Skip to main content
PLOS ONE logoLink to PLOS ONE
. 2010 Oct 1;5(10):e13054. doi: 10.1371/journal.pone.0013054

cDNA Microarray Gene Expression Profiling of Hedgehog Signaling Pathway Inhibition in Human Colon Cancer Cells

Ting Shi 1, Tapati Mazumdar 1, Jennifer DeVecchio 1, Zhong-Hui Duan 2, Akwasi Agyeman 1, Mohammad Aziz 1,¤, Janet A Houghton 1,*
Editor: Terence Lee3
PMCID: PMC2948497  PMID: 20957031

Abstract

Background

Hedgehog (HH) signaling plays a critical role in normal cellular processes, in normal mammalian gastrointestinal development and differentiation, and in oncogenesis and maintenance of the malignant phenotype in a variety of human cancers. Increasing evidence further implicates the involvement of HH signaling in oncogenesis and metastatic behavior of colon cancers. However, genomic approaches to elucidate the role of HH signaling in cancers in general are lacking, and data derived on HH signaling in colon cancer is extremely limited.

Methodology/Principal Findings

To identify unique downstream targets of the GLI genes, the transcriptional regulators of HH signaling, in the context of colon carcinoma, we employed a small molecule inhibitor of both GLI1 and GLI2, GANT61, in two human colon cancer cell lines, HT29 and GC3/c1. Cell cycle analysis demonstrated accumulation of GANT61-treated cells at the G1/S boundary. cDNA microarray gene expression profiling of 18,401 genes identified Differentially Expressed Genes (DEGs) both common and unique to HT29 and GC3/c1. Analyses using GenomeStudio (statistics), Matlab (heat map), Ingenuity (canonical pathway analysis), or by qRT-PCR, identified p21Cip1 (CDKN1A) and p15Ink4b (CDKN2B), which play a role in the G1/S checkpoint, as up-regulated genes at the G1/S boundary. Genes that determine further cell cycle progression at G1/S including E2F2, CYCLIN E2 (CCNE2), CDC25A and CDK2, and genes that regulate passage of cells through G2/M (CYCLIN A2 [CCNA2], CDC25C, CYCLIN B2 [CCNB2], CDC20 and CDC2 [CDK1], were down-regulated. In addition, novel genes involved in stress response, DNA damage response, DNA replication and DNA repair were identified following inhibition of HH signaling.

Conclusions/Significance

This study identifies genes that are involved in HH-dependent cellular proliferation in colon cancer cells, and following its inhibition, genes that regulate cell cycle progression and events downstream of the G1/S boundary.

Introduction

Hedgehog (HH) signaling plays a critical role in a variety of normal cellular processes. It is pivotal in embryogenesis, regulation of the epithelial-to-mesenchymal transition, the patterning of a diverse range of vertebrate structures in a variety of organs, maintenance of adult tissue homeostasis, tissue repair, cellular proliferation, and in cell survival [1], [2], [3], [4], [5], [6], [7], [8], [9]. The canonical HH pathway is also critical to normal mammalian gastrointestinal development, where it is involved in the coordinate regulation of differentiation of normal intestinal villi [10], [11], [12]. Thus, in the normal gastrointestinal tract, HH ligands are induced in the differentiated cells around the villous surface, generating a negative feedback loop to inhibit canonical WNT signaling in the basal cells of the crypt, thereby protecting differentiated cells from the proliferative effects of WNT [13]. Activation of the canonical HH signaling pathway comprises the binding of HH ligands to the membrane receptor Patched (PTCH1), which becomes internalized leading to the activation of the signaling molecule Smoothened (SMO) via release from PTC-mediated suppression. SMO activates the final arbiter of HH signaling, the GLI family of transcription factors that bind to the GACCACCCA-like consensus binding element in promoter sequences to transcriptionally regulate HH target genes [3], [14], [15]. GLI1 and GLI2, the transcriptional activators of HH signaling, possess distinct as well as overlapping functions that involve activator (GLI1 and GLI2) or repressor (GLI2) activities [16]; however, their roles in the regulation of HH-driven cellular proliferation, survival or cell death processes are poorly understood. Historically, GLI1 has been considered the most reliable marker of HH pathway activity, however GLI2 appears to be the primary activator of HH signaling, with GLI1 as a transcriptional target of GLI2 [3], [7], leading to augmentation of HH signaling both quantitatively as well as qualitatively [16]. An important feature of GLI proteins is that their biological activity is context-dependent, influenced by the cellular environment [17], [18].

Activation of the canonical HH signaling cascade is aberrantly activated and well known to play a critical role in oncogenesis and maintenance of the malignant phenotype in several types of human cancers. Such activation involves amplification of GLI1 or GLI2, mutations in PTC or SMO, or dysregulated gene expression [3], [4]; these malignant cells are also sensitive to the small molecule inhibitor that targets SMO, cyclopamine [4], [19], [20], [21], [22], [23]. Colon carcinomas are thought to derive from constitutive activation of WNT signaling by mutation of the APC or β-CATENIN genes, while the involvement of the HH signaling pathway is not as clear. In gastrointestinal malignancies, transcriptional up-regulation of HH ligands has been identified as the predominant activator of HH signaling in these diseases (reviewed in [3]). In addition, there is emerging evidence that HH signaling is involved in colorectal carcinogenesis [24], [25], colon carcinoma stem cell self renewal, and in the metastatic behavior of advanced colon cancers [26]. However, genomic approaches to elucidate the role of HH signaling in cancers in general are lacking, regulatory genes downstream of GLI1 and GLI2 that function in cellular proliferation, survival, and maintenance of the malignant HH phenotype remain incompletely characterized [5], and data derived on HH signaling in colon cancer is extremely limited.

Cellular proliferation is driven by progression of cells through the cell cycle consisting of sequential passage through G1, S, G2 and M phases. Cyclin-dependent kinases (CDKs) associate with cyclins to drive the cell cycle machinery [27], [28]. Thus, CDK2 associates with CYCLIN E at the G1/S transition and with CYCLIN A during S-phase, CDK4 and CDK6 bind to CYCLIN D during progression at G1/S, while CDC2 complexes with CYCLIN A at G2, and with CYCLIN B during the G2/M transition. CDC25 family members also regulate cell cycle progression through dephosphorylation of the CDKs [29], [30]. CDK inhibitors, including p21Cip1 [29], [31] and p15Ink4b [[32], bind to cyclin-CDK complexes during the cell cycle transition, in particular at G1/S (p21Cip1, p15Ink4b;[32]) and G2/M (p21Cip1;[29]), and can also induce cell cycle arrest at the G1/S boundary following cytostatic signals through functional inhibition of cyclin-CDK complexes. The E2F family of transcription factors also regulates the expression of genes required for the G1/S transition, in particular genes involved in the activation of the DNA replication machinery, and DNA repair [33].

cDNA microarray technology has provided the ability to study the expression of thousands of genes simultaneously, and is an important tool in the dissection of signal transduction pathways. For the HH signaling cascade, HH/GLI target gene expression has been examined following EGF stimulation [6] or inducible GLI1 [16] or GLI2 [9] gene activation in human keratinocytes, or in GLI1-induced cell transformation [7]. To identify unique downstream targets of the GLI genes that function in cellular proliferation in the context of colon carcinoma, we employed a small molecule inhibitor of both GLI1 and GLI2, GANT61, identified in a cell-based small molecule screen for inhibitors of GLI1-mediated transcription [34]. GANT61 acts in the nucleus to block GLI1 function, inhibits both GLI1- and GLI2- mediated transcription, and demonstrates a high degree of selectivity for HH/GLI signaling [34]. Thus, GANT61 acts downstream of cyclopamine (targeting SMO) to inhibit the final determinants of HH transcriptional regulation.

In two human colon carcinoma cell lines, HT29 and GC3/c1, inhibiting the HH signaling pathway using GANT61 decreased expression of GLI1, GLI2 and the HH ligand receptor, PTCH1, and inhibited proliferation by inducing cellular accumulation at the G1/S boundary 24 hr after treatment, determined by flow cytometric analysis. On further detailed analysis using cDNA microarray gene expression profiling and quantitative Real-Time PCR, p21Cip1 (CDKN1A) and p15Ink4b (CDKN2B), that can elicit the G1/S checkpoint, were up-regulated, while genes that further determine entry from G1 to S-phase including E2F2, CYCLIN E2 (CCNE2), CDC25A and CDK2 were decreased in expression. Concomitant with decreased G1 to S-phase progression, decreased expression of CYCLIN A2 (CCNA2), CDC25C, CYCLIN B2 (CCNB2), CDC20 and CDC2 (CDK1), that regulate the passage of cells through G2/M were also demonstrated. Additional novel genes that are involved in stress response, and the response to DNA damage, not previously identified following termination of HH signaling in human cancer cells, include the early response genes DDIT2 (GADD45G), DDIT3 (GADD153), DDIT4 (REDD1), PPP1R15A (GADD34) and ATF3 that were significantly up-regulated. Genes involved in DNA synthesis and repair (TYMS, TOP2A, TK1, POLE, POLE2), and additional novel genes involved in S-phase progression or DNA damage responses that were significantly down-regulated, include KIAA0101 (p15 [PAF]), Replication Factor C variants 2, 3, 4, 5, CDT1, the E2F transcription factors CDCA4 and TFDP1, MDC1, FANCD2, PCNA, and the genes involved in DNA repair, RAD51C (XRCC3), RAD54B, RAD51 and HELLS. This study has therefore identified genes that are regulated during the termination of HH-dependent cellular proliferation and survival in colon cancer cells, and involves genes associated with G1/S-phase arrest, DNA damage and stress responses.

Results

GANT61 induces down-regulated expression of GLI genes and accumulation of human colon carcinoma cells at G1/S

In HT29 and GC3/c1 cells treated with GANT61 (20 µM) for up to 48 hr, expression of the target genes GLI1 and GLI2 were both down-regulated, and also the HH ligand receptor PTCH1, as determined by qRT-PCR (Figure 1A). Subsequently, HT29 or GC3/c1 cells were treated, in duplicate, with GANT61 (20 µM) followed by PI staining and flow cytometric analysis for the determination of cell cycle distribution between G1, S and G2/M phases (Figure 1B). In both cell lines, cells accumulated in G1, 24 hr after treatment with GANT61. In GANT61-treated HT29 cells, a 20% increase in G1-phase cells was associated with a corresponding decrease in cells within the G2/M phase (14%) and in S-phase (5%), consistent with a G1/S checkpoint arrest. In GC3/c1 cells, an 8% increase in G1-phase cells at 24 hr after GANT61 treatment also corresponded with a similar reduction in cells in the G2/M phase of the cell cycle (Figure 1B).

Figure 1. Expression of GLI1, GLI2, PTCH1 and cell cycle distribution of HT29 and GC3/c1 cells.

Figure 1

Cells were treated for up to 48 hr with GANT61 (20 µM) or with 0.2% DMSO (vehicle control). (A) qRT-PCR of GLI1, GLI2 and PTCH1 genes from time 0 to 48 hr. (B) DNA was extracted at 24 hr after treatment, stained with propidium iodide, and subsequently analyzed for the effects of inhibition of HH signaling on phase distribution of cells within the cell cycle by flow cytometry.

cDNA microarray analyses identify changes in gene expression both common and unique to HT29 and GC3/c1 following GANT61 treatment

To delineate the changes in gene expression in HT29 and GC3/c1 human colon carcinoma cell lines in response to treatment with the GLI1/GLI2 antagonist, GANT61, the expression of 18,401 human genes was profiled in control cells treated with vehicle (0.2% DMSO) and in cells treated with GANT61 (20 µM) for 24 hr. Genes with a False Discovery Rate (FDR)-adjusted p-value of <0.001 and fold change >1.5 were considered Differentially Expressed Genes (DEGs) induced by GANT61 relative to the vehicle control, of which 1,368 genes were differentially expressed in HT29, and 1,002 genes in GC3/c1 cells (Figure 2). 755 genes or 558 genes were up-regulated, and 613 or 444 genes were down-regulated, in HT29 and GC3/c1 cells, respectively. 763 and 397 genes were differentially expressed and unique to HT29 or GC3/c1, respectively. Of the 763 DEGs unique to HT29, 459 (60%) were up-regulated and 304 (40%) were down-regulated. Similarly, of the 397 DEGs unique to GC3/c1, 262 (66%) were up-regulated and 135 (34%), down-regulated. In contrast, 605 genes representing 3.4% of all genes were differentially expressed that were common to both cell lines; of these, 296 were up-regulated (49%), and 309 (51%) were down-regulated (Figure 2). All genes common to both HT29 and GC3/c1 that were significantly up-regulated or down-regulated (p<0.001) are listed in Tables 1 and 2, respectively.

Figure 2. Venn diagram summarizing Differentially Expressed Genes (DEGs) in GANT61-treated HT29 and GC3/c1 cells.

Figure 2

Cells were treated with GANT61 (20 µM) or vehicle alone (0.2% DMSO) for 24 hr, and total RNA extracted as described in Materials and Methods. Changes in gene expression were determined by cDNA microarray gene profiling using the Illumina Human-ref8 V3.0 Bead-Chip array. Genes with a False Discovery Rate (FDR)-adjusted p-value (p<0.001) and fold change >1.5 were considered DEGs. Upper panel: up-regulated genes. Lower panel: down-regulated genes. Differential expression for the total, unique up-regulated, unique down-regulated, common up-regulated, and common down-regulated DEGs, are shown.

Table 1. Up-regulated genes (p<0.001) common in GANT61-treated HT29 and GC3/c1 cells.

