1
|
Zhang M, Bai J, Yuan H, Duan X, Yu L, Li Y, Li K, Rile S, Wang X, Wang H, Liu P, Yan J, Wang C. BRD1 deficiency affects SREBF1-related lipid metabolism through regulating H3K9ac/H3K9me3 transition to inhibit HCC progression. Cell Death Dis 2025; 16:104. [PMID: 39962068 PMCID: PMC11833140 DOI: 10.1038/s41419-025-07404-7] [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: 06/19/2024] [Revised: 01/13/2025] [Accepted: 01/29/2025] [Indexed: 02/20/2025]
Abstract
BRD1 encodes a protein containing a bromodomain, which is an essential component of histone acetyltransferase (HAT) complexes. These complexes play a crucial role in the regulation of gene transcription and the modification of chromatin structures. The aberrant expression of BRD1 is frequently observed across a range of cancer types, including hepatocellular carcinomas (HCC). However, the precise mechanisms through which BRD1 contributes to tumorigenesis, especially in HCC, remain unclear. In our investigation, we have uncovered a novel role for BRD1 as an oncogene implicated the regulation of lipid metabolism in HCC progression. Specifically, the deficiency of BRD1 impedes the proliferation and metastasis of HCC cells reducing the accumulation of lipid droplets and cholesterol levels. This effect is mediated through the SREBF1-induced downregulation of SCD1 expression in HCC cells. Mechanistically, the ablation of BRD1 disrupts acetylation level of H3K9, culminating in the subsequent trimethylation of H3K9 (H3K9me3). Notably, the H3K14ac partially colocalizes with H3K9me3 and its methyltransferase SETDB1 to from a double labeling of both H3K14ac and H3K9me3 at the SREBF1 promoter. This double labeling contributes to the creation of a repressive environment, ultimately leading to the downregulation of SREBF1 gene expression in HCC. Furthermore, the combinatorial use of a BRD1 inhibitor and simvastatin augments antitumor efficacy in vivo. Collectively, our findings underscore BRD1 as a critical regulator of SREBF1-associated lipid metabolism and a participant in HCC progression through a distinct epigenetic regulatory mechanism. These discoveries further suggest a promising epigenetic therapeutic approach for the treatment of HCC.
Collapse
Affiliation(s)
- Mingyang Zhang
- College of Life Science, Inner Mongolia University, Xi Lin Guo Le south Road 49, Yu Quan District, Hohhot, Inner Mongolia, China
| | - Jing Bai
- College of Life Science, Inner Mongolia University, Xi Lin Guo Le south Road 49, Yu Quan District, Hohhot, Inner Mongolia, China
| | - Hengye Yuan
- College of Life Science, Inner Mongolia University, Xi Lin Guo Le south Road 49, Yu Quan District, Hohhot, Inner Mongolia, China
| | - Xiaojun Duan
- College of Life Science, Inner Mongolia University, Xi Lin Guo Le south Road 49, Yu Quan District, Hohhot, Inner Mongolia, China
- School of Basic Medicine, Inner Mongolia Medical University, Xin hua Street No. 5, Hui min District, Hohhot, Inner Mongolia, China
| | - Lei Yu
- College of Life Science, Inner Mongolia University, Xi Lin Guo Le south Road 49, Yu Quan District, Hohhot, Inner Mongolia, China
| | - Yu Li
- College of Life Science, Inner Mongolia University, Xi Lin Guo Le south Road 49, Yu Quan District, Hohhot, Inner Mongolia, China
| | - Kexin Li
- College of Life Science, Inner Mongolia University, Xi Lin Guo Le south Road 49, Yu Quan District, Hohhot, Inner Mongolia, China
| | - Saqi Rile
- College of Life Science, Inner Mongolia University, Xi Lin Guo Le south Road 49, Yu Quan District, Hohhot, Inner Mongolia, China
| | - Xinran Wang
- First School of Clinical Medicine, Inner Mongolia Medical University, Xin hua Street No. 5, Hui min District, Hohhot, Inner Mongolia, China
| | - Haisheng Wang
- School of Basic Medicine, Inner Mongolia Medical University, Xin hua Street No. 5, Hui min District, Hohhot, Inner Mongolia, China
| | - Pengxia Liu
- College of Life Science, Inner Mongolia University, Xi Lin Guo Le south Road 49, Yu Quan District, Hohhot, Inner Mongolia, China
| | - Jia Yan
- School of Basic Medicine, Inner Mongolia Medical University, Xin hua Street No. 5, Hui min District, Hohhot, Inner Mongolia, China.
- Medical Experimental Center of Basic Medical School, Inner Mongolia Medical University, Xin hua Street No. 5, Hui min District, Hohhot, Inner Mongolia, China.
| | - Changshan Wang
- College of Life Science, Inner Mongolia University, Xi Lin Guo Le south Road 49, Yu Quan District, Hohhot, Inner Mongolia, China.
| |
Collapse
|
2
|
Dong W, Wang H, Li M, Li P, Ji S. Virus-induced host genomic remodeling dysregulates gene expression, triggering tumorigenesis. Front Cell Infect Microbiol 2024; 14:1359766. [PMID: 38572321 PMCID: PMC10987825 DOI: 10.3389/fcimb.2024.1359766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 03/01/2024] [Indexed: 04/05/2024] Open
Abstract
Virus-induced genomic remodeling and altered gene expression contribute significantly to cancer development. Some oncogenic viruses such as Human papillomavirus (HPV) specifically trigger certain cancers by integrating into the host's DNA, disrupting gene regulation linked to cell growth and migration. The effect can be through direct integration of viral genomes into the host genome or through indirect modulation of host cell pathways/proteins by viral proteins. Viral proteins also disrupt key cellular processes like apoptosis and DNA repair by interacting with host molecules, affecting signaling pathways. These disruptions lead to mutation accumulation and tumorigenesis. This review focuses on recent studies exploring virus-mediated genomic structure, altered gene expression, and epigenetic modifications in tumorigenesis.
Collapse
Affiliation(s)
- Weixia Dong
- Department of Basic Medicine, Zhengzhou Shuqing Medical College, Zhengzhou, Henan, China
| | - Huiqin Wang
- Department of Basic Medicine, Zhengzhou Shuqing Medical College, Zhengzhou, Henan, China
| | - Menghui Li
- Department of Basic Medicine, Zhengzhou Shuqing Medical College, Zhengzhou, Henan, China
| | - Ping Li
- Department of Basic Medicine, Zhengzhou Shuqing Medical College, Zhengzhou, Henan, China
| | - Shaoping Ji
- Department of Basic Medicine, Zhengzhou Shuqing Medical College, Zhengzhou, Henan, China
- Department of Biochemistry and Molecular Biology, Medical School, Henan University, Kaifeng, Henan, China
| |
Collapse
|
3
|
Park H, Lee S, Lee J, Moon H, Ro SW. Exploring the JAK/STAT Signaling Pathway in Hepatocellular Carcinoma: Unraveling Signaling Complexity and Therapeutic Implications. Int J Mol Sci 2023; 24:13764. [PMID: 37762066 PMCID: PMC10531214 DOI: 10.3390/ijms241813764] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/04/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
Hepatocellular Carcinoma (HCC) continues to pose a substantial global health challenge due to its high incidence and limited therapeutic options. In recent years, the Janus Kinase (JAK) and Signal Transducer and Activator of Transcription (STAT) pathway has emerged as a critical signaling cascade in HCC pathogenesis. The review commences with an overview of the JAK/STAT pathway, delving into the dynamic interplay between the JAK/STAT pathway and its numerous upstream activators, such as cytokines and growth factors enriched in pathogenic livers afflicted with chronic inflammation and cirrhosis. This paper also elucidates how the persistent activation of JAK/STAT signaling leads to diverse oncogenic processes during hepatocarcinogenesis, including uncontrolled cell proliferation, evasion of apoptosis, and immune escape. In the context of therapeutic implications, this review summarizes recent advancements in targeting the JAK/STAT pathway for HCC treatment. Preclinical and clinical studies investigating inhibitors and modulators of JAK/STAT signaling are discussed, highlighting their potential in suppressing the deadly disease. The insights presented herein underscore the necessity for continued research into targeting the JAK/STAT signaling pathway as a promising avenue for HCC therapy.
Collapse
Affiliation(s)
| | | | | | | | - Simon Weonsang Ro
- Department of Genetics and Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si 17104, Republic of Korea; (H.P.); (S.L.); (J.L.); (H.M.)
| |
Collapse
|
4
|
Pan J, Li D, Fan X, Cheng J, Jin S, Chen P, Lin H, Li Y. Aberrant DNA Methylation Patterns of Deleted in Liver Cancer 1 Isoforms in Hepatocellular Carcinoma. DNA Cell Biol 2023; 42:140-150. [PMID: 36917700 DOI: 10.1089/dna.2022.0384] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Hepatocellular carcinoma (HCC), a common primary liver cancer, is the third leading cause of death worldwide. DNA methylation changes are common in HCC and have been studied to be associated with hepatocarcinogenesis. In our study, we used the MassARRAY® EpiTYPER technology to investigate the methylation differences of deleted in liver cancer 1 (DLC1) (isoform 1 and 3) promoter between HCC tissues and corresponding adjacent noncancerous tissues and the association between methylation levels and clinicopathological features. In addition, the modified CRISPR-Cas9 system and the DNA methyltransferase inhibitor (DNMTi) were utilized to explore the functional correlation of epigenetic modifications and DLC1 gene regulation. The methylation levels of the DLC1 isoforms in HCC samples were found significantly lower than those in the adjacent noncancerous tissues (all p < 0.0001). Also, we found that the expression of DLC1 could be bidirectionally regulated by the modified CRISPR-Cas9 system and the DNMTi. Moreover, the hypomethylation of DLC1 in HCC samples was connected with the presence of satellite lesions (p = 0.0305) and incomplete tumor capsule (p = 0.0204). Receiver operator characteristic curve analysis demonstrated that the methylation levels of DLC1 could be applied to discriminate HCC patients (area under the curve = 0.728, p < 0.0001). The hypomethylation status was a key regulatory mechanism of DLC1 expression and might serve as a potential biomarker for HCC.
Collapse
Affiliation(s)
- Junhai Pan
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Duguang Li
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaoxiao Fan
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- State Key Laboratory of Modern Optical Instrumentations, Centre for Optical and Electromagnetic Research, College of Optical Science and Engineering, International Research Center for Advanced Photonics, Zhejiang University, Hangzhou, China
| | - Jiaxi Cheng
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shengxi Jin
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Peng Chen
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Hui Lin
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
- Zhejiang Engineering Research Center of Cognitive Healthcare, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou,China
| | - Yirun Li
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
5
|
Liu HH, Wang J, Zhang Y, Fan YC, Wang K. Prognostic potential of the small GTPase Ran and its methylation in hepatocellular carcinoma. Hepatobiliary Pancreat Dis Int 2022; 21:248-256. [PMID: 35367146 DOI: 10.1016/j.hbpd.2022.03.011] [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: 01/05/2022] [Accepted: 03/03/2022] [Indexed: 02/05/2023]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a common malignant tumor with high mortality. The prognostic significance of Ran, a member of Ras superfamily, remains unclear in HCC patients. METHODS Based on The Cancer Genome Atlas (TCGA) database and Tumor Immune Estimation Resource (TIMER), we analyzed the correlations among Ran expression, promoter methylation and immune cell infiltration. We also investigated the Ran expression levels in HCC tissues and normal tissues by using quantitative real-time PCR. RESULTS Ran mRNA expression was significantly increased in HCC tissues compared with the normal tissues (P < 0.001). Time-dependent receiver operating characteristic (ROC) curves showed that Ran expression had predictive value of the 1-, 3- and 5-year overall survival for HCC patients, and the areas under the curves (AUC) were 0.747, 0.634 and 0.704, respectively. Cox regression analysis showed that Ran expression was an independent prognostic factor for HCC patients (HR = 1.492, 95% CI: 1.129-1.971, P = 0.005). We also found a negative relationship between Ran mRNA expression and its promoter methylation (r = -0.36, P < 0.001). High Ran expression and promoter hypomethylation predicted worse overall survival and progression-free survival (P < 0.05) and were involved in the progression of HCC. Ran expression exhibited significant correlations with immune infiltrates and prognostic immune-related genes. CONCLUSIONS The present study provides further insight into the prognosis of HCC, and Ran could serve as a biomarker for predicting the survival of HCC patients.
