1
|
Song K, Artibani M. The role of DNA methylation in ovarian cancer chemoresistance: A narrative review. Health Sci Rep 2023; 6:e1235. [PMID: 37123549 PMCID: PMC10140645 DOI: 10.1002/hsr2.1235] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 04/07/2023] [Accepted: 04/10/2023] [Indexed: 05/02/2023] Open
Abstract
Background and Aims Ovarian cancer (OC) is the most lethal gynecological cancer. In 2018, it was responsible for over 180,000 deaths worldwide. The high mortality rate is the culmination of a lack of early diagnosis and high rates of chemotherapy resistance, which is synonymous with disease recurrence. Over the last two decades, an increasingly significant role of epigenetic mechanisms, in particular DNA methylation, has emerged. This review will discuss several of the most significant genes whose hypo/hypermethylation profiles are associated with chemoresistance. Aside from functionally elucidating and evaluating these epimutations, this review will discuss recent trials of DNA methyltransferase inhibitors (DNMTi). Finally, we will propose future directions that could enhance the feasibility of utilizing these candidate epimutations as clinical biomarkers. Methods To perform this review, a comprehensive literature search based on our keywords was conducted across the online databases PubMed and Google Scholar for identifying relevant studies published up until August 2022. Results Epimutations affecting MLH1, MSH2, and Ras-association domain family 1 isoform A (DNA damage repair and apoptosis); ATP-binding cassette subfamily B member 1 and methylation-controlled J (drug export); secreted frizzled-related proteins (Wnt/β-catenin signaling), neurocalcin delta (calcium and G protein-coupled receptor signaling), and zinc finger protein 671 all have potential as biomarkers for chemoresistance. However, specific uncertainties relating to these epimutations include histotype-specific differences, intrinsic versus acquired chemoresistance, and the interplay with complete surgical debulking. DNMTi for chemoresistant OC patients has shown some promise; however, issues surrounding their efficacy and dose-limiting toxicities remain; a personalized approach is required to maximize their effectiveness. Conclusion Establishing a panel of aberrantly methylated chemoresistance-related genes to predict chemoresponsiveness and patients' suitability to DNMTi could significantly reduce OC recurrence, while improving DNMTi therapy viability. To achieve this, a large-scale prospective genome-wide DNA methylation profile study that spans different histotypes, includes paired samples (before and after chemotherapy), and integrates transcriptomic and methylomic analysis, is warranted.
Collapse
Affiliation(s)
- Kaiyang Song
- Green Templeton CollegeUniversity of OxfordOxfordUK
| | - Mara Artibani
- Ovarian Cancer Cell Laboratory, Medical Research Council (MRC) Weatherall Institute of Molecular MedicineUniversity of OxfordOxfordUK
- Nuffield Department of Women's & Reproductive HealthUniversity of OxfordOxfordUK
| |
Collapse
|
2
|
Mashayekhi M, Asadi M, Hashemzadeh S, Vahedi A, Shanehbandi D, Al-Omar AF, Akbari M, Raeisi M. Promoter methylation levels of RASSF1 and ATIC genes are associated with lung cancer in Iranian patients. Horm Mol Biol Clin Investig 2023:hmbci-2022-0007. [PMID: 36584330 DOI: 10.1515/hmbci-2022-0007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 12/11/2022] [Indexed: 12/31/2022]
Abstract
OBJECTIVES Epigenetic alterations like methylation of tumor suppressor genes or oncogenes, in respiratory epithelium have been associated with lung cancer. Hypermethylation of genes promoter is an epigenetic event, and is responsible to tumor suppressor genes inactivation as well as oncogenes activation. This study aimed to assess the role of methylation status in promoter of RASSF1 and ATIC genes their potential implication in the pathogenesis of lung tumor in Iranian patients. METHODS In this study, we collected 100 tissue samples (50 lung cancer tissues and 50 adjacent non-cancerous lung tissues) from Iranian lung cancer patients. The genomic DNA was extracted, and methylation status of both RASSF1 and ATIC genes was investigated by methylation-sensitive high-resolution melting (MS-HRM) assay technique and Real-Time PCR. Cancer Genome Atlas (TCGA) dataset was also analyzed for further validation of the gene's methylation. RESULTS Methylation of RASSF1 gene promoter was significantly higher in lung tumor tissues. However, promoter methylation levels of ATIC gene was significantly lower in lung tumor tissues. These results were additionally confirmed by TCGA analysis. Promoter methylation of both RASSF1 and ATIC genes was significantly associated with lymph node metastasis, and clinical stage of lung cancer. The receiver operating characteristic (ROC) curve analysis indicated a high accuracy of promoter methylation in these genes as a diagnostic biomarker for lung cancer. CONCLUSIONS Methylation levels of both RASSF1 and ATIC genes promoters were associated with lung cancer pathogenesis in Iranian population, and may be a suitable biomarker for diagnosis and prognosis of lung cancer in early stage of tumorigenesis.
