1
|
Yu X, Du C, Cui Y, Jiang Y, Feng D. ELK3 Targeting AEG1 Promotes Migration and Invasion of Ovarian Cancer Cells under Hypoxia. Biol Pharm Bull 2023; 46:883-892. [PMID: 37394639 DOI: 10.1248/bpb.b22-00780] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Ovarian cancer (OC) is one of the most common tumors in female reproductive organs with a five-year survival rate of less than 45%. Metastasis is a crucial contributor to OC development. ETS transcription factor (ELK3), as a transcriptional factor, have been involved in multiple tumor development. However, its role in OC remains elusive. In this study, we observed high expression of ELK3 and AEG1 in human OC tissues. OVCAR-3 and SKOV3 cells were treated with hypoxia to mimic tumor microenvironment in vivo. We found that the expression of ELK3 was significantly increased in cells under hypoxia compared with normoxia. ELK3 knockdown inhibited cell migration and invasion abilities under hypoxia. Moreover, ELK3 knockdown decreased β-catenin expression and inhibited the activation of Wnt/β-catenin pathway in SKOV3 cells under hypoxia. Astrocyte-elevated gene-1 (AEG1) has been reported to promote OC progression. Our results showed that the mRNA level of AEG1 was decreased when ELK3 knockdown under hypoxia. Dural luciferase assay confirmed that ELK3 bound to gene AEG1 promoter (-2005-+15) and enhanced its transcriptional activity under hypoxia. Overexpression of AEG1 increased the migration and invasion abilities of SKOV3 cell with ELK3 knockdown. In the absence of ELK3, the activation of β-catenin was recovered by AEG1 overexpression. To sum up, we conclude that ELK3 promotes AEG1 expression by binding to its promoter. ELK3 could promote migration and invasion of OC cells by targeting AEG1, which provides a potential basis for therapeutic approaches to OC.
Collapse
Affiliation(s)
- Xiaoyu Yu
- Department of Pathology, Harbin Medical University Cancer Hospital
| | - Chun Du
- Department of Pathology, Harbin Medical University Cancer Hospital
| | - Yifei Cui
- Department of Pathology, Harbin Medical University Cancer Hospital
| | - Yang Jiang
- Department of Pathology, Harbin Medical University Cancer Hospital
| | - Di Feng
- Department of Pathology, Harbin Medical University Cancer Hospital
| |
Collapse
|
2
|
Song TJ, Lin XH, Huang PT, Chen YQ, Chen LM. T4 reduces cisplatin resistance by inhibiting AEG-1 gene expression in lung cancer cells. Sci Rep 2022; 12:11462. [PMID: 35794136 PMCID: PMC9259636 DOI: 10.1038/s41598-022-15643-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 06/27/2022] [Indexed: 11/09/2022] Open
Abstract
Lung cancer is the most malignant form of cancer and has the highest morbidity and mortality worldwide. Due to drug resistance, the current chemotherapy for lung cancer is not effective and has poor therapeutic effects. Tripchlorolide (T4), a natural extract from the plant Tripterygium wilfordii, has powerful immunosuppressive and antitumour effects and may become a potential therapeutic agent for lung cancer. Therefore, this study aimed to investigate the effect of T4 on reducing chemoresistance in lung cancer cells and to explore the mechanism. 1. A549 and A549/DDP cells were separately transfected with AEG-1 overexpression and AEG-1 knockdown plasmids. A549/DDP cells were divided into the A549/DDP empty group, T4 group, and T4 + AEG-1 overexpression group. A CCK-8 assay was used to evaluate the proliferation of cells in each group. RT–qPCR and Western blotting were used to detect the expression of AEG-1 and MDR-1. Expression of AEG-1 in A549 and A549/DDP cells was positively correlated with cisplatin resistance. When the AEG-1 protein was overexpressed in A549 cells, the lethal effect of cisplatin on A549 cells was attenuated (all P < 0.05). After the AEG-1 protein was knocked down in A549/DDP cells, cisplatin was applied. The lethal effect was significantly increased compared to that in the corresponding control cells (all P < 0.05). AEG-1 protein expression gradually decreased with increasing T4 concentration in A549 and A549/DDP cells. Resistance to cisplatin was reduced after the addition of T4 to A549/DDP cells (P < 0.05), and this effect was enhanced after transfection with the AEG-1 knockdown plasmid. T4 plays an important role in increasing the sensitivity of lung cancer cells to cisplatin.
