Open Access

Wnt/β‑catenin pathway activation and silencing of the APC gene in HPV‑positive human cervical cancer‑derived cells

  • Authors:
    • Erick Ayala‑Calvillo
    • Luis Humberto Mojica‑Vázquez
    • Alejandro García‑Carrancá
    • Leticia González‑Maya
  • View Affiliations

  • Published online on: October 20, 2017     https://doi.org/10.3892/mmr.2017.7853
  • Pages: 200-208
  • Copyright: © Ayala‑Calvillo et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Although persistent infections with high‑risk human papilloma virus (HPV) constitute the most significant cofactor for the development of cervical cancer, they are insufficient on their own. Mutations or epigenetic inactivation of the tumor suppressor adenomatous polyposis coli (APC), the two acting as prominent oncogenic mechanisms in a number of types of cancer, are frequently associated with aberrant activation of the Wnt/β‑catenin pathway. According to these observations, it was hypothesized that APC alteration may lead to β‑catenin deregulation and the abnormal expression of direct targets of the Wnt pathway in HPV‑infected cervical cancer cells. The present study confirmed that the stabilization of β‑catenin correlates with enhanced transcriptional activity of the β‑catenin/T‑cell factor complex in cervical cancer cell lines. Sequence analysis of the ‘hot‑spot’ in the mutation cluster region did not exhibit genetic alterations that may be associated with APC gene inactivation. In addition, it was identified that there was a good correlation with the hypermethylation status of the APC promoter 1A and the abnormal accumulation of endogenous β‑catenin in cell lines and biopsies infected with HPV16, although not HPV18. Removal of the epigenetic markers led to an increase in APC levels and a reduction of β‑catenin expression in two transcriptional targets of the Wnt pathway: Matrix metalloproteinase‑7 and vascular endothelial growth factor. The present study suggested that the increase in Wnt activity in certain cervical cancer‑derived cells may be associated with an alteration in the methylation status of the APC gene promoter 1A.

Introduction

The Wnt/β-catenin pathway serves critical functions in the sequential development of the neoplastic process from initiation, proliferation and transformation in a broad range of types of cancer. Aberrant activation of the Wnt pathway may lead to β-catenin stabilization and accumulation in the cytoplasm, allowing its translocation into the nucleus to act as a transcriptional co-activator of members of the T-cell factor (TCF)/lymphoid enhancing factor (LEF) family of transcription factors. It is known that the β-catenin/TCF-LEF complex induces the transcription of genes involved in carcinogenesis including c-myc, matrix metalloproteinase-7 (MMP-7) and the vascular endothelial growth factor (VEGF) (1). The tumor suppressor adenomatous polyposis coli (APC), an essential part of a multi-protein complex together with protein phosphatase 2A (PP2A), axin, glycogen synthase kinase 3β (GSK-3β), and casein kinase I (CK1), serves an important function in turning off the β-catenin signaling pathway. When this complex is active, GSK-3β and CK1 phosphorylate the amino terminal serine and threonine residues of β-catenin, an initial step which targets it for degradation by the proteasomal machinery (2).

Cervical carcinoma, one of the most common malignancies in women worldwide (3), is associated with persistent infection with oncogenic or high-risk human papillomavirus (HR-HPV). Although HR-HPV infection is required, it is insufficient on its own to cause cervical cancer (4). Several lines of evidence have indicated a potential association between activation of the Wnt signaling pathway and HPV-mediated cervical cancer. For instance, Uren et al (5) and Bulut et al (6) generated two models that mimicked the effects of HPV infection on the human cervical epithelium for cancer progression: i) HPV-immortalized human keratinocytes, either expressing mutated S37A β-catenin or overexpressing Wnt1; and ii) double-transgenic mice overexpressing β-catenin and HPV16-E7 oncogenes. In addition, β-catenin has been observed to be localized and expressed in the cytoplasm and nucleus in human cervical carcinoma samples (7,8) and cell lines (9), indicating the activation of the Wnt pathway. The results of these previous studies suggested that activation of the canonical Wnt pathway may represent one of the key events required for the malignant transformation of HPV-infected epithelial cells.

Previous studies have demonstrated that β-catenin deregulation is frequently associated with alterations in the APC gene, and ~80 to 90% of all APC gene mutations are confined to codons 1,286 to 1,513, a region known as the mutation cluster region (MCR), these mutations typically lead to a truncated protein lacking the β-catenin binding and regulatory sites, as is frequently observed in colon cancer (10,11). In addition, transcriptional silencing via promoter hypermethylation may impair APC function, a phenomenon of epigenetic inactivation observed in tumors in the absence of APC gene mutations. For example, in gastric cancer, a decrease in or loss of APC expression has been identified to be associated with the APC gene promoter A1 hypermethylation (12,13).

According to these observations, it has been suggested that the stabilization and activation of β-catenin in cervical cancer-derived cells are a consequence of alterations in APC gene expression. To test this hypothesis, the present study first sought to determine the transcriptional activity of β-catenin/TCF among a group of cervical cancer cell lines. Subsequently, MCR-region mutational and methylation status analyses of the APC gene and its promoter 1A, respectively, were performed and the impact of APC gene mutations and/or its epigenetic silencing on two known Wnt target genes in cervical carcinoma cell lines was analyzed.

