Open Access

EVI‑1 acts as an oncogene and positively regulates calreticulin in breast cancer

Retraction in: /10.3892/mmr.2023.13112

  • Authors:
    • Lei Wu
    • Tianyi Wang
    • Dongning He
    • Xiaoxi Li
    • Youhong Jiang
  • View Affiliations

  • Published online on: December 24, 2018     https://doi.org/10.3892/mmr.2018.9796
  • Pages: 1645-1653
  • Copyright: © Wu 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

Ecotropic viral integration site‑1 (EVI‑1) is an important transcription factor involved in oncogenesis. Aberrant EVI‑1 expression has been reported to be a characteristic of multiple types of malignancies; however, very little is known about how EVI‑1 regulates breast cancer. Current knowledge of how target genes mediate the biological function of EVI‑1 remains limited. In the present study, overexpression of EVI‑1 promoted cell proliferation, migration, and invasion, and inhibited apoptosis in breast cancer. By contrast, silencing of EVI‑1 inhibited cell proliferation, migration and invasion, and enhanced apoptosis in breast cancer. In addition, the results also revealed that the aberrant expression of EVI‑1 regulates genes associated with the apoptotic pathway in breast cancer. Furthermore, EVI‑1 was also likely to target the promoter region of calreticulin (CRT) in vitro. It was concluded that EVI‑1 can affect epithelial mesenchymal transition‑associated genes by regulating the expression of CRT in breast cancer. The results revealed that EVI‑1 may be a potential effective therapeutic target in breast cancer.

Introduction

Breast cancer is a highly prevalent malignancy in women that is associated with high rates of morbidity and mortality worldwide, and these rates have continued to increase over the past few years (1). Although breast cancer is traditionally treated using various methods including surgery, chemotherapy and endocrine therapy, recurrence and metastasis still remain a serious issue in advanced stage patients (2).

Calreticulin (CRT) is a multifunctional calcium-binding protein that is predominantly expressed in the endoplasmic reticulum (3). This protein contains three functional domains: N-, P- and C-domains (4). It is involved in a variety of cellular processes, including protein folding, calcium homeostasis and cell adhesion (5). Extensive research has revealed that the expression of CRT is markedly increased in various types of cancers (6). A previous study demonstrated that the expression of CRT was positively associated with lymph node metastasis and clinical stages in breast cancer (7). An earlier study reported epithelial-mesenchymal transition (EMT)-like changes in the cellular phenotype in CRT-overexpressing Madin-Darby canine kidney cells (8). Furthermore, the mRNA expression of the EMT marker E-cadherin was reduced when CRT was overexpressed. Overexpressing CRT regulated EMT marker characteristics in gastric cancer cells (9). These results suggested that there may be a direct association between CRT and EMT.

Ecotropic viral integration site-1 (EVI-1) has a critical role in oncogenesis as a transcription factor (10). Overexpression of EVI-1 has frequently been observed in hematological malignancies (1113) and in several types of solid tumor (1416). However, very little is known about how EVI-1 regulates the oncogenesis of breast cancer.

Previous studies have demonstrated that EVI-1 is a master regulatory element in EMT (17,18). Nayak et al (19) reported that the expression of EVI-1 was strongly correlated with E-cadherin and N-cadherin in stage IV of colon cancer. Whether EVI-1 regulates the process of EMT in breast cancer via CRT remains unknown. In the present study, the overexpression of EVI-1 promoted cell proliferation, migration and invasion, and inhibited apoptosis in breast cancer cells. In addition, EVI-1 positively regulated the expression of CRT in breast cancer. Furthermore, a novel mechanistic pathway was investigated for how EVI-1 induced CRT activation in breast cancer. The results revealed that EVI-1 may be a potential effective therapeutic target in breast cancer.

Materials and methods

Cell lines and culture

The human breast cancer cell line MDA-MB-231 was purchased from the Cell Bank of the Chinese Academy of Sciences (Shanghai, China). Cells were cultured in L-15 (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 10% fetal bovine serum (FBS; HyClone; GE Healthcare Life Sciences, Logan, UT, USA) in a humidified atmosphere at 37°C without CO2.