Collapse
Affiliation(s)
- Mahsa Mashayekhi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Milad Asadi
- Department of Basic Oncology, Health Institute of Ege University, Izmir, Türkiye
| | - Shahriar Hashemzadeh
- Tuberculosis and Lung Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Vahedi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahmad Faris Al-Omar
- Department of Basic Oncology, Health Institute of Ege University, Izmir, Türkiye
| | - Morteza Akbari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mortaza Raeisi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
3
|
Veerasamy T, Eugin Simon S, Tan KO. Emerging strategies for sensitization of therapy resistant tumors toward cancer therapeutics by targeting the Bcl-2 family, TGF-β, Wnt/β-Catenin, RASSF and miRNA regulated signaling pathways. Int J Biochem Cell Biol 2021; 137:106016. [PMID: 34082133 DOI: 10.1016/j.biocel.2021.106016] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 05/27/2021] [Accepted: 05/29/2021] [Indexed: 02/01/2023]
Abstract
Conventional chemotherapy relies on the cytotoxicity of chemo-drugs to inflict destructive effects on tumor cells. However, as most tumor cells develop resistance to chemo-drugs, small doses of chemo-drugs are unlikely to provide significant clinical benefits in cancer treatment while high doses of chemo-drugs have been shown to impact normal human cells negatively due to the non-specific nature and cytotoxicity associated with chemo-drugs. To overcome this challenge, sensitizations of tumor cells with bioactive molecules that specifically target the pro-survival and pro-apoptosis signaling pathways of the tumor cells are likely to increase the therapeutic impacts and improve the clinical outcomes by reducing the dependency and adverse effects associated with using high doses of chemo-drugs in cancer treatment. This review focuses on emerging strategies to enhance the sensitization of tumor cells toward cancer therapies based on our understanding of tumor cell biology and underlying signaling pathways.
Collapse
Affiliation(s)
- Tarmarajen Veerasamy
- Department of Biological Sciences, Sunway University, No. 5 Jalan Universiti, Bandar Sunway, 47500, Subang Jaya, Selangor, Malaysia
| | - Samson Eugin Simon
- Department of Biological Sciences, Sunway University, No. 5 Jalan Universiti, Bandar Sunway, 47500, Subang Jaya, Selangor, Malaysia
| | - Kuan Onn Tan
- Department of Biological Sciences, Sunway University, No. 5 Jalan Universiti, Bandar Sunway, 47500, Subang Jaya, Selangor, Malaysia.
| |
Collapse
|
4
|
Liu Y, Xia L, Dong L, Wang J, Xiao Q, Yu X, Zhu H. CircHIPK3 Promotes Gemcitabine (GEM) Resistance in Pancreatic Cancer Cells by Sponging miR-330-5p and Targets RASSF1. Cancer Manag Res 2020; 12:921-929. [PMID: 32104074 PMCID: PMC7023912 DOI: 10.2147/cmar.s239326] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 01/06/2020] [Indexed: 12/26/2022] Open
Abstract
Background Pancreatic cancer is one of the most common malignant diseases in the world. Gemcitabine chemotherapy remains the most important clinical treatment. However, research found that pancreatic cancer cells have chemoresistance to gemcitabine and the effect is not satisfactory. Therefore, it is urgent to find an effective early diagnosis and treatment strategy. Circular RNA is one of the most popular prognostic biomarkers in GEM-resistant PC. Materials and Methods The present study was designed to evaluate the role of circHIPK3 in PC. The expression of circHIPK3 in PC tissues and cells and its effect on proliferation, migration, invasion, EMT, and apoptosis were investigated in vitro; its effect on tumor xenografts was assessed in vivo. Used bioinformation analysis to predict which miRNAs could potentially interact with circHIPK3, mRNA, and miR-330-5p. Results RT-PCR showed that the level of circHIPK3 was increased in PC tumor tissues; moreover, circHIPK3 was also increased in GEM-resistant PC tumors tissues and GEM-resistant PC cells. Sh-circHIPK3 could knockdown circHIPK3 in PANC-1-GEM and SW-1990-GEM and could significantly inhibit cell proliferation, invasion, migration, EMT and enhance cell apoptosis, compare with control group, the tumor xenografts of circHIPK3 knockdown group were significantly smaller. CircHIPK3 served as a sponge for miR-330-5p, and miR-330-5p directly bound to the 3′ UTR of RASSF1 were revealed by dual luciferase assay and RIP in PC cells. CircHIPK3 knockdown of RASSF1 expression could neutralize the cytological function of PC cells by miR-330-5p inhibitor mediated GEM-resistance. Conclusion CircHIPK3 promotes gemcitabine (GEM) resistance in pancreatic cancer cells by targeting RASSF1 via miR-330-5p and regulates proliferation, invasive, migration, EMT, and apoptosis. Our research revealed that circHIPK3 may be a novel biomarker in GEM-resistant PC and could be used as a prognostic target.
