Microarray Analysis Identifies Differentially

Embed Size (px)

Citation preview

  • 8/17/2019 Microarray Analysis Identifies Differentially

    1/17Copyright @ 2009 by IGCS and ESGO. Unauthorized reproduction of this article is prohibited.

    Microarray Analysis Identifies DifferentiallyExpressed Genes Induced by Human Papillomavirus

    Type 18  E6  Silencing RNA 

    Wei Min, PhD,* Ma Wen-li, MD, PhD,* Sun Zhao-hui, PhD,Þ  Li Ling, PhD,* Zhang Bao, PhD,*

    and Zheng Wen-ling, MD, PhDþ

    Abstract:   The oncoprotein E6 of high-risk human papillomavirus (HPV) types promotes

    cell proliferation and contributes to carcinogenesis of HPV-positive cervical cancer cells. In

    this study, we used small interfering RNA (siRNA) technology to silence the  E6  gene in

    HPV-18 Y transformed human cervical cell line HeLa and determined the effects of  E6  gene

    knockdown on the cell by using microarray-based gene expression profiling coupled 

    with gene functional classification with bioinformatics methods. Silencing RNA prepared by

    siRNA expression cassettes against HPV-18   E6  gene could significantly inhibit  E6   gene

    expression and induce HeLa cells to apoptosis. The microarray analysis identified 359

    differentially expressed genes containing 307 up-regulated and 52 down-regulated genes. We

    analyzed the gene functions andcellular pathways in detail, including cell cycle Y related genes,

    CCNG1  and  p21; apoptosis-related genes,  CASP4,  CASP6 ,  IGFBP3, and  DFFA; ubiquitin

     proteolysis pathway Y related genes,   UBE3A   and   UBE2C ; keratinocyte differentiation Y 

    related genes, KRT4, KRT6E , and  KRT18; and antioncogenes, RECK  and  VEL . In addition,

    it can be concluded that cellular apoptosis induced by HPV-18 E6 siRNA mainly depends on

    the P53 and ubiquitin proteolysis pathway to regulate gene expression, consequently

    inhibiting cell proliferation and promoting cell apoptosis. Meanwhile, activation of 

    antioncogene and upper regulation of immunization-related genes signified the degression

    of the malignant extent of tumor cells after  E6 inhibition. Our approach, which combines the

    use of siRNA-mediated gene silencing, microarray screening, and functional classificationof differential genes, can be used in functional genomics study to elucidate the role of  E6 

    oncogene in the carcinogenesis of HPV-18 and provide some possible targets for clinical

    treatment and drug development of cervical cancer.

    Key Words:   siRNA, Microarray, HPV, E6

    ( Int J Gynecol Cancer  2009;19: 547 Y 563)

    Cervical cancer is one of the most common malignancies and the second leading cause of cancer mortality in women world-wide. Nearly 400,000 cases were diagnosed annually worldwide,80% of which occurred in developing countries.1 During the past 20 years, the links between genital human papillomavirus (HPV)

    infections and cervical cancer have been identified.

    2

    It is now agreed 

    universally that HPV causes virtually all cervical cancers.3 To date,more than 85 different HPV types have been identified, and amongthem, more than 20 high-risk HPV (HR-HPV) types have an estab-lished risk to develop into squamous cell carcinoma.4 Among theseHR-HPV types, HPV-16 and HPV-18 are found in 60% to 70% of 

    invasive cervical carcinomas, with the HPV-31, HPV-58, and HPV-52 types causing the remaining cases.5,6 Some studies have reported that patients infected with HPV-18 had worse prognoses and higher rates of disease recurrence than patients infected with HPV-16.7 Y 13

    A possible difference might as well be caused by adenocarcinomas,which are more frequently associated with HPV-18, that might havea worse prognosis.

    The transforming activity of tumor-associated HR-HPVtypes, such as HPV-16 and HPV-18, is dependent on the functionsof the viral E6  and  E7  oncogenes, and continuous E6/E7 expressionis required for the maintenance of the transformed phenotype of HPV-positive cervical cancer cells.14 E6 protein binds a number of cellular proteins including the E6-associated protein (E6AP), a protein ligase of the ubiquitin proteolysis pathway. E6 complexed 

    ORIGINAL  ARTICLE

    International Journal of Gynecological Cancer    &  Volume 19, Number 4, May 2009   547

    *Institute of Molecular Biology, Southern Medical University;  †Department of Clinical Laboratory, Guangzhou Liuhuaqiao Hospital; and   ‡SouthernChina Genomics Research Center, Guangzhou, People’s Republic of China.Address correspondence and reprint requests to Ma Wen-li, Institute of 

    Molecular Biology, Southern Medical University, Guangzhou 510515,People’s Republic of China. E-mail: [email protected].

    This work was supported by the Natural Science Foundation of GuangdongProvince, China (grant No. D07300239), and the Medical ScientificResearch Foundation of Guangdong Province, China (grant 

     No.WSTJJ20071201360103197802270749).Copyright  *  2009 by IGCS and ESGOISSN: 1048-891XDOI: 10.1111/IGC.0b013e3181a44c68

  • 8/17/2019 Microarray Analysis Identifies Differentially

    2/17Copyright @ 2009 by IGCS and ESGO. Unauthorized reproduction of this article is prohibited.

    with E6AP targets the P53 tumor suppressor protein for proteasomedegradation, resulting in genetic instability and progression toward malignancy.15 In contrast, HPV E7 is known to bind the transcrip-tional repressor hypophosphorylated retinoblastoma protein, caus-ing increased levels of active E2F transcription factors.16 This resultsin increased expression of genes involved in cell cycle progression

    and DNA synthesis.Studies have demonstrated that the expression of HPV E6

     protein is indispensable for tumor development and maintenanceof malignant phenotypes. Thus,   E6  oncogene is an ideal target of gene-specific therapy for cervical cancer. Previous studies havedemonstrated that the attempts of targeting E6/E7 by therapeuticnucleic acids including antisense RNA and oligonucleotides re-sulted in a several-fold inhibition of proliferation in cervical cancer cell lines,17 Y 19 and now, combination of the small interfering RNA(siRNA) and microarray technologies can provide a powerful toolto analyze comprehensively viral pathogenicity-related genes.

    Expression profile microarray technology should provide auseful experimental strategy to define cellular target genes for HPV-induced cell transformation by identifying transcriptionally altered genes upon silencing of endogenous E6 expression in HPV-positive

    cancer cells. Combining microarray with bioinformatics technologyto analyze the gene expression patterns of various tumors is an im- portant method in functional genomic studies. It surpasses the solegene research pattern obviously and may illuminate the gene expres-sion and regulation network of tumor cells in the whole.

    The present work used siRNA technology to silence E6  genein HPV-18 Y transformed human cervical cell line HeLa and deter-mined the effection of   E6   gene knockdown on the cell by usingmicroarray-based gene expression profiling coupled with gene func-tional classification with bioinformatics methods.

    MATERIALS AND METHODS

    Silencing RNA Expression Cassettes

    Constructed by Polymerase Chain ReactionSilencing RNAs targeting HPV-18   E6   gene were designed and synthesized according to the manufacturer’s instruction of LineSilence Complete RNAi Kit (Allele Biotechnology). The tar-get sequence for HPV-18   E6   was nucleotide site 340-358 (5¶-GACATTATTCAGACTCTGT-3¶). In addition, the negative controlmessenger-silencing RNA (m-siRNA) has the same base composi-tion but confused sequences of targeting   E6   siRNA. Both siRNAsequences were screened against the human genome by using a basiclocal alignment search tool homology search to avoid unintentionalsilencing of host cell genes. Polymerase chain reactions (PCRs)were performed using a plasmid containing the human U6 promoter as template. The upstream primer (5¶U6 universal primer) is comple-mentary to 29 nucleotides (nt) at the 5 ¶ end of the U6 promoter. Thedownstream primer (3¶U6 universal primer) contains a U6 termina-

    tor, a sense or antisense siRNA sequence, and a complementary se-quence to the last 20 nt of the U6 promoter. The sequences of thedownstream primers of  E6  siRNA and m-siRNA were synthesized (Invitrogen Corp, Carlsbad, Calif ) as follows:Antisense prime of  E6  siRNA,5¶-caaaaactgtaaa AAGACATTATTCAGACTCTGT ggtgtttcgtcctttcca

    caaga-3¶;sense prime of  E6  siRNA,5¶-caaaaactgtaaa AAACAGAGTCTGAATAATGTC ggtgtttcgtcctttcca

    caaga-3¶;Antisense prime of m-siRNA,5¶-caaaaactgtaaa AAGACTTGTATCACTAACTTC ggtgtttcgtcctttcca

    caaga-3¶;Sense prime of m-siRNA,

    5¶-caaaaactgtaaa AAGAAGTTAGTGATACAAGTC ggtgtttcgtcctttccacaaga-3¶.The PCR reactions were carried out in a final reaction volume

    of 50  KL; it contained template DNA (1 ng/ KL), 1  KL; 10  PCR  buffer, 5 KL; 4 deoxyribonucleotide triphosphates, 1  KL; Taq poly-merase (Gibco), 1 unit; upstream primer (20   Kmol/L), 1.2   KL;

    downstream primer (20 Kmol/L), 1.2 KL; and double distilled water,37.5   KL. Forty cycles of PCR amplification (2 steps) were per-formed as follows: Denature at 94-C for 30 seconds and annealand extend at 72-C for 1 minute 30 seconds. The amplified DNAfragments were fractionated by agarose 1.5% gel electrophoresisand visualized by ethidium bromide staining.

    Cell CultureHuman papillomavirus 18 Y  positive HeLa cervical carcinoma

    cells were cultured in Roswell Park Memorial Institute 1640 me-dium (Invitrogen Corp) supplemented with 10% fetal bovineserum (Invitrogen Corp) and incubated at 37-C with 5% CO2. Themedium was changed once every 2 days, and the cells were trans-ferred into 24-well plates at a density of 3 103 cells per well 1 day before transfection.

    Transfection of CellsOne day before transfection, HeLa cells were transferred into

    24-well plates at a density of 3    104 cells in 400  KL of RoswellPark Memorial Institute 1640 growth medium without antibiotics per well. We used Lipofectamine 2000 (Invitrogen Corp) as trans-fection reagent according to the protocols as follows: (1) 0.5 Kg of DNA (PCR product) was mixed thoroughly with 50  KL of serum-and antibiotic-free medium incubated for 5 minutes at room tem- perature. (2) Lipofectamine 2000 was mixed gently before use and diluted at 0.5  KL in 50  KL of serum- and antibiotic-free medium.The mixture was mixed gently and incubated for 15 minutes at room temperature. (3) After the 15-minute incubation, the diluted PCR products and the diluted Lipofectamine 2000 (total volume,

    approximately 100 KL) were combined. It was mixed gently and in-cubated for 15 minutes at room temperature to allow complexes toform. (4) The mixture of transfection was added into the well drop by drop. We set up some control groups including normal HeLacells as the blank control group and cells added with a nonspecificsiRNA sequence (m-siRNA) as the negative control group. Threewells were performed repeatedly per group. The cells were lysed after 24 hours to isolate total RNA.

    Morphological Analysis in HeLa Cells After Transfection of siRNA

    At 48 and 72 hours after transfections, the morphologicalchanges of 3 groups including normal HeLa cells, cells inter-fered with   E6   siRNA, and m-siRNA were observed under optical

    microscopy.

