Open Access

Alteration in the expression of the chemotherapy resistance‑related genes in response to chronic and acute hypoxia in pancreatic cancer

  • Authors:
    • Malek Zihlif
    • Tareq Hameduh
    • Nailya Bulatova
    • Hana Hammad
  • View Affiliations

  • Published online on: October 4, 2023     https://doi.org/10.3892/br.2023.1670
  • Article Number: 88
  • Copyright: © Zihlif 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

Pancreatic cancer is currently one of the least curable types of human cancer and remains a key health problem. One of the most important characteristics of pancreatic cancer is its ability to grow under hypoxic conditions. Hypoxia is associated with resistance of cancer cells to radiotherapy and chemotherapy. It is a major contributor to pancreatic cancer genetic instability, which local and systemic resistance that may result in poor clinical outcome. Accordingly, identifying gene expression changes in cancer resistance genes that occur under hypoxic conditions may identify a new therapeutic target. The aim of the present study was to explore the association between hypoxia and resistance to chemotherapy and determine the alteration in the expression of cancer resistance‑related genes in the presence of hypoxia. Pancreatic cancer cells (PANC‑1) were exposed to 8 h hypoxic episodes (<1% oxygen) three times/week for a total of 20 episodes (chronic hypoxia) or 72 h hypoxic episodes twice/week for a total of 10 episodes (acute hypoxia). The alterations in gene expression were examined using reverse transcription‑quantitative PCR array compared with normoxic cells. Chemoresistance of hypoxic cells toward doxorubicin was assessed using MTT cell proliferation assay. Both chronic and acute hypoxia induced chemoresistance toward doxorubicin in PANC‑1 pancreatic cancer cell line. The greatest changes occurred in estrogen Receptor Alpha Gene (ESR1) and ETS Like‑1 protein (ELK1) pathways­, in nucleic transcription factor Peroxisome proliferator‑activated receptors (PPARs) and in a cell cycle inhibitor cyclin dependent kinase inhibitor 1A (CDKN1A). The present study demonstrated that exposing cells to prolonged hypoxia results in different gene expression changes involving pleotropic pathways that serve a role in inducing resistance in pancreatic cancer.

Introduction

As the number of pancreatic adenocarcinoma cases not eligible for surgery is growing (1), along with a decreased effectiveness of chemotherapy (2), an effective treatment and a novel target for the treatment of this disease are required (2). Pancreatic cancer is the seventh most common cause of death from cancer, with 338,000 new cases diagnosed in 2012 worldwide (3). This cancer is typically fatal; despite notable declines in general cancer-associated mortality, prognosis of pancreatic cancer remains remarkably poor (4,5). Overall, median survival from the time of diagnosis is 4.6 months; in patients with metastatic disease, this is 2.8-5.7 months (6). Pancreatic cancer is clinically marked by local invasion, early metastasis and resistance to standard chemotherapy (7). In the majority of cases, the disease is diagnosed at late stages, because of the absence of early signs and symptoms and lack of markers that help to identify pancreatic cancer early (2).

One key characteristic of pancreatic cancer is its ability to grow under hypoxic conditions (8). Hypoxia is associated with an imbalance between oxygen consumption of the cancer cells and limited oxygen delivery (9). Such imbalance is more notable in highly proliferating masses of tumor cells that develop faster than the vasculature providing oxygenated blood (10). Hypoxia is the low delivery of oxygen or low oxygen partial pressure (pO2). The pO2 of normal healthy and hypoxic tumor tissue is 40.00-50.00 and 0.02-35.00 mmHg, respectively. However, other investigators define hypoxia as low amounts of oxygen that normally have pO2 of 0.7-15.0 mmHg (11). A total of 50-60% of tumors show hypoxic regions. Nevertheless, there are differences between tumors in situ, degree of hypoxia and causes, resulting in complex spatial and temporal heterogeneities in tumor hypoxia (12). Depending on the empirical observation, duration and pathophysiological process involved, the hypoxia within the tumor tissue is classified as chronic and acute types (13,14). Chronic hypoxia is present in 65-86% of tumor tissues (13,14). The causes of chronic hypoxia include poor diffusion due to the large distance between the hypoxic area and the blood vessels; poor blood delivery because of structural abnormalities in blood vessels around the tumor, such as perforation, blunt ends, tortuosity, sluggish flow and poorly perfused vascular branches; pressure within tumor due to solid stress of non-fluid parts or the interstitial pressure of the fluid parts and anemia. All these causes lead to a stable type of reduced delivery of oxygen, nutrients, growth factors and prevent delivery of anti-cancer and imaging agents (13,15).

Acute hypoxia is known as perfusion-limited or ischemic hypoxia where the blood flow to the tissue is abolished. Such temporary cessation of blood flow may be caused by vessel remodeling that causes development of blood vessels plugs and shunts, tumor cell aggregates that form physical obstructions that block the flow in the blood vessels or anemia. All of these causes result in a dynamic inhibition of delivery of oxygen, blood, chemotherapeutic and imaging agents, nutrients and growth factors (13,15).

Hypoxia has been associated with resistance of cancer cells to both radiotherapy and chemotherapy (16). Oxygen concentration alters the sensitivity of chemotherapeutic agents (17) and increased hypoxia is linked with increased resistance to apoptosis induced by gemcitabine (18). Hypoxia is a major element in empowering the pancreatic cancer genetic instability that increases local and systemic drug resistance, leading to poor clinical outcome following treatment (19).

The present study aimed to investigate the association between hypoxia and genetic instability that leads to chemotherapy resistance in pancreatic cancer. Furthermore, the present study aimed to investigate the effect of the hypoxic microenvironment on the development of genetic expression alteration, which is a precursor to therapy-associated resistance (19). The present study investigated the relationship between hypoxia and chemoresistance and the alteration in expression of cancer resistance-related genes.

Materials and methods

Cell culture condition

PANC1, a human pancreatic cancer cell line, was purchased from the American Type Culture Collection (USA). PANC1 cells were cultured in DMEM high-glucose medium (EuroClone SpA), supplemented with 10% (v/v) heat-inactivated fetal bovine serum, 2 mM L-glutamine and antibiotics (100 U/ml penicillin and 100 µg/ml streptomycin; all HyClone; Cytiva). PANC1 cells were grown in 75 cm² attached-type, filter-cap culture flasks (Membrane Solutions, LLC) at 37˚C in a humidified atmosphere containing 5% CO2. All cell culture procedures were performed in sterile conditions under a class II biological safety cabinet (Lumitos AG). All materials and disposables were disinfected with 76% ethanol before use and subculturing was performed twice/week at 80-90% confluence.

Exposure to hypoxia

An anaerobic atmosphere-generating system, AnaeroGen Compact (Oxoid Ltd.; Thermo Fisher Scientific, Inc.) was used to generate hypoxic conditions. The system is composed of a tightly sealed (by means of a plastic clip) bag and gas-generating sachet. The AnaeroGen sachet decreases oxygen levels to <1% within 30 min, as previously described (20,21). After attaching to the flask, PANC-1 cells at 70% confluency were subjected to hypoxia over 5 months. Normoxic PANC-1 cells were incubated alongside hypoxic PANC-1 cells. For chronic hypoxia, PANC-1 cells were exposed to 8 h hypoxia (1% oxygen) three times/week for a total of 40 doses at 37˚C; for acute hypoxia, cells received 72 h hypoxia once/week for a total of 20 doses at 37˚C. Chronic hypoxia is diffusion-limited with short episodes of hypoxia in the body, while acute hypoxia is perfusion-limited with long episodes of hypoxia in the body (20,21). PANC-1 cells were used because of their aggressive nature and poor responsiveness to cancer drugs such as doxorubicin, cisplatin and 5-fluorouracil (22).

