QNZ

Oxaliplatin resistance in colorectal cancer cells is mediated via activation of


Running Title: Oxaliplatin resistance in colon cancer mediated via ABCG2

Hsi-Hsien Hsu1,2, Ming-Cheng Chen3, Rathinasamy Baskaran4, Yueh-Min Lin5,6, Cecilia
Hsuan Day7, Yi-Jiun Lin8, Chuan-Chou Tu9, Viswanadha Vijaya Padma10, Wei-Wen
Kuo11,*, Chih-Yang Huang8,12,13,*

1Division of Colorectal Surgery, Mackay Memorial Hospital, Taipei, Taiwan 2Mackay Medicine, Nursing and Management College, Taipei, Taiwan
3Division of Colorectal Surgery, Taichung Veterans General Hospital, Taichung, Taiwan 4National Institute of Cancer Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
5Department of pathology, Changhua Christian Hospital, Changhua, Taiwan.
6Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli, Taiwan. 7Department of Nursing, Mei Ho University, Pingguang Road, Pingtung, Taiwan
8Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan 9Division of Chest Medicine, Department of Internal Medicine, Armed Force Taichung General Hospital, Taichung, Taiwan
10Department of Biotechnology, Bharathiar University, Coimbatore-641 046, India 11Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
12Graduate Institute of Chinese Medical Science, China Medical University, Taichung, Taiwan
13Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan * These authors contributed equally to this paper.

 

†This article has been accepted for publication and undergone full peer review but has not been through the copyediting, typesetting, pagination and proofreading process, which may lead to differences between this version and the Version of Record. Please cite this article as doi: [10.1002/jcp.26406]

 

Received 17 April 2017; Revised 30 October 2017; Accepted 12 December 2017
Journal of Cellular Physiology
This article is protected by copyright. All rights reserved
DOI 10.1002/jcp.26406

Corresponding author: Chih-Yang Huang Ph.D., Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan, No. 91, Hsueh-Shih Road, Taichung, 404, Taiwan. Tel: +886-4-22053366 ext 3313. Fax: +886-4-22032295. E-mail address: [email protected]
Abstract

Oxaliplatin (OXA), is a third generation platinum drug used as first-line chemotherapy in colorectal cancer (CRC). Cancer cells acquires resistance to anti-cancer drug and develops resistance. ATP-binding cassette (ABC) drug transporter ABCG2, one of multidrug resistance (MDR) protein which can effectively discharge a wide spectrum of chemotherapeutic agents out of cancer cells and subsequently reduce the intracellular concentration of these drugs. Role of ABCG2 and plausible molecular signaling pathways involved in Oxaliplatin-Resistant (OXA-R) colon cancer cells was evaluated in the present study. OXA resistant LoVo cells was developed by exposing the colon cells to OXA in a dose-dependent manner. Development of multi drug resistance in OXA-R cells was confirmed by exposing the resistance cells to oxaliplatin, 5-FU and doxorubicin. OXA treatment resulted in G2 phase arrest in parental LoVo cells, which was overcome by OXA-R LoVo cells. mRNA and protein expression of ABCG2 and phosphorylation of NF-κB was significantly higher in OXA-R than parental cells. Levels of ER stress markers were downregulated in OXA-R than parental cells. OXA-R LoVo cells exposed to NF-κB inhibitor QNZ effectively reduced the ABCG2 and p-NF-κB expression and increased ER stress marker expression. On other hand, invasion and migratory effect of OXA-R cells were found to be decreased, when compared to parental cells. Metastasis marker proteins also downregulated in OXA-R cells. In vivo studies in nude mice also confirms the same. ABCG2 inhibitor verapamil, downregulate ABCG2, induce ER stress markers and induces apoptosis. In vivo studies in nude mice also confirms the same. This article is protected by copyright. All rights reserved

 

 

Keywords: Multi Drug Resistance; Colon Cancer; Cancer Stem Cell; Oxaliplatin; ABCG2
Introduction
Colorectal cancer (CRC) is the third most common cancer worldwide and the first most common cause of death (Shike et al., 1990; Vormbrock and Monkemuller, 2012). Besides, many Asian countries have experienced an increase of two to four times in the incidence of colorectal cancer during the past few decades (Sung et al., 2005). In Taiwan, the incidence is also the highest one. Oxaliplatin (OXA), a first-line chemotherapy in CRC, acts as a bifunctional alkylating agent, covalently binding DNA and forming platinum-DNA adducts to inhibit DNA replication and transcription (Kelland, 2007). The intra strand cross-links formed by OXA being the most abundant lesions capable of blocking both replication and transcription of DNA, they are considered to cause the major cytotoxic lesions and being directly involved in the cancer cells death (Woynarowski et al., 2000). Despite curative surgical resected in 70-80% of patients, nearly 40% of them either present with metastases or develop recurrence and eventually die (Andre and Schmiegel, 2005). Thus, chemotherapy seems to be the only therapeutic modality which may offer some benefit. However, the failure of cancer therapy is result from the drug resistance. Despite of efficiency of anti-cancer drugs, chemoresistance develops in nearly all metastatic patients. Nevertheless, drug resistances remain a major obstacle in failure of chemotherapy.