ACCESSION NUMBER GENE SYMBOL DEFINITION Fold Change ACCESSION NUMBER GENE SYMBOL DEFINITION Fold Change
HT29 GC3/c1 HT29 GC3/c1
NM_080489.3 SDCBP2 syndecan binding protein 12.42 10.83 NM_001008213.1 OPTN optineurin 2.63 2.37
NM_001042483.1 NUPR1 nuclear protein 1 7.96 3.53 NM_000431.1 MVK mevalonate kinase 2.61 2.77
NM_021187.2 CYP4F11 cytochrome P450 7.73 4.27 NM_001550.2 IFRD1 interferon-related developmental regulator 1 2.60 1.84
NM_001001870.1 C17orf91 chromosome 17 open reading frame 91 6.79 3.82 NM_025130.2 HKDC1 hexokinase domain containing 1 2.58 4.46
NM_004864.1 GDF15 growth differentiation factor 15 6.61 3.43 NM_002461.1 MVD mevalonate (diphospho) decarboxylase 2.57 5.36
NM_004083.4 DDIT3 DNA-damage-inducible transcript 3 6.36 2.35 NM_021626.1 SCPEP1 serine carboxypeptidase 1 2.55 1.79
NM_005130.3 FGFBP1 fibroblast growth factor binding protein 1 5.86 41.19 NM_012424.2 RPS6KC1 ribosomal protein S6 kinase 2.53 2.03
NM_022060.2 ABHD4 abhydrolase domain containing 4 5.65 3.07 NM_052839.2 PANX2 pannexin 2 2.53 5.61
NM_015526.1 CLIP3 CAP-GLY domain containing linker protein 3 5.25 2.74 NM_021173.2 POLD4 polymerase (DNA-directed) 2.51 1.71
NM_000389.2 CDKN1A cyclin-dependent kinase inhibitor 1A 5.21 2.02 NM_020400.4 LPAR5 lysophosphatidic acid receptor 5 2.50 2.17
NM_002298.2 LCP1 lymphocyte cytosolic protein 1 5.02 8.60 NM_007075.3 WDR45 WD repeat domain 45 2.50 1.61
NM_019058.2 DDIT4 DNA-damage-inducible transcript 4 4.91 3.91 NM_001909.3 CTSD cathepsin D 2.50 1.68
NM_031412.2 GABARAPL1 GABA(A) receptor-associated protein like 1 4.73 4.81 NM_022157.2 RRAGC Ras-related GTP binding C 2.48 2.80
NM_033285.2 TP53INP1 tumor protein p53 inducible nuclear protein 1 4.71 2.80 NM_138573.2 NRG4 neuregulin 4 2.48 2.69
NM_001001342.1 BLOC1S2 biogenesis of lysosome-related organelles complex-1 4.68 3.95 NM_201545.1 LGALS8 lectin 2.46 2.81
NM_153282.1 HYAL1 hyaluronoglucosaminidase 1 4.57 22.29 NM_175738.3 RAB37 RAB37, member RAS oncogene family 2.40 3.53
NM_012385.1 P8 p8 protein 4.57 4.01 NM_012257.3 HBP1 HMG-box transcription factor 1 2.40 1.65
NM_016061.1 YPEL5 yippee-like 5 4.39 3.98 NM_001083617.1 RB1CC1 RB1-inducible coiled-coil 1 2.39 1.68
NM_145693.1 LPIN1 lipin 1 4.39 3.59 NM_153341.1 RNF19B ring finger protein 19B 2.38 1.61
NM_019600.1 KIAA1370 KIAA1370 4.13 2.73 NM_003129.3 SQLE squalene epoxidase 2.38 3.03
NM_021009.3 UBC ubiquitin C 4.08 7.52 NM_018045.5 BSDC1 BSD domain containing 1 2.38 2.34
NM_002084.3 GPX3 glutathione peroxidase 3 4.05 2.99 NM_003376.4 VEGFA vascular endothelial growth factor A 2.37 2.09
NM_003256.2 TIMP4 TIMP metallopeptidase inhibitor 4 4.04 2.02 NM_007028.3 TRIM31 tripartite motif-containing 31 2.37 6.54
NM_198336.1 INSIG1 insulin induced gene 1 4.02 9.90 NM_000527.2 LDLR low density lipoprotein receptor 2.35 3.83
NM_002517.2 NPAS1 neuronal PAS domain protein 1 3.95 2.76 NM_024717.3 MCTP1 multiple C2 domains, transmembrane 1 2.33 2.28
NM_001040619.1 ATF3 activating transcription factor 3 3.90 1.86 NM_198833.1 SERPINB8 serpin peptidase inhibitor 2.32 2.07
NM_006096.2 NDRG1 N-myc downstream regulated gene 1 3.89 2.63 NM_023039.2 ANKRA2 ankyrin repeat, family A (RFXANK-like) 2.31 1.82
NM_003670.1 BHLHB2 basic helix-loop-helix domain containing 3.82 3.68 NM_025047.1 ARL14 ADP-ribosylation factor-like 14 2.30 6.15
NM_002130.6 HMGCS1 3-hydroxy-3-methylglutaryl-Coenzyme A synthase 1 3.75 6.54 NM_001080391.1 SP100 SP100 nuclear antigen 2.29 1.90
NR_003086.1 HSD17B7P2 hydroxysteroid (17-beta) dehydrogenase 7 pseudogene 2 3.74 2.83 NM_002395.3 ME1 malic enzyme 1 2.29 2.31
NM_001013680.1 LOC401233 similar to HIV TAT specific factor 1 3.74 2.10 NM_001360.2 DHCR7 7-dehydrocholesterol reductase 2.29 2.73
NM_006918.4 SC5DL sterol-C5-desaturase 3.68 3.41 NM_032421.2 CLIP2 CAP-GLY domain containing linker protein 2 2.27 1.79
NM_182980.2 OSGIN1 oxidative stress induced growth inhibitor 1 3.55 4.48 NM_014039.2 C11orf54 chromosome 11 open reading frame 54 2.27 1.62
NM_025182.2 KIAA1539 KIAA1539 3.49 2.62 NM_017585.2 SLC2A6 solute carrier family 2 2.27 2.35
NM_018677.2 ACSS2 acyl-CoA synthetase short-chain family member 2 3.46 4.68 NM_014330.2 PPP1R15A protein phosphatase 1 2.26 1.63
NM_001034850.1 FAM134B family with sequence similarity 134 3.44 1.90 NM_145306.2 C10orf35 chromosome 10 open reading frame 35 2.26 2.19
NM_153742.3 CTH cystathionase (cystathionine gamma-lyase) 3.38 2.13 NM_003831.3 RIOK3 RIO kinase 3 2.25 1.93
NM_021158.3 TRIB3 tribbles homolog 3 3.34 3.71 NM_006997.2 TACC2 transforming, acidic coiled-coil containing protein 2 2.25 1.95
NM_000434.2 NEU1 sialidase 1 3.28 2.00 NM_001006932.1 RPS6KA2 ribosomal protein S6 kinase 2.24 1.68
NM_001005404.3 YPEL2 yippee-like 2 3.22 2.40 NM_000574.2 CD55 CD55 molecule 2.23 1.84
NM_004508.2 IDI1 isopentenyl-diphosphate delta isomerase 1 3.21 4.78 NM_018310.2 BRF2 subunit of RNA polymerase III transcription initiation factor 2.23 2.14
NM_003151.2 STAT4 signal transducer and activator of transcription 4 3.18 3.85 NM_001491.2 GCNT2 glucosaminyl (N-acetyl) transferase 2 2.22 4.08
NM_005165.2 ALDOC aldolase C 3.16 2.44 NM_003567.2 BCAR3 breast cancer anti-estrogen resistance 3 2.21 1.79
NM_014331.3 SLC7A11 solute carrier family 7 3.16 2.54 NM_005488.1 TOM1 target of myb1 2.20 1.89
NM_000104.2 CYP1B1 cytochrome P450 3.15 2.41 NM_004462.3 FDFT1 farnesyl-diphosphate farnesyltransferase 1 2.19 2.73
NM_002340.3 LSS lanosterol synthase 3.14 3.15 NM_019116.2 UBFD1 ubiquitin family domain containing 1 2.19 1.90
NM_078487.2 CDKN2B cyclin-dependent kinase inhibitor 2B 3.13 2.13 NM_007105.1 SLC22A18AS solute carrier family 22 2.18 1.81
NM_002201.4 ISG20 interferon stimulated exonuclease gene 20kDa 3.12 1.91 NM_182491.1 ZFAND2A zinc finger 2.17 2.17
NM_016315.2 GULP1 GULP, engulfment adaptor PTB domain containing 1 3.11 4.39 NM_181785.2 SLC46A3 solute carrier family 46 2.17 2.00
NM_017983.4 WIPI1 WD repeat domain 3.09 2.40 NM_024866.4 ADM2 adrenomedullin 2 2.16 1.77
NM_024165.1 PHF1 PHD finger protein 1 3.07 2.42 NM_021133.2 RNASEL ribonuclease L 2.15 2.28
NM_001995.2 ACSL1 acyl-CoA synthetase long-chain family member 1 3.06 2.86 NM_032548.2 ABTB1 ankyrin repeat and BTB (POZ) domain containing 1 2.14 1.58
NM_004354.1 CCNG2 cyclin G2 3.05 2.64 NM_002153.1 HSD17B2 hydroxysteroid (17-beta) dehydrogenase 2 2.14 2.98
NM_020739.2 CCPG1 cell cycle progression 1 3.04 1.88 NM_006033.2 LIPG lipase 2.13 2.86
NM_001017369.1 SC4MOL sterol-C4-methyl oxidase-like 3.03 4.04 NM_020919.2 ALS2 amyotrophic lateral sclerosis 2 2.11 1.77
NM_001031744.1 LOC158160 hypothetical protein LOC158160 3.02 2.35 NM_014701.2 KIAA0256 KIAA0256 gene product 2.11 2.30
NM_032409.2 PINK1 PTEN induced putative kinase 1 2.98 2.44 NM_001222.2 CAMK2G calcium/calmodulin-dependent protein kinase 2.11 2.12
NM_003811.2 TNFSF9 tumor necrosis factor (ligand) superfamily 2.98 3.04 NM_015271.2 TRIM2 tripartite motif-containing 2 2.11 1.79
NM_002357.2 MXD1 MAX dimerization protein 1 2.93 2.30 NM_174936.2 PCSK9 proprotein convertase subtilisin/kexin type 9 2.11 4.18
NM_000777.2 CYP3A5 cytochrome P450 2.92 1.77 NM_015713.3 RRM2B ribonucleotide reductase M2 B 2.10 1.66
NM_001098500.1 KIAA1217 KIAA1217 2.88 3.16 NM_005749.2 TOB1 transducer of ERBB2 2.09 1.58
NM_022119.3 PRSS22 protease 2.84 3.27 NM_004419.3 DUSP5 dual specificity phosphatase 5 2.08 3.61
NM_004669.2 CLIC3 Homo sapiens chloride intracellular channel 3 2.80 6.19 NM_033028.2 BBS4 Bardet-Biedl syndrome 4 2.08 2.04
NM_080491.1 GAB2 GRB2-associated binding protein 2 2.79 2.06 NM_001286.2 CLCN6 chloride channel 6 2.07 2.18
NM_178270.1 ATG4A ATG4 autophagy related 4 homolog A 2.79 1.92 XM_001132495.1 SLC26A11 PREDICTED:solute carrier family 26 2.07 1.87
NM_002770.2 PRSS2 protease, serine, 2 (trypsin 2) 2.79 1.64 NM_172070.2 ZNF650 zinc finger protein 650 2.07 1.90
NM_182608.2 ANKRD33 ankyrin repeat domain 33 2.74 1.91 NM_004567.2 PFKFB4 6-phosphofructo-2-kinase/fructose-2 2.07 1.93
NM_006705.2 GADD45G growth arrest and DNA-damage-inducible 2.74 3.01 NM_002555.3 SLC22A18 solute carrier family 22 2.06 1.62
NM_012326.2 MAPRE3 microtubule-associated protein 2.73 2.16 NM_001010990.1 HERPUD1 homocysteine-inducible, ubiquitin-like domain member 1 2.06 1.63
NM_001013251.1 SLC3A2 solute carrier family 3 2.72 2.40 NM_198867.1 ALKBH6 alkB, alkylation repair homolog 6 2.06 1.65
NM_016243.2 CYB5R1 cytochrome b5 reductase 1 2.72 2.52 NM_000271.3 NPC1 Niemann-Pick disease 2.05 2.18
NM_198129.1 LAMA3 laminin 2.65 3.36 NM_006454.2 MXD4 MAX dimerization protein 4 2.05 1.80
NM_000228.2 LAMB3 laminin 2.65 2.27 NM_021194.2 SLC30A1 solute carrier family 30 2.03 1.73
NM_006608.1 PHTF1 putative homeodomain transcription factor 1 2.64 2.38 NM_173843.1 IL1RN interleukin 1 receptor antagonist 2.03 10.01
NM_021229.3 NTN4 netrin 4 2.03 2.47 NM_152271.3 LONRF1 LON peptidase N-terminal domain and ring finger 1 1.72 3.04
NM_002064.1 GLRX glutaredoxin 2.03 1.75 NM_003314.1 TTC1 tetratricopeptide repeat domain 1 1.72 1.55
NM_005896.2 IDH1 isocitrate dehydrogenase 1 2.02 2.37 NM_005920.2 MEF2D myocyte enhancer factor 2D 1.71 1.85
NM_144498.1 OSBPL2 oxysterol binding protein-like 2 2.01 1.81 NM_016410.2 CHMP5 chromatin modifying protein 5 1.71 2.14
NM_005115.3 MVP major vault protein 1.99 1.82 NM_001748.3 CAPN2 calpain 2 1.70 2.16
NM_023938.4 C1orf116 chromosome 1 open reading frame 116 1.98 3.06 NM_004344.1 CETN2 centrin 1.70 1.54
NM_024311.2 MFSD11 major facilitator superfamily domain containing 11 1.98 1.80 NM_177947.2 ARMCX3 armadillo repeat containing, X-linked 3 1.70 1.76
NM_004433.3 ELF3 E74-like factor 3 1.97 3.72 NM_000158.2 GBE1 glucan 1.69 1.73
NM_031229.2 RBCK1 RanBP-type and C3HC4-type zinc finger containing 1 1.97 4.21 NM_052873.1 C14orf179 chromosome 14 open reading frame 179 1.69 1.52
NM_005564.3 LCN2 lipocalin 2 1.97 4.19 NM_020310.2 MNT MAX binding protein 1.69 1.51
NM_002956.2 CLIP1 CAP-GLY domain containing linker protein 1 1.96 1.62 NM_015367.2 BCL2L13 BCL2-like 13 1.69 1.88
NM_005562.1 LAMC2 laminin, gamma 2 1.96 2.01 NM_001005474.1 NFKBIZ nuclear factor of kappa light polypeptide gene enhancer 1.69 1.72
NM_000332.2 ATXN1 ataxin 1 1.96 1.86 NM_021202.1 TP53INP2 tumor protein p53 inducible nuclear protein 2 1.69 1.89
NM_001206.2 KLF9 Kruppel-like factor 9 1.96 2.19 NM_138448.2 ACYP2 acylphosphatase 2 1.68 2.03
NM_173359.3 EIF4E3 eukaryotic translation initiation factor 4E family member 3 1.95 2.01 NM_003729.1 RTCD1 RNA terminal phosphate cyclase domain 1 1.67 1.61
NM_003565.1 ULK1 unc-51-like kinase 1 1.95 1.57 NM_030912.2 TRIM8 tripartite motif-containing 8 1.67 1.70
NM_012161.2 FBXL5 F-box and leucine-rich repeat protein 5 1.95 2.27 NM_018202.3 TMEM57 transmembrane protein 57 1.67 1.88
NM_001079864.1 TAX1BP1 Tax1 (human T-cell leukemia virus type I) binding protein 1 1.93 1.66 NM_018297.2 NGLY1 N-glycanase 1 1.66 1.68
NM_005063.4 SCD stearoyl-CoA desaturase 1.93 4.44 NM_183399.1 RNF14 ring finger protein 14 1.66 1.87
NM_145279.4 MOBKL2C MOB1, Mps One Binder kinase activator-like 2C 1.92 2.58 NM_032357.2 CCDC115 coiled-coil domain containing 115 1.66 1.63
NM_004055.4 CAPN5 calpain 5 1.91 2.05 NM_172037.2 RDH10 retinol dehydrogenase 10 1.66 2.20
NM_175932.1 PSMD13 proteasome (prosome, macropain) 26S subunit 1.91 2.14 NM_012287.3 CENTB2 centaurin 1.65 1.62
NM_002885.1 RAP1GAP RAP1 GTPase activating protein 1.91 2.02 NM_018178.3 GOLPH3L golgi phosphoprotein 3-like 1.65 1.64
NM_145245.2 EVI5L ecotropic viral integration site 5-like 1.90 1.69 NM_025147.3 COQ10B coenzyme Q10 homolog B 1.64 1.54
NM_021242.3 MID1IP1 MID1 interacting protein 1 1.89 1.78 NM_006285.2 TESK1 testis-specific kinase 1 1.64 1.61
NM_006503.2 PSMC4 proteasome (prosome, macropain) 26S subunit 1.89 2.16 NM_001018102.1 GRINL1A glutamate receptor 1.64 1.92
NM_005975.2 PTK6 protein tyrosine kinase 6 1.88 1.97 NM_005476.3 GNE glucosamine (UDP-N-acetyl)-2-epimerase 1.64 2.98
NM_207304.1 MBNL2 muscleblind-like 2 1.88 1.63 NR_002204.1 FTHL11 ferritin, heavy polypeptide-like 11 on chromosome 8 1.63 1.79
NM_000859.1 HMGCR 3-hydroxy-3-methylglutaryl-Coenzyme A reductase 1.88 2.46 NM_053067.1 UBQLN1 ubiquilin 1 1.63 1.56
NM_021021.2 SNTB1 syntrophin 1.88 1.64 NM_005895.3 GOLGA3 golgi autoantigen 1.63 1.58
NM_032667.4 BSCL2 Bernardinelli-Seip congenital lipodystrophy 2 1.87 1.73 NM_037370.1 CCNDBP1 cyclin D-type binding-protein 1 1.63 1.50
NM_006022.2 TSC22D1 TSC22 domain family 1.87 2.40 NM_001042430.1 FAM164C family with sequence similarity 164 1.62 2.01
NM_012478.3 WBP2 WW domain binding protein 2 1.87 1.67 NM_004751.1 GCNT3 glucosaminyl (N-acetyl) transferase 3 1.62 3.99
NM_031476.2 CRISPLD2 cysteine-rich secretory protein LCCL domain containing 2 1.87 2.57 NM_004428.2 EFNA1 ephrin-A1 1.61 2.10
NM_017921.1 NPLOC4 nuclear protein localization 4 homolog 1.87 1.74 NM_007193.3 ANXA10 annexin A10 1.61 2.36
NM_015508.3 TIPARP TCDD-inducible poly(ADP-ribose) polymerase 1.86 1.75 NM_018180.2 DHX32 DEAH (Asp-Glu-Ala-His) box polypeptide 32 1.61 1.52
NM_001376.2 DYNC1H1 dynein, cytoplasmic 1, heavy chain 1 1.86 1.50 NM_020202.2 NIT2 nitrilase family 1.61 1.62
NM_144693.1 ZNF558 zinc finger protein 558 1.86 1.98 NM_032169.4 ACAD11 acyl-Coenzyme A dehydrogenase family 1.61 1.73
NM_003344.2 UBE2H ubiquitin-conjugating enzyme E2H 1.85 2.04 NM_147223.2 NCOA1 nuclear receptor coactivator 1 1.60 1.57
NM_152528.1 WDSUB1 WD repeat, sterile alpha motif and U-box domain containing 1 1.85 1.74 NM_002203.3 ITGA2 integrin 1.59 2.49
NM_001225.3 CASP4 caspase 4, apoptosis-related cysteine peptidase 1.85 1.97 NM_018317.1 TBC1D19 TBC1 domain family 1.59 2.18
NM_001512.2 GSTA4 glutathione S-transferase A4 1.84 2.45 NM_002061.2 GCLM glutamate-cysteine ligase 1.59 2.06
NM_020299.3 AKR1B10 aldo-keto reductase family 1 1.83 2.42 NM_019007.3 ARMCX6 armadillo repeat containing, X-linked 6 1.58 1.81
NM_016143.3 NSFL1C NSFL1 (p97) cofactor (p47) 1.83 2.61 NM_015974.2 CRYL1 crystallin, lambda 1 1.58 1.51
NM_080655.1 C9orf30 chromosome 9 open reading frame 30 1.83 1.77 NM_006149.2 LGALS4 lectin 1.58 3.12
NM_001018109.1 PIR pirin 1.83 2.67 NM_012233.1 RAB3GAP1 RAB3 GTPase activating protein subunit 1 1.57 1.78
NR_002200.1 FTHL2 ferritin, heavy polypeptide-like 2 on chromosome 1 1.83 1.62 NM_002808.3 PSMD2 proteasome (prosome, macropain) 26S subunit 1.57 1.72
NM_005485.3 PARP3 poly (ADP-ribose) polymerase family 1.82 1.77 NM_001030001.1 RPS29 ribosomal protein S29 1.57 1.61
NM_058172.3 ANTXR2 anthrax toxin receptor 2 1.82 1.88 NM_052849.2 CCDC32 coiled-coil domain containing 32 1.57 1.55
NR_002166.1 SEDLP spondyloepiphyseal dysplasia, late, pseudogene 1.81 1.88 NM_015484.4 SYF2 SYF2 homolog 1.57 1.64
NM_022449.1 RAB17 member RAS oncogene family 1.80 1.64 NM_001093771.1 TXNRD1 thioredoxin reductase 1 1.57 1.54
NM_198310.2 TTC8 tetratricopeptide repeat domain 8 1.80 1.83 NM_017707.2 ASAP3 ArfGAP with SH3 domain, ankyrin repeat and PH domain 3 1.57 1.59
NM_002786.2 PSMA1 proteasome (prosome, macropain) subunit 1.79 2.54 NM_020412.3 CHMP1B chromatin modifying protein 1B 1.56 1.83
NM_002796.2 PSMB4 proteasome (prosome, macropain) subunit 1.78 1.53 NM_017549.3 EPDR1 ependymin related protein 1 1.56 1.52
NR_002201.1 FTHL3P ferritin 1.78 2.10 NM_001080493.2 ZNF823 zinc finger protein 823 1.56 1.56
NM_020225.1 STOX2 storkhead box 2 1.78 5.98 NM_001008738.2 FNIP1 folliculin interacting protein 1 1.56 1.50
NM_080725.1 SRXN1 sulfiredoxin 1 homolog 1.78 1.67 NM_006343.2 MERTK c-mer proto-oncogene tyrosine kinase 1.56 2.12
NM_015946.4 PELO pelota homolog 1.77 1.58 NM_014851.2 KLHL21 kelch-like 21 1.56 2.02
NM_020751.1 COG6 component of oligomeric golgi complex 6 1.77 1.60 NM_002803.2 PSMC2 proteasome 1.55 1.77
NM_001080538.1 LOC441282 similar to aldo-keto reductase family 1 1.77 2.31 NM_145918.2 CTSL1 cathepsin L1 1.55 1.96
NM_004487.3 GOLGB1 golgin B1 1.76 1.82 NM_003846.1 PEX11B peroxisomal biogenesis factor 11 beta 1.55 1.92
NM_145040.2 PRKCDBP protein kinase C 1.76 3.27 NM_014905.2 GLS glutaminase 1.55 2.21
NM_201557.2 FHL2 four and a half LIM domains 2 1.76 1.74 NM_016154.3 RAB4B member RAS oncogene family 1.55 2.22
NM_015037.2 KIAA0913 KIAA0913 1.75 1.52 NM_052901.2 SLC25A25 solute carrier family 25 1.54 1.55
NM_003003.2 SEC14L1 SEC14-like 1 1.74 1.94 NM_033212.2 CCDC102A coiled-coil domain containing 102A 1.54 1.75
NM_001031835.1 PHKB phosphorylase kinase 1.74 1.63 NM_002799.2 PSMB7 proteasome (prosome, macropain) subunit 1.54 1.56
NM_016470.6 C20orf111 chromosome 20 open reading frame 111 1.74 1.54 NM_001031716.1 OBFC2A oligonucleotide/oligosaccharide-binding fold containing 2A 1.53 1.60
NM_001080791.1 C15orf57 chromosome 15 open reading frame 57 1.74 1.63 NM_015187.3 KIAA0746 KIAA0746 protein 1.53 1.62
NM_002541.2 OGDH oxoglutarate (alpha-ketoglutarate) dehydrogenase 1.74 1.55 NM_004034.1 ANXA7 annexin A7 1.52 1.75
NM_015922.1 NSDHL NAD(P) dependent steroid dehydrogenase-like 1.74 2.30 NM_002815.2 PSMD11 proteasome (prosome, macropain) 26S subunit 1.52 1.71
XM_001128220.1 PLEKHM1 pleckstrin homology domain containing, family M member 1 1.73 1.73 NM_014889.2 PITRM1 pitrilysin metallopeptidase 1 1.52 2.15
NM_004417.2 DUSP1 dual specificity phosphatase 1 1.73 1.78 NM_007126.2 VCP valosin-containing protein 1.51 1.79
NM_003971.3 SPAG9 sperm associated antigen 9 1.73 1.80 NM_032017.1 STK40 serine/threonine kinase 40 1.50 1.66
NM_016026.2 RDH11 retinol dehydrogenase 11 1.72 1.67 NM_032776.1 JMJD1C jumonji domain containing 1C 1.50 1.81

