Skip to main content
Erschienen in: Medical Oncology 1/2015

Open Access 01.01.2015 | Original Paper

Gene expression profile analysis identifies metastasis and chemoresistance-associated genes in epithelial ovarian carcinoma cells

verfasst von: Liancheng Zhu, Zhenhua Hu, Juanjuan Liu, Jian Gao, Bei Lin

Erschienen in: Medical Oncology | Ausgabe 1/2015

Abstract

The purpose of this study was to identify genes that associated with higher ability of metastasis and chemotherapic resistance in epithelial ovarian carcinoma (EOC) cells. An oligonucleotide microarray with probe sets complementary to 41,000+ unique human genes and transcripts was used to determine whether gene expression profile may differentiate three epithelial ovarian cell lines (RMG-I-C, COC1 and HO8910) from their sub-lines (RMG-I-H, COCI/DDP and HO8910/PM) with higher ability of metastasis and chemotherapic resistance. Quantitative real-time PCR and immunohistochemical staining validated the microarray results. Hierarchic cluster analysis of gene expression identified 49 genes that exhibited ≥2.0-fold change and P value ≤0.05. Highly differential expression of GCET2, NLRP4, FOXP1 and SNX29 genes was validated by quantitative PCR in all cell line samples. Finally, FOXP1 was validated at the protein level by immunohistochemistry in paraffin embedded ovarian tissues (i.e., for metastasis, 15 primary EOC and 10 omental metastasis [OM]; for chemoresistance, 13 sensitive and 13 resistant EOC). The identification of higher ability of metastasis and chemotherapic resistance-associated genes may provide a foundation for the development of new type-specific diagnostic strategies and treatment for metastasis and chemotherapic resistance in epithelial ovarian cancer.

Introduction

Epithelial ovarian cancers (EOC) are high aggressive tumors associated with high mortality and morbidity in gynecology. Although the 5-year survival rate is 90 % for women with early-stage ovarian cancer and postoperative introduction of paclitaxel drug have improved the 5-year survival rate for advanced-stage ovarian cancer, patients with this cancer have a 5-year survival rate of only 30 % [1]. Standard therapy includes cytoreductive surgery with first-line combination chemotherapy, 75 % of patients initially respond to conventional chemotherapy; however, 80 % of these women eventually relapse and die from chemotherapy resistant disease. Thus, to understand the molecular basis of epithelial ovarian cancer metastasis and chemotherapeutic resistance is of vital importance and may have the potential to improve significantly the development of more specific and effective treatment against EOC. Comprehensive, high-throughput technologies such as gene expression microarrays have provided powerful tools for this purpose.
In the present study, the whole human genome oligo microarray was used to investigate the differential expression genes (DEGs) in the human ovary cancer cell lines RMG-1-C, COC1, HO8910 and their high malignant and chemoresistant sub-cell lines RMG-I-H, COC1/DDP and HO8910/PM. Hierarchical clustering of genes by the expression level of the DEGs was performed. The potential functions of the DEGs were analyzed by Gene Ontology (GO) and pathway enrichment analyses. In addition, the interaction relationships between these DEGs were investigated by regulatory network. We hope these metastasis and chemotherapy resistance-associated genes may be used for early detection of epithelial ovarian cancer and for development of more specific chemotherapy drugs against EOC.

Materials and methods

Cell culture

The human ovarian cancer cell strain RMG-1 was the courtesy of Doctor Iwamori Masao of Kinki University in Japan. We transfected the gene of extrinsic α1,2-fucosyl transferase (α1,2-FT) into RMG-1 to create cell line RMG-1-H with high expression of Lewis (y) and α1,2-fucosyl transferase [2, 3], and we discovered that compared with the empty plasmid vector transfected cell line RMG-1-C, cell line RMG-1-H showed enhanced cellular malignant biological behaviors, such as enhanced metastasis and proliferation [4], adhesion [5] and multiple drug resistance [6].
Human ovarian cancer cell lines HO-8910 and HO-8910 PM (a highly metastatic cell line derived from HO-8910) were purchased from the Cell Bank of Type Culture Collection of the Chinese Academy of Sciences (Shanghai, China) [7], human ovarian cancer cell lines COC1 and COC1/DDP (a platinum resistance cell line derived from COC1) were purchased from the China Center for Type Culture Collection (Wuhan, China). Cells were cultured in RPMI-1640 medium supplemented with 100 units/mL penicillin/streptomycin and 10 % fetal bovine serum (FBS) and maintained in an incubator at 37 °C under a humidified atmosphere of 5 % CO2. COC1/DDP cells were cultured in RPMI-1640 medium containing 0.5 µg/mL cisplatin (Sigma, St. Louis, MO, USA) to maintain the drug resistant phenotype. The cell lines and labels in this experiment are listed in Table 1.
Table 1
Cell line samples description
Label
A
B
C
1
2
3
Cell line
RMG-I-H
COC1/DDP
HO8910/PM
RMG-I-C
COC1
HO8910

Total RNA extraction and gene chip hybridization

Total RNA was extracted from all 6 cell line samples with TRIZOL reagent (Life Technologies, Inc, Carlsbad, CA) and further purified with RNeasy Min-elute Clean-up Columns (Qiagen, Valencia, CA), as described by the manufacturers. Optical density for each sample of RNA was measured at OD 260 nm and OD 280 nm using NanoDrop ND-1000 (NanoDrop Technologies, Wilmington, DE, USA). All RNA samples isolated OD260/280 ratio should be close to 2.0 for pure RNA (ratios between 1.8 and 2.1 are acceptable). The OD A260/A230 ratio should be more than 1.8. Each isolated RNA sample was subjected to further quality check to ensure integrity of RNA with Agilent RNA 6000 Nano LabChip using Agilent 2100 Bioanalyzer (Agilent Technologies, Santa Clara, CA, USA). All RNA samples were verified to be intact with distinct 28S and 18S RNA bands at a ratio of approximately 2:1 and a RNA integrity number (RIN) > 7.
The samples were amplified and labeled using the Agilent Quick Amp labeling kit and hybridized with Agilent whole genome oligo microarray in Agilent’s SureHyb Hybridization Chambers in accordance with the manufacturer’s instructions. This array contains 41,000+ unique human genes and transcripts represented, all with public domain annotations, content sourced from RefSeq, Goldenpath Ensembl Unigene Human Genome (Build 33) and GenBank databases, over 70 % of the represented probes are validated by Agilent’s laboratory validation process, 4 × 44 K slide formats printed using Agilent’s 60-mer SurePrint technology. After hybridization and washing, the processed slides were scanned with the Agilent DNA microarray scanner (part number G2505B) using settings recommended by Agilent Technologies.

Data analysis and clustering

Agilent Feature Extraction Software (version 10.5.1.1) was used to extract the signal intensity values from each gene chip, and the resulting text files were imported into the Agilent GeneSpring GX software (version11.0) for further analysis. The 6-microarray data sets were normalized in GeneSpring GX using the Agilent FE one-color scenario (mainly median normalization), and genes marked present or marginal in all samples were chosen for data analysis. DEGs were identified through fold-change screening comparing between cell lines group A, B, C and cell lines group 1, 2 and 3. The threshold used to screen up or down-regulated genes is fold change ≥2.0 and P value ≤0.05. A scatter plot was made to visualizingly assess the variation between chips. A hierarchical clustering and volcano plot were performed to visualizingly show a distinguishable gene expression profiling among samples.

