Open Access

Gadd45β silencing impaired viability and metastatic phenotypes in cholangiocarcinoma cells by modulating the EMT pathway

  • Authors:
    • Kyaw Zwar Myint
    • Pornparn Kongpracha
    • Panthip Rattanasinganchan
    • Kawin Leelawat
    • Penpak Moolthiya
    • Kittipong Chaiyabutr
    • Rutaiwan Tohtong
  • View Affiliations

  • Published online on: December 28, 2017     https://doi.org/10.3892/ol.2017.7706
  • Pages: 3031-3041
  • Copyright: © Myint 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

Growth arrest and DNA damage-inducible-β (Gadd45β) is a stress-response protein involved in a number of processes, including cell cycle control, DNA repair, survival and death control, and stress signaling, depending on its interactions. Gadd45β expression is dysregulated in numerous types of cancer, functioning as either a tumor promoter or a tumor suppressor. However, the functions of Gadd45β in cholangiocarcinoma (CCA), particularly in metastasis, has not been studied. The immunohistochemical analysis of Gadd45β expression revealed that 75% of histological specimens from patients with CCA expressed high levels of Gadd45β, and that high Gadd45β expression was associated with metastasis. The role of Gadd45β in CCA was examined using siRNA‑mediated gene knockdown in HuCCA‑1, a human CCA cell line established from a Thai patient. The effects of Gadd45β downregulation upon cell viability and death, invasion, migration, matrix metalloproteinase (MMP) activity and epithelial‑mesenchymal transition (EMT) marker expression were investigated. Gadd45β knockdown impaired cell viability, which was associated with the induction of apoptosis. In addition, there was a marked reduction in invasion and migration, although MMP activity was unaffected. Impairment of these metastatic properties was accompanied by the decreased expression of EMT markers, including Slug, vimentin, claudin‑1 and zona occludens protein 1, whereas E‑cadherin expression was increased. The present study suggests that Gadd45β is involved in regulating the viability and the metastatic potential of CCA cells, which may be mediated by the modulation of the EMT pathway.

Introduction

Cholangiocarcinoma (CCA) is a cancer of the bile duct caused by the malignant transformation of cholangiocytes, the epithelial cells lining the bile duct. CCA is the second most common type of primary hepatic malignancy (1,2). Due to late diagnosis, resistance to chemo- and radiotherapy, and the high rates of local invasion and distant metastasis, the prognosis for CCA is very poor. The median survival time of patients with CCA is <2 years (3), and the 5-year survival rate of patients receiving curative resection is 0–40% (4). The prevalence of CCA is the highest in Asia, particularly in the northeastern region of Thailand (5). Opisthorchis viverrini infection is hypothesized to be a causal factor of CCA in Thailand, as it is markedly associated with the incidence of CCA (6).

In total, >90% of cancer-associated mortality is due to the local or distant metastasis of cancer cells (7). The epithelial-mesenchymal transition (EMT) is an important process in cancer metastasis, characterized by alterations in the gene expression and morphology of cells, which leads to a reduction of intercellular adhesion, and an increase in cell motility (811). This process is associated with a reduction of E-cadherin expression (12,13) and an increase in the expression of vimentin, an intermediate filament protein, leading to increased cell motility and promoting tumor metastasis (14,15).

Growth arrest and DNA damage-inducible-β (Gadd45β) is a stress-response protein; its expression is induced by physiological or environmental stress. The aberrant expression of Gadd45β in various types of cancer has implicated its involvement in tumorigenesis (16). Gadd45β belongs to the Gadd45 protein family (Gadd45α, Gadd45β, Gadd45γ) (17). Gadd45β may form a homodimer or heterodimer with the other Gadd45 proteins, or interact with a variety of other proteins, including proliferating cell nuclear antigen, cyclin-dependent kinase 1, p21, mitogen-activated protein kinase kinase kinase 4, mitogen-activated protein kinase kinase 7 and p38 mitogen activated protein kinase (MAPK). The function of Gadd45β differs depending on the interacting molecules, including cell cycle control, DNA repair, survival and death control, and stress signaling (1822).

Cancer cells must survive and propagate in a strenuous environment of hypoxia, nutrient competition and oxidative stress (23). It is essential for cells to acquire the ability to thrive in these stressful conditions. The function of Gadd45β as a stress-response protein in cancer is paradoxical; the downregulation of Gadd45β via promoter methylation in hepatocellular carcinoma suggests that Gadd45β may act as a tumor suppressor (24), whereas the upregulation of Gadd45β in colorectal cancer was associated with recurrence and mortality of patients with colorectal cancer (25), suggesting a tumor-promoting role. When the Gadd45β gene from normal adjacent tissue was over-expressed in colorectal cancer cell lines, apoptotic cell death was induced (25). In addition, Gadd45β was identified as upregulated in the metastasis of uveal melanoma to the liver (26), and the silencing of Gadd45β in human embryonic carcinoma cells decreased viability and invasiveness (27), suggesting that Gadd45β may contribute to the malignant phenotypes of cancer. However, the function of Gadd45β in metastasis and EMT is not yet fully characterized.

In the present study, it was identified that patients with CCA exhibit increased Gadd45β expression in tumor tissue, and that a high level of Gadd45β expression was associated with metastasis. Gadd45β expression in a CCA cell line, HuCCA-1, was suppressed using siRNA-mediated gene silencing, and the effects on cell viability, survival and death signaling pathways, migration, invasiveness, and the EMT pathway were studied. The data of the present study thus indicated that Gadd45β expression promoted the viability, migration and invasion of the HuCCA-1 cells, traits required for successful metastasis.

Materials and methods

Ethical approval

All procedures performed in the present study involving human participants were performed in accordance with the 1964 Declaration of Helsinki and its later amendments, or comparable ethical standards. The study was conducted with the approval of the Ethical Committee of Rajavithi Hospital (Bangkok, Thailand).