Collapse
Affiliation(s)
- Hui-Hui Liu
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250000, China
| | - Ju Wang
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250000, China
| | - Ying Zhang
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250000, China
| | - Yu-Chen Fan
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250000, China; Shenzhen Research Institute of Shandong University, Shenzhen 518000, China; Institute of Hepatology, Shandong University, Jinan 250000, China
| | - Kai Wang
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250000, China; Shenzhen Research Institute of Shandong University, Shenzhen 518000, China; Institute of Hepatology, Shandong University, Jinan 250000, China.
| |
Collapse
|
6
|
Sun D, Gan X, Liu L, Yang Y, Ding D, Li W, Jiang J, Ding W, Zhao L, Hou G, Yu J, Wang J, Yang F, Yuan S, Zhou W. DNA hypermethylation modification promotes the development of hepatocellular carcinoma by depressing the tumor suppressor gene ZNF334. Cell Death Dis 2022; 13:446. [PMID: 35534462 PMCID: PMC9085879 DOI: 10.1038/s41419-022-04895-6] [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: 12/18/2021] [Revised: 04/19/2022] [Accepted: 04/28/2022] [Indexed: 12/14/2022]
Abstract
DNA methylation plays a pivotal role in the development and progression of tumors. However, studies focused on the dynamic changes of DNA methylation in the development of hepatocellular carcinoma (HCC) are rare. To systematically illustrate the dynamic DNA methylation alternation from premalignant to early-stage liver cancer with the same genetic background, this study enrolled 5 HBV-related patients preceded with liver cirrhosis, pathologically identified as early-stage HCC with dysplastic nodules. Liver fibrosis tissues, dysplastic nodules and early HCC tissues from these patients were used to measure DNA methylation. Here, we report significant differences in the DNA methylation spectrum among the three types of tissues. In the early stage of HCC, DNA hypermethylation of tumor suppressor genes is predominant. Additionally, DNA hypermethylation in the early stage of HCC changes the binding ability of transcription factor TP53 to the promoter of tumor suppressor gene ZNF334, and inhibits the expression of ZNF334 at the transcription level. Furthermore, through a series of in vivo and in vitro experiments, we have clarified the exacerbation effect of tumor suppressor gene ZNF334 deletion in the occurrence of HCC. Combined with clinical data, we found that the overall survival and relapse-free survival of patients with high ZNF334 expression are significantly longer. Thus, we partly elucidated a sequential alternation of DNA methylation modification during the occurrence of HCC, and clarified the biological function and regulatory mechanism of the tumor suppressor gene ZNF334, which is regulated by related DNA methylation sites. Our study provides a new target and clinical evidence for the early diagnosis and sheds light on the precise treatment of liver cancer.
Collapse
Affiliation(s)
- Dapeng Sun
- grid.73113.370000 0004 0369 1660The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, 225 Changhai Road Shanghai, Shanghai, 200438 China
| | - Xiaojie Gan
- grid.73113.370000 0004 0369 1660The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, 225 Changhai Road Shanghai, Shanghai, 200438 China
| | - Lei Liu
- grid.73113.370000 0004 0369 1660The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, 225 Changhai Road Shanghai, Shanghai, 200438 China
| | - Yuan Yang
- grid.73113.370000 0004 0369 1660The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, 225 Changhai Road Shanghai, Shanghai, 200438 China
| | - Dongyang Ding
- grid.73113.370000 0004 0369 1660The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, 225 Changhai Road Shanghai, Shanghai, 200438 China
| | - Wen Li
- grid.73113.370000 0004 0369 1660The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, 225 Changhai Road Shanghai, Shanghai, 200438 China
| | - Junyao Jiang
- grid.428926.30000 0004 1798 2725Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou, 510530 China
| | - Wenbin Ding
- grid.73113.370000 0004 0369 1660The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, 225 Changhai Road Shanghai, Shanghai, 200438 China
| | - Linghao Zhao
- grid.73113.370000 0004 0369 1660The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, 225 Changhai Road Shanghai, Shanghai, 200438 China
| | - Guojun Hou
- grid.73113.370000 0004 0369 1660The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, 225 Changhai Road Shanghai, Shanghai, 200438 China
| | - Jian Yu
- grid.73113.370000 0004 0369 1660The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, 225 Changhai Road Shanghai, Shanghai, 200438 China
| | - Jie Wang
- grid.428926.30000 0004 1798 2725Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou, 510530 China
| | - Fu Yang
- grid.73113.370000 0004 0369 1660The department of Medical Genetics, Naval Medical University, Shanghai, 200438 China
| | - Shengxian Yuan
- grid.73113.370000 0004 0369 1660The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, 225 Changhai Road Shanghai, Shanghai, 200438 China
| | - Weiping Zhou
- grid.73113.370000 0004 0369 1660The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, 225 Changhai Road Shanghai, Shanghai, 200438 China
| |
Collapse
|
7
|
Ren SH, Qin YF, Qin H, Wang HD, Li GM, Zhu YL, Sun CL, Shao B, Zhang JY, Hao JP, Wang H. N6-Methyladenine-Related Signature for Immune Microenvironment and Response to Immunotherapy in Hepatocellular Carcinoma. Int J Gen Med 2022; 15:3525-3540. [PMID: 35386863 PMCID: PMC8978579 DOI: 10.2147/ijgm.s351815] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 02/21/2022] [Indexed: 12/31/2022] Open
Abstract
Background The prognostic value of m6A-related genes in hepatocellular carcinoma (HCC) and its correlation with the immune microenvironment still requires further investigation. Methods Consensus clustering by m6A related genes was used to classify 374 patients with HCC from The Cancer Genome Atlas (TCGA) database. Then we performed the least absolute shrinkage and selection operator (LASSO) to construct the m6A related genes model. The International Cancer Genome Consortium (ICGC) and Gene Expression Omnibus (GEO) datasets were used to verify and evaluate the model. ESTIMATE, CIBERSORTx, the expression levels of immune checkpoint genes, and TIDE were used to investigate the tumor microenvironment (TME) and the response to immunotherapy. Gene set enrichment analyses (GSEA), tumor-associated macrophages (TAMs), and gene-drug sensitivity were also analyzed. Results By expression value and regression coefficient of five m6A related genes, we constructed the risk score of each patient. The patients with a higher risk score had a considerably poorer prognosis in the primary and validated cohort. For further discussing TME and the response to immunotherapy, we divided the entire set into two groups based on the risk score. Our findings implied that the tumor-infiltrating lymphocytes (TILs) were proportional to the risk scores, which seemed to contradict that patients with higher scores had a poor prognosis. Further, we found that the high-risk group had higher expression of PD-L1, CTLA-4, and PDCD1, indicating immune dysfunction, which may be a fundamental reason for poor prognosis. This was further reinforced by the fact that the low-risk group responded better than the high-risk group to monotherapy and combination therapy. Conclusion The m6A related risk score is a new independent prognostic factor that correlates with immunotherapy response. It can provide a new therapeutic strategy for improving individual immunotherapy in HCC.
Collapse
Affiliation(s)
- Shao-Hua Ren
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.,Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Ya-Fei Qin
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.,Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Hong Qin
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.,Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Hong-da Wang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.,Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Guang-Ming Li
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.,Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Yang-Lin Zhu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.,Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Cheng-Lu Sun
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.,Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Bo Shao
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.,Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Jing-Yi Zhang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.,Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Jing-Peng Hao
- Department of Anorectal Surgery, The Second Hospital of Tianjin Medical University, Tianjin, People's Republic of China
| | - Hao Wang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.,Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| |
Collapse
|
8
|
Tang Y, Wang Y, Xu X, Sun H, Tang W. STEAP4 promoter methylation correlates with tumorigenesis of hepatocellular carcinoma. Pathol Res Pract 2022; 233:153870. [DOI: 10.1016/j.prp.2022.153870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 03/22/2022] [Accepted: 03/30/2022] [Indexed: 10/18/2022]
|
9
|
Tahoon A, El-Khateeb D, Mosbeh A, Tantawy El Sayed I, Khalil A. Significance of promoter methylation of multiple tumor suppressor genes in hepatocellular carcinoma. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2022. [DOI: 10.1186/s43042-022-00237-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Abstract
Background
Methylation of the promoter at CpG islands is a mechanism of silencing tumor suppressor genes and therefore enhances cancer progression. The study aimed to examine promoter methylation frequencies of five tumor suppressor genes in hepatocellular carcinoma and their implication on the first-year outcome of surgical resection of the tumor. Fifty specimens of hepatocellular carcinoma and the adjacent non-tumorous liver tissue were collected from the surgically resected hepatic tumor. The status of promoter methylation of tumor suppressor genes RASSF1A, CHFR, MGMT, GSTP1, and hMLH1 was investigated using methylation-specific polymerase chain reaction.
Results
The frequency of promoter methylation of these tumor suppressors genes (TSG) genes in hepatocellular carcinoma was significantly higher than non-tumorous tissue all, P < 0.05, with a methylation rate of 80% in RASSF1A, 70% in CHFR, 46% in GSTP1, 56% in MGMT, and 10% in hMLH1. Methylation of RASSF1A, CHFR, and MGMT promoter genes was significantly associated with decreased first-year postoperative survival and increased recurrence of hepatocellular carcinoma, P < 0.05.
Conclusion
Methylated RASSF1A, CHRF, and MGMT promoters indicated poor prognosis among patients with hepatocellular carcinoma and may serve as potential prognostic indicators in patients with hepatocellular carcinoma.
Collapse
|
10
|
Fan L, Xu L, Tian S, Zheng X. Identification of a novel histone phosphorylation prognostic signature in hepatocellular carcinoma based on bulk and single-cell RNA sequencing. Front Endocrinol (Lausanne) 2022; 13:965445. [PMID: 36120466 PMCID: PMC9470838 DOI: 10.3389/fendo.2022.965445] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 08/11/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the third leading cause of death in the world, characterized by high morbidity, poor prognosis and high mortality. Histone modifications regulate intracellular gene expression at the post-transcriptional level, and disturbances in the regulatory pattern of histone modifications at individual locus or across the genome can lead to tumorigenesis of HCC. In this study, we constructed a prognosis-related histone phosphorylation regulated (HPR) genes signature and elucidated whether HPR genes can predict overall survival in HCC patients. METHODS Differentially expressed genes were screened using TCGA, ICGC and GEO databases, and a new risk signature was constructed by univariate Cox regression and Lasso regression analysis. Predictive nomograms were established by multivariate Cox regression of risk scores and clinical parameters, calibration curve and decision curve analysis were used to evaluate the models. The ssGSEA methods were used to determine the effect of risk scores on the tumor immune microenvironment. Data for HCC single-cell RNA sequencing (scRNA-seq) have been downloaded from Gene Expression Omnibus (GEO) to understand the role of HPR genes in tumorigenesis. RESULTS Our analyses of nine HPR genes provided prognostic insights. Overall survival in the low-risk and high-risk groups was statistically higher, respectively (P<0.001). Cox regression analysis revealed that the risk score is a significant predictor of HCC outcomes (HR=2. 2.62, 95%CI: 1.248-5.514, P=0.011). In addition, a nomogram combining risk scores with TNM stages was constructed and tested from calibration curves and decision curves (AUC=0.780). MHC-class-I genes, iDCs, Macrophages, Tfh, Treg, Th2 were overexpressed in the high-risk group. CONCLUSION HPR genes risk score is closely related to the prognosis of HCC, tumor immune process and tumor cell progression.