Validation for gene expression by quantitative real-time polymerase chain reaction

Real-time polymerase chain reaction (RT-PCR) was performed in triplicate with primer sets and probes that were specific for 4 selected genes that were found to be significantly differentially expressed. These 4 genes were 2 up-regulated genes: GCET2, CFTR and 2 down-regulated genes: FOXP1, GARS. cDNA was synthesized using random primers (hexamers) and Oligo 18dT and Superscript II Reverse Transcriptase (Invitrogen). Real-time PCR was performed on Roche LightCycler 480 sequence detection system, using the following amplification conditions: 5 min, 95 °C; followed by 40 cycles of 15 s 95 °C, 1 min 60 °C and 20 s 72 °C. CT values were determined using the IQ5 software (Bio-Rad). The primers mostly were searched from PrimerBank (http://​pga.​mgh.​harvard.​edu/​primerbank/​ index.html). Primers of target genes are listed in Table 2. The comparative threshold cycle method was used for the calculation of amplification fold, as specified by the manufacturer. The housekeeping gene glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was used to normalize the quantity of complementary DNA that was used in the PCR reactions.
Table 2
Gene-specific primers used for validation
Gene name
GenBank accession number
Primer sequence (5′-3′)
Amplicon size (bp)
GCET2
NM_152785
F: ACCCTCATCAATCATCGGGTT
122
R: TCAGTCTCAGTTCCTCCCAAG
CFTR
NM_000492
F: TGCCCTTCGGCGATGTTTTT
127
R: GTTATCCGGGTCATAGGAAGCTA
FOXP1
NM_032682
F: TCCCGTGTCAGTGGCTATGAT
226
R: CTCTTTAGGCTGTTTTCCAGCAT
GARS
NM_002047
F: TTGGCCCAGCTTGATAACTATG
103
R: ACACTGGAGGGGATAGATCATTT
GAPDH
NM_001256799
F: ACAACTTTGGTATCGTGGAAGG
101
R: GCCATCACGCCACAGTTTC
F forward primer, R reverse primer

Immunohistochemistry on paraffin embedded tissues

To evaluate protein expression levels for 1 of the 49 genes that was found to be different regulated, in consideration of a further study, immunohistochemical staining for FOXP1 was performed on ovarian tissue samples. 29 cases of primary ovarian cancer samples and 25 cases of omental metastatic (OM) ovarian cancer samples were collected from Shengjing Hospital of China Medical University in 2013, the histopathological diagnoses were determined using the WHO criteria. In our previous studies, we have established a set of ovarian cancer chemotherapeutic sensitive and resistant paraffin embedded samples [8, 9], and we randomly selected 40 samples in sensitive and 30 samples in resistant group for FOXP1 staining. There was no statistical difference between these two groups of ovarian samples in age, pathological subtype, lymph node metastasis or residual tumor size (data not shown). FOXP1 staining was performed using JC12 mouse anti-human monoclonal antibodies (diluted 1:40, JC12 was kindly provided by Alison H. Banham, University of Oxford, UK [10]) using the Envision detection kit (Maixin. Bio China). Positive myoepithelial cell staining and negative stromal cell staining in breast carcinoma were used as internal positive and negative controls, respectively. FOXP1 nuclear expression was scored using the following system: negative = 0; weak/focal = 1; strong focal/widespread moderate staining = 2; or strong/widespread staining = 3. Tumors that scored 2 or 3 were considered positive for FOXP1 nuclear staining. Survival analysis was performed on those patients, and the overall survival (OS) time was defined from the date of surgery (earliest was in July, 2004) to the date of death or the last follow-up (Jun, 2014).

Enrichment analysis of DEGs

Gene Set Analysis Toolkit (Gestalt) tool was used to do enrichment analysis on the DEGs in three sections of function, biological process and molecular composition. Gestalt is a suite rich of analysis of biologically relevant content collecting eight species, including human, rat, mouse and other data from various different public data resources, such as NCBI, Ensemble, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG).

Construction of gene regulatory network

The gene regulatory network was visualized by Cytoscape [11]. Proteins in the network served as the “nodes,” and each pairwise protein interaction (referred to as edge) was represented by an undirected link. The property of the network was analyzed with the plug-in network analysis.

Statistics

Statistical analysis was performed using Graphpad Prism 6.0e Software for Mac OS X (GraphPad Software, La Jolla California USA, www.​graphpad.​com). Student’s t test was employed for comparison between two groups and one-way ANOVA with Tukey’s post hoc test was used for comparison between more than two groups. As to the analysis of quantitative RT-PCR result, data were expressed as mean ± SEM to compare on mRNA expression between different groups. The Chi square and Kaplan–Meier survival analysis were applied to analyze the nuclear expression of FOXP1. For these tests, a P value of <0.05 was considered statistically significant.

Results

Gene expression analysis and clustering

The expression profiles of all the samples passed the microarray quality control (Table 3); a scatter plot was constructed with a two-dimensional rectangular coordinate plane (Fig. 1).
Table 3
Sample qualification
Sample ID
OD260/280
OD260/230
Concentration (ng/µL)
RIN
28S/18S
Results
A
2.06
1.9
1,126.89
8.9
1.8
Qualified
B
2.06
2.09
1,692.74
8.7
1.9
Qualified
C
2.06
2.19
1,092.17
8.0
1.8
Qualified
1
2.07
1.95
1,431.45
8.6
1.7
Qualified
2
2.07
2
1,428.15
8.5
1.8
Qualified
3
2.07
2.14
1,024.49
8.9
2.1
Qualified
Using hierarchical clustering map analysis with probe sets, the DEGs were identified in visualization, which readily distinguished the 2 groups as shown in Fig. 2. The volcano plot of DEGs revealed a total of 49 probe sets that showed a ≥2.0-fold change and P value ≤0.05, as shown in Fig. 3. Of 49 genes, 14 genes were found to be up-regulated and 35 genes down-regulated (Table 4).
Table 4
Differentially expressed genes at least twofold higher
Gene symbol
GenBank accession
Description
Fold change
P value
Up-regulated genes
 GCET2
NM_001008756
Germinal center expressed transcript 2
3.25461
0.01955
 TMEFF1
NM_003692
Transmembrane protein with EGF-like and two follistatin-like domains 1
2.48936
0.02476
 PTTG3
NR_002734
Pituitary tumor-transforming 3 on chromosome 8
2.39659
0.00859
 CFTR
NM_000492
Cystic fibrosis transmembrane conductance regulator isoform 36
2.37741
0.02794
 MS4A6A
NM_022349
Membrane-spanning 4-domains, subfamily A, member 6A
2.32408
0.02803
 FFAR2
NM_005306
Free fatty acid receptor 2
2.26184
0.01446
 BX648831
BX648831
Poly(A) binding protein, cytoplasmic 4-like
2.25877
0.01799
 GPRC6A
NM_148963
G-protein-coupled receptor, family C, group 6, member A
2.17761
0.02608
 SLC25A42
NM_178526
Solute carrier family 25, member 42
2.13807
0.02038
 SVEP1
AK027870
Sushi, von Willebrand factor type A, EGF and pentraxin domain containing 1
2.1106
0.00132
 NLRP4
NM_134444
NLR family, pyrin domain containing 4
2.10449
0.00205
 GLP1R
NM_002062
Glucagon-like peptide 1 receptor
2.09506
0.02839
 LOC643406
BC031676
Hypothetical protein LOC643406
2.04176
0.01908
 FLJ14816
BC113708
Hypothetical protein FLJ14816
2.01485
0.04223
Down-regulated genes
 RPL28P1
XR_019242
Ribosomal protein L28 pseudogene 1
3.80897
0.01208
 RPL23A
NM_000984
Ribosomal protein L23a
3.35458
0.04575
 RPL13AP3
BC067891
Ribosomal protein L13a pseudogene 3
3.0956
0.00275
 COX19
NM_001031617
COX19 cytochrome c oxidase assembly homolog (S. cerevisiae)
2.85475
0.01561
 RBMX
NM_002139
RNA binding motif protein, X-linked
2.77763
0.04326
 LOC341412
CA455253
Hypothetical LOC341412, pseudo gene
2.764
0.0313
 LOC641784
AW302767
Similar to ribosomal protein L31, pseudo gene
2.7233
0.04737
 FOXP1
NM_032682
Forkhead box P1
2.63004
0.03159
 COL27A1
NM_032888
Collagen, type XXVII, alpha 1
2.62832
0.02947
 PTMA
NM_002823
Prothymosin, alpha
2.58023
0.04672
 CALCOCO2
NM_005831
Calcium binding and coiled-coil domain 2
2.53815
0.02005
 DNAJB6
NM_005494
DnaJ (Hsp40) homolog, subfamily B, member 6
2.53533
0.02531
 LOC391560
XR_018524
Ribosomal protein L32 pseudogene 7
2.43023
0.04685
 ZNF234
NM_006630
Zinc finger protein 234
2.37866
0.0357
 WASF2
NM_006990
WAS protein family, member 2
2.37554
0.02993
 AP3S2
NM_005829
Adaptor-related protein complex 3, sigma 2 subunit
2.36803
0.01003
 KLF2
NM_016270
Kruppel-like factor 2 (lung)
2.36563
0.03192
 ZC3H11A
NM_014827
Zinc finger CCCH-type containing 11A
2.35468
0.02414
 RPS16P9
XR_016930
Ribosomal protein S16 pseudogene 9
2.32731
0.00056
 EIF1B
NM_005875
Eukaryotic translation initiation factor 1B
2.28022
0.00834
 NR4A2
NM_006186
Nuclear receptor subfamily 4, group A, member 2
2.25709
0.03943
 LY6G6C
NM_025261
Lymphocyte antigen 6 complex, locus G6C
2.19288
0.0412
 RPS7P5
AK098605
Ribosomal protein S7 pseudogene 5
2.17098
0.00883
 RPLP0P2
NR_002775
Ribosomal protein, large, P0 pseudogene 2
2.15877
0.03239
 CCDC144A
BC034617
Coiled-coil domain containing 144A
2.15257
0.01531
 GARS
NM_002047
Glycyl-tRNA synthetase
2.14073
0.04554
 LOC388524
NM_001005472
Similar to Laminin receptor 1
2.13836
0.04665
 ZNF467
NM_207336
Zinc finger protein 467
2.12557
0.04513
 LOC732186
XR_016076
Similar to signal sequence receptor gamma subunit, pseudo gene
2.0591
0.01044
 ZBTB43
NM_014007
Zinc finger and BTB domain containing 43
2.05301
0.02564
 RPL13AP23
XR_018808
Ribosomal protein L13a pseudogene 23
2.04535
0.00653
 RPLP1P7
CH471086
Ribosomal protein, large, P1 pseudogene 7
2.04394
0.00846
 RPL31P10
XR_018695
Ribosomal protein L31 pseudogene 10
2.04042
0.03652
 SNX29
AK024473
Sorting nexin 29
2.03297
0.04401
 LOC648361
XM_001127349
Similar to 40S ribosomal protein S12, pseudogene
2.00295
0.04577