Table 2. Down-regulated genes (p<0.001) common in GANT61-treated HT29 and GC3/c1 cells.

ACCESSION NUMBER GENE SYMBOL DEFINITION Fold Change ACCESSION NUMBER GENE SYMBOL DEFINITION Fold Change
HT29 GC3/c1 HT29 GC3/c1
NM_015086.1 DDN dendrin −6.81 −2.37 NM_014791.2 MELK maternal embryonic leucine zipper kinase −2.50 −2.09
NM_001013653.2 LRRC26 leucine rich repeat containing 26 −4.77 −2.23 NM_016343.3 CENPF centromere protein F, 350/400ka −2.50 −2.71
NM_004091.2 E2F2 E2F transcription factor 2 −4.23 −2.26 NM_001789.2 CDC25A cell division cycle 25 homolog A −2.50 −2.45
NM_182687.1 PKMYT1 protein kinase, membrane associated tyrosine/threonine 1 −4.17 −4.81 NM_024037.1 C1orf135 chromosome 1 open reading frame 135 −2.49 −2.12
NM_001013653.1 LOC389816 cytokeratin associated protein −3.85 −2.32 NM_017669.2 ERCC6L excision repair cross-complementing rodent repair deficiency −2.48 −3.22
NM_020675.3 SPC25 NDC80 kinetochore complex component, homolog −3.51 −7.19 NM_031965.2 GSG2 germ cell associated 2 (haspin) -2.47 −2.79
NM_005733.1 KIF20A kinesin family member 20A −3.45 −2.21 NM_018186.2 C1orf112 chromosome 1 open reading frame 112 −2.47 −1.88
NM_002263.2 KIFC1 kinesin family member C1 −3.36 −2.74 NM_001786.2 CDC2 cell division cycle 2, G1 to S and G2 to M −2.47 −2.63
NM_006681.1 NMU neuromedin U −3.33 −3.15 NM_002105.2 H2AFX H2A histone family, member X −2.46 −2.86
NM_014783.2 ARHGAP11A Rho GTPase activating protein 11A −3.33 −3.46 NM_018492.2 PBK PDZ binding kinase −2.46 −2.21
NM_006176.1 NRGN neurogranin −3.29 −4.20 NM_198516.1 GALNTL4 UDP-N-acetyl-alpha-D-galactosamine −2.46 −1.82
NM_016095.1 GINS2 GINS complex subunit 2 −3.25 −4.51 NM_018685.2 ANLN anillin, actin binding protein −2.46 −2.44
NM_018063.3 HELLS helicase, lymphoid-specific −3.24 −3.42 NM_002915.3 RFC3 replication factor C (activator 1) 3, 38kDa −2.45 −2.59
NM_057735.1 CCNE2 cyclin E2 −3.22 −3.24 NM_007370.3 RFC5 replication factor C (activator 1) 5, 36.5kDa −2.43 −2.28
NM_014750.3 DLG7 discs, large homolog 7 −3.21 −2.48 NM_001032290.1 PSRC1 proline/serine-rich coiled-coil 1 −2.43 −2.53
NM_001168.2 BIRC5 baculoviral IAP repeat-containing 5 −3.18 −2.79 NM_000057.2 BLM Bloom syndrome −2.43 −3.28
NM_001255.2 CDC20 cell division cycle 20 homolog −3.12 −2.46 NM_012484.1 HMMR hyaluronan-mediated motility receptor −2.43 −1.82
NM_001237.2 CCNA2 cyclin A2 −3.11 −3.01 NM_145701.1 CDCA4 cell division cycle associated 4 −2.43 −2.26
NM_144508.3 CASC5 cancer susceptibility candidate 5 −3.09 −2.13 NM_024339.2 THOC6 THO complex 6 homolog −2.41 −1.74
NM_017611.2 SLC43A3 solute carrier family 43, member 3 −3.05 −2.60 NM_003258.2 TK1 thymidine kinase 1 −2.41 −3.74
NM_018154.2 ASF1B ASF1 anti-silencing function 1 homolog B −3.03 −3.06 NM_182776.1 MCM7 minichromosome maintenance complex component 7 −2.39 −2.54
NM_018410.3 HJURP Holliday junction recognition protein −3.02 −3.18 NM_178448.2 C9orf140 chromosome 9 open reading frame 140 −2.39 −2.34
NM_004217.2 AURKB aurora kinase B −3.01 −3.19 NM_001067.2 TOP2A topoisomerase (DNA) II alpha 170kDa −2.39 −3.16
NM_002692.2 POLE2 polymerase (DNA directed), epsilon 2 (p59 subunit) −3.01 −3.00 NM_178014.2 TUBB tubulin, beta −2.37 −2.27
NM_018101.2 CDCA8 cell division cycle associated 8 −2.97 −2.85 NM_018193.2 FANCI Fanconi anemia, complementation group I −2.37 −2.52
NM_002875.2 RAD51 RAD51 homolog −2.95 −2.66 NM_206833.2 CTXN1 cortexin 1 −2.36 −2.04
NM_031299.3 CDCA3 cell division cycle associated 3 −2.94 −2.82 NM_031966.2 CCNB1 cyclin B1 −2.36 −2.28
NM_181803.1 UBE2C ubiquitin-conjugating enzyme E2C −2.91 −2.80 NM_005483.2 CHAF1A chromatin assembly factor 1, subunit A (p150) −2.36 −2.21
NM_001100118.1 XRCC3 X-ray repair complementing defective repair 3 −2.90 −3.66 NM_005518.2 HMGCS2 3-hydroxy-3-methylglutaryl-Coenzyme A synthase 2 −2.35 −2.88
NM_025049.2 PIF1 PIF1 5′-to-3′ DNA helicase homolog −2.90 −2.87 NM_019013.1 FAM64A family with sequence similarity 64 −2.35 −3.13
NM_030928.2 CDT1 chromatin licensing and DNA replication factor 1 −2.86 −2.76 NM_002129.2 HMGB2 high-mobility group box 2 −2.35 −2.18
NM_016556.1 PSMC3IP PSMC3 interacting protein −2.85 −3.47 NM_001034194.1 EXOSC9 exosome component 9 −2.34 −1.69
NM_182746.1 MCM4 minichromosome maintenance complex component 4 −2.84 −2.97 NM_031423.3 NUF2 NDC80 kinetochore complex component, homolog −2.34 −2.48
NM_016937.2 POLA1 polymerase (DNA directed), alpha 1, catalytic subunit −2.84 −2.19 NM_002916.3 RFC4 replication factor C (activator 1) 4, 37kDa −2.33 −2.36
NM_007086.2 WDHD1 WD repeat and HMG-box DNA binding protein 1 −2.80 −2.53 NM_004111.4 FEN1 flap structure-specific endonuclease 1 −2.33 −1.97
NM_001761.1 CCNF cyclin F −2.80 −2.77 NM_145061.3 C13orf3 chromosome 13 open reading frame 3 −2.33 −2.53
NM_012177.2 FBXO5 F-box protein 5 −2.78 −2.95 NM_001012413.1 SGOL1 shugoshin-like 1 −2.33 −2.85
NM_018518.3 MCM10 minichromosome maintenance complex component 10 −2.78 −4.01 NM_207418.2 GCUD2 gastric cancer up-regulated-2 −2.32 −2.28
NM_003579.2 RAD54L RAD54-like −2.76 −2.98 NM_199413.1 PRC1 protein regulator of cytokinesis 1 −2.32 −2.13
NM_020242.1 KIF15 kinesin family member 15 −2.75 −3.00 NM_006461.3 SPAG5 sperm associated antigen 5 −2.31 −1.92
NM_004336.2 BUB1 BUB1 budding uninhibited by benzimidazoles 1 homolog −2.74 −2.47 NM_080668.2 CDCA5 cell division cycle associated 5 −2.29 −2.94
NM_003384.2 VRK1 vaccinia related kinase 1 −2.73 −1.89 NM_006101.1 NDC80 NDC80 homolog −2.29 −3.12
NM_018454.5 NUSAP1 nucleolar and spindle associated protein 1 −2.72 −2.40 NM_001012507.1 C6orf173 chromosome 6 open reading frame 173 −2.29 −2.38
NM_001034.1 RRM2 ribonucleotide reductase M2 polypeptide −2.72 −2.54 NM_002497.2 NEK2 NIMA (never in mitosis gene a)-related kinase 2 −2.29 −2.10
NM_012112.4 TPX2 microtubule-associated, homolog −2.72 −2.32 NM_024900.3 PHF17 PHD finger protein 17 −2.28 −1.67
NM_152259.3 C15orf42 chromosome 15 open reading frame 42 −2.70 −3.44 NM_001040668.1 BCL2L12 BCL2-like 12 −2.28 −1.93
NM_001042426.1 CENPA centromere protein A −2.70 −3.28 NM_014264.3 PLK4 polo-like kinase 4 −2.27 −1.97
NM_012415.2 RAD54B RAD54 homolog B −2.69 −1.88 NM_022809.2 CDC25C cell division cycle 25 homolog C −2.27 −2.05
NM_181471.1 RFC2 replication factor C (activator 1) 2, 40kDa −2.69 −2.50 NM_152515.2 CKAP2L cytoskeleton associated protein 2-like −2.26 −2.29
NM_005480.2 TROAP trophinin associated protein −2.69 −2.49 NM_006026.2 H1FX H1 histone family, member X −2.24 −2.08
NM_006845.2 KIF2C kinesin family member 2C −2.68 −2.81 NM_003276.1 TMPO thymopoietin −2.24 −1.95
NM_022346.3 NCAPG non-SMC condensin I complex −2.68 −2.70 NM_018136.3 ASPM asp (abnormal spindle) homolog −2.23 −2.51
NM_004260.2 RECQL4 RecQ protein-like 4 −2.68 −3.00 NM_016426.4 GTSE1 G-2 and S-phase expressed 1 −2.23 −2.44
NM_003318.3 TTK TTK protein kinase −2.67 −2.40 NM_014875.1 KIF14 kinesin family member 14 −2.23 −2.12
NM_030919.2 FAM83D family with sequence similarity 83, member D −2.66 −2.08 NM_014109.2 ATAD2 ATPase family, AAA domain containing 2 −2.23 −2.21
NM_001254.3 CDC6 cell division cycle 6 homolog −2.66 −3.05 NM_003390.2 WEE1 WEE1 homolog −2.22 −1.97
NM_001034836.1 RDM1 RAD52 motif 1 −2.65 −2.65 NM_002388.3 MCM3 minichromosome maintenance complex component 3 −2.22 −2.45
NM_004526.2 MCM2 minichromosome maintenance complex component 2 −2.65 −3.43 NM_005030.3 PLK1 polo-like kinase 1 −2.22 −2.37
NM_004701.2 CCNB2 cyclin B2 −2.65 −2.52 NM_005375.2 MYB v-myb myeloblastosis viral oncogene homolog −2.22 −2.01
NM_032818.2 C9orf100 chromosome 9 open reading frame 100 −2.64 −2.30 NM_024789.3 TMEM180 transmembrane protein 180 −2.20 −1.63
NM_006397.2 RNASEH2A ribonuclease H2, subunit A −2.64 −2.52 NM_001012716.1 C18orf56 chromosome 18 open reading frame 56 −2.20 −2.30
NM_014736.4 KIAA0101 KIAA0101 −2.61 −3.47 NM_017760.5 NCAPG2 non-SMC condensin II complex, subunit G2 −2.20 −2.11
NM_004219.2 PTTG1 pituitary tumor-transforming 1 −2.61 −2.23 NM_005192.2 CDKN3 cyclin-dependent kinase inhibitor 3 −2.20 −1.98
NM_182513.1 SPC24 SPC24, NDC80 kinetochore complex component, homolog −2.61 −3.15 NM_006231.2 POLE polymerase (DNA directed) −2.19 −3.53
NM_198948.1 NUDT1 nudix (nucleoside diphosphate linked moiety X)-type motif 1 −2.59 −3.09 NM_018124.3 RFWD3 ring finger and WD repeat domain 3 −2.19 −1.56
NM_033286.2 C15orf23 chromosome 15 open reading frame 23 −2.58 −1.77 NM_006088.5 TUBB2C tubulin, beta 2C −2.19 −1.63
NM_024053.3 CENPM centromere protein M −2.57 −3.45 NM_198434.1 AURKA aurora kinase A −2.18 −1.68
NM_006479.3 RAD51AP1 RAD51 associated protein 1 −2.57 −2.54 NM_007280.1 OIP5 Opa interacting protein 5 −2.18 −2.34
NM_006739.3 MCM5 minichromosome maintenance complex component 5 −2.56 −3.21 NM_014176.2 UBE2T ubiquitin-conjugating enzyme E2T (putative). −2.15 −2.01
NM_203401.1 STMN1 stathmin 1/oncoprotein 18 −2.56 −2.32 NM_199420.3 POLQ polymerase (DNA directed), theta −2.14 −2.45
NM_013277.2 RACGAP1 Rac GTPase activating protein 1 −2.55 −1.79 NM_001211.4 BUB1B BUB1 budding uninhibited by benzimidazoles 1 homolog beta −2.13 −2.23
NM_001005413.1 ZWINT ZW10 interactor −2.55 −2.64 NM_001025248.1 DUT deoxyuridine triphosphatase −2.13 −2.36
NM_004856.4 KIF23 kinesin family member 23 −2.54 −2.17 NM_031217.2 KIF18A kinesin family member 18A −2.13 −1.99
NM_004523.2 KIF11 kinesin family member 11 −2.53 −2.12 NM_025108.2 C16orf59 chromosome 16 open reading frame 59 −2.12 −3.48
NR_002734.1 PTTG3 pituitary tumor-transforming 3 −2.53 −2.43 NM_001042517.1 DIAPH3 diaphanous homolog 3 −2.12 −2.34
NM_006027.3 EXO1 exonuclease 1 −2.52 −3.86 NM_017998.1 C9orf40 chromosome 9 open reading frame 40 −2.11 −1.85
NM_016448.1 DTL denticleless homolog −2.51 −3.12 NM_052844.3 WDR34 WD repeat domain 34 −2.11 −2.71
NM_022770.2 GINS3 GINS complex subunit 3 (Psf3 homolog) −2.10 −1.65 NM_017918.3 CCDC109B coiled-coil domain containing 109B −1.84 −1.84
