1
|
Wang Y, Yan Y, Huo Y, Pang Y, Chan L, Wang S, Chen D, Lin D, Wang W. mRNA sequencing and CyTOF analysis revealed ASPP2 altered the response patterns of hepatocellular carcinoma HepG2 cells to usnic acid. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:1847-1856. [PMID: 36877271 DOI: 10.1007/s00210-023-02428-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 02/16/2023] [Indexed: 03/07/2023]
Abstract
In a previous study, our team found that ASPP2 overexpression increases the sensitivity of liver cancer cells to sorafenib. ASPP2 is an important target in the study of drug therapy for hepatocellular carcinoma. In this study, we demonstrated that ASPP2 altered the response of HepG2 cells to usnic acid (UA) by using mRNA sequencing and CyTOF. CCK8 assay was used to detect cytotoxicity of UA on HepG2 cells. Annexin V-RPE assay, TUNEL assay, and cleaved caspase 3 assay were performed to examine the apoptotic cell death induced by UA. Transcriptomic sequencing and a single-cell mass cytometry were used to analyze the dynamic response of HepG2shcon and HepG2shASPP2 cells to UA treatment. We have demonstrated that UA could inhibit proliferation in HepG2 cells in a concentration-dependent manner. Apoptotic cell death was significantly induced by UA in HepG2 cells, while knocking down ASPP2 could increase the resistance of HepG2 cells to UA. Data from mRNA-Seq indicated that knockout of ASPP2 in HepG2 cells affected cell proliferation, cycle, and metabolism. ASPP2 knockdown resulted in increased stemness and decreased apoptosis of HepG2 cells under the action of UA. CyTOF analysis confirmed the above results, ASPP2 knockdown increased oncoproteins in HepG2 cells and altered response patterns of HepG2 cells to UA. Our data suggested that the natural compound UA could inhibit liver cancer HepG2 cells; meanwhile, ASPP2 knockdown could affect response patterns of HepG2 cells to UA. The above results indicate that ASPP2 could be a research target in the chemoresistance of liver cancer.
Collapse
Affiliation(s)
- Yang Wang
- Beijing Institute of Hepatology Beijing You An Hospital Capital Medical University, Beijing, China
| | - Yadong Yan
- Beijing Institute of Hepatology Beijing You An Hospital Capital Medical University, Beijing, China
- Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing You An Hospital, Capital Medical University, Beijing, China
| | - Yunfei Huo
- Beijing Institute of Hepatology Beijing You An Hospital Capital Medical University, Beijing, China
| | - Yuheng Pang
- Beijing Institute of Hepatology Beijing You An Hospital Capital Medical University, Beijing, China
| | - Liujia Chan
- Beijing Institute of Hepatology Beijing You An Hospital Capital Medical University, Beijing, China
| | - Shanshan Wang
- Beijing Institute of Hepatology Beijing You An Hospital Capital Medical University, Beijing, China
| | - Dexi Chen
- Beijing Institute of Hepatology Beijing You An Hospital Capital Medical University, Beijing, China
| | - Dongdong Lin
- Xuanwu Hospital, Capital Medical University, Beijing, China.
- Beijing You An Hospital, Capital Medical University, Beijing, China.
| | - Wenjing Wang
- Beijing Institute of Hepatology Beijing You An Hospital Capital Medical University, Beijing, China.
| |
Collapse
|
2
|
Relevance of HBx for Hepatitis B Virus-Associated Pathogenesis. Int J Mol Sci 2023; 24:ijms24054964. [PMID: 36902395 PMCID: PMC10003785 DOI: 10.3390/ijms24054964] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/20/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
The hepatitis B virus (HBV) counts as a major global health problem, as it presents a significant causative factor for liver-related morbidity and mortality. The development of hepatocellular carcinomas (HCC) as a characteristic of a persistent, chronic infection could be caused, among others, by the pleiotropic function of the viral regulatory protein HBx. The latter is known to modulate an onset of cellular and viral signaling processes with emerging influence in liver pathogenesis. However, the flexible and multifunctional nature of HBx impedes the fundamental understanding of related mechanisms and the development of associated diseases, and has even led to partial controversial results in the past. Based on the cellular distribution of HBx-nuclear-, cytoplasmic- or mitochondria-associated-this review encompasses the current knowledge and previous investigations of HBx in context of cellular signaling pathways and HBV-associated pathogenesis. In addition, particular focus is set on the clinical relevance and potential novel therapeutic applications in the context of HBx.
Collapse
|
3
|
Liang B, Jiang Y, Song S, Jing W, Yang H, Zhao L, Chen Y, Tang Q, Li X, Zhang L, Bao H, Huang G, Zhao J. ASPP2 suppresses tumour growth and stemness characteristics in HCC by inhibiting Warburg effect via WNT/β-catenin/HK2 axis. J Cell Mol Med 2023; 27:659-671. [PMID: 36752127 PMCID: PMC9983321 DOI: 10.1111/jcmm.17687] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 01/09/2023] [Accepted: 01/16/2023] [Indexed: 02/09/2023] Open
Abstract
Abnormal energy metabolism is one of the characteristics of tumours. In the last few years, more and more attention is being paid to the role and regulation of tumour aerobic glycolysis. Cancer cells display enhanced aerobic glycolysis, also known as the Warburg effect, whereby tumour cells absorb glucose to produce a large amount of lactic acid and energy under aerobic conditions to favour tumour proliferation and metastasis. In this study, we report that the haploinsufficient tumour suppressor ASPP2, can inhibit HCC growth and stemness characteristics by regulating the Warburg effect through the WNT/β-catenin pathway. we performed glucose uptake, lactate production, pyruvate production, ECAR and OCR assays to verify ASPP2 can inhibit glycolysis in HCC cells. The expression of ASPP2 and HK2 was significantly inversely correlated in 80 HCC tissues. Our study reveals downregulation of ASPP2 can promote the aerobic glycolysis metabolism pathway, increasing HCC proliferation, glycolysis metabolism, stemness and drug resistance. This ASPP2-induced inhibition of glycolysis metabolism depends on the WNT/β-catenin pathway. ASPP2-regulated Warburg effect is associated with tumour progression and provides prognostic value. and suggest that may be promising as a new therapeutic strategy in HCC.
Collapse
Affiliation(s)
- Beibei Liang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Yuan Jiang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, China.,Medical Engineering Department, The Affiliated Hospital of QingDao University, Qingdao, Shandong, China
| | - Shaohua Song
- Liver Transplantation Center, Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wei Jing
- Department of Surgery, Changhai Hospital, Navy Military Medical University, Shanghai, China
| | - Hao Yang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Li Zhao
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ya Chen
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, China.,Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qiqi Tang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Xuhui Li
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Lisha Zhang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Haili Bao
- Department of Organ Transplantation, Shanghai Changzheng Hospital, Navy Military Medical University, Shanghai, China
| | - Gang Huang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Jian Zhao
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, China
| |
Collapse
|
4
|
Hashemi M, Nadafzadeh N, Imani MH, Rajabi R, Ziaolhagh S, Bayanzadeh SD, Norouzi R, Rafiei R, Koohpar ZK, Raei B, Zandieh MA, Salimimoghadam S, Entezari M, Taheriazam A, Alexiou A, Papadakis M, Tan SC. Targeting and regulation of autophagy in hepatocellular carcinoma: revisiting the molecular interactions and mechanisms for new therapy approaches. Cell Commun Signal 2023; 21:32. [PMID: 36759819 PMCID: PMC9912665 DOI: 10.1186/s12964-023-01053-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 01/15/2023] [Indexed: 02/11/2023] Open
Abstract
Autophagy is an evolutionarily conserved process that plays a role in regulating homeostasis under physiological conditions. However, dysregulation of autophagy is observed in the development of human diseases, especially cancer. Autophagy has reciprocal functions in cancer and may be responsible for either survival or death. Hepatocellular carcinoma (HCC) is one of the most lethal and common malignancies of the liver, and smoking, infection, and alcohol consumption can lead to its development. Genetic mutations and alterations in molecular processes can exacerbate the progression of HCC. The function of autophagy in HCC is controversial and may be both tumor suppressive and tumor promoting. Activation of autophagy may affect apoptosis in HCC and is a regulator of proliferation and glucose metabolism. Induction of autophagy may promote tumor metastasis via induction of EMT. In addition, autophagy is a regulator of stem cell formation in HCC, and pro-survival autophagy leads to cancer cell resistance to chemotherapy and radiotherapy. Targeting autophagy impairs growth and metastasis in HCC and improves tumor cell response to therapy. Of note, a large number of signaling pathways such as STAT3, Wnt, miRNAs, lncRNAs, and circRNAs regulate autophagy in HCC. Moreover, regulation of autophagy (induction or inhibition) by antitumor agents could be suggested for effective treatment of HCC. In this paper, we comprehensively review the role and mechanisms of autophagy in HCC and discuss the potential benefit of targeting this process in the treatment of the cancer. Video Abstract.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Niloufar Nadafzadeh
- Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mohammad Hassan Imani
- Department of Clinical Science, Faculty of Veterinary Medicine, Shahr-E Kord Branch, Islamic Azad University, Tehran, Chaharmahal and Bakhtiari, Iran
| | - Romina Rajabi
- Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Setayesh Ziaolhagh
- Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | | | - Raheleh Norouzi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Reihaneh Rafiei
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Zeinab Khazaei Koohpar
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Tonekabon Branch, Islamic Azad University, Tonekabon, Iran
| | - Behnaz Raei
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mohammad Arad Zandieh
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
- Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, Australia
- AFNP Med Austria, Vienna, Austria
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany.
| | - Shing Cheng Tan
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
5
|
Li D, Jia S, Wang S, Hu L. Glycoproteomic Analysis of Urinary Extracellular Vesicles for Biomarkers of Hepatocellular Carcinoma. Molecules 2023; 28:molecules28031293. [PMID: 36770959 PMCID: PMC9919939 DOI: 10.3390/molecules28031293] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/21/2023] [Accepted: 01/24/2023] [Indexed: 02/03/2023] Open
Abstract
Hepatocellular carcinoma (HCC) accounts for the most common form of primary liver cancer cases and constitutes a major health problem worldwide. The diagnosis of HCC is still challenging due to the low sensitivity and specificity of the serum α-fetoprotein (AFP) diagnostic method. Extracellular vesicles (EVs) are heterogeneous populations of phospholipid bilayer-enclosed vesicles that can be found in many biological fluids, and have great potential as circulating biomarkers for biomarker discovery and disease diagnosis. Protein glycosylation plays crucial roles in many biological processes and aberrant glycosylation is a hallmark of cancer. Herein, we performed a comprehensive glycoproteomic profiling of urinary EVs at the intact N-glycopeptide level to screen potential biomarkers for the diagnosis of HCC. With the control of the spectrum-level false discovery rate ≤1%, 756 intact N-glycopeptides with 154 N-glycosites, 158 peptide backbones, and 107 N-glycoproteins were identified. Out of 756 intact N-glycopeptides, 344 differentially expressed intact N-glycopeptides (DEGPs) were identified, corresponding to 308 upregulated and 36 downregulated N-glycopeptides, respectively. Compared to normal control (NC), the glycoproteins LG3BP, PIGR and KNG1 are upregulated in HCC-derived EVs, while ASPP2 is downregulated. The findings demonstrated that specific site-specific glycoforms in these glycoproteins from urinary EVs could be potential and efficient non-invasive candidate biomarkers for HCC diagnosis.
