1
|
Fan W, Li W, Li L, Qin M, Mao C, Yuan Z, Wang P, Chu B, Jiang Y. Bifunctional HDAC and DNMT inhibitor induces viral mimicry activates the innate immune response in triple-negative breast cancer. Eur J Pharm Sci 2024; 197:106767. [PMID: 38636781 DOI: 10.1016/j.ejps.2024.106767] [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: 01/22/2024] [Revised: 04/12/2024] [Accepted: 04/13/2024] [Indexed: 04/20/2024]
Abstract
Triple-negative breast cancer (TNBC) is a unique breast cancer subtype characterized by a lack of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) expression. Since TNBC lacks ER, PR, and HER2, there are currently no drugs that specifically target TNBC. Therefore, the development of new drugs or effective treatment strategies to target TNBC has become an urgent clinical need. Research has shown that the application of histone deacetylase (HDAC) inhibitors and DNA methyltransferase (DNMT) inhibitors leads to genomic and epigenomic instability. This, in turn, triggers the activation of pattern recognition receptors (PRRs) and subsequently activates downstream interferon (IFN) signalling pathways. In this study, the bifunctional HDAC and DNMT inhibitor J208 exhibited antitumour activity in TNBC cell lines. J208 effectively induced apoptosis and cell cycle arrest at the G0/G1 phase, inhibiting cell migration and invasion in TNBC. Moreover, this bifunctional inhibitor induced the expression of endogenous retroviruses (ERVs) and elicited a viral mimicry response, which increased the intracellular levels of double-stranded RNA (dsRNA) to activate the innate immune signalling pathway in TNBC. In summary, we demonstrated that the bifunctional inhibitor J208, which is designed to inhibit HDAC and DNMT, has potent anticancer effects, providing a new research basis for reactivating antitumour immunity by triggering innate immune signalling and offering a promising strategy for TNBC treatment.
Collapse
Affiliation(s)
- Weiwen Fan
- Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Wenkai Li
- Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Lulu Li
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen 518055, China
| | - Meirong Qin
- Shenzhen Institute for Drug Control, Shenzhen 518057, China
| | - Chengzhou Mao
- Department of Anatomy and Histology, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Zigao Yuan
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen 518055, China
| | - Ping Wang
- Shenzhen Institute for Drug Control, Shenzhen 518057, China.
| | - Bizhu Chu
- Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China.
| | - Yuyang Jiang
- Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China; State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen 518055, China.
| |
Collapse
|
2
|
Lv X, Li R, Li Z, Wang J. Purification of Gekko Small Peptide Fraction and Its Effect of Inducing Apoptosis of EC 9706 Esophageal Cancer Cells by Inhibiting PI3K/Akt/GLUT1 Signaling Pathway. Chem Biodivers 2021; 18:e2000720. [PMID: 33534194 DOI: 10.1002/cbdv.202000720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 02/02/2021] [Indexed: 11/07/2022]
Abstract
This study aimed to isolate and purify a cytotoxic extraction from Gekko japonicus, identify its components and determine its cytotoxic activity in vitro. We isolated and identified the most potent cytotoxic Gekko small peptide LH-20-15. The identification and analysis of peptide sequences of LH-20-15 were performed by de novo peptide sequencing, and two new peptides were found. LH-20-15 significantly inhibited the proliferation of human esophageal squamous carcinoma EC 9706 cells in a dose-dependent manner. Furthermore, LH-20-15 induced apoptosis in esophageal cancer cells by activating the mitochondrial apoptotic pathway. Further research showed that LH-20-15 inhibited the PI3 K/Akt/GLUT1 signaling pathway. In conclusion, LH-20-15 from Gekko japonicus is a peptide mixture and may inhibit EC 9706 cell proliferation and induce apoptosis by activating the mitochondrial apoptotic pathway. It also regulates glucose metabolism by targeting the PI3 K/Akt/GLUT1 signaling pathway. These small peptides could be new sources of natural cytotoxic ingredients against esophageal cancer with potential drug values.
Collapse
Affiliation(s)
- Xingzhi Lv
- Department of Pharmacology, Medical College, Henan University of Science and Technology, KaiYuan Road 263, Luoyang, 471023, Henan Province, P. R. China
| | - Ruifang Li
- Department of Pharmacology, Medical College, Henan University of Science and Technology, KaiYuan Road 263, Luoyang, 471023, Henan Province, P. R. China
| | - Zhongjie Li
- Department of Pharmacology, Medical College, Henan University of Science and Technology, KaiYuan Road 263, Luoyang, 471023, Henan Province, P. R. China
| | - Jiangang Wang
- Department of Pharmacology, Medical College, Henan University of Science and Technology, KaiYuan Road 263, Luoyang, 471023, Henan Province, P. R. China
| |
Collapse
|
3
|
Mo W, Dai Y, Chen J, Liang L, Xu S, Xu X. Long Noncoding RNA (lncRNA) MT1JP Suppresses Hepatocellular Carcinoma (HCC) in vitro. Cancer Manag Res 2020; 12:7949-7960. [PMID: 32943929 PMCID: PMC7478919 DOI: 10.2147/cmar.s253496] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 07/22/2020] [Indexed: 12/19/2022] Open
Abstract
Introduction The purpose of this study was to evaluate the effects and mechanisms of the long noncoding RNA (lncRNA) MT1JP on hepatocellular carcinoma (HCC) in vitro. Patients and Methods Thirty pairs of tumor and adjacent normal tissues were collected from HCC patients. Tissue pathology and MT1JP expression were evaluated by hematoxylin and eosin staining and in situ hybridization (ISH), respectively. The correlation between MT1JP and HCC prognosis was investigated. MTT assays, cloning, flow cytometry, transwell assays, and wound-healing assays were used to evaluate the effects of MT1JP on HCC cell lines. RT-qPCR and Western blot were used to measure the relative mRNA and protein expression levels. Results The expression of MT1JP was downregulated in HCC tumor tissues compared with that in adjacent normal tissues, while the percent survival was significantly greater in the high MT1JP expression group than in the low MT1JP expression group (P=0.0238). In vitro, overexpression of MT1JP suppressed the proliferation, invasion, and migration, reduced colony cell number, increased cell apoptosis, and induced G1-phase cell cycle arrest in Bel-7402 and Huh-7 cells. Meanwhile, the mRNA and protein expression levels of RUNX3 and P21 were significantly upregulated, whereas those of MMP2 and MMP9 were significantly downregulated, in Bel-7402 and Huh-7 cells overexpressing MT1JP (all P<0.001). Conclusion LncRNA MT1JP may function as a tumor suppressor in HCC. Overexpression of MT1JP suppressed HCC cell biological activities through the regulation of RUNX3.
