1
|
Dasgupta A, Nandi S, Gupta S, Roy S, Das C. To Ub or not to Ub: The epic dilemma of histones that regulate gene expression and epigenetic cross-talk. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2024; 1867:195033. [PMID: 38750882 DOI: 10.1016/j.bbagrm.2024.195033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 01/04/2024] [Accepted: 05/06/2024] [Indexed: 05/23/2024]
Abstract
A dynamic array of histone post-translational modifications (PTMs) regulate diverse cellular processes in the eukaryotic chromatin. Among them, histone ubiquitination is particularly complex as it alters nucleosome surface area fostering intricate cross-talk with other chromatin modifications. Ubiquitin signaling profoundly impacts DNA replication, repair, and transcription. Histones can undergo varied extent of ubiquitination such as mono, multi-mono, and polyubiquitination, which brings about distinct cellular fates. Mechanistic studies of the ubiquitin landscape in chromatin have unveiled a fascinating tapestry of events that orchestrate gene regulation. In this review, we summarize the key contributors involved in mediating different histone ubiquitination and deubiquitination events, and discuss their mechanism which impacts cell transcriptional identity and DNA damage response. We also focus on the proteins bearing epigenetic reader modules critical in discerning site-specific histone ubiquitination, pivotal for establishing complex epigenetic crosstalk. Moreover, we highlight the role of histone ubiquitination in different human diseases including neurodevelopmental disorders and cancer. Overall the review elucidates the intricate orchestration of histone ubiquitination impacting diverse cellular functions and disease pathogenesis, and provides insights into the current challenges of targeting them for therapeutic interventions.
Collapse
Affiliation(s)
- Anirban Dasgupta
- Structural Biology and Bioinformatics Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India; Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9148, USA
| | - Sandhik Nandi
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India; Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai 400094, India
| | - Sayan Gupta
- Structural Biology and Bioinformatics Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India
| | - Siddhartha Roy
- Structural Biology and Bioinformatics Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India
| | - Chandrima Das
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India; Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai 400094, India.
| |
Collapse
|
2
|
Liu C, Kuang J, Wang Y, Duan T, Min L, Lu C, Zhang T, Chen R, Wu Y, Zhu L. A functional reference map of the RNF8 interactome in cancer. Biol Direct 2022; 17:17. [PMID: 35831895 PMCID: PMC9277853 DOI: 10.1186/s13062-022-00331-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 06/20/2022] [Indexed: 12/02/2022] Open
Abstract
Background RNF8 is an E3 ligase identified as a critical DNA damage-responsive protein. Recently, multiple reports have shown that RNF8 could be used as an important therapeutic target for cancer chemo/radiotherapy. However, the understanding of RNF8 remains limited due to the lack of its interactome reference map and comprehensive analysis of RNF8 in diverse cancers, which underscores the need to map the interactome of RNF8 via high-throughput methods. Results A two-way identification method based on LC–MS was designed for the identification of the RNF8 interactome with high-specificity. By in silico analysis and in vitro validation, we identified a new reference map of the RNF8 interactome network containing many new targets, such as YBX1, DNMT1, and HDCA1, new biological functions and the gene-disease associations of RNF8. Our results revealed a close relationship between RNF8 and neurodegenerative diseases or tumor-infiltrating immune cells using bulk RNA-seq and scRNA-seq datasets. As a proof of concept of our interactome map, we validated the direct binding between RNF8 and YBX1 and showed that RNF8 catalyzed the ubiquitination of YBX1. These results demonstrated that RNF8 might be a crucial regulator of YBX1. Conclusions Our work provides a unique framework for researchers and clinicians who seek to better explore or understand RNF8-regulated biological functions in cancers. This study will hopefully facilitate the rational design and further development of anti-RNF8 therapy in cancers. Graphical abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13062-022-00331-z.
