Of the genetic changes in primary central nervous system lymphoma (PCNSL), little is known. To detect copy number alterations and differentially expressed genes in PCNSL, we analyzed a total of 12 PCNSL samples with high-resolution array-based comparative genomic hybridization and performed expression profiling in 7 of the 12 samples. The most frequent deletion found in 8 patients (66.7%) occurred in 9p21.3 containing CDKN2A. We compiled the top 96 genes (family-wise error rate, P < .05) showing the greatest differential expression between PCNSL and normal lymph node tissues. From these, we selected 8 candidate genes (NPFFR2, C4orf7, OSMR, EMCN, TPO, FNDC1, COL12A1, and MSC) in which expression changes were associated with copy number aberrations. All 8 genes showed both down-regulation in expression microarray and deletion in array-based comparative genomic hybridization analyses. These genes participate in cell signaling or cell adhesion. In addition, low mRNA expression of C4orf7 was significantly associated with poor survival (P = .0425). Using gene set enrichment analysis, we identified several signal transduction pathways, such as Janus kinase-signal transducers and activators of transcription pathway and adhesion-related pathways, which may be involved in pathogenesis of PCNSL. In conclusion, this study identified novel tumor suppressor genes that may serve as therapeutic targets of PCNSL.

Primary diffuse large B-cell lymphoma (DLBCL) of the central nervous system (CNS) is a distinct form of extranodal lymphoma, which is recognized as a DLBCL localized in the eye or brain parenchyma. This diagnosis excludes meningeal and intravascular DLBCL. The tumor cells are mainly large, with numerous apoptotic cells or widespread necrosis, which commonly hinders diagnosis in small biopsies.1,2  Because the brain is segregated from systemic circulation by the blood-brain barrier, the phenotype and cell of origin of primary central nervous system lymphoma (PCNSL) have been topics of investigation.

PCNSL has been reported to show an activated B-cell-like or germinal center B-cell-like subtype of DLBCL3-5 ; however, recent studies of genome-wide gene expression in PCNSL compared with non-CNS DLBCL suggest that PCNSL has specific signatures to be distinguished from non-CNS DLBCL and greater molecular heterogeneity.6,7  Tumor cells of PCNSL carry a heavy load of ongoing somatic hypermutation with biased use of VH genes in immunoglobulin gene analysis, suggesting an antigen-dependent proliferation.8  A recent study of genome-wide gene expression in PCNSL compared with non-CNS DLBCL showed significant differential expression of multiple extracellular matrix- and adhesion-related pathways.6  These indicate that primary CNS DLBCL is derived from peripheral circulating B cells activated by antigenic stimulation and localized in the brain through interactions between various extracellular matrix and adhesion molecules.

Of the genetic changes that lead to PCNSL, very little is known and no characteristic genetic alterations have been defined thus far. Genomic alterations commonly seen in DLBCLs, regardless of the site of origin, include abnormalities of the 3q27 region involving the BCL6 gene in up to 30% of cases and translocation of the BCL2 gene t(14;18) in 20% to 30% of cases.9,10  A MYC rearrangement is observed in up to 10% of DLBCLs and in 3% of PCNSLs11,12 ; however, higher expression of MYC in PCNSL than nodal DLBCL suggests that a mechanism other than a MYC translocation may be responsible for the MYC overexpression in PCNSL.7,12  In PCNSL, inactivation of CDKN2A by either homozygous deletion or DNA methylation is commonly observed,13,14  but this is also common in non-CNS DLBCLs. A single study using comparative genomic hybridization reported frequent losses of 6q21–22, 12q, 18q, and chromosome 22q.15 

In this study, we applied high resolution array-based comparative genomic hybridization (aCGH) to a panel of 12 PCNSL samples from patients and 13 peripheral blood samples from healthy men to obtain a genomic profile of PCNSL. In addition, gene expression analysis was performed with 7 tumor samples and 7 normal lymph node samples to identify genes differentially expressed in PCNSL, and to identify pathways specifically affected in PCNSL, a pathway analysis of the PCNSL gene expression dataset was performed. The identified genes were validated using quantitative real-time reverse-transcribed polymerase chain reaction (RT-PCR). The copy number alteration data were combined with the dataset of genes differentially expressed to confirm whether those genetic alterations had an impact on the expression level of affected genomic regions. Finally, we investigated the prognostic significance of those candidate genes using 23 additional formalin-fixed paraffin-embedded (FFPE) tissue samples of primary CNS DLBCL.

Tissues

Fresh-frozen samples were obtained from 13 patients with primary CNS DLBCL using a protocol that the Samsung Medical Center Institutional Review Board approved, in accordance with the Declaration of Helsinki. Of the samples, 8 were obtained from resection and 5 were collected from stereotactic needle biopsies. All samples were confirmed by routine histologic evaluation. The quality of frozen samples was assessed by hematoxylin and eosin and CD20 immunohistochemical stains. All samples contained more than 80% of malignant B cells except for one, which was excluded from the study. A total of 12 patients were enrolled in the study, including 6 men and 6 women. Age at diagnosis ranged from 32 to 68 years (median, 54.5 years). All 12 samples were included in aCGH analysis. Of the 12 samples, only 8 for RNA study were used for further expression array analysis because of the quantity of tissue available after aCGH. For normal control samples, we used peripheral blood from 13 healthy men for aCGH and 8 fresh-frozen normal lymph nodes for expression array.

Extraction of genomic DNA and total RNA

For aCGH, DNA was extracted from 12 fresh-frozen tumor samples using standard proteinase K/RNAse treatment and phenol-chloroform extraction. Normal DNA was obtained from the peripheral blood of 13 healthy male donors. For expression array, total RNA was prepared from 8 matched fresh tumor tissues and 8 fresh-frozen normal lymph nodes using trizol-chloroform extraction.

aCGH

aCGH was performed using the Agilent Human Genome CGH Microarray Kit 4 × 44K containing approximately 44 000 probes composed of 60-mer oligonucleotides designed by Agilent. Labeling and hybridization were performed by protocols provided by the manufacturer. Briefly, 1.5 μg of test DNA and reference DNAs were digested with AluI and Rsa I (Promega) and purified with the QIAprep Spin Miniprep kit (QIAGEN). Test sample DNA and reference DNA were labeled by random priming with either Cy3-dUTP or Cy5-dUTP using the Agilent Genomic DNA Labeling Kit PLUS. After the labeling reaction, the individually labeled samples and reference samples were combined and concentrated using Microcon YM-30 filters (Millipore). After probe denaturation and preannealing with Cot-1 DNA, hybridization was performed at 65°C with rotation for 40 hours. Four washing steps were completed with Agilent Oligo CGH washes: wash buffer 1 at room temperature for 5 minutes, wash buffer 2 at 37°C for 1 minute, an acetonitrile rinse at room temperature for 1 minute, and a 30-second wash at room temperature in Agilent's Stabilization and Drying Solution. All slides were scanned on an Agilent DNA microarray scanner. Data were obtained using Agilent Feature Extraction Software Version 9, and analyzed with Agilent CGH Analytics Version 4.0 Software, using the statistical algorithms z-score and common aberration detection method 2 with sensitivity thresholds at 2.5 and 6.0, respectively, and a moving average window of 0.2 Mb.

Gene expression microarray

Expression profiling analysis was performed using Agilent Oligo Microarray Kit 4 × 44K according to Agilent One-Color Microarray-based Gene Expression Analysis Protocol. Briefly, 0.5 μg of total RNA was amplified and labeled using Agilent Quick Amp Labeling Kit. After labeling, cRNA was purified with RNeasy Mini-spin columns (QIAGEN) and quantified using NanoDrop ND-1000 UV-VIS Spectrophotometer for cyanine 3 dye concentration, RNA absorbance ratio (260 nm/280 nm), and cRNA concentration. Cyanine 3–labeled linearly amplified cRNA was mixed with 10× blocking agent, nuclease-free water, and 25× fragmentation buffer provided in the Agilent Gene Expression Hybridization Kit (Agilent Technologies), and incubated for 30 minutes at 60°C to fragment RNA. After adding 2× GEx hybridization buffer Hi-RPM, the amplified fragmented RNA was hybridized on a 4 × 44K Whole Human Genome Oligo Microarray Chip (Agilent Technologies) containing 44 000 cDNA clones using a SureHyb DNA Microarray Hybridization Chamber in a DNA Microarray Hybridization Oven (Agilent Technologies) at 10 rpm, 65°C, for 17 hours. After hybridization, slides were washed in Gene Expression wash buffers 1 and 2 for 1 minute each and then scanned with an Agilent DNA microarray scanner (Agilent Microarray Scanner-G2565BA; Agilent Technologies). Feature Extraction Software Version 9 (Agilent Technologies), was used to extract and analyze the signals. To validate the amplification process, amplified and unamplified RNAs were labeled, hybridized, and compared.

