51
|
Khoshinani HM, Afshar S, Pashaki AS, Mahdavinezhad A, Nikzad S, Najafi R, Amini R, Gholami MH, khoshghadam A, Saidijam M. Involvement of miR-155/FOXO3a and miR-222/PTEN in acquired radioresistance of colorectal cancer cell line. Jpn J Radiol 2017; 35:664-672. [DOI: 10.1007/s11604-017-0679-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 08/16/2017] [Indexed: 12/14/2022]
|
52
|
Qiang W, Sui F, Ma J, Li X, Ren X, Shao Y, Liu J, Guan H, Shi B, Hou P. Proteasome inhibitor MG132 induces thyroid cancer cell apoptosis by modulating the activity of transcription factor FOXO3a. Endocrine 2017; 56:98-108. [PMID: 28220348 DOI: 10.1007/s12020-017-1256-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 02/01/2017] [Indexed: 12/18/2022]
Abstract
Proteasome inhibitors are promising antitumor drugs with preferable cytotoxicity in malignant cells and have exhibited clinical efficiency in several hematologic malignancies. P53-dependent apoptosis has been reported to be a major mechanism underlying. However, apoptosis can also be found in cancer cells with mutant-type p53, suggesting the involvement of p53-independent mechanism. Tumor suppressor forkhead Box O3 is another substrate of proteasomal degradation, which also functions partially through inducing apoptosis. The aim of this study was to explore the effect of proteasome inhibition on the expression and activity of forkhead Box O3 in thyroid cancer cells. Using flow cytometry, western blot, immunofluorescence staining and quantitative RT-PCR assays, we assessed proteasome inhibitor MG132-induced apoptosis in thyroid cancer cells and its effect on the expression and activity of forkhead Box O3. The resulted showed that MG132 induced significant apoptosis, and caused the accumulation of p53 protein in both p53 wild-type and mutant-type thyroid cancer cell lines, whereas the proapoptotic targets of p53 were transcriptionally upregulated only in the p53 wild-type cells. Strikingly, upon MG132 administration, the accumulation and nuclear translocation of transcription factor forkhead Box O3 as well as transcriptional upregulation of its proapoptotic target genes were found in thyroid cancer cells regardless of p53 status. Cell apoptosis was enhanced by ectopic overexpression while attenuated by silencing of forkhead Box O3. Altogether, we demonstrated that proteasome inhibitor MG132 induces thyroid cancer cell apoptosis at least partially through modulating forkhead Box O3 activity.
Collapse
Affiliation(s)
- Wei Qiang
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, The People's Republic of China
| | - Fang Sui
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, The People's Republic of China
| | - Jingjing Ma
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, The People's Republic of China
| | - Xinru Li
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, The People's Republic of China
| | - Xiaojuan Ren
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, The People's Republic of China
| | - Yuan Shao
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, The People's Republic of China
| | - Jiazhe Liu
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, The People's Republic of China
| | - Haixia Guan
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of China Medical University, Shenyang, 110001, The People's Republic of China
| | - Bingyin Shi
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, The People's Republic of China
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, The People's Republic of China
| | - Peng Hou
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, The People's Republic of China.
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, The People's Republic of China.
| |
Collapse
|
53
|
Maji P, Shah E, Paul S. RelSim: An integrated method to identify disease genes using gene expression profiles and PPIN based similarity measure. Inf Sci (N Y) 2017. [DOI: 10.1016/j.ins.2016.06.034] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
54
|
Wu N, Huang Y, Zou Z, Gimenez-Capitan A, Yu L, Hu W, Zhu L, Sun X, Sanchez JJ, Guan W, Liu B, Rosell R, Wei J. High BIM mRNA levels are associated with longer survival in advanced gastric cancer. Oncol Lett 2017; 13:1826-1834. [PMID: 28454330 DOI: 10.3892/ol.2017.5660] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 04/22/2016] [Indexed: 02/06/2023] Open
Abstract
Chemotherapy drugs, including 5-fluorouracil (5-FU), oxaliplatin and docetaxel, are commonly used in the treatment of gastric cancer (GC). Apoptosis-relevant genes may be associated with drug resistance. In the present study, the messenger RNA (mRNA) expression levels of B-cell lymphoma 2 interacting mediator of cell death (BIM), astrocyte elevated gene-1 (AEG-1) and AXL receptor tyrosine kinase (AXL) were investigated in 131 advanced GC samples, and the expression levels of these genes were correlated with patients' overall survival (OS). All 131 patients received first-line FOLFOX combination chemotherapy with folinic acid and 5-FU, in which 56 patients were further treated with second-line docetaxel-based chemotherapy. A correlation between the mRNA expression levels of BIM and AEG-1 was observed (rs=0.30; P=0.002). There was no association between the mRNA expression levels of any of the individual genes analyzed and OS in patients only receiving first-line FOLFOX chemotherapy. In a subgroup of patients receiving docetaxel-based second-line chemotherapy, those with high or intermediate levels of BIM exhibited a median OS of 18.2 months [95% confidence interval (CI), 12.8-23.6], compared with 9.6 months (95% CI, 8.9-10.3) in patients with low BIM levels (P=0.008). However, there was no correlation between the mRNA expression levels of AEG-1 or AXL and OS. The risk of mortality was higher in patients with low BIM mRNA levels than in those with high or intermediate BIM mRNA levels (hazard ratio, 2.61; 95% CI, 1.21-5.62; P=0.010). Therefore, BIM may be considered as a biomarker to identify whether patients could benefit from docetaxel-based second-line chemotherapy in GC.
Collapse
Affiliation(s)
- Nandie Wu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Department of Oncology, The Affiliated Drum Tower Hospital of Nanjing University, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| | - Ying Huang
- The Comprehensive Cancer Centre of Drum Tower Hospital, Department of Oncology, The Affiliated Drum Tower Hospital of Nanjing University, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| | - Zhengyun Zou
- The Comprehensive Cancer Centre of Drum Tower Hospital, Department of Oncology, The Affiliated Drum Tower Hospital of Nanjing University, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| | - Ana Gimenez-Capitan
- Pangaea Biotech, Department of Oncology, USP Dexeus University Institute, Barcelona 08001, Spain
| | - Lixia Yu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Department of Oncology, The Affiliated Drum Tower Hospital of Nanjing University, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| | - Wenjing Hu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Department of Oncology, The Affiliated Drum Tower Hospital of Nanjing University, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| | - Lijing Zhu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Department of Oncology, The Affiliated Drum Tower Hospital of Nanjing University, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| | - Xia Sun
- The Comprehensive Cancer Centre of Drum Tower Hospital, Department of Oncology, The Affiliated Drum Tower Hospital of Nanjing University, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| | - Jose Javier Sanchez
- Department of Preventive Medicine and Public Health, Autonomous University of Madrid, Madrid 28001, Spain
| | - Wenxian Guan
- Department of General Surgery, The Affiliated Drum Tower Hospital of Nanjing University, Medical School of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| | - Baorui Liu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Department of Oncology, The Affiliated Drum Tower Hospital of Nanjing University, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| | - Rafael Rosell
- Pangaea Biotech, Department of Oncology, USP Dexeus University Institute, Barcelona 08001, Spain.,Department of Medical Oncology, Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Badalona, Barcelona 08916, Spain
| | - Jia Wei
- The Comprehensive Cancer Centre of Drum Tower Hospital, Department of Oncology, The Affiliated Drum Tower Hospital of Nanjing University, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| |
Collapse
|
55
|
Voulgari E, Bakandritsos A, Galtsidis S, Zoumpourlis V, Burke BP, Clemente GS, Cawthorne C, Archibald SJ, Tuček J, Zbořil R, Kantarelou V, Karydas AG, Avgoustakis K. Synthesis, characterization and in vivo evaluation of a magnetic cisplatin delivery nanosystem based on PMAA-graft-PEG copolymers. J Control Release 2016; 243:342-356. [PMID: 27793687 DOI: 10.1016/j.jconrel.2016.10.021] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 10/15/2016] [Accepted: 10/23/2016] [Indexed: 12/11/2022]
Abstract
The development of anticancer drug delivery systems which retain or enhance the cytotoxic properties of the drug to tumorous tissues, while reducing toxicity to other organs is of key importance. We investigated different poly(methacrylic acid)-g-poly(ethyleneglycol methacrylate) polymers as in situ coating agents for magnetite nanocrystallites. The obtained magnetic nano-assemblies were in turn thoroughly characterized for their structural, colloidal and physicochemical properties (drug loading capacity/release, magnetic field triggered drug release, cell uptake and localization) in order to select the best performing system. With the focus on in vivo validation of such magnetic drug delivery systems for first time, we selected cisplatin as the drug, since it is a potent anticancer agent which exhibits serious side effects due to lack of selectivity. In addition, cisplatin would offer facile determination of the metal content in the animal tissues for biodistribution studies. Alongside post-mortem Pt determination in the tissues, the biodistribution of the drug nanocarriers was also monitored in real time with PET-CT (positron emission tomography/computed tomography) with and without the presence of magnetic field gradients; using a novel chelator-free method, the nanoparticles were radiolabeled with 68Ga without having to alter their structure with chemical modifications for conjugation of radiochelators. The ability to be radiolabeled in such a straightforward but very robust way, along with their measured high MRI response, renders them attractive for dual imaging, which is an important functionality for translational investigations. Their anticancer properties were evaluated in vitro and in vivo, in a cisplatin resistant HT-29 human colon adenocarcinoma model, with and without the presence of magnetic field gradients. Enhanced anticancer efficacy and reduced toxicity was recorded for the cisplatin-loaded nanocarriers in comparison to the free cisplatin, particularly when a magnetic field gradient was applied at the tumor site. Post mortem and real-time tissue distribution studies did not reveal increased cisplatin concentration in the tumor site, suggesting that the enhanced anticancer efficacy of the cisplatin-loaded nanocarriers is driven by mechanisms other than increased cisplatin accumulation in the tumors.
