Open Access

Conversion of epithelial-to-mesenchymal transition to mesenchymal-to-epithelial transition is mediated by oxygen concentration in pancreatic cancer cells

Retraction in: /10.3892/ol.2022.13227

  • Authors:
    • Shuo Chen
    • Xi Chen
    • Wei Li
    • Tao Shan
    • Wan Run Lin
    • Jiancang Ma
    • Xijuan Cui
    • Wenbin Yang
    • Gang Cao
    • Yiming Li
    • Li Wang
    • Ya'an Kang
  • View Affiliations

  • Published online on: March 8, 2018     https://doi.org/10.3892/ol.2018.8219
  • Pages: 7144-7152
  • Copyright: © Chen 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

Tumor metastasis is accompanied by a two‑stage process of epithelial-to-mesenchymal transition (EMT) and mesenchymal-to-epithelial transition (MET). Currently, the exact mechanisms underlying EMT‑MET conversion are unclear. In the present study, the mechanisms by which primary sites (hypoxic) and homing sites (normoxic or hyperoxic) participate in EMT‑MET conversion were evaluated. Pancreatic cancer cells were grown under different oxygenation conditions. Cell morphology and epithelial (E)‑cadherin and vimentin expression were examined. Transwell chambers were used to examine tumor invasiveness, and scratch assays were performed to examine cell migration. Reverse transcription‑polymerase chain reaction and western blot analysis were used to quantitate the mRNA and protein expression of E‑cadherin, vimentin, Snail and hypoxia‑inducible factor (HIF)‑1α. BxPc‑3 and Panc‑1 cells grown under hypoxic conditions demonstrated increased partial EMT, reduced E‑cadherin expression, and increased vimentin expression, compared with cells grown under normoxic or hyperoxic conditions. Cells grown under hypoxic conditions also indicated increased migration and invasiveness. HIF‑1α mRNA and protein expression was increased in cells grown under hypoxic conditions. These changes were reversed when a specific inhibitor of the HIF‑1α receptor was used to block HIF‑1α signaling. Differences in oxygen concentration at primary sites and homing sites are important in the EMT‑MET process, and the underlying mechanism may involve HIF-1α-Snail signaling.

Introduction

An accurate understanding of the mechanism underlying pancreatic cancer metastasis is necessary for effective therapy (1). Tumor metastasis refers to a multi-step process of tumor cell migration from the primary site to distant sites (2). These steps include invasion, intravasation, migration, extravasation and colonization. Epithelial-to-mesenchymal transition (EMT) serves an important role in promoting tumor metastasis (2); however, EMT may be reversible, as metastatic lesions and primary lesions in the tumor tissue are structurally similar. Therefore, a two-step tumor migration theory has been proposed in which primary epithelial tumors initially undergo EMT for invasion (2). Following these metastatic tumor cells arriving at distant sites, they undergo a mesenchymal-to-epithelial transition (MET), reversing the conversion into metastatic lesions (3). Currently, the temporal mechanisms regulating EMT-MET conversion are unclear.

Standard cell and tissue culture pO2 and atmospheric pO2 are 160 mmHg, whilst the pO2 inside the body and various organs is below this value. For example, pO2 is 24 mmHg in the brain, 24 mmHg in the liver, 66 mmHg in the spleen, and 25 mmHg in the kidney (4). During oncogenesis and development of solid tumors, the microenvironment is in a state of significant hypoxia (5). Hyperoxia is used to describe am increase in oxygen or partial oxygen critical values, whereas hypoxia describes their decrease (4). Hypoxia induces tumor EMT and promotes metastasis through the hypoxia-inducible factor (HIF)-1α pathway (6). High oxygen pressure may inhibit the growth of breast cancer cells and gliomas (7,8), and cells considered to be adapted to hypoxia cannot adapt to sudden environmental changes. Moen et al (9) indicated that a hyperoxic environment could change the plasticity of breast cancer cells, causing a conversion from EMT to MET and decreasing invasiveness. Hypoxia-induced EMT in pancreatic cancer involves a number of underlying mechanisms (10). HIF-1α expression under hypoxia in CD133+ pancreatic cancer cells is correlated with tumor cell migration through EMT gene expression (11). Chen et al (12) demonstrated that hypoxia induced EMT in pancreatic cancer cells though TWIST interaction with Ring1B and EZH2 in vitro and in nude mice. Lei et al (13) indicated that hedgehog signaling regulates hypoxia-induced EMT and invasion in pancreatic cancer cells in a ligand-independent manner.

