1
|
Xia Q, Liu G, Lin W, Zhang J. microRNA-2117 Negatively Regulates Liver Cancer Stem Cells Expansion and Chemoresistance Via Targeting SOX2. Mol Carcinog 2024. [PMID: 39400383 DOI: 10.1002/mc.23824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 09/11/2024] [Accepted: 09/18/2024] [Indexed: 10/15/2024]
Abstract
Cancer stem cells (CSCs) are involved in the regulation of tumor initiation, progression, recurrence, and chemoresistance. However, the role of microRNAs (miRNAs) in liver CSCs has not been fully understood. Here we show that miR-2117 is downregulated in liver CSCs and predicts the poor prognosis of hepatocellular carcinoma (HCC) patients. Biofunction studies found that knockdown miR-2117 facilitates liver CSCs self-renewal and tumorigenesis. Conversely, forced miR-2117 expression suppresses liver CSCs self-renewal and tumorigenesis. Mechanistically, we find that transcription factor SOX2 is required for miR-2117-mediated liver CSCs expansion. The correlation between miR-2117 and SOX2 was confirmed in human HCC tissues. More importantly, miR-2117 overexpression HCC cells are more sensitive to CDDP treatment. Analysis of patients' cohort further demonstrates that miR-2117 may predict transcatheter arterial chemoembolization benefits in HCC patients. Our findings revealed the crucial role of miR-2117 in liver CSCs expansion, rendering miR-2117 as an optimal therapeutic target for HCC.
Collapse
Affiliation(s)
- Qing Xia
- Department of General Surgery, Hwa Mei Hospital (Ningbo No.2 Hospital), University of Chinese Academy of Sciences, Ningbo, China
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, China
- Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo, China
| | - Guanghua Liu
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Interventional Radiology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wenbo Lin
- Department of Orthopedic Surgery, Changzheng Hospital, Navy Medical University, Shanghai, China
| | - Jin Zhang
- Department of General Surgery, Third Affiliated Hospital of Second Military Medical University, Shanghai, China
| |
Collapse
|
2
|
Li T, Hu H, Song Y, Shi Y, Hu D, Shen W, Ning B. DDR1 is a Novel Biomarker and Potential Therapeutic Target for the Combination Treatment of Liver Hepatocellular Carcinoma. Cancer Control 2024; 31:10732748241286257. [PMID: 39284684 PMCID: PMC11406661 DOI: 10.1177/10732748241286257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024] Open
Abstract
AIM This study aimed to investigate the role of discoidin domain receptor tyrosine kinase 1 (DDR1) in liver hepatocellular carcinoma (LIHC) and to evaluate its prognostic value on patient response to combination therapy. METHODS In the current retrospective study, we examined the protein expression of DDR1 in various cancers by standard immunohistochemical (IHC) methods and evaluated its clinical significance in LIHC personalized treatment. Multiple online databases, including The Cancer Genome Atlas (TCGA), TIMER, GEO, ROC Plotter, and Genomics of Drug Sensitivity in Cancer (GDSC), were used. RESULTS DDR1 protein expression was higher in LIHC than in other nine examined cancer types. Additionally, DDR1 exhibited higher expression levels in adjacent normal tissues compared to HBs-positive LIHC tissues. Analysis at single-cell resolution revealed that DDR1 was expressed primarily in epithelial cells but not in stromal and immune cells, and DDR1 expression was lower in HBs-positive LIHC cells in comparison with normal hepatocytes. Correlation of DDR1 upregulation and sorafenib resistance was observed in the patient cohort. Moreover, DDR1 expression positively correlated with the expression of inflammatory response-related genes, ECM-related genes, and collagen formation-related genes, but negatively correlated with the infiltration of CD8+ T cells, NK cells, and dendritic cells in LIHC. CONCLUSIONS Our findings suggest that DDR1 expression might be induced by collagen production-related cellular events involved in liver injury and repair, and that DDR1 overexpression might contribute to the resistance to LIHC targeted therapy and immunotherapy, highlighting DDR1 as a potential prognostic biomarker and therapeutic target.
