1
|
Restored microRNA-519a enhances the radiosensitivity of non-small cell lung cancer via suppressing EphA2. Gene Ther 2022; 29:588-600. [PMID: 33414521 DOI: 10.1038/s41434-020-00213-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 10/29/2020] [Accepted: 11/30/2020] [Indexed: 01/09/2023]
Abstract
Accumulating evidence has demonstrated that microRNA-519a (miR-519a) acts as the tumor suppressor in various cancers, but little is known regarding its intrinsic regulatory mechanisms in non-small cell lung cancer (NSCLC). Here, we aimed to investigate the role of miR-519a-targeted ephrinA2 receptor (EphA2) in radiosensitivity of NSCLC. MiR-519a and EphA2 expression in NSCLC and paracancerous tissues were detected using RT-qPCR and western blot analysis. A549 cell line was cultured and radiation-resistant cell line A549R was constructed using fractionated X-ray irradiation of these cells at 60 Gy. Colony formation ability and radioresistance of parent strain A549 and resistant strain A549R were detected with restored miR-519a and depleted EphA2. MTT assay was used to measure cell proliferation, flow cytometry was performed for determination of cell cycle distribution and apoptosis. The migration and invasion abilities were assessed by Transwell assay. The target relationship between miR-519a and EphA2 was verified. Results suggested that miR-519a was downregulated and EphA2 was upregulated in NSCLC tissues and cells, and miR-519a targeted EphA2. MiR-519a expression declined, while EphA2 expression elevated in A549R cells versus A549 cells. Upregulated miR-519a and downregulated EphA2 suppressed D0, Dq, survival fraction (SF2) and N-value, arrested cells at G0/G1 phase, advanced the apoptosis and attenuated migration, proliferation, and invasion of A549 and A549R cells. Overexpression of EphA2 reversed the promotion of upregulated miR-519a on radiosensitivity of NSCLC cells. Our results revealed that miR-519a enhances radiosensitivity of NSCLC by inhibiting EphA2 expression. Moreover, miR-519a serves as a target for NSCLC treatment.
Collapse
|
2
|
Shatnawi A, Abu Rabe DI, Frigo DE. Roles of the tumor suppressor inhibitor of growth family member 4 (ING4) in cancer. Adv Cancer Res 2021; 152:225-262. [PMID: 34353439 DOI: 10.1016/bs.acr.2021.05.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Inhibitor of growth family member 4 (ING4) is best known as a tumor suppressor that is frequently downregulated, deleted, or mutated in many cancers. ING4 regulates a broad array of tumor-related processes including proliferation, apoptosis, migration, autophagy, invasion, angiogenesis, DNA repair and chromatin remodeling. ING4 alters local chromatin structure by functioning as an epigenetic reader of H3K4 trimethylation histone marks (H3K4Me3) and regulating gene transcription through directing histone acetyltransferase (HAT) and histone deacetylase (HDAC) protein complexes. ING4 may serve as a useful prognostic biomarker for many cancer types and help guide treatment decisions. This review provides an overview of ING4's central functions in gene expression and summarizes current literature on the role of ING4 in cancer and its possible use in therapy.
Collapse
Affiliation(s)
- Aymen Shatnawi
- Department of Pharmaceutical and Administrative Sciences, University of Charleston School of Pharmacy, Charleston, WV, United States.