The oxygen environment may be a dynamic switch for plasticity regulation in cells (14), but whether this can be used to explain the secondary mechanism underlying tumor metastasis remains unknown. In the present study, in vitro hypoxic simulation and moderate hyperoxic environments were used to investigate the effect of oxygen concentration on EMT and MET phenotypes in tumor cells. The results provided insights into the mechanisms involved in pancreatic cancer cell metastasis, thereby providing a basis for novel treatment.

Materials and methods

Materials

RIPA cracking liquid kits were obtained from Beyotime Institute of Biotechnology (Shanghai, China). Dulbecco's modified Eagle's medium (DMEM) and fetal calf serum were purchased from GE Healthcare Life Sciences (Logan, UT, USA). Transwell chambers were purchased from Merck KGaA (Darmstadt, Germany). Matrigel and One-Step Reverse transcription-polymerase chain reaction (RT-PCR) kits were obtained from BD Biosciences (Franklin Lakes, NJ, USA). Epithelial (E)-cadherin (cat. no. sc-71007), vimentin (cat. no. sc-80975), HIF-1α (cat. no. sc-13515), Snail (cat. no. sc-393172) and β-actin (cat. no. sc-517582) antibodies were purchased from Santa Cruz Biotechnology, Inc. (Dallas, TX, USA). The HIF-1α-specific blocker, YC-1, was purchased from Sigma (Shanghai, China). Human pancreatic cancer cell lines, BxPc-3 and Panc-1, were obtained from the American Type Culture Collection (Manassas, VA, USA).

Cell cultures and treatments

BxPc-3 and Panc-1 cells were maintained in DMEM (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with penicillin (100 U/ml), streptomycin (100 µg/ml), 0.1 mM nonessential amino acids, 0.2 mM glutamine, 1 mM pyruvate and 10% heat-inactivated fetal bovine serum. Cells grown to 80% confluency were exposed to hypoxia (5% oxygen), normoxia (21% oxygen) and moderate hyperoxia (30% oxygen), and incubated in 5% CO2 humidified atmosphere at 37°C for two days. Cells were incubated in 5% CO2 DMEM without serum for at 37°C one day prior to harvest for use in further experiments. In the invasion and migration experiments, cells were cultured in DMEM without fetal bovine serum.

Cell proliferation assay

Cell proliferation was assessed by the MTT assay. A 96-well plate was seeded with 5×103 cells, 200 µl DMEM was added to each well, and the plate was incubated overnight at 37°C. The cells were cultured for 24 h following transfection, and then MTT reagent (QiYi Biological Technology Co., Ltd., Shanghai, China) (5 mg/ml) was added. The supernatant was discarded following 4 h of incubation. Then, 150 µl dimethyl sulfoxide was added to each well and the absorbance (570 nm) was measured. The assay was repeated three times.

Immunofluorescence

For immunofluorescence experiments, Panc-1 cells were cultured onto glass cover slips inside 6-well plates. Following treatment, cells were rinsed with phosphate buffered saline (PBS) and fixed in 4% formaldehyde in PBS for 15 min at 25°C. Thereafter, cells were treated with 0.2% Triton X-100 (Beijing SolarBio Science & Technology Co., Ltd., Beijing, China) in PBS for an additional 15 min at 4°C. Following blocking with 1% bovine serum albumin (QiYi Biological Technology Co., Ltd.) in PBS for 1 h at 25°C, cells were incubated for 2 h at room temperature (RT) with the primary antibodies against E-cadherin and vimentin (1:100 dilution in blocking solution). Following three washes with PBS, cells were incubated with a fluorescein isothiocyanate-goat anti-rabbit IgG (1:100; GB22303; Boster Bioengineering Co., Ltd.) for 30 min at 37°C. Finally, slides were incubated with 1 µg/ml DAPI (Sigma-Aldrich; Merck KGaA) for 10 min at 25°C, washed with PBS three times, and mounted for visualization using an inverted fluorescent microscope (magnification, ×400; model AXIO; Zeiss AG, Oberkochen, Germany) with appropriate emission/excitation parameters.