Collapse
Affiliation(s)
- Wenhui Mo
- Department of Gastroenterology, Shidong Hospital of Shanghai, Shanghai 200433, People's Republic of China
| | - Ying Dai
- Department of Gastroenterology, Shidong Hospital of Shanghai, Shanghai 200433, People's Republic of China
| | - Jianqing Chen
- Department of Gastroenterology, Shidong Hospital of Shanghai, Shanghai 200433, People's Republic of China
| | - Liwei Liang
- Department of Gastroenterology, Shidong Hospital of Shanghai, Shanghai 200433, People's Republic of China
| | - Shuqi Xu
- Department of Gastroenterology, Shidong Hospital of Shanghai, Shanghai 200433, People's Republic of China
| | - Xuanfu Xu
- Department of Gastroenterology, Shidong Hospital of Shanghai, Shanghai 200433, People's Republic of China
| |
Collapse
|
4
|
Ouyang Q, Cui Y, Yang S, Wei W, Zhang M, Zeng J, Qu F. lncRNA MT1JP Suppresses Biological Activities of Breast Cancer Cells in vitro and in vivo by Regulating the miRNA-214/RUNX3 Axis. Onco Targets Ther 2020; 13:5033-5046. [PMID: 32581560 PMCID: PMC7280253 DOI: 10.2147/ott.s241503] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Accepted: 04/01/2020] [Indexed: 12/12/2022] Open
Abstract
Introduction The purpose of our research was to evaluate MT1JP in breast cancer. Material and Methods For clinical purpose, tissues were collected, and a correlation analysis ofMT1JP and miRNA-214 gene expressions was conducted. Using an in vitro study, MDA-MB-231 and MCF-7 cell lines were used as research objects in our research. Colony, flow cytometry, TUNEL, transwell, adhesion and wound healing assay were used to discuss the biological activities of the cells. In an in vivo study, tumor weight and volume were measured, and cell apoptosis was measured by TUNEL assay. The relative mechanism's proteins were evaluated by Western blotting or immunohistochemistry assay. Results Compared with adjacent tissues, MT1JP and miRNA-214 gene expressions were significantly different (P<0.001, respectively). By in vitro and in vivo studies, the biological activities of the cells were significantly decreased in MDA-MB-231 and MCF-7 cell lines with MT1JP overexpression. The relative mechanism was correlated with miRNA-214/RUNX3 axis. Conclusion The overexpression of MT1JP suppresses the biological activities of breast cancer cells by regulation miRNA-214/RUNX3 axis in vitro and vivo study.
Collapse
Affiliation(s)
- Qianwen Ouyang
- Department of Breast Surgery, The Third Hospital of Nanchang, Jiangxi, China Jiangxi Province Key Laboratory for Breast Diseases, Nanchang, Jiangxi 330009, People's Republic of China
| | - Yanru Cui
- Department of Physiology, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, People's Republic of China
| | - Shixin Yang
- Department of Breast Surgery, The Third Hospital of Nanchang, Jiangxi, China Jiangxi Province Key Laboratory for Breast Diseases, Nanchang, Jiangxi 330009, People's Republic of China
| | - Wensong Wei
- Department of Breast Surgery, The Third Hospital of Nanchang, Jiangxi, China Jiangxi Province Key Laboratory for Breast Diseases, Nanchang, Jiangxi 330009, People's Republic of China
| | - Mingyue Zhang
- Department of Pharmacology, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, People's Republic of China
| | - Jie Zeng
- Department of Breast Surgery, The Third Hospital of Nanchang, Jiangxi, China Jiangxi Province Key Laboratory for Breast Diseases, Nanchang, Jiangxi 330009, People's Republic of China
| | - Fei Qu
- Department of Pharmacology, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, People's Republic of China
| |
Collapse
|
5
|
Patties I, Kallendrusch S, Böhme L, Kendzia E, Oppermann H, Gaunitz F, Kortmann RD, Glasow A. The Chk1 inhibitor SAR-020106 sensitizes human glioblastoma cells to irradiation, to temozolomide, and to decitabine treatment. J Exp Clin Cancer Res 2019; 38:420. [PMID: 31639020 PMCID: PMC6805470 DOI: 10.1186/s13046-019-1434-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 10/01/2019] [Indexed: 12/30/2022] Open
Abstract
Background Glioblastoma is the most common and aggressive brain tumour in adults with a median overall survival of only 14 months after standard therapy with radiation therapy (IR) and temozolomide (TMZ). In a novel multimodal treatment approach we combined the checkpoint kinase 1 (Chk1) inhibitor SAR-020106 (SAR), disrupting homologue recombination, with standard DNA damage inducers (IR, TMZ) and the epigenetic/cytotoxic drug decitabine (5-aza-2′-deoxycitidine, 5-aza-dC). Different in vitro glioblastoma models are monitored to evaluate if the impaired DNA damage repair may chemo/radiosensitize the tumour cells. Methods Human p53-mutated (p53-mut) and -wildtype (p53-wt) glioblastoma cell lines (p53-mut: LN405, T98G; p53-wt: A172, DBTRG) and primary glioblastoma cells (p53-mut: P0297; p53-wt: P0306) were treated with SAR combined with TMZ, 5-aza-dC, and/or IR and analysed for induction of apoptosis (AnnexinV and sub-G1 assay), cell cycle distribution (nuclear PI staining), DNA damage (alkaline comet or gH2A.X assay), proliferation inhibition (BrdU assay), reproductive survival (clonogenic assay), and potential tumour stem cells (nestinpos/GFAPneg fluorescence staining). Potential treatment-induced neurotoxicity was evaluated on nestin-positive neural progenitor cells in a murine entorhinal-hippocampal slice culture model. Results SAR showed radiosensitizing effects on the induction of apoptosis and on the reduction of long-term survival in p53-mut and p53-wt glioblastoma cell lines and primary cells. In p53-mut cells, this effect was accompanied by an abrogation of the IR-induced G2/M arrest and an enhancement of IR-induced DNA damage by SAR treatment. Also TMZ and 5-aza-dC acted radioadditively albeit to a lesser extent. The multimodal treatment achieved the most effective reduction of clonogenicity in all tested cell lines and did not affect the ratio of nestinpos/GFAPneg cells. No neurotoxic effects were detected when the number of nestin-positive neural progenitor cells remained unchanged after multimodal treatment. Conclusion The Chk1 inhibitor SAR-020106 is a potent sensitizer for DNA damage-induced cell death in glioblastoma therapy strongly reducing clonogenicity of tumour cells. Selectively enhanced p53-mut cell death may provide stronger responses in tumours defective of non-homologous end joining (NHEJ). Our results suggest that a multimodal therapy involving DNA damage inducers and DNA repair inhibitors might be an effective anti-tumour strategy with a low risk of neurotoxicity.
