Skip to main content
Neoplasia (New York, N.Y.) logoLink to Neoplasia (New York, N.Y.)
. 2008 Aug;10(8):757–772. doi: 10.1593/neo.07914

Genomic Deletions Correlate with Underexpression of Novel Candidate Genes at Six Loci in Pediatric Pilocytic Astrocytoma1

Nicola Potter *, Aikaterini Karakoula *, Kim P Phipps , William Harkness , Richard Hayward , Dominic N P Thompson , Thomas S Jacques ‡,§, Brian Harding §, David G T Thomas , Rodger W Palmer #, Jeremy Rees *, John Darling **, Tracy J Warr *
PMCID: PMC2481566  PMID: 18670637

Abstract

The molecular pathogenesis of pediatric pilocytic astrocytoma (PA) is not well defined. Previous cytogenetic and molecular studies have not identified nonrandom genetic aberrations. To correlate differential gene expression and genomic copy number aberrations (CNAs) in PA, we have used Affymetrix GeneChip HG_U133A to generate gene expression profiles of 19 pediatric patients and the SpectralChip 2600 to investigate CNAs in 11 of these tumors. Hierarchical clustering according to expression profile similarity grouped tumors and controls separately. We identified 1844 genes that showed significant differential expression between tumor and normal controls, with a large number clearly influencing phosphatidylinositol and mitogen-activated protein kinase signaling in PA. Most CNAs identified in this study were single-clone alterations. However, a small region of loss involving up to seven adjacent clones at 7q11.23 was observed in seven tumors and correlated with the underexpression of BCL7B. Loss of four individual clones was also associated with reduced gene expression including SH3GL2 at 9p21.2-p23, BCL7A (which shares 90% sequence homology with BCL7B) at 12q24.33, DRD1IP at 10q26.3, and TUBG2 and CNTNAP1 at 17q21.31. Moreover, the down-regulation of FOXG1B at 14q12 correlated with loss within the gene promoter region in most tumors. This is the first study to correlate differential gene expression with CNAs in PA.

Introduction

The most prevalent brain tumors in the pediatric population are astrocytomas, which most frequently manifest as pilocytic astrocytoma (PA), World Health Organization (WHO) grade I [1]. Pilocytic astrocytomas are commonly located in the cerebellum and can often be completely surgically resected, resulting in excellent long-term survival [2]. However, a small number of PA recur after resection and may occasionally undergo malignant transformation [3].

The genetic events that contribute to the development of PA are poorly defined. Cytogenetic analysis of pediatric PA has shown that most of these tumors (>70%) have a normal karyotype. Numerical and structural abnormalities of chromosomes 5, 6, 7, 8, and 9 have been reported, although definitive, nonrandom aberrations have not yet been identified [4–8]. Loss of the p53 locus on chromosome 17p has been reported in some PA studies but not consistently [9–11]. Similarly, although TP53 gene mutations were found in 7 of 20 pediatric PA, other studies have failed to confirm these findings [12,13].

Global expression technology can now generate detailed gene expression profiles of tumors, and statistical algorithm-based classifications can be used to identify subgroups with clinical or biologic significance [14,15]. Array gene expression profiles of 21 pediatric PA have shown that genes involved in neurogenesis, cell adhesion, synaptic transmission, central nervous system development, potassium ion transport, protein dephosphorylation, and cell differentiation were significantly deregulated. The same study also demonstrated that the tumors clustered into two groups could be distinguished by the extent of myelin basic protein staining [16]. A similar approach has also been used to identify an expression signature that can stratify PA according to tumor location. A signature of 36 genes showed differential expression between supratentorial PA and those arising in the posterior fossa [17].

Array-based comparative genomic hybridization (aCGH) has recently been used to define genome-wide aberrations at a resolution of 1 Mb in adult malignant astrocytoma [18–24]. Only one previous study has used array technology to investigate chromosome copy number aberrations (CNAs) in PA [25]. Whole-chromosome aberrations were only observed in tumors arising in patients older than 10 years, in which the most common aberrations were gain of chromosomes 5 and 7, present in 13% and 17% of cases, respectively. Whole-chromosome aberrations of chromosome 15 were present in 6% of cases and of chromosomes 4, 6, 9, 10, 11, 12, and 20 in 3% of cases [25]. Single-nucleotide polymorphic allelic arrays have also been used to identify regions of allelic imbalance in low-grade pediatric gliomas including six PA. No detectable loss of heterozygosity was found at any of the 11,562 single-nucleotide polymorphic loci investigated in the PA [26]. It is also possible that epigenetic events such as aberrant promoter hypermethylation are common genomic alterations in pediatric low-grade astrocytoma and that they may have a greater impact on tumor development [27].

The aim of the present study was to correlate aberrant gene expression in pediatric PA with CNAs or gene promoter hypermethylation to identify and characterize cellular pathways that are involved in tumor development and growth. We have used the Affymetrix GeneChip HG_U133A (Affymetrix, Santa Clara, CA) to generate gene expression profiles and to identify aberrantly expressed genes in 19 pediatric PA biopsies. In 11 of 19 cases, we also identified CNAs using the aCGH SpectralChip 2600 (Spectral Genomics, Houston, TX). Both approaches were validated by real-time quantitative polymerase chain reaction (RT-QPCR). The methylation status of the promoter regions of six genes, showing significant underexpression in the PA compared to the normal controls, was examined using methylation-specific PCR (MSP) and bisulfite sequencing to investigate alternative mechanisms of transcriptional silencing in PA. These genes have previously been reported as being methylated in astrocytoma or in other tumors or cancer [28–33].

Materials and Methods

Tumor Samples and RNA Preparation

Pilocytic astrocytoma tumor tissue was obtained with informed consent from 19 pediatric patients directly from the operating theater and was stored in liquid nitrogen until ready for use. All tumor samples were directly adjacent to tumor tissue processed for routine histologic evaluation and were first examined macroscopically to ensure that no frankly normal tissue was present. They were diagnosed according to the WHO classification and were reviewed by two neuropathologists [1]. The patients' mean age at diagnosis was 5.8 years (range, 1.75–11.5 years), with a female/male ratio of 8:11. At the time of this study, patient follow-up was available from diagnosis for 17 children who remain alive (follow-up ranged from 1 to 160 months from diagnosis; Table W1).

Total RNA was isolated from the 19 tumor samples using TRIzol reagent (Invitrogen Ltd, Paisley, UK) followed by cleanup using an RNeasy mini spin column (Qiagen, Crawley, UK). The quality and quantity of each sample were assessed using the Agilent 2100 Bioanalyser (Agilent Technologies Ltd, West Lothian, UK). Control RNA, consisting of four pooled normal whole-brain total RNA samples from young adults between 21 and 29 years of age, were obtained from AMS Biotechnology (Abingdon, Oxfordshire, UK) as previously described by Wong et al. [16].

cRNA Synthesis, Array Hybridization, and Array Scanning

Total RNA from 19 PA and 4 pooled normal whole-brain samples was used to prepare biotinylated target RNA following the manufacturer's instructions (Affymetrix). Briefly, 8 µg of total RNA was used to generate first-strand cDNA using a T7-linked oligo (dT) primer. Second-stand synthesis was then completed followed by a final transcription step with biotinylated UTP and CTP to label and amplify cRNA. The labeled cRNA was hybridized overnight to the HG_U133A array (Affymetrix). The arrays were washed and stained with streptavidin-phycoerythrin and were scanned using an Affymetrix GeneChip Scanner 3000. Array background, Q values, and mean intensities were within acceptable ranges for all arrays.

Affymetrix Array Data Analysis

Analysis was carried out using GeneSpring version 7.2 (Silicon Genetics, Redwood City, CA). The samples were analyzed in one experiment, and data transformation normalization was completed. Measurements less than 0.01 (set as a standard cutoff) were readjusted to 0.01, and per-chip normalizations were completed dividing all measurements on each array by the 50th percentile. The resulting expression levels were centered on 1 for each chip. The experiment was then normalized to the control samples (per gene normalization).

Identification of Aberrantly Expressed Genes

Data filters were applied after the raw expression data had been normalized, to remove unreliable and unnecessary data from further analysis including genes that were constant in both control and tumor samples and Affymetrix standard control genes. Unsupervised hierarchical clustering with the remaining 10,653 informative probe sets was used to cluster the tumors according to gene expression profile similarity. A t test and Bonferroni multiple correction test (P < .05) were used to identify genes that showed a significant difference in expression between PA and normal controls. The variance statistic was derived from the multiple samples in each condition. A filter tool was then used to identify genes that showed a greater than twofold change in expression between the two conditions. From the 10,653 reliable expression results, 2273 probe sets that showed a significant and greater than twofold change in expression between PA and control samples, as described by Sowar et al. [34], were identified. These represented 1844 specific genes that were used for further analysis by Onto-Tools [35,36]. This software generates functional profiles of differentially expressed genes and assesses the impact of their alterations on biologic processes and pathways. The pathways used by Onto-Tools originate from the Kyoto Encyclopedia of Genes and Genomes (KEGG) database [37].

Validation of Differential Gene Expression by RT-QPCR Analysis

Real-time quantitative polymerase chain reaction analysis was completed for six genes that showed a twofold and significant difference in expression between PA and normal controls to validate the Affymetrix array results. RNA was available for RT-QPCR analysis from 16 of 19 tumors and 4 normal controls. cDNA was synthesized from 1 µg of total RNA using the QuantiTect Reverse Transcription Kit (Qiagen) following the manufacturer's instructions. Gene-specific oligonucleotide probes and primer pairs were obtained from Assays on Demand (Applied Biosystems, Warrington, UK) for CCNA1, SPINT2, MAPK1, MATK, TIMP1, and MCL1. All probes were designed across exon-exon boundaries. The TaqMan Universal PCR Master Mix No AmpErase UNG was used in a 25-µl reaction volume after cycling conditions recommended by the manufacturer. β-Actin was used as an endogenous control. All reactions were completed as single-well triplicates using the ABI Prism 7000 Sequence Detection System. No-template and no-amplification controls were included in each experiment. The comparative CT method (PE Applied Biosystems, Warrington, UK) was used to determine the relative ratio of expression for each gene.

DNA Preparation and aCGH

Tumor tissue was available for aCGH analysis from 11 of 19 PA. Genomic DNA was isolated from the same tissue mass as total RNA using DNAMini Kit following the manufacturer's instructions (Qiagen). Array-based comparative genomic hybridization was carried out using the SpectralChip 2600 following manufacturer's instructions (Spectral Genomics). The SpectralChip 2600 is a human BAC array that generates genome-wide molecular profiles at a resolution of 1 Mb allowing quantitative analysis. Each of the 2600 complementary BAC elements are printed on a glass slide, fluorescent-labeled normal and tumor DNA are cohybridized to the array, and dye ratios are calculated for each clone. Briefly, 1 µg of tumor and pooled sex-mismatched reference DNA (Promega, Southampton, UK) were differentially labeled with Cy5-dCTP or Cy3-dCTP using random prime labeling (Bioprime Labeling Kit; Invitrogen). Labeled tumor and reference DNA were cohybridized to the SpectralChip at 37°C for 16 hours in a hybridization chamber (Corning, Inc., Corning, NY). Arrays were washed at 50°C for 20 minutes in 50% formamide/2x SSC followed by 2x SSC/0.1% Tween 20 for 20 minutes and 0.2x SSC for 10 minutes. Arrays were then washed in 80% ethanol and blown dry using nitrogen gas. Finally, the arrays were scanned using a microarray scanner (GenePix Personal 4100A;Molecular Devices, CA). Each experiment was duplicated swapping the dye labels between tumor and reference DNA.

Array-Based Comparative Genomic Hybridization Analysis

The scanned images were analyzed using GenePix Pro 6 software (Molecular Devices). Image spots were defined by an automatic grid feature that correlates the BAC clone position on the array with clone details including genomic locations. The spots were adjusted where necessary, and the dye intensities were calculated. Further analysis was completed using Formatter Software (Digital Scientific, Cambridge, UK; MIDAS Team) [38]. Preliminary analysis involved the removal of defective spots, that is, those without a signal, and background corrections were applied. A flip-dye consistency check involving the cross-log ratios of each signal from the duplicated clone spots on each array and from the dye swap experiment was also completed, and those signals that showed a greater than ±2SD variation between replicates were discarded [39,40].