Collapse
Affiliation(s)
- Tianxing Li
- Clinical Cancer Institute, Center for Translational Medicine, Naval Medical University, Shanghai, China
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Hao Hu
- Department of Pathology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yuhao Song
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Yihai Shi
- Department of Gastroenterology, Shanghai Pudong New Area Gongli Hospital, Shanghai, China
| | - Dingtao Hu
- Clinical Cancer Institute, Center for Translational Medicine, Naval Medical University, Shanghai, China
| | - Weifeng Shen
- The Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Beifang Ning
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
3
|
Corneillie L, Lemmens I, Weening K, De Meyer A, Van Houtte F, Tavernier J, Meuleman P. Virus-Host Protein Interaction Network of the Hepatitis E Virus ORF2-4 by Mammalian Two-Hybrid Assays. Viruses 2023; 15:2412. [PMID: 38140653 PMCID: PMC10748205 DOI: 10.3390/v15122412] [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: 09/14/2023] [Revised: 12/01/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
Throughout their life cycle, viruses interact with cellular host factors, thereby influencing propagation, host range, cell tropism and pathogenesis. The hepatitis E virus (HEV) is an underestimated RNA virus in which knowledge of the virus-host interaction network to date is limited. Here, two related high-throughput mammalian two-hybrid approaches (MAPPIT and KISS) were used to screen for HEV-interacting host proteins. Promising hits were examined on protein function, involved pathway(s), and their relation to other viruses. We identified 37 ORF2 hits, 187 for ORF3 and 91 for ORF4. Several hits had functions in the life cycle of distinct viruses. We focused on SHARPIN and RNF5 as candidate hits for ORF3, as they are involved in the RLR-MAVS pathway and interferon (IFN) induction during viral infections. Knocking out (KO) SHARPIN and RNF5 resulted in a different IFN response upon ORF3 transfection, compared to wild-type cells. Moreover, infection was increased in SHARPIN KO cells and decreased in RNF5 KO cells. In conclusion, MAPPIT and KISS are valuable tools to study virus-host interactions, providing insights into the poorly understood HEV life cycle. We further provide evidence for two identified hits as new host factors in the HEV life cycle.
Collapse
Affiliation(s)
- Laura Corneillie
- Laboratory of Liver Infectious Diseases, Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Irma Lemmens
- VIB-UGent Center for Medical Biotechnology, Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Karin Weening
- Laboratory of Liver Infectious Diseases, Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Amse De Meyer
- Laboratory of Liver Infectious Diseases, Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Freya Van Houtte
- Laboratory of Liver Infectious Diseases, Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Jan Tavernier
- VIB-UGent Center for Medical Biotechnology, Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Philip Meuleman
- Laboratory of Liver Infectious Diseases, Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
4
|
Xu H, Ma Z, Mo X, Chen X, Xu F, Wu F, Chen H, Zhou G, Xia H, Zhang C. Inducing Synergistic DNA Damage by TRIP13 and PARP1 Inhibitors Provides a Potential Treatment for Hepatocellular Carcinoma. J Cancer 2022; 13:2226-2237. [PMID: 35517402 PMCID: PMC9066198 DOI: 10.7150/jca.66020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 12/20/2021] [Indexed: 12/26/2022] Open
Abstract
Thyroid hormone receptor interactor 13 (TRIP13), an AAA-ATPase, participates in the development of many cancers. This study explores the function of TRIP13 and synergistic effects of TRIP13 and PARP1 inhibitors in hepatocellular carcinoma (HCC). The dose-dependent effects of TRIP13 and PARP1 inhibitors on HCC cells proliferation or migration were investigated by the CCK-8 and Transwell assays. Using siRNA or lentivirus to knock down TRIP13, we tested HCC cell and tumor growth in vitro and in vivo. The DNA damage caused by TRIP13 and PARP1 inhibitors was measured by the phosphorylation of H2AX, one of the DNA damage biomarkers. The phosphorylation of H2AX was increased after treatment with DCZ0415 or TRIP13 knockdown. Combining DCZ0415 with PARP1 inhibitor, Olaparib induced synergistic anti-HCC activity. We also found that the overexpression of TRIP13 is significantly associated with early recurrent HCC and poor survival. Up-regulation of TRIP13 in HCC was regulated by transcription factor SP1. In conclusion, our study demonstrated that DCZ0415 targeting TRIP13 impaired non-homologous end-joining repair to inhibit HCC progression and had a synergistic effect with PARP1 inhibitor Olaparib in HCC, suggesting a potential treatment of HCC.
