Targeting the ∆133p53 isoform can restore chemosensitivity in 5-fluorouracil-resistant cholangiocarcinoma cells

  • Authors:
    • Nichapavee Nutthasirikul
    • Chariya Hahnvajanawong
    • Anchalee Techasen
    • Temduang Limpaiboon
    • Chanvit Leelayuwat
    • Siri Chau-In
    • Patcharee Jearanaikoon
  • View Affiliations

  • Published online on: October 5, 2015     https://doi.org/10.3892/ijo.2015.3188
  • Pages: 2153-2164
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Lack of the normal p53 transactivation domain, ∆133p53 isoform exhibits anti-p53 function. Many studies report the correlation between ∆133p53 expression and poor survival in various cancers, including cholangiocarcinoma (CCA), which is a cancer of the bile ducts. CCA almost always results in short survival times. The relevance of ∆133p53 to drug resistance in CCA is not yet well understood. This study aimed to demonstrate the association between ∆133p53 and 5-fluorouracil (5-FU) resistance in CCA. ∆133p53 protein was highly expressed in CCA patients with poor outcome compared to favorable outcome but was not statistically significant. However, a significant correlation was found between normalized ∆133p53 levels and 5-FU resistance which was defined by an ex vivo histoculture drug response assay (P=0.019). Two stable 5-FU-resistant CCA cell lines, KKU-M139R (IC50 38.8 µM) and KKU-M214R (IC50 39.5 µM), were used as a model to evaluate the role of ∆133p53. Increased ∆133p53 was correlated with 5-FU in a dose-dependent manner. The transient knockdown of ∆133p53 expression can restore drug sensitivity in both resistant CCA cells with 11- to 45-fold reduction of IC50 compared to control. Upon ∆133p53 silencing, apoptotic signaling was enhanced by the upregulation of Bax and downregulation of Bcl-2. Additionally, p21 and p27 were upregulated, resulting in cell cycle arrest at G2. Inhibition of colony formation and prolong doubling time were also observed. Our findings demonstrated that chemosensitivity can be modulated via targeting of ∆133p53 suggesting the potential use of ∆133p53 as a candidate for targeting therapy in CCA.

Introduction

Cholangiocarcinoma (CCA) is a cancer arising from bile duct epithelium. The highest prevalence of CCA in the world has been reported in the northeastern regions of Thailand, where liver fluke infection is highly endemic (13). The etiology of the disease is related to chronic biliary inflammation caused by the consumption of raw or undercooked fish that is infected with the liver fluke Opisthorchis viverrini, together with foods containing N-nitroso compounds (4,5). Only CCA patients with early stage disease are curable by surgical treatment (6). However, the early stage disease is asymptomatic and difficult to diagnose. Treatment for advance stage CCA is rather unsuccessful. Most CCA patients have different chemotherapeutic responses even at the same stage, leading to poor clinical outcomes with short survival times of ~5–7 months (7). 5-Fluorouracil (5-FU) is recommended in low resource countries as the first line drug of choice for the treatment of a variety of solid tumors including CCA (8). However, 5-FU is rather ineffective treatment for CCA, with a response rate <10% (9). Several reports have noted the effectiveness of other anticancer drugs in CCA such as cisplatin (10,11), doxorubicin (10) and gemcitabine (12). Unfortunately, the survival time was not significantly improved which was ~6–9 months (9). Therefore, the use of drug combinations rather than single drug has been proposed for obtaining better response in CCA. A response rate of ≤40% was obtained with the triple combination of cisplatin, epirubicin and 5-FU (13). However, using more anticancer drugs may produce additional adverse effects and acquired drug resistance in patients.

5-FU is an inhibitor of thymidylate synthase leading to inhibition of RNA and DNA synthesis of tumor cells. DNA damage can trigger the induction of p53-dependent cell cycle arrest and apoptosis, resulting in tumor cell death (14,15). Although downregulation of p53 mRNA, a p73 family member, has been reported in 5-FU-resistant CCA cell lines by Namwat et al (16) no information regarding p53 role in 5-FU-resistant CCA cells has been reported. The p53 gene contains two alternative promoters (P1 and P2). The P1 promoter generates the full-length TAp53 and Δ40p53, a 40 amino acids deleted p53 variant via alternative splicing and initiation of translation within intron 2 (17). The assembly of full-length p53 molecules as a tetramer leads to normal function as a transcription factor. The P2 promoter encodes larger amino-terminal truncated proteins (133 and 160 deleted amino acids), Δ133p53 (17) and Δ160p53 (18), which exhibit anti-apoptotic properties when oligomerized with TAp53 resulting in loss of the transactivation function. The N-terminal deleted p53 protein variant (ΔNp53) has been reported to disrupt wild-type p53 function (19). Hence, the control of promoter usage in p53 is proposed as an auto-regulation mechanism for p53 functions (20). In addition to the ΔNp53 isoforms, three alternate isoforms of p53; α, β and γ at the carboxyl terminal are encoded by alternative splicing. A full-length p53 or TAp53α is encoded from the normal splice site, whereas TAp53β and TAp53γ are encoded from two different alternative splicing sites of intron 9 at carboxyl end (17). To date, any impact of β and γ isoforms on tumor suppressor activities remains unclear. An increase in Δ133p53 expression has also been reported in renal cell (21), acute myeloid leukemia (22), ovarian cancer (23), breast (17), head and neck (24), melanoma (25), colon cancer (19). The correlation between Δ133p53 and tumor progression has been found in colon carcinomas (19). Our previous study also found the relationship between overexpression of defective p53 (mutant p53 and Δ133p53) with poor prognosis in CCA (26). Upregulation of Δ133p53 mRNA level and ratio disruption of the p53 isoforms encoded from P2/P1 (Δ133p53/TAp53) was correlated with poor survival outcome of CCA patients. The major factors affecting the patients survival outcome may be the contribution from drug resistance. Reports of Δ133p53 effects on drug resistance are few. Recently, upregulation of Δ133p53α in response to a low dose of doxorubicin has been noted in osteosarcoma and colon cancer cell lines (27). However, the role of Δ133p53 isoforms in drug resistance in CCA remains unclear.