Iterative linear regression global normalizations were completed for each sample reducing the SD of spot log2 ratios. In this approach, the SD of a given sample was continuously recalculated, removing outliers, until the correlation coefficients converged to a constant value. The final value is referred to as the modified SD (MSD). The outlier range used for this was ±2SD [41]. Those clones that showed a log2 ratio in excess of 3MSD were deemed as gained or lost. This approach assumes that data variations are random, and constant nonchanging results form most data. Theoretically, only 0.7% of data values will occur outside the threshold; therefore, any deviations exceeding 3SD are considered true positives [39,40].

Validation of DNA Copy Number by RT-QPCR Analysis

To validate the SpectralChip 2600 array experiments, RT-QPCR analysis to determine relative DNA copy number was completed at five genomic regions and/or gene loci including 1p36.32, 14q12, the promoter region of FOXG1B at 14q12, BCL7B at 7q11.23, and BLC7A at 12q24.33. Primer and oligonucleotide probe sets were designed within gene exon boundaries using primer design Web server and software packages (http://frodo.wi.mit.edu/cgi-bin/primer3/primer3_www.cgi) and Primer Express and were purchased from MWG-Biotech (Milton, Keynes, UK). β-Actin, at 7p12, was used as an endogenous control because this region was not altered in any of the PA in this study (see Table W2 for specific primer and probe sequences and concentrations). Probes for the genes of interest were labeled with the FAM fluorescent report, and the endogenous control probe was labeled with VIC. Sufficient DNA was available from 10 of 11 tumors investigated using the SpectralChip 2600 array and 4 additional tumors, for which expression array analysis had been completed IN2356, IN2825, IN2921, and IN3156). DNA extracted from blood samples of four normal individuals and one pooled male and one pooled female DNA samples (Promega) were used as normal reference controls.

The RT-QPCR experiments were carried out using the ABI Prism 7000 Sequence Detection System. Reactions were completed as single-well triplicates in a final volume of 25 µl consisting of 1x QuantiTect Probe PCRMix (Qiagen), probe and primers at the required concentrations, and 25 ng of genomic DNA. No-template and no-amplification controls were also included for each locus investigated. The thermal cycling profile consisted of a 15-minute heat activation step at 95°C, followed by 40 cycles at 95°C for 10 seconds and 60°C for 1 minute.

Before genomic quantification, primer limitation experiments were carried out to determine optimum primer concentrations for each experiment (see User Bulletin #2, Applied Biosystems, Warrington, UK). Primer concentrations that generated the highest magnitude of signal intensity (ΔRn) but lowest cycle threshold (CT) at a given threshold were selected. The standard curve method (PE Applied Biosystems, Warrington, UK) was used to determine the relative copy number for each locus. A standard curve was included on each 96-well plate for control and target regions consisting of serially diluted normal genomic DNA (pooled blood from 20 healthy individuals). Only standard curves with correlation coefficients of 0.98 and higher were used for genomic copy number analysis. Tumors with values less than or equal to the copy number of controls minus the 95% reference range of controls (or 2x SD of controls) were considered to have genomic loss, whereas a value greater or equal to the copy number of controls plus 95% reference range of controls (or 2x SD of controls) was regarded as gain [42,43].

Methylation-Specific PCR and Sequencing

The methylation status of the promoter regions of six genes was investigated in 17 of 19 PA using MSP. Sequencing was used to confirm these results in two tumors and one normal brain control. The MSP and sequencing primer pairs for this study were either adapted or designed according to MSP principles [44,45] with the aid of the CpG island identification Web server (http://www.uscnorris.com/cpgislands2/), primer design Web server and software packages (http://frodo.wi.mit.edu/cgi-bin/primer3/primer3_www.cgi), and Primer Express (see Tables W3 and W4 for MSP and sequencing primer information).

After sodium bisulfite modification of tumor and normal DNA as described previously, MSP was carried out in a 25-µl volume with HotStart Taq (Qiagen) [44]. After an initial step of 95°C for 15 minutes, DNA was amplified by 35 cycles for 30 seconds at 95°C, for 30 seconds at the optimal annealing temperature for each primer pair, and for 30 seconds at 72°C, followed by a final extension step at 72°C for 5 minutes. Polymerase chain reaction analysis for sequencing was carried out in a 50-µl volume with HotStart Taq (Qiagen). After a 15-minute incubation at 95°C, DNA was amplified by 38 cycles at 94°C for 15 seconds, optimal annealing temperatures for primer pairs for 30 seconds and at 72°C for 45 seconds, followed by a final extension at 72°C for 5 minutes. A blank control containing all MSP components except the sample DNA and two positive controls (normal DNA and universally methylated DNA, both of which were bisulfite-treated as described previously) was included in each experiment. Products were separated using a 1.5% agarose gel containing ethidium bromide and 1x Tris-borate-EDTA buffer. The products were visualized by UV light.

Polymerase chain reaction amplifications of DNA for sequencing were performed using the BigDye Terminator v1.1 cycle sequencing kit following manufacturer's instructions (Applied Biosystems, Foster City, CA). Sequencing was carried out using an ABI 3730x1 DNA Analyser, and raw data were analyzed with SeqScape software v2.5 (Applied Biosystems, Foster City, CA).

Results

Gene Expression Profiles of Pediatric PA

Using the Affymetrix GeneChip HG_U133A, we have studied the expression pattern of approximately 22,000 genes in pediatric PA compared to normal brain (see Table W5 for all Affymetrix GeneChip raw data). Unsupervised hierarchical clustering using 10,653 reliable gene expression results was used to generate a dendrogram illustrating the similarity in expression profiles of the 19 PA (Figure 1). The tumor and normal control samples clustered independently. However, the PA did not group or order according to patient age, sex, or tumor location, and no consistent expression profiles could be associated with these clinical parameters or patient outcome. Moreover, no further subgroups were clearly defined, in contrast to the previous findings [16,17]. Wong et al. [16] described a molecular signature of 89 genes that distinguished two subgroups of PA representing tumors with different potentials of progression. A list of the 89 genes was not available in the literature. However, 16 of these 89 genes were described by Wong et al. [16] and were involved in biologic processes that significantly altered between the two subgroups. In total, 13 of these 16 genes showed reliable data in the present study and were used to cluster the 19 PA. The tumors did not cluster into subgroups, and a continuous increase or decrease in the expression of these 13 genes was seen between tumors that are least similar (Figure W1, a and b).

Figure 1.

Figure 1

Unsupervised hierarchical clustering of 19 pediatric PA and 4 normal brain controls using 10,653 reliable gene expression results. The dendrogram color saturation is proportional to the magnitude of the difference from the mean, ranging from red (overexpressed) to green (underexpressed). Patient ages are in years at diagnosis. Tumor labels in green indicate a male patient, and pink a female patient. CERE indicates cerebellum; CHI, chiasmatic; OP, optic pathway; PF, posterior fossa.

From 10,653 reliable expression results, 1844 genes showed a twofold and significant change in expression in PA compared to the normal controls. Of these, 1002 were up-regulated and 842 were down-regulated. Genes that showed a greater than 10-fold change in expression and were either involved in a KEGG pathway or have been well characterized were investigated further (Table 1). SERPINA3, a protease inhibitor, showed the largest up-regulation in gene expression with an 80.89-fold change in the tumors compared to normal brain. FOXG1B, a member of the forkhead family of transcription factors, showed the largest down-regulation in gene expression with a 325.73-fold change.

Table 1.

Differentially Expressed Genes That Show a 10-Fold Minimum Change in Expression from the Normal and Are Involved in Specific Pathways or Are Well Characterized with Functions That Can Be Linked to Tumor Development.

Rank Gene Symbol Gene Description Fold Change in Expression Chromosome Location
1 SERPINA3 Serine (or cysteine) proteinase inhibitor, clade A (alpha-1 antiproteinase, antitrypsin), member 3 80.89 14q32.1
2 CHI3L1 Chitinase 3-like 1 (cartilage glycoprotein-39) 56.98 1q32.1
4 CHAD2 (4, 11) Chondroadherin 43.68 17q21.33
6 TIMP4 Tissue inhibitor of metalloproteinase 4 41.47 3p25
7 WEE11 (38) WEE1 homolog (Schizosaccharomyces pombe) 41.03 11p15.3–p15.1
11 T1A-2 Lung type-I cell membrane-associated glycoprotein 35.21 1p36
12 ABCC3 ATP-binding cassette, subfamily C (CFTR/MRP), member 3 33.72 17q22
15 IL1RAP2 (1, 47) Interleukin 1 receptor accessory protein 26.58 3q28
17 CSPG4 Chondroitin sulfate proteoglycan 4 (melanoma-associated) 23.77 15q23
19 AGTRL11 (32) Angiotensin II receptor-like 1 22.36 11q12
22 CD44 Homo sapiens CD44 isoform RC (CD44) mRNA, complete cds 19.88 11p13
24 MYT1 Myelin transcription factor 1 18.49 20q13.33
26 TAZ Transcriptional coactivator with PDZ-binding motif (TAZ) 17.53 3q23–q24
31 TIMP1 Tissue inhibitor of metalloproteinase 1 (erythroid potentiating activity, collagenase inhibitor) 16.23 Xp11.3–p11.23
32 HLA-DPA13 (1, 2, 32) Major histocompatibility complex, class II, DP alpha 1 15.79 6p21.3
36 S100A10 S100 calcium binding protein A10 (annexin II ligand, calpactin I, light polypeptide (p11)) 15.27 1q21
38 TLR21 (14) Toll-like receptor 2 14.66 4q32
40 TFPI1 (12) ESTs, weakly similar to cytokine receptor-like factor 2; cytokine receptor CRL2 precusor (Homo sapiens) 14.09 2q32
44 POSTN Osteoblast-specific factor 2 (fasciclin I-like) 12.60 13q13.1
46 CHI3L2 Chitinase 3-like 2 12.22 1p13.3
48 CCL3 Chemokine (C-C motif) ligand 3 11.89 17q11–q21
49 MSR1 Macrophage scavenger receptor 1 11.87 8p22
50 SRPX Sushi-repeat-containing protein, X chromosome 11.29 Xp21.1
51 CSPG21 (2) Chondroitin sulfate proteoglycan 2 (versican) 10.86 5q14.3
56 CD151 CD151 antigen 10.61 11p15.5
-74 FGF132 (5, 6) Fibroblast growth factor 13 -10.34 Xq26.3
-72 MEF2C1 (5) MADS box transcription enhancer factor 2, polypeptide C (myocyte enhancer factor 2C) -10.48 5q14
-70 WNT10B2 (28, 46) wingless-type MMTV integration site family, member 10B -10.50 12q13
-69 NPY1R1 (32) Neuropeptide Y receptor Y1 -10.96 4q31.3–q32
-67 ATP2B21 (19) Homo sapiens, similar to nuclear localization signals binding protein 1, clone IMAGE:4933343, mRNA -11.22 3p25.3
-66 PRKCB115 (3–5, 7, 8, 10, 16–19, 22, 28, 36, 39, 41) Protein kinase C, beta 1 -11.24 16p11.2
-65 MAL mal, T-cell differentiation protein -11.55 2cen–q13
-62 CACNA2D31 (5) Calcium channel, voltage-dependent, alpha 2/delta 3 subunit -11.89 3p21.1
-60 GRIN13 (7, 19, 32) Glutamate receptor, ionotropic, N-methyl d-aspartate 1 -12.15 9q34.3
-59 HTR2A3 (10, 19, 32) 5-Hydroxytryptamine (serotonin) receptor 2A -12.22 13q14–q21
-55 SNAP251 (43) Synaptosomal-associated protein, 25 kDa -13.04 20p12–p11.2
-54 PTK2B4 (8, 16, 17, 19) PTK2B protein tyrosine kinase 2 beta -13.42 8p21.1
-52 VIP1 (32) Vasoactive intestinal peptide -14.27 6q26–q27
-50 VAMP11 (43) Vesicle-associated membrane protein 1 (synaptobrevin 1) -15.17 12p
-48 NPY2 (32, 35) Neuropeptide Y -15.63 7p15.1
-45 ENPP2 Ectonucleotide pyrophosphatase/phosphodiesterase 2 (autotaxin) -17.09 8q24.1
-44 MBP Myelin basic protein -17.48 18q23
-39 HTR2C3 (10, 1, 32) 5-Hydroxytryptamine (serotonin) receptor 2C -19.23 Xq24
-37 P2RX52 (19, 23) Purinergic receptor P2X, ligand-gated ion channel, 5 -19.57 17p13
-32 MATK2 (6, 15) Megakaryocyte-associated tyrosine kinase -23.75 19p13.3
-31 CRH2 (22, 32) Corticotropin-releasing hormone -24.27 8q13
-22 NEFH2 (25, 31) Neurofilament, heavy polypeptide 200 kDa -27.40 22q12.2
-21 ICAM5 Intercellular adhesion molecule 5, telencephalin -27.70 19p13.2
-19 EGR4 Early growth response 4 -28.90 2p13
-15 CAMK2A5 (7, 8, 19, 20, 28) Calcium/calmodulin-dependent protein kinase (CaM kinase) II alpha -36.76 5q33.1
-13 ATP2B21 (19) ATPase, Ca2+ transporting, plasma membrane 2 -39.22 3p26–p25
-11 SST1 (32) Somatostatin -43.86 3q28
-10 ITPKA2 (3, 19) Inositol 1,4,5-trisphosphate 3-kinase A -49.50 15q14–q21
-6 GABRA51 (32) γ-Aminobutyric acid (GABA) A receptor, alpha 5 -101.01 15q11.2–q12
-3 CACNG3 1 (5) Calcium channel, voltage-dependent, gamma subunit 3 -136.99 16p12–p13.1
-2 CCK 1 (32) Cholecystokinin -146.63 3p22–p21.3
-1 FOXG1B Forkhead box G1B -325.73 14q12–q13

The number of pathways that each gene is involved in is given in bold after the gene symbol. The numbers inside the parentheses correspond to the specific pathways in Table 2. Genes highlighted in red are involved in multiple pathways listed in Table 3.