Collapse
Affiliation(s)
- Tian-Jiao Song
- Department of Emergency, Fujian Provincial Hospital, 134 East Street, Fuzhou, 350001, Fujian, People's Republic of China.,Shengli Clinical Medical College of Fujian Medical University, 134 East Street, Fuzhou, 350001, Fujian, People's Republic of China.,Fujian Provincial Key Laboratory of Emergency Medicine, 134 East Street, Fuzhou, 350001, Fujian, People's Republic of China
| | - Xiao-Hong Lin
- Department of Respiratory Medicine, The Affiliated Hospital of Putian University, No.999 Dongzhen East Road, Pu'tian, 351100, Fujian, People's Republic of China
| | - Ping-Ting Huang
- Department of Respiratory Medicine, Affiliated Union Hospital of Fujian Medical University, No.29 Xinquan Road, Fuzhou, 350001, Fujian, People's Republic of China
| | - Yu-Qing Chen
- Department of Respiratory Medicine, Affiliated Union Hospital of Fujian Medical University, No.29 Xinquan Road, Fuzhou, 350001, Fujian, People's Republic of China
| | - Li-Min Chen
- Department of Respiratory Medicine, Affiliated Union Hospital of Fujian Medical University, No.29 Xinquan Road, Fuzhou, 350001, Fujian, People's Republic of China.
| |
Collapse
|
3
|
Xiang Y, Duan Y, Peng Z, Huang H, Ding W, Chen E, Liu Z, Dou C, Li J, Ou J, Wan Q, Yang B, He Z. Microparticles from Hyperphosphatemia-Stimulated Endothelial Cells Promote Vascular Calcification Through Astrocyte-Elevated Gene-1. Calcif Tissue Int 2022; 111:73-86. [PMID: 35195734 DOI: 10.1007/s00223-022-00960-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 02/08/2022] [Indexed: 12/13/2022]
Abstract
Endothelial microparticles (EMPs) can be released in chronic kidney disease (CKD). Plasma concentration of high inorganic phosphate (HP) is considered as a decisive determinant of vascular calcification in CKD. We therefore explored the role of HP-induced EMPs (HP-EMPs) in the vascular calcification and its potential mechanism. We observed the shape of HP-EMPs captured by vascular smooth muscle cells (VSMCs) dynamically changed from rare dots, rosettes, to semicircle or circle. Our results demonstrated that HP-EMPs could directly promote VSMC calcification, or accelerate HP-induced calcification through signal transducers and activators of transcription 3 (STAT3)/bone morphogenetic protein-2 (BMP2) signaling pathway. AEG-1 activity was increased through HP-EMPs-induced VSMC calcification, in arteries from uremic rats, or from uremic rats treated with HP-EMPs. AEG-1 deficiency blocked, whereas AEG-1 overexpression exacerbated, the calcium deposition of VSMCs. AEG-1, a target of miR-153-3p, could be suppressed by agomiR-153-3p. Notably, VSMC-specific enhance of miR-153-3p by tail vein injection of aptamer-agomiR-153-3p decreased calcium deposition in both uremia rats treated with HP-EMPs or not. HP-EMPs could directly induce VSMCs calcification and accelerate Pi-induced calcification, and AEG-1 may act as crucial regulator of HP-EMPs-induced vascular calcification. This study sheds light on the therapeutic agents that influence HP-EMPs production or AEG-1 activity, which may be of benefit to treat vascular calcification.
Collapse
Affiliation(s)
- Yazhou Xiang
- Department of Nephrology, The First Affiliated Hospital, Hengyang Medical School, University of South China, 69 Chuanshan Road, Hengyang, Hunan, 421001, People's Republic of China
| | - Yingjie Duan
- Department of Nephrology, The First Affiliated Hospital, Hengyang Medical School, University of South China, 69 Chuanshan Road, Hengyang, Hunan, 421001, People's Republic of China
| | - Zhong Peng
- Department of Gastroenterology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Hong Huang
- Institute of Clinical Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Wenjun Ding
- Institute of Clinical Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - En Chen
- Clinical Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Zilong Liu
- Department of Stomatology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Chengyun Dou
- Clinical Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Jianlong Li
- Department of Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Jihong Ou
- Department of Nephrology, The First Affiliated Hospital, Hengyang Medical School, University of South China, 69 Chuanshan Road, Hengyang, Hunan, 421001, People's Republic of China
| | - Qingsong Wan
- Department of Nephrology, The First Affiliated Hospital, Hengyang Medical School, University of South China, 69 Chuanshan Road, Hengyang, Hunan, 421001, People's Republic of China
| | - Bo Yang
- Department of Nephrology, The First Affiliated Hospital, Hengyang Medical School, University of South China, 69 Chuanshan Road, Hengyang, Hunan, 421001, People's Republic of China
| | - Zhangxiu He
- Department of Nephrology, The First Affiliated Hospital, Hengyang Medical School, University of South China, 69 Chuanshan Road, Hengyang, Hunan, 421001, People's Republic of China.
