1
|
EBV infection is associated with histone bivalent switch modifications in squamous epithelial cells. Proc Natl Acad Sci U S A 2019; 116:14144-14153. [PMID: 31235597 DOI: 10.1073/pnas.1821752116] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Epstein-Barr virus (EBV) induces histone modifications to regulate signaling pathways involved in EBV-driven tumorigenesis. To date, the regulatory mechanisms involved are poorly understood. In this study, we show that EBV infection of epithelial cells is associated with aberrant histone modification; specifically, aberrant histone bivalent switches by reducing the transcriptional activation histone mark (H3K4me3) and enhancing the suppressive mark (H3K27me3) at the promoter regions of a panel of DNA damage repair members in immortalized nasopharyngeal epithelial (NPE) cells. Sixteen DNA damage repair family members in base excision repair (BER), homologous recombination, nonhomologous end-joining, and mismatch repair (MMR) pathways showed aberrant histone bivalent switches. Among this panel of DNA repair members, MLH1, involved in MMR, was significantly down-regulated in EBV-infected NPE cells through aberrant histone bivalent switches in a promoter hypermethylation-independent manner. Functionally, expression of MLH1 correlated closely with cisplatin sensitivity both in vitro and in vivo. Moreover, seven BER members with aberrant histone bivalent switches in the EBV-positive NPE cell lines were significantly enriched in pathway analysis in a promoter hypermethylation-independent manner. This observation is further validated by their down-regulation in EBV-infected NPE cells. The in vitro comet and apurinic/apyrimidinic site assays further confirmed that EBV-infected NPE cells showed reduced DNA damage repair responsiveness. These findings suggest the importance of EBV-associated aberrant histone bivalent switch in host cells in subsequent suppression of DNA damage repair genes in a methylation-independent manner.
Collapse
|
2
|
You Y, Wang Q, Li H, Ma Y, Deng Y, Ye Z, Bai F. Zoledronic acid exhibits radio-sensitizing activity in human pancreatic cancer cells via inactivation of STAT3/NF-κB signaling. Onco Targets Ther 2019; 12:4323-4330. [PMID: 31239706 PMCID: PMC6556542 DOI: 10.2147/ott.s202516] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 04/09/2019] [Indexed: 01/28/2023] Open
Abstract
Background: Although pancreatic cancer is typically radio-sensitive, local treatment failure and metastasis are commonly caused by the development of resistance to radiotherapy. In the current study, the radio-sensitizing actions of zoledronic acid (ZOL) on pancreatic cancer cells were investigated. Materials and methods: Three human pancreatic cancer cell lines were exposed to ZOL, ionizing radiation (IR), or a combination of both, and the effects of the respective drug regimens on cell proliferation and invasion were examined. Results: Combined treatment with low doses of ZOL plus IR efficiently increased cell death and attenuated cell invasion compared with the individual use of ZOL or IR. These effects of ZOL were associated with inactivation of signal transducer and activator of transcription 3 (STAT3) and nuclear factor-κB (NF-κB). Conclusion: Collectively, these data suggest that ZOL in combination with IR is a promising therapeutic strategy for enhancing radio-sensitivity in pancreatic cancer cells via downregulation of the STAT3/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Yanjie You
- Department of Gastroenterology, Ningxia Hui Autonomous Region People's Hospital, Yinchuan, Ningxia Hui Autonomous Region 750021, People's Republic of China
| | - Qiang Wang
- Department of Science and Education, Ningxia Hui Autonomous Region People's Hospital, Yinchuan 750021, People's Republic of China
| | - Haijun Li
- Department of Radiation Oncology, The Second People's Hospital of Neijiang, Neijiang, Sichuan 641003, People's Republic of China
| | - Yuhong Ma
- Department of Gastroenterology, Ningxia Hui Autonomous Region People's Hospital, Yinchuan, Ningxia Hui Autonomous Region 750021, People's Republic of China