Materials and methods

Cell cultures and clinical samples

Human cervical cancer-derived cell lines CaSki, SiHa and HeLa, in addition to C33A and the colon cancer cell line SW480 were obtained from the American Type Culture Collection (Manassas, VA, USA). The gastric cancer cell line KATOIII was provided by Dr Angel Zarain-Herzberg (Facultad de Medicina-UNAM, CDMX, México). Human foreskin fibroblasts (HFF) were provided by Dr. María Luisa Villarreal Ortega (Centro de Investigaciones en Biotecnología-UAEM, Cuernavaca, Morelos, México). HFF and CaSki cells were grown in RPMI-1640 (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA); SiHa, HeLa, C33A, SW480 and KATOIII cells were grown in Dulbecco's modified Eagle's medium/F-12 (Invitrogen; Thermo Fisher Scientific, Inc.), and all the cultured cells were supplemented with 10% (v/v) fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc.) and 2 mM L-glutamine (Invitrogen; Thermo Fisher Scientific, Inc.). Cells were cultured at 37°C in a humidified atmosphere containing 5% CO2. Normal tissue samples and tumors from the uterine cervix were obtained from archives of the Department of Gynecology at the Hospital General Manuel Gea González and the Department of Pathology at the Instituto Nacional de Cancerología-SSA (CDMX, México). This set of samples has been characterized previously for clinical pathological parameters, type of HPV and β-catenin protein status (7). All the samples were frozen in liquid nitrogen immediately following resection and stored at −70°C until processing.

Reporter assay

The TOPflash/FOPflash TCF reporter system (Upstate Biotechnology, Inc., Lake Placid, NY, USA) was used to directly measure the levels of Wnt signaling in human cervical cancer cell lines. A total of 4,000 cells were cultured in 96-well plate and transfected, with either TOPflash or FOPflash (100 ng) and the internal control plasmid pRL-Renilla (5 ng; Promega Corporation, Madison, WI, USA) expressing Renilla luciferase, using Lipofectamine reagent (Invitrogen; Thermo Fisher Scientific, Inc.). The TOPflash and FOPflash reporters contain two sets of three copies of wild-type (TOP) or mutant (FOP) β-catenin/TCF binding sites, respectively, in addition to the thymidine kinase minimal promoter upstream of the firefly luciferase open reading frame. Luciferase activity was measured with Dual-Glo Luciferase Assay System (Promega Corporation) 48 h following transfection, according to the manufacturer's protocol. Firefly luciferase activity was normalized to Renilla luciferase activity. All results are expressed as TOPflash/FOPflash correlation mean ± standard deviation for independent triplicate cultures.

Mutational analysis of APC

Genomic DNA was isolated from cells using the standard Proteinase K treatment followed by phenol/chloroform extraction (14). Briefly, the cells were incubated in the lysis buffer (Tris 10 mM, EDTA 20 mM, SDS 0.5% and Proteinase K 100 µM/ml) for 20 min at 45°C. The lysate was extracted by phenol/chloroform (1:1, v/v), DNA was precipitated with isopropanol (1:1, v/v), then washed with 75% ethanol and the pellet DNA was dissolved in water. The mutation cluster region of the APC gene was amplified as three overlapping fragments (codons 1032-1703) in a nested polymerase chain reaction (PCR) strategy. In addition to the previously described primers (15), new primers were used to adapt the size of different amplicons (Table I). PCR was performed in a 50-µl volume containing 100 ng genomic DNA and 0.25 µM each primer using Taq DNA Polymerase-recombinant (Fermentas; Thermo Fisher Scientific, Inc.). The PCR reactions were run at 95°C for 5 min followed by 35 cycles at 95°C for 40 sec, followed by 54°C, 63°C and 60°C for 60 sec as the annealing step of the three overlapping fragments, respectively, and finally at 72°C for 60 sec. The PCR products were purified using the Qiaquick PCR purification kit (Qiagen GmbH, Hilden, Germany). Mutational analysis by direct sequencing was performed using the BigDye Terminator version 3.1 Cycle Sequencing kit (Applied Biosystems; Thermo Fisher Scientific, Inc.). All sequencing analyses were performed at least twice on two independent PCR products.

Table I.

Primer sequences used in the present study.

Table I.

Primer sequences used in the present study.

Gene and methodSense 5′-3′Antisense 5′-3′Product length (bp)
Mutational analysis
  Fragment A CCCCTCGAGTCAGATGAGCAGTTG CCGGATCCCTGCTTCCTGTGTCG796
  Fragment B CCCCTCGAGCAGCTCCATCCAAG CCGGATCCCCATCTGGAGTAC809
  Fragment C CCCCTCGAGCCAGATAGCCCTGG CCGGATCCCCTCCTTGAGCCTC839
MSP
  APCprom1AUNMet GTGTTTTATTGTGGAGTGTGGGTT CCAATCAACAAACTCCCAACAA108
  APCprom1AMet TATTGCGGAGTGCGGGTC TCGACGAACTCCCGACGAA  98
RT-PCR
  MMP7 TGTTAAACTCCCGCGTCATA GCGTTCATCCTCATCGAAGT379
  VEGF GAGGGCAGAATCATCACGAA AACGCTCCAGGACTTATACC395
  β-globin CAACTTCATCCACGTTCACC GAAGAGCCAAGGACAGGTAC260
  APC-exon 15 CCCCTCGAGTCAGATGAGCAGTTG CCGGATCCCTGCTTCCTGTGTCG796

[i] bp, base pair; RT-PCR, reverse transcription-polymerase chain reaction; MMP-7, matrix metalloproteinase-7; VEGF, vascular endothelial growth factor; APC, adenomatous polyposis coli.