Immunohistochemical (IHC) staining

Paraffinized tissue samples from 28 patients with CCA who had undergone surgical treatment at Rajavithi Hospital between January 2010 and October 2010 were selected for retrospective analysis. Standard IHC technique was used for the detection of Gadd45β in paraffinized sections on glass slides. Polyclonal Gadd45β antibody (HPA029816-100UL; 1:500 dilution; Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) was hybridized with the sections overnight at 4°C, followed by incubation with biotinylated goat anti-rabbit IgG at RT for 1 h (E0432; dilution 1:500; Dako; Agilent Technologies, Inc., Santa Clara, CA, USA). Subsequently avidin-biotin-peroxidase conjugate was added (ABC Elite; Vector Laboratories, Burlingame, CA, USA) for 30 min at room temperature and the immunohistochemical reaction was developed with freshly prepared reagents from a Histofine SAB-PO kit (Nichirei, Inc., Tokyo, Japan). The samples were visualized at ×400 magnification using an Olympus BX53 microscope (Olympus Corporation, Tokyo, Japan) and categorized into four grades according to the intensity and percentage of positively stained cells: Negative or <5%, grade 0; weak or 5–25%, grade 1; moderate or 25–50%, grade 2; and strong or >50%, grade 3. IHC grade 0 (3 cases) and grade 1 (4 cases), which exhibited weak and/or <25% staining, were grouped as low expression, whereas grade 2 (5 cases) and grade 3 (16 cases), which exhibited moderate to strong and/or >25% staining, were grouped as high expression of Gadd45β for the purpose of statistical analysis.

Cell line and culture condition

The human CCA cell line, HuCCA-1 (28), developed from a Thai patient with CCA, was used for the study. The cells were provided by Professor Satitaya Sirisinha (Mahidol University, Bangkok, Thailand). These cells were grown in HAM's F-12 medium supplemented with 10% fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) at 37°C in 5% CO2 humidified atmosphere.

Small interfering (si)RNA-mediated Gadd45β gene silencing

Commercial Gadd45β siRNA [GADD45β siRNA(h): sc-37416], a pool of 3 different siRNA duplexes to the Gadd45β gene (NCBI RefSeq. 12759252), was purchased from Santa Cruz Biotechnology, Inc. (Dallas, TX, USA). Silencer® Negative control siRNA no. 1 (cat. no. 4404021; Ambion; Thermo Fisher Scientific, Inc.) was used as negative control siRNA. The cells were washed with PBS and detached using 0.25% Trypsin/EDTA (Gibco; Thermo Fisher Scientific, Inc.). Cells were seeded at 2×105 cells/well into a 6 well plate for 24 h, then transfected with 40 nM siRNA using Lipofectamine® RNAiMax reagent (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. The medium containing transfection reagents was changed for 10% FBS supplemented HAM's-F12 at 24 h after transfection.

Western blot analysis

At 48 h post-transfection, total cellular protein was extracted and prepared following a protocol described previously (29). Each protein sample of 40 µg was separated via SDS-PAGE (12% gel; 120 V for 2 h) followed by an electroblotting transfer of the protein to a nitrocellulose membrane at 30 V at 4°C for 15 h. The membranes were blocked in 1.5% skimmed milk at room temperature for 1 h, the membranes were incubated with the primary antibodies at 4°C overnight on a rocking platform. The next day, the membranes were washed with TBS with Tween-20 prior to incubation with horseradish peroxidase-conjugated secondary antibodies for 1 h at room temperature. Following 3 washes with TBST for 10 min, ECL Plus Western Blotting Detection system (Bio-Rad Laboratories, Inc., Hercules, CA, USA) was used to visualize the immunoreactive bands in G:Box ChemiXL 1.4 (Syngene; Synoptics, Cambridge, UK).

Rabbit polyclonal Gadd45β antibodies were purchased from Abcam (Cambridge, UK) and goat polyclonal GAPDH antibodies from Santa Cruz Biotechnology, Inc. The rest of the antibodies used in the study were purchased from Cell Signaling Technology, Inc. (Danvers, MA, USA); the details regarding all antibodies used in western blotting are included in Table I. Densitometry analysis was performed using ImageJ software (version 1.49v; National Institutes of Health, Bethesda, MD, USA) (30). The density of the protein of interest relative to GAPDH was determined in HuCCA-1 cells transfected with Gadd45β or negative control siRNA. The activation of Akt, p38 MAPK and extracellular signal-regulated kinase (ERK)1/2 signaling pathways was examined using phospho-specific antibodies at 48 h after siRNA transfection. The densitometry measurements for total Akt, ERK1/2 and p38 MAPK were used for the normalization of protein loading. The phospho-protein/total protein density ratio was determined in HuCCA-1 cells transfected with Gadd45β siRNA or negative control siRNA. The phospho-protein/total protein density ratio of cells transfected with negative control siRNA was designated as 100%.

Table I.

The primary antibodies used for western blotting in the present study.

Table I.

The primary antibodies used for western blotting in the present study.

AntibodyCat. no.Clone no.Type of antibodyOrganismCompanyDilution
Gadd45βab105060 PolyclonalRabbitAbcam1:2,000
GAPDHsc-48166I-19PolyclonalGoatSanta Cruz Biotechnology, Inc.1:2,000
Cleaved PARP (Asp214)#5625D64E10MonoclonalRabbitCell Signaling Technology, Inc.1:1,000
Total PARP#9542 MonoclonalRabbitCell Signaling Technology, Inc.1:1,000
Cleaved caspase-3 (Asp175)#96645A1EMonoclonalRabbitCell Signaling Technology, Inc.1:1,000
Full-length caspase 3#9662 MonoclonalRabbitCell Signaling Technology, Inc.1:1,000
Phospho-p38 MAPK (Thr180/Tyr182)#9211 MonoclonalRabbitCell Signaling Technology, Inc.1:1,000
p38 MAPK#9212 MonoclonalRabbitCell Signaling Technology, Inc.1:1,000
Phospho-Akt (Ser473)#4058193H12MonoclonalRabbitCell Signaling Technology, Inc.1:1,000
Akt#9272 MonoclonalRabbitCell Signaling Technology, Inc.1:1,000
Phospho-ERK1/2 (Thr202/Tyr204)#9101 MonoclonalRabbitCell Signaling Technology, Inc.1:1,000
ERK1/2#9102 MonoclonalRabbitCell Signaling Technology, Inc.1:1,000
Vimentin#5741D21H3MonoclonalRabbitCell Signaling Technology, Inc.1:1,000
Claudin-1#13255D5H1DMonoclonalRabbitCell Signaling Technology, Inc.1:1,000
β-catenin#8480D10A8MonoclonalRabbitCell Signaling Technology, Inc.1:1,000
ZO-1#8193D7D12MonoclonalRabbitCell Signaling Technology, Inc.1:1,000
Slug#9585C19G7MonoclonalRabbitCell Signaling Technology, Inc.1:1,000
E-cadherin#319524E10MonoclonalRabbitCell Signaling Technology, Inc.1:1,000

[i] Gadd45β, growth arrest and DNA damage-inducible-β; PARP, Poly (ADP-ribose) polymerase; p38 MAPK, p38 mitogen-activated protein kinase; ERK, extracellular signal-regulated kinase; ZO-1, zona occludens protein 1.