Cell proliferation assay

The CellTiter assay using Non-Radioactive Cell Proliferation Assay kit® (Promega Corporation) was applied according to the manufacturer's instructions to detect the anti-proliferative effects of doxorubicin on PANC-1 cells and resistance resulting from the hypoxia shots. A total of 7x103 cells was seeded into each well of a coated 96-well plate for 24 h at 37˚C (Greiner Bio-One GmbH). Both hypoxic and normoxic cells were seeded in triplicate in DMEM (without doxorubicin) for ≥24 h at 37˚C. The media were aspirated from the wells. Afterwards, doxorubicin was added to each well at descending concentrations starting with 100.000, 50, 25, 12.5, 6.25, 3.125. 1.56, 0.78, 0.39, 0.19, .095, 0.047, 0.023, 0.012, 0.06, 0.03 µM. Cells were incubated at 37˚C for 72 h. A total of 15 µl MTT reagent was added to each well. The plates were incubated at 37˚C for 4 h, after which 100 µl solubilization/stop solution was added to each well. Optical density at 570 nm wavelength was recorded 1 h later using a 96-well plate reader (Biotek ELx808™ Absorbance Microplate Reader). Measurements were performed in triplets. Results were analyzed using the GraphPad PRISM®5.0 software (GraphPad Software, Inc.; Dotmatics). The half-maximal inhibitory concentration (IC50) values, defined as the drug concentration at which 50% of cells are viable, were calculated from the logarithmic trend of the cytotoxicity graphs.

MTT proliferation assay was conducted for cells exposed to chronic hypoxia after the 10th, 20th, 30th and 40th hypoxic shots, while for cells exposed to acute hypoxia, it was conducted after 10th, 15th and 20th hypoxic shots. The MTT proliferation assay was also conducted in parallel for control PANC-1 cells incubated under normoxic condition. Although doxorubicin is not commonly used in treatment of pancreatic cancer, it has been used in many studies to illustrate the hypoxic resistance phenotype (21,23).

RNA extraction and cDNA synthesis

Total RNA was extracted from PANC-1 cells after every 10 shots of hypoxia as well as from control (normoxic) cells. All RNA samples were stored at -80˚C until cDNA synthesis. RNA was isolated using an RNeasy® Mini kit (Qiagen GmbH) following the manufacturer's instructions. RNase-free DNase (Qiagen GmbH) was used according to the manufacturer's instructions to ensure complete genomic DNA elimination. Purity of isolated RNA was determined by measuring ratio of the optical density of the samples at 260 and 280 nm. (Biotek ELx808™ Absorbance Microplate Reader).

cDNA strands were synthesized using RT2 First Strand kit (Qiagen GmbH), according to the manufacturer's instructions; aliquots containing 1 µg total RNA were used from each sample. The OD260/OD280 ratio was calculated for purity.

Reverse transcription-quantitative PCR

RNA used in cDNA synthesis was selected at the time points coinciding with the maximum resistance according to the MTT colorimetric test, namely, the 20th shot of the chronic hypoxia and the 10th shot of the acute hypoxia models. RNA of normoxic PANC-1 cells was also used for comparison. The effect of hypoxia on gene expression of PANC-1 pancreatic cancer cell line was studied using a 96-well Real time 2 Profiler PCR array (cat. no. 330231; Qiagen, GmbH) at 37˚C. In this array, 96-well plates contain primers assays for 84 genes known to respond to low oxygen concentration, in addition to 12 genes for quality control. Primers were provided by Qiagen GmbH (sequences not available). cDNA was mixed with RT2 SYBR® green master mix (Qiagen, Inc.) and nuclease-free water (Bio Basic, Inc.). Then, 20 µl mix was placed in every well and the plate was centrifuged (Hettich Holding GmbH & Co.) at 1,000 x g for 1 min to remove air bubbles at room temperature. qPCR was performed using the CFX (Bio-Rad Laboratories, Inc.) thermocycler as follows: Initial denaturation at 95˚C for 10 min, followed by 40 cycles of 95˚C for 15 sec and 60˚C for 1 min.

Fold change is calculated by using the ∆∆Cq method (24) β-actin, β-2-microglobulin, GAPDH, hypoxanthine phosphoribosyltransferase 1 and ribosomal protein lateral stalk subunit P0 (RPLP0) were used as a control for normalization.

Statistical analysis

Data were analyzed using unpaired Student's t test (two-tail distribution and equal variances) for gene expression. Data are presented as mean and SD. Each group contained ≥3 samples. The analysis was performed using RT2 Profiler PCR Array Data Analysis Webportal (sabiosciences.com/pcr/arrayanalysis.php).

Results

Effect of hypoxia on PANC-1 cell resistance to doxorubicin

The resistance to doxorubicin was used to confirm induction of the hypoxic phenotype. IC50 values of doxorubicin against PANC1 cells increased >2-fold after 20 shots of chronic hypoxia compared with the normoxic cells (Table I). IC50 was similar to that of the control after 40 shots of chronic hypoxia. Following acute hypoxia of 10 cycles, the IC50 increased to ~6-fold that of the control and was similar to that of the control after 20 cycles of acute hypoxia (Table II).

Table I

IC50 of doxorubicin in chronic hypoxic PANC-1 cells.

Table I

IC50 of doxorubicin in chronic hypoxic PANC-1 cells.

GroupMean IC50, µM
Normoxia, passage 80.29±0.01
Chronic hypoxia (10 shots), passage 150.34±0.010
Chronic hypoxia (20 shots), passage 270.70±0.05
Chronic hypoxia (40 shots), passage 420.38±0.06

[i] IC50, half-maximal inhibitory concentration.

Table II

IC50 of doxorubicin in acute hypoxic PANC-1 cells.

Table II

IC50 of doxorubicin in acute hypoxic PANC-1 cells.

GroupMean IC50, µM
Normoxia, passage 100.21±0.04
Acute hypoxia (10 shots), passage 211.40±0.15
Acute hypoxia (20 shots), passage 320.40±0.03

[i] IC50, half-maximal inhibitory concentration.

Gene expression in PANC-1 cells exposed to acute hypoxia

To investigate the effect of hypoxia on gene expression and its association with development of resistance, an arbitrary cut-off point of 2-fold was selected to show notable up- and downregulation of genes. Compared with passage-matched normoxic cells, cells exposed to 10 episodes of acute hypoxia demonstrated that 23 genes were profoundly upregulated (Table III) and three were profoundly downregulated (Table IV). Most genes were significantly upregulated, except Xeroderma pigmentosum, complementation group C (XPC), ATP binding cassette subfamily B member 1 (ABCB1), retinoid X receptor β (RXRB), Bcl-2-like protein 1 (BCL2L1) and ELK1. Of the upregulated genes, ESR1 exhibited the greatest change (8.52-fold), while ABCG2 was the most downregulated gene (-2.87-fold;). A total of eight genes involved in drug inactivation were upregulated in acute hypoxia: CYP2E1 (7.45-fold), CYP1A1 (5.16-fold), CYP1A2 (5.14-fold), SULT1E1 (4.85-fold), CYP3A4 (3.56-fold), CYP2B6 (3.06-fold), CYP3A5 (2.77-fold) and CYP2D6 (2.22-fold).

Table III

Genes profoundly upregulated in acute hypoxia PANC-1 cells.

Table III

Genes profoundly upregulated in acute hypoxia PANC-1 cells.

GeneUpregulation (fold-change)P-value
ESR18.5200.016
CYP2E17.4500.001
ABCB16.5900.209
CYP1A15.1600.005
CYP1A25.1400.044
SULT1E14.8500.405
CDKN2D4.3600.005
XPC4.1100.084
CDKN1A4.080<0.001
CYP3A43.5600.055
ERBB23.3700.012
FOS3.250<0.001
CYP2B63.0600.054
PPARA2.9600.003
CYP3A52.7700.161
NAT22.6700.043
ELK12.5900.406
BCL2L12.3800.187
PPARD2.3500.077
ARNT2.3000.058
RXRB2.2900.107
CYP2D62.2200.050
ABCC52.1400.096

Table IV

Genes profoundly downregulated in acute hypoxia PANC-1 cells.

Table IV

Genes profoundly downregulated in acute hypoxia PANC-1 cells.

GeneDownregulation (fold-change)P-value
ABCG2-2.870.000
ABCC2-2.200.000
PPARG-2.180.002
Gene expression in PANC-1 cells exposed to chronic hypoxia

Compared with passage-matched normoxic cells, in cells exposed to 20 episodes of chronic hypoxia, nine genes were profoundly upregulated (Table V) and 11 were profoundly downregulated (Table VI). All genes were significantly upregulated except CYP1A2. Although CYP1A2 upregulated 3.97 fold, it did not reach statistical significance. Peroxisome proliferator-activated receptor D (PPARD), Erb-B2 Receptor Tyrosine Kinase 4 (ERBB4) and XPC were not significantly downregulated. ESR1 demonstrated the highest upregulation and ABC demonstrated the most marked downregulation.