Cell migration experiments

Cell migration was evaluated in a scratch test. BxPc-3 and Panc-1 cells (10×105) were seeded into a 24-well plate containing 1.5 ml DMEM in each well. The cells were grown to a confluent layer (48 h) and then a scratch was produced in each well by using a pipette tip. Subsequently, the cells were washed gently with PBS three times. An image was taken at 0 h. The cells were then exposed to hypoxia, normoxia or moderate hyperoxia at 37°C in a 5% CO2 atmosphere and images were taken again following 24 h. The 24 h time point was selected to decrease the potential impact of proliferation on the closing of the scratch. NIH Image Pro Plus 5.0 image analysis software (NIH, Bethesda, MD, USA) was used to standardize and present the results. Results were expressed as a migration index-that is, the distance migrated by normoxia or moderate hyperoxia relative to the distance migrated by hypoxia treated cells. Three independent experiments were performed.

Cell invasion experiments

Cell invasion was examined using Transwell assays. Following incubation for 48 h, 3×104 cells were transferred to the top of the Matrigel-coated invasion chambers (BD Biosciences) in serum-free DMEM. DMEM containing 10% fetal bovine serum was added to the lower chamber. Following 24 h, the non-invading cells were removed and the invading cells were fixed using 95% ethanol for 30 min at 25°C. Cells were then stained with 0.1% crystal violet for 5 min at 25°C and images were captured at ×100 magnification under an inverted phase contrast microscope (Olympus CKX31/41; Olympus Corporation, Tokyo, Japan). Three independent experiments were performed.

RT-PCR

Total RNA was extracted from the cells using TRIzol® reagent (Thermo Fisher Scientific, Inc.). A total of 2 µg RNA was reversed transcribed into first-strand cDNA using the RevertAid First Strand cDNA Synthesis kit (Thermo Fisher Scientific, Inc.). PCR primer sequences were as follows: E-cadherin forward, 5′-CAATGGTGTCCATGTGAACA-3′ and reverse, 5′-CCTCCTACCCTCCTGTTCG-3′; vimentin forward, 5′-CGCTTCGCCAACTACAT-3′ and reverse, 5′-AGGGCATCCACTTCACAG-3′; β-actin forward, 5′-ATCGTGCGTGACATTAAGGAGAAG-3′ and reverse, 5′-AGGAAGGAAGGCTGGAAGAGTG-3′; Snail1 forward, 5′-AAGGATCTCCAGGCTCGAAAG-3′ and reverse, 5′-GCTTCGGATGTGCATCTTGA-3′. cDNA synthesis was performed at 42°C for 1 h, followed by denaturation at 94°C for 5 min. The PCR procedure consisted of 22 cycles with the following conditions: 94°C for 30 sec, 55°C for 30 sec and 72°C for 30 sec. Following the last cycle, the reaction was incubated at 72°C for 10 min. The housekeeping gene, β-actin, was used as an internal reference.

Western blot analysis

A total of 5×105 cells in the logarithmic growth phase were added to 0.5 ml pre-chilled cell lysis buffer (Beijing Dingguo Changsheng Biotechnology, Co., Ltd., Beijing, China) and incubated on ice for 30 min. Following centrifugation, the supernatant was collected and protein concentrations were measured using a BCA assay. The proteins (50 µg) were separated by 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis and blotted onto a nitrocellulose membrane by semi-dry transfer. Membranes were blocked with TBST containing 5% skim milk for 1 h at 37°C and incubated with the primary antibodies against E-cadherin, vimentin, HIF-1α, Snail and β-actin (dilution, 1:2,000) for 12 h at 4°C. On the following day, the membranes were incubated at RT for 2 h with a horseradish peroxidase-conjugated secondary monoclonal anti mouse IgG antibody (1:2,000; cat. no. sc-2005; Santa Cruz Biotechnology, Inc., Dallas, TX, USA). An enhanced chemiluminescence kit (GE Healthcare Life Sciences) was used for staining.