Validation of gene expression results by using quantitative RT-PCR

Four highly differentially expressed genes (i.e., GCET2, CFTR, FOXP1 and SNX29) were selected for quantitative RT-PCR analysis as shown in Fig. 4. These results were in good agreement with the microarray data, confirming the reliability of the microarray results.

Validation of protein expression by immunohistochemical staining

To confirm gene expression results at the protein level, immunohistochemistry for FOXP1 was carried out on all paraffin embedded samples. For metastasis, as shown in Table 5, FOXP1 nuclear positive staining in EOC was detected in 12 of 29 EOC samples (41.4 %), while only 4 of 25 OM samples (16.0 %) showed positive nuclear staining for FOXP1 (P = 0.042). For chemotherapeutic resistance, as shown in Table 6 and depicted in Fig. 5, FOXP1 nuclear positive staining in sensitive group was detected in 17 of 40 sensitive samples (42.5 %), while only 5 of 30 resistant samples (16.7 %) showed positive nuclear staining for FOXP1 (P = 0.021). A Kaplan–Meier survival analysis was applied to further investigate the effect of FOXP1 protein on ovarian cancer patients, as shown in Fig. 6, positive nuclear staining of FOXP1 was an independent risk factor and strongly correlated with prognosis.
Table 5
FOXP1 protein expression in 29 primary epithelial ovarian cancer (EOC) and 25 omental metastasis (OM) epithelial ovarian cancer
 
Cases
Nucleus staining
+
EOC
29
17
12
OM
25
21
4
Table 6
FOXP1 protein expression in 40 chemotherapy-sensitive epithelial ovarian cancers and 30 resistant ovarian epithelial cancers
 
Cases
Nucleus staining
+
Sensitive
40
23
17
Resistant
30
25
5

GO function analysis and Signal pathway result of differential genes

Significant bioprocesses of the DEGs, gene expression, biopolymer biosynthetic process, macromolecule biosynthetic process, cAMP-mediated signaling, nucleic acid metabolic process, transcription and so on (Table 7). A total of 176 KEGG pathways were enriched for the 49 DEGs, including 20 significantly enriched pathways (Table 8), such as P450 hydroxylations, HIF-1-alpha transcription factor network, mechanism of acetaminophen activity and toxicity, cystic fibrosis transmembrane conductance regulator (CFTR) and beta 2 adrenergic receptor (B2AR) pathway, alpha6beta4integrin, negative regulation of the PI3K/AKT network.
Table 7
Classification of the up-regulated and down- regulated genes involved in the significant bioprocesses
GO Term
P value
Count in selection
% Count in selection
Count in total
% Count in total
Gene expression
9.36E−04
11
39.285713
2,187
14.157172
Biopolymer biosynthetic process
0.001659173
10
35.714287
1,972
12.765407
Macromolecule biosynthetic process
0.0031013
10
35.714287
2,141
13.859399
cAMP-mediated signaling
0.00382362
2
7.142857
51
0.33013982
Nucleobase, nucleoside, nucleotide and nucleic acid metabolic process
0.00942933
10
35.714287
2,495
16.150957
Cyclic-nucleotide-mediated signaling
0.010541212
2
7.142857
86
0.55670637
Transcription
0.014451807
7
25
1,483
9.599948
Biosynthetic process
0.014984685
10
35.714287
2,668
17.270844
G-protein-coupled receptor activity
0.016115764
4
14.285714
546
3.5344381
Translation
0.021661116
3
10.714286
332
2.1491456
Nucleic acid binding
0.030367257
10
35.714287
2,970
19.22579
Biopolymer metabolic process
0.030960111
12
42.857143
3,891
25.187727
Receptor activity
0.036230754
6
21.428572
1,398
9.049715
G-protein-coupled receptor protein signaling pathway
0.036383796
4
14.285714
702
4.5442777
Second-messenger-mediated signaling
0.042707212
2
7.142857
182
1.178146
Ribonucleoprotein complex
0.046174083
3
10.714286
447
2.8935785
Rhodopsin-like receptor activity
0.04668641
3
10.714286
449
2.9065251
Structural constituent of ribosome
0.0483463
2
7.142857
195
1.2622993
Macromolecule metabolic process
0.049876012
13
46.42857
4,646
30.07509
Table 8
Analysis of the differential gene pathways
Pathway
Number of entities
Matched with technology
Matched with entity list
P value
P450 Hydroxylations
19
4
2
0.002811053
HIF-1-alpha transcription factor network
88
73
6
0.002937696
Mechanism of acetaminophen activity and toxicity
12
5
2
0.004168975
Hypoxic and oxygen homeostasis regulation of HIF-1-alpha
111
86
6
0.006581206
Cystic fibrosis transmembrane conductance regulator (CFTR) and beta 2 adrenergic receptor (B2AR) pathway
14
3
1
0.006656855
De novo synthesis of IMP
32
4
1
0.011070482
Cytochrome p450
54
9
2
0.01444615
Phase 1 functionalization
87
9
2
0.01444615
il12 and stat4 dependent signaling pathway in th1 development
13
10
2
0.015267268
alpha6beta4integrin
53
50
4
0.01597487
IL4-mediated signaling events
84
52
4
0.018223463
Purine metabolism
100
9
1
0.022020191
Xenobiotics
60
15
2
0.0333969
Stathmin and breast cancer resistance to antimicrotubule agents
18
2
1
0.03385132
Negative regulation of the PI3 K/AKT network
12
2
1
0.03385132
TCR
140
125
6
0.035289083
Gap-filling DNA repair synthesis and ligation in GG-NER
7
2
1
0.038461793
Gap-filling DNA repair synthesis and ligation in TC-NER
7
2
1
0.038461793
FOXA1 transcription factor network
53
40
3
0.04229567
Nucleotide metabolism
198
22
1
0.049933493

Establishment of regulatory network for the DEGs

In order to further investigate the global expression occurring and to define how individual up- or down-regulated genes interact with each other to have a coordinated role, we identified potential networks for these DEGs (Fig. 7). Among the 49 DEGs, 20 were involved in the establishment of regulation network, of which 4 were up-regulated and 16 were down-regulated. A total of 21 transcription factors (TFs) were predicted, in which UBC and EP200 are the most connected predicted hub genes.