NM_003503.2 CDC7 cell division cycle 7 homolog −2.09 −1.97 NM_005782.2 THOC4 THO complex 4 −1.84 −1.63
NM_001071.1 TYMS thymidylate synthetase −2.08 −3.41 NM_003035.2 STIL SCL/TAL1 interrupting locus −1.84 −1.95
NM_005441.2 CHAF1B chromatin assembly factor 1, subunit B (p60) −2.08 −2.25 NM_005342.2 HMGB3 high-mobility group box 3 −1.83 −1.60
NM_014865.2 NCAPD2 non-SMC condensin I complex, subunit D2 −2.07 −2.03 NM_018132.3 CENPQ centromere protein Q −1.83 −2.10
NM_001798.2 CDK2 cyclin-dependent kinase 2 −2.07 −1.88 NM_024516.2 C16orf53 chromosome 16 open reading frame 53 −1.81 −1.86
NM_022720.5 DGCR8 DiGeorge syndrome critical region gene 8 −2.07 −1.57 NM_015895.3 GMNN geminin, DNA replication inhibitor −1.81 −1.87
NM_145018.2 C11orf82 chromosome 11 open reading frame 82 −2.06 −2.28 NM_002689.2 POLA2 polymerase (DNA directed), alpha 2 (70kD subunit) −1.81 −2.11
NM_016195.2 KIF20B kinesin family member 20B −2.06 −1.63 NM_000154.1 GALK1 galactokinase 1 −1.80 −1.84
NM_001008393.1 LOC201725 hypothetical protein −2.05 −1.98 NM_004499.3 HNRNPAB heterogeneous nuclear ribonucleoprotein A/B −1.80 −2.31
NM_000465.1 BARD1 BRCA1 associated RING domain 1 −2.05 −2.42 NM_024656.2 GLT25D1 glycosyltransferase 25 domain containing 1 −1.80 −1.62
NM_003173.2 SUV39H1 suppressor of variegation 3-9 homolog 1 −2.05 −1.89 NM_199250.1 C19orf48 chromosome 19 open reading frame 48 −1.79 −1.83
NM_015426.2 WDR51A WD repeat domain 51A −2.05 −1.60 NM_153329.2 ALDH16A1 aldehyde dehydrogenase 16 family, member A1 −1.79 −1.98
NM_020342.1 SLC39A10 solute carrier family 39 −2.05 −3.15 NM_032637.2 SKP2 S-phase kinase-associated protein 2 (p45) −1.79 −2.00
NM_144999.2 LRRC45 leucine rich repeat containing 45 −2.04 −3.33 NM_006191.2 PA2G4 proliferation-associated 2G4, 38kDa −1.78 −1.84
NM_000946.2 PRIM1 primase, DNA, polypeptide 1 (49kDa) −2.04 −2.29 NM_001033580.1 MYO19 myosin XIX −1.78 −3.61
NM_016183.3 MRTO4 turnover 4 homolog −2.04 −1.68 NM_017915.2 C12orf48 chromosome 12 open reading frame 48 −1.78 −1.92
NM_017518.5 UCHL5IP UCHL5 interacting protein −2.04 −1.75 NM_003362.2 UNG uracil-DNA glycosylase −1.76 −1.55
NM_020394.2 ZNF695 zinc finger protein 695 −2.03 −2.78 NM_014985.2 CEP152 centrosomal protein 152kDa −1.75 −2.37
NM_203394.2 E2F7 E2F transcription factor 7 −2.02 −2.03 NM_001333.2 CTSL2 cathepsin L2 −1.75 −1.70
NM_173529.3 C18orf54 chromosome 18 open reading frame 54 −2.02 −1.99 NM_016310.2 POLR3K polymerase (RNA) III (DNA directed) polypeptide K, 12.3 kDa −1.75 −1.59
NM_000156.4 GAMT guanidinoacetate N-methyltransferase −2.02 −1.64 NM_006406.1 PRDX4 peroxiredoxin 4 −1.74 −1.93
NM_007299.2 BRCA1 breast cancer 1, early onset −2.02 −2.11 NM_005484.2 PARP2 poly (ADP-ribose) polymerase family, member 2 −1.74 −1.53
NM_014641.1 MDC1 mediator of DNA damage checkpoint 1 −2.01 −1.57 NM_001039091.1 PRPS2 phosphoribosyl pyrophosphate synthetase 2 −1.73 −1.60
NM_032485.4 MCM8 minichromosome maintenance complex component 8 −2.01 −3.18 NM_015703.3 RRP7A ribosomal RNA processing 7 homolog A −1.73 −1.71
NM_024660.2 TMEM149 transmembrane protein 149 −2.01 −2.07 NM_015032.1 PDS5B PDS5, regulator of cohesion maintenance, homolog B −1.73 −1.78
NM_015414.2 RPL36 ribosomal protein L36 −2.00 −2.40 NM_000992.2 RPL29 ribosomal protein L29 −1.73 −2.53
NM_021734.3 SLC25A19 solute carrier family 25, member 19 −2.00 −1.67 NM_173608.1 C14orf80 chromosome 14 open reading frame 80 −1.72 −1.73
NM_003504.3 CDC45L CDC45 cell division cycle 45-like −2.00 −2.84 NM_001042550.1 SMC2 structural maintenance of chromosomes 2 −1.72 −1.92
NM_014708.3 KNTC1 kinetochore associated 1 −1.99 −1.86 NM_014285.4 EXOSC2 exosome component 2 −1.72 −1.70
NM_018131.3 CEP55 centrosomal protein 55kDa −1.98 −2.01 NM_138961.1 ESAM endothelial cell adhesion molecule −1.71 −2.21
NM_007243.1 NRM nurim −1.98 −2.22 NM_145159.1 JAG2 jagged 2 −1.70 −1.89
NM_003517.2 HIST2H2AC histone cluster 2, H2ac −1.98 −2.54 NM_006392.2 NOL5A nucleolar protein 5A (56kDa with KKE/D repeat) −1.69 −1.70
NM_001002800.1 SMC4 structural maintenance of chromosomes 4 −1.98 −1.94 NM_033661.3 WDR4 WD repeat domain 4 −1.69 −1.84
NM_001033505.1 CSTF3 cleavage stimulation factor, 3′ pre-RNA, subunit 3, 77kD −1.97 −1.96 NM_032358.2 CCDC77 coiled-coil domain containing 77 −1.68 −1.54
NM_181716.2 PRR6 proline rich 6 −1.96 −1.85 NM_018693.2 FBXO11 F-box protein 11 −1.68 −1.93
NM_018663.1 PXMP2 peroxisomal membrane protein 2, 22kDa −1.96 −2.02 NM_001026.3 RPS24 ribosomal protein S24 −1.68 −1.94
NM_005915.4 MCM6 minichromosome maintenance complex component 6 −1.96 −2.25 NM_016292.1 TRAP1 TNF receptor-associated protein 1 −1.67 −1.51
NM_005189.1 CBX2 chromobox homolog 2 −1.96 −1.68 NM_017613.2 DONSON downstream neighbor of SON −1.66 −1.61
NM_001379.1 DNMT1 DNA (cytosine-5-)-methyltransferase 1 −1.96 −1.84 NM_006047.4 RBM12 RNA binding motif protein 12 −1.66 −1.91
NM_001002018.1 HCFC1R1 host cell factor C1 regulator 1 (XPO1 dependent) −1.96 −1.79 NM_012140.3 SLC25A10 solute carrier family 25 (dicarboxylate transporter), member 10 −1.66 −1.55
NM_000234.1 LIG1 ligase I, DNA, ATP-dependent −1.96 −2.81 NM_001025238.1 TSPAN4 tetraspanin 4 −1.66 −1.94
NM_138443.2 CCDC5 coiled-coil domain containing 5 (spindle associated) −1.95 −1.68 NM_001008735.1 HMG1L1 high-mobility group (nonhistone chromosomal) protein 1-like 1 −1.66 −1.62
NM_020315.4 PDXP pyridoxal (pyridoxine, vitamin B6) phosphatase −1.95 −2.04 NM_005956.2 MTHFD1 methylenetetrahydrofolate dehydrogenase (NADP+dependent) 1 −1.65 −1.69
NM_032117.2 MND1 meiotic nuclear divisions 1 homolog −1.95 −2.63 NM_145014.1 HYLS1 hydrolethalus syndrome 1 −1.65 −1.62
NM_194255.1 SLC19A1 solute carrier family 19 (folate transporter), member 1 −1.95 −1.93 NM_004309.3 ARHGDIA Rho GDP dissociation inhibitor (GDI) alpha −1.64 −1.75
NM_058216.1 RAD51C RAD51 homolog C −1.94 −2.13 NM_182649.1 PCNA proliferating cell nuclear antigen −1.63 −1.99
NM_018725.3 IL17RB interleukin 17 receptor B −1.94 −2.25 NM_032118.2 WDR54 WD repeat domain 54 −1.63 −1.77
NM_024844.3 NUP85 nucleoporin 85kDa −1.93 −1.53 NM_015697.6 COQ2 coenzyme Q2 homolog −1.63 −1.85
NM_015190.3 DNAJC9 DnaJ (Hsp40) homolog, subfamily C, member 9 −1.93 −1.64 NM_032982.2 CASP2 caspase 2 −1.63 −1.67
NM_004237.2 TRIP13 thyroid hormone receptor interactor 13 −1.93 −2.23 NM_001042762.1 FIGNL1 fidgetin-like 1 −1.63 −1.85
NM_000179.1 MSH6 mutS homolog 6 −1.93 −1.83 NM_001100417.1 UBR7 ubiquitin protein ligase E3 component n-recognin 7 (putative) −1.61 −1.64
NM_001009936.1 PHF19 PHD finger protein 19 −1.93 −2.42 NM_003146.2 SSRP1 structure specific recognition protein 1 −1.60 −1.66
NM_003609.2 HIRIP3 HIRA interacting protein 3 −1.92 −2.01 NM_152308.1 C16orf75 chromosome 16 open reading frame 75 −1.60 −1.99
NM_022145.3 CENPK centromere protein K −1.92 −2.25 NM_001024662.1 RPL6 ribosomal protein L6 −1.60 −1.56
NM_022908.1 NT5DC2 5′-nucleotidase domain containing 2 −1.92 −1.81 NM_182620.3 FAM33A family with sequence similarity 33, member A −1.59 −2.17
NM_006342.1 TACC3 transforming, acidic coiled-coil containing protein 3 −1.92 −2.27 NM_033120.2 NKD2 naked cuticle homolog 2 −1.59 −1.65
NM_001080450.1 KIAA1553 KIAA1553 −1.92 −1.65 NM_003472.2 DEK DEK oncogene (DNA binding) −1.58 −1.91
NM_001018115.1 FANCD2 Fanconi anemia, complementation group D2 −1.92 −2.39 NM_032015.3 RNF26 ring finger protein 26 −1.58 −1.70
NM_001033.2 RRM1 ribonucleotide reductase M1 polypeptide −1.92 −1.87 NM_018455.3 CENPN centromere protein N −1.57 −2.01
NM_001078174.1 SLC29A1 solute carrier family 29 (nucleoside transporters) −1.91 −2.19 NM_003091.3 SNRPB small nuclear ribonucleoprotein polypeptides B and B1 −1.56 −1.52
NM_153485.1 NUP155 nucleoporin 155kDa −1.91 −1.65 NM_003863.2 DPM2 dolichyl-phosphate mannosyltransferase polypeptide 2 −1.56 −1.77
NM_002482.2 NASP nuclear autoantigenic sperm protein (histone-binding) −1.91 −1.96 NM_017916.1 PIH1D1 PIH1 domain containing 1 −1.55 −1.97
NM_176880.4 NR2C2AP nuclear receptor 2C2-associated protein −1.90 −1.87 NM_020810.1 TRMT5 tRNA methyltransferase 5 homolog −1.55 −1.54
NM_021953.2 FOXM1 forkhead box M1 −1.90 −2.28 NM_058219.2 EXOSC6 exosome component 6 −1.55 −1.65
NM_015721.2 GEMIN4 gem (nuclear organelle) associated protein 4 −1.90 −1.54 NM_002882.2 RANBP1 RAN binding protein 1 −1.54 −1.88
NM_018188.2 ATAD3A ATPase family, AAA domain containing 3A −1.89 −1.70 NM_007111.3 TFDP1 transcription factor Dp-1 −1.54 −1.51
NM_145060.3 C18orf24 chromosome 18 open reading frame 24 −1.89 −2.45 NM_021709.1 SIVA CD27-binding (Siva) protein −1.54 −1.71
NM_018365.1 MNS1 meiosis-specific nuclear structural 1 −1.89 −2.12 NM_015140.2 TTLL12 tubulin tyrosine ligase-like family, member 12 −1.53 −1.61
NM_012310.3 KIF4A kinesin family member 4A −1.89 −2.38 NM_022044.2 SDF2L1 stromal cell-derived factor 2-like 1 −1.53 −1.70
NM_015261.2 NCAPD3 non-SMC condensin II complex, subunit D3 −1.88 −1.88 NM_002346.1 LY6E lymphocyte antigen 6 complex, locus E −1.52 −1.56
NM_013296.3 GPSM2 G-protein signalling modulator 2 −1.87 −1.63 NM_022490.1 POLR1E polymerase (RNA) I polypeptide E, 53kDa −1.52 −1.77
NM_002691.1 POLD1 polymerase (DNA directed), delta 1, catalytic subunit −1.87 −2.22 NM_032799.4 ZDHHC12 zinc finger, DHHC-type containing 12 −1.51 −1.64
NM_203467.1 PPIL5 peptidylprolyl isomerase (cyclophilin)-like 5 −1.87 −1.79 NM_001017980.2 LOC203547 hypothetical protein −1.51 −1.54
NM_015201.3 BOP1 block of proliferation 1 −1.87 −2.29 NM_003801.3 GPAA1 glycosylphosphatidylinositol anchor attachment protein 1 homolog −1.51 −1.89
NM_018353.3 C14orf106 chromosome 14 open reading frame 106 −1.86 −1.54 NM_016594.1 FKBP11 FK506 binding protein 11, 19 kDa −1.50 −1.94
NM_080626.5 BRI3BP BRI3 binding protein −1.84 −1.72 NM_022754.4 SFXN1 sideroflexin 1 −1.50 −1.88

