1
|
Li Z, Xu Y, Hu Y, He Z, Zhang Z, Zhou J, Zhou T, Wang H. The critical role of SETDB1-mediated CCND1/PI3K/AKT pathway via p53-RS di-methylation at K370 in the proliferation of WRL68 cells induced by nicotine. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 282:116686. [PMID: 38971100 DOI: 10.1016/j.ecoenv.2024.116686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/15/2024] [Accepted: 07/02/2024] [Indexed: 07/08/2024]
Abstract
Constituents of cigarette smoke are known to be carcinogens. Additionally, there is mounting evidence that the liver is an organ susceptible to tobacco carcinogenicity. Nicotine, the primary constituent of tobacco, plays a role in cancer progression. In our previous study, it was found that nicotine enhances the proliferation of a human normal fetal hepatic (WRL68) cell due to the activation of p53 mutation at Ser249 (p53-RS)/STAT1/CCND1 signaling pathway. Here, we further elucidated the mechanism of regulating this pathway. Firstly, dose-dependent increase of SETDB1 protein level in WRL68 cells upon exposure to nicotine (1.25, 2.5, and 5 μM), significantly enhanced cellular proliferation. In addition, the upregulation of SETDB1 protein was necessary for the nuclear translocation of p53-RS to establish a ternary complex with STAT1 and SETDB1, which facilitated p53-RS di-methylation at K370 (p53-RS/K370me2). After that, the activation of CCND1/PI3K/AKT pathway was initiated when STAT1 stability was enhanced by p53-RS/K370me2, ultimately resulting in cell proliferation. Altogether, the study revealed that the increase in SETDB1 expression could potentially have a significant impact on the activation of CCND1/PI3K/AKT pathway through p53-RS/K370me2, leading to the proliferation of WRL68 cells induced by nicotine, which could contribute to hepatocellular carcinoma for smokers. Besides, the results of this study provided a foundation for the development of anticancer therapies for cancers associated with tobacco use.
Collapse
Affiliation(s)
- Zihan Li
- School of Public Health, Jiangxi Medical College, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, PR China; Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, PR China; Chongqing Research Institute of Nanchang University, Tai Bai Road, Tongnan, Chongqing 402679, PR China
| | - Yuqin Xu
- School of Public Health, Jiangxi Medical College, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, PR China; Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, PR China; Chongqing Research Institute of Nanchang University, Tai Bai Road, Tongnan, Chongqing 402679, PR China
| | - Yuxin Hu
- School of Public Health, Jiangxi Medical College, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, PR China; Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, PR China; Chongqing Research Institute of Nanchang University, Tai Bai Road, Tongnan, Chongqing 402679, PR China
| | - Zihan He
- School of Public Health, Jiangxi Medical College, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, PR China; Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, PR China; Chongqing Research Institute of Nanchang University, Tai Bai Road, Tongnan, Chongqing 402679, PR China
| | - Zhongwei Zhang
- School of Public Health, Jiangxi Medical College, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, PR China; Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, PR China; Chongqing Research Institute of Nanchang University, Tai Bai Road, Tongnan, Chongqing 402679, PR China
| | - Jianming Zhou
- School of Public Health, Jiangxi Medical College, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, PR China; Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, PR China; Chongqing Research Institute of Nanchang University, Tai Bai Road, Tongnan, Chongqing 402679, PR China
| | - Tong Zhou
- School of Public Health, Jiangxi Medical College, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, PR China; Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, PR China; Chongqing Research Institute of Nanchang University, Tai Bai Road, Tongnan, Chongqing 402679, PR China
| | - Huai Wang
- School of Public Health, Jiangxi Medical College, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, PR China; Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, PR China; Chongqing Research Institute of Nanchang University, Tai Bai Road, Tongnan, Chongqing 402679, PR China.
| |
Collapse
|
2
|
Qin S, Gao K, Tian Z. Comprehensive characterization of differential glycation in hepatocellular carcinoma using tissue proteomics with stable isotopic labeling. Anal Bioanal Chem 2024; 416:4531-4541. [PMID: 38922433 DOI: 10.1007/s00216-024-05392-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 05/17/2024] [Accepted: 06/06/2024] [Indexed: 06/27/2024]
Abstract
Glycation is a non-enzymatic posttranslational modification coming from the reaction between reducing sugars and free amino groups in proteins, where early glycation products (fructosyl-lysine, FL) and advanced glycation end products (AGEs) are formed. The occurrence of glycation and accumulation of AGEs have been closely associated with hepatocellular carcinoma (HCC). Here, we reported the characterization of differential glycation in HCC using tissue proteomics with stable isotopic labeling; early glycation-modified peptides were enriched with boronate affinity chromatography (BAC), and AGEs-modified peptides were fractionated with basic reversed-phase separation. By this integrated approach, 3717 and 1137 early and advanced glycated peptides corresponding to 4007 sites on 1484 proteins were identified with a false discovery rate (FDR) of no more than 1%. One hundred fifty-five sites were modified with both early and advanced end glycation products. Five early and 7 advanced glycated peptides were quantified to be differentially expressed in HCC tissues relative to paired adjacent tissues. Most (8 out of 10) of the proteins corresponding to the differential glycated peptides have previously been reported with dysregulation in HCC. The results together may deepen our knowledge of glycation as well as provide insights for therapeutics.
Collapse
Affiliation(s)
- Shanshan Qin
- School of Chemical Science & Engineering, Shanghai Key Laboratory of Chemical Assessment and Sustainability, Tongji University, Shanghai, 200092, China
| | - Ke Gao
- Department of Liver Surgery and Transplantation, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhixin Tian
- School of Chemical Science & Engineering, Shanghai Key Laboratory of Chemical Assessment and Sustainability, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
3
|
Zhao J, Huo Q, Zhang J, Sun K, Guo J, Cheng F, Hu X, Xu Q. UCHL3 promotes hepatocellular carcinoma progression by stabilizing EEF1A1 through deubiquitination. Biol Direct 2024; 19:53. [PMID: 38965582 PMCID: PMC11225194 DOI: 10.1186/s13062-024-00495-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 06/23/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) ranks as the second leading cause of global cancer-related deaths and is characterized by a poor prognosis. Eukaryotic translation elongation factor 1 alpha 1 (EEF1A1) have been proved to play important roles in various human cancers, whereas the deubiquitination of EEF1A1 was poorly understood. METHODS The binding and regulatory relationship between Ubiquitin carboxyl-terminal hydrolase L3 (UCHL3) and EEF1A1 was validated using clinical tissue samples, reverse transcription quantitative real-time fluorescence quantitative PCR (RT-qPCR), Western blotting, co-immunoprecipitation, and immunofluorescence, as well as ubiquitin detection and cyclohexamide tracking experiments. Finally, the impact of the UCHL3/EEF1A1 axis on HCC malignant behavior was analyzed through functional experiments and nude mouse models. RESULTS UCHL3 was found to have a high expression level in HCC tissues. Tissue samples from 60 HCC patients were used to evaluate the correlation between UCHL3 and EEF1A1. UCHL3 binds to EEF1A1 through the lysine site, which reduces the ubiquitination level of EEF1A1. Functional experiments and nude mouse models have demonstrated that the UCHL3/EEF1A1 axis promotes the migration, stemness, and drug resistance of HCC cells. Reducing the expression of EEF1A1 can reverse the effect of UCHL3 on the malignant behavior of HCC cells. CONCLUSION Our findings revealed that UCHL3 binds and stabilizes EEF1A1 through deubiquitination. UCHL3 and EEF1A1 formed a functional axis in facilitating the malignant progression of HCC, proving new insights for the anti-tumor targeted therapy for HCC.
Collapse
Affiliation(s)
- Jie Zhao
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Qiang Huo
- Department of General Surgery, Zhoushan Dinghai Central Hospital (Dinghai District of Zhejiang Provincial People's Hospital), Zhoushan, China
| | - Ji Zhang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Kexiang Sun
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Jinhui Guo
- Qingdao Medical College, Qingdao University, Qingdao, China
| | - Feng Cheng
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China.
| | - Xiaoge Hu
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, China.
| | - Qiuran Xu
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, China.
| |
Collapse
|
4
|
Zhang W, Wang J, Shan C. The eEF1A protein in cancer: Clinical significance, oncogenic mechanisms, and targeted therapeutic strategies. Pharmacol Res 2024; 204:107195. [PMID: 38677532 DOI: 10.1016/j.phrs.2024.107195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/09/2024] [Accepted: 04/22/2024] [Indexed: 04/29/2024]
Abstract
Eukaryotic elongation factor 1A (eEF1A) is among the most abundant proteins in eukaryotic cells. Evolutionarily conserved across species, eEF1A is in charge of translation elongation for protein biosynthesis as well as a plethora of non-translational moonlighting functions for cellular homeostasis. In malignant cells, however, eEF1A becomes a pleiotropic driver of cancer progression via a broad diversity of pathways, which are not limited to hyperactive translational output. In the past decades, mounting studies have demonstrated the causal link between eEF1A and carcinogenesis, gaining deeper insights into its multifaceted mechanisms and corroborating its value as a prognostic marker in various cancers. On the other hand, an increasing number of natural and synthetic compounds were discovered as anticancer eEF1A-targeting inhibitors. Among them, plitidepsin was approved for the treatment of multiple myeloma whereas metarrestin was currently under clinical development. Despite significant achievements in these two interrelated fields, hitherto there lacks a systematic examination of the eEF1A protein in the context of cancer research. Therefore, the present work aims to delineate its clinical implications, molecular oncogenic mechanisms, and targeted therapeutic strategies as reflected in the ever expanding body of literature, so as to deepen mechanistic understanding of eEF1A-involved tumorigenesis and inspire the development of eEF1A-targeted chemotherapeutics and biologics.
Collapse
Affiliation(s)
- Weicheng Zhang
- The State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, People's Republic of China.
| | - Jiyan Wang
- The State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, People's Republic of China
| | - Changliang Shan
- The State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, People's Republic of China.
| |
Collapse
|
5
|
Chen J, Ning D, Du P, Liu Q, Mo J, Liang H, Zhang W, Zhang M, Jiang L, Zhang B, Chen X. USP11 potentiates HGF/AKT signaling and drives metastasis in hepatocellular carcinoma. Oncogene 2024; 43:123-135. [PMID: 37973952 DOI: 10.1038/s41388-023-02847-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 09/06/2023] [Accepted: 09/18/2023] [Indexed: 11/19/2023]
Abstract
USP11 is a member of the ubiquitin-specific protease family and plays a crucial role in tumor progression in various cancers. However, the precise mechanism by which USP11 promotes EMT and metastasis in hepatocellular carcinoma (HCC) is not fully understood. In this study, we demonstrated that the USP11 expression was dramatically upregulated in HCC tissues and cell lines. Increased USP11 expression was closely associated with tumor number, vascular invasion, and poor prognosis. Functional experiments demonstrated that USP11 markedly promoted metastasis and EMT in HCC via induction of the transcription factor Snail. Mechanistically, USP11 interacted with and deubiquitinated eEF1A1 on Lys439, thereby inhibiting its ubiquitin-mediated degradation. Subsequently, the elevated expression of eEF1A1 resulted in its binding to SP1, which in turn drove the binding of SP1 to its target HGF gene promoter to increase its transcription. This led to an enhanced expression of HGF and the activation of the downstream PI3K/AKT signaling pathway. We demonstrated that USP11 promotes EMT and metastasis in HCC via eEF1A1/SP1/HGF dependent-EMT. Our findings suggest that the USP11/ eEF1A1/SP1/HGF axis contributes to metastasis in HCC, and therefore, could be considered as a potential therapeutic target for the treatment of HCC.