Gene expression analysis

To identify genes differentially expressed between the tumors and normal lymph nodes, the family-wise error rate (FWER) for control of multiple-testing error was calculated using software R (Version 2.9.1; http://www.r-project.org) after normalization by the variance-stabilizing transformation method (VSN) proposed by Huber et al.16  Significantly different genes (FWER, P < .05) identified by expression array analysis were then imported into DAVID (the database for annotation, visualization and integrated discovery) bioinformatics resources17,18  to extract biologic features and meaning associated with large gene lists. To identify genomic alterations in the different genes expressed, the identified genes were combined with aCGH data obtained by Agilent CGH Analytics Version 4.0 Software, to correlate common aberrant regions from aCGH. In addition, to identify pathways specifically affected in PCNSL, a pathway analysis of the PCNSL gene expression dataset was performed using gene set enrichment analysis19,20  with KEGG/Biocarta pathway gene sets.

Quantitative real-time RT PCR

To validate expression data, cDNA was synthesized from 2 μg of total RNA from 6 fresh tumor samples and 5 normal reactive lymph node samples using Superscript III RNA Reverse Transcriptase (Invitrogen) according to the manufacturer's protocol. Real-time RT-PCR was performed using the ABI 7900 HT Fast Real-Time PCR System (Applied Biosystems). The NPFFR2, C4orf7, OSMR, EMCN, TPO, FNDC1, COL12A1, and MSC genes and a reference gene (GAPDH) were amplified and detected using dsDNA-specific CYBR Green dye (Applied Biosystems). PCR reactions were prepared in a final volume of 10 μL, with 5 μL of 2 × Power SYBR Green PCR Master Mix (Applied Biosystems), 0.5 μL of cDNA, and 0.2 μL each of the 10 pmol/μL forward and reverse primers. Thermal cycling commenced with DNA polymerase activation at 95°C for 10 minutes and continued with 45 cycles of denaturation at 95°C for 15 seconds, followed by annealing and extension at 60°C for 1 minute. Each measurement was performed in duplicate. PCR product quality was monitored using post-PCR melt-curve analysis. Threshold cycle (Ct), the fractional cycle number at which the amount of amplified target reached a fixed threshold, was determined. The concentration ratios of each of the 8 genes to GAPDH were calculated using 2−ΔCtCt = ΔCtested genes − ΔCreference gene).

Primers were as follows: COL12A1: forward (5′-AGTGCATCAACGGGATACAG), reverse (5′-ACATGCAATTCCCTCTCTGC); C4orf7: forward (5′-CATATCCATTTCGCCCACTT), reverse (5′-TTTCGCTAGG AAGGGGAGTT); FNDC1: forward (5′-TCCCCAATGACCTGAAGAAG), reverse (5′-GCTTTGTCCCAGTCCACAAT); NPFFR2: forward (5′-GCCCCTGTGG ACTCTAATGA), reverse (5′-TGGGATTGAC ACTGCTGTTG); MSC: forward (5′-CGCTTAAATCGGACTGGAAC), reverse (5′-GGGAAGGGTCAAGGAGAATC); EMCN: forward (5′-TCACACCAACAACTGGAACAA), reverse (5′-TCAGTGGTTGTGGCTTTCAA); OSMR: forward (5′-TCTGAGCTCCCTTTGGAATG), reverse (5′-GGAACTCCAGTTGCTCCAGA); TPO: forward (5′-GAGGGATTACATCCCCAGGA), reverse (5′-GAACACGTTGGACACAGTGG); and GAPDH: forward (5′-GCACCGTCAAGGCTGAGAA), reverse (5′-AGCATCGCCCCACTTGATT).

Statistical analysis

Differences in mRNA expression levels between tumors and normal samples were calculated using the Mann-Whitney U test. Overall survival was determined using the Kaplan-Meier method, and survival curves were compared using the log-rank test. Survival was measured from the date of surgery, and all patients were followed through April 2010. All tests were 2-sided, and statistical significance was set at a threshold of P less than .05. Statistical analyses were performed using SPSS Version 18 (SPSS Inc).

Copy number aberrations in PCNSL

Data have been deposited in Gene Expression Omnibus (accession number GSE25298). All of the 12 PCNSLs showed frequent genomic aberrations, whereas all normal samples from healthy men showed few genomic aberrations (Figure 1). Regions of recurrent gain (≥ 50%) were located on chromosomes 1p36.33, 3q21.3, 3q22.1, 3q29, 7p22.3, 7q11.23, 7q22.1, 7q36.1, 8q24.3, 9q34.11, 9q34.3, 10q26.3, 11p15.5, 11q13.1, 11q23.3, 14q32.33, 15q24.1, 16p13.3, 16p11.2, 16q24.3, 17p13.3, 17q25.3, 19p13.3, 19q13.2, 19q13.31–33, 19q13.41–43, 20q13.33, 21q22.3, and 22q13.33. Regions of most frequent gain were 17p13.3 containing TMEM93 and P2RX5, and 22q13.33 containing IL17REL and MLC1 in 9 patients (75%). Regions of recurrent loss (≥ 50%) were located on chromosomes 6p21.32, 6q14.1–3, 6q15, 6q16.1–3, 6q21, 6q22.1–2, 6q22.31–33, 6q23.1–3, 6q24.1–2, 6q26, 8p22, 8p21.3, 9p21.3, 14q32.33, p11.31, and 22q11.23. Deletions were most frequently observed in 9p21.3 (8 patients; 66.7%), which contains CDKN2A.

Figure 1

Genome-wide frequency plots of copy number alterations identified in 12 PCNSLs and 13 normal peripheral blood samples.

Figure 1

Genome-wide frequency plots of copy number alterations identified in 12 PCNSLs and 13 normal peripheral blood samples.

Close modal

Expression signatures of PCNSL compared with normal lymph node

Data have been deposited in Gene Expression Omnibus (accession number GSE25297). From the raw data from 8 tumors and 8 normal lymph nodes, we excluded one tumor and one normal sample that showed inadequate quality. After normalization using the VSN method, we detected 96 significant genes (FWER, P < .05) that were differentially expressed in the tumor group compared with the normal group. Clustering of these top 96 genes showed a distinct separation between the 2 groups (Figure 2). The genes included 27 up-regulated genes (Table 1) and 69 down-regulated genes (Table 2). Functional annotation analysis using DAVID bioinformatics resources produced 111 enriched biologic terms. Many of these terms are related to signaling, extracellular matrix (ECM) properties, and cellular adhesion. A functional annotation group, including OSMR, CCND3, TPO, and IL3RA, is associated with Janus kinase (JAK)-signal transducers and activators of transcription (STAT) signaling. Biologic terms that met the significance threshold of P less than .01 are summarized in Table 3.

Figure 2

Clustering of the top 96 genes (P < .05, FWER) differentially expressed between the tumor group and normal group shows a distinct separation between the groups.

Figure 2

Clustering of the top 96 genes (P < .05, FWER) differentially expressed between the tumor group and normal group shows a distinct separation between the groups.