The Onto-Tools software was used to rank the KEGG pathways influenced by the 1844 differentially expressed genes according to the extent of differential gene expression. Of the 53 pathways that were ranked, only those of interest are displayed in Table 2 (see Table W6 for all pathways involved in this analysis). The pathway most affected by differential gene expression (rank 1) was antigen processing and presentation, in which most genes are significantly up-regulated and located at 6p21.3.

Table 2.

The Pathway Express Results Ranking the KEGG Pathways Incorporating the 1844 Differentially Expressed Genes in PA.

Rank Unique Pathway ID KEGG Database Pathway Name Impact Factor Genes in Pathway Pathway Genes in Input (%)
1 1:04612 Antigen processing and presentation 76.6 86 24.4
2 1:04514 Cell adhesion molecules (CAMs) 24.1 132 25.8
3 1:04070 Phosphatidylinositol signaling system 23.2 79 27.8
4 1:04510 Focal adhesion 14.7 194 25.8
5 1:04010 MAPK signaling pathway 14.3 273 22.7
6 1:04810 Regulation of actin cytoskeleton 12.9 206 23.3
9 1:04210 Apoptosis 9.3 84 27.4
10 1:04540 Gap junction 9.3 99 25.3
11 1:04512 ECM-receptor interaction 9.1 87 26.4
12 1:04610 Complement and coagulation cascades 8.8 69 27.5
14 1:04620 Toll-like receptor signaling pathway 8.1 91 25.3
16 1:04670 Leukocyte transendothelial migration 7.8 117 23.1
17 1:04650 Natural killer cell mediated cytotoxicity 7.3 128 21.1
18 1:04662 B-cell receptor signaling pathway 7.2 63 27.0
19 1:04020 Calcium signaling pathway 7.0 176 18.2
24 1:04910 Insulin signaling pathway 5.7 135 20.7
25 1:01510 Neurodegenerative disorders 5.7 35 28.6
26 1:05210 Colorectal cancer 5.6 77 23.4
28 1:04310 Wnt signaling pathway 5.5 147 18.4
32 1:04080 Neuroactive ligand-receptor interaction 4.7 302 11.3
33 1:04660 T cell receptor signaling pathway 4.4 93 20.4
35 1:04920 Adipocytokine signaling pathway 4.0 69 20.3
36 1:04530 Tight junction 3.9 119 16.8
37 1:04520 Adherens junction 3.8 77 20.8
38 1:04110 Cell cycle 3.5 112 16.1
39 1:04370 Vascular endothelial growth factor (VEGF) signaling pathway 3.0 72 16.7
42 1:04150 mTOR signaling pathway 2.6 49 18.4
43 1:04130 SNARE interactions in vesicular transport 2.5 36 13.9
44 1:04350 TGFβ signaling pathway 2.3 84 15.5
45 1:04630 Jak-STAT signaling pathway 2.2 153 13.7
46 1:04340 Hedgehog signaling pathway 1.6 57 12.3
47 1:04060 Cytokine-cytokine receptor interaction 1.6 256 11.7
51 1:04140 Regulation of autophagy 0.8 29 3.4
52 1:04330 Notch signaling pathway 0.8 46 8.7

The pathways are ranked according to impact factor, taking into account the fold change, the number of genes disrupted in a pathway, and the influence each gene exerts on a pathway.

To investigate if particular differentially expressed genes could influence several pathways and have an overall impact on tumor development, those genes involved in five or more pathways were identified (Table 3). Although MAPK1 is involved in 17 pathways, this gene only showed a 2.01-fold down-regulation in expression. In contrast, genes with the largest fold changes are involved in fewer pathways. Only two genes, CAMK2A and PRKCB1, showed a greater than 10-fold change in expression and are involved in more than five pathways (Tables 1 and 3), suggesting that those genes with the highest degree of disregulation in pediatric PA may not have the greatest impact on cellular pathways.

Table 3.

The Pathway Express Results of Differentially Expressed Genes Involved in ≥5 Pathways.

Gene Symbol Gene Description Fold Change in Expression Number of Pathways Gene Involved in Chromosome Location
MAPK1 Mitogen-activated protein kinase 1 -2.01 17 (4–8, 10, 17, 21, 22, 24, 26, 37, 39–42, 44) 22q11.21
PIK3CB Phosphoinositide-3-kinase, catalytic, beta polypeptide -2.05 16 (3, 4, 6, 9, 14, 16–18, 24, 26, 33, 39–42, 45) 3q22.3
PIK3R1 Phosphoinositide-3-kinase, regulatory subunit 1 (p85 alpha) -2.13 16 (3, 4, 6, 9, 14, 16–18, 24, 26, 33, 39–42, 45) 5q13.1
PRKCB1 Protein kinase C, beta 1 -11.24 15 (3–5, 7, 8, 10, 16–19, 22, 28, 36, 39, 41) 16p11.2
AKT3 v-akt murine thymoma viral oncogene homolog 3 (protein kinase B, gamma) -2.54 14 (4, 5, 9, 14, 18, 24, 26, 33, 35, 36, 39, 41, 42, 45) 1q43–q44
MAP2K1 Mitogen-activated protein kinase kinase 1 -4.17 12 (4–8, 10, 17, 22, 24, 26, 39, 41) 15q22.1–q22.33
MAPK9 Mitogen-activated protein kinase 9 -3.48 11 (4, 5, 8, 14, 15, 24, 26, 28, 35, 40, 41) 5q35
MAPK10 Mitogen-activated protein kinase 10 -2.28 11 (4, 5, 8, 14, 15, 24, 26, 28, 35, 40, 41) 4q22.1–q23
PRKACB Protein kinase, cAMP-dependent, catalytic, beta -2.55 11 (5, 7–10, 19, 20, 24, 28, 46, 48) 1p36.1
PPP3CA Protein phosphatase 3 (formerly 2B), catalytic subunit, alpha isoform (calcineurin A alpha) -4.13 11 (5, 7, 9, 17–19, 21, 28, 31, 33, 39) 4q21–q24
PPP3CB Protein phosphatase 3 (formerly 2B), catalytic subunit, beta isoform (calcineurin A beta) -3.65 10 (5, 7, 9, 17–19, 21, 28, 31, 33, 39) 10q21–q22
PRKX Protein kinase, X-linked 2.08 10 (5, 7, 8, 10, 19, 20, 24, 28, 46, 48) Xp22.3
PPP3R1 Protein phosphatase 3 (formerly 2B), regulatory subunit B, 19 kDa, alpha isoform (calcineurin B, type I) -3.05 10 (5, 7, 9, 17–19, 21, 28, 33, 39) 2p15
IKBKB Inhibitor of kappa light polypeptide gene enhancer in B cells, kinase beta 2.75 9 (5, 9, 14, 15, 18, 24, 33, 35, 40) 8p11.2
JUN v-jun sarcoma virus 17 oncogene homolog (avian) 2.49 9 (4, 5, 8, 14, 15, 18, 26, 28, 33) 1p32-p31
PAK1 p21/Cdc42/Rac1-activated kinase 1 (STE20 homolog, yeast) -7.87 7 (4–6, 15, 17, 21, 33) 11q13–q14
TP53 Tumor protein p53 (Li-Fraumeni syndrome) 3.14 7 (5, 9, 26, 28, 29, 31, 38) 17p13.1
PLCB1 Phospholipase C, beta 1 (phosphoinositide-specific) -2.73 7 (3, 7, 8, , 10, 19, 22, 28) 20p12
CALM1 Calmodulin 1 (phosphorylase kinase, delta) -2.22 7 (3, 7, 8, 19, 20, 24, 29) 14q24–q31
PDGFRA Platelet-derived growth factor receptor, alpha polypeptide 5.76 7 (4–6, 10, 19, 26, 47) 4q11–q13
CALM3 Calmodulin 3 (phosphorylase kinase, delta) -2.31 7 (3, 7, 8, 19, 20, 24, 29) 19q13.2–q13.3
ITGB1 Integrin, beta 1 (fibronectin receptor, beta polypeptide, antigen CD29 includes MDF2, MSK12) 2.26 6 (2, 4, 6, 11, 16, 21) 10p11.2
ITPR1 Inositol 1,4,5-triphosphate receptor, type 1 -8.62 6 (3, 7, 8, 10, 19, 22) 3p26–p25
ITPR2 Inositol 1,4,5-triphosphate receptor, type 2 5.42 6 (3, 7, 8, 10, 19, 22) 12p11
ROCK1 Rho-associated, coiled-coil containing protein kinase 1 2.42 6 (4, 6, 16, 21, 28, 44) 18q11.1
NFKBIA Nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha 2.17 6 (9, 14, 15, 18, 33, 35) 14q13
ROCK2 Rho-associated, coiled-coil containing protein kinase 2 -2.40 6 (4, 6, 16, 21, 28, 44) 2p24
CCND1 Cyclin D1 3.77 5 (4, 26, 28, 38, 45) 11q13
CAMK2A Calcium/calmodulin-dependent protein kinase (CaM kinase) II alpha -36.77 5 (7, 8, 19, 20, 28) 5q33.1
CAMK2B Calcium/calmodulin-dependent protein kinase (CaM kinase) II beta -8.77 5 (7, 8, 19, 20, 28) 7p14.3-p14.1
GNAI3 Guanine nucleotide binding protein (G protein), alpha inhibiting activity polypeptide 3 2.72 5 (10, 16, 21, 22, 36) 3p21
TGFB1 Transforming growth factor, beta 1 (Camurati-Engelmann disease) 2.32 5 (5, 26, 38, 44, 47) 19q13.1
ACTN1 Actinin, alpha 1 2.26 5 (4, 6, 16, 36, 37) 14q22–q24
PAK2 p21 (CDKN1A)-activated kinase 2 2.23 5 (4–6, 21, 33) 3q29
FOS v-fos FBJ murine osteosarcoma viral oncogene homolog 5.10 5 (5, 14, 18, 26, 33) 14q24.3
ACTN2 Actinin, alpha 2 -2.52 5 (4, 6, 16, 36, 37) 1q42–q43
MAP2K4 Mitogen-activated protein kinase kinase 4 -3.30 5 (5, 8, 14, 15, 41) 17p11.2
PLA2G6 Phospholipase A2, group VI (cytosolic, calcium-independent) -2.02 5 (5, 8, 2, 39, 41) 22q13.1
PTPN6 Protein tyrosine phosphatase, nonreceptor type 6 2.21 5 (17, 18, 33, 37, 45) 12p13
FAS Fas (TNF receptor superfamily, member 6) 5.90 5 (5, 9, 13, 17, 47) 10q24.1
CCND2 Cyclin D2 2.02 5 (4, 26, 28, 38, 45) 12p13
PLA2G12A Phospholipase A2, group XIIA 3.99 5 (5, 8, 22, 39, 41) 4q25
GNAS GNAS complex locus -4.93 5 (8, 10, 19, 22, 48) 20q13.3
GNAI1 Guanine nucleotide binding protein (G protein), alpha inhibiting activity polypeptide 1 -2.25 5 (10, 16, 21, 22, 36) 7q21
CAMK2G Calcium/calmodulin-dependent protein kinase (CaM kinase) II gamma -3.40 5 (7, 8, 19, 20, 28) 10q22

The numbers inside the parentheses correspond to the specific pathways in Table 2. Genes highlighted in red are also shown in Table 1.