This study attempts to demonstrate the association between Δ133p53 overexpression and chemoresistance in CCA. 5-FU sensitivity in clinical tissues of CCA was classified using an ex vivo histoculture drug response assay (HDRA) and clinical treatment outcome. Two 5-FU-resistant CCA cell lines were established in this study and used as a model to evaluate the role of Δ133p53 isoform in chemosensitivity.

Materials and methods

Clinical samples

A total of 22 tumor samples and 10 normal adjacent tissues were collected from intrahepatic cholangiocarcinoma (ICC) patients who were admitted to Srinagarind Hospital, Faculty of Medicine, Khon Kaen University, Thailand. The project was approved by the Khon Kaen University Ethics Committee in human research (HE571044). All patients gave written informed consent. Fresh tumor tissues were tested for Histoculture Drug Response Assay. Paralleled tissues were kept under liquid nitrogen until used for protein extraction.

Histoculture drug response assay (HDRA)

Tumor tissues were classified as 5-FU sensitive and 5-FU resistant based on results obtained from an ex vivo histoculture drug response assay (HDRA), using the median of inhibition index (% II) as previously described (28). In brief, fresh tumor tissues were washed and a 3-mm diameter punch was used aseptically to take samples that were placed on collagen gel sponges. Each was cultured at 37ºC in a 6-well plate containing RPMI medium supplemented with 2.5% v/v penicillin-streptomycin-fungizone (PSF; Invitrogen, Carlsbad, CA, USA) and 5-FU at 200 μM. For a control sample, no 5-FU was added in the culture medium. After 4 days of culture, the viability of tumor cells in the cultured tissues was examined using TUNEL staining. TUNEL-positive cells were identified as dead cells. The efficacy of 5-FU was calculated and expressed as the % II using the following formula: % II = (1 − % viable tumor cells in 5-FU-treated tumor tissue/% living tumor cells in control tissue) × 100.

CCA cell lines and cell culture

Two CCA cell lines, KKU-M139 and KKU-M214 were established from primary tumors of human intrahepatic CCA at the Liver Fluke and Cholangiocarcinoma Research Center, Khon Kaen University Thailand (16,29,30). Both were cultured at 37ºC in RPMI medium (Gibco BRL, Grand Island, NY, USA) supplemented with 10% fetal bovine serum (FBS), 1% v/v penicillin-streptomycin solution (Gibco, BRL) under 5% CO2 atmosphere. These CCA cells were named as KKU-M139P and KKU-M214P, to identify the parental cell lines at the beginning of drug resistance induction.

Establishment of 5-FU-resistant CCA cell lines

5-FU-resistant cell lines were generated from the parental cell lines KKU-M139P and KKU-M214P by stepwise increases of the concentration of 5-FU (Boryung Pharm, Korea) as described previously (16). In brief, 1×105 cells were seeded in 25-cm2 flasks and cultured without 5-FU for 24 h. Subsequently, cells were exposed with 5-FU at 6 μM for KKU-M139 (1X IC50 value of KKU-M139P) and 4 μM for KKU-M214 (1X IC50 value of KKU-M214P) for 72 h. Cells were then cultured in a drug-free medium until they reached 70% confluence. These cells were continuously maintained in 1X IC50 for several passages until these cells were stable before being subjected into 2X IC50. The 5-FU-resistant clones were finally obtained after continuous selection by several passages for 18 months. The IC50 of the resistant clones was checked by the SRB assay (31). The resistant clones were then passaged into a drug-free medium for 2 weeks before being stored as a stock of the resistant cell lines (KKU-M139R and KKU-M214R) at −80ºC. KKU-M139R and KKU-M214R were cultured in 5-FU-free medium for ≥2 weeks to eliminate potential long-term effects of 5-FU unrelated to drug resistance prior to being performed in all experiments.

Transient silencing of Δ133p53 by siRNA

Expression of Δ133p53 in KKU-M139R and KKU-M214R cells was suppressed using a siRNA technique. The sequences of two specific siRNA targeting human Δ133p53 (Δ133p53a and Δ133p53b) as described previously (18), were purchased from Ambion (Austin, TX, USA). The cells (2×106 cells/well) were seeded in a 6-well plate and cultured overnight before being transfected separately with 100 pM of siΔ133p53a and siΔ133p53b, while siGFP (Green fluorescence protein, Applied Biosystems/Ambion, Carlsbad, CA, USA) was used as a siRNA control. Transfection was carried out using Lipofectamine RNAiMAX (Invitrogen) according to the manufacturer's instructions. After 24 h of transfection, culture medium was added and the plates were incubated at 37ºC for a further 48 h. At 72 h-post transfection, total proteins were extracted using TRIzol reagent (Invitrogen). The level of Δ133p53 protein was determined by western blot analysis using β-actin as a loading control.

Measurement of IC50 by Sulforhodamine B (SRB) assays

The parental CCA cells (KKU-M139P and KKU-M214P) and the 5-FU-resistant cells (KKU-M139R and KKU-M214R) were seeded at 1×104 cells/well in triplicate into a 96-well culture plate and incubated at 37ºC for 24 h. All cell lines were then treated with various concentrations of 5-FU ranging from 2–128 μM in triplicate for 72 h, while 0.9% saline was used as a negative control. The cytotoxicity was performed using a sulforhodamine B (SRB) assay as previously described (31). The cells were fixed using a 10% cold trichloroacetic acid (TCA), washed and air-dried at room temperature. SRB (Sigma-Aldrich, MO, USA) solution (100 μl/well) was added and followed by three quick rinses with 1% acetic acid to remove unbound dye. SRB was solubilized in a 10 mM Tris base solution and the absorbance at 490 nm was measured using a microplate reader (Tecan Ltd., Reading, UK). Percentage of cell viability was calculated [(mean ODsample − mean ODday0/mean ODnegative control − mean ODday0) × 100] and used to generate the curve by which IC50 was calculated. Resistance index was defined as a ratio of the IC50 value of drug resistant cells to parental cells.

Population doubling time (PDT)

To assess cell growth, the population doubling time (PDT) of the cells was assessed in triplicate. Cells (2×105) were cultured in a drug free medium supplemented with 10% FBS at 37ºC in a humidified 5% CO2 atmosphere. When reaching 70% confluence, the cells were trypsinized, stained with trypan blue and counted on a hemocytometer. PDT was calculated using the following formula as described previously (32): (T-T0) log2/logN-logN0, where N0 is the initial cell number, N is the final cell number, T is the time interval between N0 and N, and T0 is the initial time.