Close modal
Table 1

Up-regulated genes in PCNSL

PCNSL versus normal LN (present study)
PCNSL versus non-CNS DLBCL
Probe IDGeneP (FWER)RankGene by Tun et al6 Gene by Tun et al6 Gene by Rubenstein et al7 
A_23_P2181 CYB5R2 .001 SPP1 RGS13 MYC 
A_24_P157370 IL 17RB .002 TF FCGR3A MINA 
A_24_P366315 KIF21A .003 DDR1 CNP EIF5 
A_32_P85405 Not found .006 NM_178012 FBP1 EIF4A 
A_23_P3371 RASGRF1 .006 10 SERPINA3 BC000525 CUL5 
A_32_P125808 Not found .008 14 S-100B BMP7 E2F5 
A_23_P97473 MRPL20 .009 17 C9orf58 NM_013332 BUB3 
A_24_P880000 Not found .011 20 BC020630 C1QB PIM1 
A_32_P23525 Not found .012 22 CRYAB IRF4 JAK1 
A_23_P325642 PDIA2 .015 23 NM_018584 DECR2 XBP1 
A_23_P77201 C15orf41 .015 26 AF034208  ATF6 
A_23_P3379 RASGRF1 .016 29 CXCL13  PDCD4 
A_23_P420610 FCHO2 .017 31 NM_152680  FAF1 
A_24_P392265 EXOC5 .023 38 TCL1A  CFLAR0 
A_23_P205007 IPO5 .025 42 DKK3  ITGAE 
A_24_P31165 GFAP .026 45 FOXG1B  CTSD 
A_23_P170761 PDLIM5 .026 46 UGT2B17   
A_24_P281923 DLAT .026 48 TACSTD1   
A_23_P329133 GOT2 .026 50 FEZ1   
A_24_P288754 PIGA .027 52 CHI3L1   
A_23_P68998 MIOX .030 61 CA2   
A_23_P216080 HOOK3 .031 67 MGST1   
A_32_P89691 SORD .033 73 DNAJC12   
A_23_P117411 FOXG1 .039 78 AK097976   
A_23_P106575 GOT2 .040 79 NM_018357   
A_23_P29153 RTDR1 .042 82 HBA2   
A_23_P91081 TACSTD1 .045 89 NM_024897   
PCNSL versus normal LN (present study)
PCNSL versus non-CNS DLBCL
Probe IDGeneP (FWER)RankGene by Tun et al6 Gene by Tun et al6 Gene by Rubenstein et al7 
A_23_P2181 CYB5R2 .001 SPP1 RGS13 MYC 
A_24_P157370 IL 17RB .002 TF FCGR3A MINA 
A_24_P366315 KIF21A .003 DDR1 CNP EIF5 
A_32_P85405 Not found .006 NM_178012 FBP1 EIF4A 
A_23_P3371 RASGRF1 .006 10 SERPINA3 BC000525 CUL5 
A_32_P125808 Not found .008 14 S-100B BMP7 E2F5 
A_23_P97473 MRPL20 .009 17 C9orf58 NM_013332 BUB3 
A_24_P880000 Not found .011 20 BC020630 C1QB PIM1 
A_32_P23525 Not found .012 22 CRYAB IRF4 JAK1 
A_23_P325642 PDIA2 .015 23 NM_018584 DECR2 XBP1 
A_23_P77201 C15orf41 .015 26 AF034208  ATF6 
A_23_P3379 RASGRF1 .016 29 CXCL13  PDCD4 
A_23_P420610 FCHO2 .017 31 NM_152680  FAF1 
A_24_P392265 EXOC5 .023 38 TCL1A  CFLAR0 
A_23_P205007 IPO5 .025 42 DKK3  ITGAE 
A_24_P31165 GFAP .026 45 FOXG1B  CTSD 
A_23_P170761 PDLIM5 .026 46 UGT2B17   
A_24_P281923 DLAT .026 48 TACSTD1   
A_23_P329133 GOT2 .026 50 FEZ1   
A_24_P288754 PIGA .027 52 CHI3L1   
A_23_P68998 MIOX .030 61 CA2   
A_23_P216080 HOOK3 .031 67 MGST1   
A_32_P89691 SORD .033 73 DNAJC12   
A_23_P117411 FOXG1 .039 78 AK097976   
A_23_P106575 GOT2 .040 79 NM_018357   
A_23_P29153 RTDR1 .042 82 HBA2   
A_23_P91081 TACSTD1 .045 89 NM_024897   

LN indicates lymph node.

Table 2

Down-regulated genes in PCNSL

PCNSL versus normal LN (present study)
PCNSL versus non-CNS DLBCL
Probe IDGeneP (FWER)RankProbe IDGeneP (FWER)RankGene by Tun et al6 Gene by Rubenstein et al7 
A_23_P140190 FAM30A .001 A_32_P167239 AFAP1L1 .029 56 CX3CR1 ZFP36 
A_24_P95723 KIAA0125 .001 A_23_P43283 GPR124 .030 57 AK091178 MLL 
A_24_P70183 MYH11 .002 A_32_P76137 Not found .030 58 D42043 CHES1 
A_24_P76868 Not found .002 A_24_P375761 STGC3 .030 59 SQLE EPHB3 
A_23_P206920 MYH11 .006 A_24_P110618 Not found .030 60 P2RY8 RPS6KA3 
A_23_P76992 PGF .008 11 A_23_P90710 DES .030 62 LXN DDR2 
A_24_P52697 H19 .008 12 A_23_P218858 ABI3BP .031 63 LPP ADAM12 
A_23_P123853 CCL19 .008 13 A_23_P310956 COL6A2 .031 64 S56205 MMP2 
A_24_P183128 PLAC8 .008 15 A_23_P382065 EMCN .031 65 Z24727 COL4A2 
A_32_P172320 CCND3 .009 16 A_24_P270044 ASAM .031 66 STEAP COL5A1 
A_32_P145968 Not found .011 18 A_24_P99963 CSNK1G2 .032 68 COL6A1 VCAM1 
A_23_P78980 B3GNT3 .011 19 A_23_P112470 CCL21 .032 69 COL12A1 CDH1 
A_23_P200741 DPT .012 21 A_24_P86968 CD3E .032 70 AK022110 ITGAX 
A_32_P444104 LOC284454 .015 24 A_23_P158096 COL27A1 .032 71 NR2F2  
A_23_P45821 Not found .015 25 A_24_P15062 ZNF490 .033 72 NNMT  
A_23_P12686 GFRA1 .015 27 A_24_P257224 TPO .033 74 AK091153  
A_23_P81219 PLAC8 .016 28 A_23_P150064 MMRN2 .035 75 BC040354  
A_23_P149281 EPHA2 .016 30 A_24_P361816 IGLV6–57 .038 76 VEGFC  
A_23_P345581 FOSL2 .018 32 A_23_P407840 FNDC1 .038 77 LAMA4  
A_23_P155900 NPFFR2 .018 33 A_24_P101642 LOC401847 .042 80 NM_015714  
A_23_P381261 ADCY4 .020 34 A_23_P215634 IGFBP3 .042 81 CAMKK2  
A_23_P96955 FAM167B .022 35 A_23_P43276 GPR124 .043 83 FBN1  
A_23_P362694 C4orf7 .022 36 A_32_P12454 Not found .043 84 AJ001381  
A_24_P788878 FAM148B .023 37 A_24_P324787 KANK2 .043 85 LUM  
A_23_P48936 SMAD3 .024 39 A_23_P52161 NUAK2 .043 86 LOXL1  
A_24_P940288 PGS1 .024 40 A_23_P208595 LDLR .043 87 COL1A2  
A_23_P215634 IGFBP3 .024 41 A_23_P48936 SMAD3 .043 88   
A_23_P48936 SMAD3 .025 43 A_23_P214168 COL12A1 .045 90   
A_24_P71280 GPR157 .026 44 A_24_P216456 MAP1LC3C .045 91   
A_24_P207907 R3HDM2 .026 47 A_23_P209799 MYO7B .046 92   
A_23_P156289 OSMR .026 49 A_23_P127627 DGKZ .047 93   
A_23_P407695 FAM151A .027 51 A_32_P210642 EGFL7 .047 94   
A_23_P1083 GJA4 .027 53 A_23_P141055 TGFB1I1 .047 95   
A_32_P217750 IL3RA .027 54 A_23_P256948 MSC .048 96   
A_23_P58588 SLIT3 .028 55       
PCNSL versus normal LN (present study)
PCNSL versus non-CNS DLBCL
Probe IDGeneP (FWER)RankProbe IDGeneP (FWER)RankGene by Tun et al6 Gene by Rubenstein et al7 
A_23_P140190 FAM30A .001 A_32_P167239 AFAP1L1 .029 56 CX3CR1 ZFP36 
A_24_P95723 KIAA0125 .001 A_23_P43283 GPR124 .030 57 AK091178 MLL 
A_24_P70183 MYH11 .002 A_32_P76137 Not found .030 58 D42043 CHES1 
A_24_P76868 Not found .002 A_24_P375761 STGC3 .030 59 SQLE EPHB3 
A_23_P206920 MYH11 .006 A_24_P110618 Not found .030 60 P2RY8 RPS6KA3 
A_23_P76992 PGF .008 11 A_23_P90710 DES .030 62 LXN DDR2 
A_24_P52697 H19 .008 12 A_23_P218858 ABI3BP .031 63 LPP ADAM12 
A_23_P123853 CCL19 .008 13 A_23_P310956 COL6A2 .031 64 S56205 MMP2 
A_24_P183128 PLAC8 .008 15 A_23_P382065 EMCN .031 65 Z24727 COL4A2 
A_32_P172320 CCND3 .009 16 A_24_P270044 ASAM .031 66 STEAP COL5A1 
A_32_P145968 Not found .011 18 A_24_P99963 CSNK1G2 .032 68 COL6A1 VCAM1 
A_23_P78980 B3GNT3 .011 19 A_23_P112470 CCL21 .032 69 COL12A1 CDH1 
A_23_P200741 DPT .012 21 A_24_P86968 CD3E .032 70 AK022110 ITGAX 
A_32_P444104 LOC284454 .015 24 A_23_P158096 COL27A1 .032 71 NR2F2  
A_23_P45821 Not found .015 25 A_24_P15062 ZNF490 .033 72 NNMT  
A_23_P12686 GFRA1 .015 27 A_24_P257224 TPO .033 74 AK091153  
A_23_P81219 PLAC8 .016 28 A_23_P150064 MMRN2 .035 75 BC040354  
A_23_P149281 EPHA2 .016 30 A_24_P361816 IGLV6–57 .038 76 VEGFC  
A_23_P345581 FOSL2 .018 32 A_23_P407840 FNDC1 .038 77 LAMA4  
A_23_P155900 NPFFR2 .018 33 A_24_P101642 LOC401847 .042 80 NM_015714  
A_23_P381261 ADCY4 .020 34 A_23_P215634 IGFBP3 .042 81 CAMKK2  
A_23_P96955 FAM167B .022 35 A_23_P43276 GPR124 .043 83 FBN1  
A_23_P362694 C4orf7 .022 36 A_32_P12454 Not found .043 84 AJ001381  
A_24_P788878 FAM148B .023 37 A_24_P324787 KANK2 .043 85 LUM  
A_23_P48936 SMAD3 .024 39 A_23_P52161 NUAK2 .043 86 LOXL1  
A_24_P940288 PGS1 .024 40 A_23_P208595 LDLR .043 87 COL1A2  
A_23_P215634 IGFBP3 .024 41 A_23_P48936 SMAD3 .043 88   
A_23_P48936 SMAD3 .025 43 A_23_P214168 COL12A1 .045 90   
A_24_P71280 GPR157 .026 44 A_24_P216456 MAP1LC3C .045 91   
A_24_P207907 R3HDM2 .026 47 A_23_P209799 MYO7B .046 92   
A_23_P156289 OSMR .026 49 A_23_P127627 DGKZ .047 93   
A_23_P407695 FAM151A .027 51 A_32_P210642 EGFL7 .047 94   
A_23_P1083 GJA4 .027 53 A_23_P141055 TGFB1I1 .047 95   
A_32_P217750 IL3RA .027 54 A_23_P256948 MSC .048 96   
A_23_P58588 SLIT3 .028 55       