Collapse
Affiliation(s)
- Yunfei Liu
- Department of Hepatobiliary and Pancreatic Surgery‖, Third Xiangya Hospital, Central South University, Changsha 410006, People's Republic of China
| | - Li Xia
- Department of Endocrinology, Third Xiangya Hospital, Central South University, Changsha, 410006, People's Republic of China
| | - Luo Dong
- Department of Hepatobiliary and Pancreatic Surgery‖, Third Xiangya Hospital, Central South University, Changsha 410006, People's Republic of China
| | - Jiale Wang
- Department of Hepatobiliary and Pancreatic Surgery‖, Third Xiangya Hospital, Central South University, Changsha 410006, People's Republic of China
| | - Qiangsheng Xiao
- Department of Hepatobiliary and Pancreatic Surgery‖, Third Xiangya Hospital, Central South University, Changsha 410006, People's Republic of China
| | - Xiao Yu
- Department of Hepatobiliary and Pancreatic Surgery‖, Third Xiangya Hospital, Central South University, Changsha 410006, People's Republic of China
| | - Hongwei Zhu
- Department of Gastroenterology and Hepatology, Third Xiangya Hospital, Central South University, Changsha, 410006, People's Republic of China
| |
Collapse
|
5
|
Dubois F, Keller M, Hoflack J, Maille E, Antoine M, Westeel V, Bergot E, Quoix E, Lavolé A, Bigay-Game L, Pujol JL, Langlais A, Morin F, Zalcman G, Levallet G. Role of the YAP-1 Transcriptional Target cIAP2 in the Differential Susceptibility to Chemotherapy of Non-Small-Cell Lung Cancer (NSCLC) Patients with Tumor RASSF1A Gene Methylation from the Phase 3 IFCT-0002 Trial. Cancers (Basel) 2019; 11:cancers11121835. [PMID: 31766357 PMCID: PMC6966477 DOI: 10.3390/cancers11121835] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 11/12/2019] [Accepted: 11/19/2019] [Indexed: 12/27/2022] Open
Abstract
RASSF1 gene methylation predicts longer disease-free survival (DFS) and overall survival (OS) in patients with early-stage non-small-cell lung cancer treated using paclitaxel-based neo-adjuvant chemotherapy compared to patients receiving a gemcitabine-based regimen, according to the randomized Phase 3 IFCT (Intergroupe Francophone de Cancérologie Thoracique)-0002 trial. To better understand these results, this study used four human bronchial epithelial cell (HBEC) models (HBEC-3, HBEC-3-RasV12, A549, and H1299) and modulated the expression of RASSF1A or YAP-1. Wound-healing, invasion, proliferation and apoptosis assays were then carried out and the expression of YAP-1 transcriptional targets was quantified using a quantitative polymerase chain reaction. This study reports herein that gemcitabine synergizes with RASSF1A, silencing to increase the IAP-2 expression, which in turn not only interferes with cell proliferation but also promotes cell migration. This contributes to the aggressive behavior of RASSF1A-depleted cells, as confirmed by a combined knockdown of IAP-2 and RASSF1A. Conversely, paclitaxel does not increase the IAP-2 expression but limits the invasiveness of RASSF1A-depleted cells, presumably by rescuing microtubule stabilization. Overall, these data provide a functional insight that supports the prognostic value of RASSF1 gene methylation on survival of early-stage lung cancer patients receiving perioperative paclitaxel-based treatment compared to gemcitabine-based treatment, identifying IAP-2 as a novel biomarker indicative of YAP-1-mediated modulation of chemo-sensitivity in lung cancer.
Collapse
Affiliation(s)
- Fatéméh Dubois
- Normandie Université, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP CYCERON, 14074 Caen, France; (F.D.); (M.K.); (E.M.); (E.B.)
- Department of Pathology, CHU de Caen, 14033 Caen, France
| | - Maureen Keller
- Normandie Université, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP CYCERON, 14074 Caen, France; (F.D.); (M.K.); (E.M.); (E.B.)
- Normandie Université, UNICAEN, UPRES-EA2608, 14032 Caen, France
| | - Julien Hoflack
- Normandie Université, UNICAEN, UPRES-EA2608, 14032 Caen, France
| | - Elodie Maille
- Normandie Université, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP CYCERON, 14074 Caen, France; (F.D.); (M.K.); (E.M.); (E.B.)
- Normandie Université, UNICAEN, INSERM UMR 1086 ANTICIPE, 14032 Caen, France
| | - Martine Antoine
- Department of Pathology, Hôpital Tenon, AP-HP, 75020 Paris, France;
| | - Virginie Westeel
- Department of Pneumology, University Hospital of Besançon, University Bourgogne Franche-Comté, 25000 Besançon, France;
| | - Emmanuel Bergot
- Normandie Université, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP CYCERON, 14074 Caen, France; (F.D.); (M.K.); (E.M.); (E.B.)