Modulation of canonical signaling pathways following inhibition of HH signaling

Genes with significant changes in expression following GANT61 treatment were assigned to different canonical signaling pathways and subjected to Ingenuity Pathway Analysis (IPA), where the resulting 1,368 DEGs in HT29 and 1,002 DEGs in GC3/c1 were mapped to networks defined by the IPA database (Figure 3). For the mapped DEGs including both up- and down- regulated genes, the 15 most significantly altered canonical pathways in HT29 demonstrated –log(p-value) ranging from 2.045 to 9.025, and in GC3/c1 from 2.32 to 7.509. Of the 15 pathways involving genes significantly down-regulated, 12 were common to both cell lines. The 3 common pathways with the greatest differential down-regulated expression include genes involved in the DNA damage response, cell cycle checkpoint control, and mitosis. Other pathways down-regulated involved the G1/S and G2/M DNA damage checkpoints, DNA precursor metabolism, and cell signaling involving different pathways including those involved in cancers, which also demonstrate 3 signatures unique to either HT29 or GC3/c1 (Figure 3). Of the 15 pathways involving genes that are the most significantly up-regulated, 8 are common to both HT29 and GC3/c1, and 7 represent unique pathways for each cell line, demonstrating more diversity in patterns of up-regulated gene expression (Figure 3). The up-regulated pathways common to both cell lines include the metabolism-related such as steroids, pyruvate, glycolysis glutathione, or glycerolipid, and not directly related to the control of cellular proliferation.

Figure 3. The top 15 canonical signaling pathways influenced by inhibition of GLI1/GLI2 function in HT29 and GC3/c1 cells.