Colony forming assay

KKU-M139R and KKU-M214R cells were pre-treated with either siΔ133p53 or siRNA control. The cells were seeded at 200 cells/well in a 6-well plate containing 2 ml RPMI culture medium supplemented with 10% v/v FBS and cultured at 37ºC in a humidified 5% CO2 atmosphere for 5 days. The cells were washed twice with PBS, stained with H&E and the colonies were counted.

Analysis of apoptosis by Annexin V/PI staining

The analysis of Annexin V binding was carried out with the Annexin V-FITC Detection Kit I (eBioscience, San Diego, CA, USA) according to the manufacturer's instructions. Briefly, cells were incubated with or without siΔ133p53 or a control scramble RNA for 48 h. Cells were collected, washed twice with cold PBS, centrifuged at 1,800 rpm for 3 min, and resuspended in 1X binding buffer at a concentration of 106 cells/ml. Then 100 μl of the solution (105 cells) was transferred to a 5-ml culture tube; 5 μl of Annexin V-FITC and 5 μl of PI were added. Cells were incubated for 15 min at room temperature in the dark. Furthermore, 200 μl of 1X binding buffer were added to each tube, and samples were analyzed by FACScan flow cytometry (BD FACSCanto II; BD, USA). For each sample, 10,000 ungated events were acquired. Annexin V+/PI cells represented the early apoptotic populations, and Annexin V+/PI+ cells the late apoptotic populations.

Cell cycle analysis by flow cytometry

KKU-M139R and KKU-M214R cells (105 cells/ml) were incubated with or without siΔ133p53 or a siRNA control for 48 h. The cells were collected, washed with cold PBS, fixed in cold 100% ethanol, treated with DNase-free RNase, and stained with 40 μg/ml of propidium iodide (PI). The distribution of the cells between phases of the cell cycle was deduced from the DNA content on a FACScan flow cytometer (BD FACSCanto II; BD, USA). For each sample, 10,000 gated events were acquired.

Western blot analyses

Protein was extracted from CCA tissues and cell lines using TRIzol (Invitrogen) and 40 μg aliquots were fractionated on 15% polyacrylamide gel electrophoresis. Primary antibodies; CM-1 (1:100, Signet, Emeryville, CA, USA), p21 (1:400), p27 (1:400), Bcl-2 (1:200), Bax (1:200), and p73 (ab-4) (Santa Cruz Biotechnology, Santa Cruz, CA, USA) as well as a loading control, β-actin (1:4,000, Sigma Chemical Co.) were used followed by the secondary antibody peroxidase-labeled anti-rabbit (1:10,000, Abcam, UK) The proteins were detected by chemiluminescence using the ECL Plus system (GE Healthcare, UK). Protein band intensity was calculated by Scion image program, and normalized to β-actin.

Immunocytochemical staining of p53

The paraffin-embedded sections (5 μm) of CCA cell pellets fixed with 10% formalin solution were deparaffinized. Antigen retrieval was performed in boiling 0.01 M citrate buffer (pH 6.0) as described previously (26). Endogenous peroxidase was inactivated with 100 μl of 3% H2O2. Non-specific binding was further treated with a blocking buffer containing phosphate-buffered saline with Tween-20 (PBS-T), 30% casein and 5% FBS. For p53 protein detection, mutant p53 was detected with the primary antibody clone DO-7, 1:100 (Dako, Glostrup, Denmark), which recognizes an epitope between amino acids 1–45 of human p53. The slides were incubated overnight at room temperature with primary antibody. Proteins were detected using the EnVision system (Dako) for 1 h at room temperature. Color was developed with DAB solution (Dako) and nuclei were counterstained with hematoxylin. Positive staining was observed as brown color in blue/gray nuclei. Positive with DO-7 antibody indicates overexpression of the mutated p53 due to its stability (26,3335).

Statistical analyses

Statistical analyses were performed using SPSS for Windows version 15 (SPSS, Inc., IL, USA). The Mann-Whitney U test was used for comparison of two groups. Data are expressed as mean ± SD from three independent experiments. Statistically significant differences are indicated in the figures as *P<0.05, **P<0.01, ***P<0.001.

Results

Assessment of 5-FU sensitivity in CCA samples

5-FU sensitivity in 22 CCA patients was classified based on the results obtained from histoculture drug response assays (HDRA) and treatment outcome data. For the HDRA-based classification, we used the median of the % inhibition index (% II) which was 36.5% and this identified 11 patients as 5-FU-sensitive and 11 as 5-FU-resistant. Only 12 of 22 CCA patients underwent complete course of chemotherapy. The treatment outcome was further followed up every 6 months for ≥12 months. Poor response (n=7) and favorable response (n=5) were defined in terms of having tumor progression before and after 6 months after treatment, respectively. All clinical data are summarized in Table I.

Table I

Clinicopathological data of 22 CCA patients.

Table I

Clinicopathological data of 22 CCA patients.

Chemosensitivityb

No.SexAgeSurvival timeaStageChemotherapyHDRAClinical outcome
1M53LongIVBTreatedSensitivePoor response
2M64LongIITreatedSensitiveFavorable response
3M52LongIIITreatedSensitiveFavorable response
4M61ShortIITreatedSensitivePoor response
5F51ShortIVAUntreatedSensitiveNA
6F65ShortIIITreatedSensitivePoor response
7M58ShortIIIAUntreatedSensitiveNA
8M70ShortIVAUntreatedSensitiveNA
9M69LongIVATreatedSensitivePoor response
10F51ShortIITreatedSensitiveFavorable response
11F64LongIIITreatedSensitiveFavorable response
12M57LongIVATreatedResistantPoor response
13M53ShortIIIAUntreatedResistantNA
14F58LongIVATreatedResistantFavorable response
15F51LongIIIATreatedResistantPoor response
16F64ShortIVAUntreatedResistantNA
17M63ShortIIIUntreatedResistantNA
18M59LongIIIUntreatedResistantNA
19F50ShortIVAUntreatedResistantNA
20M69ShortIVBUntreatedResistantNA
21M62LongIIIATreatedResistantPoor response
22M69LongIVAUntreatedResistantNA

a Long and short survival was classified by the median cut off at 52.43 weeks.

b Chemosensitivity was categorized according to HDRA and clinical outcome.