Validation of Differential Gene Expression by RT-QPCR Analysis

To validate the expression results obtained from the Affymetrix array experiments, the expression of six genes that showed a two-fold and significant difference in expression in PA compared to the normal controls was assessed using RT-QPCR. The array data demonstrated that CCNA1, SPINT2, MAPK1, and MATK were significantly down-regulated and TIMP1 and MCL1 were significantly up-regulated. Real-time quantitative polymerase chain reaction confirmed these changes in gene expression in PA compared to normal brain (Table W7).

Genomic Aberrations Found in Pediatric PA

Array-based comparative genomic hybridization was carried out using the SpectralChip 2600 to identify regions of chromosome gain and loss in 11 pediatric PA. The array profile from the most altered tumor, IN2368, is illustrated in Figure 2. In total, 97 individual BAC clones were found to be either gained or lost in one or more tumors. Of these, 26 were gained, 64 were lost, and 7 were either gained or lost in different tumors (Figure 3; see Table W8, a–l, for details of these 97 altered clones and the combined dye swap raw data for each sample). Several chromosome regions were frequently altered in PA. BAC clones mapping to 1p36.32, 14q12, 19p13.2, and 22q13.33 showed gain, loss, or both in 91%, 82%, 64%, and 91% of tumors, respectively. Other regions less frequently altered included the following: 7q11.2 and 8q21 in 45% of tumors; 7q34, 8q24.3, 10q21.3, and 17q21 in 36% of tumors; and 6q12, 6q13, 17q21.3, and 19p13.3 in 27% of tumors. A small region containing two adjacent clones at 1p13 was lost in one tumor and a region that varied in size containing up to seven adjacent clones at 7q11.23 was lost in seven tumors. Large-scale copy number variants (LCVs) in specific chromosome regions have been characterized in the normal population. Of 97 clones altered in PA of this study, 37 are located in regions of known LCV (Figure 3).

Figure 2.

Figure 2

Combined dye swap aCGH profile for the tumor with the most aberrations, IN2368. Iterative linear regression global normalizations were used to reduce the SD of spot log2 ratios with an outlier range of ±2SD. Clones that showed a log2 ratio in excess of ±3SD were deemed to be gained or lost (indicated by the red dotted line).

Figure 3.

Figure 3

Results of aCGH analysis illustrating regions of chromosome gain and loss in pediatric PA. Circles to the right of each chromosome ideogram show the number of individual tumors with copy gains (red) and losses (blue) for each clone among the 11 PA studied. Clones in a region of copy number variation in the normal population are indicated by a green ring. Adjacent clones lost in the same tumor are indicated by a yellow box. Multiple clones in a region are indicated by two arrows. Correlations between clone CNAs and differential gene expression in the same region are indicated by a purple box.

Validation of DNA Copy Number by RT-QPCR Analysis

To validate the results obtained from the SpectralChip 2600 array, RT-QPCR analysis was used to determine the relative genomic copy number at five genomic regions and/or gene loci (see Table W9 for standard curve method raw data). This approach confirmed CNAs (both loss and gain) of the BAC clone RP4-703E10 at 1p36.32 in 80% of cases (Tables 4 and 5). Loss of BLC7A at 12q24.33 was verified by RT-QPCR in the single tumor, which demonstrated loss of the equivalent BAC clone (RP11-87C12), and in 11 additional tumors, indicating the superior sensitivity of RT-QPCR analysis in the detection of small CNAs [46]. BCL7B lies within a small deletion of up to seven adjacent clones at 7q11.23. Real-time quantitative polymerase chain reaction analysis confirmed loss of this gene in five of six tumors and detected loss in seven additional cases. Loss of clone RP11-125A5 in the region of LCV at 14q12 was verified by RTQPCR analysis in only two of eight tumors demonstrating loss by BAC array analysis, similar to the findings of the recent study by Qiao et al. [47].

Table 4.

SD and SEM Are Derived from the Control Values from Normal DNA Obtained from Blood from Four Individuals and One Male and One Female Commercially Available Pooled DNA Samples (see Table W8 for Raw Data).

Equation of Standard Curve SD Normalized SEM Normalized 95% Reference Range Deletion ≤ Average Control Copy Number Minus 95% Reference Range Gain ≥ Average Control Copy Number Plus 95% Reference Range
β-actin y = -3.4929x + 30.726
R2 = 0.9928
1p36.32 y = -3.3914x + 29.223 0.05 0.02 0.09 0.91 1.09
R2 = 0.998
BLC7B y = -3.4301x + 28.386 0.06 0.03 0.12 0.88 1.12
R2 = 0.9814
BLC7A y = -3.1059x + 28.599 0.05 0.02 0.10 0.90 1.10
R2 = 0.9934
14q12 y = -3.2777x + 28.844 0.12 0.05 0.23 0.77 1.23
R2 = 0.9979
FOXG1B y = -3.0543x + 30.212 0.12 0.05 0.24 0.76 1.24
R2 = 0.9935

Table 5.

Relative Copy Number for Each Gene or Region Was Calculated as Follows: Deletion: ≤ Copy Number of Controls Minus 95% Reference Range of Controls (or 2SD of Controls), Gain: ≥ Copy Number of Controls Plus 95% Reference Range of Controls (or 2SD of Controls).

Tumor 1p36.32 (RP4-703E10) BCL7B* BCL7A (RP11-87C12) 14q12 (RP11-125A5) FOXG1B Promoter
BAC array/RT-QPCR BAC array/RT-QPCR BAC array/RT-QPCR BAC array/RT-QPCR RT-QPCR
IN1740 Loss/0.54 No CNA/0.38 No CNA/0.53 No CNA/0.71 0.50
IN2356 ND/0.85 ND/0.88ND/0.79 ND/0.62 1.07
IN2368 No CNA/1.05 Loss/0.31 No CNA/0.85 Loss/0.89 1.12
IN2524 Gain/1.22 Loss/1.08 No CNA/1.01 Loss/0.33 1.30
IN2631 Gain/1.17 No CNA/0.24 No CNA/1.10 Loss/1.10 0.95
IN2674 Loss/0.85 Loss/0.48 No CNA/0.51 Loss/0.85 1.04
IN2788 Loss/0.37 Loss/0.23 No CNA/0.29 No CNA/0.74 0.32
IN2825 ND/0.99 ND/0.66 ND/0.67 ND/0.96 0.69
IN2921 ND/0.93 ND1.03 ND/0.34 ND/0.90 0.38
IN2940 Loss/1.10 No CNA/0.59 No CNA/0.44 Loss/1.01 0.49
IN2969 Gain/0.61 No CNA/0.52 No CNA/0.48 Loss/0.91 0.55
IN3013 Loss/0.46 Loss/0.44 No CNA/0.35 Loss/0.47 0.32
IN3115 Gain/1.12 Loss/0.33 Loss/0.38 Loss/0.98 1.06
IN3156 ND/0.33 ND/0.35 ND/0.34 ND/0.28 0.48

BAC clone alterations for each region are indicated by loss or gain. Blue values correspond to deletions detected by RT-QPCR. Red values correspond to gains detected by RT-QPCR.

*

BCL7B is not located under a specific clone but maps within the small region of loss encompassing up to seven clones at 7q11.23. ND indicates not done.

FOXG1B maps adjacent to clone RP11-125A5 at 14q12.

Correlation of Genomic and Expression Array Data

Reliable gene expression data were not available for all genes located in the same regions as the 97 BAC clones which were lost and/or gained. Overall, loss of four single clones correlated with down-regulation of genes mapping to the same region. These comprised SH3GL2 at 9p21.2-p23 (clone lost in one tumor), DRD1IP at 10q26.3 (clone lost in one tumor), BCL7A at 12q24.33 (clone lost in one tumor), and TUBG2 and CNTNAP1 at 17q21.31 (clone lost in two tumors). In seven tumors, there was also loss of a region at 7q11.23 containing up to seven adjacent clones that mapped to 71,658,093–73,945,118 (Figure 4). This region contains 47 genes in total, but only 17 were present on the Affymetrix GeneChip HG_U133A. Of these genes, seven showed unreliable expression data, nine were expressed at a normal level, and BCL7B was significantly down-regulated in all tumors compared to normal controls (P < .05). In addition, FOXG1B maps adjacent to the clone RP11-125A5 at 14q12, which was determined to be lost by aCGH and showed significant down-regulation in all PA compared to normal controls (P < .01). Real-time quantitative polymerase chain reaction analysis of relative DNA copy number of the FOXG1B promoter demonstrated genomic loss in 8 of 14 tumors.

Figure 4.

Figure 4

Loss of between two and seven contiguous BAC clones at 7q11.23 in seven PA. The rows of colored spots represent individual tumors and single BAC clones losses at 7q11.23. The distance between each spot is representative of the BAC clone binding locations on DNA.

Critically, although clone loss at these regions was not seen in every tumor, all samples showed a twofold and significant down-regulation in the specific genes at these loci listed in Table 6. No correlations were found between clone gain and gene overexpression.

Table 6.

BAC Clone Losses and Differential Gene Expression in the Same Region in Pediatric PA.

Clone Location Clone Name Clone Alteration Gene Located in Clone Region Fold Change in Expression Description
7q11.23 RP11-35P20, B315H11, CITB-51J22, B270D13, B39H04, RP11-137E8, RP11-89A20 Loss BCL7B -2.14 B-cell CLL/lymphoma 7B
9p21.2–p23 RP11-163F8 Loss SH3GL2 -6.94 SH3-domain GRB2-like 2
10q26.3 RP11-122K13 Loss DRD1IP -35.09 Dopamine receptor D1 interacting protein
12q24.33 RP11-87C12 Loss BCL7A -2.98 B-cell CLL/lymphoma 7A
14q12 RP11-125A5 Loss FOXG1B -325.73 Forkhead box G1B
17q21.31 AC100793.8 Loss TUBG2 -2.28 Tubulin, gamma 2
CNTNAP1 -2.99 Contactin-associated protein 1

Methylation-Specific PCR and Sequencing

Methylation-specific PCR products were only generated by primer sets specific for unmethylated promoter regions, indicating that the promoter regions of CDKN1C, PRDM2, SPINT2, REPRIMO, CCNA1, and DAPK1 were not methylated in the 17 PA analyzed. Sequencing of each gene in IN3115 and IN2940 and normal brain confirmed that promoter region methylation was not present in either PA or the normal samples (see Figure W2, a and b, for gel electrophoresis and sequencing results).

Discussion

This investigation has focused on the correlation between genomic CNAs at a 1-Mb resolution and global differential gene expression in PA. Overall, these data remain descriptive with the aims of identifying novel candidate target genes and characterizing cellular pathways that are involved in tumor development and growth. Future functional studies are needed to evaluate these targets in pediatric PA, but as discussed, evidence supports a role for these candidates in tumorigenesis.

Subgroups and Molecular Signatures in Pediatric PA

Unsupervised hierarchical clustering of PA in this study grouped tumor and control samples separately but further subgroups within PA were not identified. Wong et al. [16] previously demonstrated that two subgroups of PA could be distinguished by a molecular signature and suggested that these subgroups represented PA with different potentials of progression. Although it has been documented that pediatric PA that undergo partial excision may reoccur in the same location, malignant progression is rare [3,48]. In the PA included in our study, 13 of 89 genes from the molecular signature for which there were reliable data showed a continuous change in expression rather than a significant differential expression between two PA subgroups. However, the authors recognize that only a proportion of the genes could be investigated here in a smaller sample group compared to that of Wong et al. [16]. Sharma et al. [17] also reported a molecular signature of 36 genes with differential expression between supratentorial PA and posterior fossa PA and suggested that glial tumors have an intrinsic, lineage-specific signature that reflects the region of the brain from which the nonmalignant tumor cell precursors originated. In the present tumor group, most tumors were located in the posterior fossa and, consequently, a comparison was not possible.

The characterization of molecular signatures that can distinguish PA arising in different locations or tumor subgroups emphasizes the intrinsic genetic heterogeneity of PA and the need for large sample numbers to identify valid subgroups with clinical or prognostic relevance.