Collapse
Affiliation(s)
- Jin Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Clinical Medicine Research Center for Hepatic Surgery of Hubei Province; Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, Hubei, 430030, PR China
| | - Deng Ning
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Clinical Medicine Research Center for Hepatic Surgery of Hubei Province; Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, Hubei, 430030, PR China
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Pengcheng Du
- Department of Biliary and Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Qiumeng Liu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Clinical Medicine Research Center for Hepatic Surgery of Hubei Province; Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, Hubei, 430030, PR China
| | - Jie Mo
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Clinical Medicine Research Center for Hepatic Surgery of Hubei Province; Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, Hubei, 430030, PR China
| | - Huifang Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Clinical Medicine Research Center for Hepatic Surgery of Hubei Province; Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, Hubei, 430030, PR China
| | - Wanguang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Clinical Medicine Research Center for Hepatic Surgery of Hubei Province; Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, Hubei, 430030, PR China
| | - Mingzhi Zhang
- Department of Cancer Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Li Jiang
- Department of Biliary and Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China.
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Clinical Medicine Research Center for Hepatic Surgery of Hubei Province; Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, Hubei, 430030, PR China.
| | - Xiaoping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Clinical Medicine Research Center for Hepatic Surgery of Hubei Province; Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, Hubei, 430030, PR China.
| |
Collapse
|
6
|
Yang F, Wu J, Zhao M, Zheng H, Suo J, Liu X, Zheng D. MicroRNA PC-3p-2869 Regulates Antler Growth and Inhibits Proliferation and Migration of Human Osteosarcoma and Chondrosarcoma Cells by Targeting CDK8, EEF1A1, and NTN1. Int J Mol Sci 2023; 24:10840. [PMID: 37446017 DOI: 10.3390/ijms241310840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/25/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
MicroRNAs (miRNAs) play a crucial role in maintaining the balance between the rapid growth and suppression of tumorigenesis during antler regeneration. This study investigated the role of a novel miRNA, PC-3p-2869 (miR-PC-2869), in antler growth and its therapeutic potential in human osteosarcoma and chondrosarcoma. Stem-loop RT-qPCR showed that miR-PC-2869 was expressed extensively in diverse layers of antler tissues. Overexpression of miR-PC-2869 suppressed the proliferation and migration of antler cartilage cells. Similarly, heterologous expression of miR-PC-2869 reduced the proliferation, colony formation, and migration of osteosarcoma cell line MG63 and U2OS and chondrosarcoma cell line SW1353. Moreover, 18 functional target genes of miR-PC-2869 in humans were identified based on the screening of the reporter library. Among them, 15 target genes, including CDK8, EEF1A1, and NTN1, possess conserved miR-PC-2869-binding sites between humans and red deer (Cervus elaphus). In line with this, miR-PC-2869 overexpression decreased the expression levels of CDK8, EEF1A1, and NTN1 in MG63, SW1353, and antler cartilage cells. As expected, the knockdown of CDK8, EEF1A1, or NTN1 inhibited the proliferation and migration of MG63, SW1353, and antler cartilage cells, demonstrating similar suppressive effects as miR-PC-2869 overexpression. Furthermore, we observed that CDK8, EEF1A1, and NTN1 mediated the regulation of c-myc and cyclin D1 by miR-PC-2869 in MG63, SW1353, and antler cartilage cells. Overall, our work uncovered the cellular functions and underlying molecular mechanism of antler-derived miR-PC-2869, highlighting its potential as a therapeutic candidate for bone cancer.
Collapse
Affiliation(s)
- Fan Yang
- Laboratory of Genetics and Molecular Biology, College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, China
| | - Jin Wu
- Laboratory of Genetics and Molecular Biology, College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, China
| | - Mindie Zhao
- Laboratory of Genetics and Molecular Biology, College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, China
| | - Han Zheng
- Biotechnology Program, Division of Biology and Medicine, Brown University, Providence, RI 02912, USA
| | - Jingyuan Suo
- Laboratory of Genetics and Molecular Biology, College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, China
| | - Xuedong Liu
- Laboratory of Genetics and Molecular Biology, College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, China
| | - Dong Zheng
- Laboratory of Genetics and Molecular Biology, College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, China
| |
Collapse
|
7
|
Luo BH, Huang JQ, Huang CY, Tian P, Chen AZ, Wu WH, Ma XM, Yuan YX, Yu L. Screening of Lymphoma Radiotherapy-Resistant Genes with CRISPR Activation Library. Pharmgenomics Pers Med 2023; 16:67-80. [PMID: 36743888 PMCID: PMC9897072 DOI: 10.2147/pgpm.s386085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 12/05/2022] [Indexed: 02/03/2023] Open
Abstract
Objective The objective of this study was to screen lymphoma radiotherapy-resistant genes using CRISPR activation (CRISPRa). Methods The Human CRISPRa library virus was packaged and then transfected into lymphoma cells to construct an activation library cell line, which was irradiated at the minimum lethal radiation dose to screen radiotherapy-resistant cells. Radiotherapy-resistant cell single-guide RNA (sgRNA) was first amplified by quantitative polymerase chain reaction (qPCR) in the coding region and then subject to next-generation sequencing (NGS) and bioinformatics analysis to screen radiotherapy-resistant genes. Certain radiotherapy-resistant genes were then selected to construct activated cell lines transfected with a single gene so as to further verify the relationship between gene expression and radiotherapy resistance. Results A total of 16 radiotherapy-resistant genes, namely, C20orf203, MTFR1, TAF1L, MYADM, NIPSNAP1, ZUP1, RASL11A, PSMB2, PSMA6, OR8H3, TMSB4Y, CD300LF, EEF1A1, ATP6AP1L, TRAF3IP2, and SNRNP35, were screened based on the NGS results and bioinformatics analysis of the radiotherapy-resistant cells. Activated cell lines transfected with a single gene were constructed using 10 radiotherapy-resistant genes. The qPCR findings showed that, when compared with the control group, the experimental group had significantly up-regulated mRNA expression of MTFR1, NIPSNAP1, ZUP1, PSMB2, PSMA6, EEF1A1, TMSB4Y and TAF1L (p < 0.05). No significant difference in the mRNA expression of AKT3 or TRAF3IP2 (p > 0.05) was found between the two groups (p > 0.05). Conclusion The 16 genes screened are potential lymphoma radiotherapy-resistant genes. It was initially determined that the high expression of 8 genes was associated with lymphoma radiotherapy resistance, and these genes could serve as the potential biomarkers for predicting lymphoma radiotherapy resistance or as new targets for therapy.
Collapse
Affiliation(s)
- Bi-Hua Luo
- Department of Hematology, Longyan First Hospital Affiliated Fujian Medical University, Fujian, People’s Republic of China
| | - Jian-Qing Huang
- Department of Hematology, Longyan First Hospital Affiliated Fujian Medical University, Fujian, People’s Republic of China
| | - Chun-Yu Huang
- Department of Hematology, Longyan First Hospital Affiliated Fujian Medical University, Fujian, People’s Republic of China
| | - Pan Tian
- Department of Hematology, Longyan First Hospital Affiliated Fujian Medical University, Fujian, People’s Republic of China
| | - Ai-Zhen Chen
- Department of Hematology, Longyan First Hospital Affiliated Fujian Medical University, Fujian, People’s Republic of China
| | - Wei-Hao Wu
- Department of Hematology, Longyan First Hospital Affiliated Fujian Medical University, Fujian, People’s Republic of China
| | - Xiao-Mei Ma
- Department of Hematology, Longyan First Hospital Affiliated Fujian Medical University, Fujian, People’s Republic of China
| | - Yue-Xing Yuan
- Department of Hematology, Longyan First Hospital Affiliated Fujian Medical University, Fujian, People’s Republic of China
| | - Lian Yu
- Department of Hematology, Longyan First Hospital Affiliated Fujian Medical University, Fujian, People’s Republic of China,Correspondence: Lian Yu, Department of Hematology, Longyan First Hospital Affiliated Fujian Medical University, No. 105 of Jiuyibei Road, Xinluo District, Fujian, 364000, People’s Republic of China, Tel +86 13859572936, Email
| |
Collapse
|
8
|
Liu S, Chen X, Huang K, Xiong X, Shi Y, Wang X, Pan X, Cong Y, Sun Y, Ge L, Xu J, Jia X. Long noncoding RNA RFPL1S-202 inhibits ovarian cancer progression by downregulating the IFN-β/STAT1 signaling. Exp Cell Res 2023; 422:113438. [PMID: 36435219 DOI: 10.1016/j.yexcr.2022.113438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 11/06/2022] [Accepted: 11/23/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND RFPL1S was first identified as one of the pseudogenes located in the intrachromosomal duplications within 22q12-13. Our previous study found that one of the predicted transcripts of lncRNA RFPL1S, ENST00000419368.1 (GRCh37/hg19), also named as RFPL1S-202 in Ensembl website, is significantly downregulated in the chemoresistant ovarian cancer cells. However, its function and underlying mechanism have not been studied. METHODS Quantitative Real-time PCR was used to analyze the expression. Cell Counting Kit-8, transwell, flow cytometry analysis and tail vein injected mouse model were used to test the function. RNA-sequencing, RNA pull down, western blot, ELISA and RNA-Binding Protein Immunoprecipitation were performed for studying the mechanism. 5' and 3' rapid amplification of complementary DNA ends were performed to analyze the full length of RFPL1S-202. RESULTS RFPL1S-202 is significantly downregulated in epithelial ovarian cancer tissues and cell lines. Gain- and loss-of-function study indicated that RFPL1S-202 could enhance cisplatin or paclitaxel in cytotoxicity, inhibit cell proliferation, invasion and migration of ovarian cancer cells in vitro, and inhibit the liver metastasis of ovarian cancer cells in vivo. Mechanistically, RFPL1S-202 could physically interact with DEAD-Box Helicase 3 X-linked (DDX3X) protein, and decrease the expression of p-STAT1 and the IFN inducible genes by increasing the m6A modification of IFNB1. RFPL1S-202 is a spliced and polyadenylated non-coding RNA with a full length of 1071 bp. CONCLUSIONS Our study suggested that the predicted lncRNA RFPL1S-202 exerts a tumor- suppressive function in oarian cancer chemoresistance and progression by interacting with DDX3X and down-regulating the IFN-β-STAT1 signaling pathway.
Collapse
Affiliation(s)
- Siyu Liu
- Department of Gynecology, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, 210004, China.
| | - Xiyi Chen
- Department of Gynecology, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, 210004, China
| | - Ke Huang
- Department of Gynecology, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, 210004, China
| | - Xueyou Xiong
- Department of Gynecology, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, 210004, China
| | - Yaqian Shi
- Department of Gynecology, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, 210004, China
| | - Xusu Wang
- Department of Gynecology, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, 210004, China
| | - Xinxing Pan
- Department of Gynecology, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, 210004, China
| | - Yu Cong
- Department of Gynecology, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, 210004, China
| | - Yu Sun
- Department of Gynecology, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, 210004, China
| | - Lili Ge
- Department of Gynecology, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, 210004, China.
| | - Juan Xu
- Department of Gynecology, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, 210004, China.
| | - Xuemei Jia
- Department of Gynecology, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, 210004, China.
| |
Collapse
|
9
|
Effects of SENP1-induced deSUMOylation of STAT1 on proliferation and invasion in nasopharyngeal carcinoma. Cell Signal 2023; 101:110530. [PMID: 36417976 DOI: 10.1016/j.cellsig.2022.110530] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/13/2022] [Accepted: 11/18/2022] [Indexed: 11/21/2022]
Abstract
Nasopharyngeal carcinoma (NPC) is the most common nasopharyngeal squamous cell carcinoma, and recurrence and metastasis are still difficult problems in its current treatment. This study aimed to investigate the effect of SUMO modification of STAT1 protein on the proliferation and invasion of NPC, and to reveal the underlying mechanism. Two gene expression profiles (GSE12452 and GSE53819) of 49 nasopharyngeal carcinomas and 28 normal controls were analyzed to identify differentially expressed genes. In total, 448 up-regulated genes and 622 down-regulated genes were identified. In addition, 16 SUMO-related molecules in the NPC dataset GSE102349 with survival data were analyzed, and it was found that the high expression of SENP1 and SENP2 was closely related to the poor prognosis of NPC. GO and GSEA analysis suggested that immune-related biological processes, IFN-γ-STAT signaling pathway and protein modification-related molecules were significantly enriched in NPC, resulting in poor survival prognosis. In order to verify the results of bioinformatics analysis and explore its underlying molecular mechanisms, western blot, Immunofluorescence, Immunoprecipitation and Immunohistochemistry are conducted in NPC cells, animals and clinical samples. SENP1 and STAT protein levels were increased in NPC tissues. SENP1 inhibited SUMOylation of STAT1, thereby promoting the protein level of STAT1 and the nuclear translocation. SENP1 promoted the proliferation and invasion of NPC by inducing STAT1. Overall, SENP1-induced deSUMOylation of STAT1, resulting in an increased proliferation and invasion of nasopharyngeal carcinoma.