| |
Collapse
|
4
|
Zhao X, Sun Y, Sun X, Li J, Shi X, Liang Z, Ma Y, Zhang X. AEG-1 Knockdown Sensitizes Glioma Cells to Radiation Through Impairing Homologous Recombination Via Targeting RFC5. DNA Cell Biol 2021; 40:895-905. [PMID: 34042508 DOI: 10.1089/dna.2020.6287] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Radiotherapy is the most important adjuvant treatment for glioma; however, radioresistance is the major cause for inevitable recurrence and poor survival of glioma patients. Thus, this study aims to investigate the effect of astrocyte elevated gene-1 (AEG-1) on the radiosensitivity of glioma cells. Immunohistochemistry assay found that AEG-1 was generally overexpressed in glioma tissues and was correlated with poor clinicopathological features of glioma patients. AEG-1 knockdown inhibited proliferation of glioma cells. And γ-H2AX foci assay, colony formation assay, and flow cytometry analysis demonstrated that AEG-1 depletion enhanced radiosensitivity and promoted apoptosis as well as cell cycle arrest in G2 phase of glioma cells treated by ionizing radiation. Moreover, replication factor C5 (RFC5) was screened as the target of AEG-1 by using Affymetrix human gene expression array, and RFC5 expression was downregulated in AEG-1 knockdown glioma cells. Mechanistically, AEG-1 knockdown impaired homologous recombination repair activity induced by radiation through inhibiting RFC5 expression. Furthermore, the Kaplan-Meier analysis and multivariate Cox regression analysis indicated that high levels of AEG-1 and RFC5 were related to poor prognosis of glioma patients treated with radiotherapy. Taken together, our findings indicate that AEG-1 may serve as a reliable radiosensitizing target for glioma radiotherapy.
Collapse
Affiliation(s)
- Xu Zhao
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yuchen Sun
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xuanzi Sun
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jing Li
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xiaobo Shi
- Department of Radiation Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zhinan Liang
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yuan Ma
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xiaozhi Zhang
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
5
|
Manna D, Sarkar D. Multifunctional Role of Astrocyte Elevated Gene-1 (AEG-1) in Cancer: Focus on Drug Resistance. Cancers (Basel) 2021; 13:cancers13081792. [PMID: 33918653 PMCID: PMC8069505 DOI: 10.3390/cancers13081792] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 03/31/2021] [Accepted: 04/04/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Chemotherapy is a major mode of treatment for cancers. However, cancer cells adapt to survive in stressful conditions and in many cases, they are inherently resistant to chemotherapy. Additionally, after initial response to chemotherapy, the surviving cancer cells acquire new alterations making them chemoresistant. Genes that help adapt the cancer cells to cope with stress often contribute to chemoresistance and one such gene is Astrocyte elevated gene-1 (AEG-1). AEG-1 levels are increased in all cancers studied to date and AEG-1 contributes to the development of highly aggressive, metastatic cancers. In this review, we provide a comprehensive description of the mechanism by which AEG-1 augments tumor development with special focus on its ability to regulate chemoresistance. We also discuss potential ways to inhibit AEG-1 to overcome chemoresistance. Abstract Cancer development results from the acquisition of numerous genetic and epigenetic alterations in cancer cells themselves, as well as continuous changes in their microenvironment. The plasticity of cancer cells allows them to continuously adapt to selective pressures brought forth by exogenous environmental stresses, the internal milieu of the tumor and cancer treatment itself. Resistance to treatment, either inherent or acquired after the commencement of treatment, is a major obstacle an oncologist confronts in an endeavor to efficiently manage the disease. Resistance to chemotherapy, chemoresistance, is an important hallmark of aggressive cancers, and driver oncogene-induced signaling pathways and molecular abnormalities create the platform for chemoresistance. The oncogene Astrocyte elevated gene-1/Metadherin (AEG-1/MTDH) is overexpressed in a diverse array of cancers, and its overexpression promotes all the hallmarks of cancer, such as proliferation, invasion, metastasis, angiogenesis and chemoresistance. The present review provides a comprehensive description of the molecular mechanism by which AEG-1 promotes tumorigenesis, with a special emphasis on its ability to regulate chemoresistance.