- Department of Pulmonology & Thoracic Oncology, CHU de Caen, 14033 Caen, France
| | - Elisabeth Quoix
- Department of Pneumology, University Hospital, 67000 Strasbourg, France;
| | - Armelle Lavolé
- Sorbonne Université, GRC n 04, Theranoscan, AP-HP, Service de Pneumologie, Hôpital Tenon, 75020 Paris, France;
| | - Laurence Bigay-Game
- Pneumology Department, Toulouse-Purpan, University Hospital Toulouse, 31300 Toulouse, France;
| | - Jean-Louis Pujol
- Département d’Oncologie Thoracique, CHU Montpellier, Univ. Montpellier, 34595 Montpellier, France;
| | - Alexandra Langlais
- Intergroupe Francophone de Cancérologie Thoracique (IFCT), 75009 Paris, France; (A.L.); (F.M.)
| | - Franck Morin
- Intergroupe Francophone de Cancérologie Thoracique (IFCT), 75009 Paris, France; (A.L.); (F.M.)
| | - Gérard Zalcman
- U830 INSERM “Genetics and Biology of Cancers, A.R.T Group”, Curie Institute, 75005 Paris, France
- Department of Thoracic Oncology & CIC1425, Hôpital Bichat-Claude Bernard, Assistance Publique Hôpitaux de Paris, Université Paris-Diderot, 75018 Paris, France
- Correspondence: (G.Z.); (G.L.); Tel.: +33-(0)140-257-502 (G.Z.); +33-(0)231-063-134 (G.L.)
| | - Guénaëlle Levallet
- Normandie Université, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP CYCERON, 14074 Caen, France; (F.D.); (M.K.); (E.M.); (E.B.)
- Department of Pathology, CHU de Caen, 14033 Caen, France
- Correspondence: (G.Z.); (G.L.); Tel.: +33-(0)140-257-502 (G.Z.); +33-(0)231-063-134 (G.L.)
| |
Collapse
|
6
|
Xiaochuan B, Qianfeng J, Min X, Xiao L. RASSF1 promotes cardiomyocyte apoptosis after acute myocardial infarction and is regulated by miR-125b. J Cell Biochem 2019; 121:489-496. [PMID: 31595551 DOI: 10.1002/jcb.29236] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 06/05/2019] [Indexed: 02/01/2023]
Abstract
Cardiomyocyte apoptosis is a common pathological injury in association with acute myocardial infarction (AMI). In the current study, the relationship between Ras-association domain family 1 (RASSF1) and cardiomyocyte apoptosis was investigated. RASSF1 was significantly over expressed in infarcted myocardial tissues as well as in cardiomyocytes induced by hypoxia. Inhibition of RASSF1 expression alleviated cardiomyocytes apoptosis induced by hypoxia in vitro and reduced cardiomyocytes apoptosis after AMI in vivo. RASSF1 expression was directly modulated by miR-125b, which was further confirmed by luciferase reporter assay. The current study verified that the miR-125b/RASSF1 axis was involved in cardiomyocytes apoptosis. To sum up, these results suggest that RASSF1 downregulation alleviated infarction-induced cardiomyocytes apoptosis and was regulated by miR-125b.
Collapse
Affiliation(s)
- Bai Xiaochuan
- Department of Cardiovascular Medicine of the First People's Hospital of Zunyi City, Guizhou, China
| | - Jiang Qianfeng
- Department of Cardiovascular Medicine of the First People's Hospital of Zunyi City, Guizhou, China
| | - Xu Min
- Department of Cardiovascular Medicine of the First People's Hospital of Zunyi City, Guizhou, China
| | - Liang Xiao
- Department of Cardiovascular Medicine of the First People's Hospital of Zunyi City, Guizhou, China
| |
Collapse
|
7
|
Mirza-Aghazadeh-Attari M, Ostadian C, Saei AA, Mihanfar A, Darband SG, Sadighparvar S, Kaviani M, Samadi Kafil H, Yousefi B, Majidinia M. DNA damage response and repair in ovarian cancer: Potential targets for therapeutic strategies. DNA Repair (Amst) 2019; 80:59-84. [PMID: 31279973 DOI: 10.1016/j.dnarep.2019.06.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 06/01/2019] [Accepted: 06/15/2019] [Indexed: 12/24/2022]
Abstract
Ovarian cancer is among the most lethal gynecologic malignancies with a poor survival prognosis. The current therapeutic strategies involve surgery and chemotherapy. Research is now focused on novel agents especially those targeting DNA damage response (DDR) pathways. Understanding the DDR process in ovarian cancer necessitates having a detailed knowledge on a series of signaling mediators at the cellular and molecular levels. The complexity of the DDR process in ovarian cancer and how this process works in metastatic conditions is comprehensively reviewed. For evaluating the efficacy of therapeutic agents targeting DNA damage in ovarian cancer, we will discuss the components of this system including DDR sensors, DDR transducers, DDR mediators, and DDR effectors. The constituent pathways include DNA repair machinery, cell cycle checkpoints, and apoptotic pathways. We also will assess the potential of active mediators involved in the DDR process such as therapeutic and prognostic candidates that may facilitate future studies.