    Apoptosis Analysis by Hypoploidy AnalysisFirst, HeLa cells (1   106) after transfection were harvested 

    and washed twice using 0.01 mmol/L phosphate-buffered saline(PBS). Then, the cells were fixed using 4 mL of cold 70% ethanolat 4-C for a minimum of 4 hours and then washed twice withPBS. Next, the cells were resuspended in 500  KL of PBS, stained  by adding 200  KL of propidium iodide (50  Kg/mL; Sigma) alongwith 20  KL of RNase (1 mg/mL, Sigma) in a 37-C water bath for 15 Y 20 min. Finally, the apoptosis cells were determined by Eliteflow cytometry (Beckman-Coulter Inc) and analyzed using Start VERITY Mod LT software.

     Min et al    International Journal of Gynecological Cancer    &  Volume 19, Number 4, May 2009

    548   *  2009 IGCS and ESGO

  • 8/17/2019 Microarray Analysis Identifies Differentially

    3/17Copyright @ 2009 by IGCS and ESGO. Unauthorized reproduction of this article is prohibited.

    Cell Viability Assay Cell viability was measured using the 3-(4,5-dimethylthiazol-

    2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assay as described  previously.20 A total of 2   104 cells per well were plated in a 96-well plate. After 24 hours of plating, the untreated cells and thecells transfected with  E6  siRNA or negative control m-siRNA were

    incubated with 10  KL of MTT (10 mg/mL) at 37-C. As a tetrazo-lium salt, MTT can be converted by living cells into blue formazancrystals. The medium was removed from the wells 4 hours after MTT addition, 200  KL of dimethyl sulfoxide was added to dissolvethe farmazan crystals, and then the absorbance ( A) values weremeasured in an automatic microplate reader (MicroQuant; Bio-Tek Instruments, Inc) at the wavelength of 570 nm. Each assay was performed in triplicate.

    Semiquantitative Real-Time PCR Analysis of E6  Messenger RNA Expression

    Relative quantitative multiplex real-time PCR (RT-PCR)was performed using the housekeeping gene glyceraldehyde-3- phosphate dehydrogenase (GAPDH ) internal control as follows.Total RNA was extracted using the Trizol reagent (Invitrogen) ac-cording to manufacturer’s instructions. For RT-PCR, 2  Kg of deoxy-ribonuclease I (DNase I) Y treated total RNAwas used as the templatefor single-stranded complementary DNA (cDNA) synthesis reac-tions using oligo(deoxythymidine) primer and reverse transcriptase(Gibco) for 60 minutes at 42-C. Multiplex PCR reactions were per-formed using Taq polymerase (Takara Co, Japan) with primers for  both the  GAPDH   internal control and   E6  gene. To avoid satura-tion or plateau effects, the linear range of amplification efficiencyfor each gene studied was determined by comparing the amplifi-cation products from identical reactions removed from the ther-mal cycle every other cycle. The number of cycles that yielded anamplification-efficiency in the middle of this linear range was used in subsequent reactions. The primers for amplification of  E6   wereforward 5¶-CAACACGGCGACCCTACA-3¶ and reverse 5¶-GGATTCAACGGTTTCTGG-3¶   with an annealing temperature of 58-C,

    yielding an amplicon of 330 base pairs (bp). In addition,  GAPDH messenger RNA (mRNA) was amplified using the forward primer 5¶-CAACGGATTTGGTCGTATT-3¶ and the reverse 5¶-CACAGTCTTCTGGGTGGC-3 ¶, yielding an amplicon of 550 bp. The PCR prod-ucts were fractionated by agarose 1.5% gel electrophoresis and vi-sualized by ethidium bromide staining.

    Oligonucleotide Array Hybridizationand Data Analysis

    Fluorescent-labeled single-stranded complementary RNA(cRNA) samples were prepared for hybridization as follows. TotalRNA was extracted from cells interfered with  E6   siRNA and cellswith nonspecific siRNA after 24 hours using the Trizol reagent (Invitrogen) and purified on RNeasy columns (Qiagen, Inc, Valen-

    cia, Calif). The integrity and high quality of RNA samples wereconfirmed by using agarose electrophoresis and the lab-on-a-chipsystem (Agilent Technologies Inc, Santa Clara, Calif ). Fluorescent cRNA samples were synthesized using Agilent Low RNA Input Fluorescent Linear Amplification Kit using 50 to 500 ng of totalRNA as starting material. A primer that contains poly(deoxythymi-dine) and a T7 polymerase promoter, was annealed to the poly A +RNA. Reverse transcriptase was added to the reaction to synthesizethe first and second strands of cDNA. At this point, double-stranded cDNA had been synthesized. Next, cRNA was synthesized usingT7 RNA polymerase, which simultaneously incorporated cyanine3 Y  or cyanine 5 Y labeled cytosine triphosphate (CTP). The cRNA of the E6  siRNA group was labeled with Cy3 and m-siRNA with Cy5.Labeled cRNA samples were cleaned using the RNeasy Mini kit 

    (Qiagen) and eluted with 30  KL of diethyl pyrocarbonate Y treated water. Labeled cRNA samples were fragmented to a mean size of 100 to 200 bases by incubating at 60-C for 30 minutes in 25fragmentation buffer contained in the Agilent Human 1A OligoMicroarray Kit (Agilent Technologies Inc).

    The array hybridization, washing, and scanning procedures

    were performed according to the Agilent protocol using labeled cRNA on Agilent Human 1A Oligo Microarray containing morethan 20,000 60-mer probes corresponding to more than 18,000human genes and expressed sequence tags. In addition, the Agilent G2565BA dual-laser microarray scanner scanned the oligo array.Two independent duplicates were performed.

    Spot quantitation, normalization, and application of a platform-specific error model were performed using Agilent’s Fea-ture Extractor software. This allows for the calculation of meanratios between expression levels of each gene in the analyzed sam- ple pair, SD, and P values for each experiment.

    Quantitative RT-PCRQuantitative RT-PCR applying SYBR Green dye was used to

    validate the differential expression of 4 genes found by microarray

    and   E6   gene inhibition. The total RNA from the cells that inter-fered with  E6  siRNA and m-siRNA 24 hours after transfection wasanalyzed in the RT-PCR. Total RNA samples were treated withRNAse-free DNase treatment and removal reagents (Ambion, Tex).Reactions were performed in quadruplicate on the Cycle Real-TimePCR System (BioRad). We used a 2-step RT-PCR method to am- plify target genes and internal control gene (GAPDH ) from HeLacells of both the siRNA transfection and the control groups. Single-stranded cDNAs as standard substances were synthesized from500 ng of total RNA of HeLa cells using a reverse transcription sys-tem. The cDNA samples were serially diluted (1, 1:10, 1:100, and 1:1000) and subjected to SYBR Green fluorescent quantitationPCR amplification with SYBR Premix Ex Taq with forward and reverse primers specific to the target genes or  GAPDH . Then thestandard curves of the target genes and  GAPDH  were achieved ac-cording to the cycle threshold values of each amplification curves.The relative expression levels of 4 genes including   CCNG1,   CD KN1A, and   IFBP3   were testified with RT-PCR under the follow-ing conditions: Denaturation at 94-C for 10 seconds followed  by 45 PCR cycles (denaturation at 94-C for 5 seconds and primersannealing and extension at 60-C for 20 seconds). The gene-specific primer pairs and product sizes were listed in Table 1, showing that all primers had the same melting temperature.

    Gene Function AnalysisWe analyzed the gene functions and cellular pathways of 

    some differentially expressed genes in detail, including cell cycle Y related genes,   CCNG1   and   p21; apoptosis-related genes,  CASP4,CASP6 ,   IGFBP3, and   DFFA; ubiquitin proteolysis pathway Y related genes,   UBE3A   and   UBE2C ; keratinocyte differentiation Y related genes, KRT4, KRT6E , and  KRT18; and antioncogene, RECK and  VEL.

    RESULTS

    Morphological Changes in HeLa Cells After Transfection of siRNA

    We first determined if the transient transfection of  E6  siRNAcould induce the morphological changes in HeLa cells by phasecontrast microscope. As shown in Figure 1, visible morphologicalchanges were prominent at 48 hours after the original transfectionincluding cell swelling and rounding and cytoplasmic vacuole ap- pearance. By 72 hours after transfection, there were striking mor- phological differences between the E6  siRNA Y treated cells and the

    International Journal of Gynecological Cancer    &  Volume 19, Number 4, May 2009   Microarray Analysis Identifies Genes 

    *  2009 IGCS and ESGO   549

  • 8/17/2019 Microarray Analysis Identifies Differentially

    4/17Copyright @ 2009 by IGCS and ESGO. Unauthorized reproduction of this article is prohibited.

    control cells. The cytoplasmic shrinkage and condensation of nu-clear chromatin were observed in the   E6   siRNA Y treated cells. In

    addition, DNA fragmentation apoptosis assay detected DNA strand  breaks in cells after 48 hours in the  E6  siRNA group, whereas thecontrol group had no the same change (data not shown).

    E6  siRNA Causes Low Viability of HeLa CellsTo examine if the suppression of   E6   and m-siRNA would 

    affect cell viability, MTT assay were carried out on hours 0, 12, 24,48, and 72 after transfection using an equal number of HeLa cells.Figure 2 showed that inhibition of   E6   expression by   E6   siRNAdecreased the viability of HeLa cell compared with that in thecontrol and the m-siRNA groups ( P  G  0.01). The marked decreasein cell viabilities was observed from 48 hours after transfection.All experiments were repeated at least thrice. The data were ana-

    lyzed with the software package SPSS 10.0. The  W2 test was per-formed to compare frequencies between the 2 groups. P  G  0.05 were

    considered significant statistically.

    Increased Apoptosis in HeLa Cells Inducedby  E6  siRNA

    The number of apoptotic cells was determined by analysisof 1     106 HeLa cells stained by propidium iodide (Fig. 3). The percentage of apoptotic cells at 24, 48, and 72 hours after  E6  siRNAinterference were 26.4%, 40.3%, and 55.8%, respectively. In ad-dition, the percentage of apoptotic cells in normal cells and m-siRNA Y infected cells were 6% and 10% at 48 hours, respectively.We conclude that siRNA directed against HPV-18 E6 specificallykills HPV-18 Y  positive cancer cells with high efficiency through theinduction of apoptotic cell death.

    Detection of Inhibition of  E6  mRNA

    Expression in HeLa Cells by RT-PCRRelative quantitative RT-PCR was used for analysis of   E6 

    RNA expression in HeLa cells. The amplified DNA fragments were

    FIGURE 1.  Morphological changes in HeLa cells after transfection of siRNA under optical microscope (originalmagnification 40). A, Control group. B, Forty-eight hoursafter transfection of m-siRNA group. C, Forty-eight hours after transfection of the siRNA group. D, Seventy-two hoursafter transfection of the siRNA group.

    FIGURE 2.  Effect of  E6  siRNA on the viability of HeLa cells.Data shown are the mean (SD) results of a representativeexperiment performed in triplicate. *P  G  0.05 and **P  G  0.01.