Collapse
Affiliation(s)
- Dejun Li
- Center for Supramolecular Chemical Biology, School of Life Sciences, Jilin University, Changchun 130012, China
- Prenatal Diagnosis Center, Reproductive Medicine Center, The First Hospital of Jilin University, Changchun 130021, China
| | - Shengnan Jia
- Department of Hepatopancreatobiliary Medicine, The Second Hospital, Jilin University, Changchun 130041, China
- Correspondence: (S.J.); (L.H.)
| | - Shuyue Wang
- Center for Supramolecular Chemical Biology, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Lianghai Hu
- Center for Supramolecular Chemical Biology, School of Life Sciences, Jilin University, Changchun 130012, China
- Correspondence: (S.J.); (L.H.)
| |
Collapse
|
6
|
Yang Y, Zhang Y, Yang J, Zhang M, Tian T, Jiang Y, Liu X, Xue G, Li X, Zhang X, Li S, Huang X, Li Z, Guo Y, Zhao L, Bao H, Zhou Z, Song J, Yang G, Xuan L, Shan H, Zhang Z, Lu Y, Yang B, Pan Z. Interdependent Nuclear Co-Trafficking of ASPP1 and p53 Aggravates Cardiac Ischemia/Reperfusion Injury. Circ Res 2023; 132:208-222. [PMID: 36656967 PMCID: PMC9855749 DOI: 10.1161/circresaha.122.321153] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE ASPP1 (apoptosis stimulating of p53 protein 1) is critical in regulating cell apoptosis as a cofactor of p53 to promote its transcriptional activity in the nucleus. However, whether cytoplasmic ASPP1 affects p53 nuclear trafficking and its role in cardiac diseases remains unknown. This study aims to explore the mechanism by which ASPP1 modulates p53 nuclear trafficking and the subsequent contribution to cardiac ischemia/reperfusion (I/R) injury. METHODS AND RESULTS The immunofluorescent staining showed that under normal condition ASPP1 and p53 colocalized in the cytoplasm of neonatal mouse ventricular cardiomyocytes, while they were both upregulated and translocated to the nuclei upon hypoxia/reoxygenation treatment. The nuclear translocation of ASPP1 and p53 was interdependent, as knockdown of either ASPP1 or p53 attenuated nuclear translocation of the other one. Inhibition of importin-β1 resulted in the cytoplasmic sequestration of both p53 and ASPP1 in neonatal mouse ventricular cardiomyocytes with hypoxia/reoxygenation stimulation. Overexpression of ASPP1 potentiated, whereas knockdown of ASPP1 inhibited the expression of Bax (Bcl2-associated X), PUMA (p53 upregulated modulator of apoptosis), and Noxa, direct apoptosis-associated targets of p53. ASPP1 was also increased in the I/R myocardium. Cardiomyocyte-specific transgenic overexpression of ASPP1 aggravated I/R injury as indicated by increased infarct size and impaired cardiac function. Conversely, knockout of ASPP1 mitigated cardiac I/R injury. The same qualitative data were observed in neonatal mouse ventricular cardiomyocytes exposed to hypoxia/reoxygenation injury. Furthermore, inhibition of p53 significantly blunted the proapoptotic activity and detrimental effects of ASPP1 both in vitro and in vivo. CONCLUSIONS Binding of ASPP1 to p53 triggers their nuclear cotranslocation via importin-β1 that eventually exacerbates cardiac I/R injury. The findings imply that interfering the expression of ASPP1 or the interaction between ASPP1 and p53 to block their nuclear trafficking represents an important therapeutic strategy for cardiac I/R injury.
Collapse
Affiliation(s)
- Ying Yang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Heilongjiang, China (Y.Y., Y.Z., J.Y., M.Z., T.T., Y.J., X.L., G.X., X.L., X.Z., S.L., X.H., Z.L., Y.G., L.Z., H.B., Z. Zhou, J.S., G.Y., L.X., H.S., Y.L., B.Y., Z.P.).,Department of Cardiology, Xiamen Key Laboratory of Cardiac Electrophysiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, China (Y.Y.)
| | - Yang Zhang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Heilongjiang, China (Y.Y., Y.Z., J.Y., M.Z., T.T., Y.J., X.L., G.X., X.L., X.Z., S.L., X.H., Z.L., Y.G., L.Z., H.B., Z. Zhou, J.S., G.Y., L.X., H.S., Y.L., B.Y., Z.P.)
| | - Jiqin Yang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Heilongjiang, China (Y.Y., Y.Z., J.Y., M.Z., T.T., Y.J., X.L., G.X., X.L., X.Z., S.L., X.H., Z.L., Y.G., L.Z., H.B., Z. Zhou, J.S., G.Y., L.X., H.S., Y.L., B.Y., Z.P.)
| | - Manman Zhang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Heilongjiang, China (Y.Y., Y.Z., J.Y., M.Z., T.T., Y.J., X.L., G.X., X.L., X.Z., S.L., X.H., Z.L., Y.G., L.Z., H.B., Z. Zhou, J.S., G.Y., L.X., H.S., Y.L., B.Y., Z.P.)
| | - Tao Tian
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Heilongjiang, China (Y.Y., Y.Z., J.Y., M.Z., T.T., Y.J., X.L., G.X., X.L., X.Z., S.L., X.H., Z.L., Y.G., L.Z., H.B., Z. Zhou, J.S., G.Y., L.X., H.S., Y.L., B.Y., Z.P.)
| | - Yuan Jiang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Heilongjiang, China (Y.Y., Y.Z., J.Y., M.Z., T.T., Y.J., X.L., G.X., X.L., X.Z., S.L., X.H., Z.L., Y.G., L.Z., H.B., Z. Zhou, J.S., G.Y., L.X., H.S., Y.L., B.Y., Z.P.).,Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China (Y.J.)
| | - Xuening Liu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Heilongjiang, China (Y.Y., Y.Z., J.Y., M.Z., T.T., Y.J., X.L., G.X., X.L., X.Z., S.L., X.H., Z.L., Y.G., L.Z., H.B., Z. Zhou, J.S., G.Y., L.X., H.S., Y.L., B.Y., Z.P.)
| | - Genlong Xue
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Heilongjiang, China (Y.Y., Y.Z., J.Y., M.Z., T.T., Y.J., X.L., G.X., X.L., X.Z., S.L., X.H., Z.L., Y.G., L.Z., H.B., Z. Zhou, J.S., G.Y., L.X., H.S., Y.L., B.Y., Z.P.)
| | - Xingda Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Heilongjiang, China (Y.Y., Y.Z., J.Y., M.Z., T.T., Y.J., X.L., G.X., X.L., X.Z., S.L., X.H., Z.L., Y.G., L.Z., H.B., Z. Zhou, J.S., G.Y., L.X., H.S., Y.L., B.Y., Z.P.)
| | - Xiaofang Zhang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Heilongjiang, China (Y.Y., Y.Z., J.Y., M.Z., T.T., Y.J., X.L., G.X., X.L., X.Z., S.L., X.H., Z.L., Y.G., L.Z., H.B., Z. Zhou, J.S., G.Y., L.X., H.S., Y.L., B.Y., Z.P.)
| | - Shangxuan Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Heilongjiang, China (Y.Y., Y.Z., J.Y., M.Z., T.T., Y.J., X.L., G.X., X.L., X.Z., S.L., X.H., Z.L., Y.G., L.Z., H.B., Z. Zhou, J.S., G.Y., L.X., H.S., Y.L., B.Y., Z.P.)
| | - Xiang Huang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Heilongjiang, China (Y.Y., Y.Z., J.Y., M.Z., T.T., Y.J., X.L., G.X., X.L., X.Z., S.L., X.H., Z.L., Y.G., L.Z., H.B., Z. Zhou, J.S., G.Y., L.X., H.S., Y.L., B.Y., Z.P.)
| | - Zheng Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Heilongjiang, China (Y.Y., Y.Z., J.Y., M.Z., T.T., Y.J., X.L., G.X., X.L., X.Z., S.L., X.H., Z.L., Y.G., L.Z., H.B., Z. Zhou, J.S., G.Y., L.X., H.S., Y.L., B.Y., Z.P.)
| | - Yang Guo
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Heilongjiang, China (Y.Y., Y.Z., J.Y., M.Z., T.T., Y.J., X.L., G.X., X.L., X.Z., S.L., X.H., Z.L., Y.G., L.Z., H.B., Z. Zhou, J.S., G.Y., L.X., H.S., Y.L., B.Y., Z.P.)
| | - Lexin Zhao
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Heilongjiang, China (Y.Y., Y.Z., J.Y., M.Z., T.T., Y.J., X.L., G.X., X.L., X.Z., S.L., X.H., Z.L., Y.G., L.Z., H.B., Z. Zhou, J.S., G.Y., L.X., H.S., Y.L., B.Y., Z.P.)
| | - Hairong Bao
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Heilongjiang, China (Y.Y., Y.Z., J.Y., M.Z., T.T., Y.J., X.L., G.X., X.L., X.Z., S.L., X.H., Z.L., Y.G., L.Z., H.B., Z. Zhou, J.S., G.Y., L.X., H.S., Y.L., B.Y., Z.P.)
| | - Zhiwen Zhou
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Heilongjiang, China (Y.Y., Y.Z., J.Y., M.Z., T.T., Y.J., X.L., G.X., X.L., X.Z., S.L., X.H., Z.L., Y.G., L.Z., H.B., Z. Zhou, J.S., G.Y., L.X., H.S., Y.L., B.Y., Z.P.)
| | - Jiahui Song
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Heilongjiang, China (Y.Y., Y.Z., J.Y., M.Z., T.T., Y.J., X.L., G.X., X.L., X.Z., S.L., X.H., Z.L., Y.G., L.Z., H.B., Z. Zhou, J.S., G.Y., L.X., H.S., Y.L., B.Y., Z.P.)
| | - Guohui Yang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Heilongjiang, China (Y.Y., Y.Z., J.Y., M.Z., T.T., Y.J., X.L., G.X., X.L., X.Z., S.L., X.H., Z.L., Y.G., L.Z., H.B., Z. Zhou, J.S., G.Y., L.X., H.S., Y.L., B.Y., Z.P.)
| | - Lina Xuan
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Heilongjiang, China (Y.Y., Y.Z., J.Y., M.Z., T.T., Y.J., X.L., G.X., X.L., X.Z., S.L., X.H., Z.L., Y.G., L.Z., H.B., Z. Zhou, J.S., G.Y., L.X., H.S., Y.L., B.Y., Z.P.)