Table V

Genes profoundly upregulated in chronic hypoxia PANC-1 cells.

Table V

Genes profoundly upregulated in chronic hypoxia PANC-1 cells.

GeneUpregulation (fold-change)P-value
ESR14.710.003
CYP1A23.970.109
SULT1E13.90.001
CYP2C192.910.002
CYP1A12.70.002
ABCB12.630.002
CYP3A42.540.002
CDKN1A2.20.003
CYP2B62.090.002

Table VI

Genes profoundly downregulated in chronic hypoxia PANC-1 cells.

Table VI

Genes profoundly downregulated in chronic hypoxia PANC-1 cells.

GeneDownregulation (fold-change)P-value
ABCC1-2.8100.034
ELK1-2.7500.226
XPC-2.6900.912
FOS-2.6900.009
ABCG2-2.510<0.001
RARG-2.4600.078
ERBB4-2.4400.421
ABCC3-2.3900.001
CDKN1B-2.3500.046
TP53-2.1900.042
PPARD-2.1800.076

Discussion

The extent and duration of oxygenation is not consistent between tumors and even in different areas of the same tumor (25). Oxygen concentration is not constant in the same area (26,27). It is proposed that numerous fluctuations in oxygenation occur in the tumor microenvironment. These fluctuations are complex and dynamic, changing within minutes to hours. These fluctuations are known as intermittent hypoxia (25,28). Hypoxia occurs in a transient and heterogeneous manner. The tumor microenvironment exhibits frequent cycles of hypoxia and reoxygenation (29); therefore, the present study used two intermittent hypoxic models that may mimic the real cancer hypoxic microenvironment. Cells showed desensitization with continuous exposure to acute and chronic hypoxia. This may be because cancer cells adapt to hypoxia after several hypoxic episodes (21).

The increase of doxorubicin IC50 in PAN1 cells after 20 shots of chronic hypoxia and 10 shots of acute hypoxia is an unexplained phenomenon that has been reported in previous investigations (29-33). The upregulated genes in both acute and chronic hypoxia are associated with drug resistance mechanisms, namely, drug efflux, drug metabolism inactivation, drug targets and signaling transduction molecules, repair of drug-induced DNA damage and evasion of apoptosis. The present results showed that in acute hypoxia, there was a profound upregulation in genes associated with active drug efflux, namely, ABCB1 (6.59-fold) and ABCC5 (2.14-fold). This upregulation explains decreased intracellular doxorubicin concentration in resistant MCF-7 cells (30). ABCB1 gene encodes the P-glycoprotein, a broad-spectrum multidrug efflux pump that is considered to be a key contributor in the development of multidrug resistance (31,32). ABCB1 gene was upregulated 2.63 fold in chronic hypoxia. A previous study in G3361 melanoma cells showed that cells with higher expression of ABCC5 showed higher doxorubicin resistance (33). Another proposed mechanism of resistance is related to hypoxia and extracellular acidity. Such acidity has a direct effect on the activity and/or uptake of certain anticancer drugs. Drugs that are weak bases such as doxorubicin, have a greater proportion of molecules in the charged form under acidic conditions, which decreases their ability to cross the plasma membrane and to be taken up into the cell, leading to decreased activity (34,35).

A total of eight genes involved in drug inactivation were upregulated in acute hypoxia. CYP2E1 expression is correlated with increased reactive oxygen species (ROS) generation in breast cancer and other types of cells (32). ROS trigger autophagy, DNA damage, impaired protein-folding and chemoresistance in cancer cells that undergo hypoxic oxidative stress (36). ROS induce chemoresistance by stimulating P-gp function and expression in human colon cancer Caco-2 cells (37). This is consistent with the present upregulation of the ABCB1 and ABCC5 genes involved in drug efflux and also with previous data that low levels of ROS downregulate P-gp expression whereas high levels of ROS result in upregulation of P-gp in multicellular prostate tumor spheroids (38). The oxidative state induced by hypoxia followed by reoxygenation and accompanied by accumulation of ROS increases peroxisome proliferator-activated receptor (PPAR) γ coactivator 1-α (PGC-1α) that induces chemoresistance by enzymatic deactivation; PGC-1α a protein acts symbiotically with PPARA and PPARD that are upregulated as a result (39).

CYP1A1 is a factor in the aryl hydrocarbon receptor (AhR)/CYP1A1 signaling pathway which is responsible for chemoresistance in cancer cells; CYP1A1 expression is induced by AhR, a helix-loop-helix transcription factor, and can bind a number of native or foreign ligands; AhR is upregulated by hypoxia (40). Combined used of AhR antagonist and 5-flurouracil increases sensitivity to chemotherapy. On the other hand, the effect of doxorubicin was not changed, leading to a suggestion that doxorubicin acts on cancer cells independently of the mechanism involving AhR despite the fact that an AhR antagonist decreases the expression of CYP1A1(41).

CYP1A2 a is phase 1 metabolism enzyme highly expressed in human liver (42). To the best of our knowledge, there are no reported data about the role of CYP1A2 in chemoresistance. Here, CYP1A2 was upregulated in chronic hypoxia up to 3.97-fold.

CYP3A5 is highly expressed in pancreatic cancer cells. Its expression can be induced by paclitaxel or erlotinib; knocking down the CYP3A5 gene, notably increases response to drugs (43). CYP3A4 participates in chemoresistance in colon cancer stem cells (44). The interplay between CYP3A4 and P-glycoprotein as a factor that limits oral drug bioavailability is well-established, especially in the intestinal mucosa, which may reduce the drug concentration that reaches the cancer cell (45,46). P-glycoprotein can work in concert with CYP3A4 to increase drug metabolism by controlling the access of the drug to the intracellular metabolizing enzyme such as CYP1A1, CYP1A2, CYP3A4 and CYP3A5(47). Here, CYP3A4 gene was upregulated up to 3.56-fold in acute hypoxia and up to 2.54-fold in chronic hypoxia, along with ABCB1, which indicates possible interplay between CYP3A4 and P-gp in development of resistance in pancreatic cancer cells.

The present study showed that CYP2B6 gene was upregulated in acute hypoxia up to 3.06-fold and in chronic hypoxia up to 2.09-fold. CYP2B6 enzyme is associated with tamoxifen activation and mutations in CYP2B6 are associated with poor response to cyclophosphamide-based therapy in patients with breast cancer, in addition to inhibiting the anticancer effect of tamoxifen (48). Another report suggested the role of CYP2B6 polymorphism in increasing relapse after treatment with cyclophosphamide in patients with lymphoma (49,50). CYP2C19 was upregulated in chronic hypoxia up to 2.91-fold, but not in acute hypoxia. This enzyme is responsible for cyclophosphamide and ifosfamide activation. CYP2C19 polymorphism affects pharmacokinetic profiles of nelfinavir in patients with locally advanced pancreatic cancer (51). A previous study reported hypoxia has no effect on the regulation of CYP2C19 gene while other studies suggest that hypoxia downregulates this gene in a normal liver rat model (51,52), in contrast to the present results.

The present study showed that ESR1 gene was upregulated 8.52-fold in acute hipoxia and up to 4.71-fold in chronic hypoxia. The high presence of estrogen receptors in pancreatic cancer cells was firstly described by Greenway et al (53). Konduri et al (54) suggested that the ratio of ESR-β/ESR-α may predict a response to estrogen-associated therapy in the treatment of pancreatic cancer. A previous study has demonstrated that ESR1 expression in breast cancer cells is correlated with therapeutic efficacy of chemotherapy (55). Chemoresistance is associated with high expression of ESR-1 in breast cancer cells (56,57). Apoptosis-related molecules or signal pathways such as BCL-2 and p53 may be involved in ESR1-mediated chemoresistance (58).

The present study also showed that SULT1E1 was upregulated in acute hypoxia up to 4.85-fold and in chronic hypoxia up to 3.90-fold. The present data are in alignment with induction of hepatic estrogen Sulfotransferase (EST) mediated by oxidative stress in mice (59). Overexpression of SULT1E1 in MCF-7 is associated with arrested cell cycle and apoptosis (60). Karle et al (61) demonstrated elevated activity of this enzyme in MCF7 cells resistant to doxorubicin. The SULT1E1 levels are correlated with the levels of Estrogen Receptor α (ERα) in ovarian cancer cells (62).