Collapse
Affiliation(s)
- Chuanyang Liu
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, 410073, Hunan, China
| | - Jingyu Kuang
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, 410073, Hunan, China.
| | - Yuxuan Wang
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, 410073, Hunan, China
| | - Ting Duan
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Lu Min
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, 410073, Hunan, China
| | - Chenyu Lu
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, 410073, Hunan, China
| | - Tianyi Zhang
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, 410073, Hunan, China
| | - Ruifen Chen
- Joint Logistic Support Force 921th Hospital, Changsha, 410073, Hunan, China
| | - Ying Wu
- Department of Critical Care Medicine, Second Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Lingyun Zhu
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, 410073, Hunan, China.
| |
Collapse
|
3
|
Xu Y, Hu Y, Xu T, Yan K, Zhang T, Li Q, Chang F, Guo X, Peng J, Li M, Zhao M, Zhen H, Xu L, Zheng D, Li L, Shao G. RNF8-mediated regulation of Akt promotes lung cancer cell survival and resistance to DNA damage. Cell Rep 2021; 37:109854. [PMID: 34686341 DOI: 10.1016/j.celrep.2021.109854] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 08/18/2021] [Accepted: 09/28/2021] [Indexed: 01/21/2023] Open
Abstract
Despite the tremendous success of targeted and conventional therapies for lung cancer, therapeutic resistance is a common and major clinical challenge. RNF8 is a ubiquitin E3 ligase that plays essential roles in the DNA damage response; however, its role in the pathogenesis of lung cancer is unclear. Here, we report that RNF8 is overexpressed in lung cancer and positively correlates with the expression of p-Akt and poor survival of patients with non-small-cell lung cancer. In addition, we identify RNF8 as the E3 ligase for regulating the activation of Akt by K63-linked ubiquitination under physiological and genotoxic conditions, which leads to lung cancer cell proliferation and resistance to chemotherapy. Together, our study suggests that RNF8 could be a very promising target in precision medicine for lung cancer.
Collapse
Affiliation(s)
- Yongjie Xu
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yumeng Hu
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Tao Xu
- The Affiliated Hospital of Qingdao University, Qingdao 266021, China
| | - Kaowen Yan
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Ting Zhang
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Qin Li
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Fen Chang
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Xueyuan Guo
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Jingyu Peng
- State Key Laboratory of Membrane Biology, Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing 100871, China
| | - Mo Li
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China
| | - Min Zhao
- Department of Oncology, Hebei Chest Hospital, Research Center of Hebei Lung Cancer Prevention and Treatment, Shijiazhuang, Hebei 050041, China
| | - Hongying Zhen
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Luzheng Xu
- Medical and Health Analysis Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Duo Zheng
- Department of Cell Biology and Genetics, Shenzhen University School of Medicine, Shenzhen 518055, China
| | - Li Li
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China.
| | - Genze Shao
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China.
| |
Collapse
|
4
|
Xiao N, Li C, Liao W, Yin J, Zhang S, Zhang P, Yuan L, Hong M. FOXG1 mediates the radiosensitivity of glioma cells through regulation of autophagy. Int J Radiat Biol 2021; 97:139-148. [PMID: 33201747 DOI: 10.1080/09553002.2021.1846816] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND/AIM Upregulation of Forkhead box G1 (FOXG1) has recently been observed in many cancers, while its effect on radiosensitivity in glioma is still unclear. In this study, we hypothesized that FOXG1 be a major player in radioresistance of glioma as well as the underlying mechanism. METHODS Immunohistochemistry (IHC) was conducted to assess FOXG1 expression in glioma tissues and glioma-adjacent tissues. Western Blot was implemented to detect the expression of autophagy-related proteins. CCK-8, colony formation and flow cytometry assays were implemented to assess cell viability, proliferation and apoptosis, respectively. Transmission electron microscope (TEM) was used to observe autophagic vesicles. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) assay was applied to detect the expression of FOXG1. RESULTS The present study demonstrated that FOXG1 was highly expressed in glioma tissues. FOXG1 expression level was up-regulated in glioma cells following exposure to X-ray irradiation. FOXG1 can attenuate radiosensitivity of glioma cells. Moreover, it revealed that FOXG1 attenuate radiosensitivity of glioma cells by promoting autophagy. CONCLUSIONS The present study suggests that FOXG1 is a pivotal molecule for circumventing radiation-induced cell death in malignant glioma cells through the regulation of autophagy, and it may be a target for the treatment of human brain glioma.