Collapse
|
10
|
Chen F, Yang L, Peng X, Mao M, Liu X, Song J, Hu J. Histone deacetylase 2 regulates STAT1-dependent upregulation of atypical chemokine receptor 3 to induce M2 macrophage migration and immune escape in hepatocellular carcinoma. Mol Immunol 2022; 151:204-217. [PMID: 36179603 DOI: 10.1016/j.molimm.2022.09.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 09/05/2022] [Accepted: 09/07/2022] [Indexed: 10/31/2022]
Abstract
Atypical chemokine receptor 3 (ACKR3) has been linked to the tumor microenvironment. This work investigates the effects of ACKR3 and its regulatory molecules on the chemotactic migration of tumor-associated macrophages (TAMs) in hepatocellular carcinoma (HCC). RT-qPCR and western blot assays identified elevated ACKR3 and HDAC2 levels in HCC tissues and cells. Knockdown or overexpression of ACKR3 was induced in HCC cells through vectors of lentivirus plasmids, and then the conditioned medium of the HCC cells was collected to stimulate TAMs. The stimulated TAMs were co-cultured with CD3+ T cells. ACKR3 knockdown in HCC cells inhibited migration of TAMs and promoted M1 polarization, which restored the activity of T cells. Histone deacetylase 2 (HDAC2) recruited signal transducer and activator of transcription 1 (STAT1) to the ACKR3 promoter to activate ACKR3 transcription. HDAC2 silencing suppressed nuclear translocation of STAT1 and decreased ACKR3 expression. HDAC2 knockdown in HCC cells similarly suppressed TAM migration, promoted M1 polarization, and restored T cell function, but these changes were inversed by ACKR3 upregulation. HDAC2 or ACKR3 silencing weakened tumor growth and immune escape in mice. In conclusion, this study demonstrates that HDAC2 upregulates ACKR3 via STAT1 to induce migration of M2 macrophages and immune escape in HCC.
Collapse
Affiliation(s)
- Fu Chen
- The Seventh Department of General Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang 110000, Liaoning, PR China
| | - Liang Yang
- The Third Department of General Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang 110000, Liaoning, PR China
| | - Xueqiang Peng
- The Third Department of General Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang 110000, Liaoning, PR China
| | - Minghuan Mao
- Department of Urology Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang 110000, Liaoning, PR China
| | - Xiaodan Liu
- The Fifth Department of General Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang 110000, Liaoning, PR China
| | - Jianbo Song
- Interventional Department, the Fourth Affiliated Hospital of China Medical University, Shenyang 110000, Liaoning, PR China
| | - Jingyao Hu
- The Seventh Department of General Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang 110000, Liaoning, PR China.
| |
Collapse
|
11
|
Fan A, Zhao X, Liu H, Li D, Guo T, Zhang J, Duan L, Cheng H, Nie Y, Fan D, Zhao X, Lu Y. eEF1A1 promotes colorectal cancer progression and predicts poor prognosis of patients. Cancer Med 2022; 12:513-524. [PMID: 35607944 PMCID: PMC9844609 DOI: 10.1002/cam4.4848] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 04/17/2022] [Accepted: 04/18/2022] [Indexed: 01/26/2023] Open
Abstract
Colorectal cancer (CRC) is a major leading cause of cancer mortality worldwide in which dysregulated protein synthesis plays an etiologic role. The eukaryotic elongation factor 1 A1 (eEF1A1) exerts significant effects on protein synthesis by contributing to peptide chain extension. Whereas its role in CRC remains to be investigated. In this study, we found that the mRNA and protein levels of eEF1A1 were significantly upregulated in CRC cell lines and tissues. Elevated expression of eEF1A1 was correlated with shorter overall survival in 94 CRC patients. The inhibition of proliferation and cell cycle block were observed in CRC cells after eEF1A1 downregulation. Mechanistically, weighted gene correlation network analysis and further Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis suggested that mitogen-activated protein kinases (MAPKs) signaling pathways were significantly enriched in high-eEF1A1 expression group, and the levels of phosphorylated p38/JNK/ERK MAPK were dramatically decreased after eEF1A1 downregulation. Overexpression of eEF1A1 in CRC correlated with a poor prognosis. Collectively, this study determined the oncogenic role of eEF1A1 in CRC proliferation and tumorigenesis. eEF1A1 might be a promising therapeutic target and prognostic biomarker in CRC.
Collapse
Affiliation(s)
- A‐hui Fan
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anChina
| | - Xiaojuan Zhao
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular BiologyFourth Military Medical UniversityXi'anChina
| | - Hao Liu
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anChina
| | - Danxiu Li
- Department of Gastroenterology, Tangdu HospitalFourth Military Medical UniversityXi'anChina
| | - Tongtong Guo
- Department of Cell Biology, College of Life ScienceNorthwest UniversityXi'anChina
| | - Jiehao Zhang
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anChina
| | - Lili Duan
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anChina
| | - Hao Cheng
- Department of Gastroenterology, Tangdu HospitalFourth Military Medical UniversityXi'anChina
| | - Yongzhan Nie
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anChina
| | - Daiming Fan
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anChina
| | - Xiaodi Zhao
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anChina
| | - Yuanyuan Lu
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive DiseasesFourth Military Medical UniversityXi'anChina
| |
Collapse
|
12
|
Cui H, Li H, Wu H, Du F, Xie X, Zeng S, Zhang Z, Dong K, Shang L, Jing C, Li L. A novel 3'tRNA-derived fragment tRF-Val promotes proliferation and inhibits apoptosis by targeting EEF1A1 in gastric cancer. Cell Death Dis 2022; 13:471. [PMID: 35585048 PMCID: PMC9117658 DOI: 10.1038/s41419-022-04930-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 05/03/2022] [Accepted: 05/09/2022] [Indexed: 12/14/2022]
Abstract
At present, it is commonly believed that tRFs and tiRNAs are formed by the specific and selective shear of tRNAs under certain pressure stimulation, rather than by random degradation of tRNA. tRFs and tiRNAs have been reported to contribute to the biological process of a variety of human cancers. However, the evidence for the mechanisms of tRFs and tiRNAs in the occurrence and development of gastric cancer (GC) is still insufficient. Here, we aimed to explore the carcinogenic roles of tRFs and tiRNAs in GC with RNA-sequencing technique, and found a novel 3'tRNA-derived fragment tRF-Val was significantly upregulated in GC tissues and cell lines. tRF-Val expression was positively correlated with tumor size and the depth of tumor invasion in GC tissues. Functionally, tRF-Val promoted proliferation and invasion, and inhibited apoptosis in GC cells. Mechanistically, tRF-Val directly bound to the chaperone molecule EEF1A1, mediated its transport into the nucleus and promoted its interaction with MDM2 (a specific p53 E3 ubiquitin ligase), thus inhibiting the downstream molecular pathway of p53 and promoting GC progression. These findings provided a new potential therapeutic target for GC and a new explanation for the occurrence of GC.
Collapse
Affiliation(s)
- Huaiping Cui
- grid.27255.370000 0004 1761 1174Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, 250021 Jinan, Shandong China ,grid.460018.b0000 0004 1769 9639Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250021 Jinan, Shandong China ,grid.460018.b0000 0004 1769 9639Shandong Provincial Laboratory of Translational Medicine Engineering for Digestive Tumors, Shandong Provincial Hospital, 250021 Jinan, Shandong China
| | - Han Li
- grid.452422.70000 0004 0604 7301Department of Gastrointestinal Surgery, the First Affiliated Hospital of Shandong First Medical University, Shandong Provincial Qianfoshan Hospital, 250013 Jinan, Shandong China
| | - Hao Wu
- grid.27255.370000 0004 1761 1174Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, 250021 Jinan, Shandong China
| | - Fengying Du
- grid.27255.370000 0004 1761 1174Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, 250021 Jinan, Shandong China
| | - Xiaozhou Xie
- grid.460018.b0000 0004 1769 9639Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250021 Jinan, Shandong China
| | - Shujie Zeng
- grid.27255.370000 0004 1761 1174Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, 250021 Jinan, Shandong China
| | - Zihao Zhang
- grid.27255.370000 0004 1761 1174Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, 250021 Jinan, Shandong China
| | - Kangdi Dong
- grid.460018.b0000 0004 1769 9639Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250021 Jinan, Shandong China
| | - Liang Shang
- grid.27255.370000 0004 1761 1174Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, 250021 Jinan, Shandong China ,grid.460018.b0000 0004 1769 9639Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250021 Jinan, Shandong China ,grid.460018.b0000 0004 1769 9639Shandong Provincial Laboratory of Translational Medicine Engineering for Digestive Tumors, Shandong Provincial Hospital, 250021 Jinan, Shandong China
| | - Changqing Jing
- grid.27255.370000 0004 1761 1174Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, 250021 Jinan, Shandong China ,grid.460018.b0000 0004 1769 9639Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250021 Jinan, Shandong China ,grid.460018.b0000 0004 1769 9639Shandong Provincial Laboratory of Translational Medicine Engineering for Digestive Tumors, Shandong Provincial Hospital, 250021 Jinan, Shandong China
| | - Leping Li
- grid.27255.370000 0004 1761 1174Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, 250021 Jinan, Shandong China ,grid.460018.b0000 0004 1769 9639Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250021 Jinan, Shandong China ,grid.460018.b0000 0004 1769 9639Shandong Provincial Laboratory of Translational Medicine Engineering for Digestive Tumors, Shandong Provincial Hospital, 250021 Jinan, Shandong China
| |
Collapse
|
13
|
Shen L, Gu P, Qiu C, Ding WT, Zhang L, Cao WY, Li ZY, Yan B, Sun X. Lysophosphatidylcholine acyltransferase 1 promotes epithelial-mesenchymal transition of hepatocellular carcinoma via the Wnt/β-catenin signaling pathway. Ann Hepatol 2022; 27:100680. [PMID: 35108614 DOI: 10.1016/j.aohep.2022.100680] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 02/04/2023]
Abstract
INTRODUCTION AND OBJECTIVES Hepatocellular carcinoma (HCC) is one of the most malignant digestive tumors, and its insidious onset and rapid progression are the main reasons for the difficulty in effective treatment. Lysophosphatidylcholine acyltransferase 1 (LPCAT1) is a key enzyme that regulates phospholipid metabolism of the cell membrane. However, the mechanism by which LPCAT1 regulates HCC metastasis remains unknown. This study aimed to explore its biological function and potential mechanisms concerning migration and invasion in HCC. MATERIALS AND METHODS LPCAT1 expression in HCC tissues and its association with clinical outcomes were investigated by western blotting and bioinformatic methods, respectively. The role of LPCAT1 in migration and invasion was assessed via Transwell assays. The expression pattern of epithelial-mesenchymal transition (EMT) markers was quantified by western blotting. The biological behaviors of LPCAT1 in vivo were evaluated using xenograft tumor models and caudal vein metastatic models. Signaling pathways related to LPCAT1 were predicted using gene set enrichment analysis (GSEA) and further confirmed by western blotting. RESULTS LPCAT1 expression was significantly upregulated in HCC tissues and indicated a poor prognosis of HCC patients. Several EMT-related markers were found to be regulated by LPCAT1. HCC cells overexpressing LPCAT1 exhibited remarkably high migration and invasion capacities, upregulated expression of mesenchymal markers and reduced E-cadherin expression. In vivo, LPCAT1 promoted HCC pulmonary metastasis. Furthermore, the Wnt/β-catenin signaling pathway was confirmed to be activated by LPCAT1. CONCLUSIONS LPCAT1 could serve as a promising biomarker of HCC and as a novel therapeutic target for the treatment of metastatic HCC.