Collapse
Affiliation(s)
- Lei Fan
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, China
| | - Ling Xu
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, China
| | - Shan Tian
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Zheng
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Xin Zheng,
| |
Collapse
|
11
|
Cai Q, Gan C, Tang C, Wu H, Gao J. Mechanism and Therapeutic Opportunities of Histone Modifications in Chronic Liver Disease. Front Pharmacol 2021; 12:784591. [PMID: 34887768 PMCID: PMC8650224 DOI: 10.3389/fphar.2021.784591] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 11/08/2021] [Indexed: 02/05/2023] Open
Abstract
Chronic liver disease (CLD) represents a global health problem, accounting for the heavy burden of disability and increased health care utilization. Epigenome alterations play an important role in the occurrence and progression of CLD. Histone modifications, which include acetylation, methylation, and phosphorylation, represent an essential part of epigenetic modifications that affect the transcriptional activity of genes. Different from genetic mutations, histone modifications are plastic and reversible. They can be modulated pharmacologically without changing the DNA sequence. Thus, there might be chances to establish interventional solutions by targeting histone modifications to reverse CLD. Here we summarized the roles of histone modifications in the context of alcoholic liver disease (ALD), metabolic associated fatty liver disease (MAFLD), viral hepatitis, autoimmune liver disease, drug-induced liver injury (DILI), and liver fibrosis or cirrhosis. The potential targets of histone modifications for translation into therapeutics were also investigated. In prospect, high efficacy and low toxicity drugs that are selectively targeting histone modifications are required to completely reverse CLD and prevent the development of liver cirrhosis and malignancy.
Collapse
Affiliation(s)
- Qiuyu Cai
- Laboratory of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, China
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Can Gan
- Laboratory of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, China
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Chengwei Tang
- Laboratory of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, China
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Hao Wu
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Jinhang Gao
- Laboratory of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, China
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
12
|
Qing X, Xu W, Zong J, Du X, Peng H, Zhang Y. Emerging treatment modalities for systemic therapy in hepatocellular carcinoma. Biomark Res 2021; 9:64. [PMID: 34419152 PMCID: PMC8380325 DOI: 10.1186/s40364-021-00319-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 08/05/2021] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) has long been a major global clinical problem as one of the most common malignant tumours with a high rate of recurrence and mortality. Although potentially curative therapies are available for the early and intermediate stages, the treatment of patients with advanced HCC remains to be resolved. Fortunately, the past few years have shown the emergence of successful systemic therapies to treat HCC. At the molecular level, HCC is a heterogeneous disease, and current research on the molecular characteristics of HCC has revealed numerous therapeutic targets. Targeted agents based on signalling molecules have been successfully supported in clinical trials, and molecular targeted therapy has already become a milestone for disease management in patients with HCC. Immunotherapy, a viable approach for the treatment of HCC, recognizes the antigens expressed by the tumour and treats the tumour using the immune system of the host, making it both selective and specific. In addition, the pipeline for HCC is evolving towards combination therapies with promising clinical outcomes. More drugs designed to focus on specific pathways and immune checkpoints are being developed in the clinic. It has been demonstrated that some drugs can improve the prognosis of patients with HCC in first- or second-line settings, and these drugs have been approved by the Food and Drug Administration or are nearing approval. This review describes targeting pathways and systemic treatment strategies in HCC and summarizes effective targeted and immune-based drugs for patients with HCC and the problems encountered.
Collapse
Affiliation(s)
- Xin Qing
- Department of General Surgery, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China
| | - Wenjing Xu
- Department of General Surgery, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China
| | - Jingjing Zong
- Department of General Surgery, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China
| | - Xuanlong Du
- Department of General Surgery, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China
| | - Hao Peng
- Department of General Surgery, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China
| | - Yewei Zhang
- Department of General Surgery, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China.
| |
Collapse
|
13
|
Histone Methyltransferase G9a-Promoted Progression of Hepatocellular Carcinoma Is Targeted by Liver-Specific Hsa-miR-122. Cancers (Basel) 2021; 13:cancers13102376. [PMID: 34069116 PMCID: PMC8157135 DOI: 10.3390/cancers13102376] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/11/2021] [Accepted: 05/11/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Targeting epigenetic alterations in hepatocellular carcinoma (HCC) provides therapeutic options in addition to traditional treatments. The aim of our study was to evaluate the potential of targeting histone methyltransferase G9a in the development of a therapeutic target. We confirmed the prognostic values of mRNA and protein levels of G9a expression in HCC respectively from public database and tissue microarray. We also confirmed the aggressive phenotypes supported by G9a in both HBV+ and HBV− HCC cells. The identification of a regulation axis between liver-specific tumor suppressor miR-122 and G9a further supported the important roles of G9a during the tumorigenesis and progression of HCC. Combination of lower miR-122 and higher G9a levels may provide prognostic potential for poor clinical outcomes and therapeutic potential for epigenetic targeting therapies. Abstract Hepatocellular carcinoma (HCC) accounts for the majority of primary liver cancers, which is the second most lethal tumor worldwide. Epigenetic deregulation is a common trait observed in HCC. Recently, increasing evidence suggested that the G9a histone methyltransferase might be a novel regulator of HCC development. However, several HCC cell lines were recently noted to have HeLa cell contamination or to have been derived from non-hepatocellular origin, suggesting that functional validation of G9a in proper HCC models is still required. Herein, we first confirmed that higher G9a messenger RNA and protein expression levels were correlated with poor overall survival (OS) and disease-free survival (DFS) rates of HCC patients from The Cancer Genome Atlas (TCGA) dataset and our recruited HCC cohort. In an in vitro functional evaluation of HCC cells, HCC36 (hepatitis B virus-positive (HBV+) and Mahlavu (HBV−)) cells showed that G9a participated in promoting cell proliferation, colony formation, and migration/invasion abilities. Moreover, orthotopic inoculation of G9a-depleted Mahlavu cells in NOD-SCID mice also resulted in a significantly decreased tumor burden compared to the control group. Furthermore, after surveying microRNA (miRNA; miR) prediction databases, we identified the liver-specific miR-122 as a G9a-targeting miRNA. In various HCC cell lines, we observed that miR-122 expression levels tended to be inversely correlated to G9a expression levels. In clinical HCC specimens, a significant inverse correlation of miR-122 and G9a mRNA expression levels was also observed. Functionally, the colony formation and invasive ability were attenuated in miR-122-overexpressing HCC cells. HCC patients with low miR-122 and high G9a expression levels had the worst OS and DFS rates compared to others. Together, our results confirmed the importance of altered G9a expression during HCC progression and discovered that a novel liver-specific miR-122-G9a regulatory axis exists.
Collapse
|
14
|
Garmpis N, Damaskos C, Garmpi A, Georgakopoulou VE, Sarantis P, Antoniou EA, Karamouzis MV, Nonni A, Schizas D, Diamantis E, Koustas E, Farmaki P, Syllaios A, Patsouras A, Kontzoglou K, Trakas N, Dimitroulis D. Histone Deacetylase Inhibitors in the Treatment of Hepatocellular Carcinoma: Current Evidence and Future Opportunities. J Pers Med 2021; 11:223. [PMID: 33809844 PMCID: PMC8004277 DOI: 10.3390/jpm11030223] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/16/2021] [Accepted: 03/19/2021] [Indexed: 02/05/2023] Open
Abstract
Hepatocellular carcinoma (HCC) remains a major health problem worldwide with a continuous increasing prevalence. Despite the introduction of targeted therapies like the multi-kinase inhibitor sorafenib, treatment outcomes are not encouraging. The prognosis of advanced HCC is still dismal, underlying the need for novel effective treatments. Apart from the various risk factors that predispose to the development of HCC, epigenetic factors also play a functional role in tumor genesis. Histone deacetylases (HDACs) are enzymes that remove acetyl groups from histone lysine residues of proteins, such as the core nucleosome histones, in this way not permitting DNA to loosen from the histone octamer and consequently preventing its transcription. Considering that HDAC activity is reported to be up-regulated in HCC, treatment strategies with HDAC inhibitors (HDACIs) showed some promising results. This review focuses on the use of HDACIs as novel anticancer agents and explains the mechanisms of their therapeutic effects in HCC.
Collapse
Affiliation(s)
- Nikolaos Garmpis
- Second Department of Propedeutic Surgery, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (E.A.A.); (K.K.); (D.D.)
- N.S. Christeas Laboratory of Experimental Surgery and Surgical Research, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Christos Damaskos
- N.S. Christeas Laboratory of Experimental Surgery and Surgical Research, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
- Renal Transplantation Unit, Laiko General Hospital, 11527 Athens, Greece
| | - Anna Garmpi
- First Department of Propedeutic Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Vasiliki E. Georgakopoulou
- Department of Pulmonology, Laiko General Hospital, 11527 Athens, Greece;
- First Department of Pulmonology, Sismanogleio Hospital, 15126 Athens, Greece
| | - Panagiotis Sarantis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.S.); (M.V.K.); (E.K.)
| | - Efstathios A. Antoniou
- Second Department of Propedeutic Surgery, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (E.A.A.); (K.K.); (D.D.)
- N.S. Christeas Laboratory of Experimental Surgery and Surgical Research, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Michalis V. Karamouzis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.S.); (M.V.K.); (E.K.)
| | - Afroditi Nonni
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Dimitrios Schizas
- First Department of Surgery, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.S.); (A.S.)
| | - Evangelos Diamantis
- Department of Endocrinology and Diabetes Center, G. Gennimatas General Hospital, 11527 Athens, Greece;
| | - Evangelos Koustas
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.S.); (M.V.K.); (E.K.)
| | - Paraskevi Farmaki
- First Department of Pediatrics, Agia Sofia Children’s Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Athanasios Syllaios
- First Department of Surgery, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.S.); (A.S.)
| | - Alexandros Patsouras
- Second Department of Internal Medicine, Tzanio General Hospital, 18536 Piraeus, Greece;
| | - Konstantinos Kontzoglou
- Second Department of Propedeutic Surgery, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (E.A.A.); (K.K.); (D.D.)
- N.S. Christeas Laboratory of Experimental Surgery and Surgical Research, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Nikolaos Trakas
- Department of Biochemistry, Sismanogleio Hospital, 15126 Athens, Greece;
| | - Dimitrios Dimitroulis
- Second Department of Propedeutic Surgery, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (E.A.A.); (K.K.); (D.D.)
| |
Collapse
|
15
|
Maternal hepatitis B or C carrier status and long-term risk for offspring neurological morbidity: a population-based cohort study. J Dev Orig Health Dis 2021; 13:115-119. [PMID: 33472720 DOI: 10.1017/s2040174420001397] [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: 11/07/2022]
Abstract
Hepatitis B and hepatitis C (HBV/HCV) are important global public health concerns. We aimed to evaluate the association between maternal HBV/HCV carrier status and long-term offspring neurological hospitalisations. A population-based cohort analysis compared the risk for long-term childhood neurological hospitalisations in offspring born to HBV/HCV carrier vs. non-carrier mothers in a large tertiary medical centre between 1991 and 2014. Childhood neurological diseases, such as cerebral palsy, movement disorders or developmental disorders, were pre-defined based on ICD-9 codes as recorded in hospital medical files. Offspring with congenital malformations and multiple gestations were excluded from the study. A Kaplan-Meier survival curve was constructed to compare cumulative neurological hospitalisations over time, and a Cox proportional hazards model was used to control for confounders. During the study period (1991-2014), 243,682 newborns met the inclusion criteria, and 777 (0.3%) newborns were born to HBV/HCV mothers. The median follow-up was 10.51 years (0-18 years). The offspring from HBV/HCV mothers had higher incidence of neurological hospitalisations (4.5 vs. 3.1%, hazard ratio (HR) = 1.91, 95% CI 1.37-2.67). Similarly, the cumulative incidence of neurological hospitalisations was higher in children born to HBV/HCV carrier mothers (Kaplan-Meier survival curve log-rank test p < 0.001). The increased risk remained significant in a Cox proportional hazards model, which adjusted for gestational age, mode of delivery and pregnancy complications (adjusted HR = 1.40, 1.01-1.95, p = 0.049). We conclude that maternal HBV or HCV carrier status is an independent risk factor for the long-term neurological hospitalisation of offspring regardless of gestational age and other adverse perinatal outcomes.