    TABLE 1.  Gene-specific primer pairs and product sizes

    No. Gene Sequence (5¶ Y 3¶) Temperature,   -C GC, % Product, bp

    1   CCNG1   TGACAAGCCTGAGAAGGTAAACTG 63.7 45.8 148

    TTGAAGCTGTGGGAAGACTGATAG 63.6 45.8

    2   p21   TAACTCTGAGGACACGCATTTGG 64.9 47.8 120

    TGAGTAGAAGAATCGTCGGTTGC 64.2 47.8

    3   IGFBP3   GGGTGTCTGATCCCAAGTTCCA 64.0 50.0 129

    AGGAGAAGTTCTGGGTATCTGTGC 65.0 54.5

    4   UBE3A   GCCATTGTTGCTGCTTC 62.9 54.5 103

    TGGGCTCTTCATCATCTTC 63.2 50.0

    5 HPV-18, E6    CCGTTGAATCCAGCAGA 67.3 54.5 130

    TGCGTCGTTGGAGTCGT 65.0 50.0

    6   GAPDH    GCACCGTCAAGGCTGAGAAC 63.3 60.0 142

    ATGGTGGTGAAGACGCCAGT 62.9 55.0

     Min et al    International Journal of Gynecological Cancer    &  Volume 19, Number 4, May 2009

    550   *  2009 IGCS and ESGO

  • 8/17/2019 Microarray Analysis Identifies Differentially

    5/17Copyright @ 2009 by IGCS and ESGO. Unauthorized reproduction of this article is prohibited.

    fractionated by agarose 1.5% gel electrophoresis and visualized byethidium bromide staining (Fig. 4). Gels were photographed and analyzed semiquantitatively with a laboratory image and analysissystem (UVP Inc). Optical density scanning was used to measuregray value, and  GAPHD   was used to assay the relative expressionof HPV-18   E6   mRNA. The results were expressed as the ratio

     between the gray value of the  E6  gene fragment (330 bp) and thegray value of the GAPHD amplified fragment (550 bp). It wascalculated that HPV-18 E6  mRNA expression in the cells treated by E6   siRNA at 24, 48, and 72 hours were significantly reduced byapproximately 57%, 78%, and 40%, respectively, as compared withthat of the negative and blank control groups, whereas the latter 2groups had similar expression levels. Therefore, it can be concluded that interference with siRNA against HPV-18   E6   gene can sig-nificantly inhibit   E6   gene expression and induce HeLa cells toapoptosis.

    Oligonucleotide Array Analysis of mRNALevels in Cells Interfered by  E6  siRNA

    To identify genes up-regulated or down-regulated by HPV-18 E6 , we used Agilent Human 1A 60mer oligo microarray and defined 

    the differential expression genes with a criterion (log ratio,  P o 0.01and  P R 1.5-fold change in mRNA levels). Among the whole 18716genes and expressed sequence tags, 359 differentially expressed genes were identified, containing 307 up-regulated genes and 52down-regulated genes. These genes were principally classified intoseveral biological process Y related functions using the Panther ana-lytical system, including (1) cell cycle; (2) apoptosis; (3) cell proli-feration and differentiation; (4) protein biosynthesis, metabolism,and modification; (5) nucleobases, nucleoside, nucleotide, and nucleicacid metabolism; (6) signal transduction; (7) immune and defense;

    (8) transcription regulation; and so on. The differentially expressed genes involved in cell cycle, cell proliferation and differentiation, ap-optosis, and immune and defense were listed in Table 2.

    Activities of genes were summarized according to the arraysupplier in combination with extensive information mainly from NCBI PubMed resources and used for grouping the genes. Cy3/Cy5ratio represented the proportion of the fluorescence intensity of sig-nals in E6  siRNA Y transfected cells to the control ones and reflected the change extent of each gene ( Table 3).

    Validation of Gene Expression ChangesWith RT-PCR

    Five genes including HPV-18  E6 ,  CCNG1,  p21,  IFBP3, and UBE3A were testified with RT-PCR. It was shown that the  E6   levelof   E6   siRNA Y treated cells was 39% to that of the control cells.Therefore, it proved that  E6  siRNA significantly inhibited  E6  geneexpression again. In addition, the results of the other 4 genes werein concord with microarray, signifying the high reliability of themicroarray results (Fig. 5).

    DISCUSSIONSilencing RNA is a highly specific tool for targeted gene

    knockdown, and it has advantages over the antisense oligo-DNA or ribozyme because it can be introduced into cells with a high effi-ciency and exert its gene-silencing effect at a concentration severalorders lower. Today, it is generally accepted that RNA interferenceis an effective, feasible, and stable approach for exploring genefunction and identifying and validating new drug targets in func-tional genomic studies. Using HPV-transformed tumor cells as amodel system, we demonstrate that siRNA targeting the viral E6  oncogene represent a very efficient molecular tool to kill virus- positive cancer cells specifically through induction of apoptosis.It is accordant with other previous study, showing that the sur-vival of HPV-positive cancer cells is strictly dependent on the anti-apoptotic function of E6 protein.21 Moreover, our findings indicatethat the molecular targeting of  E6  by siRNAs represents a promi-sing novel approach for the development of specific treatment stra-tegies against HPV-positive cancers and dysplasias.22

    In view of the central role of viral  E6  oncogene expressionfor HPV-associated carcinogenesis, it will be important to identifydownstream regulatory pathways that are affected by E6   oncogene.This should help to gain insights into the cellular pathways targeted during viral transformation. In addition, these analyses may be use-ful to identify novel molecular markers for the diagnosis or the prog-nostic evaluation of cervical cancer.

    E6 is the major transforming proteins of high-risk HPV typesand is known to have the ability to alter cellular differentiation,

    FIGURE 3.  Apoptosis analysis of HeLa cells interfered by  E6 siRNA by hypoploidy analysis. The  x -axis represents themeasurement of fluorescent intensity of Elite flow cytometry,with a resolution of 1024 in units of channels. The  y -axis is therelative numbers of cells. The line (A  Y E) within each figuremeans a threshold gate for detection of apoptosis cells. A,Normal cells. B, Cells interfered by nonspecific siRNA. C, Cellsinterfered by E6  siRNA for 24 hours. D, Cells interfered by  E6 siRNA for 48 hours. F, Cells interfered by E6 siRNA for 72 hours.

    FIGURE 4.  Agarose gel electrophoresis of RT-PCR products of HPV-18 E6 gene (330 bp) and cellular  GAPDH   housekeepinggene (550 bp). M, DL 2000; lane 1, blank control; lane 2,24 hours after  E6  siRNA transfection; lane 3, 48 hours after E6  siRNA transfection; lane 4, 72 hours after  E6  siRNA transfection; lane 5, m-siRNA group.

    International Journal of Gynecological Cancer    &  Volume 19, Number 4, May 2009   Microarray Analysis Identifies Genes 

    *  2009 IGCS and ESGO   551

  • 8/17/2019 Microarray Analysis Identifies Differentially

    6/17Copyright @ 2009 by IGCS and ESGO. Unauthorized reproduction of this article is prohibited.

    TABLE 2.  List of partial differentially expressed genes regulated by  E6  siRNA 

    No. UniGene ID Symbol Gene Name Fold Changes*

    Cell cycle

    1 Hs.510225   RPS6KA5   Ribosomal protein S6 kinase, 90 kd, polypeptide 5 7.718

    2 Hs.115242   DRG1   Developmentally regulated GTP-binding protein 1 5.445

    3 Hs.79101   CCNG1   Cyclin G1 5.244

    4 Hs.464419   FBXO6    F-box protein 6 5.228

    5 Hs.285051   CCPG1   Cell cycle progression 1 4.409

    6 Hs.239   FOXM1   Forkhead box M1 4.064

    7 Hs.559215   MAD2L1   MAD2 mitotic arrest deficient  Y like 1 (yeast) 4.036

    8 Hs.73625   KIF20A   Kinesin family member 20A 3.700

    9 Hs.100426   BRMS1   Breast cancer metastasis suppressor 1 3.178

    10 Hs.491682   PRKDC    Protein kinase, DNA-activated, catalytic polypeptide 3.169

    11 Hs.445758   E2F5   E2F transcription factor 5, p130 binding 2.954

    12 Hs.370771   CDKN1A   Cyclin-dependent kinase inhibitor 1A (p21, Cip1) 2.879

    13 Hs.132161   FOXK2   Forkhead box K2 2.452

    14 Hs.169487   MAFB   V-maf   musculoaponeurotic fibrosarcoma oncogene homolog B (avian) 2.38615 Hs.263812   NUDC    Nuclear distribution gene C homolog ( Aspergillus nidulans) 2.353

    16 Hs.3887   PSMD1   Proteasome (prosome, macropain) 26S subunit, non-ATPase, 1 2.346

    17 Hs.275243   S100A6    S100 calcium-binding protein A6 (calcyclin) 2.296

    18 Hs.498248   EXO1   Exonuclease 1 1.992

    19 Hs.436035   TUBA6    Tubulin, alpha 6 1.608

    20 Hs.122511   CETN1   Centrin, EF-hand protein, 1 1.522

    23 Hs.306791   POLD2   Polymerase (DNA directed), delta 2, regulatory subunit 50 kd 0.629

    24 Hs.520046   GPSM3   G-protein polymerase modulator 3(AGS3-like,  Caenorhabditis elegans)

    0.599

    25 Hs.449410   FOXH1   Forkhead box H1 0.564

    26 Hs.93002   UBE2C    Ubiquitin-conjugating enzyme E2C 0.342

    27 Hs.128073   CETN3   Centrin, EF-hand protein, 3 (CDC31 homolog, yeast) 0.178

    Cell proliferation and differentiation28 Hs.233119   ME2   Inositol polyphosphate-5-phosphatase F 6.842

    29 Hs.473082   ZFP64   Zinc finger protein 64 homolog (mouse) 6.659

    30 Hs.467740   LPIN1   Lipin 1 4.428

    31 Hs.26988   EFNB3   Ephrin-B3 3.000

    32 Hs.497200   PLA2G4A   Phospholipase A2, group IVA (cytosolic, calcium dependent) 2.596

    33 Hs.386294   ZNF195   Zinc finger protein 195 2.451

    34 Hs.459927   PTMA   Prothymosin, alpha (gene sequence 28) 2.317

    35 Hs.504613   PTMS    Parathymosin 2.317

    36 Hs.252229   MAFG    V-maf   musculoaponeurotic fibrosarcoma oncogene homolog G 1.726

    39 Hs.190495   GPNMB   Glycoprotein (transmembrane) nmb 0.582

    40 Hs.140720   FRAT2   Frequently rearranged in advanced T-cell lymphomas 2 0.262

    Apoptosis

    41 Hs.484782   DFFA   DNA fragmentation factor, 45 kd, alpha polypeptide 6.092

    42 Hs.66180   NAP1L2   Nucleosome assembly protein 1 Y like 2 5.912

    43 Hs.103755   RIPK2   Receptor-interacting serine-threonine kinase 2 5.460

    44 Hs.3280   CASP6    Caspase 6, apoptosis-related cysteine peptidase 4.509

    45 Hs.516075   TIA1   TIA1 cytotoxic granule Y associated RNA-binding protein 4.381

    46 Hs.379970   RASSF2   Ras association (RalGDS/AF-6) domain family 2 3.436

    47 Hs.25155   NET1   Neuroepithelial cell Y transforming gene 1 2.692

    48 Hs.445898   MYBL1   V-myb myeloblastosis viral oncogene homolog (avian) Y like 1 2.525

    49 Hs.129708   TNFSF14   Tumor necrosis factor (ligand) superfamily, member 14 2.322

    50 Hs.450230   IGFBP3   Insulin-like growth factor  Y  binding protein 3 2.290

     Min et al    International Journal of Gynecological Cancer    &  Volume 19, Number 4, May 2009

    552   *  2009 IGCS and ESGO

  • 8/17/2019 Microarray Analysis Identifies Differentially

    7/17Copyright @ 2009 by IGCS and ESGO. Unauthorized reproduction of this article is prohibited.

    reactivate host DNA synthesis, and stimulate cell cycle progres-sion.23 In the current work, the differentially expressed genes that are regulated by inhibition of HPV-18  E6   in HeLa cells had beenidentified. We found by microarray analysis that at least 359 geneswere up-regulated or down-regulated after transfection of specific

     E6  siRNA ( P  G

     0.01). In the analysis of microarray results, 4 genesincluding CCNG1,  p21,   IGFBP3, and  UBE3A were chosen for RT-PCR validation. Among all differentially expressed genes, the 4genes were closely related to the expression of P53, which is clearlyaccepted as the target of   E6   oncogene. Therefore, we only chosethese 4 genes in the preliminary analysis to verify the reliability of microarray results.