{ label (or @symbol) needed for fn[@id='tfn3-ijo-47-06-2153'] } NA, not applicable is defined as an incomplete drug treatment. HDRA, histoculture drug response assay.

Levels of the ΔNp53 isoform are increased significantly in 5-FU-resistant CCA samples

Western blot analysis of 22 tumors and 10 normal paired tissues using CM-1 antibody revealed the presence of various p53 isoforms (Fig. 1). At least 3 isoforms; TAp53, Δ40p53 and Δ133p53 were observed in tumor tissues, in which two different sizes of Δ133p53 at 35 kDa corresponding to Δ133p53α and 28 kDa to Δ133p53β, γ were defined (Fig. 1A). Interestingly, no Δ133p53 isoform was observed in the normal tissues (Fig. 1A). The box-plot analysis of Δ133p53 protein expression normalized to β-actin and 5-FU sensitivity classified by HDRA showed significantly increased Δ133p53 in 5-FU-resistant cases compared to sensitive ones (P=0.019) (Fig. 1B). It seemed that Δ133p53 protein was highly expressed in CCA patients with poor outcome compared to CCA cases with favorable outcome but was not statistically significant (P=0.264) (Fig. 1C).

Characteristics of 5-FU-resistant CCA cell lines

Two resistant CCA cell lines; KKU-M139R and KKU-M214R were successfully induced from the parental cells; KKU-M139P and KKU-M214P. Drug toxicity is presented in Fig. 2. The IC50 values of 5-FU in KKU-M139P, KKU-M139R, KKU-M214P and KKU-M214R cells were 6.2±2.09, 38.8 ±7.11, 3.9±2.04 and 39.5±3.45 μM, respectively. The resistance index calculated from the ratio of IC50 of resistant to parental CCA cell lines was 6.26 and 10.12 for KKU-M139 and KKU-M214, respectively. Furthermore, population doubling times of both resistant cell lines were shorter than those of parental cell lines as summarized in Table II.

Table II

Population doubling time (PDT) and IC50 of 5-FU at 72-h culture of KKU-M139 and KKU-M214 cell lines.

Table II

Population doubling time (PDT) and IC50 of 5-FU at 72-h culture of KKU-M139 and KKU-M214 cell lines.

Cell linesPopulation doubling time (PDT) (h) (mean ± SD)IC50 of 5-FU (μM) (mean ± SD)Resistant index (IC50 of resistant/parental cells)
KKU-M139P30.13±0.866.2±2.096.26
KKU-M139R17.81±0.7438.8±7.11
KKU-M214P40.16±1.123.9±2.0410.12
KKU-M214R22.92±1.0339.5±3.45
Upregulation of the Δ133p53 isoform induced by 5-FU

The expression of p53 isoforms was assessed in both parental and resistant CCA cell lines in response to 5-FU concentration covering their IC50 values. When challenged with 5-FU with 5 and 10 μM, TAp53, Δ40p53 and Δ133p53 protein isoforms were markedly increased in both parental cell lines (KKU-M139P and KKU-M214P) in a dose-dependent manner (Fig. 3). This finding indicates the increased usage of both P1 and P2 promoters under 5-FU stress. In contrast to parental cells, only Δ133p53 protein was upregulated in both resistant cell lines (KKU-M139R and KKU-M214R) induced by 5-FU (Fig. 3) suggesting the enhancement of P2 promoter usage. Upon 5-FU challenge with 20 and 50 μM, KKU-M214R showed rapid response to 5-FU in which the upregulation of Δ133p53 was detected at a lower dose (20 μM of 5-FU) compared to KKU-M139R (50 μM of 5-FU).

Silencing of Δ133p53 promotes apoptosis and cell cycle arrest in 5-FU resistant CCA cells

Silencing of Δ133p53 in both KKU-M214R and KKU-M139R was used to investigate the role of Δ133p53 in 5-FU-resistant CCA cells. The expression of Δ133p53 was successfully suppressed with both siΔ133p53a and siΔ133p53b compared to the siRNA control (Fig. 4A and B) with normal apparent morphology (Fig. 4C). The effect of silenced Δ133p53 on apoptotic and cell cycle markers in KKU-M139R and KKU-M214R was investigated. The suppression of Δ133p53 protein resulted in significant upregulation of Bax expression in both KKU-M214R and KKU-M139R (P<0.01) as well as downregulation of Bcl-2 in KKU-M214R (P<0.001) and KKU-M139R (P<0.01) (Fig. 4D). Accordingly, Annexin V/PI staining showed significantly increased cell apoptosis in the silenced siΔ133p53 of both KKU-M139R and KKU-M214R (siΔ133p53a at P<0.01 and siΔ133p53b at P<0.001, respectively) compared with control (Fig. 5).

Additionally p21 and p27 proteins were significantly upregulated with P<0.01 and P<0.001 in M139R and KKU-M214R compared to the siRNA control (Fig. 4A and 4D). Moreover, cells were significantly arrested at G2 in both siΔ133p53a and siΔ133p53b treated KKU-M139R and KKU-M214R compared to the siRNA controls (Fig. 6). Interestingly, significant upregulation of p73 was observed in both types of silenced CCA cells (Fig. 4A and D). Moreover, no mutated p53 was revealed in siΔ133p53 treated (KKU-M139R and KKU-M214R) or siRNA control compared to their parental cells (KKU-M139P and KKU-M214P) using immunostaining with DO-7 (Fig. 7). These results imply that the induced p73 protein might help mediating apoptosis upon the absence of wild-type TAp53 (Fig. 3A) and mutated p53 (Fig. 7). Moreover, the induced cell cycle arrest by Δ133p53 silencing leads to growth retardation, as shown by the prolonged PDT (Fig. 8A) and the inhibition of colony forming capability (Fig. 8B). Hence, suppression of Δ133p53 expression affects certain tumor characteristics of these CCA resistant cells.

