Open Access

Rotenone restrains colon cancer cell viability, motility and epithelial‑mesenchymal transition and tumorigenesis in nude mice via the PI3K/AKT pathway

Retraction in: /10.3892/ijmm.2023.5340

  • Authors:
    • Wenbo Xiao
    • Yongwei Liu
    • Maolin Dai
    • Yu Li
    • Renqun Peng
    • Shuangjiang Yu
    • Hao Liu
  • View Affiliations

  • Published online on: June 10, 2020     https://doi.org/10.3892/ijmm.2020.4637
  • Pages: 700-708
  • Copyright: © Xiao 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

Rotenone, a natural hydrophobic pesticide, has been reported to display anticancer activity in a variety of cancer cells. However, the mechanism of rotenone on colon cancer (CC) cell migration, invasion and metastasis is still unknown. In the present study, the cytotoxicity of rotenone on CC cells were detected by the Cell Counting Kit‑8 assay and confirmed by clone formation assay. The effects of rotenone on CC cell invasion and migration activity were determined in vitro by Transwell invasion and wound healing assays, respectively. In addition, to reveal whether rotenone affected the epithelial‑mesenchymal‑transition (EMT) process, reverse transcription‑quantitative PCR, western blotting and immunofluorescence assays were used to detect the expression of EMT markers. The expression levels of the key markers of the PI3K/AKT pathway after rotenone treatment alone or in combination with a PI3K/AKT signaling activator in CC were also detected by western blotting. Finally, the in vivo antitumor effects of rotenone were evaluated in a subcutaneous xenotransplant tumor model treated with an intraperitoneal injection of rotenone. The results of the present study demonstrated that rotenone treatment induced CC cell cytotoxicity and greater effects were observed with increasing concentrations and inhibited cell proliferation compared with untreated cells. In vitro cell function assays revealed that rotenone inhibited CC cell migration, invasion and EMT compared with untreated cells. Mechanically, the phosphorylation levels of AKT and mTOR were downregulated in rotenone‑treated CC cells compared with untreated cells. Additionally, AKT and mTOR phosphorylation levels were increased by the PI3K/AKT signaling activator insulin‑like growth factor 1 (IGF‑1), which was reversed by rotenone treatment. The cell function assays confirmed that the IGF‑1‑activated cell proliferation, migration and invasion were decreased by rotenone treatment. These results indicated that rotenone affected CC cell proliferation and metastatic capabilities by inhibiting the PI3K/AKT/mTOR signaling pathway. In addition, rotenone inhibited tumor growth and metastatic capability of CC, which was confirmed in a xenograft mouse model. In conclusion, the present study revealed that rotenone inhibited CC cell viability, motility, EMT and metastasis in vitro and in vivo by inhibiting the PI3K/AKT/mTOR signaling pathway.

Introduction

Colon carcinoma (CC) was the most commonly diagnosed cancer and the third most deadly disease in the world in 2015 (1). Despite extensive research and clinic trials in the diagnosis and treatment of CC, conventional approaches, including cytotoxic chemotherapy, have failed to substantially increase the therapeutic capacity (2). As a result, the 5-year overall survival rate of patients with CC undergoing chemotherapy is only 10% (3). Liver, lymph node and lung metastases have been reported in more than half of CC cases (4). Therefore, it is crucial to understand the carcinogenesis of CC and identify more effective treatments.

Rotenone, a natural hydrophobic pesticide isolated from the roots and barks of the Derris and Lonchorcarpus species (such as sweet potato and sandalwood seeds), has been reported to present anticancer activity in a variety of cancer cells (5). Previous studies have indicated that deguelin, a rotenoid, exerts a chemopreventative effect in decreasing the occurrence of tobacco-induced lung tumorigenesis (6). The partial mechanisms of rotenone anticarcinogenesis have been described as the suppression of cyclooxygenase-2 (5), downregulation of ornithine decarboxylase (7) and inhibition of the PI3K/AKT pathway (8). In addition, low-dose rotenone inhibits the migration and invasion of oral cancer cells by regulating tumor nuclear factor-κB (NF-κB) activity and matrix metallpproteinase-2 (9,10). A number of studies have confirmed that rotenone induces apoptosis in vitro and in vivo in a variety of types of cancer including breast and colorectal cancer and hepatocellular carcinoma (11,12). Rotenone has been demonstrated to affect the apoptosis of CC cells, which results in cell cycle arrest in the G1-S phase (12,13). However, the mechanism and pathways of the antitumor effect of rote-none on CC cell migration, invasion and metastasis is still unknown.