LN indicates lymph node.

Table 3

Enriched functional annotation terms associated with top 96 genes

CategoryTerm%P*GenesBonferroni*
SP_PIR_KEYWORDS Signal 35.6 < .0001 OSMR, MMRN2, EMCN, C4orf7, DLAT, IL3RA, TACSTD1, SLIT3, ABI3BP, COL6A2, COL27A1, EGFL7, EPHA2, CD3E, DPT, PDIA2, CCL21, LDLR, COL12A1, ASAM, GFRA1, GPR124, CCL19, IGFBP3, PGF, TPO 0.0018 
UP_SEQ_FEATURE Signal peptide 32.9 < .0001 ABI3BP, MMRN2, EMCN, COL6A2, COL27A1, C4orf7, EPHA2, EGFL7, DLAT, IL3RA, TACSTD1, CD3E, DPT, PDIA2, LDLR, CCL21, GFRA1, COL12A1, CCL19, GPR124, PGF, SLIT3, IGFBP3, TPO 0.3724 
IPR008957 Fibronectin, type III-like fold 8.2 .0004 ABI3BP, OSMR, FNDC1, COL12A1, EPHA2, IL3RA 0.9106 
GO:0009987 Cellular process 69.9 .0018 EXOC5, MAP1LC3C, DLAT, ADCY4, MSC, FOSL2, DGKZ, SLIT3, COL6A2, RASGRF1, MIOX, PIGA, NUAK2, GPR157, CCL21, LDLR, COL12A1, GPR124, IGFBP3, GFAP, OSMR, EMCN, IL3RA, PGS1, SORD, B3GNT3, DES, KIF21A, HOOK3, GOT2, MYH11, CSNK1G2, COL27A1, EGFL7, EPHA2, FOXG1, NPFFR2, CYB5R2, CD3E, DPT, CCND3, PDIA2, TGFB1I1, GJA4, SMAD3, GFRA1, CCL19, ZNF490, PGF, TPO, MRPL20 0.9999 
GO:0048513 Organ development 16.4 .0028 CD3E, MYH11, EMCN, PDLIM5, SMAD3, GJA4, COL12A1, PGF, FOXG1, EGFL7, IGFBP3, SLIT3 1.0000 
IPR003961 Fibronectin, type III 6.9 .0037 ABI3BP, OSMR, FNDC1, COL12A1, EPHA2 1.0000 
GO:0048731 System development 19.2 .0041 MYH11, EMCN, PDLIM5, EGFL7, EPHA2, FOXG1, CD3E, GJA4, SMAD3, COL12A1, GFRA1, PGF, SLIT3, IGFBP3 1.0000 
IPR001881 EGF-like calcium-binding 5.5 .0055 LDLR, EGFL7, SLIT3, TPO 1.0000 
SM00060 FN3 6.9 .0059 ABI3BP, OSMR, FNDC1, COL12A1, EPHA2 0.9731 
SP_PIR_KEYWORDS Secreted 16.4 .0067 DPT, MMRN2, EMCN, COL6A2, CCL21, COL12A1, C4orf7, CCL19, PGF, EGFL7, IGFBP3, SLIT3 0.9992 
GO:0044421 Extracellular region part 12.3 .0077 DPT, MMRN2, COL6A2, CCL21, COL27A1, COL12A1, CCL19, EGFL7, SLIT3 0.9988 
GO:0006519 Amino acid and derivative Metabolic process 8.2 .0080 GOT2, CD3E, CCND3, IGFBP3, DLAT, TPO 1.0000 
SM00179 EGF_CA 5.5 .0085 LDLR, EGFL7, SLIT3, TPO 0.9944 
GO:0005581 Collagen 4.1 .0096 COL6A2, COL27A1, COL12A1 0.9998 
CategoryTerm%P*GenesBonferroni*
SP_PIR_KEYWORDS Signal 35.6 < .0001 OSMR, MMRN2, EMCN, C4orf7, DLAT, IL3RA, TACSTD1, SLIT3, ABI3BP, COL6A2, COL27A1, EGFL7, EPHA2, CD3E, DPT, PDIA2, CCL21, LDLR, COL12A1, ASAM, GFRA1, GPR124, CCL19, IGFBP3, PGF, TPO 0.0018 
UP_SEQ_FEATURE Signal peptide 32.9 < .0001 ABI3BP, MMRN2, EMCN, COL6A2, COL27A1, C4orf7, EPHA2, EGFL7, DLAT, IL3RA, TACSTD1, CD3E, DPT, PDIA2, LDLR, CCL21, GFRA1, COL12A1, CCL19, GPR124, PGF, SLIT3, IGFBP3, TPO 0.3724 
IPR008957 Fibronectin, type III-like fold 8.2 .0004 ABI3BP, OSMR, FNDC1, COL12A1, EPHA2, IL3RA 0.9106 
GO:0009987 Cellular process 69.9 .0018 EXOC5, MAP1LC3C, DLAT, ADCY4, MSC, FOSL2, DGKZ, SLIT3, COL6A2, RASGRF1, MIOX, PIGA, NUAK2, GPR157, CCL21, LDLR, COL12A1, GPR124, IGFBP3, GFAP, OSMR, EMCN, IL3RA, PGS1, SORD, B3GNT3, DES, KIF21A, HOOK3, GOT2, MYH11, CSNK1G2, COL27A1, EGFL7, EPHA2, FOXG1, NPFFR2, CYB5R2, CD3E, DPT, CCND3, PDIA2, TGFB1I1, GJA4, SMAD3, GFRA1, CCL19, ZNF490, PGF, TPO, MRPL20 0.9999 
GO:0048513 Organ development 16.4 .0028 CD3E, MYH11, EMCN, PDLIM5, SMAD3, GJA4, COL12A1, PGF, FOXG1, EGFL7, IGFBP3, SLIT3 1.0000 
IPR003961 Fibronectin, type III 6.9 .0037 ABI3BP, OSMR, FNDC1, COL12A1, EPHA2 1.0000 
GO:0048731 System development 19.2 .0041 MYH11, EMCN, PDLIM5, EGFL7, EPHA2, FOXG1, CD3E, GJA4, SMAD3, COL12A1, GFRA1, PGF, SLIT3, IGFBP3 1.0000 
IPR001881 EGF-like calcium-binding 5.5 .0055 LDLR, EGFL7, SLIT3, TPO 1.0000 
SM00060 FN3 6.9 .0059 ABI3BP, OSMR, FNDC1, COL12A1, EPHA2 0.9731 
SP_PIR_KEYWORDS Secreted 16.4 .0067 DPT, MMRN2, EMCN, COL6A2, CCL21, COL12A1, C4orf7, CCL19, PGF, EGFL7, IGFBP3, SLIT3 0.9992 
GO:0044421 Extracellular region part 12.3 .0077 DPT, MMRN2, COL6A2, CCL21, COL27A1, COL12A1, CCL19, EGFL7, SLIT3 0.9988 
GO:0006519 Amino acid and derivative Metabolic process 8.2 .0080 GOT2, CD3E, CCND3, IGFBP3, DLAT, TPO 1.0000 
SM00179 EGF_CA 5.5 .0085 LDLR, EGFL7, SLIT3, TPO 0.9944 
GO:0005581 Collagen 4.1 .0096 COL6A2, COL27A1, COL12A1 0.9998 
*