Cell viability assays
Live/Dead® cell viability assay

HuCCA-1 cells were grown on coverslips prior to transfection with siRNA. At 48 h post-transfection, cell viability was assessed using the Live/Dead® Viability/Cytotoxicity kit for mammalian cells (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. Briefly, a working solution, containing 2 µM calcein AM and 4 µM ethidium homodimer (EthD-1), was prepared in PBS. The cells were washed with pre-warmed PBS, and 150 µl of Live/Dead solution was applied to each cover slip. Following incubation at 37°C in a 5% CO2 humidified atmosphere for 30 min, the reagents were removed and the cover slips were washed with PBS, mounted on the glass-slides, and viewed using a Nikon Eclipse TE2000-U microscope (Nikon Corporation, Tokyo, Japan) using B-2A (green, calcein) and G-2A (red, EthD-1) excitation filters. Images were recorded at ×100 total magnification using ACT-1C software (version 1.02; Nikon Corporation). Live cells were stained with calcein-AM (green), whereas dead cells were stained with ethidium homodimer (red).

WST-1 assay

A total of 5,000 HuCCA-1 cells were seeded in 96-well plates for 24 h prior to siRNA transfection. At 24, 48 and 72 h post-transfection, 10 µl of WST-1 reagent (Roche Diagnostics, Basel, Switzerland) was added into each well. The plates were incubated at 37°C in a humidified atmosphere containing 5% CO2 for 2 h. Then the absorbance at 450 nm was measured.

Hoechst staining

At 48 h post-transfection, the culture media was carefully removed to avoid the loss of detached cells. HuCCA-1 cells were then fixed in 2% w/v paraformaldehyde solution for 30 min at room temperature prior to permeabilization using 0.03% v/v Triton X-100 for 30 min. Nuclear staining was performed with Hoechst 333258 dye at room temperature (5 µg/ml; Thermo Fisher Scientific, Inc.) for 2 h, followed by visualization at ×400 total magnification with an Olympus IX83 live cell fluorescence microscope (Olympus Corporation).

Gelatin zymography

To analyze the activity of MMPs, the gelatinase activity of the enzymes secreted into the medium was determined using gelatin zymography assay, following a previously described protocol (29). The gelatinolytic activity of MMPs was observed as clear bands on the blue background of the Coomassie-stained gel. Densitometry analysis was performed using ImageJ software (30).

Cell invasion and migration assays

Cells were harvested at 48 h post-transfection for in vitro invasion and migration assays using transwell chambers, as described by Li et al (31). Briefly, HuCCA-1 cells (1×105 cells/well) transfected with Gadd45β siRNA or negative control siRNA were seeded into the upper chamber of the transwells coated with Matrigel for the invasion assay and with no coating for the migration assay, and analyzed after 6 h of incubation at 37°C. HAM's F-12 supplemented with 10% FBS, used as chemoattractant, was added to the lower chamber. Cells were seeded in the upper chamber in serum free medium. Three independent experiments were performed in duplicate transwells.

Statistical analysis

Data is presented as the mean ± standard deviation and every experiment was performed at least three times. The χ2 test was used for clinicopathological data analysis, and experimental data analysis was performed using a paired t-test. P<0.05 was considered to indicate a statistically significant difference.

Results

High levels of Gadd45β expression are associated with metastatic incidence in CCA

Gadd45β expression in the histological specimens of CCA tissue from 28 patients was examined with IHC staining. The expression of Gadd45β was detected in all tumor tissues (n=28), and 75% (n=21) exhibited higher Gadd45β expression than the surrounding stroma (Fig. 1A-D). Although Gadd45β expression was previously reported to be moderate in the liver, gall bladder and pancreas (32), the expression level in cholangiocytes or CCA tissue has not previously been reported, to the best of our knowledge. A high level of Gadd45β expression was associated with the incidence of metastasis in patients (P=0.035), although no statistically significant association was observed between the level of Gadd45β expression and age, sex, lymph node involvement or differentiation status (Table I).

Gadd45β knockdown reduces the viability of HuCCA-1 cells

To investigate the functional importance of Gadd45β in CCA, Gadd45β expression was suppressed using siRNA-mediated silencing in HuCCA-1, a CCA cell line that expresses moderate levels of Gadd45β. Gadd45β siRNA transfection decreased the expression of Gadd45β in HuCCA-1 to 19.13±2.66% relative to negative control siRNA transfection (Fig. 2A and B).

During Gadd45β silencing experiments, a decrease in the confluence of Gadd45β-silenced HuCCA-1 cells was observed compared with negative control siRNA-transfected cells (Fig. 3A and B). Furthermore, there was an increase in the number of cells floating in the medium of Gadd45β-silenced HuCCA-1 cells, indicating that silencing of Gadd45β perturbed the viability of HuCCA-1 cells (data not shown). A Live/Dead® cell viability assay confirmed that there was an increase in the number of dead cells in HuCCA-1 cells when Gadd45β was silenced (Fig. 3C and D). Cell viability was assessed using a WST-1 assay, in which the WST-1 reagent was added directly into the cells without removing the culture medium, preventing the loss of detached viable cells. Consistent with visual observation, Gadd45β silencing significantly decreased the viability of HuCCA-1 cells (Fig. 3E).

Gadd45β silencing kills HuCCA-1 cells via apoptosis

The mode of cell death in HuCCA-1 cells upon Gadd45β silencing was examined by staining the nuclear DNA with Hoechst 333258. Upon silencing of Gadd45β, HuCCA-1 cells were observed to exhibit nuclear condensation and DNA fragmentation, characteristics of apoptotic cells (33). The nuclei of live cells were homogenously stained in blue, whereas those of apoptotic cells appeared condensed and fragmented. Gadd45β-silenced HuCCA-1 cells exhibited an increased rate of apoptotic cells compared with negative control cells (24.87±5.96% vs. 9.18±3.59%; Fig. 3F-H). Consistently, western blot analysis revealed that poly (ADP-ribose) polymerase (PARP) cleavage and caspase-3 cleavage were observed, indicating that HuCCA-1 cells died via apoptosis when Gadd45β was silenced (Fig. 3I) (34).