Collapse
|
16
|
Xu G, Zhou X, Xing J, Xiao Y, Jin B, Sun L, Yang H, Du S, Xu H, Mao Y. Identification of RASSF1A promoter hypermethylation as a biomarker for hepatocellular carcinoma. Cancer Cell Int 2020; 20:547. [PMID: 33292241 PMCID: PMC7653745 DOI: 10.1186/s12935-020-01638-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 11/02/2020] [Indexed: 02/07/2023] Open
Abstract
Background RAS association domain family protein 1A (RASSF1A) promoter hypermethylation is suggested to be linked to hepatocellular carcinoma (HCC), but the results remained controversial. Methods We evaluated how RASSF1A promoter hypermethylation affects HCC risk and its clinicopathological characteristics through meta-analysis. Data on DNA methylation in HCC and relevant clinical data were also collected based on The Cancer Genome Atlas (TCGA) database to investigate the prognostic role of RASSF1A promoter hypermethylation in HCC. Results Forty-four articles involving 4777 individuals were enrolled in the pooled analyses. The RASSF1A promoter methylation rate was notably higher in the HCC cases than the non-tumor cases and healthy individuals, and was significantly related to hepatitis B virus (HBV) infection-positivity and large tumor size. Kaplan–Meier survival analysis revealed that HCC cases with RASSF1A promoter hypermethylation had worse outcomes. Receiver operating characteristic curves confirmed that RASSF1A promoter methylation may be a marker of HCC-related prognoses. Conclusions RASSF1A promoter hypermethylation is a promising biomarker for the diagnosis of HCC from tissue and peripheral blood, and is an emerging therapeutic target against HCC.
Collapse
Affiliation(s)
- Gang Xu
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC and Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xiaoxiang Zhou
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC and Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Jiali Xing
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC and Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yao Xiao
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC and Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Bao Jin
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC and Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Lejia Sun
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC and Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Huayu Yang
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC and Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Shunda Du
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC and Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Haifeng Xu
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC and Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Yilei Mao
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC and Chinese Academy of Medical Sciences, Beijing, 100730, China.
| |
Collapse
|
17
|
Lian B, Li H, Liu Y, Chai D, Gao Y, Zhang Y, Zhou J, Li J. Expression and promoter methylation status of OPCML and its functions in the inhibition of cell proliferation, migration, and invasion in breast cancer. Breast Cancer 2020; 28:448-458. [PMID: 33108608 DOI: 10.1007/s12282-020-01179-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 10/16/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Opioid binding protein/cell adhesion molecule-like (OPCML) has been demonstrated to be a tumor suppressor gene, as it has been shown in previous studies to play a tumor-suppressive role in a variety of cancers. However, the role of OPCML in breast tumorigenesis remains unclear. METHODS In this study, we analyzed OPCML expression in breast cancers and adjacent non-tumor tissue samples and examined its molecular function in the breast cancer-derived cell lines MDA-MB-231 and MCF7. RESULTS We found that OPCML was downregulated in most breast cancer samples but that this protein was expressed in most adjacent non-tumor samples. The loss or downregulation of OPCML is associated with hypermethylation of its promoter. Methylation of the OPCML promoter was detected in all breast cancer cell lines and primary tumors but was not detected in surgical margin tissues and normal breast tissues. Furthermore, functional assays showed that ectopic OPCML expression could inhibit breast tumor cell proliferation in vivo and in vitro and further suppresses tumor cell migration and invasion. CONCLUSION Our results show that OPCML exerts its tumor-suppressive functions in human breast cancer cells. Moreover, the promoter-specific hypermethylation of OPCML plays an important role in human breast cancer development.
Collapse
Affiliation(s)
- Bin Lian
- Department of Oncology Surgery, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Hong Li
- Department of Oncology Surgery, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yaobang Liu
- Department of Oncology Surgery, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Dahai Chai
- Department of Oncology Surgery, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yali Gao
- Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yangyang Zhang
- Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Jia Zhou
- Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Jinping Li
- Department of Oncology Surgery, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China.
| |
Collapse
|
18
|
Liu Q, Lu F, Chen Z. Identification of MT1E as a novel tumor suppressor in hepatocellular carcinoma. Pathol Res Pract 2020; 216:153213. [PMID: 32956919 DOI: 10.1016/j.prp.2020.153213] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 09/06/2020] [Accepted: 09/08/2020] [Indexed: 11/26/2022]
Abstract
BACKGROUND Metallothioneins (MTs) involves in the tumorigenesis and prognosis of various cancers. The biological function and methylation status of MT1E in hepatocellular carcinoma (HCC) remain to be elucidated. METHODS We analyzed differentially expressed genes (DEGs) in tumor tissue samples and normal samples from The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) database, and identified the expression levels of MT1E in the HCC. Then, the expression levels and methylation status of MT1E in HCC tissues and cells were validated by qRT-PCR and methylation-specific PCR (MSP). Also, MTT, colony formation, transwell assays, and flow cytometry, as well as xenograft model, were used to assess the biological roles of MT1E in HCC. RESULTS Downregulated expression of MT1E was found in HCC tissues, and was notably correlated with an aberrant methylation level of the gene promoter. Moreover, our study verified that MT1E suppressed cell growth in vitro and vivo. Further study demonstrated that MT1E could induce apoptosis and suppress the metastasis of HCC cells. CONCLUSIONS Our results suggested that epigenetic silencing of MT1E due to promoter hypermethylation could play a vital role in HCC.
Collapse
Affiliation(s)
- Qichen Liu
- Departmentof General Surgery, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215000, China; Departmentof General Surgery, Binhai Country People's Hospital, Binhai, Jiangsu, 224500, China
| | - Feng Lu
- Departmentof General Surgery, Binhai Country People's Hospital, Binhai, Jiangsu, 224500, China
| | - Zhong Chen
- Departmentof General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226000, China.
| |
Collapse
|
19
|
Matsushita J, Suzuki T, Okamura K, Ichihara G, Nohara K. Identification by TCGA database search of five genes that are aberrantly expressed and involved in hepatocellular carcinoma potentially via DNA methylation changes. Environ Health Prev Med 2020; 25:31. [PMID: 32703154 PMCID: PMC7376645 DOI: 10.1186/s12199-020-00871-8] [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: 04/02/2020] [Accepted: 07/05/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Various treatments for hepatocellular carcinoma (HCC) are utilized in clinical practice; however, the prognosis is still poor on account of high recurrence rates. DNA methylation levels of CpG islands around promoters (promoter CpGis) inversely regulate gene expression and closely involved in carcinogenesis. As a new strategy, several chemicals globally inhibiting DNA methylation have been developed aiming at reducing DNA methylation levels and maintaining the expression of tumor suppressor genes. On the other hand, since these drugs nonspecifically modify DNA methylation, they can cause serious adverse effects. In order to ameliorate the methods by targeting specific CpGs, information of cancer-related genes that are regulated by DNA methylation is required. METHODS We searched candidate genes whose expressions were regulated by DNA methylation of promoter CpGi and which are involved in HCC cases. To do so, we first identified genes whose expression were changed in HCC by comparing gene expressions of 371 HCC tissues and 41 non-tumor tissues using the Cancer Genome Atlas (TCGA) database. The genes were further selected for poor prognosis by log-rank test of Kaplan-Meier plot and for cancer relevance by Pubmed search. Expression profiles of upregulated genes in HCC tissues were assessed by Gene Ontology (GO) analysis. Finally, using DNA methylation data of TCGA database, we selected genes whose promoter DNA methylation levels were inversely correlated with gene expression. RESULTS We found 115 genes which were significantly up- or downregulated in HCC tissues and were associated with poor prognosis and cancer relevance. The upregulated genes were significantly enriched in cell division, cell cycle, and cell proliferation. Among the upregulated genes in HCC, we identified hypomethylation of CpGis around promoters of FANCB, KIF15, KIF4A, ERCC6L, and UBE2C. In addition, TCGA data showed that the tumor suppressor gene P16 is unexpectedly overexpressed in many types of cancers. CONCLUSIONS We identified five candidate genes whose expressions were regulated by DNA methylation of promoter CpGi and associate with cancer cases and poor prognosis in HCC. Modification of site-specific DNA methylation of these genes enables a different approach for HCC treatment with higher selectivity and lower adverse effects.
Collapse
Affiliation(s)
- Junya Matsushita
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies, Tsukuba, Japan.,Graduate School of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Takehiro Suzuki
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies, Tsukuba, Japan
| | - Kazuyuki Okamura
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies, Tsukuba, Japan
| | - Gaku Ichihara
- Graduate School of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Keiko Nohara
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies, Tsukuba, Japan.
| |
Collapse
|
20
|
Zhou C, Huang Y, Chen Y, Xie Y, Wen H, Tan W, Wang C. miR-602 Mediates the RASSF1A/JNK Pathway, Thereby Promoting Postoperative Recurrence in Nude Mice with Liver Cancer. Onco Targets Ther 2020; 13:6767-6776. [PMID: 32764964 PMCID: PMC7368130 DOI: 10.2147/ott.s243651] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 06/19/2020] [Indexed: 12/25/2022] Open
Abstract
Purpose At present, there are few studies on the mechanisms underlying postoperative recurrence of liver cancer, and the mechanism of action of miR-602 in postoperative recurrence of liver tumors is not clear. Our goals were to investigate the effects of miR-602 on the expression of the Ras-associated domain family 1A (RASSF1A) gene and the regulation of primary and recurrent hepatic tumors to clarify the molecular mechanisms of miR-602 in postoperative hepatocellular carcinoma. Methods We constructed a mouse liver orthotopic tumor model and a mouse liver recurrent tumor model. We measured the expression levels of the RASSF1A gene and then analyzed the effects of miR-602 on the regulation of RASSF1A. We transiently transfected the miR-602 gene into cells that stably overexpressed RASSF1A and examined relevant indicators to elucidate the mechanisms by which miR-602 regulates the RASSF1A/c-Jun N-terminal kinase (JNK) pathway in recurrence and dormancy in liver cancer. Results RASSF1A expression was inversely related to that of JNK, activating transcription factor 2 (ATF-2), and c-Jun in SMMC7721 cells stably transfected with the RASSF1A gene and in recurrent mouse tumor tissues. After transient transfection of cells with miR-602 mimic or miR-602 inhibitor, the expression of miR-602 was inversely related to that of RASSF1A. Conclusion MiR-602 might inhibit the JNK signaling pathway by inhibiting the expression of RASSF1A, thereby promoting recurrence of liver cancer after surgery. The low expression levels of miR-602 in liver cancer tissues were closely related to postoperative recurrence; they could be used as a marker to judge the prognosis of patients with liver cancer.