Cell transfection

MDA-MB-231 breast cancer cells were cultured in a 6-well plate for 24 h at 37°C and then transfected with plasmids pcDNA3.1-EVI-1 and pSilencer-2.1-EVI-1 [EVI-1-short hairpin RNA (shRNA)]; both synthesized by Jrdun Biotechnology Co., Ltd., Shanghai, China). The following primers were used to amplify the EVI-1 sequence for cloning into the pcDNA vector: pcDNA3.1-EVI-1 forward, 5′-CCGGAATTCATGATCTTAGACGAATTTTACA-3′; pcDNA3.1-EVI-1 reverse, 5′-CGCGGATCCTCATACGTGGGGATAGCACTGGA-3′. The short hairpin RNA sequence (shRNA) in pSilencer2.1-EVI-1 (EVI-1-shRNA) was forward, 5′-CCTACGATCAGTCCTACCA-3′ and reverse, 5′-TGGTAGGACTGATCGTAGG-3′. For EVI-1 expression, 2.5 µg pcDNA3.1 control vector/pcDNA3.1-EVI-1 or pSilencer-2.1 control vector/pSilencer-2.1-EVI-1 were transfected into cells using Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) into 6-well plates (3×105 cells/well). For CRT expression, a small interfering RNA (siRNA) targeting CRT was used (synthesized by Jrdun Biotechnology Co., Ltd.), which had the following sequence: siRNA-CRT, 5′-GGAGCAGUUUCUGGACGGATT-3′. siRNA-NC was 5′-UUCUCCGAACGUGUCACGUTT-3′. siRNAs (90 pmol) were transfected into MDA-MB-231 cells (6-well plates; 3×105 cells/well) using the Lipofectamine® RNAiMAX Transfection Reagent (Thermo Fisher Scientific, Inc.) following the manufacturer's instructions (incubated for 1 day at 37°C).

Dual luciferase reporter system

The MDA-MB-231 cells were transfected psiCHECK2-CRT-WT and pcDAN3.1 empty vector, psiCHECK2-CRT-WT (the fragments of the promoter region of CRT) pcDAN3.1-EVI-1, psiCHECK2-CRT-MUT and pcDAN3.1 empty vector, or pcDAN3.1-EVI-1 and psiCHECK2-CRT-MUT using Lipofectamine® 2000 (Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. For all plasmids, 2 µg was used fro transfection and were synthesized by Jrdun Biotechnology Co., Ltd. After 48 h, the luciferase activity was measured using a Dual-Luciferase reporter assay system according to the manufacturer's instructions (Promega Corporation, Madison, WI, USA). Firefly luciferase activities were normalized by Renilla luciferase activities to control for transfection efficiency.

Electrophoresis mobility shift assay (EMSA)

EMSA was performed using the LightShift EMSA kit (Pierce; Thermo Fisher Scientific, Inc.). Nuclear protein extracts from breast cancer cells were prepared using NE-PER™ Nuclear and Cytoplasmic Extraction Reagents (Thermo Fisher Scientific, Inc.). Protein concentration was determined using the bicinchoninic acid assay. Oligonucleotides were synthesized for the CRT EVI-1 binding site and the mutation, and labeled with biotin by Beijing Genomics Institute (Shenzhen, China). The oligonucleotide sequences were as follows: EVI-1 binding site from CRT promoter region, 5′-GCTGGTTCTCAAATGCAAGATAAGAGCTGG-3′; and EVI-1 binding site-mutant from CRT promoter region 5′-GCTGGTTCTCATCGATCTGATAAGAGCTGG-3′. The binding reaction mixtures (2 µl 5X Gel shift buffer, 2 µl nuclear extracts, 1 µl labeled DNA probe, nuclease-free water to 10 µl) were incubated according to the manufacturer's instruction. For competition experiments, the corresponding unlabeled probe (cold probe) was used at 100-fold excess concentrations when compared with the labeled probe in the binding reaction. Reaction products were separated by 5% non-denaturing polyacrylamide gels in 0.5X Tris/Borate/EDTA buffer, and then the bands were transferred to a nylon membrane. The nylon membrane was visualized using the LightShift™ Chemiluminescent EMSA Kit (Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions.

Cell viability assay

Cell viability was assessed by a Cell Counting Kit-8 (CCK-8; Beyotime Institute of Biotechnology, Haimen, China) assay according to the manufacturer's instructions. Transfected cells were seeded into a 96-well plate at 5,000 cells/well. After transfection for 24, 48 and 72 h, 10 µl CCK-8 assay solution was added to each well and incubated for 1 h. Absorbance was then measured at 450 nm using a microplate reader.

Flow cytometry analysis of cell apoptosis

Cells were collected after transfection for 48 h, then an Annexin V-FITC Apoptosis Detection Kit (Beyotime Institute of Biotechnology) was used to assess apoptosis according to the manufacturer's instructions. The rate of apoptosis was analyzed by flow cytometry with BD FACSDiva software version 8.0 (BD Biosciences, San Jose, CA, USA).