Collapse
Affiliation(s)
| | - Aristides Bakandritsos
- Department of Materials Science, University of Patras, Patras 26500, Greece; Regional Centre for Advanced Technologies and Materials, Department of Physical Chemistry, Faculty of Science, Palacky University in Olomouc, 17.listopadu 1192/12, 771 46 Olomouc, Czech Republic
| | - Sotiris Galtsidis
- Institute of Biology, Medicinal Chemistry & Biotechnology, NHRF, Athens, Greece
| | | | - Benjamin P Burke
- Department of Chemistry and Positron Emission Tomography Research Centre, University of Hull, Cottingham Road, Hull HU6 7RX, UK
| | - Gonçalo S Clemente
- Department of Chemistry and Positron Emission Tomography Research Centre, University of Hull, Cottingham Road, Hull HU6 7RX, UK
| | - Christopher Cawthorne
- Department of Chemistry and Positron Emission Tomography Research Centre, University of Hull, Cottingham Road, Hull HU6 7RX, UK
| | - Stephen J Archibald
- Department of Chemistry and Positron Emission Tomography Research Centre, University of Hull, Cottingham Road, Hull HU6 7RX, UK
| | - Jiři Tuček
- Regional Centre for Advanced Technologies and Materials, Department of Physical Chemistry, Faculty of Science, Palacky University in Olomouc, 17.listopadu 1192/12, 771 46 Olomouc, Czech Republic
| | - Radek Zbořil
- Regional Centre for Advanced Technologies and Materials, Department of Physical Chemistry, Faculty of Science, Palacky University in Olomouc, 17.listopadu 1192/12, 771 46 Olomouc, Czech Republic
| | - Vasiliki Kantarelou
- Institute of Nuclear and Particle Physics, NCSR "Demokritos", Athens, Greece
| | | | | |
Collapse
|
56
|
Nestal de Moraes G, Bella L, Zona S, Burton MJ, Lam EWF. Insights into a Critical Role of the FOXO3a-FOXM1 Axis in DNA Damage Response and Genotoxic Drug Resistance. Curr Drug Targets 2016; 17:164-77. [PMID: 25418858 PMCID: PMC5403963 DOI: 10.2174/1389450115666141122211549] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 11/06/2014] [Accepted: 11/19/2014] [Indexed: 11/22/2022]
Abstract
FOXO3a and FOXM1 are two forkhead transcription factors with antagonistic roles in cancer and DNA damage response. FOXO3a functions like a typical tumour suppressor, whereas FOXM1 is a potent oncogene aberrantly overexpressed in genotoxic resistant cancers. FOXO3a not only represses FOXM1 expression but also its transcriptional output. Recent research has provided novel insights into a central role for FOXO3a and FOXM1 in DNA damage response. The FOXO3a-FOXM1 axis plays a pivotal role in DNA damage repair and the accompanied cellular response through regulating the expression of genes essential for DNA damage sensing, mediating, signalling and repair as well as for senescence, cell cycle and cell death control. In this manner, the FOXO3a-FOXM1 axis also holds the key to cell fate decision in response to genotoxic therapeutic agents and controls the equilibrium between DNA repair and cell termination by cell death or senescence. As a consequence, inhibition of FOXM1 or reactivation of FOXO3a in cancer cells could enhance the efficacy of DNA damaging cancer therapies by decreasing the rate of DNA repair and cell survival while increasing senescence and cell death. Conceptually, targeting FOXO3a and FOXM1 may represent a promising molecular therapeutic option for improving the efficacy and selectivity of DNA damage agents, particularly in genotoxic agent resistant cancer. In addition, FOXO3a, FOXM1 and their downstream transcriptional targets may also be reliable diagnostic biomarkers for predicting outcome, for selecting therapeutic options, and for monitoring treatments in DNA-damaging agent therapy.
Collapse
Affiliation(s)
| | | | | | | | - Eric W-F Lam
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK.
| |
Collapse
|
57
|
Fanale D, Castiglia M, Bazan V, Russo A. Involvement of Non-coding RNAs in Chemo- and Radioresistance of Colorectal Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 937:207-28. [DOI: 10.1007/978-3-319-42059-2_11] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
58
|
Jeung YJ, Kim HG, Ahn J, Lee HJ, Lee SB, Won M, Jung CR, Im JY, Kim BK, Park SK, Son MJ, Chung KS. Shikonin induces apoptosis of lung cancer cells via activation of FOXO3a/EGR1/SIRT1 signaling antagonized by p300. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:2584-2593. [PMID: 27452907 DOI: 10.1016/j.bbamcr.2016.07.005] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 06/23/2016] [Accepted: 07/19/2016] [Indexed: 01/23/2023]
Abstract
Shikonin derivatives exert powerful cytotoxic effects including induction of apoptosis. Here, we demonstrate the cytotoxic efficacy of shikonin in vivo in xenograft models, which did not affect body weight as well as its reduction of cell viability in vitro using several non-small cell lung cancer (NSCLC) cell lines. We found that inhibition of AKT by shikonin activated the forkhead box (FOX)O3a/early growth response protein (EGR)1 signaling cascade and enhanced the expression of the target gene Bim, leading to apoptosis in lung cancer cells. Overexpression of wild-type or a constitutively active mutant of FOXO3a enhanced shikonin-induced Bim expression. The NAD+-dependent histone deacetylase sirtuin (SIRT)1 amplified the pro-apoptotic effect by deacetylating FOXO3a, which induced EGR1 binding to the Bim promoter and activated Bim expression. Meanwhile, PI3K/AKT activity was enhanced, whereas that of FOXO3a was reduced and p300 was upregulated by treatment with a sublethal dose of shikonin. FOXO3a acetylation was enhanced by p300 overexpression, while shikonin-induced Bim expression was suppressed by p300 overexpression, which promoted cell survival. FOXO3a acetylation was increased by p300 overexpression and treatment with SIRT1 inhibitor, improving cell survival. In addition, shikonin-induced FOXO3a nuclear localization was blocked by AKT activation and SIRT1 inhibition, which blocked Bim expression and conferred resistance to the cytotoxic effects of shikonin. The EGR1 increase induced by shikonin was restored by pretreatment with SIRT1 inhibitor. These results suggest that shikonin induces apoptosis in some lung cancer cells via activation of FOXO3a/EGR1/SIRT1 signaling, and that AKT and p300 negatively regulate this process via Bim upregulation.
Collapse
Affiliation(s)
- Yun-Ji Jeung
- Biomedical Translational Research Center, KRIBB, Daejeon 34141, Republic of Korea; Department of Biochemistry, Chungnam National University Medical School, Daejeon 301-747, Republic of Korea
| | - Han-Gyeul Kim
- Biomedical Translational Research Center, KRIBB, Daejeon 34141, Republic of Korea; Functional Genomics, Korea University of Science and Technology (UST), Daejeon, 305-806, Republic of Korea
| | - Jiwon Ahn
- Biomedical Translational Research Center, KRIBB, Daejeon 34141, Republic of Korea
| | - Ho-Joon Lee
- Biomedical Translational Research Center, KRIBB, Daejeon 34141, Republic of Korea
| | - Sae-Bhom Lee
- Biomedical Translational Research Center, KRIBB, Daejeon 34141, Republic of Korea
| | - Misun Won
- Biomedical Translational Research Center, KRIBB, Daejeon 34141, Republic of Korea
| | - Cho-Rock Jung
- Biomedical Translational Research Center, KRIBB, Daejeon 34141, Republic of Korea
| | - Joo-Young Im
- Biomedical Translational Research Center, KRIBB, Daejeon 34141, Republic of Korea
| | - Bo-Kyung Kim
- Biomedical Translational Research Center, KRIBB, Daejeon 34141, Republic of Korea
| | - Seung-Kiel Park
- Department of Biochemistry, Chungnam National University Medical School, Daejeon 301-747, Republic of Korea
| | - Myung Jin Son
- Stem Cell Research Center, KRIBB, Daejeon 34141, Republic of Korea; Functional Genomics, Korea University of Science and Technology (UST), Daejeon, 305-806, Republic of Korea.
| | - Kyung-Sook Chung
- Biomedical Translational Research Center, KRIBB, Daejeon 34141, Republic of Korea; Functional Genomics, Korea University of Science and Technology (UST), Daejeon, 305-806, Republic of Korea.
| |
Collapse
|
59
|
Salinomycin decreases doxorubicin resistance in hepatocellular carcinoma cells by inhibiting the β-catenin/TCF complex association via FOXO3a activation. Oncotarget 2016; 6:10350-65. [PMID: 25871400 PMCID: PMC4496360 DOI: 10.18632/oncotarget.3585] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Accepted: 02/13/2015] [Indexed: 12/17/2022] Open
Abstract
Doxorubicin is a conventional and effective chemotherapy drug against hepatocellular carcinoma (HCC). However, during long-term doxorubicin monotherapy, HCC cells may eventually develop acquired-resistance to doxorubicin which results in recurrence and a poor prognosis. Salinomycin, an ionophore antibiotic, was recently reported to selectively kill human cancer stem cells (CSCs) which were response for chemoresistance. In this study, salinomycin was found to exert synergistic cytotoxicity with doxorubicin in HCC cells and be capable of inhibiting doxorubicin-induced epithelial-mesenchymal transition (EMT), an important cellular process involved in the acquired chemoresistance of tumors. Further experiments revealed that FOXO3a, a multifunctional transcription factor that can be activated by salinomycin, was vital in mediating doxorubicin-induced EMT. In addition, activated FOXO3a disturbed the interaction between β-catenin and TCF and inhibited the expression of β-catenin/TCF target genes (ZEB1, c-Myc and CyclinD1), which played important roles in doxorubicin-induced EMT in HCC cells. Finally, the enhanced curative efficacy of combination treatment of doxorubicin and salinomycin for HCC was confirmed in established xenograft models. In summary, the present study identifies a new doxorubicin-based chemotherapy for advanced HCC and provides a potential anti-cancer strategy targeting FOXO3a and related cell pathway molecules.
Collapse
|
60
|
Liu HB, Gao XX, Zhang Q, Liu J, Cui Y, Zhu Y, Liu YF. Expression and prognostic implications of FOXO3a and Ki67 in lung adenocarcinomas. Asian Pac J Cancer Prev 2015; 16:1443-8. [PMID: 25743813 DOI: 10.7314/apjcp.2015.16.4.1443] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
To investigate the significance of FOXO3a and Ki67 in human lung adenocarcinomas. Envision immunohistochemical staining and Western blotting were used to examine the protein expression of FOXO3a in 127 cases of human lung adenocarcinoma specimens. The positive rate in lung adenocarcinoma (55.9%) was lower than that in normal tissues (80%). We found that the expression of FOXO3a was closely related with the degree of differentiation, TNM staging, lymph node metastasis and survival. In addition, significant differences in the different pathological types of lung adenocarcinoma cases (P<0.01). The FOXO3a positive rate of the acini as the main type (APA) (86.7%) and the lepidic as the main type (LPA) (82.4%) was higher than the solid as the main type (SPA) (50.0%), the papilla as the main type (PPA) (42.9%) and the micropapilla as the main type (MPA) (9.4%). Moreover, the expression of FOXO3a was negatively related with Ki67 expression. Our results suggested that the expression of FOXO3a is closely correlated with the aggressiveness of lung adenocarcinoma. It was indicated that disregulation of FOXO3a might play key roles in the occurrence and development of lung adenocarcinoma and joint detection of the two markers might play an important role in diagnosing tumors.
Collapse
Affiliation(s)
- Hong-Bin Liu
- Department of Pathology, The First People's Hospital of Nantong, Nantong, China E-mail :
| | | | | | | | | | | | | |
Collapse
|
61
|
Tarrado-Castellarnau M, Cortés R, Zanuy M, Tarragó-Celada J, Polat IH, Hill R, Fan TWM, Link W, Cascante M. Methylseleninic acid promotes antitumour effects via nuclear FOXO3a translocation through Akt inhibition. Pharmacol Res 2015; 102:218-34. [PMID: 26375988 DOI: 10.1016/j.phrs.2015.09.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 08/28/2015] [Accepted: 09/10/2015] [Indexed: 01/22/2023]
Abstract
Selenium supplement has been shown in clinical trials to reduce the risk of different cancers including lung carcinoma. Previous studies reported that the antiproliferative and pro-apoptotic activities of methylseleninic acid (MSA) in cancer cells could be mediated by inhibition of the PI3K pathway. A better understanding of the downstream cellular targets of MSA will provide information on its mechanism of action and will help to optimize its use in combination therapies with PI3K inhibitors. For this study, the effects of MSA on viability, cell cycle, metabolism, apoptosis, protein and mRNA expression, and reactive oxygen species production were analysed in A549 cells. FOXO3a subcellular localization was examined in A549 cells and in stably transfected human osteosarcoma U2foxRELOC cells. Our results demonstrate that MSA induces FOXO3a nuclear translocation in A549 cells and in U2OS cells that stably express GFP-FOXO3a. Interestingly, sodium selenite, another selenium compound, did not induce any significant effects on FOXO3a translocation despite inducing apoptosis. Single strand break of DNA, disruption of tumour cell metabolic adaptations, decrease in ROS production, and cell cycle arrest in G1 accompanied by induction of apoptosis are late events occurring after 24h of MSA treatment in A549 cells. Our findings suggest that FOXO3a is a relevant mediator of the antiproliferative effects of MSA. This new evidence on the mechanistic action of MSA can open new avenues in exploiting its antitumour properties and in the optimal design of novel combination therapies. We present MSA as a promising chemotherapeutic agent with synergistic antiproliferative effects with cisplatin.