    Among them, some alterations in gene expression (eg,  p21and  IGFBP3) were dependent on p53 degradation, which is involved in diverse cellular processes, including regulation of the cell cycle,apoptosis, senescence, DNA repair, cell differentiation, and an-giogenesis. One key downstream effector of p53 is p21Waf/Cip(hereafter referred to as p21), a cyclin-dependent kinase/cyclininhibitor whose up-regulation by p53 leads to cell cycle arrest.24 Be-cause p53 up-regulates expression of the cyclin-dependent kinase/ cyclin inhibitor p21, we anticipated that cells with   E6   siRNAinterference and concomitant degradation of p53 would have higher levels of p21. In addition, the microarray analysis and RT-PCR validated that the p21 transcript was up-regulated by 3-folds inapoptotic cells induced by HPV-18  E6  siRNA.

    The differences between the ability of the low-risk and HR-HPV types to induce immortalization and transformation may welllie in their abilities to interact with the cell cycle components,resulting in the loss of multiple cell cycle checkpoints that are im- portant in maintaining host genome fidelity, thus leading to poten-tial accumulation of genetic abnormalities.25 The E6 and E7 proteinsof HR-HPV bind to cell cycle regulatory proteins and interfere with both G1/S and G2/M cell cycle checkpoints much more effectivelythan the low-risk HPV. In addition, the HR-HPV proteins can (1) up-

    regulate expression of cyclins A and B in association with im-mortalization; (2) up-regulate cyclin E expression, shown recently toinduce genetic instability; and (3) abrogate cyclin D1 expression,important in the Rb pathway.26

    CCNG1   (cyclin G1) is also one of the target genes of the

    transcription factor p53 and act as a mediator of p53 functions suchas growth inhibition, DNA repair, and apoptosis.27 Other reportsshowed, however, that cyclin G1 contributes to G2/M arrest of cellsin response to DNA damage and plays a role in apoptosis. 28 Several p53-induced target genes can promote apoptosis, although the ex- pression of each gene alone is usually insufficient to cause sig-nificant cell death. Apoptotic target genes may need to act in concert,activating parallel apoptotic pathways, to cause a full apoptoticresponse, and this might explain why disruption of the   CCNG1gene alone did not seem to affect apoptosis. Overexpression of CCNG1 resulted in increased sensitivity to apoptosis induced by  E6 siRNA.29 In our experiment, the expression of   CCNG1   was ob-viously increased by 5.244 times, and it signified that  CCNG1 might  promote cell apoptosis with the p53  gene.

    Inhibition of apoptosis is a mechanism of survival for virallyor chemically transformed malignant cells. One of the earliest and most consistent observed features of apoptosis is the induction of a series of cytosolic proteases, that is, caspases.30 Active caspasescleave numerous intracellular proteins and contribute to apoptoticcell death.31 At present, the fact that  CASP6  and  CASP4  were ac-tivated during the inhibition of E6 expression could further explainthe antiapoptosis capacity of the E6 protein in transformed cells.Insulin-like growth factor  Y  binding protein 3 ( IGFBP-3) is one of target genes of p53, an extracellular protein responsible for thecarriage of IGF-I but can act independently of IGF-I, inhibiting cellgrowth and enhancing apoptosis. Hollowood et al32 found that anautocrine/paracrine feedback loop existed between IGFBP-3 and  p53, which may provide the social control necessary to maintainnormal tissue homeostasis.

    TABLE 2. (Continued)

    No. UniGene ID Symbol Gene Name Fold Changes*

    51 Hs.435136   TXN    Thioredoxin 2.199

    52 Hs.276876   TM2D1   TM2 domain containing 1 2.102

    53 Hs.446427   OAZ1   Ornithine decarboxylase antizyme 1 2.07154 Hs.94011   NDNL2   Necdin-like 2 2.027

    55 Hs.302015   FKSG2   Apoptosis inhibitor 1.837

    56 Hs.8375   TRAF4   TNF receptor  Y associated factor 4 1.768

    57 Hs.138378   CASP4   Caspase 4, apoptosis-related cysteine peptidase 1.754

    58 Hs.502775   HRASLS3   HRAS-like suppressor 3 1.537

    59 Hs.436657   CLU    Clusterin 0.607

    60 Hs.224137   ENDOG    Mitochondrial endonuclease G 0.574

    Defense/immune response

    61 Hs.495985   TCIRG1   T-cell, immune regulator 1, ATPase, H + transporting, lysosomal 8.243

    62 Hs.512152   HLA-G    HLA-G histocompatibility antigen, class I, G 2.654

    63 Hs.337557   XTP7    Protein 7 transactivated by hepatitis B virus X antigen (HbxAg) 2.320

    64 Hs.374596   TPT1   Tumor protein, translationally controlled 1 1.812

    65 Hs.534255   B2M    A2-microglobulin 1.736

    66 Hs.515369   TYROBP    TYRO protein tyrosine kinase binding protein 0.575

    67 Hs.464987   SFTPA2   Surfactant, pulmonary-associated protein A2 0.530

    68 Hs.193122   FCAR   Fc fragment of IgA, receptor for 0.523

    *Mean of 2 independent experiments.ATPase, Adenosine triphosphatase; GTP, guanosine triphosphate; MAD2, mitotic arrest deficient 2.

    International Journal of Gynecological Cancer    &  Volume 19, Number 4, May 2009   Microarray Analysis Identifies Genes 

    *  2009 IGCS and ESGO   553

  • 8/17/2019 Microarray Analysis Identifies Differentially

    8/17Copyright @ 2009 by IGCS and ESGO. Unauthorized reproduction of this article is prohibited.

    TABLE 3.  List of differentially expressed genes in Panther biological processes categories

    No. UniGene ID Symbol Gene Name Ratio

    Cell cycle

    1 Hs.510225   RPS6KA5   Ribosomal protein S6 kinase, 90 kd, polypeptide 5 7.718

    2 Hs.115242   DRG1   Developmentally regulated GTP-binding protein 1 5.445

    3 Hs.79101   CCNG1   Cyclin G1 5.244

    4 Hs.464419   FBXO6    F-box protein 6 5.228

    5 Hs.285051   CCPG1   Cell cycle progression 1 4.409

    6 Hs.239   FOXM1   Forkhead box M1 4.064

    7 Hs.559215   MAD2L1   MAD2 mitotic arrest deficient  Y like 1 (yeast) 4.036

    8 Hs.73625   KIF20A   Kinesin family member 20A 3.700

    9 Hs.100426   BRMS1   Breast cancer metastasis suppressor 1 3.178

    10 Hs.491682   PRKDC    Protein kinase, DNA-activated, catalytic polypeptide 3.169

    11 Hs.445758   E2F5   E2F transcription factor 5, p130-binding 2.954

    12 Hs.370771   CDKN1A   Cyclin-dependent kinase inhibitor 1A (p21, Cip1) 2.879

    13 Hs.132161   FOXK2   Forkhead box K2 2.452

    14 Hs.169487   MAFB   V-maf   musculoaponeurotic fibrosarcoma oncogene homolog B (avian) 2.38615 Hs.263812   NUDC    Nuclear distribution gene C homolog ( Aspergillus nidulans) 2.353

    16 Hs.3887   PSMD1   Proteasome (prosome, macropain) 26S subunit, non-ATPase, 1 2.346

    17 Hs.275243   S100A6    S100 calcium-binding protein A6 (calcyclin) 2.296

    18 Hs.498248   EXO1   Exonuclease 1 1.992

    19 Hs.436035   TUBA6    Tubulin, alpha 6 1.608

    20 Hs.122511   CETN1   Centrin, EF-hand protein, 1 1.522

    21 Hs.465498   TXNL4A   Thioredoxin-like 4A 1.475

    22 Hs.344400   MPHOSPH6    M-phase phosphoprotein 6 1.440

    23 Hs.306791   POLD2   Polymerase (DNA directed), delta 2, regulatory subunit 50 kd 0.629

    24 Hs.520046   GPSM3   G-protein polymerase modulator 3 (AGS3-like,  Caenorhabditis elegans) 0.599

    25 Hs.449410   FOXH1   Forkhead box H1 0.564

    26 Hs.93002   UBE2C    Ubiquitin-conjugating enzyme E2C 0.342

    27 Hs.128073   CETN3   Centrin, EF-hand protein, 3 (CDC31 homolog, yeast) 0.178Cell proliferation and differentiation

    28 Hs.233119   ME2   Inositol polyphosphate-5-phosphatase F 6.842

    29 Hs.473082   ZFP64   Zinc finger protein 64 homolog (mouse) 6.659

    30 Hs.467740   LPIN1   Lipin 1 4.428

    31 Hs.26988   EFNB3   Ephrin-B3 3.000

    32 Hs.497200   PLA2G4A   Phospholipase A2, group IVA (cytosolic, calcium-dependent) 2.596

    33 Hs.386294   ZNF195   Zinc finger protein 195 2.451

    34 Hs.459927   PTMA   Prothymosin, alpha (gene sequence 28) 2.317

    35 Hs.504613   PTMS    Parathymosin 2.317

    36 Hs.252229   MAFG    V-maf   musculoaponeurotic fibrosarcoma oncogene homolog G 1.726

    37 Hs.440829   CEBPD   CCAAT/enhancer binding protein (C/EBP), delta 1.436

    38 Hs.158287   SDC3   Syndecan 3 ( N -syndecan) 0.733

    39 Hs.190495   GPNMB   Glycoprotein (transmembrane) nmb 0.582

    40 Hs.140720   FRAT2   Frequently rearranged in advanced T-cell lymphomas 2 0.262

    Protein biosynthesis

    41 Hs.22867   EIF2C1   Eukaryotic translation initiation factor 2C, 1 5.654

    42 Hs.515070   EEF2   Eukaryotic translation elongation factor 2 2.275

    43 Hs.421608   EEF1B2   Eukaryotic translation elongation factor 1 beta 2 2.041

    44 Hs.434248   PLEC1   Plectin 1, intermediate filament  Y  binding protein 500 kd 2.028

    45 Hs.88977   EEF1E1   Eukaryotic translation elongation factor 1 epsilon 1 1.890

    46 Hs.530734   MRPL16    Mitochondrial ribosomal protein L16 1.772

    47 Hs.491988   TRAM1   Translocation-associated membrane protein 1 1.460

     Min et al    International Journal of Gynecological Cancer    &  Volume 19, Number 4, May 2009

    554   *  2009 IGCS and ESGO

  • 8/17/2019 Microarray Analysis Identifies Differentially

    9/17Copyright @ 2009 by IGCS and ESGO. Unauthorized reproduction of this article is prohibited.