| | - Hongli Shan
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Heilongjiang, China (Y.Y., Y.Z., J.Y., M.Z., T.T., Y.J., X.L., G.X., X.L., X.Z., S.L., X.H., Z.L., Y.G., L.Z., H.B., Z. Zhou, J.S., G.Y., L.X., H.S., Y.L., B.Y., Z.P.).,Shanghai Frontiers Science Research Center for Druggability of Cardiovascular noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, China (H.S.)
| | - Zhiren Zhang
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China (Z. Zhang, Z.P.)
| | - Yanjie Lu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Heilongjiang, China (Y.Y., Y.Z., J.Y., M.Z., T.T., Y.J., X.L., G.X., X.L., X.Z., S.L., X.H., Z.L., Y.G., L.Z., H.B., Z. Zhou, J.S., G.Y., L.X., H.S., Y.L., B.Y., Z.P.)
| | - Baofeng Yang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Heilongjiang, China (Y.Y., Y.Z., J.Y., M.Z., T.T., Y.J., X.L., G.X., X.L., X.Z., S.L., X.H., Z.L., Y.G., L.Z., H.B., Z. Zhou, J.S., G.Y., L.X., H.S., Y.L., B.Y., Z.P.)
| | - Zhenwei Pan
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Heilongjiang, China (Y.Y., Y.Z., J.Y., M.Z., T.T., Y.J., X.L., G.X., X.L., X.Z., S.L., X.H., Z.L., Y.G., L.Z., H.B., Z. Zhou, J.S., G.Y., L.X., H.S., Y.L., B.Y., Z.P.).,Research Unit of Noninfectious Chronic Diseases in Frigid Zone, Chinese Academy of Medical Sciences, 2019 Research Unit 070, Harbin, Heilongjiang, China (Z.P.).,NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China (Z. Zhang, Z.P.)
| |
Collapse
|
7
|
Shimada H, Kohno T, Konno T, Okada T, Saito K, Shindo Y, Kikuchi S, Tsujiwaki M, Ogawa M, Matsuura M, Saito T, Kojima T. The Roles of Tricellular Tight Junction Protein Angulin-1/Lipolysis-Stimulated Lipoprotein Receptor (LSR) in Endometriosis and Endometrioid-Endometrial Carcinoma. Cancers (Basel) 2021; 13:6341. [PMID: 34944960 PMCID: PMC8699113 DOI: 10.3390/cancers13246341] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 12/04/2021] [Accepted: 12/05/2021] [Indexed: 12/11/2022] Open
Abstract
Tight junction proteins play roles beyond permeability barriers functions and control cell proliferation and differentiation. The relation between tight junctions and the signal transduction pathways affects cell growth, invasion and migration. Abnormality of tight junction proteins closely contributes to epithelial mesenchymal transition (EMT) and malignancy of various cancers. Angulin-1/lipolysis-stimulated lipoprotein receptor (LSR) forms tricellular contacts that has a barrier function. Downregulation of angulin-1/LSR correlates with the malignancy in various cancers, including endometrioid-endometrial carcinoma (EEC). These alterations have been shown to link to not only multiple signaling pathways such as Hippo/YAP, HDAC, AMPK, but also cell metabolism in ECC cell line Sawano. Moreover, loss of angulin-1/LSR upregulates claudin-1, and loss of apoptosis stimulating p53 protein 2 (ASPP2) downregulates angulin-1/LSR. Angulin-1/LSR and ASPP2 concentrate at both midbody and centrosome in cytokinesis. In EEC tissues, angulin-1/LSR and ASPP2 are reduced and claudin-2 is overexpressed during malignancy, while in the tissues of endometriosis changes in localization of angulin-1/LSR and claudin-2 are seen. This review highlights how downregulation of angulin-1/LSR promotes development of endometriosis and EEC and discusses about the roles of angulin-1/LSR and its related proteins, including claudins and ASPP2.
Collapse
Affiliation(s)
- Hiroshi Shimada
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; (H.S.); (T.K.); (T.O.); (K.S.); (Y.S.)
- Departments of Obstetrics, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; (M.O.); (M.M.); (T.S.)
| | - Takayuki Kohno
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; (H.S.); (T.K.); (T.O.); (K.S.); (Y.S.)
| | - Takumi Konno
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; (H.S.); (T.K.); (T.O.); (K.S.); (Y.S.)
| | - Tadahi Okada
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; (H.S.); (T.K.); (T.O.); (K.S.); (Y.S.)
- Departments of Obstetrics, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; (M.O.); (M.M.); (T.S.)
| | - Kimihito Saito
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; (H.S.); (T.K.); (T.O.); (K.S.); (Y.S.)
- Departments of Obstetrics, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; (M.O.); (M.M.); (T.S.)
| | - Yuma Shindo
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; (H.S.); (T.K.); (T.O.); (K.S.); (Y.S.)
| | - Shin Kikuchi
- Department of Anatomy, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan;
| | - Mitsuhiro Tsujiwaki
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan;
| | - Marie Ogawa
- Departments of Obstetrics, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; (M.O.); (M.M.); (T.S.)
| | - Motoki Matsuura
- Departments of Obstetrics, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; (M.O.); (M.M.); (T.S.)
| | - Tsuyoshi Saito
- Departments of Obstetrics, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; (M.O.); (M.M.); (T.S.)
| | - Takashi Kojima
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; (H.S.); (T.K.); (T.O.); (K.S.); (Y.S.)
| |
Collapse
|
8
|
Zhang D, Guo S, Schrodi SJ. Mechanisms of DNA Methylation in Virus-Host Interaction in Hepatitis B Infection: Pathogenesis and Oncogenetic Properties. Int J Mol Sci 2021; 22:9858. [PMID: 34576022 PMCID: PMC8466338 DOI: 10.3390/ijms22189858] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 09/10/2021] [Accepted: 09/10/2021] [Indexed: 12/11/2022] Open
Abstract
Hepatitis B virus (HBV), the well-studied oncovirus that contributes to the majority of hepatocellular carcinomas (HCC) worldwide, can cause a severe inflammatory microenvironment leading to genetic and epigenetic changes in hepatocyte clones. HBV replication contributes to the regulation of DNA methyltransferase gene expression, particularly by X protein (HBx), and subsequent methylation changes may lead to abnormal transcription activation of adjacent genes and genomic instability. Undoubtedly, the altered expression of these genes has been known to cause diverse aspects of infected hepatocytes, including apoptosis, proliferation, reactive oxygen species (ROS) accumulation, and immune responses. Additionally, pollutant-induced DNA methylation changes and aberrant methylation of imprinted genes in hepatocytes also complicate the process of tumorigenesis. Meanwhile, hepatocytes also contribute to epigenetic modification of the viral genome to affect HBV replication or viral protein production. Meanwhile, methylation levels of HBV integrants and surrounding host regions also play crucial roles in their ability to produce viral proteins in affected hepatocytes. Both host and viral changes can provide novel insights into tumorigenesis, individualized responses to therapeutic intervention, disease progress, and early diagnosis. As such, DNA methylation-mediated epigenetic silencing of cancer-related genes and viral replication is a compelling therapeutic goal to reduce morbidity and mortality from liver cancer caused by chronic HBV infection. In this review, we summarize the most recent research on aberrant DNA methylation associated with HBV infection, which is involved in HCC development, and provide an outlook on the future direction of the research.
Collapse
Affiliation(s)
- Dake Zhang
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Shicheng Guo
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA;
| | - Steven J. Schrodi
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA;
- Computation and Informatics in Biology and Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
9
|
Bousali M, Papatheodoridis G, Paraskevis D, Karamitros T. Hepatitis B Virus DNA Integration, Chronic Infections and Hepatocellular Carcinoma. Microorganisms 2021; 9:1787. [PMID: 34442866 PMCID: PMC8398950 DOI: 10.3390/microorganisms9081787] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/13/2021] [Accepted: 08/18/2021] [Indexed: 12/16/2022] Open
Abstract
Hepatitis B Virus (HBV) is an Old World virus with a high mutation rate, which puts its origins in Africa alongside the origins of Homo sapiens, and is a member of the Hepadnaviridae family that is characterized by a unique viral replication cycle. It targets human hepatocytes and can lead to chronic HBV infection either after acute infection via horizontal transmission usually during infancy or childhood or via maternal-fetal transmission. HBV has been found in ~85% of HBV-related Hepatocellular Carcinomas (HCC), and it can integrate the whole or part of its genome into the host genomic DNA. The molecular mechanisms involved in the HBV DNA integration is not yet clear; thus, multiple models have been described with respect to either the relaxed-circular DNA (rcDNA) or the double-stranded linear DNA (dslDNA) of HBV. Various genes have been found to be affected by HBV DNA integration, including cell-proliferation-related genes, oncogenes and long non-coding RNA genes (lincRNAs). The present review summarizes the advances in the research of HBV DNA integration, focusing on the evolutionary and molecular side of the integration events along with the arising clinical aspects in the light of WHO's commitment to eliminate HBV and viral hepatitis by 2030.
Collapse
Affiliation(s)
- Maria Bousali
- Bioinformatics and Applied Genomics Unit, Department of Microbiology, Hellenic Pasteur Institute, 11521 Athens, Greece;
| | - George Papatheodoridis
- Department of Gastroenterology, “Laiko” General Hospital of Athens, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Dimitrios Paraskevis
- Department of Hygiene Epidemiology and Medical Statistics, School of Medicine, National and Kapodistrian University of Athens, 15772 Athens, Greece;
| | - Timokratis Karamitros
- Bioinformatics and Applied Genomics Unit, Department of Microbiology, Hellenic Pasteur Institute, 11521 Athens, Greece;
- Laboratory of Medical Microbiology, Department of Microbiology, Hellenic Pasteur Institute, 11521 Athens, Greece
| |
Collapse
|
10
|
Pietropaolo V, Prezioso C, Moens U. Role of Virus-Induced Host Cell Epigenetic Changes in Cancer. Int J Mol Sci 2021; 22:ijms22158346. [PMID: 34361112 PMCID: PMC8346956 DOI: 10.3390/ijms22158346] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 07/30/2021] [Accepted: 08/02/2021] [Indexed: 12/12/2022] Open
Abstract
The tumor viruses human T-lymphotropic virus 1 (HTLV-1), hepatitis C virus (HCV), Merkel cell polyomavirus (MCPyV), high-risk human papillomaviruses (HR-HPVs), Epstein-Barr virus (EBV), Kaposi’s sarcoma-associated herpes virus (KSHV) and hepatitis B virus (HBV) account for approximately 15% of all human cancers. Although the oncoproteins of these tumor viruses display no sequence similarity to one another, they use the same mechanisms to convey cancer hallmarks on the infected cell. Perturbed gene expression is one of the underlying mechanisms to induce cancer hallmarks. Epigenetic processes, including DNA methylation, histone modification and chromatin remodeling, microRNA, long noncoding RNA, and circular RNA affect gene expression without introducing changes in the DNA sequence. Increasing evidence demonstrates that oncoviruses cause epigenetic modifications, which play a pivotal role in carcinogenesis. In this review, recent advances in the role of host cell epigenetic changes in virus-induced cancers are summarized.