Oxaliplatin was widely used in combination therapies with other anticancer drugs such as Irinotecan, 5-fluorouracil (5-FU), folinic acid (FA) and leucovorin (LV) (Cavanna et al.,
2006; Peinert et al., 2010; Wasserman et al., 2001) for treating different solid tumors. Therefore Multidrug-resistances (MDR) become a critical factor in the failure of many forms of chemotherapy, which result from structurally, chemically and functionally cross-resistance to many other unrelated agents (Umsumarng et al., 2013).

Metastasis of cancer cell is a procedure that takes multiple steps, including initial migration and invasion, dissemination of aggressive tumor cells and skipping immune system, result in growth or colonization of micro-metastatic lesions and even turn to macro-metastases. In colon cancer, primary tumor and their corresponding metastasis, exhibit an association to Epithelial-to-Mesenchymal Transition (EMT) phenotype. ABC transportersare important for mediating cellular resistance to chemotherapeutic drugs (Dean et al., 2001; Paumi et al., 2009). Mechanism of resistant chemotherapy is the expression of ABC transporters that use the energy of ATP hydrolysis to transport a wide variety of substrates across the cell membrane (Robey et al., 2007). These transporters are characterized into three efflux transporters includes the multi-drug resistance proteins (MDRs), multi-drugs resistance-associated proteins (MRPs), and breast cancer resistance protein (BCRP/ABCG2). ABCG2 have been associated with the side population (SP) phenotype and stem cell activity (Fatima et al., 2012). SP cells could repopulate both SP and non-SP cells suggesting that these cells possess the hallmark properties of cancer stem cells (CSCs), namely self-renewal and differentiation (Abdullah and Chow, 2013). However, it appears that the ABCG2 transporter
is the most specific marker of stem cells in the adult hematopoietic system (Zhou et al., 2002). Intriguingly, CSCs are also supposed to be responsible for the acquisition of multi-drug resistance. Therefore, side population and chemoresistance suggest a close link between ABCG2 and CSCs. ABCG2 is also involved in the development of irinotecan resistance in vitro and in vivo in colon cancer (Candeil et al., 2004). Expression level of ABCG2 increased in plasma cell lines that ABCG2 work as a surviving factor in plasma cells under ER stress (Nakamichi et al., 2009) through phosphorylation of PERK and eIF2α (Teske et al., 2011). During ER stress, PERK induces auto-phosphorylation of its kinase domain and increases its activity to Phosphorylate alpha subunit of eukaryotic translation-initiation factor 2 (eIF2α), leading to its inactivation and result in a rapid reduction of translational initiation and attenuation of protein synthesis.

Present study was designed to evaluate plausible role of ABCG2, multi drug resistance protein and associated pathway involved in OXA-R LoVo cells. Our data show that oxaliplatin resistant LoVo colon cells are resistant to oxaliplatin, 5-FU and doxorubicin. Multi drug resistance acquired by overexpression of ABCG2 and p-NF-κB and downregulation of ER stress protein thereby preventing apoptosis.
Materials and Methods Cell culture

LoVo cells were cultivated in Dulbecco’s minimum essential medium (D5523, Sigma, Saint Louis, Missouri, USA) containing 10% FBS (Characterized Fetal Bovine Serum, Hyclone, long, Utah USA), 1% penicillin (Invitrogen Corp., California, USA).
Whole Cell Extract
Total proteins from the cells were extracted in a cell lysis buffer (50 mM Tris-base, 0.5 M NaCl, 1 mM EDTA, 1% NP40, 1% Glycerol, 1 mM – Mercaptoethanol, Proteinase k inhibitor). After centrifugation the protein in the supernatant were collected and stored at
-80°C.
MTT (Thiazolyl blue tetrazilium bromide) assay
The cell viability was measured using MTT [3-(4, 5-Dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium-bromide] assay (Sigma) after treatment. Cells were plated in triplicate in 12-well plates and treated with increasing concentrations of drugs parental LoVo cell (Oxaliplatin 5, 10, 15, 20, 25 µg/ml – 24 h), OXA-R LoVo cells (Oxaliplatin 10, 15, 25, 35, 45, 55, 65 µg/ml – 24 h). After 24 h of incubation, 0.5 mg/ml of MTT was added to each well for at least 4-5 h. The blue MTT formazan crystals were then dissolved in 500 μl of DMSO. The absorbance at 570 nm was measured by ELISA reader. Cell viability was expressed in percentage compare to control.
Lowry Protein Assay
Protein were reacted to alkaline cupric sulfate in the presence of tartrate (2% Na-K tartrate: 1% CuSO4.5H2O: 2% Na2CO3 in 0.1 N NaOH = 1:1:98). During 10 min incubation at room temperature, a tetradentate copper complex forms from four peptide bonds and one atom of copper. Then, Folin phenol reagent was added. When the color of solution mixture changed into blue is due to the tetradentate copper complex transfers electrons to the phospho molybdic/phospho tungstic acid complex. After 30 min room temperature incubation, absorbance of targets was detected at OD value 750 nm by ELISA reader.

Antibodies and reagents.