Collapse
|
6
|
Yang J, Tian X, Yang J, Cui J, Jiang S, Shi R, Liu Y, Liu X, Xu W, Xie W, Jia X, Bade R, Zhang T, Zhang M, Gong K, Yan S, Yang Z, Shao G. 5-Aza-2'-deoxycytidine, a DNA methylation inhibitor, induces cytotoxicity, cell cycle dynamics and alters expression of DNA methyltransferase 1 and 3A in mouse hippocampus-derived neuronal HT22 cells. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2017; 80:1222-1229. [PMID: 28880816 DOI: 10.1080/15287394.2017.1367143] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Epigenetic processes such as DNA methylation are essential for processes of gene expression in normal mammalian development. DNA methyltransferases (DNMT) are responsible for initiating and maintaining DNA methylation. It is known that 5-Aza-CdR, an inhibitor of DNMT induces cytotoxicity by reducing DNMT activity in various tumor cell lines. However, disturbances in neuronal DNA methylation may also play a role in altered brain functions. Thus, it was of interest to determine whether alterations in DNA methylation might be associated with neuronal functions by using 5-Aza-CdR, on mouse hippocampus-derived neuronal HT22 cell line. In particular, the aim of this study was to investigate the effects of 5-Aza-CdR on cell growth inhibition, cell cycle arrest, apoptosis as well as the expression levels of DNMT in HT22 cells. HT22 cells were incubated with 5 or 20 μmol/L 5-Aza-CdR for 24 h. Data showed that 5-Aza-CdR at both concentrations significantly inhibited proliferation of HT22 cells and exacerbated cytoplasmic vacuolization. Flow cytometry analysis demonstrated that 5-Aza-CdR treatment at both concentrations decreased early apoptosis but enhanced late apoptosis. Cell cycle analysis illustrated that 5-Aza-CdR treatment induced S phase arrest. Further, incubation with 5-Aza-CdR produced a down-regulation in expression of mRNA and protein DNMT1 and 3A but no marked changes were noted in DNMT 3B and p21 expression. In addition, DNMT1 activity was significantly decreased at both 5-Aza-CdR concentrations. Evidence indicates that 5-Aza-CdR induced cytotoxicity was associated with altered mRNA and protein expression of DNMT 1 and 3A associated with reduced DNMT1 activity in HT22 cells which might affect brain functions.
Collapse
Affiliation(s)
- Jing Yang
- a Department of Neurobiology and Center of Stroke , Beijing Institute for Brain Disorders, Capital Medical University , Beijing , P.R.C
- b Inner Mongolia Key laboratory of Hypoxic Translational Medicine , Baotou Medical College , Inner Mongolia , P.R.C
- c Beijing key laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital , Capital Medical University , Beijing , P.R.C
| | - Xiaoli Tian
- b Inner Mongolia Key laboratory of Hypoxic Translational Medicine , Baotou Medical College , Inner Mongolia , P.R.C
- c Beijing key laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital , Capital Medical University , Beijing , P.R.C
| | - Jie Yang
- b Inner Mongolia Key laboratory of Hypoxic Translational Medicine , Baotou Medical College , Inner Mongolia , P.R.C
- c Beijing key laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital , Capital Medical University , Beijing , P.R.C
| | - Junhe Cui
- b Inner Mongolia Key laboratory of Hypoxic Translational Medicine , Baotou Medical College , Inner Mongolia , P.R.C
- c Beijing key laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital , Capital Medical University , Beijing , P.R.C
| | - Shuyuan Jiang
- b Inner Mongolia Key laboratory of Hypoxic Translational Medicine , Baotou Medical College , Inner Mongolia , P.R.C
- c Beijing key laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital , Capital Medical University , Beijing , P.R.C
| | - Rui Shi
- b Inner Mongolia Key laboratory of Hypoxic Translational Medicine , Baotou Medical College , Inner Mongolia , P.R.C
- c Beijing key laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital , Capital Medical University , Beijing , P.R.C
| | - You Liu
- b Inner Mongolia Key laboratory of Hypoxic Translational Medicine , Baotou Medical College , Inner Mongolia , P.R.C
- c Beijing key laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital , Capital Medical University , Beijing , P.R.C
| | - Xiaolei Liu
- b Inner Mongolia Key laboratory of Hypoxic Translational Medicine , Baotou Medical College , Inner Mongolia , P.R.C
- c Beijing key laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital , Capital Medical University , Beijing , P.R.C
| | - Wenqiang Xu
- b Inner Mongolia Key laboratory of Hypoxic Translational Medicine , Baotou Medical College , Inner Mongolia , P.R.C
- c Beijing key laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital , Capital Medical University , Beijing , P.R.C
| | - Wei Xie
- b Inner Mongolia Key laboratory of Hypoxic Translational Medicine , Baotou Medical College , Inner Mongolia , P.R.C
- c Beijing key laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital , Capital Medical University , Beijing , P.R.C
| | - Xiaoe Jia
- b Inner Mongolia Key laboratory of Hypoxic Translational Medicine , Baotou Medical College , Inner Mongolia , P.R.C
- c Beijing key laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital , Capital Medical University , Beijing , P.R.C
| | - Rengui Bade
- b Inner Mongolia Key laboratory of Hypoxic Translational Medicine , Baotou Medical College , Inner Mongolia , P.R.C
- c Beijing key laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital , Capital Medical University , Beijing , P.R.C
| | - Tao Zhang
- b Inner Mongolia Key laboratory of Hypoxic Translational Medicine , Baotou Medical College , Inner Mongolia , P.R.C