Collapse
Affiliation(s)
- Ning Xiao
- Health Management Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Churong Li
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Wenjun Liao
- West China Hospital, Sichuan University, Chengdu, China
| | - Jun Yin
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Shichuan Zhang
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Peng Zhang
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lan Yuan
- School of Medical and Life Sciences/Reproductive & Women-Children Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Min Hong
- Health Management Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
5
|
Kuang J, Min L, Liu C, Chen S, Gao C, Ma J, Wu X, Li W, Wu L, Zhu L. RNF8 Promotes Epithelial-Mesenchymal Transition in Lung Cancer Cells via Stabilization of Slug. Mol Cancer Res 2020; 18:1638-1649. [PMID: 32753472 DOI: 10.1158/1541-7786.mcr-19-1211] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 03/11/2020] [Accepted: 07/22/2020] [Indexed: 11/16/2022]
Abstract
RNF8 (ring finger protein 8), a RING finger E3 ligase best characterized for its role in DNA repair and sperm formation via ubiquitination, has been found to promote tumor metastasis in breast cancer recently. However, whether RNF8 also plays a role in other types of cancer, especially in lung cancer, remains unknown. We show here that RNF8 expression levels are markedly increased in human lung cancer tissues and negatively correlated with the survival time of patients. Overexpression of RNF8 promotes the EMT process and migration ability of lung cancer cells, while knockdown of RNF8 demonstrates the opposite effects. In addition, overexpression of RNF8 activates the PI3K/Akt signaling pathway, knockdown of RNF8 by siRNA inhibits this activation, and pharmacologic inhibition of PI3K/Akt in RNF8-overexpressing cells also reduces the expression of EMT markers and the ability of migration. Furthermore, RNF8 is found to directly interact with Slug and promoted the K63-Ub of Slug, and knockdown of Slug disrupts RNF8-dependent EMT in A549 cells, whereas overexpression of Slug rescues RNF8-dependent MET in H1299 cells, and depletion of RNF8 expression by shRNA inhibits metastasis of lung cancer cells in vivo. Taken together, these results indicate that RNF8 is a key regulator of EMT process in lung cancer and suggest that inhibition of RNF8 could be a useful strategy for lung cancer treatment. IMPLICATIONS: This study provides a new mechanistic insight into the novel role of RNF8 and identifies RNF8 as a potential new therapeutic target for the treatment of lung cancer.
Collapse
Affiliation(s)
- Jingyu Kuang
- Department of Biology and Chemistry, College of Liberal Arts and Sciences, National University of Defense Technology, Changsha, Hunan, China
| | - Lu Min
- Department of Biology and Chemistry, College of Liberal Arts and Sciences, National University of Defense Technology, Changsha, Hunan, China
| | - Chuanyang Liu
- Department of Biology and Chemistry, College of Liberal Arts and Sciences, National University of Defense Technology, Changsha, Hunan, China
| | - Si Chen
- Department of pathology, Hunan Provincial People's Hospital, Changsha, Hunan, China
| | - Changsong Gao
- Department of Biology and Chemistry, College of Liberal Arts and Sciences, National University of Defense Technology, Changsha, Hunan, China
| | - Jiaxin Ma
- Department of Biology and Chemistry, College of Liberal Arts and Sciences, National University of Defense Technology, Changsha, Hunan, China
| | - Xiaomin Wu
- Department of Biology and Chemistry, College of Liberal Arts and Sciences, National University of Defense Technology, Changsha, Hunan, China
| | - Wenying Li
- Department of Biology and Chemistry, College of Liberal Arts and Sciences, National University of Defense Technology, Changsha, Hunan, China
| | - Lei Wu
- Department of Biology and Chemistry, College of Liberal Arts and Sciences, National University of Defense Technology, Changsha, Hunan, China. .,Hunan Engineering Research Center for Intelligent Decision Making and Big Data on Industrial Development, Hunan University of Science and Technology, Xiangtan, Hunan, China
| | - Lingyun Zhu
- Department of Biology and Chemistry, College of Liberal Arts and Sciences, National University of Defense Technology, Changsha, Hunan, China.