P value and Bonferroni were provided by DAVID bioinformatics resources.

Pathways specific to PCNSL

The pathway activities that most clearly distinguished PCNSL from normal lymph nodes (false discovery rate [FDR] Q value < 0.05) mediate functions, such as cytokine-receptor interaction, cell adhesion and migration, apoptosis, and signal transduction via Bad, Notch, JAK-STAT, and NF-κB pathways (Table 4). Expression signature analysis revealed that some component genes in the JAK-STAT pathway, including OSMR, CCND3, and TPO, were down-regulated in PCNSL. Other genes in the JAK-STAT path-way, such as STAT3, JAK1, AKT3, IL22RA1, and IL12A, were up-regulated in PCNSL, whereas SOCS1 and SOCS3, which inhibit JAK-STAT signaling, were down-regulated (Figure 3A). Gene expression changes that influence cell adhesion clearly distinguished the normal and tumor groups (Figure 3B).

Table 4

Selection of pathways differentially expressed in PCNSL tissues compared with normal lymph node tissues

Pathway nameNominal PFDR Q
Cytokine cytokine receptor interaction < .001 <0.001 
Cell adhesion molecules < .001 0.002 
ECM receptor interaction < .001 0.014 
Leukocyte transendothelial migration < .001 0.016 
Focal adhesion < .001 0.017 
Apoptosis .002 0.027 
Signal transduction pathway   
    Bad pathway < .001 0.008 
    Notch signaling pathway < .001 0.010 
    JAK-STAT signaling pathway < .001 0.015 
    NF-κB pathway .007 0.026 
Pathway nameNominal PFDR Q
Cytokine cytokine receptor interaction < .001 <0.001 
Cell adhesion molecules < .001 0.002 
ECM receptor interaction < .001 0.014 
Leukocyte transendothelial migration < .001 0.016 
Focal adhesion < .001 0.017 
Apoptosis .002 0.027 
Signal transduction pathway   
    Bad pathway < .001 0.008 
    Notch signaling pathway < .001 0.010 
    JAK-STAT signaling pathway < .001 0.015 
    NF-κB pathway .007 0.026 
Figure 3

Representative molecular pathways differentially expressed in PCNSL compared with normal lymph node (LN). The genes involved in JAK-STAT (A) and cell adhesion (B) pathways were clustered according to phenotype (ie, as normal LN or PCNSL).

Figure 3

Representative molecular pathways differentially expressed in PCNSL compared with normal lymph node (LN). The genes involved in JAK-STAT (A) and cell adhesion (B) pathways were clustered according to phenotype (ie, as normal LN or PCNSL).

Close modal

Gene expression changes associated with copy number aberrations

The regions that contained significant common aberrations in copy number were correlated to a dataset of the top 96 differentially expressed genes (FWER, P < .05) obtained from expression array. Eight genes (NPFFR2, C4orf7, OSMR, EMCN, TPO, FNDC1, COL12A1, and MSC) were identified in this correlational analysis. All 8 genes showed both down-regulation in expression microarray and deletion in aCGH in 2 or more patients (> 15%). The same 8 genes showed neither deletion nor amplification in all 13 normal samples from healthy men. The FNDC1 and COL12A1 genes most frequently showed deletions, which were identified in 5 patients (41.7%). The data for each patient are summarized in Table 5.

Table 5

Deletion (Del) status in individual patients of 8 genes showing down-regulation by expression array

Case no.NPFFR2C4orf7OSMREMCNTPOFNDC1COL12A1MSC
Del Del Del Del Del — Del Del 
— — — — — Del — — 
Del Del — — — — — — 
— — — — Del — — — 
— — — — — — Del — 
Del Del Del Del — Del Del Del 
— — — — — — — — 
— — — — — — — — 
10 — — — — — — — — 
11 — — Del — — Del Del — 
12 — — — — — Del Del — 
13 — — — — — Del — — 
Normal (n = 13) — — — — — — — — 
Case no.NPFFR2C4orf7OSMREMCNTPOFNDC1COL12A1MSC
Del Del Del Del Del — Del Del 
— — — — — Del — — 
Del Del — — — — — — 
— — — — Del — — — 
— — — — — — Del — 
Del Del Del Del — Del Del Del 
— — — — — — — — 
— — — — — — — — 
10 — — — — — — — — 
11 — — Del — — Del Del — 
12 — — — — — Del Del — 
13 — — — — — Del — — 
Normal (n = 13) — — — — — — — — 

— indicates no Del or amplification.

Quantitative real-time RT-PCR for NPFFR2, C4orf7, OSMR, EMCN, TPO, FNDC1, COL12A1, and MSC

Eleven fresh-frozen tissue samples (6 tumors and 5 normal lymph nodes) were assayed for RNA expression levels, and the Ct was calculated for all 8 genes. The mRNA expression levels for each of the 8 genes were normalized to the reference gene (GAPDH). The concentration ratios of each of the 8 genes over GAPDH were calculated.

All 8 genes, except for TPO, showed significantly lower mRNA expression in the PCNSL group than in the normal lymph node group (Mann-Whitney U test, P < .05; Figure 4). The TPO mRNA expression level in the tumor group was not significantly lower than that in the normal group (mean concentration ratio = 0.001 [tumor] and 0.093 [normal], P = .1). The C4orf7 showed the most striking difference (mean concentration ratio = 0.007 [tumor] and 12.433 [normal], P = .006).