 

The following antibodies were used in this study: p-EGFR (#2231, Cell Signaling Technology), p-PI3K (#4228, Cell Signaling Technology), P-Akt (sc-7985, Santa Cruz Biotechnology), p-NF-κB (#3033L, Cell Signaling Technology), β-Actin (sc-47778, Santa Cruz Biotechnology), E-Cadherin (sc-8426, Santa Cruz Biotechnology), β-Catenin (sc-7963, Santa Cruz Biotechnology), FAK (sc-1688, Santa Cruz Biotechnology), Snai1 (sc-10433, Santa Cruz Biotechnology), RAC1 (ab33186, Abcam, Cambridge, UK), Raptor (#2280, Cell Signaling Technology), Rictor (#2114, Cell Signaling Technology), ABCG2 (#4477s, Cell Signaling Technology), p-PERK (sc-32577, Santa Cruz Biotechnology), p-eIF2α (#9721s, Cell Signaling Technology), Caspase 12 (#2202, Cell Signaling Technology), Caspase 3 (#9665s, Cell Signaling Technology), α-tubulin (sc-5286, Santa Cruz Biotechnology).
Western blotting
Cells were harvested in lysis buffer (250 mM sucrose, 50 mM Tris-HCl, 5 mM imidazole, 2.5 mM EDTA, 2.5 mM DTT, 0.1% TritonX-100, pH 7.4) and concentration of sample proteins were determined using Lowry protein assay. Briefly, equal amount (35 μg) of protein were heated after adding the appropriate amount of 5x sample buffer (5 mM EDTA, 162 mM DTT, 5% SDS, 50% glycerol, 0.5 l bromophenol blue, 188 mM Tris, pH 8.8). The samples were separated on 10% SDS-polyacrylamide gels (PAGE) and subsequently transferred onto nitrocellulose filters using the Bio-Rad electro transfer system (Bio-Rad Laboratories, Munich, Germany). The blot was then incubated with specific primary antibody for overnight at 4°C, washed thrice with TBST and incubated with appropriate secondary antibody for 1 h at room temperature. Finally the blots were developed using a custom-made ECL detection system (2.5 mM luminol, 0.4 mM para-coumaric acid, 10 mM Tris base, 0.15 l H2O2, pH 8.5).
Wound healing assay
Cells were seeded on 10 cm culture plates, an artificial “wound” carefully created on surface were fulfilled with by using a 200 µl pipette tip scratch on the sub confluent cell monolayer. Observation of the level of cell migration was started at 0 h as control cultured in 1% serums supplemented DMEM by a phase contrast microscope.
Transwell migration assay and invasion assay
In one horizontal divided two spaces side-by-side by a 6-well culture plate with 8 μm pore size polycarbonate membrane contained chamber. Cell suspension loaded into chamber with serum free culture medium then invading cells migrate through as well as attach to the bottom of membrane in serum contained side. Therefore non-invading cells remain above. Invasion assay is almost the same as migration assay except for the upper chamber was coding with one layer matrix gel to mimic the ECM of tissue.
Flow cytometry
Cell cycles are analyzed individually based on their shapes, sizes and DNA content. Cells were seeded in serum free medium, then cells were harvest at different time points 0, 24, 48 h (exposed to serum and oxaliplatin separately and without as control). Next, the collected cells were fixed by 70% ethanol over-night in -20℃. After that ethanol was discard, cells washed with PBS and finally stained with PI (1 mg/ml PI, 1% Triton, 2 mg/ml RNAse) in 37℃ for 30 min.
Reverse-transcription polymerase chain reaction (RT-PCR)
Total RNA was extracted using the Direct-zol RNA MiniPrep Kit (Zymo Research Corporation, Irvine, CA, USA) according to the manufacturer’s instructions. Briefly, 2 μg of total RNA was incubated with 0.5 μg of oligo dT (MD Bio., Taipei, Taiwan) at 70 °C for 15 min. Then, the RNA was mixed with buffer containing 0.25 mM dNTPs (MD Bio.), 20 U
of RNasin I Plus RNase Inhibitor (Promega, San Luis Obispo, CA, USA) and 20 U of M-MLV Reverse Transcriptase (Promega) and incubated at 42 °C for 90 min to allow for cDNA synthesis. This mixture was then used for specific cDNA amplification in a GeneAmp PCR system 2400 (Perkin Elmer, Waltham, MA, USA). Sense primer for ABCG2:
5′-TCCATATCGTGGAATGCTGA-3′; antisense primer:
5′-TTTCAGCCGTGGAACTCTTT-3. Sense primer for GAPDH:

 

5′-TCGGAGTCAACGGATTTGG-3′;

5′-GAATTTGCCATGGGTGGAAT-3′ were used.

 

Gelatin zymography
antisense primer:

 
107 cells were seeded in serum free media for 48 h. Supernatants were harvest, mixed with 5x loading buffer (SDS, 0.5M Tris-HCl, DDW, Glycerol, Bromophenol blue). The samples were separated by electrophoresis with 0.1% Gelatin in 8% SDS-PAGE for 90 min. Then washed by washing buffer 2.5% (Triton X-100) for 30 min twice. Next, stain the gel by mixture (Coomassie Brilliant Blue, Amino Black, Methanol and Acetic acid) to binding the glycerol.