Methylation analysis

DNA methylation patterns in the CpG islands of the APC gene promoter A1 were determined by methylation-specific PCR (MSP), according to protocols described previously (16). Genomic DNA (1 µg) was treated with sodium bisulfite using the EZ DNA methylation kit (Zymo Research Corp., Irvine, CA, USA). For the MSP analysis of APC gene promoter 1A, the primers for the unmethylated reaction amplify a 108-bp PCR product, and the primers for the methylated reaction amplify a 98-bp product; all primer sequences used are listed in Table I. PCR amplification was carried out at 95°C for 5 min followed by 35 cycles at 95°C for 60 sec, 60°C for 40 sec and 72°C for 60 sec. PCR was run in a 50-µl volume containing 100 ng genomic DNA, 0.25 µM each primer using Taq DNA Polymerase-recombinant (Fermentas; Thermo Fisher Scientific, Inc.). Each experiment was repeated twice.

Treatment with 5-aza-2′-deoxycytidine (Aza) and trichostatin-A (TSA)

Cells were cultured at 60% confluence and incubated for 48 h in a medium containing 3 µM DNA methyltransferase (DNMT) inhibitor Aza (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany). The medium was refreshed every 24 h. The cells were treated with the histone deacetylase (HDAC) inhibitor TSA (Sigma-Aldrich; Merck KGaA) at 0.5 and 3 µM of decitabine for further 24 h to complete 72 h of treatment. The medium was removed and the cells were processed to perform assays by reverse transcription (RT)-PCR and western blot analysis.

RNA isolation and RT-PCR

Total RNA was extracted from cells by using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. For RT-PCR, total RNA (5 µg) was used for cDNA synthesis by reverse transcription using a RevertAid H. Minus First Strand cDNA Synthesis kit (Fermentas; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol, in a total volume of 20 µl. The PCR reactions of APC, VEGF, MMP7 and β-globin genes were performed using the specific primers presented in Table I, and standard PCR conditions were used: 95°C for 5 min, followed by 35 cycles at 95°C for 60 sec, annealing at 60°C, 62°C, 58°C and 55°C for each gene, respectively, for 60 sec, and extension at 72°C for 60 sec. The PCR products were visualized by electrophoresis in 2% agarose gels.

Western blot analysis

Cells were homogenized in lysis buffer (50 mM Tris-HCl, 150 mM NaCl, 1% NP40, 0.5% sodium deoxycholate and 0.1% SDS), containing protease inhibitors cocktail (Roche Diagnostics GmbH, Mannheim, Germany). The protein concentration was determined by the bicinchoninic acid assay (Pierce; Thermo Fisher Scientific, Inc.). Total proteins (25 µg) was separated by a 7% SDS-PAGE and transferred onto polyvinylidene fluoride membranes (EMD Millipore, Billerica, MA, USA). The antibodies and dilutions used included anti-β-catenin (cat. no. C19220; 1:2,000; BD Biosciences, San Jose, CA, USA) and anti-β-actin (cat. no. 81178, 1:100; Santa Cruz Biotechnology Inc., Dallas, TX, USA), both incubated for 5 h at room temperature and following extensive washing the membranes were incubated with anti-mouse immunoglobulin G-horseradish peroxidase-conjugated antibody (cat. no. Nr7074, 1:5,000; Cell Signaling Technology, Inc., Danvers, MA, USA) for 1 h at room temperature and revealed with Western Lightning Chemiluminescence Reagent Plus (PerkinElmer, Inc., Waltham, MA, USA). Membranes were probed for β-actin to normalize for loading.

Statistical analysis

Data were analysed using the GraphPad Prism 5.0 statistical program (GraphPad Software, Inc., La Jolla, CA, USA) and statistical differences were evaluated using one-way analysis of variance followed by Tukey's multiple comparisons test. P<0.05 was considered to indicate a statistically significant difference.

Results

β-catenin expression and transcriptional activity of the β-catenin/TCF complex in cervical cancer cell lines

β-catenin levels in different cell lines were analyzed using western blotting and there was abundant β-catenin in CaSki cells, low levels in HeLa cells, intermediate levels in SiHa and none detected in C33A cancer-derived cells (Fig. 1A). The transcriptional activity of the β-catenin/TCF complex in these four cell lines (CaSki, SiHa, HeLa and C33A) was subsequently analyzed using a luciferase reporter assay with TCF reporter constructs (TOPflash and FOPflash). The results demonstrated that the activity of the TCF reporter TOPflash was increased compared with the FOPflash reporter in all cell lines except C33A cells. The transcriptional activity of the β-catenin/TCF complex was higher in CaSki cells, with higher β-catenin levels, compared with the transcriptional activity observed in SiHa and Hela cells (Fig. 1B). The CaSki cells, infected with HPV16, thus exhibited the highest levels of β-catenin expression and transcriptional activity. These results demonstrated that the activation of the Wnt/β-catenin signaling pathway responded in a concentration-dependent manner to the levels of β-catenin in these cell lines.