Collapse
Affiliation(s)
- Cheng Zhou
- School of Medicine, South China University of Technology, Guangzhou, People's Republic of China.,Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Geriatric Institute, Guangzhou, People's Republic of China
| | - Yajing Huang
- Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China
| | - Yongxu Chen
- School of Medicine, South China University of Technology, Guangzhou, People's Republic of China.,Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Geriatric Institute, Guangzhou, People's Republic of China
| | - Yingjie Xie
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, People's Republic of China
| | - Huihong Wen
- Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China
| | - Wei Tan
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Geriatric Institute, Guangzhou, People's Republic of China
| | - Changjun Wang
- School of Medicine, South China University of Technology, Guangzhou, People's Republic of China.,Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Geriatric Institute, Guangzhou, People's Republic of China
| |
Collapse
|
21
|
Afaloniati H, Angelopoulou K, Giakoustidis A, Hardas A, Pseftogas A, Makedou K, Gargavanis A, Goulopoulos T, Iliadis S, Papadopoulos V, Papalois A, Mosialos G, Poutahidis T, Giakoustidis D. HDAC1/2 Inhibitor Romidepsin Suppresses DEN-Induced Hepatocellular Carcinogenesis in Mice. Onco Targets Ther 2020; 13:5575-5588. [PMID: 32606772 PMCID: PMC7304783 DOI: 10.2147/ott.s250233] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 05/27/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a frequently diagnosed cancer and a leading cause of cancer-related death worldwide. Its rapid progression, combined with the limited treatment options at late stages, imposes the need for early detection and aggressive intervention. Based on the knowledge that hepatocarcinogenesis is significantly influenced by histone acetylation, we directed our search for novel HCC therapeutics among histone deacetylation inhibitors (HDACi). The aim of the present study was to investigate the effect of HDAC1/2 inhibitor Romidepsin in the well-established mouse model of diethylnitrosamine (DEN)-induced HCC. MATERIALS AND METHODS C56BL/6 mice were treated with Romidepsin at the critical point of 10 months after DEN challenge and their livers were examined 2 months later using histopathology and morphometry. Protein levels were assessed in serum using ELISA and in liver tissues using Western blot and immunohistochemistry (in-situ detection). Gene expression was quantified using real-time PCR. RESULTS Romidepsin suppressed cancer progression. This effect was associated with decreased proliferation and increased apoptosis of cancer cells. The cell cycle regulator CK2a, the anti-inflammatory molecule PPAR-γ, and the tumor suppressors PTEN and CYLD were upregulated in treated HCC. By contrast, the expression of PI3K, NF-κB p65 and c-Jun was reduced. In line with this result, the levels of two major apoptosis regulators, ie, BAD and the multifunctional protein c-Met, were lower in the blood serum of treated mice compared to the untreated mice with HCC. CONCLUSION These findings suggest that Romidepsin, a drug currently used in the treatment of lymphoma, could also be considered in the management of early-stage HCC.
Collapse
Affiliation(s)
- Hara Afaloniati
- Laboratory of Biochemistry and Toxicology, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Katerina Angelopoulou
- Laboratory of Biochemistry and Toxicology, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Alexander Giakoustidis
- First Department of Surgery, Medical School, Aristotle University of Thessaloniki, General Hospital Papageorgiou, Thessaloniki, Greece
| | - Alexandros Hardas
- Laboratory of Pathology, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Athanasios Pseftogas
- School of Biology, Faculty of Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Kali Makedou
- Department of Biological Chemistry, Medical School, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Athanasios Gargavanis
- First Department of Surgery, Medical School, Aristotle University of Thessaloniki, General Hospital Papageorgiou, Thessaloniki, Greece
| | - Thomas Goulopoulos
- First Department of Surgery, Medical School, Aristotle University of Thessaloniki, General Hospital Papageorgiou, Thessaloniki, Greece
| | - Stavros Iliadis
- Department of Biological Chemistry, Medical School, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Vasileios Papadopoulos
- First Department of Surgery, Medical School, Aristotle University of Thessaloniki, General Hospital Papageorgiou, Thessaloniki, Greece
| | - Apostolos Papalois
- Experimental, Educational and Research Center, ELPEN, Pikermi, Attica, Greece
| | - George Mosialos
- School of Biology, Faculty of Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Theofilos Poutahidis
- Laboratory of Pathology, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Dimitrios Giakoustidis
- First Department of Surgery, Medical School, Aristotle University of Thessaloniki, General Hospital Papageorgiou, Thessaloniki, Greece
| |
Collapse
|
22
|
Liu A, Wu Q, Peng D, Ares I, Anadón A, Lopez-Torres B, Martínez-Larrañaga MR, Wang X, Martínez MA. A novel strategy for the diagnosis, prognosis, treatment, and chemoresistance of hepatocellular carcinoma: DNA methylation. Med Res Rev 2020; 40:1973-2018. [PMID: 32525219 DOI: 10.1002/med.21696] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 04/29/2020] [Accepted: 05/17/2020] [Indexed: 12/11/2022]
Abstract
The cancer mortality rate of hepatocellular carcinoma (HCC) is the second highest in the world and the therapeutic options are limited. The incidence of this deadly cancer is rising at an alarming rate because of the high degree of resistance to chemo- and radiotherapy, lack of proper, and adequate vaccination to hepatitis B, and lack of consciousness and knowledge about the disease itself and the lifestyle of the people. DNA methylation and DNA methylation-induced epigenetic alterations, due to their potential reversibility, open the access to develop novel biomarkers and therapeutics for HCC. The contribution to these epigenetic changes in HCC development still has not been thoroughly summarized. Thus, it is necessary to better understand the new molecular targets of HCC epigenetics in HCC diagnosis, prevention, and treatment. This review elaborates on recent key findings regarding molecular biomarkers for HCC early diagnosis, prognosis, and treatment. Currently emerging epigenetic drugs for the treatment of HCC are summarized. In addition, combining epigenetic drugs with nonepigenetic drugs for HCC treatment is also mentioned. The molecular mechanisms of DNA methylation-mediated HCC resistance are reviewed, providing some insights into the difficulty of treating liver cancer and anticancer drug development.
Collapse
Affiliation(s)
- Aimei Liu
- National Reference Laboratory of Veterinary Drug Residues and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University (HZAU), Wuhan, China
| | - Qinghua Wu
- College of Life Science, Yangtze University, Jingzhou, China.,Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Dapeng Peng
- National Reference Laboratory of Veterinary Drug Residues and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University (HZAU), Wuhan, China
| | - Irma Ares
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Research Institute Hospital 12 de Octubre (i+12), Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Arturo Anadón
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Research Institute Hospital 12 de Octubre (i+12), Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Bernardo Lopez-Torres
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Research Institute Hospital 12 de Octubre (i+12), Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - María-Rosa Martínez-Larrañaga
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Research Institute Hospital 12 de Octubre (i+12), Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Xu Wang
- National Reference Laboratory of Veterinary Drug Residues and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University (HZAU), Wuhan, China.,Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Research Institute Hospital 12 de Octubre (i+12), Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - María-Aránzazu Martínez
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Research Institute Hospital 12 de Octubre (i+12), Universidad Complutense de Madrid (UCM), Madrid, Spain
| |
Collapse
|
23
|
Wang Q, Xue J, Ren Q, Li X, Qiu X. Long-chain non-coding RNA GACAT1 promotes development and progression of breast cancer by targeting microRNA-875-3p. Oncol Lett 2020; 19:2547-2553. [PMID: 32194758 PMCID: PMC7039160 DOI: 10.3892/ol.2020.11260] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 12/20/2019] [Indexed: 12/19/2022] Open
Abstract
Role of long-chain non-coding ribonucleic acid (lncRNA) GACAT1 in the development of breast cancer and its possible mechanism were investigated. The levels of GACAT1, microRNA-875-3p and Stonin2 (STON2) in breast cancer tissues and adjacent normal tissues were detected by quantitative real-time polymerase chain reaction (qRT-PCR). The level of GACAT1 in breast cancer cell lines was further explored. The effects of GACAT1 and microRNA-875-3p on cell proliferation and cycle were detected by cell counting kit-8 (CCK-8) and flow cytometry. The binding relationship between microRNA-875-3p and STON2, microRNA-875-3p and GACAT1 was examined by a dual luciferase reporting assay. It was confirmed by rescue experiments whether GACAT1 canregulate the level of STON2 by binding to microRNA-875-3p. GACAT1 level was clearly enhanced in breast cancer tissues compared to that of the adjacent tissues. Similar result was observed in breast cancer cell lines. Upregulation of GACAT1 promoted the proliferation and cycle of breast cancer cells including MCF-7 and BCap-37. The dual luciferase reporting assay results indicated that GACAT1 had a binding relationship with microRNA-875-3p. Further experiments confirmed that microRNA-875-3p was conspicuously downregulated in breast cancer tissues, and upregulation of microRNA-875-3p could inhibit the proliferation ability of MCF-7 and BCap-37 cells, and partially reversed the promoting effect of GACAT1 on cell cycle. Through bioinformatics prediction and dual luciferase reporter gene experiments, we found that STON2 might be a target gene of microRNA-875-3p. Overexpression of STON2 could partially abolish the effect of microRNA-875-3p on cell proliferation and cycle of MCF-7 and BCap-37 cells. GACAT1 can participate in the progression of breast cancer by promoting the proliferation and cycle of breast cancer cells. The mechanism may be through the regulation of the level of STON2 by adsorbing microRNA-875-3p.
Collapse
Affiliation(s)
- Qinghua Wang
- Department of Breast Surgery, Linyi Cancer Hospital, Linyi, Shandong 276000, P.R. China
| | - Jie Xue
- Department of Breast Surgery, Linyi Cancer Hospital, Linyi, Shandong 276000, P.R. China
| | - Qingfang Ren
- Department of Medical Oncology, Linyi Cancer Hospital, Linyi, Shandong 276000, P.R. China
| | - Xiaona Li
- Department of Breast Surgery, Linyi Cancer Hospital, Linyi, Shandong 276000, P.R. China
| | - Xiaoli Qiu
- Department of Breast Surgery, Linyi Cancer Hospital, Linyi, Shandong 276000, P.R. China
| |
Collapse
|
24
|
García-Gutiérrez L, McKenna S, Kolch W, Matallanas D. RASSF1A Tumour Suppressor: Target the Network for Effective Cancer Therapy. Cancers (Basel) 2020; 12:cancers12010229. [PMID: 31963420 PMCID: PMC7017281 DOI: 10.3390/cancers12010229] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 01/12/2020] [Accepted: 01/14/2020] [Indexed: 02/06/2023] Open
Abstract
The RASSF1A tumour suppressor is a scaffold protein that is involved in cell signalling. Increasing evidence shows that this protein sits at the crossroad of a complex signalling network, which includes key regulators of cellular homeostasis, such as Ras, MST2/Hippo, p53, and death receptor pathways. The loss of expression of RASSF1A is one of the most common events in solid tumours and is usually caused by gene silencing through DNA methylation. Thus, re-expression of RASSF1A or therapeutic targeting of effector modules of its complex signalling network, is a promising avenue for treating several tumour types. Here, we review the main modules of the RASSF1A signalling network and the evidence for the effects of network deregulation in different cancer types. In particular, we summarise the epigenetic mechanism that mediates RASSF1A promoter methylation and the Hippo and RAF1 signalling modules. Finally, we discuss different strategies that are described for re-establishing RASSF1A function and how a multitargeting pathway approach selecting druggable nodes in this network could lead to new cancer treatments.
Collapse
Affiliation(s)
- Lucía García-Gutiérrez
- Systems Biology Ireland, University College Dublin, Belfield, Dublin 4, Ireland; (L.G.-G.); (S.M.); (W.K.)
| | - Stephanie McKenna
- Systems Biology Ireland, University College Dublin, Belfield, Dublin 4, Ireland; (L.G.-G.); (S.M.); (W.K.)
| | - Walter Kolch
- Systems Biology Ireland, University College Dublin, Belfield, Dublin 4, Ireland; (L.G.-G.); (S.M.); (W.K.)