Among genes associated with cell cycle and cell death inhibition mechanisms, the most upregulated genes in acute hypoxia were CDKN2D (4.36-fold), RXRB (2.29-fold) and CDKN1A (4.08-fold), which was also upregulated in chronic hypoxia up to 2.20-fold. CDKN2D, upregulated in acute hypoxia, encodes cyclin-dependent kinase 4 inhibitor D protein, a member of the INK4 family of cyclin-dependent kinase inhibitors. These function as cell growth regulators that control cell cycle G1 progression, which may explain slow proliferation rate in pancreatic cancer after shots of hypoxia (63). RXRB, upregulated in acute hypoxia, encodes RXRs, nucleic receptor transcription factors. These factors bind retinoid, natural and synthetic molecules structurally and/or functionally associated with vitamin A and regulate cell differentiation, proliferation and survival (64). An association has been suggested between RXRB and activation of pancreatic stellate cells that induce chemoresistance (65). RXRB heterodimers with PPAR initiate biological responses including oxidative stress response, which supports the present results (66). CDKN1A gene, upregulated in both hypoxia models, is a major inhibitor of p53-dependent apoptosis (67); expression of this gene protects cells from doxorubicin-induced apoptosis (68). High cytoplasmic expression of p21 induces resistance to cisplatin in testicular cancer cell (68).

PPAR genes were upregulated in acute hypoxia: PPARA 2.96-fold and PPARD 2.35-fold. PPARs are ligand-activated transcription factors that belong to the nuclear hormone receptor superfamily (69). Activation of PPARA leads to inhibition of apoptosis and increased oxidative stress in hepatocellular cells (70). PPARA also serves as an inflammation inducer in cancer and a factor in providing a suitable microenvironment for tumor growth. PPARA antagonist can inhibit growth in pancreatic cancer cells (70,71). PPARD is a component of the angiogenetic switch in pancreatic cancer and high expression levels are associated with tumor progression and distant metastases (72,73). In breast cancer cells, PPARD protein expression is increased in response to hypoxic and metabolic starvation; PPARD helps breast cancer cells to survive by decreasing oxidative stress and enhancing survival signaling responses (74). The present PPARD upregulation supports the aforementioned findings and the upregulation of this gene is an adaption to hypoxia.

XPC gene was upregulated in acute hypoxia up to 4.11-fold. Hypoxia and hypoxia-inducible factor 1α (HIF-1α) are actively involved in XPC regulation and XPC is associated with increased sensitivity to oxidative DNA damage (75,76). It was shown that activation of the ataxia-telangiectasia mutated (ATM) gene which is recruited to the damaged DNA through XPC prevents cisplatin-induced apoptosis via nucleotide excision repair (77). XPC is associated with chemoresistance in non-small cell lung carcinoma (NSCLC).

ErbB2 (also known as HER2 or neu) was upregulated in acute hypoxia. This gene encodes a 185 kDa transmembrane glycoprotein, which belongs to the epidermal growth factor receptor (EGFR) family (78). ErbB2 is commonly overexpressed in cancers, including pancreatic cancer (79,80).

FOS is a proto-oncogene that serves an important role in many cellular functions such as proliferation, apoptosis, angiogenesis, epidermal-mesenchymal transition (EMT), invasion and metastasis. It was shown that this gene is affected by hypoxia (81,82). In human ovarian carcinoma cells, cisplatin resistance is associated with upregulation of the c-FOS gene (82). The present upregulation in the FOS gene in acute hypoxia up to 3.25-fold is consistent with the aforementioned findings.

ELK1 acts as transcription activator (83) and was upregulated in acute hypoxia up to 2.59-fold. ELK1 mRNA expression is upregulated in chemoresistant specimens of patients with serous epithelial ovarian cancer; patients with high nuclear expression of ELK1 have significantly shorter survival time (83). One of the pathways responsible for chemoresistance in cancer cells is the Ras/Raf/MEK/ERK pathway. ERKs activate transcription factors such as ELK1 that are associated with drug resistance (84). ELK1 activation is required to induce FOS transcription after ERK stimulation; both of these genes were upregulated in the present study, hence ELK1 represents a key link between signal transduction pathways and initiation of gene transcription (85).

AhR nuclear translocator (ARNT) attaches to HIF-1α to form the HIF-1, a master regulator of oxygen homeostasis with pleiotropic effects, including inducing chemoresistance (86). ARNT overexpression is associated with drug-resistant properties of cancer cells by upregulation of MDR1, which prevents the action of many chemotherapeutic drugs such as doxorubicin and cisplatin in different types of cancer (87). These data are consistent with the present upregulation of ARNT up to 2.30-fold in acute hypoxia.

N-acetyltransferase (NAT2) is an enzyme that both activates and deactivates aryl amine, hydrazine drugs and carcinogens; here, it was upregulated up to 2.67-fold in acute hypoxia. This is consistent with a study that found significant changes in the activity and protein and mRNA expression of NAT2 in rats in high-altitude hypoxia as a result of a whole metabolism change in cancer cells (49).

Bcl2L1 is an anti-apoptotic protein that regulates the production of ROS and release of cytochrome C by mitochondria, both of which are key inducers of cell apoptosis (88). Bcl2L1 was upregulated up to 2.38-fold for acute hypoxia in the present study. A previous study found that Bcl2L1 expression confers resistance to chemotherapy-induced apoptosis resulting from treatment with cisplatin, paclitaxel, topotecan and gemcitabine in ovarian cancer cells (88).

Here, three genes were downregulated in acute hypoxia: ABCG2 by 2.87-fold, ABCC2 by 2.20-fold and PPARG by 2.18-fold. The role of PPARG in pancreatic cancer remains controversial but cellular studies have demonstrated that PPARG inactivation increases pancreatic cancer cell growth and attenuates their migration and invasive capacity (89-93). PPARG agonists could be a promising pharmacological approach for the treatment of colorectal cancer (93). Synergy has been demonstrated between PPARG ligands, a platinum-based agent in two NSCLC-derived cell lines (94). In vitro studies demonstrate that PPARG activation decreases pancreatic cancer cell growth and a number of ligands have been designed as potential drugs that can be combined with gemcitabine (95,96). Therefore, downregulation of this gene may explain increased resistance and growth in the pancreatic cancer cell line.

The genes encoding efflux pumps ABCG2 and ABCC2 propagate resistance to chemotherapy (97); the hypoxic chemoresistance effect is induced by the regulation of ABCG2 via activation of the ERK1/2/HIF1a pathway (97), which contradicts the present results. ABCG2 was also downregulated in chronic hypoxia -2.51-fold, along with ABCC1 to -2.81-fold. On the other hand, ABCC3 was downregulated -2.39-fold only in chronic hypoxia, which conflicts with the role of these pumps in the initiation of chemoresistance in pancreatic cancer (98).

By contrast with acute hypoxia, the following genes were downregulated in chronic hypoxia: ELK1 by 2.75-fold, XPC by 2.69-fold, FOS by 2.69-fold and PPARD by 2.18-fold. This suggests that the adaptive mechanism of resistance in chronic hypoxia is different from that in acute hypoxia.

Tumor suppressive gene TP53 was downregulated in chronic hypoxia -2.19-fold; this gene acts as a DNA repair tool. Although TP53 is not a clinical marker for drug resistance, studies have correlated overexpression of mutated p53 with reduced or abolished resistance to standard medications (99,100) in pancreatic cancer, and increased expression of mutated TP53 leads to chemoresistance (99).

CDKN1B was downregulated in acute hypoxia -2.35-fold; this gene encodes a protein that controls the cell cycle progression at G1 and decreases cell cycle proliferation rate and is considered as a tumor suppressor gene (100).