Collapse
Affiliation(s)
- Valeria Pietropaolo
- Department of Public Health and Infectious Diseases, “Sapienza” University, 00185 Rome, Italy;
- Correspondence: (V.P.); (U.M.)
| | - Carla Prezioso
- Department of Public Health and Infectious Diseases, “Sapienza” University, 00185 Rome, Italy;
- IRCSS San Raffaele Roma, Microbiology of Chronic Neuro-Degenerative Pathologies, 00161 Rome, Italy
| | - Ugo Moens
- Molecular Inflammation Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø—The Arctic University of Norway, 9037 Tromsø, Norway
- Correspondence: (V.P.); (U.M.)
| |
Collapse
|
11
|
Wang S, Sun Y, Wang Y, Wang A, Kou B, Che Y, Chen D, Zhang Y, Shi Y. ASPP2 inhibits hepatitis B virus replication by preventing nucleus translocation of HSF1 and attenuating the transactivation of ATG7. J Cell Mol Med 2021; 25:6899-6908. [PMID: 34085409 PMCID: PMC8278078 DOI: 10.1111/jcmm.16699] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 04/22/2021] [Accepted: 05/12/2021] [Indexed: 12/13/2022] Open
Abstract
Hepatitis B virus (HBV) is a kind of virus with the capability to induce autophagy, thereby facilitating its replication. Reducing hepatocyte autophagy is proved to be a useful way to inhibit HBV replication. Herein, we reported that p53-binding protein 2 (apoptosis-stimulating protein of p53-2, ASPP2) could attenuate HBV-induced hepatocyte autophagy in a p53-independent manner. Mechanistically, overexpressed ASPP2 binds to HSF1 in cytoplasm of HBV-infected cells, which prevents the translocation of HSF1 to nuclei, thereby inhibiting the transactivation of Atg7. By regulating the transcription of Atg7, ASPP2 reduces hepatocyte autophagy, thereby inhibiting HBV replication. Therefore, ASPP2 is a key regulator of cell autophagy, and overexpression of ASPP2 could be a novel method to inhibit HBV replication in hepatocytes.
Collapse
Affiliation(s)
- Shanshan Wang
- Capital Medical University Affiliated Beijing You’an HospitalBeijingChina
- Beijing Institute of HepatologyBeijingChina
| | - Yu Sun
- Minimally Invasive Interventional Centre of OncologyBeijing You’an HospitalCapital Medical UniversityBeijingChina
| | - Yang Wang
- Capital Medical University Affiliated Beijing You’an HospitalBeijingChina
- Beijing Institute of HepatologyBeijingChina
| | - Anna Wang
- Capital Medical University Affiliated Beijing You’an HospitalBeijingChina
| | - Buxin Kou
- Capital Medical University Affiliated Beijing You’an HospitalBeijingChina
- Beijing Institute of HepatologyBeijingChina
| | - Yang Che
- Capital Medical University Affiliated Beijing You’an HospitalBeijingChina
- Beijing Institute of HepatologyBeijingChina
| | - Dexi Chen
- Capital Medical University Affiliated Beijing You’an HospitalBeijingChina
- Beijing Institute of HepatologyBeijingChina
| | - Yulin Zhang
- Capital Medical University Affiliated Beijing You’an HospitalBeijingChina
| | - Ying Shi
- Capital Medical University Affiliated Beijing You’an HospitalBeijingChina
- Beijing Institute of HepatologyBeijingChina
| |
Collapse
|
12
|
Lu X, Li J, Lou H, Cao Z, Fan X. Genome-Wide DNA Methylation Alterations and Potential Risk Induced by Subacute and Subchronic Exposure to Food-Grade Nanosilica in Mice. ACS NANO 2021; 15:8225-8243. [PMID: 33938728 DOI: 10.1021/acsnano.0c07323] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The intensive application of nanomaterials in the food industry has raised concerns about their potential risks to human health. However, limited data are available on the biological safety of nanomaterials in food, especially at the epigenetic level. This study examined the implications of two types of synthetic amorphous silica (SAS), food-grade precipitated silica (S200) and fumed silica Aerosil 200F (A200F), which are nanorange food additives. After 28-day continuous and intermittent subacute exposure to these SAS via diet, whole-genome methylation levels in mouse peripheral leukocytes and liver were significantly altered in a dose- and SAS type-dependent manner, with minimal toxicity detected by conventional toxicological assessments, especially at a human-relevant dose (HRD). The 84-day continuous subchronic exposure to all doses of S200 and A200F induced liver steatosis where S200 accumulated in the liver even at HRD. Genome-wide DNA methylation sequencing revealed that the differentially methylated regions induced by both SAS were mainly located in the intron, intergenic, and promoter regions after 84-day high-dose continuous exposure. Bioinformatics analysis of differentially methylated genes indicated that exposure to S200 or A200F may lead to lipid metabolism disorders and cancer development. Pathway validation experiments indicated both SAS types as potentially carcinogenic. While S200 inhibited the p53-mediated apoptotic pathway in mouse liver, A200F activated the HRAS-mediated MAPK signaling pathway, which is a key driver of hepatocarcinogenesis. Thus, caution must be paid to the risk of long-term exposure to food-grade SAS, and epigenetic parameters should be included as end points during the risk assessment of food-grade nanomaterials.
Collapse
Affiliation(s)
- Xiaoyan Lu
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Junying Li
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - He Lou
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zeya Cao
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xiaohui Fan
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Center in Zhejiang University, State Key Laboratory of Component-Based Chinese Medicine, Hangzhou 310058, China
| |
Collapse
|
13
|
Jiang Y, Han Q, Zhao H, Zhang J. The Mechanisms of HBV-Induced Hepatocellular Carcinoma. J Hepatocell Carcinoma 2021; 8:435-450. [PMID: 34046368 PMCID: PMC8147889 DOI: 10.2147/jhc.s307962] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 05/06/2021] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a common malignancy, and the hepatitis B virus (HBV) is its major pathogenic factor. Over the past decades, it has been confirmed that HBV infection could promote disease progression through a variety of mechanisms, ultimately leading to the malignant transformation of liver cells. Many factors have been identified in the pathogenesis of HBV-associated HCC (HBV-HCC), including HBV gene integration, genomic instability caused by mutation, and activation of cancer-promoting signaling pathways. As research in the progression of HBV-HCC progresses, the role of many new mechanisms, such as epigenetics, exosomes, autophagy, metabolic regulation, and immune suppression, is also being continuously explored. The occurrence of HBV-HCC is a complex process caused by interactions across multiple genes and multiple steps, where the synergistic effects of various cancer-promoting mechanisms accelerate the process of disease evolution from inflammation to tumorigenesis. In this review, we aim to provide a brief overview of the mechanisms involved in the occurrence and development of HBV-HCC, which may contribute to a better understanding of the role of HBV in the occurrence and development of HCC.
Collapse
Affiliation(s)
- Yu Jiang
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, Shandong Province, People's Republic of China
| | - Qiuju Han
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, Shandong Province, People's Republic of China
| | - Huajun Zhao
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, Shandong Province, People's Republic of China
| | - Jian Zhang
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, Shandong Province, People's Republic of China
| |
Collapse
|
14
|
Sekiba K, Otsuka M, Koike K. Potential of HBx Gene for Hepatocarcinogenesis in Noncirrhotic Liver. Semin Liver Dis 2021; 41:142-149. [PMID: 33984871 DOI: 10.1055/s-0041-1723033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Current treatments for hepatitis B virus (HBV) using nucleos(t)ide analogs cannot eliminate the risk of hepatocellular carcinoma (HCC) development. As HBV-associated HCC can develop even in the absence of liver cirrhosis, HBV is regarded to possess direct oncogenic potential. HBV regulatory protein X (HBx) has been identified as a primary mediator of HBV-mediated hepatocarcinogenesis. A fragment of the HBV genome that contains the coding region of HBx is commonly integrated into the host genome, resulting in the production of aberrant proteins and subsequent hepatocarcinogenesis. Besides, HBx interferes with the host DNA or deoxyribonucleic acid damage repair pathways, signal transduction, epigenetic regulation of gene expression, and cancer immunity, thereby promoting carcinogenesis in the noncirrhotic liver. However, numerous molecules and pathways have been implicated in the development of HBx-associated HCC, suggesting that the mechanisms underlying HBx-mediated hepatocarcinogenesis remain to be elucidated.
Collapse
Affiliation(s)
- Kazuma Sekiba
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan
| | - Motoyuki Otsuka
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kazuhiko Koike
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
15
|
Zeisel MB, Guerrieri F, Levrero M. Host Epigenetic Alterations and Hepatitis B Virus-Associated Hepatocellular Carcinoma. J Clin Med 2021; 10:jcm10081715. [PMID: 33923385 PMCID: PMC8071488 DOI: 10.3390/jcm10081715] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/06/2021] [Accepted: 04/12/2021] [Indexed: 12/16/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most frequent primary malignancy of the liver and a leading cause of cancer-related deaths worldwide. Although much progress has been made in HCC drug development in recent years, treatment options remain limited. The major cause of HCC is chronic hepatitis B virus (HBV) infection. Despite the existence of a vaccine, more than 250 million individuals are chronically infected by HBV. Current antiviral therapies can repress viral replication but to date there is no cure for chronic hepatitis B. Of note, inhibition of viral replication reduces but does not eliminate the risk of HCC development. HBV contributes to liver carcinogenesis by direct and indirect effects. This review summarizes the current knowledge of HBV-induced host epigenetic alterations and their association with HCC, with an emphasis on the interactions between HBV proteins and the host cell epigenetic machinery leading to modulation of gene expression.
Collapse
Affiliation(s)
- Mirjam B. Zeisel
- Cancer Research Center of Lyon (CRCL), UMR Inserm 1052 CNRS 5286 Mixte CLB, Université de Lyon 1 (UCBL1), 69003 Lyon, France;
- Correspondence: (M.B.Z.); (M.L.)
| | - Francesca Guerrieri
- Cancer Research Center of Lyon (CRCL), UMR Inserm 1052 CNRS 5286 Mixte CLB, Université de Lyon 1 (UCBL1), 69003 Lyon, France;
| | - Massimo Levrero
- Cancer Research Center of Lyon (CRCL), UMR Inserm 1052 CNRS 5286 Mixte CLB, Université de Lyon 1 (UCBL1), 69003 Lyon, France;
- Hospices Civils de Lyon, Hôpital Croix Rousse, Service d’Hépato-Gastroentérologie, 69004 Lyon, France
- Correspondence: (M.B.Z.); (M.L.)
| |
Collapse
|
16
|
Abstract
Hepatitis B virus (HBV) is a major cause of hepatocellular carcinoma (HCC). There are approximately 250 million people in the world that are chronically infected by this virus, resulting in nearly 1 million deaths every year. Many of these patients die from severe liver diseases, including HCC. HBV may induce HCC through the induction of chronic liver inflammation, which can cause oxidative stress and DNA damage. However, many studies also indicated that HBV could induce HCC via the alteration of hepatocellular physiology that may involve genetic and epigenetic changes of the host DNA, the alteration of cellular signaling pathways, and the inhibition of DNA repair mechanisms. This alteration of cellular physiology can lead to the accumulation of DNA damages and the promotion of cell cycles and predispose hepatocytes to oncogenic transformation.