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
- Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - David Matallanas
- Systems Biology Ireland, University College Dublin, Belfield, Dublin 4, Ireland; (L.G.-G.); (S.M.); (W.K.)
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
- Correspondence:
| |
Collapse
|
25
|
Zhang H, Li G, Zhang Y, Shi J, Yan B, Tang H, Chen S, Zhang J, Wen P, Wang Z, Pang C, Li J, Guo W, Zhang S. Targeting BET Proteins With a PROTAC Molecule Elicits Potent Anticancer Activity in HCC Cells. Front Oncol 2020; 9:1471. [PMID: 31993368 PMCID: PMC6971110 DOI: 10.3389/fonc.2019.01471] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 12/09/2019] [Indexed: 12/23/2022] Open
Abstract
Background and Aim: Bromodomain and extraterminal domain (BET) family proteins are epigenetic regulators involved in human malignances. Targeting BET proteins for degradation using proteolysis-targeting chimera (PROTAC) recently has drawn increasing attention in the field of cancer therapeutics. BET proteins have been found to be overexpressed in HCC cells and tumor tissues. However, the biological activity of BET-PROTACs in hepatocellular carcinoma (HCC) remains unclear. In this study, we investigated anti-HCC activity of BETd-260, a BET-PROTAC molecule using in vitro and in vivo models. Methods: BETd-260-mediated anti-HCC activity was investigated by cell viability, apoptosis assays. Efficacy was examined with a cell lines-derived HCC xenograft model in mice. Anticancer mechanism was investigated by RT-PCR, western blotting and immunohistochemical staining. Results: BETd-260 potently suppressed cell viability and robustly induced apoptosis in HCC cells. BETd-260 reciprocally modulated the expression of several apoptotic genes in HCC cells, i.e., suppressing the expression of anti-apoptotic Mcl-1, Bcl-2, c-Myc, and X-linked inhibitor of apoptosis (XIAP), whereas increasing the expression of pro-apoptotic Bad. BETd-260 treatment led to disruption of mitochondrial membrane integrity, and triggered apoptosis via intrinsic signaling in HCC cells. BETd-260 triggered apoptosis in HCC xenograft tissue and profoundly inhibited the growth of HCC xenograft tumors in mice. Conclusion: Our data suggest that pharmacological targeting of BET for degradation may be a novel therapeutic strategy for the treatment of HCC.
Collapse
Affiliation(s)
- Huapeng Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Zhengzhou Key Laboratory of Hepatobiliary & Pancreatic Diseases and Organ Transplantation, Zhengzhou, China.,Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, China
| | - Gongquan Li
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Zhengzhou Key Laboratory of Hepatobiliary & Pancreatic Diseases and Organ Transplantation, Zhengzhou, China.,Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, China
| | - Yi Zhang
- Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jihua Shi
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Zhengzhou Key Laboratory of Hepatobiliary & Pancreatic Diseases and Organ Transplantation, Zhengzhou, China.,Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, China
| | - Bing Yan
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Zhengzhou Key Laboratory of Hepatobiliary & Pancreatic Diseases and Organ Transplantation, Zhengzhou, China.,Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, China
| | - Hongwei Tang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Zhengzhou Key Laboratory of Hepatobiliary & Pancreatic Diseases and Organ Transplantation, Zhengzhou, China.,Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, China
| | - Sanyang Chen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Zhengzhou Key Laboratory of Hepatobiliary & Pancreatic Diseases and Organ Transplantation, Zhengzhou, China.,Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, China
| | - Jiakai Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Zhengzhou Key Laboratory of Hepatobiliary & Pancreatic Diseases and Organ Transplantation, Zhengzhou, China.,Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, China
| | - Peihao Wen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Zhengzhou Key Laboratory of Hepatobiliary & Pancreatic Diseases and Organ Transplantation, Zhengzhou, China.,Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, China
| | - Zhihui Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Zhengzhou Key Laboratory of Hepatobiliary & Pancreatic Diseases and Organ Transplantation, Zhengzhou, China.,Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, China
| | - Chun Pang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Zhengzhou Key Laboratory of Hepatobiliary & Pancreatic Diseases and Organ Transplantation, Zhengzhou, China.,Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, China
| | - Jie Li
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Zhengzhou Key Laboratory of Hepatobiliary & Pancreatic Diseases and Organ Transplantation, Zhengzhou, China.,Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, China
| | - Wenzhi Guo
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Zhengzhou Key Laboratory of Hepatobiliary & Pancreatic Diseases and Organ Transplantation, Zhengzhou, China.,Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, China
| | - Shuijun Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Zhengzhou Key Laboratory of Hepatobiliary & Pancreatic Diseases and Organ Transplantation, Zhengzhou, China.,Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, China
| |
Collapse
|
26
|
Yoles I, Sheiner E, Abu-Freha N, Wainstock T. Maternal hepatitis B or C status and the long-term risk of gastrointestinal morbidity for offspring: A population-based cohort study. Liver Int 2019; 39:2046-2051. [PMID: 31319010 DOI: 10.1111/liv.14193] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 07/09/2019] [Accepted: 07/11/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND More than 360 million people have chronic hepatitis B or C (HBV/HCV) infection worldwide, many of which are women at childbearing age. While the risk of perinatal HBV/HCV has been well established, the long-term implications on offspring health, have been less studied. We aimed to evaluate the association between maternal HBV/HCV carrier status and long-term gastrointestinal (GI) morbidities in offspring. AIMS & METHODS A population-based cohort analysis compared the risk for long-term childhood GI morbidities in children born to HBV/HCV carrier mothers vs the risk in those who were born to noncarriers. Childhood GI morbidities were predefined based on ICD-9 codes, as recorded in hospital medical files. Children with congenital malformations and multiple gestations were excluded from the analysis. A Kaplan-Meier survival curve was constructed to compare the cumulative GI morbidities over time, and a Cox proportional hazards model was used to control for confounders. RESULTS During the study period (1991-2014), 242 342 newborns met the inclusion criteria: 771 (0.3%) were born to HBV/HCV mothers and 241 571 (99.7%) were not. The median follow-up was 10.51 years (0-18 years). Offspring to HBV/HCV mothers had a higher incidence of GI diseases (9.3% vs 5.4%, OR = 1.82; 95% CI 1.43-2.32; Kaplan-Meier log-rank = 0.001). The increased risk remained significant in the Cox proportional hazards models, which adjusted for gestational age, mode of delivery and pregnancy complications (adjusted HR = 2.26, 95% CI: 1.79-2.85; P < .001). CONCLUSION Maternal HBV or HCV carrier status is an independent risk factor for long-term the GI morbidity of offspring.
Collapse
Affiliation(s)
- Israel Yoles
- The Central District, Clalit Health Services, Rishon Le Tzion, Israel.,Department of Obstetrics and Gynecology, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Eyal Sheiner
- Department of Obstetrics and Gynecology, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Naim Abu-Freha
- Gastroenterology and Hepatology Department, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Tamar Wainstock
- Department of Public Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
27
|
Yu L, Xie R, Tian T, Zheng L, Tang L, Cai S, Ma Z, Yang T, Han B, Yang Q. Suberoylanilide hydroxamic acid upregulates histone acetylation and activates endoplasmic reticulum stress to induce apoptosis in HepG2 liver cancer cells. Oncol Lett 2019; 18:3537-3544. [PMID: 31516571 PMCID: PMC6732958 DOI: 10.3892/ol.2019.10705] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 06/26/2019] [Indexed: 01/07/2023] Open
Abstract
Suberoylanilide hydroxamic acid (SAHA) is a histone deacetylase inhibitor that has demonstrated clinical activity against various solid tumors. The aim of the present study was to explore the effects of SAHA on the apoptosis of HepG2 liver cancer cells, as well as the potential mechanisms involved in histone acetylation and endoplasmic reticulum (ER) stress. HepG2 cells were treated with various doses of SAHA (0, 1, 6 and 12 µM), and apoptosis was measured by flow cytometry. The levels of ER stress-associated molecules, including 78 kDa glucose-regulated protein (GRP78), PRKR-like endoplasmic reticulum kinase (PERK), phosphorylated (p)-PERK, activating transcription factor 4 (ATF4) and C/EBP-homologous protein (CHOP), were quantitated by western blot analysis and reverse transcription-quantitative PCR assay. The expression levels of acetylated histone H4 (acH4, acH4 lysine (K)5 and acH4K12) were detected by western blot analysis. The effects of SAHA on the acetylation of H4 in the promoter regions of GRP78, ATF4 and CHOP were evaluated by chromatin immunoprecipitation assays. Following treatment with higher doses of SAHA (6 and 12 µM) for 48 h, the proliferation of HepG2 cells was significantly suppressed. SAHA induced dose-dependent apoptosis and increased both protein and mRNA expression levels of GRP78, ATF4 and CHOP in HepG2 cells. The protein expression of PERK was markedly decreased by treatment with SAHA, whereas the p-PERK expression level was notably increased, which resulted in increased p-PERK/PERK ratio. Furthermore, the acetylation levels of H4 in the promoter regions of GRP78, ATF4 and CHOP were significantly increased in HepG2 cells exposed to 6 µM SAHA for 36 h. Thus, SAHA induces apoptosis in HepG2 cells by activating the ER stress-mediated apoptotic signaling pathway, at least partially by enhancing the acetylation of histone H4 on the promoter regions of ER-stress associated genes, including GRP78, ATF4 and CHOP.
Collapse
Affiliation(s)
- Lei Yu
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Department of Pathology, Hospital of Maternal and Childhood Health, Guiyang, Guizhou 550000, P.R. China
| | - Rujia Xie
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Tian Tian
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Lu Zheng
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Lei Tang
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Shuang Cai
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Zihua Ma
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Ting Yang
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Bing Han
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Qin Yang
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| |
Collapse
|
28
|
Wang L, Wang FS. Clinical immunology and immunotherapy for hepatocellular carcinoma: current progress and challenges. Hepatol Int 2019; 13:521-533. [PMID: 31352593 DOI: 10.1007/s12072-019-09967-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 06/27/2019] [Indexed: 12/16/2022]
Abstract
At the time of hepatocellular carcinoma (HCC) diagnosis, patients are most often at an advanced stage; however, the current treatment regimens remain unsatisfactory. Thus, novel and more powerful therapeutic approaches for advanced HCC are urgently required. Exacerbation of immunotolerant signals and/or escaping immunosurveillance leads to the development of HCC, which appears to be a rational reason to use immunotherapy to restore anticancer immunity. Several novel immunotherapeutic methods, including the use of immune checkpoint inhibitors, new types of immune cell adoption [e.g., chimeric antigen receptor T cell (CAR-T), TCR gene-modified T cells and stem cells], and microRNAs have been used in clinical trials for the treatment of HCC. However, some crucial issues remain to be addressed for such novel immunotherapy techniques. Finally, immunotherapy is now standing on the threshold of great advances in the fight against HCC.