Cell migration and invasion assays

The migration and invasion assays were performed using Transwell inserts. For the migration assay, at 48 h after transfection the indicated cells were starved for 24 h, 1×105 cells resuspended in serum-free medium and then added to the upper chamber of Transwell plates (Corning Incorporated, Corning, NY, USA). The lower chamber was filled with L15 medium and 10% FBS (Gibco; Thermo Fisher Scientific, Inc.). After incubation for 24 h, the cells attached to the lower surface of the membrane were fixed in 95% ethanol at 37°C for 30 min, and stained with 0.5% crystal violet for 30 min at room temperature; cells were then counted in five randomly selected fields under a light microscope (Olympus Corporation, Tokyo, Japan). For the cell invasion assay, the indicated cells were plated in Transwell polycarbonate membrane inserts precoated with a layer of diluted Matrigel (BD Biosciences). The remaining experimental procedures were consistent with those described for cell migration experiments.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA was isolated from MDA-MB-231 breast cancer cells using RNAiso Plus (Takara Biotechnology Co., Ltd., Dalian, China) according to manufacturer's instructions. cDNA was synthesized using the PrimeScript™ RT reagent Kit with gDNA Eraser (Takara Biotechnology Co., Ltd.), the temperature protocol for reverse transcription was as follows: 37°C for 15 min; 85°C for 5 sec and 4°C for 5 min. RT-qPCR was conducted using SYBR® Premix Ex Taq™ II (Takara Biotechnology Co., Ltd.). The expression of target genes was normalized to GAPDH. The reaction thermocycling conditions for qPCR were as follows: 95°C for 30 sec, followed by 40 cycles of 95°C for 5 sec, and 60°C for 31 sec. The sequences of the primers used for RT-qPCR are presented in Table I. All of the qPCR data were processed using the 2−ΔΔCq method (20).

Table I.

Primers used for reverse transcritpion-quantitative polymerase chain reaction.

Table I.

Primers used for reverse transcritpion-quantitative polymerase chain reaction.

PrimerForward (5′ to 3′)Reverse (5′ to 3′)
EVI-1 TTATAGAGCGATACAAGGGGGAG CGCCGTCTGATTATCTTGATGAG
Calreticulin AAGGAGCAGTTTCTGGACGG GCCGACAGAGCATAAAAGCG
Snail 1 TGCTGTCCCCGGCGATATT GTAGCTGCCCTGGTAGGTT
Slug TCCCTCGCGTGAGGTGAAGCA TCTGTCTCTGGAGCCAGGTGC
E-cadherin TGCAGGGGCAGCCATCTCCT TTCCCCCAGCGTCCTCCACC
N-cadherin CGGCCGCTGCCACCACAGTT AGTCCCCACGCTGCTCTTCT
BAX GGTCGCTTGTGGCCTTTTTC TGCTGCATTGTTCCCATAGAG
Caspase-3 ATGGAGCGAATCAATGGACTC CTGTACCAGACCGAGATGTCA
BCL-2 GACTGGGGGAGGATTGTGG CCGGTTCAGGTACTCAGTCA
GAPDH CTCCTCCTGGCCTCGCTGT GCTGTCACCTTCACCGTTCC

[i] EVI-1, ecotropic viral integration site-1; Snail 1, zinc finger protein SNAI1; Slug, zinc finger protein SNAI2; BAX, apoptosis regulator BAX; BCL-2, apoptosis regulator Bcl-2.

Western blot analysis

Total protein was isolated from MDA-MB-231 breast cancer cells using radioimmunoprecipitation assay lysis buffer (Beyotime Institute of Biotechnology) and protein concentration was measured using a bicinchoninic acid assay. Equal amounts of protein (20 µg/lane) separated by SDS-PAGE on 8% gels and then transferred to polyvinylidene difluoride membranes. The membranes were blocked with 5% nonfat milk for 2 h at room temperature and then incubated with rabbit polyclonal anti-EVI-1 (1:1,000; cat. no. ab28457), rabbit polyclonal anti-CRT (1:1,000; cat. no. ab2907), rabbit polyclonal anti-E-cadherin (1:1,000; cat. no. ab15148), rabbit polyclonal anti-N-cadherin (1:1,000; cat. no. ab18203), rabbit polyclonal anti-apoptosis regulator BAX (BAX; 1:1,000; cat. no. ab53154), rabbit polyclonal anti-caspase-3 (1:1,000; cat. no. ab13847), rabbit polyclonal anti-zinc finger protein SNAI1 (Snail 1; 1:1,000; cat. no. ab110490), rabbit polyclonal anti-zinc finger protein SNAI2 (Slug; 1:1,000; cat. no. ab27568) and rabbit polyclonal anti-apoptosis regulator Bcl-2 (BCL-2; 1:1,000; cat. no. ab59348) primary antibodies at 4°C overnight. GAPDH (1:3,000; cat. no. ab9485) was used as the internal control to ensure equal protein loading. All antibodies were purchased from Abcam (Cambridge, UK). The signal was developed with Pierce™ ECL Western Blotting Substrate (Thermo Fisher Scientific, Inc.) following incubation with the corresponding goat anti-rabbit horseradish peroxidase-conjugated (IgG H&L) secondary antibody at room temperature for 1 h (1:2,000; cat. no. ab205718; Abcam).