Control Samples in Pediatric PA Microarray Analysis

The use of various controls to identify differentially expressed genes between normal and diseased samples in microarray experiments can directly influence those genes deemed to show differential gene expression, as reported by Zorn et al. [49] who investigated the choice of normal controls in the study of ovarian carcinoma. The normal controls used in previous microarray studies of brain tumors vary greatly. Studies comparing tumor expression profiles to normal brain have used total RNA from tissue samples obtained from normal brain regions including tissue from a lobectomy after edema, hippocampus tissue from a patient with epilepsy, subcortical white matter, cortical brain, the cortex of temporal lobe, and postmortem cortex, and medulla tissues [50–54]. Groups who have not had direct access to normal brain have also commercially purchased total RNA samples of adult and fetal normal brain for use as controls, including Wong et al. [16,50].

The control samples of this study are pooled adult normal wholebrain samples from a commercial source. This removes the chance of tumor contamination within the normal sample and using pooled normal samples reduces expression variations present in each individual. The decision to use adult normal total brain as a control was reached after detailed investigations of both fetal and adult brain from specific brain regions and whole brain (Figure W3).

Hierarchical clustering of adult and fetal normal controls independently of the tumors suggested that either fetal or adult normal controls were suitable for the study of tumor biopsy samples. Furthermore, 1345 common differentially expressed probe sets were identified in the PA when using either adult or fetal normal controls. However, it was noted that fetal brain is in an exponential or linear growth phase and that this may affect gene expression and signaling pathways, masking aspects of the tumor profile if used as a control [55]. Increased proliferation, cell signaling, and reduced apoptosis are associated with fetal development and are characteristics of tumor cells. Furthermore, on investigation of the cellular pathways influenced by differential gene expression in the PA when adult or fetal normal controls were used and in the normal fetal controls when compared to adult normal controls, it became apparent that genes of the Wnt signaling pathway, cell cycle, and, to a lesser extent, the colorectal cancer pathway were up-regulated in the fetal normal controls compared to the adult normal controls and masked some differential gene expression in the PA. Moreover, the Wnt signaling pathway has been implicated in fetal development [56–60].

Variation was also seen between the expression profiles of total brain, cerebellum and corpus callosum from both adult and fetal origin. Furthermore, the tumors of this study are located in brain regions other than the cerebellum and corpus callosum, and it was felt inappropriate to compare tumors from different locations to normal controls taken from these regions, particularly because unique molecular signatures have been characterized from PA located in supratentorial and posterior fossa regions as previously discussed [17]. Consequently, to standardize the normal control group and reduce sample heterogeneity, four pooled adult whole-brain samples from between three and five individuals were selected as controls. Although variation can be seen between the four control samples, it is critical to note that if any one of these four is removed and the analysis repeated, the overall expression analysis results are unchanged with only minor differences in exact fold change values.

Genes Involved in Pediatric PA Development

To investigate pediatric PA development, those genes that showed a greater than 10-fold change in expression and were either involved in a KEGG pathway or were well characterized were identified. The overexpression of SERPINA3 and CHI3L1 provides evidence that the tumor cells are of astrocytic lineage. SERPINA3 is specifically expressed in astrocytes and is involved in the expression of astrocytic markers such as glial fibrillary acidic protein (GFAP) [61,62]. GFAP was found to be 1.98-fold up-regulated in this study. CHI3L1 is another astrocytic marker that has been used previously as a diagnostic indicator in astrocytoma [63,64].

Several genes listed in Table 1 have been investigated in other grades of astrocytoma including T1A-2, ABCC3, CD44, MYT1, TIMP1, HLA-DPA1, TFPI, CSPG2, CD151, WNT10B, PTK2B, ENPP2, MBP, MATK, SST, CCK, and FOXG1B. T1A-2 expression was found to be significantly higher in glioblastoma multiforme (GBM) compared to anaplastic astrocytoma or diffuse astrocytoma, and it is a marker of malignant progression [65]. The large fold change in PA compared to normal brain suggests that this gene is involved in promoting PA development.

Other genes from this group have functions associated with a range of tumor characteristics. For example, increased expression of ABCC3 from the multidrug-resistant protein family of ATP-dependent efflux pumps may mediate drug resistance in PA [66]. There is evidence to suggest that CSPG2 protects from oxidative stress induced apoptosis and promotes tumor growth and angiogenesis [67]. Interestingly, CPGS4 from the same family was found to be up-regulated in diffuse and malignant astrocytoma in response to platelet-derived growth factor (PDGF) and increases tumor cell proliferation and invasion [68,69]. In the present study, CSPG2 and CPGS4 demonstrated a 10.86- and 23.77-fold up-regulation in expression, respectively, in addition to PDGFRA that was also overexpressed by 5.84-fold.

The remaining genes listed in Table 1 either have been associated with the development of other tumors and cancers or have functions that could promote or inhibit tumor development. POSTN, for example, is overexpressed in more than 80% of human colon cancers and demonstrates the highest fold change expression in oral squamous cell carcinoma. POSTN enhances cancer growth by preventing stress-induced apoptosis and enhancing endothelial cell survival promoting angiogenesis [70,71].

Pathways with Differential Gene Expression in Pediatric PA

The Onto-Tools software ranked the KEGG pathways influenced by the 1844 differentially expressed genes according to the extent of disruption. In PA, the KEGG pathway antigen processing and presentation was most affected by differential gene expression. With the exception of HSP90A, all identified genes involved in this pathway were up-regulated, including CD74, HLA-DPA1, HLA-DRA, HLA-DMA, HLA-DRB1, HLA-DMB, HLA-DQB1, HLA-G, CTSS, HLA-B, KLRC3, HLA-F, HLA-A, PSME2, PDIA3, HLA-E, IFI30, TAP1, and B2M. A previous study noted an increase in the expression of genes involved in immune response in PA compared to diffuse astrocytoma, oligodendrogliomas, and normal brain [72]. An increase in HLA-DPA1 expression in PA compared to anaplastic astrocytoma was also observed in a second study [73]. Furthermore, HLA-DR overexpression has been associated with good prognosis in large-bowel carcinoma and serous ovarian tumors [74,75]. The up-regulation of immune response genes in PA may contribute to the benign behavior of this tumor type.

The KEGG phosphatidylinositol (PI3K) signaling system and the mitogen-activated protein kinase (MAPK) signaling pathway are ranked as the third and fifth, respectively, according to the extent of differential gene expression in PA (Table 2). Altered signaling in these pathways through increased receptor stimulation and disrupted tumor suppressor or oncogene function has been characterized in other grades of astrocytoma particularly primary and secondary adult GBM [76].

Genomic Aberrations Previously Identified in Pediatric PA

Few studies have identified nonrandom whole-chromosome or arm aberrations in PA of the pediatric population. As discussed in the introduction, Jones et al. [25] only identified CNAs in children older than 10 years using aCGH. Furthermore, using analog CGH, Sanoudou et al. [5] identified five PA (12%) with whole-chromosome aberrations, and all were from patients 7 years or older (four patients were 10 years or older). Of the tumors in this study, 59% were from patients older than 7 years and 41% were from patients older than 10 years. Possibly, whole-chromosome or arm aberrations maybe more common in PA of older children. In our study, five cases (45%) were from patients 7 years or older and only one case was from a patient older than 10 years. This may explain why we have not identified any whole-chromosome or arm aberrations in the tumors of this patient cohort.

Genomic Loss in Regions of LCVs in Pediatric PA

Of 97 BAC clones altered in PA, 40% were located in regions of LCV present in the normal population [77–84]. The role of LCVs (which involve gain or loss of hundreds of kilobases of genomic DNA) in phenotypically normal individuals is not clear [77]. However, LCVs at 14q12 were found to be present in DNA from 50% of chronic myeloid leukemia and pediatric solid tumors compared to an incidence of 10% in somatic DNA of healthy control individuals, suggesting that acquired or inherited LCVs at 14q12 may be associated with the onset or progression of neoplasia [85]. In our study, the incidence of 14q12 aberrations in PA was 79%, as identified by combined aCGH and RT-QPCR analyses. Further investigations are necessary to establish the incidence of LCVs in somatic DNA of PA patients to determine whether 14q12 aberrations are inherited or are acquired during tumor development.

Correlation of CNAs and Gene Expression in Pediatric PA

Loss of individual clones at 9p21.2-p23, 10q26.3, 12q24.33, and 17q21.31 and a small region of up to seven adjacent clones at 7q11.23 correlated with differential gene expression. Crucially, although BAC array analysis did not identify genomic loss in all tumors, the down-regulation of genes including SH3GL2 at 9p21.2-p23, DRD1IP at 10q26.3, BCL7A at 12q24.33, TUB2 and CNTNAP1 at 17q21.31, and BCL7B at 7q11.23 was seen in all cases. Furthermore, RT-QPCR analysis of relative DNA copy number of BCL7B at 7q11.23 and BCL7A at 12q24.33 identified loss in 12 (86%) of 14 tumors. BCL7B and BCL7A are from the same gene family and share 90% homology. BCL7B maps to a region deleted in the congenital disorder Williams syndrome, although little is known about its function [86]. BCL7A is a putative tumor suppressor gene, which is hypermethylated in the primary cutaneous T-cell lymphoma [87]. Earlier studies have demonstrated the involvement of the BCL7A locus in a recurrent break point in B-cell lymphomas and a complex translocation and rearrangement in specific cell lines [88]. Expression profiling of mycosis fungoides, a common cutaneous T-cell lymphoma and B-cell lymphoma, identified the down-regulation of BCL7A as part of molecular signatures that could distinguish mycosis fungoides and inflammatory dermatoses and distinct types of diffuse large B-cell lymphoma [89,90]. The loss of BCL7A has also been associated with disrupted NF-κB signaling and unfavorable prognosis in patients with B-cell lymphomas, suggesting that this gene has tumor suppressor properties [91].

The clone aberrations detected at 14q12 by BAC array analysis could only be confirmed by RT-QPCR in two cases. A previous study also failed to confirm RP11-125A5 loss [47]. This region is a common site for duplication and deletion events in hepatoblastoma, which may explain the discrepancies between CNA detected by BAC array and RT-QPCR analysis in the PA of this study [92]. However, FOXG1B maps adjacent to clone RP11-125A5 and shows significant down-regulation in all PA. Real-time quantitative polymerase chain reaction analysis of relative DNA copy number of the FOXG1B promoter demonstrated genomic loss in almost 60% of tumors examined, indicating that genomic loss is an important mechanism in the underexpression of this gene.

FOXG1B is an oncogenic transformer and transcriptional repressor, which interacts with Groucho, Hes, and Smad proteins [93,94]. More importantly, FOXG1B has been proposed as a major FoxO forkhead transcription factor that acts as a signal transducer between the Smad, PI3K, and FoxG1B pathways. The activity of these pathway interactions has been shown to be involved in the resistance to transforming growth factor beta (TGFβ)-mediated cytostasis during telencephalic neuroepithelium development and in GBM cells [95]. The involvement of FOXG1B in PI3K (the pathway most affected by the differential gene expression in PA) highlights the impact FOXG1B disruption may have on intracellular signaling. The location of this gene in a region of LCV at 14q12 also suggests that some individuals may be predisposed to PA development.

Reduction of SH3GL2 expression correlated with loss at 9p21.2-p23. A recent study indicates that SH3GL2 is involved in the development and progression of laryngeal squamous cell carcinoma and expression levels correlate with pathologic classification [96]. The expression of SH3GL2 also correlates with distinct stages of intestinaltype gastric cancer, although a functional role in tumorigenesis has not yet been determined [97].

TUBG2, located at 17q21.31, shares 97.3% amino acid identity with TUBG1, and the two genes are coexpressed in a variety of tissues. This suggests that the functions of TUBG1 and TUBG2 are similar, both having a significant role in the organization of the microtubule cytoskeleton [98]. CNTNAP1, also located at 17q21.31, is transcribed predominantly in the brain, and the architecture of the protein extracellular domain is similar to that of neurexins. CNTNAP1 plays an important role in the creation and maintenance of paranodal regions of myelinated axons, enabling recruitment and activation of intracellular signaling pathways in neurons [99]. DRD1IP maps to 10q26.3 and encodes a brain-specific dopamine receptor 1-interacting protein involved in calcium signaling [100]. A role for these genes in tumor development has yet to be established.