Attenuation of Δ133p53 levels enhances the chemosensitivity of 5-FU-resistant CCA cells

The chemosensitivity of KKU-M139R and KKU-M214R cells transfected with siΔ133p53a and siΔ133p53b was re-assessed using an SRB assay of cell numbers (Table III). Strikingly, the IC50 of KKU-M139R-siΔ133p53a and KKU-M139R-Δ133p53b cells to 5-FU was decreased 12- and 45-fold, as compared with the IC50 of the siRNA control cells. Similar effects were also observed in KKU-M214R with 11- and 20-fold decreases, respectively. The IC50 of silenced CCA cells was lower than those of parental cells suggesting that silencing of Δ133p53 can re-sensitize 5-FU resistance in CCA cell lines.

Table III

The IC50 of 5-FU at 72-h culture of KKU-M139R and KKU-M214R cell lines after siRNA treatment.

Table III

The IC50 of 5-FU at 72-h culture of KKU-M139R and KKU-M214R cell lines after siRNA treatment.

Cell lines5-FU IC50 at 72 h (μM) (mean ± SD)Fold reduction of IC50 a
KKU-M139R
 Mock38.83±7.11-
 Control31.47±4.98-
 siΔ133p53a2.7±1.1811.66
 siΔ133p53b0.7±0.9444.96
KKU-M214R
 Mock39.5±3.45-
 Control35.24±3.91-
 siΔ133p53a3.2±2.8411.01
 siΔ133p53b1.8±1.4819.58

a Fold reduction, IC50 of silenced cells/control.

Discussion

A variety of ΔNp53 isoforms generated from both P1 and P2 promoters were reported in clinical tumors, however, limited evidence of chemoresistance has been noted. This study is the first to demonstrate the correlation between high levels of Δ133p53 expression in clinical CCA tissues with 5-FU resistant based HDRA. A limited sample sizing, resulting from an incomplete treatment, affected the statistical testing for correlation in clinical treatment. Even though Δ133p53 expression showed no significant correlation with clinical treatment outcome, Δ133p53 level seemed to increase in patients with poor response to treatment. This result suggested that Δ133p53 might be involved on drug responsiveness. Aoubala et al reported the upregulation of Δ133p53α with response to a low dose of doxorubicin in osteosarcoma and colon cancer (27). However, p53 function can be inactivated by either mutation or Δ133p53 overexpression, thus, the mutant p53 should also be considered in clinical CCA. The incidence of p53 mutation has been reported in clinical CCA samples as up to 41–44% (26,36). Defective p53 due to mutation has been reported with drug resistance to 5-FU-based therapy in colorectal cancer (3739). A correlation between p53 mutation and platinum-based chemotherapy resistance, early relapse, and shortened overall survival was reported in ovarian cancer patients (40). Of note, the incidence of ΔNp53 overexpression without mutant p53 in CCA has been previously found at 54% (26). Therefore, the 5-FU-resistant CCA cell lines KKU-M139R and KKU-M214R were used as an in vitro model to address the impact of Δ133p53 on 5-FU resistance without the influence of p53 mutation. These 5-FU-resistant CCA cell lines contain only Δ133p53 protein without the full length p53 (TAp53) or the mutated p53 as shown by negative western blotting with CM-1 and negative DO-7 staining. The IC50 of 5-FU resistance remained stable even cultured in drug free medium for ≥4 weeks, supporting the claim for stable resistant clone.

For both parental CCA cell lines, 5-FU can enhance the upregulation of both TAp53 and Δ133p53 in a dose-dependent manner. The enhancement of both P1 and P2 promoter usage might provide an advantage of apoptosis evasion via p53 inactivation which enabling an acquired 5-FU resistance upon drug exposure. Similar finding of Δ133p53 upregulation in response to 5-FU was revealed in both resistant cells except TAp53 existence. The enhancement of P2 promoter regardless of TAp53 may result from continuous selective pressure upon induction of 5-FU resistance. The rapid response to lower dose of 5-FU found in KKU-M214R, may explain the higher resistance index of KKU-M214R (10.12-fold) than that of KKU-M139R (6.26-fold). Targeting of Δ133p53 in 5-FU resistant cells by siRNA is therefore verified the role of Δ133p53 on chemoresistance in these 5-FU-resistant CCA cell lines which was successfully obtained by both siΔ133p53a and siΔ133p53b with 75–90% suppression. Interestingly, the targeting of Δ133p53 helps restoring the 5-FU sensitivity with markedly reduced IC50 compared to that of parental cells. The molecular underlying mechanism of 5-FU resensitization can be explained by an increase of cell apoptosis via an upregulation of pro-apoptotic BAX and downregulation of anti-apoptotic Bcl-2. Moreover, G2 arrest was induced by upregulation of p21 and p27 in comparison with siRNA control cells. This evidence is relevant to the antitumor activity of 5-FU which is known to be involved in the induction of p53-dependent cell cycle arrest and apoptosis (37,39,41).

Regardless of TAp53, the suppressed Δ133p53 can explain only the withdrawal of p53 inactivation, but is unable to provide clues for p53 activation. The increase of p73 expression observed by western blotting in both types of silenced Δ133p53 CCA cells might be responsible for p53 function restoration. p73, as a p53 family member, has been shown to possess the capability to restore p53 function via p21 activation in a neuroblastoma cell line (42). The p73 protein can activate upstream transcriptional regulation of p21 and p27, resulting in cell cycle arrest in G2 (4345). In human lung adenocarcinoma, p73 overexpression can enhance chemosensitivity by apoptosis induction (46,47). Namwat et al (16), reported the association between downregulation of TAp73 mRNA and 5-FU-resistant CCA cell lines. Evasion of apoptosis and cell cycle arrest are evident as a common mechanism of 5-FU resistance in various cancers such as colorectal cancer (48), breast cancer (49), and CCA (16,29). Recently, evasion of both intrinsic and extrinsic apoptotic pathway has been reported in gemcitabine-resistant CCA cell lines (32). Thus, Δ133p53 may exert a signature of chemoresistant cells to evade p53-dependent cell apoptosis and cell cycle arrest.