- c Beijing key laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital , Capital Medical University , Beijing , P.R.C
| | - Ming Zhang
- b Inner Mongolia Key laboratory of Hypoxic Translational Medicine , Baotou Medical College , Inner Mongolia , P.R.C
- c Beijing key laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital , Capital Medical University , Beijing , P.R.C
| | - Kerui Gong
- d Department of Oral and Maxillofacial Surgery , University of California San Francsico , San Francisco , USA
| | - Shaochun Yan
- b Inner Mongolia Key laboratory of Hypoxic Translational Medicine , Baotou Medical College , Inner Mongolia , P.R.C
- c Beijing key laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital , Capital Medical University , Beijing , P.R.C
| | - Zhanjun Yang
- b Inner Mongolia Key laboratory of Hypoxic Translational Medicine , Baotou Medical College , Inner Mongolia , P.R.C
- c Beijing key laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital , Capital Medical University , Beijing , P.R.C
| | - Guo Shao
- a Department of Neurobiology and Center of Stroke , Beijing Institute for Brain Disorders, Capital Medical University , Beijing , P.R.C
- b Inner Mongolia Key laboratory of Hypoxic Translational Medicine , Baotou Medical College , Inner Mongolia , P.R.C
- c Beijing key laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital , Capital Medical University , Beijing , P.R.C
| |
Collapse
|
7
|
Naz A, Cui Y, Collins CJ, Thompson DH, Irudayaraj J. PLGA-PEG nano-delivery system for epigenetic therapy. Biomed Pharmacother 2017; 90:586-597. [PMID: 28407579 DOI: 10.1016/j.biopha.2017.03.093] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 03/22/2017] [Accepted: 03/27/2017] [Indexed: 12/31/2022] Open
Abstract
Efficient delivery of cytidine analogues such as Azacitidine (AZA) into solid tumors constitutes a primary challenge in epigenetic therapies. We developed a di-block nano-vector based on poly(lactic-co-glycolic acid) (PLGA) and poly(ethylene glycol) (PEG) for stabilization of the conjugated AZA under physiological conditions. With equimolar drug content, our nano-conjugate could elicit a better anti-proliferative effect over free drug in breast cancer both in vitro and in vivo, through reactivation of p21 and BRCA1 to restrict cell proliferation. In addition, we applied single-molecule fluorescence tools to characterize the intracellular behavior of the AZA-PLGE-PEG nano-micelles at a finer spatiotemporal resolution. Our results suggest that the nano-micelles could effectively enrich in cancer cells and may not be limited by nucleoside transporters. Afterwards, the internalized nano-micelles exhibit pH-dependent release and resistance to active efflux. Altogether, our work describes a delivery strategy for DNA demethylating agents with nanoscale tunability, providing a cost-effective option for pharmaceutics.
Collapse
Affiliation(s)
- Asia Naz
- Bindley Bioscience Center and Purdue Center for Cancer Research, Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN 47907, USA; Department of Pharmaceutical Chemistry, University of Karachi, Karachi 75270, Pakistan
| | - Yi Cui
- Bindley Bioscience Center and Purdue Center for Cancer Research, Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN 47907, USA.
| | | | - David H Thompson
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Joseph Irudayaraj
- Bindley Bioscience Center and Purdue Center for Cancer Research, Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
8
|
Andrade AF, Borges KS, Suazo VK, Geron L, Corrêa CAP, Castro-Gamero AM, de Vasconcelos EJR, de Oliveira RS, Neder L, Yunes JA, Dos Santos Aguiar S, Scrideli CA, Tone LG. The DNA methyltransferase inhibitor zebularine exerts antitumor effects and reveals BATF2 as a poor prognostic marker for childhood medulloblastoma. Invest New Drugs 2016; 35:26-36. [PMID: 27785591 DOI: 10.1007/s10637-016-0401-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 10/20/2016] [Indexed: 12/11/2022]
Abstract
Medulloblastoma (MB) is the most common solid tumor among pediatric patients and corresponds to 20 % of all pediatric intracranial tumors in this age group. Its treatment currently involves significant side effects. Epigenetic changes such as DNA methylation may contribute to its development and progression. DNA methyltransferase (DNMT) inhibitors have shown promising anticancer effects. The agent Zebularine acts as an inhibitor of DNA methylation and shows low toxicity and high efficacy, being a promising adjuvant agent for anti-cancer chemotherapy. Several studies have reported its effects on different types of tumors; however, there are no studies reporting its effects on MB. We analyzed its potential anticancer effects in four pediatric MB cell lines. The treatment inhibited proliferation and clonogenicity, increased the apoptosis rate and the number of cells in the S phase (p < 0.05), as well as the expression of p53, p21, and Bax, and decreased cyclin A, Survivin and Bcl-2 proteins. In addition, the combination of zebularine with the chemotherapeutic agents vincristine and cisplatin resulted in synergism and antagonism, respectively. Zebularine also modulated the activation of the SHH pathway, reducing SMO and GLI1 levels and one of its targets, PTCH1, without changing SUFU levels. A microarray analysis revealed different pathways modulated by the drug, including the Toll-Like Receptor pathway and high levels of the BATF2 gene. The low expression of this gene was associated with a worse prognosis in MB. Taken together, these data suggest that Zebularine may be a potential drug for further in vivo studies of MB treatment.
