1
|
Liu X, Xie X, Wang K, Liu X, Gong J, Yang Z, Li J. Raddeanin A suppresses intracellular 5Methylcytosine DNA modification engaged the metastasis of hepatocellular carcinoma. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119036. [PMID: 39515681 DOI: 10.1016/j.jep.2024.119036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/10/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Anemonoides Raddeana (Rege) Holubhe is commonly employed in clinical practice as a traditional Chinese medicine for the treatment of conditions such as rheumatism and limb numbness. Raddeanin A (RA), an active compound derived from this Traditional Chinese Medicine (TCM), demonstrates specific anticancer properties against many tumorigeneses. However, the molecular mechanism underlying its effects on hepatocellular carcinoma (HCC) remains unexplored. AIM OF THE STUDY The aim of this study is to investigate the inhibitory effects of RA in human HCC stimulated cells and its impact on DNA methylation in tumor cells, as well as to elucidate the molecular mechanisms underlying RA's anti-tumor activity. MATERIALS AND METHODS The inhibitory effects of RA on QGY-7703 and HepG2 cells were evaluated. The IC50 values were determined by employing non-linear sigmoidal curve fitting to analyze the normalized response. The impact of RA was investigated in cells overexpressing DNMT3A and DNMT3B. The effects of RA on cell cycle progression and apoptosis were assessed. Furthermore, the influence of RA on cellular methylation was determined, along with its effects on the expression levels of DNMT3A, DNMT3B, Bcl-2, Bax, and Caspase-3. RESULTS The findings demonstrate that RA induces cell cycle arrest at the G0/G1 phase and promotes apoptosis in hepatocellular carcinoma cells. Furthermore, RA effectively inhibits the invasion and migration of human HCC stimulated cells. The expression of DNMT3A and DNMT3B is downregulated by RA, effectively suppressing the intracellular 5mC DNA modification level. Moreover, the overexpression of these enzymes in RA-treated human HCC stimulated cells significantly impacts the overall 5mC level and hinders tumor metastasis by restricting migration and invasion. CONCLUSION The RA compound acts as an antagonist against HCC by reducing intracellular DNA 5mC levels through mechanisms mediated by methyltransferase. Moreover, RA demonstrates the capacity to induce apoptosis in tumor cells, thereby exerting its anti-tumor effects. The findings of this study provide valuable insights for enhancing the pharmacodynamic efficacy of RA in HCC treatment.
Collapse
Affiliation(s)
- Xin Liu
- Key Laboratory of Chinese Medicine Analysis, School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Department of General Surgery, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, China; Postgraduate School, Jilin Normal University, Siping, 136000, China
| | - Xiaoyan Xie
- Department of Pharmacy, The 3rd Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Kangyu Wang
- Key Laboratory of Chinese Medicine Analysis, School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Department of General Surgery, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, China
| | - Xiaokang Liu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China
| | - Jiyu Gong
- Key Laboratory of Chinese Medicine Analysis, School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Zizhao Yang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Department of General Surgery, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, China.
| | - Jiannan Li
- Key Laboratory of Chinese Medicine Analysis, School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, China.
| |
Collapse
|
2
|
Liu J, Ruan M, Liu Y, Hong X, Zhang L, Zhang Q. Identification of 3-(9H-carbazol-9-yl)-2-(1,3-dioxoisoindolin-2-yl)propanoic acids as promising DNMT1 inhibitors. Eur J Med Chem 2024; 274:116538. [PMID: 38823264 DOI: 10.1016/j.ejmech.2024.116538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/12/2024] [Accepted: 05/26/2024] [Indexed: 06/03/2024]
Abstract
DNA methyltransferase 1 (DNMT1) is the primary enzyme responsible for maintaining DNA methylation patterns during cellular division, crucial for cancer development by suppressing tumor suppressor genes. In this study, we retained the phthalimide structure of N-phthaloyl-l-tryptophan (RG108) and substituted its indole ring with nitrogen-containing aromatic rings of varying sizes. We synthesized 3-(9H-carbazol-9-yl)-2-(1,3-dioxoisoindolin-2-yl)propanoic acids and confirmed them as DNMT1 inhibitors through protein affinity testing, radiometric method using tritium labeled SAM, and MTT assay. Preliminary structure-activity relationship analysis revealed that introducing substituents on the carbazole ring could enhance inhibitory activity, with S-configuration isomers showing greater activity than R-configuration ones. Notably, S-3-(3,6-di-tert-butyl-9H-carbazol-9-yl)-2-(1,3-dioxoisoindolin-2-yl)propanoic acid (7r-S) and S-3-(1,3,6-trichloro-9H-carbazol-9-yl)-2-(1,3-dioxoisoindolin-2-yl)propanoic acid (7t-S) exhibited significant DNMT1 enzyme inhibition activity, with IC50 values of 8.147 μM and 0.777 μM, respectively (compared to RG108 with an IC50 above 250 μM). Moreover, they demonstrated potential anti-proliferative activity on various tumor cell lines including A2780, HeLa, K562, and SiHa. Transcriptome analysis and KEGG pathway enrichment of K562 cells treated with 7r-S and 7t-S identified differentially expressed genes (DEGs) related to apoptosis and cell cycle pathways. Flow cytometry assays further indicated that 7r-S and 7t-S induce apoptosis in K562 cells and arrest them in the G0/G1 phase in a concentration-dependent manner. Molecular docking revealed that 7t-S may bind to the methyl donor S-adenosyl-l-methionine (SAM) site in DNMT1 with an orientation opposite to RG108, suggesting potential for deeper penetration into the DNMT1 pocket and laying the groundwork for further modifications.
Collapse
Affiliation(s)
- Jingyi Liu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Minli Ruan
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Yueqin Liu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Xiaoqian Hong
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Lijun Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Qian Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, 201203, China.
| |
Collapse
|
3
|
Yan L, Geng Q, Cao Z, Liu B, Li L, Lu P, Lin L, Wei L, Tan Y, He X, Li L, Zhao N, Lu C. Insights into DNMT1 and programmed cell death in diseases. Biomed Pharmacother 2023; 168:115753. [PMID: 37871559 DOI: 10.1016/j.biopha.2023.115753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/15/2023] [Accepted: 10/17/2023] [Indexed: 10/25/2023] Open
Abstract
DNMT1 (DNA methyltransferase 1) is the predominant member of the DNMT family and the most abundant DNMT in various cell types. It functions as a maintenance DNMT and is involved in various diseases, including cancer and nervous system diseases. Programmed cell death (PCD) is a fundamental mechanism that regulates cell proliferation and maintains the development and homeostasis of multicellular organisms. DNMT1 plays a regulatory role in various types of PCD, including apoptosis, autophagy, necroptosis, ferroptosis, and others. DNMT1 is closely associated with the development of various diseases by regulating key genes and pathways involved in PCD, including caspase 3/7 activities in apoptosis, Beclin 1, LC3, and some autophagy-related proteins in autophagy, glutathione peroxidase 4 (GPX4) and nuclear receptor coactivator 4 (NCOA4) in ferroptosis, and receptor-interacting protein kinase 1-receptor-interacting protein kinase 3-mixed lineage kinase domain-like protein (RIPK1-RIPK3-MLKL) in necroptosis. Our study summarizes the regulatory relationship between DNMT1 and different types of PCD in various diseases and discusses the potential of DNMT1 as a common regulatory hub in multiple types of PCD, offering a perspective for therapeutic approaches in disease.
Collapse
Affiliation(s)
- Lan Yan
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qi Geng
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhiwen Cao
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bin Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Li Li
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Peipei Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lin Lin
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lini Wei
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yong Tan
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaojuan He
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Li Li
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ning Zhao
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
4
|
Wang LL, Li RT, Zang ZH, Song YX, Zhang YZ, Zhang TF, Wang FZ, Hao GP, Cao L. 6-Methoxydihydrosanguinarine exhibits cytotoxicity and sensitizes TRAIL-induced apoptosis of hepatocellular carcinoma cells through ROS-mediated upregulation of DR5. Med Oncol 2023; 40:266. [PMID: 37566135 DOI: 10.1007/s12032-023-02129-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 07/18/2023] [Indexed: 08/12/2023]
Abstract
6-methoxydihydrosanguinarine (6-MS), a natural benzophenanthridine alkaloid extracted from Macleaya cordata (Willd.) R. Br, has shown to trigger apoptotic cell death in cancer cells. However, the exact mechanisms involved have not yet been clarified. The current study reveals the underlying mechanisms of 6-MS-induced cytotoxicity in hepatocellular carcinoma (HCC) cells and investigates whether 6-MS sensitizes TNF-related apoptosis inducing ligand (TRAIL)-induced apoptosis. 6-MS was shown to suppress cell proliferation and trigger cell cycle arrest, DNA damage, and apoptosis in HCC cells. Mechanisms analysis indicated that 6-MS promoted reactive oxygen species (ROS) generation, JNK activation, and inhibits EGFR/Akt signaling pathway. DNA damage and apoptosis induced by 6-MS were reversed following N-acetyl-l-cysteine (NAC) treatment. The enhancement of PARP cleavage caused by 6-MS was abrogated by pretreatment with JNK inhibitor SP600125. Furthermore, 6-MS enhanced TRAIL-mediated HCC cells apoptosis by upregulating the cell surface receptor DR5 expression. Pretreatment with NAC attenuated 6-MS-upregulated DR5 protein expression and alleviated cotreatment-induced viability reduction, cleavage of caspase-8, caspase-9, and PARP. Overall, our results suggest that 6-MS exerts cytotoxicity by modulating ROS generation, EGFR/Akt signaling, and JNK activation in HCC cells. 6-MS potentiates TRAIL-induced apoptosis through upregulation of DR5 via ROS generation. The combination of 6-MS with TRAIL may be a promising strategy and warrants further investigation.