Statistical analysis

Statistical analysis was performed using SPSS (version 13.0; SPSS, Inc., Chicago, IL, USA). Each experiment was performed at least three times. The data are expressed as mean ± standard error of the mean, and analyzed using Student's t-test or a one-way analysis of variance followed by a Bonferroni post hoc test, where P<0.05 (two-tailed) was considered to indicate a statistically significant difference.

Results

Effects of different oxygen concentrations on cell proliferation

The MTT method was used to investigate cell viability changes during hypoxia, normoxia and moderate hyperoxia. Compared with the hypoxia and normoxia groups, cells exposed to moderate hyperoxia demonstrated no significant differences in proliferation or cell death (P>0.05), indicating that the oxygen concentration used in the moderate hyperoxia group was suitable for subsequent experiments (Fig. 1).

Effects of different oxygen concentrations on partial EMT

A light microscope was used to observe cell morphology and examine whether hypoxia could induce EMT in cancer cells. Following hypoxic treatment, the cancer cells indicated a change in morphology from a round to spindle or long-spindle shape, with a disorderly, non-directional and radial appearance (Fig. 2A). Laser confocal microscopy was used to examine the fluorescence intensity in Panc-1 cells. Cancer cells grown in a hypoxic environment demonstrated higher vimentin fluorescence and lower E-cadherin fluorescence, compared with cells in normoxia and hyperoxia groups (Fig. 2B). Western blot analysis results indicated that the protein expression of vimentin in the hypoxia group was higher than in the normoxia and hyperoxia groups. E-cadherin protein expression was lower in the hypoxia group than in the normoxia and hyperoxia groups (Fig. 2C and D). This indicated that hypoxia can increase partial EMT changes in pancreatic cancer cells, whilst normoxia or hyperoxia can reverse the conversion of EMT to MET.

Changes in the migration capacity of pancreatic cancer cells in different oxygen concentrations

Scratch assays were conducted to verify whether hypoxia-induced EMT in pancreatic cells could promote metastasis. Following treatment of cells for 48 h, Panc-1 and BxPc-3 cells exposed to normoxia or hyperoxia demonstrated reduced migration capacity, compared with hypoxia-treated cells (Fig. 3). The migration index of normoxia or moderate hyperoxia treated cells was significantly (P<0.05) reduced, compared with hypoxia treated cells. This result demonstrated that hypoxia induces EMT transformation in pancreatic cancer cells, and it may also promote tumor metastasis; whilst, exposure to normoxia or hyperoxia causes the reversal of EMT and reduction in cell migration ability.

Changes in the invasive capacity of pancreatic cancer cells in different oxygen concentrations

Transwell experiments were carried out to confirm whether hypoxia-induced EMT transformation in pancreatic cancer cells promotes invasiveness. The number of cells penetrating the membrane significantly (P<0.05) increased under hypoxic conditions, compared with normoxic or hyperoxic conditions (Fig. 4). This indicated that tumor invasiveness is increases following hypoxia-induced EMT transformation in pancreatic cancer cells.

Effects of oxygen concentration on Snail mRNA and protein expression in pancreatic cancer cells

Snail is an upstream transcription factor that regulates the expression of the EMT markers, E-cadherin and vimentin (15). To confirm whether hypoxia affects Snail expression in pancreatic cancer cells, RT-PCR and western blot analysis were conducted to quantify Snail mRNA and protein expression. Snail mRNA and protein expression were significantly higher in hypoxic conditions, compared with that in normoxic and hyperoxic conditions (P<0.05; Fig. 5). These results indicated that hypoxia-induced upregulation of Snail expression is a key factor in EMT induction.