| |
Collapse
|
6
|
Xu L, Wang Y, Liu Q, Luo H, Zhong X, Li Y. [Role of Autophagy in the Radiosensitivity of Human Lung Adenocarcinoma A549 Cells]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2017; 19:799-804. [PMID: 27978864 PMCID: PMC5973450 DOI: 10.3779/j.issn.1009-3419.2016.12.01] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
背景与目的 放射治疗是肺癌最重要的治疗手段之一,然而却因放疗抵抗极易导致肿瘤的复发和转移。放疗可诱导肿瘤细胞自噬发生,最新研究也报道,自噬可能与DNA损伤修复过程相关。本研究旨在探讨通过雷帕霉素上调A549细胞自噬,能否增加细胞放疗敏感性,其过程是否与DNA损伤修复过程相关。 方法 以人肺腺癌A549细胞作为实验对象,实验设对照组(N)、单纯放疗组(IR)、雷帕霉素联合放疗组(R+RAPA)。采用Western blot检测γ-H2AX蛋白质、Rad51蛋白质、Ku70/80蛋白质、p62蛋白质、LC3蛋白质表达;电镜检测自噬体形成;细胞克隆形成实验检测细胞存活分数(survival fraction, SF)值。 结果 与单纯放疗组相比,放疗联合雷帕霉素组自噬活性增加,且Rad51、Ku80蛋白质表达减少,细胞增殖能力下降。 结论 通过雷帕霉素上调自噬可增加肺癌细胞放疗敏感性,其机制可能与抑制DNA损伤修复过程相关。
Collapse
Affiliation(s)
- Liyao Xu
- Department of Oncology, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Yong Wang
- Department of Oncology, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Qin Liu
- Department of Respirology, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Hui Luo
- Department of Oncology, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Xiaojun Zhong
- Department of Oncology, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Yong Li
- Department of Oncology, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| |
Collapse
|
7
|
RNF8 identified as a co-activator of estrogen receptor α promotes cell growth in breast cancer. Biochim Biophys Acta Mol Basis Dis 2017; 1863:1615-1628. [PMID: 28216286 DOI: 10.1016/j.bbadis.2017.02.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 01/20/2017] [Accepted: 02/09/2017] [Indexed: 02/07/2023]
Abstract
The ring finger protein 8 (RNF8), a key component of protein complex crucial for DNA-damage response, consists of a forkhead-associated (FHA) domain and a really interesting new gene (RING) domain that enables it to function as an E3 ubiquitin ligase. However, the biological functions of RNF8 in estrogen receptor α (ERα)-positive breast cancer and underlying mechanisms have not been fully defined. Here, we have explored RNF8 as an associated partner of ERα in breast cancer cells, and co-activates ERα-mediated transactivation. Accordingly, RNF8 depletion inhibits the expression of endogenous ERα target genes. Interestingly, our results have demonstrated that RNF8 increases ERα stability at least partially if not all via triggering ERα monoubiquitination. RNF8 functionally promotes breast cancer cell proliferation. RNF8 is highly expressed in clinical breast cancer samples and the expression of RNF8 positively correlates with that of ERα. Up-regulation of ERα-induced transactivation by RNF8 might contribute to the promotion of breast cancer progression by allowing enhancement of ERα target gene expression. Our study describes RNF8 as a co-activator of ERα increases ERα stability via post-transcriptional pathway, and provides a new insight into mechanisms for RNF8 to promote cell growth of ERα-positive breast cancer.