Discussion

Ovarian cancer has the highest mortality rate of all gynecologic cancers, and 75 % of patients diagnosed with ovarian cancer are already at an advanced stage. Chemotherapy is important in treating and preventing the recurrence of ovarian cancer; however, resistance is an obstacle to overcome and finding new treatment strategies has become increasingly valuable. High-throughput technologies for assaying gene expression, such as high-density oligonucleotide and cDNA microarrays, may offer the potential to identify clinically relevant genes highly differentially expressed between different cell lines. Thus, this study showed the first communication of an investigation that involved the genome-wide examination of differences in gene expression between ovarian cancer cell lines and their sub-lines with enhanced metastasis and chemotherapic resistance. We identified 49 genes that were expressed differentially between Group A, B, C and Group 1, 2, 3, and the average change in expression level between the two groups was at least twofold. The known functions of some of these genes can provide insights with the highly metastasis and chemotherapic resistance for ovarian cancer, although others are still useful for a further research.
GCET2 is found to be the most up-regulated genes in the more enhanced metastasis and chemoresistance cell group, and it is also known as human germinal center associated lymphoma (HGAL) gene, is specifically expressed in germinal center B-lymphocytes and germinal center-derived B cell lymphomas [12], but its function is largely unknown. The GCET2 gene is located on chromosome 3q13 and encodes a 178-amino acid (aa) protein with 51 % identity and 62 % similarity to the murine M17 protein [13]. GCET2 is a cytoplasmatic protein that may also associate with cell membrane. GCET2 expression is associated with improved survival in diffuse large B cell lymphoma (DLBCL) and classic Hodgkin lymphoma patients [14]. In vitro studies in human lymphocytes demonstrated that HGAL increased the binding of myosin to F-actin and inhibits the ability of myosin to translocate actin by reducing the maximal velocity of myosin head/actin movement [15]. In vitro HGAL enhances BCR signaling by binding and increasing Syk activation, in vivo older HGAL transgenic animals progressively developed polyclonal lymphoid hyperplasia and reactive AA amyloidosis [16], these finding suggests that GCET2 may play a role in humoral immune responses. No articles about expression and function of GCET2 on ovarian tissue have been published until now, and our findings make a possible insight of this gene in the study of ovarian cancer, especially about the aspects of metastasis and chemoresistance.
Cystic fibrosis transmembrane conductance regulator (CFTR, ABC35 or ABCC7) was found among the most up-regulated genes in more metastasis and chemoresistance cell line group, and it participates in the beta-adrenergic-dependent CFTR expression pathway. Loss of function mutations of this gene causes the autosomal recessive lethal disease cystic fibrosis (CF) and congenital bilateral aplasia of the vas deferens. There is an increasing interest in the association of cancer incidence with the genetic variations in the CFTR gene. Large cohort studies in North American and European patients with CF found that there was a marked increase in the risk of malignancies affecting the gastrointestinal tract, even to 17 times higher risk of digestive cancer with most cases arising in the bowel [17]. Meanwhile, mutations and low-penetrance polymorphisms in the CFTR gene have been found in patients with various cancers, including pancreatic cancer [18], breast cancer [19], cervical cancer [20], melanoma [21], prostate cancer [22] and lung cancer [23, 24]. On the other hand, CFTR has been suggested to interact with various cancer-related kinases [25]. It encodes a member of the ATP-binding cassette (ABC) transporter superfamily. ABC proteins transport various molecules across extracellular and intracellular membranes. ABC genes are divided into seven distinct subfamilies (ABC1, MDR/TAP, MRP, ALD, OABP, GCN20, White). Meanwhile, CFTR is a member of the MRP subfamily that is involved in multidrug resistance. The encoded protein is a cAMP-activated Cl channel lining the luminal/apical surfaces of epithelial cells in airway, gut, and exocrine glands, and there is a functional coupling between CFTR and MRP2 that may be mediated by PDZ protein [24]. Taken together, our gene expression profile that show a significant up-regulated result of CFTR in more metastasis and chemoresistant ovarian cancer cell lines are consistent with previous findings, a further research on the mechanism of CFTR on ovarian cancer or selective inhibition of CFTR or its pathway may give a insight in therapeutic effects against metastatic and chemoresistant of ovarian cancer.
RBMX gene, also known as HnRNP G, is a member of heterogeneous nuclear ribonucleoprotein (hnRNP) family and can collaborate with hTra2-beta1 (human transformer-2-beta1) as sequence-specific transacting factors to exert antagonistic effects on alternative splicing which is recognized as a pivotal mechanism in regulation of gene expression and associated to tumorigenesis and metastasis of a wide variety of human cancers [26]. It is proposed that the ratio of hnRNP G/hTra2-beta1 influenced cellular splicing preference [27, 28]. Some researches revealed that hnRNP G-protein showed as tumor suppressor in endometrial carcinoma [27] and oral squamous cancer [29], its activity was elicited by transactivating tumor suppressor Txnip gene [30]. A recent research showed that high frequency of hnRNP G-protein reduction and loss of expression in precancerous and human oral squamous cell carcinoma tissue specimens, suggesting that reduction in hnRNP G may play an important role in the early pathogenesis of oral squamous cell carcinomas [31].
GPRC6A encodes an orphan G-protein-coupled receptor, mediates the non-genomic effects of testosterone and other anabolic steroids in multiple tissue, and it is a potential target for developing antagonists and agonists that would have broad applications in health and disease [32], including cancer. A genome-wide association study on prostate cancer identified GPRC6A was one of the five novel genetic loci associated with prostate cancer in Japanese and Chinese Han population [33, 34], and the same result was verified by a genome-wide testing of putative functional exonic variants in a multiethnic population [35]. GPRC6A is expressed at higher levels in human prostate cancer cells and prostate cancer tissues and small interfering RNA knockdown of GPRC6A attenuates these response in human prostate cancer cell lines [36]. GPRC6A is also coupled to signaling pathways, such as phosphatidylinositol 3-kinase that are known to be deregulated in prostate cancer [32]. On all accounts, nearly all researches about GPRC6A associated with cancer focused on prostate cancer, no investigation about this gene on ovarian cancer has been published.
FOXP1, as one of the down-regulated genes, drew our attention for a further research. FOXP1 is a FOX family member consisting of the winged-helix DNA-binding domain and the N-terminal transcriptional repression domain, and it is widely expressed and plays a key role in the development of various human tissues [37, 38]. FOXP1 represses its target genes by forming homodimers or heterodimers with FOXP2 and FOXP4 [39], it has been suggested to be both a tumor suppressor candidate and potential oncogene, because of its differential expression levels in distinctive types of tumors, including B cell lymphomas [40], breast cancer [41, 42], endometrial cancer [43], prostate cancer [44], non-small cell lung cancer [38] and renal cell carcinoma [45], the loss of FOXP1 in breast cancer has been associated with shorter survival times [42]. Until now, no article about FOXP1 expression in ovarian cancer has been published, and we made the first investigation of FOXP1 protein expression in ovarian tissue and found that nuclear staining of FOXP1 decreased as the metastasis increased, a significant decrease in FOXP1 expression in the resistance group, nuclear FOXP1 expression were independent risk factors strongly correlated with prognosis of ovarian cancer, above all, FOXP1 may serve as a good marker for late stage ovarian cancer and chemoresistance EOC patients, high expression of FOXP1 in nucleus is associated with improved survival in patients with ovarian cancer.
There are some pseudogenes which shows significant expression difference in enhanced metastasis and chemoresistant ovarian cancer cell lines, in which BC031676 and BC113708 are up-regulated, and RPL28P1, RPL23A, RPL13AP3, LOC341412, LOC641784, LOC391560, RPS16P9, LOC732186, RPL13AP23, RPLP1P7, RPL31P10, LOC648361 are down-regulated, and most of them are ribosomal protein pseudogenes. Pseudogenes are DNA sequences similar to genes encoding functional proteins but are presumed to be nonfunctional due to mutations and truncation by premature stop codons [46]. Ribosomal protein (RP) pseudogenes constitute the largest family of pseudogenes (approximately 2000 RP processed pseudogenes), and they are constitutively expressed at reasonably stable levels and are very highly conserved [47]. Although pseudogenes have long been considered as nonfunctional genomic sequences, during recent two decades, especially with the broad applications of next-generation sequencing technologies, emerging evidences have confirmed that some pseudogenes have acquired diverse functions in regulating development and diseases, especially in cancers [48]. Some pseudogenes are specifically expressed in certain cancers or diseases. It has been shown that the pseudogene of PTEN, PTENP1, was selectively lost in some human cancer cells, resulting in decreased expression of PTEN and abnormal proliferation of cancer cells [49]. The expression of MYLKP1, a duplicated pseudogene of MYLK, can decrease the stability of MYLK mRNA at the posttranscriptional level and stimulate cell proliferation [50]. Recently, a study provided a systematic approach to analyze expressed pseudogenes, enabling comparisons of cancer versus benign tissues in multiple solid tumors, which overcome the limitations of previous analyses of pseudogene expression. They observed 218 pseudogenes expressed only in cancer samples, of which 178 were observed in multiple cancers, and 40 were found to have highly specific expression in a single cancer type only, finally they described ATP8A2-J and CXADR-J pseudogenes preferentially associated with distinct subsets of breast cancer and prostate cancer patients, respectively [51]. Besides cancers, pseudogenes also involve in the development of other diseases, such as HMGA1-p [52]. Although the regulatory functions of pseudogenes seem to be striking, the functional studies of pseudogenes are still in its early stage. The pseudogenes in our study should not be useless, their functions and relationships with ovarian cancer, especially the enhanced metastasis and chemoresistance should be investigated in near further.
In conclusion, this study has identified potential DEGs responsible for enhanced metastasis and chemoresistance in ovarian cancer cell lines. Among the 49 DEGs, 14 genes were up-regulated and 35 genes were down-regulated. Prospective investigations using a combination of genomic and proteomic approaches are required to validate the functionality of these targets identified.