Gadd45β silencing reduces Akt activity in HuCCA-1

To determine which signaling pathways were involved, three of the principal pathways involved in the regulation of cell viability and proliferation, the Akt, ERK and p38 MAPK pathways, were studied. Akt is associated with cell proliferation, survival and apoptosis inhibition (35). p38 MAPK is a stress-activated protein kinase pathway important for cell proliferation, differentiation, survival and migration (36,37). ERK1/2 activation has been demonstrated to protect cells from apoptosis (38).

Gadd45β silencing resulted in a marked reduction of Akt phosphorylation (P=0.0237), whereas no significant alteration in p38 MAPK or ERK1/2 phosphorylation was observed (Fig. 4A-D). The decrease in Akt activity may be responsible for the reduction of viability in HuCCA-1 upon Gadd45β silencing.

Gadd45β silencing reduces the invasion and migration of HuCCA-1 cells

A hallmark of malignancy is the ability to invade local tissues or to spread to distant sites (39). During this process, the cancer cells must acquire the ability to invade and migrate through connective tissue barriers, including the basement membrane, surrounding matrix and existing blood vessels (40). IHC data analysis of patient samples revealed that there was an association between the high expression of Gadd45β and metastasis (Table II). Furthermore, Gadd45β was demonstrated to be important for the viability of HuCCA-1 cells in the present study; viability is required by the cancer cells for successful metastasis (41). Therefore, the effect of Gadd45β silencing on the migration and invasion abilities of HuCCA-1 cells was investigated using in vitro transwell migration and invasion assays. Gadd45β silencing decreased the migration of HuCCA-1 cells to 35.67±5.60%, and invasion to 39.59±11.09%, compared with the negative control siRNA (Fig. 5A and B).

Table II.

Association between Gadd45β expression and the clinicopathological features of patients with cholangiocarcinoma.

Table II.

Association between Gadd45β expression and the clinicopathological features of patients with cholangiocarcinoma.

Gadd45β expression

Clinicopathological parameterTotalLowHighP-value
Total, n (%)287 (25.0)21 (75.0)
Age, years 0.378
  Mean ± standard deviation59.68±10.41
  <60, n (%)12 (42.9)4 (14.3)8 (28.6)
  ≥60, n (%)16 (57.1)3 (10.7)13 (46.4)
Sex, n (%) 0.512
  Male13 (46.4)4 (14.3)9 (32.1)
  Female15 (53.6)3 (10.7)12 (42.9)
Tumor size, cm 0.385a
  Mean ± standard deviation2.90±2.4
  <5, n (%)25 (92.6)7 (25.9)18 (66.7)
  ≥5, n (%)2 (7.4)0 (0)2 (7.4)
Lymph node metastasis, n (%) 0.190
  Absent14 (50.0)5 (17.9)9 (32.1)
  Present14 (50.0)2 (7.1)12 (42.9)
Distant metastasis, n (%) 0.035c
  Absent19 (67.9)7 (25.0)12 (42.9)
  Present9 (32.1)0 (0)9 (32.1)
Differentiation status, n (%) 0.696b
  Well11 (52.4)3 (14.3)8 (38.1)
  Moderate10 (47.6)2 (9.5)8 (38.1)
Type of surgery, n (%) 0.076
  R014 (50.0)6 (21.0)8 (28.6)
  R17 (25.0)1 (3.6)6 (21.4)
  R27 (25.0)0 (0)7 (25.0)

a Tumor size data was not available in one case

b cell differentiation status data was not available in 7 cases

c P<0.05. Gadd45β, growth arrest and DNA damage-inducible-β.

Cancer cells may secrete matrix metalloproteinases (MMPs) or stimulate surrounding stromal cells to secrete MMPs, consequently leading to digestion of the extracellular matrix, promoting local invasion and metastasis (42,43). However, gelatin zymography analysis revealed that Gadd45β silencing did not significantly alter the activity of MMP-2 (P=0.5609) and MMP-9 (P=0.1886) secreted by HuCCA-1 cells (Fig. 5C-E).

Gadd45β silencing reverses the EMT changes in HuCCA-1

Epithelial-mesenchymal transition is an important process during cancer metastasis, denoted by acquisition of mesenchymal phenotypes, including increased motility and invasiveness (8,9,4446). Since Gadd45β silencing decreased the invasion and migration of HuCCA-1 cells, properties associated with EMT, including morphological changes and EMT marker expression, were examined. It was demonstrated that Gadd45β silencing may have induced HuCCA-1 cells to undergo morphological changes, from spindle-shaped and fibroblast-like in appearance, to a flattened epithelial-like phenotype (Fig. 6A and B). The expression patterns of the proteins involved in the EMT pathway were further investigated with western blotting. It was identified that Gadd45β silencing affected the expression of multiple EMT markers (Fig. 7A-G). Epithelial marker E-cadherin expression was increased (P=0.0430), whereas the expression of the mesenchymal markers, Vimentin (P=0.0085) and Slug (P=0.0399), was decreased by Gadd45β silencing. However, there was no change in β-catenin expression (P=0.6006; Fig. 7A and D).

Notably, proteins associated with tight junctions were also affected by Gadd45β silencing. Claudin-1 and zona occludens protein 1 (ZO-1) constitute the tight junctions between cells (47,48); one of the mechanisms involved in cancer invasion is collective migration, in which the expression levels of tight-junction proteins are important to allow cells to move as a cluster (49). There was a decrease in claudin-1 (P=0.0100; Fig. 7A and F) and the ZO-1 expression (P=0.0087; Fig. 7A and G) upon Gadd45β silencing.

Discussion

The IHC staining of paraffin embedded tissue samples from patients with CCA revealed that the majority of CCA tissue samples expressed increased Gadd45β relative to non-tumor tissue, and that the high level of Gadd45β in CCA was associated with an increased incidence of metastasis in patients. These findings suggest that Gadd45β may be of functional importance in CCA. As cancer cells are continuously exposed to stressful environments including hypoxia, competition for nutrients and oxidative stress during growth (23), Gadd45β appears to enable CCA cells to cope with stress and to thrive in the harsh tumor microenvironment.