Figure 3

The top 15 canonical signaling pathways, determined by IPA, that were significantly up-regulated or down-regulated by GANT61 treatment in HT29 and GC3/c1 cells, are shown. The 1,368 DEGs in HT29 and 1,002 DEGs in GC3/c1 were mapped to the IPA- defined network. The significance p-values that determine the probability that the association between the genes in the dataset and the canonical pathway is by chance alone were calculated by Fisher's exact test, and are expressed as –log (p-value). A. Pathways with enriched down-regulated genes. B. Pathways with enriched up-regulated genes. Blue: Pathways common to both HT29 and GC3/c1. Red: Pathways unique to either HT29 or GC3/c1. Yellow squares: Ratio of the number of DEGs that map to a specific canonical pathway divided by total number of genes that make up that pathway.

Genes that demonstrate differential fold change patterns in GANT61-treated HT29 and GC3/c1 cells

Fold change patterns of most highly DEGs in HT29 and GC3/c1 were selected, analyzed and displayed in a heat map to evaluate and compare similarity and differences in differential expression between the two cell lines treated with GANT61 (Figure 4). In addition to genes with diverse functions that are not directly related to HH-dependent proliferation, up-regulated genes that influence the G1/S transition and subsequent cell cycle progression, and that are common to both cell lines, include CDKN1A, and the DNA-damage-inducible transcripts 3 and 4 (DDIT3 and DDIT4). A considerably greater number of genes involved in cellular proliferation and cell cycle transition through the G1/S boundary, S-phase progression, and the G2/M transition, were significantly down-regulated in expression, and common to both cell lines. These include CDC6 (involved at G1/S), three genes that drive entry into and passage through S-phase (CCNE2, E2F2) and G2 (CCNA2), genes involved in DNA replication and repair (TYMS, POLE, TOP2A, TK1, POLE2), and two genes that regulate mitosis (AURKB, CDC20; Figure 4, asterisks).

Figure 4. Heat map showing fold change patterns of most highly DEGs in GANT61-treated human colon carcinoma cell lines.

Figure 4

The heat map was generated in Matlab (Mathworks), and compares fold change patterns of the most highly DEGs in HT29 and GC3/c1 cells after GANT61 treatment. The most highly DEGs demonstrated a differential expression p-value of p<0.001 between vehicle control (0.2% DMSO) and GANT61-treated cells. Left panel (red): up-regulated genes. Right panel (green): down-regulated genes. Genes denoted with asterisks define those genes with specific roles in G1/S transition, S-phase progression, DNA replication or repair, or regulation of the G2- or M- phase transitions. Fold changes of all down-regulated DEGs and all but one up-regulated DEG are ≤8 (central color spectrum bar).

Of the 296 up-regulated genes, in addition to the genes comparably represented in the heat map that include DDIT3 (GADD153) and DDIT4 (REDD1), additional novel DNA damage-inducible transcripts were also identified and include DDIT2 (GADD45G), PPP1R15A (GADD34) and ATF3 (Table 1). TP53INP1, which can regulate cell cycle arrest, and TP53INP2, identified in cell death responses, were also up-regulated. Of the 309 genes significantly down-regulated in response to GANT61, novel genes identified include KIAA0101 (p15[PAF]), Replication Factor C variants 2, 3, 4, 5, CDT1, the E2F transcription factors CDCA4 and TFDP1, MDC1, PCNA, FANCD2, and the genes involved in DNA repair, RAD51C (XRCC3), RAD54B, RAD51 and HELLS (Table 2).

Differentially expressed genes involved in the G1/S and G2/M transitions

To further evaluate the genes involved in control of cell cycle progression in human colon carcinoma cells following GANT61 treatment, 10 genes involved in the G1/S or G2/M transitions, identified by IPA, were selected for further examination. Genes required at the G1/S boundary for G1/S transition, or for the induction of a G1/S checkpoint following cytostatic signals, include the two cyclin-dependent kinase inhibitors, p21Cip1 (CDKN1A) and p15Ink4B (CDKN2B), which were up-regulated by 5.2- and 3.1- fold, 24 hr after GANT61 administration (Table 3). Additional genes required for the G1/S transition that were down-regulated include the E2 transcription factor E2F2 (-4.2-fold), and other critical genes that were down-regulated by 2.1- to 3.2- fold, include CYCLIN E (CCNE2), CDK2 and CDC25A. At G2/M, GANT61 induced down-regulated expression of CCNA2 (CYCLIN A2), CYCLIN B1 (CCNB1), CYCLIN B2 (CCNB2), CDK1 (CDC2), and CDC25C by 2.3- to 3.1- fold (Table 3).

Table 3. Gene expression changes from cDNA arrays at G1/S and G2/M.

Cell Cycle Phase Accession Number Gene Symbol Gene Name Fold Change
HT29 GC3/c1
G1/S NM_000389.2 CDKN1A cyclin-dependent kinase inhibitor 1A (p21Cip1) +5.21 +2.02
NM_078487.2 CDKN2B cyclin-dependent kinase inhibitor 2B (p15Ink4b) +3.13 +2.13
NM_004091.2 E2F2 E2F transcription factor 2 −4.24 −2.26
NM_057735.1 CCNE2 cyclin E2 −3.22 −3.24
NM_001789.2 CDC25A cell division cycle 25 homolog A −2.50 −2.45
NM_001798.2 CDK2 cyclin−dependent kinase 2 −2.07 −1.88
G2/M NM_001237.2 CCNA2 cyclin A2 −3.11 −3.01
NM_004701.2 CCNB2 cyclin B2 −2.65 −2.52
NM_001786.2 CDK1 cyclin-dependent kinase 1 −2.47 −2.63
NM_031966.2 CCNB1 cyclin B1 −2.36 −2.28
NM_022809.2 CDC25C cell division cycle 25 homolog C −2.27 −2.05

To determine the robustness of cDNA microarray gene expression profiling following treatment of HT29 and GC3/c1 cells with GANT61 (20 µM) for 24 hr, qRT-PCR was employed to determine changes in expression of the selected group of 10 DEGs determined from the cDNA microarrays. The genes involved and primers synthesized are shown in Table 4. qRT-PCR was performed on cDNA generated using total RNA independently isolated from GANT61-treated HT29 and GC3/c1 cells for 0 hr, 16 hr, 24 hr, 38 hr and 48 hr after treatment. GAPDH was used to normalize all qRT-PCR data. Genes determined by qRT-PCR included the expression of E2F2, CCNE2, CDC25A and CDK2 at G1/S, which were down-regulated, up-regulation of CDKN1A and CDKN2B at G1/S, and down-regulation of CCNA2, CDC25C, CCNB2, and CDK1 at G2/M (Figure 5). The up-regulated or down-regulated changes in gene expression following GANT61 treatment and determined by cDNA microarray profiling, were confirmed by qRT-PCR (Figure 5).

Table 4. Sequences of primers used in quantitative Real-Time PCR.

Accession # Gene Symbol Strand Primer Sequence Product Size (bp)
NM_078487.2 CDKN2B Plus 5′-TCTCCGTTGGCCGGAGGTCA-3′ 95
Minus 5′-TGGCAGGGTCTGCGCAGTTG-3′
NM_004701.2 CCNB2 Plus 5′-CTAACGGCGCCTCGTACGCT-3′ 54
Minus 5′-CAGGGAGGGACGCGGACTGA-3′
NM_057749.1 CCNE2 Plus 5′-GAGCGGTAGCTGGTCTGGCG-3′ 94
Minus 5′-GGGCTGGGGCTGCTGCTTAG-3′
NM_001789.2 CDC25A Plus 5′-CGTGGCTGCCTGCACTCTCA-3′ 159
Minus 5′-GGCTGTCACAGGTGACTGGGG-3′
NM_001798.3 CDK2 Plus 5′-TTTGCTGAGATGGTGACTCGCCG-3′ 159
Minus 5′-CCGGGCCCACTTGGGGAAAC-3′
NM_004091.2 E2F2 Plus 5′-CCGGCAGAAGCTGTGTGGGG-3′ 97
Minus 5′-GGCCTCCCTAGGCCCAGCTT-3′
NM_001237.3 CCNA2 Plus 5′-AAAAGGCAGCGCCCGTCCAA-3′ 89
Minus 5′-CTGCTGCTGCGCTAGACCCC-3′
NM_005631.3 CDKN1A Plus 5′-CTGCGCCAGCTGAGGTGTGA-3′ 189
Minus 5′-GCTGCTCGCTGTCCACTGGG-3′
NM_022809.2 CDC25C Plus 5′-GTGCATTTAGCTGGGATGACAATGGAA-3′ 189
Minus 5′-GGCCACTTCTGCTCACCTTTGC-3′
NM_001786.2 CDK1 Plus 5′-ACTGGCTGATTTTGGCCTTGCC-3′ 118
Minus 5′-TGAGTAACGAGCTGACCCCAGCAA-3′
NM_005269 GLI1 Plus 5′GCCCAGACAGAGGCCCACTC-3′ 547
Minus 5′CTGCAGCCATCCCAACGGCA-3′
NM_005270 GLI2 Plus 5′-CACCGCTGCTCAAAGAGAA-3′ 227
Minus 5′-TCTCCACGCCACTGTCATT-3′
NM_000264 PTCH1 Plus 5′-CCACAGAAGCGCTCCTACA-3′ 214
Minus 5′-CTGTAATTTCGCCCCTTCC-3′

Figure 5. Selected DEGs from cDNA array gene expression profiling analyzed by qRT- PCR in HT29 (A) or GC3/c1 (B).

Figure 5

Cells were treated with vehicle alone (0.2% DMSO) or GANT61 (20 µM) for 16 hr, 24 hr, 38 hr, or 48 hr. Total RNA was extracted and qRT-PCR was performed as described in Materials and Methods using the primer sets listed in Table 2. Data represent the mean±SD of 4 determinations, and GAPDH was used to normalize the relative mRNA levels.

Discussion

The HH signaling pathway is activated in a variety of human cancers following mutations in genes that regulate canonical HH signaling, including the receptor PTC, and the HH signaling molecule, SMO, and can also be activated via transcriptional up-regulation of the HH ligands (reviewed in [3]). This pathway is becoming of increasing importance due to gaining insight into its prominent role in many developmental processes, and in the maintenance of the malignant phenotype in a wide variety of human cancers, whose growth has been found to be prevented by selective inhibition of constitutive HH pathway activity [14], [35], [36]. Tumors of the brain, prostate, skin, pancreas, and kidney have demonstrated the requirement for HH-GLI signaling, and have responded to inhibition of the HH signaling target molecule SMO by cyclopamine or SMOshRNA [4], [19], [20], [21], [22], [23].

The transcriptional activators in HH signaling comprise members of the GLI family of transcription factors, GLI1 and GLI2, which have both distinct as well as overlapping functions [16]. Activation of the GLI proteins is an intricate process that involves modifications and interactions of a number of positive and negative pathway regulators and is not fully understood [1], [14], [35]. Target genes regulated by the HH signaling pathway differ between tissues and cell types, as well as being influenced by the presence or absence of regulatory factors co-expressed with GLI proteins that eventually determine the transcriptional programs activated by HH signaling [6], [37]. Thus, oncogenic signaling pathways converge on canonical HH signaling at the level of the GLI transcription factors and additionally on target genes downstream of GLI1 and GLI2 to further drive the HH signaling pathway in cellular survival in malignancies [6], [7], [20], [38], [39], [40]. The HH signaling phenotype is therefore significantly influenced and ultimately determined by the co-expression of additional regulatory factors, and hence by the cellular context of gene expression.

HH signaling plays a role in the differentiation program of normal intestinal villi [11], [12], [41], and it has been suggested recently that human colon cancer epithelial cells display a HH-GLI signaling axis in the process of carcinogenesis [24], [25]. Expression of HH-GLI pathway components was consistently demonstrated in an analysis of 40 primary human colon carcinomas and tumors metastatic to the liver [26], consistent with findings of previous investigators [25], [42], [43]. Thus, using qRT-PCR, the expression of GLI1, PTCH1, GLI2 and SHH was determined in all human colon carcinomas examined. The requirement for both GLI1 and GLI2 for sustained proliferation and survival of human colon carcinoma cell lines in vitro, including HT29, was demonstrated using siRNA technology [26]. In addition, knockdown of SMO by SMOshRNA prevented the growth of HT29 cells in SCID mice, while wt HT29 subcutaneous xenografts responded to cyclopamine by reduction in tumor volume [26]. Thus, canonical activation of GLI1 and GLI2 via SMO is important for the survival and proliferation of human colon carcinoma cells in vivo.

In the current study, the function of both GLI1 and GLI2 downstream of SMO was inhibited in the presence of GANT61, a small molecule inhibitor that was identified from a cell-based screen to specifically inhibit GLI1-mediated transcription, but that also inhibited the function of GLI2 [34]. This agent was selected to specifically inhibit the final arbiters of HH signaling, the GLI transcription factors, in elucidation of the downstream target genes that determine HH-dependent proliferation in human colon carcinoma cells. Two cell lines, well characterized in our laboratories, HT29 and GC3/c1, were treated with GANT61 (20 µM) for 24 hr, and the expression of GLI1, GLI2 and PTCH1 mRNA was down-regulated. Further, the effects on cellular proliferation as determined by the distribution of cells within the cell cycle and flow cytometric analysis demonstrated accumulation of cells in G1 following treatment, with a concomitant decrease of cells from the G2/M compartment, and in the case of HT29, also from S-phase, suggesting the induction of a G1/S checkpoint.