APC gene mutations in cervical cancer cell lines

To investigate the existence of sequence variation in APC, one of the most important members of the β-catenin degradation complex, the MCR region, was evaluated via PCR and direct sequencing. As a positive control, DNA extracted from the colon cancer cell line SW480 was used, homozygous for a mutation at codon 1338 that generates a truncated APC protein (17), leading to abnormal accumulation/delocalization of β-catenin. The sequence analysis of the MCR did not reveal any mutations that were associated with the activation of APC in the cervical cancer cell lines CaSki, SiHa, HeLa and C33A (Fig. 2). None of the known ‘hot-spots’ in codons 1061, 1338 and 1450 were mutated, the most common sites of mutation in colorectal cancers (18). The mutation at codon 1338 (CAG/TAG) in SW480 cell line was detected (Fig. 2), confirming the single base specificity of the sequence analysis detection level. These results suggested that APC mutations are not responsible for the β-catenin accumulation observed in the cervical cancer cell lines analyzed.

Methylation of the APC gene promoter 1A in cervical cancer cell lines and biopsies

It has been demonstrated that APC downregulation may be associated with epigenetic rather than genetic alterations in other tumor cell types (19,20). To investigate that possibility, the methylation status of the APC gene promoter 1A in cervical cancer cell lines was analyzed by MSP. The gastric cancer cell line KATOIII, which exhibits bi-allelic methylation (12) and HFF, without methylation, were used as positive and negative controls, respectively. The APC gene promoter 1A did not exhibit DNA methylation at CpG dinucleotides in the C33A, HeLa and SiHa cell lines. However, CaSki cells demonstrated heterozygous signals, indicating methylated and unmethylated alleles of the APC gene promoter 1A. (Fig. 3A).

To further investigate a possible association between the methylated status of the APC promoter, the levels of expression of β-catenin or its aberrant localization and HPV infection, the same MSP analysis was applied to a small group of biopsies from tumors of the uterine cervix. Different samples, with variations in the delocalization/accumulation of β-catenin and the types of HPV (HPV-16 or −18), were analyzed (7). Notably, the MSP results demonstrated that methylated alleles (homozygous) or unmethylated/methylated alleles (heterozygous) of the APC gene promoter 1A were only amplified in cervical tumor samples that exhibited abnormal subcellular localization of β-catenin and were infected by the HPV16 type (Fig. 3B). These observations indicated a clear association between the methylation status of the APC gene promoter 1A and the abnormal localization of β-catenin observed in cervical carcinoma specimens infected by HR-HPV16.

APC expression and Aza/TSA treatment

To determine how the methylation patterns of APC gene promoter 1A identified in certain cervical cancer cell lines may affect APC transcript levels, their abundance was examined by quantifying exon 15 of APC by RT-PCR analysis. Abundant APC transcript expression in C33A, SiHa and Hela cell lines and HFF was identified where the APC promoter 1A was not methylated, whereas the KATOIII cell line, which demonstrated bi-allelic methylation, exhibited undetectable levels of APC mRNA. As expected, a marked reduction of APC transcripts in CaSki cells, which presented a heterozygous methylation pattern, was identified (Fig. 4A). These observations indicated that methylation of the APC gene promoter 1A led to the repression of APC expression. To further confirm this role of the methylation of APC gene promoter 1A in the transcriptional repression of the APC gene, KATOIII cells carrying fully methylated APC alleles and all cervical cancer cell lines were treated with the DNA methyltransferase inhibitor, Aza, together with TSA. Following treatment, the KATOIII cells demonstrated high levels of APC transcripts, a result associated with the demethylation of APC gene promoter 1A and the chromatin remodeling by histone acetylation (13). Of note, CaSki cells also demonstrated a marked increase in APC transcript expression following treatment (Fig. 4B), whereas the HFF, C33A, SiHa and HeLa cells did not demonstrate changes in APC expression levels. Taken together, these results highlighted that the levels of APC gene expression were negatively regulated by the methylation status of the promoter 1A in CaSki and KATOIII cells.

β-catenin expression and Wnt signaling pathway activity

It was observed that low levels of APC were linked to the methylation status of its promoter 1A. Since APC is a direct regulator of β-catenin, it was hypothesized that the reactivation of APC gene expression may lead to a reduction in β-catenin accumulation and a decrease in Wnt signaling pathway activity. To test this hypothesis, the expression of endogenous β-catenin and the expression of two β-catenin/TCF complex target genes in CaSki cells were evaluated prior to and following treatment with Aza and TSA. Prior to treatment, CaSki cells demonstrated abundant β-catenin protein levels, while following treatment; the β-catenin levels were significantly reduced. Consistently, alterations in β-catenin protein expression levels were observed in KATOIII cells, prior to and following treatment with Aza and TSA. In HFF, the negative control, the β-catenin protein levels did not alter following treatment (Fig. 5A). To further confirm the link between the presence of high β-catenin levels and high β-catenin/TCF complex activity, RT-PCR analysis was used to evaluate the expression of VEGF and MMP-7, two well-known target genes of the Wnt canonical pathway. The VEGF and MMP-7 mRNA levels were notably reduced following treatment in CaSki and KATOIII cells, while no alteration was observed in HFF (Fig. 5B). Taken together, these results confirmed that the methylation of the APC gene promoter 1A in CaSki cells had a direct effect on the expression of β-catenin and on the activity of the β-catenin/TCF complex, leading to increased levels of at least two Wnt target genes: VEGF and MMP-7. This suggested that this epigenetic modification of the APC gene promoter 1A may directly contribute to the expression of Wnt signaling pathway target genes involved in tumor growth and invasion.