Collapse
Affiliation(s)
- Míriam Tarrado-Castellarnau
- Department of Biochemistry and Molecular Biology, Faculty of Biology, Universitat de Barcelona, Av Diagonal 643, Barcelona 08028, Spain; Institute of Biomedicine of Universitat de Barcelona (IBUB) and CSIC-Associated Unit, Barcelona, Spain.
| | - Roldán Cortés
- Department of Biochemistry and Molecular Biology, Faculty of Biology, Universitat de Barcelona, Av Diagonal 643, Barcelona 08028, Spain; Institute of Biomedicine of Universitat de Barcelona (IBUB) and CSIC-Associated Unit, Barcelona, Spain.
| | - Miriam Zanuy
- Department of Biochemistry and Molecular Biology, Faculty of Biology, Universitat de Barcelona, Av Diagonal 643, Barcelona 08028, Spain; Institute of Biomedicine of Universitat de Barcelona (IBUB) and CSIC-Associated Unit, Barcelona, Spain.
| | - Josep Tarragó-Celada
- Department of Biochemistry and Molecular Biology, Faculty of Biology, Universitat de Barcelona, Av Diagonal 643, Barcelona 08028, Spain; Institute of Biomedicine of Universitat de Barcelona (IBUB) and CSIC-Associated Unit, Barcelona, Spain.
| | - Ibrahim H Polat
- Department of Biochemistry and Molecular Biology, Faculty of Biology, Universitat de Barcelona, Av Diagonal 643, Barcelona 08028, Spain; Institute of Biomedicine of Universitat de Barcelona (IBUB) and CSIC-Associated Unit, Barcelona, Spain.
| | - Richard Hill
- Centre for Biomedical Research (CBMR), University of Algarve, Campus of Gambelas, Building 8, Room 2.22, 8005-139 Faro, Portugal; Regenerative Medicine Program, Department of Biomedical Sciences and Medicine University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal; Brain Tumour Research Centre, School of Pharmacy and Biomedical Sciences, University of Portsmouth, PO1 2DT, United Kingdom.
| | - Teresa W M Fan
- Department of Toxicology, Markey Cancer Center and Center for Environmental and Systems Biochemistry (CESB), University of Kentucky, Lexington, KY 40536, USA.
| | - Wolfgang Link
- Centre for Biomedical Research (CBMR), University of Algarve, Campus of Gambelas, Building 8, Room 2.22, 8005-139 Faro, Portugal; Regenerative Medicine Program, Department of Biomedical Sciences and Medicine University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal.
| | - Marta Cascante
- Department of Biochemistry and Molecular Biology, Faculty of Biology, Universitat de Barcelona, Av Diagonal 643, Barcelona 08028, Spain; Institute of Biomedicine of Universitat de Barcelona (IBUB) and CSIC-Associated Unit, Barcelona, Spain.
| |
Collapse
|
62
|
Epibrassinolide alters PI3K/MAPK signaling axis via activating Foxo3a-induced mitochondria-mediated apoptosis in colon cancer cells. Exp Cell Res 2015; 338:10-21. [DOI: 10.1016/j.yexcr.2015.08.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 08/04/2015] [Accepted: 08/25/2015] [Indexed: 12/27/2022]
|
63
|
Benard A, Janssen CM, van den Elsen PJ, van Eggermond MCJA, Hoon DSB, van de Velde CJH, Kuppen PJK. Chromatin status of apoptosis genes correlates with sensitivity to chemo-, immune- and radiation therapy in colorectal cancer cell lines. Apoptosis 2015; 19:1769-78. [PMID: 25292014 DOI: 10.1007/s10495-014-1042-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The apoptosis pathway of programmed cell death is frequently deregulated in cancer. An intact apoptosis pathway is required for proper response to anti-cancer treatment. We investigated the chromatin status of key apoptosis genes in the apoptosis pathway in colorectal cancer cell lines in relation to apoptosis induced by chemo-, immune- or radiation therapy. Using chromatin immunoprecipitation (ChIP), we measured the presence of transcription-activating histone modifications H3Ac and H3K4me3 and silencing modifications H3K9me3 and H3K27me3 at the gene promoter regions of key apoptosis genes Bax, Bcl2, Caspase-9, Fas (CD95) and p53. Cell lines DLD1, SW620, Colo320, Caco2, Lovo and HT29 were treated with cisplatin, anti-Fas or radiation. The apoptotic response was measured by flow cytometry using propidium iodide and annexin V-FITC. The chromatin status of the apoptosis genes reflected the activation status of the intrinsic (Bax, Bcl2, Caspase-9 and p53) and extrinsic (Fas) pathways. An active intrinsic apoptotic pathway corresponded to sensitivity to cisplatin and radiation treatment of cell lines DLD1, SW620 and Colo320. An active Fas promoter corresponded to an active extrinsic apoptotic pathway in cell line DLD1. mRNA expression data correlated with the chromatin status of the apoptosis genes as measured by ChIP. In conclusion, the results presented in this study indicate that the balance between activating and silencing histone modifications, reflecting the chromatin status of apoptosis genes, can be used to predict the response of tumor cells to different anti-cancer therapies and could provide a novel target to sensitize tumors to obtain adequate treatment responses.
Collapse
Affiliation(s)
- Anne Benard
- Department of Surgery, K6-R, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
64
|
Wang F, Marshall CB, Ikura M. Forkhead followed by disordered tail: The intrinsically disordered regions of FOXO3a. INTRINSICALLY DISORDERED PROTEINS 2015; 3:e1056906. [PMID: 28232890 DOI: 10.1080/21690707.2015.1056906] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 05/22/2015] [Indexed: 12/22/2022]
Abstract
Forkhead box Class O is one of 19 subfamilies of the Forkhead box family, comprising 4 human transcription factors: FOXO1, FOXO3a, FOXO4, and FOXO6, which are involved in many crucial cellular processes. FOXO3a is a tumor suppressor involved in multiple physiological and pathological processes, and plays essential roles in metabolism, cell cycle arrest, DNA repair, and apoptosis. In its role as a transcription factor, the FOXO3a binds a consensus Forkhead response element DNA sequence, and recruits transcriptional coactivators to activate gene transcription. FOXO3a has additional functions, such as regulating p53-mediated apoptosis and activating kinase ATM. With the exception of the structured DNA-binding forkhead domain, most of the FOXO3a sequence comprises intrinsically disordered regions (IDRs), including 3 regions (CR1-3) that are conserved within the FOXO subfamily. Numerous studies have demonstrated that these IDRs directly mediate many of the diverse functions of FOXO3a. These regions contain post-translational modification and protein-protein interaction sites that integrate upstream signals to maintain homeostasis. Thus, the FOXO3a IDRs are emerging as key mediators of diverse regulatory processes, and represent an important target for the future development of therapeutics for FOXO3a-related diseases.
Collapse
Affiliation(s)
- Feng Wang
- The Campbell Family Cancer Research Institute, Princess Margaret Cancer Center, Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada; Present affiliation: Department of Biochemistry; Vanderbilt University School of Medicine; Nashville, TN USA
| | - Christopher B Marshall
- The Campbell Family Cancer Research Institute, Princess Margaret Cancer Center, Department of Medical Biophysics, University of Toronto , Toronto, Ontario, Canada
| | - Mitsuhiko Ikura
- The Campbell Family Cancer Research Institute, Princess Margaret Cancer Center, Department of Medical Biophysics, University of Toronto , Toronto, Ontario, Canada
| |
Collapse
|
65
|
Abstract
Cisplatin is one of the major chemotherapeutic agents used against different human cancers. A better understanding of the downstream cellular targets of cisplatin will provide information on its mechanism of action. FOXO3a is a member of the FOXO transcription factor family, which modulates the expression of genes involved in cell cycle arrest, apoptosis, and other cellular processes. In this study, we have investigated the effects of cisplatin in a panel of lung cancer cell lines. The results showed that cisplatin inhibited the proliferation of these lung cancer cell lines by inhibiting the PI3K/AKT pathway, with evidence of decreasing phosphorylation of PI3K and AKT under cisplatin treatment, and constitutively activating AKT1 could reduce cisplatin-induced cell apoptosis. More importantly, cisplatin significantly inhibited FOXO3a phosphorylation (at Thr32, AKT phosphorylation site) and induced FOXO3a nuclear accumulation, which in turn increased the expression of FOXO3a-dependent apoptotic protein Bim. Knockdown of FOXO3a expression using small interfering RNA attenuated cisplatin-induced apoptosis. Furthermore, activation of FOXO3a induced cell apoptosis irrespective of p53 status, whereas p53 may act as FOXO3a downstream molecules involved in cisplatin-induced cell apoptosis. Together, our findings suggested that FOXO3a is a relevant mediator of the cytotoxic effects of cisplatin in lung cancer cells.
Collapse
|
66
|
Osman AMM, Al-Malki HS, Al-Harthi SE, El-Hanafy AA, Elashmaoui HM, Elshal MF. Modulatory role of resveratrol on cytotoxic activity of cisplatin, sensitization and modification of cisplatin resistance in colorectal cancer cells. Mol Med Rep 2015; 12:1368-74. [PMID: 25815689 DOI: 10.3892/mmr.2015.3513] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Accepted: 08/29/2014] [Indexed: 12/22/2022] Open
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-associated mortality worldwide. Cisplatin (CIS) is one of the most active cytotoxic agents in current use and it has proven efficacy against various human malignancies. However, its clinical usefulness has been restricted by detrimental side effects, including nephrotoxicity and myelosuppression. The aim of the present study was to attempt to decrease the required dose of CIS, in order to minimize its side effects, and increase its capability to arrest, delay or reverse carcinogenesis. In addition, the present study aimed to ameliorate CIS-resistance in CRC cells, using the natural compound resveratrol (RSVL). RSVL (3,4', 5-trihydroxy-trans-stilbene) is a naturally occurring polyphenol present in the roots of white hellebore (Veratrum grandiflorum O. Loes) and extracted from >70 other plant species. RSVL can exert antioxidant and anti-inflammatory activities, and it has been shown to be active in the regulation of numerous cellular events associated with carcinogenesis. The present study evaluated the effects of RSVL on sensitization of both parent and CIS-resistant HCT-116 CRC cells to the action of cisplatin. The CIS was administered at a dose of 5 and 20 µg/ml, and CIS cytotoxicity, apoptosis, cell cycle and cisplatin cellular uptake were examined in the presence and absence of RSVL (15 µg/ml). RSVL treatment showed anti-proliferative effects and enhanced the cytotoxic effects of cis against the growth of both parent and CIS-resistant HCT-116 CRC cells, with a half maximal inhibitory concentration of 4.20 µg/ml and 4.72 µg/ml respectively. RSVL also induced a significant increase in the early apoptosis fraction and enhanced the subsequent apoptotic effects of CIS. The cellular uptake of CIS was significantly increased in the presence of RSVL, as compared with CIS treatment alone, and RSVL treatment sensitized the CIS-resistant HCT-116 cells. In conclusion, RSVL treatment increased the cytotoxic activity of CIS against the growth of both parent and CIS-resistant HCT-116 CRC cells.