Collapse
Affiliation(s)
- Ling Shen
- Clinical Medical School, Shanghai General Hospital of Nanjing Medical University, Shanghai, China; Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peng Gu
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Qiu
- Institute of Gallstone Disease, Center of Gallbladder Disease, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wen-Tao Ding
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Zhang
- Intervention Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wan-Yue Cao
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zu-Yin Li
- Department of Hepatobiliary Surgery, Peking University Organ Transplantation Institute, Peking University People's Hospital, Beijing, China
| | - Bin Yan
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xing Sun
- Clinical Medical School, Shanghai General Hospital of Nanjing Medical University, Shanghai, China; Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
14
|
Ning X, Shi G, Ren S, Liu S, Ding J, Zhang R, Li L, Xie Q, Xu W, Meng F, Ma R. OUP accepted manuscript. Oncologist 2022; 27:e64-e75. [PMID: 35305106 PMCID: PMC8842331 DOI: 10.1093/oncolo/oyab015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 11/18/2021] [Indexed: 11/17/2022] Open
Abstract
Background The glioblastoma-amplified sequence (GBAS) is a newly identified gene that is amplified in approximately 40% of glioblastomas. This article probes into the expression, prognostic significance, and possible pathways of GBAS in ovarian cancer (OC). Method Immunohistochemical methods were used to evaluate the expression level of GBAS in OC and its relationship with clinicopathological characteristics and prognosis. Glioblastoma-amplified sequence shRNA was designed to transfect into OC cell lines to silence GBAS expression, then detect the proliferation, apoptosis, and migration ability of the cell. Furthermore, an in vitro tumor formation experiment in mice was constructed to prove the effect of GBAS expression on the growth of OC in vivo. To further study the regulation mechanism of GBAS, we performed co-immunoprecipitation (Co-IP) and shotgun LC-MS mass spectrometry identification. Results Immunohistochemistry indicated that GBAS was markedly overexpressed in OC compared with normal ovarian tissue and was associated with lymph node metastasis. Inhibition of GBAS expression can significantly reduce OC cell proliferation, colony formation, promote cell apoptosis, and reduce the ability of cell migration and invasion. In vivo tumor formation experiments showed that the size and weight of tumors in mice after GBAS expression knockdown was significantly smaller. Glioblastoma-amplified sequence may be combined with elongation factor 1 alpha 1 (eEF1A1) to achieve its regulation in OC. Bioinformatics analysis data indicate that GBAS may be a key regulator of mitochondria-associated pathways, therefore controlling cancer progression. MicroRNA-27b, MicroRNA-23a, and MicroRNA-590 may directly targeting GBAS affects the biological behavior of OC cells. Conclusion The glioblastoma-amplified sequence may regulate the proliferation and metastasis of OC cells by combining with eEF1A1.
Collapse
Affiliation(s)
- Xin Ning
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Guangyue Shi
- Department of Oncology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Sujing Ren
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Shuang Liu
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Jing Ding
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Ruichun Zhang
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Lianwei Li
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Qin Xie
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Wei Xu
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Fanling Meng
- Corresponding author: Fanling Meng, Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin 150081, China. Tel: +86 451 85718069;
| | - Rong Ma
- Corresponding author: Rong Ma, Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin 150081, China. Tel: +86 451 85718058;
| |
Collapse
|
15
|
Gong T, Shuang Y. Expression and Clinical Value of Eukaryotic Translation Elongation Factor 1A1 (EEF1A1) in Diffuse Large B Cell Lymphoma. Int J Gen Med 2021; 14:7247-7258. [PMID: 34737619 PMCID: PMC8559353 DOI: 10.2147/ijgm.s324645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 07/29/2021] [Indexed: 12/02/2022] Open
Abstract
Background The eukaryotic translation elongation factor 1A1 (EEF1A1) participates in protein translation and has been reported to be involved in tumor progression such as hepatocellular carcinoma. Diffuse large B-cell lymphoma (DLBCL) is the most common lymphoid malignancy in adults. In the present study, we aimed to detect the expression of EEF1A1 in DLBCL and to analyze its relationship with prognosis. Methods We reviewed medical records of DLBCL patients in our hospital and evaluated their expression level of EEF1A1 in tumor tissues using immunohistochemical (IHC) assay. The Chi-square method was used for correlation analysis. The Kaplan–Meier method with Log rank test was used for univariate analysis. Cox proportional hazards model was used for multivariate analysis. Cellular and mice models were introduced to validate its oncogenic role. Results EEF1A1 expression in tumor cells was higher in certain DLBCL cases. Patients with higher EEF1A1 expression were more likely to have advanced tumor stage and poorer 5-year overall survival (OS) rates. EEF1A1 expression in tumor cells was an independent risk predictor for OS (P < 0.05). Cellular assays demonstrated that EEF1A1-shRNA significantly inhibited lymphoma cell proliferation. The study of xenografts further verified the effect of EEF1A1-shRNA on suppressing tumor growth in vivo. Conclusion EEF1A1 positivity predicts short survival in DLBCL patients. For patients with higher EEF1A1 expression, more strategy such as anti-EEF1A1 antibody treatment should be developed.
Collapse
Affiliation(s)
- Tiejun Gong
- Institute of Hematology and Oncology, Harbin the First Hospital, Harbin, 150010, People's Republic of China
| | - Yuerong Shuang
- Department of Lymphatic Hematology and Oncology, Jiangxi Cancer Hospital, Nanchang, 330029, People's Republic of China
| |
Collapse
|
16
|
Bracic Tomazic S, Schatz C, Haybaeck J. Translational Regulation in Hepatocellular Carcinogenesis. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:4359-4369. [PMID: 34703211 PMCID: PMC8523516 DOI: 10.2147/dddt.s255582] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 10/04/2021] [Indexed: 12/17/2022]
Abstract
The mortality of hepatocellular carcinoma (HCC) is distributed unevenly worldwide. One of the major causes is hepatitis B or hepatitis C virus infection and the development and progression of liver cirrhosis. The carcinogenesis of HCC is among others regulated via the mTOR (mechanistic target of rapamycin) signaling pathway and represents a possible method of targeted treatment. The aim of our article was to address the most recent clinical advances and findings of basic studies on the mTOR signaling pathway and the involved factors. Risk factors play a key role in dysregulation of the signaling pathway, where both mTORCs are upregulated and protein synthesis is altered. eIFs and, to a lesser extent, eEFs play an essential role in this process. Whether the factor will be upregulated or downregulated, among others, depends on hepatitis B/C virus infection. The amount of a particular factor in a patient sample lets us know whether HCC recurrence will occur, what is the likelihood of chemoresistance, and what outcome is predicted for patients with an increased value. Our analysis shows that in addition to mTOR, eIF3, eIF4, and eIF5 play an important role, as they can serve as biomarkers for non- and virus-related HCC.
Collapse
Affiliation(s)
- Suzana Bracic Tomazic
- Department of Pathology, Hospital Graz II, Graz, 8020, Austria.,Faculty of Medicine, University of Maribor, Maribor, 2000, Slovenia
| | - Christoph Schatz
- Institute of Pathology, Neuropathology and Molecular Pathology, Medical University of Innsbruck, Innsbruck, 6020, Austria
| | - Johannes Haybaeck
- Institute of Pathology, Neuropathology and Molecular Pathology, Medical University of Innsbruck, Innsbruck, 6020, Austria.,Diagnostic & Research Center for Molecular BioMedicine, Institute of Pathology, Medical University Graz, Graz, 8010, Austria
| |
Collapse
|
17
|
Hou Y, Liu R, Xia M, Sun C, Zhong B, Yu J, Ai N, Lu JJ, Ge W, Liu B, Chen X. Nannocystin ax, an eEF1A inhibitor, induces G1 cell cycle arrest and caspase-independent apoptosis through cyclin D1 downregulation in colon cancer in vivo. Pharmacol Res 2021; 173:105870. [PMID: 34500061 DOI: 10.1016/j.phrs.2021.105870] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/24/2021] [Accepted: 09/02/2021] [Indexed: 12/28/2022]
Abstract
Colorectal cancer (CRC) is one of the most common causes of cancer-related death worldwide. Nannocystin ax (NAN), a 21-membered cyclodepsipeptide initially isolated from myxobacteria of the Nannocystis genus, was found to target the eukaryotic elongation factor 1A (eEF1A). The current study was designed to evaluate the anticancer effect and underlying mechanisms of NAN with in vitro and in vivo models. Results showed that NAN induced G1 phase cell cycle arrest and caspase-independent apoptosis in HCT116 and HT29 human CRC cells. NAN significantly downregulated cyclin D1 level in a short time, but NAN did not affect the transcription level and ubiquitin-dependent degradation of cyclin D1. Furthermore, NAN treatment directly targeted eEF1A and partially decreased the synthesis of new proteins, contributing to the downregulation of cyclin D1. Besides, NAN significantly suppressed tumor growth in the zebrafish xenograft model. In conclusion, NAN triggered G1 phase cell cycle arrest through cyclin D1 downregulation and eEF1A-targeted translation inhibition and promoted caspase-independent apoptosis in CRC cells.
Collapse
Affiliation(s)
- Ying Hou
- Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Rong Liu
- Key Laboratory of Green Chemistry & Technology of Ministry of Education, College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Mengwei Xia
- Key Laboratory of Green Chemistry & Technology of Ministry of Education, College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Chong Sun
- Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Bingling Zhong
- Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Jie Yu
- Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Nana Ai
- Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Jin-Jian Lu
- Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Wei Ge
- Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Bo Liu
- Key Laboratory of Green Chemistry & Technology of Ministry of Education, College of Chemistry, Sichuan University, Chengdu 610064, China.
| | - Xiuping Chen
- Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macau, China.
| |
Collapse
|
18
|
Sopko B, Tejral G, Bitti G, Abate M, Medvedikova M, Hajduch M, Chloupek J, Fajmonova J, Skoric M, Amler E, Erban T. Glyphosate Interaction with eEF1α1 Indicates Altered Protein Synthesis: Evidence for Reduced Spermatogenesis and Cytostatic Effect. ACS OMEGA 2021; 6:14848-14857. [PMID: 34151066 PMCID: PMC8209799 DOI: 10.1021/acsomega.1c00449] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 05/18/2021] [Indexed: 06/13/2023]
Abstract
The broad-spectrum herbicide, glyphosate, is considered safe for animals because it selectively affects the shikimate pathway that is specific to plants and microorganisms. We sought a previously unknown mechanism to explain the concerns that glyphosate exposure can negatively affect animals, including humans. Computer modeling showed a probable interaction between glyphosate and eukaryotic translation elongation factor 1 subunit alpha 1 (eEF1α1), which was confirmed by microcalorimetry. Only restricted, nondisrupted spermatogenesis in rats was observed after chronic glyphosate treatments (0.7 and 7 mg/L). Cytostatic and antiproliferative effects of glyphosate in GC-1 and SUP-B15 cells were indicated. Meta-analysis of public health data suggested a possible effect of glyphosate use on sperm count. The in silico, in vitro, and in vivo experimental results as well as the metastatistics indicate side effects of chronic glyphosate exposure. Together, these findings indicate that glyphosate delays protein synthesis through an interaction with eEF1α1, thereby suppressing spermatogenesis and cell growth.