Acknowledgments

This work was supported by the National Natural Science Foundation of China (Grant Nos.: 81072118, 81172491, 81101527), the Education Department Doctor Project Fund (Grant Nos.: 20112104110016, 20112104120019) and the Free Researchers’ Plan of Shengjing Hospital Affiliated to China Medical University (Grant No. 200807).

Conflict of interest

The authors declare no conflict of interest.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution License which permits any use, distribution, and reproduction in any medium, provided the original author(s) and the source are credited.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Liu CM. Cancer of the ovary. N Engl J Med. 2005;352:1268–9; author reply-9. Liu CM. Cancer of the ovary. N Engl J Med. 2005;352:1268–9; author reply-9.
2.
Zurück zum Zitat Iwamori M, Tanaka K, Kubushiro K, Lin B, Kiguchi K, Ishiwata I, et al. Alterations in the glycolipid composition and cellular properties of ovarian carcinoma-derived RMG-1 cells on transfection of the alpha1,2-fucosyltransferase gene. Cancer Sci. 2005;96:26–30.PubMedCrossRef Iwamori M, Tanaka K, Kubushiro K, Lin B, Kiguchi K, Ishiwata I, et al. Alterations in the glycolipid composition and cellular properties of ovarian carcinoma-derived RMG-1 cells on transfection of the alpha1,2-fucosyltransferase gene. Cancer Sci. 2005;96:26–30.PubMedCrossRef
3.
Zurück zum Zitat Lin B, Hao YY, Wang DD, Zhu LC, Zhang SL, Saito M, et al. Transfection of alpha1, 2-fucosyltransferase gene increases the antigenic expression of Lewis y in ovarian cancer cell line RMG-I. Zhongguo yi xue ke xue yuan xue bao Acta Academiae Medicinae Sinicae. 2008;30:284–9.PubMed Lin B, Hao YY, Wang DD, Zhu LC, Zhang SL, Saito M, et al. Transfection of alpha1, 2-fucosyltransferase gene increases the antigenic expression of Lewis y in ovarian cancer cell line RMG-I. Zhongguo yi xue ke xue yuan xue bao Acta Academiae Medicinae Sinicae. 2008;30:284–9.PubMed
4.
Zurück zum Zitat Liu JJ, Lin B, Hao YY, Li FF, Liu DW, Qi Y, et al. Lewis(y) antigen stimulates the growth of ovarian cancer cells via regulation of the epidermal growth factor receptor pathway. Oncol Rep. 2010;23:833–41.PubMedCrossRef Liu JJ, Lin B, Hao YY, Li FF, Liu DW, Qi Y, et al. Lewis(y) antigen stimulates the growth of ovarian cancer cells via regulation of the epidermal growth factor receptor pathway. Oncol Rep. 2010;23:833–41.PubMedCrossRef
5.
Zurück zum Zitat Yan LM, Lin B, Zhu LC, Hao YY, Qi Y, Wang CZ, et al. Enhancement of the adhesive and spreading potentials of ovarian carcinoma RMG-1 cells due to increased expression of integrin alpha5beta1 with the Lewis Y-structure on transfection of the alpha1,2-fucosyltransferase gene. Biochimie. 2010;92:852–7.PubMedCrossRef Yan LM, Lin B, Zhu LC, Hao YY, Qi Y, Wang CZ, et al. Enhancement of the adhesive and spreading potentials of ovarian carcinoma RMG-1 cells due to increased expression of integrin alpha5beta1 with the Lewis Y-structure on transfection of the alpha1,2-fucosyltransferase gene. Biochimie. 2010;92:852–7.PubMedCrossRef
6.
Zurück zum Zitat Gao S, Liu Q, Wang X, Lin B, Zhang S. Effects of Lewis Y antigen on the gene expression of multiple drug resistance-associated proteins in human ovarian cancer RMG-I-H cells. Med Oncol. 2010;27:960–7.PubMedCrossRef Gao S, Liu Q, Wang X, Lin B, Zhang S. Effects of Lewis Y antigen on the gene expression of multiple drug resistance-associated proteins in human ovarian cancer RMG-I-H cells. Med Oncol. 2010;27:960–7.PubMedCrossRef
7.
Zurück zum Zitat Shenhua X, Lijuan Q, Hanzhou N, Xinghao N, Chihong Z, Gu Z, et al. Establishment of a highly metastatic human ovarian cancer cell line (HO-8910PM) and its characterization. J Exp Clin Cancer Res CR. 1999;18:233–9. Shenhua X, Lijuan Q, Hanzhou N, Xinghao N, Chihong Z, Gu Z, et al. Establishment of a highly metastatic human ovarian cancer cell line (HO-8910PM) and its characterization. J Exp Clin Cancer Res CR. 1999;18:233–9.
8.
Zurück zum Zitat Gao J, Hu Z, Liu D, Liu J, Liu C, Hou R, et al. Expression of Lewis y antigen and integrin alphav, beta3 in ovarian cancer and their relationship with chemotherapeutic drug resistance. J Exp Clin Cancer Res CR. 2013;32:36.CrossRef Gao J, Hu Z, Liu D, Liu J, Liu C, Hou R, et al. Expression of Lewis y antigen and integrin alphav, beta3 in ovarian cancer and their relationship with chemotherapeutic drug resistance. J Exp Clin Cancer Res CR. 2013;32:36.CrossRef
9.
Zurück zum Zitat Hu Z, Gao S, Gao J, Hou R, Liu C, Liu J, et al. Elevated levels of Lewis y and integrin alpha5beta1 correlate with chemotherapeutic drug resistance in epithelial ovarian carcinoma. Int J Mol Sci. 2012;13:15588–600.PubMedCentralPubMedCrossRef Hu Z, Gao S, Gao J, Hou R, Liu C, Liu J, et al. Elevated levels of Lewis y and integrin alpha5beta1 correlate with chemotherapeutic drug resistance in epithelial ovarian carcinoma. Int J Mol Sci. 2012;13:15588–600.PubMedCentralPubMedCrossRef
10.
11.
Zurück zum Zitat Diaz-Montana JJ, Diaz-Diaz N. Development and use of the Cytoscape app GFD-Net for measuring semantic dissimilarity of gene networks. F1000 Res. 2014;3:142. Diaz-Montana JJ, Diaz-Diaz N. Development and use of the Cytoscape app GFD-Net for measuring semantic dissimilarity of gene networks. F1000 Res. 2014;3:142.
12.
Zurück zum Zitat Pan Z, Shen Y, Ge B, Du C, McKeithan T, Chan WC. Studies of a germinal centre B-cell expressed gene, GCET2, suggest its role as a membrane associated adapter protein. Br J Haematol. 2007;137:578–90.PubMedCentralPubMedCrossRef Pan Z, Shen Y, Ge B, Du C, McKeithan T, Chan WC. Studies of a germinal centre B-cell expressed gene, GCET2, suggest its role as a membrane associated adapter protein. Br J Haematol. 2007;137:578–90.PubMedCentralPubMedCrossRef
13.
