, Covas DT, Pitteri SJ, et al: Proteomic analysis of epithelial to mesenchymal transition (EMT) reveals cross-talk between SNAIL and HDAC1 proteins in breast cancer cells. Mol Cell Proteomics. 15:906–917. 2016. View Article : Google Scholar :

46 

Triscott J, Lee C, Foster C, Manoranjan B, Pambid MR, Berns R, Fotovati A, Venugopal C, O'Halloran K, Narendran A, et al: Personalizing the treatment of pediatric medulloblastoma: Polo-like kinase 1 as a molecular target in high-risk children. Cancer Res. 73:6734–6744. 2013. View Article : Google Scholar : PubMed/NCBI

47 

Bobola MS, Silber JR, Ellenbogen RG, Geyer JR, Blank A and Goff RD: O6-methylguanine-DNA methyltransferase, O6-benzylguanine, and resistance to clinical alkylators in pediatric primary brain tumor cell lines. Clin Cancer Res. 11:2747–2755. 2005. View Article : Google Scholar : PubMed/NCBI

48 

Murat A, Migliavacca E, Gorlia T, Lambiv WL, Shay T, Hamou MF, de Tribolet N, Regli L, Wick W, Kouwenhoven MC, et al: Stem cell-related 'self-renewal' signature and high epidermal growth factor receptor expression associated with resistance to concomitant chemoradiotherapy in glioblastoma. J Clin Oncol. 26:3015–3024. 2008. View Article : Google Scholar : PubMed/NCBI

49 

Kurscheid S, Bady P, Sciuscio D, Samarzija I, Shay T, Vassallo I, Criekinge WV, Daniel RT, van den Bent MJ, Marosi C, et al: Chromosome 7 gain and DNA hypermethylation at the HOXA10 locus are associated with expression of a stem cell related HOX-signature in glioblastoma. Genome Biol. 16:162015. View Article : Google Scholar : PubMed/NCBI

50 

Perkins EJ, Bao W, Guan X, Ang CY, Wolfinger RD, Chu TM, Meyer SA and Inouye LS: Comparison of transcriptional responses in liver tissue and primary hepatocyte cell cultures after exposure to hexahydro-1,3,5-trinitro-1,3,5-triazine. BMC Bioinformatics. 7(Suppl 4): S222006. View Article : Google Scholar

51 

Snuderl M, Batista A, Kirkpatrick ND, Ruiz de Almodovar C, Riedemann L, Walsh EC, Anolik R, Huang Y, Martin JD, Kamoun W, et al: Targeting placental growth factor/neuropilin 1 pathway inhibits growth and spread of medulloblastoma. Cell. 152:1065–1076. 2013. View Article : Google Scholar : PubMed/NCBI

52 

Sengupta S, Weeraratne SD, Sun H, Phallen J, Rallapalli SK, Teider N, Kosaras B, Amani V, Pierre-Francois J, Tang Y, et al: α5-GABAA receptors negatively regulate MYC-amplified medulloblastoma growth. Acta Neuropathol. 127:593–603. 2014. View Article : Google Scholar

53 

Tang W, Jiang Y, Mu X, Xu L, Cheng W and Wang X: MiR-135a functions as a tumor suppressor in epithelial ovarian cancer and regulates HOXA10 expression. Cell Signal. 26:1420–1426. 2014. View Article : Google Scholar : PubMed/NCBI

54 

Hemmesi K, Squadrito ML, Mestdagh P, Conti V, Cominelli M, Piras IS, Sergi LS, Piccinin S, Maestro R, Poliani PL, et al: miR-135a inhibits cancer stem cell-driven medulloblastoma development by directly repressing Arhgef6 expression. Stem Cells. 33:1377–1389. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2017
Volume 51 Issue 6

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
Bonfim-Silva R, Ferreira Melo FU, Thomé CH, Abraham KJ, De Souza FL, Ramalho FS, Machado HR, De Oliveira RS, Cardoso AA, Covas DT, Covas DT, et al: Functional analysis of HOXA10 and HOXB4 in human medulloblastoma cell lines. Int J Oncol 51: 1929-1940, 2017
APA
Bonfim-Silva, R., Ferreira Melo, F.U., Thomé, C.H., Abraham, K.J., De Souza, F.L., Ramalho, F.S. ... Fontes, A.M. (2017). Functional analysis of HOXA10 and HOXB4 in human medulloblastoma cell lines. International Journal of Oncology, 51, 1929-1940. https://doi.org/10.3892/ijo.2017.4151
MLA
Bonfim-Silva, R., Ferreira Melo, F. U., Thomé, C. H., Abraham, K. J., De Souza, F. L., Ramalho, F. S., Machado, H. R., De Oliveira, R. S., Cardoso, A. A., Covas, D. T., Fontes, A. M."Functional analysis of HOXA10 and HOXB4 in human medulloblastoma cell lines". International Journal of Oncology 51.6 (2017): 1929-1940.
Chicago
Bonfim-Silva, R., Ferreira Melo, F. U., Thomé, C. H., Abraham, K. J., De Souza, F. L., Ramalho, F. S., Machado, H. R., De Oliveira, R. S., Cardoso, A. A., Covas, D. T., Fontes, A. M."Functional analysis of HOXA10 and HOXB4 in human medulloblastoma cell lines". International Journal of Oncology 51, no. 6 (2017): 1929-1940. https://doi.org/10.3892/ijo.2017.4151