Collapse
Affiliation(s)
- Haojun Xu
- School of Basic Medical Sciences &Key Laboratory of Antibody Technique of National Health Commission & Jiangsu Antibody Drug Engineering Research Center, Nanjing Medical University, Nanjing 211166, China
| | - Zhijie Ma
- School of Basic Medical Sciences &Key Laboratory of Antibody Technique of National Health Commission & Jiangsu Antibody Drug Engineering Research Center, Nanjing Medical University, Nanjing 211166, China
| | - Xiao Mo
- Department of Pathology, The first people's hospital of Foshan, Foshan 528041, China.,School of Basic Medical Sciences &Key Laboratory of Antibody Technique of National Health Commission & Jiangsu Antibody Drug Engineering Research Center, Nanjing Medical University, Nanjing 211166, China
| | - Xiaoli Chen
- Sir Run Run Hospital, Nanjing Medical University, Nanjing 211166, China
| | - Fanggui Xu
- Sir Run Run Hospital, Nanjing Medical University, Nanjing 211166, China
| | - Fubing Wu
- Sir Run Run Hospital, Nanjing Medical University, Nanjing 211166, China
| | - Hongjin Chen
- School of Basic Medical Sciences &Key Laboratory of Antibody Technique of National Health Commission & Jiangsu Antibody Drug Engineering Research Center, Nanjing Medical University, Nanjing 211166, China
| | - Guoren Zhou
- Jiangsu Cancer Hospital & The Affiliated Cancer Hospital of Nanjing Medical University& Jiangsu Institute of Cancer Research, Nanjing 2100092, China
| | - Hongping Xia
- School of Basic Medical Sciences &Key Laboratory of Antibody Technique of National Health Commission & Jiangsu Antibody Drug Engineering Research Center, Nanjing Medical University, Nanjing 211166, China.,Sir Run Run Hospital, Nanjing Medical University, Nanjing 211166, China
| | - Chengfei Zhang
- Sir Run Run Hospital, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
5
|
Ye W, Shi Z, Zhou Y, Zhang Z, Zhou Y, Chen B, Zhang Q. Autophagy-Related Signatures as Prognostic Indicators for Hepatocellular Carcinoma. Front Oncol 2022; 12:654449. [PMID: 35402224 PMCID: PMC8987527 DOI: 10.3389/fonc.2022.654449] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 02/21/2022] [Indexed: 01/13/2023] Open
Abstract
Background Hepatocellular carcinoma (HCC) is the most common and deadly type of liver cancer. Autophagy is the process of transporting damaged or aging cellular components into lysosomes for digestion and degradation. Accumulating evidence implies that autophagy is a key factor in tumor progression. The aim of this study was to determine a panel of novel autophagy-related prognostic markers for liver cancer. Methods We conducted a comprehensive analysis of autophagy-related gene (ARG) expression profiles and corresponding clinical information based on The Cancer Genome Atlas (TCGA) and International Cancer Genome Consortium (ICGC) databases. The univariate Cox proportional regression model was used to screen candidate autophagy-related prognostic genes. In addition, a multivariate Cox proportional regression model was used to identify five key prognostic autophagy-related genes (ATIC, BAX, BIRC5, CAPNS1, and FKBP1A), which were used to construct a prognostic signature. Real-time qPCR analysis was used to evaluate the expression levels of ARGs in 20 surgically resected HCC samples and matched tumor-adjacent normal tissue samples. In addition, the effect of FKBP1A on autophagy and tumor progression was determined by performing in vitro and in vivo experiments. Results Based on the prognostic signature, patients with liver cancer were significantly divided into high-risk and low-risk groups in terms of overall survival (OS). A subsequent multivariate Cox regression analysis indicated that the prognostic signature remained an independent prognostic factor for OS. The prognostic signature possessing a better area under the curve (AUC) displayed better performance in predicting the survival of patients with HCC than other clinical parameters. Furthermore, FKBP1A was overexpressed in HCC tissues, and knockdown of FKBP1A impaired cell proliferation, migration, and invasion through the PI3K/AKT/mTOR signaling pathway. Conclusion This study provides a prospective biomarker for monitoring outcomes of patients with HCC.