Figure 4

Significantly lower mRNA expression was detected for NPFFR2, C4orf7, OSMR, EMCN, FNDC1, COL12A1, and MSC in the PCNSL group than in the normal lymph node group (P < .05, Mann-Whitney U test) by quantitative real-time RT-PCR. TPO expression did not differ statistically between groups.

Figure 4

Significantly lower mRNA expression was detected for NPFFR2, C4orf7, OSMR, EMCN, FNDC1, COL12A1, and MSC in the PCNSL group than in the normal lymph node group (P < .05, Mann-Whitney U test) by quantitative real-time RT-PCR. TPO expression did not differ statistically between groups.

Close modal

Prognostic significance of NPFFR2, C4orf7, OSMR, EMCN, TPO, FNDC1, COL12A1, and MSC

To estimate the prognostic significance of the 8 differentially expressed genes, we investigated an independent set of 23 FFPE tissue samples from primary CNS DLBCL patients. All patients received chemotherapy regimens based on high-dose methotrexate after diagnosis. Among them, 22 patients received additional whole-brain radiation therapy. From these 23 FFPE samples, we extracted total RNA and synthesized the corresponding cDNA. Quantitative real-time RT-PCR was performed as described in “Methods,” and concentration ratios were calculated for each gene. To identify a cut-off point for the mRNA concentration ratio that is most meaningful with respect to prognosis, we tested various levels of expression using the Kaplan-Meier method with log-rank test. Among the 8 genes, low mRNA expression of C4orf7, when the cut-off point was set as less than or equal to 0.5, was significantly associated with poor overall survival (P = .0425, Figure 5). The median and mean values of concentration ratio of C4orf7 in 23 FFPE tumor samples were 0.58 and 0.71, respectively (range, 0.11-2.49). Significant associations with survival were not found for the 7 other genes (P > .05).

Figure 5

Low mRNA expression of C4orf7 was associated with poor overall survival in 23 patients with PCNSL.

Figure 5

Low mRNA expression of C4orf7 was associated with poor overall survival in 23 patients with PCNSL.

Close modal

In this study, we observed 96 genes showing the greatest difference in expression between PCNSL and normal lymphoid tissue. From these, we selected 8 candidate genes based on gene expression changes associated with copy number aberrations. Quantitative real-time RT-PCR confirmed down-regulation of all 8 genes in tumor tissue compared with normal lymphoid tissue. FNDC1, EMCN, and COL12A1 are associated with ECM and cell adhesion. Our results suggest that ECM and cell adhesion contribute to the pathogenesis of PCNSL. This finding converges with prior studies that have found that the gene set that is most significantly affected in PCNSL encodes components of the ECM-receptor pathway.6  These results support the model that an interaction between the CNS microenvironment and lymphoma cells strongly influences PCNSL development.

The other genes (ie, OSMR, C4orf7, NPFFR2, TPO, and MSC) mediate cell signaling. MSC, also called activated B-cell factor-1, is a transcriptional repressor that inhibits the transactivational capacity of TCF3/E47 and may participate in antigen-dependent B-cell differentiation. MSC is frequently silenced by promoter methylation in follicular lymphoma, DLBCL, and Burkitt lymphoma.21 NPFFR2 is a receptor protein for NPAF and NPFF neuropeptides and acts through association with G proteins that activate a phosphatidylinositol-calcium second messenger system. C4orf7 is expressed by follicular dendritic cells and activates leukocytes during an ongoing immune response. These leukocytes may then bind to the surface of B-lymphoma cells, but not T-lymphoma cells, which is consistent with a function for C4orf7 as a secreted B cell-specific mediator. In this study, low expression of C4orf7 mRNA was significantly associated with poor survival in 23 patients with primary CNS DLBCL. C4orf7 has been previously identified as playing a role in the lymphatic metastasis of human breast cancer.22  However, the specific mechanism for the prognostic effect of C4orf7 in PCNSL remains unclear, and further studies with a larger sample are needed.

OSMR is activated by oncostatin M (OSM), which is a member of the IL6 family of cytokines. OSM inhibits the growth of many tumor cell types, such as neuroblastoma, fibrosarcoma, and ovarian, lung, stomach, and breast carcinoma cell lines.23  Thus, OSMR, as the OSM receptor, may act as a tumor suppressor. In the present study, we found that OSMR was significantly down-regulated in PCNSL with deletion, which is consistent with the idea of a tumor suppressor role for OSMR in the pathogenesis of PCNSL. On the other hand, binding of OSM to OSMR triggers the activation of multiple members of the JAK family,24  which in turn leads to the activation of transcription factors known as STATs.25  Moreover, OSMR participates in STAT3 (and possibly STAT1 and STAT5) activation and mediates OSM-specific signaling events. Aberrant STAT3 and STAT5 expression in lymphoma may promote tumor progression.26  In this study, STAT1 was generally down-regulated in PCNSL, which correlates with OSMR down-regulation. STAT3, however, was up-regulated in PCNSL. Thus, factors other than OSMR may influence STAT3 expression in this type of tumor. For example, we also observed down-regulation of SOCS1 and SOCS3 expression. SOCS is known as inhibitor of JAK-STAT pathway, and a study has explored the negative relationship between SOCS3 and the STAT3 pathway.27 

In the aCGH analysis, we identified 9p21.3 containing CDKN2A as the region most frequently affected by deletion. CDKN2A is frequently mutated or deleted in a wide variety of tumors, which is evidence of its role as a tumor suppressor gene. Sulong et al28  observed CDKN2A deletion as a significant secondary abnormality in childhood acute lymphoblastic leukemia strongly correlated with phenotype and genotype. Schwindt et al29  reported frequent deletion at the CDKN2A locus in PCNSL, and they suggest that both deletion and DNA methylation may be considered as mechanisms of CDKN2A inactivation in PCNSL. Collectively, these findings indicate the importance of CDKN2A deletion in the pathogenesis of PCNSL.

Deletion of 6q also appears to be a common and important finding in the pathogenesis of PCNSL as well as in systemic DLBCL. Weber et al15  reported 6q as the region most frequently affected by losses of genomic material and mapped a commonly deleted region to 6q21-q22. Boonstra et al30  also described frequent 6q deletion in PCNSL, and they suggested that one or more suppressor genes involved in the pathogenesis of PCNSL may be localized in the region 6q16–22. Recently, Cady et al12  reported that del 6q22 is common in PCNSL, and it has been associated with inferior overall survival. In the present study, aCGH analysis of PCNSL frequently detected deletions in the region of chromosome 6q22, but no significantly down-regulated genes matched in this region. Among several genes showing deletion in this region, RSPO3, which is located at 6q22, showed deletion in 50% of the cases and also demonstrated decreased expression with borderline statistical significance (unadjusted P < .001 but FWER P > .05) in this study.

Using the expression array, we detected 96 significant genes (FWER, P < .05) from approximately 44 000 genes. To select significant genes, there are 2 standard statistical measurements, FWER and FDR, in multiple testing problems. FWER is one of the most conservative methods and provides much better protection against false positives than FDR, although this method has the disadvantage that some biologically significant genes could be missed because of its statistical conservatism. For example, SPP1 (osteopontin) has been reported as the gene most significantly up-regulated in PCNSL.6  In our study, SPP1 was not included in the top up-regulated genes; however, we identified that SPP1 was significantly up-regulated in PCNSL when the FDR method was applied (FDR Q < 0.05). Although we may have missed identifying a few biologically significant genes using an FWER correction, we wanted to protect against identifying false positives.