CDKN1B protein expression is reduced in ~60% of human cancer cases which is indicative of poor prognosis and chemotherapy resistance (101,102). Decreased expression of p27 is associated with acquired resistance to docetaxel in breast cancer cells (103), which is consistent with the present findings; further research is needed to understand the role of p27 in inducing chemoresistance in pancreatic cancer. Retinoic acid receptors (RARs) are nucleic receptor transcription factors that bind retinoids, natural and synthetic molecules structurally and/or functionally related to vitamin A, and regulate cell differentiation, proliferation and survival (103). RARG mediates the growth inhibitory response of retinoids in numerous types of cancer cells (104). In colorectal cancer, RARG knockdown results in downregulation of MDR1 and suppression of the Wnt/β-catenin pathway, leading to sensitivity to chemotherapy, suggesting that overexpression of RARG contributes to the multidrug chemoresistance of colorectal cancer cells (105). Here, RARG gene was downregulated -2.46-fold. To the best of our knowledge, no previous study has investigated the role of RARG in pancreatic cancer chemoresistance.

ERBB4 was downregulated -2.44-fold, which, along with increased cell proliferation, is in accordance with previous data indicating that the lack of HER-4 expression may increase the metastatic capacity of pancreatic cancer cells (106-108). HER-4 may also be of potential prognostic value and deserves further attention (106-108).

The present study provided evidence that exposing cells to prolonged periods of hypoxia results in different genetic expression changes. Pleotropic pathways, including ESR1 and ELK1 pathways, and nucleic transcription receptors such as CDKN1A and PPARs are involved in resistance in pancreatic cancer but more investigation of gene expression is needed to determine gene interactions.

Acknowledgements

Not applicable.

Funding

Funding: The present study was supported by Deanship of Scientific Research at The University of Jordan, Amman, Jordan (grant no. 19/2016/2256).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

TH performed the experiments. MZ conceived the study. NB interpreted data, HH designed the study. All authors have read and approved the final manuscript. TH and MZ confirm the authenticity of all the raw data.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Buanes TA: Role of surgery in pancreatic cancer. World J Gastroenterol. 23:3765–3770. 2017.PubMed/NCBI View Article : Google Scholar

2 

Oberstein PE and Olive KP: Pancreatic cancer: Why is it so hard to treat? Therap Adv Gastroenterol. 6:321–337. 2013.PubMed/NCBI View Article : Google Scholar

3 

Rawla P, Sunkara T and Gaduputi V: Epidemiology of pancreatic cancer: global trends, etiology and risk factors. World J Oncol. 10:10–27. 2019.PubMed/NCBI View Article : Google Scholar

4 

Jemal A, Siegel R, Ward E, Hao Y, Xu J and Thun MJ: Cancer statistics, 2009. CA Cancer J Clin. 59:225–249. 2009.PubMed/NCBI View Article : Google Scholar

5 

McGuigan A, Kelly P, Turkington RC, Jones C, Coleman HG and McCain RS: Pancreatic cancer: A review of clinical diagnosis, epidemiology, treatment and outcomes. World J Gastroenterol. 24:4846–4861. 2018.PubMed/NCBI View Article : Google Scholar

6 

Carrato A, Falcone A, Ducreux M, Valle JW, Parnaby A, Djazouli K, Alnwick-Allu K, Hutchings A, Palaska C and Parthenaki I: A systematic review of the burden of pancreatic cancer in Europe: Real-World Impact on survival, quality of life and costs. J Gastrointest Cancer. 46:201–211. 2015.PubMed/NCBI View Article : Google Scholar

7 

Jemal A, Bray F, Center MM, Ferlay J, Ward E and Forman D: Global cancer statistics. CA Cancer J Clin. 61:69–90. 2011.PubMed/NCBI View Article : Google Scholar

8 

Erkan M, Kurtoglu M and Kleeff J: The role of hypoxia in pancreatic cancer: A potential therapeutic target? Expert Rev Gastroenterol Hepatol. 10:301–316. 2016.PubMed/NCBI View Article : Google Scholar

9 

Vaupel P and Harrison L: Tumor hypoxia: Causative factors, compensatory mechanisms, and cellular response. Oncologist 9 Suppl. 5:S4–S9. 2004.PubMed/NCBI View Article : Google Scholar

10 

Semenza GL: Hypoxia-inducible factors in physiology and medicine. Cell. 148:399–408. 2012.PubMed/NCBI View Article : Google Scholar

11 

Vaupel P and Mayer A: Hypoxia in cancer: Significance and impact on clinical outcome. Cancer Metastasis Rev. 26:225–239. 2007.PubMed/NCBI View Article : Google Scholar

12 

Höckel M and Vaupel P: Tumor Hypoxia: Definitions and current clinical, biologic, and molecular aspects. J Natl Cancer Inst. 93:266–276. 2001.PubMed/NCBI View Article : Google Scholar

13 

Saxena K and Jolly MK: Acute vs. Chronic vs. Cyclic Hypoxia: Their differential dynamics, molecular mechanisms, and effects on tumor progression. Biomolecules. 9(339)2019.PubMed/NCBI View Article : Google Scholar

14 

Bayer C and Vaupel P: Acute versus chronic hypoxia in tumors: Controversial data concerning time frames and biological consequences. Strahlenther Onkol. 188:616–627. 2012.PubMed/NCBI View Article : Google Scholar

15 

Bayer C, Shi K, Astner ST, Maftei CA and Vaupel P: Acute versus chronic hypoxia: Why a simplified classification is simply not enough. Int J Radiat Oncol Biol Phys. 80:965–968. 2011.PubMed/NCBI View Article : Google Scholar

16 

Whipple C and Korc M: Targeting angiogenesis in pancreatic cancer: Rationale and pitfalls. Langenbecks Arch Surg. 393:901–910. 2008.PubMed/NCBI View Article : Google Scholar

17 

Littlewood TJ, Bajetta E, Nortier JW, Vercammen E and Rapoport B: Epoetin Alfa Study Group. Effects of epoetin alfa on hematologic parameters and quality of life in cancer patients receiving nonplatinum chemotherapy: Results of a randomized, double-blind, placebo-controlled trial. J Clin Oncol. 19:2865–2874. 2001.PubMed/NCBI View Article : Google Scholar

18 

Yokoi K and Fidler IJ: Hypoxia increases resistance of human pancreatic cancer cells to apoptosis induced by gemcitabine. Clin Cancer Res. 10:2299–2306. 2004.PubMed/NCBI View Article : Google Scholar

19 

Luoto KR, Kumareswaran R and Bristow RG: Tumor hypoxia as a driving force in genetic instability. Genome Integr. 4(5)2013.PubMed/NCBI View Article : Google Scholar

20 

Dewhirst MW, Kimura H, Rehmus SW, Braun RD, Papahadjopoulos D, Hong K and Secomb TW: Microvascular studies on the origins of perfusion-limited hypoxia. Br J Cancer. (Suppl 27):S247–S251. 1996.PubMed/NCBI

21 

Jarrar Y, Zihlif M, Al Bawab AQ and Sharab A: Effects of intermittent hypoxia on expression of glucose metabolism genes in MCF7 breast cancer cell line. Curr Cancer Drug Targets. 20:216–222. 2020.PubMed/NCBI View Article : Google Scholar

22 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018.PubMed/NCBI View Article : Google Scholar

23 

Chenevier-Gobeaux C, Simonneau C, Lemarechal H, Bonnefont-Rousselot D, Poiraudeau S, Rannou F, Ekindjian OG, Anract P and Borderie D: Effect of hypoxia/reoxygenation on the cytokine-induced production of nitric oxide and superoxide anion in cultured osteoarthritic synoviocytes. Osteoarthritis Cartilage. 21:874–881. 2013.PubMed/NCBI View Article : Google Scholar

24 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 25:402–408. 2001.PubMed/NCBI View Article : Google Scholar

25 

Mellor HR, Snelling S, Hall MD, Modok S, Jaffar M, Hambley TW and Callaghan R: The influence of tumour microenvironmental factors on the efficacy of cisplatin and novel platinum(IV) complexes. Biochem Pharmacol. 70:1137–1146. 2005.PubMed/NCBI View Article : Google Scholar

26 

Toffoli S and Michiels C: Intermittent hypoxia is a key regulator of cancer cell and endothelial cell interplay in tumours. FEBS J. 275:2991–3002. 2008.PubMed/NCBI View Article : Google Scholar