Collapse
Affiliation(s)
- Wen Ye
- Department of Breast Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhehao Shi
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yilin Zhou
- College of Engineering, Boston University, Boston, MA, United States
| | - Zhongjing Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yi Zhou
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Bicheng Chen
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
- *Correspondence: Qiyu Zhang, ; Bicheng Chen,
| | - Qiyu Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
- *Correspondence: Qiyu Zhang, ; Bicheng Chen,
| |
Collapse
|
6
|
Significant Diagnostic and Prognostic Value of FLAD1 and Related MicroRNAs in Breast Cancer after a Pan-Cancer Analysis. DISEASE MARKERS 2021; 2021:6962526. [PMID: 34336008 PMCID: PMC8321750 DOI: 10.1155/2021/6962526] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 05/30/2021] [Accepted: 06/24/2021] [Indexed: 12/09/2022]
Abstract
The identification of biomarkers plays an important role in the diagnosis and prognosis of cancers. In this study, we explored the diagnostic and prognostic value of the FLAD1 expression across pan-cancer analysis from online databases (Oncomine, cBioPortal, Breast Cancer Gene-Expression Miner, UALCAN, GEO, BCIP, TNMplot, ENCORI, Kaplan-Meier Plotter, and LinkedOmics). We found that FLAD1 was overexpressed in a number of different kinds of cancers, especially in breast cancer, and higher FLAD1 expression level was associated with the HER+, p53 mutant, node-involved, NPI stage 3, basal-like, and triple-negative groups compared with the other subgroups of breast cancer. The FLAD1 expression levels were higher in patients that were 21–40 years old than those in patients of other ages and were higher in the African-American group than in the Caucasian group. We also analyzed the FLAD1-related microRNAs and their prognostic values in breast cancer. This study highlights the significance of FLAD1 in cancers and provides evidence for its potential as a biomarker for the diagnosis and prognosis of cancers.
Collapse
|
7
|
Yang Q, Qi M, Chen Y, Tian S, Liao F, Dong W. ASPM is a Novel Candidate Gene Associated with Colorectal Cancer Cell Growth. DNA Cell Biol 2021; 40:921-935. [PMID: 34042518 DOI: 10.1089/dna.2020.6457] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most prevalent diseases worldwide; however, the molecular mechanisms involved in CRC remain unclear. Thus, we aimed to explore a novel biomarker for CRC. In this study, we screened 361 differentially expressed genes; 152 downregulated genes; and 209 upregulated genes) through analysis of the GSE44861, GSE110223, GSE110224, and GSE113513 CRC datasets. Next, ASPM, CCNA2, CCNB1, CEP55, KIF20A, MAD2L1, MELK, RRM2, TOP2A, TPX2, TRIP13, and TTK were identified as hub genes associated with the cell cycle in CRC through comprehensive bioinformatics analysis using the Cytoscape and Metascape software, the Database for Annotation, Visualization, and Integrated Discovery (DAVID), and the Oncomine and Gene Expression Profiling Interactive Analysis 2 (GEPIA2) databases. Furthermore, ASPM mRNA expression in CRC tissues was verified in Oncomine, The Cancer Genome Atlas and our data, and ASPM was found to be significantly upregulated in CRC tissues compared with that in the noncancer colon tissues. Functionally, we showed that overexpression of ASPM significantly promoted the proliferation and inhibited apoptosis; silencing of ASPM suppressed the proliferation of CRC cells by affecting the cell cycle G1/S transition by reducing cyclin E1 expression, and inducing apoptosis. Overall, our findings indicated that ASPM plays a crucial role in the regulation of CRC cell proliferation, and ASPM is a potential candidate diagnostic tool and therapeutic target for CRC.