Collapse
Affiliation(s)
- Abdel-Moneim M Osman
- Pharmacology Department, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Hamdan S Al-Malki
- Pharmacology Department, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Sameer E Al-Harthi
- Pharmacology Department, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Amr A El-Hanafy
- Department of Biological Science, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Hassan M Elashmaoui
- Department of Biological Science, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohamed F Elshal
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
67
|
Yu S, Yu Y, Sun Y, Wang X, Luo R, Zhao N, Zhang W, Li Q, Cui Y, Wang Y, Li W, Liu T. Activation of FOXO3a suggests good prognosis of patients with radically resected gastric cancer. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:2963-2970. [PMID: 26045805 PMCID: PMC4440114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Accepted: 02/23/2015] [Indexed: 06/04/2023]
Abstract
OBJECTIVE This study sought to investigate the role of the forkhead transcription factor FOXO3a in the prognosis of stage II/III gastric cancer patients. MATERIALS AND METHODS A single-institution cohort of 289 patients with stage II/III gastric cancer was studied. Formalin-fixed paraffin-embedded tumor and adjacent normal specimens were used for tissue microarray construction. Tissue sections were immunostained with FOXO3a. Microscopic evaluation to assess the presence and localization of FOXO3a in tumor and adjacent normal tissues was performed. Results were analyzed for association with clinicopathological characters and overall survival (OS). RESULTS FOXO3a expression was significantly higher in tumor tissues compared with adjacent normal tissues, and nuclear FOXO3a staining was observed to be more common in tumor samples than adjacent normal tissues. Poorer prognosis was seen in patients with tumors harboring lower expression of FOXO3a and also patients with adjacent normal tissues harboring higher expression of FOXO3a. High expression of FOXO3a in tumor tissues served as a good prognostic marker with multivariate hazard ratio (HR) of 0.737 (95% CI, 0.574 to 0.947; P=0.017) for OS. CONCLUSION The expression of FOXO3a was upregulated and activated in gastric cancer tissues, and was significantly associated with a favorable prognosis in stage II/III gastric cancer patients.
Collapse
Affiliation(s)
- Shan Yu
- Department of Medical Oncology, Zhongshan Hospital, Fudan UniversityShanghai, People’s Republic of China
| | - Yiyi Yu
- Department of Medical Oncology, Zhongshan Hospital, Fudan UniversityShanghai, People’s Republic of China
| | - Yihong Sun
- Department of General Surgery, Zhongshan Hospital, Fudan UniversityShanghai, People’s Republic of China
| | - Xuefei Wang
- Department of General Surgery, Zhongshan Hospital, Fudan UniversityShanghai, People’s Republic of China
| | - Rongkui Luo
- Department of Pathology, Zhongshan Hospital, Fudan UniversityShanghai, People’s Republic of China
| | - Naiqing Zhao
- Department of Biostatistics, Fudan UniversityShanghai, People’s Republic of China
| | - Wen Zhang
- Department of Medical Oncology, Zhongshan Hospital, Fudan UniversityShanghai, People’s Republic of China
| | - Qian Li
- Department of Medical Oncology, Zhongshan Hospital, Fudan UniversityShanghai, People’s Republic of China
| | - Yuehong Cui
- Department of Medical Oncology, Zhongshan Hospital, Fudan UniversityShanghai, People’s Republic of China
| | - Yan Wang
- Department of Medical Oncology, Zhongshan Hospital, Fudan UniversityShanghai, People’s Republic of China
| | - Wei Li
- Department of Medical Oncology, Zhongshan Hospital, Fudan UniversityShanghai, People’s Republic of China
| | - Tianshu Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan UniversityShanghai, People’s Republic of China
| |
Collapse
|
68
|
Huang W, Wu QD, Zhang M, Kong YL, Cao PR, Zheng W, Xu JH, Ye M. Novel Hsp90 inhibitor FW-04-806 displays potent antitumor effects in HER2-positive breast cancer cells as a single agent or in combination with lapatinib. Cancer Lett 2015; 356:862-71. [DOI: 10.1016/j.canlet.2014.10.040] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 10/30/2014] [Accepted: 10/30/2014] [Indexed: 10/24/2022]
|
69
|
Yang YC, Tang YA, Shieh JM, Lin RK, Hsu HS, Wang YC. DNMT3B overexpression by deregulation of FOXO3a-mediated transcription repression and MDM2 overexpression in lung cancer. J Thorac Oncol 2014; 9:1305-15. [PMID: 25122426 DOI: 10.1097/jto.0000000000000240] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
INTRODUCTION DNA methyltransferase 3B (DNMT3B) contributes to de novo DNA methylation and its overexpression promotes tumorigenesis. However, whether DNMT3B is upregulated by transcriptional deregulation remains unclear. METHODS We studied the transcriptional repression of DNMT3B by forkhead O transcription factor 3a (FOXO3a) in lung cancer cell, animal, and clinical models. RESULTS The results of luciferase reporter assay showed that FOXO3a negatively regulated DNMT3B promoter activity by preferentially interacting with the binding element FOXO3a-E (+166 to +173) of DNMT3B promoter. Ectopically overexpressed FOXO3a or combined treatment with doxorubicin to induce FOXO3a nuclear accumulation further bound at the distal site, FOXO3a-P (-249 to -242) by chromatin-immunoprecipitation assay. Knockdown of FOXO3a resulted in an open chromatin structure and high DNMT3B mRNA and protein expression. Abundant FOXO3a repressed DNMT3B promoter by establishing a repressed chromatin structure. Note that FOXO3a is a degradation substrate of MDM2 E3-ligase. Cotreatment with doxorubicin and MDM2 inhibitor, Nutlin-3, further enforced abundant nuclear accumulation of FOXO3a resulting in decrease expression of DNMT3B leading to synergistic inhibition of tumor growth and decrease of methylation status on tumor suppressor genes in xenograft specimens. Clinically, lung cancer patients with DNMT3B high, FOXO3a low, and MDM2 high expression profile correlated with poor prognosis examined by immunohistochemistry and Kaplan-Meier survival analysis. CONCLUSIONS We reveal a new mechanism that FOXO3a transcriptionally represses DNMT3B expression and this regulation can be attenuated by MDM2 overexpression in human lung cancer model. Cotreatment with doxorubicin and Nutlin-3 is a novel therapeutic strategy through epigenetic modulation.
Collapse
Affiliation(s)
- Yi-Chieh Yang
- *Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan; †Institute of Basic Medical Science, College of Medicine, National Cheng Kung University, Tainan, Taiwan; ‡Division of Chest Medicine, Department of Internal Medicine, Chi Mei Medical Center, Tainan; The Center of General Education, Chia Nan University of Pharmacy & Science, Tainan, Taiwan; §Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei, Taiwan; and ‖Division of Thoracic Surgery, Taipei Veterans General Hospital; Institute of Emergency and Critical Care Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
70
|
Queiroz EAIF, Puukila S, Eichler R, Sampaio SC, Forsyth HL, Lees SJ, Barbosa AM, Dekker RFH, Fortes ZB, Khaper N. Metformin induces apoptosis and cell cycle arrest mediated by oxidative stress, AMPK and FOXO3a in MCF-7 breast cancer cells. PLoS One 2014; 9:e98207. [PMID: 24858012 PMCID: PMC4032293 DOI: 10.1371/journal.pone.0098207] [Citation(s) in RCA: 199] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 04/30/2014] [Indexed: 12/23/2022] Open
Abstract
Recent studies have demonstrated that the anti-diabetic drug, metformin, can exhibit direct antitumoral effects, or can indirectly decrease tumor proliferation by improving insulin sensitivity. Despite these recent advances, the underlying molecular mechanisms involved in decreasing tumor formation are not well understood. In this study, we examined the antiproliferative role and mechanism of action of metformin in MCF-7 cancer cells treated with 10 mM of metformin for 24, 48, and 72 hours. Using BrdU and the MTT assay, it was found that metformin demonstrated an antiproliferative effect in MCF-7 cells that occurred in a time- and concentration- dependent manner. Flow cytometry was used to analyze markers of cell cycle, apoptosis, necrosis and oxidative stress. Exposure to metformin induced cell cycle arrest in G0-G1 phase and increased cell apoptosis and necrosis, which were associated with increased oxidative stress. Gene and protein expression were determined in MCF-7 cells by real time RT-PCR and western blotting, respectively. In MCF-7 cells metformin decreased the activation of IRβ, Akt and ERK1/2, increased p-AMPK, FOXO3a, p27, Bax and cleaved caspase-3, and decreased phosphorylation of p70S6K and Bcl-2 protein expression. Co-treatment with metformin and H2O2 increased oxidative stress which was associated with reduced cell number. In the presence of metformin, treating with SOD and catalase improved cell viability. Treatment with metformin resulted in an increase in p-p38 MAPK, catalase, MnSOD and Cu/Zn SOD protein expression. These results show that metformin has an antiproliferative effect associated with cell cycle arrest and apoptosis, which is mediated by oxidative stress, as well as AMPK and FOXO3a activation. Our study further reinforces the potential benefit of metformin in cancer treatment and provides novel mechanistic insight into its antiproliferative role.
Collapse
Affiliation(s)
- Eveline A. I. F. Queiroz
- Pharmacology Department, Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
- Department of Physiological Sciences, State University of Londrina, Londrina, Paraná, Brazil
| | - Stephanie Puukila
- Biology Department, Lakehead University, Thunder Bay, Ontario, Canada
| | - Rosangela Eichler
- Pharmacology Department, Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Sandra C. Sampaio
- Pharmacology Department, Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Heidi L. Forsyth
- Northern Ontario School of Medicine, Lakehead University, Thunder Bay, Ontario, Canada
| | - Simon J. Lees
- Northern Ontario School of Medicine, Lakehead University, Thunder Bay, Ontario, Canada
- Biology Department, Lakehead University, Thunder Bay, Ontario, Canada
| | - Aneli M. Barbosa
- Biorefining Research Institute, Lakehead University, Thunder Bay, Ontario, Canada
| | - Robert F. H. Dekker
- Biorefining Research Institute, Lakehead University, Thunder Bay, Ontario, Canada
| | - Zuleica B. Fortes
- Pharmacology Department, Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
- * E-mail: (ZBF); (NK)
| | - Neelam Khaper
- Northern Ontario School of Medicine, Lakehead University, Thunder Bay, Ontario, Canada
- Biology Department, Lakehead University, Thunder Bay, Ontario, Canada
- * E-mail: (ZBF); (NK)
| |
Collapse
|
71
|
Inhibition of EGFR-induced glucose metabolism sensitizes chondrosarcoma cells to cisplatin. Tumour Biol 2014; 35:7017-24. [DOI: 10.1007/s13277-014-1902-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 03/27/2014] [Indexed: 12/19/2022] Open
|
72
|
Cortés R, Tarrado-Castellarnau M, Talancón D, López C, Link W, Ruiz D, Centelles JJ, Quirante J, Cascante M. A novel cyclometallated Pt(ii)–ferrocene complex induces nuclear FOXO3a localization and apoptosis and synergizes with cisplatin to inhibit lung cancer cell proliferation. Metallomics 2014; 6:622-33. [DOI: 10.1039/c3mt00194f] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
73
|
Forkhead box proteins: the tuning forks in cancer development and treatment. Mol Oncol 2013. [DOI: 10.1017/cbo9781139046947.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
74
|
Targeted therapy against chemoresistant colorectal cancers: Inhibition of p38α modulates the effect of cisplatin in vitro and in vivo through the tumor suppressor FoxO3A. Cancer Lett 2013; 344:110-118. [PMID: 24215867 DOI: 10.1016/j.canlet.2013.10.035] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 10/22/2013] [Accepted: 10/22/2013] [Indexed: 12/26/2022]
Abstract
Chemoresistance is a major obstacle to effective therapy against colorectal cancer (CRC) and may lead to deadly consequences. The metabolism of CRC cells depends highly on the p38 MAPK pathway, whose involvement in maintaining a chemoresistant behavior is currently being investigated. Our previous studies revealed that p38α is the main p38 isoform in CRC cells. Here we show that p38α pharmacological inhibition combined with cisplatin administration decreases colony formation and viability of cancer cells and strongly increases Bax-dependent apoptotic cell death by activating the tumor suppressor protein FoxO3A. Our results indicate that FoxO3A activation up-regulates transcription of its target genes (p21, PTEN, Bim and GADD45), which forces both chemosensitive and chemoresistant CRC cells to undergo apoptosis. Additionally, we found that FoxO3A is required for apoptotic cell death induction, as confirmed by RNA interference experiments. In animal models xenografted with chemoresistant HT29 cells, we further confirmed that the p38-targeted dual therapy strategy produced an increase in apoptosis in cancer tissue leading to tumor regression. Our study uncovers a major role for the p38-FoxO3A axis in chemoresistance, thereby suggesting a new therapeutic approach for CRC treatment; moreover, our results indicate that Bax status may be used as a predictive biomarker.