Collapse
Affiliation(s)
- Mohammad Mirza-Aghazadeh-Attari
- Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Caspian Ostadian
- Department of Biology, Faculty of Science, Urmia University, Urmia, Iran
| | - Amir Ata Saei
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Ainaz Mihanfar
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Saber Ghazizadeh Darband
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 171 77, Sweden; Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Shirin Sadighparvar
- Neurophysiology Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Mojtaba Kaviani
- School of Nutrition and Dietetics, Acadia University, Wolfville, Nova Scotia, Canada
| | | | - Bahman Yousefi
- Molecular MedicineResearch Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Maryam Majidinia
- Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
8
|
Chen T, Sun Z, Liu F, Wang Q. RASSF1A and SIRT6 in non-small cell lung cancer: Relationship with clinical outcome. Oncol Lett 2017; 14:5759-5764. [PMID: 29204192 PMCID: PMC5707562 DOI: 10.3892/ol.2017.6172] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 04/26/2017] [Indexed: 12/13/2022] Open
Abstract
This study investigated the expression of RASSF1A and SIRT6 in non-small cell lung cancer (NSCLC) and its relationship with clinical prognosis. The expression in 122 cases of NSCLC tissues (NSCLC group) and 122 cases of normal lung tissues (NOR group) during the same period were detected by immunohistochemical Super Pic Ture™ Polymer two-step method, and the relationship between its expression and the clinicopathological features and prognosis of patients was analyzed. The positive expression rates of RASSF1A and SIRT6 in NSCLC group were lower than those in the normal group (55.74 vs. 84.43% and 52.46 vs. 82.49%, P<0.01). The differences in expression intensity of RASSF1A in NSCLC tissues between different tumor pathological types, tumor differentiation degrees and lymph node metastases were statistically significant, and the differences in expression intensity of SIRT6 between different TNM stages, tumor differentiation degrees and lymph node metastases were statistically significant. There was a positive correlation between the expression of RASSF1A and SIRT6 in NSCLC group (r=0.532, P<0.01). The 3-year survival rate of patients with high-expression of RASSF1A was higher than in those with low-expression of RASSF1A (81.33 vs. 39.45%, log-rank χ2=19,102, P<0.01); the 3-year survival rate of patients with high-expression of SIRT6 was higher than in those with low-expression of SIRT6 (83.51 vs. 42.43%, log-rank χ2=17,180, P<0.01). The low expression of RASSF1A and SIRT6 and lymph node metastasis were the risk factors affecting the prognosis of NSCLC patients. There is a better correlation between the expression of RASSF1A and SIRT6 in NSCLC tissues, and the detection of their expression is of great significance in the judgement of clinicopathological features and prognosis of NSCLC patients.
Collapse
Affiliation(s)
- Tao Chen
- The Second Department of Surgery, Chinese Medicine Hospital in Linyi City, Linyi, Shandong 276002, P.R. China
| | - Zhaojun Sun
- The Second Department of Surgery, Chinese Medicine Hospital in Linyi City, Linyi, Shandong 276002, P.R. China
| | - Fengling Liu
- The Second Department of Surgery, Chinese Medicine Hospital in Linyi City, Linyi, Shandong 276002, P.R. China
| | - Qiang Wang
- The Second Department of Surgery, Chinese Medicine Hospital in Linyi City, Linyi, Shandong 276002, P.R. China
| |
Collapse
|
9
|
Lakshminarasimhan R, Andreu-Vieyra C, Lawrenson K, Duymich CE, Gayther SA, Liang G, Jones PA. Down-regulation of ARID1A is sufficient to initiate neoplastic transformation along with epigenetic reprogramming in non-tumorigenic endometriotic cells. Cancer Lett 2017; 401:11-19. [PMID: 28483516 DOI: 10.1016/j.canlet.2017.04.040] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 04/25/2017] [Accepted: 04/26/2017] [Indexed: 12/28/2022]
Abstract
The chromatin remodeler AT-Rich Interactive Domain 1A (ARID1A) is frequently mutated in ovarian clear cell carcinoma (OCCC) and endometriosis precursor lesions. Here, we show that knocking down ARID1A in an immortalized endometriosis cell line is sufficient to induce phenotypic changes indicative of neoplastic transformation as evidenced by higher efficiency of anchorage-independent growth, increased propensity to adhere to collagen, and greater capacity to invade basement membrane extract in vitro. ARID1A knockdown is associated with expression dysregulation of 99 target genes, and many of these expression changes are also observed in primary OCCC tissues. Further, pathway analysis indicates these genes fall within networks highly relevant to tumorigenesis including integrin and paxillin pathways. We demonstrate that the down-regulation of ARID1A does not markedly alter global chromatin accessibility or DNA methylation but unexpectedly, we find strong increases in the active H3K27ac mark in promoter regions and decreases of H3K27ac at potential enhancers. Taken together, these data provide evidence that ARID1A mutation can be an early stage event in the oncogenic transformation of endometriosis cells giving rise to OCCC.