HT29 and GC3/c1 cells were subsequently treated with GANT61 (20 µM) for 24 hr, RNA was extracted, and changes in gene expression were determined by Illumina cDNA microarray profiling. Following statistical analyses, 1,368 genes in HT29 and 1,002 genes in GC3/c1, were determined to be significantly modulated by GANT61 treatment (FC>1.5; p<0.001). For genes that were up-regulated in expression, 296 genes were common to both cell lines, and for down-regulated genes, 309 genes were common to both cell lines. The blockade of cells at the G1/S boundary is evidenced by up-regulated expression of p21Cip1 and p15Ink4b that in part regulate the G1/S transition. p15Ink4b is a member of the Ink4 family of CDK inhibitors, is induced in response to cytostatic signals [32], [44], and complexes with CYCLIN D/CDK4 or CYCLIN D/CDK6 to mediate G1-phase arrest at the G1/S transition in certain systems [30], [32]. p21Cip1 can bind a broad range of cyclin-CDK complexes, with a preference for those containing CDK2 (reviewed in [31], [45], and during a normal cell cycle, facilitates active cyclin-CDK complex formation to promote cell proliferation. However when overexpressed, p21Cip1 forms an inhibitory complex with CYCLIN E/CDK2, leading to G1- and consequently S- phase arrest, thereby forming the G1/S checkpoint [46], [47]. The scheduled timing of expression of CYCLINS E, A and B that drive cell cycle progression, is reflected in the major changes in the phases of the cell cycle. CYCLIN E is expressed at maximal levels in cells undergoing the G1 to S transition, declining during S-phase progression, such that G2/M cells are CYCLIN E-negative (reviewed in [30]). CYCLIN A is expressed in late G1, demonstrates pronounced expression during S-phase, and increases as the cells advance towards G2, with degradation in early mitosis; B type cyclins begin to be expressed in late S-phase, and drive the cells though G2- and M- phases of the cell cycle [30]. CDK2 controls the G1/S transition by complexing with CYCLIN E, and is activated by CDC25A, which dephosphorylates CDK2 [48]. CDC2 controls cellular entry into mitosis at the G2/M transition, thereby forming complexes with CYCLINS A and B, is activated by CDC25C, and is down-regulated in late M-phase [29]. All of these genes are down-regulated in expression in response to the inhibition of GLI1/GLI2 function (Table 2). Thus, signals elicited to promote cellular accumulation at G1/S lead to the repression of genes that regulate further cell cycle progression, and p21Cip1 expression has been shown to deplete the expression of genes that regulate DNA replication and repair, and mitosis [49], [50], [51]. In the current study these genes include CDC6 (active at the G1/S transition and essential for the initiation of DNA replication), TYMS, TOP2A, TK1, POLE and POLE2 (S-phase), and AURKB and CDC20 (mitosis [52], [53]), determined by heat map analysis. A schematic representation of the genes involved in GANT61-induced inhibition of cell cycle progression at G1/S, S-phase progression, and regulation during G2- and M-phases, identified from cDNA microarrays, heat map analysis, and by qRT-PCR, is shown in Figure 6, and involves 5 of the 12 common signaling pathways determined by IPA analysis.

Figure 6. Schematic representation of genes involved in GANT61-induced inhibition of cell cycle progression.

Figure 6

From cDNA microarray, heat map, and qRT-PCR analyses, genes involved at different phases of the cell cycle including the G1/S transition, and progression through S- G2- and M- phases, are shown. The genes identified include CDK inhibitors, members of the CDK and CDC families, cyclins, genes involved in DNA replication and repair, and genes that regulate the mitotic spindle, and involve 5 of the 12 common signaling pathways determined by IPA analysis. Red: Up-regulated genes. Green: Down-regulated genes. Light shade→dark shade, increasing differential expression.

Comprehensive cDNA microarray gene profiling analysis of genes that determine the HH signaling phenotype has been conducted only in non-cancer cell models. In these systems, GLI activation has been stimulated by EGF treatment [6], stable GLI1 or HA-RAS expression [7], or expression of constitutively activated GLI2 [16]. In these studies, CYCLIN D [6], [7], GADD153 [7], CDKN2B, CDKN1A, CDK2, PCNA, TOP2A, CCNB1, XRCC1 [16], have been identified as genes activated downstream of GLI. In GANT61-treated human colon carcinoma cells, novel DNA damage-inducible transcripts DDIT3 (GADD153), DDIT4 (REDD1), DDIT2 (GADD45G), PPP1R15A (GADD34) and ATF3 were significantly up-regulated concomitant with the arrest of cells at G1/S. TP53INP1, involved in cell cycle regulation, and TP53INP2, linked to cell death responses, were also up-regulated. Additional novel genes involved in S-phase progression and DNA damage response that were significantly down-regulated include KIAA0101 (p15[PAF]), Replication Factor C variants 2, 3, 4, 5, CDT1, the E2F transcription factors CDCA4 and TFDP1, MDC1, PCNA, FANCD2, and the genes involved in DNA repair, RAD51C (XRCC3), RAD54B, RAD51 and HELLS.

In summary, we have compared gene expression profiles in two human colon carcinoma cell lines after targeting the function of the transcriptional regulators of HH signaling, GLI1 and GLI2, using the small molecule inhibitor GANT61. Data are consistent with accumulation of cells at the G1/S boundary, as evidenced from flow cytometric analysis, cDNA microarray gene profiling, and qRT- PCR. GANT61-treated cells demonstrated up-regulated expression of the CDK inhibitors p21Cip1 and p15Ink4b that function at the G1/S boundary, and down-regulated expression of additional key genes that determine the G1/S transition, initiation of DNA replication, S-phase progression, DNA repair, and subsequent transition through the G2/M phases. Inhibition of the transcriptional regulation of HH signaling in human colon carcinoma cells therefore directly involves genes that regulate cell cycle transition through G1/S, cell cycle progression, and proliferation, and genes involved in stress-induced and DNA damage responses.

Materials and Methods

Human colon carcinoma cell lines

HT29 was purchased from ATCC (Manassas, VA), while GC3/c1 was established in culture by our group from a human colon adenocarcinoma xenograft model [54]; both cell lines express mutant p53 alleles. Cell lines were maintained in the presence of folate-free RPMI 1640 medium containing 10% dFBS and 80 nM [6RS] 5-methyltetrahydrofolate.

Flow cytometric analysis

HT29 and GC3/c1 cells were plated at a density of 100,000 cells/well in six-well plates. After overnight attachment, cells were treated with GANT61 (20µM; Enzo Life Sciences, Germany) or vehicle control (DMSO, 0.2%), in duplicate, for 24 hr, followed by washing ×1 with PBS, trypsinization, and centrifugation. Cells were fixed with 70% ethanol at RT, 20–30 min, stored at −20°C overnight, centrifuged for 5 min at 200×g to remove ethanol, and washed ×1 in PBS. The cells were resuspended in PBS, and low molecular weight DNA was extracted using DNA extraction buffer (0.2M Na2HPO4 and 0.1M citric acid, pH 7.8) for 5 min. The extracted DNA was centrifuged and resuspended in DNA staining solution containing propidium iodide (50 µg) and DNAse-free RNAse (2 mg), and allowed to incubate for 30 min in the dark at RT. Distribution of cells throughout the cell cycle was analyzed using a FACSCalibur flow cytometer, and data were analyzed using CellQuest software.

RNA isolation

Cells were seeded at 7×106 cells/10 cm plate overnight at ≈60% confluency, and subsequently treated with either vehicle control (0.2% DMSO) or GANT61 (20 µM), in duplicate, for 24 hr. Cells were subsequently harvested and RNA was isolated using the RNeasy® Mini Kit (Qiagen, Valencia, CA) following the manufacturer's protocol. Integrity of the RNA was determined by spectrophotometry and electrophoresis.

cDNA microarray analysis

RNA (250 ng) was reverse transcribed into cRNA and biotin-UTP labeled using the Illumina® TotalPrep RNA Amplification Kit (Ambion, Applied Biosystems, Foster City, CA) according to the manufacturer's protocol. cRNA was quantified using a nanodrop spectrophotometer, and the cRNA quality (size distribution) further analyzed on a 1% agarose gel. Biotinylated cRNAs were hybridized to the Illumina Human-ref8 V3.0 BeadChip (Illumina, San Diego, CA) that represents 18,401 genes, using standard protocols. The arrays were washed and subsequently scanned using an Illumina BeadArray Reader.

Raw signal intensities of gene expression data were processed and analyzed using GenomeStudio (Illumina), with background subtraction and average normalization performed on the average signal intensities, and p-values were calculated. Raw data were exported from GenomeStudio into Excel (available as Data S1 and Data S2), and the fold change in gene expression calculated, by dividing the average gene expression signal intensity of treated samples (GANT61) with that of the vehicle control (DSMO). Differential gene expression (DEG) analysis between control and GANT61-treated samples, was subsequently conducted using the Illumina customer model, which applies multiple testing corrections that determines the false discovery rate (FDR; [55]. Thus, FDR-adjusted differential scores and p-values for each gene/probe set between treated and control samples were generated. Genes with a FDR-adjusted p-value of p<0.001 and fold change ≥1.5 were considered to be DEGs and were subjected to Venn analysis and Ingenuity Pathway analysis.

These differentially expressed genes were, uploaded and mapped to the library of canonical pathways of the Ingenuity Pathways Analysis (IPA) database for pathway analysis (Ingenuity® Systems, http://www.ingenuity.com, Mountain View, CA). The mapped datasets containing either up-regulated or down-regulated genes in HT29 or GC3/c1 cells with corresponding expression values were subjected to core analysis that includes overall canonical pathway enrichment analysis. The significance of enrichment of genes mapped to different canonical pathways was calculated by the Fischer's exact test (p-value) to determine the probability that the association between the genes and the canonical pathway could be explained by chance alone. Further, the ratio between the number of identified genes in a particular pathway and total number of genes that make up that pathway provides an estimation of the extent of pathway involvement. The enriched canonical pathways were ranked by −log (p-value) as shown in a histogram of pathway vs. –log (p-value). Datasets containing both up-regulated and down-regulated genes were also analyzed in selected pathways pertaining to cell cycle progression at G1/S and G2/M.

Fold change patterns of most highly DEGs in HT29 and GC3/c1 were compared by Heat map analysis using Matlab (Mathworks) software. DEGs analyzed and displayed in the heat map demonstrated a differential expression p-value of p<0.001 between control and GANT61 treated cells in both cell lines.

Quantitative Real-Time PCR (qRT-PCR)

The expression levels of selected genes identified by cDNA microarray expression profiling at 24 hr following GANT61 treatment, were validated by qRT-PCR. Thus, HT29 or GC3/c1 cells were either untreated (vehicle control, 0.2% DMSO), or treated with GANT61 (20 µM) for 0 hr, 16 hr, 24 hr, 38 hr or 48 hr at 37°C, dissolved in DMSO-containing medium. Total RNA (1 µg) was employed to prepare cDNA via reverse transcription using the iScript Select cDNA Synthesis Kit (Biorad) Reverse Transcription System according to manufacturers instructions and analyzed using an Applied Biosystems 7500 PCR Detection System (Applied Biosystems Inc.). All amplifications were primed by pairs of chemically synthesized 18- to 24- mer oligonucleotides designed using freely available primer design software (Primer-BLAST, NCBI) to generate target amplicons of 50–547 bp. All reactions were performed in a final volume of 15 µl. qRT-PCR reaction conditions were as follows: activation at 95°C for 10 min with 40 cycles of denaturation at 95°C for 15 s, primer annealing and extension at 60°C for 1 min and ramping back to 95°C. Melt curve analysis of all samples was routinely performed to ascertain that only the expected products had been generated. A fluorescence reading determined the extent of amplification at the end of each cycle. mRNA expression levels of target genes were normalized to the expression of glyceraldehyde phosphate dehydrogenase (GAPDH) and quantified using the comparative CT method [56]. Q-RT-PCR for each gene was determined in duplicate, and each experiment was repeated at least twice.

Supporting Information

Data S1

HT29 Raw Microarray Data.

(11.39 MB XLS)

Data S2

GC3/c1 Raw Microarray Data.

(11.06 MB XLS)

Acknowledgments

The authors would like to acknowledge the Lerner Research Institute Genomic Core Facility for performing oligonucleotide hybridization.

Footnotes

Competing Interests: The authors have declared that no competing interests exist.

Funding: Financial support from National Cancer Institute awards RO1 CA 32613 (JAH), RO1 108929 (JAH), and from the Cleveland Clinic. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