Collapse
Affiliation(s)
- Jiyoung Lee
- Department of Molecular Microbiology and Immunology, University of Southern California Keck School of Medicine, 2011 Zonal Avenue, HMR-401, Los Angeles, CA, 90033, USA
| | - Kuen-Nan Tsai
- Department of Molecular Microbiology and Immunology, University of Southern California Keck School of Medicine, 2011 Zonal Avenue, HMR-401, Los Angeles, CA, 90033, USA
| | - Jing-Hsiung James Ou
- Department of Molecular Microbiology and Immunology, University of Southern California Keck School of Medicine, 2011 Zonal Avenue, HMR-401, Los Angeles, CA, 90033, USA.
| |
Collapse
|
17
|
Patel KD, Barasiya YV, Patel JB, Patel PS. Apoptosis stimulating protein of p53 (ASPP) 1 and ASPP2 m-RNA expression in oral cancer. Arch Oral Biol 2020; 119:104920. [PMID: 32987288 DOI: 10.1016/j.archoralbio.2020.104920] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 09/09/2020] [Accepted: 09/09/2020] [Indexed: 12/25/2022]
Abstract
OBJECTIVE The present study was carried out to unfold the clinical significance of apoptosis stimulating protein of p53 (ASPP) 1 and ASPP2 expression in oral cancer (OC). METHODS Tissue specimens (malignant and their corresponding adjacent normal) from 40 pathologically confirmed OC patients treated at the Institute were included in the study. ASPP1 and ASPP2 expression were examined using semi-quantitative RT-PCR. RESULTS The results indicated lower ASPP1 expression in OC tissues as compared to adjacent normal tissues (p = 0.085). Stratified analysis as per tumor site revealed significant down-regulation of ASPP1 in tongue cancer tissues (p = 0.005). Receiver operating characteristic curve depicted significant discriminatory efficacy in distinguishing tongue cancer tissues and adjacent normal tissues (p = 0.019). Moreover, ASPP1 expression was remarkably declined in stage II, III and IV OC tumors than stage I OC tumors (p = 0.007, 0.092 and 0.013, respectively). A similar trend was observed in buccal mucosa tumors on further analysis. ASPP2 expression was lower in moderately differentiated OC tumors as compared to well differentiated OC tumors (p = 0.061). Significantly reduced ASPP2 expression was observed in tongue cancer tumors without invasion in contrast to tumors with perineural invasion (p = 0.007). Besides, ASPP1 and ASPP2 expression was positively inter-correlated in tongue tissues (r = 0.325, p = 0.091). CONCLUSIONS Lower ASPP1 expression in tongue cancer during malignant transformation has significance in cancer initiation. Association of reduced ASPP1 and ASPP2 expression with advanced disease stage and moderate differentiation suggests their role in OC progression. Thus, down-regulation of ASPP1 and ASPP2 may serve as potential diagnostic and prognostic indicators in OC.
Collapse
Affiliation(s)
- Kinjal D Patel
- The Gujarat Cancer & Research Institute, Civil Hospital Campus, Asarwa, Ahmedabad, 380 016, Gujarat, India
| | - Yesha V Barasiya
- The Gujarat Cancer & Research Institute, Civil Hospital Campus, Asarwa, Ahmedabad, 380 016, Gujarat, India
| | - Jayendra B Patel
- The Gujarat Cancer & Research Institute, Civil Hospital Campus, Asarwa, Ahmedabad, 380 016, Gujarat, India
| | - Prabhudas S Patel
- The Gujarat Cancer & Research Institute, Civil Hospital Campus, Asarwa, Ahmedabad, 380 016, Gujarat, India.
| |
Collapse
|
18
|
Mir-30b-5p Promotes Proliferation, Migration, and Invasion of Breast Cancer Cells via Targeting ASPP2. BIOMED RESEARCH INTERNATIONAL 2020; 2020:7907269. [PMID: 32420372 PMCID: PMC7210518 DOI: 10.1155/2020/7907269] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 01/06/2020] [Accepted: 01/16/2020] [Indexed: 12/18/2022]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtypes of breast cancer, which has few effective targeted therapies. Various sources of evidence confirm that microRNAs (miRNAs) contribute to the progression and metastasis of human breast cancer. However, the molecular mechanisms underlying the changes in miRNAs expression and the regulation of miRNAs functions have not been well clarified. In this study, we found that the expression of miR-30b-5p was upregulated in breast cancer tissues and breast cancer cell lines, compared to paracancer tissues and normal breast cell lines. Moreover, induced overexpression of miR-30b-5p promoted the MDA-MB-231 and HCC 1937 cell growth, migration, and invasion and reduced the cellular apoptosis. Further studies confirmed that miR-30b-5p could directly target ASPP2 and then activate the AKT signaling pathway. Our results suggested that miR-30b-5p could act as a tumor promoter in TNBC. The newly identified miR-30b-5p/ASPP2/AKT axis represents a novel therapeutic strategy for treating TNBC.
Collapse
|
19
|
Liu D, Ertay A, Hill C, Zhou Y, Li J, Zou Y, Qiu H, Yuan X, Ewing RM, Lu X, Xiong H, Wang Y. ASPP1 deficiency promotes epithelial-mesenchymal transition, invasion and metastasis in colorectal cancer. Cell Death Dis 2020; 11:224. [PMID: 32269211 PMCID: PMC7142079 DOI: 10.1038/s41419-020-2415-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/17/2020] [Accepted: 03/18/2020] [Indexed: 01/06/2023]
Abstract
The apoptosis-stimulating protein of p53 (ASPP) family of proteins can regulate apoptosis by interacting with the p53 family and have been identified to play an important role in cancer progression. Previously, we have demonstrated that ASPP2 downregulation can promote invasion and migration by controlling β-catenin-dependent regulation of ZEB1, however, the role of ASPP1 in colorectal cancer (CRC) remains unclear. We analyzed data from The Cancer Genome Atlas (TCGA) and coupled this to in vitro experiments in CRC cell lines as well as to experimental pulmonary metastasis in vivo. Tissue microarrays of CRC patients with information of clinical-pathological parameters were also used to investigate the expression and function of ASPP1 in CRC. Here, we report that loss of ASPP1 is capable of enhancing migration and invasion in CRC, both in vivo and in vitro. We demonstrate that depletion of ASPP1 could activate expression of Snail2 via the NF-κB pathway and in turn, induce EMT; and this process is further exacerbated in RAS-mutated CRC. ASPP1 could be a prognostic factor in CRC, and the use of NF-κB inhibitors may provide new strategies for therapy against metastasis in ASPP1-depleted CRC patients.
Collapse
Affiliation(s)
- Dian Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Ayse Ertay
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Charlotte Hill
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Yilu Zhou
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
- Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Juanjuan Li
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Yanmei Zou
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Hong Qiu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Rob M Ewing
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
- Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Xin Lu
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7DQ, UK
| | - Hua Xiong
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China.
| | - Yihua Wang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China.
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK.
- Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK.
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, SO16 6YD, UK.
| |
Collapse
|
20
|
ASPP2 suppression promotes malignancy via LSR and YAP in human endometrial cancer. Histochem Cell Biol 2020; 154:197-213. [PMID: 32266459 DOI: 10.1007/s00418-020-01876-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/29/2020] [Indexed: 12/19/2022]
Abstract
Apoptosis-stimulating p53 protein 2 (ASPP2) is an apoptosis inducer that acts via binding with p53 and epithelial polarity molecule PAR3. Lipolysis-stimulated lipoprotein receptor (LSR) is an important molecule at tricellular contacts, and loss of LSR promotes cell migration and invasion via Yes-associated protein (YAP) in human endometrial cancer cells. In the present study, to find how ASPP2 suppression promotes malignancy in human endometrial cancer, we investigated its mechanisms including the relationship with LSR. In endometriosis and endometrial cancers (G1 and G2), ASPP2 was observed as well as PAR3 and LSR in the subapical region. ASPP2 decreased in G3 endometrial cancer compared to G1. In human endometrial cancer cell line Sawano, ASPP2 was colocalized with LSR and tricellulin at tricellular contacts and binding to PAR3, LSR, and tricellulin in the confluent state. ASPP2 suppression promoted cell migration and invasion, decreased LSR expression, and induced expression of phosphorylated YAP, claudin-1, -4, and -7 as effectively as the loss of LSR. Knockdown of YAP prevented the upregulation of pYAP, cell migration and invasion induced by the ASPP2 suppression. Treatment with a specific antibody against ASPP2 downregulated ASPP2 and LSR, affected F-actin at tricellular contacts, upregulated expression of pYAP and claudin-1, and induced cell migration and invasion via YAP. In normal human endometrial epithelial cells, ASPP2 was in part colocalized with LSR at tricellular contacts and knockdown of ASPP2 or LSR induced expression of claudin-1 and claudin-4. ASPP2 suppression promoted cell invasion and migration via LSR and YAP in human endometrial cancer cells.
Collapse
|
21
|
ASPP2 inhibits tumor growth by repressing the mevalonate pathway in hepatocellular carcinoma. Cell Death Dis 2019; 10:830. [PMID: 31685796 PMCID: PMC6828733 DOI: 10.1038/s41419-019-2054-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 09/18/2019] [Accepted: 10/01/2019] [Indexed: 12/13/2022]
Abstract
Cancer is, fundamentally, a disorder of cell growth and proliferation, which requires adequate supplies of energy and nutrients. In this study, we report that the haplo-insufficient tumor suppressor ASPP2, a p53 activator, negatively regulates the mevalonate pathway to mediate its inhibitory effect on tumor growth in hepatocellular carcinoma (HCC). Gene expression profile analysis revealed that the expression of key enzymes in the mevalonate pathway were increased when ASPP2 was downregulated. HCC cells gained higher cholesterol levels and enhanced tumor-initiating capability in response to the depletion of ASPP2. Simvastatin, a mevalonate pathway inhibitor, efficiently abrogated ASPP2 depletion-induced anchorage-independent cell proliferation, resistance to chemotherapy drugs in vitro, and tumor growth in xenografted nude mice. Mechanistically, ASPP2 interacts with SREBP-2 in the nucleus and restricts the transcriptional activity of SREBP-2 on its target genes, which include key enzymes involved in the mevalonate pathway. Moreover, clinical data revealed better prognosis in patients with high levels of ASPP2 and low levels of the mevalonate pathway enzyme HMGCR. Our findings provide functional and mechanistic insights into the critical role of ASPP2 in the regulation of the mevalonate pathway and the importance of this pathway in tumor initiation and tumor growth, which may provide a new therapeutic opportunity for HCC.