Collapse
Affiliation(s)
- Augusto Faria Andrade
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Avenida Bandeirantes 3900, 14048-900, Ribeirão Preto, SP, Brazil.
| | - Kleiton Silva Borges
- Department of Pediatrics - Ribeirão Preto Medical School, University of São Paulo (USP), São Paulo, Brazil
| | - Veridiana Kiill Suazo
- Department of Pediatrics - Ribeirão Preto Medical School, University of São Paulo (USP), São Paulo, Brazil
| | - Lenisa Geron
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Avenida Bandeirantes 3900, 14048-900, Ribeirão Preto, SP, Brazil
| | | | | | | | | | - Luciano Neder
- Department of Pathology, Ribeirão Preto Medical School, USP, São Paulo, Brazil
| | | | | | - Carlos Alberto Scrideli
- Department of Pediatrics - Ribeirão Preto Medical School, University of São Paulo (USP), São Paulo, Brazil
| | - Luiz Gonzaga Tone
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Avenida Bandeirantes 3900, 14048-900, Ribeirão Preto, SP, Brazil.,Department of Pediatrics - Ribeirão Preto Medical School, University of São Paulo (USP), São Paulo, Brazil
| |
Collapse
|
9
|
Cheng YW, Lin FCF, Chen CY, Hsu NY. Environmental exposure and HPV infection may act synergistically to induce lung tumorigenesis in nonsmokers. Oncotarget 2016; 7:19850-62. [PMID: 26918347 PMCID: PMC4991423 DOI: 10.18632/oncotarget.7628] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 02/16/2016] [Indexed: 11/25/2022] Open
Abstract
Most studies of lung tumorigenesis have focused on smokers rather than nonsmokers. In this study, we used human papillomavirus (HPV)-positive and HPV-negative lung cancer cells to test the hypothesis that HPV infection synergistically increases DNA damage induced by exposure to the carcinogen benzo[a]pyrene (B[a]P), and contributes to lung tumorigenesis in nonsmokers. DNA adduct levels induced by B[a]P in HPV-positive cells were significantly higher than in HPV-negative cells. The DNA adduct formation was dependent on HPV E6 oncoprotein expression. Gene and protein expression of two DNA repair genes, XRCC3 and XRCC5, were lower in B[a]P-treated E6-positive cells than in E6-negative lung cancer cells. The reduced expression was also detected immunohistochemically and was caused by increased promoter hypermethylation. Moreover, mutations of p53 and epidermal growth factor receptor (EGFR) genes in lung cancer patients were associated with XRCC5 inactivation. In sum, our study indicates that HPV E6-induced promoter hypermethylation of the XRCC3 and XRCC5 DNA repair genes and the resultant decrease in their expression increases B[a]P-induced DNA adducts and contributes to lung tumorigenesis in nonsmokers.
Collapse
Affiliation(s)
- Ya-Wen Cheng
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Cancer Center, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - Frank Cheau-Feng Lin
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Surgery, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chih-Yi Chen
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Surgery, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Nan-Yung Hsu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Cancer Center, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Surgery, Taipei Medical University Hospital, Taipei, Taiwan
| |
Collapse
|
10
|
Cui Y, Naz A, Thompson DH, Irudayaraj J. Decitabine nanoconjugate sensitizes human glioblastoma cells to temozolomide. Mol Pharm 2015; 12:1279-88. [PMID: 25751281 DOI: 10.1021/mp500815b] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
In this study, we developed and characterized a delivery system for the epigenetic demethylating drug, decitabine, to sensitize temozolomide-resistant human glioblastoma multiforme (GBM) cells to alkylating chemotherapy. A poly(lactic-co-glycolic acid) (PLGA) and poly(ethylene glycol) (PEG) based nanoconjugate was fabricated to encapsulate decitabine and achieved a better therapeutic response in GBM cells than that with the free drug. After synthesis, the highly efficient uptake process and intracellular dynamics of this nanoconjugate were monitored by single-molecule fluorescence tools. Our experiments demonstrated that, under an acidic pH due to active glycolysis in cancer cells, the PLGA-PEG nanovector could release the conjugated decitabine at a faster rate, after which the hydrolyzed lactic acid and glycolic acid would further acidify the intracellular microenvironment, thus providing positive feedback to increase the effective drug concentration and realize growth inhibition. In temozolomide-resistant GBM cells, decitabine can potentiate the cytotoxic DNA alkylation by counteracting cytosine methylation and reactivating tumor suppressor genes, such as p53 and p21. Owing to the excellent internalization and endolysosomal escape enabled by the PLGA-PEG backbone, the encapsulated decitabine exhibited a better anti-GBM potential than that of free drug molecules. Hence, the synthesized nanoconjugate and temozolomide could act in synergy to deliver a more potent and long-term antiproliferative effect against malignant GBM cells.