Discussion

It has been widely accepted that abnormal activation of the Wnt signaling pathway leads to the stabilization and accumulation of β-catenin in the cytoplasm, then to its translocation to the nucleus, where it promotes the transcription of multiple genes involved in tumor growth and invasion (2,21,22). In certain types of cancer, including colorectal and hepatocellular carcinomas, deregulation of this pathway occurs almost invariably through mutation of the two key regulators: APC and β-catenin. These mutations lead to the reduced regulatory activity of APC and enhanced protein stability of β-catenin, respectively (23,24). No mutations in CTNNB1 (β-catenin gene) have yet been detected in cervical neoplasia or cancer-derived cell lines (8,9). Therefore, the present study aimed to identify mutations in the APC gene. However, no significant mutations were observed at the ‘hot-spot’ codons (1061, 1338 and 1450) within the MCR, the most common site of alterations that usually lead to a truncated protein lacking β-catenin binding and regulation sites (25).

A number of studies have suggested aberrant methylation in promoter regions as an epigenetic mechanism leading to the deregulation of tumor suppressor genes. Esteller et al (16) first demonstrated the mono- or bi-allelic methylation of APC gene promoter 1A in tumors of the gastrointestinal tract, including colon, gastric, pancreatic, esophageal and hepatic carcinomas. Subsequent studies have detected a similar status of promoter 1A methylation in several cancers, although in gastric cancer it has been identified with particularly high frequencies, in >50% of the cases (12,13,26). Fu et al (27) suggested that the hypermethylation of APC gene promoter 1A led to moderate activation of Wnt signaling pathway in colorectal cancer, instead of the usual mutations involving the APC and β-catenin genes. In apparent contradiction with these results, studies in cervical cancer samples have reported a great diversity in the APC gene promoter 1A methylation status (28,29), although none evaluated the association between APC promoter methylation, APC gene expression and the aberrant activation of the Wnt signaling pathway. To reconcile these data, the methylation patterns of the APC gene promoter 1A were evaluated in cell lines and biopsies from cervical cancer, in order to determine the frequency of this epigenetic regulation. These samples exhibited different levels of accumulation/delocalization of β-catenin and were either infected by HR-HPV type 16 or 18. The results demonstrated that the methylation frequency ranged from 1/4 (25%) to 3/9 (~33%) in cell lines and biopsies, respectively. These results are in conflict with a previous report, which demonstrated a 90% frequency of APC promoter 1A methylation in biopsies from cervical cancer (30). However, the results of the present study are comparable to other studies, which have proposed that the methylation of the APC promoter 1A, analyzed by MSP or by quantitative MSP, is not as common in cervical cancer cells, ranging between 12 and 35% (3133). Notably, the results of the present study indicated a strong correlation between the methylation status of the APC gene promoter 1A and the delocalization/accumulation of β-catenin observed in biopsies that were infected by HPV16. Furthermore, alleles of APC promoter 1A which were completely unmethylated (homozygous) were observed in C33A (HPV negative), and HeLa cells (HPV18). The mechanism through which HPV16 promotes Wnt signaling requires further investigation. However, knocking down HPV16 E6/E7 reduced the Wnt signal in CaSki (HPV16) cells, while overexpressing HPV16 E6 and/or E7 increased the Wnt signal in C33A cells (34).

To confirm the role of the methylation of promoter 1A in the transcriptional regulation of the APC gene, APC gene expression was examined over the same panel of cervical cancer cell lines. The results demonstrated no significant difference in APC transcript levels in C33A, SiHa and Hela cell lines when compared with HFF, where the APC gene promoter 1A was not methylated. The absence of APC mRNA in the KATOIII cell line, which presented bi-allelic hypermethylation, was confirmed (12), and it was identified that CaSki cells, which demonstrated a heterogeneous methylation pattern, exhibited a marked reduction of APC mRNA levels. The RT-PCR and MSP analyses thus demonstrated a correlation between APC expression levels and the APC gene promoter 1A methylation status in the cell lines. Additionally, since some studies have demonstrated that the lack of expression of tumor suppressor genes may be reversed by treatment with epigenetic silencing inhibitors (inhibitor of DNMT and HDAC) in cancer cells (35,36), this strategy was employed to evaluate the recovery of APC gene expression. The results demonstrated a significant increase in APC mRNA levels following treatment with Aza and TSA in CaSki cells, whereas no significant alteration was observed in C33A, SiHa and HeLa cells. Similar results were observed in melanoma and gastric cancer cell lines carrying a mono- or bi-allelic methylation of APC gene promoter 1A, in which the low or absent APC expression levels were modified following Aza and TSA treatment, leading to its active transcription and recovery of APC expression (13,37). These results indicated that the presence of promoter methylation suppressed APC gene transcription and contributed to inactivating the APC tumor suppressor function in CaSki cells.