Collapse
Affiliation(s)
- Bruno Sopko
- Crop
Research Institute, Prague 161 06, Czechia
- Department
of Medical Chemistry and Clinical Biochemistry, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague 150 06, Czechia
- Laboratory
of Tissue Engineering, Institute of Experimental
Medicine, Academy of Sciences of the Czech Republic, Prague 142 20, Czechia
- Biomedicine
and Advanced Biomaterials Department, University Center for Energy
Efficient Buildings, The Czech Technical
University in Prague, Prague, Bustehrad 273 43, Czechia
| | - Gracian Tejral
- Laboratory
of Tissue Engineering, Institute of Experimental
Medicine, Academy of Sciences of the Czech Republic, Prague 142 20, Czechia
- Biomedicine
and Advanced Biomaterials Department, University Center for Energy
Efficient Buildings, The Czech Technical
University in Prague, Prague, Bustehrad 273 43, Czechia
- Department
of Biophysics, 2nd Faculty of Medicine, Charles University, Prague 150 06, Czechia
| | - Guissepe Bitti
- Laboratory
of Tissue Engineering, Institute of Experimental
Medicine, Academy of Sciences of the Czech Republic, Prague 142 20, Czechia
- Biomedicine
and Advanced Biomaterials Department, University Center for Energy
Efficient Buildings, The Czech Technical
University in Prague, Prague, Bustehrad 273 43, Czechia
| | - Marianna Abate
- Department
of Precision Medicine, University of Campania
“Luigi Vanvitelli”, Naples 80131, Italy
| | - Martina Medvedikova
- Institute
of Molecular and Translation Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc 779 00, Czechia
| | - Marian Hajduch
- Institute
of Molecular and Translation Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc 779 00, Czechia
| | - Jan Chloupek
- Department
of Pharmacology and Pharmacy, Faculty of Veterinary Medicine, University of Veterinary and Pharmaceutical Sciences
Brno, Brno 612 42, Czechia
| | - Jolana Fajmonova
- Department
of Pharmacology and Pharmacy, Faculty of Veterinary Medicine, University of Veterinary and Pharmaceutical Sciences
Brno, Brno 612 42, Czechia
| | - Misa Skoric
- Department
of Pathological Morphology and Parasitology, Faculty of Veterinary
Medicine, University of Veterinary and Pharmaceutical
Sciences Brno, Brno 612 42, Czechia
| | - Evzen Amler
- Biomedicine
and Advanced Biomaterials Department, University Center for Energy
Efficient Buildings, The Czech Technical
University in Prague, Prague, Bustehrad 273 43, Czechia
- Department
of Biophysics, 2nd Faculty of Medicine, Charles University, Prague 150 06, Czechia
| | - Tomas Erban
- Crop
Research Institute, Prague 161 06, Czechia
| |
Collapse
|
19
|
Ji W, Peng Z, Sun B, Chen L, Zhang Q, Guo M, Su C. LpCat1 Promotes Malignant Transformation of Hepatocellular Carcinoma Cells by Directly Suppressing STAT1. Front Oncol 2021; 11:678714. [PMID: 34178664 PMCID: PMC8220817 DOI: 10.3389/fonc.2021.678714] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 05/18/2021] [Indexed: 01/17/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a malignant cancer with rapid proliferation and high metastasis ability. To explore the crucial genes that maintain the aggressive behaviors of cancer cells is very important for clinical gene therapy of HCC. LpCat1 was reported to be highly expressed and exert pro-tumorigenic effect in a variety of cancers, including HCC. However, its detailed molecular mechanism remained unclear. In this study, we confirmed that LpCat1 was up-regulated in HCC tissues and cancer cell lines. The overexpressed LpCat1 promoted the proliferation, migration and invasion of HCC cells, and accelerated cell cycle progression, while knocking down LpCat1 significantly inhibited cell proliferation, migration and invasion in vitro and in vivo, and arrested HCC cells at G0/G1 phase. Moreover, we proved for the first time that LpCat1 directly interacted with STAT1 which was generally recognized as a tumor suppressor in HCC. High levels of LpCat1 in HCC could inhibit STAT1 expression, up-regulate CyclinD1, CyclinE, CDK4 and MMP-9, and decrease p27kip1 to promote cancer progression. Conversely, down-regulation of LpCat1 would cause the opposite changes to repress the viability and motility of HCC cells. Consequently, we concluded that LpCat1 was a contributor to progression and metastasis of HCC by interacting with STAT1.
Collapse
Affiliation(s)
- Weidan Ji
- Department of Molecular Oncology, Eastern Hepatobiliary Surgical Hospital & National Centre for Liver Cancer, Navy Military Medical University, Shanghai, China
| | - Zhangxiao Peng
- Department of Molecular Oncology, Eastern Hepatobiliary Surgical Hospital & National Centre for Liver Cancer, Navy Military Medical University, Shanghai, China
| | - Bin Sun
- Department of Molecular Oncology, Eastern Hepatobiliary Surgical Hospital & National Centre for Liver Cancer, Navy Military Medical University, Shanghai, China
| | - Lei Chen
- Department of Molecular Oncology, Eastern Hepatobiliary Surgical Hospital & National Centre for Liver Cancer, Navy Military Medical University, Shanghai, China
| | - Qin Zhang
- Department of Molecular Oncology, Eastern Hepatobiliary Surgical Hospital & National Centre for Liver Cancer, Navy Military Medical University, Shanghai, China
| | - Minggao Guo
- Department of General Surgery, Shanghai Sixth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Changqing Su
- Department of Molecular Oncology, Eastern Hepatobiliary Surgical Hospital & National Centre for Liver Cancer, Navy Military Medical University, Shanghai, China
| |
Collapse
|
20
|
Ao J, Ma Z, Li R, Zhang S, Gao X, Zhang M. Phospho-Tudor-SN coordinates with STAT5 to regulate prolactin-stimulated milk protein synthesis and proliferation of bovine mammary epithelial cells. Anim Biotechnol 2021; 33:1161-1169. [PMID: 33849380 DOI: 10.1080/10495398.2021.1879824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Tudor staphylococcal nuclease (Tudor-SN) participates in milk synthesis and cell proliferation in response to prolactin (PRL) and plays a regulatory role on mTOR phosphorylation. However, the complicated molecular mechanism of Tudor-SN regulating milk protein synthesis and cell proliferation still remains to be illustrated. In present study, we observed that the proteins level of phosphorylated Tudor-SN and phosphorylated STAT5 were simultaneously enhanced upon PRL treatment in bovine mammary epithelial cells (BMECs). Tudor-SN overexpression and knockdown experiment showed that Tudor-SN positively regulated the synthesis of milk protein, cell proliferation and the phosphorylation of STAT5, which was dependent on Tudor-SN phosphorylation. STAT5 knockdown experiment showed that Tudor-SN stimulated mTOR pathway through regulating STAT5 activation, which was required for PRL to activate the mTOR pathway. Thus, these results demonstrate the primary mechanism of Tudor-SN coordinating with STAT5 to regulate milk protein synthesis and cell proliferation under stimulation of PRL in BMECs, which may provide some new perspectives for increasing milk production.
Collapse
Affiliation(s)
- Jinxia Ao
- College of Life Science, Northeast Agricultural University, Heilongjiang, Harbin, China
| | - Zonghua Ma
- College of Life Science, Northeast Agricultural University, Heilongjiang, Harbin, China
| | - Rui Li
- College of Life Science, Northeast Agricultural University, Heilongjiang, Harbin, China
| | - Sen Zhang
- College of Life Science, Northeast Agricultural University, Heilongjiang, Harbin, China
| | - Xuejun Gao
- College of Animal Science, Yangtze University, Jingzhou, Hubei, China
| | - Minghui Zhang
- College of Life Science, Northeast Agricultural University, Heilongjiang, Harbin, China.,College of Animal Science, Yangtze University, Jingzhou, Hubei, China
| |
Collapse
|
21
|
Pawlonka J, Rak B, Ambroziak U. The regulation of cyclin D promoters - review. Cancer Treat Res Commun 2021; 27:100338. [PMID: 33618151 DOI: 10.1016/j.ctarc.2021.100338] [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] [Received: 12/17/2020] [Revised: 02/06/2021] [Accepted: 02/15/2021] [Indexed: 11/25/2022]
Abstract
Cyclins are key regulators of cell cycle progression and survival. Particularly cyclins D (cyclin D1, D2, and D3) act in response to the mitogenic stimulation and are pivotal mediators between proliferative pathways and the nuclear cell cycle machinery. Dysregulation of cyclins expression results in impaired development, abnormal cell growth or tumorigenesis. In this review we summarize current knowledge about regulatory role of the cyclin D promoters, transcriptional factors: regulators, co-activators and adaptor proteins necessary to their activation. We focused on the intracellular signaling pathways vital to cell growth, differentiation and apoptosis including transcription factor families: activator protein 1 (AP1), nuclear factor (NFκB), signal transducer and activator of transcription (STAT), cAMP response element-binding protein (CREB) and Sp/NF-Y, with a special insight into the tissue specific cyclin representation.
Collapse
Affiliation(s)
- Jan Pawlonka
- Department of Internal Medicine and Endocrinology, Medical University of Warsaw, Warsaw
| | - Beata Rak
- Department of Internal Medicine and Endocrinology, Medical University of Warsaw, Warsaw; Department of Genomic Medicine, Medical University of Warsaw, Warsaw.
| | - Urszula Ambroziak
- Department of Internal Medicine and Endocrinology, Medical University of Warsaw, Warsaw
| |
Collapse
|
22
|
Mei N, Zhao N, Tian T, Jiao M, Li C. Biological features, gene expression profile, and mechanisms of drug resistance of two- and three-dimensional hepatocellular carcinoma cell cultures. Pharmacol Res Perspect 2021; 9:e00715. [PMID: 33486902 PMCID: PMC7827916 DOI: 10.1002/prp2.715] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 12/21/2020] [Indexed: 12/21/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a common malignant tumor with insidious onset and rapid progression. Its treatment is often difficult owing to tumor resistance. In this study, we aimed to understand the different biological characteristics, gene expression profiles, and drug resistance mechanisms of HCC cells cultured under different conditions. A conventional adherence method and a liquid overlay technique were used to prepare two- and three-dimensional cultures of Bel-7402 and 5-fluorouracil (5-Fu)-resistant Bel-7402 (Bel-7402/5-Fu) cells. Morphological characteristics were assessed via microscopy, and cell cycle distribution and apoptotic rate were obtained using flow cytometry. Cell sensitivity to different concentrations of drugs was detected with 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays. Gene expression profiles and signal transduction pathways of Bel-7402 and Bel-7402/5-Fu cells under different culture conditions were determined using gene chips. Cells in three-dimensional culture were suspended and they grew into dense multicellular spheroid (MCS) structures, aggregating with each other. In contrast to cells in the two-dimensional culture, cell cycle arrest was observed in MCSs. The sensitivity of Bel-7402 cells in the two-dimensional culture to drugs at high concentrations was significantly higher than that of cells in the three-dimensional culture (p < .05). The apoptotic rate of Bel-7402 and Bel-7402/5-Fu cells was also higher in the two-dimensional culture (p < .05). Signal transduction pathway analysis showed that after Bel-7402 cells acquired resistance to 5-Fu, CCND1, MCM2, and MCM3 gene expression was upregulated in the G1 to S cell cycle control signal transduction pathway, CDKN1C and CCNG2 gene expression was downregulated, and MCM2 and MCM3 gene expression was upregulated in the DNA replication signal transduction pathway. Therefore, the liquid overlay technique is a simple, low-cost procedure to successfully construct three-dimensional culture models of HCC. This study provides new information and methods for exploring the molecular mechanisms of liver cancer resistance, clinical treatment, development of molecular information, and interventional prevention.