Collapse
|
75
|
Zhang L, Pickard K, Jenei V, Bullock MD, Bruce A, Mitter R, Kelly G, Paraskeva C, Strefford J, Primrose J, Thomas GJ, Packham G, Mirnezami AH. miR-153 supports colorectal cancer progression via pleiotropic effects that enhance invasion and chemotherapeutic resistance. Cancer Res 2013; 73:6435-47. [PMID: 23950211 DOI: 10.1158/0008-5472.can-12-3308] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Although microRNAs (miRNA) have been broadly studied in cancer, comparatively less is understood about their role in progression. Here we report that miR-153 has a dual role during progression of colorectal cancer by enhancing cellular invasiveness and platinum-based chemotherapy resistance. miRNA profiling revealed that miR-153 was highly expressed in a cellular model of advanced stage colorectal cancer. Its upregulation was also noted in primary human colorectal cancer compared with normal colonic epithelium and in more advanced colorectal cancer stages compared with early stage disease. In colorectal cancer patients followed for 50 months, 21 of 30 patients with high levels of miR-153 had disease progression compared with others in this group with low levels of miR-153. Functional studies revealed that miR-153 upregulation increased colorectal cancer invasiveness and resistance to oxaliplatin and cisplatin both in vitro and in vivo. Mechanistic investigations indicated that miR-153 promoted invasiveness indirectly by inducing matrix metalloprotease enzyme 9 production, whereas drug resistance was mediated directly by inhibiting the Forkhead transcription factor Forkhead box O3a (FOXO3a). In support of the latter finding, we found that levels of miR-153 and FOXO3a were inversely correlated in matched human colorectal cancer specimens. Our findings establish key roles for miR-153 overexpression in colorectal cancer progression, rationalizing therapeutic strategies to target expression of this miRNA for colorectal cancer treatment.
Collapse
Affiliation(s)
- Lei Zhang
- Authors' Affiliations: University of Southampton Cancer Sciences Division, Somers Cancer Research Building; Department of Colorectal Surgery, Southampton University Hospital NHS Trust, Southampton; Bioinformatics Unit, London Research Institute, Cancer Research UK, London; and School of Cellular and Molecular Medicine, University of Bristol, Medical Sciences Building, Bristol, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Bullock MD, Bruce A, Sreekumar R, Curtis N, Cheung T, Reading I, Primrose JN, Ottensmeier C, Packham GK, Thomas G, Mirnezami AH. FOXO3 expression during colorectal cancer progression: biomarker potential reflects a tumour suppressor role. Br J Cancer 2013; 109:387-94. [PMID: 23828518 PMCID: PMC3721407 DOI: 10.1038/bjc.2013.355] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 03/17/2013] [Accepted: 03/22/2013] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND In previous studies, the Forkhead/winged-helix-box-class-O3 (FOXO3) transcription factor has displayed both tumour suppressive and metastasis-promoting properties.To clarify its role in human colorectal cancer (CRC) progression, we examined in vivo FOXO3 expression at key points of the metastatic cascade. METHODS Formalin-fixed paraffin-embedded resection specimens from normal colon, adenomas, primary CRC specimens of different pathological stage and CRC specimens with matched liver metastases were used to generate three separate custom-designed tissue microarray (TMA) representations of metastatic progression. Triplicate cores, immunostained for FOXO3 were scored semiquantitatively by two investigators. RESULTS The FOXO3 expression is significantly reduced in CRC specimens compared with normal tissue, and progressive FOXO3 downregulation is associated with advancing pathological stage. In addition, recurrent stage I/II primary tumours show a significantly lower FOXO3 expression compared with stage-matched non-recurrent tumours. When stratified according to high and low FOXO3 expression, mean disease-free survival in the low-expressing group was 28 months (95% CI 15.8-50.6) compared with 64 months (95% CI 52.9-75.4) in the high-expressing group. CONCLUSION We have demonstrated an association between low FOXO3 expression and CRC progression in vivo using purpose-designed TMAs. Forkhead/winged-helix-box-class-O3 may represent a novel biomarker of nodal and distant disease spread with clinical utility in CRC.
Collapse
Affiliation(s)
- M D Bullock
- Cancer Research UK Centre, University of Southampton, Somers Cancer Sciences Building, Southampton General Hospital, University Hospital Southampton, Tremona Road, Southampton, UK.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
77
|
van Boxtel R, Gomez-Puerto C, Mokry M, Eijkelenboom A, van der Vos KE, Nieuwenhuis EES, Burgering BMT, Lam EWF, Coffer PJ. FOXP1 acts through a negative feedback loop to suppress FOXO-induced apoptosis. Cell Death Differ 2013; 20:1219-29. [PMID: 23832113 DOI: 10.1038/cdd.2013.81] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 05/21/2013] [Accepted: 05/23/2013] [Indexed: 12/27/2022] Open
Abstract
Transcriptional activity of Forkhead box transcription factor class O (FOXO) proteins can result in a variety of cellular outcomes depending on cell type and activating stimulus. These transcription factors are negatively regulated by the phosphoinositol 3-kinase (PI3K)-protein kinase B (PKB) signaling pathway, which is thought to have a pivotal role in regulating survival of tumor cells in a variety of cancers. Recently, it has become clear that FOXO proteins can promote resistance to anti-cancer therapeutics, designed to inhibit PI3K-PKB activity, by inducing the expression of proteins that provide feedback at different levels of this pathway. We questioned whether such a feedback mechanism may also exist directly at the level of FOXO-induced transcription. To identify critical modulators of FOXO transcriptional output, we performed gene expression analyses after conditional activation of key components of the PI3K-PKB-FOXO signaling pathway and identified FOXP1 as a direct FOXO transcriptional target. Using chromatin immunoprecipitation followed by next-generation sequencing, we show that FOXP1 binds enhancers that are pre-occupied by FOXO3. By sequencing the transcriptomes of cells in which FOXO is specifically activated in the absence of FOXP1, we demonstrate that FOXP1 can modulate the expression of a specific subset of FOXO target genes, including inhibiting expression of the pro-apoptotic gene BIK. FOXO activation in FOXP1-knockdown cells resulted in increased cell death, demonstrating that FOXP1 prevents FOXO-induced apoptosis. We therefore propose that FOXP1 represents an important modulator of FOXO-induced transcription, promoting cellular survival.
Collapse
Affiliation(s)
- R van Boxtel
- Department of Cell Biology, University Medical Center Utrecht, Heidelberglaan 100, Utrecht 3584 CX, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Yung MMH, Chan DW, Liu VWS, Yao KM, Ngan HYS. Activation of AMPK inhibits cervical cancer cell growth through AKT/FOXO3a/FOXM1 signaling cascade. BMC Cancer 2013; 13:327. [PMID: 23819460 PMCID: PMC3702529 DOI: 10.1186/1471-2407-13-327] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 07/01/2013] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Although advanced-stage cervical cancer can benefit from current treatments, approximately 30% patients may fail after definitive treatment eventually. Therefore, exploring alternative molecular therapeutic approaches is imperatively needed for this disease. We have recently shown that activation of AMP-activated protein kinase (AMPK), a metabolic sensor, hampers cervical cancer cell growth through blocking the Wnt/β-catenin signaling activity. Here, we report that activated AMPK (p-AMPK) also inhibits cervical cancer cell growth by counteracting FOXM1 function. METHODS Effect of the activation of AMPK on FOXM1 expression was examined by hypoxia and glucose deprivation, as well as pharmacological AMPK activators such as A23187, AICAR and metformin. RT Q-PCR and Western blot analysis were employed to investigate the activities of AMPK, FOXM1 and AKT/FOXO3a signaling. RESULTS Consistent with our previous findings, the activation of AMPK by either AMPK activators such as AICAR, A23187, metformin, glucose deprivation or hypoxia significantly inhibited the cervical cancer cell growth. Importantly, we found that activated AMPK activity was concomitantly associated with the reduction of both the mRNA and protein levels of FOXM1. Mechanistically, we showed that activated AMPK was able to reduce AKT mediated phosphorylation of p-FOXO3a (Ser253). Interestingly, activated AMPK could not cause any significant changes in FOXM1 in cervical cancer cells in which endogenous FOXO3a levels were knocked down using siRNAs, suggesting that FOXO3a is involved in the suppression of FOXM1. CONCLUSION Taken together, our results suggest the activated AMPK impedes cervical cancer cell growth through reducing the expression of FOXM1.
Collapse
Affiliation(s)
- Mingo Ming Ho Yung
- Departments of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, People's Republic of China
| | | | | | | | | |
Collapse
|
79
|
Gomes AR, Zhao F, Lam EWF. Role and regulation of the forkhead transcription factors FOXO3a and FOXM1 in carcinogenesis and drug resistance. CHINESE JOURNAL OF CANCER 2013; 32:365-70. [PMID: 23706767 PMCID: PMC3845605 DOI: 10.5732/cjc.012.10277] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The FOXO3a and FOXM1 forkhead transcription factors are key players in cancer initiation, progression, and drug resistance. Recent research shows that FOXM1 is a direct transcriptional target of FOXO3a, a vital downstream effector of the PI3K-AKT-FOXO signaling cascade. In addition, FOXM1 and FOXO3a also antagonize each other's activity by competitively binding to the same target genes, which are involved in chemotherapeutic drug sensitivity and resistance. Understanding the role and regulation of the FOXO-FOXM1 axis will provide insight into chemotherapeutic drug action and resistance in patients, and help to identify novel therapeutic approaches as well as diagnostic and predictive biomarkers.