In the present study, the functional importance of Gadd45β in CCA was determined using siRNA-mediated gene silencing in HuCCA-1, a cell line established from a patient with CCA. Gadd45β silencing decreased the proliferation and induced apoptosis in HuCCA-1 cells, as demonstrated by nuclear condensation and fragmentation, together with the activation of caspase-3 and PARP cleavage. The impairment of cell proliferation together with an increased rate of apoptosis in Gadd45β-silenced cells suggests that Gadd45β serves a pro-survival role in CCA. These data are in agreement with previous reports on genotoxic stress-induced apoptosis in hematopoietic cells of Gadd45β-deficient mice (50). Additionally, Gadd45β has been demonstrated to promote the survival of mouse embryo fibroblasts in response to tumor necrosis factor-α (51) and of B cells during Fas-induced apoptosis (52).

To study the underlying molecular mechanisms by which Gadd45β promotes viability in HuCCA-1, key cellular survival and death signaling pathways were studied, since the activation of death signals or the reduction of survival signals reduces the viability of cells. The results of the present study demonstrated that Gadd45β silencing significantly decreased the Akt activity, although p38 MAPK and ERK1/2 activity was unchanged. Activation of the Akt/PI3K pathway has been associated with growth and metastasis (53); therefore, the decrease in p-Akt may be responsible for the reduced growth and induction of apoptosis in HuCCA-1 cells upon Gadd45β silencing. The altered cell signaling activities appeared to shift the balance from pro-survival to pro-apoptotic following the silencing of Gadd45β.

EMT is an important biological process that allows malignant tumor cells to acquire migratory and invasive phenotypes; prerequisites for cancer invasion and metastasis. It is indicated by the acquisition of fibroblast-like morphology, with a reduction in intercellular adhesion and increased cell motility (1118). The IHC data from the present study indicated that Gadd45β may be involved in the metastasis of CCA. Gadd45β silencing of HuCCA-1 cells induced a marked reduction in invasiveness and migration, although the activity of MMPs was not affected. A number of reports have implicated Gadd45β in the cell migration ability, an important characteristic of metastatic cancer cells. For example Salerno et al (54) demonstrated that the granulocytes of Gadd45β-deficient mice displayed the impairment of lipopolysacchiride-stimulated chemotactic migration. Furthermore, Kodama and Negishi (55) demonstrated that the ectopic expression of Gadd45β increased, whereas siRNA-mediated silencing of Gadd45β decreased, pregnane X receptor-induced cell migration in hepatocellular carcinoma cells.

Examination of the expression of EMT markers revealed that Gadd45β silencing resulted in reduction in Slug expression, with a consequent increase in E-cadherin and decrease in vimentin expression. High Slug expression has been demonstrated to induce EMT in cancer cells (56) and the effects are orchestrated by an increase in vimentin expression (57). As Slug expression is negatively regulated by GS3Kβ (58), which in turn is inactivated by pAkt (59), the decrease in Akt activity following Gadd45β silencing may have led to the downregulation of Slug expression, with the consequent suppression of vimentin expression and an increase in the invasiveness and migration of HuCCA-1 cells.

Furthermore, a decrease in the expression of the tight junction proteins, claudin-1 and ZO-1, was induced by Gadd45β silencing. Claudins and ZO-1 are integral proteins of the tight junctions that seal adjacent epithelial cells (47,48). Although the conventional roles of claudins and ZO-1 are to control paracellular ion flux and maintain cell polarity, accumulating evidence has demonstrated that these proteins also exhibit non-junctional functions, including in cell motility, and may function in the EMT, apoptosis resistance, invasion and metastasis of cancer cells (6062). Claudins have been demonstrated to promote collective migration, where groups of cells move in a coordinated manner and remain connected via cell-cell junctions. Additionally, claudin expression has been associated with increased MMP activity and cell survival (62). Claudin-1 expression has been demonstrated to be associated with invasive and metastatic phenotypes in a range of types of cancer including colon, liver, oral squamous cell carcinoma and melanoma (63). ZO-1 is a component of the ‘junctional plaque’ at the cytoplasmic surface of the tight junctions, which links integral membrane proteins with the cytoskeleton (64). It was identified in a previous study that ZO-1 can translocate from the cell membrane to the nucleus and may be involved in cell signaling (65). The upregulation of ZO-1 has been reported in melanoma, and the silencing of ZO-1 led to a marked reduction in the rate of cell invasion (66). Thus, the identified association between a decrease in claudin-1 and ZO-1 expression with impaired cell invasion and migration in the present study appears to be consistent with these studies.

In conclusion, the silencing of Gadd45β significantly decreased the viability, metastatic phenotypes and EMT markers of HuCCA-1 cells, suggesting that Gadd45β serves a function in the metastatic process in CCA. This is in accord with the IHC data of patients with CCA, in which Gadd45β expression was associated with metastasis. However, a limitation of this study was that the experiments were performed in only one CCA cell line. Further studies should be performed in more CCA cell lines with more stable knock down systems, as transient siRNA transfection has a limited gene silencing duration. The study of Gadd45β expression, function and Gadd45β-mediated signaling pathways in CCA will be important in understanding cancer biology, in which the cancer cells are dynamically evolving with accumulated mutations. Since the cancer cells must thrive in a competitive environment, a stress response protein like Gadd45β may be critical. The understanding or manipulation of Gadd45β-dependent survival signaling and EMT pathways may be beneficial as a potential therapeutic option, adjuvant to the conventional therapies for CCA.

Acknowledgements

The authors thank Professor Satitaya Sirisinha (Mahidol University, Bangkok, Thailand) for providing the HuCCA-1 cell line and the Central Instrument Facility, Center of Nanoimaging, Faculty of Science of Mahidol University. The authors express their thanks to Professor Tavan Janvilisri and Ms. Phorutai Pearngam for assistance in the study of EMT pathways. The present study was supported by Mahidol University (grant no. 18/2555).