Hypoxia regulates Snail through HIF-1α

HIF-1α serves an important role as a transcriptional regulator in tumor EMT (16,17). Western blot analysis was used to evaluate HIF-1α expression to confirm whether Snail upregulation in cancer cells under hypoxic conditions is associated with HIF-1α. Fig. 6A depicted that HIF-1α protein expression under hypoxia was significantly higher than that under normoxia or hyperoxia (P<0.05). This result demonstrated that the HIF-1α pathway is active in cancer cells under hypoxia. To confirm that the HIF-1α pathway is associated with EMT in hypoxic tumor cells, cells were pre-treated with the HIF-1α-specific inhibitor, YC-1, for 24 h prior to exposure to different oxygen concentrations, and the Snail protein expression was then measured. Following treatment with YC-1, there was no difference in expression of Snail, E-cadherin and vimentin between the hypoxia, normoxia or hyperoxia groups under different oxygen environments (Fig. 6B and C). These results confirmed that the HIF-1α-Snail regulatory axis is important in inducing EMT in hypoxic pancreatic cancer cells. Correspondingly, deactivation of the HIF-1α-Snail regulatory axis under normoxia or hyperoxia may induce MET transformation.

Discussion

EMT provides cells with migration and invasion capacities, enabling cancer cells to penetrate the basement membrane and enter into circulation through angiogenesis and intravasation, thereby forming circulating tumor cells (CTCs) (18). There are three cell phenotypes that exist during the process of EMT: Epithelial phenotype (E); mesenchymal phenotype (M); and a partial EMT state (P), containing epithelial and mesenchymal features (19). E-cadherin is considered to be a guardian of the epithelial phenotype in various cell types, whilst vimentin is a canonical molecular marker of EMT events (18). The EMT core transcription factors, Snail1 and Snail2, can inhibit the transcription of E-cadherin via directly binding to E-boxes on the E-cadherin promoter (19). In addition to promoting CTC production, EMT in tumor cells can also aid in survival (20,21), whist CTCs are transformed into metastatic lesions through MET (22); however, the exact mechanisms underlying EMT-MET temporal regulation remain unclear. The present study demonstrated that the hypoxic microenvironment in primary sites promotes EMT, upregulates vimentin, and downregulates E-cadherin expression through the HIF-1α-Snail axis, resulting in increased invasiveness. Conversely, in normoxic and hyperoxic environments, these changes are reversed. These results support the hypothesis of the present study that when tumor cells adapted to an oxygen-poor environment move to an oxygen-rich homing site, hypoxic induction is lost and the activity of the HIF-1α-Snail transcriptional regulatory axis is downregulated, leading to P-to-E transition and formation of novel metastatic foci. A novel hypoxic microenvironment is then created when these metastatic lesions grow, resulting in repeated cycles.

Involvement of the MET process in metastatic sites has been postulated to resolve the contradiction of EMT involvement, since numerous metastatic lesions and their primary tumor counterparts share a similar epithelial nature. Although the role of MET in metastatic tumor formation is gradually being elucidated, the exact mechanisms underlying this process, including where and how MET takes place and how it facilitates the formation of metastases, remain largely elusive (23). Previous research has focused mainly on factors that contribute to metastasis initiation, including EGF, Wnt and Akt. Relatively few studies have examined the formation of secondary tumors (23). Another plausible mechanism has been proposed to explain the changes in cell phenotype. There are numerous signals from the activated stroma in primary tumors that promote EMT. Tumor cells that leave the primary site may revert to an epithelial state due to the absence of these EMT-inducing signals at their novel site (24). This proposed mechanism is in line with the hypothesis that indicates that differences in oxygen concentration in the microenvironment may be important in the EMT-MET process.