Collapse
Affiliation(s)
- Ana R Gomes
- Department of Surgery and Cancer, Imperial College London, London, UK
| | | | | |
Collapse
|
80
|
Kim A, Lee JE, Lee SS, Kim C, Lee SJ, Jang WS, Park S. Coexistent mutations of KRAS and PIK3CA affect the efficacy of NVP-BEZ235, a dual PI3K/MTOR inhibitor, in regulating the PI3K/MTOR pathway in colorectal cancer. Int J Cancer 2013; 133:984-96. [PMID: 23475782 DOI: 10.1002/ijc.28073] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 01/08/2013] [Indexed: 12/29/2022]
Abstract
Colorectal cancer (CRC) with mutational activation of KRAS is observed frequently. In addition, PIK3CA mutations commonly coexist with KRAS mutations and lead to additive activation of the PI3K/MTOR signaling pathway. Here, we investigated how CRC cells that harbor KRAS and PIK3CA mutations affect sensitivity to inhibition of PI3K/MTOR with NVP-BEZ235 (BEZ235). We selected CRC patient samples and assessed their mutational status. CRC patients with KRAS or PIK3CA mutations show activation of AKT and MTOR, particularly when KRAS and PIK3CA mutations coexist. Suppression of PI3K/MTOR by BEZ235 results in a growth inhibitory effect and enhanced apoptosis via BIM activation in KRAS mutant cells. Mutational activation of KRAS when accompanied by a PIK3CA mutation converges at PI3K/MTOR pathway activation, resulting in resistance to BEZ235. BIM knockdown blocked the apoptotic response to BEZ235 in KRAS mutant cells, suggesting that PI3K inhibition leads to BIM accumulation. Moreover, BEZ235 treatment resulted in induction of FOXO3A activity and its induced transcription of BIM activation, which sensitized cells to cytotoxic agents leading to apoptosis in double mutant cells in vitro and in vivo. Taken together, our data suggest that targeting PI3K/MTOR sensitizes cells to apoptosis, implying that activation of PI3K/MTOR signaling via KRAS or PIK3CA mutation is an important pathway in CRC cell growth. Based on these results, coexistent KRAS and PIK3CA mutations confer resistance to BEZ235 via suppression of BIM-induced apoptosis, suggesting that combined treatment with conventional chemoagents is a potential strategy in the clinic.
Collapse
Affiliation(s)
- Areumnuri Kim
- Laboratory of Experimental Pathology, Korea Institute of Radiological and Medical Science, Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|
81
|
Carbajo-Pescador S, Steinmetz C, Kashyap A, Lorenz S, Mauriz JL, Heise M, Galle PR, González-Gallego J, Strand S. Melatonin induces transcriptional regulation of Bim by FoxO3a in HepG2 cells. Br J Cancer 2012; 108:442-9. [PMID: 23257900 PMCID: PMC3566813 DOI: 10.1038/bjc.2012.563] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Background: Melatonin induces apoptosis in many different cancer cell lines, including hepatocellular carcinoma cells. However, the responsible pathways have not been clearly elucidated. A member of the forkhead transcription factors' family, FoxO3a, has been implicated in the expression of the proapoptotic protein Bim (a Bcl-2-interacting mediator of cell death). In this study, we used human HepG2 liver cancer cells as an in vitro model to investigate whether melatonin treatment induces Bim through regulation by the transcription factor FoxO3a. Methods: Cytotoxicity of melatonin was compared in HepG2 hepatoblastoma cells and primary human hepatocytes. Proapoptotic Bim expression was analysed by reverse transcriptase–polymerase chain reaction and western blot. Reporter gene assays and chromatin immunoprecipitation assays were performed to analyse whether FoxO3a transactivates the Bim promoter. Small interfering RNA (siRNA) was used to study the role of FoxO3a in Bim expression. Immunofluorescence was performed to analyse FoxO3a localisation in HepG2 cells. Results: Melatonin treatment induces apoptosis in HepG2 cells, but not in primary human hepatocytes. The proapoptotic effect was mediated by increased expression of the BH3-only protein Bim. During melatonin treatment, we observed increased transcriptional activity of the forkhead-responsive element and could demonstrate that FoxO3a binds to a specific sequence within the Bim promoter. Furthermore, melatonin reduced phosphorylation of FoxO3a at Thr32 and Ser253, and induced its increased nuclear localisation. Moreover, silencing experiments with FoxO3a siRNA prevented Bim upregulation. Conclusion: This study shows that melatonin can induce apoptosis in HepG2 hepatocarcinoma cells through the upregulation of proapoptotic Bim mediated by nuclear translocation and activation of the transcription factor FoxO3a.
Collapse
Affiliation(s)
- S Carbajo-Pescador
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd) and Institute of Biomedicine, University of León, León, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Upregulation of the antiapoptotic factor Livin contributes to cisplatin resistance in colon cancer cells. Tumour Biol 2012. [PMID: 23188704 DOI: 10.1007/s13277-012-0596-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
The antiapoptotic factor Livin has been considered critical for tumor progression and poor prognosis for variant types of tumors. However, there are only limited reports regarding its expression and biological functions in colon cancer. Here, we examined Livin expression in four colon cancer cell lines (HCT116, RKO, KM12C, and SW620) in the presence or absence of cisplatin that was used as a model reagent. We found the different response to cisplatin was related to endogenous Livin expression level. From among a panel of apoptosis-related factors (p53, Bcl-2, Bcl-XL, BAX, and survivin), the expression of Livin was upregulated after cisplatin treatment in a dose-dependent manner. Both immunocytochemistry and nuclear cytoplasmic fractionation indicated Livin remained in the cytoplasm after treatment with cisplatin. In an attempt to explore the mechanism, we found the elevated expression of Livin was not due to the decreased degradation by proteosome but was enhanced at the mRNA level. Besides, cisplatin treatment activated the mammalian target of rapamycin (mTOR) pathway as shown by increased phosphorylation of Akt1, mTOR, S6K, and 4E-BP1, together with the elevated Livin. The PI3K inhibitor LY294002 inhibited both the phosphorylation of mTOR and upregulation of Livin. The stable overexpression of Livin inhibited the activation of caspase-3 and led to resistance to cisplatin, while the knockdown of Livin by siRNA rendered colon cancer cells more sensitive to cisplatin. Our study, along with others, highlighted the potential of Livin for cancer therapy in colon cancer.
Collapse
|
83
|
Zhao F, Lam EWF. Role of the forkhead transcription factor FOXO-FOXM1 axis in cancer and drug resistance. Front Med 2012; 6:376-80. [PMID: 23124885 DOI: 10.1007/s11684-012-0228-0] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Accepted: 09/26/2012] [Indexed: 01/04/2023]
Abstract
The forkhead transcription factors FOXO and FOXM1 have pivotal roles in tumorigenesis and in mediating chemotherapy sensitivity and resistance. Recent research shows that the forkhead transcription factor FOXM1 is a direct transcriptional target repressed by the forkhead protein FOXO3a, a vital downstream effector of the PI3K-AKT-FOXO signaling pathway. Intriguingly, FOXM1 and FOXO3a also compete for binding to the same gene targets, which have a role in chemotherapeutic drug action and sensitivity. An understanding of the role and regulation of the FOXO-FOXM1 axis will impact directly on our knowledge of chemotherapeutic drug action and resistance in patients, and provide new insights into the design of novel therapeutic strategy and reliable biomarkers for prediction of drug sensitivity.
Collapse
Affiliation(s)
- Fung Zhao
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, UK
| | | |
Collapse
|
84
|
He L, Yang X, Cao X, Liu F, Quan M, Cao J. Casticin induces growth suppression and cell cycle arrest through activation of FOXO3a in hepatocellular carcinoma. Oncol Rep 2012; 29:103-8. [PMID: 23064420 DOI: 10.3892/or.2012.2076] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Accepted: 08/27/2012] [Indexed: 11/06/2022] Open
Abstract
Casticin, a polymethoxyflavone, has been reported to exert anticancer activities. The objectives of this study were to examine the molecular mechanisms by which casticin induces the growth inhibition and cell cycle arrest in human hepatocellular carcinoma (HCC) cells. The HCC cell lines Hep G2 and PLC/PRF/5 were cultured in vitro. The growth inhibitory effects of casticin were evaluated using clonogenic assays. The distribution of phases in the cell cycle was analyzed using flow cytometry (FCM) analysis with propidium iodide (PI) staining. Multiple molecular techniques, such as western blotting and gene transfection, were used to explore the molecular mechanisms of action. Our data demonstrated that casticin significantly inhibited cell viability and colony formation in HCC cells. Furthermore, it induced cell cycle arrest in the G2/M phase. Casticin inhibited phosphorylation of the FOXO3a protein and decreased the expression of FoxM1 and its downstream genes, such as cyclin-dependent kinase (CDK1), cdc25B and cyclin B and increased the expression of p27KIP1. Silencing of FOXO3a expression by small interfering RNA (siRNA) transfection clearly attenuated the inhibitory effects of casticin on FOXM1 expression and cell growth. Our findings provided clear evidence that casticin induces growth suppression and cell cycle arrest through inhibition of FOXO3a phosphorylation causing inactivation of FOXM1 in HCC cells.
Collapse
Affiliation(s)
- Lihua He
- Medical College, Hunan Normal University, Changsha 410013, PR China
| | | | | | | | | | | |
Collapse
|
85
|
Stem cell transcription factor NANOG controls cell migration and invasion via dysregulation of E-cadherin and FoxJ1 and contributes to adverse clinical outcome in ovarian cancers. Oncogene 2012; 32:3500-9. [PMID: 22945654 DOI: 10.1038/onc.2012.363] [Citation(s) in RCA: 121] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Revised: 05/24/2012] [Accepted: 07/04/2012] [Indexed: 12/26/2022]
Abstract
Ovarian cancer is the most lethal of all gynecological malignancies, and the identification of novel prognostic and therapeutic targets for ovarian cancer is crucial. It is believed that only a small subset of cancer cells are endowed with stem cell properties, which are responsible for tumor growth, metastatic progression and recurrence. NANOG is one of the key transcription factors essential for maintaining self-renewal and pluripotency in stem cells. This study investigated the role of NANOG in ovarian carcinogenesis and showed overexpression of NANOG mRNA and protein in the nucleus of ovarian cancers compared with benign ovarian lesions. Increased nuclear NANOG expression was significantly associated with high-grade cancers, serous histological subtypes, reduced chemosensitivity, and poor overall and disease-free survival. Further analysis showed NANOG is an independent prognostic factor for overall and disease-free survival. Moreover, NANOG was highly expressed in ovarian cancer cell lines with metastasis-associated property and in clinical samples of metastatic foci. Stable knockdown of NANOG impeded ovarian cancer cell proliferation, migration and invasion, which was accompanied by an increase in mRNA expression of E-cadherin, caveolin-1, FOXO1, FOXO3a, FOXJ1 and FOXB1. Conversely, ectopic NANOG overexpression enhanced ovarian cancer cell migration and invasion along with decreased E-cadherin, caveolin-1, FOXO1, FOXO3a, FOXJ1 and FOXB1 mRNA expression. Importantly, we found Nanog-mediated cell migration and invasion involved its regulation of E-cadherin and FOXJ1. This is the first report revealing the association between NANOG expression and clinical outcome of patients with ovarian cancers, suggesting NANOG to be a potential prognostic marker and therapeutic molecular target in ovarian cancer.