Collapse
|
6
|
AlMourgi M, Alzahrani R, Elsawy WH, Gharib AF, El Askary A, Amin HS, Raafat N. Astrocyte elevated gene-1: A potential molecular non-invasive marker for prognosis of esophageal cancer patients. Meta Gene 2020. [DOI: 10.1016/j.mgene.2020.100812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
7
|
Zhang Y, Zhao F. MicroRNA‑758 inhibits tumorous behavior in tongue squamous cell carcinoma by directly targeting metadherin. Mol Med Rep 2019; 19:1883-1890. [PMID: 30628702 DOI: 10.3892/mmr.2019.9805] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 11/30/2018] [Indexed: 11/05/2022] Open
Abstract
Numerous microRNAs (miRNAs) are dysregulated in tongue squamous cell carcinoma (TSCC), and their dysregulation has been demonstrated to have a strong correlation with TSCC progression via regulation of their targets. Therefore, miRNAs have potential use in the diagnosis and treatment of patients with TSCC. In the present study, miRNA‑758 (miR‑758) expression in TSCC tissues and cell lines was detected through reverse transcription‑quantitative polymerase chain reaction, and the effects of miR‑758 on TSCC cell proliferation and invasion were investigated by using Cell Counting kit‑8 and Transwell invasion assays. A luciferase reporter assay was performed to determine the target interaction between miR‑758 and metadherin (MTDH) in TSCC cells. The results revealed that miR‑758 was downregulated in TSCC tissues and cell lines. miR‑758 overexpression restricted the proliferation and invasion of TSCC cells. Additionally, MTDH was verified as a direct target gene of miR‑758 in TSCC cells. Furthermore, MTDH was observed to be upregulated in TSCC tissues, and the upregulation of MTDH was inversely correlated with miR‑758 expression. Moreover, restored MTDH expression significantly counteracted the suppressive effects of miR‑758 overexpression on TSCC cells. These results suggested that miR‑758 may prevent TSCC progression and development by directly targeting MTDH, thereby providing evidence that miR‑758 is a novel therapeutic target for the treatment of patients with TSCC.
Collapse
Affiliation(s)
- Yulan Zhang
- Department of Stomatology, Affiliated Hospital of Inner Mongolia University for The Nationalities, Tongliao, Inner Mongolia 028000, P.R. China
| | - Fuquan Zhao
- Department of Cardiovascular Intervention, Affiliated Hospital of Inner Mongolia University for The Nationalities, Tongliao, Inner Mongolia 028000, P.R. China
| |
Collapse
|
8
|
Yang X, Song S. Silencing of Astrocyte elevated gene-1 (AEG-1) inhibits proliferation, migration, and invasiveness, and promotes apoptosis in pancreatic cancer cells. Biochem Cell Biol 2018; 97:165-175. [PMID: 30359541 DOI: 10.1139/bcb-2018-0181] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
To investigate the role of Astrocyte elevated gene-1 (AEG-1) in the development and progress of pancreatic cancer, short hairpin RNA (shRNA) was inserted into the RNA interference vector to knock-down the endogenous AEG-1 in two pancreatic cancer cell lines: AsPC-1 and PANC-1. Our results showed that silencing of AEG-1 suppressed the proliferation, colony formation ability, and cell stemness of AsPC-1 and PANC-1 cells, and inhibited their G1-to-S phase transition. Results from apoptosis assay showed that knock-down of AEG-1 led to cell apoptosis. The expression of anti-apoptotic Bcl-2 was downregulated and that of the pro-apoptotic Bax and cleaved caspase-3 was upregulated in AEG-1-silenced pancreatic cancer cells. Further, the capability of AEG-1-silenced cells to migrate and to invade through the Matrigel-coated membrane was weaker, and the expression of matrix metallopeptidase 2 (MMP-2) and MMP-9 were decreased. Moreover, the AKT-β-catenin signaling pathway was inhibited in the cells with knock-down of AEG-1. In addition, the growth of xenograft tumors formed by AsPC-1 and PANC-1 cells was suppressed by AEG-1 shRNA. In conclusion, our study demonstrates that pancreatic cancer cells require AEG-1 to maintain their survival and metastasis, suggesting AEG-1 as a potential target for the treatment of pancreatic cancers.