Collapse
Affiliation(s)
- Nan Mei
- Department of Medical OncologyThe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’anShaanxi ProvincePeople’s Republic of China
| | - Ni Zhao
- Department of Medical OncologyThe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’anShaanxi ProvincePeople’s Republic of China
| | - Tao Tian
- Department of Medical OncologyThe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’anShaanxi ProvincePeople’s Republic of China
| | - Min Jiao
- Department of Medical OncologyThe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’anShaanxi ProvincePeople’s Republic of China
| | - Chunli Li
- Department of Medical OncologyThe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’anShaanxi ProvincePeople’s Republic of China
| |
Collapse
|
23
|
Ai J, Tan G, Wang T, Li W, Gao R, Song Y, Xiong S, Qing X. Transcription factor STAT1 promotes the proliferation, migration and invasion of nasopharyngeal carcinoma cells by upregulating LINC01160. Future Oncol 2020; 17:57-69. [PMID: 33263259 DOI: 10.2217/fon-2020-0618] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Aim: To investigate the role of LINC01160 in nasopharyngeal carcinoma (NPC). Materials & methods: Using NPC cells CNE-2 and HNE-2 in vitro, we performed quantitative PCR to determine mRNA expression and western blotting to determine protein expression. CCK-8, transwell, flow cytometry and wound healing assays were done to examine the function of LINC01160 and STAT1. Chromatin immunoprecipitation PCR (ChIP-PCR) confirmed that STAT1 combines with the LINC01160 promoter region. Xenograft experiments were used to verify the role of STAT1 and LINC01160 in vivo. Results: LINC01160 is upregulated in NPC and can promote a malignant cell phenotype. STAT1 is a transcription factor of LINC01160 and can promote a malignant cell phenotype through upregulating LINC01160 expression. Conclusion: STAT1 can promote a malignant cell phenotype by upregulating LINC01160.
Collapse
Affiliation(s)
- Jingang Ai
- Department of Otolaryngology-Head & Neck Surgery, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Guolin Tan
- Department of Otolaryngology-Head & Neck Surgery, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Tiansheng Wang
- Department of Otolaryngology-Head & Neck Surgery, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Wei Li
- Department of Otolaryngology-Head & Neck Surgery, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Ru Gao
- Department of Otolaryngology-Head & Neck Surgery, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Yexun Song
- Department of Otolaryngology-Head & Neck Surgery, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Shanshan Xiong
- Department of Otolaryngology-Head & Neck Surgery, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Xiang Qing
- Department of Otolaryngology-Head & Neck Surgery, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| |
Collapse
|
24
|
Xiao S, Wang Y, Ma Y, Liu J, Tang C, Deng A, Fang C. Dimethylation of eEF1A at Lysine 55 Plays a Key Role in the Regulation of eEF1A2 on Malignant Cell Functions of Acute Myeloid Leukemia. Technol Cancer Res Treat 2020; 19:1533033820914295. [PMID: 32347192 PMCID: PMC7225831 DOI: 10.1177/1533033820914295] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE This study aimed to explore whether eukaryotic translation elongation factor 1 alpha 2 affected cell proliferation, migration, and apoptosis via regulating the dimethylation of eukaryotic translation elongation factor 1 alpha at lysine 55 in acute myeloid leukemia. METHODS The expressions of eukaryotic translation elongation factor 1 alpha 2 and dimethylation of eukaryotic translation elongation factor 1 alpha at lysine 55 in acute myeloid leukemia cell lines and human normal bone marrow mononuclear cells (as control) were assessed. Control CRISPR-Cas9 lentivirus, eukaryotic translation elongation factor 1 alpha 2 knockout CRISPR-Cas9 lentivirus, vector plasmid, eukaryotic translation elongation factor 1 alpha 2 wild type overexpression plasmid, and eukaryotic translation elongation factor 1 alpha 2 with a K55R substitution overexpression plasmid were transfected into AML-193 and Kasumi-1 cells combined or alone, and were accordingly divided into 4 groups (Sgcontrol + vector group, SgeEF1A2 + vector group, SgeEF1A2 + eEF1A2WT group, and SgeEFIA2 + eEF1A2K55R group). RESULTS Eukaryotic translation elongation factor 1 alpha 2 and dimethylation of eukaryotic translation elongation factor 1 alpha at lysine 55 expressions were higher in AML-193, Kasumi-1, and KG-1 cell lines compared to the control. In AML-193 and Kasumi-1 cells, the knockout and compensated experiments revealed that eukaryotic translation elongation factor 1 alpha 2 promoted cell proliferation and migration but repressed apoptosis. Additionally, the knockout of eukaryotic translation elongation factor 1 alpha 2 decreased dimethylation of eukaryotic translation elongation factor 1 alpha at lysine 55 expression, meanwhile, eukaryotic translation elongation factor 1 alpha 2 wild type overexpression enhanced while eukaryotic translation elongation factor 1 alpha 2 with a K55R substitution overexpression did not influence the dimethylation of eukaryotic translation elongation factor 1 alpha at lysine 55 expression. Furthermore, eukaryotic translation elongation factor 1 alpha 2 wild type overexpression promoted cell proliferation, enhanced migration, and decreased apoptosis, but eukaryotic translation elongation factor 1 alpha 2 with a K55R substitution overexpression did not influence these cellular functions in AML-193 and Kasumi-1 cells, suggesting the implication of dimethylation of eukaryotic translation elongation factor 1 alpha at lysine 55 in eukaryotic translation elongation factor 1 alpha 2 mediated oncogenesis of acute myeloid leukemia. CONCLUSION Eukaryotic translation elongation factor 1 alpha 2 and its dimethylated product may serve as therapeutic targets, and these findings may provide support for exploring novel strategies in acute myeloid leukemia treatment.
Collapse
Affiliation(s)
- Shan Xiao
- Department of Pharmacy, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yanping Wang
- Department of Pediatrics, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yuwen Ma
- Department of Pharmacy, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Jue Liu
- Department of Pharmacy, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Can'e Tang
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Aiping Deng
- Department of Pharmacy, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Chunxiang Fang
- Department of Pharmacy, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| |
Collapse
|
25
|
Yu L, Ye F, Li YY, Zhan YZ, Liu Y, Yan HM, Fang Y, Xie YW, Zhang FJ, Chen LH, Ding Y, Chen KL. Histone methyltransferase SETDB1 promotes colorectal cancer proliferation through the STAT1-CCND1/CDK6 axis. Carcinogenesis 2020; 41:678-688. [PMID: 31306481 DOI: 10.1093/carcin/bgz131] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 06/16/2019] [Accepted: 07/12/2019] [Indexed: 12/14/2022] Open
Abstract
Upregulation of histone methyltransferase SET domain bifurcated 1 (SETDB1) is associated with poor prognosis in cancer patients. However, the mechanism of oncogenicity of SETDB1 in cancer is hitherto unknown. Here, we show that SETDB1 is upregulated in human colorectal cancer (CRC) where its level correlates with poor clinical outcome. Ectopic SETDB1 promotes CRC cell proliferation, whereas SETDB1 attenuation inhibits this process. Flow cytometry reveals that SETDB1 promotes proliferation by driving the CRC cell cycle from G0/G1 phase to S phase. Mechanistically, SETDB1 binds directly to the STAT1 promoter region resulting in increased STAT1 expression. Functional characterization reveals that STAT1-CCND1/CDK6 axis is a downstream effector of SETDB1-mediated CRC cell proliferation. Furthermore, SETDB1 upregulation is sufficient to accelerate in vivo proliferation in xenograft animal model. Taken together, our results provide insight into the upregulation of SETDB1 within CRC and can lead to novel treatment strategies targeting this cell proliferation-promoting gene.
Collapse
Affiliation(s)
- Lu Yu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Feng Ye
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yi-Yi Li
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yi-Zhi Zhan
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yang Liu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hong-Mei Yan
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuan Fang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yu-Wen Xie
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Feng-Jiao Zhang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Long-Hua Chen
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yi Ding
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ke-Li Chen
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,HuiQiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
26
|
Wang H, Chen L, Zhou T, Zhang Z, Zeng C. Nicotine Promotes WRL68 Cells Proliferation Due to the Mutant p53 Gain-of-Function by Activating CDK6-p53-RS-PIN1-STAT1 Signaling Pathway. Chem Res Toxicol 2020; 33:2361-2373. [PMID: 32820905 DOI: 10.1021/acs.chemrestox.0c00119] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The liver is an important organ with many tasks, such as dealing with drugs, alcohol and other toxins to remove them from the body. Nicotine is the more abundant component in cigarette smoking, which is first metabolized in liver and increases the risk of developing hepatocellular carcinoma (HCC). Also, genotoxic potential of nicotine has been extensively studied in vitro. However, the carcinogenic action of nicotine on the HCC needs to be elucidated. The current study demonstrated that chronic exposure to nicotine significantly promotes human normal fetal hepatic cell line (WRL68 cells) proliferation in a time- and concentration-dependent manner resulting from G0/G1-S-phase transition. Also remarkably, nicotine induced the level of p53 mutation at Ser249 (p53-RS). Note as well that the level of STAT1 protein was increased along with p53-RS owing to the prolonged half-life of STAT1. Furthermore, it is suggested that CDK6-dependent binding between phosphorylation of p53-RS at Ser249 and PIN1 by nicotine treatment leads to the nucleus translocation, followed by interacting with STAT1 and subsequent activation of STAT1 via the improvement of its stability, which is involved in cellular growth and colony formation after nicotine treatment. Simply put, these findings indicated that nicotine induces mutant p53 gain-of function (GOF), activating CDK6-p53-RS-PIN1-STAT1 signaling pathway and promoting cell proliferation, which could contribute to HCC for smokers.