Collapse
Affiliation(s)
- Ranjani Lakshminarasimhan
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Claudia Andreu-Vieyra
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Kate Lawrenson
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA; Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Suite 290W, Los Angeles, CA, USA
| | - Christopher E Duymich
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Simon A Gayther
- Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Suite 290W, Los Angeles, CA, USA; Center for Bioinformatics and Functional Genomics, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Gangning Liang
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | - Peter A Jones
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Biochemistry & Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Van Andel Research Institute, Grand Rapids, MI 49503, USA.
| |
Collapse
|
10
|
Boyanapalli SSS, Li W, Fuentes F, Guo Y, Ramirez CN, Gonzalez XP, Pung D, Kong ANT. Epigenetic reactivation of RASSF1A by phenethyl isothiocyanate (PEITC) and promotion of apoptosis in LNCaP cells. Pharmacol Res 2016; 114:175-184. [PMID: 27818231 DOI: 10.1016/j.phrs.2016.10.021] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 10/21/2016] [Accepted: 10/24/2016] [Indexed: 12/23/2022]
Abstract
Epigenetic silencing of tumor suppressor genes is a phenomenon frequently observed in multiple cancers. Ras-association domain family 1 isoform A (RASSF1A) is a well-characterized tumor suppressor that belongs to the Ras-association domain family. Several studies have demonstrated that hypermethylation of the RASSF1A promoter is frequently observed in lung, prostate, and breast cancers. Phenethyl isothiocyanate (PEITC), a phytochemical abundant in cruciferous vegetables, possesses chemopreventive activities; however, its potential involvement in epigenetic mechanisms remains elusive. The present study aimed to examine the role of PEITC in the epigenetic reactivation of RASSF1A and the induction of apoptosis in LNCaP cells. LNCaP cells were treated for 5days with 0.01% DMSO, 2.5 or 5μM PETIC or 2.5μM azadeoxycytidine (5-Aza) with 0.5μM trichostatin A (TSA). We evaluated the effects of these treatments on CpG demethylation using methylation-specific polymerase chain reaction (MSP) and bisulfite genomic sequencing (BGS). CpG demethylation was significantly enhanced in cells treated with 5μM PEITC and 5-Aza+TSA; therefore, the latter treatment was used as a positive control in subsequent experiments. The decrease in RASSF1A promoter methylation correlated with an increase in expression of the RASSF1A gene in a dose-dependent manner. To confirm that promoter demethylation was mediated by DNA methyltransferases (DNMTs), we analyzed the expression levels of DNMTs and histone deacetylases (HDACs) at the gene and protein levels. PEITC reduced DNMT1, 3A and 3B protein levels in a dose-dependent manner, and 5μM PEITC significantly reduced DNMT3A and 3B protein levels. HDAC1, 2, 4 and 6 protein expression was also inhibited by 5μM PEITC. The combination of 5-Aza and TSA, a DNMT inhibitor and a HDAC inhibitor, respectively, was used as a positive control as this treatment significantly inhibited both HDACs and DNMTs. The function of RASSF1A reactivation in promoting apoptosis and inducing G2/M cell cycle arrest was analyzed using flow-cytometry analysis with Annexin V and propidium iodide (PI). Growth inhibition effect on LNCaP cells were investigated by colony formation assay. In addition, we analyzed p21, caspase-3 and 7, Bax, and Cyclin B1 protein levels. Flow-cytometry analysis of cells stained with PI alone demonstrated that 5μM PEITC promotes early apoptosis and G2/M cell cycle arrest. Flow cytometry analysis of cells stained with Annexin V and PI also demonstrated an increased proportion of cells in early apoptosis in cells treated with 5μM PEITC or 5-Aza with TSA. PEITC and efficiently inhibit colony numbers and total area. In addition, 5μM PEITC significantly enhanced p21, caspase-3, 7 and Bax levels and reduced Cyclin B1 expression compared with the control group. Collectively, the results of our study suggest that PEITC induces apoptosis in LNCaP cells potentially by reactivating RASSF1A via epigenetic mechanisms.
Collapse
Affiliation(s)
- Sarandeep S S Boyanapalli
- Center for Cancer Prevention Research, Department of Pharmaceutics, Ernest-Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road Piscataway, NJ, 08854, United States
| | - Wenji Li
- Center for Cancer Prevention Research, Department of Pharmaceutics, Ernest-Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road Piscataway, NJ, 08854, United States
| | - Francisco Fuentes
- Center for Cancer Prevention Research, Department of Pharmaceutics, Ernest-Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road Piscataway, NJ, 08854, United States
| | - Yue Guo
- Center for Cancer Prevention Research, Department of Pharmaceutics, Ernest-Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road Piscataway, NJ, 08854, United States
| | - Christina N Ramirez
- Center for Cancer Prevention Research, Department of Pharmaceutics, Ernest-Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road Piscataway, NJ, 08854, United States
| | - Ximena-Parades Gonzalez
- Center for Cancer Prevention Research, Department of Pharmaceutics, Ernest-Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road Piscataway, NJ, 08854, United States
| | - Douglas Pung
- Center for Cancer Prevention Research, Department of Pharmaceutics, Ernest-Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road Piscataway, NJ, 08854, United States
| | - Ah-Ng Tony Kong
- Center for Cancer Prevention Research, Department of Pharmaceutics, Ernest-Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road Piscataway, NJ, 08854, United States.