Collapse
Affiliation(s)
- Lin-Lin Wang
- School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, People's Republic of China
| | - Ruo-Tong Li
- Department of Pathology, Tai' an Central Hospital, Taian, 271000, People's Republic of China
| | - Zi-Heng Zang
- School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, People's Republic of China
| | - Yun-Xuan Song
- School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, People's Republic of China
| | - Yu-Zhe Zhang
- School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, People's Republic of China
| | - Teng-Fei Zhang
- School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, People's Republic of China
| | - Feng-Ze Wang
- School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, People's Republic of China
- Department of Pathology, The Second Affiliated Hospital of Shandong First Medical University, Taian, 271000, People's Republic of China
| | - Gang-Ping Hao
- School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, People's Republic of China.
| | - Lu Cao
- Department of Pathology, The Second Affiliated Hospital of Shandong First Medical University, Taian, 271000, People's Republic of China.
| |
Collapse
|
5
|
Bahrami A, Ferns GA. Diagnostic, Prognostic, and Therapeutic Value of miR-148b in Human Cancers. Curr Mol Med 2022; 22:860-869. [PMID: 34961461 DOI: 10.2174/1566524021666211213123315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 07/06/2021] [Accepted: 11/05/2021] [Indexed: 11/22/2022]
Abstract
MicroRNAs (miRs) is a class of conserved, small, noncoding RNA molecules that modulate gene expression post-transcriptionally. miR-148b is a member of miR- 148/152 family generally known to be a tumor suppressor via its effect on different signaling pathways and regulatory genes. Aberrant expression of miR-148b has recently been shown to be responsible for tumorigenesis of several different cancer types. This review discusses the current evidence regarding the involvement of miR-148b expression in human cancers and its potential clinical importance for tumor diagnosis, prognosis, and therapeutics.
Collapse
Affiliation(s)
- Afsane Bahrami
- Clinical Research Development Unit, Imam Reza Hospital, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Division of Medical Education, Brighton & Sussex Medical School, Brighton, Sussex, UK
| |
Collapse
|
6
|
Ozyerli-Goknar E, Bagci-Onder T. Epigenetic Deregulation of Apoptosis in Cancers. Cancers (Basel) 2021; 13:3210. [PMID: 34199020 PMCID: PMC8267644 DOI: 10.3390/cancers13133210] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/09/2021] [Accepted: 06/11/2021] [Indexed: 12/11/2022] Open
Abstract
Cancer cells possess the ability to evade apoptosis. Genetic alterations through mutations in key genes of the apoptotic signaling pathway represent a major adaptive mechanism of apoptosis evasion. In parallel, epigenetic changes via aberrant modifications of DNA and histones to regulate the expression of pro- and antiapoptotic signal mediators represent a major complementary mechanism in apoptosis regulation and therapy response. Most epigenetic changes are governed by the activity of chromatin modifying enzymes that add, remove, or recognize different marks on histones and DNA. Here, we discuss how apoptosis signaling components are deregulated at epigenetic levels, particularly focusing on the roles of chromatin-modifying enzymes in this process. We also review the advances in cancer therapies with epigenetic drugs such as DNMT, HMT, HDAC, and BET inhibitors, as well as their effects on apoptosis modulation in cancer cells. Rewiring the epigenome by drug interventions can provide therapeutic advantage for various cancers by reverting therapy resistance and leading cancer cells to undergo apoptotic cell death.
Collapse
Affiliation(s)
- Ezgi Ozyerli-Goknar
- Brain Cancer Research and Therapy Laboratory, Koç University School of Medicine, Istanbul 34450, Turkey;
- Research Center for Translational Medicine, Koç University, Istanbul 34450, Turkey
| | - Tugba Bagci-Onder
- Brain Cancer Research and Therapy Laboratory, Koç University School of Medicine, Istanbul 34450, Turkey;
- Research Center for Translational Medicine, Koç University, Istanbul 34450, Turkey
| |
Collapse
|
7
|
Gao L, Wu ZX, Assaraf YG, Chen ZS, Wang L. Overcoming anti-cancer drug resistance via restoration of tumor suppressor gene function. Drug Resist Updat 2021; 57:100770. [PMID: 34175687 DOI: 10.1016/j.drup.2021.100770] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 06/08/2021] [Accepted: 06/13/2021] [Indexed: 02/07/2023]
Abstract
The cytotoxic anti-cancer drugs cisplatin, paclitaxel, doxorubicin, 5-fluorouracil (5-FU), as well as targeted drugs including imatinib, erlotinib, and nivolumab, play key roles in clinical cancer treatment. However, the frequent emergence of drug resistance severely comprosises their anti-cancer efficacy. A number of studies indicated that loss of function of tumor suppressor genes (TSGs) is involved in the development of cancer drug resistance, apart from decreased drug influx, increased drug efflux, induction of anti-apoptosis mechanisms, alterations in tumor microenvironment, drug compartmentalization, enhanced DNA repair and drug inactivation. TSGs are involved in the pathogenesis of tumor formation through regulation of DNA damage repair, cell apoptosis, autophagy, proliferation, cell cycle progression, and signal transduction. Our increased understanding of TSGs in the past decades demonstrates that gene mutation is not the only reason that leads to the inactivation of TSGs. Loss of function of TSGs may be based on the ubiquitin-proteasome pathway, epigenetic and transcriptional regualtion, post-translation modifications like phosphorylation as well as cellular translocation of TSGs. As the above processes can constitute"druggable targets", these mechanisms provide novel therapeutic approaches in targeting TSGs. Some small molecule compounds targeting these approaches re-activated TSGs and reversed cancer drug resistance. Along this vein, functional restoration of TSGs is a novel and promising approach to surmount cancer drug resistance. In the current review, we draw a scenario based on the role of loss of function of TSGs in drug resistance, on mechanisms leading to inactivation of TSGs and on pharmacological agents acting on these mechanisms to overcome cancer drug resistance. This review discusses novel therapeutic strategies targeting TSGs and offers possible modalities to conquer cancer drug resistance.
Collapse
Affiliation(s)
- Lingyue Gao
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China; Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Zhuo-Xun Wu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, NY, 11439, USA
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, NY, 11439, USA.
| | - Lihui Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China; Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Shenyang, PR China.
| |
Collapse
|
8
|
Ozyerli-Goknar E, Sur-Erdem I, Seker F, Cingöz A, Kayabolen A, Kahya-Yesil Z, Uyulur F, Gezen M, Tolay N, Erman B, Gönen M, Dunford J, Oppermann U, Bagci-Onder T. The fungal metabolite chaetocin is a sensitizer for pro-apoptotic therapies in glioblastoma. Cell Death Dis 2019; 10:894. [PMID: 31772153 PMCID: PMC6879621 DOI: 10.1038/s41419-019-2107-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 09/12/2019] [Accepted: 10/16/2019] [Indexed: 01/19/2023]
Abstract
Glioblastoma Multiforme (GBM) is the most common and aggressive primary brain tumor. Despite recent developments in surgery, chemo- and radio-therapy, a currently poor prognosis of GBM patients highlights an urgent need for novel treatment strategies. TRAIL (TNF Related Apoptosis Inducing Ligand) is a potent anti-cancer agent that can induce apoptosis selectively in cancer cells. GBM cells frequently develop resistance to TRAIL which renders clinical application of TRAIL therapeutics inefficient. In this study, we undertook a chemical screening approach using a library of epigenetic modifier drugs to identify compounds that could augment TRAIL response. We identified the fungal metabolite chaetocin, an inhibitor of histone methyl transferase SUV39H1, as a novel TRAIL sensitizer. Combining low subtoxic doses of chaetocin and TRAIL resulted in very potent and rapid apoptosis of GBM cells. Chaetocin also effectively sensitized GBM cells to further pro-apoptotic agents, such as FasL and BH3 mimetics. Chaetocin mediated apoptosis sensitization was achieved through ROS generation and consequent DNA damage induction that involved P53 activity. Chaetocin induced transcriptomic changes showed induction of antioxidant defense mechanisms and DNA damage response pathways. Heme Oxygenase 1 (HMOX1) was among the top upregulated genes, whose induction was ROS-dependent and HMOX1 depletion enhanced chaetocin mediated TRAIL sensitization. Finally, chaetocin and TRAIL combination treatment revealed efficacy in vivo. Taken together, our results provide a novel role for chaetocin as an apoptosis priming agent and its combination with pro-apoptotic therapies might offer new therapeutic approaches for GBMs.
Collapse
Affiliation(s)
- Ezgi Ozyerli-Goknar
- Brain Cancer Research and Therapy Laboratory, Koç University School of Medicine, 34450, Istanbul, Turkey
| | - Ilknur Sur-Erdem
- Brain Cancer Research and Therapy Laboratory, Koç University School of Medicine, 34450, Istanbul, Turkey
| | - Fidan Seker
- Brain Cancer Research and Therapy Laboratory, Koç University School of Medicine, 34450, Istanbul, Turkey
| | - Ahmet Cingöz
- Brain Cancer Research and Therapy Laboratory, Koç University School of Medicine, 34450, Istanbul, Turkey
| | - Alisan Kayabolen
- Brain Cancer Research and Therapy Laboratory, Koç University School of Medicine, 34450, Istanbul, Turkey
| | - Zeynep Kahya-Yesil
- Brain Cancer Research and Therapy Laboratory, Koç University School of Medicine, 34450, Istanbul, Turkey
| | - Fırat Uyulur
- Department of Computational Biology, Koç University, 34450, Istanbul, Turkey
| | - Melike Gezen
- Molecular Biology, Genetics and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul, Turkey
| | - Nazife Tolay
- Molecular Biology, Genetics and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul, Turkey
| | - Batu Erman
- Molecular Biology, Genetics and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul, Turkey
| | - Mehmet Gönen
- Department of Industrial Engineering, College of Engineering, Koç University, İstanbul, Turkey
| | - James Dunford
- Botnar Research Centre, NIHR Biomedical Research Centre Oxford, University of Oxford, Oxford, OX3 7LD, UK
| | - Udo Oppermann
- Botnar Research Centre, NIHR Biomedical Research Centre Oxford, University of Oxford, Oxford, OX3 7LD, UK
- Structural Genomics Consortium, University of Oxford, Oxford, OX3 7DQ, UK
- FRIAS, Freiburg Institute of Advanced Studies, University of Freiburg, 79104, Freiburg, Germany
| | - Tugba Bagci-Onder
- Brain Cancer Research and Therapy Laboratory, Koç University School of Medicine, 34450, Istanbul, Turkey.