Collapse
Affiliation(s)
| | - Asia Naz
- ‡Department of Pharmaceutical Chemistry, University of Karachi, Karachi 75270, Pakistan
| | | | | |
Collapse
|
11
|
Wang S, Liu H, Akhtar J, Chen HX, Wang Z. Alteration of runt-related transcription factor 3 gene expression and biologic behavior of esophageal carcinoma TE-1 cells after 5-azacytidine intervention. Asian Pac J Cancer Prev 2014; 14:5427-33. [PMID: 24175838 DOI: 10.7314/apjcp.2013.14.9.5427] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
5-Azacytidine (5-azaC) was originally identified as an anticancer drug (NSC102876) which can cause hypomethylation of tumor suppressor genes. To assess its effects on runt-related transcription factor 3 (RUNX3), expression levels and the promoter methylation status of the RUNX3 gene were assessed. We also investigated alteration of biologic behavior of esophageal carcinoma TE-1 cells. MTT assays showed 5-azaC inhibited the proliferation of TE-1 cells in a time and dose-dependent way. Although other genes could be demethylated after 5-azaC intervention, we focused on RUNX3 gene in this study. The expression level of RUNX3 mRNA increased significantly in TE-1 cells after treatment with 5-azaC at hypotoxic levels. RT-PCR showed 5-azaC at 50 μM had the highest RUNX3-induction activity. Methylation-specific PCR indicated that 5-azaC induced RUNX3 expression through demethylation. Migration and invasion of TE-1 cells were inhibited by 5-azaC, along with growth of Eca109 xenografts in nude mice. In conclusion, we demonstrate that the RUNX3 gene can be reactivated by the demethylation reagent 5-azaC, which inhibits the proliferation, migration and invasion of esophageal carcinoma TE-1 cells.
Collapse
Affiliation(s)
- Shuai Wang
- Department of Thoracic Surgery, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China E-mail : wz620226@ hotmail.com
| | | | | | | | | |
Collapse
|
12
|
PTEN methylation involved in benzene-induced hematotoxicity. Exp Mol Pathol 2014; 96:300-6. [DOI: 10.1016/j.yexmp.2014.03.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Accepted: 03/13/2014] [Indexed: 12/19/2022]
|
13
|
Wang S, Liu H, Wang Z, Chen HX. Effects of 5-azacytidine on RUNX3 gene expression and the biological behavior of esophageal carcinoma cells. Mol Med Rep 2014; 9:1259-65. [PMID: 24535051 DOI: 10.3892/mmr.2014.1945] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Accepted: 01/28/2014] [Indexed: 11/05/2022] Open
Abstract
The present study investigated the effects of 5-azacytidine (5-azaC) on the expression level of the human runt-related transcription factor 3 (RUNX3) gene and the biological behavior of esophageal carcinoma Eca109 cells. The effect of the demethylation reagent 5-azaC on the viability of Eca109 cells was detected by the MTT assay, which demonstrated that 5-azaC inhibited the viability of Eca109 cells in a time- and dose-dependent manner. Although demethylation of other genes may occur following treatment with 5-azaC, we focused on the RUNX3 gene. When treated with 5-azaC at hypoxic levels, the expression of RUNX3 increased and the methylation degree of the RUNX3 gene was decreased significantly in Eca109 cells. 5-azaC at 50 µM demonstrated the highest RUNX3-induction activity, inducing RUNX3 mRNA and protein expression, and decreasing the degree of methylation of the RUNX3 gene. Methylation specific PCR indicated that 5-azaC induced RUNX3 expression through demethylation. The abilities of migration and invasion of Eca109 cells were inhibited by 5-azaC. The growth of Eca109 cells treated with 5-azaC in vivo was detected by a tumorigenesis experiment. 5-azaC inhibited the growth of Eca109 xenografts in nude mice. Taken together, our findings demonstrated that the RUNX3 gene is hypermethylated in Eca109 cells and that 5-azaC induces the expression of the RUNX3 gene by demethylation, which inhibits the proliferation, migration and invasion of Eca109 cells.
Collapse
Affiliation(s)
- Shuai Wang
- Department of Thoracic Surgery, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Hong Liu
- Department of Otorhinolaryngology, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Zhou Wang
- Department of Thoracic Surgery, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Hua-Xia Chen
- Department of Thoracic Surgery, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
14
|
You BR, Park WH. Zebularine inhibits the growth of A549 lung cancer cells via cell cycle arrest and apoptosis. Mol Carcinog 2013; 53:847-57. [PMID: 23661569 DOI: 10.1002/mc.22042] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Accepted: 04/04/2013] [Indexed: 01/03/2023]
Abstract
Zebularine (Zeb) is a DNA methyltransferase (DNMT) inhibitor to that has an anti-tumor effect. Here, we evaluated the anti-growth effect of Zeb on A549 lung cancer cells in relation to reactive oxygen species (ROS) levels. Zeb inhibited the growth of A549 cells with an IC50 of approximately 70 µM at 72 h. Cell cycle analysis indicated that Zeb induced an S phase arrest in A549 cells. Zeb also induced A549 cell death, which was accompanied by the loss of mitochondrial membrane potential (MMP; ΔΨm ), Bcl-2 decrease, Bax increase, p53 increase and activation of caspase-3 and -8. In contrast, Zeb mildly inhibited the growth of human pulmonary fibroblast (HPF) normal cells and lead to a G1 phase arrest. Zeb did not induce apoptosis in HPF cells. In relation to ROS level, Zeb increased ROS level in A549 cells and induced glutathione (GSH) depletion. The well-known antioxidant, N-acetyl cysteine (NAC) prevented the death of Zeb-treated A549 cells. Moreover, Zeb increased the level of thioredoxin reductase 1 (TrxR1) in A549 cells. While the overexpression of TrxR1 attenuated death and ROS level in Zeb-treated A549 cells, the downregulation of TrxR1 intensified death and ROS level in these cells. In conclusion, Zeb inhibited the growth of A549 lung cancer cells via cell cycle arrest and apoptosis. The inhibition was influenced by ROS and TrxR1 levels.