Collapse
Affiliation(s)
- Qian Yang
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, P.R. China.,Key Laboratory of Hubei Province for Digestive System Disease, Wuhan, P.R. China.,Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, P.R. China
| | - Mingming Qi
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, P.R. China.,Key Laboratory of Hubei Province for Digestive System Disease, Wuhan, P.R. China.,Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, P.R. China
| | - Yongyu Chen
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, P.R. China.,Key Laboratory of Hubei Province for Digestive System Disease, Wuhan, P.R. China
| | - Shan Tian
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, P.R. China
| | - Fei Liao
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, P.R. China
| | - Weiguo Dong
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, P.R. China.,Key Laboratory of Hubei Province for Digestive System Disease, Wuhan, P.R. China
| |
Collapse
|
8
|
Aspartame induces cancer stem cell enrichment through p21, NICD and GLI1 in human PANC-1 pancreas adenocarcinoma cells. Food Chem Toxicol 2021; 153:112264. [PMID: 33992720 DOI: 10.1016/j.fct.2021.112264] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 04/30/2021] [Accepted: 05/10/2021] [Indexed: 11/21/2022]
Abstract
This study aimed to investigate the molecular effects of the common natural sugar glucose and artificial sweetener aspartame on cancer stem cell (CSC) population and cancer aggressiveness of PANC-1 human pancreas adenocarcinoma cells. According to our findings while aspartame exposure significantly increased the CSC population, high glucose had no effect on it. The epithelial-mesenchymal transition marker N-cadherin increased only in the aspartame group. The findings indicate that a high level of glucose exposure does not effect the invasion and migration of PANC-1 cells, while aspartame increases both of these aggressiveness criteria. The findings also suggest that a high concentration of glucose maintains CSC population through induction of nuclear Oct3/4 and differentiation to parental cells via increasing cytoplasmic c-myc. Aspartame exposure to PANC-1 cells activated AKT and deactivated GSK3β by increasing levels of ROS and cytoplasmic Ca+2, respectively, through T1R2/T1R3 stimulation. Then p-GSK3β(Ser9) boosted the CSC population by increasing pluripotency factors Oct3/4 and c-myc via NICD, GLI1 and p21. In the aspartame group, T1R1 silencing further increased the CSC population but decreased cell viability and suppressed the p21, NICD and GLI activation. The presence and amount of T1R subunits in the membrane fraction of PANC-1 cells are demonstrated for the first time in this study, as is the regulatory effect of T1R1's on CSC population. In conclusion, the present study demonstrated that long-term aspartame exposure increases CSC population and tumor cell aggressiveness through p21, NICD, GLI1. Moreover, while aspartame had no tumorigenic effect, it could potentially advance an existing tumor.
Collapse
|
9
|
Zhang X, Yin S, Ma K. Bioinformatics analysis of different candidate genes involved in hepatocellular carcinoma induced by HepG2 cells or tumor cells of patients. J Int Med Res 2021; 48:300060520932112. [PMID: 32567431 PMCID: PMC7309404 DOI: 10.1177/0300060520932112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE Hepatocellular carcinoma (HCC) is a common cancer with a high mortality rate; the molecular mechanism involved in HCC remain unclear. We aimed to provide insight into HCC induced with HepG2 cells and identify genes and pathways associated with HCC, as well as potential therapeutic targets. METHODS Dataset GSE72581 was downloaded from the Gene Expression Omnibus, including samples from mice injected in liver parenchyma with HepG2 cells, and from mice injected with cells from patient tumor explants. Differentially expressed genes (DEGs) between the two groups of mice were analyzed. Then, gene ontology and Kyoto Encyclopedia of Gene and Genomes pathway enrichment analyses were performed. The MCODE plug-in in Cytoscape was applied to create a protein-protein interaction (PPI) network of DEGs. RESULTS We identified 1,405 DEGs (479 upregulated and 926 downregulated genes), which were enriched in complement and coagulation cascades, peroxisome proliferator-activated receptor signaling pathway, and extracellular matrix-receptor interaction. The top 4 modules and top 20 hub genes were identified from the PPI network, and associations with overall survival were determined using Kaplan-Meier analysis. CONCLUSION This preclinical study provided data on molecular targets in HCC that could be useful in the clinical treatment of HCC.