| | - Dina I Abu Rabe
- Integrated Bioscience Program, North Carolina Central University, Durham, NC, United States
| | - Daniel E Frigo
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, United States; Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
3
|
Tang X, Ding Y, Wang X, Wang X, Zhao L, Bi H. miR-650 promotes non-small cell lung cancer cell proliferation and invasion by targeting ING4 through Wnt-1/β-catenin pathway. Oncol Lett 2019; 18:4621-4628. [PMID: 31611970 PMCID: PMC6781663 DOI: 10.3892/ol.2019.10805] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 07/30/2019] [Indexed: 12/14/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is the most frequent cancer worldwide with a poor 5-year survival. miR-650 acts as an oncogene and regulates tumor progress in various cancers. Molecular mechanisms of miR-650 in NSCLC cell proliferation and invasion was studied. The mRNA levels of miR-650 and special genes were calculated using RT-qPCR. MTT and transwell assays were applied to measure the proliferative and invasive ability. Kaplan-Meier method was used to assess the survival of NSCLC patients. miR-650 was upregulated in NSCLC and upregulation of miR-650 was associated with a poor overall survival of NSCLC, while the results of ING4 demonstrated the opposite results. miR-650 promoted proliferation and invasion through Wnt-1/β-catenin pathway by targeting inhibitor of growth 4 (ING4) in A549 cells. ING4 was a direct target gene of miR-650 and the expression of ING4 was mediated by exogenous altering the expression of miR-650. Remarkably, alterations of ING4 expression eliminated the functions of miR-650 on the proliferation and metastasis of NSCLC. miR-650 enhanced A549 cell proliferation and invasion through Wnt-1/β-catenin pathway by directly targeting the 3'-UTR of ING4 mRNA. The newly identified miR-650/ING4 axis provides a novel insight into the pathogenesis of NSCLC.
Collapse
Affiliation(s)
- Xiangqin Tang
- Department of Oncology, Jining No. 1 People's Hospital, Jining, Shandong 272011, P.R. China
| | - Yanjun Ding
- Department of General Surgery, People's Hospital of Chiping, Chiping, Shandong 252100, P.R. China
| | - Xiaoqing Wang
- First Ward, Department of Neurology, The Affiliated Central Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Xiuzhen Wang
- Department of Clinical Nutrition, People's Hospital of Zhangqiu Area, Jinan, Shandong 250200, P.R. China
| | - Lin Zhao
- Department of Respiratory Medicine, People's Hospital of Rizhao, Rizhao, Shandong 276826, P.R. China
| | - Hongmei Bi
- Department of Respiratory Medicine, The Third People's Hospital of Qingdao, Qingdao, Shandong 266041, P.R. China
| |
Collapse
|
4
|
The essential role of tumor suppressor gene ING4 in various human cancers and non-neoplastic disorders. Biosci Rep 2019; 39:BSR20180773. [PMID: 30643005 PMCID: PMC6356015 DOI: 10.1042/bsr20180773] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 12/19/2018] [Accepted: 01/13/2019] [Indexed: 12/21/2022] Open
Abstract
Inhibitor of growth 4 (ING4), a member of the ING family discovered in 2003, has been shown to act as a tumor suppressor and is frequently down-regulated in various human cancers. Numerous published in vivo and in vitro studies have shown that ING4 is responsible for important cancer hallmarks such as pathologic cell cycle arrest, apoptosis, autophagy, contact inhibition, and hypoxic adaptation, and also affects tumor angiogenesis, invasion, and metastasis. These characteristics are typically associated with regulation through chromatin acetylation by binding histone H3 trimethylated at lysine 4 (H3K4me3) and through transcriptional activity of transcription factor P53 and NF-κB. In addition, emerging evidence has indicated that abnormalities in ING4 expression and function play key roles in non-neoplastic disorders. Here, we provide an overview of ING4-modulated chromosome remodeling and transcriptional function, as well as the functional consequences of different genetic variants. We also present the current understanding concerning the role of ING4 in the development of neoplastic and non-neoplastic diseases. These studies offer inspiration for pursuing novel therapeutics for various cancers.