27 

Magagnin MG, Koritzinsky M and Wouters BG: Patterns of tumor oxygenation and their influence on the cellular hypoxic response and hypoxia-directed therapies. Drug Resist Updat. 9:185–197. 2006.PubMed/NCBI View Article : Google Scholar

28 

Hill SA and Chaplin DJ: Detection of microregional fluctuations in erythrocyte flow using laser Doppler microprobes. Adv Exp Med Biol. 388:367–371. 1996.PubMed/NCBI View Article : Google Scholar

29 

Dewhirst MW: Intermittent hypoxia furthers the rational for HypoxiaInducible Factor-1 Targeting. Cancer Res. 67:854–855. 2007.PubMed/NCBI View Article : Google Scholar

30 

Bindra RS and Glazer PM: Genetic instability and the tumor microenvironment: Towards the concept of microenvironment-induced mutagenesis. Mutat Res. 569:75–85. 2005.PubMed/NCBI View Article : Google Scholar

31 

AbuHammad S and Zihlif M: Gene expression alterations in doxorubicin resistant MCF7 breast cancer cell line. Genomics. 101:213–220. 2013.PubMed/NCBI View Article : Google Scholar

32 

Chou CW, Wang CC, Wu CP, Lin YJ, Lee YC, Cheng YW and Hsieh CH: Tumor cycling hypoxia induces chemoresistance in glioblastoma multiforme by upregulating the expression and function of ABCB1. Neuro Oncol. 14:1227–1238. 2012.PubMed/NCBI View Article : Google Scholar

33 

Hodges LM, Markova SM, Chinn LW, Gow JM, Kroetz DL, Klein TE and Altman RB: Very important pharmacogene summary: ABCB1 (MDR1, P-glycoprotein). Pharmacogenet Genomics. 21:152–161. 2011.PubMed/NCBI View Article : Google Scholar

34 

Frank NY, Margaryan A, Huang Y, Schatton T, Waaga-Gasser AM, Gasser M, Sayegh MH, Sadee W and Frank MH: ABCB5-mediated doxorubicin transport and chemoresistance in human malignant melanoma. Cancer Res. 65:4320–4333. 2005.PubMed/NCBI View Article : Google Scholar

35 

Wojtkowiak JW, Verduzco D, Schramm KJ and Gillies RJ: Drug resistance and cellular adaptation to tumor acidic pH microenvironment. Mol Pharm. 8:2032–2038. 2011.PubMed/NCBI View Article : Google Scholar

36 

Leung T, Rajendran R, Singh S, Garva R, Krstic-Demonacos M and Demonacos C: Cytochrome P450 2E1 (CYP2E1) regulates the response to oxidative stress and migration of breast cancer cells. Breast Cancer Res. 15(R107)2013.PubMed/NCBI View Article : Google Scholar

37 

Duan R, Hu N, Liu HY, Li J, Guo HF, Liu C, Liu L and Liu XD: Biphasic regulation of P-glycoprotein function and expression by NO donors in Caco-2 cells. Acta Pharmacol Sin. 33:767–774. 2012.PubMed/NCBI View Article : Google Scholar

38 

Wartenberg M, Ling FC, Muschen M, Klein F, Acker H, Gassmann M, Petrat K, Putz V, Hescheler J and Sauer H: Regulation of the multidrug resistance transporter P-glycoprotein in multicellular tumor spheroids by hypoxia-inducible factor (HIF-1) and reactive oxygen species. FASEB J. 17:503–505. 2003.PubMed/NCBI View Article : Google Scholar

39 

Kim B, Jung JW, Jung J, Han Y, Suh DH, Kim HS, Dhanasekaran DN and Song YS: PGC1α induced by reactive oxygen species contributes to chemoresistance of ovarian cancer cells. Oncotarget. 8:60299–60311. 2017.PubMed/NCBI View Article : Google Scholar

40 

Al-Dhfyan A, Alhoshani A and Korashy HM: Aryl hydrocarbon receptor/cytochrome P450 1A1 pathway mediates breast cancer stem cells expansion through PTEN inhibition and β-Catenin and Akt activation. Mol Cancer. 16(14)2017.PubMed/NCBI View Article : Google Scholar

41 

Zhao B, Degroot DE, Hayashi A, He G and Denison MS: CH223191 is a ligand-selective antagonist of the Ah (Dioxin) receptor. Toxicol Sci. 117:393–403. 2010.PubMed/NCBI View Article : Google Scholar

42 

Xie C, Pogribna M, Word B, Lyn-Cook L Jr, Lyn-Cook BD and Hammons GJ: In vitro analysis of factors influencing CYP1A2 expression as potential determinants of interindividual variation. Pharmacol Res Perspect. 5(e00299)2017.PubMed/NCBI View Article : Google Scholar

43 

Noll EM, Eisen C, Stenzinger A, Espinet E, Muckenhuber A, Klein C, Vogel V, Klaus B, Nadler W, Rosli C, et al: CYP3A5 mediates basal and acquired therapy resistance in different subtypes of pancreatic ductal adenocarcinoma. Nat Med. 22:278–287. 2016.PubMed/NCBI View Article : Google Scholar

44 

Olszewski U, Liedauer R, Ausch C, Thalhammer T and Hamilton G: Overexpression of CYP3A4 in a COLO 205 colon cancer stem cell model in vitro. Cancers (Basel). 3:1467–1479. 2011.PubMed/NCBI View Article : Google Scholar

45 

Cummins CL, Jacobsen W and Benet LZ: Unmasking the dynamic interplay between intestinal P-glycoprotein and CYP3A4. J Pharmacol Exp Ther. 300:1036–1045. 2002.PubMed/NCBI View Article : Google Scholar

46 

Eagling VA, Profit L and Back DJ: Inhibition of the CYP3A4-mediated metabolism and P-glycoprotein-mediated transport of the HIV-1 protease inhibitor saquinavir by grapefruit juice components. Br J Clin Pharmacol. 48:543–552. 1999.PubMed/NCBI View Article : Google Scholar

47 

Kivisto KT, Niemi M, Schaeffeler E, Pitkala K, Tilvis R, Fromm MF, Schwab M, Eichelbaum M and Strandberg T: Lipid-lowering response to statins is affected by CYP3A5 polymorphism. Pharmacogenetics. 14:523–525. 2004.PubMed/NCBI View Article : Google Scholar

48 

Pan ST, Li ZL, He ZX, Qiu JX and Zhou SF: Molecular mechanisms for tumour resistance to chemotherapy. Clin Exp Pharmacol Physiol. 43:723–737. 2016.PubMed/NCBI View Article : Google Scholar

49 

Bachanova V, Shanley R, Malik F, Chauhan L, Lamba V, Weisdorf DJ, Burns LJ and Lamba JK: Cytochrome P450 2B6*5 increases relapse after cyclophosphamide-containing conditioning and autologous transplantation for lymphoma. Biol Blood Marrow Transplant. 21:944–948. 2015.PubMed/NCBI View Article : Google Scholar

50 

Zembutsu H, Nakamura S, Akashi-Tanaka S, Kuwayama T, Watanabe C, Takamaru T, Takei H, Ishikawa T, Miyahara K, Matsumoto H, et al: Significant Effect of Polymorphisms in CYP2D6 on response to tamoxifen therapy for breast cancer: A prospective multicenter study. Clin Cancer Res. 23:2019–2026. 2017.PubMed/NCBI View Article : Google Scholar

51 

Kattel K, Evande R, Tan C, Mondal G, Grem JL and Mahato RI: Impact of CYP2C19 polymorphism on the pharmacokinetics of nelfinavir in patients with pancreatic cancer. Br J Clin Pharmacol. 80:267–275. 2015.PubMed/NCBI View Article : Google Scholar

52 

Li X, Wang X, Li Y, Yuan M, Zhu J, Su X, Yao X, Fan X and Duan Y: Effect of exposure to acute and chronic high-altitude hypoxia on the activity and expression of CYP1A2, CYP2D6, CYP2C9, CYP2C19 and NAT2 in rats. Pharmacology. 93:76–83. 2014.PubMed/NCBI View Article : Google Scholar

53 

Greenway B, Iqbal MJ, Johnson PJ and Williams R: Oestrogen receptor proteins in malignant and fetal pancreas. Br Med J (Clin Res Ed). 283:751–753. 1981.PubMed/NCBI View Article : Google Scholar