Collapse
|
22
|
CDH1 gene as a prognostic biomarker in HCV (genotype 4) induced hepatocellular carcinoma in the Egyptian patients. GENE REPORTS 2019. [DOI: 10.1016/j.genrep.2019.100452] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
23
|
Zhou Y, Millott R, Kim HJ, Peng S, Edwards RA, Skene-Arnold T, Hammel M, Lees-Miller SP, Tainer JA, Holmes CFB, Glover JNM. Flexible Tethering of ASPP Proteins Facilitates PP-1c Catalysis. Structure 2019; 27:1485-1496.e4. [PMID: 31402222 DOI: 10.1016/j.str.2019.07.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 06/14/2019] [Accepted: 07/22/2019] [Indexed: 12/15/2022]
Abstract
ASPP (apoptosis-stimulating proteins of p53) proteins bind PP-1c (protein phosphatase 1) and regulate p53 impacting cancer cell growth and apoptosis. Here we determine the crystal structure of the oncogenic ASPP protein, iASPP, bound to PP-1c. The structure reveals a 1:1 complex that relies on interactions of the iASPP SILK and RVxF motifs with PP-1c, plus interactions of the PP-1c PxxPxR motif with the iASPP SH3 domain. Small-angle X-ray scattering analyses suggest that the crystal structure undergoes slow interconversion with more extended conformations in solution. We show that iASPP, and the tumor suppressor ASPP2, enhance the catalytic activity of PP-1c against the small-molecule substrate, pNPP as well as p53. The combined results suggest that PxxPxR binding to iASPP SH3 domain is critical for complex formation, and that the modular ASPP-PP-1c interface provides dynamic flexibility that enables functional binding and dephosphorylation of p53 and other diverse protein substrates.
Collapse
Affiliation(s)
- Yeyun Zhou
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Robyn Millott
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Hyeong Jin Kim
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Shiyun Peng
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Ross A Edwards
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Tamara Skene-Arnold
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Michal Hammel
- Molecular Biophysics & Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Susan P Lees-Miller
- Department of Biochemistry and Molecular Biology, Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - John A Tainer
- Molecular Biophysics & Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA; Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Charles F B Holmes
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada.
| | - J N Mark Glover
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada.
| |
Collapse
|
24
|
Chauhan R, Shimizu Y, Watashi K, Wakita T, Fukasawa M, Michalak TI. Retrotransposon elements among initial sites of hepatitis B virus integration into human genome in the HepG2-NTCP cell infection model. Cancer Genet 2019; 235-236:39-56. [PMID: 31064734 DOI: 10.1016/j.cancergen.2019.04.060] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 04/11/2019] [Accepted: 04/18/2019] [Indexed: 02/07/2023]
Abstract
Integration of hepatitis B virus (HBV) DNA into host's genome is evident in all stages and models of HBV infection. Investigations of the initial virus-host junctions have been just recently initiated since their nature may promote liver oncogenesis immediately following infection. We examined the time-frame and host sites at which HBV integrates in HepG2 cells overexpressing sodium taurocholate co-transporting polypeptide (NTCP) receptor mediating HBV entry. HepG2-NTCP cells were analyzed from 15 min to 13 days post-infection (p.i.). The results showed that except for 15 min p.i., HBV-host integrations were detected at all time points thereafter. At 30 min p.i., virus junctions with retrotransposon SINE and with neuroblastoma breakpoint family member 1 gene were detected. At one-hour p.i., HBV integration with retrotransposon THE-1B-LTR was identified, while virus insertions into proline-rich protein and protein kinase cGMP-dependent type 1 encoding genes were found at 3 h p.i. Fusion with runt-related transcription factor 1 was detected at 24 h p.i. and merges with 9 different genes at 13 day p.i. The data showed that retrotransposon elements are frequent among first-hit sites of HBV insertion. This may suggest a mechanism by which HBV DNA may spread across host's genome from earliest stages of infection.
Collapse
Affiliation(s)
- Ranjit Chauhan
- Molecular Virology and Hepatology Research Group, Division of BioMedical Sciences, Faculty of Medicine, Health Science Centre, Memorial University, St. John's, NL, Canada
| | - Yoshimi Shimizu
- Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Koichi Watashi
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
| | - Takaji Wakita
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
| | - Masayoshi Fukasawa
- Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Tomasz I Michalak
- Molecular Virology and Hepatology Research Group, Division of BioMedical Sciences, Faculty of Medicine, Health Science Centre, Memorial University, St. John's, NL, Canada.
| |
Collapse
|
25
|
Schittenhelm MM, Walter B, Tsintari V, Federmann B, Bajrami Saipi M, Akmut F, Illing B, Mau-Holzmann U, Fend F, Lopez CD, Kampa-Schittenhelm KM. Alternative splicing of the tumor suppressor ASPP2 results in a stress-inducible, oncogenic isoform prevalent in acute leukemia. EBioMedicine 2019; 42:340-351. [PMID: 30952616 PMCID: PMC6491939 DOI: 10.1016/j.ebiom.2019.03.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 03/10/2019] [Accepted: 03/11/2019] [Indexed: 01/01/2023] Open
Abstract
Background Apoptosis-stimulating Protein of TP53-2 (ASPP2) is a tumor suppressor enhancing TP53-mediated apoptosis via binding to the TP53 core domain. TP53 mutations found in cancers disrupt ASPP2 binding, arguing for an important role of ASPP2 in TP53-mediated tumor suppression. We now identify an oncogenic splicing variant, ASPP2κ, with high prevalence in acute leukemia. Methods An mRNA screen to detect ASPP2 splicing variants was performed and ASPP2κ was validated using isoform-specific PCR approaches. Translation into a genuine protein isoform was evaluated after establishing epitope-specific antibodies. For functional studies cell models with forced expression of ASPP2κ or isoform-specific ASPP2κ-interference were created to evaluate proliferative, apoptotic and oncogenic characteristics of ASPP2κ. Findings Exon skipping generates a premature stop codon, leading to a truncated C-terminus, omitting the TP53-binding sites. ASPP2κ translates into a dominant-negative protein variant impairing TP53-dependent induction of apoptosis. ASPP2κ is expressed in CD34+ leukemic progenitor cells and functional studies argue for a role in early oncogenesis, resulting in perturbed proliferation and impaired induction of apoptosis, mitotic failure and chromosomal instability (CIN) – similar to TP53 mutations. Importantly, as expression of ASPP2κ is stress-inducible it defines a novel class of dynamic oncogenes not represented by genomic mutations. Interpretation Our data demonstrates that ASPP2κ plays a distinctive role as an antiapoptotic regulator of the TP53 checkpoint, rendering cells to a more aggressive phenotype as evidenced by proliferation and apoptosis rates – and ASPP2κ expression results in acquisition of genomic mutations, a first initiating step in leukemogenesis. We provide proof-of-concept to establish ASPP2κ as a clinically relevant biomarker and a target for molecule-defined therapy. Fund Unrestricted grant support from the Wilhelm Sander Foundation for Cancer Research, the IZKF Program of the Medical Faculty Tübingen, the Brigitte Schlieben-Lange Program and the Margarete von Wrangell Program of the State Ministry Baden-Wuerttemberg for Science, Research and Arts and the Athene Program of the excellence initiative of the Eberhard-Karls University, Tübingen.
Collapse
Affiliation(s)
- Marcus Matthias Schittenhelm
- Department of Oncology, Hematology, Rheumatology, Immunology and Pulmology, University Hospital Tübingen, Germany
| | - Bianca Walter
- Department of Oncology, Hematology, Rheumatology, Immunology and Pulmology, University Hospital Tübingen, Germany
| | - Vasileia Tsintari
- Department of Oncology, Hematology, Rheumatology, Immunology and Pulmology, University Hospital Tübingen, Germany
| | - Birgit Federmann
- Institute of General and Molecular Pathology and Pathological Anatomy, University Hospital Tübingen, Germany
| | - Mihada Bajrami Saipi
- Department of Oncology, Hematology, Rheumatology, Immunology and Pulmology, University Hospital Tübingen, Germany
| | - Figen Akmut
- Department of Oncology, Hematology, Rheumatology, Immunology and Pulmology, University Hospital Tübingen, Germany
| | - Barbara Illing
- Department of Oncology, Hematology, Rheumatology, Immunology and Pulmology, University Hospital Tübingen, Germany
| | | | - Falko Fend
- Institute of General and Molecular Pathology and Pathological Anatomy, University Hospital Tübingen, Germany
| | - Charles Darin Lopez
- Department of Hematology and Medical Oncology, Oregon Health and Science University, Portland, OR, United States of America
| | | |
Collapse
|
26
|
Xu Q, Gu S, Liang J, Lin Z, Zheng S, Yan J. The Biological Function of Hepatitis B Virus X Protein in Hepatocellular Carcinoma. Oncol Res 2019; 27:509-514. [PMID: 29891022 PMCID: PMC7848407 DOI: 10.3727/096504018x15278771272963] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the major malignant tumors that lead to death. Chronic hepatitis B virus infection is an important risk factor for HCC initiation. HBx protein, encoded by the HBV X gene, is a significant factor that promotes HBV-related HCC, although the exact molecular mechanism remains unclear. This article summarizes the pathological roles and related mechanisms of HBx in HCC. HBx plays a carcinogenic role by promoting cell proliferation, metastasis, and angiogenesis and inhibiting apoptosis in HCC. A detailed study of the biological functions of HBx will help to elucidate the mechanism of hepatocarcinogenesis and lead to the development of novel therapeutic targets for the treatment of HBV-related HCC.