| | - Yanhong Deng
- Department of Gastroenterology, Ningxia Hui Autonomous Region People's Hospital, Yinchuan, Ningxia Hui Autonomous Region 750021, People's Republic of China
| | - Zhengcai Ye
- Endoscopy Center, Ningxia Hui Autonomous Region People's Hospital, Yinchuan 750021, People's Republic of China
| | - Feihu Bai
- Department of Gastroenterology, Ningxia Hui Autonomous Region People's Hospital, Yinchuan, Ningxia Hui Autonomous Region 750021, People's Republic of China
| |
Collapse
|
3
|
Wang L, Gao H, Yang X, Liang X, Tan Q, Chen Z, Zhao C, Gu Z, Yu M, Zheng Y, Huang Y, Zhu L, Jacob TJC, Wang L, Chen L. The apoptotic effect of Zoledronic acid on the nasopharyngeal carcinoma cells via ROS mediated chloride channel activation. Clin Exp Pharmacol Physiol 2018; 45:1019-1027. [PMID: 29884989 DOI: 10.1111/1440-1681.12979] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 05/12/2018] [Accepted: 05/29/2018] [Indexed: 01/09/2023]
Abstract
Zoledronic acid (ZA), a third-generation bisphosphonate, has been applied for treatment of bone metastases caused by malignant tumors. Recent studies have found its anti-cancer effects on various tumor cells. One of the mechanisms of anti-cancer effects of ZA is induction of apoptosis. However, the mechanisms of ZA-induced apoptosis in tumor cells have not been clarified clearly. In this study, we investigated the roles of chloride channels in ZA-induced apoptosis in nasopharyngeal carcinoma CNE-2Z cells. Apoptosis and chloride current were induced by ZA and suppressed by chloride channel blockers. After the knockdown of ClC-3 expression by ClC-3 siRNA, ZA-induced chloride current and apoptosis were significantly suppressed, indicating that the chloride channel participated in ZA-induced apoptosis may be ClC-3. When reactive oxygen species (ROS) generation was inhibited by the antioxidant N-acetyl-L-cysteine (L-NAC), ZA-induced apoptosis and chloride current were blocked accordingly, suggesting that ZA induces apoptosis through promoting ROS production and subsequently activating chloride channel.
Collapse
Affiliation(s)
- Liang Wang
- Division of Oncology, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Hong Gao
- Department of Pharmacology, Medical College, Jinan University, Guangzhou, China
| | - Xiaoya Yang
- Department of Physiology, Medical College, Jinan University, Guangzhou, China.,Department of Physiology, Guangzhou Health Science College, Guangzhou, China
| | - Xiechou Liang
- Department of Physiology, Medical College, Jinan University, Guangzhou, China
| | - Qiuchan Tan
- Department of Physiology, Medical College, Jinan University, Guangzhou, China
| | - Zhanru Chen
- Department of Physiology, Medical College, Jinan University, Guangzhou, China
| | - Chan Zhao
- Department of Physiology, Medical College, Jinan University, Guangzhou, China
| | - Zhuoyu Gu
- Department of Pharmacology, Medical College, Jinan University, Guangzhou, China
| | - Meisheng Yu
- Department of Pharmacology, Medical College, Jinan University, Guangzhou, China
| | - Yanfang Zheng
- Department of Pharmacology, Medical College, Jinan University, Guangzhou, China
| | - Yanqing Huang
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Linyan Zhu
- Department of Pharmacology, Medical College, Jinan University, Guangzhou, China
| | - Tim J C Jacob
- Cardiff School of Biosciences, Cardiff University, Cardiff, Wales, UK
| | - Liwei Wang
- Department of Physiology, Medical College, Jinan University, Guangzhou, China
| | - Lixin Chen
- Department of Pharmacology, Medical College, Jinan University, Guangzhou, China
| |
Collapse
|
4
|
Zoledronic acid induces micronuclei formation, mitochondrial-mediated apoptosis and cytostasis in kidney cells. Life Sci 2018; 203:305-314. [PMID: 29729261 DOI: 10.1016/j.lfs.2018.04.059] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 03/11/2018] [Accepted: 04/30/2018] [Indexed: 11/21/2022]
Abstract