Collapse
Affiliation(s)
- Lifeng Wang
- Treatment and Research Center for Infectious Diseases, The Fifth Medical Center of PLA General Hospital, 100 Western 4th Ring Road, Beijing, 100039, China
| | - Fu-Sheng Wang
- Treatment and Research Center for Infectious Diseases, The Fifth Medical Center of PLA General Hospital, 100 Western 4th Ring Road, Beijing, 100039, China.
| |
Collapse
|
29
|
Chen Z, Zuo X, Pu L, Zhang Y, Han G, Zhang L, Wu J, Wang X. circLARP4 induces cellular senescence through regulating miR-761/RUNX3/p53/p21 signaling in hepatocellular carcinoma. Cancer Sci 2019; 110:568-581. [PMID: 30520539 PMCID: PMC6361555 DOI: 10.1111/cas.13901] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/22/2018] [Accepted: 11/25/2018] [Indexed: 12/31/2022] Open
Abstract
Circular RNAs (circRNAs), a novel class of non-coding RNAs, have emerged as indispensable modulators in human malignancies. Aberrant cellular senescence is a phenotype observed in various cancers. The association of circRNAs with cellular senescence in tumors is yet to determined. Here, we investigated the role of circLARP4 in cellular senescence and cell proliferation in hepatocellular carcinoma (HCC). Downregulated circLARP4 level was observed in HCC tissues and cell lines. Low expression level of circLARP4 independently predicted poor survival outcome. Gain-of-function and loss-of-function assays demonstrated that circLARP4 suppressed HCC cell proliferation, mediated cell cycle arrest and induced senescence in vitro. Levels of p53 and p21, 2 key regulatory molecules in cellular senescence, were increased in circLARP4-overexpressed HCC cells and decreased in circLARP4-silenced HCC cells. In vivo experiments further confirmed the tumor-suppressing activity of circLARP4. Further mechanistic studies showed that circLARP4 dampened HCC progression by sponging miR-761, thereby promoting the expression level of RUNX3 and activating the downstream p53/p21 signaling. Our study revealed the role of circLARP4/miR-761/RUNX3/p53/p21 signaling in HCC progression, providing a potential survival predictor and therapeutic candidate for HCC.
Collapse
Affiliation(s)
- Zhiqiang Chen
- Hepatobiliary CenterThe First Affiliated Hospital of Nanjing Medical UniversityKey Laboratory of Liver TransplantationChinese Academy of Medical SciencesNHC Key Laboratory of Living Donor Liver TransplantationNanjingChina
| | - Xueliang Zuo
- Hepatobiliary CenterThe First Affiliated Hospital of Nanjing Medical UniversityKey Laboratory of Liver TransplantationChinese Academy of Medical SciencesNHC Key Laboratory of Living Donor Liver TransplantationNanjingChina
- Department of Gastrointestinal SurgeryThe First Affiliated HospitalYijishan Hospital of Wannan Medical CollegeWuhuChina
| | - Liyong Pu
- Hepatobiliary CenterThe First Affiliated Hospital of Nanjing Medical UniversityKey Laboratory of Liver TransplantationChinese Academy of Medical SciencesNHC Key Laboratory of Living Donor Liver TransplantationNanjingChina
| | - Yao Zhang
- Hepatobiliary CenterThe First Affiliated Hospital of Nanjing Medical UniversityKey Laboratory of Liver TransplantationChinese Academy of Medical SciencesNHC Key Laboratory of Living Donor Liver TransplantationNanjingChina
| | - Guoyong Han
- Hepatobiliary CenterThe First Affiliated Hospital of Nanjing Medical UniversityKey Laboratory of Liver TransplantationChinese Academy of Medical SciencesNHC Key Laboratory of Living Donor Liver TransplantationNanjingChina
| | - Long Zhang
- Hepatobiliary CenterThe First Affiliated Hospital of Nanjing Medical UniversityKey Laboratory of Liver TransplantationChinese Academy of Medical SciencesNHC Key Laboratory of Living Donor Liver TransplantationNanjingChina
| | - Jindao Wu
- Hepatobiliary CenterThe First Affiliated Hospital of Nanjing Medical UniversityKey Laboratory of Liver TransplantationChinese Academy of Medical SciencesNHC Key Laboratory of Living Donor Liver TransplantationNanjingChina
- State Key Laboratory of Reproductive MedicineNanjing Medical UniversityNanjingChina
| | - Xuehao Wang
- Hepatobiliary CenterThe First Affiliated Hospital of Nanjing Medical UniversityKey Laboratory of Liver TransplantationChinese Academy of Medical SciencesNHC Key Laboratory of Living Donor Liver TransplantationNanjingChina
| |
Collapse
|
30
|
Mekky MA, Salama RH, Abdel-Aal MF, Ghaliony MA, Zaky S. Studying the frequency of aberrant DNA methylation of APC, P14, and E-cadherin genes in HCV-related hepatocarcinogenesis. Cancer Biomark 2018; 22:503-509. [PMID: 29865038 DOI: 10.3233/cbm-171156] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Data about the molecular pathogenesis of hepatitis C-related hepatocellular carcinoma (HCC) are still challenging. OBJECTIVES Therefore, we tried to investigate the epigenetic study of three nominated genes (APC, P14, and E-cadherin) in the pathogenesis of HCV-related HCC in Egyptian. METHODS Between March 2016 and March 2017, the DNA methylation, and quantification using (epigenetic ELISA kit) for E-cadherin, APC, and P14 genes were studied in three groups of patients: HCV related liver cirrhosis without HCC group (LC-group; n= 20), HCC on top of HCV-related cirrhosis (HCC-group; n= 20), and a third apparently healthy control group (control-group; n= 10). RESULTS E-cad methylation showed non-significant differences between groups. P14 methylation was occurred only in HCC-group (45%). APC methylation was the highest in HCC group (70%). Methylation level was high in HCC group in comparison to both LC and control groups (P< 0.001). DNA methylation at a cutoff point > 2.9 ng/ml predicts HCC in LC-group with 90% sensitivity and 80% specificity and at level > 2.3 ng/ml had 95% sensitivity and 90% specificity in control-group. The pooled sensitivity, specificity, positive and negative predictive values and accuracy were 90%, 60%, 69.2, 85.7 and 75% respectively. CONCLUSION Aberrant DNA methylation of multiple genes is associated with disease progression in HCV related cirrhosis. Moreover, early detection of promotor methylation of these may sever as good biomarker for early detection and therapeutic targets in high risk patients.
Collapse
Affiliation(s)
- Mohamed A Mekky
- Department of Tropical Medicine and Gastroenterology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Rgaa H Salama
- Department of Biochemistry, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Mahmoud F Abdel-Aal
- Department of Tropical Medicine and Gastroenterology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Mohamed A Ghaliony
- Department of Tropical Medicine and Gastroenterology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Saad Zaky
- Department of Tropical Medicine and Gastroenterology, Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
31
|
Wang J, Xia C, Pu M, Dai B, Yang X, Shang R, Yang Z, Zhang R, Tao K, Dou K. Silencing of CDCA5 inhibits cancer progression and serves as a prognostic biomarker for hepatocellular carcinoma. Oncol Rep 2018; 40:1875-1884. [PMID: 30015982 PMCID: PMC6111608 DOI: 10.3892/or.2018.6579] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 07/09/2018] [Indexed: 12/15/2022] Open
Abstract
Cell division cycle associated 5 (CDCA5) has been associated with the progression of several types of cancers. However, its possible role and mechanism in hepatocellular carcinoma (HCC) remain unknown. In the present study, immunohistochemical staining and real-time PCR were used to assess CDCA5 protein and mRNA levels in clinical samples. Statistical analysis was performed to explore the clinical correlation between CDCA5 protein expression and clinicopathological features and overall survival in HCC patients. Cell counting and colony formation assays were employed to analyse the effect of CDCA5 on cell proliferation, and flow cytometry was used to study the role of CDCA5 in cell cycle progression and apoptosis. Moreover, subcutaneous xenograft tumour models were implemented to predict the efficacy of targeting CDCA5 in HCC in vivo. We found that CDCA5 expression was significantly higher in HCC tumour tissues, was associated with clinicopathological characteristics, and predicted poor overall survival in HCC patients. Silencing of CDCA5 with small interfering RNA (siRNA) inhibited cell proliferation and induced G2/M cell cycle arrest in vitro. The xenograft growth assay revealed that CDCA5 downregulation impeded HCC growth in vivo. Further study indicated that CDCA5 depletion decreased the levels of ERK1/2 and AKT phosphorylation in vitro and in vivo. Taken together, these results indicate that CDCA5 may act as a novel prognostic biomarker and therapeutic target for HCC.
Collapse
Affiliation(s)
- Jianlin Wang
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Congcong Xia
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Meng Pu
- Department of Hepatobiliary Surgery, The Air Force General Hospital of the People's Liberation Army, Beijing 100142, P.R. China
| | - Bin Dai
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Xisheng Yang
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Runze Shang
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Zhen Yang
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Ruohan Zhang
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Kaishan Tao
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Kefeng Dou
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| |
Collapse
|
32
|
Huang Y, Wei L, Zhao RC, Liang WB, Zhang J, Ding XQ, Li ZL, Sun CJ, Li B, Liu QY, He JY, Yu XQ, Gao B, Chen MM, Sun AM, Qin Y. Predicting hepatocellular carcinoma development for cirrhosis patients via methylation detection of heparocarcinogenesis-related genes. J Cancer 2018; 9:2203-2210. [PMID: 29937940 PMCID: PMC6010674 DOI: 10.7150/jca.24024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Accepted: 04/04/2018] [Indexed: 02/05/2023] Open
Abstract
Background: Most hepatocellular carcinoma (HCC) patients have undergone a progression from chronic hepatitis, then liver cirrhosis (LC), and finally to carcinoma. The objective of this study was to elucidate risk factors to predict HCC development for cirrhosis patients. Methods: Multiple methylated specific PCR (MSP) was applied to determine methylation status of heparocarcinogenesis-related genes in 396 tissue and plasma specimens and multivariate cox model was used to analyze the relationship between risk variables and HCC development among cirrhosis patients, followed up in a median period of 30 months. Results: Among 105 LC cases, HCC incidence rate at 30 months was 30.48% (32/105), which were statistically associated with patients' age and aberrant methylation of p16, SFRP, and LINE1 (p<0.05). Receiver operating characteristic (ROC) curve showed the overall predictive accuracy reached the highest (90.7%) if the four risk variables were concurrent to predict HCC development. Moreover, along with the growth of age from 0-40, 40-55, to 55-70 years or the increased number of aberrantly-methylated gene from 0-1 to 2-3, the HCC incidence rate of cirrhosis patients rised from 10.00%, 12.28% to 82.14% and 17.44% to 89.47%, separately. Thus, based on combined analysis with diverse age and number of aberrantly-methylated gene, 105 cases were divided into five groups and computed their respective HCC incidecne rate to categorize them into different risk groups. Of note, A significant lifting of HCC incidence rate in the high-risk group (40-55 years coupled with 2-3 aberrantly-methylated genes, 55-70 years coupled with 0-1 aberrantly-methylated gene, 55-70 years coupled with 2-3 aberrantly-methylated genes; n=33) was observed compared with the low-risk group (0-40 years coupled with 0-1 aberrantly-methylated gene, 40-55 years coupled with 0-1 aberrantly-methylated gene; (n=72) (p<0.01). Conclusions: Ultimately, high-risk cirrhosis patients with 55-over years or 2-3 aberrantly-methylated genes should be paid more attention to be regularly screened with HCC development.