Statistical analysis

All experiments were repeated three times independently, and the data are presented as the mean ± standard deviation. One-way analysis of variance followed by Bonferroni's multiple comparisons procedure was used to compare multiple groups and paired t-test was used to compare two groups. Analysis was performed using SPSS 19.0 statistical software (IBM Corp., Armonk, NY, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

EVI-1 may target the CRT promoter region in vitro

The expression of EVI-1 was markedly upregulated following transfection with pcDNA3.1-EVI-1, whereas EVI-1 was downregulated after transfection with pSilencer-2.1-EVI-1 (Fig. 1A). Subsequently, the expression of CRT was analyzed at the mRNA and protein levels. CRT mRNA expression was increased following transfection with pcDNA3.1-EVI-1; however, pSilencer-2.1-EVI-1 reduced CRT mRNA expression (Fig. 1B). In addition, a similar effect on CRT protein expression was observed (Fig. 1C). To further demonstrate that CRT was a target of EVI-1, dual luciferase reporter and EMSA assays were performed. Luciferase activity was increased in cells co-transfected with pcDNA3.1-EVI-1 and the psiCHECK2-CRT-WT reporter vector. However, pcDNA3.1-EVI-1 had no effect on the luciferase activity of cells co-transfected with the psiCHECK2-CRT-MUT reporter vector (Fig. 1D). Based on the EMSA gel shifts, a protein bound to the CRT promoter region, and this protein may be EVI-1. No specific gel shift was observed in lane 1; however, a marked shift was observed in lane 2. In addition, no shift was observed with the 100-fold excess of the unlabeled EVI-1 probe (cold probe) in lane 3, and there was no obvious interaction between the nuclear extract and the mutated EVI-1 probe in lane 4 (Fig. 1E). In summary, these results indicated that EVI-1 positively regulated CRT expression, and EVI-1 is likely to regulate its expression by binding to the promoter region of CRT in vitro experiment.

EVI-1 promotes cell proliferation and inhibits cell apoptosis in human breast cancer

The overexpression of EVI-1 markedly increased cell proliferation, while downregulation of EVI-1 inhibited cell proliferation (Fig. 2A). Overexpression of EVI-1 also decreased the apoptosis of breast cancer cells, while EVI-1 silencing increased the rate of apoptosis in breast cancer cells (Fig. 2B). Subsequently, expression of apoptotic pathway-associated genes, including caspase-3, Bax and Bcl-2, was investigated. As indicated in Fig. 2C and D, a marked decrease in caspase-3 and Bax expression, and a marked increase in Bcl-2 expression was observed when EVI-1 was overexpressed. By contrast, knockdown of EVI-1 increased the expression of caspase-3 and Bax, and reduced the expression of Bcl-2.

EVI-1 promotes cell migration and invasion in human breast cancer

The present study further investigated the effect of EVI-1 on cell migration and invasion. The results revealed that overexpression of EVI-1 effectively promoted cell migration and invasion, and silencing EVI-1 expression markedly suppressed cell migration and invasion in human breast cancer cells compared with the respective empty vector controls (Fig. 3).

EVI-1 regulates EMT-related gene expression in breast cancer

Previous studies have indicated that the abnormal expression of CRT regulates the expression of EMT-associated genes (8,9), and the present study revealed that EVI-1 positively regulated CRT expression. As indicated in Fig. 4A and B, knockdown of CRT using siRNA-CRT decreased the expression of Snail 1, Slug and N-cadherin, and increase the expression of E-cadherin.

To determine whether EVI-1 can regulate EMT-associated gene expression, RT-qPCR and western blot analyses were performed to investigate the expression of EMT-associated genes. As demonstrated in Fig. 4C and D, a marked increase in Snail 1, Slug, and N-cadherin expression, and a marked decrease in E-cadherin expression were observed when EVI-1 was upregulated. By contrast, silencing EVI-1 decreased the expression of Snail 1, Slug and N-cadherin, and increased the expression of E-cadherin. These results suggested that EVI-1 may influence the process of EMT in breast cancer cells.