During the development of solid tumors, the most significant change in the microenvironment is hypoxia (5). It has been demonstrated that the hypoxic microenvironment of pancreatic cancers is closely associated with high invasiveness. Under hypoxic conditions, HIF-1α is an important regulatory factor maintaining steady-state oxygen levels in the cells (25). Hypoxia can induce EMT by causing plasticity changes in tumor cells through HIF-1α (6). Huang et al (26) determined that hypoxia upregulates the expression of Slug through HIF-1α, causing EMT and enabling cells to become highly invasive and metastatic. These results are consistent with those of the present study. A previous study indicated that hyperoxia can induce partial MET in a mouse tumorigenesis model, indicating that hyperoxia serves an important role in the MET process at the tumor homing sites (27). A highlight of the present study was the continuous observation of MET in oxygen-rich environments, indicating that differences in oxygen concentration are essential for EMT to MET conversion. Differences between the hypoxic primary sites and hyperoxic homing sites may offer a novel perspective for studies on EMT-MET conversion. Further investigation is required for an in-depth understanding of the underlying mechanisms involved.

EMT increases the migration and invasiveness of tumor cells, enabling them to penetrate through the basement membrane, and promote their intravasation to form CTCs (19). Tsuji et al (28) subcutaneously injected cells containing p12CKD2-AP1 into mice and only cells that had undergone EMT could invade into the neighboring tissues and blood vessels, demonstrating that EMT is a precursor of CTC production. Following CTCs metastasizing to distant sites and adapting to the surrounding matrix environment, they undergo MET conversion and acquire proliferative characteristics to form metastatic lesions (29). The reversible and instantaneous EMT-MET conversion requires further analysis. In-depth investigation of the production and conversion mechanisms underlying EMT in CTCs would improve the understanding of tumor oncogenesis, development and metastasis, as well as provide a basis for novel targeted tumor therapies.

To conclude, epithelial tumors require dynamic EMT-MET conversion to undergo micro-metastases, and this process is mediated by differences in oxygen concentration in the microenvironment. Improving the understanding of the molecular regulation of the dynamic EMT-MET process during tumor metastasis may provide effective strategies for eradicating tumor metastases.

Acknowledgements

Not applicable.

Funding

This project was supported by the National Natural Science Foundation of China (grant no. 81402583), Natural Science Foundation of Shaanxi Province (grant no. 2014JQ4165), Xi'an Jiaotong University Education Foundation (grant no. xjj2014077) and the Hospital Fund of the Second Affiliated Hospital of the Health Science Center, Xi'an Jiaotong University [grant nos. RC(XM)201402 and YJ(QN)201521].

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contributions

TS, SC, YK and XCh conceived and designed the experiments. TS, SC, WeL, WY, GC and WaL performed the experiments. XCu, YL, LW and JM analyzed the data.

Ethics approval and consent to participate

This article does not contain any studies with human participants or animals performed by any of the authors. Informed consent was obtained from all individual participants included in the study.

Consent for publication

This article does not contain any studies with human participants or animals performed by any of the authors. There is no need for consent for publication.

Competing interests

The authors declare that they have no competing interests.

References

1 

Shi H, Li J and Fu D: Process of hepatic metastasis from pancreatic cancer: Biology with clinical significance. J Cancer Res Clin Oncol. 142:1137–1161. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Tsai JH and Yang J: Epithelial-mesenchymal plasticity in carcinoma metastasis. Genes Dev. 27:2192–2206. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Thiery JP, Acloque H, Huang RY and Nieto MA: Epithelial-mesenchymal transitions in development and disease. Cell. 139:871–890. 2009. View Article : Google Scholar : PubMed/NCBI

4 

Welford SM and Giaccia AJ: Hypoxia and senescence: The impact of oxygenation on tumor suppression. Mol Cancer Res. 9:538–544. 2011. View Article : Google Scholar : PubMed/NCBI

5 

Ikeda Y, Hisano H, Nishikawa Y and Nagasaki Y: Targeting and treatment of tumor hypoxia by newly designed prodrug possessing high permeability in solid tumors. Mol Pharm. 13:2283–2289. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Yang SW, Zhang ZG, Hao YX, Zhao YL, Qian F, Shi Y, Li PA, Liu CY and Yu PW: HIF-1α induces the epithelial-mesenchymal transition in gastric cancer stem cells through the Snail pathway. Oncotarget. 8:9535–9545. 2017.PubMed/NCBI

7 

Sletta Yttersian K, Tveitarås MK, Lu N, Engelsen AST, Reed RK, Garmann-Johnsen A and Stuhr L: Oxygen-dependent regulation of tumor growth and metastasis in human breast cancer xenografts. PloS One. 12:e01832542017. View Article : Google Scholar : PubMed/NCBI