In the expression array analysis, we found that TACSTD1 (EPCAM), a pan-epithelial differentiation antigen commonly expressed on carcinomas, was significantly up-regulated. Tun et al6  also present this gene as a potentially important contributor to PCNSL. Another interesting up-regulated gene is HOOK3. This microtubule-binding protein interacts with scavenger receptor A, which is implicated in cell adhesion, innate immunity, and atherogenesis.31  Fusion of RET to the HOOK3 gene may lead to formation of an oncogene.32  Among the down-regulated genes, a few additional tumor suppressor genes were identified. Kank was found as a candidate tumor suppressor gene for renal cell carcinoma.33  Loss of Smad expression may be associated with development of some T-cell lymphomas.34  Smad3 expression may have a critical role in tumor suppression in the early stages of gastric carcinogenesis.35 ABI3BP (TARSH) mRNA expression is significantly down-regulated in all lung cancer cells and in follicular thyroid carcinoma.36,37 IGFBP-3 acts as tumor suppressor; IGFBP3 down-regulation is associated with shorter disease-specific survival in early non-small cell lung cancer and is an early event during head and neck carcinogenesis.38,39 

In this study, we used normal peripheral blood and normal lymph node tissue samples as the control group to identify genomic alterations in PCNSL. Until now, there have been only 3 studies of gene expression profiling PCNSL, which have all used systemic DLBCL as a control comparison5-7  because one of major issues in studying PCNSL is characterizing how it differs from systemic DLBCL. These previous studies suggest that PCNSL is indeed significantly different from DLBCL.6,7  Compared with systemic DLBCL, PCNSL appears to have a specific signature in the differential expression of ECM- and adhesion-related pathways, including candidate genes, such as SPP1, DDR1, CXCL13, MUM1, TCL1A, and CHI3L1.6  PCNSL also has a higher expression of c-MYC and Pim-1, and it may be that disruption of the IL-4 signaling pathway involving JAK1 and STAT6 could potentially facilitate apoptosis in PCNSL.7  Furthermore, several genes are differentially expressed between PCNSL and systemic DLBCL, including IGM, FN, SERPINA3, SPP1, TF, and HBA1.5,40  In this study, we identified a few overlapped genes, including TACSTD1, FOXG1, SPP1, COL12A1, COL6A, FN, and JAK1, despite using normal lymphoid tissue as control. These genes may be key genes to understand PCNSL biology. This study suggests that JAK-STAT pathway may have a significant role in pathogenesis of PCNSL, in addition to an ECM and cell-adhesion related pathway. The results are in agreement with those of Rubenstein et al,7  who also identified increased expression of JAK-1, suggesting a significant role of the JAK-STAT pathway in the pathogenesis of PCNSL.

Recently, Richter et al41  reported that PCNSL and DLBCL do not differ in their DNA methylation pattern; however, they identified 194 genes differentially methylated between PCNSL and normal hematopoietic controls. Among 194 genes, there were several genes, specifically MYH11, IGFBP3, and DES, which overlapped with findings from our results examining differentially expressed genes. These genes were unmethylated in normal controls but methylated in PCNSL,41  and these genes were significantly down-regulated in our study.

However, there are many genes that do not overlap, which may be the result of the methodologic differences among studies. Montesinos-Rongen et al40  described several reasons for this, including different platforms used, different algorithms applied, different methods to eliminate the background signature, low number of cases, and sample-induced bias (resection vs biopsy). Besides these, different control samples used and heterogeneity of patients may also result in different genes found among the studies. For example, some patients with PCNSL were given steroid before biopsy because their condition was very serious and rapidly progressed, or malignant lymphoma was not suspected from either clinical or radiologic standpoints. Such glucocorticoid treatment could affect gene expression, especially of apoptosis-related genes associated with glucocorticoid. In the expression array analysis of the current study, 2 of 7 patients received steroid treatment before surgery. Although the whole gene expression pattern of these 2 patients showed no clustering distinct from the non–glucocorticoid-treated group, there may have been potential influence in gene expression, which will need to be evaluated in larger studies.

In conclusion, we identified pathways involved in signal transduction and cell adhesion that may contribute to PCNSL pathogenesis. By combining aCGH with expression array analysis, we identified several candidate genes involved in cell signaling, ECM activity, and cell adhesion, which may perform tumor suppressor gene functions in the pathogenesis of PCNSL. Although the molecular and cellular pathways that lead to PCNSL still remain largely unknown and existing evidence appears to be mixed, this study identified novel tumor suppressor genes that may serve as therapeutic targets of PCNSL.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

The authors thank Kyusam Choi of Samsung Cancer Research Institute in Seoul for his excellent technical support in real-time RT-PCR from formalin-fixed paraffin-embedded tissues.

This work was supported by the Samsung Biomedical Research Institute (grant SBRI C-A8-202), Korea Healthcare Technology R&D Project, and Ministry for Health & Welfare, Republic of Korea (A092255).

Contribution: C.O.S. performed a pathology review and research, analyzed data, collected clinical data, and wrote the manuscript; S.C.K. analyzed data; S.K., K.K., and M.S. performed research and obtained samples; H.J.S. and D.-H.N. obtained samples and collected clinical data; S.J.K. and W.S.K. collected clinical data; Y.-L.S. and J.Y.K. performed a pathology review; S.-H.K. performed research; and Y.-H.K. designed research, performed a pathology review and research, analyzed data, obtained samples, and wrote the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Young-Hyeh Ko, Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 50 Irwon-dong Gangnam-gu, Seoul 135-710, South Korea; e-mail: [email protected].

1
Choi
 
YL
Suh
 
YL
Kim
 
D
Ko
 
YH
Sung
 
CO
Lee
 
JI
Malignant lymphoma of the central nervous system: difficult histologic diagnosis after glucocorticoid therapy prior to biopsy.
Clin Neuropathol
2006
, vol. 
25
 
1
(pg. 
29
-
36
)
2
Commins
 
DL
Pathology of primary central nervous system lymphoma.
Neurosurg Focus
2006
, vol. 
21
 
5
pg. 
E2
 
3
Bhagavathi
 
S
Sharathkumar
 
A
Hunter
 
S
et al. 
Activated B-cell immunophenotype might be associated with poor prognosis of primary central nervous system lymphomas.
Clin Neuropathol
2008
, vol. 
27
 
1
(pg. 
13
-
20
)
4
Camilleri-Broet
 
S
Criniere
 
E
Broet
 
P
et al. 
A uniform activated B-cell-like immunophenotype might explain the poor prognosis of primary central nervous system lymphomas: analysis of 83 cases.
Blood
2006
, vol. 
107
 
1
(pg. 
190
-
196
)
5
Montesinos-Rongen
 
M
Brunn
 
A
Bentink
 
S
et al. 
Gene expression profiling suggests primary central nervous system lymphomas to be derived from a late germinal center B cell.
Leukemia
2008
, vol. 
22
 
2
(pg. 
400
-
405
)
6
Tun
 
HW
Personett
 
D
Baskerville
 
KA
et al. 
Pathway analysis of primary central nervous system lymphoma.
Blood
2008
, vol. 
111
 
6
(pg. 
3200
-
3210
)
7
Rubenstein
 
JL
Fridlyand
 
J
Shen
 
A
et al. 
Gene expression and angiotropism in primary CNS lymphoma.
Blood
2006
, vol. 
107
 
9
(pg. 
3716
-
3723
)
8
Malumbres
 
R
Davis
 
J
Ruiz
 
P
Lossos
 
IS
Somatically mutated immunoglobulin IGHV@ genes without intraclonal heterogeneity indicate a postgerminal centre origin of primary intraocular diffuse large B-cell lymphomas.
Br J Haematol
2007
, vol. 
138
 
6
(pg. 
749
-
755
)
9
Lipford
 
E
Wright
 
JJ
Urba
 
W
et al. 
Refinement of lymphoma cytogenetics by the chromosome 18q21 major breakpoint region.
Blood
1987
, vol. 
70
 
6
(pg. 
1816
-
1823
)
10
Weiss
 
LM
Warnke
 
RA
Sklar
 
J
Cleary
 
ML
Molecular analysis of the t(14;18) chromosomal translocation in malignant lymphomas.
N Engl J Med
1987
, vol. 
317
 
19
(pg. 
1185
-
1189
)
11
Yunis
 
JJ
Mayer
 
MG
Arnesen
 
MA
Aeppli
 
DP
Oken
 
MM
Frizzera
 
G
bcl-2 and other genomic alterations in the prognosis of large-cell lymphoma.
N Engl J Med
1989
, vol. 
320
 
16
(pg. 
1047
-
1054
)
12
Cady
 
FM
O'Neill
 
BP
Law
 
ME
et al. 
Del(6)(q22) and BCL6 rearrangements in primary CNS lymphoma are indicators of an aggressive clinical course.
J Clin Oncol
2008
, vol. 
26
 