Discussion

EVI-1, an oncogene located on chromosome 3q26, specifically binds to promoter DNA sequences and has a role in transcriptional regulation (10). Multiple studies have revealed that EVI-1 is upregulated in leukemia, ependymoma, ovarian cancer, breast cancer and colon cancer, and the downregulation of EVI-1 expression inhibited the proliferation of tumor cells (1113). In addition, increased EVI-1 expression has also been revealed to be a risk factor for poor prognosis in patients with leukemia (2123). However, whether EVI-1 exerts specific regulatory effects on breast cancer has not been reported thoroughly. In the present study, overexpression of EVI-1 increased the proliferative ability of breast cancer.

c-Jun N-terminal protein kinase (JNK) has a critical role in the process of cell apoptosis and the mechanism of JNK-mediated apoptosis is associated with the regulated expression of apoptosis-associated proteins, including Bax and Bcl-2 (2426). However, EVI-1 inhibits the activity of JNK and the subsequent cell apoptosis by interfering with the interactions between JNK and its physiological substrates (27). It was hypothesized that EVI-1 may regulate breast cancer apoptosis and the present study demonstrated that cell apoptosis was inhibited by the overexpression of EVI-1 and enhanced by the downregulation of EVI-1. Bcl-2 has a major role in the signal transduction pathways of cell apoptosis (28). Bcl-2 and Bax are the most representative genes for inhibiting and promoting apoptosis, and Bax is the main regulator of Bcl-2 (29). Bcl-2 can bind to Bax to form a heterodimer, and the increased expression of Bax can antagonize the effect of Bcl-2 and promote cell apoptosis (3032). Caspase-3 is the key mediator of cell apoptosis and has a role in a variety of apoptotic signaling pathways (3335). Previous studies have reported that overexpression of Bcl-2 can effectively inhibit the activation of caspase-3, thus inhibiting the occurrence of apoptosis (36). In the present study, RT-qPCR and western blotting revealed that EVI-1 overexpression/silencing altered the expression of the aforementioned proteins associated with apoptosis, and subsequently the process of apoptosis, in breast cancer. When EVI-1 was highly expressed, the expression of Bax and caspase-3 was reduced, and Bcl-2 was increased.

A number of previous studies investigating CRT demonstrated that the abnormal expression of CRT leads to changes in certain biological processes, including cell invasion and proliferation, and may be associated with the cancer occurrence, development and prognosis (3,6,3739). However, research on the association between EVI-1 and CRT is rarely reported. EMSA is an invaluable tool to study interaction of proteins with DNA. The experiment can simulate the specific binding of protein and DNA in vitro. EMSA has some limitations in the process of reconstructing the binding between proteins and DNA in vivo. However, EMSA is still a good method to predict the binding between EVI-1 and CRT in vitro in this study. By using EMSA and dual luciferase assays, the present study revealed that EVI-1 may bind to the promoter region of CRT, and positively regulate its expression. Thus, it is reasonable to suggest that EVI-1 may affect some of the biological functions of breast cancer by regulating the expression of CRT.

EMT is a process by which epithelial cells gain the phenotype of mesenchymal cells, and the close connection between cells with the extracellular matrix and neighboring cells weakens or is completely abolished; consequently, cell migration and invasion is enhanced (40). In the process of EMT, the expression of several proteins, including E-cadherin, N-cadherin, Snail 1 and Slug, is altered (4144). Previous studies have revealed that the overexpression of CRT can inhibit the expression of E-cadherin and enhance cell migration abilities (8,45). Liu et al (9) reported that the overexpression of CRT promoted cell invasion and metastasis in gastric cancer by regulating the expression of Snail 1 and E-cadherin. In the present study, the results demonstrated that increased expression of EVI-1 significantly reduced the expression of E-cadherin and enhanced the expression of N-cadherin, Snail 1 and Slug. When the expression of EVI-1 was suppressed, the expression of E-cadherin was increased, and N-cadherin, Snail 1 and Slug were reduced. It was speculated that EVI-1 may regulate EMT-associated genes via the CRT pathway in breast cancer.

In conclusion, the results of the present study revealed that EVI-1 overexpression promotes cell proliferation, migration and invasion, and inhibits apoptosis in breast cancer cells. EVI-1 is likely to bind to CRT and positively regulate the expression of CRT in vitro environment; it was concluded that EVI-1 may affect EMT-associated genes by regulating the expression of CRT in breast cancer.

Acknowledgements

The authors would like to thank Professor Ping Ma (Molecular Oncology Laboratory of Cancer Research Institute, The First Affiliated Hospital of China Medical University, Shenyang, China) for designing the study and for their excellent technical assistance.

Funding

No funding was received.

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

LW designed the study, performed the experiments and drafted the manuscript. TW, DH and XL analyzed and interpreted the experimental data. YJ conceived the study and participated in its design and coordination.