54 

Konduri S, Schwarz MA, Cafasso D and Schwarz RE: Androgen receptor blockade in experimental combination therapy of pancreatic cancer. J Surg Res. 142:378–386. 2007.PubMed/NCBI View Article : Google Scholar

55 

Mutoh K, Tsukahara S, Mitsuhashi J, Katayama K and Sugimoto Y: Estrogen-mediated post transcriptional down-regulation of P-glycoprotein in MDR1-transduced human breast cancer cells. Cancer Sci. 97:1198–1204. 2006.PubMed/NCBI View Article : Google Scholar

56 

Wang L, Jiang Z, Sui M, Shen J, Xu C and Fan W: The potential biomarkers in predicting pathologic response of breast cancer to three different chemotherapy regimens: A case control study. BMC Cancer. 9(226)2009.PubMed/NCBI View Article : Google Scholar

57 

Jeong JH, Jung SY, Park IH, Lee KS, Kang HS, Kim SW, Kwon Y, Kim EA, Ko KL, Nam BH, et al: Predictive factors of pathologic complete response and clinical tumor progression after preoperative chemotherapy in patients with stage II and III breast cancer. Invest New Drugs. 30:408–416. 2012.PubMed/NCBI View Article : Google Scholar

58 

Levine B, Sinha S and Kroemer G: Bcl-2 family members: Dual regulators of apoptosis and autophagy. Autophagy. 4:600–606. 2008.PubMed/NCBI View Article : Google Scholar

59 

Falany CN: Enzymology of human cytosolic sulfotransferases. FASEB J. 11:206–216. 1997.PubMed/NCBI View Article : Google Scholar

60 

Xu Y, Liu X, Guo F, Ning Y, Zhi X, Wang X, Chen S, Yin L and Li X: Effect of estrogen sulfation by SULT1E1 and PAPSS on the development of estrogen-dependent cancers. Cancer Sci. 103:1000–1009. 2012.PubMed/NCBI View Article : Google Scholar

61 

Karle JM, Cowan KH, Chisena CA and Cysyk RL: Uracil nucleotide synthesis in a human breast cancer cell line (MCF-7) and in two drug-resistant sublines that contain increased levels of enzymes of the de novo pyrimidine pathway. Mol Pharmacol. 30:136–141. 1986.PubMed/NCBI

62 

Mungenast F, Aust S, Vergote I, Vanderstichele A, Sehouli J, Braicu E, Mahner S, Castillo-Tong DC, Zeillinger R and Thalhammer T: Clinical significance of the estrogen-modifying enzymes steroid sulfatase and estrogen sulfotransferase in epithelial ovarian cancer. Oncol Lett. 13:4047–4054. 2017.PubMed/NCBI View Article : Google Scholar

63 

Tavera-Mendoza LE, Wang TT and White JH: p19INK4D and cell death. Cell Cycle. 5:596–598. 2006.PubMed/NCBI View Article : Google Scholar

64 

Polvani S, Tarocchi M, Tempesti S and Galli A: Nuclear receptors and pathogenesis of pancreatic cancer. World J Gastroenterol. 20:12062–12081. 2014.PubMed/NCBI View Article : Google Scholar

65 

Brun PJ, Wongsiriroj N and Blaner WS: Retinoids in the pancreas. Hepatobiliary Surg Nutr. 5:1–14. 2016.PubMed/NCBI View Article : Google Scholar

66 

Gartel AL and Tyner AL: The role of the cyclin-dependent kinase inhibitor p21 in apoptosis. Mol Cancer Ther. 1:639–649. 2002.PubMed/NCBI

67 

Bunz F, Hwang PM, Torrance C, Waldman T, Zhang Y, Dillehay L, Williams J, Lengauer C, Kinzler KW and Vogelstein B: Disruption of p53 in human cancer cells alters the responses to therapeutic agents. J Clin Invest. 104:263–269. 1999.PubMed/NCBI View Article : Google Scholar

68 

Koster R, di Pietro A, Timmer-Bosscha H, Gibcus JH, van den Berg A, Suurmeijer AJ, Bischoff R, Gietema JA and de Jong S: Cytoplasmic p21 expression levels determine cisplatin resistance in human testicular cancer. J Clin Invest. 120:3594–3605. 2010.PubMed/NCBI View Article : Google Scholar

69 

Tachibana K, Yamasaki D, Ishimoto K and Doi T: The role of PPARs in cancer. PPAR Res. 2008(102737)2008.PubMed/NCBI View Article : Google Scholar

70 

Hua AM: PPAR-alpha: A Novel Target in Pancreatic Cancer. ProQuest LLC, Ann Arbor, MI, 2012.

71 

Gao J, Liu Q, Xu Y, Gong X, Zhang R, Zhou C, Su Z, Jin J, Shi H, Shi J and Hou Y: PPARα induces cell apoptosis by destructing Bcl2. Oncotarget. 6:44635–44642. 2015.PubMed/NCBI View Article : Google Scholar

72 

Abdollahi A, Schwager C, Kleeff J, Esposito I, Domhan S, Peschke P, Hauser K, Hahnfeldt P, Hlatky L, Debus J, et al: Transcriptional network governing the angiogenic switch in human pancreatic cancer. Proc Natl Acad Sci USA. 104:12890–12895. 2007.PubMed/NCBI View Article : Google Scholar

73 

Gou Q, Gong X, Jin J, Shi J and Hou Y: Peroxisome proliferator-activated receptors (PPARs) are potential drug targets for cancer therapy. Oncotarget. 8:60704–60709. 2017.PubMed/NCBI View Article : Google Scholar

74 

Wang X, Wang G, Shi Y, Sun L, Gorczynski R, Li YJ, Xu Z and Spaner DE: PPAR-delta promotes survival of breast cancer cells in harsh metabolic conditions. Oncogenesis. 5(e232)2016.PubMed/NCBI View Article : Google Scholar

75 

Melis JP, Luijten M, Mullenders LH and van Steeg H: The role of XPC: Implications in cancer and oxidative DNA damage. Mutat Res. 728:107–117. 2011.PubMed/NCBI View Article : Google Scholar

76 

Rezvani HR, Mahfouf W, Ali N, Chemin C, Ged C, Kim AL, de Verneuil H, Taïeb A, Bickers DR and Mazurier F: Hypoxia-inducible factor-1α regulates the expression of nucleotide excision repair proteins in keratinocytes. Nucleic Acids Res. 38:797–809. 2010.PubMed/NCBI View Article : Google Scholar

77 

Colton SL, Xu XS, Wang YA and Wang G: The involvement of ataxia-telangiectasia mutated protein activation in nucleotide excision repair-facilitated cell survival with cisplatin treatment. J Biol Chem. 281:27117–27125. 2006.PubMed/NCBI View Article : Google Scholar

78 

Tan M and Yu D: Molecular mechanisms of erbB2-mediated breast cancer chemoresistance. Adv Exp Med Biol. 608:119–129. 2007.PubMed/NCBI View Article : Google Scholar

79 

Stoecklein NH, Luebke AM, Erbersdobler A, Knoefel WT, Schraut W, Verde PE, Stern F, Scheunemann P, Peiper M, Eisenberger CF, et al: Copy number of chromosome 17 but not HER2 amplification predicts clinical outcome of patients with pancreatic ductal adenocarcinoma. J Clin Oncol. 22:4737–4745. 2004.PubMed/NCBI View Article : Google Scholar

80 

Gusterson BA, Gelber RD, Goldhirsch A, Price KN, Save-Soderborgh J, Anbazhagan R, Styles J, Rudenstam CM, Golouh R, Reed R, et al: Prognostic importance of c-erbB-2 expression in breast cancer. International (Ludwig) breast cancer study group. J Clin Oncol. 10:1049–1056. 1992.PubMed/NCBI View Article : Google Scholar

81 

Tulchinsky E: Fos family members: Regulation, structure and role in oncogenic transformation. Histol Histopathol. 15:921–928. 2000.PubMed/NCBI View Article : Google Scholar

82 

Moorehead RA and Singh G: Influence of the proto-oncogene c-fos on cisplatin sensitivity. Biochem Pharmacol. 59:337–345. 2000.PubMed/NCBI View Article : Google Scholar