Collapse
Affiliation(s)
- Xiang Zhang
- Medical School, Yan'an University, Yan'an, P. R. China
| | - Songna Yin
- Medical School, Yan'an University, Yan'an, P. R. China
| | - Ke Ma
- Shandong Co-Innovation Center of Classic TCM formula, Shandong University of Traditional Chinese Medicine, Jinan, P. R. China
| |
Collapse
|
10
|
Yu Z, Feng H, Zhuo Y, Li M, Zhu X, Huang L, Zhang X, Zhou Z, Zheng C, Jiang Y, Le F, Yu DY, Cheng AS, Sun X, Gao Y. Bufalin inhibits hepatitis B virus-associated hepatocellular carcinoma development through androgen receptor dephosphorylation and cell cycle-related kinase degradation. Cell Oncol (Dordr) 2020; 43:1129-1145. [PMID: 32623699 DOI: 10.1007/s13402-020-00546-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/23/2020] [Indexed: 12/30/2022] Open
Abstract
PURPOSE Hepatitis B virus (HBV)-associated hepatocellular carcinoma (HCC), which has a male predominance, lacks effective therapeutic options. Previously, the cardiac glycoside analogue bufalin has been found to inhibit HBV infection and HCC development. As yet, however, its molecular role in HBV-associated HCC has remained obscure. METHODS Colony formation and soft agar assays, xenograft and orthotopic mouse models and HBV X protein (HBx) transgenic mice with exposure to diethylnitrosamine were used to evaluate the effect of bufalin on HBV-associated HCC growth and tumorigenicity. HBx-induced oncogenic signaling regulated by bufalin was assessed using PCR array, chromatin immunoprecipitation, site-directed mutagenesis, luciferase reporter, transcription and protein expression assays. Synergistic HCC therapeutic effects were examined using combinations of bufalin and sorafenib. RESULTS We found that bufalin exerted a more profound effect on inhibiting the proliferation of HBV-associated HCC cells than of non HBV-associated HCC cells. Bufalin significantly inhibited HBx-induced malignant transfromation in vitro and tumorigenicity in vivo. Androgen receptor (AR) signaling was found to be a target of bufalin resistance to HBV-associated hepatocarcinogenesis. We also found that bufalin induced both AR dephosphorylation and cell cycle-related kinase (CCRK) degradation to inhibit β-catenin/TCF signaling, which subsequently led to cell cycle arrest via cyclin D1 down-regulation and p21 up-regulation, resulting in HCC regression. Furthermore, we found that bufalin reduced > 60% diethylnitrosamine-induced hepatocarcinogenesis in HBx transgenic mice, and improved the sensitivity of refractory HBV-associated HCC cells to sorafenib treatment. CONCLUSION Our results indicate that bufalin acts as a potential anti-HCC therapeutic candidate to block HBx-induced AR/CCRK/β-catenin signaling by targeting AR and CCRK, which may provide a novel strategy for the treatment of HBV-associated HCC.
Collapse
Affiliation(s)
- Zhuo Yu
- Liver Disease Department, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No.528. Zhangheng Road, Pudong New District, Shanghai, People's Republic of China.
| | - Hai Feng
- Department of pharmacology, School of Pharmacy, Harbin Medical University, Harbin, People's Republic of China
| | - Yunhui Zhuo
- Liver Disease Department, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No.528. Zhangheng Road, Pudong New District, Shanghai, People's Republic of China
| | - Man Li
- Laboratory of Cellular Immunity, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Xiaojun Zhu
- Liver Disease Department, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No.528. Zhangheng Road, Pudong New District, Shanghai, People's Republic of China
| | - Lingying Huang
- Liver Disease Department, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No.528. Zhangheng Road, Pudong New District, Shanghai, People's Republic of China
| | - Xin Zhang
- Laboratory of Cellular Immunity, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Zhenhua Zhou
- Laboratory of Cellular Immunity, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Chao Zheng
- Liver Disease Department, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No.528. Zhangheng Road, Pudong New District, Shanghai, People's Republic of China
| | - Yun Jiang
- Liver Disease Department, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No.528. Zhangheng Road, Pudong New District, Shanghai, People's Republic of China
| | - Fan Le
- Liver Disease Department, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No.528. Zhangheng Road, Pudong New District, Shanghai, People's Republic of China
| | - Dae-Yeul Yu
- Disease Model Research Laboratory, Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 305-806, Republic of Korea
| | - Alfred Szelok Cheng
- School of Biomedical Sciences, State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, SAR, People's Republic of China
| | - Xuehua Sun
- Liver Disease Department, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No.528. Zhangheng Road, Pudong New District, Shanghai, People's Republic of China.