Collapse
|
8
|
Tumor invasion and metastasis regulated by microRNA-184 and microRNA-574-5p in small-cell lung cancer. Oncotarget 2016; 6:44609-22. [PMID: 26587830 PMCID: PMC4792579 DOI: 10.18632/oncotarget.6338] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Accepted: 11/06/2015] [Indexed: 12/12/2022] Open
Abstract
Small-cell lung cancer (SCLC) is a highly aggressive neuroendocrine tumor that has an extremely poor clinical prognosis. Metastasis is the key event in SCLC progression, but its mechanism has not been fully elucidated. MicroRNAs (miRNAs) have been proven to participate in cancer processes, but their function in SCLC has not been thoroughly studied either. Here, we performed microarray and quantitative real-time PCR (qRT-PCR) analysesto identify the miRNAsassociated with metastasis and prognosis in SCLC as well as the correlation between serum and tissue. We also explored these miRNAs' promising molecular mechanisms by 3′UTR reporter assay and immunoblotting. We showed thatmiR-184 significantly attenuated the metastasis of SCLC, whereas miR-574–5p enhanced it. Both miRNAs were found to participate in β-catenin signaling by suppressing protein tyrosine phosphatase receptor type U (PTPRU)orendothelial PAS domain protein 1 (EPAS1). Furthermore, miR-574–5p was verified as an independent prognostic risk factor for SCLC. Taken together, our findings providea comprehensive analysis of the miRNA expression pattern in SCLC and indicate that miRNAs may serve as potential therapeutic and prognostic predictors in SCLC.
Collapse
|
9
|
Zhou R, Zhou X, Yin Z, Guo J, Hu T, Jiang S, Liu L, Dong X, Zhang S, Wu G. MicroRNA-574-5p promotes metastasis of non-small cell lung cancer by targeting PTPRU. Sci Rep 2016; 6:35714. [PMID: 27761023 PMCID: PMC5071770 DOI: 10.1038/srep35714] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 10/04/2016] [Indexed: 02/06/2023] Open
Abstract
Dysregulation of microRNAs (miRNAs) has been associated with malignant behavior in a variety of cancers. Our previous study demonstrated that miRNA expression profiles are predictors for patients with advanced non-small cell lung cancer (NSCLC). We also showed that miRNAs are involved in small-cell lung cancer metastasis. Here, we used qRT-PCR to re-analyze our previous microarray results using serum samples from 75 patients with NSCLC. Surprisingly, we found that miR-574-5p and miR-874 were overexpressed in patients with metastatic advanced NSCLC but not in patients with non-metastatic advanced NSCLC. Additionally, miR-574-5p expression was correlated between matched serum and tissue samples from 68 patients. However, these 2 miRNAs are not prognostic factors for NSCLC. Transwell and wound-healing assays showed that miR-574-5p promotes the migration and invasion of NSCLC cells. Furthermore, miR-574-5p enhanced the tyrosine phosphorylation of β-catenin by repressing PTPRU expression in vitro. In conclusion, this study explored the expression of miR-574-5p in clinical samples and its molecular mechanisms in the metastasis of advanced NSCLC.
Collapse
Affiliation(s)
- Rui Zhou
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Xiaoshu Zhou
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Zhongyuan Yin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Jing Guo
- Department of Oncology of the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ting Hu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Shun Jiang
- Department of Oncology, Second Xiangya Hospital of Central South University, Changsha, China
| | - Li Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Xiaorong Dong
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Sheng Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Gang Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| |
Collapse
|
10
|
Li Y, Liu F, Wang Y, Li D, Guo F, Xu L, Zeng Z, Zhong X, Qian K. Rapamycin-induced autophagy sensitizes A549 cells to radiation associated with DNA damage repair inhibition. Thorac Cancer 2016; 7:379-86. [PMID: 27385978 PMCID: PMC4930955 DOI: 10.1111/1759-7714.12332] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 12/14/2015] [Indexed: 01/09/2023] Open
Abstract
Background Autophagy has been reported to increase in cancer cells after radiation. However, it remains unknown whether increased autophagy as a result of radiation affects DNA damage repair and sensitizes cancer cells. In this study, the radiosensitization effect of rapamycin, a mammalian target of rapamycin inhibitor that induces autophagy, on human lung adenocarcinoma A549 cells was investigated. Methods A549 cells were treated with different concentrations of rapamycin. Cell viability was evaluated by methyl‐thiazolyl‐tetrazolium assay. Survival fraction values of A549 cells after radiotherapy were detected by colony formation assay. Autophagosome was observed by a transmission electron microscope. Furthermore, Western blot was employed to examine alterations in autophagy protein LC3 and p62, DNA damage protein γ–H2AX, and DNA damage repair proteins Rad51, Ku70, and Ku80. Rad51, Ku70, and Ku80 messenger ribonucleic acid (mRNA) expression levels were examined by real‐time polymerase chain reaction. Results Rapamycin suppressed A549 cell proliferation in dose and time‐dependent manners. An inhibitory concentration (IC)10 dose of rapamycin could induce autophagy in A549 cells. Rapamycin combined with radiation significantly decreased the colony forming ability of cells, compared with rapamycin or radiation alone. Rapamycin and radiation combined increased γ–H2AX expression levels and decreased Rad51 and Ku80 expression levels, compared with single regimens. However, rapamycin treatment did not induce any change in Rad51, Ku70, and Ku80 mRNA levels, regardless of radiation. Conclusions These findings indicate that increasing autophagy sensitizes lung cancer cells to radiation.