Collapse
Affiliation(s)
- Xing Yang
- Department of Pancreatobiliary Surgery, The First Affiliated Hospital of China Medical University, Shenyang 110001, People's Republic of China.,Department of Pancreatobiliary Surgery, The First Affiliated Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Shaowei Song
- Department of Pancreatobiliary Surgery, The First Affiliated Hospital of China Medical University, Shenyang 110001, People's Republic of China.,Department of Pancreatobiliary Surgery, The First Affiliated Hospital of China Medical University, Shenyang 110001, People's Republic of China
| |
Collapse
|
9
|
Hui X, Zhang S, Wang Y. miR‑454‑3p suppresses cell migration and invasion by targeting CPEB1 in human glioblastoma. Mol Med Rep 2018; 18:3965-3972. [PMID: 30106109 DOI: 10.3892/mmr.2018.9386] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 06/28/2018] [Indexed: 11/05/2022] Open
Abstract
MicroRNAs (miRNA/miRs) serve crucial roles in the progression of human glioblastoma (GBM); however, the exact regulatory mechanisms of miRNAs in human GBM remain unclear. The present study aimed to investigate the roles of miR‑454‑3p in human GBM. Reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) analysis was performed to examine the expression of miR‑454‑3p in glioma tissues and adjacent tissues. Human GBM cell lines (LN‑229, A172 and GL15) and a normal human astrocyte cells (HA1800) were used for analysis. In addition, RT‑qPCR and western blotting were applied for mRNA and protein expression analysis, respectively. The cell proliferation was measured using a Cell Counting kit‑8 assay. Furthermore, scratch and Transwell assays were employed for the analysis of cell migration and invasion. A luciferase reporter assay was used to verify the target of miR‑454‑3p. The results revealed that miR‑454‑3p was downregulated in the glioma tissues and GBM cell lines, including LN‑229, A172 and GL15. Additionally, the overexpression of miR‑454‑3p significantly suppressed the proliferation, migration and invasion of LN‑229 cells. Furthermore, cytoplasmic polyadenylation element‑binding protein 1 (CPEB1) was confirmed as a direct target of miR‑454‑3p. These findings indicated that the overexpression of miR‑454‑3p inhibited cell proliferation, migration and invasion by downregulating CPEB1. Therefore, miR‑454‑3p may act as a tumor suppressor and represent an effective therapeutic strategy in GBM.
Collapse
Affiliation(s)
- Xiaobo Hui
- Department of Neurosurgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Shiming Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Yanping Wang
- Department of Neurosurgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| |
Collapse
|
10
|
Long J, Menggen Q, Wuren Q, Shi Q, Pi X. Long Noncoding RNA Taurine-Upregulated Gene1 (TUG1) Promotes Tumor Growth and Metastasis Through TUG1/Mir-129-5p/Astrocyte-Elevated Gene-1 (AEG-1) Axis in Malignant Melanoma. Med Sci Monit 2018; 24:1547-1559. [PMID: 29543785 PMCID: PMC5866625 DOI: 10.12659/msm.906616] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 10/10/2017] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Malignant melanoma is a class of malignant tumors derived from melanocytes. lncRNAs have been considered as pro-/anti-tumor factors in progression of cancers. The function of lncRNA TUG1 on growth of melanoma was investigated in this study. MATERIAL AND METHODS The TUG1 and miR-129-5p expression were examined via qRT-PCR. The protein expression was investigated by Western blotting assay. Luciferase reporter assay was used to assess if lncRNA TUG1 can bind to miR-129-5p and if miR-129-5p can target AEG1 mRNA. CCK-8 and apoptosis assay were used to detect cell growth and apoptosis. The metastasis of melanoma cells was detected by wound-healing and Transwell assays. The effects of TUG1 on growth of melanoma in vivo and cell chemoresistance were investigated via xenograft animal experiment and CCK-8 assay. RESULTS The expression of TUG1 and AEG1 was elevated and the miR-129-5p level was decreased in melanoma specimens and cell lines. Downregulation of either TUG1 or AEG1 suppressed cell growth and metastasis. miR-129-5p can bind directly to AEG1 and TUG1 can directly sponge miR-129-5p. Inhibition of TUG1 expression suppressed the expression of Bcl-2, MMP-9, and cyclin D1, and raised the level of cleaved caspase3 by modulating AEG1 level in melanoma cells. Inhibition of TUG1 reduced the growth of tumors in vivo and improved the chemosensitivity of A375 cells to cisplatin and 5-FU. CONCLUSIONS Reduction of TUG1 level suppressed cell growth and metastasis by regulating AEG1 expression mediated by targeting miR-129-5p. Suppression of lnc TUG1 may be a promising therapeutic strategy in the treatment of malignant melanoma.