Collapse
Affiliation(s)
- Qiaodong Xu
- Department of Biliary-Pancreatic Minimally Invasive Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, P.R. China
| | - Songgang Gu
- Department of Biliary-Pancreatic Minimally Invasive Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, P.R. China
| | - Jiahong Liang
- Department of Biliary-Pancreatic Minimally Invasive Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, P.R. China
| | - Zhihua Lin
- Department of Biliary-Pancreatic Minimally Invasive Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, P.R. China
| | - Shaodong Zheng
- Department of Biliary-Pancreatic Minimally Invasive Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, P.R. China
| | - Jiang Yan
- Department of Biliary-Pancreatic Minimally Invasive Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, P.R. China
| |
Collapse
|
27
|
Shi H, Zhang Y, Ji J, Xu P, Shi H, Yue X, Ren F, Chen Y, Duan Z, Chen D. Deficiency of apoptosis-stimulating protein two of p53 promotes liver regeneration in mice by activating mammalian target of rapamycin. Sci Rep 2018; 8:17927. [PMID: 30560875 PMCID: PMC6298958 DOI: 10.1038/s41598-018-36208-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 11/09/2018] [Indexed: 12/13/2022] Open
Abstract
Although liver regeneration has been intensively studied in various ways, the mechanisms underlying liver regeneration remain elusive. Apoptosis-stimulating protein two of p53 (ASPP2) was discovered as a binding partner of p53 and plays an important role in regulating cell apoptosis and growth. However, the role of ASPP2 in hepatocyte proliferation and liver regeneration has not been reported. The expression profile of ASPP2 was measured in a mouse model with 70% partial hepatectomy (PHX). Liver regeneration and hepatocyte proliferation were detected in wild-type (ASPP2+/+) and ASPP2 haploinsufficient (ASPP2+/-) mice with PHX. The mammalian target of rapamycin (mTOR) and autophagy pathways were analyzed in the ASPP2+/+ and ASPP2+/- mice with PHX. After rapamycin or 3-methyladenine (3-MA) treatment, hepatocyte proliferation and liver regeneration were analyzed in the ASPP2+/+ and ASPP2+/- mice with PHX. ASPP2 expression was shown to be upregulated at the early stage and downregulated at the late stage. Compared to the ASPP2+/+ mice, liver regeneration was enhanced in ASPP2+/- mice with 70% PHX. In addition, compared to the ASPP2+/+ mice, the mTORC1 pathway was significantly upregulated and the autophagic pathway was downregulated in ASPP2+/-mice with 70% PHX. Inhibition of the mTORC1 pathway significantly suppressed liver regeneration in ASPP2+/- mice with 70% PHX. In contrast, disruption of the autophagic pathway further enhanced liver regeneration in ASPP2+/- mice with 70% PHX. ASPP2 deficiency can promote liver regeneration through activating the mTORC1 pathway, which further regulates downstream molecules, such as those related to autophagy and p70S6K expression in mouse model post-PHX.
Collapse
Affiliation(s)
- Hongbo Shi
- Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China.
- Beijing Institute of Hepatology, Capital Medical University, Beijing, 100069, China.
- Beijing Precision Medicine and Transformation Engineering Technology Research Center of Hepatitis and Liver Center, Beijing, 100069, China.
| | - Yizhi Zhang
- Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Jing Ji
- Shanxi Provincial People's Hospital Affiliated to Shanxi Medical University, Shanxi, China
| | - Ping Xu
- Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
- Beijing Institute of Hepatology, Capital Medical University, Beijing, 100069, China
- Beijing Precision Medicine and Transformation Engineering Technology Research Center of Hepatitis and Liver Center, Beijing, 100069, China
| | - Honglin Shi
- Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
- Beijing Institute of Hepatology, Capital Medical University, Beijing, 100069, China
- Beijing Precision Medicine and Transformation Engineering Technology Research Center of Hepatitis and Liver Center, Beijing, 100069, China
| | - Xiujuan Yue
- Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
- Beijing Institute of Hepatology, Capital Medical University, Beijing, 100069, China
- Beijing Precision Medicine and Transformation Engineering Technology Research Center of Hepatitis and Liver Center, Beijing, 100069, China
| | - Feng Ren
- Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
- Beijing Institute of Hepatology, Capital Medical University, Beijing, 100069, China
- Beijing Precision Medicine and Transformation Engineering Technology Research Center of Hepatitis and Liver Center, Beijing, 100069, China
| | - Yu Chen
- Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Zhongping Duan
- Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
- Beijing Institute of Hepatology, Capital Medical University, Beijing, 100069, China
| | - Dexi Chen
- Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China.
- Beijing Institute of Hepatology, Capital Medical University, Beijing, 100069, China.
- Beijing Precision Medicine and Transformation Engineering Technology Research Center of Hepatitis and Liver Center, Beijing, 100069, China.
| |
Collapse
|
28
|
Charostad J, Astani A, Goudarzi H, Faghihloo E. DNA methyltransferases in virus-associated cancers. Rev Med Virol 2018; 29:e2022. [PMID: 30511446 DOI: 10.1002/rmv.2022] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 10/24/2018] [Accepted: 10/24/2018] [Indexed: 12/19/2022]
Abstract
Human tumor viruses are either casually linked or contribute in the development of human cancers. Viruses can stimulate oncogenesis through affecting diverse biological pathways in human cells. Growing data have demonstrated frequent involvement of one of the most characteristic parts of cellular epigenetic machinery, DNA methylation, in the oncogenesis. DNA methylation of cellular genes is catalyzed by DNA methyltransferases (DNMTs) as a key effector enzyme in this process. Dysregulation of DNMTs can cause aberrant gene methylation in promoter of cancer-related genes including tumor suppressor genes, resulting in gene silencing. In this regard, the role of tumor viruses is remarkable. Here, in this review, we used published information to elucidate whether tumor viruses are able to manipulate DNMT regulation, and if so, what are its consequences in the process of oncogenesis. This essay also aims to shed light on which cellular pathways have been engaged by viruses to induce DNMTs.
Collapse
Affiliation(s)
- Javad Charostad
- Department of Virology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Akram Astani
- Zoonotic Diseases Research Center, School of Public Health, Sahid Sadoughi University of Medical Sciences, Yazd, Iran.,Department of Microbiology, Shahid Sadoghi University of Medical Science, Yazd, Iran
| | - Hossein Goudarzi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ebrahim Faghihloo
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
29
|
Zhou C, Bi F, Yuan J, Yang F, Sun S. Gain of UBE2D1 facilitates hepatocellular carcinoma progression and is associated with DNA damage caused by continuous IL-6. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:290. [PMID: 30482241 PMCID: PMC6260761 DOI: 10.1186/s13046-018-0951-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 11/07/2018] [Indexed: 02/07/2023]
Abstract
Background Hepatocellular carcinoma (HCC) is the most common type of liver cancer with increasing incidence and poor prognosis. Ubiquitination regulators are reported to play crucial roles in HCC carcinogenesis. UBE2D1, one of family member of E2 ubiquitin conjugating enzyme, mediates the ubiquitination and degradation of tumor suppressor protein p53. However, the expression and functional roles of UBE2D1 in HCC was unknown. Methods Immunohistochemistry (IHC), western blotting, and real-time PCR were used to detect the protein, transcription and genomic levels of UBE2D1 in HCC tissues with paired nontumor tissues, precancerous lesions and hepatitis liver tissues. Four HCC cell lines and two immortalized hepatic cell lines were used to evaluate the functional roles and underlying mechanisms of UBE2D1 in HCC initiation and progression in vitro and in vivo. The contributors to UBE2D1 genomic amplification were first evaluated by performing a correlation analysis between UBE2D1 genomic levels with clinical data of HCC patients, and then evaluated in HCC and hepatic cell lines. Results Expression of UBE2D1 was significantly increased in HCC tissues and precancerous lesions and was associated with reduced survival of HCC patients. Upregulation of UBE2D1 promoted HCC growth in vitro and in vivo by decreasing the p53 in ubiquitination-dependent pathway. High expression of UBE2D1 was attributed to the recurrent genomic copy number gain, which was associated with high serum IL-6 level of HCC patients. Further experiments showed that continuous IL-6 activated the DNA damage response and genomic instability by repressing DNA damage checkpoint protein RAD51B. Moreover, continuous IL-6 could significantly facilitate the HCC growth especially with the genomic gain of UBE2D1. Conclusions Our findings showed that UBE2D1 played a crucial role in HCC progression, and suggested a novel pattern of continuous IL-6 to promote cancers by inducing the genomic alterations of specific oncogenes. Electronic supplementary material The online version of this article (10.1186/s13046-018-0951-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chuanchuan Zhou
- Department of Medical Genetics, Second Military Medical University, Shanghai, 200433, China.,Reproductive Medicine Center, Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510000, China
| | - Fengrui Bi
- Department of Medical Genetics, Second Military Medical University, Shanghai, 200433, China
| | - Jihang Yuan
- Department of Medical Genetics, Second Military Medical University, Shanghai, 200433, China
| | - Fu Yang
- Department of Medical Genetics, Second Military Medical University, Shanghai, 200433, China
| | - Shuhan Sun
- Department of Medical Genetics, Second Military Medical University, Shanghai, 200433, China.
| |
Collapse
|
30
|
Bioinformatics identification of crucial genes and pathways associated with hepatocellular carcinoma. Biosci Rep 2018; 38:BSR20181441. [PMID: 30341252 PMCID: PMC6239270 DOI: 10.1042/bsr20181441] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 10/10/2018] [Accepted: 10/10/2018] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a major cause of cancer-related death worldwide. Up to date, HCC pathogenesis has not been fully understood. The aim of the present study was to identify crucial genes and pathways associated with HCC by bioinformatics methods. The differentially expressed genes (DEGs) between 14 HCC tissues and corresponding non-cancerous tissues were identified using limma package. Gene Ontology (GO) and KEGG pathway enrichment analysis of DEGs were performed by clusterProfiler package. The protein-protein interaction (PPI) network of DEGs was constructed and visualized by STRING database and Cytoscape software, respectively. The crucial genes in PPI network were identified using a Cytoscape plugin, CytoNCA. Furthermore, the effect of the expression level of the crucial genes on HCC patient survival was analyzed by an interactive web-portal, UALCAN. A total of 870 DEGs including 237 up-regulated and 633 down-regulated genes were identified in HCC tissues. KEGG pathway analysis revealed that DEGs were mainly enriched in complement and coagulation cascades pathway, chemical carcinogenesis pathway, retinol metabolism pathway, fatty acid degradation pathway, and valine, leucine and isoleucine degradation pathway. PPI network analysis showed that CDK1, CCNB1, CCNB2, MAD2L1, ACACB, IGF1, TOP2A, and EHHADH were crucial genes. Survival analysis suggested that the high expression of CDK1, CCNB1, CCNB2, MAD2L1, and TOP2A significantly decreased the survival probability of HCC patients. In conclusion, the identification of the above crucial genes and pathways will not only contribute to elucidating the pathogenesis of HCC, but also provide prognostic markers and therapeutic targets for HCC.