| |
Collapse
|
9
|
Jing W, Song N, Liu YP, Qu XJ, Qi YF, Li C, Hou KZ, Che XF, Yang XH. DNMT3a promotes proliferation by activating the STAT3 signaling pathway and depressing apoptosis in pancreatic cancer. Cancer Manag Res 2019; 11:6379-6396. [PMID: 31372043 PMCID: PMC6635825 DOI: 10.2147/cmar.s201610] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 05/09/2019] [Indexed: 12/18/2022] Open
Abstract
Background Although aberrant DNA methyltransferase 3a (DNMT3a) expression is important to the tumorigenesis of pancreatic ductal adenocarcinoma (PDAC), the role of DNMT3a in PDAC prognosis is not clarified yet due to the limited studies and lacking of underlying molecular mechanism. Methods The expression of DNMT3a was examined by immunohistochemistry in PDAC tissues. Gene expression profiles assays were conducted to explore the impact of DNMT3a on biological processes and signal pathways. Cell cycle and apoptosis were measured by flow cytometry. Western blotting and real-time qPCR assays were used to explore the impact of DNMT3a on expression of protein and mRNA related to cell cycle, STAT3 signaling pathway and apoptosis. Results DNMT3a was overexpressed and closely associated with poor outcomes of PDAC. DNMT3a knockdown restrained PDAC cell proliferation, induced cell cycle arrest and promoted apoptosis in vitro. Affymetrix GeneChip Human Transcriptome Array identified that the cell cycle-related process was most significantly associated with DNMT3a. DNMT3a knockdown induced G1-S phase transition arrest by decreasing the expression of cyclin D1, which was mediated by the reduction of IL8 and the subsequent inactivation of STAT3 signaling pathway. Furthermore, exogenous apoptosis was also promoted after DNMT3a knockdown, probably via up-regulation of DNA transcription and expression in CASP8. Conclusion These findings indicate that DNMT3a plays an important role in PDAC progression. DNMT3a may serve as a prognostic biomarker and a therapeutic strategy candidate in PDAC.
Collapse
Affiliation(s)
- Wei Jing
- The First Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Na Song
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, People's Republic of China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Yun-Peng Liu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, People's Republic of China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Xiu-Juan Qu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, People's Republic of China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Ya-Fei Qi
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Ce Li
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, People's Republic of China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Ke-Zuo Hou
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, People's Republic of China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Xiao-Fang Che
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, People's Republic of China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Xiang-Hong Yang
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| |
Collapse
|
10
|
Elmallah MIY, Micheau O. Epigenetic Regulation of TRAIL Signaling: Implication for Cancer Therapy. Cancers (Basel) 2019; 11:cancers11060850. [PMID: 31248188 PMCID: PMC6627638 DOI: 10.3390/cancers11060850] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 06/11/2019] [Accepted: 06/18/2019] [Indexed: 12/20/2022] Open
Abstract
One of the main characteristics of carcinogenesis relies on genetic alterations in DNA and epigenetic changes in histone and non-histone proteins. At the chromatin level, gene expression is tightly controlled by DNA methyl transferases, histone acetyltransferases (HATs), histone deacetylases (HDACs), and acetyl-binding proteins. In particular, the expression level and function of several tumor suppressor genes, or oncogenes such as c-Myc, p53 or TRAIL, have been found to be regulated by acetylation. For example, HATs are a group of enzymes, which are responsible for the acetylation of histone proteins, resulting in chromatin relaxation and transcriptional activation, whereas HDACs by deacetylating histones lead to chromatin compaction and the subsequent transcriptional repression of tumor suppressor genes. Direct acetylation of suppressor genes or oncogenes can affect their stability or function. Histone deacetylase inhibitors (HDACi) have thus been developed as a promising therapeutic target in oncology. While these inhibitors display anticancer properties in preclinical models, and despite the fact that some of them have been approved by the FDA, HDACi still have limited therapeutic efficacy in clinical terms. Nonetheless, combined with a wide range of structurally and functionally diverse chemical compounds or immune therapies, HDACi have been reported to work in synergy to induce tumor regression. In this review, the role of HDACs in cancer etiology and recent advances in the development of HDACi will be presented and put into perspective as potential drugs synergizing with TRAIL's pro-apoptotic potential.
Collapse
Affiliation(s)
- Mohammed I Y Elmallah
- INSERM, Université Bourgogne Franche-Comté, LNC UMR1231, F-21079 Dijon, France.
- Chemistry Department, Faculty of Science, Helwan University, Ain Helwan 11795 Cairo, Egypt.
| | - Olivier Micheau
- INSERM, Université Bourgogne Franche-Comté, LNC UMR1231, F-21079 Dijon, France.
| |
Collapse
|
11
|
Ponnusamy M, Liu F, Zhang YH, Li RB, Zhai M, Liu F, Zhou LY, Liu CY, Yan KW, Dong YH, Wang M, Qian LL, Shan C, Xu S, Wang Q, Zhang YH, Li PF, Zhang J, Wang K. Long Noncoding RNA CPR (Cardiomyocyte Proliferation Regulator) Regulates Cardiomyocyte Proliferation and Cardiac Repair. Circulation 2019; 139:2668-2684. [PMID: 30832495 DOI: 10.1161/circulationaha.118.035832] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND The adult mammalian cardiomyocytes lose their proliferative capacity, which is responsible for cardiac dysfunction and heart failure following injury. The molecular mechanisms underlying the attenuation of adult cardiomyocyte proliferation remain largely unknown. Because long noncoding RNAs (lncRNAs) have a critical role in the development of cardiovascular problems, we investigated whether lncRNAs have any role in the regulation of cardiomyocyte proliferation and cardiac repair. METHODS Using bioinformatics and initial analysis, we identified an lncRNA, named CPR (cardiomyocyte proliferation regulator), that has a potential regulatory role in cardiomyocyte proliferation. For in vivo experiments, we generated CPR knockout and cardiac-specific CPR-overexpressing mice. In isolated cardiomyocytes, we used adenovirus for silencing (CPR-small interfering RNA) or overexpressing CPR. To investigate the mechanisms of CPR function in cardiomyocyte proliferation, we performed various analyses including quantitative reverse transcription-polymerase chain reaction, Western blot, histology, cardiac function (by echocardiography), transcriptome analyses (microarray assay), RNA pull-down assay, and chromatin immunoprecipitation assay. RESULTS CPR level is comparatively higher in the adult heart than in the fetal stage. The silencing of CPR significantly increased cardiomyocyte proliferation in postnatal and adult hearts. Moreover, CPR deletion restored the heart function after myocardial injury, which was evident from increased cardiomyocyte proliferation, improvement of myocardial function, and reduced scar formation. In contrast, the neonatal cardiomyocyte proliferation and cardiac regeneration were remarkably suppressed in CPR-overexpressing mice or adeno-associated virus serotype 9-CPR-overexpressing heart. These results indicate that CPR acts as a negative regulator of cardiomyocyte proliferation and regeneration. Next, we found that CPR targets minichromosome maintenance 3, an initiator of DNA replication and cell cycle progression, to suppress cardiomyocyte proliferation. CPR silenced minichromosome maintenance 3 expression through directly interacting and recruiting DNMT3A to its promoter cysteine-phosphate-guanine sites, as evident from decreased minichromosome maintenance 3 promoter methylation and increased minichromosome maintenance 3 expression in CPR knocked-down cardiomyocytes and CPR knockout mouse heart. These results were confirmed in CPR-overexpressing cardiomyocytes and CPR-overexpressing mouse heart. CONCLUSIONS Together, our findings identified that CPR is a suppressor of cardiomyocyte proliferation and indicated that lncRNAs take part in the regulation of cardiomyocyte proliferation and cardiac repair. Our study provides an lncRNA-based therapeutic strategy for effective cardiac repair and regeneration.
Collapse
Affiliation(s)
- Murugavel Ponnusamy
- Center for Developmental Cardiology, Institute for Translational Medicine, College of Medicine, Qingdao University, China (M.P., L.-Y.Z., C.-Y.L., K.-W.L., Y.-H.D., M.W., L.-L.Q., C.S., S.X., Q.W., Yan-Hui Zhang, P.-F.L., K.W.)
| | - Fang Liu
- Center of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, and Department of Anatomy, Guilin Medical University, China (Fang Liu)
| | - Yu-Hui Zhang
- State Key Laboratory of Cardiovascular Disease, Heart Failure Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China (Yu-Hui Zhang, M.Z., J.Z.)
| | - Rui-Bei Li
- School of Professional Studies, Northwestern University, Chicago, IL (R.-B.L.)
| | - Mei Zhai
- State Key Laboratory of Cardiovascular Disease, Heart Failure Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China (Yu-Hui Zhang, M.Z., J.Z.)
| | - Fei Liu
- Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine at Scott & White, Temple, TX (Fei Liu)
| | - Lu-Yu Zhou
- Center for Developmental Cardiology, Institute for Translational Medicine, College of Medicine, Qingdao University, China (M.P., L.-Y.Z., C.-Y.L., K.-W.L., Y.-H.D., M.W., L.-L.Q., C.S., S.X., Q.W., Yan-Hui Zhang, P.-F.L., K.W.)