Collapse
Affiliation(s)
- Jianwen Long
- Department of Dermatology, First Clinical Medicine School, Hubei University of Chinese Medicine, Wuhan, Hubei, P.R. China
| | - Qiqige Menggen
- Department of Dermatology, Mongolian Medicine Hospital of Bortala Mongolia Autonomous Prefecture, Bola, Bortala, P.R. China
| | - Qimige Wuren
- Department of Dermatology, Mongolian Medicine Hospital of Bortala Mongolia Autonomous Prefecture, Bola, Bortala, P.R. China
| | - Quan Shi
- Department of Dermatology, Hubei Provincial Academy of Traditional Chinese Medicine, Wuhan, Hubei, P.R. China
- Department of Dermatology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, Hubei, P.R. China
| | - Xianming Pi
- Department of Dermatology, Hubei Provincial Academy of Traditional Chinese Medicine, Wuhan, Hubei, P.R. China
- Department of Dermatology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, Hubei, P.R. China
| |
Collapse
|
11
|
Chen L, Ma G, Cao X, An X, Liu X. MicroRNA-331 Inhibits Proliferation and Invasion of Melanoma Cells by Targeting Astrocyte-Elevated Gene-1. Oncol Res 2018; 26:1429-1437. [PMID: 29510779 PMCID: PMC7844642 DOI: 10.3727/096504018x15186047251584] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Melanoma is characterized by aggressive invasion, early metastasis, and resistance to existing chemotherapeutic agents. Accumulated studies have reported that microRNA (miRNA) is a potentially robust molecular tool for developing future therapeutic technologies. Therefore, examining the expression patterns, biological roles, and associated mechanisms of cancer-related miRNAs in melanoma is essential for developing novel therapeutic targets for patients with this disease. In this study, miRNA-331 (miR-331) was underexpressed in melanoma tissues and cell lines. Functional assays revealed that the enforced expression of miR-331 inhibited cell proliferation and invasion. In addition, astrocyte-elevated gene-1 (AEG-1) was identified as a novel target of miR-331 through bioinformatics analysis, reverse transcription quantitative polymerase chain reaction analysis, Western blot analysis, dual-luciferase reporter assay, and Spearman’s correlation analysis. Furthermore, reintroduction of AEG-1 partially abrogated the inhibitory effects of miR-331 overexpression on the proliferation and invasion of melanoma cells. Moreover, miR-331 suppressed the activation of the PTEN/AKT signaling pathway in melanoma by inhibiting AEG-1. In short, miR-331 may play tumor-suppressive roles in melanoma by directly targeting AEG-1 and regulating the PTEN/AKT signaling pathway, suggesting that miR-331 could be investigated as a therapeutic strategy for patients with this malignancy.