    TABLE 3. (Continued)

    No. UniGene ID Symbol Gene Name Ratio

    Protein metabolism and modification

    48 Hs.108332   UBE2D2   Ubiquitin-conjugating enzyme E2D 2 (UBC4/5 homolog, yeast) 6.998

    49 Hs.472119   MKKS    McKusick-Kaufman syndrome 6.98150 Hs.193226   UGCGL2   UDP-glucose ceramide glucosyltransferase Y like 2 6.954

    51 Hs.233950   SPINT1   Serine peptidase inhibitor, Kunitz type 1 5.285

    52 Hs.356769   MAN2B1   Mannosidase, alpha, class 2B, member 1 4.950

    53 Hs.180758   GALNACT-2   Chondroitin sulfate GalNAcT-2 4.835

    54 Hs.175322   USP13   Ubiquitin-specific peptidase 13 (isopeptidase T-3) 4.710

    55 Hs.163776   UBE2J1   Ubiquitin-conjugating enzyme E2, J1 (UBC6 homolog, yeast) 4.610

    56 Hs.744   FDX1   Ferredoxin 1 4.444

    57 Hs.440833   PKN2   Protein kinase N2 4.319

    58 Hs.531176   SARS    Seryl-tRNA synthetase 3.779

    59 Hs.49774   PTPRM    Protein tyrosine phosphatase, receptor type, M 3.370

    60 Hs.438231   TFPI2   Tissue factor pathway inhibitor 2 3.336

    61 Hs.491682   PRKDC    Protein kinase, DNA-activated, catalytic polypeptide 3.169

    62 Hs.121676   ZFYVE19   Zinc finger, FYVE domain containing 19 3.157

    63 Hs.73986   CLK2   CDC-like kinase 2 2.882

    64 Hs.178748   ADAM21   ADAM metallopeptidase domain 21 2.839

    65 Hs.523438   TRIM68   Tripartite motif  Y containing 68 2.746

    66 Hs.110364   PPIC    Peptidylprolyl isomerase C (cyclophilin C) 2.594

    67 Hs.72026   PRSS21   Protease, serine, 21 (testisin) 2.525

    68 Hs.127407   GALNT7    UDP- N -acetyl-alpha-d-galactosamine:polypeptide N -acetylgalactosaminyltransferase7 (GalNAc-T7)

    2.496

    69 Hs.14511   SCO1   SCO cytochrome oxidase deficient homolog 1 (yeast) 2.492

    70 Hs.158688   EIF5B   Eukaryotic translation initiation factor 5B 2.480

    71 Hs.233952   PSMA7    Proteasome (prosome, macropain) subunit, alpha type, 7 2.323

    72 Hs.524648   LTA4H    Leukotriene A4 hydrolase 2.311

    73 Hs.520348   UBC    Ubiquitin C 2.253

    74 Hs.18349   MRPL15   Mitochondrial ribosomal protein L15 2.119

    75 Hs.1197   HSPE1   Heat shock 10-kd protein 1 (chaperonin 10) 2.104

    76 Hs.434248   PLEC1   Plectin 1, intermediate filament  Y  binding protein, 500 kd 2.028

    77 Hs.520028   HSPA1A   Heat shock 70-kd protein 1A 2.002

    78 Hs.381167   SERPINB1   Serpin peptidase inhibitor, clade B (ovalbumin), member 1 1.993

    79 Hs.522394   HSPA5   Heat shock 70-kd protein 5 (glucose-regulated protein, 78 kd) 1.989

    80 Hs.423163   SLC35A1   Solute carrier family 35 (CMP Y sialic acid transporter), member A1 1.952

    81 Hs.523936   PRCP    GCRG-P224 1.830

    82 Hs.47099   GALNT12   UDP- N -acetyl-alpha-d-galactosamine:polypeptide N -acetylgalactosaminyltransferase12 (GalNAc-T12)

    1.808

    83 Hs.128420   VPS4A   Vacuolar protein sorting 4A (yeast) 1.782

    84 Hs.368985   TRIP12   Thyroid hormone receptor interactor 12 1.761

    85 Hs.57732   MAPK11   Mitogen-activated protein kinase 11 1.76186 Hs.356190   UBB   Ubiquitin B 1.745

    87 Hs.20013   SYF2   SYF2 homolog, RNA splicing factor (Saccharomyces cerevisiae) 1.684

    88 Hs.7879   IFRD1   Interferon-related developmental regulator 1 1.635

    89 Hs.191887   SEC61B   Sec61 beta subunit 1.554

    90 Hs.405410   OGT O-Linked   N -acetylglucosamine (GlcNAc) transferase (UDP- N -acetylglucosamine: polypeptide- N -acetylglucosaminyl transferase)

    1.535

    91 Hs.521937   PPP1R16A   Protein phosphatase 1, regulatory (inhibitor) subunit 16A 1.495

    92 Hs.524690   PPIE    Peptidylprolyl isomerase E (cyclophilin E) 1.483

    93 Hs.466743   MAP3K10   Mitogen-activated protein kinase kinase kinase 10 1.435

    (Continued on next page)

    International Journal of Gynecological Cancer    &  Volume 19, Number 4, May 2009   Microarray Analysis Identifies Genes 

    *  2009 IGCS and ESGO   555

  • 8/17/2019 Microarray Analysis Identifies Differentially

    10/17Copyright @ 2009 by IGCS and ESGO. Unauthorized reproduction of this article is prohibited.

    TABLE 3. (Continued)

    No. UniGene ID Symbol Gene Name Ratio

    94 Hs.477879   H2AFX    H2A histone family, member X 1.398

    95 Hs.232375   ACAT1   Acetyl-coenzyme A acetyltransferase 1 (acetoacetyl coenzyme A thiolase) 1.367

    96 Hs.130988   DYRK1B   Dual-specificity tyrosine-(Y)-phosphorylation regulated kinase 1B 0.73997 Hs.380742   PLXNB3   Plexin B3 0.708

    98 Hs.484138   FBXW11   F-box and WD-40 domain protein 11 0.655

    99 Hs.567354   UBE3A   Ubiquitin protein ligase E3A (human papilloma virus E6AP, Angelman syndrome) 0.532

    100 Hs.407856   SPINK1   Serine peptidase inhibitor, Kazal type 1 0.491

    101 Hs.162241   UCHL3   Ubiquitin carboxyl-terminal esterase L3 (ubiquitin thiolesterase) 0.441

     Nucleobase, nucleoside, nucleotide, and nucleic acid metabolism

    102 Hs.534460   DUS2L   Dihydrouridine synthase 2 Y like, SMM1 homolog (Saccharomyces cerevisiae) 10.023

    103 Hs.200596   LCMT2   Leucine carboxyl methyltransferase 2 5.615

    104 Hs.459857   CARHSP1   Calcium-regulated heat stable protein 1, 24 kd 5.532

    105 Hs.478708   OPA1   Optic atrophy 1 (autosomal dominant) 5.389

    106 Hs.242635   RAD50   RAD50 homolog (Saccharomyces cerevisiae) 4.875

    107 Hs.65734   ARNTL   Aryl hydrocarbon receptor nuclear translocator  Y like 4.774

    108 Hs.505004   TCEA2   Transcription elongation factor A (SII), 2 4.672

    109 Hs.473087   CTPS    CTP synthase 4.337

    110 Hs.437582   MCM8   MCM8 minichromosome maintenance deficient 8 (Saccharomyces cerevisiae) 4.247

    111 Hs.193163   BIN1   Bridging integrator 1 3.987

    112 Hs.429666   CEBPG    CCAAT/enhancer binding protein (C/EBP), gamma 3.637

    113 Hs.98367   SOX17    SRY (sex-determining region Y)-box 17 3.585

    114 Hs.437056   SUPT5H    Suppressor of Ty 5 homolog (Saccharomyces cerevisiae) 3.337

    115 Hs.509140   BAZ1A   Bromodomain adjacent to zinc f inger domain, 1A 3.256

    116 Hs.446318   HOXA7    Homeobox A7 3.256

    117 Hs.469872   ERCC3   Excision repair cross-complementing rodent repair deficiency, complementationgroup 3 (xeroderma pigmentosum group B complementing)

    3.197

    118 Hs.170568   TATDN1   TatD DNase domain containing 1 3.176

    119 Hs.491682   PRKDC    Protein kinase, DNA-activated, catalytic polypeptide 3.169

    120 Hs.323213   YIPF2   Yip1 domain family, member 2 2.934

    121 Hs.293818   NEIL2   Nei-like 2 ( Escherichia coli) 2.824

    122 Hs.368410   CBX2   Chromobox homolog 2 (Pc class homolog, Drosophila) 2.643

    123 Hs.21160   ME1   Malic enzyme 1, NADP(+)-dependent, cytosolic 2.483

    124 Hs.130098   DDX23   DEAD (Asp-Glu-Ala-Asp) box polypeptide 23 2.451

    125 Hs.463456   NME2   Methionine sulfoxide reductase B3 2.321

    126 Hs.408067   HIST2H2AC    Histone 2, H2ac 2.147

    127 Hs.235069   RECQL   RecQ protein-like (DNA helicase Q1-like) 2.099

    128 Hs.522767   SLC25A5   Solute carrier family 25 (mitochondrial carrier; adenine translocator), member 5 2.086

    129 Hs.498248   EXO1   Exonuclease 1 1.992

    130 Hs.515255   LSM4   LSM4 homolog, U6 small nuclear RNA associated (Saccharomyces cerevisiae) 1.967

    131 Hs.525629   MTA1   Metastasis associated 1 1.869

    132 Hs.124027   SEPHS1   Selenophosphate synthetase 1 1.811133 Hs.429   ATP5G3   ATP synthase, H + transporting, mitochondrial F0 complex, subunit C3 (subunit 9) 1.948

    134 Hs.326387   MORF4L2   Mortality factor 4 like 2 1.794

    135 Hs.557550   NPM1   TRK-fused gene 1.605

    136 Hs.529798   BTF3   Basic transcription factor 3 1.548

    137 Hs.369056   SP100   SP100 nuclear antigen 1.521

    138 Hs.477481   MCM2   MCM2 minichromosome maintenance deficient 2, mitotin (Saccharomyces cerevisiae) 1.499

    139 Hs.68714   SFRS1   Splicing factor, arginine/serine-rich 1 (splicing factor 2, alternate splicing factor) 1.497

    140 Hs.440829   CEBPD   CCAAT/enhancer binding protein (C/EBP), delta 1.436

    141 Hs.346868   EBNA1BP2   EBNA1 binding protein 2 1.430

    142 Hs.477879   H2AFX    H2A histone family, member X 1.398

     Min et al    International Journal of Gynecological Cancer    &  Volume 19, Number 4, May 2009

    556   *  2009 IGCS and ESGO

  • 8/17/2019 Microarray Analysis Identifies Differentially

    11/17Copyright @ 2009 by IGCS and ESGO. Unauthorized reproduction of this article is prohibited.