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 

Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J and Jemal A: Global cancer statistics, 2012. CA Cancer J Clin. 65:87–108. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Ghislain I, Zikos E, Coens C, Quinten C, Balta V, Tryfonidis K, Piccart M, Zardavas D, Nagele E, Bjelic-Radisic V, et al: Health-related quality of life in locally advanced and metastatic breast cancer: Methodological and clinical issues in randomised controlled trials. Lancet Oncol. 17:e294–e304. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Goitea VE and Hallak ME: Calreticulin and arginylated calreticulin have different susceptibilities to proteasomal degradation. J Biol Chem. 290:16403–16414. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Holmström MO, Ocias LF, Kallenbach K, Kjær L, Kristensen TK, Pallisgaard N, Petersen BL, Skov V, de Stricker K, Larsen TS, et al: New disease markers within the chronic myeloproliferative neoplasms. Ugeskr Laeger. 1772015.(In Danish).

5 

Wang WA, Groenendyk J and Michalak M: Calreticulin signaling in health and disease. Int J Biochem Cell Biol. 44:842–846. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Zamanian M, Veerakumarasivam A, Abdullah S and Rosli R: Calreticulin and cancer. Pathol Oncol Res. 19:149–154. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Lwin ZM, Guo C, Salim A, Yip GW, Chew FT, Nan J, Thike AA, Tan PH and Bay BH: Clinicopathological significance of calreticulin in breast invasive ductal carcinoma. Mod Pathol. 23:1559–1566. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Hayashida Y, Urata Y, Muroi E, Kono T, Miyata Y, Nomata K, Kanetake H, Kondo T and Ihara Y: Calreticulin represses E-cadherin gene expression in Madin-Darby canine kidney cells via Slug. J Biol Chem. 281:32469–32484. 2006. View Article : Google Scholar : PubMed/NCBI

9 

Liu SH, Lee WJ, Lai DW, Wu SM, Liu CY, Tien HR, Chiu CS, Peng YC, Jan YJ, Chao TH, et al: Honokiol confers immunogenicity by dictating calreticulin exposure, activating ER stress and inhibiting epithelial-to-mesenchymal transition. Mol Oncol. 9:834–849. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Wieser R: The oncogene and developmental regulator EVI1: Expression, biochemical properties, and biological functions. Gene. 396:346–357. 2007. View Article : Google Scholar : PubMed/NCBI

11 

Daghistani M, Marin D, Khorashad JS, Wang L, May PC, Paliompeis C, Milojkovic D, De Melo VA, Gerrard G, Goldman JM, et al: EVI-1 oncogene expression predicts survival in chronic-phase CML patients resistant to imatinib treated with second-generation tyrosine kinase inhibitors. Blood. 116:6014–6017. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Gröschel S, Lugthart S, Schlenk RF, Valk PJ, Eiwen K, Goudswaard C, van Putten WJ, Kayser S, Verdonck LF, Lübbert M, et al: High EVI1 expression predicts outcome in younger adult patients with acute myeloid leukemia and is associated with distinct cytogenetic abnormalities. J Clin Oncol. 28:2101–2107. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Vázquez I, Maicas M, Cervera J, Agirre X, Marin-Béjar O, Marcotegui N, Vicente C, Lahortiga I, Gomez-Benito M, Carranza C, et al: Down-regulation of EVI1 is associated with epigenetic alterations and good prognosis in patients with acute myeloid leukemia. Haematologica. 96:1448–1456. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Patel JB, Appaiah HN, Burnett RM, Bhat-Nakshatri P, Wang G, Mehta R, Badve S, Thomson MJ, Hammond S, Steeg P, et al: Control of EVI-1 oncogene expression in metastatic breast cancer cells through microRNA miR-22. Oncogene. 30:1290–1301. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Koos B, Bender S, Witt H, Mertsch S, Felsberg J, Beschorner R, Korshunov A, Riesmeier B, Pfister S, Paulus W and Hasselblatt M: The transcription factor evi-1 is overexpressed, promotes proliferation, and is prognostically unfavorable in infratentorial ependymomas. Clin Cancer Res. 17:3631–3637. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Deng X, Cao Y, Liu Y, Li F, Sambandam K, Rajaraman S, Perkins AS, Fields AP, Hellmich MR, Townsend CM Jr, et al: Overexpression of Evi-1 oncoprotein represses TGF-β signaling in colorectal cancer. Mol Carcinog. 52:255–264. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Venkov CD, Link AJ, Jennings JL, Plieth D, Inoue T, Nagai K, Xu C, Dimitrova YN, Rauscher FJ and Neilson EG: A proximal activator of transcription in epithelial-mesenchymal transition. J Clin Invest. 117:482–491. 2007. View Article : Google Scholar : PubMed/NCBI