Promoter Hypermethylation in Pediatric PA

Down-regulation of gene expression through promoter hypermethylation is an important mechanism of transcriptional silencing in many tumors. Hypermethylation of the six genes investigated in this study has been detected in more than 30% of gastric carcinoma, lung cancer, and renal cell carcinoma. Moreover, CCNA1 was hypermethylated in 100% of colorectal adenoma [28–33]. All six genes demonstrated a twofold and significant down-regulation in PA compared to normal brain, but hypermethylation of gene promoter regions was not detected. Only one previous study has investigated hypermethylation in pediatric PA involving MGMT, GSTP1, DAPK1, p14ARF, THBS-1, TIMP-3, p73, p16INK4A, RB-1, and TP53. Hypermethylation of p16INK4A was most frequent occurring in 46% of cases. However, five of the remaining genes were hypermethylated in less than 8% of cases [101]. It is possible that methylation is not the predominant mechanism of gene silencing in PA development but that other epigenetic processes such as histone modification and microRNA play a more significant role.

We have clearly demonstrated that PA have distinct expression profiles compared to normal whole brain.We have identified two key signaling pathways (PI3K and MAPK) that contribute toward tumor development. Large regions of chromosome alterations were not identified in PA, although 97 individual BAC clones and a small region on chromosome 7 were lost and/or gained. This is the first study to establish a correlation between small regions of genomic CNA and differential gene expression in PA. The down-regulation of gene family members BCL7B and BCL7A due to independent CNAs suggests a tumor suppressor role for these genes in PA. Furthermore, we have demonstrated that genomic alterations in a region of LCV at 14q12 may predispose individuals to tumor development. The accompanying loss of FOXG1B expression in all samples together with its involvement in PI3K signaling provides compelling evidence that this gene may play a significant role in the development of pediatric PA.

Supplementary Material

Supplementary Figures and Tables
neo1008_0757SD1.pdf (755.7KB, pdf)

Acknowledgments

The authors thank Ali's Dream and the Rosetrees Trust for funding this study.

Footnotes

1

Supplementary data are available at the following links on request to the authors: http://www.ion.ucl.ac.uk/paper_data_np/SupplementaryData.doc, http://www.ion.ucl.ac.uk/paper_data_np/SupplementaryDataTables5an d8.xls.