    TABLE 3. (Continued)

    No. UniGene ID Symbol Gene Name Ratio

    143 Hs.288487   SMS    Spermine synthase 1.384

    144 Hs.387804   PABPC1   Poly(A)-binding protein, cytoplasmic 1 1.386

    145 Hs.79110   NCL   U23 small nucleolar RNA 1.352146 Hs.535499   RARA   Retinoic acid receptor, alpha 0.646

    147 Hs.306791   POLD2   Polymerase (DNA directed), delta 2, regulatory subunit, 50 kd 0.629

    148 Hs.567358   WRN    Werner syndrome 0.617

    149 Hs.14839   POLR2G    Polymerase (RNA) II (DNA directed) polypeptide G 0.600

    150 Hs.255932   XRN2   5¶-3¶ exoribonuclease 2 0.579

    151 Hs.227049   CTPS2   CTP synthase II 0.533

    152 Hs.70937   HIST1H3H    Histone 1, H3h 0.421

    153 Hs.375179   ADSSL1   Adenylosuccinate synthase Y like 1 0.220

    Signal transduction

    154 Hs.553838   OR10C1   Olfactory receptor, family 10, subfamily C, member 1 8.210

    155 Hs.415172   RABL4   RAB, member of RAS oncogene family Y like 4 7.718

    156 Hs.191762   NOXO1   NADPH oxidase organizer 1 5.308

    157 Hs.472861   CDH22   Cadherin-like 22 5.259

    158 Hs.282326   DSCR1   Down syndrome critical region gene 1 4.941

    159 Hs.247787   OPN1MW    Opsin 1 (cone pigments), medium-wave sensitive (color blindness, deutan) 4.869

    160 Hs.518149   TNR   Tenascin R (restrictin, janusin) 4.691

    161 Hs.553592   OR6N1   Olfactory receptor, family 6, subfamily N, member 1 4.070

    162 Hs.379970   RASSF2   Ras association (RalGDS/AF-6) domain family 2 3.436

    163 Hs.49774   PTPRM    Protein tyrosine phosphatase, receptor type, M 3.370

    164 Hs.321541   RAB11A   RAB11A, member RAS oncogene family 3.263

    165 Hs.74034   CAV1   Caveolin 1, caveolae protein, 22 kd 3.085

    166 Hs.26988   EFNB3   Ephrin-B3 3.000

    167 Hs.27018   RASL12   RAS-like, family 12 2.893

    168 Hs.178748   ADAM21   ADAM metallopeptidase domain 21 2.839

    169 Hs.7879   IFRD1   Interferon-related developmental regulator 1 2.786170 Hs.158348   HCRT    Hypocretin (orexin) neuropeptide precursor 2.614

    171 Hs.558543   PPCS    Phosphopantothenoylcysteine synthetase 2.393

    172 Hs.259461   PALM2-AKAP2   Paralemmin 2 2.324

    173 Hs.450230   IGFBP3   Insulin-like growth factor  Y  binding protein 3 2.290

    174 Hs.301540   SPR   Sepiapterin reductase (7,8-dihydrobiopterin:NADP + oxidoreductase) 2.886

    175 Hs.567639   SPSB4   SplA/ryanodine receptor domain and SOCS box containing 4 1.844

    176 Hs.57732   MAPK11   Mitogen-activated protein kinase 11 1.761

    177 Hs.501293   BSG    Basigin (Ok blood group) 1.742

    178 Hs.54483   NMI    N-myc (and STAT) interactor 1.709

    179 Hs.294603   CNIH    Cornichon homolog (Drosophila) 1.544

    180 Hs.119689   CGA   Glycoprotein hormones, alpha polypeptide 1.512

    181 Hs.209983   STMN1   Stathmin 1/oncoprotein 18 1.385

    182 Hs.247838   CCL24   Chemokine (C-C motif) ligand 24 0.718183 Hs.522484   OLFM1   Olfactomedin 1 0.618

    184 Hs.316997   EPS8   Epidermal growth factor receptor pathway substrate 8 0.561

    185 Hs.175934   GABRA1   F-Aminobutyric acid (GABA) A receptor, alpha 1 0.531

    186 Hs.193122   FCAR   Fc fragment of IgA, receptor for 0.523

    187 Hs.351812   CLEC4C    C-type lectin domain family 4, member C 0.490

    188 Hs.407587   GNRHR   Gonadotropin-releasing hormone receptor 0.472

    189 Hs.3945   C20orf45   Chromosome 20 open reading frame 45 0.467

    (Continued on next page)

    International Journal of Gynecological Cancer    &  Volume 19, Number 4, May 2009   Microarray Analysis Identifies Genes 

    *  2009 IGCS and ESGO   557

  • 8/17/2019 Microarray Analysis Identifies Differentially

    12/17Copyright @ 2009 by IGCS and ESGO. Unauthorized reproduction of this article is prohibited.

    TABLE 3. (Continued)

    No. UniGene ID Symbol Gene Name Ratio

    Apoptosis

    190 Hs.484782   DFFA   DNA fragmentation factor, 45 kd, alpha polypeptide 6.092

    191 Hs.66180   NAP1L2   Nucleosome assembly protein 1 Y 

    like 2 5.912192 Hs.103755   RIPK2   Receptor-interacting serine-threonine kinase 2 5.460

    193 Hs.3280   CASP6    Caspase 6, apoptosis-related cysteine peptidase 4.509

    194 Hs.516075   TIA1   TIA1 cytotoxic granule Y associated RNA-binding protein 4.381

    195 Hs.379970   RASSF2   Ras association (RalGDS/AF-6) domain family 2 3.436

    196 Hs.25155   NET1   Neuroepithelial cell transforming gene 1 2.692

    197 Hs.445898   MYBL1   V-myb myeloblastosis viral oncogene homolog (avian) Y like 1 2.525

    198 Hs.129708   TNFSF14   Tumor necrosis factor (ligand) superfamily, member 14 2.322

    199 Hs.450230   IGFBP3   Insulin-like growth factor  Y  binding protein 3 2.290

    200 Hs.435136   TXN    Thioredoxin 2.199

    201 Hs.276876   TM2D1   TM2 domain containing 1 2.102

    202 Hs.446427   OAZ1   Ornithine decarboxylase antizyme 1 2.071

    203 Hs.94011   NDNL2   Necdin-like 2 2.027

    204 Hs.302015   FKSG2   Apoptosis inhibitor 1.837

    205 Hs.8375   TRAF4   TNF receptor  Y associated factor 4 1.768

    206 Hs.138378   CASP4   Caspase 4, apoptosis-related cysteine peptidase 1.754

    207 Hs.502775   HRASLS3   HRAS-like suppressor 3 1.537

    208 Hs.448588   NGFRAP1   Nerve growth factor receptor (TNFRSF16) associated protein 1 1.395

    209 Hs.436657   CLU    Clusterin 0.607

    210 Hs.224137   ENDOG    Mitochondrial endonuclease G 0.574

    Defense/immune response

    211 Hs.495985   TCIRG1   T-cell, immune regulator 1, ATPase, H + transporting, lysosomal 8.243

    V0 subunit A3

    212 Hs.512152   HLA-G    HLA-G histocompatibility antigen, class I, G 2.654

    213 Hs.337557   XTP7    Protein 7 transactivated by hepatitis B virus X antigen (HbxAg) 2.320

    214 Hs.374596   TPT1   Tumor protein, translationally controlled 1 1.812215 Hs.534255   B2M    A2-microglobulin 1.736

    216 Hs.524690   PPIE    Peptidylprolyl isomerase E (cyclophilin E) 1.483

    217 Hs.466743   MAP3K10   Mitogen-activated protein kinase kinase kinase 10 1.435

    218 Hs.247838   CCL24   Chemokine (C-C motif) ligand 24 0.718

    219 Hs.515369   TYROBP    TYRO protein tyrosine kinase Y  binding protein 0.575

    220 Hs.464987   SFTPA2   Surfactant, pulmonary-associated protein A2 0.530

    221 Hs.193122   FCAR   Fc fragment of IgA, receptor for 0.523

    Cell structure and motility

    222 Hs.371139   KRT4   Keratin 4 5.623

    223 Hs.478708   OPA1   Optic atrophy 1 (autosomal dominant) 5.389

    224 Hs.474053   COL6A1   Collagen, type VI, alpha 1 3.939

    225 Hs.500916   INA   Internexin neuronal intermediate filament protein, alpha 3.688

    226 Hs.558758   KRT6E    Keratin 6E 3.525

    227 Hs.435326   ACTL6A   Actin-like 6A 2.855

    228 Hs.512842   MFAP5   Microfibrillar-associated protein 5 2.456

    229 Hs.406013   KRT18   Keratin 18 2.322

    230 Hs.526500   DNAH3   Dynein, axonemal, heavy polypeptide 3 2.320

    231 Hs.524390   K-ALPHA-1   Tubulin, alpha, ubiquitous 2.312

    232 Hs.434248   PLEC1   Plectin 1, intermediate filament  Y  binding protein, 500 kd 2.028

    233 Hs.128420   VPS4A   Vacuolar protein sorting 4A (yeast) 1.782

    234 Hs.437403   PPA1   Pyrophosphatase (inorganic) 1 1.728

    235 Hs.433512   ACTR3   ARP3 actin-related protein 3 homolog (yeast) 1.467

     Min et al    International Journal of Gynecological Cancer    &  Volume 19, Number 4, May 2009

    558   *  2009 IGCS and ESGO

  • 8/17/2019 Microarray Analysis Identifies Differentially

    13/17Copyright @ 2009 by IGCS and ESGO. Unauthorized reproduction of this article is prohibited.

    TABLE 3. (Continued)

    No. UniGene ID Symbol Gene Name Ratio

    236 Hs.368525   PDLIM1   PDZ and LIM domain 1 (elfin) 0.711

    237 Hs.497893   ENAH    Enabled homolog (Drosophila) 0.698

    238 Hs.467142   MYH14   Myosin, heavy polypeptide 14 0.685239 Hs.84359   GABARAP    GABA(A) receptor-associated protein 0.640

    Oncogenesis

    240 Hs.536850   FBXL4   F-box and leucine-rich repeat protein 4 3.392

    241 Hs.421597   VHL   Von Hippel Y Lindau tumor suppressor 2.322

    242 Hs.351316   TM4SF1   Transmembrane 4 L 6 Y family member 1 1.422

    243 Hs.534597   MAGEA2   Melanoma antigen family A, 2B 1.400

    244 Hs.116479   LOXL2   Lysyl oxidase Y like 2 0.489

    Transcription/regulation of transcription

    245 Hs.505004   TCEA2   Transcription elongation factor A (SII), 2 4.672

    246 Hs.486507   TBPL1   TBP-like 1 4.486

    247 Hs.410406   SMARCAD1   SWI/SNF-related, matrix-associated actin-dependent regulator of chromatin,subfamily a, containing DEAD/H box 1

    3.727

    248 Hs.98367   SOX17    SRY (sex-determining region Y)-box 17 3.585249 Hs.128067   WASPIP    Wiskott-Aldrich syndrome protein-interacting protein 3.575