| | - Cui-Yun Liu
- Center for Developmental Cardiology, Institute for Translational Medicine, College of Medicine, Qingdao University, China (M.P., L.-Y.Z., C.-Y.L., K.-W.L., Y.-H.D., M.W., L.-L.Q., C.S., S.X., Q.W., Yan-Hui Zhang, P.-F.L., K.W.)
| | - Kao-Wen Yan
- Center for Developmental Cardiology, Institute for Translational Medicine, College of Medicine, Qingdao University, China (M.P., L.-Y.Z., C.-Y.L., K.-W.L., Y.-H.D., M.W., L.-L.Q., C.S., S.X., Q.W., Yan-Hui Zhang, P.-F.L., K.W.)
| | - Yan-Han Dong
- Center for Developmental Cardiology, Institute for Translational Medicine, College of Medicine, Qingdao University, China (M.P., L.-Y.Z., C.-Y.L., K.-W.L., Y.-H.D., M.W., L.-L.Q., C.S., S.X., Q.W., Yan-Hui Zhang, P.-F.L., K.W.)
| | - Man Wang
- Center for Developmental Cardiology, Institute for Translational Medicine, College of Medicine, Qingdao University, China (M.P., L.-Y.Z., C.-Y.L., K.-W.L., Y.-H.D., M.W., L.-L.Q., C.S., S.X., Q.W., Yan-Hui Zhang, P.-F.L., K.W.)
| | - Li-Li Qian
- Center for Developmental Cardiology, Institute for Translational Medicine, College of Medicine, Qingdao University, China (M.P., L.-Y.Z., C.-Y.L., K.-W.L., Y.-H.D., M.W., L.-L.Q., C.S., S.X., Q.W., Yan-Hui Zhang, P.-F.L., K.W.)
| | - Chan Shan
- Center for Developmental Cardiology, Institute for Translational Medicine, College of Medicine, Qingdao University, China (M.P., L.-Y.Z., C.-Y.L., K.-W.L., Y.-H.D., M.W., L.-L.Q., C.S., S.X., Q.W., Yan-Hui Zhang, P.-F.L., K.W.)
| | - Sheng Xu
- Center for Developmental Cardiology, Institute for Translational Medicine, College of Medicine, Qingdao University, China (M.P., L.-Y.Z., C.-Y.L., K.-W.L., Y.-H.D., M.W., L.-L.Q., C.S., S.X., Q.W., Yan-Hui Zhang, P.-F.L., K.W.)
| | - Qi Wang
- Center for Developmental Cardiology, Institute for Translational Medicine, College of Medicine, Qingdao University, China (M.P., L.-Y.Z., C.-Y.L., K.-W.L., Y.-H.D., M.W., L.-L.Q., C.S., S.X., Q.W., Yan-Hui Zhang, P.-F.L., K.W.)
| | - Yan-Hui Zhang
- Center for Developmental Cardiology, Institute for Translational Medicine, College of Medicine, Qingdao University, China (M.P., L.-Y.Z., C.-Y.L., K.-W.L., Y.-H.D., M.W., L.-L.Q., C.S., S.X., Q.W., Yan-Hui Zhang, P.-F.L., K.W.)
| | - Pei-Feng Li
- Center for Developmental Cardiology, Institute for Translational Medicine, College of Medicine, Qingdao University, China (M.P., L.-Y.Z., C.-Y.L., K.-W.L., Y.-H.D., M.W., L.-L.Q., C.S., S.X., Q.W., Yan-Hui Zhang, P.-F.L., K.W.)
| | - Jian Zhang
- State Key Laboratory of Cardiovascular Disease, Heart Failure Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China (Yu-Hui Zhang, M.Z., J.Z.)
| | - Kun Wang
- Center for Developmental Cardiology, Institute for Translational Medicine, College of Medicine, Qingdao University, China (M.P., L.-Y.Z., C.-Y.L., K.-W.L., Y.-H.D., M.W., L.-L.Q., C.S., S.X., Q.W., Yan-Hui Zhang, P.-F.L., K.W.)
| |
Collapse
|
12
|
Elucidation for modulation of death receptor (DR) 5 to strengthen apoptotic signals in cancer cells. Arch Pharm Res 2019; 42:88-100. [DOI: 10.1007/s12272-018-01103-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 12/17/2018] [Indexed: 12/15/2022]
|
13
|
Nucleosidic DNA demethylating epigenetic drugs – A comprehensive review from discovery to clinic. Pharmacol Ther 2018; 188:45-79. [DOI: 10.1016/j.pharmthera.2018.02.006] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
14
|
Sanna L, Marchesi I, Melone MAB, Bagella L. The role of enhancer of zeste homolog 2: From viral epigenetics to the carcinogenesis of hepatocellular carcinoma. J Cell Physiol 2018; 233:6508-6517. [PMID: 29574790 DOI: 10.1002/jcp.26545] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 02/16/2018] [Indexed: 12/17/2022]
Abstract
Nowadays, epigenetics covers a crucial role in different fields of science. The enhancer of zeste homolog 2 (EZH2), the catalytic subunit of the Polycomb Repressive Complex 2 (PRC2), is a big proponent of how epigenetic changes can affect the initiation and progression of several diseases. Through its catalytic activity, responsible for the tri-methylation of lysine 27 of the histone H3 (H3K27me3), EZH2 is a good target for both diagnosis and therapy of different pathologies. A large number of studies have demonstrated its crucial role in cancer initiation and progression. Nevertheless, only recently its function in virus diseases has been uncovered; therefore, EZH2 can be an important promoter of viral carcinogenesis. This review explores the role of EZH2 in viral epigenetics based on recent progress that demonstrated the role of this protein in virus environment. In particular, the review focuses on EZH2 behavior in Hepatitis B Virus, analyzing its role in the rise of Hepatocellular Carcinoma.
Collapse
Affiliation(s)
- Luca Sanna
- Department of Biomedical Science, and National Institute of Biostructures and Biosystems, University of Sassari, Sassari, Italy
| | - Irene Marchesi
- Department of Biomedical Science, and National Institute of Biostructures and Biosystems, University of Sassari, Sassari, Italy
| | - Mariarosa A B Melone
- Department of Medical, Surgical, Neurological, Metabolic Sciences and Aging, Second Division of Neurology, Center for Rare Neurological e Neuromuscular Diseases and Interuniversity Center for Research in Neurosciences, University of Campania Luigi Vanvitelli, Naples, Italy.,Sbarro Institute for Cancer Research and Molecular Medicine, Department of Biology, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania
| | - Luigi Bagella
- Department of Biomedical Science, and National Institute of Biostructures and Biosystems, University of Sassari, Sassari, Italy.,Sbarro Institute for Cancer Research and Molecular Medicine, Department of Biology, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
15
|
Modulations of DNMT1 and HDAC1 are involved in the OTA-induced cytotoxicity and apoptosis in vitro. Chem Biol Interact 2017; 278:170-178. [PMID: 29080797 DOI: 10.1016/j.cbi.2017.10.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Revised: 09/29/2017] [Accepted: 10/20/2017] [Indexed: 12/16/2022]
Abstract
Ochratoxin A (OTA) as a fungal metabolite is reported to induce cytotoxicity and apoptosis through the mechanism of oxidative stress. Oxidative stress could induce the epigenetic enzymes modifications. However, whether epigenetic enzymes modifications are involved in OTA-induced cytotoxicity and apoptosis has not been reported until now. Therefore, the objectives of this study were to verify OTA-induced cytotoxicity and apoptosis and to investigate the potential role of epigenetic enzymes in OTA-induced cytotoxicity and apoptosis in PK15 cells. The results demonstrated that OTA at 4 μg/ml treatment for 12 h and 24 h induced cytotoxicity and apoptosis as demonstrated by decreasing cell viability, increasing LDH release, Annexin V/PI staining, Bcl-2/Bax mRNA ratio and apoptotic nuclei in PK15 cells. OTA treatment up-regulated ROS production and down-regulated GSH levels. In addition, OTA treatment activated the epigenetics related enzymes DNA methyltransferase 1 (DNMT1) and Histone deacetylase 1 (HDAC1). Adding DNMT1 inhibitor (5-Aza-2dc) or HDAC1 inhibitor (LBH589) depressed the up-regulation of DNMT1 or HDAC1 expression, the decreases of GSH levels and increases of ROS production induced by OTA, respectively. Furthermore, inhibition of DNMT1 or HDAC1 by their inhibitor reversed the decreases of cell viability and increases of LDH activity and apoptosis induced by OTA, respectively. In conclusion, the observed effects indicate that the critical modulation of DNMT1 and HDAC1 is related to OTA-induced cytotoxicity and apoptosis.
Collapse
|
16
|
Jiang C, Gong F. DNA Methyltransferase 1: A Potential Gene Therapy Target for Hepatocellular Carcinoma? Oncol Res Treat 2016; 39:448-52. [PMID: 27487275 DOI: 10.1159/000447414] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 05/25/2016] [Indexed: 11/19/2022]
Abstract
BACKGROUND DNA methyltransferase 1 (DNMT1) mutants display altered methylation patterns that may contribute to oncogenesis. We hypothesized that the silencing or inhibition of DNMT1 may affect the malignancy of hepatocellular carcinoma (HCC) cells. METHODS The HCC cell line KYN2 was used to construct 3 experimental groups: i) a DNMT1-siRNA group transfected with a green fluorescent protein (GFP) lentiviral vector to silence endogenous DNMT1 gene expression, which was confirmed by real-time quantitative polymerase chain reaction, ii) a 5-Aza-CdR group transfected with a null GFP lentiviral vector and treated with the DNMT1 inhibitor 5-aza-2'-deoxycytidine (5-Aza-CdR), and iii) a control group transfected with a null GFP lentiviral vector. Cellomics ArrayScan VTI imaging and MTT assays were conducted to assess cell proliferation. Cell cycle phase arrest and apoptosis were assayed by flow cytometry. Colony formation was assessed by fluorescence microscopy. RESULTS DNMT1 mRNA expression was significantly inhibited in DNMT1-silenced cells relative to control cells (p < 0.05), indicating successful transfection and gene expression knockdown. Cell proliferation was significantly inhibited in DNMT1-siRNA and 5-Aza-CdR cells relative to control cells (p < 0.05). G1-to-S phase shifts were significantly increased in DNMT1-siRNA and 5-Aza-CdR cells relative to control cells (p < 0.05). Apoptosis was significantly increased in DMNT1-siRNA and 5-Aza-CdR cells relative to control cells (p < 0.05). DMNT1-siRNA and 5-Aza-CdR cells displayed significantly reduced colony formation relative to control cells (p < 0.05). Notably, 5-Aza-CdR had more pronounced effects upon all these parameters than DNMT1 silencing. CONCLUSION DNMT1 activity appears to positively contribute to the malignancy of HCC cells.