Epithelial-mesenchymal transition (EMT) is a process by which epithelial cells lose polarity and adhesiveness and thus transform into mesenchymal cells (14). Emerging evidence has demonstrated that EMT is of vital importance in tumor cell invasion and metastasis (15-17). Rotenone has been reported to target NF-κB to induce EMT reversion and apoptosis in pancreatic cancer (16). In addition, rotenone can prevent the metastasis and EMT of human non-small cell lung cancer cells by modulating NIMA-related kinase 2 (18). However, the effects and underlying mechanisms of rotenone on CC metastasis and EMT require further study. The present study aimed to determine the effects of rotenone on CC cell viability, motility, metastasis and EMT in vitro and in vivo, and to identify the underlying mechanisms associated with thePI3K/AKT/mTOR signaling pathway in CC cells.

Materials and methods

Cell lines and regents

Colon cancer cell lines SW480 and SW620 as well as the normal human colon cells CRL-1790 were obtained from The Cell Bank of Type Culture Collection of the Chinese Academy of Sciences and cultured in DMEM (HyClone; GE Healthcare Life Sciences) supplemented with 10% FBS (Sigma-Aldrich; Merck KGaA) and penicillin/streptomycin (Gibco; Thermo Fisher Scientific, Inc.) at 37°C with 5% CO2. Rotenone was obtained from Sigma-Aldrich; Merck KGaA, and a stock of 10 mM was diluted in DMSO Hybri-Max (Sigma-Aldrich; Merck KGaA). Colon cells were treated with 0, 0.01, 0.05, 0.1, 1, 5, 10 and 20 µM rotenone for 48 h as previously described (11).

Ethynyl-2-deoxyuridine (EdU) incorporation assay

EdU incorporation assay was performed using an EdU Apollo DNA In Vitro kit (Guangzhou RiboBio Co., Ltd.) according to the manufacturer's instructions. Following treatment with rote-none, the cells were incubated with 50 µM EdU as previously described (19,20). Subsequently, the cells were incubated with 1X Hoechst 33342 solution and observed under a confocal microscope (magnification, ×400). The EdU incorporation rate was expressed as the ratio of EdU-positive cells to Hoechst-positive cells.

In vitro cell viability assay

Cell viability was determined by the Cell Counting Kit-8 (CCK-8) assay (Dojindo Molecular Technologies, Inc.). SW480 and SW620 cells (4×103) were seeded into a 96-well plate and cultured overnight in an incubator at 37°C. The cells were treated with 10 µM rotenone alone or in presence of10 nMPI3K/AKT signaling activator insulin-like growth factor 1 (IGF-1) in DMEM for 24 h. CCK-8 solution (10 µl) was added into each well and incu-bated at 37°C for 3 h. The OD value of the reaction solution at 450 nm was evaluated using an Anthos 2010 microplate reader (Biochrom, Ltd.).

In vitro clone formation assay

SW480 and SW620 cells (100 cells/well) were seeded in 6-well plates and incubated overnight at 37°C, followed by treatment with 10 µM rotenone alone or with 10 nMIGF-1 at 37°C. After a 2-week incubation, the cell colonies were stained with 0.5% crystal violet for 10 min at room temperature, the 6-well plate was slowly immersed in tap water to remove the redundant stain and allowed to dry at room temperature for 2 days. The cell colonies (>50 cells) were counted using ImageJ software version 1.48u (National Institutes of Health).