Collapse
Affiliation(s)
- Yong Li
- Department of Medical Oncology The First Affiliated Hospital of Nanchang University Nanchang Jiangxi China
| | - Fen Liu
- Critical Care Medicine The First Affiliated Hospital of Nanchang University Nanchang Jiangxi China
| | - Yong Wang
- Department of Medical Oncology The First Affiliated Hospital of Nanchang University Nanchang Jiangxi China
| | - Donghai Li
- Department of Neurosurgery The First Affiliated Hospital of Nanchang University Nanchang Jiangxi China
| | - Fei Guo
- The institute of Burn Research The First Affiliated Hospital of Nanchang University Nanchang Jiangxi China
| | - Liyao Xu
- Department of Medical Oncology The First Affiliated Hospital of Nanchang University Nanchang Jiangxi China
| | - Zhengguo Zeng
- Critical Care Medicine The First Affiliated Hospital of Nanchang University Nanchang Jiangxi China
| | - Xiaojun Zhong
- Department of Medical Oncology The First Affiliated Hospital of Nanchang University Nanchang Jiangxi China
| | - Kejian Qian
- Critical Care Medicine The First Affiliated Hospital of Nanchang University Nanchang Jiangxi China
| |
Collapse
|
11
|
Song L, Ma L, Cong F, Shen X, Jing P, Ying X, Zhou H, Jiang J, Fu Y, Yan H. Radioprotective effects of genistein on HL-7702 cells via the inhibition of apoptosis and DNA damage. Cancer Lett 2015; 366:100-11. [PMID: 26095601 DOI: 10.1016/j.canlet.2015.06.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Revised: 05/06/2015] [Accepted: 06/15/2015] [Indexed: 02/07/2023]
Abstract
Radiation induced normal tissue damage is the most important limitation for the delivery of a high potentially curative radiation dose. Genistein (GEN), one of the main soy isoflavone components, has drawn wide attention for its bioactivity in alleviating radiation damage. However, the effects and molecular mechanisms underlying the radioprotective effects of GEN remain unclear. In the present study, we showed that low concentration of GEN (1.5 µM) protected L-02 cells against radiation damage via inhibition of apoptosis, alleviation of DNA damage and chromosome aberration, down-regulation of GRP78 and up-regulation of HERP, HUS1 and hHR23A. In contrast, high concentration of GEN (20 µM) demonstrated radiosensitizing characteristics through the promotion of apoptosis and chromosome aberration, impairment of DNA repair, up-regulation of GRP78, and down-regulation of HUS1, SIRT1, RAD17, RAD51 and RNF8. These findings shed light on using low, but not high-concentration GEN, as a potential candidate for adjuvant therapy to alleviate radiation-induced injuries to human recipients of ionizing radiation.