Collapse
Affiliation(s)
- Changke Jiang
- Department of Pediatrics, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | | |
Collapse
|
17
|
Butcher LM, Ito M, Brimpari M, Morris TJ, Soares FAC, Ährlund-Richter L, Carey N, Vallier L, Ferguson-Smith AC, Beck S. Non-CG DNA methylation is a biomarker for assessing endodermal differentiation capacity in pluripotent stem cells. Nat Commun 2016; 7:10458. [PMID: 26822956 PMCID: PMC4740175 DOI: 10.1038/ncomms10458] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 12/11/2015] [Indexed: 01/07/2023] Open
Abstract
Non-CG methylation is an unexplored epigenetic hallmark of pluripotent stem cells. Here we report that a reduction in non-CG methylation is associated with impaired differentiation capacity into endodermal lineages. Genome-wide analysis of 2,670 non-CG sites in a discovery cohort of 25 phenotyped human induced pluripotent stem cell (hiPSC) lines revealed unidirectional loss (Δβ=13%, P<7.4 × 10(-4)) of non-CG methylation that correctly identifies endodermal differentiation capacity in 23 out of 25 (92%) hiPSC lines. Translation into a simplified assay of only nine non-CG sites maintains predictive power in the discovery cohort (Δβ=23%, P<9.1 × 10(-6)) and correctly identifies endodermal differentiation capacity in nine out of ten pluripotent stem cell lines in an independent replication cohort consisting of hiPSCs reprogrammed from different cell types and different delivery systems, as well as human embryonic stem cell (hESC) lines. This finding infers non-CG methylation at these sites as a biomarker when assessing endodermal differentiation capacity as a readout.
Collapse
Affiliation(s)
- Lee M Butcher
- UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6BT, UK
- Department of Surgery and Cancer, Imperial College London, London W12 0NN, UK
| | - Mitsuteru Ito
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| | - Minodora Brimpari
- Anne McLaren Laboratory, Department of Surgery, Wellcome Trust and Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge CB2 0SZ, UK
| | - Tiffany J Morris
- UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6BT, UK
- Cambridge Epigenetix, Jonas Webb Building, Babraham Campus, Cambridge CB22 3AT, UK
| | - Filipa A C Soares
- Anne McLaren Laboratory, Department of Surgery, Wellcome Trust and Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge CB2 0SZ, UK
- Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK
| | - Lars Ährlund-Richter
- Division of Paediatric Oncology, Department of Women's and Children's Health, Karolinska Institutet,171 76 Stockholm, Sweden
| | - Nessa Carey
- PraxisUnico, The Jeffreys Building, St John's Innovation Park, Cambridge CB4 0DE, UK
| | - Ludovic Vallier
- Anne McLaren Laboratory, Department of Surgery, Wellcome Trust and Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge CB2 0SZ, UK
- Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK
| | | | - Stephan Beck
- UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6BT, UK
| |
Collapse
|
18
|
Cacan E, Greer SF, Garnett-Benson C. Radiation-induced modulation of immunogenic genes in tumor cells is regulated by both histone deacetylases and DNA methyltransferases. Int J Oncol 2015; 47:2264-75. [PMID: 26458736 DOI: 10.3892/ijo.2015.3192] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 07/16/2015] [Indexed: 11/06/2022] Open
Abstract
Radiation treatment is a pivotal therapy for several cancer types, including colorectal cancer. It has been shown that sublethal doses of radiation modulate gene expression, making tumor cells more susceptible to T-cell-mediated immune attack. We have recently shown that low dose radiation enhances expression of multiple death receptors (Fas, DR4 and DR5) and co-stimulatory molecules (4-1BBL and OX-40L) in colorectal cancer (CRC) cells; however, it is unclear how ionizing radiation (IR) enhances expression of these molecules mechanistically. In the present study, we elucidate the molecular mechanisms by which radiation controls expression of these molecules in CRC. Here we report that, enhanced expression of these genes following radiation treatment of CRC cells is due, in part, to changes in DNA methylation and histone acetylation. We observed that radiation (5 Gy) significantly increased histone acetylation at the promoter regions of 4-1BBL, Fas and DR5 but not OX-40L. However, radiation did not induce changes in the global levels of acetylated histone H3 suggesting specificity of IR-induced changes. Furthermore, evaluation of epigenetic controlling enzymes revealed that IR did not alter overall cellular levels of HDACs (HDAC1, HDAC2 or HDAC3) or DNMTs (DNMT1, DNMT3a, or DNMT3b). Instead, radiation decreased binding of HDAC2 and HDAC3 at the promoter regions of Fas and 4-1BBL, respectively. Radiation also resulted in reduced DNMT1 at both the Fas and 4-1BBL promoter regions but not a control gene. We conclude that single dose radiation can influence the expression of immune response relevant genes in colorectal tumor cells by altering the binding of epigenetic enzymes, and modulating histone acetylation, at specific gene promoters.
Collapse
Affiliation(s)
- Ercan Cacan
- Department of Biology, Georgia State University, Atlanta, GA 30302, USA
| | - Susanna F Greer
- Department of Biology, Georgia State University, Atlanta, GA 30302, USA
| | | |
Collapse
|
19
|
Nayeb-Hashemi H, Desai A, Demchev V, Bronson RT, Hornick JL, Cohen DE, Ukomadu C. Targeted disruption of fibrinogen like protein-1 accelerates hepatocellular carcinoma development. Biochem Biophys Res Commun 2015. [PMID: 26225745 DOI: 10.1016/j.bbrc.2015.07.078] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Fibrinogen like protein-1 (Fgl1) is a predominantly liver expressed protein that has been implicated as both a hepatoprotectant and a hepatocyte mitogen. Fgl1 expression is decreased in hepatocellular carcinoma (HCC) and its loss correlates with a poorly differentiated phenotype. To better elucidate the role of Fgl1 in hepatocarcinogenesis, we treated mice wild type or null for Fgl1 with diethyl nitrosamine and monitored for incidence of hepatocellular cancer. We find that mice lacking Fgl1 develop HCC at more than twice the rate of wild type mice. We show that hepatocellular cancers from Fgl1 null mice are molecularly distinct from those of the wild type mice. In tumors from Fgl1 null mice there is enhanced activation of Akt and downstream targets of the mammalian target of rapamycin (mTOR). In addition, there is paradoxical up regulation of putative hepatocellular cancer tumor suppressors; tripartite motif-containing protein 35 (Trim35) and tumor necrosis factor super family 10b (Tnfrsf10b). Taken together, these findings suggest that Fgl1 acts as a tumor suppressor in hepatocellular cancer through an Akt dependent mechanism and supports its role as a potential therapeutic target in HCC.
Collapse
Affiliation(s)
- Hamed Nayeb-Hashemi
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine. Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Anal Desai
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine. Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Valeriy Demchev
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine. Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Roderick T Bronson
- Department of Microbiology and Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Jason L Hornick
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - David E Cohen
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine. Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Chinweike Ukomadu
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine. Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
20
|
Wu PH, Chen XM, Liu XQ, He JL, Feng Q, Lan X, Zhang X, Geng YQ, Wang YX, Ding YB. Activation of tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) receptor gene expression following DNA demethylation in placental choriocarcinoma and transformed cell lines. Reprod Fertil Dev 2015; 28:RD14408. [PMID: 26014898 DOI: 10.1071/rd14408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2014] [Accepted: 04/29/2015] [Indexed: 11/23/2022] Open
Abstract
We characterised DNA methylation and gene expression of four tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) receptors DR4, DR5, DcR1 and DcR2 in three choriocarcinoma (JAR, JEG-3, BeWo) and two transformed (HTR-8/SVneo and HPT-8) cell lines. DR4 mRNA was detected in JAR, JEG-3, BeWo and HTR-8/SVneo cells, whereas DR5 was present in all detected cells. DcR1 transcripts were expressed only in JAR, JEG-3 and BeWo cells, whereas DcR2 transcripts were detected only in HTR-8/SVneo and HPT-8 cells. Hypermethylated DR4 promoter was observed in JAR, JEG-3, BeWo and HTR-8/SVneo cells, hypermethylated DcR1 promoter in HTR-8/SVneo and HPT-8 cells and hypermethylated DcR2 promoter in JAR, JEG-3 and BeWo cells. Restoration of DR4, DcR1 and DcR2 expression with decreased DNA methylation of these genes was induced by the DNA demethylation agent 5-aza-2'-deoxycytidine (5-aza-CdR) in trophoblast cells, whereas DR5 expression did not exhibit any change. Significant negative correlation between the expression and DNA methylation of these genes was also observed. In all tested cell lines, only HPT-8 demonstrated sensitivity to TRAIL-induced apoptosis. Combined treatment with 5-aza-CdR and TRAIL resulted in apoptosis in JAR, JEG-3, BeWo and HTR-8/SVneo cells but not in HPT-8 cells. The results indicate that DNA methylation is associated with TRAIL receptor expression and might be involved in trophoblast apoptosis.