18 

Venkov C, Plieth D, Ni T, Karmaker A, Bian A, George AL Jr and Neilson EG: Transcriptional networks in epithelial-mesenchymal transition. PLoS One. 6:e253542011. View Article : Google Scholar : PubMed/NCBI

19 

Nayak KB, Sajitha IS, Kumar TRS and Chakraborty S: Ecotropic viral integration site 1 promotes metastasis independent of epithelial mesenchymal transition in colon cancer cells. Cell Death Dis. 9:182018. View Article : Google Scholar : PubMed/NCBI

20 

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. View Article : Google Scholar : PubMed/NCBI

21 

Goyama S and Kurokawa M: Evi-1 as a critical regulator of leukemic cells. Int J Hematol. 91:753–757. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Lugthart S, van Drunen E, van Norden Y, van Hoven A, Erpelinck CA, Valk PJ, Beverloo HB, Löwenberg B and Delwel R: High EVI1 levels predict adverse outcome in acute myeloid leukemia: Prevalence of EVI1 overexpression and chromosome 3q26 abnormalities underestimated. Blood. 111:4329–4337. 2008. View Article : Google Scholar : PubMed/NCBI

23 

Barjesteh van Waalwijk van Doorn-Khosrovani S, Erpelinck C, van Putten WL, Valk PJ, van der Poel-van de Luytgaarde S, Hack R, Slater R, Smit EM, et al: High EVI1 expression predicts poor survival in acute myeloid leukemia: A study of 319 de novo AML patients. Blood. 101:837–845. 2003. View Article : Google Scholar : PubMed/NCBI

24 

Dhanasekaran DN and Reddy EP: JNK signaling in apoptosis. Oncogene. 27:6245–6251. 2008. View Article : Google Scholar : PubMed/NCBI

25 

Aoki H, Kang PM, Hampe J, Yoshimura K, Noma T, Matsuzaki M and Izumo S: Direct activation of mitochondrial apoptosis machinery by c-Jun N-terminal kinase in adult cardiac myocytes. J Biol Chem. 277:10244–10250. 2002. View Article : Google Scholar : PubMed/NCBI

26 

Yang CB, Pei WJ, Zhao J, Cheng YY, Zheng XH and Rong JH: Bornyl caffeate induces apoptosis in human breast cancer MCF-7 cells via the ROS- and JNK-mediated pathways. Acta Pharmacol Sin. 35:113–123. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Kurokawa M, Mitani K, Yamagata T, Takahashi T, Izutsu K, Ogawa S, Moriguchi T, Nishida E, Yazaki Y and Hirai H: The evi-1 oncoprotein inhibits c-Jun N-terminal kinase and prevents stress-induced cell death. Embo J. 19:2958–2968. 2000. View Article : Google Scholar : PubMed/NCBI

28 

Garrison SP, Phillips DC, Jeffers JR, Chipuk JE, Parsons MJ, Rehg JE, Opferman JT, Green DR and Zambetti GP: Genetically defining the mechanism of Puma- and Bim-induced apoptosis. Cell Death Differ. 19:642–649. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Rossé T, Olivier R, Monney L, Rager M, Conus S, Fellay I, Jansen B and Borner C: Bcl-2 prolongs cell survival after Bax-induced release of cytochrome c. Nature. 391:496–499. 1998. View Article : Google Scholar : PubMed/NCBI

30 

Brooks C and Dong Z: Regulation of mitochondrial morphological dynamics during apoptosis by Bcl-2 family proteins: A key in Bak? Cell Cycle. 6:3043–3047. 2007. View Article : Google Scholar : PubMed/NCBI

31 

Yang J, Liu X, Bhalla K, Kim CN, Ibrado AM, Cai J, Peng TI, Jones DP and Wang X: Prevention of apoptosis by Bcl-2: Release of cytochrome c from mitochondria blocked. Science. 275:1129–1132. 1997. View Article : Google Scholar : PubMed/NCBI

32 

Cheng EH, Kirsch DG, Clem RJ, Ravi R, Kastan MB, Bedi A, Ueno K and Hardwick JM: Conversion of Bcl-2 to a Bax-like death effector by caspases. Science. 278:1966–1968. 1997. View Article : Google Scholar : PubMed/NCBI

33 

Zhang Y, Goodyer C and LeBlanc A: Selective and protracted apoptosis in human primary neurons microinjected with active caspase-3, −6, −7, and −8. J Neurosci. 20:8384–8389. 2000. View Article : Google Scholar : PubMed/NCBI