Collectively, the silencing of Δ133p53 and increased p73 expression may modulate the chemosensitivity of 5-FU resistance in CCA cell lines. Low incidence of p73 mutation has been reported in CCA (50), downregulation via p73 methylation has been frequently found in various cancers (5154) including CCA (55). Data on the alteration of p73 for chemo-resistance in CCA is still limited. The status of p73 should be investigated in further study.

In conclusion, this study is the first to demonstrate the important role of Δ133p53 in 5-FU resistance in CCA. The attenuation of p53 by molecular targeting of Δ133p53 may modulate the chemosensitivity in CCA, hence the potential for use of Δ133p53 as a candidate for targeted therapy.

Acknowledgements

This study was supported by the Higher Education Research Promotion and National Research University Project of Thailand, Office of the Higher Education Commission, through the Health Cluster (SHeP-GMS), Khon Kaen University (Grant no. H-2553-Ph.D-06); the Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University.

References

1 

Vatanasapt V, Sriamporn S and Vatanasapt P: Cancer control in Thailand. Jpn J Clin Oncol. 32(Suppl): S82–S91. 2002. View Article : Google Scholar : PubMed/NCBI

2 

Sriamporn S, Pisani P, Pipitgool V, Suwanrungruang K, Kamsa-ard S and Parkin DM: Prevalence of Opisthorchisviverrini infection and incidence of cholangiocarcinoma in Khon Kaen, Northeast Thailand. Trop Med Int Health. 9:588–594. 2004. View Article : Google Scholar : PubMed/NCBI

3 

Patel T: Worldwide trends in mortality from biliary tract malignancies. BMC Cancer. 2:102002. View Article : Google Scholar : PubMed/NCBI

4 

Thamavit W, Bhamarapravati N, Sahaphong S, Vajrasthira S and Angsubhakorn S: Effects of dimethylnitrosamine on induction of cholangiocarcinoma in Opisthorchis viverrini-infected Syrian golden hamsters. Cancer Res. 38:4634–4639. 1978.PubMed/NCBI

5 

Thamavit W, Kongkanuntn R, Tiwawech D and Moore MA: Level of Opisthorchis infestation and carcinogen dose-dependence of cholangiocarcinoma induction in Syrian golden hamsters. Virchows Arch B Cell Pathol Incl Mol Pathol. 54:52–58. 1987. View Article : Google Scholar : PubMed/NCBI

6 

Patel T: Cholangiocarcinoma. Nat Clin Pract Gastroenterol Hepatol. 3:33–42. 2006. View Article : Google Scholar : PubMed/NCBI

7 

Shaib Y and El-Serag HB: The epidemiology of cholangiocarcinoma. Semin Liver Dis. 24:115–125. 2004. View Article : Google Scholar : PubMed/NCBI

8 

Patel T: Cholangiocarcinoma - controversies and challenges. Nat Rev Gastroenterol Hepatol. 8:189–200. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Thongprasert S: The role of chemotherapy in cholangiocarcinoma. Ann Oncol. 16(Suppl 2): ii93–ii96. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Patt YZ, Hassan MM, Lozano RD, Waugh KA, Hoque AM, Frome AI, Lahoti S, Ellis L, Vauthey JN, Curley SA, et al: Phase II trial of cisplatin, interferon alpha-2b, doxorubicin, and 5-fluorouracil for biliary tract cancer. Clin Cancer Res. 7:3375–3380. 2001.PubMed/NCBI

11 

Lee MA, Woo IS, Kang JH, Hong YS and Lee KS: Epirubicin, cisplatin, and protracted infusion of 5-FU (ECF) in advanced intrahepatic cholangiocarcinoma. J Cancer Res Clin Oncol. 130:346–350. 2004. View Article : Google Scholar : PubMed/NCBI

12 

Sookprasert A, Chindaprasert J and Wirasorn K: Systemic therapy for locally advanced and metastatic cholangiocarcinoma. Asian Pac J Cancer Prev. 13(Suppl): S3–S6. 2012.

13 

Choi CW, Choi IK, Seo JH, Kim BS, Kim JS, Kim CD, Um SH, Kim JS and Kim YH: Effects of 5-fluorouracil and leucovorin in the treatment of pancreatic-biliary tract adenocarcinomas. Am J Clin Oncol. 23:425–428. 2000. View Article : Google Scholar : PubMed/NCBI

14 

Longley DB, Harkin DP and Johnston PG: 5-fluorouracil: Mechanisms of action and clinical strategies. Nat Rev Cancer. 3:330–338. 2003. View Article : Google Scholar : PubMed/NCBI

15 

Noordhuis P, Holwerda U, Van der Wilt CL, Van Groeningen CJ, Smid K, Meijer S, Pinedo HM and Peters GJ: 5-Fluorouracil incorporation into RNA and DNA in relation to thymidylate synthase inhibition of human colorectal cancers. Ann Oncol. 15:1025–1032. 2004. View Article : Google Scholar : PubMed/NCBI

16 

Namwat N, Amimanan P, Loilome W, Jearanaikoon P, Sripa B, Bhudhisawasdi V and Tassaneeyakul W: Characterization of 5-fluorouracil-resistant cholangiocarcinoma cell lines. Chemotherapy. 54:343–351. 2008. View Article : Google Scholar : PubMed/NCBI

17 

Bourdon JC, Fernandes K, Murray-Zmijewski F, Liu G, Diot A, Xirodimas DP, Saville MK and Lane DP: p53 isoforms can regulate p53 transcriptional activity. Genes Dev. 19:2122–2137. 2005. View Article : Google Scholar : PubMed/NCBI

18 

Marcel V, Perrier S, Aoubala M, Ageorges S, Groves MJ, Diot A, Fernandes K, Tauro S and Bourdon JC: Δ160p53 is a novel N-terminal p53 isoform encoded by Δ133p53 transcript. FEBS Lett. 584:4463–4468. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Fujita K, Mondal AM, Horikawa I, Nguyen GH, Kumamoto K, Sohn JJ, Bowman ED, Mathe EA, Schetter AJ, Pine SR, et al: p53 isoforms Delta133p53 and p53beta are endogenous regulators of replicative cellular senescence. Nat Cell Biol. 11:1135–1142. 2009. View Article : Google Scholar : PubMed/NCBI

20 

Lu X: Tied up in loops: Positive and negative autoregulation of p53. Cold Spring Harb Perspect Biol. 2:a0009842010. View Article : Google Scholar : PubMed/NCBI

21 

Song W, Huo SW, Lü JJ, Liu Z, Fang XL, Jin XB and Yuan MZ: Expression of p53 isoforms in renal cell carcinoma. Chin Med J (Engl). 122:921–926. 2009.