Collapse
Affiliation(s)
- Huai Wang
- School of Public Health, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, P. R. of China.,Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, P. R. of China
| | - Lu Chen
- Wuhan Taisheng Biological Technology Co., Ltd., No. 10 West Yezhihu Road, Wuhan, Hubei 430074, P. R. of China
| | - Tong Zhou
- School of Public Health, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, P. R. of China.,Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, P. R. of China
| | - Zhongwei Zhang
- School of Public Health, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, P. R. of China.,Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, P. R. of China
| | - Canwei Zeng
- Wuhan Taisheng Biological Technology Co., Ltd., No. 10 West Yezhihu Road, Wuhan, Hubei 430074, P. R. of China
| |
Collapse
|
27
|
Song X, Yang W, Wu C, Han Y, Lu Y. USP9X promotes the proliferation, invasion and metastasis of liver cancer cells through regulating the JAK2/STAT3 signaling. Oncol Lett 2020; 20:2897-2905. [PMID: 32782606 PMCID: PMC7400992 DOI: 10.3892/ol.2020.11824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Accepted: 05/27/2020] [Indexed: 12/25/2022] Open
Abstract
X-linked ubiquitin-specific peptidase 9 (USP9X) serves important roles in the development and progression of various human cancers. However, its role and molecular mechanism in liver cancer require further elucidation. In the present study, USP9X was found to be upregulated in liver cancer tissues. At the same time, overexpression of USP9X promoted the proliferation, invasiveness and migration of liver cancer cells, which were subsequently suppressed by USP9X silencing. On a molecular level, the results revealed that USP9X knockdown suppressed elements of the Janus kinase 2 (JAK2)/STAT3 signaling pathway, including JAK2, STAT3, matrix metalloproteinase-2 and c-Myc. By contrast, overexpression of USP9X had the opposite effect. In conclusion, the results of the present study suggest that USP9X is upregulated in patients with liver cancer, which may accelerate the proliferation, invasiveness and migration of liver cancer cells by regulating the JAK2/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Xingchao Song
- Department of General Surgery, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu 221000, P.R. China.,Department of General Surgery, Xuzhou No. 1 People's Hospital, Xuzhou, Jiangsu 221000, P.R. China
| | - Weibin Yang
- Department of General Surgery, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu 221000, P.R. China.,Department of General Surgery, Xuzhou No. 1 People's Hospital, Xuzhou, Jiangsu 221000, P.R. China
| | - Chao Wu
- Department of General Surgery, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu 221000, P.R. China.,Department of General Surgery, Xuzhou No. 1 People's Hospital, Xuzhou, Jiangsu 221000, P.R. China
| | - Yamin Han
- Department of General Surgery, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu 221000, P.R. China.,Department of General Surgery, Xuzhou No. 1 People's Hospital, Xuzhou, Jiangsu 221000, P.R. China
| | - Yaowu Lu
- Department of General Surgery, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu 221000, P.R. China.,Department of General Surgery, Xuzhou No. 1 People's Hospital, Xuzhou, Jiangsu 221000, P.R. China
| |
Collapse
|
28
|
Zhang M, Ma Z, Li R, Guo S, Qiu Y, Gao X. Proteomic Analysis Reveals Proteins and Pathways Associated with Lactation in Bovine Mammary Epithelial Cell-Derived Exosomes. J Proteome Res 2020; 19:3211-3219. [DOI: 10.1021/acs.jproteome.0c00176] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Minghui Zhang
- College of Animal Science, Yangtze University, Nanhuan Road 1, Jingzhou, 434020 Hubei, China
| | - Zonghua Ma
- College of Life Science, Northeast Agricultural University, Changjiang Road 600, Xiangfang District, Harbin, 150030 Heilongjiang, China
| | - Rui Li
- College of Life Science, Northeast Agricultural University, Changjiang Road 600, Xiangfang District, Harbin, 150030 Heilongjiang, China
| | - Siqi Guo
- College of Life Science, Northeast Agricultural University, Changjiang Road 600, Xiangfang District, Harbin, 150030 Heilongjiang, China
| | - Youwen Qiu
- College of Life Science, Northeast Agricultural University, Changjiang Road 600, Xiangfang District, Harbin, 150030 Heilongjiang, China
| | - Xuejun Gao
- College of Animal Science, Yangtze University, Nanhuan Road 1, Jingzhou, 434020 Hubei, China
| |
Collapse
|
29
|
Sun R, Zhai R, Ma C, Miao W. Combination of aloin and metformin enhances the antitumor effect by inhibiting the growth and invasion and inducing apoptosis and autophagy in hepatocellular carcinoma through PI3K/AKT/mTOR pathway. Cancer Med 2020; 9:1141-1151. [PMID: 31830378 PMCID: PMC6997051 DOI: 10.1002/cam4.2723] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 10/21/2019] [Accepted: 11/09/2019] [Indexed: 01/27/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a devastating and highly metastatic cancer worldwide. Metformin (MET) is the priority drug for treatment of type 2 diabetes; however, it possesses multiple biological effects like anticancer and hepatoprotective activity. Herein, we examined the effects of aloin (barbaloin) and MET as well as combination treatment in HCC cell line in vitro and in vivo. As a result, aloin and MET alone exhibited inhibitory effects on proliferation and invasion of HepG2 and Bel-7402 cells. Specially, combination treatment of aloin and MET showed enhanced inhibitory effects in vitro. Aloin and MET alone induced apoptosis and autophagy in vitro. Similarly, aloin and MET cooperated to promote apoptosis and autophagy in HepG2 and Bel-7402 cells. In the HepG2 xenograft models, aloin in combination with MET confine tumor growth and facilitate apoptosis and autophagy. Both the in vitro and in vivo results showed that aloin and MET alone as well as combination treatment activated the PI3K/AKT/mTOR pathway. Overall, our research demonstrated that the concomitant treatment with aloin and MET enhances the antitumor effect by inhibiting the growth and invasion as well as inducing apoptosis and autophagy in HCC through PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Ruijie Sun
- Department of Hepatobiliary SurgeryJining First People 's HospitalJiningShandongChina
| | - Ruiren Zhai
- Department of Cancer CenterTumor Center Shandong Sunshine HospitalWeifangShandongChina
| | - Changlin Ma
- Department of Hepatobiliary SurgeryJining First People 's HospitalJiningShandongChina
| | - Wei Miao
- Department of Health CareJining First People's HospitalJiningShandongChina
| |
Collapse
|
30
|
Liu P, Zhu C, Luo J, Lan S, Su D, Wang Q, Wei Z, Cui W, Xu C, Yang X. Par6 regulates cell cycle progression through enhancement of Akt/PI3K/GSK-3β signaling pathway activation in glioma. FASEB J 2020; 34:1481-1496. [PMID: 31914615 DOI: 10.1096/fj.201901629rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 11/05/2019] [Accepted: 11/12/2019] [Indexed: 02/05/2023]
Abstract
As the key factor of the polarity protein complex, Par6 not only regulates polarization processes, but also plays important roles in tumor metastasis and progression in many epithelium malignancy tumors. Here, we showed that Par6 is an essential component in glioma tumorigenesis. Our results indicated the aberrant expression of Par6 in malignant glioma tissues and cell lines. We found that the regulation of Par6 expression induces cell proliferation and tumor growth in vivo and in vitro. Additionally, RNA-seq revealed the effects of Par6 were associated with cyclin D1-regulated cell cycle progression in glioma cells. Moreover, our results demonstrated that the regulation of Par6 can enhance the activation of Akt/PI3K signaling pathway, and subsequently upregulate the expression level of GSK-3β protein, which then regulate cyclin D1-mediated cell cycle regulation. Furthermore, we found that TGF-β-induced the upregulation of Par6 expression may be involved in this process. The pathological analysis confirmed the correlation between Par6 expression and the prognosis in human glioma tissues, suggesting the regulation of Par6 expression regulates glioma tumorigenesis and progression. Thus, our findings showed that Par6 might be a potential biomarker for the diagnosis and providing a therapeutic strategy for the treatment of malignant glioma.
Collapse
Affiliation(s)
- Pei Liu
- Neuroscience Center, Shantou University Medical College, Shantou, China
| | - Chenchen Zhu
- Neuroscience Center, Shantou University Medical College, Shantou, China
| | - Juanjuan Luo
- Neuroscience Center, Shantou University Medical College, Shantou, China
| | - Sheng Lan
- Neuroscience Center, Shantou University Medical College, Shantou, China
| | - Dongsheng Su
- Neuroscience Center, Shantou University Medical College, Shantou, China
| | - Qiongjin Wang
- Neuroscience Center, Shantou University Medical College, Shantou, China
| | - Zhe Wei
- Faculty of Medicine and Health, Lishui University, Lishui, China
| | - Wei Cui
- Department of Pharmacology, College of Life Science and Biopharmaceutical, Shenyang Pharmaceutical University, Shenyang, China
| | - Chuan Xu
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaojun Yang
- Neuroscience Center, Shantou University Medical College, Shantou, China
| |
Collapse
|
31
|
Joung EK, Kim J, Yoon N, Maeng LS, Kim JH, Park S, Kang K, Kim JS, Ahn YH, Ko YH, Byun JH, Hong JH. Expression of EEF1A1 Is Associated with Prognosis of Patients with Colon Adenocarcinoma. J Clin Med 2019; 8:jcm8111903. [PMID: 31703307 PMCID: PMC6912729 DOI: 10.3390/jcm8111903] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 10/31/2019] [Accepted: 11/04/2019] [Indexed: 01/06/2023] Open
Abstract
Background: The prognostic role of the translational factor, elongation factor-1 alpha 1 (EEF1A1), in colon cancer is unclear. Objectives: The present study aimed to investigate the expression of EEF1A in tissues obtained from patients with stage II and III colon cancer and analyze its association with patient prognosis. Methods: A total of 281 patients with colon cancer who underwent curative resection were analyzed according to EEF1A1 expression. Results: The five-year overall survival in the high-EEF1A1 group was 87.7%, whereas it was 65.6% in the low-EEF1A1 expression group (hazard ratio (HR) 2.47, 95% confidence interval (CI) 1.38–4.44, p = 0.002). The five-year disease-free survival of patients with high EEF1A1 expression was 82.5%, which was longer than the rate of 55.4% observed for patients with low EEF1A1 expression (HR 2.94, 95% CI 1.72–5.04, p < 0.001). Univariate Cox regression analysis indicated that age, preoperative carcinoembryonic antigen level, adjuvant treatment, total number of metastatic lymph nodes, and EEF1A1 expression level were significant prognostic factors for death. In multivariate analysis, expression of EEF1A1 was an independent prognostic factor associated with death (HR 3.01, 95% CI 1.636–5.543, p < 0.001). EEF1A1 expression was also an independent prognostic factor for disease-free survival in multivariate analysis (HR 2.54, 95% CI 1.459–4.434, p < 0.001). Conclusions: Our study demonstrated that high expression of EEF1A1 has a favorable prognostic effect on patients with colon adenocarcinoma.
Collapse
Affiliation(s)
- Eun kyo Joung
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
| | - Jiyoung Kim
- Department of Pathology, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (J.K.); (N.Y.); (L.-s.M.)
| | - Nara Yoon
- Department of Pathology, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (J.K.); (N.Y.); (L.-s.M.)
| | - Lee-so Maeng
- Department of Pathology, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (J.K.); (N.Y.); (L.-s.M.)
| | - Ji Hoon Kim
- Department of General Surgery, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
| | | | - Keunsoo Kang
- Department of Microbiology, College of Natural Sciences, Dankook University, Cheonan 31116, Korea;
| | - Jeong Seon Kim
- Department of Molecular Medicine and Tissue Injury Defense Research Center, College of Medicine, Ewha Womans University, Seoul 03760, Korea; (J.S.K.); (Y.-H.A.)
| | - Young-Ho Ahn
- Department of Molecular Medicine and Tissue Injury Defense Research Center, College of Medicine, Ewha Womans University, Seoul 03760, Korea; (J.S.K.); (Y.-H.A.)
| | - Yoon Ho Ko
- Division of Oncology, Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, The Catholic University of Korea, Seoul 03312, Korea;
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Jae Ho Byun
- Division of Oncology, Department of Internal Medicine, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Correspondence: (J.H.B.); (J.H.H.)
| | - Ji Hyung Hong
- Division of Oncology, Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, The Catholic University of Korea, Seoul 03312, Korea;
- Correspondence: (J.H.B.); (J.H.H.)
| |
Collapse
|
32
|
An JC, Shi HB, Hao WB, Zhu K, Ma B. miR-944 inhibits lung adenocarcinoma tumorigenesis by targeting STAT1 interaction. Oncol Lett 2019; 17:3790-3798. [PMID: 30881499 PMCID: PMC6403514 DOI: 10.3892/ol.2019.10045] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 10/22/2018] [Indexed: 12/19/2022] Open
Abstract
Lung adenocarcinoma (LAC) is a leading cause of cancer-associated mortalities, particularly in developed countries. The aberrant expression of microRNAs (miRNAs) has been proven to regulate numerous diseases in the past two decades. miRNAs have been identified in almost all human cancer types. In the present study, the role of miR-944 in LAC proliferation was examined. It was identified that miR-944 was downregulated in LAC tissues and cells, and miR-944 overexpression inhibited A549 and H1299 cell proliferation, as determined by the Cell Counting Kit-8 and colony formation assay. Signal transducer and activator of transcription 1 (STAT1) was upregulated in LAC tissues and cells. Kaplan-Meier analysis demonstrated that the 5-year overall survival in patients with high STAT1 levels was significantly reduced, compared with patients with negative and low STAT1 expression. STAT1 was the direct target of miR-944. Additionally, a miR-944 mimic inhibited A549 cell growth in vitro. Collectively, these data demonstrate that miR-944 serves a pivotal role in LAC tumor growth by targeting STAT1. The data obtained indicated that miR-944 may be a novel biomarker and could result in potential therapies for LAC.