Collapse
Affiliation(s)
- Yuan Huang
- Department of Biochemistry and Molecular Biology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Ling Wei
- Department of Biochemistry and Molecular Biology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Rong-Ce Zhao
- Division of Liver Transplantation, Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province,China
| | - Wei-Bo Liang
- Department of Forensic Genetics, West China School of Basic Science and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Jing Zhang
- West China School of Public Health, Sichuan University, Chengdu 610041, China
| | - Xue-Qin Ding
- West China School of Public Health, Sichuan University, Chengdu 610041, China
| | - Zhi-Long Li
- Department of Forensic Genetics, West China School of Basic Science and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Cheng-Jun Sun
- West China School of Public Health, Sichuan University, Chengdu 610041, China
| | - Bo Li
- Division of Liver Transplantation, Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province,China
| | - Qiu-Ying Liu
- Department of Biochemistry and Molecular Biology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Jing-Yang He
- Department of Biochemistry and Molecular Biology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Xiao-Qin Yu
- Department of Biochemistry and Molecular Biology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Bo Gao
- Analytical & testing center, Sichuan University, Chengdu, Sichuan Province, China
| | - Ming-Mei Chen
- Department of Biochemistry and Molecular Biology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Ai-Min Sun
- Analytical & testing center, Sichuan University, Chengdu, Sichuan Province, China
| | - Yang Qin
- Department of Biochemistry and Molecular Biology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| |
Collapse
|
33
|
Shin S, Kim M, Lee SJ, Park KS, Lee CH. Trichostatin A Sensitizes Hepatocellular Carcinoma Cells to Enhanced NK Cell-mediated Killing by Regulating Immune-related Genes. Cancer Genomics Proteomics 2018; 14:349-362. [PMID: 28871002 DOI: 10.21873/cgp.20045] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 07/18/2017] [Accepted: 07/20/2017] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND/AIM Hepatocellular carcinoma (HCC) is the second leading cause of cancer-related death worldwide. The ability of HCC to avoid immune detection is considered one of the main factors making it difficult to cure. Abnormal histone deacetylation is thought to be one of the mechanisms for HCC immune escape, making histone deacetylases (HDACs) attractive targets for HCC treatment. Here, we investigated the effect of trichostatin A (TSA), a highly potent HDAC inhibitor, on HCC (HepG2) gene expression and function. MATERIALS AND METHODS A genome wide-transcriptional microarray was used to identify genes regulated by TSA in HepG2 cells. Gene Ontology was used to identify pathways regulated by TSA, and these changes were confirmed by qPCR. The effect of TSA on natural killer (NK) cell-mediated killing of HCC cell lines were analyzed by both flow cytometry and LDH cytotoxicity assay. A study was also conducted in a Balb/c nude mice xenograft model to assess the anti-tumor activity of TSA. RESULTS TSA regulated the transcription of numerous innate immunity & tumor antigen recognition-associated genes, such as ULBP1 and RAET1G, in HCC cells. In vivo, TSA reduced tumor cell growth in an NK cell-dependent manner. In vitro, TSA treatment of HepG2 cells rendered them more susceptible to NK cell-mediated killing while increasing the expression of NKGD2 ligands, including ULBP1/2/3 and MICA/B. TSA also induced direct killing of HCC cells by stimulating apoptosis. CONCLUSION TSA likely increases killing of HCC cells indirectly by increasing NK cell-directed killing and directly by increasing apoptosis.
Collapse
Affiliation(s)
- Sangsu Shin
- Department of Animal Biotechnology, Kyungpook National University, Sangju, Republic of Korea
| | - Miok Kim
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon, Republic of Korea.,Bio & Drug Discovery Division, Center for Drug Discovery Technology, Korea Research Institute of Chemical Technology (KRICT), Daejeon, Republic of Korea
| | - Seon-Jin Lee
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Kang-Seo Park
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Chang Hoon Lee
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon, Republic of Korea .,Bio & Drug Discovery Division, Center for Drug Discovery Technology, Korea Research Institute of Chemical Technology (KRICT), Daejeon, Republic of Korea
| |
Collapse
|
34
|
Horiuchi Y, Lin J, Shinojima Y, Fujiwara K, Moriyama M, Nagase H. Characterizing key nucleotide polymorphisms of hepatitis C virus-disease associations via mass-spectrometric genotyping. Int J Oncol 2017; 52:441-452. [PMID: 29207078 DOI: 10.3892/ijo.2017.4209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 10/16/2017] [Indexed: 11/06/2022] Open
Abstract
As more than 80% of hepatocellular carcinoma patients in Japan also suffer from hepatitis C virus infections some time in their medical history, identifying genetic aberrations associated to hepatitis C virulence in these individuals remains a high priority in the diagnosis and treatment of hepatocellular carcinoma. From the BioBank Japan Project, we acquired 480 subjects of hepatocellular carcinoma, chronic hepatitis and liver cirrhosis, and genotyped 131 clinically relevant host single nucleotide polymorphisms to survey the potential association between certain risk alleles and genes to a patient's predisposition to hepatitis C and liver cancer. Among those polymorphisms, we found 12 candidates with statistical significance to support association with hepatitis C virus susceptibility and genetic predisposition to hepatocellular carcinoma. SNPs in genes such as XPC, FANCA, KDR and BRCA2 also suggested likely connections between hepatitis C virus susceptibility and the contraction of liver diseases. Single nucleotide polymorphisms reported here provided suggestions for genes as biomarkers and elucidated insights briefing the linkage of hepatitis C virulence to the alteration of healthy liver genomic landscape as well as liver disease progression.
Collapse
Affiliation(s)
- Yuta Horiuchi
- Institute of Gastroenterology, Nihon University School of Medicine, Itabashi-ku, Tokyo 173-8610, Japan
| | - Jason Lin
- Laboratory of Cancer Genetics, Chiba Cancer Center Research Institute, Chuo-ku, Chiba 260-8717, Japan
| | - Yui Shinojima
- Department of Cancer Genetics, Nihon University School of Medicine, Itabashi-ku, Tokyo 173-8610, Japan
| | - Kyoko Fujiwara
- Department of Cancer Genetics, Nihon University School of Medicine, Itabashi-ku, Tokyo 173-8610, Japan
| | - Mitsuhiko Moriyama
- Institute of Gastroenterology, Nihon University School of Medicine, Itabashi-ku, Tokyo 173-8610, Japan
| | - Hiroki Nagase
- Laboratory of Cancer Genetics, Chiba Cancer Center Research Institute, Chuo-ku, Chiba 260-8717, Japan
| |
Collapse
|
35
|
Zhang SQ, Yang Z, Cai XL, Zhao M, Sun MM, Li J, Feng GX, Feng JY, Ye LH, Niu JQ, Zhang XD. miR-511 promotes the proliferation of human hepatoma cells by targeting the 3'UTR of B cell translocation gene 1 (BTG1) mRNA. Acta Pharmacol Sin 2017; 38:1161-1170. [PMID: 28603285 DOI: 10.1038/aps.2017.62] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 02/27/2017] [Indexed: 12/17/2022]
Abstract
Aberrant expression of miR-511 is involved in the development of cancer, but the role of miR-511 in hepatocellular carcinoma (HCC) is not well documented. In this study, we explored the molecular mechanisms of miR-511 in hepatocarcinogenesis. Our results of bioinformatics analysis suggested that B cell translocation gene 1 (BTG1), a member of anti-proliferative gene family, was one of the putative targets of miR-511. The expression levels of miR-511 were significantly higher in 30 clinical HCC tissues than in corresponding peritumor tissues, and were negatively correlated with those of BTG1 in the HCC tissues (r=-0.6105, P<0.01). In human hepatoma cell lines HepG2 and H7402, overexpression of miR-511 dose-dependently inhibited the expression of BTG1, whereas knockdown of miR-511 dose-dependently increased the expression of BTG1. Luciferase reporter gene assays verified that miR-511 targeted the 3'UTR of BTG1 mRNA. In the hepatoma cells, overexpression of miR-511 significantly decreased BTG1-induced G1 phase arrest, which was rescued by overexpression of BTG1. Furthermore, overexpression of miR-511 promoted the proliferation of the hepatoma cells, which was rescued by overexpression of BTG1. Conversely, knockdown of miR-511 inhibited cell proliferation, which was reversed by knockdown of BTG1. In conclusion, miR-511 promotes the proliferation of human hepatoma cells in vitro by targeting the 3'UTR of BTG1 mRNA.
Collapse
|
36
|
Pu M, Wang J, Huang Q, Zhao G, Xia C, Shang R, Zhang Z, Bian Z, Yang X, Tao K. High MRPS23 expression contributes to hepatocellular carcinoma proliferation and indicates poor survival outcomes. Tumour Biol 2017; 39:1010428317709127. [PMID: 28714366 DOI: 10.1177/1010428317709127] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Hepatocellular carcinoma is one of the most prevalent neoplasms and the leading cause of cancer-related mortality worldwide. Mitochondrial ribosomal protein S23 is encoded by a nuclear gene and participates in mitochondrial protein translation. Mitochondrial ribosomal protein S23 overexpression has been found in many types of cancer. In this study, we explored mitochondrial ribosomal protein S23 expression in primary hepatocellular carcinoma tissues compared with matched adjacent non-tumoral liver tissues using mitochondrial ribosomal protein S23 messenger RNA and protein levels collected from public databases and clinical samples. Immunohistochemistry was performed to analyze the relationship between mitochondrial ribosomal protein S23 and various clinicopathological features. The results indicated that mitochondrial ribosomal protein S23 was significantly overexpressed in hepatocellular carcinoma. High mitochondrial ribosomal protein S23 expression was correlated with the tumor size and tumor–metastasis–node stage. Moreover, patients with high mitochondrial ribosomal protein S23 expression levels presented poorer survival rates. Mitochondrial ribosomal protein S23 was an independent prognostic factor for survival, especially at the early stage of hepatocellular carcinoma. In addition, the downregulation of mitochondrial ribosomal protein S23 decreased the proliferation of hepatocellular carcinoma in vitro and in vivo. In conclusion, we verified for the first time that mitochondrial ribosomal protein S23 expression was upregulated in hepatocellular carcinoma. High mitochondrial ribosomal protein S23 levels can predict poor clinical outcomes in hepatocellular carcinoma, and this protein plays a key role in tumor proliferation. Therefore, mitochondrial ribosomal protein S23 may be a potential therapeutic target for hepatocellular carcinoma.
Collapse
Affiliation(s)
- Meng Pu
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Jianlin Wang
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Qike Huang
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Ge Zhao
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Congcong Xia
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Runze Shang
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Zhuochao Zhang
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Zhenyuan Bian
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Xishegn Yang
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Kaishan Tao
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| |
Collapse
|
37
|
Arai E, Yotani T, Kanai Y. DNA and Histone Methylation in Liver Cancer. CANCER DRUG DISCOVERY AND DEVELOPMENT 2017:437-460. [DOI: 10.1007/978-3-319-59786-7_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
38
|
Jiang Y, Chen J, Yue C, Zhang H, Chen T. Trichloroethylene-Induced DNA Methylation Changes in Male F344 Rat Liver. Chem Res Toxicol 2016; 29:1773-1777. [DOI: 10.1021/acs.chemrestox.6b00257] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Yan Jiang
- Department of Physiology, School of Biology
and Basic Medical Sciences, ‡Jiangsu Key Laboratory
of Preventive and Translational Medicine for Geriatric Diseases, §Department of Toxicology,
School of Public Health, Soochow University, Suzhou 215123, China
| | - Jiahong Chen
- Department of Physiology, School of Biology
and Basic Medical Sciences, ‡Jiangsu Key Laboratory
of Preventive and Translational Medicine for Geriatric Diseases, §Department of Toxicology,
School of Public Health, Soochow University, Suzhou 215123, China
| | - Cong Yue
- Department of Physiology, School of Biology
and Basic Medical Sciences, ‡Jiangsu Key Laboratory
of Preventive and Translational Medicine for Geriatric Diseases, §Department of Toxicology,
School of Public Health, Soochow University, Suzhou 215123, China
| | - Hang Zhang
- Department of Physiology, School of Biology
and Basic Medical Sciences, ‡Jiangsu Key Laboratory
of Preventive and Translational Medicine for Geriatric Diseases, §Department of Toxicology,
School of Public Health, Soochow University, Suzhou 215123, China
| | - Tao Chen
- Department of Physiology, School of Biology
and Basic Medical Sciences, ‡Jiangsu Key Laboratory
of Preventive and Translational Medicine for Geriatric Diseases, §Department of Toxicology,
School of Public Health, Soochow University, Suzhou 215123, China
| |
Collapse
|