Collapse
Affiliation(s)
- Bo Ra You
- Department of Physiology, Medical School, Research Institute for Endocrine Sciences, Chonbuk National University, Jeonju, Republic of Korea
| | | |
Collapse
|
15
|
Ping SY, Shen KH, Yu DS. Epigenetic regulation of vascular endothelial growth factor a dynamic expression in transitional cell carcinoma. Mol Carcinog 2012; 52:568-79. [PMID: 22392726 DOI: 10.1002/mc.21892] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2011] [Revised: 01/30/2012] [Accepted: 02/06/2012] [Indexed: 11/11/2022]
Abstract
Vascular endothelial growth factor A (VEGF-A) is a key mediator in the neovascularization of cancers. We have found that VEGF-A was expressed at significantly higher levels in high-grade transitional cell carcinoma (TCC) cells than low-grade TCC cells in our previous study. In the present study, promoter methylation pattern was assessed and quantified by bisulfite genomic sequencing (BGS) and specific VEGF-A CpG sites in low-grade, but not in high-grade, TCC cells were observed. Reporter assays indicated that hypermethylation of nine CpG sites can inhibit the transcriptional activity of the VEGF-A gene. Subsequent chromatin immunoprecipitation (ChIP) assay revealed down-regulation of transcription activity of VEGF-A with increasing binding of methyl-CpG-binding protein 2 (MBD2) and trimethyl-histone H3 (Lys9) proteins to these CpG sites in low-grade TCC cells during hypermethylation. Furthermore, treatment of low-grade TCC cells with DNA methyltransferase inhibitor and histone deacetylase inhibitor can restore the expression of VEGF-A and promote the invasive ability of low-grade TCC cells. Hypermethylation with lower expression levels of VEGF-A in low-grade TCC tumors than high-grade TCC tumors was also confirmed in clinical specimens by reverse transcriptase-PCR and pyrosequencing analyses. Our findings are the first results indicating that VEGF-A expression is suppressed in low-grade TCC tumors by promoter hypermethylation. This offers a new perspective on the role of VEGF-A in TCC tumor behavior.
Collapse
Affiliation(s)
- Szu-Yuan Ping
- Graduate Institute of Life Science, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | | | | |
Collapse
|
16
|
Liu QY, Chen DW, Xie LP, Zhang RQ, Wang HZ. Decitabine, independent of apoptosis, exerts its cytotoxic effects on cell growth in melanoma cells. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2011; 32:423-429. [PMID: 22004962 DOI: 10.1016/j.etap.2011.08.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2011] [Accepted: 08/14/2011] [Indexed: 05/31/2023]
Abstract
Decitabine is a synthesized cytosine analog that is a potent inhibitor of DNA methylation. There have been a few reports on the in vitro anti-melanoma effect of decitabine or its functional mechanisms. We investigated the anti-proliferation effect of decitabine on the cultured murine melanoma cell line K1735M2. MTT assay showed that decitabine had strong inhibition on melanoma K1735M2 in a time- and dose-dependent manner in vitro. Morphological observation showed that decitabine could induce melanoma K1735M2 cells to produce dendrite-like structures with the increase of decitabine concentration and incubation time. Decitabine could effectively induce K1735M2 cells to differentiate in vitro. Additionally, decitabine could induce a dose-dependent G2/M cell cycle arrest in K1735M2 cells. We provided experimental evidences that the anti-proliferation effect of decitabine on murine K1735M2 melanoma cells was associated predominately with G2/M cell cycle arrest and the induction of differentiation rather than apopotosis.
Collapse
Affiliation(s)
- Qian-Ying Liu
- School of Life Science, Tsinghua University, Beijing 100084, China
| | | | | | | | | |
Collapse
|
17
|
Ruiz-Magaña MJ, Rodríguez-Vargas JM, Morales JC, Saldivia MA, Schulze-Osthoff K, Ruiz-Ruiz C. The DNA methyltransferase inhibitors zebularine and decitabine induce mitochondria-mediated apoptosis and DNA damage in p53 mutant leukemic T cells. Int J Cancer 2011; 130:1195-207. [PMID: 21455989 DOI: 10.1002/ijc.26107] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2010] [Accepted: 03/15/2011] [Indexed: 12/23/2022]
Abstract
DNA methyltransferase (DNMT)-inhibiting nucleoside analogs reactivate the expression of tumor suppressor genes and apoptosis-related genes silenced by methylation, thus favoring the induction of apoptosis in tumor cells. Moreover, induction of DNA damage seems to contribute to their antitumor effect. However, the apoptotic signaling pathway induced by these demethylating drugs is not well understood. Here, we have investigated the induction of apoptosis by two nucleoside DNMT inhibitors, decitabine and zebularine, in leukemic T cells. Both inhibitors induced caspase-dependent apoptosis in Jurkat, CEM-6 and MOLT-4 leukemia T cell lines, all with mutant p53, whereas resting and activated normal T lymphocytes were highly resistant to these demethylating agents. Although decitabine and zebularine showed different ability to induce apoptosis and cell cycle arrest among the three cell lines, they similarly activated the intrinsic apoptotic pathway by inducing mitochondrial alterations such as Bak activation, loss of transmembrane potential and generation of reactive oxygen species (ROS). Accordingly, Bcl-2- and Bcl-x(L) -overexpressing Jurkat cells, as well as caspase-9-deficient Jurkat cells, were resistant to apoptosis induced by decitabine and zebularine. Interestingly, ROS production seemed to be necessary for the induction of apoptosis. Apoptotic events, such as Bak and caspase activation, started as soon as 20 hr after treatment with either decitabine or zebularine. In addition, progression of apoptosis triggered by both DNMT inhibitors was paralleled by the induction of DNA damage. Our results suggest that the mitochondrial apoptotic pathway activated by decitabine and zebularine in p53 mutant leukemic T cells depends mainly on the induction of DNA damage.