Collapse
Affiliation(s)
- Jing Chun An
- Department of Respiratory Medicine, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161000, P.R. China
| | - Han-Bing Shi
- Department of Respiratory Medicine, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161000, P.R. China
| | - Wen-Bo Hao
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161000, P.R. China
| | - Kun Zhu
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161000, P.R. China
| | - Bo Ma
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161000, P.R. China
| |
Collapse
|
33
|
Network Pharmacology and Bioinformatics Approach Reveals the Therapeutic Mechanism of Action of Baicalein in Hepatocellular Carcinoma. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:7518374. [PMID: 30891079 PMCID: PMC6390240 DOI: 10.1155/2019/7518374] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 01/09/2019] [Indexed: 12/29/2022]
Abstract
Liver cancer is the fourth leading cause of cancer death worldwide, and hepatocellular carcinoma (HCC) accounts for the greatest proportion of these deaths. Baicalein, a flavonoid isolated from the root of Scutellariae radix, is considered a potential candidate to treat HCC. However, the underlying molecular mechanisms remain poorly understood. In the present study, a network pharmacological approach was combined with microarray data (GSE95504) acquired from the Gene Expression Omnibus database to reveal the therapeutic mechanisms of action of baicalein at a systemic level. We identified 38 baicalein targets and 76 differently expressed genes (DEGs) following treatment with baicalein, including 55 upregulated and 21 downregulated genes. The DEGs were significantly enriched in the biological functions of apoptosis, endoplasmic reticulum stress, and PERK-mediated unfolded protein response. Protein-protein interaction (PPI) network construction and topological screening revealed a core module of PPIs including two baicalein targets, TP53 and CDK1, and two downregulated DEGs, HSPA1A and HSPA1B. Expression and survival data for these genes in the module derived from Gene Expression Profiling Interactive Analysis (GEPIA) were subjected to Kaplan–Meier analysis of overall survival and disease-free survival. Overexpression of CDK1, BRCA1, TUBB, HSPA1A, HSPA1B, and HSPA4 was associated with significantly worse overall survival, while overexpression of CDK1, CLU7, BRCA1, and TUBB was associated with significantly worse disease-free survival. These data suggest that baicalein exerts therapeutic effects against HCC via a PPI network involving TP53, CDK1, HSPA1A, and HSPA1B.
Collapse
|
34
|
Liu Z, Tian Z, Cao K, Zhang B, Wen Q, Zhou X, Yang W, Wang T, Shi H, Wang R. TSG101 promotes the proliferation, migration and invasion of hepatocellular carcinoma cells by regulating the PEG10. J Cell Mol Med 2018; 23:70-82. [PMID: 30450735 PMCID: PMC6307771 DOI: 10.1111/jcmm.13878] [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: 04/20/2018] [Revised: 07/11/2018] [Accepted: 08/03/2018] [Indexed: 01/19/2023] Open
Abstract
The tumour susceptibility gene 101 (TSG101) is reported to play important roles in the development and progression of several human cancers. However, its potential roles and underlined mechanisms in human hepatocellular carcinoma (HCC) are still needed to be further clarified. In the present study, we reported that knock down of TSG101 suppressed the proliferation, migration and invasion of HCC cells, while overexpression of TSG101 facilitated them. Molecularly, the results revealed that knock down of TSG101 significantly decreased the cell cycle related regulatory factor p53 and p21. In another point, knock down of TSG101 also obviously decreased the level of metallopeptidase inhibitor TIMP1 (Tissue inhibitors of metalloproteinases 1), which results in inhibition of MMP2, MMP7 and MMP9. In contrast, overexpression of TSG101 had opposite effects. The iTRAQ proteomics analysis identified that oncogenic protein PEG10 (Paternally expressed gene 10) might be a potential downstream target of TSG101. Further investigation showed that TSG101 interacted with PEG10 and protected it from proteasomal degradation thereby regulating the expression of p53, p21 and MMPs. Finally, we found that both TSG101 and PEG10 proteins are up-regulated and presented a direct correlation in HCC patients. In conclusion, these results suggest that TSG101 is up-regulated in human HCC patients, which may accelerate the proliferation, migration and invasion of HCC cells through regulating PEG10.
Collapse
Affiliation(s)
- Zhiyi Liu
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China.,The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zilu Tian
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China.,The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Kuan Cao
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of General Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Bin Zhang
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of General Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Quan Wen
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of General Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xinyu Zhou
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Weibin Yang
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China.,The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Tao Wang
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China.,The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hengliang Shi
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of General Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Renhao Wang
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of General Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
35
|
Murphy N, Shen J, Shih A, Liew A, Khalili H, Yaskiv O, Katona K, Lee A, Zhu XH. Paraneoplastic Syndrome Secondary to Treatment Emergent Neuroendocrine Tumor in Metastatic Castration-resistant Prostate Cancer: A Unique Case. Clin Genitourin Cancer 2018; 17:e56-e60. [PMID: 30279116 DOI: 10.1016/j.clgc.2018.09.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 09/01/2018] [Indexed: 12/16/2022]
Affiliation(s)
- Neal Murphy
- Department of Internal Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY
| | - Janice Shen
- Department of Internal Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY
| | - Andrew Shih
- Feinstein Institute for Medical Research, Manhasset, NY
| | - Anthony Liew
- Feinstein Institute for Medical Research, Manhasset, NY
| | | | - Oksana Yaskiv
- Northwell Health Department of Pathology, New Hyde Park, NY
| | - Kyle Katona
- Department of Internal Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY
| | - Annette Lee
- Feinstein Institute for Medical Research, Manhasset, NY; Northwell Health Cancer Institute, Lake Success, NY; Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY
| | - Xin-Hua Zhu
- Feinstein Institute for Medical Research, Manhasset, NY; Northwell Health Cancer Institute, Lake Success, NY.
| |
Collapse
|
36
|
Contradictory mRNA and protein misexpression of EEF1A1 in ductal breast carcinoma due to cell cycle regulation and cellular stress. Sci Rep 2018; 8:13904. [PMID: 30224719 PMCID: PMC6141510 DOI: 10.1038/s41598-018-32272-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 09/05/2018] [Indexed: 02/08/2023] Open
Abstract
Encoded by EEF1A1, the eukaryotic translation elongation factor eEF1α1 strongly promotes the heat shock response, which protects cancer cells from proteotoxic stress, following for instance oxidative stress, hypoxia or aneuploidy. Unexpectedly, therefore, we find that EEF1A1 mRNA levels are reduced in virtually all breast cancers, in particular in ductal carcinomas. Univariate and multivariate analyses indicate that EEF1A1 mRNA underexpression independently predicts poor patient prognosis for estrogen receptor-positive (ER+) cancers. EEF1A1 mRNA levels are lowest in the most invasive, lymph node-positive, advanced stage and postmenopausal tumors. In sharp contrast, immunohistochemistry on 100 ductal breast carcinomas revealed that at the protein level eEF1α1 is ubiquitously overexpressed, especially in ER+ , progesterone receptor-positive and lymph node-negative tumors. Explaining this paradox, we find that EEF1A1 mRNA levels in breast carcinomas are low due to EEF1A1 allelic copy number loss, found in 27% of tumors, and cell cycle-specific expression, because mRNA levels are high in G1 and low in proliferating cells. This also links estrogen-induced cell proliferation to clinical observations. In contrast, high eEF1α1 protein levels protect tumor cells from stress-induced cell death. These observations suggest that, by obviating EEF1A1 transcription, cancer cells can rapidly induce the heat shock response following proteotoxic stress, and survive.
Collapse
|
37
|
Chen J, Song W, Du Y, Li Z, Xuan Z, Zhao L, Chen J, Zhao Y, Tuo B, Zheng S, Song P. Inhibition of KLHL21 prevents cholangiocarcinoma progression through regulating cell proliferation and motility, arresting cell cycle and reducing Erk activation. Biochem Biophys Res Commun 2018; 499:433-440. [PMID: 29574153 DOI: 10.1016/j.bbrc.2018.03.152] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 03/20/2018] [Indexed: 01/06/2023]
Abstract
Kelch-like family member 21 (KLHL21) is involved in cell mitosis and motility. Nevertheless, the clinical significance and biological function of KLHL21 in cholangiocarcinoma (CCA) are elusive. This is the first study to describe a pivotal role for KLHL21 in the progression of CCA. The expression of KLHL21 was elevated in CCA tissues compared with paired normal bile duct tissues. In addition, immunohistochemical and statistical analyses demonstrated that the expression of KLHL21 correlated inversely with tumor histological grade (p < 0.05) and the overall survival of patients (p < 0.01). In CCA cells, we found that the inhibition of KLHL21 significantly reduced proliferation, migration and invasion. Further results indicated that inhibition of KLHL21 triggered G0/G1 cell cycle arrest, leading to the increased expression of P21 and P27 and decreased expression of Cyclin E1, which eventually resulted in proliferation suppression in CCA cells. Furthermore, KLHL21 knockdown alleviated the activation of the Erk signaling pathway via decreasing the expression of phospho-Erk1/2. Our data demonstrated that KLHL21 plays an essential role in the tumorigenesis and progression of CCA, implying that it might serve as a potential therapeutic target for CCA treatment.
Collapse
Affiliation(s)
- Jian Chen
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, China; NHFPC Key Laboratory of Combined Multi-organ Transplantation, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, China; Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou 310003, China; Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, China
| | - Wenfeng Song
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, China; NHFPC Key Laboratory of Combined Multi-organ Transplantation, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, China; Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou 310003, China; Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, China
| | - Yehui Du
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, China; NHFPC Key Laboratory of Combined Multi-organ Transplantation, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, China; Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou 310003, China; Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, China
| | - Zequn Li
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, China; NHFPC Key Laboratory of Combined Multi-organ Transplantation, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, China; Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou 310003, China; Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, China
| | - Zefeng Xuan
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, China; NHFPC Key Laboratory of Combined Multi-organ Transplantation, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, China; Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou 310003, China; Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, China
| | - Long Zhao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, China; NHFPC Key Laboratory of Combined Multi-organ Transplantation, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, China; Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou 310003, China; Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, China
| | - Jun Chen
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, China; NHFPC Key Laboratory of Combined Multi-organ Transplantation, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, China; Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou 310003, China; Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, China
| | - Yongchao Zhao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, China; NHFPC Key Laboratory of Combined Multi-organ Transplantation, China; Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China
| | - Biguang Tuo
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical College, Zunyi 563003, China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, China; NHFPC Key Laboratory of Combined Multi-organ Transplantation, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, China; Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou 310003, China; Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, China.
| | - Penghong Song
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, China; NHFPC Key Laboratory of Combined Multi-organ Transplantation, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, China; Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou 310003, China; Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, China.
| |
Collapse
|
38
|
GRP78 Promotes Hepatocellular Carcinoma proliferation by increasing FAT10 expression through the NF-κB pathway. Exp Cell Res 2018; 365:1-11. [PMID: 29458176 DOI: 10.1016/j.yexcr.2018.02.007] [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] [Received: 01/06/2018] [Revised: 02/06/2018] [Accepted: 02/13/2018] [Indexed: 12/21/2022]
Abstract
Glucose-regulated protein 78(GRP78) and the ubiquitin-like protein FAT10 each promote proliferation in hepatocellular carcinoma(HCC). However, the relationship of GRP78 and FAT10 in HCC proliferation are still not known. In this study, we found that GRP78 and FAT10 were significantly overexpressed in HCC tissues compare with adjacent non-cancerous tissues, and a positive correlation was found between their expression and associated proliferation characteristics. High expression of GRP78 and FAT10 were positively correlated with tumor proliferation and poor prognosis in HCC. Moreover, GRP78 knockdown reduced FAT10 expression and suppressed HCC proliferation in vitro and in vivo. The effects of GRP78 knockdown were rescued by FAT10 up-regulation, whereas FAT10 knockdown reduced HCC proliferation enhanced by GRP78 up-regulation. Furthermore, GRP78 modulated FAT10 expression by regulating the NF-κB pathway, direct activation of the NF-κB pathway increased the expression of FAT10, a gene counteracting the tumor suppressor p53. Taken together, these results suggest that this newly identified GRP78-NF-κB-FAT10 axis will provide novel insight into the understanding of the regulatory mechanisms of proliferation in human HCC.
Collapse
|