Conversely, previous studies with human cervical carcinoma samples observed an abnormal accumulation/delocalization of β-catenin, which constitutes a hallmark of the activated Wnt pathway (7,8). The present study confirmed through western blot analyses and luciferase reporter assays that the abnormal expression of β-catenin is associated, in a concentration-dependent manner, with the transcriptional activity of the β-catenin/TCF complex in CaSki, SiHa and HeLa cervical cancer cells infected with HR-HPV16 or 18. In CaSki cells (HPV16), with the greatest β-catenin expression, the strongest transcriptional activity was observed, whereas neither expression of β-catenin, nor β-catenin/TCF transcriptional activity was observed in non-HPV infected C33A cervical cancer cells (38,39). To determine the role of the APC promoter methylation in the regulation of Wnt/β-catenin signaling, the expression of endogenous β-catenin and the expression of β-catenin/TCF complex target genes were evaluated following treatment with Aza and TSA. In CaSki cells, a reduction of β-catenin levels was observed, in addition to a decrease in the transcripts of two β-catenin/TCF target genes, VEGF and MMP-7. These results suggested that stimulating the expression of the APC gene via treatment with epigenetic silencing inhibitors reduced the abnormal activation of the Wnt pathway in CaSki cells. This conclusion is further supported by a study from Svedlund et al (40), which demonstrated that the treatment of a primary PC cell culture with the DNA hypomethylating agent Aza induced APC expression and reduced β-catenin levels. Taken together, these data suggested that the re-expression of the APC gene in CaSki cells appears to be sufficient to assemble a functional complex of Axin/PP2A/GSK-3β/CK1/APC, capable of phosphorylating the β-catenin protein, eventually leading to its degradation and switching off the Wnt/β-catenin signaling pathway (41).

In conclusion, the present study proposed that methylation-dependent silencing of the APC gene promoter 1A is a mechanism that contributes to the activation of Wnt signaling pathway in cervical cancer cells infected by high risk HPV16. The reduction of APC levels induced by hypermethylation of the APC gene promoter 1A, rather than direct mutations in the APC or β-catenin genes, leads to accumulation of β-catenin, which in turn increases the Wnt/β-catenin transcriptional activity. This mechanism may lead to an increase in the transcription of genes involved in cancer development. Therefore, the development of an epigenetic therapy to reactivate the expression of APC could be advantageous for the treatment of cervical cancer cells-infected with HPV16. However, further studies need to be performed to confirm the above results.

References

1 

Polakis P: Wnt signaling in cancer. Cold Spring Harb Perspect Biol. 4:a0080522012. View Article : Google Scholar : PubMed/NCBI

2 

Munemitsu S, Albert I, Souza B, Rubinfeld B and Polakis P: Regulation of intracellular betacatenin levels by the adenomatous polyposis coli (APC) tumor-suppressor protein. Proc Natl Acad Sci USA. 92:3046–3050. 1995. View Article : Google Scholar : PubMed/NCBI

3 

Walboomers JM, Jacobs MV, Manos MM, Bosch FX, Kummer JA, Shah KV, Snijders PJ, Peto J, Meijer CJ and Muñoz N: Human papillomavirus is a necessary cause of invasive cervical cancer worldwide. J Pathol. 189:12–19. 1999. View Article : Google Scholar : PubMed/NCBI

4 

Moody CA and Laimins LA: Human papillomavirus oncoproteins: Pathways to transformation. Nat Rev Cancer. 10:550–560. 2010. View Article : Google Scholar : PubMed/NCBI

5 

Uren A, Fallen S, Yuan H, Usubütün A, Kücükali T, Schlegel R and Toretsky JA: Activation of the canonical Wnt pathway during genital keratinocyte transformation: A model for cervical cancer progression. Cancer Res. 65:6199–6206. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Bulut G, Fallen S, Beauchamp EM, Drebing LE, Sun J, Berry DL, Kallakury B, Crum CP, Toretsky JA, Schlegel R and Üren A: Beta-catenin accelerates human papilloma virus type-16 mediated cervical carcinogenesis in transgenic mice. PLoS One. 6:e272432011. View Article : Google Scholar : PubMed/NCBI

7 

Rodríguez-Sastre M, González-Maya L, Delgado R, Mohar A and García-Carrancá A: Abnormal distribution of E-cadherin and β-catenin in different histologic types of cancer of the uterine cervix. Gynecol Oncol. 97:330–336. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Shinohara A, Yokoyama Y, Wan X, Takahashi Y, Mori Y, Takami T, Shimokawa K and Tamaya T: Cytoplasmic/nuclear expression without mutation of exon 3 of the beta-catenin gene is frequent in the development of the neoplasm of the uterine cervix. Gynecol Oncol. 82:450–455. 2001. View Article : Google Scholar : PubMed/NCBI

9 

Pereira-Suárez A, Meraz MA, Lizano M, Estrada-Chávez C, Hernández F, Olivera P, Pérez E, Padilla P, Yaniv M, Thierry F and García-Carrancá A: Frequent alterations of the beta-catenin protein in in cancer of the uterine cervix. Tumor Biol. 23:45–53. 2002. View Article : Google Scholar

10 

Nakamura Y, Nishisho I, Kinzler KW, Vogelstein B, Miyoshi Y, Miki Y, Ando H, Horii A and Nagase H: Mutations of the adenomatous polyposis coli gene in familial polyposis coli patients and sporadic colorectal tumors. Princess Takamatsu Symp. 22:285–292. 1991.PubMed/NCBI

11 

Horii A, Nakatsuru S, Miyoshi Y, Ichii S, Nagase H, Kato Y, Yanagisawa A and Nakamura Y: The APC gene, responsible for familial adenomatous polyposis, is mutated in human gastric cancer. Cancer Res. 152:3231–3233. 1992.