Collapse
|
21
|
Fang QL, Yin YR, Xie CR, Zhang S, Zhao WX, Pan C, Wang XM, Yin ZY. Mechanistic and biological significance of DNA methyltransferase 1 upregulated by growth factors in human hepatocellular carcinoma. Int J Oncol 2015; 46:782-90. [PMID: 25420499 DOI: 10.3892/ijo.2014.2776] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 10/30/2014] [Indexed: 11/06/2022] Open
Abstract
Dysregulation of growth factor signaling plays a pivotal role in controlling the malignancy phenotype and progression of hepatocellular carcinoma (HCC). However, the precise oncogenic mechanisms underlying transcription regulation of certain tumor suppressor genes (TSGs) by growth factors are poorly understood. In the present study, we report a novel insulin-like growth factor 1 (IGF1) pathway that mediates de novo DNA methylation and TSG (such as DLC1 and CHD5) silencing by upregulation of the DNA methyltransferase 1 (DNMT1) via an AKT/β-transducin repeat-containing protein (βTrCP)-mediated ubiquitin-proteasome pathway in HCC. Analysis of DNA methylation in CpG islands of target genes revealed high co-localization of DNMT1 and DNMT3B on the promoters of TSGs associated with enhanced CpG hypermethylation. Our results point to a novel epigenetic mechanism for growth factor-mediated repression of TSG transcription that involves DNA methylation.
Collapse
Affiliation(s)
- Qin-Liang Fang
- Department of Hepatobiliary Surgery, Zhongshan Hospital of Xiamen University, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Xiamen, Fujian 361004, P.R. China
| | - Yi-Rui Yin
- Department of Hepatobiliary Surgery, Zhongshan Hospital of Xiamen University, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Xiamen, Fujian 361004, P.R. China
| | - Cheng-Rong Xie
- Department of Hepatobiliary Surgery, Zhongshan Hospital of Xiamen University, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Xiamen, Fujian 361004, P.R. China
| | - Sheng Zhang
- Department of Hepatobiliary Surgery, Zhongshan Hospital of Xiamen University, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Xiamen, Fujian 361004, P.R. China
| | - Wen-Xiu Zhao
- Department of Hepatobiliary Surgery, Zhongshan Hospital of Xiamen University, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Xiamen, Fujian 361004, P.R. China
| | - Chao Pan
- Department of Pathology, Zhongshan Hospital of Xiamen University, Xiamen, Fujian 361004, P.R. China
| | - Xiao-Min Wang
- Department of Hepatobiliary Surgery, Zhongshan Hospital of Xiamen University, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Xiamen, Fujian 361004, P.R. China
| | - Zhen-Yu Yin
- Department of Hepatobiliary Surgery, Zhongshan Hospital of Xiamen University, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Xiamen, Fujian 361004, P.R. China
| |
Collapse
|
22
|
Fayyaz S, Yaylim I, Turan S, Kanwal S, Farooqi AA. Hepatocellular carcinoma: targeting of oncogenic signaling networks in TRAIL resistant cancer cells. Mol Biol Rep 2014; 41:6909-17. [PMID: 25037270 DOI: 10.1007/s11033-014-3577-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 07/01/2014] [Indexed: 01/18/2023]
Abstract
Apoptotic response in hepatocellular carcinoma (HCC) cells is impaired because of interconnectivity of proteins into complexes and signaling networks that are highly divergent in time and space. TNF-related apoptosis-inducing ligand (TRAIL) has emerged as an attractive anticancer agent reported to selectively induce apoptosis in cancer cells. Although diametrically opposed roles of TRAIL are reported both as an inducer of apoptosis and regulator of metastasis, overwhelmingly accumulating experimental evidence highlighting apoptosis inducing activity of TRAIL is directing TRAIL into clinical trials. Insights from TRAIL mediated signaling in HCC research are catalyzing new lines of study that should not only explain molecular mechanisms of disease but also highlight emerging paradigms in restoration of TRAIL mediated apoptosis in resistant cancer cells. It is becoming progressively more understandable that phytochemicals derived from edible plants have shown potential in modelling their interactions with their target proteins. Rapidly accumulating in vitro and in-vivo evidence indicates that phytonutrients have anticancer activity in rodent models of hepatocellular carcinoma. In this review we bring to limelight how phytonutrients restore apoptosis in hepatocellular carcinoma cells by rebalancing pro-apoptotic and anti-apoptotic proteins. Evidence has started to emerge, that reveals how phytonutrients target pharmacologically intractable proteins to suppress cancer. Target-based small-molecule discovery has entered into the mainstream research in the pharmaceutical industry and a better comprehension of the genetics of patients will be essential for identification of responders and non-responders.
Collapse
Affiliation(s)
- Sundas Fayyaz
- Laboratory for Translational Oncology and Personalized Medicine, Rashid Latif Medical College, Lahore, Pakistan
| | | | | | | | | |
Collapse
|
23
|
Farooqi AA, Yaylim I, Ozkan NE, Zaman F, Halim TA, Chang HW. Restoring TRAIL mediated signaling in ovarian cancer cells. Arch Immunol Ther Exp (Warsz) 2014; 62:459-74. [PMID: 25030086 DOI: 10.1007/s00005-014-0307-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 06/26/2014] [Indexed: 02/08/2023]
Abstract
Ovarian cancer has emerged as a multifaceted and genomically complex disease. Genetic/epigenetic mutations, suppression of tumor suppressors, overexpression of oncogenes, rewiring of intracellular signaling cascades and loss of apoptosis are some of the deeply studied mechanisms. In vitro and in vivo studies have highlighted different molecular mechanisms that regulate tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) mediated apoptosis in ovarian cancer. In this review, we bring to limelight, expansion in understanding systematical characterization of ovarian cancer cells has led to the rapid development of new drugs and treatments to target negative regulators of TRAIL mediated signaling pathway. Wide ranging synthetic and natural agents have been shown to stimulate mRNA and protein expression of death receptors. This review is compartmentalized into programmed cell death protein 4, platelet-derived growth factor signaling and miRNA control of TRAIL mediated signaling to ovarian cancer. Mapatumumab and PRO95780 have been tested for efficacy against ovarian cancer. Use of high-throughput screening assays will aid in dissecting the heterogeneity of this disease and increasing a long-term survival which might be achieved by translating rapidly accumulating information obtained from molecular and cellular studies to clinic researches.
Collapse
Affiliation(s)
- Ammad Ahmad Farooqi
- Laboratory for Translational Oncology and Personalized Medicine, RLMC, 35 km Ferozepur Road, Lahore, Pakistan,
| | | | | | | | | | | |
Collapse
|
24
|
Wongtrakoongate P, Li J, Andrews PW. Aza-deoxycytidine induces apoptosis or differentiation via DNMT3B and targets embryonal carcinoma cells but not their differentiated derivatives. Br J Cancer 2014; 110:2131-8. [PMID: 24603304 PMCID: PMC3992495 DOI: 10.1038/bjc.2014.128] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 02/13/2014] [Accepted: 02/14/2014] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Teratocarcinoma is a malignant male germ cell tumour, which contains stem cells and differentiated cancer tissues. DNMT3B has been shown to be highly expressed in human teratocarcinoma stem cells, and to mediate cytotoxicity of Aza-deoxycytidine (Aza-dC) in a pluripotent stem cell line NTERA2. METHODS We have established DNMT3B or POU5F1 (hereafter referred to as OCT4) knockdown in teratocarcinoma stem cells N2102Ep and TERA1 and in the pluripotent NTERA2 by a doxycycline-inducible system, and tested the cytotoxicity induced by Aza-dC. RESULTS Silencing of DNMT3B led to apoptosis of human teratocarcinoma stem cells N2102Ep and TERA1. Further, we found that induction of apoptosis or differentiation in NTERA2 and human embryonic stem cells by Aza-dC requires DNMT3B. To test whether Aza-dC inhibits proliferation of differentiated teratocarcinoma cells, we depleted OCT4 expression in N2102Ep and TERA1 cells treated with Aza-dC. Treatment with Aza-dC reduced cell number of differentiated cells to a lesser extent than their undifferentiated parental stem cells. Moreover, in contrast to the stem cells, Aza-dC failed to induce apoptosis of differentiated cells. CONCLUSIONS Our finding suggests that DNMT3B acts as an antiapoptotic gene in teratocarcinoma stem cells, and mediates apoptosis and differentiation of human pluripotent stem cells induced by Aza-dC, and that Aza-dC specifically induces apoptosis of teratocarcinoma stem cells.
Collapse
Affiliation(s)
- P Wongtrakoongate
- Centre for Stem Cell Biology, University of Sheffield, Sheffield S10 2TN, UK
| | - J Li
- Centre for Stem Cell Biology, University of Sheffield, Sheffield S10 2TN, UK
| | - P W Andrews
- Centre for Stem Cell Biology, University of Sheffield, Sheffield S10 2TN, UK
| |
Collapse
|
25
|
Wongtrakoongate P, Li J, Andrews PW. DNMT3B inhibits the re-expression of genes associated with induced pluripotency. Exp Cell Res 2014; 321:231-9. [PMID: 24333507 DOI: 10.1016/j.yexcr.2013.11.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 11/28/2013] [Accepted: 11/30/2013] [Indexed: 12/11/2022]
Abstract
DNMT3B is a de novo DNA methyltransferase that is highly expressed in mouse and human embryonic stem (ES) cells and has been shown to be essential for differentiation of mouse ES cells toward different lineages. In the present study, we found that DNMT3B is rapidly down-regulated in human ES cells during retinoic acid (RA)-induced differentiation compared with DNMT3A2, which is also highly expressed in ES cells. Silencing of DNMT3B in human ES cells by an inducible shRNAi system leads to a reduction of clonal ability of the stem cells, while expression of OCT4 and NANOG is unchanged. By contrast, the germline-specific genes VASA and SCP3 and the surface antigen BE12 are down regulated following DNMT3B knockdown. Upon retinoic acid-induced differentiation, we found that depletion of DNMT3B leads to a decrease in expression of the surface antigen A2B5 and of neural tube-associated genes PAX7 and BRN3A. Consistent with its importance in stem cell differentiation, we observed that silencing of DNMT3B facilitates the generation of cells that bear the hallmarks of pluripotency. Our findings suggest a role of DNMT3B in controlling the differentiation of human ES cells and in the generation of iPS cells.