    250 Hs.437056   SUPT5H    Suppressor of Ty 5 homolog (Saccharomyces cerevisiae) 3.337

    251 Hs.509140   BAZ1A   Bromodomain adjacent to zinc f inger domain, 1A 3.256

    252 Hs.264345   ZNF675   Zinc finger protein 675 2.949

    253 Hs.463456   NME2   Methionine sulfoxide reductase B3 2.321

    254 Hs.118964   GATAD2A   GATA zinc f inger domain containing 2A 2.018

    Development processes

    255 Hs.99141   COBL   Cordon-bleu homolog (mouse) 5.458

    256 Hs.159028   BTN2A1   Butyrophilin, subfamily 2, member A1 3.348

    257 Hs.494163   GDA   Guanine deaminase 3.148

    258 Hs.7879   IFRD1   Interferon-related developmental regulator 1 2.786

    259 Hs.326387   MORF4L2   Mortality factor 4 Y like 2 1.794

    260 Hs.501293   BSG    Basigin (Ok blood group) 1.742

    261 Hs.322901   SAS10   Disrupter of silencing 10 1.653

    262 Hs.288487   SMS    Spermine synthase 1.384

    263 Hs.438779   STARD6    START domain containing 6 0.526

    Transport 

    264 Hs.491611   SLC20A2   Solute carrier family 20 (phosphate transporter), member 2 4.028

    265 Hs.443826   MGC4399   PNC1 protein 3.473

    266 Hs.162121   COPA   Coatomer protein complex, subunit alpha 2.465

    267 Hs.477789   ATP1B3   ATPase, Na + /K + transporting, beta 3 polypeptide 2.322

    268 Hs.73769   FOLR1   Folate receptor 1 (adult) 2.317

    269 Hs.568347   FTH1   Ferritin, heavy polypeptide 1 2.314

    270 Hs.179522   SLC2A8   Solute carrier family 2 (facilitated glucose transporter), member 8 2.223

    271 Hs.567337   SLC22A3   Solute carrier family 22 (extraneuronal monoamine transporter), member 3 2.189272 Hs.78888   DBI    Diazepam-binding inhibitor (GABA receptor modulator, acyl-coenzyme A Y  binding

     protein)2.139

    273 Hs.200600   SCAMP3   Secretory carrier membrane protein 3 2.020

    274 Hs.290404   SLC25A3   Solute carrier family 25 (mitochondrial carrier; phosphate carrier), member 3 1.899

    275 Hs.491597   VDAC3   Voltage-dependent anion channel 3 1.800

    276 Hs.9573   ABCF1   ATP-binding cassette, subfamily F (GCN20), member 1 1.742

    277 Hs.467701   ODC1   Ornithine decarboxylase 1 1.640

    278 Hs.500761   SLC16A3   Solute carrier family 16 (monocarboxylic acid transporters), member 3 1.377

    (Continued on next page)

    International Journal of Gynecological Cancer    &  Volume 19, Number 4, May 2009   Microarray Analysis Identifies Genes 

    *  2009 IGCS and ESGO   559

  • 8/17/2019 Microarray Analysis Identifies Differentially

    14/17Copyright @ 2009 by IGCS and ESGO. Unauthorized reproduction of this article is prohibited.

    TABLE 3. (Continued)

    No. UniGene ID Symbol Gene Name Ratio

    Cell communication

    279 Hs.49774   PTPRM    Protein tyrosine phosphatase, receptor type, M 3.370

    280 Hs.405410   OGT O-linked   N -acetylglucosamine (GlcNAc) transferase (UDP- N -acetylglucosamine: polypeptide- N -acetylglucosaminyl transferase)

    1.535

    281 Hs.209983   STMN1   Stathmin 1/oncoprotein 18 1.385

    Biological process unclassified 

    282 Hs.258576   CLDN12   Claudin 12 7.828

    283 Hs.437599   HPS5   Hermansky-Pudlak syndrome 5 7.463

    284 Hs.400625   GRWD1   Glutamate-rich WD repeat containing 1 7.298

    285 Hs.162032   HBP1   HMG-box transcription factor 1 6.995

    286 Hs.524082   NOD9   NOD9 protein 6.918

    287 Hs.527412   ASAH1 N  -Acylsphingosine amidohydrolase (acid ceramidase) 1 6.660

    288 Hs.496191   TINF2   TERF1 (TRF1)-interacting nuclear factor 2 5.268

    289 Hs.107622   SRFBP1   Serum response factor binding protein 1 5.314

    290 Hs.388918   RECK    Reversion-inducing cysteine-rich protein with kazal motifs 5.033

    291 Hs.32018   SNAPAP    SNAP-associated protein 4.997

    292 Hs.157106   JMJD2C    Jumonji domain containing 2C 4.798

    293 Hs.533736   RBM7    RNA-binding motif protein 7 4.744

    294 Hs.195710   ZNF503   Zinc finger protein 503 4.583

    295 Hs.523715   VPS37C    Vacuolar protein sorting 37C (yeast) 4.358

    296 Hs.493739   UBAP2   Ubiquitin-associated protein 2 4.220

    297 Hs.468702   COMMD1   Copper metabolism (Murr1) domain containing 1 4.070

    298 Hs.385998   WDHD1   WD repeat and HMG-box DNA-binding protein 1 4.018

    299 Hs.83916   NDUFA5   NADH dehydrogenase (ubiquinone) 1 alpha subcomplex, 5, 13 kd 3.982

    300 Hs.104320   GOLGA5   Golgi autoantigen, golgin subfamily a, 5 3.885

    301 Hs.514330   EME1   Essential meiotic endonuclease 1 homolog 1 (Schizosaccharomyces pombe) 3.851

    302 Hs.513315   NUDT16L1   Nudix (nucleoside diphosphate linked moiety X)-type motif 16 Y like 1 3.543

    303 Hs.534398   COMMD3   COMM domain containing 3 3.443304 Hs.127432   DTWD1   DTW domain containing 1 3.072

    305 Hs.500756   GOT1   Glutamic-oxaloacetic transaminase 1, soluble (aspartate aminotransferase 1) 3.028

    306 Hs.57898   RG9MTD1   RNA (guanine-9-) methyltransferase domain containing 1 2.998

    307 Hs.117780   KCNS1   Potassium voltage-gated channel, delayed-rectifier, subfamily S, member 1 2.989

    308 Hs.443650   JARID1B   Jumonji, AT-rich interactive domain 1B (RBP2-like) 2.805

    309 Hs.7570   CNO   Cappuccino homolog (mouse) 2.797

    310 Hs.533543   F8A1   Coagulation factor VIII-associated (intronic transcript) 1 2.776

    311 Hs.444770   SH3KBP1   SH3-domain kinase-binding protein 1 2.761

    312 Hs.2430   VPS72   Vacuolar protein sorting 72 (yeast) 2.723

    313 Hs.515317   IMPACT    Impact homolog (mouse) 2.680

    314 Hs.103561   ARL6IP4   ADP-ribosylation Y like factor 6 interacting protein 4 2.637

    315 Hs.499620   GEMIN4   Gem (nuclear organelle) associated protein 4 2.449

    316 Hs.527874   PSRC2   Proline/serine-rich coiled-coil 2 2.489

    317 Hs.152173   ANAPC4   Anaphase promoting complex subunit 4 2.438

    318 Hs.129614   TMEM27    Transmembrane protein 27 2.402

    319 Hs.483305   HINT1   Histidine triad nucleotide binding protein 1 2.317

    320 Hs.356440   CCDC72   Coiled-coil domain containing 72 2.316

    321 Hs.177530   ATP5E    ATP synthase, H + transporting, mitochondrial F1 complex, epsilon subunit 2.316

    322 Hs.101007   DCUN1D3   DCN1, defective in cullin neddylation 1, domain containing 3 (Saccharomycescerevisiae)

    2.093

    323 Hs.434207   HARS2   Histidyl-tRNA synthetase 2 2.069

    324 Hs.483136   COMMD10   COMM domain containing 10 2.062

     Min et al    International Journal of Gynecological Cancer    &  Volume 19, Number 4, May 2009

    560   *  2009 IGCS and ESGO

  • 8/17/2019 Microarray Analysis Identifies Differentially

    15/17Copyright @ 2009 by IGCS and ESGO. Unauthorized reproduction of this article is prohibited.

    The function of the HPV E6 protein that is most clearly linked tocarcinogenesis is the targeted degradation of p53, which is de- pendent on the E6AP ubiquitin ligase, the member of a family of ubiquitin ligases (or E3 enzymes). Kelley et al33 found that   E6 and   E6AP   siRNAs elicited nearly identical alterations in thetranscriptional profile of HeLa, Caski, and SiHa cell line. Some of the expression alterations were apparent secondary effects of p53stabilization, whereas the basis of most other changes was not rec-oncilable with previously proposed E6 functions. They concluded that E6AP mediates a broad spectrum of E6 functions, includingvirtually all functions that impact on the transcriptional program of HPV-positive cell lines. It is noticeable that in our study, theexpression of  E6AP   gene was down-regulated (0.532 times) after  E6  was inhibited. Therefore, we presume that both genes can affect each other and show a coincident trend and play a role in p53 stab-

    ilization together. The phenomenon had not been reported before,so further validation and study are needed.

    Another differentially expressed gene was the ubiquitin ligaseUBE2C . It is known that ubiquitin-dependent proteolysis by the26S proteasome plays a pivotal role in cell cycle progression and in tumorigenesis. Among the  E2  gene family, the expression levelof  UBE2C   was extremely low in many of the normal tissues but  prominent in most cancerous cell lines. Intriguingly,  UBE2C   wasexpressed at high levels in primary tumors derived from the lung,stomach, uterus, and bladder as compared with their correspondingnormal tissues, suggesting that  UBE2C   is involved in tumorigen-esis or progression of the tumor.34

    Other groups have shown by microarray analysis that ex- pression of E6 down-regulated a large number of genes involved in keratinocyte differentiation, including several genes such as

    TABLE 3. (Continued)

    No. UniGene ID Symbol Gene Name Ratio

    325 Hs.502745   FADS2   Fatty acid desaturase 2 1.994

    326 Hs.494691   PFN1   Profilin 1 1.990

    327 Hs.503546   FADS1   Fatty acid desaturase 1 1.905328 Hs.505735   NACA   Nascent-polypeptide Y associated complex alpha polypeptide 1.824

    329 Hs.511138   TMEM87A   Transmembrane protein 87A 1.777

    330 Hs.345694   KCMF1   Potassium channel modulatory factor 1 1.754

    331 Hs.83758   CKS2   CDC28 protein kinase regulatory subunit 2 1.729

    332 Hs.308122   ITPK1   Inositol 1,3,4-triphosphate 5/6 kinase 1.728

    333 Hs.546285   RPLP0   Ribosomal protein, large, P0 1.718

    334 Hs.98484   ILDR1   Immunoglobulin-like domain containing receptor 1 1.716

    335 Hs.201641   BASP1   Brain abundant, membrane attached signal protein 1 1.677

    336 Hs.416998   MRPL18   Mitochondrial ribosomal protein L18 1.663

    337 Hs.418241   MT2A   Metallothionein 2A 1.656

    338 Hs.71465   SQLE    Squalene epoxidase 1.652

    339 Hs.69855   CSDE1   Cold shock domain containing E1, RNA binding 1.555

    340 Hs.483765   SCGB3A2   Secretoglobin, family 3A, member 2 1.544

    341 Hs.304613   NDUFB4   NADH dehydrogenase (ubiquinone) 1 beta subcomplex, 4, 15 kd 1.500

    342 Hs.406515   NQO1   NAD(P)H dehydrogenase, polymer 1 1.487

    343 Hs.50252   MRPL32   Mitochondrial ribosomal protein L32 1.479

    344 Hs.472185   NDUFS5   NADH dehydrogenase (ubiquinone) Fe-S protein 5, 15 kd (NADH-coenzymeQ reductase)

    1.466

    345 Hs.438064   FN5   FN5 protein 1.436

    346 Hs.91161   PFDN4   Prefoldin subunit 4 1.434

    347 Hs.346868   EBNA1BP2   EBNA1 binding protein 2 1.430

    348 Hs.14559   CEP55   Centrosomal protein, 55 kd 1.410

    349 Hs.484991   HIST1H2BO   Histone 1, H2bo 0.711

    350 Hs.121592   AP1S2   Adaptor-related protein complex 1, sigma 2 subunit 0.701

    351 Hs.5836   MRPS23   Mitochondrial ribosomal protein S23 0.699

    352 Hs.21691   GPR75   G protein-coupled receptor 75 0.667

    353 Hs.502378   LENG8   Leukocyte receptor cluster (LRC) member 8 0.661

    354 Hs.202001   KIAA1012   Hypothetical protein LOC284242 0.661

    355 Hs.513044   CSPG4   Chondroitin sulfate proteoglycan 4 (melanoma associated) 0.641

    356 Hs.399984   WDR75   WD repeat domain 75 0.585

    357 Hs.436527   ANAPC1   Anaphase promoting complex subunit 1 0.580

    358 Hs.54056   FAM53C    Cell division cycle 25C 0.551

    359 Hs.493796   RUSC2   RUN and SH3 domain containing 2 0.535

    CDC, Cell division cycle; CMP, cytidine 5¶-monophosphate: UDP, uridine 5¶-diphosphate; tRNA, transfer RNA.