| |
Collapse
|
11
|
Amato E, Barbi S, Fassan M, Luchini C, Vicentini C, Brunelli M, Malleo G, Scarpa A, Malpeli G. RASSF1 tumor suppressor gene in pancreatic ductal adenocarcinoma: correlation of expression, chromosomal status and epigenetic changes. BMC Cancer 2016; 16:11. [PMID: 26754001 PMCID: PMC4710004 DOI: 10.1186/s12885-016-2048-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 01/06/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The Ras Association Domain Family Member 1 (RASSF1) is one of the most frequently reported methylation-inactivated tumor suppressor genes in primary pancreatic ductal adenocarcinomas (PDAC). Limited information is still available about the impact of RASSF1 gene silencing on the expression of its different isoforms in neoplastic cells. METHODS A series of 96 primary PDAC, with known clinico-pathological parameters, was tested for RASSF1 methylation status by methylation-specific PCR, RASSF1 locus copy number alterations by fluorescence in situ hybridization, and Rassf1a protein expression by immunohistochemistry. A further series of 14 xenografted primary PDAC and 8 PDAC-derived cell lines were tested to obtain a detailed methylation mapping of CpG islands A and C of the RASSF1 locus by pyrosequencing and to evaluate the expression of Rassf1 variants by qRT-PCR. RESULTS Methylation of CpG island A of the RASSF1 gene was observed in 35% of the tumors and allelic loss of RASSF1 locus was seen in 30 disomic and in 20 polysomic cases (52%). Rassf1a immunohistochemical expression was downregulated in half of primary PDAC, and this downregulation was neither correlated with methylation of RASSF1 promoter nor with RASSF1 copy number alterations. RASSF1 status did not influence patients' prognosis. The expression of the seven RASSF1 isoforms in xenografts and cell lines showed that RASSF1A, RASSF1B, and RASSF1C isoforms were present in all xenografts and cell lines, whereas RASSF1D, RASSF1E, and RASSF1F isoforms were variably expressed among samples. RASSF1G was never expressed in either xenografts or cell lines. The variable expression of RASSF1 isoforms in PDAC xenografts and cell lines was not dependent on RASSF1 methylation status of CpG islands A and C. CONCLUSIONS RASSF1 alterations occurring in PDAC mainly consist in variations of expression of the different isoforms. Different genetic mechanisms seem to contribute to RASSF1 deregulation in this setting, but RASSF1 methylation does not seem to substantially affect RASSF1 isoforms expression.
Collapse
Affiliation(s)
- Eliana Amato
- ARC-NET Centre for Applied Research on Cancer, Department of Pathology and Diagnostics, The Hospital and University of Verona, Verona, Italy.
| | - Stefano Barbi
- ARC-NET Centre for Applied Research on Cancer, Department of Pathology and Diagnostics, The Hospital and University of Verona, Verona, Italy.
| | - Matteo Fassan
- ARC-NET Centre for Applied Research on Cancer, Department of Pathology and Diagnostics, The Hospital and University of Verona, Verona, Italy.
| | - Claudio Luchini
- ARC-NET Centre for Applied Research on Cancer, Department of Pathology and Diagnostics, The Hospital and University of Verona, Verona, Italy.
- Department of Pathology, The Hospital and University of Verona, Verona, Italy.
| | - Caterina Vicentini
- ARC-NET Centre for Applied Research on Cancer, Department of Pathology and Diagnostics, The Hospital and University of Verona, Verona, Italy.
| | - Matteo Brunelli
- ARC-NET Centre for Applied Research on Cancer, Department of Pathology and Diagnostics, The Hospital and University of Verona, Verona, Italy.
| | - Giuseppe Malleo
- Department of Surgery and Oncology, The Hospital and University of Verona, Verona, Italy.
| | - Aldo Scarpa
- ARC-NET Centre for Applied Research on Cancer, Department of Pathology and Diagnostics, The Hospital and University of Verona, Verona, Italy.
- Department of Pathology, The Hospital and University of Verona, Verona, Italy.
| | - Giorgio Malpeli
- ARC-NET Centre for Applied Research on Cancer, Department of Pathology and Diagnostics, The Hospital and University of Verona, Verona, Italy.