Zurück zum Zitat Schenten D, Egert A, Pasparakis M, Rajewsky K. M17, a gene specific for germinal center (GC) B cells and a prognostic marker for GC B-cell lymphomas, is dispensable for the GC reaction in mice. Blood. 2006;107:4849–56.PubMedCentralPubMedCrossRef Schenten D, Egert A, Pasparakis M, Rajewsky K. M17, a gene specific for germinal center (GC) B cells and a prognostic marker for GC B-cell lymphomas, is dispensable for the GC reaction in mice. Blood. 2006;107:4849–56.PubMedCentralPubMedCrossRef
14.
Zurück zum Zitat Natkunam Y, Lossos IS, Taidi B, Zhao S, Lu X, Ding F, et al. Expression of the human germinal center-associated lymphoma (HGAL) protein, a new marker of germinal center B-cell derivation. Blood. 2005;105:3979–86.PubMedCentralPubMedCrossRef Natkunam Y, Lossos IS, Taidi B, Zhao S, Lu X, Ding F, et al. Expression of the human germinal center-associated lymphoma (HGAL) protein, a new marker of germinal center B-cell derivation. Blood. 2005;105:3979–86.PubMedCentralPubMedCrossRef
15.
Zurück zum Zitat Lu X, Kazmierczak K, Jiang X, Jones M, Watt J, Helfman DM, et al. Germinal center-specific protein human germinal center associated lymphoma directly interacts with both myosin and actin and increases the binding of myosin to actin. FEBS J. 2011;278:1922–31.PubMedCentralPubMedCrossRef Lu X, Kazmierczak K, Jiang X, Jones M, Watt J, Helfman DM, et al. Germinal center-specific protein human germinal center associated lymphoma directly interacts with both myosin and actin and increases the binding of myosin to actin. FEBS J. 2011;278:1922–31.PubMedCentralPubMedCrossRef
16.
Zurück zum Zitat Romero-Camarero I, Jiang X, Natkunam Y, Lu X, Vicente-Duenas C, Gonzalez-Herrero I, et al. Germinal centre protein HGAL promotes lymphoid hyperplasia and amyloidosis via BCR-mediated Syk activation. Nat Commun. 2013;4:1338.PubMedCentralPubMedCrossRef Romero-Camarero I, Jiang X, Natkunam Y, Lu X, Vicente-Duenas C, Gonzalez-Herrero I, et al. Germinal centre protein HGAL promotes lymphoid hyperplasia and amyloidosis via BCR-mediated Syk activation. Nat Commun. 2013;4:1338.PubMedCentralPubMedCrossRef
17.
Zurück zum Zitat Maisonneuve P, Marshall BC, Knapp EA, Lowenfels AB. Cancer risk in cystic fibrosis: a 20-year nationwide study from the United States. J Natl Cancer Inst. 2013;105:122–9.PubMedCrossRef Maisonneuve P, Marshall BC, Knapp EA, Lowenfels AB. Cancer risk in cystic fibrosis: a 20-year nationwide study from the United States. J Natl Cancer Inst. 2013;105:122–9.PubMedCrossRef
18.
Zurück zum Zitat McWilliams R, Highsmith WE, Rabe KG, de Andrade M, Tordsen LA, Holtegaard LM, et al. Cystic fibrosis transmembrane regulator gene carrier status is a risk factor for young onset pancreatic adenocarcinoma. Gut. 2005;54:1661–2.PubMedCentralPubMedCrossRef McWilliams R, Highsmith WE, Rabe KG, de Andrade M, Tordsen LA, Holtegaard LM, et al. Cystic fibrosis transmembrane regulator gene carrier status is a risk factor for young onset pancreatic adenocarcinoma. Gut. 2005;54:1661–2.PubMedCentralPubMedCrossRef
19.
Zurück zum Zitat Zhang JT, Jiang XH, Xie C, Cheng H, Da Dong J, Wang Y, et al. Downregulation of CFTR promotes epithelial-to-mesenchymal transition and is associated with poor prognosis of breast cancer. Biochim Biophys Acta. 2013;1833:2961–9.PubMedCrossRef Zhang JT, Jiang XH, Xie C, Cheng H, Da Dong J, Wang Y, et al. Downregulation of CFTR promotes epithelial-to-mesenchymal transition and is associated with poor prognosis of breast cancer. Biochim Biophys Acta. 2013;1833:2961–9.PubMedCrossRef
20.
Zurück zum Zitat Peng X, Wu Z, Yu L, Li J, Xu W, Chan HC, et al. Overexpression of cystic fibrosis transmembrane conductance regulator (CFTR) is associated with human cervical cancer malignancy, progression and prognosis. Gynecol Oncol. 2012;125:470–6.PubMedCrossRef Peng X, Wu Z, Yu L, Li J, Xu W, Chan HC, et al. Overexpression of cystic fibrosis transmembrane conductance regulator (CFTR) is associated with human cervical cancer malignancy, progression and prognosis. Gynecol Oncol. 2012;125:470–6.PubMedCrossRef
21.
Zurück zum Zitat Warren N, Holmes JA, Al-Jader L, West RR, Lewis DC, Padua RA. Frequency of carriers of cystic fibrosis gene among patients with myeloid malignancy and melanoma. BMJ (Clinical research ed). 1991;302:760–1.CrossRef Warren N, Holmes JA, Al-Jader L, West RR, Lewis DC, Padua RA. Frequency of carriers of cystic fibrosis gene among patients with myeloid malignancy and melanoma. BMJ (Clinical research ed). 1991;302:760–1.CrossRef
22.
Zurück zum Zitat Qiao D, Yi L, Hua L, Xu Z, Ding Y, Shi D, et al. Cystic fibrosis transmembrane conductance regulator (CFTR) gene 5T allele may protect against prostate cancer: a case-control study in Chinese Han population. J Cyst Fibros. 2008;7:210–4.PubMedCrossRef Qiao D, Yi L, Hua L, Xu Z, Ding Y, Shi D, et al. Cystic fibrosis transmembrane conductance regulator (CFTR) gene 5T allele may protect against prostate cancer: a case-control study in Chinese Han population. J Cyst Fibros. 2008;7:210–4.PubMedCrossRef
23.
Zurück zum Zitat Li Y, Sun Z, Wu Y, Babovic-Vuksanovic D, Li Y, Cunningham JM, et al. Cystic fibrosis transmembrane conductance regulator gene mutation and lung cancer risk. Lung Cancer (Amsterdam, Netherlands). 2010;70:14–21.CrossRef Li Y, Sun Z, Wu Y, Babovic-Vuksanovic D, Li Y, Cunningham JM, et al. Cystic fibrosis transmembrane conductance regulator gene mutation and lung cancer risk. Lung Cancer (Amsterdam, Netherlands). 2010;70:14–21.CrossRef
24.
Zurück zum Zitat Li C, Schuetz JD, Naren AP. Tobacco carcinogen NNK transporter MRP2 regulates CFTR function in lung epithelia: implications for lung cancer. Cancer Lett. 2010;292:246–53.PubMedCentralPubMedCrossRef Li C, Schuetz JD, Naren AP. Tobacco carcinogen NNK transporter MRP2 regulates CFTR function in lung epithelia: implications for lung cancer. Cancer Lett. 2010;292:246–53.PubMedCentralPubMedCrossRef
25.
Zurück zum Zitat Pagano MA, Tibaldi E, Gringeri E, Brunati AM. Tyrosine phosphorylation and liver regeneration: a glance at intracellular transducers. IUBMB Life. 2012;64:27–35.PubMedCrossRef Pagano MA, Tibaldi E, Gringeri E, Brunati AM. Tyrosine phosphorylation and liver regeneration: a glance at intracellular transducers. IUBMB Life. 2012;64:27–35.PubMedCrossRef