AIMS Zoledronic acid (ZA), a FDA approved drug has used widely in the treatment of bone metastasis complications, has been linked to renal toxicity with unclear mechanism. The present study is aimed at investigating the genotoxic and cytotoxic effects of ZA in renal epithelial cells. MAIN METHODS The genotoxic effect of ZA in Vero and MDCK cells determined by cytokinesis block micronucleus (CBMN) assay. The cytotoxic effect assessed by analysing cell cycle profile, cell death and mitochondrial membrane potential by flow cytometry using propidium iodide, AnnexinV-FITC/PI and JC1 dye staining, respectively, BAX and Bcl-2 expression by Western blotting and caspase activity by spectrofluorimetry. KEY FINDINGS The cytotoxic effect of ZA based on MTT assay revealed variable sensitivities of Vero and MDCK cells, with IC50 values of 7.41 and 109.58 μM, respectively. The CBMN assay has shown prominent dose-dependent (IC10-50) induction of micronuclei formation in both cells, indicating ZA's clastogenic and aneugenic potential. Further, the ZA treatment led the cells to apoptosis, evident from dose-dependent increase in the percentage of cells in subG1 phase and display of membranous phosphatidylserine translocation. Studies also confirmed apoptosis through mitochondria, evident from the prominent increase in BAX/Bcl-2 ratio, mitochondrial membrane depolarization and caspase-3/7 activity. In addition, ZA reduces cytokinetic activity of renal cells, evident from dose-wise lowered replicative indices. SIGNIFICANCE The study depict ZA's potential genotoxic effect along with cytotoxic effect in renal epithelial cells, could be key factors for the development of renal complications associated with it, which prompts renal safety measures in lieu with ZA usage.
Collapse
|
5
|
Du C, Wang Y, Li H, Huang Y, Jiang O, You Y, Luo F. Zoledronic acid augments the radiosensitivity of cancer cells through perturbing S- and M-phase cyclins and p21 CIP1 expression. Oncol Lett 2017; 14:4237-4242. [PMID: 28943933 DOI: 10.3892/ol.2017.6710] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Accepted: 06/02/2017] [Indexed: 02/05/2023] Open
Abstract
Radiotherapy and adjuvant chemotherapy have become the standard treatments for multiple types of cancer. Although cancer cells are usually sensitive to radiotherapy, metastasis and local failure still occur mainly due to developed resistance to radiotherapy. Thus, it is critical to improve therapeutics for cancer treatment. The present study demonstrated that third-generation bisphosphonate zoledronic acid (ZOL), even at a low concentration, augments the radiosensitivity of cancer cells exposed to ionizing radiation (IR) by inducing S-phase arrest and subsequently promoting apoptosis. This function of ZOL was associated with elevated levels of cyclin A and cyclin B in the S and M phases, as well as decreased p21CIP1 expression. In addition, ZOL also inhibited malignant the invasiveness of cancer cells. Notably, these effects could be enhanced concurrently with IR. The present data indicated that combined treatment with ZOL plus IR may be a novel technique to augment the radiosensitivity of cancer cells.
Collapse
Affiliation(s)
- Chi Du
- Department of Medical Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 610004, P.R. China.,Department of Oncology, The Second People's Hospital of Neijiang, Luzhou Medical College, Neijiang, Sichuan 641003, P.R. China
| | - Yuyi Wang
- Department of Medical Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 610004, P.R. China
| | - Haijun Li
- Department of Oncology, The Second People's Hospital of Neijiang, Luzhou Medical College, Neijiang, Sichuan 641003, P.R. China
| | - Yi Huang
- Department of Oncology, The Second People's Hospital of Neijiang, Luzhou Medical College, Neijiang, Sichuan 641003, P.R. China
| | - Ou Jiang
- Department of Oncology, The Second People's Hospital of Neijiang, Luzhou Medical College, Neijiang, Sichuan 641003, P.R. China
| | - Yanjie You
- Pathological Examinations and Research Center, Luohe, Henan 462002, P.R. China.,Department of Pharmacy, Luohe Medical College, Luohe, Henan 462002, P.R. China
| | - Feng Luo
- Department of Medical Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 610004, P.R. China
| |
Collapse
|