Collapse
Affiliation(s)
- Lihua Song
- Key Laboratory of Urban Agriculture (South), Ministry of Agriculture, Research Center for Food Safety and Nutrition, Bor S. Luh Food Safety Research Center, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Lijun Ma
- Department of Oncology, Shanghai Tongren Hospital, Shanghai Jiaotong University, Shanghai 200336, China
| | - Fengsong Cong
- School of Life Science and Technology, Shanghai Jiao Tong University, Shanghai 200020, China
| | - Xiuhua Shen
- Nutrition Department, School of Medicine, Shanghai Jiao Tong University, Shanghai 200020, China
| | - Pu Jing
- Key Laboratory of Urban Agriculture (South), Ministry of Agriculture, Research Center for Food Safety and Nutrition, Bor S. Luh Food Safety Research Center, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xiong Ying
- Key Laboratory of Urban Agriculture (South), Ministry of Agriculture, Research Center for Food Safety and Nutrition, Bor S. Luh Food Safety Research Center, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Haiyue Zhou
- Key Laboratory of Urban Agriculture (South), Ministry of Agriculture, Research Center for Food Safety and Nutrition, Bor S. Luh Food Safety Research Center, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jing Jiang
- Key Laboratory of Urban Agriculture (South), Ministry of Agriculture, Research Center for Food Safety and Nutrition, Bor S. Luh Food Safety Research Center, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yongye Fu
- Department of Laboratory Medicine, Changhai Hosipital, Second Military Medical University, Shanghai 200433, China
| | - Hongli Yan
- Department of Laboratory Medicine, Changhai Hosipital, Second Military Medical University, Shanghai 200433, China.
| |
Collapse
|
12
|
WANG MAOXIN, CHEN XIANMING, CHEN HUI, ZHANG XIAN, LI JIANZHONG, GONG HONGXUN, SHIYAN CHEN, YANG FAN. RNF8 plays an important role in the radioresistance of human nasopharyngeal cancer cells in vitro. Oncol Rep 2015; 34:341-9. [DOI: 10.3892/or.2015.3958] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Accepted: 04/20/2015] [Indexed: 11/06/2022] Open
|
13
|
Zhou C, Liu Z, Liu Y, Fu W, Ding X, Liu J, Yu Y, Zhang Q. Gene silencing of porcine MUC13 and ITGB5: candidate genes towards Escherichia coli F4ac adhesion. PLoS One 2013; 8:e70303. [PMID: 23922972 PMCID: PMC3726385 DOI: 10.1371/journal.pone.0070303] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2013] [Accepted: 06/17/2013] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Integrin beta-5 (ITGB5) and mucin 13 (MUC13) genes are highly expressed on the apical surface of intestinal epithelia and are thought to be candidate genes for controlling the expression of the receptor for enterotoxigenic Escherichia coli (ETEC) F4ac. Human MUC13 protein has an expected role in protecting intestinal mucosal surfaces and porcine ITGB5 is a newly identified potential receptor for ETEC F4ac. METHODOLOGY/PRINCIPAL FINDINGS To test the hypothesis that ITGB5 and MUC13 both play key roles in protection of the intestinal mucosa against pathogenic bacterium, porcine intestinal epithelial cells (IPEC-J2) were transfected with ITGB5-targeting, MUC13-targeting or negative control small interfering RNA (siRNA), respectively. Firstly, we measured mRNA expression levels of mucin genes (MUC4, MUC20), pro-inflammatory genes (IL8, IL1A, IL6, CXCL2), anti-inflammatory mediator SLPI, and PLAU after RNAi treatments with and without ETEC infection. Secondly, we compared the adhesions of ETEC to the pre- and post-knockdown IPEC-J2 cells of ITGB5 and MUC13, respectively. We found that ITGB5 and MUC13 knockdown both had small but significant effects in attenuating the inflammation induced by ETEC infection, and both increased bacterial adhesion in response to F4ac ETEC exposure. CONCLUSIONS/SIGNIFICANCE Our current study first reported that ITGB5 and MUC13 are important adhesion molecules of mucosal epithelial signaling in response to Escherichia coli in pigs. These data suggest that both ITGB5 and MUC13 play key roles in defending the attachment and adhesion of ETEC to porcine jejunal cells and in maintaining epithelial barrier and immunity function.
Collapse
Affiliation(s)
- Chuanli Zhou
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, P.R. China
| | - Zhengzhu Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, P.R. China
- Department of Animal Science, College of Animal Science and Technology, Hebei Normal University of Science and Technology, Changli, P.R. China
| | - Yang Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, P.R. China
| | - Weixuan Fu
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, P.R. China
| | - Xiangdong Ding
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, P.R. China
| | - Jianfeng Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, P.R. China
| | - Ying Yu
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, P.R. China
- * E-mail: (YY); (QZ)
| | - Qin Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, P.R. China
- * E-mail: (YY); (QZ)
| |
Collapse
|