Mouse xenograft study

 

The experimental design was approved by the Institutional Animal Care and Use
Committee of China Medical University, Taichung, Taiwan, ROC. Male nude mice (8 weeks
old) were divided into 2 treatment groups with 6 animals per group. Parental-LoVo cells and Oxaliplatin-resistance LoVo cells (1×106 cells per injection) suspended in matrigel: serum free medium (1:1; BD Biosciences, Oxford, United Kingdom) subcutaneously into their right flanks. Tumors were allowed to reach 50~100 mm3 before beginning of treatment. 5 μM Ko143 were injected into each tumors per cm3 of nude mice at 23rd day and sacrificed at 30th days. Expression level of caspase-3 was evaluated in the resected tumor.

Statistical analysis

 

All experiments were repeated at least three times and statistical analysis was performed using SigmaPlot 10.0 software. Student t-test was used to analyze each numeric data. In all cases, differences at p<0.05 were regarded as statistically significant, ones at p<0.01, p<0.005 or p<0.001 were considered higher statistical significances.
Results

Establishment and characterization of oxaliplatin-resistant cell lines
To establish stable colon cancer cell lines resistant to oxaliplatin, LoVo cell lines were exposed to oxaliplatin in dose-dependent manner from 0-25 μg/ml for 24 h (Fig. 1A). This was MTT-1. LoVo cell lines exposed to 15 μg/ml oxaliplatin resulted around 50% cell death (IC50 of oxaliplatin). Cells treated with 15 μg/ml oxaliplatin (24 h) allowed to reached 80% confluence, and passaged twice in this same concentration (15 μg/ml) of oxaliplatin. The
same procedure was repeated at increased doses of oxaliplatin (20 and 25 μg/ml) until a cell population was selected that demonstrated at least a 3-fold greater IC50 (45 μg/ml) to oxaliplatin than the parental cell lines. Another MTT experiment was conducted at doses of 10, 15, 25, 35, 45, 55, 65 μg/ml (MTT-2) for 24 h (Fig. 1B). In order to compare resistance between parental cell and resistant cell, both cells were treated with oxaliplatin in different dose from 5-25 μM for 24 h. Cell viability was significantly decreased in parental cell in a dose-dependent manner. However, there are no significantly decreased cell viability (in lower concentration) was observed in oxaliplatin-R-LoVo cells (Fig. 1C).
Oxaliplatin-resistant LoVo colon cancer cells develops multidrug resistance
Resistance to oxaliplatin in LoVo colon cancer cell line was confirmed previously. Promoting incidence of multidrug resistance to chemotherapeutic agents has becoming a major cause of the failure of cancer therapy. To examine the multidrug resistance of colon LoVo cancer cell line, both cells (oxaliplatin-resistant cell and parental cell) were treated with 5-FU and doxorubicin separately and the cell viability was measured by MTT assay. After the treatment with 5-FU, cell viability was significantly decreased compare to control in parental cell (Fig. 2A). Besides, the cell viability was obviously decreased in parental cell compare to resistant cell (Fig. 2B).
Cell survival pathway proteins were highly expressed in oxaliplatin-resistant cells
To compare the proliferation and survival between two cell lines, we use examine the expression levels of p-EGFR, p-PI3K, p-Akt and p-NF-κB by Western blot. The expression of p-EGFR, p-PI3K, p-Akt and p-NF-κB were higher in oxaliplatin-resistant cell than parental cells (Fig. 3A). Flow cytometry analysis was performed to determine the cell cycle distribution. G2/M phase accumulation was observed in OXA-R cells when compared to parental cells (Fig. 4A). Cell cycle distribution analysis reveals that oxaliplatin treatment in parental LoVo cell resulted in G2/M phase cell cycle arrest (Fig. 4A, B).
Migration and Invasion analysis of LoVo parental and oxaliplatin-resistant LoVo cell
Migration and invasion are critical steps in initial progression of cancer that facilitate metastasis. Western blot showed that the expression levels of migration markers β-catenin, FAK, snail, RAC1, Raptor, Ritor except for E-cadherin were elevated in parental cells when compare to oxaliplatin-R-LoVo cell line (Fig. 5A). To further compare the migration and invasion ability of two cell line, we performed wound-healing assay, Transwell migration assay and Transwell invasion assay. All the data showed that the migration and invasion ability of parental cells were stronger than oxaliplatin-R-LoVo cell lines (Fig. 5B, C, D). On the other hand, Gelatin zymography showed that the activity of MMP2 were significantly higher in parental cells compare to oxaliplatin R-LoVo cell line and the activity of MMP9 were not different between two cell line (Fig. 5E). IVIS was performed to localize the tumor
cells in nude mice. IVIS data showed that the higher levels of GFP is in the same area as tumor site in both parental cells and oxaliplatin-R-LoVo cell induced tumor nude mice model. But, in the mice injected with parental LoVo cells showed migration of metastatic cancer cells to other parts also (Fig. 5F, G). Taken together, our study shows that oxaliplatin resistant LoVo cell processes strong proliferation instead of metastasis both in vitro and in vivo.