Collapse
|
5
|
Wang L, Xia Y, Chen T, Zeng Y, Li L, Hou Y, Li W, Liu Z. Sanyang Xuedai enhances the radiosensitivity of human non-small cell lung cancer cells via increasing iNOS/NO production. Biomed Pharmacother 2018; 102:618-625. [PMID: 29602129 DOI: 10.1016/j.biopha.2018.03.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Revised: 02/28/2018] [Accepted: 03/05/2018] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE In this research, we aimed at finding out how San Yang Xue Dai (SYKT) promotes the radiosensitivity of non-small cell lung cancer (NSCLC) cell line NCI-H460. METHODS Survival rate of NSCLC cells (A549, NCI-H460, NCI-H1650 and NCI-H1975) after the SYKT treatment or irradiation (IR) was calculated by the MTT assay. The radiosensitization of SYKT (0.5 g/mL and 1.0 g/mL) on cell line NCI-H460 and the radioresistant cell line NCI-H460R was studied by MTT assay and clone formation assay. The protein expression levels of iNOS, Cyclin B1 and CDC2 were determined by western blot, and the expression of NO was measured by Griess method. Finally, cell cycle and apoptotic rate of NSCLC cell line NCI-H460 were accessed by flow cytometry assay. BrdU staining was also applied to detect the cell proliferation after IR with or without SYKT treatment. RESULTS The IC10 value of SYKT for NCI-H460 cells was 1.03 g/mL. After 1.0 g/mL SYKT treatment, the radiosensitivity of NCI-H460R cells was enhanced. The level of iNOS in the cells was found decreased after IR. We also found that SYKT could enhance iNOS and NO expressions while inhibit cyclin B1 and CDC2 expressions in radiation resistant cells. Combining β-irradiation with SYKT caused cell cycle arrest in G2/M phase and increased cell apoptosis. CONCLUSION SYKT resensitized radioresistant NCI-H460R cells via increasing cell apoptosis and cell cycle arrest. This was due to an elevated NO level caused by accumulating iNOS and effects of SYKT on radiosensitization of NSCLC should be further investigated in clinical application.
Collapse
Affiliation(s)
- Li Wang
- Department of Radiotherapy Oncology, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming 650118, Yunnan, China
| | - Yaoxiong Xia
- Department of Radiotherapy Oncology, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming 650118, Yunnan, China
| | - Ting Chen
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming 650118, Yunnan, China
| | - Yueqin Zeng
- Institute of Molecular and Clinical Medicine, Kunming Medical University, Kunming 650000, Yunnan, China
| | - Lan Li
- Department of Radiotherapy Oncology, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming 650118, Yunnan, China
| | - Yu Hou
- Department of Radiotherapy Oncology, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming 650118, Yunnan, China
| | - Wenhui Li
- Department of Radiotherapy Oncology, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming 650118, Yunnan, China.
| | - Zhijie Liu
- Institute of Molecular and Clinical Medicine, Kunming Medical University, Kunming 650000, Yunnan, China.
| |
Collapse
|
6
|
Zhang Q, Chen X, Luo Y, Ren H, Qiao T. Fuzi Enhances Anti-Tumor Efficacy of Radiotherapy on Lung Cancer. J Cancer 2017; 8:3945-3951. [PMID: 29187869 PMCID: PMC5705996 DOI: 10.7150/jca.22162] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Accepted: 08/25/2017] [Indexed: 12/23/2022] Open
Abstract
In traditional Chinese medicine, Fuzi is widely used as an antitumor agent or an adjuvant medication combined with radiotherapy and chemotherapy, but its mechanism remains unclear. In this study, we investigated anti-tumor and immunoregulation efficacy of Fuzi combined with radiotherapy in mice with Lewis lung cancer (LLC). We found that Fuzi combined with radiotherapy significantly inhibited the growth of LLC, promoted the apoptosis of cancer cells, and prolonged the survival of mice with LLC. Mechanistically, we found that Fuzi decreased the proportion of Treg cells, reduced serum levels of cytokines such as interleukin (IL)-10 and transforming growth factor (TGF)-β, and downregulated the expression of programmed death ligand-1 in mice with LLC subjected to radiotherapy. This study suggests that Fuzi has immunomodulation function to act as radiosensitizer and improve radiotherapy against lung cancer.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Oncology, Jinshan Hospital, Fudan University, Shanghai 201508, China
| | - Xue Chen
- Department of Oncology, Jinshan Hospital, Fudan University, Shanghai 201508, China
| | - Youjun Luo
- Department of Oncology, Jinshan Hospital, Fudan University, Shanghai 201508, China
| | - Huaping Ren
- Department of Chinese Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, China
| | - Tiankui Qiao
- Department of Oncology, Jinshan Hospital, Fudan University, Shanghai 201508, China
| |
Collapse
|