83 

Shuang T, Wang M, Zhou Y, Shi C and Wang D: NF-κB1, c-Rel, and ELK1 inhibit miR-134 expression leading to TAB1 upregulation in paclitaxel-resistant human ovarian cancer. Oncotarget. 8:24853–24868. 2017.PubMed/NCBI View Article : Google Scholar

84 

Zheng HC: The molecular mechanisms of chemoresistance in cancers. Oncotarget. 8:59950–59964. 2017.PubMed/NCBI View Article : Google Scholar

85 

Janknecht R, Ernst WH, Pingoud V and Nordheim A: Activation of ternary complex factor Elk-1 by MAP kinases. EMBO J. 12:5097–5104. 1993.PubMed/NCBI View Article : Google Scholar

86 

Doktorova H, Hrabeta J, Khalil MA and Eckschlager T: Hypoxia-induced chemoresistance in cancer cells: The role of not only HIF-1. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub. 159:166–177. 2015.PubMed/NCBI View Article : Google Scholar

87 

Chan YY, Kalpana S, Chang WC, Chang WC and Chen BK: Expression of aryl hydrocarbon receptor nuclear translocator enhances cisplatin resistance by upregulating MDR1 expression in cancer cells. Mol Pharmacol. 84:591–602. 2013.PubMed/NCBI View Article : Google Scholar

88 

Williams J, Lucas PC, Griffith KA, Choi M, Fogoros S, Hu YY and Liu JR: Expression of Bcl-xL in ovarian carcinoma is associated with chemoresistance and recurrent disease. Gynecol Oncol. 96:287–295. 2005.PubMed/NCBI View Article : Google Scholar

89 

Adrian TE, Hennig R, Friess H and Ding X: The role of PPARgamma receptors and leukotriene B(4) receptors in mediating the effects of LY293111 in pancreatic cancer. PPAR Res. 2008(827096)2008.PubMed/NCBI View Article : Google Scholar

90 

Polvani S, Tarocchi M, Tempesti S, Bencini L and Galli A: Peroxisome proliferator activated receptors at the crossroad of obesity, diabetes, and pancreatic cancer. World J Gastroenterol. 22:2441–2459. 2016.PubMed/NCBI View Article : Google Scholar

91 

Farrow B and Evers BM: Activation of PPARgamma increases PTEN expression in pancreatic cancer cells. Biochem Biophys Res Commun. 301:50–53. 2003.PubMed/NCBI View Article : Google Scholar

92 

Aires V, Brassart B, Carlier A, Scagliarini A, Mandard S, Limagne E, Solary E, Martiny L, Tarpin M and Delmas D: A role for peroxisome proliferator-activated receptor gamma in resveratrol-induced colon cancer cell apoptosis. Mol Nutr Food Res. 58:1785–1794. 2014.PubMed/NCBI View Article : Google Scholar

93 

Reddy RC, Srirangam A, Reddy K, Chen J, Gangireddy S, Kalemkerian GP, Standiford TJ and Keshamouni VG: Chemotherapeutic drugs induce PPAR-gamma expression and show sequence-specific synergy with PPAR-gamma ligands in inhibition of non-small cell lung cancer. Neoplasia. 10:597–603. 2008.PubMed/NCBI View Article : Google Scholar

94 

Chi T, Wang M, Wang X, Yang K, Xie F, Liao Z and Wei P: PPAR-γ modulators as current and potential cancer treatments. Front Oncol. 11(737776)2021.PubMed/NCBI View Article : Google Scholar

95 

Eibl G: The role of PPAR-gamma and its interaction with COX-2 in pancreatic cancer. PPAR Res. 2008(326915)2008.PubMed/NCBI View Article : Google Scholar

96 

Adamska A and Falasca M: ATP-binding cassette transporters in progression and clinical outcome of pancreatic cancer: What is the way forward? World J Gastroenterol. 24:3222–3238. 2018.PubMed/NCBI View Article : Google Scholar

97 

He X, Wang J, Wei W, Shi M, Xin B, Zhang T and Shen X: Hypoxia regulates ABCG2 activity through the activivation of ERK1/2/HIF-1α and contributes to chemoresistance in pancreatic cancer cells. Cancer Biol Ther. 17:188–198. 2016.PubMed/NCBI View Article : Google Scholar

98 

Konig J, Hartel M, Nies AT, Martignoni ME, Guo J, Buchler MW, Friess H and Keppler D: Expression and localization of human multidrug resistance protein (ABCC) family members in pancreatic carcinoma. Int J Cancer. 115:359–367. 2005.PubMed/NCBI View Article : Google Scholar

99 

Hientz K, Mohr A, Bhakta-Guha D and Efferth T: The role of p53 in cancer drug resistance and targeted chemotherapy. Oncotarget. 8:8921–8946. 2017.PubMed/NCBI View Article : Google Scholar

100 

Fiorini C, Cordani M, Padroni C, Blandino G, Di Agostino S and Donadelli M: Mutant p53 stimulates chemoresistance of pancreatic adenocarcinoma cells to gemcitabine. Biochim Biophys Acta. 1853:89–100. 2015.PubMed/NCBI View Article : Google Scholar

101 

Chu IM, Hengst L and Slingerland JM: The Cdk inhibitor p27 in human cancer: Prognostic potential and relevance to anticancer therapy. Nat Rev Cancer. 8:253–267. 2008.PubMed/NCBI View Article : Google Scholar

102 

Alderton G: Priming resistance. Nat Rev Cancer. 7(162)2007.

103 

Brown I, Shalli K, McDonald SL, Moir SE, Hutcheon AW, Heys SD and Schofield AC: Reduced expression of p27 is a novel mechanism of docetaxel resistance in breast cancer cells. Breast Cancer Res. 6:R601–R607. 2004.PubMed/NCBI View Article : Google Scholar

104 

Germain P, Staels B, Dacquet C, Spedding M and Laudet V: Overview of nomenclature of nuclear receptors. Pharmacol Rev. 58:685–704. 2006.PubMed/NCBI View Article : Google Scholar

105 

Soprano KJ and Soprano DR: Retinoic acid receptors and cancer. J Nutr. 132:3809S–3813S. 2002.PubMed/NCBI View Article : Google Scholar

106 

Huang GL, Song W, Zhou P, Fu QR, Lin CL, Chen QX and Shen DY: Oncogenic retinoic acid receptor γ knockdown reverses multi-drug resistance of human colorectal cancer via Wnt/β-catenin pathway. Cell Cycle. 16:685–692. 2017.PubMed/NCBI View Article : Google Scholar

107 

Williams CC, Allison JG, Vidal GA, Burow ME, Beckman BS, Marrero L and Jones FE: The ERBB4/HER4 receptor tyrosine kinase regulates gene expression by functioning as a STAT5A nuclear chaperone. J Cell Biol. 167:469–478. 2004.PubMed/NCBI View Article : Google Scholar

108 

Thybusch-Bernhardt A, Beckmann S and Juhl H: Comparative analysis of the EGF-receptor family in pancreatic cancer: Expression of HER-4 correlates with a favourable tumor stage. Int J Surg Investig. 2:393–400. 2001.PubMed/NCBI

Related Articles

Journal Cover

December-2023
Volume 19 Issue 6

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zihlif M, Hameduh T, Bulatova N and Hammad H: Alteration in the expression of the chemotherapy resistance‑related genes in response to chronic and acute hypoxia in pancreatic cancer. Biomed Rep 19: 88, 2023.
APA
Zihlif, M., Hameduh, T., Bulatova, N., & Hammad, H. (2023). Alteration in the expression of the chemotherapy resistance‑related genes in response to chronic and acute hypoxia in pancreatic cancer. Biomedical Reports, 19, 88. https://doi.org/10.3892/br.2023.1670
MLA
Zihlif, M., Hameduh, T., Bulatova, N., Hammad, H."Alteration in the expression of the chemotherapy resistance‑related genes in response to chronic and acute hypoxia in pancreatic cancer". Biomedical Reports 19.6 (2023): 88.
Chicago
Zihlif, M., Hameduh, T., Bulatova, N., Hammad, H."Alteration in the expression of the chemotherapy resistance‑related genes in response to chronic and acute hypoxia in pancreatic cancer". Biomedical Reports 19, no. 6 (2023): 88. https://doi.org/10.3892/br.2023.1670