Collapse
Affiliation(s)
- Patompon Wongtrakoongate
- Centre for Stem Cell Biology, University of Sheffield, Alfred Denny Building, Western Bank, S10 2TN, United Kingdom.
| | - Jianliang Li
- Centre for Stem Cell Biology, University of Sheffield, Alfred Denny Building, Western Bank, S10 2TN, United Kingdom
| | - Peter W Andrews
- Centre for Stem Cell Biology, University of Sheffield, Alfred Denny Building, Western Bank, S10 2TN, United Kingdom.
| |
Collapse
|
26
|
Xia L, Wang TD, Shen SM, Zhao M, Sun H, He Y, Xie L, Wu ZX, Han SF, Wang LS, Chen GQ. Phosphoproteomics study on the activated PKCδ-induced cell death. J Proteome Res 2013; 12:4280-301. [PMID: 23879269 DOI: 10.1021/pr400089v] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The proteolytic activation of protein kinase Cδ (PKCδ) generates a catalytic fragment called PKCδ-CF, which induces cell death. However, the mechanisms underlying PKCδ-CF-mediated cell death are largely unknown. On the basis of an engineering leukemic cell line with inducible expression of PKCδ-CF, here we employ SILAC-based quantitative phosphoproteomics to systematically and dynamically investigate the overall phosphorylation events during cell death triggered by PKCδ-CF expression. Totally, 3000 phosphorylation sites were analyzed. Considering the fact that early responses to PKCδ-CF expression initiate cell death, we sought to identify pathways possibly related directly with PKCδ by further analyzing the data set of phosphorylation events that occur in the initiation stage of cell death. Interacting analysis of this data set indicates that PKCδ-CF triggers complicated networks to initiate cell death, and motif analysis and biochemistry verification reveal that several kinases in the downstream of PKCδ conduct these networks. By analysis of the specific sequence motif of kinase-substrate, we also find 59 candidate substrates of PKCδ from the up-regulated phosphopeptides, of which 12 were randomly selected for in vitro kinase assay and 9 were consequently verified as substrates of PKCδ. To our greatest understanding, this study provides the most systematic analysis of phosphorylation events initiated by the cleaved activated PKCδ, which would vastly extend the profound understanding of PKCδ-directed signal pathways in cell death. The MS data have been deposited to the ProteomeXchange with identifier PXD000225.
Collapse
Affiliation(s)
- Li Xia
- The Department of Pathophysiology and Shanghai Universities E-Institute for Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine (SJTU-SM) , Shanghai, P.R. China , 200025
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Exposure to Penicillium mycotoxins alters gene expression of enzymes involved in the epigenetic regulation of bovine macrophages (BoMacs). Mycotoxin Res 2013; 29:235-43. [DOI: 10.1007/s12550-013-0174-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 07/02/2013] [Accepted: 07/05/2013] [Indexed: 11/26/2022]
|
28
|
Sato A, Saito Y, Sugiyama K, Sakasegawa N, Muramatsu T, Fukuda S, Yoneya M, Kimura M, Ebinuma H, Hibi T, Ikeda M, Kato N, Saito H. Suppressive effect of the histone deacetylase inhibitor suberoylanilide hydroxamic acid (SAHA) on hepatitis C virus replication. J Cell Biochem 2013; 114:1987-96. [DOI: 10.1002/jcb.24541] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Accepted: 03/05/2013] [Indexed: 01/16/2023]
Affiliation(s)
- Ayami Sato
- Division of Pharmacotherapeutics; Keio University Faculty of Pharmacy; Tokyo; 1058512; Japan
| | | | - Kazuo Sugiyama
- Department of Internal Medicine; Keio University School of Medicine; Tokyo; 1608582; Japan
| | - Noriko Sakasegawa
- Division of Pharmacotherapeutics; Keio University Faculty of Pharmacy; Tokyo; 1058512; Japan
| | - Toshihide Muramatsu
- Division of Pharmacotherapeutics; Keio University Faculty of Pharmacy; Tokyo; 1058512; Japan
| | - Shinya Fukuda
- Division of Pharmacotherapeutics; Keio University Faculty of Pharmacy; Tokyo; 1058512; Japan
| | - Mikiko Yoneya
- Division of Pharmacotherapeutics; Keio University Faculty of Pharmacy; Tokyo; 1058512; Japan
| | - Masaki Kimura
- Division of Pharmacotherapeutics; Keio University Faculty of Pharmacy; Tokyo; 1058512; Japan
| | - Hirotoshi Ebinuma
- Department of Internal Medicine; Keio University School of Medicine; Tokyo; 1608582; Japan
| | - Toshifumi Hibi
- Department of Internal Medicine; Keio University School of Medicine; Tokyo; 1608582; Japan
| | - Masanori Ikeda
- Department of Molecular Biology; Okayama University Graduate School of Medicine and Dentistry; Okayama; 7008558; Japan
| | - Nobuyuki Kato
- Department of Molecular Biology; Okayama University Graduate School of Medicine and Dentistry; Okayama; 7008558; Japan
| | | |
Collapse
|
29
|
DNA methylation and apoptosis resistance in cancer cells. Cells 2013; 2:545-73. [PMID: 24709797 PMCID: PMC3972670 DOI: 10.3390/cells2030545] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2013] [Revised: 06/27/2013] [Accepted: 06/28/2013] [Indexed: 01/13/2023] Open
Abstract
Apoptosis is a cell death programme primordial to cellular homeostasis efficiency. This normal cell suicide program is the result of the activation of a cascade of events in response to death stimuli. Apoptosis occurs in normal cells to maintain a balance between cell proliferation and cell death. A deregulation of this balance due to modifications in the apoptosic pathway leads to different human diseases including cancers. Apoptosis resistance is one of the most important hallmarks of cancer and some new therapeutical strategies focus on inducing cell death in cancer cells. Nevertheless, cancer cells are resistant to treatment inducing cell death because of different mechanisms, such as DNA mutations in gene coding for pro-apoptotic proteins, increased expression of anti-apoptotic proteins and/or pro-survival signals, or pro-apoptic gene silencing mediated by DNA hypermethylation. In this context, aberrant DNA methylation patterns, hypermethylation and hypomethylation of gene coding for proteins implicated in apoptotic pathways are possible causes of cancer cell resistance. This review highlights the role of DNA methylation of apoptosis-related genes in cancer cell resistance.
Collapse
|
30
|
Benegiamo G, Vinciguerra M, Guarnieri V, Niro GA, Andriulli A, Pazienza V. Hepatitis delta virus induces specific DNA methylation processes in Huh-7 liver cancer cells. FEBS Lett 2013; 587:1424-8. [PMID: 23523924 DOI: 10.1016/j.febslet.2013.03.021] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 03/05/2013] [Accepted: 03/11/2013] [Indexed: 12/11/2022]
Abstract
Hepatitis delta virus (HDV) is a small, defective RNA virus that can infect only individuals carrying hepatitis B virus. HBV/HDV co-infection results in more severe liver disease than HBV single infection and more rapid progression to cirrhosis and hepatocellular carcinoma (HCC). The epigenetic events involved in hepatocyte transformation towards malignancy in this context are poorly known. Here we report that, in Huh-7 cells, HDV induces DNMT3b expression and is associated to E2F1 transcription factor hypermethylation. Moreover our cell cycle analysis showed that HDV induces G2/M arrest. These findings suggest that HDV could play a role in HCC development at least in part by altering DNA methylation events. A better understanding of the molecular mechanisms involved in HDV-related carcinogenesis could help to identify new therapeutic targets.
Collapse
Affiliation(s)
- Giorgia Benegiamo
- Gastroenterology Unit, IRCCS Casa Sollievo della Sofferenza Hospital, San Giovanni Rotondo (FG), Italy
| | | | | | | | | | | |
Collapse
|
31
|
Zhao C, Yin P, Mei C, Li N, Yao W, Li X, Qi J, Fan K, Li Z, Wang L, Shi Y, Qiu S, Fan J, Zha X. Down-regulation of DNA methyltransferase 3B in staurosporine-induced apoptosis and its mechanism in human hepatocarcinoma cell lines. Mol Cell Biochem 2013; 376:111-9. [PMID: 23397112 DOI: 10.1007/s11010-012-1556-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Accepted: 12/19/2012] [Indexed: 11/24/2022]
Abstract
Abnormal DNA methylation is one of the important characteristics in tumor cells. Apoptosis plays an essential role in cell survival and processing. It is not clear whether DNA methyltransferases (DNMTs) change in apoptosis and how DNMTs are regulated in apoptosis. In this study, we found that SMMC-7721 or BEL-7404 cells were induced to apoptosis by STS, meanwhile the DNMT3B protein and mRNA level were decreased. To explore the mechanism of DNMT3B down-regulation, we found that the mRNA decay was not changed and core promoter activity of DNMT3B gene was decreased in STS-induced apoptosis. In order to figure out the signal molecule involved in transcriptional regulation of DNMT3B gene by STS, p-JNK, p-ERK, and p-p38 were examined. In STS-induced apoptosis p-JNK level was increased, and p-ERK and p-p38 were decreased. Furthermore, the inhibitor of p-JNK significantly alleviated the decline of DNMT3B protein. We also found that the siRNA of DNMT3B strengthened the cleavage of PARP and pro-caspase-3 as well as up-regulated the p16 gene expression in STS-treated cells. We concluded here that STS-regulated DNMT3B gene expression via p-JNK and down-regulation of DNMT3B-mediated STS-induced apoptosis through the up-regulation p16 expression.