26.
Zurück zum Zitat Fischer DC, Noack K, Runnebaum IB, Watermann DO, Kieback DG, Stamm S, et al. Expression of splicing factors in human ovarian cancer. Oncol Rep. 2004;11:1085–90.PubMed Fischer DC, Noack K, Runnebaum IB, Watermann DO, Kieback DG, Stamm S, et al. Expression of splicing factors in human ovarian cancer. Oncol Rep. 2004;11:1085–90.PubMed
27.
Zurück zum Zitat Ouyang YQ, zur Hausen A, Orlowska-Volk M, Jager M, Bettendorf H, Hirschfeld M, et al. Expression levels of hnRNP G and hTra2-beta1 correlate with opposite outcomes in endometrial cancer biology. Int J Cancer J Int du Cancer. 2011;128:2010–9.CrossRef Ouyang YQ, zur Hausen A, Orlowska-Volk M, Jager M, Bettendorf H, Hirschfeld M, et al. Expression levels of hnRNP G and hTra2-beta1 correlate with opposite outcomes in endometrial cancer biology. Int J Cancer J Int du Cancer. 2011;128:2010–9.CrossRef
28.
Zurück zum Zitat Nasim MT, Chernova TK, Chowdhury HM, Yue BG, Eperon IC. HnRNP G and Tra2beta: opposite effects on splicing matched by antagonism in RNA binding. Hum Mol Genet. 2003;12:1337–48.PubMedCrossRef Nasim MT, Chernova TK, Chowdhury HM, Yue BG, Eperon IC. HnRNP G and Tra2beta: opposite effects on splicing matched by antagonism in RNA binding. Hum Mol Genet. 2003;12:1337–48.PubMedCrossRef
29.
Zurück zum Zitat Shin KH, Kang MK, Kim RH, Christensen R, Park NH. Heterogeneous nuclear ribonucleoprotein G shows tumor suppressive effect against oral squamous cell carcinoma cells. Clin Cancer Res. 2006;12:3222–8.PubMedCrossRef Shin KH, Kang MK, Kim RH, Christensen R, Park NH. Heterogeneous nuclear ribonucleoprotein G shows tumor suppressive effect against oral squamous cell carcinoma cells. Clin Cancer Res. 2006;12:3222–8.PubMedCrossRef
30.
Zurück zum Zitat Shin KH, Kim RH, Kim RH, Kang MK, Park NH. hnRNP G elicits tumor-suppressive activity in part by upregulating the expression of Txnip. Biochem Biophys Res Commun. 2008;372:880–5.PubMedCentralPubMedCrossRef Shin KH, Kim RH, Kim RH, Kang MK, Park NH. hnRNP G elicits tumor-suppressive activity in part by upregulating the expression of Txnip. Biochem Biophys Res Commun. 2008;372:880–5.PubMedCentralPubMedCrossRef
31.
Zurück zum Zitat Shin KH, Kim RH, Yu B, Kang MK, Elashoff D, Christensen R, et al. Expression and mutation analysis of heterogeneous nuclear ribonucleoprotein G in human oral cancer. Oral Oncol. 2011;47:1011–6.PubMedCentralPubMedCrossRef Shin KH, Kim RH, Yu B, Kang MK, Elashoff D, Christensen R, et al. Expression and mutation analysis of heterogeneous nuclear ribonucleoprotein G in human oral cancer. Oral Oncol. 2011;47:1011–6.PubMedCentralPubMedCrossRef
33.
Zurück zum Zitat Takata R, Akamatsu S, Kubo M, Takahashi A, Hosono N, Kawaguchi T, et al. Genome-wide association study identifies five new susceptibility loci for prostate cancer in the Japanese population. Nat Genet. 2010;42:751–4.PubMedCrossRef Takata R, Akamatsu S, Kubo M, Takahashi A, Hosono N, Kawaguchi T, et al. Genome-wide association study identifies five new susceptibility loci for prostate cancer in the Japanese population. Nat Genet. 2010;42:751–4.PubMedCrossRef
34.
Zurück zum Zitat Long QZ, Du YF, Ding XY, Li X, Song WB, Yang Y, et al. Replication and fine mapping for association of the C2orf43, FOXP4, GPRC6A and RFX6 genes with prostate cancer in the Chinese population. PLoS One. 2012;7:e37866.PubMedCentralPubMedCrossRef Long QZ, Du YF, Ding XY, Li X, Song WB, Yang Y, et al. Replication and fine mapping for association of the C2orf43, FOXP4, GPRC6A and RFX6 genes with prostate cancer in the Chinese population. PLoS One. 2012;7:e37866.PubMedCentralPubMedCrossRef
35.
Zurück zum Zitat Haiman CA, Han Y, Feng Y, Xia L, Hsu C, Sheng X, et al. Genome-wide testing of putative functional exonic variants in relationship with breast and prostate cancer risk in a multiethnic population. PLoS Genet. 2013;9:e1003419.PubMedCentralPubMedCrossRef Haiman CA, Han Y, Feng Y, Xia L, Hsu C, Sheng X, et al. Genome-wide testing of putative functional exonic variants in relationship with breast and prostate cancer risk in a multiethnic population. PLoS Genet. 2013;9:e1003419.PubMedCentralPubMedCrossRef
37.
Zurück zum Zitat Green MR, Gandhi MK, Courtney MJ, Marlton P, Griffiths L. Relative abundance of full-length and truncated FOXP1 isoforms is associated with differential NFkappaB activity in Follicular Lymphoma. Leuk Res. 2009;33:1699–702.PubMedCrossRef Green MR, Gandhi MK, Courtney MJ, Marlton P, Griffiths L. Relative abundance of full-length and truncated FOXP1 isoforms is associated with differential NFkappaB activity in Follicular Lymphoma. Leuk Res. 2009;33:1699–702.PubMedCrossRef
38.
Zurück zum Zitat Feng J, Zhang X, Zhu H, Wang X, Ni S, Huang J. High expression of FoxP1 is associated with improved survival in patients with non-small cell lung cancer. Am J Clin Pathol. 2012;138:230–5.PubMedCrossRef Feng J, Zhang X, Zhu H, Wang X, Ni S, Huang J. High expression of FoxP1 is associated with improved survival in patients with non-small cell lung cancer. Am J Clin Pathol. 2012;138:230–5.PubMedCrossRef
39.
Zurück zum Zitat Santos ME, Athanasiadis A, Leitao AB, DuPasquier L, Sucena E. Alternative splicing and gene duplication in the evolution of the FoxP gene subfamily. Mol Biol Evol. 2011;28:237–47.PubMedCentralPubMedCrossRef Santos ME, Athanasiadis A, Leitao AB, DuPasquier L, Sucena E. Alternative splicing and gene duplication in the evolution of the FoxP gene subfamily. Mol Biol Evol. 2011;28:237–47.PubMedCentralPubMedCrossRef
40.
Zurück zum Zitat Hoeller S, Schneider A, Haralambieva E, Dirnhofer S, Tzankov A. FOXP1 protein overexpression is associated with inferior outcome in nodal diffuse large B-cell lymphomas with non-germinal centre phenotype, independent of gains and structural aberrations at 3p14.1. Histopathology. 2010;57:73–80.PubMedCrossRef Hoeller S, Schneider A, Haralambieva E, Dirnhofer S, Tzankov A. FOXP1 protein overexpression is associated with inferior outcome in nodal diffuse large B-cell lymphomas with non-germinal centre phenotype, independent of gains and structural aberrations at 3p14.1. Histopathology. 2010;57:73–80.PubMedCrossRef