Glossary

Abbreviations

Abbreviations:

CCA

cholangiocarcinoma

ZO-1

zona occludens protein 1

MMP

matrix metalloproteinase

EMT

epithelial-mesenchymal transition

siRNA

small interfering RNA

References

1 

Blechacz B and Gores GJ: Cholangiocarcinoma: Advances in pathogenesis, diagnosis, and treatment. Hepatology. 48:308–321. 2008. View Article : Google Scholar : PubMed/NCBI

2 

Welzel TM, McGlynn KA, Hsing AW, O'Brien TR and Pfeiffer RM: Impact of classification of hilar cholangiocarcinomas (Klatskin tumors) on the incidence of intra- and extrahepatic cholangiocarcinoma in the United States. J Natl Cancer Inst. 98:873–875. 2006. View Article : Google Scholar : PubMed/NCBI

3 

Farley DR, Weaver AL and Nagorney DM: ‘Natural history’ of unresected cholangiocarcinoma: Patient outcome after noncurative intervention. Mayo Clin Proc. 70:pp. 425–429. 1995; View Article : Google Scholar : PubMed/NCBI

4 

Anderson CD, Pinson CW, Berlin J and Chari RS: Diagnosis and treatment of cholangiocarcinoma. Oncologist. 9:43–57. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Sripa B, Brindley PJ, Mulvenna J, Laha T, Smout MJ, Mairiang E, Bethony JM and Loukas A: The tumorigenic liver fluke Opisthorchis viverrini-multiple pathways to cancer. Trends Parasitol. 28:395–407. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Sripa B, Kaewkes S, Sithithaworn P, Mairiang E, Laha T, Smout M, Pairojkul C, Bhudhisawasdi V, Tesana S, Thinkamrop B, et al: Liver fluke induces cholangiocarcinoma. PLoS Med. 4:e2012007. View Article : Google Scholar : PubMed/NCBI

7 

Weigelt B, Peterse JL and van't Veer LJ: Breast cancer metastasis: Markers and models. Nat Rev Cancer. 5:591–602. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Heerboth S, Housman G, Leary M, Longacre M, Byler S, Lapinska K, Willbanks A and Sarkar S: EMT and tumor metastasis. Clin Transl Med. 4:62015. View Article : Google Scholar : PubMed/NCBI

9 

Nieto MA: Epithelial plasticity: A common theme in embryonic and cancer cells. Science. 342:12348502013. View Article : Google Scholar : PubMed/NCBI

10 

Moreno-Bueno G, Portillo F and Cano A: Transcriptional regulation of cell polarity in EMT and cancer. Oncogene. 27:6958–6969. 2008. View Article : Google Scholar : PubMed/NCBI

11 

Peinado H, Olmeda D and Cano A: Snail, Zeb and bHLH factors in tumour progression: An alliance against the epithelial phenotype? Nat Rev Cancer. 7:415–428. 2007. View Article : Google Scholar : PubMed/NCBI

12 

Adhikary A, Chakraborty S, Mazumdar M, Ghosh S, Mukherjee S, Manna A, Mohanty S, Nakka KK, Joshi S, De A, et al: Inhibition of epithelial to mesenchymal transition by E-cadherin up-regulation via repression of slug transcription and inhibition of E-cadherin degradation: Dual role of scaffold/matrix attachment region-binding protein 1 (SMAR1) in breast cancer cells. J Biol Chem. 289:25431–25444. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Techasen A, Loilome W, Namwat N, Khuntikeo N, Puapairoj A, Jearanaikoon P, Saya H and Yongvanit P: Loss of E-cadherin promotes migration and invasion of cholangiocarcinoma cells and serves as a potential marker of metastasis. Tumour Biol. 35:8645–8652. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Mendez MG, Kojima S and Goldman RD: Vimentin induces changes in cell shape, motility, and adhesion during the epithelial to mesenchymal transition. FASEB J. 24:1838–1851. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Chaw SY, Majeed AA, Dalley AJ, Chan A, Stein S and Farah CS: Epithelial to mesenchymal transition (EMT) biomarkers-E-cadherin, beta-catenin, APC and Vimentin-in oral squamous cell carcinogenesis and transformation. Oral Oncol. 48:997–1006. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Qiu W, David D, Zhou B, Chu PG, Zhang B, Wu M, Xiao J, Han T, Zhu Z, Wang T, et al: Down-regulation of growth arrest DNA damage-inducible gene 45beta expression is associated with human hepatocellular carcinoma. Am J Pathol. 162:1961–1974. 2003. View Article : Google Scholar : PubMed/NCBI

17 

Zumbrun SD, Hoffman B and Liebermann DA: Distinct mechanisms are utilized to induce stress sensor gadd45b by different stress stimuli. J Cell Biochem. 108:1220–1231. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Liebermann DA and Hoffman B: Gadd45 in stress signaling. J Mol Signal. 3:152008. View Article : Google Scholar : PubMed/NCBI

19 

Vairapandi M, Balliet AG, Fornace AJ Jr, Hoffman B and Liebermann DA: The differentiation primary response gene MyD118, related to GADD45, encodes for a nuclear protein which interacts with PCNA and p21WAF1/CIP1. Oncogene. 12:2579–2594. 1996.PubMed/NCBI

20 

Smith ML, Ford JM, Hollander MC, Bortnick RA, Amundson SA, Seo YR, Deng CX, Hanawalt PC and Fornace AJ Jr: p53-mediated DNA repair responses to UV radiation: Studies of mouse cells lacking p53, p21, and/or gadd45 genes. Mol Cell Biol. 20:3705–3714. 2000. View Article : Google Scholar : PubMed/NCBI

21 

Jónsson ZO and Hübscher U: Proliferating cell nuclear antigen: More than a clamp for DNA polymerases. Bioessays. 19:967–975. 1997. View Article : Google Scholar : PubMed/NCBI

22 

Kelman Z and Hurwitz J: Protein-PCNA interactions: A DNA-scanning mechanism? Trends Biochem Sci. 23:236–238. 1998. View Article : Google Scholar : PubMed/NCBI

23 

Leprivier G, Rotblat B, Khan D, Jan E and Sorensen PH: Stress-mediated translational control in cancer cells. Biochim Biophys Acta. 1849:845–860. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Qiu W, Zhou B, Zou H, Liu X, Chu PG, Lopez R, Shih J, Chung C and Yen Y: Hypermethylation of growth arrest DNA damage-inducible gene 45 beta promoter in human hepatocellular carcinoma. Am J Pathol. 165:1689–1699. 2004. View Article : Google Scholar : PubMed/NCBI

25 

Wang L, Xiao X, Li D, Chi Y, Wei P, Wang Y, Ni S, Tan C, Zhou X and Du X: Abnormal expression of GADD45B in human colorectal carcinoma. J Transl Med. 10:2152012. View Article : Google Scholar : PubMed/NCBI

26 

Meir T, Dror R, Yu X, Qian J, Simon I, Pe'er J and Chowers I: Molecular characteristics of liver metastases from uveal melanoma. Invest Ophthalmol Vis Sci. 48:4890–4896. 2007. View Article : Google Scholar : PubMed/NCBI