Oxaliplatin resistance in colon cancer cell is mediated through activation of ABCG2 to alleviate ER stress induced apoptosis.

To confirm the mechanism of oxaliplatin resistance in LoVo colon cancer cell line is regulated by ABCG2, we use MTT assay and found that the cell viability was significantly decreased after treating verapamil, an inhibitor of ABCG2, in dose dependent manner from 0-150 μM (Fig. 6A). Western and RT-PCR results showed that both protein level and RNA levels of ABCG2 were visibly elevated in oxaliplatin-R-LoVo cell line compare to parental cell as well as the ER stress markers, p-PERK, caspase 12 were significantly decreased in oxaliplatin-R-LoVo cell lines (Fig. 6B, C). Next, we check whether inhibition of ABCG2 could reverse drug resistance in OXA-R LoVo cells with verapamil, an inhibitor of ABCG2. After treating with the inhibitor of ABCG2, verapamil, at dose range from 0-200 μM the expression of ABCG2 was significantly decreased (Fig. 6D). On the other hand in animal model, Ko143 injection reduced the tumor size (Fig. 6E&F) and effectively induce apoptosis in the tumor of nude mice, which was induced by parental and oxaliplatin-R LoVo cell (Fig.
6G). NF-κB plays an significant role in multi drug resistance. We have also shown that OXA-R induced p- NF-κB expression (Fig.3). Further to confirm the role of NF-κB in oxaliplatin-resistant-LoVo colon cancer model, NF-κB inhibition study was performed. In this experiment, both cells were treated with QNZ, selective inhibitor NF-κB, at different dose range from 0-1000 nM and cell viability was measured. QNZ treatment decreased the cell viability more effectively in resistant cell than that of parental cell (Fig. 7A). Next we check the effect of QNZ on NF-κB and ABCG2. Inhibition of p-NF-κB was observed in both parental and OXA-R LoVo cells, but the inhibition was profound in OXA-R cells. Relatively, it could also induce apoptosis in both cell types which was evident from the cleaved caspase 3 expression. (Fig. 7B).
Discussion
ABCG2 is a kind of cancer stem cell marker, reported in various cancer types including hepatocellular carcinoma (Zhang et al., 2013), breast cancer (Britton et al., 2012) and colon cancer (Liu et al., 2010). Previous report are there, which showed a strong correlation with ABCG2 for maintenance of side population (SP) feature (Xu et al., 2012). In addition, Hoechst dye exclusion and FACS of cancer cell lines, identify and track cancer stem-like side populations of cancer cells characterized by drug resistance (Wu et al., 2013). First, SP cells have specific stem cell features, particularly self-renewal and differentiation potential. Second, SP cells play a key role in the drug resistance of tumors (Relation et al., 2017). These
studies reveal the association between ABCG2 and SP phenotype and its stem cell properties. In corroborate with these statement, we found the overexpression of ABCG2 in oxaliplatin-resistant cell in order to escape from the apoptosis induced by ER stress.

Cancer stem cells have been observed in many human solid malignancies, breast, brain, colon, prostate and pancreas (Al-Hajj et al., 2003; Collins et al., 2005; Li et al., 2007; Singh et al., 2003). Tumor microenvironments have been proven for regulation of cancer stem cell by stimulation of CSC features. In addition, the maintenance of CSC, microenvironment is speculated to be related to metastasis by induction of the epithelial-mesenchymal transition, resulting in dissemination and invasion of tumor cells (Borovski et al., 2011). Microenvironment promotes cancer progression through metastasis and drug resistance (Kuo et al., 2014). Thus, the microenvironment seems to be of crucial importance for primary tumor growth as well as metastasis formation. ABCG2 might also play a protective role in the survival of CSCs. It has been reported that, ABCG2 over expression linked to poor clinical outcomes especially in patients with lung carcinoma esophageal squamous cell carcinoma and acute myeloid leukemia (Wu et al., 2017). NF-κB is one of the stem cell regulatory pathways frequently dysregulated in tumor cells (Iliopoulos et al., 2009). NF-κB is a crucial factor in oxaliplatin resistance via autocrine signaling through IL-1 and IL-8 (Wilson et al., 2008) and participate in regulators of inflammation lead to chemoresistance to anti-cancer drugs (Abdullah and Chow, 2013). NF-κB activate additionally secretion of IL-6 and IL-8 to
generate a positive feed-back loop between immune cell and tumor cell that stimulates the cancer stem cell components, accelerating metastasis and therapeutic resistance (Korkaya et al., 2011). Surprisingly, in our study both the level of migration and invasion in oxaliplatin-resistant cells is lower than parental cells. Indeed, the level of phosphorylation of NF-κB is significantly higher in oxaliplatin-resistant cells. Moreover, after treating with QNZ, (inhibitor of NF-κB) and verapamil (ABCG2 inhibitor) the cell viability and levels of both ABCG2 and NF-κB were decreased in oxaliplatin-resistant cells compare to parental cells. The PI3K/Akt/mTOR signaling pathway is involved in cell proliferation, diff erentiation, survival and prevent apoptosis (Shen et al., 2017). We found that cell proliferation was higher in oxaliplatin-R-LoVo cell lines through phosphorylation of EGFR, PI3K, Akt, NF-κB signaling. The level of migration in resistant cells were suppressed compare to parental cell, but do strongly proliferation strongly suggest microenvironment seems to be an important factor to progression of oxaliplatin chemoresistance in colon cancer (Fig. 8).