29
(pg. 
4814
-
4819
)
13
Chu
 
LC
Eberhart
 
CG
Grossman
 
SA
Herman
 
JG
Epigenetic silencing of multiple genes in primary CNS lymphoma.
Int J Cancer
2006
, vol. 
119
 
10
(pg. 
2487
-
2491
)
14
Cobbers
 
JM
Wolter
 
M
Reifenberger
 
J
et al. 
Frequent inactivation of CDKN2A and rare mutation of TP53 in PCNSL.
Brain Pathol
1998
, vol. 
8
 
2
(pg. 
263
-
276
)
15
Weber
 
T
Weber
 
RG
Kaulich
 
K
et al. 
Characteristic chromosomal imbalances in primary central nervous system lymphomas of the diffuse large B-cell type.
Brain Pathol
2000
, vol. 
10
 
1
(pg. 
73
-
84
)
16
Huber
 
W
von Heydebreck
 
A
Sultmann
 
H
Poustka
 
A
Vingron
 
M
Variance stabilization applied to microarray data calibration and to the quantification of differential expression.
Bioinformatics
2002
, vol. 
18
 
suppl 1
(pg. 
S96
-
S104
)
17
Dennis
 
G
Sherman
 
BT
Hosack
 
DA
et al. 
DAVID: Database for Annotation, Visualization, and Integrated Discovery.
Genome Biol
2003
, vol. 
4
 
5
pg. 
P3
 
18
Huang da
 
W
Sherman
 
BT
Lempicki
 
RA
Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources.
Nat Protoc
2009
, vol. 
4
 
1
(pg. 
44
-
57
)
19
Mootha
 
VK
Lindgren
 
CM
Eriksson
 
KF
et al. 
PGC-1alpha-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes.
Nat Genet
2003
, vol. 
34
 
3
(pg. 
267
-
273
)
20
Subramanian
 
A
Tamayo
 
P
Mootha
 
VK
et al. 
Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles.
Proc Natl Acad Sci U S A
2005
, vol. 
102
 
43
(pg. 
15545
-
15550
)
21
Ushmorov
 
A
Leithauser
 
F
Ritz
 
O
Barth
 
TF
Moller
 
P
Wirth
 
T
ABF-1 is frequently silenced by promoter methylation in follicular lymphoma, diffuse large B-cell lymphoma and Burkitt's lymphoma.
Leukemia
2008
, vol. 
22
 
10
(pg. 
1942
-
1944
)
22
Wu
 
M
Han
 
L
Shi
 
Y
et al. 
Development and characterization of a novel method for the analysis of gene expression patterns in lymphatic endothelial cells derived from primary breast tissues.
J Cancer Res Clin Oncol
2010
, vol. 
136
 
6
(pg. 
863
-
872
)
23
Grant
 
SL
Begley
 
CG
The oncostatin M signalling pathway: reversing the neoplastic phenotype?
Mol Med Today
1999
, vol. 
5
 
9
(pg. 
406
-
412
)
24
Chen
 
SH
Benveniste
 
EN
Oncostatin M: a pleiotropic cytokine in the central nervous system.
Cytokine Growth Factor Rev
2004
, vol. 
15
 
5
(pg. 
379
-
391
)
25
Schaefer
 
LK
Wang
 
S
Schaefer
 
TS
Oncostatin M activates stat DNA binding and transcriptional activity in primary human fetal astrocytes: low- and high-passage cells have distinct patterns of stat activation.
Cytokine
2000
, vol. 
12
 
11
(pg. 
1647
-
1655
)
26
Ferrajoli
 
A
Faderl
 
S
Ravandi
 
F
Estrov
 
Z
The JAK-STAT pathway: a therapeutic target in hematological malignancies.
Curr Cancer Drug Targets
2006
, vol. 
6
 
8
(pg. 
671
-
679
)
27
Zhu
 
BM
Ishida
 
Y
Robinson
 
GW
et al. 
SOCS3 negatively regulates the gp130-STAT3 pathway in mouse skin wound healing.
J Invest Dermatol
2008
, vol. 
128
 
7
(pg. 
1821
-
1829
)
28
Sulong
 
S
Moorman
 
AV
Irving
 
JA
et al. 
A comprehensive analysis of the CDKN2A gene in childhood acute lymphoblastic leukemia reveals genomic deletion, copy number neutral loss of heterozygosity, and association with specific cytogenetic subgroups.
Blood
2009
, vol. 
113
 
1
(pg. 
100
-
107
)
29
Schwindt
 
H
Vater
 
I
Kreuz
 
M
et al. 
Chromosomal imbalances and partial uniparental disomies in primary central nervous system lymphoma.
Leukemia
2009
, vol. 
23
 
10
(pg. 
1875
-
1884
)
30
Boonstra
 
R
Koning
 
A
Mastik
 
M
van den Berg
 
A
Poppema
 
S
Analysis of chromosomal copy number changes and oncoprotein expression in primary central nervous system lymphomas: frequent loss of chromosome arm 6q.
Virchows Arch
2003
, vol. 
443
 
2
(pg. 
164
-
169
)
31
Sano
 
H
Ishino
 
M
Kramer
 
H
et al. 
The microtubule-binding protein Hook3 interacts with a cytoplasmic domain of scavenger receptor A.
J Biol Chem
2007
, vol. 
282
 
11
(pg. 
7973
-
7981
)
32
Ciampi
 
R
Giordano
 
TJ
Wikenheiser-Brokamp
 
K
Koenig
 
RJ
Nikiforov
 
YE
HOOK3-RET: a novel type of RET/PTC rearrangement in papillary thyroid carcinoma.
Endocr Relat Cancer
2007
, vol. 
14
 
2
(pg. 
445
-
452
)
33
Sarkar
 
S
Roy
 
BC
Hatano
 
N
Aoyagi
 
T
Gohji
 
K
Kiyama
 
R
A novel ankyrin repeat-containing gene (Kank) located at 9p24 is a growth suppressor of renal cell carcinoma.
J Biol Chem
2002
, vol. 
277
 
39
(pg. 
36585
-
36591
)
34
Go
 
JH
Altered expression of Smad proteins in T or NK-cell lymphomas.
Cancer Res Treat
2008
, vol. 
40
 
4
(pg. 
197
-
201
)
35
Han
 
SU
Kim
 
HT
Seong
 
DH
et al. 
Loss of the Smad3 expression increases susceptibility to tumorigenicity in human gastric cancer.
Oncogene
2004
, vol. 
23
 
7
(pg. 
1333
-
1341
)
36
Cerutti
 
JM
Delcelo
 
R
Amadei
 
MJ
et al. 
A preoperative diagnostic test that distinguishes benign from malignant thyroid carcinoma based on gene expression.
J Clin Invest
2004
, vol. 
113
 
8
(pg. 
1234
-
1242
)
37
Terauchi
 
K
Shimada
 
J
Uekawa
 
N
Yaoi
 
T
Maruyama
 
M
Fushiki
 
S
Cancer-associated loss of TARSH gene expression in human primary lung cancer.
J Cancer Res Clin Oncol
2006
, vol. 
132
 
1
(pg. 
28
-
34
)
38
Papadimitrakopoulou
 
VA
Brown
 
EN
Liu
 
DD
et al. 
The prognostic role of loss of insulin-like growth factor-binding protein-3 expression in head and neck carcinogenesis.
Cancer Lett
2006
, vol. 
239
 
1
(pg. 
136
-
143
)
39
Chang
 
YS
Kong
 
G
Sun
 
S
et al. 
Clinical significance of insulin-like growth factor-binding protein-3 expression in stage I non-small cell lung cancer.
Clin Cancer Res
2002
, vol. 
8
 
12
(pg. 
3796
-
3802
)
40
Montesinos-Rongen
 
M
Siebert
 
R
Deckert
 
M
Primary lymphoma of the central nervous system: just DLBCL or not?
Blood
2009
, vol. 
113
 
1
(pg. 
7
-
10
)
41
Richter
 
J
Ammerpohl
 
O
Martín-Subero
 
JI
et al. 
Array-based DNA methylation profiling of primary lymphomas of the central nervous system.
BMC Cancer
2009
, vol. 
9
 pg. 
455
 
Sign in via your Institution