    International Journal of Gynecological Cancer    &  Volume 19, Number 4, May 2009   Microarray Analysis Identifies Genes 

    *  2009 IGCS and ESGO   561

  • 8/17/2019 Microarray Analysis Identifies Differentially

    16/17Copyright @ 2009 by IGCS and ESGO. Unauthorized reproduction of this article is prohibited.

    small proline-rich protein genes, elafin, stratum corneum chymo-tryptic enzyme, involucrin, and keratinocyte transglutaminase, that  play a role in the formation of the cornified envelope.35,36 Most of these genes were down-regulated regardless of p53 status. We alsoobserved that keratin family members   KRT18   (2.322 times), KRT4 (5.623 times), and  KRT6E  (3.525 times) were all up-regulated after   E6   was inhibited, signifying that HeLa cells may show atrend toward normal keratinocyte differentiation after  E6  oncogeneinhibition.

    Expression of E6 has also been shown to down-regulate ex- pression of genes involved in immune responses such as interferon-inducible genes. It has been reported that HPV-16, HPV-31, E6,and E7 can down-regulate genes involved in immune responsessuch as the interferon-regulated gene  Stat-1.37 The differential ex- pression of the interferon-inducible gene  Staf50   and several genesinvolved in inflammation such as TCIRG1, HLA-G , and  XTP7  wereobserved in our experiment, suggesting that HPV-18 E6 also playsa role in regulation of immune/inflammatory response.

    From the present work, it can be concluded that cellular ap-optosis induced by HPV-18  E6  siRNA mainly depends on the P53and ubiquitin proteolysis pathway to regulate gene expression, con-sequently inhibiting cell proliferation and promoting cell apoptosis.Meanwhile , the activation of antioncogene and upper regulation of immunization-related genes signified the degression of the malig-nant extent of tumor cells after   E6  interferences. In addition, theapproach of using transcriptional profiles to reveal pathways affected  by E6 proves useful.

    ACKNOWLEDGMENTThe authors thank Dapeng Ding and Jueyu Zhou for con-

    tributions to the microarray experiments.

    REFERENCES1. van Doorn LJ, Kleter B, Quint WG. Molecular detection and genotyping

    of human papillomavirus.  Expert Rev Mol Diagn. 2001;1:394 Y 402.

    2. van der Graaf Y, Molijn A, Doornewaard H, et al. Human papillomavirus and the long-term risk of cervical neoplasia. Am J  Epidemiol . 2002;156:158 Y 164.

    3. Walboomers JM, Jacobs MV, Manos MM, et al. Human papillomavirusis a necessary cause of invasive cervical cancer worldwide.  J Pathol .1999;189:1 Y 3.

    4. zur Hausen H. Papillomaviruses in human cancers. Proc Assoc Am Physicians. 1999;111:581 Y 587.

    5. Bosch FX, Manos MM, Munoz N, et al. Prevalence of human papillomavirus in cervical cancer: a worldwide perspective. International

     biological study on cervical cancer (IBSCC) Study Group. J Natl Cancer Inst . 1995;87:796 Y 802.

    6. Pina-Sanchez P, Hernandez-Hernandez D, Lopez-Romero R, et al.Human papillomavirus Y specific viral types are common in Mexicanwomen affected by cervical lesions. Int J Gynecol Cancer . 2006;16:1 Y 7.

    7. Tong SY, Lee YS, Park JS, et al. Human papillomavirus genotype asa prognostic factor in carcinoma of the uterine cervix. Int J Gynecol 

    Cancer . 2007;17:1307 Y 1313.

    8. Burger RA, Monk BJ, Kurosaki T, et al. Human papillomavirus type 18:association with poor prognosis in early stage cervical cancer.  J Natl Cancer Inst . 1996;88:1361 Y 1368.

    9. Schwartz SM, Daling JR, Shera KA, et al. Human papillomavirus and  prognosis of invasive cervical cancer: a population-based study. J ClinOncol . 2001;19:1906 Y 1915.

    10. Lombard I, Vincent-Salomon A, Validire P, et al. Human papillomavirusgenotype as a major determinant of the course of cervical cancer.

     J Clin Oncol . 1998;16:2613 Y 2619.

    11. Zhang J, Rose BR, Thompson CH, et al. Associations betweenoncogenic human papillomaviruses and local invasive patterns incervical cancer.  Gynecol Oncol . 1995;57:70 Y 77.

    12. Rose BR, Thompson CH, Simpson JM, et al. Human papillomavirusdeoxyribonucleic acid as a prognostic indicator in early-stage cervicalcancer: a possible role for type 18.  Am J Obstet Gynecol . 1995;173:

    1461 Y 1468.

    13. Im SS, Wilczynski SP, Burger RA, et al. Early stage cervical cancerscontaining human papillomavirus type 18 DNA have more nodalmetastasis and deeper stromal invasion.  Clin Cancer Res. 2003;9:4145 Y 4150.

    14. Spitkovsky D, Aengeneyndt F, Braspenning J, et al. p53-independent growth regulation of cervical cancer cells by the papillomavirus E6oncogene. Oncogene. 1996;13:1027 Y 1035.

    15. Scheffner M, Whitaker NJ. Human papillomavirus-induced carcinogenesis and the ubiquitin-proteasome system.  Semin Cancer 

     Biol . 2003;13:59 Y 67.

    16. Phelps WC, Yee CL, Munger K, et al. The human papillomavirustype 16 E7 gene encodes transactivation and transformation functionssimilar to those of adenovirus E1A.  Cell . 1988;53:539 Y 547.

    17. He Y, Huang L. Growth inhibition of human papillomavirus 16DNA-positive mouse tumor by antisense RNA transcribed from U6

     promoter. Cancer Res. 1997;57:3993 Y 3999.

    18. Tan TM, Ting RC. In vitro and in vivo inhibition of human papillomavirus type 16 E6 and E7 genes.  Cancer Res. 1995;55:4599 Y 4605.

    19. Sima N, Wang S, Wang W, et al. Antisense targeting human papillomavirus type 16 E6 and E7 genes contributes to apoptosisand senescence in SiHa cervical carcinoma cells. Gynecol Oncol .2007;106:299 Y 304.

    20. Blagosklonny MV, el-Deiry WS. In vitro evaluation of a p53-expres sing adenovirus as an anti-can cer drug.   Int J Cancer .1996;67:386 Y 392.

    21. Butz K, Ristriani T, Hengstermann A, et al. siRNA targeting of theviral E6 oncogene efficiently kills human papillomavirus-positivecancer cells. Oncogene. 2003;22:5938 Y 5945.

    22. Hallez S, Simon P, Maudoux F, et al. Phase I/II trial of immunogenicityof a human papillomavirus (HPV) type 16 E7 protein-based vaccine

    in women with oncogenic HPV-positive cervical intraepithelialneoplasia.  Cancer Immunol Immunother . 2004;53:642 Y 650.

    23. Mantovani F, Banks L. The human papillomavirus E6 protein and its contribution to malignant progression.  Oncogene. 2001;20:7874 Y 7887.

    24. el-Deiry WS. p21/p53, cellular growth control and genomic integrity.Curr Top Microbiol Immunol . 1998;227:121 Y 137.

    25. Syrjänen SM, Syrjänen KJ. New concepts on the role of human papillomavirus in cell cycle regulation. Ann Med . 1999;31:175 Y 187.

    26. Southern SA, Herrington CS. Disruptions of cell cycle control by human papillomaviruses with special reference to cervical carcinoma. Int J Gynecol Cancer . 2000;10:263 Y 274.

    27. Tamura K, Kanaoka Y, Jinno S, et al. Cyclin G: a new mammalian cyclinwith homology to fission yeast Cig1. Oncogene. 1993;8:2113 Y 2118.

    FIGURE 5.  Comparison of 5 differential expression genesbetween the microarray and RT-PCR analyses.

     Min et al    International Journal of Gynecological Cancer    &  Volume 19, Number 4, May 2009

    562   *  2009 IGCS and ESGO

  • 8/17/2019 Microarray Analysis Identifies Differentially

    17/17

    28. Kimura SH, Ikawa M, Ito A, et al. Cyclin G1 is involved in G2/M arrest in response to DNA damage and in growth control after damagerecovery.  Oncogene. 2001;20:3290 Y 3300.

    29. Zhao L, Samuels T, Winckler S, et al. Cyclin G1 has growth inhibitoryactivity linked to the ARF-Mdm2-p53 and pRb tumor suppressor 

     pathways. Mol Cancer Res. 2003;1:195 Y 206.

    30. Zimmermann KC, Bonzon C, Green DR. The machinery of programmed cell death. Pharmacol Ther . 2001;92:57 Y 70.

    31. Afford S, Randhawa S. Apoptosis. Mol Pathol . 2000;53:55 Y 63.

    32. Hollowood AD, Lai T, Perks CM, et al. IGFBP-3 prolongs the p53response and enhances apoptosis following UV irradiation. Int J Cancer .2000;88:336 Y 341.

    33. Kelley ML, Keiger KE, Lee CJ, et al. The global transcriptional effects

    of the human papillomavirus E6 protein in cervical carcinoma cell linesare mediated by the E6AP ubiquitin ligase.  J Virol . 2005;9:3737 Y 3747.

    34. Okamoto Y, Ozaki T, Miyazaki K, et al. UbcH10 is the cancer-related E2 ubiquitin-conjugating enzyme.  Cancer Res. 2003;63:4167 Y 4173.

    35. Duffy CL, Phillips SL, Klingelhutz AJ. Microarray analysis identifiesdifferentiation-associated genes regulated by humanpapillomavirus type16 E6. Virology. 2003;314:196 Y 205.

    36. Chang YE, Laimins LA. Microarray analysis identifiesinterferon-inducible genes and Stat-1 as major transcriptional targets of human papillomavirus type 31. J Virol . 2000;74:4174 Y 4182.

    37. Wall SJ, Edward DR. Quantitative reverse transcription-polymerasechain reaction (TR-PCR): a comparison of prime-dropping, competitive,and real-time RT-PCRs.  Anal Biochem. 2002;300:269 Y 273.

    International Journal of Gynecological Cancer    &  Volume 19, Number 4, May 2009   Microarray Analysis Identifies Genes 

    *  2009 IGCS and ESGO 563