Collapse
|
86
|
Madia F, Grossi V, Peserico A, Simone C. Updates from the Intestinal Front Line: Autophagic Weapons against Inflammation and Cancer. Cells 2012; 1:535-57. [PMID: 24710489 PMCID: PMC3901109 DOI: 10.3390/cells1030535] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Revised: 07/04/2012] [Accepted: 08/01/2012] [Indexed: 12/11/2022] Open
Abstract
The intestine lies at the interface between the organism and its environment and responds to infection/inflammation in a multi-leveled manner, potentially leading to chronic inflammatory pathologies and cancer formation. Indeed, the immune response at the intestinal epithelium has been found to be involved in the origin and development of colorectal cancer, which is the third most commonly diagnosed neoplastic disease. Among the mechanisms induced upon inflammation, autophagy appears as a defensive strategy for the clearance of invading microbes and intracellular waste components. Autophagy has also been found to play an important role in colorectal cancer, where it seems to have a pro-survival or pro-death function depending on the stage of the neoplastic process. In this paper we discuss the dual role of autophagy in colorectal cancer and review evidence showing that modulation of autophagy affects the immune response and cancer biology. The study of key players involved in autophagy might contribute to the design of new approaches for colorectal cancer, consisting in combined therapies capable of modifying cancer-specific metabolism rather than simply evoking a generic apoptotic and/or autophagic response, thus enhancing the efficacy of currently used drugs and treatments.
Collapse
Affiliation(s)
- Federica Madia
- Laboratory of Signal-dependent Transcription, Department of Translational Pharmacology, Consorzio Mario Negri Sud, Santa Maria Imbaro (CH) 66030, Italy.
| | - Valentina Grossi
- Laboratory of Signal-dependent Transcription, Department of Translational Pharmacology, Consorzio Mario Negri Sud, Santa Maria Imbaro (CH) 66030, Italy.
| | - Alessia Peserico
- Laboratory of Signal-dependent Transcription, Department of Translational Pharmacology, Consorzio Mario Negri Sud, Santa Maria Imbaro (CH) 66030, Italy.
| | - Cristiano Simone
- Laboratory of Signal-dependent Transcription, Department of Translational Pharmacology, Consorzio Mario Negri Sud, Santa Maria Imbaro (CH) 66030, Italy.
| |
Collapse
|
87
|
Tezil T, Bodur C, Kutuk O, Basaga H. IKK-β mediates chemoresistance by sequestering FOXO3; a critical factor for cell survival and death. Cell Signal 2012; 24:1361-8. [PMID: 22313691 DOI: 10.1016/j.cellsig.2012.01.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Revised: 01/24/2012] [Accepted: 01/24/2012] [Indexed: 10/14/2022]
Abstract
Chemotherapeutic drugs proved only 50% successful in breast cancer because of cell type-dependent resistance mechanisms. FOXO3 is known to be involved in the regulation of several cell death-related genes; however, the extent of FOXO3 regulation in chemoresistance is still not fully understood. Here, we show that FOXO3 critically mediates cisplatin chemosensitivity of MCF-7 breast cancer cells which express higher levels of FOXO3 compared to resistant MDA-MB-231 cells. Administration of cisplatin induces apoptosis in MCF-7 cells in a FOXO3-dependent manner as indicated by RNA interference. On the other hand, IKK-β (IκB kinase) appears to inhibit FOXO3 action after cisplatin treatment and promotes chemoresistance in MDA-MB-231 cells. IKK-β directly interacts and sequesters FOXO3 in the cytosol preventing its nuclear localization. Moreover, cisplatin treatment induces autophagosome formation through LC-3 conversion while inhibiting the cleavage of caspase 9 and caspase 3 in MDA-MB-231 cells manipulated to overexpress FOXO3. In brief, our findings demonstrate that in addition to cellular level of active FOXO3, cisplatin chemoresistance is also regulated by IKK-β sequestration of FOXO3 in cytosol.
Collapse
Affiliation(s)
- Tugsan Tezil
- Sabanci University, Biological Sciences and Bioengineering Program, 34956, Tuzla, Istanbul, Turkey.
| | | | | | | |
Collapse
|
88
|
Yao LM, He JP, Chen HZ, Wang Y, Wang WJ, Wu R, Yu CD, Wu Q. Orphan receptor TR3 participates in cisplatin-induced apoptosis via Chk2 phosphorylation to repress intestinal tumorigenesis. Carcinogenesis 2011; 33:301-11. [DOI: 10.1093/carcin/bgr287] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
89
|
Obrador-Hevia A, Serra-Sitjar M, Rodríguez J, Villalonga P, Fernández de Mattos S. The tumour suppressor FOXO3 is a key regulator of mantle cell lymphoma proliferation and survival. Br J Haematol 2011; 156:334-45. [PMID: 22107151 DOI: 10.1111/j.1365-2141.2011.08951.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The FOXO3 (Forkhead/winged helix box class O 3) transcription factor is a crucial regulator of haematopoietic cell fate that controls proliferation and apoptosis, among other processes. Despite the central role of FOXO3 as a tumour suppressor and phosphatidylinositol 3-kinase (PI3K)/AKT effector, little is known about its involvement in mantle cell lymphoma (MCL) biology. This study investigated the expression and activity of FOXO3 in MCL cell lines and in primary cultures. We analysed the expression of key FOXO regulators and targets, and studied the effect of modulators of FOXO function on cell viability and apoptosis. FOXO3 was constitutively inactivated in MCL cell lines, and showed cytoplasmic localization in patient-derived cells. PI3K and AKT, but not mammalian target of rapamycin (mTOR), inhibitors induced FOXO3 nuclear translocation and activation in correlation with their impact on MCL proliferation and survival. Moreover, FOXO3-defective cells were resistant to PI3K/AKT inhibitors. Reactivation of FOXO function with a nuclear export inhibitor had a profound effect on cell viability, consistent with FOXO3 nuclear accumulation. Interestingly, inhibition of FOXO3 nuclear export enhanced the effect of doxorubicin. Taken together, our results confirm that FOXO3 is a relevant regulator of proliferation and apoptosis in MCL, and suggest that reactivation of FOXO3 function might be a useful therapeutic strategy in MCL patients.
Collapse
Affiliation(s)
- Antònia Obrador-Hevia
- Cancer Cell Biology Group, Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, Palma, Illes Balears, Crta Valldemossa km 7Æ5. E-07122 Palma, Illes Balears, Spain
| | | | | | | | | |
Collapse
|
90
|
Koo CY, Muir KW, Lam EWF. FOXM1: From cancer initiation to progression and treatment. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2011; 1819:28-37. [PMID: 21978825 DOI: 10.1016/j.bbagrm.2011.09.004] [Citation(s) in RCA: 314] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Revised: 09/20/2011] [Accepted: 09/21/2011] [Indexed: 10/17/2022]
Abstract
The Forkhead box protein M1 (FOXM1) transcription factor is a regulator of myriad biological processes, including cell proliferation, cell cycle progression, cell differentiation, DNA damage repair, tissue homeostasis, angiogenesis and apoptosis. Elevated FOXM1 expression is found in cancers of the liver, prostate, brain, breast, lung, colon, pancreas, skin, cervix, ovary, mouth, blood and nervous system, suggesting it has an integral role in tumorigenesis. Recent research findings also place FOXM1 at the centre of cancer progression and drug sensitivity. In this review the involvement of FOXM1 in various aspects of cancer, in particular its role and regulation within the context of cancer initiation, progression, and cancer drug response, will be summarised and discussed.
Collapse
Affiliation(s)
- Chuay-Yeng Koo
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, UK
| | | | | |
Collapse
|
91
|
Weidinger C, Krause K, Mueller K, Klagge A, Fuhrer D. FOXO3 is inhibited by oncogenic PI3K/Akt signaling but can be reactivated by the NSAID sulindac sulfide. J Clin Endocrinol Metab 2011; 96:E1361-71. [PMID: 21752881 DOI: 10.1210/jc.2010-2453] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Overactivation of the phosphatidylinositol 3-kinase (PI3K)/Akt pathway has emerged as a pivotal trigger of thyroid carcinogenesis. Recent findings from other tumor entities revealed that PI3K/Akt-driven carcinogenesis critically involves the inactivation of the tumor-suppressive transcription factor Forkhead box O (FOXO)-3. However, little is known about FOXO3 in the thyroid context. AIMS We sought to investigate the influence of the thyroid oncogenes BRAFV600E, H-RASV12, and p110α (H1074R) on the regulation of the PI3K downstream target FOXO3 in vitro. Furthermore, the impact of the expression of phosphatase and tensin homolog deleted from chromosome 10 (PTEN) on the transcriptional activity of FOXO3 was assessed in a panel of 44 human thyroid tumors. Finally, we tried to modulate FOXO3 activity pharmacologically with help of the nonsteroidal antiinflammatory drug sulindac sulfide (SS). RESULTS We found that the overexpression of p110α H1074R results in the inactivation of FOXO3 via its nuclear exclusion. In vivo, we observed a direct correlation between PTEN expression and the transcriptional activation of FOXO3. In vitro, we found that stimulation with SS reversed PI3K/Akt-driven inactivation of FOXO3, resulting in its nuclear relocation and a combined induction of the antiproliferative FOXO target genes Gadd45α and p27(kip1) and the proapoptotic FOXO target gene Bim in benign (FRTL-5) and malignant human thyrocytes (FTC-133). In agreement with this, SS promoted the cell cycle arrest and apoptosis in thyroid cells, which could be amplified by the transfection of exogenous FOXO3. CONCLUSION Our data suggest that deregulation of proapoptotic FOXO3 represents a central step in PI3K/Akt-mediated thyroid carcinogenesis. Thus, SS might represent an attractive pharmacological tool for targeting thyroid neoplasia with aberrant PI3K/Akt/FOXO3 signaling.
Collapse
Affiliation(s)
- Carl Weidinger
- Clinic of Endocrinology, Department of Medicine, University Hospital Essen, Hufelandstr 55, D-45147 Essen, Germany
| | | | | | | | | |
Collapse
|
92
|
FOXO3a represses VEGF expression through FOXM1-dependent and -independent mechanisms in breast cancer. Oncogene 2011; 31:1845-58. [PMID: 21860419 PMCID: PMC3232453 DOI: 10.1038/onc.2011.368] [Citation(s) in RCA: 128] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Vascular endothelial growth factor (VEGF) plays a central role in breast cancer development and progression, but the mechanisms that control its expression are poorly understood. Breast cancer tissue microarrays revealed an inverse correlation between the Forkhead transcription factor FOXO3a and VEGF expression. Using the lapatinib-sensitive breast cancer cell lines BT474 and SKBR3 as model systems, we tested the possibility that VEGF expression is negatively regulated by FOXO3a. Lapatinib treatment of BT474 or SKBR3 cells resulted in nuclear translocation and activation of FOXO3a, followed by a reduction in VEGF expression. Transient transfection and inducible expression experiments showed that FOXO3a represses the proximal VEGF promoter whereas another forkhead member, FOXM1, induces VEGF expression. Chromatin immunoprecipitation and oligonucleotide pull-down assays demonstrated that both FOXO3a and FOXM1 bind a consensus Forkhead response element (FHRE) in the VEGF promoter. Upon lapatinib stimulation, activated FOXO3a displaces FOXM1 bound to the FHRE before recruiting histone deacetylase 2 (HDAC2) to the promoter, leading to decreased histones H3 and H4 acetylation, and concomitant transcriptional inhibition of VEGF. These results show that FOXO3a-dependent repression of target genes in breast cancer cells, such as VEGF, involves competitive displacement of DNA-bound FOXM1 and active recruitment of transcriptional repressor complexes.