Collapse
Affiliation(s)
- Li Chen
- Department of Dermatology, Heilongjiang Provincial Hospital, Harbin, Heilongjiang, P.R. China
| | - Guozhang Ma
- Department of Dermatology, Heilongjiang Provincial Hospital, Harbin, Heilongjiang, P.R. China
| | - Xiaohui Cao
- Department of Dermatology, Heilongjiang Provincial Hospital, Harbin, Heilongjiang, P.R. China
| | - Xiaoxia An
- Department of Dermatology, Heilongjiang Provincial Hospital, Harbin, Heilongjiang, P.R. China
| | - Xiguang Liu
- Department of Dermatology, Heilongjiang Provincial Hospital, Harbin, Heilongjiang, P.R. China
| |
Collapse
|
12
|
Zhang Y, Wang Y, Wang J. MicroRNA-584 inhibits cell proliferation and invasion in non-small cell lung cancer by directly targeting MTDH. Exp Ther Med 2017; 15:2203-2211. [PMID: 29434826 DOI: 10.3892/etm.2017.5624] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 11/21/2017] [Indexed: 12/14/2022] Open
Abstract
Lung cancer is the third most frequent human malignant tumour and the leading cause of cancer-associated mortality worldwide. Emerging lines of evidence have demonstrated that microRNAs (miRNAs) are upregulated or downregulated in non-small cell lung cancer (NSCLC), and this phenomenon is involved in the regulation of various processes during tumorigenesis and progression, including tumour groWTh, apoptosis, cell invasion, and tumour metastasis. Therefore, understanding the molecular mechanism that associates abnormally expressed miRNAs with NSCLC formation and development may lead to the identification of novel diagnostic, and therapeutic targets for patients with NSCLC. miRNA-584 (miR-584) functions as a tumour suppressor in several types of cancer. However, the expression pattern, detailed biological function and underlying molecular mechanism of miR-584 in NSCLC remain unclear. Therefore, the present study detected the expression of miR-584 in NSCLC, investigated its role in NSCLC cells and determined its underlying molecular mechanism. In the current study, it was demonstrated that miR-584 was downregulated in NSCLC tissues and cell lines. Low miR-584 expression was correlated with tumour size, tumour node metastasis stage and distant metastasis. Overexpression of miR-584 inhibited cell proliferation and invasion in NSCLC. Additionally, metadherin was identified as a direct target gene of miR-584 in NSCLC as confirmed by a series of experiments. Moreover, upregulation of miR-584 was involved in the regulation of the phosphatase and tensin homolog/Akt serine/threonine kinase signalling pathway in NSCLC. Thus, miR-584 may serve as a tumor-suppressor, and the results of the present study provide a reference for future research into the potential mechanisms underlying NSCLC progression.
Collapse
Affiliation(s)
- Yixiang Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Yanjun Wang
- Department of Thoracic Surgery, Dalian University Affiliated Xinhua Hospital, Dalian, Liaoning 116011, P.R. China
| | - Jinguang Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| |
Collapse
|
13
|
Li H, Zhao J. let-7d suppresses proliferation and invasion and promotes apoptosis of meningioma by targeting AEG-1. Onco Targets Ther 2017; 10:4895-4904. [PMID: 29070952 PMCID: PMC5640403 DOI: 10.2147/ott.s141008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND let-7d has been indicated to act as a tumor suppressor in various cancers. However, the function and molecular mechanism of let-7d in meningioma progression have not been elucidated. MATERIALS AND METHODS Quantitative real-time polymerase chain reaction (qRT-PCR) was performed to detect the expression levels of let-7d and AEG-1 mRNA in meningioma tissues and cell lines. The protein level of AEG-1 was measured by Western blot analysis. MTT assay, Transwell invasion assay and flow cytometry analysis were carried out to determine the proliferation, invasion and apoptosis of IOMM-Lee and CH-157MN cells, respectively. Target gene of let-7d was verified by luciferase reporter analysis. RESULTS let-7d expression was downregulated, and AEG-1 expression was upregulated in meningioma tumor tissues. let-7d overexpression suppressed proliferation and invasion and induced apoptosis in IOMM-Lee and CH-157MN cells. Moreover, AEG-1 was a direct target of let-7d. Restoration of AEG-1 expression reversed let-7d-mediated suppression of the proliferation and invasion and let-7d-induced apoptosis in IOMM-Lee and CH-157MN cells. CONCLUSION let-7d repressed proliferation and invasion and promoted apoptosis of meningioma cells by targeting AEG-1. The present study provided a better understanding of the meningioma pathogenesis and a promising therapeutic target for meningioma patients.
Collapse
Affiliation(s)
- Hui Li
- Department of Neurology, Xinxiang Central Hospital, Xinxiang, China
| | - Jianmin Zhao
- Department of Neurology, Xinxiang Central Hospital, Xinxiang, China
| |
Collapse
|
14
|
In vitro and in vivo anti-uveal melanoma activity of JSL-1, a novel HDAC inhibitor. Cancer Lett 2017; 400:47-60. [DOI: 10.1016/j.canlet.2017.04.028] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 04/08/2017] [Accepted: 04/14/2017] [Indexed: 12/27/2022]
|