In vitro scratch assay

SW480 and SW620 cells were seeded into 6-well plates (~1×106 cells/well) and incubated at 37°C with 5% CO2 for 24 h until ~100% confluence. Subsequently, the cells were serum starved and treated with 10 µM rote-none for 3 h at room temperature. Wounds were created by scratching the cell monolayers with a 10-µl pipette tip, and the cells were incubated in DMEM containing 1% fetal calf serum (Sigma-Aldrich; Merck KGaA) for 24 h, followed by removal of medium. Images were captured to estimate the cell migration at 0 and 24 h. The scratch width of the cells on both sides of the scratch was measured, and the relative migration rate of cells was calculated as the relative migration distance divided by the scratch width at 0 h.

In vitro Transwell invasion assay

For cell invasive ability assessment, Transwell chambers (Corning, Inc.) were precoated with Matrigel (BD Biosciences) at 4°C overnight. First, 1×105 SW480 and SW620 cells were treated with 10 µM rotenone and incubated for 24 h in advance on the upper chamber with 8-µm pores with 500 µl serum-free DMEM. The lower chambers were filled with 700 µl DMEM containing 10% FBS. After 24 h, the cells remaining on the upper side of the membrane were removed carefully with a cotton swab, and the cells on the underside of the membrane were incubated with 0.1% crystal violet for 30 min at room temperature and counted under a light microscope in three random high-power fields (magnification, ×200).

Reverse transcription-quantitative PCR (RT-qPCR)

Total RNAs from CC cell lines were acquired using the TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.), and the concentration and purity of the RNA were determined. RNA was reverse-transcribed into cDNA using a RevertAid Fist Strand cDNA Synthesis kit (Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. QPCR was performed using the ABI Q6 detection system (Applied Biosystems; Thermo Fisher Scientific, Inc.) with 1.0 µl cDNA and a SYBR® Green Real-Time PCR Master mix (Takara Biotechnology Co., Ltd.). The PCR amplification included initial denaturation at 95°C for 3 min, followed by 40 cycles of 94°C for 30 sec, 60°C for 30 sec and 70°C for 30 sec. Relative quantification and calculations were performed using the comparative quantitation cycle method (2−ΔΔCq) (21). GAPDH was used as the internal reference. The primer sequences used were as follows: E-cadherin forward, 5′-CCATTCCCAATGAGGCTGGT-3′ and reverse, 5′-GGCTTTTCTGTGACATCCGC-3′; vimentin forward, 5′-ACATGGTGGAAACCGAGGAT-3′ and reverse, 5′-TCCATTTCCCGCATTTGGT-3′; Snail forward, 5′-ACATGGTGGAAACCGAGGAT-3′ and reverse, 5′-GGTGGTGGAAGGAATAACGC-3′; and GAPDH forward, 5′-CGCTCCACCTTCAAGTATGC-3′ and reverse, 5′-GTCCACCACCCTGTTGCTGTAG-3′.

Western blotting

Protein samples were preprocessed with RIPA lysis buffer (Beijing Solarbio Science & Technology Co., Ltd.). The protein (30 µg/lane) was separated by 10% SDS-PAGE and transferred to PVDF membranes (EMD Millipore). The membranes were blocked with 10% non-fat milk in 1X TBS + 0.1% Tween-20 (TBST) for 2 h at room temperature. Subsequently, the membranes were incubated with primary antibodies against cleaved caspase-3 (1:1,000; ab2302), Bax (1:1,000; ab32503), Bcl-2 (1:1,000; ab32124), E-cadherin (1:50; ab1416), vimentin (1:1,000; ab92547), Snail (1:1,000; ab229701), AKT (1:10,000; ab179463), phosphor (p)-AKT (T308; 1:1,000; ab38449), mTOR (1:2,000; ab2732), p-mTOR (1:1,000; ab109268) and anti-GAPDH (1:10,000; ab181602) at 4°C overnight. All primary antibodies were obtained from Abcam. Subsequently, the membranes were washed three times with TBST and incubated with a horse-radish peroxidase-conjugated goat-anti-mouse IgG (1:1,000; A7007; Beyotime Institute of Biotechnology) for 1 h at room temperature. Specific protein bands were developed using an enhanced chemiluminescence reagent (Thermo Fisher Scientific, Inc.) and visualized using a ChemiDoc MP system (Bio-Rad Laboratories, Inc.).