12 

Tsuchiya T, Tamura G, Sato K, Endoh Y, Sakata K, Jin Z, Motoyama T, Usuba O, Kimura W, Nishizuka S, et al: Distinct methylation patterns of two APC gene promoters in normal and cancerous gastric epithelia. Oncogene. 19:3642–3646. 2000. View Article : Google Scholar : PubMed/NCBI

13 

Hosoya K, Yamashita S, Ando T, Nakajima T, Itoh F and Ushijima T: Adenomatous polyposis coli 1A is likely to be methylated as a passenger in human gastric carcinogenesis. Cancer Lett. 285:182–189. 2009. View Article : Google Scholar : PubMed/NCBI

14 

Javadi A, Shamaei M, Ziazi Mohammadi L, Pourabdollah M, Dorudinia A, Seyedmehdi SM and Karimi S: Qualification study of two genomic DNA extraction methods in different clinical samples. Tanaffos. 13:41–47. 2014.PubMed/NCBI

15 

Miyoshi Y, Ando H, Nagase H, Nishisho I, Horii A, Miki Y, Mori T, Utsunomiya J, Baba S, Petersen G, et al: Germ-line mutations of the APC gene in 53 familial adenomatous polyposis patients. Proc Natl Acad Sci USA. 89:4452–4456. 1992. View Article : Google Scholar : PubMed/NCBI

16 

Esteller M, Sparks A, Toyota M, Sanchez-Cespedes M, Capella G, Peinado MA, Gonzalez S, Tarafa G, Sidransky D, Meltzer SJ, et al: Analysis of adenomatous polyposis coli promoter hypermethylation in human cancer. Cancer Res. 60:4366–4371. 2000.PubMed/NCBI

17 

Rowan AJ, Lamlum H, Ilyas M, Wheeler J, Straub J, Papadopoulou A, Bicknell D, Bodmer WF and Tomlinson IP: APC mutations in sporadic colorectal tumors: A mutational ‘hotspot’ and interdependence of the ‘two hits’. Proc Natl Acad Sci USA. 97:3352–3357. 2000. View Article : Google Scholar : PubMed/NCBI

18 

Minde DP, Anvarian Z, Rüdiger SG and Maurice MM: Messing up disorder: How do missense mutations in the tumor suppressor protein APC lead to cancer? Mol Cancer. 10:1012011. View Article : Google Scholar : PubMed/NCBI

19 

Hiltunen M, Alhonen L, Koistinaho J, Myöhänen S, Pääkkönen M, Marin S, Kosma VM and Jänne J: Hypermethylation of the APC (adenomatous polyposis coli) gene promoter region in human colorectal carcinoma. Int J Cancer. 70:644–648. 1997. View Article : Google Scholar : PubMed/NCBI

20 

Virmani A, Rathi A, Sathyanarayana U, Padar A, Huang C, Cunnigham H, Farinas A, Milchgrub S, Euhus D, Gilcrease M, et al: Aberrant methylation of the adenomatous polyposis coli (APC) gene promoter 1A in breast and lung carcinomas. Clin Cancer Res. 7:1998–2004. 2001.PubMed/NCBI

21 

Aguiler O, Muñoz A, Esteller M and Fraga M: Epigenetic alterations of the Wnt/beta-catenin pathway in human disease. Endocr Metab Immune Disord Drug Targets. 7:13–21. 2007. View Article : Google Scholar : PubMed/NCBI

22 

Kikuchi A: Tumor formation by genetic mutations in the components of the Wnt signaling pathway. Cancer Sci. 94:225–229. 2003. View Article : Google Scholar : PubMed/NCBI

23 

Morin PJ, Sparks AB, Korinek V, Barker N, Clevers H, Vogelstein B and Kinzler KW: Activation of beta-catenin-Tcf signaling in colon cancer by mutations in beta-catenin or APC. Science. 275:1787–1790. 1997. View Article : Google Scholar : PubMed/NCBI

24 

Cieply B, Zeng G, Proverbs-Singh T, Geller DA and Monga SP: Unique phenotype of hepatocellular cancers with exon-3 mutations in beta-catenin gene. Hepatology. 49:821–31. 2009. View Article : Google Scholar : PubMed/NCBI

25 

Roberts DM, Pronobis MI, Poulton JS, Waldmann JD, Stephenson EM, Hanna S and Peifer M: Deconstructing the ßcatenin destruction complex: Mechanistic roles for the tumor suppressor APC in regulating Wnt signaling. Mol Biol Cell. 22:1845–1863. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Bai A, Tong J, To K, Chan M, Man E, Lo K, Lee J, Sung J and Leung W: Promoter hypermethylation of tumor-related genes in the progression of colorectal neoplasia. Int J Cancer. 112:846–853. 2004. View Article : Google Scholar : PubMed/NCBI

27 

Fu X, Li J, Li K, Tian X and Zhang Y: Hypermethylation of APC promoter 1A is associated with moderate activation of Wnt signalling pathway in a subset of colorectal serrated adenomas. Histopathology. 55:554–563. 2009. View Article : Google Scholar : PubMed/NCBI

28 

Kang S, Kim J, Kang G, Lee S, Park N, Song Y, Park S, Kang S and Lee H: Comparison of DNA hypermethylation patterns in different types of uterine cancer: Cervical squamous cell carcinoma, cervical adenocarcinoma and endometrial adenocarcinoma. Int J Cancer. 118:2168–2171. 2006. View Article : Google Scholar : PubMed/NCBI