22 

Anensen N, Oyan AM, Bourdon JC, Kalland KH, Bruserud O and Gjertsen BT: A distinct p53 protein isoform signature reflects the onset of induction chemotherapy for acute myeloid leukemia. Clin Cancer Res. 12:3985–3992. 2006. View Article : Google Scholar : PubMed/NCBI

23 

Hofstetter G, Berger A, Fiegl H, Slade N, Zori A, Holzer B, Schuster E, Mobus VJ, Reimer D, Daxenbichler G, et al: Alternative splicing of p53 and p73: The novel p53 splice variant p53delta is an independent prognostic marker in ovarian cancer. Oncogene. 29:1997–2004. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Boldrup L, Bourdon JC, Coates PJ, Sjöström B and Nylander K: Expression of p53 isoforms in squamous cell carcinoma of the head and neck. Eur J Cancer. 43:617–623. 2007. View Article : Google Scholar : PubMed/NCBI

25 

Avery-Kiejda KA, Zhang XD, Adams LJ, Scott RJ, Vojtesek B, Lane DP and Hersey P: Small molecular weight variants of p53 are expressed in human melanoma cells and are induced by the DNA-damaging agent cisplatin. Clin Cancer Res. 14:1659–1668. 2008. View Article : Google Scholar : PubMed/NCBI

26 

Nutthasirikul N, Limpaiboon T, Leelayuwat C, Patrakitkomjorn S and Jearanaikoon P: Ratio disruption of the 133p53 and TAp53 isoform equilibrium correlates with poor clinical outcome in intrahepatic cholangiocarcinoma. Int J Oncol. 42:1181–1188. 2013.PubMed/NCBI

27 

Aoubala M, Murray-Zmijewski F, Khoury MP, Fernandes K, Perrier S, Bernard H, Prats AC, Lane DP and Bourdon JC: p53 directly transactivates Δ133p53α, regulating cell fate outcome in response to DNA damage. Cell Death Differ. 18:248–258. 2011. View Article : Google Scholar :

28 

Hahnvajanawong C, Chaiyagool J, Seubwai W, Bhudhisawasdi V, Namwat N, Khuntikeo N, Sripa B, Pugk hem A and Tassaneeyakul W: Orotate phosphoribosyl transferase mRNA expression and the response of cholangiocarcinoma to 5-fluorouracil. World J Gastroenterol. 18:3955–3961. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Tepsiri N, Chaturat L, Sripa B, Namwat W, Wongkham S, Bhudhisawasdi V and Tassaneeyakul W: Drug sensitivity and drug resistance profiles of human intrahepatic cholangiocarcinoma cell lines. World J Gastroenterol. 11:2748–2753. 2005. View Article : Google Scholar : PubMed/NCBI

30 

Thanasai J, Limpaiboon T, Jearanaikoon P, Sripa B, Pairojkul C, Tantimavanich S and Miwa M: Effects of thymidine phosphorylase on tumor aggressiveness and 5-fluorouracil sensitivity in cholangiocarcinoma. World J Gastroenterol. 16:1631–1638. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Voigt W: Sulforhodamine B assay and chemosensitivity. Methods Mol Med. 110:39–48. 2005.PubMed/NCBI

32 

Wattanawongdon W, Hahnvajanawong C, Namwat N, Kanchanawat S, Boonmars T, Jearanaikoon P, Leelayuwat C, Techasen A and Seubwai W: Establishment and characterization of gemcitabine-resistant human cholangiocarcinoma cell lines with multidrug resistance and enhanced invasiveness. Int J Oncol. 47:398–410. 2015.PubMed/NCBI

33 

Nakano Y, Naoe T, Kiyoi H, Kitamura K, Minami S, Miyawaki S, Asou N, Kuriyama K, Kusumoto S, Shimazaki C, et al: Prognostic value of p53 gene mutations and the product expression in de novo acute myeloid leukemia. Eur J Haematol. 65:23–31. 2000. View Article : Google Scholar : PubMed/NCBI

34 

Albaric O, Bret L, Amardeihl M and Delverdier M: Immunohistochemical expression of p53 in animal tumors: A methodological study using four anti-human p53 antibodies. Histol Histopathol. 16:113–121. 2001.PubMed/NCBI

35 

Limpaiboon T, Sripa B, Wongkham S, Bhudhisawasdi V, Chau-in S and Teerajetgul Y: Anti-p53 antibodies and p53 protein expression in cholangiocarcinoma. Hepatogastroenterology. 51:25–28. 2004.PubMed/NCBI

36 

Ong CK, Subimerb C, Pairojkul C, Wongkham S, Cutcutache I, Yu W, McPherson JR, Allen GE, Ng CC, Wong BH, et al: Exome sequencing of liver fluke-associated cholangiocarcinoma. Nat Genet. 44:690–693. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Liang JT, Huang KC, Cheng YM, Hsu HC, Cheng AL, Hsu CH, Yeh KH, Wang SM and Chang KJ: P53 overexpression predicts poor chemosensitivity to high-dose 5-fluorouracil plus leucovorin chemotherapy for stage IV colorectal cancers after palliative bowel resection. Int J Cancer. 97:451–457. 2002. View Article : Google Scholar : PubMed/NCBI

38 

Elsaleh H, Powell B, McCaul K, Grieu F, Grant R, Joseph D and Iacopetta B: P53 alteration and microsatellite instability have predictive value for survival benefit from chemotherapy in stage III colorectal carcinoma. Clin Cancer Res. 7:1343–1349. 2001.PubMed/NCBI

39 

Kaeser MD, Pebernard S and Iggo RD: Regulation of p53 stability and function in HCT116 colon cancer cells. J Biol Chem. 279:7598–7605. 2004. View Article : Google Scholar