Immunofluorescence

SW480 and SW620 cells (1×105 cells/dish) were fixed in 4% paraformaldehyde at 25°C for 15 min and treated with 0.1% Triton X-100 for 15 min. After blocking with 5% FBS for 1 h at room temperature, the cells were incubated with an anti-vimentin antibody (1:1,000; ab92547) for 2 h at room temperature. After washing three times with PBS, the cells were fixed with an anti-quenching sealer Fluoromount-G (Southern Biotech) and observed under fluorescence microscopyin three random high-power fields (magnification, ×400).

Mice and xenograft tumor model establishment

Male BALB/c athymic nude mice (6-week-old) were obtained from the Chongqing Medical University Animal Center and maintained in specific pathogen-free grade filter-top cages with a 12-h light/dark cycle in a controlled temperature (24±1°C) and 55% humidity, and received sterile rodent chow and water ad libitum. Rotenone (3 mg/kg; treated group) or vehicle (untreated group) was administered by intraperitoneal injection once a day for three days prior to inoculation of the cells into mice (n=6 per treatment group) as previously described (20). In addition, the nude mice were subcutaneously injected with 1×107 SW480 cells in 0.1 ml DMEM in the right axillary region. Animals were monitored three times a week to observe tumor growth. Tumor sizes were measured using a caliper, and the tumor volumes were calculated as follows: Tumor volume (mm3)=1/2 × length × width2. When tumors reached sufficient size (length ~18 mm, width ~13 mm and volume ~1,500 mm3), animals were euthanized by an overdose of pentobarbital (intraperitoneal, 120 mg/kg) on day 28, and tumor tissues were obtained. The protocol was approved by the Laboratory Animal Management Committee of Chongqing Medical University.

Statistical analysis

Data were analyzed using SPSS 19.0 (IBM Corp.) and are presented as the mean ± SD of three independent experiments. Student's t-test was used for comparisons between two groups. Multiple comparisons were performed by two-way ANOVA followed by Tukey's post hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

Rotenone induces CC cell cytotoxicity

To identify the cytotoxicity of rotenone on CC cells, human CC cell lines SW480 and SW620 and normal human colon CRL-1790 cells were treated with rotenone for 2 days and analyzed by the CCK-8 assay. The viability of CC cells decreased as the concentration of rotenone increased; by contrast, rotenone exhibited no significant effects on CRL-1790 cell viability (Fig. 1A). In subsequent experiments, cells were treated with 10 µM rote-none. In the colony formation assay, the numbers of colonies in the rotenone-treated wells were significantly lower compared with those in the untreated wells (Fig. 1B). In addition, EdU incorporation assays were used to investigate the effects of rotenone on CC cell proliferation. The results showed that rotenone notably inhibited CC cell proliferation compared with the untreated cells (Fig. 1C). The rotenone-mediated reduction of CC cell proliferation was also confirmed by western blot assay, as the protein level of Bcl-2 was decreased, whereas the levels of Bax and cleaved caspase-3 were increased following rotenone treatment compared with the untreated cells (Fig. 1D).

Rotenone inhibits CC cell migration and invasion in vitro

To determine the effects of rotenone on CC cell migration and invasion in vitro, wound healing and Transwell assays were respectively used with SW480 and SW620 cells treated with 10 µM rotenone. As demonstrated in Fig. 2A, 24-h rotenone incubation inhibited the migration of CC cells compared with that of the untreated cells. In addition, following incubation with rotenone for 24 h, the invasive ability of CC cells was decreased when compared with the untreated control group (Fig. 2C).