Collapse
Affiliation(s)
- Chao Zhao
- Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Hernandez JM, Siegel EM, Riggs B, Eschrich S, Elahi A, Qu X, Ajidahun A, Berglund A, Coppola D, Grady WM, Giuliano AR, Shibata D. DNA methylation profiling across the spectrum of HPV-associated anal squamous neoplasia. PLoS One 2012; 7:e50533. [PMID: 23226306 PMCID: PMC3511539 DOI: 10.1371/journal.pone.0050533] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Accepted: 09/27/2012] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Changes in host tumor genome DNA methylation patterns are among the molecular alterations associated with HPV-related carcinogenesis. However, there is little known about the epigenetic changes associated specifically with the development of anal squamous cell cancer (SCC). We sought to characterize broad methylation profiles across the spectrum of anal squamous neoplasia. METHODOLOGY/PRINCIPAL FINDINGS Twenty-nine formalin-fixed paraffin embedded samples from 24 patients were evaluated and included adjacent histologically normal anal mucosa (NM; n = 3), SCC-in situ (SCC-IS; n = 11) and invasive SCC (n = 15). Thirteen women and 11 men with a median age of 44 years (range 26-81) were included in the study. Using the SFP(10) LiPA HPV-typing system, HPV was detected in at least one tissue from all patients with 93% (27/29) being positive for high-risk HPV types and 14 (93%) of 15 invasive SCC tissues testing positive for HPV 16. Bisulfite-modified DNA was interrogated for methylation at 1,505 CpG loci representing 807 genes using the Illumina GoldenGate Methylation Array. When comparing the progression from normal anal mucosa and SCC-IS to invasive SCC, 22 CpG loci representing 20 genes demonstrated significant differential methylation (p<0.01). The majority of differentially methylated gene targets occurred at or close to specific chromosomal locations such as previously described HPV methylation "hotspots" and viral integration sites. CONCLUSIONS We have identified a panel of differentially methlylated CpG loci across the spectrum of HPV-associated squamous neoplasia of the anus. To our knowledge, this is the first reported application of large-scale high throughput methylation analysis for the study of anal neoplasia. Our findings support further investigations into the role of host-genome methylation in HPV-associated anal carcinogenesis with implications towards enhanced diagnosis and screening strategies.
Collapse
Affiliation(s)
- Jonathan M. Hernandez
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - Erin M. Siegel
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - Bridget Riggs
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - Steven Eschrich
- Department of Biomedical Informatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - Abul Elahi
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - Xiaotao Qu
- Department of Biomedical Informatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - Abidemi Ajidahun
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - Anders Berglund
- Department of Biomedical Informatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - Domenico Coppola
- Department of Anatomic Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - William M. Grady
- Division of Gastroenterology, University of Washington, Seattle, Washington, United States of America
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Anna R. Giuliano
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - David Shibata
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| |
Collapse
|
33
|
Liu X, Guo XY, Xu XZ, Wu M, Zhang X, Li Q, Ma PP, Zhang Y, Wang CY, Geng FJ, Qin CH, Liu L, Shi WH, Wang YC, Yu Y. Novel single nucleotide polymorphisms of the bovine methyltransferase 3b gene and their association with meat quality traits in beef cattle. GENETICS AND MOLECULAR RESEARCH 2012; 11:2569-77. [PMID: 22843074 DOI: 10.4238/2012.june.29.1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
DNA methylation is essential for adipose deposition in mammals. We screened SNPs of the bovine DNA methyltransferase 3b (DNMT3b) gene in Snow Dragon beef, a commercial beef cattle population in China. Nine SNPs were found in the population and three of six novel SNPs were chosen for genotyping and analyzing a possible association with 16 meat quality traits. The frequencies of the alleles and genotypes of the three SNPs in Snow Dragon beef were similar to those in their terminal-paternal breed, Wagyu. Association analysis disclosed that SNP1 was not associated with any of the traits; SNP2 was significantly associated with lean meat color score and chuck short rib score, and SNP3 had a significant effect on dressing percentage and back-fat thickness in the beef population. The individuals with genotype GG for SNP2 had a 25.7% increase in lean meat color score and a 146% increase in chuck short rib score, compared with genotype AA. The cattle with genotype AG for SNP3 had 35.7 and 24% increases in dressing percentage and 28.8 and 29.2% increases in back-fat thickness, compared with genotypes GG and AA, respectively. Genotypic combination analysis revealed significant interactions between SNP1 and SNP2 and between SNP2 and SNP3 for the traits rib-eye area and live weight. We conclude that there is considerable evidence that DNMT3b is a determiner of beef quality traits.
Collapse
Affiliation(s)
- X Liu
- Key Laboratory of Agricultural Animal and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, P.R. China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Wu Y, Liu GL, Liu SH, Wang CX, Xu YL, Ying Y, Mao P. MicroRNA-148b enhances the radiosensitivity of non-Hodgkin's Lymphoma cells by promoting radiation-induced apoptosis. JOURNAL OF RADIATION RESEARCH 2012; 53:516-25. [PMID: 22843616 PMCID: PMC3393342 DOI: 10.1093/jrr/rrs002] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2011] [Revised: 02/17/2012] [Accepted: 02/17/2012] [Indexed: 05/20/2023]
Abstract
Growing evidence has demonstrated that microRNAs (miRNAs) play an important role in regulating cellular radiosensitivity. This study aimed to explore the role of miRNAs in non-Hodgkin's lymphoma (NHL) radiosensitivity. Microarray was employed to compare the miRNA expression profiles in B cell lymphoma cell line Raji before and after a 2-Gy dose of radiation. A total of 20 differentially expressed miRNAs were identified including 10 up-regulated and 10 down-regulated (defined as P < 0.05). Among the differentially expressed miRNAs, miR-148b was up-regulated 1.53-fold in response to radiation treatment. A quantitative real-time polymerase chain reaction (PCR) assay confirmed the up-regulation of miR-148b after radiation. Transient transfection experiments showed that miR-148b was up-regulated by miR-148b mimic and down-regulated by miR-148b inhibitor in the Raji cells. A proliferation assay showed that miR-148b could inhibit the proliferation of Raji cells before and after radiation. A clonogenic assay demonstrated that miR-148b sensitized Raji cells to radiotherapy. MiR-148b did not affect the cell cycle profile of post-radiation Raji cells compared with controls. An apoptosis assay showed that miR-148b enhanced apoptosis of Raji cells after irradiation. Taken together, these results demonstrate that miR-148b increased the radiosensitivity of NHL cells probably by promoting radiation-induced apoptosis, which suggests that miR-148b plays an important role in the response of NHL to ionizing radiation.
Collapse
Affiliation(s)
- Yong Wu
- Department of Hematology, Guangzhou First Municipal People's Hospital Affiliated to Guangzhou Medical College, 1 Panfu Road, Guangzhou, People's Republic of China, 510180
- Department of Oncology, Guangzhou First Municipal People's Hospital Affiliated to Guangzhou Medical College, 1 Panfu Road, Guangzhou, People's Republic of China, 510180
| | - Guo-Long Liu
- Department of Oncology, Guangzhou First Municipal People's Hospital Affiliated to Guangzhou Medical College, 1 Panfu Road, Guangzhou, People's Republic of China, 510180
| | - Si-Hong Liu
- Department of Surgery, Guangzhou First Municipal People's Hospital Affiliated to Guangzhou Medical College, 1 Panfu Road, Guangzhou, People's Republic of China, 510180
| | - Cai-Xia Wang
- Department of Hematology, Guangzhou First Municipal People's Hospital Affiliated to Guangzhou Medical College, 1 Panfu Road, Guangzhou, People's Republic of China, 510180
| | - Yan-Li Xu
- Department of Hematology, Guangzhou First Municipal People's Hospital Affiliated to Guangzhou Medical College, 1 Panfu Road, Guangzhou, People's Republic of China, 510180
| | - Yi Ying
- Department of Hematology, Guangzhou First Municipal People's Hospital Affiliated to Guangzhou Medical College, 1 Panfu Road, Guangzhou, People's Republic of China, 510180
| | - Ping Mao
- Department of Hematology, Guangzhou First Municipal People's Hospital Affiliated to Guangzhou Medical College, 1 Panfu Road, Guangzhou, People's Republic of China, 510180
- Corresponding author: Tel: +86(20)81048163; Fax: +86(20)81048163;
| |
Collapse
|
35
|
A New Player in the Development of TRAIL Based Therapies for Hepatocarcinoma Treatment: ATM Kinase. Cancers (Basel) 2012; 4:354-78. [PMID: 24213315 PMCID: PMC3712690 DOI: 10.3390/cancers4020354] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Revised: 03/15/2012] [Accepted: 03/26/2012] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common cancers worldwide. HCCs are genetically and phenotypically heterogeneous tumors characterized by very poor prognosis, mainly due to the lack, at present, of effective therapeutic options, as these tumors are rarely suitable for radiotherapy and often resistant to chemotherapy protocols. In the last years, agonists targeting the Tumor Necrosis Factor Related Apoptosis Inducing Ligand (TRAIL) death receptor, has been investigated as a valuable promise for cancer therapy, based on their selectivity for malignant cells and low toxicity for healthy cells. However, many cancer models display resistance to death receptor induced apoptosis, pointing to the requirement for the development of combined therapeutic approaches aimed to selectively sensitize cancer cells to TRAIL. Recently, we identified ATM kinase as a novel modulator of the ability of chemotherapeutic agents to enhance TRAIL sensitivity. Here, we review the biological determinants of HCC responsiveness to TRAIL and provide an exhaustive and updated analysis of the molecular mechanisms exploited for combined therapy in this context. The role of ATM kinase as potential novel predictive biomarker for combined therapeutic approaches based on TRAIL and chemotherapeutic drugs will be closely discussed.
Collapse
|