8 

Stuhr LE, Raa A, Oyan AM, Kalland KH, Sakariassen PO, Petersen K, Bjerkvig R and Reed RK: Hyperoxia retards growth and induces apoptosis, changes in vascular density and gene expression in transplanted gliomas in nude rats. J Neurooncol. 85:191–202. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Moen I, Øyan AM, Kalland KH, Tronstad KJ, Akslen LA, Chekenya M, Sakariassen PØ, Reed RK and Stuhr LE: Hyperoxic treatment induces mesenchymal-to-epithelial transition in a rat adenocarcinoma model. PloS One. 4:e63812009. View Article : Google Scholar : PubMed/NCBI

10 

Chang Q, Jurisica I, Do T and Hedley DW: Hypoxia predicts aggressive growth and spontaneous metastasis formation from orthotopically grown primary xenografts of human pancreatic cancer. Cancer Res. 71:3110–3120. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Maeda K, Ding Q, Yoshimitsu M, Kuwahata T, Miyazaki Y, Tsukasa K, Hayashi T, Shinchi H, Natsugoe S and Takao S: CD133 modulate HIF-1α expression under hypoxia in EMT phenotype pancreatic cancer stem-like cells. Int J Mol Sci. 17:E10252016. View Article : Google Scholar : PubMed/NCBI

12 

Chen S, Chen JZ, Zhang JQ, Chen HX, Yan ML, Huang L, Tian YF, Chen YL and Wang YD: Hypoxia induces TWIST-activated epithelial-mesenchymal transition and proliferation of pancreatic cancer cells in vitro and in nude mice. Cancer Lett. 383:73–84. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Lei J, Ma J, Ma Q, Li X, Liu H, Xu Q, Duan W, Sun Q, Xu J, Wu Z and Wu E: Hedgehog signaling regulates hypoxia induced epithelial to mesenchymal transition and invasion in pancreatic cancer cells via a ligand-independent manner. Mol Cancer. 12:662013. View Article : Google Scholar : PubMed/NCBI

14 

De Bock K, Mazzone M and Carmeliet P: Antiangiogenic therapy, hypoxia, and metastasis: Risky liaisons, or not? Nat Rev Clin Oncol. 8:393–404. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Batlle E, Sancho E, Francí C, Domínguez D, Monfar M, Baulida J and De Herreros García A: The transcription factor snail is a repressor of E-cadherin gene expression in epithelial tumour cells. Nat Cell Biol. 2:84–89. 2000. View Article : Google Scholar : PubMed/NCBI

16 

Barriga EH, Maxwell PH, Reyes AE and Mayor R: The hypoxia factor Hif-1alpha controls neural crest chemotaxis and epithelial to mesenchymal transition. J Cell Biol. 201:759–776. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Zhang W, Shi X, Peng Y, Wu M, Zhang P, Xie R, Wu Y, Yan Q, Liu S and Wang J: HIF-1α promotes epithelial-mesenchymal transition and metastasis through direct regulation of ZEB1 in colorectal cancer. PloS One. 10:e01296032015. View Article : Google Scholar : PubMed/NCBI

18 

Chen T, You Y, Jiang H and Wang ZZ: Epithelial-mesenchymal transition (EMT): A biological process in the development, stem cell differentiation and tumorigenesis. J Cell Physiol. 232:3261–3272. 2017. View Article : Google Scholar : PubMed/NCBI

19 

Chaffer CL, Juan San BP, Lim E and Weinberg RA: EMT, cell plasticity and metastasis. Cancer Metast Rev. 35:645–654. 2016. View Article : Google Scholar

20 

Pradella D, Naro C, Sette C and Ghigna C: EMT and stemness: Flexible processes tuned by alternative splicing in development and cancer progression. Mol Cancer. 16:82017. View Article : Google Scholar : PubMed/NCBI