Rotenone inhibits CC cell EMT in vitro

To determine whether rotenone affected the EMT process in CC cells, RT-qPCR and western blotting assays were performed to detect the mRNA and protein levels of the EMT markers E-cadherin, vimentin and Snail. SW480 and SW620 cells treated with 10 µM rotenone for 24 h exhibited upregulated E-cadherin and downregulated Snail and vimentin mRNA and protein levels compared with the untreated cells (Fig. 3A and B). In addition, the detection of vimentin expression by fluorescent immunocytostaining was also consistent with the above results as rotenone-treated cells exhibited lower fluorescence levels compared with the untreated controls (Fig. 3C). Therefore, these results indicated that rotenone contributed to EMT in CC cells.

Rotenone suppresses CC cell proliferation, migration, invasion and EMT via the PI3K/AKT signaling pathway

To further investigate the molecular mechanism and pathway by which rotenone suppressed CC development and metastasis, the involvement of the PI3K/AKT signaling pathway in the anticancer effect of rotenone was determined. As presented in Fig. 4A, p-AKT (Ser473) and p-mTOR protein expression were downregulated in rotenone-treated CC cells. These results suggested that rotenone may exert its antitumor effect by inhibiting the activity of the PI3K/AKT/mTOR signaling pathway. To address this hypothesis, rotenone-treated or untreated CC cells were incubated with a PI3K/AKT signaling activatorIGF-1. The results demonstrated that the protein levels of p-AKT and p-mTOR were upregulated byIGF-1 compared with the untreated control cells, whereas rotenone reversed this effect (Fig. 4B). In addition, the proliferative, migratory and invasive abilities of rotenone and IGF-1 co-treated CC cells were determined. Of note, the SW480 and SW620 cell proliferation ofIGF-1-treatedcells was reversed by rotenone treatment, which was confirmed by the CCK-8 assay (Fig. 4C). Similarly, the wound healing and Transwell assays indicated that the IGF-1-induced CC cell migratory and invasive abilities were decreased following rotenone treatment compared with those of cells treated with IGF-1 alone (Fig. 4D and E). In addition, co-treatment with IGF-1 and rotenone resulted in the upregulation of E-cadherin and downregulation of vimentin and Snail protein expression in CC cells compared with that in cells treated with IGF-1 alone (Fig. 4F). In summary, these results suggested that rotenone decreased CC proliferation, migration, invasion and EMT in vitro by inhibiting the PI3K/AKT/mTOR pathway.

Rotenone inhibits CC tumor growth and metastasis in vivo in a xenograft mouse model

To confirm the in vitro anti-tumor effect of rotenone, the in vivo effects of rotenone were evaluated using the subcutaneous xenotransplant tumor model. Mice were injected intraperitoneally once a day with 3 mg/kg/day rotenone as previously described (22) for three days, and SW480 tumor cells were inoculated into nude mice. Rotenone-treated mice exhibited significantly inhibited tumor growth compared with the untreated control group (Fig. 5A). Immunohistochemical staining of the xenograft tissue sections revealed decreased expression of vimentin and increased expression of caspase-3, Bcl-2, E-cadherin after rotenone treatment (Fig. 5B). Subsequently, rotenone-mediated inhibition of mouse tumor tissue proliferative and metastatic abilities in vivo was confirmed by western blot assay, as the protein level of Bcl-2 was downregulated, whereas those of Bax and cleaved caspase-3 were upregulated following rotenone treatment compared with the tissues from untreated mice. The expression levels of vimentin were decreased, whereas the levels of E-cadherin were increased following rotenone treatment compared with those detected in the untreated mice. Of note, p-AKT expression was downregulated by rotenone compared with that in the untreated mouse tissues (Fig. 5C). These findings revealed that rotenone inhibited CC cell proliferation and metastasis in vivo.

Discussion

CC is a common malignant gastrointestinal tumor; ~20% of patients suffer from liver metastasis after treatment, and the 3-year survival rate of these patients is 5% (23,24). Therefore, it is important to explore novel effective therapeutic options for CC treatment. The present study investigated the effects of rotenone on CC cell proliferation and metastasis in vitro and in vivo, and revealed that rotenone may exert anti-tumor growth effects in CC.