34 

Cryns V and Yuan J: Proteases to die for. Genes Dev. 12:1551–1570. 1998. View Article : Google Scholar : PubMed/NCBI

35 

Yang Y, Duan W, Liang Z, Yi W, Yan J, Wang N, Li Y, Chen W, Yu S, Jin Z and Yi D: Curcumin attenuates endothelial cell oxidative stress injury through Notch signaling inhibition. Cell Signal. 25:615–629. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Lončarević-Vasiljković N, Milanović D, Pešić V, Tešić V, Brkić M, Lazić D, Avramović V and Kanazir S: Dietary restriction suppresses apoptotic cell death, promotes Bcl-2 and Bcl-xl mRNA expression and increases the Bcl-2/Bax protein ratio in the rat cortex after cortical injury. Neurochem Int. 96:69–76. 2016. View Article : Google Scholar : PubMed/NCBI

37 

Sazawal S, Singh N, Mahapatra M and Saxena R: Calreticulin mutation profile in Indian patients with primary myelofibrosis. Hematology. 20:567–570. 2015. View Article : Google Scholar : PubMed/NCBI

38 

Chiang WF, Hwang TZ, Hour TC, Wang LH, Chiu CC, Chen HR, Wu YJ, Wang CC, Wang LF, Chien CY, et al: Calreticulin, an endoplasmic reticulum-resident protein, is highly expressed and essential for cell proliferation and migration in oral squamous cell carcinoma. Oral Oncol. 49:534–541. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Raghavan M, Wijeyesakere SJ, Peters LR and Del Cid N: Calreticulin in the immune system: Ins and outs. Trends Immunol. 34:13–21. 2013. View Article : Google Scholar : PubMed/NCBI

40 

Li X, Wang X, Tan Z, Chen S and Guan F: Role of glycans in cancer cells undergoing epithelial-mesenchymal transition. Front Oncol. 6:332016. View Article : Google Scholar : PubMed/NCBI

41 

Morra L and Moch H: Periostin expression and epithelial-mesenchymal transition in cancer: A review and an update. Virchows Arch. 459:465–475. 2011. View Article : Google Scholar : PubMed/NCBI

42 

Xiong H, Hong J, Du W, Lin YW, Ren LL, Wang YC, Su WY, Wang JL, Cui Y, Wang ZH and Fang JY: Roles of STAT3 and ZEB1 proteins in E-cadherin down-regulation and human colorectal cancer epithelial-mesenchymal transition. J Biol Chem. 287:5819–5832. 2012. View Article : Google Scholar : PubMed/NCBI

43 

Jiang Y, Dey S and Matsunami H: Calreticulin: Roles in cell-surface protein expression. Membranes (Basel). 4:630–641. 2014. View Article : Google Scholar : PubMed/NCBI

44 

Lu Z, Wang J, Zheng T, Liang Y, Yin D, Song R, Pei T, Pan S, Jiang H and Liu L: FTY720 inhibits proliferation and epithelial-mesenchymal transition in cholangiocarcinoma by inactivating STAT3 signaling. BMC Cancer. 14:7832014. View Article : Google Scholar : PubMed/NCBI

45 

Ihara Y, Inai Y and Ikezaki M: Alteration of integrin-dependent adhesion and signaling in EMT-like MDCK cells established through overexpression of calreticulin. J Cell Biochem. 112:2518–2528. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2019
Volume 19 Issue 3

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wu L, Wang T, He D, Li X and Jiang Y: EVI‑1 acts as an oncogene and positively regulates calreticulin in breast cancer Retraction in /10.3892/mmr.2023.13112. Mol Med Rep 19: 1645-1653, 2019
APA
Wu, L., Wang, T., He, D., Li, X., & Jiang, Y. (2019). EVI‑1 acts as an oncogene and positively regulates calreticulin in breast cancer Retraction in /10.3892/mmr.2023.13112. Molecular Medicine Reports, 19, 1645-1653. https://doi.org/10.3892/mmr.2018.9796
MLA
Wu, L., Wang, T., He, D., Li, X., Jiang, Y."EVI‑1 acts as an oncogene and positively regulates calreticulin in breast cancer Retraction in /10.3892/mmr.2023.13112". Molecular Medicine Reports 19.3 (2019): 1645-1653.
Chicago
Wu, L., Wang, T., He, D., Li, X., Jiang, Y."EVI‑1 acts as an oncogene and positively regulates calreticulin in breast cancer Retraction in /10.3892/mmr.2023.13112". Molecular Medicine Reports 19, no. 3 (2019): 1645-1653. https://doi.org/10.3892/mmr.2018.9796