29 

Yang N, Nijhuis E, Volders H, Eijsink J, Lendvai A, Zhang B, Hollema H, Schuuring E, Wisman G and van der Zeea A: Gene promoter methylation patterns throughout the process of cervical carcinogenesis. Cell Oncol. 32:131–143. 2010.PubMed/NCBI

30 

Zambrano P, Segura-Pacheco B, Perez-Cardenas E, Cetina L, Revilla-Vazquez A, Taja-Chayeb L, Chavez-Blanco A, Angeles E, Cabrera G, Sandoval K, et al: A phase I study of hydralazine to demethylate and reactivate the expression of tumor suppressor genes. BMC Cancer. 5:442005. View Article : Google Scholar : PubMed/NCBI

31 

Dong S, Kim H, Rha S and Sidransky D: Promoter hypermethylation of multiple genes in carcinoma of the uterine cervix. Clin Cancer Res. 7:982–1986. 2001.

32 

Wisman GB, Nijhuis ER, Hoque MQ, Reesink-Peters N, Koning AJ, Volders HH, Buikema HJ, Boezen HM, Hollema H, Schuuring E, et al: Assessment of gene promoter hypermethylation for detection of cervical neoplasia. Int J Cancer. 119:1908–1914. 2006. View Article : Google Scholar : PubMed/NCBI

33 

van der Meide WF, Snellenberg S, Meijer CJ, Baalbergen A, Helmerhorst TJ, van der Sluis WB, Snijders PJ and Steenbergen RD: Promoter methylation analysis of WNT/β-catenin signaling pathway regulators to detect adenocarcinoma or its precursor lesion of the cervix. Gynecol Oncol. 123:116–122. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Ma C, Zeng C, Jin L, Yang Y, Li P, Chen L and Wang J: GSK3β mediates the carcinogenic effect of HPV16 in cervical cancer. Sci Rep. 5:165552015. View Article : Google Scholar : PubMed/NCBI

35 

Cameron EE, Bachman KE, Myöhänen S, Herman JG and Baylin SB: Synergy of demethylation and histone deacetylase inhibition in the re-expression of genes silenced in cancer. Nat Genet. 21:103–107. 1999. View Article : Google Scholar : PubMed/NCBI

36 

Yang X, Phillips DL, Ferguson AT, Nelson WG, Herman JG and Davidson NE: Synergistic activation of functional estrogen receptor (ER)-alpha by DNA methyltransferase and histone deacetylase inhibition in human ER-alpha-negative breast cancer cells. Cancer Res. 61:7025–7029. 2001.PubMed/NCBI

37 

Worm J, Christensen C, Grønbaek K, Tulchinsky E and Guldberg P: Genetic and epigenetic alterations of the APC gene in malignant melanoma. Oncogene. 23:5215–5226. 2004. View Article : Google Scholar : PubMed/NCBI

38 

Chung MT, Lai HC, Sytwu HK, Yan MD, Shih YL, Chang CC, Yu MH, Liu HS, Chu DW and Lin YW: SFRP1 and SFRP2 suppress the transformation and invasion abilities of cervical cancer cells through Wnt signal pathway. Gynecol Oncol. 112:646–653. 2009. View Article : Google Scholar : PubMed/NCBI

39 

Korinek V, Barker N, Morin PJ, van Wichen D, de Weger R, Kinzler KW, Vogelstein B and Clevers H: Constitutive transcriptional activation by a beta-catenin-Tcf complex in APC−/−colon carcinoma. Science. 275:1784–1787. 1997. View Article : Google Scholar : PubMed/NCBI

40 

Svedlund J, Aurén M, Sundström M, Dralle H, Akerström G, Björklund P and Westin G: Aberrant WNT/β-catenin signaling in parathyroid carcinoma. Mol Cancer. 9:2942010. View Article : Google Scholar : PubMed/NCBI

41 

Huang M, Wang Y, Sun D, Zhu H, Yin Y, Zhang W, Yang S, Quan L, Bai J, Wang S, et al: Identification of genes regulated by Wnt/beta-catenin pathway and involved in apoptosis via microarray analysis. BMC Cancer. 6:2212006. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2018
Volume 17 Issue 1

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ayala‑Calvillo E, Mojica‑Vázquez LH, García‑Carrancá A and González‑Maya L: Wnt/β‑catenin pathway activation and silencing of the APC gene in HPV‑positive human cervical cancer‑derived cells. Mol Med Rep 17: 200-208, 2018
APA
Ayala‑Calvillo, E., Mojica‑Vázquez, L.H., García‑Carrancá, A., & González‑Maya, L. (2018). Wnt/β‑catenin pathway activation and silencing of the APC gene in HPV‑positive human cervical cancer‑derived cells. Molecular Medicine Reports, 17, 200-208. https://doi.org/10.3892/mmr.2017.7853
MLA
Ayala‑Calvillo, E., Mojica‑Vázquez, L. H., García‑Carrancá, A., González‑Maya, L."Wnt/β‑catenin pathway activation and silencing of the APC gene in HPV‑positive human cervical cancer‑derived cells". Molecular Medicine Reports 17.1 (2018): 200-208.
Chicago
Ayala‑Calvillo, E., Mojica‑Vázquez, L. H., García‑Carrancá, A., González‑Maya, L."Wnt/β‑catenin pathway activation and silencing of the APC gene in HPV‑positive human cervical cancer‑derived cells". Molecular Medicine Reports 17, no. 1 (2018): 200-208. https://doi.org/10.3892/mmr.2017.7853