Collapse
|
93
|
Lööf J, Pfeifer D, Ding Z, Sun XF, Zhang H. Effects of ΔNp73β on cisplatin treatment in colon cancer cells. Mol Carcinog 2011; 51:628-35. [PMID: 21837762 DOI: 10.1002/mc.20835] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2010] [Revised: 05/14/2011] [Accepted: 07/05/2011] [Indexed: 11/11/2022]
Abstract
p73 can activate transcription of p53-responsive genes, thereby inhibiting cell growth. An alternative promoter in the TP73 gene gives rise to an N-terminally truncated isoform of p73, ΔNp73, which lacks the transactivation domain of the full length TAp73 protein. TAp73 is considered pro-apoptotic, and ΔNp73 anti-apoptotic. In this study, we overexpressed ΔNp73β in p53 wild type and p53 mutant colon cancer cell lines and further exposed the cells to cancer therapeutic drug cisplatin. The results showed that cisplatin decreased the protein expression levels of ΔNp73β in a dose-dependent manner, and both TAp73 and p53 were upregulated after cisplatin treatment. Further, clonogenic potential and cell viability were decreased, and apoptotic cells increased, in p53 mutant and in p53 wild type cells. Cellular viability was significantly higher in ΔNp73β-cells than mock-transfected cells. However, ΔNp73β overexpression did not affect the cellular susceptibility to cisplatin. In conclusion, the overexpression of ΔNp73β increases viability in p53 wild type and p53 mutant colon cancer cells, and cisplatin induces the degradation of ΔNp73β in a dose-dependent manner.
Collapse
Affiliation(s)
- Jasmine Lööf
- Division of Tumor Biology, Systems Biology Research Centre, University of Skövde, Skövde, Sweden
| | | | | | | | | |
Collapse
|
94
|
Edvardsson K, Ström A, Jonsson P, Gustafsson JÅ, Williams C. Estrogen receptor β induces antiinflammatory and antitumorigenic networks in colon cancer cells. Mol Endocrinol 2011; 25:969-79. [PMID: 21493669 DOI: 10.1210/me.2010-0452] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Several studies suggest estrogen to be protective against the development of colon cancer. Estrogen receptor β (ERβ) is the predominant estrogen receptor expressed in colorectal epithelium and is the main candidate to mediate the protective effects. We have previously shown that expression of ERβ reduces growth of colorectal cancer in xenografts. Little is known of the actions of ERβ and its effect on gene transcription in colon cancers. To dissect the processes that ERβ mediates and to investigate cell-specific mechanisms, we reexpressed ERβ in three colorectal cancer cell lines (SW480, HT29, and HCT-116) and conducted genome-wide expression studies in combination with gene-pathway analyses and cross-correlation to ERβ-chromatin-binding sites. Although induced gene regulation was cell specific, overrepresentation analysis of functional classes indicated that the same biological themes, including apoptosis, cell differentiation, and regulation of the cell cycle, were affected in all three cell lines. Novel findings include a strong ERβ-mediated down-regulation of IL-6 and downstream networks with significant implications for inflammatory mechanisms involved in colon carcinogenesis. We also discovered cross talk between the suggested nuclear receptor coregulator PROX1 and ERβ, demonstrating that ERβ both regulates and shares target genes with PROX1. The influence of ERβ on apoptosis was further explored using functional studies, which suggested an increased DNA-repair capacity. We conclude that reexpression of ERβ induces transcriptome changes that, through several parallel pathways, converge into antitumorigenic capabilities in all three cell lines. We propose that enhancing ERβ action has potential as a novel therapeutic approach for prevention and/or treatment of colon cancer.
Collapse
Affiliation(s)
- Karin Edvardsson
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, Texas 77204-5056, USA
| | | | | | | | | |
Collapse
|
95
|
A gene signature distinguishing CD133hi from CD133- colorectal cancer cells: essential role for EGR1 and downstream factors. Pathology 2011; 43:220-7. [DOI: 10.1097/pat.0b013e328344e391] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
96
|
Myatt SS, Brosens JJ, Lam EWF. Sense and sensitivity: FOXO and ROS in cancer development and treatment. Antioxid Redox Signal 2011; 14:675-87. [PMID: 20649462 DOI: 10.1089/ars.2010.3383] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Forkhead box O (FOXO) transcription factors are at the center of an emerging paradigm that links longevity, cell fate, and tumor development. Key to these processes is the ability of FOXO to regulate, and be regulated by, oxidative stress. Perturbation of the mechanisms that tightly couple reactive oxygen species (ROS) production, oxidative stress signaling, and FOXO activity to the subsequent cellular response is a pivotal step in cancer development and progression. Consequently, the ROS-FOXO pathway is a major therapeutic target in cancer, not only as it mediates the cellular response to chemotherapy, but also because it underpins drug resistance. As the intimate and reciprocal relation between FOXO and ROS is being unravelled, new opportunities arise to develop more-effective cancer treatments that circumvent resistance to the conventional cytotoxic drugs.
Collapse
Affiliation(s)
- Stephen S Myatt
- Cancer Research-UK Labs, Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, London, England, United Kingdom
| | | | | |
Collapse
|
97
|
Olmos Y, Brosens JJ, Lam EWF. Interplay between SIRT proteins and tumour suppressor transcription factors in chemotherapeutic resistance of cancer. Drug Resist Updat 2011; 14:35-44. [DOI: 10.1016/j.drup.2010.12.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2010] [Revised: 12/07/2010] [Accepted: 12/08/2010] [Indexed: 12/20/2022]
|
98
|
Akt-FOXO3a signaling axis dysregulation in human oral squamous cell carcinoma and potent efficacy of FOXO3a-targeted gene therapy. Oral Oncol 2010; 47:16-21. [PMID: 21109483 DOI: 10.1016/j.oraloncology.2010.10.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2010] [Revised: 10/17/2010] [Accepted: 10/18/2010] [Indexed: 02/01/2023]
Abstract
Phosphoinositide-3-kinase (PI3K)/Akt pathway has been shown to be activated in oral squamous cell carcinoma (OSCC). Activation of Akt suppresses FOXO transcription factor-mediated growth arrest and apoptosis in various cancers. We investigated FOXO3a and phosphor(p)-Akt expression and potential efficacy of FOXO3a-targeted gene therapy in OSCC. Low expression of FOXO3a was negatively associated with overexpression of p-Akt and histological grade using immunohistochemistry. Akt-FOXO3a axis was also examined by detection of FOXO3a expression after induction or inhibition of Akt activity in Tca8113 OSCC cells. Transfection of a constitutively active form of FOXO3a (FOXO3a(3A)) in OSCC cells induced significant G₁-phase arrest and apoptosis as compared with control and transfection of a wild-type FOXO3a (FOXO3a(WT)). Stable FOXO3a(3A) transfectant OSCC cells also revealed the most potent growth inhibition effect in vivo. Furthermore, the downstream effects of FOXO3a activation were found to be inhibition of CDK4/6 and cyclin D1, and accumulation of p27 and Bim. We also found that transcription of FOXO1 and FOXO4 were stimulated by FOXO3a. Our results suggest that FOXO3a activity may be important in tumorigenesis and development of OSCC. Akt-FOXO3a axis inhibition-mediated constitutively active FOXO3a induces significant growth inhibition and FOXO3a activation may present a potent target-based therapeutic strategy for OSCC therapy.
Collapse
|
99
|
Fang L, Wang H, Zhou L, Yu D. FOXO3a reactivation mediates the synergistic cytotoxic effects of rapamycin and cisplatin in oral squamous cell carcinoma cells. Toxicol Appl Pharmacol 2010; 251:8-15. [PMID: 21092744 DOI: 10.1016/j.taap.2010.11.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2010] [Revised: 11/08/2010] [Accepted: 11/09/2010] [Indexed: 11/15/2022]
Abstract
FOXO3a, a well-known transcriptional regulator, controls a wide spectrum of biological processes. The phosphoinositide-3-kinase (PI3K)/Akt signaling pathway inactivates FOXO3a via phosphorylation-induced nuclear exclusion and degradation. A loss or gain of FOXO3a activity has been correlated with efficiency of chemotherapies in various cancers including oral squamous cell carcinoma (OSCC). Therefore, in the current study, we have investigated the FOXO3a activity modulating and antitumor effects of rapamycin and cisplatin in OSCC cells. Cisplatin inhibited proliferation and induced apoptosis in a dose-dependent way in OSCC Tca8113 cells. Rapamycin alone had no effect on cell proliferation and apoptosis. Rapamycin downregulated the expression of S-phase kinase associated protein-2 (Skp2) and increased the FOXO3a protein stability but induced the upregulation of feedback Akt activation-mediated FOXO3a phosphorylation. Cisplatin decreased the phosphorylation of FOXO3a via Akt inhibition. Rapamycin combined with cisplatin as its feedback Akt activation inhibitor revealed the most dramatic FOXO3a nuclear localization and reactivation with the prevention of its feedback loop and exposed significant synergistic effects of decreased cell proliferation and increased apoptosis in vitro and decreased tumor size in vivo. Furthermore, the downstream effects of FOXO3a reactivation were found to be accumulation of p27 and Bim. In conclusion, rapamycin/cisplatin combination therapy boosts synergistic antitumor effects through the significant FOXO3a reactivation in OSCC cells. These results may represent a novel mechanism by which rapamycin/cisplatin combination therapy proves to be a potent molecular-targeted strategy for OSCC.
Collapse
Affiliation(s)
- Liang Fang
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 QingChun Road, Hangzhou 310003, PR China
| | | | | | | |
Collapse
|
100
|
Meng J, Fang B, Liao Y, Chresta CM, Smith PD, Roth JA. Apoptosis induction by MEK inhibition in human lung cancer cells is mediated by Bim. PLoS One 2010; 5:e13026. [PMID: 20885957 PMCID: PMC2946397 DOI: 10.1371/journal.pone.0013026] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2010] [Accepted: 08/31/2010] [Indexed: 11/19/2022] Open
Abstract
AZD6244 (ARRY-142886) is an inhibitor of MEK1/2 and can inhibit cell proliferation or induce apoptosis in a cell-type dependent manner. The precise molecular mechanism of AZD6244-induced apoptosis is not clear. To investigate mechanisms of AZD6244 induced apoptosis in human lung cancer, we determined the molecular changes of two subgroups of human lung cancer cell lines that are either sensitive or resistant to AZD6244 treatment. We found that AZD6244 elicited a large increase of Bim proteins and a smaller increase of PUMA and NOXA proteins, and induced cell death in sensitive lung cancer cell lines, but had no effect on other Bcl-2 related proteins in those cell lines. Knockdown of Bim by siRNA greatly increased the IC50 and reduced apoptosis for AZD6244 treated cells. We also found that levels of endogenous p-Thr32-FOXO3a and p-Ser253-FOXO3a were lower in AZD6244-sensitive cells than in AZD6244-resistant cells. In the sensitive cells, AZD6244 induced FOXO3a nuclear translocation required for Bim activation. Moreover, the silencing of FOXO3a by siRNA abrogated AZD6244-induced cell apoptosis. In addition, we found that transfection of constitutively active AKT up-regulated p-Thr32-FOXO3a and p-Ser253-FOXO3a expression and inhibited AZD6244-induced Bim expression in sensitive cells. These results show that Bim plays an important role in AZD6244-induced apoptosis in lung cancer cells and that the PI3K/AKT/FOXO3a pathway is involved in Bim regulation and susceptibility of lung cancer cells to AZD6244. These results have implications in the development of strategies to overcome resistance to MEK inhibitors.
Collapse
Affiliation(s)
- Jieru Meng
- Department of Thoracic and Cardiovascular Surgery, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America.
| | | | | | | | | | | |
Collapse
|