Rotenone has been demonstrated to display antitumor effects through the induction of apoptosis in various types of cancer cells (11,25). Agarwal et al (26) have demonstrated that rotenone can promote HeLa cell apoptosis by inhibiting p-AKT, p-ERK and activating the expression of caspase-3 and Bax. Additionally, rotenone exerts anticancer effects by inducing G1 phase arrest and apoptosis in thyroid papillary carcinoma-derived cell line (25). Metastasis is the main cause of death in patients with advanced stage CC (27). EMT serves an important role in metastasis (28). It has been reported the expression of EMT markers are increased in CC (29,30). However, the effect of rotenone on CC cell viability, motility, invasion and EMT has not been previously determined. The results of the present study demonstrated that rotenone exerted its anticancer effects by modulating cell viability, motility, invasion and EMT, which was also confirmed in an in vivo mouse model.

Acute exposure to rotenone has been reported to induce oxidative stress, which impairs autophagic flux by modulating the PI3K/AKT/mTORC1 signaling pathway in human lung cancer cells (13). However, it is unclear whether the inhibitory effect of rotenone on CC depends on the PI3K/AKT signaling pathway. The results of the present study revealed that rote-none treatment inhibited the phosphorylation of AKT and mTOR in CC cells. In addition, rotenone treatment reversed the activation of PI3K/AKT/mTOR signaling pathway and the promotion of cell proliferation, migration and invasion that induced by PI3K/AKT signaling activator IGF-1 in CC cells. Of note, AKT protein expression was downregulated, whereas the expression of p-AKT was upregulated in rotenone-treated mice, which indicated that rotenone may exert its antitumor effects by inhibiting the PI3K/AKT signaling pathway. However, the detailed downstream mechanism requires further study using an in vivo mouse model or in a clinical study.

In conclusion, rotenone restrained the viability, motility, invasion and EMT process of CC cells in vitro, and inhibited tumorigenesis in nude mice in vivo, which was associated with its inhibitory effect on the PI3K/AKT pathway. The results described in the present study may provide new evidence for the anticancer effect of rotenone in CC.

Acknowledgments

Not applicable.

Funding

This study was supported by the Chongqing 2018 Rongchang District Medical and Health Institution Talent Training Program (grant no. 425, rongweifa 2018).

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

WBX and HL conceived the study. WBX, MLD, YWL and RQP performed the experiments and acquired the data. WBX, MLD, YWL, RQP, SJY and YL analyzed and interpreted the data. WBX, MLD, YWL and RQP drafted and revised the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The animal experiments were approved by the Laboratory Animal Management Committee of Chongqing Medical University.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D and Bray F: Cancer incidence and mortality worldwide: Sources methods and major patterns in GLOBOCAN 2012. Int J Cancer. 136:E359–E386. 2015. View Article : Google Scholar

2 

Jaganathan SK, Vellayappan MV, Narasimhan G and Supriyanto E: Role of pomegranate and citrus fruit juices in colon cancer prevention. World J Gastroenterol. 20:4618–4625. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Chen J, Elfiky A, Han M, Chen C and Saif MW: The role of Src in colon cancer and its therapeutic implications. Clin Colorectal Cancer. 13:5–13. 2014. View Article : Google Scholar

4 

Aggarwal BB, Takada Y and Oommen OV: From chemoprevention to chemotherapy: Common targets and common goals. Expert Opin Investig Drugs. 13:1327–1338. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Lee HY, Suh YA, Kosmeder JW, Pezzuto JM, Hong WK and Kurie JM: Deguelin-induced inhibition of cyclooxygenase-2 expression in human bronchial epithelial cells. Clin Cancer Res. 10:1074–1079. 2004. View Article : Google Scholar : PubMed/NCBI

6 

Dell'Eva R, Ambrosini C, Minghelli S, Noonan DM, Albini A and Ferrari N: The Akt inhibitor deguelin, is an angiopreventive agent also acting on the NF-kappaB pathway. Carcinogenesis. 28:404–413. 2007. View Article : Google Scholar