Conflict of Interest

 

The authors declared that there are no conflicts of interests.
References
Abdullah LN, Chow EK. 2013. Mechanisms of chemoresistance in cancer stem cells. Clinical and translational medicine 2(1):3.
Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. 2003. Prospective identification of tumorigenic breast cancer cells. Proceedings of the National Academy of Sciences of the United States of America 100(7):3983-3988.
Andre N, Schmiegel W. 2005. Chemoradiotherapy for colorectal cancer. Gut 54(8):1194-1202.
Borovski T, De Sousa EMF, Vermeulen L, Medema JP. 2011. Cancer stem cell niche: the place to be. Cancer research 71(3):634-639.
Britton KM, Eyre R, Harvey IJ, Stemke-Hale K, Browell D, Lennard TW, Meeson AP. 2012. Breast cancer, side population cells and ABCG2 expression. Cancer letters 323(1):97-105.
Candeil L, Gourdier I, Peyron D, Vezzio N, Copois V, Bibeau F, Orsetti B, Scheffer GL, Ychou M, Khan QA, Pommier Y, Pau B, Martineau P, Del Rio M. 2004. ABCG2 overexpression in colon cancer cells resistant to SN38 and in irinotecan-treated metastases. International journal of cancer 109(6):848-854.
Cavanna L, Artioli F, Codignola C, Lazzaro A, Rizzi A, Gamboni A, Rota L, Rodino C, Boni F, Iop A, Zaniboni A. 2006. Oxaliplatin in combination with 5-fluorouracil (5-FU) and leucovorin (LV) in patients with metastatic gastric cancer (MGC). American journal of clinical oncology 29(4):371-375.
Collins AT, Berry PA, Hyde C, Stower MJ, Maitland NJ. 2005. Prospective identification of tumorigenic prostate cancer stem cells. Cancer research 65(23):10946-10951.
Dean M, Rzhetsky A, Allikmets R. 2001. The human ATP-binding cassette (ABC) transporter superfamily. Genome research 11(7):1156-1166.
Fatima S, Zhou S, Sorrentino BP. 2012. Abcg2 expression marks tissue-specific stem cells in
multiple organs in a mouse progeny tracking model. Stem cells 30(2):210-221. Iliopoulos D, Hirsch HA, Struhl K. 2009. An epigenetic switch involving NF-B, Lin28, Let-7
MicroRNA, and IL6 links inflammation to cell transformation. Cell 139(4):693-706. Kelland L. 2007. The resurgence of platinum-based cancer chemotherapy. Nature reviews
Cancer 7(8):573-584.
Korkaya H, Liu S, Wicha MS. 2011. Regulation of cancer stem cells by cytokine networks: attacking cancer’s inflammatory roots. Clinical cancer research : an official journal of the American Association for Cancer Research 17(19):6125-6129.
Kuo CT, Chiang CL, Chang CH, Liu HK, Huang GS, Huang RY, Lee H, Huang CS, Wo AM. 2014. Modeling of cancer metastasis and drug resistance via biomimetic nano-cilia and microfluidics. Biomaterials 35(5):1562-1571.
Li C, Heidt DG, Dalerba P, Burant CF, Zhang L, Adsay V, Wicha M, Clarke MF, Simeone DM. 2007. Identification of pancreatic cancer stem cells. Cancer research
67(3):1030-1037.
Liu HG, Pan YF, You J, Wang OC, Huang KT, Zhang XH. 2010. Expression of ABCG2 and its significance in colorectal cancer. Asian Pacific journal of cancer prevention : APJCP 11(4):845-848.
Nakamichi N, Morii E, Ikeda J, Qiu Y, Mamato S, Tian T, Fukuhara S, Aozasa K. 2009. Synergistic effect of interleukin-6 and endoplasmic reticulum stress inducers on the high level of ABCG2 expression in plasma cells. Laboratory investigation; a journal of technical methods and pathology 89(3):327-336.
Paumi CM, Chuk M, Snider J, Stagljar I, Michaelis S. 2009. ABC transporters in Saccharomyces cerevisiae and their interactors: new technology advances the biology of the ABCC (MRP) subfamily. Microbiology and molecular biology reviews : MMBR
73(4):577-593.
Peinert S, Grothe W, Stein A, Muller LP, Ruessel J, Voigt W, Schmoll HJ, Arnold D. 2010. Safety and efficacy of weekly 5-fluorouracil/folinic acid/oxaliplatin/irinotecan in the first-line treatment of gastrointestinal cancer. Therapeutic advances in medical oncology 2(3):161-174.