41.
Zurück zum Zitat Ijichi N, Ikeda K, Horie-Inoue K, Inoue S. FOXP1 and estrogen signaling in breast cancer. Vitam Horm. 2013;93:203–12.PubMedCrossRef Ijichi N, Ikeda K, Horie-Inoue K, Inoue S. FOXP1 and estrogen signaling in breast cancer. Vitam Horm. 2013;93:203–12.PubMedCrossRef
42.
Zurück zum Zitat Bates GJ, Fox SB, Han C, Launchbury R, Leek RD, Harris AL, et al. Expression of the forkhead transcription factor FOXP1 is associated with that of estrogen receptor-beta in primary invasive breast carcinomas. Breast Cancer Res Treat. 2008;111:453–9.PubMedCrossRef Bates GJ, Fox SB, Han C, Launchbury R, Leek RD, Harris AL, et al. Expression of the forkhead transcription factor FOXP1 is associated with that of estrogen receptor-beta in primary invasive breast carcinomas. Breast Cancer Res Treat. 2008;111:453–9.PubMedCrossRef
43.
Zurück zum Zitat Giatromanolaki A, Koukourakis MI, Sivridis E, Gatter KC, Harris AL, Banham AH. Loss of expression and nuclear/cytoplasmic localization of the FOXP1 forkhead transcription factor are common events in early endometrial cancer: relationship with estrogen receptors and HIF-1alpha expression. Mod Pathol. 2006;19:9–16.PubMedCrossRef Giatromanolaki A, Koukourakis MI, Sivridis E, Gatter KC, Harris AL, Banham AH. Loss of expression and nuclear/cytoplasmic localization of the FOXP1 forkhead transcription factor are common events in early endometrial cancer: relationship with estrogen receptors and HIF-1alpha expression. Mod Pathol. 2006;19:9–16.PubMedCrossRef
44.
Zurück zum Zitat Takayama K, Horie-Inoue K, Ikeda K, Urano T, Murakami K, Hayashizaki Y, et al. FOXP1 is an androgen-responsive transcription factor that negatively regulates androgen receptor signaling in prostate cancer cells. Biochem Biophys Res Commun. 2008;374:388–93.PubMedCrossRef Takayama K, Horie-Inoue K, Ikeda K, Urano T, Murakami K, Hayashizaki Y, et al. FOXP1 is an androgen-responsive transcription factor that negatively regulates androgen receptor signaling in prostate cancer cells. Biochem Biophys Res Commun. 2008;374:388–93.PubMedCrossRef
45.
Zurück zum Zitat Toma MI, Weber T, Meinhardt M, Zastrow S, Grimm MO, Fussel S, et al. Expression of the Forkhead transcription factor FOXP1 is associated with tumor grade and Ki67 expression in clear cell renal cell carcinoma. Cancer Invest. 2011;29:123–9.PubMedCrossRef Toma MI, Weber T, Meinhardt M, Zastrow S, Grimm MO, Fussel S, et al. Expression of the Forkhead transcription factor FOXP1 is associated with tumor grade and Ki67 expression in clear cell renal cell carcinoma. Cancer Invest. 2011;29:123–9.PubMedCrossRef
46.
Zurück zum Zitat Pink RC, Wicks K, Caley DP, Punch EK, Jacobs L, Carter DR. Pseudogenes pseudo-functional or key regulators in health and disease. RNA (New York, NY). 2011;17:792–8.CrossRef Pink RC, Wicks K, Caley DP, Punch EK, Jacobs L, Carter DR. Pseudogenes pseudo-functional or key regulators in health and disease. RNA (New York, NY). 2011;17:792–8.CrossRef
47.
Zurück zum Zitat Balasubramanian S, Zheng D, Liu YJ, Fang G, Frankish A, Carriero N, et al. Comparative analysis of processed ribosomal protein pseudogenes in four mammalian genomes. Genome Biol. 2009;10:R2.PubMedCentralPubMedCrossRef Balasubramanian S, Zheng D, Liu YJ, Fang G, Frankish A, Carriero N, et al. Comparative analysis of processed ribosomal protein pseudogenes in four mammalian genomes. Genome Biol. 2009;10:R2.PubMedCentralPubMedCrossRef
48.
Zurück zum Zitat Li W, Yang W, Wang XJ. Pseudogenes: pseudo or real functional elements? J Genet Genom. 2013;40:171–7.CrossRef Li W, Yang W, Wang XJ. Pseudogenes: pseudo or real functional elements? J Genet Genom. 2013;40:171–7.CrossRef
49.
Zurück zum Zitat Poliseno L, Salmena L, Zhang J, Carver B, Haveman WJ, Pandolfi PP. A coding-independent function of gene and pseudogene mRNAs regulates tumour biology. Nature. 2010;465:1033–8.PubMedCentralPubMedCrossRef Poliseno L, Salmena L, Zhang J, Carver B, Haveman WJ, Pandolfi PP. A coding-independent function of gene and pseudogene mRNAs regulates tumour biology. Nature. 2010;465:1033–8.PubMedCentralPubMedCrossRef
50.
Zurück zum Zitat Han YJ, Ma SF, Yourek G, Park YD, Garcia JG. A transcribed pseudogene of MYLK promotes cell proliferation. FASEB J. 2011;25:2305–12.PubMedCrossRef Han YJ, Ma SF, Yourek G, Park YD, Garcia JG. A transcribed pseudogene of MYLK promotes cell proliferation. FASEB J. 2011;25:2305–12.PubMedCrossRef
51.
Zurück zum Zitat Kalyana-Sundaram S, Kumar-Sinha C, Shankar S, Robinson DR, Wu YM, Cao X, et al. Expressed pseudogenes in the transcriptional landscape of human cancers. Cell. 2012;149:1622–34.PubMedCentralPubMedCrossRef Kalyana-Sundaram S, Kumar-Sinha C, Shankar S, Robinson DR, Wu YM, Cao X, et al. Expressed pseudogenes in the transcriptional landscape of human cancers. Cell. 2012;149:1622–34.PubMedCentralPubMedCrossRef
52.
Zurück zum Zitat Chiefari E, Iiritano S, Paonessa F, Le Pera I, Arcidiacono B, Filocamo M, et al. Pseudogene-mediated posttranscriptional silencing of HMGA1 can result in insulin resistance and type 2 diabetes. Nature Commun. 2010;1:40.CrossRef Chiefari E, Iiritano S, Paonessa F, Le Pera I, Arcidiacono B, Filocamo M, et al. Pseudogene-mediated posttranscriptional silencing of HMGA1 can result in insulin resistance and type 2 diabetes. Nature Commun. 2010;1:40.CrossRef
Metadaten
Titel
Gene expression profile analysis identifies metastasis and chemoresistance-associated genes in epithelial ovarian carcinoma cells
verfasst von
Liancheng Zhu
Zhenhua Hu
Juanjuan Liu
Jian Gao
Bei Lin
Publikationsdatum
01.01.2015
Verlag
Springer US
Erschienen in
Medical Oncology / Ausgabe 1/2015
Print ISSN: 1357-0560
Elektronische ISSN: 1559-131X
DOI
https://doi.org/10.1007/s12032-014-0426-5

Weitere Artikel der Ausgabe 1/2015

Medical Oncology 1/2015 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.