References

  • 1.Kleihues P, Cavenee WK. Pathology and Genetics of Tumours of the Central Nervous System. Lyon, France: IARC Press; [Google Scholar]
  • 2.Gajjar A, Sanford RA, Heideman R, Jenkins JJ, Walter A, Li Y, Langston JW, Muhlbauer M, Boyett JM, Kun LE. Low-grade astrocytoma: a decade of experience at St. Jude Children's Research Hospital. J Clin Oncol. 1997;8:2792–2799. doi: 10.1200/JCO.1997.15.8.2792. [DOI] [PubMed] [Google Scholar]
  • 3.Fernandez C, Figarella-Branger D, Girard N, Bouvier-Labit C, Gouvernet J, Paz PA, Lena G. Pilocytic astrocytomas in children: prognostic factors—a retrospective study of 80 cases. Neurosurgery. 2003;3:544–553. doi: 10.1227/01.neu.0000079330.01541.6e. [DOI] [PubMed] [Google Scholar]
  • 4.Rickert CH, Paulus W. Comparative genomic hybridization in central and peripheral nervous system tumors of childhood and adolescence. J Neuropathol Exp Neurol. 2004;5:399–417. doi: 10.1093/jnen/63.5.399. [DOI] [PubMed] [Google Scholar]
  • 5.Sanoudou D, Tingby O, Ferguson-Smith MA, Collins VP, Coleman N. Analysis of pilocytic astrocytoma by comparative genomic hybridization. Br J Cancer. 2000;6:1218–1222. doi: 10.1054/bjoc.1999.1066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Shlomit R, Ayala AG, Michal D, Ninett A, Frida S, Boleslaw G, Gad B, Gideon R, Shlomi C. Gains and losses of DNA sequences in childhood brain tumors analyzed by comparative genomic hybridization. Cancer Genet Cytogenet. 2000;1:67–72. doi: 10.1016/s0165-4608(00)00218-1. [DOI] [PubMed] [Google Scholar]
  • 7.White FV, Anthony DC, Yunis EJ, Tarbell NJ, Scott RM, Schofield DE. Nonrandom chromosomal gains in pilocytic astrocytomas of childhood. Hum Pathol. 1995;9:979–986. doi: 10.1016/0046-8177(95)90087-x. [DOI] [PubMed] [Google Scholar]
  • 8.Zattara-Cannoni H, Gambarelli D, Lena G, Dufour H, Choux M, Grisoli F, Vagner-Capodano AM. Are juvenile pilocytic astrocytomas benign tumors? A cytogenetic study in 24 cases. Cancer Genet Cytogenet. 1998;2:157–160. doi: 10.1016/s0165-4608(97)00455-x. [DOI] [PubMed] [Google Scholar]
  • 9.Phelan CM, Liu L, Ruttledge MH, Muntzning K, Ridderheim PA, Collins VP. Chromosome 17 abnormalities and lack of TP53 mutations in paediatric central nervous system tumours. Hum Genet. 1995;6:684–690. doi: 10.1007/BF00210300. [DOI] [PubMed] [Google Scholar]
  • 10.von Deimling A, Louis DN, Menon AG, von Ammon K, Petersen I, Ellison D, Wiestler OD, Seizinger BR. Deletions on the long arm of chromosome 17 in pilocytic astrocytoma. Acta Neuropathol (Berl) 1993;1:81–85. doi: 10.1007/BF00454903. [DOI] [PubMed] [Google Scholar]
  • 11.Willert JR, Daneshvar L, Sheffield VC, Cogen PH. Deletion of chromosome arm 17p DNA sequences in pediatric high-grade and juvenile pilocytic astrocytomas. Genes Chromosomes Cancer. 1995;3:165–172. doi: 10.1002/gcc.2870120303. [DOI] [PubMed] [Google Scholar]
  • 12.Hayes VM, Dirven CM, Dam A, Verlind E, Molenaar WM, Mooij JJ, Hofstra RM, Buys CH. High frequency of TP53 mutations in juvenile pilocytic astrocytomas indicates role of TP53 in the development of these tumors. Brain Pathol. 1999;3:463–467. doi: 10.1111/j.1750-3639.1999.tb00535.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Patt S, Gries H, Giraldo M, Cervos-Navarro J, Martin H, Janisch W, Brockmoller J. p53 gene mutations in human astrocytic brain tumors including pilocytic astrocytomas. Hum Pathol. 1996;6:586–589. doi: 10.1016/s0046-8177(96)90166-5. [DOI] [PubMed] [Google Scholar]
  • 14.Bertucci F, Finetti P, Rougemont J, Charafe-Jauffret E, Cervera N, Tarpin C, Nguyen C, Xerri L, Houlgatte R, Jacquemier J, et al. Gene expression profiling identifies molecular subtypes of inflammatory breast cancer. Cancer Res. 2005;6:2170–2178. doi: 10.1158/0008-5472.CAN-04-4115. [DOI] [PubMed] [Google Scholar]
  • 15.Tay ST, Leong SH, Yu K, Aggarwal A, Tan SY, Lee CH, Wong K, Visvanathan J, Lim D, Wong WK, et al. A combined comparative genomic hybridization and expression microarray analysis of gastric cancer reveals novel molecular subtypes. Cancer Res. 2003;12:3309–3316. [PubMed] [Google Scholar]
  • 16.Wong KK, Chang YM, Tsang YT, Perlaky L, Su J, Adesina A, Armstrong DL, Bhattacharjee M, Dauser R, Blaney SM, et al. Expression analysis of juvenile pilocytic astrocytomas by oligonucleotide microarray reveals two potential subgroups. Cancer Res. 2005;1:76–84. [PubMed] [Google Scholar]
  • 17.Sharma MK, Mansur DB, Reifenberger G, Perry A, Leonard JR, Aldape KD, Albin MG, Emnett RJ, Loeser S, Watson MA, et al. Distinct genetic signatures among pilocytic astrocytomas relate to their brain region origin. Cancer Res. 2007;3:890–900. doi: 10.1158/0008-5472.CAN-06-0973. [DOI] [PubMed] [Google Scholar]
  • 18.Cai WW, Mao JH, Chow CW, Damani S, Balmain A, Bradley A. Genome-wide detection of chromosomal imbalances in tumors using BAC microarrays. Nat Biotechnol. 2002;4:393–396. doi: 10.1038/nbt0402-393. [DOI] [PubMed] [Google Scholar]
  • 19.Misra A, Pellarin M, Nigro J, Smirnov I, Moore D, Lamborn KR, Pinkel D, Albertson DG, Feuerstein BG. Array comparative genomic hybridization identifies genetic subgroups in grade 4 human astrocytoma. Clin Cancer Res. 2005;8:2907–2918. doi: 10.1158/1078-0432.CCR-04-0708. [DOI] [PubMed] [Google Scholar]
  • 20.Nigro JM, Misra A, Zhang L, Smirnov I, Colman H, Griffin C, Ozburn N, Chen M, Pan E, Koul D, et al. Integrated array-comparative genomic hybridization and expression array profiles identify clinically relevant molecular subtypes of glioblastoma. Cancer Res. 2005;5:1678–1686. doi: 10.1158/0008-5472.CAN-04-2921. [DOI] [PubMed] [Google Scholar]
  • 21.Rossi MR, La DJ, Matsui S, Nowak NJ, Hawthorn L, Cowell JK. Novel amplicons on the short arm of chromosome 7 identified using high resolution array CGH contain over expressed genes in addition to EGFR in glioblastoma multiforme. Genes Chromosomes Cancer. 2005;4:392–404. doi: 10.1002/gcc.20256. [DOI] [PubMed] [Google Scholar]
  • 22.Ruano Y, Mollejo M, Ribalta T, Fiano C, Camacho FI, Gomez E, de Lope AR, Hernandez-Moneo JL, Martinez P, Melendez B. Identification of novel candidate target genes in amplicons of glioblastoma multiforme tumors detected by expression and CGH microarray profiling. Mol Cancer. 2006;5:39. doi: 10.1186/1476-4598-5-39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Vranova V, Necesalova E, Kuglik P, Cejpek P, Pesakova M, Budinska E, Relichova J, Veselska R. Screening of genomic imbalances in glioblastoma multiforme using high-resolution comparative genomic hybridization. Oncol Rep. 2007;2:457–464. [PubMed] [Google Scholar]
  • 24.Wrensch M, McMillan A, Wiencke J, Wiemels J, Kelsey K, Patoka J, Jones H, Carlton V, Miike R, Sison J, et al. Nonsynonymous coding singlenucleotide polymorphisms spanning the genome in relation to glioblastoma survival and age at diagnosis. Clin Cancer Res. 2007;1:197–205. doi: 10.1158/1078-0432.CCR-06-1199. [DOI] [PubMed] [Google Scholar]
  • 25.Jones DT, Ichimura K, Liu L, Pearson DM, Plant K, Collins VP. Genomic analysis of pilocytic astrocytomas at 0.97 Mb resolution shows an increasing tendency toward chromosomal copy number change with age. J Neuropathol Exp Neurol. 2006;11:1049–1058. doi: 10.1097/01.jnen.0000240465.33628.87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Wong KK, Tsang YT, Chang YM, Su J, Di Francesco AM, Meco D, Riccardi R, Perlaky L, Dauser RC, Adesina A, et al. Genome-wide allelic imbalance analysis of pediatric gliomas by single nucleotide polymorphic allele array. Cancer Res. 2006;23:11172–11178. doi: 10.1158/0008-5472.CAN-06-2438. [DOI] [PubMed] [Google Scholar]
  • 27.Costello JF, Plass C, Cavenee WK. Aberrant methylation of genes in low-grade astrocytomas. Brain Tumor Pathol. 2000;2:49–56. doi: 10.1007/BF02482735. [DOI] [PubMed] [Google Scholar]
  • 28.Kikuchi T, Toyota M, Itoh F, Suzuki H, Obata T, Yamamoto H, Kakiuchi H, Kusano M, Issa JP, Tokino T, et al. Inactivation of p57KIP2 by regional promoter hypermethylation and histone deacetylation in human tumors. Oncogene. 2002;17:2741–2749. doi: 10.1038/sj.onc.1205376. [DOI] [PubMed] [Google Scholar]
  • 29.Morris MR, Gentle D, Abdulrahman M, Maina EN, Gupta K, Banks RE, Wiesener MS, Kishida T, Yao M, Teh B, et al. Tumor suppressor activity and epigenetic inactivation of hepatocyte growth factor activator inhibitor type 2/SPINT2 in papillary and clear cell renal cell carcinoma. Cancer Res. 2005;11:4598–4606. doi: 10.1158/0008-5472.CAN-04-3371. [DOI] [PubMed] [Google Scholar]
  • 30.Oshimo Y, Oue N, Mitani Y, Nakayama H, Kitadai Y, Yoshida K, Chayama K, Yasui W. Frequent epigenetic inactivation of RIZ1 by promoter hypermethylation in human gastric carcinoma. Int J Cancer. 2004;2:212–218. doi: 10.1002/ijc.20090. [DOI] [PubMed] [Google Scholar]
  • 31.Takahashi T, Suzuki M, Shigematsu H, Shivapurkar N, Echebiri C, Nomura M, Stastny V, Augustus M, Wu CW, Wistuba II, et al. Aberrant methylation of Reprimo in human malignancies. Int J Cancer. 2005;4:503–510. doi: 10.1002/ijc.20910. [DOI] [PubMed] [Google Scholar]
  • 32.Toyooka S, Toyooka KO, Miyajima K, Reddy JL, Toyota M, Sathyanarayana UG, Padar A, Tockman MS, Lam S, Shivapurkar N, et al. Epigenetic down-regulation of death-associated protein kinase in lung cancers. Clin Cancer Res. 2003;8:3034–3041. [PubMed] [Google Scholar]
  • 33.Xu XL, Yu J, Zhang HY, Sun MH, Gu J, Du X, Shi DR, Wang P, Yang ZH, Zhu JD. Methylation profile of the promoter CpG islands of 31 genes that may contribute to colorectal carcinogenesis. World J Gastroenterol. 2004;23:3441–3454. doi: 10.3748/wjg.v10.i23.3441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Sowar K, Straessle J, Donson AM, Handler M, Foreman NK. Predicting which children are at risk for ependymoma relapse. J Neurooncol. 2006;1:41–46. doi: 10.1007/s11060-005-9072-2. [DOI] [PubMed] [Google Scholar]
  • 35.Draghici S, Khatri P, Bhavsar P, Shah A, Krawetz SA, Tainsky MA. Onto-Tools, the toolkit of the modern biologist: Onto-Express, Onto-Compare, Onto-Design and Onto-Translate. Nucleic Acids Res. 2003;13:3775–3781. doi: 10.1093/nar/gkg624. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Khatri P, Sellamuthu S, Malhotra P, Amin K, Done A, Draghici S. Recent additions and improvements to the Onto-Tools. Nucleic Acids Res. 2005;33(Web Server issue):W762–W765. doi: 10.1093/nar/gki472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Kanehisa M, Goto S. KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 2000;1:27–30. doi: 10.1093/nar/28.1.27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Saeed AI, Sharov V, White J, Li J, Liang W, Bhagabati N, Braisted J, Klapa M, Currier T, Thiagarajan M, et al. TM4: a free, open-source system for microarray data management and analysis. Biotechniques. 2003;2:374–378. doi: 10.2144/03342mt01. [DOI] [PubMed] [Google Scholar]
  • 39.Yang YH, Dudoit S, Luu P, Lin DM, Peng V, Ngai J, Speed TP. Normalization for cDNA microarray data: a robust composite method addressing single and multiple slide systematic variation. Nucleic Acids Res. 2002;4:e15. doi: 10.1093/nar/30.4.e15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Quackenbush J. Microarray data normalization and transformation. Nat Genet. 2002;32 Suppl:496–501. doi: 10.1038/ng1032. [DOI] [PubMed] [Google Scholar]
  • 41.Finkelstein DB, Gollub J, Ewing R, Sterky F, Somerville S, Cherry JM. Iterative linear regression by sector: renormalization of cDNA microarray data and cluster analysis weighted by cross homology. In: Lin SM, Johnson KF, editors. Methods of Microarray Analysis. 2002. [Google Scholar]
  • 42.Senchenko V, Liu J, Braga E, Mazurenko N, Loginov W, Seryogin Y, Bazov I, Protopopov A, Kisseljov FL, Kashuba V, et al. Deletion mapping using quantitative real-time PCR identifies two distinct 3p21.3 regions affected in most cervical carcinomas. Oncogene. 2003;19:984–2992. doi: 10.1038/sj.onc.1206429. [DOI] [PubMed] [Google Scholar]
  • 43.Suarez-Merino B, Hubank M, Revesz T, Harkness W, Hayward R, Thompson D, Darling JL, Thomas DG, Warr TJ. Microarray analysis of pediatric ependymoma identifies a cluster of 112 candidate genes including four transcripts at 22q12.1-q13.3. Neuro Oncol. 2005;1:20–31. doi: 10.1215/S1152851704000596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Olek A, Oswald J, Walter J. A modified and improved method for bisulphite based cytosine methylation analysis. Nucleic Acids Res. 1996;24:5064–5066. doi: 10.1093/nar/24.24.5064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Warnecke PM, Stirzaker C, Melki JR, Millar DS, Paul CL, Clark SJ. Detection and measurement of PCR bias in quantitative methylation analysis of bisulphite-treated DNA. Nucleic Acids Res. 1997;21:4422–4426. doi: 10.1093/nar/25.21.4422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Coe BP, Ylstra B, Carvalho B, Meijer GA, Macaulay C, Lam WL. Resolving the resolution of array CGH. Genomics. 2007;5:647–653. doi: 10.1016/j.ygeno.2006.12.012. [DOI] [PubMed] [Google Scholar]
  • 47.Qiao Y, Liu X, Harvard C, Nolin SL, Brown WT, Koochek M, Holden JJ, Lewis ME, Rajcan-Separovic E. Large-scale copy number variants (CNVs): distribution in normal subjects and FISH/real-time qPCR analysis. BMC Genomics. 2007;8:167. doi: 10.1186/1471-2164-8-167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Krieger MD, Gonzalez-Gomez I, Levy ML, McComb JG. Recurrence patterns and anaplastic change in a long-term study of pilocytic astrocytomas. Pediatr Neurosurg. 1997;1:1–11. doi: 10.1159/000121218. [DOI] [PubMed] [Google Scholar]
  • 49.Zorn KK, Jazaeri AA, Awtrey CS, Gardner GJ, Mok SC, Boyd J, Birrer MJ. Choice of normal ovarian control influences determination of differentially expressed genes in ovarian cancer expression profiling studies. Clin Cancer Res. 2003;13:4811–4818. [PubMed] [Google Scholar]
  • 50.Godard S, Getz G, Delorenzi M, Farmer P, Kobayashi H, Desbaillets I, Nozaki M, Diserens AC, Hamou MF, Dietrich PY, et al. Classification of human astrocytic gliomas on the basis of gene expression: a correlated group of genes with angiogenic activity emerges as a strong predictor of subtypes. Cancer Res. 2003;20:6613–6625. [PubMed] [Google Scholar]