7 

Gerhäuser C, Mar W, Lee SK, Suh N, Luo Y, Kosmeder J, Luyengi L, Fong HH, Kinghorn AD, Moriarty RM, et al: Rotenoids mediate potent cancer chemopreventive activity through transcriptional regulation of ornithine decarboxylase. Nat Med. 1:260–266. 1995. View Article : Google Scholar : PubMed/NCBI

8 

Chun KH, Kosmeder JW II, Sun S, Pezzuto JM, Lotan R, Hong WK and Lee HY: Effects of deguelin on the phosphatidylinositol 3-kinase/Akt pathway and apoptosis in premalignant human bronchial epithelial cells. J Natl Cancer Inst. 95:291–302. 2003. View Article : Google Scholar : PubMed/NCBI

9 

Shang HS, Chang JB, Lin JH, Lin JP, Hsu SC, Liu CM, Liu JY, Wu PP, Lu HF, Au MK and Chung JG: Deguelin inhibits the migration and invasion of U-2 OS human osteosarcoma cells via the inhibition of matrix metalloproteinase-2/-9 in vitro. Molecules. 19:16588–16608. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Liu YP, Lee JJ, Lai TC, Lee CH, Hsiao YW, Chen PS, Liu WT, Hong CY, Lin SK, Ping Kuo MY, et al: Suppressive function of low-dose deguelin on the invasion of oral cancer cells by downregulating tumor necrosis factor alpha-induced nuclear factor-kappa B signaling. Head Neck. 38(Suppl 1): E524–E534. 2016. View Article : Google Scholar

11 

Deng YT, Huang HC and Lin JK: Rotenone induces apoptosis in MCF-7 human breast cancer cell-mediated ROS through JNK and p38 signaling. Mol Carcinog. 49:141–151. 2010.

12 

Lee JH, Lee DH, Lee HS, Choi JS, Kim KW and Hong SS: Deguelin inhibits human hepatocellular carcinoma by antiangiogenesis and apoptosis. Oncol Rep. 20:129–134. 2008.PubMed/NCBI

13 

Hu W, Tian H, Yue W, Li L, Li S, Gao C, Si L, Qi L, Lu M, Hao B and Shan S: Rotenone induces apoptosis in human lung cancer cells by regulating autophagic flux. IUBMB Life. 68:388–393. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Chen Y and Ding YY: LINC00467 enhances head and neck squamous cell carcinoma progression and epithelial-mesenchymal transition process via miR-299-5p/ubiquitin specific protease-48 axis. J Gene Med. e318411–Mar;2020.Epub ahead of print.

15 

Iwadate Y: Epithelial-mesenchymal transition in glioblastoma progression. Oncol Lett. 11:1615–1620. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Boreddy SR and Srivastava SK: Deguelin suppresses pancreatic tumor growth and metastasis by inhibiting epithelial-to-mesen-chymal transition in an orthotopic model. Oncogene. 32:3980–3991. 2013. View Article : Google Scholar

17 

Wang X, Liu H, Wang X and An Y: Clinical significance of migration and invasion inhibitor protein expression in non-small-cell lung cancer. Oncol Lett. 8:2417–2422. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Zhao D, Han W, Liu X, Cui D and Chen Y: Deguelin inhibits epithelial-to-mesenchymal transition and metastasis of human non-small cell lung cancer cells by regulating NIMA-related kinase 2. Thorac Cancer. 8:320–327. 2017. View Article : Google Scholar : PubMed/NCBI

19 

Miao HL, Lei CJ, Qiu ZD, Liu ZK, Li R, Bao ST and Li MY: MicroRNA-520c-3p inhibits hepatocellular carcinoma cell proliferation and invasion through induction of cell apoptosis by targeting glypican-3. Hepatol Res. 44:338–348. 2014. View Article : Google Scholar

20 

Zeng C, Pan F, Jones LA, Lim MM, Griffin EA, Sheline YI, Mintun MA, Holtzman DM and Mach RH: Evaluation of 5-ethynyl-2′-deoxyuridine staining as a sensitive and reliable method for studying cell proliferation in the adult nervous system. Brain Res. 1319:21–32. 2010. View Article : Google Scholar : PubMed/NCBI