40 

Reles A, Wen WH, Schmider A, Gee C, Runnebaum IB, Kilian U, Jones LA, El-Naggar A, Minguillon C, Schönborn I, et al: Correlation of p53 mutations with resistance to platinum-based chemotherapy and shortened survival in ovarian cancer. Clin Cancer Res. 7:2984–2997. 2001.PubMed/NCBI

41 

Longley DB, Boyer J, Allen WL, Latif T, Ferguson PR, Maxwell PJ, McDermott U, Lynch M, Harkin DP and Johnston PG: The role of thymidylate synthase induction in modulating p53-regulated gene expression in response to 5- fluorouracil and antifolates. Cancer Res. 62:2644–2649. 2002.PubMed/NCBI

42 

Goldschneider D, Blanc E, Raguénez G, Barrois M, Legrand A, Le Roux G, Haddada H, Bénard J and Douc-Rasy S: Differential response of p53 target genes to p73 overexpression in SH-SY5Y neuroblastoma cell line. J Cell Sci. 117:293–301. 2004. View Article : Google Scholar

43 

Fang L, Lee SW and Aaronson SA: Comparative analysis of p73 and p53 regulation and effector functions. J Cell Biol. 147:823–830. 1999. View Article : Google Scholar : PubMed/NCBI

44 

Fulco M, Costanzo A, Merlo P, Mangiacasale R, Strano S, Blandino G, Balsano C, Lavia P and Levrero M: p73 is regulated by phosphorylation at the G2/M transition. J Biol Chem. 278:49196–49202. 2003. View Article : Google Scholar : PubMed/NCBI

45 

Zhu J, Jiang J, Zhou W and Chen X: The potential tumor suppressor p73 differentially regulates cellular p53 target genes. Cancer Res. 58:5061–5065. 1998.PubMed/NCBI

46 

He Y, Fan S, Jiang Y, Chen J, Li Z, Zhou P and Zhou Y: Effect of exogenous p73 gene on chemosensitivity of wild-type p53 human lung adenocarcinoma cell A549. Zhongguo Fei Ai Za Zhi. 7:331–335. 2004.(In Chinese). PubMed/NCBI

47 

He Y, Fan SZ, Jiang YG, Chen JM, Li ZP, Zhou P and Zhou YG: Effect of p73 gene on chemosensitivity of human lung adenocarcinoma cells H1299. Ai Zheng. 23:645–649. 2004.(In Chinese). PubMed/NCBI

48 

Adamsen BL, Kravik KL, Clausen OP and De Angelis PM: Apoptosis, cell cycle progression and gene expression in TP53-depleted HCT116 colon cancer cells in response to short-term 5- fluorouracil treatment. Int J Oncol. 31:1491–1500. 2007.PubMed/NCBI

49 

Guo X, Goessl E, Jin G, Collie-Duguid ES, Cassidy J, Wang W and O'Brien V: Cell cycle perturbation and acquired 5-fluorouracil chemoresistance. Anticancer Res. 28A:9–14. 2008.

50 

Levrero M, De Laurenzi V, Costanzo A, Gong J, Wang JY and Melino G: The p53/p63/p73 family of transcription factors: Overlapping and distinct functions. J Cell Sci. 113:1661–1670. 2000.PubMed/NCBI

51 

Jha AK, Nikbakht M, Jain V, Sehgal A, Capalash N and Kaur J: Promoter hypermethylation of p73 and p53 genes in cervical cancer patients among north Indian population. Mol Biol Rep. 39:9145–9157. 2012. View Article : Google Scholar : PubMed/NCBI

52 

Zhang YL, Guo XR, Shen DH, Cheng YX, Liang XD, Chen YX and Wang Y: Expression and promotor methylation of p73 gene in ovarian epithelial tumors. Zhonghua Bing Li Xue Za Zhi. 41:33–38. 2012.PubMed/NCBI

53 

House MG, Wistuba II, Argani P, Guo M, Schulick RD, Hruban RH, Herman JG and Maitra A: Progression of gene hypermethylation in gallstone disease leading to gallbladder cancer. Ann Surg Oncol. 10:882–889. 2003. View Article : Google Scholar : PubMed/NCBI

54 

Kawano S, Miller CW, Gombart AF, Bartram CR, Matsuo Y, Asou H, Sakashita A, Said J, Tatsumi E and Koeffler HP: Loss of p73 gene expression in leukemias/lymphomas due to hypermethylation. Blood. 94:1113–1120. 1999.PubMed/NCBI

55 

Yang B, House MG, Guo M, Herman JG and Clark DP: Promoter methylation profiles of tumor suppressor genes in intrahepatic and extrahepatic cholangiocarcinoma. Mod Pathol. 18:412–420. 2005. View Article : Google Scholar

Related Articles

Journal Cover

December-2015
Volume 47 Issue 6

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Nutthasirikul N, Hahnvajanawong C, Techasen A, Limpaiboon T, Leelayuwat C, Chau-In S and Jearanaikoon P: Targeting the ∆133p53 isoform can restore chemosensitivity in 5-fluorouracil-resistant cholangiocarcinoma cells. Int J Oncol 47: 2153-2164, 2015
APA
Nutthasirikul, N., Hahnvajanawong, C., Techasen, A., Limpaiboon, T., Leelayuwat, C., Chau-In, S., & Jearanaikoon, P. (2015). Targeting the ∆133p53 isoform can restore chemosensitivity in 5-fluorouracil-resistant cholangiocarcinoma cells. International Journal of Oncology, 47, 2153-2164. https://doi.org/10.3892/ijo.2015.3188
MLA
Nutthasirikul, N., Hahnvajanawong, C., Techasen, A., Limpaiboon, T., Leelayuwat, C., Chau-In, S., Jearanaikoon, P."Targeting the ∆133p53 isoform can restore chemosensitivity in 5-fluorouracil-resistant cholangiocarcinoma cells". International Journal of Oncology 47.6 (2015): 2153-2164.
Chicago
Nutthasirikul, N., Hahnvajanawong, C., Techasen, A., Limpaiboon, T., Leelayuwat, C., Chau-In, S., Jearanaikoon, P."Targeting the ∆133p53 isoform can restore chemosensitivity in 5-fluorouracil-resistant cholangiocarcinoma cells". International Journal of Oncology 47, no. 6 (2015): 2153-2164. https://doi.org/10.3892/ijo.2015.3188