- Department of Surgery and Oncology, The Hospital and University of Verona, Verona, Italy.
| |
Collapse
|
12
|
The tumor suppressive role of RASSF1A in osteosarcoma through the Wnt signaling pathway. Tumour Biol 2016; 37:8869-77. [PMID: 26750098 DOI: 10.1007/s13277-015-4660-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 12/15/2015] [Indexed: 01/10/2023] Open
Abstract
Ras-association domain family 1 isoform A (RASSF1A) is a tumor suppressor gene and its expression is lost in numerous types of cancer cells, including primary osteosarcoma cells. However, its functional significance in osteosarcoma has not been well defined. The messenger RNA (mRNA) expression of RASSF1A in osteosarcoma tissues and corresponding non-tumoral tissues was measured by real-time PCR. Overexpression of RASSF1A was established by an adenoviral vector expressing RASSF1A. Cell migration and invasion were analyzed in transwells. Apoptosis and cell cycle were analyzed using flow cytometry. Wnt/β-catenin activity was measured by TCF reporter dual-luciferase assay. Cell viability was measured by MTT assay. Protein expression was detected by Western blot. RASSF1A mRNA expression was significantly lower in osteosarcoma tissues than that in the corresponding non-tumoral tissues. The lowered RASSF1A expression correlated with the clinical severity of osteosarcoma. rAd-RASSF1A injection significantly inhibited the growth of xenograft MNNG/HOS tumors in mice. Overexpression of RASSF1A resulted in significant inhibition of the proliferation, migration, and invasion; induced apoptosis; and arrested cell cycle at G0/G1 phase in both the MNNG/HOS and SaOS2 cells. Overexpression of RASSF1A inhibited the Wnt/β-catenin activity, decreased phosphorylation of Akt/glycogen synthase kinase-3-β (GSK3-β), and increased phosphorylation of mammalian sterile 20-like kinase 1 (MST1). Overexpression of RASSF1A downregulated the cyclin D1, c-Myc, and matrix metalloproteinase-7 (MMP-7) protein levels. RASSF1A functions as a tumor suppressor in osteosarcoma and exerts anti-cancer roles through regulating Akt/GSK-3-Wnt/β-catenin signaling.
Collapse
|
13
|
Du Z, Ma K, Sun X, Li A, Wang H, Zhang L, Lin F, Feng X, Song J. Methylation of RASSF1A gene promoter and the correlation with DNMT1 expression that may contribute to esophageal squamous cell carcinoma. World J Surg Oncol 2015; 13:141. [PMID: 25886188 PMCID: PMC4403718 DOI: 10.1186/s12957-015-0557-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 03/23/2015] [Indexed: 01/10/2023] Open
Abstract
Background Esophageal squamous cell carcinoma is one of the most common malignancies in the world. Studies have confirmed that there are many genes abnormally hypermethylated in esophageal squamous cell carcinoma. The objective is to detect methylation of the RASSF1A gene promoter and the expression of the DNA methyltransferase 1 (DNMT1) protein in esophageal cancer tissue and discuss their relationship with esophageal squamous cell carcinoma. Methods The CpG island methylation status of RASSF1A genes were analyzed in 100 cases of tumor specimens as well as their adjacent tissues which was used for methylation-specific polymerase chain reaction (MSP). The expression of DNMT1 protein was determined by immunohistochemistry. Difference between measurement data and categorical data was compared through analysis of t test and chi-square test. All the statistics were taken with a bilateral test. The difference was statistically significant (P < 0.05). Results The promoter methylation of the RASSF1A gene promoter has been detected in 45 out of 100 (45%) esophageal squamous carcinoma cases, while methylation of RASSF1A gene has been detected in 2 out of 100 adjacent normal tissues (2%). The RASSF1A gene promoter was highly methylated in cancer tissues, and there were significant differences between normal esophagus tissues and esophageal squamous carcinoma (P < 0.05). The expression of DNMT1 protein has been detected in 61 out of 100 (61%) esophageal squamous carcinoma cases, including 41 cases in the above 45 methylated samples of RASSF1A gene promoter, and none in adjacent tissues. DNMT1 proteins are highly expressed in cancer tissues, and there were significant differences (P < 0.05). In positive cases for methylation of RASSF1A, the DNMT1 protein had been detected in 41 out of 45 (91%), while in non-methylated cancer cases, 20 out of 55(36.3%), and the difference is significant (P < 0.05). Conclusions Esophageal squamous carcinoma tumorigenesis may be related with hypermethylation of DNMT1 and RASSF1A promoter CpG island due to their high expression and also their hypermethylation.
Collapse
Affiliation(s)
- Zhenzong Du
- Department of Cardiothoracic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China.
| | - Kui Ma
- Department of Cardiothoracic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China.
| | - Xiaolin Sun
- Department of Cardiothoracic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China.
| | - Angui Li
- Department of Cardiothoracic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China.
| | - Haiyong Wang
- Department of Cardiothoracic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China.
| | - Lifei Zhang
- Department of Cardiothoracic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China.
| | - Feng Lin
- Department of Cardiothoracic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China.
| | - Xiaoyan Feng
- Department of Cardiothoracic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China.
| | - Jianfei Song
- Department of Cardiothoracic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, China.
| |
Collapse
|