Collapse
|
18
|
Irradiation, cisplatin, and 5-azacytidine upregulate cytomegalovirus promoter in tumors and muscles: implementation of non-invasive fluorescence imaging. Mol Imaging Biol 2011; 13:43-52. [PMID: 20396957 PMCID: PMC3023030 DOI: 10.1007/s11307-010-0300-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Purpose The cytomegalovirus (CMV) promoter is one of the most commonly used promoters for expression of transgenes in mammalian cells. The aim of our study was to evaluate the role of methylation and upregulation of the CMV promoter by irradiation and the chemotherapeutic agent cisplatin in vivo using non-invasive fluorescence in vivo imaging. Procedures Murine fibrosarcoma LPB and mammary carcinoma TS/A cells were stably transfected with plasmids encoding CMV and p21 promoter-driven green fluorescent protein (GFP) gene. Solid TS/A tumors were induced by subcutaneous injection of fluorescent tumor cells, while leg muscles were transiently transfected with plasmid encoding GFP under the control of the CMV promoter. Cells, tumors, and legs were treated either by DNA methylation inhibitor 5-azacytidine, irradiation, or cisplatin. GFP expression was determined using a fluorescence microplate reader in vitro and by non-invasive fluorescence imaging in vivo. Results Treatment of cells, tumors, and legs with 5-azacytidine (re)activated the CMV promoter. Furthermore, treatment with irradiation or cisplatin resulted in significant upregulation of GFP expression both in vitro and in vivo. Conclusions Observed alterations in the activity of the CMV promoter limit the usefulness of this widely used promoter as a constitutive promoter. On the other hand, inducibility of CMV promoters can be beneficially used in gene therapy when combined with standard cancer treatment, such as radiotherapy and chemotherapy.
Collapse
|
19
|
Epigenetic modification involved in benzene-induced apoptosis through regulating apoptosis-related genes expression. Cell Biol Int 2011; 35:391-6. [DOI: 10.1042/cbi20100256] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
20
|
Epigenetic mediated transcriptional activation of PARP-1 participates in silica-associated malignant transformation of human bronchial epithelial cells. Toxicol Lett 2010; 193:236-41. [DOI: 10.1016/j.toxlet.2010.01.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2009] [Revised: 12/22/2009] [Accepted: 01/21/2010] [Indexed: 01/13/2023]
|
21
|
Vinken M, Snykers S, Fraczek J, Decrock E, Leybaert L, Rogiers V, Vanhaecke T. DNA methyltransferase 3a expression decreases during apoptosis in primary cultures of hepatocytes. Toxicol In Vitro 2009; 24:445-51. [PMID: 19833194 DOI: 10.1016/j.tiv.2009.10.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2009] [Revised: 10/01/2009] [Accepted: 10/07/2009] [Indexed: 02/08/2023]
Abstract
Although it has been acknowledged that DNA methylation is a key factor in the epigenetic control of liver homeostasis, its role in the occurrence of hepatocyte cell death has yet been poorly documented. We therefore have investigated the expression pattern of the effectors of DNA methylation, namely DNA methyltransferase (DNMT) isoenzymes, during Fas-mediated apoptotic cell death in primary hepatocyte cultures. Cell death was assessed by in situ stainings with Annexin V, Hoechst 33342 and Propidium iodide, and measurement of caspase 3-like activity and lactate dehydrogenase release. Similar to the hepatic in vivo situation, DNMT1, DNMT2 and DNMT3b could not be detected, whereas relatively high levels of DNMT3a protein were observed in the in vitro setting, as studied by immunoblotting. Upon induction of cell death, a progressive decrease in DNMT3a protein amount was noticed, reaching a minimum level towards the final stages of the cell death process. This was preceded by parallel changes in DNMT3a mRNA production, measured by qRT-PCR analysis, which became already evident during the early stages of apoptosis. We conclude that downregulated DNMT3a protein production during Fas-mediated hepatocyte apoptosis results from inhibition of DNMT3a gene transcription. This finding further substantiates the existence of an epigenetic signature of apoptosis.
Collapse
Affiliation(s)
- Mathieu Vinken
- Department of Toxicology, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Laarbeeklaan 103, B-1090 Brussels, Belgium.
| | | | | | | | | | | | | |
Collapse
|
22
|
Xiong H, Qiu H, Zhuang L, Xiong H, Jiang R, Chen Y. Effects of 5-Aza-CdR on the proliferation of human breast cancer cell line MCF-7 and on the expression of Apaf-1 gene. ACTA ACUST UNITED AC 2009; 29:498-502. [PMID: 19662370 DOI: 10.1007/s11596-009-0421-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2008] [Indexed: 12/21/2022]
Abstract
Hypermethylation in the promoter region of tumor suppressor genes is a common mechanism of gene silencing, which tends to occur in cancer. The effects of 5-Aza-2'-deoxycytidine (5-Aza-CdR), a specific DNA methyltransferase inhibitor, on the cell proliferation of human breast cancer cell line MCF-7 and on the expression of Apaf-1 gene were investigated. Human MCF-7 cells were incubated with increasing concentrations of 5-Aza-CdR for 12 to 120 h. The growth inhibition rates of MCF-7 cells were detected by MTT assay. Changes of cell cycle distribution and apoptotic rates of MCF-7 cells were determined by flow cytometry. The expressions of DNA methyltransferase 3b mRNA and Apaf-1 mRNA were measured by reverse transcription polymerase chain reaction (RT-PCR). Meanwhile, the expression of Apaf-1 protein was detected by Western blotting. The results showed that 5-Aza-CdR significantly inhibited the growth of MCF-7 cells and the growth inhibition rate of MCF-7 cells was significantly enhanced with the concentration of 5-Aza-CdR and the action time. Flow cytometry indicated that 5-Aza-CdR could significantly induce G(1)/S cell cycle arrest and increase the apoptosis rate of MCF-7 cells. The mRNA and protein expressions of Apaf-1 were up-regulated in MCF-7 cells treated with 5-Aza-CdR, which was accompanied by down-regulation of DNA methyltransferase 3b mRNA. It is concluded that 5-Aza-CdR might retard the growth of tumor cells and promote the apoptosis of MCF-7 breast cancer cells by inhibiting the expression of DNA methyltransferase 3b and re-activating the Apaf-1 gene expression.
Collapse
Affiliation(s)
- Huihua Xiong
- Cancer Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | | | | | | | | | | |
Collapse
|