21 

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

22 

Kang HW, Kim JM, Cha MY, Jung HC, Song IS and Kim JS: Deguelin, an Akt inhibitor, down-regulates NF-κB signaling and induces apoptosis in colon cancer cells and inhibits tumor growth in mice. Dig Dis Sci. 57:2873–2882. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Xia ZS, Wu D, Zhong W, Lu XJ, Yu T and Chen QK: Wip1 gene silencing enhances the chemosensitivity of human colon cancer cells. Oncol Lett. 14:1875–1883. 2017. View Article : Google Scholar : PubMed/NCBI

24 

Ding YL, Wang QS, Zhao WM and Xiang L: Expression of smoothened protein in colon cancer and its prognostic value for postoperative liver metastasis. Asian Pac J Cancer Prev. 13:4001–4005. 2012. View Article : Google Scholar : PubMed/NCBI

25 

Goncalves AP, Videira A, Maximo V and Soares P: Synergistic growth inhibition of cancer cells harboring the RET/PTC1 oncogene by staurosporine and rotenone involves enhanced cell death. J Biosci. 36:639–648. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Agarwal NR, Maurya N, Pawar JS and Ghosh I: A combined approach against tumorigenesis using glucose deprivation and mitochondrial complex 1 inhibition by rotenone. Cell Biol Int. 40:821–831. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Matsumoto R, Mori S, Kita Y, Toda H, Sasaki K, Arigami T, Matsushita D, Kurahara H, Maemura K and Natsugoe S: Multiple liver metastases with synchronous gastric and transverse colon cancer diagnosed by gastric perforation successfully treated by SOX plus bevacizumab and completely resected by surgery: A case report. Surg Case Rep. 6:512020. View Article : Google Scholar : PubMed/NCBI

28 

Li S, Hou X, Wu C, Han L, Li Q, Wang J and Luo S: MiR-645 promotes invasiveness, metastasis and tumor growth in colorectal cancer by targeting EFNA5. Biomed Pharmacother. 125:1098892020. View Article : Google Scholar : PubMed/NCBI

29 

Valastyan S and Weinberg RA: Tumor metastasis: Molecular insights and evolving paradigms. Cell. 147:275–292. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Bates RC and Mercurio AM: The epithelial-mesenchymal tansition (EMT) and colorectal cancer progression. Cancer Biol Ther. 4:365–370. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2020
Volume 46 Issue 2

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Xiao W, Liu Y, Dai M, Li Y, Peng R, Yu S and Liu H: Rotenone restrains colon cancer cell viability, motility and epithelial‑mesenchymal transition and tumorigenesis in nude mice via the PI3K/AKT pathway Retraction in /10.3892/ijmm.2023.5340. Int J Mol Med 46: 700-708, 2020
APA
Xiao, W., Liu, Y., Dai, M., Li, Y., Peng, R., Yu, S., & Liu, H. (2020). Rotenone restrains colon cancer cell viability, motility and epithelial‑mesenchymal transition and tumorigenesis in nude mice via the PI3K/AKT pathway Retraction in /10.3892/ijmm.2023.5340. International Journal of Molecular Medicine, 46, 700-708. https://doi.org/10.3892/ijmm.2020.4637
MLA
Xiao, W., Liu, Y., Dai, M., Li, Y., Peng, R., Yu, S., Liu, H."Rotenone restrains colon cancer cell viability, motility and epithelial‑mesenchymal transition and tumorigenesis in nude mice via the PI3K/AKT pathway Retraction in /10.3892/ijmm.2023.5340". International Journal of Molecular Medicine 46.2 (2020): 700-708.
Chicago
Xiao, W., Liu, Y., Dai, M., Li, Y., Peng, R., Yu, S., Liu, H."Rotenone restrains colon cancer cell viability, motility and epithelial‑mesenchymal transition and tumorigenesis in nude mice via the PI3K/AKT pathway Retraction in /10.3892/ijmm.2023.5340". International Journal of Molecular Medicine 46, no. 2 (2020): 700-708. https://doi.org/10.3892/ijmm.2020.4637