References

  • 1.Lum L, Beachy PA. The Hedgehog response network: sensors, switches, and routers. Science. 2004;304:1755–1759. doi: 10.1126/science.1098020. [DOI] [PubMed] [Google Scholar]
  • 2.Hooper JE, Scott MP. Communicating with Hedgehogs. Nat Rev Mol Cell Biol. 2005;6:306–317. doi: 10.1038/nrm1622. [DOI] [PubMed] [Google Scholar]
  • 3.Katoh Y, Katoh M. Hedgehog target genes: mechanisms of carcinogenesis induced by aberrant hedgehog signaling activation. Curr Mol Med. 2009;9:873–886. doi: 10.2174/156652409789105570. [DOI] [PubMed] [Google Scholar]
  • 4.Ruiz i Altaba A, Mas C, Stecca B. The Gli code: an information nexus regulating cell fate, stemness and cancer. Trends Cell Biol. 2007;17:438–447. doi: 10.1016/j.tcb.2007.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Yu M, Gipp J, Yoon JW, Iannaccone P, Walterhouse D, et al. Sonic hedgehog-responsive genes in the fetal prostate. J Biol Chem. 2009;284:5620–5629. doi: 10.1074/jbc.M809172200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Kasper M, Schnidar H, Neill GW, Hanneder M, Klingler S, et al. Selective modulation of Hedgehog/GLI target gene expression by epidermal growth factor signaling in human keratinocytes. Mol Cell Biol. 2006;26:6283–6298. doi: 10.1128/MCB.02317-05. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Yoon JW, Kita Y, Frank DJ, Majewski RR, Konicek BA, et al. Gene expression profiling leads to identification of GLI1-binding elements in target genes and a role for multiple downstream pathways in GLI1-induced cell transformation. J Biol Chem. 2002;277:5548–5555. doi: 10.1074/jbc.M105708200. [DOI] [PubMed] [Google Scholar]
  • 8.Katoh Y, Katoh M. Integrative genomic analyses on GLI2: mechanism of Hedgehog priming through basal GLI2 expression, and interaction map of stem cell signaling network with P53. Int J Oncol. 2008;33:881–886. doi: 10.3892/ijo_00000076. [DOI] [PubMed] [Google Scholar]
  • 9.Regl G, Kasper M, Schnidar H, Eichberger T, Neill GW, et al. The zinc-finger transcription factor GLI2 antagonizes contact inhibition and differentiation of human epidermal cells. Oncogene. 2004;23:1263–1274. doi: 10.1038/sj.onc.1207240. [DOI] [PubMed] [Google Scholar]
  • 10.Varnat F, Zacchetti G, Ruiz i Altaba A. Hedgehog pathway activity is required for the lethality and intestinal phenotypes of mice with hyperactive Wnt signaling. Mech Dev. 127:73–81. doi: 10.1016/j.mod.2009.10.005. [DOI] [PubMed] [Google Scholar]
  • 11.Alinger B, Kiesslich T, Datz C, Aberger F, Strasser F, et al. Hedgehog signaling is involved in differentiation of normal colonic tissue rather than in tumor proliferation. Virchows Arch. 2009;454:369–379. doi: 10.1007/s00428-009-0753-7. [DOI] [PubMed] [Google Scholar]
  • 12.van den Brink GR. Linking pathways in colorectal cancer. Nat Genet. 2004;36:1038–1039. doi: 10.1038/ng1004-1038. [DOI] [PubMed] [Google Scholar]
  • 13.Katoh Y, Katoh M. Hedgehog signaling pathway and gastrointestinal stem cell signaling network (review). Int J Mol Med. 2006;18:1019–1023. [PubMed] [Google Scholar]
  • 14.Kasper M, Regl G, Frischauf AM, Aberger F. GLI transcription factors: mediators of oncogenic Hedgehog signalling. Eur J Cancer. 2006;42:437–445. doi: 10.1016/j.ejca.2005.08.039. [DOI] [PubMed] [Google Scholar]
  • 15.Thiyagarajan S, Bhatia N, Reagan-Shaw S, Cozma D, Thomas-Tikhonenko A, et al. Role of GLI2 transcription factor in growth and tumorigenicity of prostate cells. Cancer Res. 2007;67:10642–10646. doi: 10.1158/0008-5472.CAN-07-2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Eichberger T, Sander V, Schnidar H, Regl G, Kasper M, et al. Overlapping and distinct transcriptional regulator properties of the GLI1 and GLI2 oncogenes. Genomics. 2006;87:616–632. doi: 10.1016/j.ygeno.2005.12.003. [DOI] [PubMed] [Google Scholar]
  • 17.Ruiz i Altaba A. Gli proteins encode context-dependent positive and negative functions: implications for development and disease. Development. 1999;126:3205–3216. doi: 10.1242/dev.126.14.3205. [DOI] [PubMed] [Google Scholar]
  • 18.Nguyen V, Chokas AL, Stecca B, Ruiz i Altaba A. Cooperative requirement of the Gli proteins in neurogenesis. Development. 2005;132:3267–3279. doi: 10.1242/dev.01905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Sanchez P, Hernandez AM, Stecca B, Kahler AJ, DeGueme AM, et al. Inhibition of prostate cancer proliferation by interference with SONIC HEDGEHOG-GLI1 signaling. Proc Natl Acad Sci U S A. 2004;101:12561–12566. doi: 10.1073/pnas.0404956101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Stecca B, Mas C, Clement V, Zbinden M, Correa R, et al. Melanomas require HEDGEHOG-GLI signaling regulated by interactions between GLI1 and the RAS-MEK/AKT pathways. Proc Natl Acad Sci U S A. 2007;104:5895–5900. doi: 10.1073/pnas.0700776104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Feldmann G, Dhara S, Fendrich V, Bedja D, Beaty R, et al. Blockade of hedgehog signaling inhibits pancreatic cancer invasion and metastases: a new paradigm for combination therapy in solid cancers. Cancer Res. 2007;67:2187–2196. doi: 10.1158/0008-5472.CAN-06-3281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Sarangi A, Valadez JG, Rush S, Abel TW, Thompson RC, et al. Targeted inhibition of the Hedgehog pathway in established malignant glioma xenografts enhances survival. Oncogene. 2009;28:3468–3476. doi: 10.1038/onc.2009.208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Dormoy V, Danilin S, Lindner V, Thomas L, Rothhut S, et al. The sonic hedgehog signaling pathway is reactivated in human renal cell carcinoma and plays orchestral role in tumor growth. Mol Cancer. 2009;8:123. doi: 10.1186/1476-4598-8-123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Yoshikawa K, Shimada M, Miyamoto H, Higashijima J, Miyatani T, et al. Sonic hedgehog relates to colorectal carcinogenesis. J Gastroenterol. 2009;44:1113–1117. doi: 10.1007/s00535-009-0110-2. [DOI] [PubMed] [Google Scholar]
  • 25.Bian YH, Huang SH, Yang L, Ma XL, Xie JW, et al. Sonic hedgehog-Gli1 pathway in colorectal adenocarcinomas. World J Gastroenterol. 2007;13:1659–1665. doi: 10.3748/wjg.v13.i11.1659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Varnat F, Duquet A, Malerba M, Zbinden M, Mas C, et al. Human colon cancer epithelial cells harbour active HEDGEHOG-GLI signalling that is essential for tumour growth, recurrence, metastasis and stem cell survival and expansion. EMBO Mol Med. 2009;1:338–351. doi: 10.1002/emmm.200900039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Nurse P. A long twentieth century of the cell cycle and beyond. Cell. 2000;100:71–78. doi: 10.1016/s0092-8674(00)81684-0. [DOI] [PubMed] [Google Scholar]
  • 28.Berger JH, Bardeesy N. Modeling INK4/ARF tumor suppression in the mouse. Curr Mol Med. 2007;7:63–75. doi: 10.2174/156652407779940477. [DOI] [PubMed] [Google Scholar]
  • 29.Stark GR, Taylor WR. Control of the G2/M transition. Mol Biotechnol. 2006;32:227–248. doi: 10.1385/MB:32:3:227. [DOI] [PubMed] [Google Scholar]
  • 30.McDonald ER, 3rd, El-Deiry WS. Cell cycle control as a basis for cancer drug development (Review). Int J Oncol. 2000;16:871–886. [PubMed] [Google Scholar]
  • 31.Harper JW, Elledge SJ, Keyomarsi K, Dynlacht B, Tsai LH, et al. Inhibition of cyclin-dependent kinases by p21. Mol Biol Cell. 1995;6:387–400. doi: 10.1091/mbc.6.4.387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Choi S, Kim TW, Singh SV. Ginsenoside Rh2-mediated G1 phase cell cycle arrest in human breast cancer cells is caused by p15 Ink4B and p27 Kip1-dependent inhibition of cyclin-dependent kinases. Pharm Res. 2009;26:2280–2288. doi: 10.1007/s11095-009-9944-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Polager S, Kalma Y, Berkovich E, Ginsberg D. E2Fs up-regulate expression of genes involved in DNA replication, DNA repair and mitosis. Oncogene. 2002;21:437–446. doi: 10.1038/sj.onc.1205102. [DOI] [PubMed] [Google Scholar]
  • 34.Lauth M, Bergstrom A, Shimokawa T, Toftgard R. Inhibition of GLI-mediated transcription and tumor cell growth by small-molecule antagonists. Proc Natl Acad Sci U S A. 2007;104:8455–8460. doi: 10.1073/pnas.0609699104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Ingham PW, McMahon AP. Hedgehog signaling in animal development: paradigms and principles. Genes Dev. 2001;15:3059–3087. doi: 10.1101/gad.938601. [DOI] [PubMed] [Google Scholar]
  • 36.Ruiz i Altaba A, Sanchez P, Dahmane N. Gli and hedgehog in cancer: tumours, embryos and stem cells. Nat Rev Cancer. 2002;2:361–372. doi: 10.1038/nrc796. [DOI] [PubMed] [Google Scholar]
  • 37.Ingham PW, Placzek M. Orchestrating ontogenesis: variations on a theme by sonic hedgehog. Nat Rev Genet. 2006;7:841–850. doi: 10.1038/nrg1969. [DOI] [PubMed] [Google Scholar]
  • 38.Riobo NA, Lu K, Ai X, Haines GM, Emerson CP., Jr Phosphoinositide 3-kinase and Akt are essential for Sonic Hedgehog signaling. Proc Natl Acad Sci U S A. 2006;103:4505–4510. doi: 10.1073/pnas.0504337103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Riobo NA, Haines GM, Emerson CP., Jr Protein kinase C-delta and mitogen-activated protein/extracellular signal-regulated kinase-1 control GLI activation in hedgehog signaling. Cancer Res. 2006;66:839–845. doi: 10.1158/0008-5472.CAN-05-2539. [DOI] [PubMed] [Google Scholar]
  • 40.Schnidar H, Eberl M, Klingler S, Mangelberger D, Kasper M, et al. Epidermal growth factor receptor signaling synergizes with Hedgehog/GLI in oncogenic transformation via activation of the MEK/ERK/JUN pathway. Cancer Res. 2009;69:1284–1292. doi: 10.1158/0008-5472.CAN-08-2331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Varnat F, Zacchetti G, Ruiz IAA. Hedgehog pathway activity is required for the lethality and intestinal phenotypes of mice with hyperactive Wnt signaling. Mech Dev. 2009 doi: 10.1016/j.mod.2009.10.005. [DOI] [PubMed] [Google Scholar]
  • 42.Monzo M, Moreno I, Artells R, Ibeas R, Navarro A, et al. Sonic hedgehog mRNA expression by real-time quantitative PCR in normal and tumor tissues from colorectal cancer patients. Cancer Lett. 2006;233:117–123. doi: 10.1016/j.canlet.2005.03.001. [DOI] [PubMed] [Google Scholar]
  • 43.Oniscu A, James RM, Morris RG, Bader S, Malcomson RD, et al. Expression of Sonic hedgehog pathway genes is altered in colonic neoplasia. J Pathol. 2004;203:909–917. doi: 10.1002/path.1591. [DOI] [PubMed] [Google Scholar]
  • 44.Koyama M, Matsuzaki Y, Yogosawa S, Hitomi T, Kawanaka M, et al. ZD1839 induces p15INK4b and causes G1 arrest by inhibiting the mitogen-activated protein kinase/extracellular signal-regulated kinase pathway. Mol Cancer Ther. 2007;6:1579–1587. doi: 10.1158/1535-7163.MCT-06-0814. [DOI] [PubMed] [Google Scholar]
  • 45.Gartel AL, Tyner AL. The role of the cyclin-dependent kinase inhibitor p21 in apoptosis. Mol Cancer Ther. 2002;1:639–649. [PubMed] [Google Scholar]
  • 46.Niculescu AB, 3rd, Chen X, Smeets M, Hengst L, Prives C, et al. Effects of p21(Cip1/Waf1) at both the G1/S and the G2/M cell cycle transitions: pRb is a critical determinant in blocking DNA replication and in preventing endoreduplication. Mol Cell Biol. 1998;18:629–643. doi: 10.1128/mcb.18.1.629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Ogryzko VV, Wong P, Howard BH. WAF1 retards S-phase progression primarily by inhibition of cyclin-dependent kinases. Mol Cell Biol. 1997;17:4877–4882. doi: 10.1128/mcb.17.8.4877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Sandhu C, Donovan J, Bhattacharya N, Stampfer M, Worland P, et al. Reduction of Cdc25A contributes to cyclin E1-Cdk2 inhibition at senescence in human mammary epithelial cells. Oncogene. 2000;19:5314–5323. doi: 10.1038/sj.onc.1203908. [DOI] [PubMed] [Google Scholar]
  • 49.Abbas T, Dutta A. p21 in cancer: intricate networks and multiple activities. Nat Rev Cancer. 2009;9:400–414. doi: 10.1038/nrc2657. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Chang BD, Watanabe K, Broude EV, Fang J, Poole JC, et al. Effects of p21Waf1/Cip1/Sdi1 on cellular gene expression: implications for carcinogenesis, senescence, and age-related diseases. Proc Natl Acad Sci U S A. 2000;97:4291–4296. doi: 10.1073/pnas.97.8.4291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Chang BD, Broude EV, Fang J, Kalinichenko TV, Abdryashitov R, et al. p21Waf1/Cip1/Sdi1-induced growth arrest is associated with depletion of mitosis-control proteins and leads to abnormal mitosis and endoreduplication in recovering cells. Oncogene. 2000;19:2165–2170. doi: 10.1038/sj.onc.1203573. [DOI] [PubMed] [Google Scholar]
  • 52.Keen N, Taylor S. Mitotic drivers–inhibitors of the Aurora B Kinase. Cancer Metastasis Rev. 2009;28:185–195. doi: 10.1007/s10555-009-9184-9. [DOI] [PubMed] [Google Scholar]
  • 53.Ge S, Skaar JR, Pagano M. APC/C- and Mad2-mediated degradation of Cdc20 during spindle checkpoint activation. Cell Cycle. 2009;8:167–171. doi: 10.4161/cc.8.1.7606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Tillman DM, Izeradjene K, Szucs KS, Douglas L, Houghton JA. Rottlerin sensitizes colon carcinoma cells to tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis via uncoupling of the mitochondria independent of protein kinase C. Cancer Res. 2003;63:5118–5125. [PubMed] [Google Scholar]
  • 55.Reiner A, Yekutieli D, Benjamini Y. Identifying differentially expressed genes using false discovery rate controlling procedures. Bioinformatics. 2003;19:368–375. doi: 10.1093/bioinformatics/btf877. [DOI] [PubMed] [Google Scholar]
  • 56.Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 2001;25:402–408. doi: 10.1006/meth.2001.1262. [DOI] [PubMed] [Google Scholar]

Associated Data

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

Supplementary Materials

Data S1

HT29 Raw Microarray Data.

(11.39 MB XLS)

Data S2

GC3/c1 Raw Microarray Data.

(11.06 MB XLS)


Articles from PLoS ONE are provided here courtesy of PLOS

RESOURCES