21 

Zhou P, Li B, Liu F, Zhang M, Wang Q, Liu Y, Yao Y and Li D: The epithelial to mesenchymal transition (EMT) and cancer stem cells: Implication for treatment resistance in pancreatic cancer. Mol Cancer. 16:522017. View Article : Google Scholar : PubMed/NCBI

22 

Burdick MM, Henson KA, Delgadillo LF, Choi YE, Goetz DJ, Tees DF and Benencia F: Expression of E-selectin ligands on circulating tumor cells: Cross-regulation with cancer stem cell regulatory pathways? Front Oncol. 2:1032012. View Article : Google Scholar : PubMed/NCBI

23 

Yao D, Dai C and Peng S: Mechanism of the mesenchymal-epithelial transition and its relationship with metastatic tumor formation. Mol Cancer Res. 9:1608–1620. 2011. View Article : Google Scholar : PubMed/NCBI

24 

Serrano-Gomez SJ, Maziveyi M and Alahari SK: Regulation of epithelial-mesenchymal transition through epigenetic and post-translational modifications. Mol Cancer. 15:182016. View Article : Google Scholar : PubMed/NCBI

25 

Choudhry H and Harris AL: Advances in hypoxia-inducible factor biology. Cell Metab. 27:281–298. 2018. View Article : Google Scholar : PubMed/NCBI

26 

Huang CH, Yang WH, Chang SY, Tai SK, Tzeng CH, Kao JY, Wu KJ and Yang MH: Regulation of membrane-type 4 matrix metalloproteinase by SLUG contributes to hypoxia-mediated metastasis. Neoplasia. 11:1371–1382. 2009. View Article : Google Scholar : PubMed/NCBI

27 

Moen I and Stuhr LE: Hyperbaric oxygen therapy and cancer-a review. Target Oncol. 7:233–242. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Tsuji T, Ibaragi S, Shima K, Hu MG, Katsurano M, Sasaki A and Hu GF: Epithelial-mesenchymal transition induced by growth suppressor p12CDK2-AP1 promotes tumor cell local invasion but suppresses distant colony growth. Cancer Res. 68:10377–10386. 2008. View Article : Google Scholar : PubMed/NCBI

29 

Hong Y and Zhang Q: Phenotype of circulating tumor cell: Face-off between epithelial and mesenchymal masks. Tumour Biol. 37:5663–5674. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2018
Volume 15 Issue 5

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
Chen S, Chen X, Li W, Shan T, Lin WR, Ma J, Cui X, Yang W, Cao G, Li Y, Li Y, et al: Conversion of epithelial-to-mesenchymal transition to mesenchymal-to-epithelial transition is mediated by oxygen concentration in pancreatic cancer cells Retraction in /10.3892/ol.2022.13227. Oncol Lett 15: 7144-7152, 2018
APA
Chen, S., Chen, X., Li, W., Shan, T., Lin, W.R., Ma, J. ... Kang, Y. (2018). Conversion of epithelial-to-mesenchymal transition to mesenchymal-to-epithelial transition is mediated by oxygen concentration in pancreatic cancer cells Retraction in /10.3892/ol.2022.13227. Oncology Letters, 15, 7144-7152. https://doi.org/10.3892/ol.2018.8219
MLA
Chen, S., Chen, X., Li, W., Shan, T., Lin, W. R., Ma, J., Cui, X., Yang, W., Cao, G., Li, Y., Wang, L., Kang, Y."Conversion of epithelial-to-mesenchymal transition to mesenchymal-to-epithelial transition is mediated by oxygen concentration in pancreatic cancer cells Retraction in /10.3892/ol.2022.13227". Oncology Letters 15.5 (2018): 7144-7152.
Chicago
Chen, S., Chen, X., Li, W., Shan, T., Lin, W. R., Ma, J., Cui, X., Yang, W., Cao, G., Li, Y., Wang, L., Kang, Y."Conversion of epithelial-to-mesenchymal transition to mesenchymal-to-epithelial transition is mediated by oxygen concentration in pancreatic cancer cells Retraction in /10.3892/ol.2022.13227". Oncology Letters 15, no. 5 (2018): 7144-7152. https://doi.org/10.3892/ol.2018.8219