27 

Inowa T, Hishikawa K, Matsuzaki Y, Isagawa T, Takeuchi T, Aburatani H, Kitamura T and Fujita T: GADD45β determines chemoresistance and invasive growth of side population cells of human embryonic carcinoma. Stem Cells Int. 2010:7829672010. View Article : Google Scholar : PubMed/NCBI

28 

Sirisinha S, Tengchaisri T, Boonpucknavig S, Prempracha N, Ratanarapee S and Pausawasdi A: Establishment and characterization of a cholangiocarcinoma cell line from a Thai patient with intrahepatic bile duct cancer. Asian Pac J Allergy Immunol. 9:153–157. 1991.PubMed/NCBI

29 

Pongcharoen P, Jinawath A and Tohtong R: Silencing of CD44 by siRNA suppressed invasion, migration and adhesion to matrix, but not secretion of MMPs, of cholangiocarcinoma cells. Clin Exp Metastasis. 28:827–839. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Schneider CA, Rasband WS and Eliceiri KW: NIH Image to ImageJ: 25 years of image analysis. Nat Methods. 9:671–675. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Li YH and Zhu C: A modified Boyden chamber assay for tumor cell transendothelial migration in vitro. Clin Exp Metastasis. 17:423–429. 1999. View Article : Google Scholar : PubMed/NCBI

32 

Uhlen M, Fagerberg L, Hallström BM, Lindskog C, Oksvold P, Mardinoglu A, Sivertsson Å, Kampf C, Sjöstedt E, Asplund A, et al: Proteomics. Tissue-based map of the human proteome. Science. 347:12604192015. View Article : Google Scholar : PubMed/NCBI

33 

Kerr JF, Wyllie AH and Currie AR: Apoptosis: A basic biological phenomenon with wide-ranging implications in tissue kinetics. Br J Cancer. 26:239–257. 1972. View Article : Google Scholar : PubMed/NCBI

34 

Tewari M, Quan LT, O'Rourke K, Desnoyers S, Zeng Z, Beidler DR, Poirier GG, Salvesen GS and Dixit VM: Yama/CPP32 beta, a mammalian homolog of CED-3, is a CrmA-inhibitable protease that cleaves the death substrate poly(ADP-ribose) polymerase. Cell. 81:801–809. 1995. View Article : Google Scholar : PubMed/NCBI

35 

Kumar A, Rajendran V, Sethumadhavan R and Purohit R: Akt kinase pathway: A leading target in cancer research. ScientificWorldJournal. 2013:7561342013. View Article : Google Scholar : PubMed/NCBI

36 

Nebreda AR and Porras A: p38 MAP kinases: Beyond the stress response. Trends Biochem Sci. 25:257–260. 2000. View Article : Google Scholar : PubMed/NCBI

37 

Kyriakis JM and Avruch J: Mammalian mitogen-activated protein kinase signal transduction pathways activated by stress and inflammation. Physiol Rev. 81:807–869. 2001. View Article : Google Scholar : PubMed/NCBI

38 

Erhardt P, Schremser EJ and Cooper GM: B-Raf inhibits programmed cell death downstream of cytochrome c release from mitochondria by activating the MEK/Erk pathway. Mol Cell Biol. 19:5308–5315. 1999. View Article : Google Scholar : PubMed/NCBI

39 

Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

40 

Talmadge JE and Fidler IJ: AACR centennial series: The biology of cancer metastasis: Historical perspective. Cancer Res. 70:5649–5669. 2010. View Article : Google Scholar : PubMed/NCBI

41 

Mehlen P and Puisieux A: Metastasis: A question of life or death. Nat Rev Cancer. 6:449–458. 2006. View Article : Google Scholar : PubMed/NCBI

42 

Davies KJ: The complex interaction of matrix metalloproteinases in the migration of cancer cells through breast tissue Stroma. Int J Breast Cancer. 2014:8390942014. View Article : Google Scholar : PubMed/NCBI

43 

Roomi MW, Monterrey JC, Kalinovsky T, Rath M and Niedzwiecki A: Patterns of MMP-2 and MMP-9 expression in human cancer cell lines. Oncol Rep. 21:1323–1333. 2009.PubMed/NCBI

44 

Micalizzi DS, Farabaugh SM and Ford HL: Epithelial-mesenchymal transition in cancer: Parallels between normal development and tumor progression. J Mammary Gland Biol Neoplasia. 15:117–134. 2010. View Article : Google Scholar : PubMed/NCBI

45 

Powell DR, Blasky AJ, Britt SG and Artinger KB: Riding the crest of the wave: Parallels between the neural crest and cancer in epithelial-to-mesenchymal transition and migration. Wiley Interdiscip Rev Syst Biol Med. 5:511–522. 2013. View Article : Google Scholar : PubMed/NCBI

46 

Wallerand H, Cai Y, Wainberg ZA, Garraway I, Lascombe I, Nicolle G, Thiery JP, Bittard H, Radvanyi F and Reiter RR: Phospho-Akt pathway activation and inhibition depends on N-cadherin or phospho-EGFR expression in invasive human bladder cancer cell lines. Urol Oncol. 28:180–188. 2010. View Article : Google Scholar : PubMed/NCBI

47 

Furuse M, Fujita K, Hiiragi T, Fujimoto K and Tsukita S: Claudin-1 and −2: Novel integral membrane proteins localizing at tight junctions with no sequence similarity to occludin. J Cell Biol. 141:1539–1550. 1998. View Article : Google Scholar : PubMed/NCBI

48 

Stevenson BR, Siliciano JD, Mooseker MS and Goodenough DA: Identification of ZO-1: A high molecular weight polypeptide associated with the tight junction (zonula occludens) in a variety of epithelia. J Cell Biol. 103:755–766. 1986. View Article : Google Scholar : PubMed/NCBI

49 

Rørth P: Collective cell migration. Annu Rev Cell Dev Biol. 25:407–429. 2009. View Article : Google Scholar : PubMed/NCBI

50 

Gupta M, Gupta SK, Balliet AG, Hollander MC, Fornace AJ, Hoffman B and Liebermann DA: Hematopoietic cells from Gadd45a- and Gadd45b-deficient mice are sensitized to genotoxic-stress-induced apoptosis. Oncogene. 24:7170–7179. 2005. View Article : Google Scholar : PubMed/NCBI

51 

De Smaele E, Zazzeroni F, Papa S, Nguyen DU, Jin R, Jones J, Cong R and Franzoso G: Induction of gadd45beta by NF-kappaB downregulates pro-apoptotic JNK signalling. Nature. 414:308–313. 2001. View Article : Google Scholar : PubMed/NCBI