  • 51.Huang H, Colella S, Kurrer M, Yonekawa Y, Kleihues P, Ohgaki H. Gene expression profiling of low-grade diffuse astrocytomas by cDNA arrays. Cancer Res. 2000;24:6868–6874. [PubMed] [Google Scholar]
  • 52.Khatua S, Peterson KM, Brown KM, Lawlor C, Santi MR, LaFleur B, Dressman D, Stephan DA, MacDonald TJ. Overexpression of the EGFR/FKBP12/HIF-2alpha pathway identified in childhood astrocytomas by angiogenesis gene profiling. Cancer Res. 2003;8:1865–1870. [PubMed] [Google Scholar]
  • 53.Rickman DS, Bobek MP, Misek DE, Kuick R, Blaivas M, Kurnit DM, Taylor J, Hanash SM. Distinctive molecular profiles of high-grade and lowgrade gliomas based on oligonucleotide microarray analysis. Cancer Res. 2001;18:6885–6891. [PubMed] [Google Scholar]
  • 54.Shai R, Shi T, Kremen TJ, Horvath S, Liau LM, Cloughesy TF, Mischel PS, Nelson SF. Gene expression profiling identifies molecular subtypes of gliomas. Oncogene. 2003;31:4918–4923. doi: 10.1038/sj.onc.1206753. [DOI] [PubMed] [Google Scholar]
  • 55.Samuelsen GB, Larsen KB, Bogdanovic N, Laursen H, Graem N, Larsen JF, Pakkenberg B. The changing number of cells in the human fetal forebrain and its subdivisions: a stereological analysis. Cereb Cortex. 2003;2:115–122. doi: 10.1093/cercor/13.2.115. [DOI] [PubMed] [Google Scholar]
  • 56.Katoh M, Kirikoshi H, Saitoh T, Sagara N, Koike J. Alternative splicing of the WNT-2B/WNT-13 gene. Biochem Biophys Res Commun. 2000;1:209–216. doi: 10.1006/bbrc.2000.3252. [DOI] [PubMed] [Google Scholar]
  • 57.McMahon AP, Bradley A. The Wnt-1 (int-1) proto-oncogene is required for development of a large region of the mouse brain. Cell. 1990;6:1073–1085. doi: 10.1016/0092-8674(90)90385-r. [DOI] [PubMed] [Google Scholar]
  • 58.Parr BA, Shea MJ, Vassileva G, McMahon AP. Mouse Wnt genes exhibit discrete domains of expression in the early embryonic CNS and limb buds. Development. 1993;1:247–261. doi: 10.1242/dev.119.1.247. [DOI] [PubMed] [Google Scholar]
  • 59.Hoang BH, Thomas JT, bdul-Karim FW, Correia KM, Conlon RA, Luyten FP, Ballock RT. Expression pattern of two Frizzled-related genes, Frzb-1 and Sfrp-1, during mouse embryogenesis suggests a role for modulating action of Wnt family members. Dev Dyn. 1998;3:364–372. doi: 10.1002/(SICI)1097-0177(199807)212:3<364::AID-AJA4>3.0.CO;2-F. [DOI] [PubMed] [Google Scholar]
  • 60.Smalley MJ, Dale TC. Wnt signalling in mammalian development and cancer. Cancer Metastasis Rev. 1999;2:215–230. doi: 10.1023/a:1006369223282. [DOI] [PubMed] [Google Scholar]
  • 61.Gopalan SM, Wilczynska KM, Konik BS, Bryan L, Kordula T. Nuclear factor-1-X regulates astrocyte-specific expression of the alpha1-antichymotrypsin and glial fibrillary acidic protein genes. J Biol Chem. 2006;19:13126–13133. doi: 10.1074/jbc.M601194200. [DOI] [PubMed] [Google Scholar]
  • 62.Gopalan SM, Wilczynska KM, Konik BS, Bryan L, Kordula T. Astrocyte-specific expression of the alpha1-antichymotrypsin and glial fibrillary acidic protein genes requires activator protein-1. J Biol Chem. 2006;4:1956–1963. doi: 10.1074/jbc.M510935200. [DOI] [PubMed] [Google Scholar]
  • 63.Rousseau A, Nutt CL, Betensky RA, Iafrate AJ, Han M, Ligon KL, Rowitch DH, Louis DN. Expression of oligodendroglial and astrocytic lineage markers in diffuse gliomas: use of YKL-40, ApoE, ASCL1, and NKX2-2. J Neuropathol Exp Neurol. 2006;12:1149–1156. doi: 10.1097/01.jnen.0000248543.90304.2b. [DOI] [PubMed] [Google Scholar]
  • 64.Nutt CL, Betensky RA, Brower MA, Batchelor TT, Louis DN, Stemmer-Rachamimov AO. YKL-40 is a differential diagnostic marker for histologic subtypes of high-grade gliomas. Clin Cancer Res. 2005;6:2258–2264. doi: 10.1158/1078-0432.CCR-04-1601. [DOI] [PubMed] [Google Scholar]
  • 65.Mishima K, Kato Y, Kaneko MK, Nishikawa R, Hirose T, Matsutani M. Increased expression of podoplanin in malignant astrocytic tumors as a novel molecular marker of malignant progression. Acta Neuropathol (Berl) 2006;5:483–488. doi: 10.1007/s00401-006-0063-y. [DOI] [PubMed] [Google Scholar]
  • 66.Bronger H, Konig J, Kopplow K, Steiner HH, Ahmadi R, Herold-Mende C, Keppler D, Nies AT. ABCC drug efflux pumps and organic anion uptake transporters in human gliomas and the blood-tumor barrier. Cancer Res. 2005;24:11419–11428. doi: 10.1158/0008-5472.CAN-05-1271. [DOI] [PubMed] [Google Scholar]
  • 67.Zheng PS, Wen J, Ang LC, Sheng W, Viloria-Petit A, Wang Y, Wu Y, Kerbel RS, Yang BB. Versican/PG-M G3 domain promotes tumor growth and angiogenesis. FASEB J. 2004;6:754–756. doi: 10.1096/fj.03-0545fje. [DOI] [PubMed] [Google Scholar]
  • 68.Johansson FK, Goransson H, Westermark B. Expression analysis of genes involved in brain tumor progression driven by retroviral insertional mutagenesis in mice. Oncogene. 2005;24:3896–3905. doi: 10.1038/sj.onc.1208553. [DOI] [PubMed] [Google Scholar]
  • 69.Chekenya M, Rooprai HK, Davies D, Levine JM, Butt AM, Pilkington GJ. The NG2 chondroitin sulfate proteoglycan: role in malignant progression of human brain tumours. Int J Dev Neurosci. 1999;5–6:421–435. doi: 10.1016/s0736-5748(99)00019-2. [DOI] [PubMed] [Google Scholar]
  • 70.Bao S, Ouyang G, Bai X, Huang Z, Ma C, Liu M, Shao R, Anderson RM, Rich JN, Wang XF. Periostin potently promotes metastatic growth of colon cancer by augmenting cell survival via the Akt/PKB pathway. Cancer Cell. 2004;4:329–339. doi: 10.1016/s1535-6108(04)00081-9. [DOI] [PubMed] [Google Scholar]
  • 71.Siriwardena BS, Kudo Y, Ogawa I, Kitagawa M, Kitajima S, Hatano H, Tilakaratne WM, Miyauchi M, Takata T. Periostin is frequently overexpressed and enhances invasion and angiogenesis in oral cancer. Br J Cancer. 2006;10:1396–1403. doi: 10.1038/sj.bjc.6603431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Huang H, Hara A, Homma T, Yonekawa Y, Ohgaki H. Altered expression of immune defense genes in pilocytic astrocytomas. J Neuropathol Exp Neurol. 2005;10:891–901. doi: 10.1097/01.jnen.0000183345.19447.8e. [DOI] [PubMed] [Google Scholar]
  • 73.Hunter S, Young A, Olson J, Brat DJ, Bowers G, Wilcox JN, Jaye D, Mendrinos S, Neish A. Differential expression between pilocytic and anaplastic astrocytomas: identification of apolipoprotein D as a marker for low-grade, non-infiltrating primary CNS neoplasms. J Neuropathol Exp Neurol. 2002;3:275–281. doi: 10.1093/jnen/61.3.275. [DOI] [PubMed] [Google Scholar]
  • 74.Lovig T, Andersen SN, Thorstensen L, Diep CB, Meling GI, Lothe RA, Rognum TO. Strong HLA-DR expression in microsatellite stable carcinomas of the large bowel is associated with good prognosis. Br J Cancer. 2002;7:756–762. doi: 10.1038/sj.bjc.6600507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Tamiolakis D, Kotini A, Venizelos J, Jivannakis T, Simopoulos C, Papadopoulos N. Prognostic significance of HLA-DR antigen in serous ovarian tumors. Clin Exp Med. 2003;2:113–118. doi: 10.1007/s10238-003-0013-0. [DOI] [PubMed] [Google Scholar]
  • 76.Soni D, King JA, Kaye AH, Hovens CM. Genetics of glioblastoma multiforme: mitogenic signaling and cell cycle pathways converge. J Clin Neurosci. 2005;1:1–5. doi: 10.1016/j.jocn.2004.04.001. [DOI] [PubMed] [Google Scholar]
  • 77.Iafrate AJ, Feuk L, Rivera MN, Listewnik ML, Donahoe PK, Qi Y, Scherer SW, Lee C. Detection of large-scale variation in the human genome. Nat Genet. 2004;9:949–951. doi: 10.1038/ng1416. [DOI] [PubMed] [Google Scholar]
  • 78.Locke DP, Sharp AJ, McCarroll SA, McGrath SD, Newman TL, Cheng Z, Schwartz S, Albertson DG, Pinkel D, Altshuler DM, et al. Linkage disequilibrium and heritability of copy-number polymorphisms within duplicated regions of the human genome. Am J Hum Genet. 2006;2:275–290. doi: 10.1086/505653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Sebat J, Lakshmi B, Troge J, Alexander J, Young J, Lundin P, Maner S, Massa H, Walker M, Chi M, et al. Large-scale copy number polymorphism in the human genome. Science. 2004;5683:525–528. doi: 10.1126/science.1098918. [DOI] [PubMed] [Google Scholar]
  • 80.Fiegler H, Redon R, Andrews D, Scott C, Andrews R, Carder C, Clark R, Dovey O, Ellis P, Feuk L, et al. Accurate and reliable high-throughput detection of copy number variation in the human genome. Genome Res. 2006;12:1566–1574. doi: 10.1101/gr.5630906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Khaja R, Zhang J, MacDonald JR, He Y, Joseph-George AM, Wei J, Rafiq MA, Qian C, Shago M, Pantano L, et al. Genome assembly comparison identifies structural variants in the human genome. Nat Genet. 2006;12:1413–1418. doi: 10.1038/ng1921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Redon R, Ishikawa S, Fitch KR, Feuk L, Perry GH, Andrews TD, Fiegler H, Shapero MH, Carson AR, Chen W, et al. Global variation in copy number in the human genome. Nature. 2006;7118:444–454. doi: 10.1038/nature05329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Conrad DF, Andrews TD, Carter NP, Hurles ME, Pritchard JK. A high-resolution survey of deletion polymorphism in the human genome. Nat Genet. 2006;1:75–81. doi: 10.1038/ng1697. [DOI] [PubMed] [Google Scholar]
  • 84.Sharp AJ, Locke DP, McGrath SD, Cheng Z, Bailey JA, Vallente RU, Pertz LM, Clark RA, Schwartz S, Segraves R, et al. Segmental duplications and copy-number variation in the human genome. Am J Hum Genet. 2005;1:78–88. doi: 10.1086/431652. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Braude I, Vukovic B, Prasad M, Marrano P, Turley S, Barber D, Zielenska M, Squire JA. Large scale copy number variation (CNV) at 14q12 is associated with the presence of genomic abnormalities in neoplasia. BMC Genomics. 2006;7:138. doi: 10.1186/1471-2164-7-138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Jadayel DM, Osborne LR, Coignet LJ, Zani VJ, Tsui LC, Scherer SW, Dyer MJ. The BCL7 gene family: deletion of BCL7B in Williams syndrome. Gene. 1998;1–2:35–44. doi: 10.1016/s0378-1119(98)00514-9. [DOI] [PubMed] [Google Scholar]
  • 87.van Doorn R, Zoutman WH, Dijkman R, de Menezes RX, Commandeur S, Mulder AA, van der Velden PA, Vermeer MH, Willemze R, Yan PS, et al. Epigenetic profiling of cutaneous T-cell lymphoma: promoter hypermethylation of multiple tumor suppressor genes including BCL7a, PTPRG, and p73. J Clin Oncol. 2005;17:886–3896. doi: 10.1200/JCO.2005.11.353. [DOI] [PubMed] [Google Scholar]
  • 88.Zani VJ, Asou N, Jadayel D, Heward JM, Shipley J, Nacheva E, Takasuki K, Catovsky D, Dyer MJ. Molecular cloning of complex chromosomal translocation t(8;14;12)(q24.1;q32.3;q24.1) in a Burkitt lymphoma cell line defines a new gene (BCL7A) with homology to caldesmon. Blood. 1996;8:3124–3134. [PubMed] [Google Scholar]
  • 89.Tracey L, Villuendas R, Dotor AM, Spiteri I, Ortiz P, Garcia JF, Peralto JL, Lawler M, Piris MA. Mycosis fungoides shows concurrent deregulation of multiple genes involved in the TNF signaling pathway: an expression profile study. Blood. 2003;3:1042–1050. doi: 10.1182/blood-2002-11-3574. [DOI] [PubMed] [Google Scholar]
  • 90.Alizadeh AA, Eisen MB, Davis RE, Ma C, Lossos IS, Rosenwald A, Boldrick JC, Sabet H, Tran T, Yu X, et al. Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling. Nature. 2000;6769:503–511. doi: 10.1038/35000501. [DOI] [PubMed] [Google Scholar]
  • 91.Martinez-Delgado B, Melendez B, Cuadros M, Alvarez J, Castrillo JM, Ruiz De La Parte A, Mollejo M, Bellas C, Diaz R, Lombardia L, et al. Expression profiling of T-cell lymphomas differentiates peripheral and lymphoblastic lymphomas and defines survival related genes. Clin Cancer Res. 2004;15:4971–4982. doi: 10.1158/1078-0432.CCR-04-0269. [DOI] [PubMed] [Google Scholar]
  • 92.Adesina AM, Nguyen Y, Guanaratne P, Pulliam J, Lopez-Terrada D, Margolin J, Finegold M. FOXG1 is overexpressed in hepatoblastoma. Hum Pathol. 2007;3:400–409. doi: 10.1016/j.humpath.2006.09.003. [DOI] [PubMed] [Google Scholar]
  • 93.Rodriguez C, Huang LJ, Son JK, McKee A, Xiao Z, Lodish HF. Functional cloning of the proto-oncogene brain factor-1 (BF-1) as a Smadbinding antagonist of transforming growth factor-beta signaling. J Biol Chem. 2001;32:30224–30230. doi: 10.1074/jbc.M102759200. [DOI] [PubMed] [Google Scholar]
  • 94.Yao J, Lai E, Stifani S. The winged-helix protein brain factor 1 interacts with groucho and hes proteins to repress transcription. Mol Cell Biol. 2001;6:1962–1972. doi: 10.1128/MCB.21.6.1962-1972.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Seoane J, Le HV, Shen L, Anderson SA, Massague J. Integration of Smad and forkhead pathways in the control of neuroepithelial and glioblastoma cell proliferation. Cell. 2004;2:211–223. doi: 10.1016/s0092-8674(04)00298-3. [DOI] [PubMed] [Google Scholar]
  • 96.Shang C, Fu WN, Guo Y, Huang DF, Sun KL. Study of the SH3-domain GRB2-like 2 gene expression in laryngeal carcinoma. Chin Med J (Engl) 2007;5:385–388. [PubMed] [Google Scholar]
  • 97.Sun XJ, Sun KL, Zheng ZH, Fu WN, Hao DM, Xu HM, Li XM. Gene expression patterns in gastric cancer. Zhonghua Yi Xue Yi Chuan Xue Za Zhi. 2006;2:142–146. [PubMed] [Google Scholar]
  • 98.Wise DO, Krahe R, Oakley BR. The gamma-tubulin gene family in humans. Genomics. 2000;2:164–170. doi: 10.1006/geno.2000.6247. [DOI] [PubMed] [Google Scholar]
  • 99.Geurts JJ, Wolswijk G, Bo L, vanderValk P, Polman CH, Troost D, Aronica E. Altered expression patterns of group I and II metabotropic glutamate receptors in multiple sclerosis. Brain. 2003;126(Pt 8):1755–1766. doi: 10.1093/brain/awg179. [DOI] [PubMed] [Google Scholar]
  • 100.Laurin N, Misener VL, Crosbie J, Ickowicz A, Pathare T, Roberts W, Malone M, Tannock R, Schachar R, Kennedy JL, et al. Association of the calcyon gene (DRD1IP) with attention deficit/hyperactivity disorder. Mol Psychiatry. 2005;12:1117–1125. doi: 10.1038/sj.mp.4001737. [DOI] [PubMed] [Google Scholar]
  • 101.Gonzalez-Gomez P, Bello MJ, Lomas J, Arjona D, Alonso ME, Aminoso C, De Campos JM, Vaquero J, Sarasa JL, Casartelli C, et al. Epigenetic changes in pilocytic astrocytomas and medulloblastomas. Int J Mol Med. 2003;5:655–660. [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary Figures and Tables
neo1008_0757SD1.pdf (755.7KB, pdf)

Articles from Neoplasia (New York, N.Y.) are provided here courtesy of Neoplasia Press

RESOURCES