Relation T, Dominici M, Horwitz EM. 2017. Concise Review: An (Im)Penetrable Shield: How the Tumor Microenvironment Protects Cancer Stem Cells. Stem Cells
35(5):1123-1130.
Robey RW, Polgar O, Deeken J, To KW, Bates SE. 2007. ABCG2: determining its relevance in clinical drug resistance. Cancer metastasis reviews 26(1):39-57.
Shen Z, Xu L, Li J, Zhang N. 2017. Capilliposide C Sensitizes Esophageal Squamous Carcinoma Cells to Oxaliplatin by Inducing Apoptosis Through the PI3K/Akt/mTOR Pathway. Med Sci Monit 23:2096-2103.
Shike M, Winawer SJ, Greenwald PH, Bloch A, Hill MJ, Swaroop SV. 1990. Primary prevention of colorectal cancer. The WHO Collaborating Centre for the Prevention of Colorectal Cancer. Bulletin of the World Health Organization 68(3):377-385.
Singh SK, Clarke ID, Terasaki M, Bonn VE, Hawkins C, Squire J, Dirks PB. 2003. Identification of a cancer stem cell in human brain tumors. Cancer research 63(18):5821-5828.
Sung JJ, Lau JY, Goh KL, Leung WK, Asia Pacific Working Group on Colorectal C. 2005. Increasing incidence of colorectal cancer in Asia: implications for screening. The Lancet Oncology 6(11):871-876.
Teske BF, Wek SA, Bunpo P, Cundiff JK, McClintick JN, Anthony TG, Wek RC. 2011. The eIF2 kinase PERK and the integrated stress response facilitate activation of ATF6 during endoplasmic reticulum stress. Molecular biology of the cell 22(22):4390-4405.
Umsumarng S, Pintha K, Pitchakarn P, Sastraruji K, Sastraruji T, Ung AT, Jatisatienr A, Pyne SG, Limtrakul P. 2013. Inhibition of P-glycoprotein mediated multidrug resistance by stemofoline derivatives. Chemical & pharmaceutical bulletin 61(4):399-404.
Vormbrock K, Monkemuller K. 2012. Difficult colon polypectomy. World journal of
gastrointestinal endoscopy 4(7):269-280.
Wasserman E, Sutherland W, Cvitkovic E. 2001. Irinotecan plus oxaliplatin: a promising combination for advanced colorectal cancer. Clinical colorectal cancer 1(3):149-153.
Wilson C, Purcell C, Seaton A, Oladipo O, Maxwell PJ, O’Sullivan JM, Wilson RH, Johnston PG, Waugh DJ. 2008. Chemotherapy-induced CXC-chemokine/CXC-chemokine receptor signaling in metastatic prostate cancer cells confers resistance to oxaliplatin through potentiation of nuclear factor-B transcription and evasion of apoptosis. The Journal of pharmacology and experimental therapeutics 327(3):746-759.
Woynarowski JM, Faivre S, Herzig MC, Arnett B, Chapman WG, Trevino AV, Raymond E, Chaney SG, Vaisman A, Varchenko M, Juniewicz PE. 2000. Oxaliplatin-induced damage of cellular DNA. Molecular pharmacology 58(5):920-927.
Wu CP, Hsiao SH, Murakami M, Lu MJ, Li YQ, Hsieh CH, Ambudkar SV, Wu YS. 2017. Tyrphostin RG14620 selectively reverses ABCG2-mediated multidrug resistance in cancer cell lines. Cancer Lett 409:56-65.
Wu CP, Zhou L, Xie M, Du HD, Tian J, Sun S, Li JY. 2013. Identification of cancer stem-like side population cells in purified primary cultured human laryngeal squamous cell carcinoma epithelia. PloS one 8(6):e65750.
Xu XT, Xu Q, Tong JL, Zhu MM, Nie F, Chen X, Xiao SD, Ran ZH. 2012. MicroRNA expression profiling identifies miR-328 regulates cancer stem cell-like SP cells in colorectal cancer. British journal of cancer 106(7):1320-1330.
Zhang G, Wang Z, Luo W, Jiao H, Wu J, Jiang C. 2013. Expression of Potential Cancer Stem Cell Marker ABCG2 is Associated with Malignant Behaviors of Hepatocellular Carcinoma. Gastroenterology research and practice 2013:782581.
Zhou S, Morris JJ, Barnes Y, Lan L, Schuetz JD, Sorrentino BP. 2002. Bcrp1 gene expression is required for normal numbers of side population stem cells in mice, and confers relative protection to mitoxantrone in hematopoietic cells in vivo. Proceedings of the National Academy of Sciences of the United States of America 99(19):12339-12344.
Figure legends:

Fig. 1. Oxaliplatin induces chemoresistance in LoVo colon cancer cell lines. (A) IC50 of oxaliplatin was determined by treating parental LoVo cancer cells in a dose dependent manner. (B) LoVo cells were challenged with oxaliplatin IC50 dose, resistance to oxaliplatin was increased compared to the control. (C) Both parental and oxaliplatin-R cells were treated with varying concentration of oxaliplatin for 24 h analyzed for cell viability. Each vertical bar shows the mean ± SD, of cell viability in each dose of oxaliplatin *p˂0.05, **p˂0.005, ***p˂0.001.

Fig. 2. Assessment of multi-drug resistance in oxaliplatin-resistance LoVo cell lines. (A) 5-FU and (B) Doxorubicin were used to evaluate the multidrug resistance in oxaliplatin-R-LoVo cells. Both cell types were exposed to 5-FU and doxorubicin for 24 h, cell viability was measured by MTT assay and compared between parental LoVo cells and oxaliplatin-R cells in each dose. Each vertical bar shows the mean ± SD, of cell viability in each dose of doxorubicin and 5-FU. *p˂0.05, **p˂0.005, ***p˂0.001.

Fig. 3. Proliferation rate of oxaliplatin-R-LoVo cell lines was higher than the parental LoVo cell lines. (A) Expression levels of proliferation markers EGFR, Akt, PI3K and NF-κB were measured by western blot in parental LoVo cell lines and oxaliplatin-R-LoVo cell lines. (B) Densitometric values of phosphorylated EGFR, Akt, PI3K and NF-κB expressions were plotted. Each vertical bar shows the mean ± SD. Statistical differences were marked as *p˂0.05, **p˂0.005, ***p˂0.001.

Fig. 4. Oxaliplatin resistant cells overcome oxaliplatin induced G2/M phase cell cycle arrest. (A) Flow cytometry analysis was performed to determine the cell cycle distribution in LoVo cell line and oxaliplatin R-LoVo cell line. (B) Cells were treated with oxaliplatin for 48 h and cell cycle analysis was carried out. (C) Flow cytometric quantification of all phases of
the cell cycle were shown in both LoVo cell line and oxaliplatin R-LoVo cell line for 48 h. (D) Treatment with oxaliplatin for 48 h in oxaliplatin-R-LoVo cell line increased the G2 phase.

Fig. 5. Migration ability and Invasion ability of parental LoVo cell lines and oxaliplatin-R-LoVo cell lines. (A) Migration markers (β-catenine, FAK, Snai1, RAC1, Raptor and Rictor) were decreased in oxaliplatin resistant cells, except for E-cadherin. (B) Wound healing assay showed that the migration ability was lowed in oxaliplatin-R-LoVo cells compared to control. (C) & (D) Transwell assay showed that invasion ability of oxaliplatin resistant cells were decreased. (E) MMP 2 and MMP 9 expressions of parental and oxaliplatin-R-LoVo cell lines was analyzed by Zelatin zymography. (F) Oxaliplatin-R-LoVo cell lines and (G) parental LoVo cell lines were transfected green fluorescent protein (GFP) then subcutaneously injected into nude mice, analyzed by International Veterinary Information Service (IVIS).

Fig. 6. Resistance to oxaliplatin is mediated by ABCG2 in LoVo colon cancer cells. (A) Parental LoVo cell lines and oxaliplatin-R-LoVo cell lines were treated of verapamil in dose-dependent manner (0-150 μM) and cell viability were measured by MTT assay. (B) Protein levels of ABCG2, p-PERK and Caspase12 in both cell type was analysed by western blotting. Each vertical bar shows the mean ± SD. *p˂0.05, **p˂0.005, ***p˂0.001. (C) ABCG2 mRNA expression was analyzed in both in parental and oxaliplatin-resistant cells by RT-PCR. (D) Parental LoVo cell line and oxaliplatin-R-LoVo cell line were treated with different concentrations of Verapamil (0-150 μM) and analyzed for ABCG2, p-NF-κB, p-PERK, p-eIF2α, caspase-12 & 3 expression. (E) The weight and (F) size of nude mices were record from the day, after parental LoVo cell line and oxaliplatin-R-LoVo cell line cell were injected into nude mice. Treatment of Ko143 was started at day 23 in each day till sacrificed at day 29. (G) Tumors were successfully induced by subcutaneous injection of after parental LoVo cell line and oxaliplatin-R-LoVo cell line into nude mice then 5 μM Ko143
were injected into each tumors per cm3 at day 23 and the expression level of caspase-3 was evaluated in the resected tumor.

Fig. 7. NF-κB affect oxaliplatin Resistance via ABCG2. (A) Parental LoVo cell line and oxaliplatin-R-LoVo cell line were treated with different concentrations of QNZ (0-1000 nM) and cell viability were measured by MTT assay. Each vertical bar shows the mean ± SD of cell viability in each dose of QNZ. *p˂0.05, **p˂0.005, ***p˂0.001. (B) Expression levels of ABCG2 and p-NF-κB in parental and resistance cells were analysed by western blot after treating with 200 nM of QNZ.

Fig. 8. Graphic abstract: NF-κB affect oxaliplatin resistance via ABCG2.

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 
Figure 1

 

 

 

 

 

 

 

 

 

 

 

Figure 2

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 
Figure 3

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 
Figure 4

 

 

 

 

 

 

 

 

 

 

 

 

 

 
Figure 5

 

 

 

 

 

 

 

 

 

 

 

 

 

 
Figure 6

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Figure 7

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Figure 8

Leave a Reply

Your email address will not be published. Required fields are marked *

*

You may use these HTML tags and attributes: <a href="" title=""> <abbr title=""> <acronym title=""> <b> <blockquote cite=""> <cite> <code> <del datetime=""> <em> <i> <q cite=""> <strike> <strong>