52 

Zazzeroni F, Papa S, Algeciras-Schimnich A, Alvarez K, Melis T, Bubici C, Majewski N, Hay N, De Smaele E, Peter ME and Franzoso G: Gadd45 beta mediates the protective effects of CD40 costimulation against Fas-induced apoptosis. Blood. 102:3270–3279. 2003. View Article : Google Scholar : PubMed/NCBI

53 

Yothaisong S, Dokduang H, Techasen A, Namwat N, Yongvanit P, Bhudhisawasdi V, Puapairoj A, Riggins GJ and Loilome W: Increased activation of PI3K/Akt signaling pathway is associated with cholangiocarcinoma metastasis and PI3K/mTOR inhibition presents a possible therapeutic strategy. Tumour Biol. 34:3637–3648. 2013. View Article : Google Scholar : PubMed/NCBI

54 

Salerno DM, Tront JS, Hoffman B and Liebermann DA: Gadd45a and Gadd45b modulate innate immune functions of granulocytes and macrophages by differential regulation of p38 and JNK signaling. J Cell Physiol. 227:3613–3620. 2012. View Article : Google Scholar : PubMed/NCBI

55 

Kodama S and Negishi M: Pregnane X receptor PXR activates the GADD45beta gene, eliciting the p38 MAPK signal and cell migration. J Biol Chem. 286:3570–3578. 2011. View Article : Google Scholar : PubMed/NCBI

56 

Medici D, Hay ED and Olsen BR: Snail and Slug promote epithelial-mesenchymal transition through beta-catenin-T-cell factor-4-dependent expression of transforming growth factor-beta3. Mol Biol Cell. 19:4875–4887. 2008. View Article : Google Scholar : PubMed/NCBI

57 

Vuoriluoto K, Haugen H, Kiviluoto S, Mpindi JP, Nevo J, Gjerdrum C, Tiron C, Lorens JB and Ivaska J: Vimentin regulates EMT induction by Slug and oncogenic H-Ras and migration by governing Axl expression in breast cancer. Oncogene. 30:1436–1448. 2011. View Article : Google Scholar : PubMed/NCBI

58 

Kim JY, Kim YM, Yang CH, Cho SK, Lee JW and Cho M: Functional regulation of Slug/Snail2 is dependent on GSK-3β-mediated phosphorylation. FEBS J. 279:2929–2939. 2012. View Article : Google Scholar : PubMed/NCBI

59 

Cross DA, Alessi DR, Vandenheede JR, McDowell HE, Hundal HS and Cohen P: The inhibition of glycogen synthase kinase-3 by insulin or insulin-like growth factor 1 in the rat skeletal muscle cell line L6 is blocked by wortmannin, but not by rapamycin: Evidence that wortmannin blocks activation of the mitogen-activated protein kinase pathway in L6 cells between Ras and Raf. Biochem J. 303:21–26. 1994. View Article : Google Scholar : PubMed/NCBI

60 

Bezdekova M, Brychtova S, Sedlakova E, Langova K, Brychta T and Belej K: Analysis of Snail-1, E-cadherin and claudin-1 expression in colorectal adenomas and carcinomas. Int J Mol Sci. 13:1632–1643. 2012. View Article : Google Scholar : PubMed/NCBI

61 

Stebbing J, Filipovic A and Giamas G: Claudin-1 as a promoter of EMT in hepatocellular carcinoma. Oncogene. 32:4871–4872. 2013. View Article : Google Scholar : PubMed/NCBI

62 

Oliveira SS and Morgado-Diaz JA: Claudins: Multifunctional players in epithelial tight junctions and their role in cancer. Cell Mol Life Sci. 64:17–28. 2007. View Article : Google Scholar : PubMed/NCBI

63 

Kwon MJ: Emerging roles of claudins in human cancer. Int J Mol Sci. 14:18148–18180. 2013. View Article : Google Scholar : PubMed/NCBI

64 

Bauer H, Zweimueller-Mayer J, Steinbacher P, Lametschwandtner A and Bauer HC: The dual role of zonula occludens (ZO) proteins. J Biomed Biotechnol. 2010:4025932010. View Article : Google Scholar : PubMed/NCBI

65 

Gottardi CJ, Arpin M, Fanning AS and Louvard D: The junction-associated protein, zonula occludens-1, localizes to the nucleus before the maturation and during the remodeling of cell-cell contacts. Proc Natl Acad Sci USA. 93:pp. 10779–10784. 1996; View Article : Google Scholar : PubMed/NCBI

66 

Smalley KS, Brafford P, Haass NK, Brandner JM, Brown E and Herlyn M: Up-regulated expression of zonula occludens protein-1 in human melanoma associates with N-cadherin and contributes to invasion and adhesion. Am J Pathol. 166:1541–1554. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2018
Volume 15 Issue 3

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Myint KZ, Kongpracha P, Rattanasinganchan P, Leelawat K, Moolthiya P, Chaiyabutr K and Tohtong R: Gadd45β silencing impaired viability and metastatic phenotypes in cholangiocarcinoma cells by modulating the EMT pathway. Oncol Lett 15: 3031-3041, 2018
APA
Myint, K.Z., Kongpracha, P., Rattanasinganchan, P., Leelawat, K., Moolthiya, P., Chaiyabutr, K., & Tohtong, R. (2018). Gadd45β silencing impaired viability and metastatic phenotypes in cholangiocarcinoma cells by modulating the EMT pathway. Oncology Letters, 15, 3031-3041. https://doi.org/10.3892/ol.2017.7706
MLA
Myint, K. Z., Kongpracha, P., Rattanasinganchan, P., Leelawat, K., Moolthiya, P., Chaiyabutr, K., Tohtong, R."Gadd45β silencing impaired viability and metastatic phenotypes in cholangiocarcinoma cells by modulating the EMT pathway". Oncology Letters 15.3 (2018): 3031-3041.
Chicago
Myint, K. Z., Kongpracha, P., Rattanasinganchan, P., Leelawat, K., Moolthiya, P., Chaiyabutr, K., Tohtong, R."Gadd45β silencing impaired viability and metastatic phenotypes in cholangiocarcinoma cells by modulating the EMT pathway". Oncology Letters 15, no. 3 (2018): 3031-3041. https://doi.org/10.3892/ol.2017.7706