| | - Yueqiu Gao
- Liver Disease Department, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No.528. Zhangheng Road, Pudong New District, Shanghai, People's Republic of China. .,Laboratory of Cellular Immunity, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China.
| |
Collapse
|
11
|
Nia A, Dhanasekaran R. Genomic Landscape of HCC. CURRENT HEPATOLOGY REPORTS 2020; 19:448-461. [PMID: 33816052 PMCID: PMC8015384 DOI: 10.1007/s11901-020-00553-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/23/2020] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Hepatocellular carcinoma (HCC) is a leading cause of cancer related mortality in the world and it has limited treatment options. Understanding the molecular drivers of HCC is important to develop novel biomarkers and therapeutics. PURPOSE OF REVIEW HCC arises in a complex background of chronic hepatitis, fibrosis and liver regeneration which lead to genomic changes. Here, we summarize studies that have expanded our understanding of the molecular landscape of HCC. RECENT FINDINGS Recent technological advances in next generation sequencing (NGS) have elucidated specific genetic and molecular programs involved in hepatocarcinogenesis. We summarize the major somatic mutations and epigenetic changes have been identified in NGS-based studies. We also describe promising molecular therapies and immunotherapies which target specific genetic and epigenetic molecular events. SUMMARY The genomic landscape of HCC is incredibly complex and heterogeneous. Promising new developments are helping us decipher the molecular drivers of HCC and leading to new therapies.
Collapse
|
12
|
Hu P, Pan Y, Wang C, Zhang W, Huang H, Wang J, Zhang N. FLAD1 is up-regulated in Gastric Cancer and is a potential prediction of prognosis. Int J Med Sci 2020; 17:1763-1772. [PMID: 32714079 PMCID: PMC7378663 DOI: 10.7150/ijms.48162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 06/23/2020] [Indexed: 12/28/2022] Open
Abstract
Background: Gastric cancer (GC) is a common malignancy throughout the world. Biomarkers for prognosis and risk evaluation of GC are rapidly discovered. We investigated the prognostic role of FLAD1, an important protein-coding gene that affects cell cycle and survival. Methods: The expression of FLAD1 at mRNA levels in GC tumor tissues and normal tissues was mined and analyzed in Oncomine database and verified in 10 pairs of GS tissues and their adjacent normal tissues in our center by RT qPCR. The FLAD1 protein expression were detected in 106 paraffin-embedded GC tissues by immunohistochemistry (IHC). Statistical analyses were applied to evaluate the clinical significance of FLAD1. The prognostic value of FLAD1 mRNA expression was also analyzed using the Kaplan-Meier plotter (www.kmplot.com). Results: Statistics obtained from online database suggested FLAD1 mRNA was overexpressed in GC tissues. The results were further validated in 10 pairs of GS tissues and adjacent normal tissues in our center (p=0.021). IHC and survival analysis of GC samples from 106 patients showed FLAD1 was overexpressed in 63/106 (59.4%) patients and was associated to higher TNM stage (p=0.026). Multivariate analysis revealed FLAD1 was an independent prognostic factor for GC (p < 0.001). Furthermore, FLAD1 mRNA was associated to unfavorable overall survival (OS), first progression (FP), and post-progression survival (PPS) of GC (p<0.001). Conclusion: FLAD1 in GC is overexpressed at both mRNA and protein level and could be a potential biomarker for GC prognosis.
Collapse
Affiliation(s)
- Pan Hu
- Breast Cancer Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510000, P.R. China
| | - Yuhang Pan
- Department of Pathology, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou510000, P. R. China
| | - Chenyang Wang
- Department of Urologic Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, 400030, P.R. China
| | - Wenhui Zhang
- Joint Surgery/Orthopedic Trauma Department, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510000, P. R. China
| | - He Huang
- General Surgery Department, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510000, P.R. China
| | - Jiani Wang
- Breast Cancer Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510000, P.R. China
| | - Nana Zhang
- Department of Pathology, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou510000, P. R. China
| |
Collapse
|