Collapse
|
31
|
Tian L, Deng Z, Xu L, Yang T, Yao W, Ji L, Lu Y, Zhang J, Liu Y, Wang J. Downregulation of ASPP2 promotes gallbladder cancer metastasis and macrophage recruitment via aPKC-ι/GLI1 pathway. Cell Death Dis 2018; 9:1115. [PMID: 30389910 PMCID: PMC6214900 DOI: 10.1038/s41419-018-1145-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 09/28/2018] [Accepted: 10/09/2018] [Indexed: 12/28/2022]
Abstract
Gallbladder cancer (GBC) is a highly malignant bile duct cancer with poor prognosis due to early invasion and metastasis. However, the molecular mechanisms through which GBC cells interact with the tumor microenvironment (TME) remain poorly understood. Here, we examined the role of the tumor suppressor apoptosis-stimulating of p53 protein 2 (ASPP2) in regulating GBC invasion and metastasis and macrophage recruitment. The clinicopathological significance of ASPP2 expression was measured by immunohistochemical analysis in 72 patients with GBC. Lentivirus-mediated knockdown or overexpression of ASPP2 was used to investigate the biological functions and molecular mechanisms of ASPP2 in GBC cells. Our data showed that downregulation of ASPP2 in patients with GBC was linked to poor prognosis. Knockdown of ASPP2 induced epithelial–mesenchymal transition (EMT) in GBC cells and influenced the TME. Mechanistically, we further confirmed that ASPP2 affected the expression and protein binding between atypical protein kinase C (aPKC)-ι and glioma-associated oncogene homolog 1 (GLI1). ASPP2 also induced C−C motif chemokine ligand (CCL) 2, CCL5, and tumor necrosis factor-α secretion by cancer cells, thereby promoting macrophage recruitment. The latter also induced EMT-like changes in GBC. Furthermore, ASPP2 deficiency regulated GLI1 transcriptional activity via the noncanonical Hedgehog (Hh) pathway and aPKC-ι/GLI1 signaling loop and promoted GLI1 nuclear localization and binding to the promoters of target genes. Our findings revealed that downregulation of ASPP2 promoted GBC invasion and metastasis through the aPKC-ι/GLI1 pathway and enhanced macrophage recruitment. Thus, ASPP2/aPKC-ι/GLI1 pathway may be a potential therapeutic target for the treatment of GBC.
Collapse
Affiliation(s)
- Li Tian
- Department of Biliary and Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, Hubei, China
| | - Zhengdong Deng
- Department of Biliary and Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, Hubei, China
| | - Lei Xu
- Department of Biliary and Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, Hubei, China
| | - Tao Yang
- Department of Biliary and Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, Hubei, China
| | - Wei Yao
- Department of Biliary and Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, Hubei, China
| | - Lei Ji
- Department of Biliary and Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, Hubei, China
| | - Yun Lu
- Department of Biliary and Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, Hubei, China
| | - Jian Zhang
- Department of Biliary and Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, Hubei, China
| | - Yan Liu
- Department of Geriatrics, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, Hubei, China
| | - Jianming Wang
- Department of Biliary and Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, Hubei, China.
| |
Collapse
|
32
|
Li H, Wang X, Zhang C, Cheng Y, Yu M, Zhao K, Ge W, Cai A, Zhang Y, Han F, Hu Y. HDAC1-induced epigenetic silencing of ASPP2 promotes cell motility, tumour growth and drug resistance in renal cell carcinoma. Cancer Lett 2018; 432:121-131. [DOI: 10.1016/j.canlet.2018.06.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 06/04/2018] [Accepted: 06/06/2018] [Indexed: 12/17/2022]
|
33
|
Liu D, Li R, Guo X, Pang L, Zang Y, Liu K, Chen D. DNA damage regulated autophagy modulator 1 recovers the function of apoptosis-stimulating of p53 protein 2 on inducing apoptotic cell death in Huh7.5 cells. Oncol Lett 2018; 15:9333-9338. [PMID: 29844830 DOI: 10.3892/ol.2018.8453] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 03/02/2018] [Indexed: 12/18/2022] Open
Abstract
Overexpression of apoptosis-stimulating of p53 protein 2 (ASPP2) can induce apoptotic cell death in hepatoma cells, which contributes to a killing effect of ASPP2 on treating hepatocellular carcinoma (HCC). In the present study, ASPP2 overexpression failed to induce apoptotic cell death in the HCC Huh7.5 cell line, but promoted autophagy development by inhibiting AKT/mTOR pathway. Inhibition of autophagy using 3-methyladenosine recovered the function of ASPP2 on inducing apoptotic cell death, indicating that ASPP2-induced autophagy has an anti-apoptotic role in Huh7.5 cells. A previous study demonstrated that ASPP2-induced autophagy could induce apoptosis in a CHOP- and DRAM-dependent manner, in which CHOP is involved in the initiation of autophagy and DRAM allows autophagy to induce apoptosis. In the present study, CHOP and DRAM were not involved in ASPP2-induced autophagy; however, the induction of DRAM overexpression recovered the apoptosis-inducing function of ASPP2, indicating that DRAM overexpression switches the role of ASPP2-induced autophagy from anti-apoptotic to pro-apoptotic in Huh7.5 cells. Thus, in combination with DRAM, ASPP2 may better perform its pro-apoptotic role by preventing the occurrence of anti-apoptotic autophagy.
Collapse
Affiliation(s)
- Dongjie Liu
- Capital Medical University Affiliated to Beijing You An Hospital, Beijing 100069, P.R. China.,Beijing Institute of Hepatology, Beijing 100069, P.R. China
| | - Rui Li
- Department of Acupuncture and Mini-invasive Oncology, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing 100029, P.R. China
| | - Xianghua Guo
- Capital Medical University Affiliated to Beijing You An Hospital, Beijing 100069, P.R. China.,Beijing Institute of Hepatology, Beijing 100069, P.R. China
| | - Lijun Pang
- Capital Medical University Affiliated to Beijing You An Hospital, Beijing 100069, P.R. China.,Beijing Institute of Hepatology, Beijing 100069, P.R. China
| | - Yunjin Zang
- Capital Medical University Affiliated to Beijing You An Hospital, Beijing 100069, P.R. China.,Organ Transplantation Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Kai Liu
- Capital Medical University Affiliated to Beijing You An Hospital, Beijing 100069, P.R. China.,Beijing Institute of Hepatology, Beijing 100069, P.R. China
| | - Dexi Chen
- Capital Medical University Affiliated to Beijing You An Hospital, Beijing 100069, P.R. China.,Beijing Institute of Hepatology, Beijing 100069, P.R. China.,Organ Transplantation Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| |
Collapse
|
34
|
Wu T, Song H, Xie D, Zhao B, Xu H, Wu C, Hua K, Deng Y, Ji C, Hu J, Fang L. Silencing of ASPP2 promotes the proliferation, migration and invasion of triple-negative breast cancer cells via the PI3K/AKT pathway. Int J Oncol 2018; 52:2001-2010. [PMID: 29568874 DOI: 10.3892/ijo.2018.4331] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 03/08/2018] [Indexed: 11/06/2022] Open
Abstract
Apoptosis-stimulating p53 protein 2 (ASPP2) is an apoptosis inducer that acts via binding with p53 and then enhancing the transcriptional activities toward pro‑apoptosis genes. ASPP2 has recently been reported to serve a major role in p53‑independent pathways. Triple‑negative breast cancer (TNBC) is a type of breast cancer that is more aggressive and highly lethal when p53 is mutated. In the present study, the mRNA level of ASPP2 was found to be suppressed in breast tumors compared with that in adjacent normal breast tissues, and the expression of ASPP2 was also decreased in a series of breast cancer cell lines compared with that in MCF‑10A normal breast cells. Downregulation of ASPP2 by specific small interfering RNA (siRNA) transfection was able to promote cell growth, reduce cell apoptosis, and contribute to cell migration and invasion. Furthermore, downregulation of ASPP2 promoted cell epithelial‑mesenchymal transition (EMT) in MDA‑MB‑231 and HCC‑1937 TNBC cells. Furthermore, it was found that when ASPP2 siRNA was transfected into MDA‑MB‑231 and HCC‑1937 cells, the expression of phosphoinositide‑3‑kinase regulatory subunit 1 (p85α) decreased and phosphorylation of protein kinase B (AKT) increased, which are key molecular regulators in the phosphatidylinositol 3-kinase (PI3K)/AKT pathway. In conclusion, the present data indicated that ASPP2 had a crucial influence on the proliferation and metastasis in TNBC, and that the functional mechanism may be p53‑independent to a great extent. ASPP2 and its link with the PI3K/AKT pathway deserve further investigation and may provide novel insights into therapeutic targets for TNBC.
Collapse
Affiliation(s)
- Tianqi Wu
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Hongming Song
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Dan Xie
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Bingkun Zhao
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Hui Xu
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Chenyang Wu
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Kaiyao Hua
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Yijun Deng
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Changle Ji
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Jiashu Hu
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Lin Fang
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| |
Collapse
|
35
|
Wang X, Cheng Y, Zhu Y, Li H, Ge W, Wu X, Zhao K, Yuan J, Li Z, Jiang S, Han Z, Jiang Q, Wu Q, Liu T, Zhang C, Yu M, Hu Y. Epigenetic silencing of ASPP1 confers 5‐FU resistance in clear cell renal cell carcinoma by preventing p53 activation. Int J Cancer 2017; 141:1422-1433. [DOI: 10.1002/ijc.30852] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 05/07/2017] [Accepted: 06/21/2017] [Indexed: 12/11/2022]
Affiliation(s)
- Xingwen Wang
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin Heilongjiang150001 China
- Shenzhen Graduate School of Harbin Institute of TechnologyXili University CityNanshanShenzhen Guangdong518055 China
| | - Yiwei Cheng
- The First Affiliated HospitalHarbin Medical UniversityHarbin Heilongjiang150081 China
| | - YiFu Zhu
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin Heilongjiang150001 China
| | - Huayi Li
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin Heilongjiang150001 China
| | - Wenjie Ge
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin Heilongjiang150001 China
- Shenzhen Graduate School of Harbin Institute of TechnologyXili University CityNanshanShenzhen Guangdong518055 China
| | - Xiaoliang Wu
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin Heilongjiang150001 China
| | - Kunming Zhao
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin Heilongjiang150001 China
| | - Jinyang Yuan
- The First Affiliated HospitalHarbin Medical UniversityHarbin Heilongjiang150081 China
| | - Zhenglin Li
- School of Chemical Engineering and TechnologyHarbin Institute of TechnologyHarbin Heilongjiang150001 China
| | - Shijian Jiang
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin Heilongjiang150001 China
| | - Zhengbin Han
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin Heilongjiang150001 China
| | - Qinghua Jiang
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin Heilongjiang150001 China
| | - Qiong Wu
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin Heilongjiang150001 China
| | - Tao Liu
- Shenzhen Luohu People's Hospital, Shenzhen Zhongxun Precision Medicine Research InstituteShenzhen Guangdong518001 China
| | - Cheng Zhang
- The First Affiliated HospitalHarbin Medical UniversityHarbin Heilongjiang150081 China
| | - Miao Yu
- School of Chemical Engineering and TechnologyHarbin Institute of TechnologyHarbin Heilongjiang150001 China
| | - Ying Hu
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin Heilongjiang150001 China
- Shenzhen Graduate School of Harbin Institute of TechnologyXili University CityNanshanShenzhen Guangdong518055 China
| |
Collapse
|
36
|
Xu W, Yu J, Wong VWS. Mechanism and prediction of HCC development in HBV infection. Best Pract Res Clin Gastroenterol 2017; 31:291-298. [PMID: 28774411 DOI: 10.1016/j.bpg.2017.04.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 04/28/2017] [Indexed: 02/07/2023]
Abstract
Chronic hepatitis B virus (HBV) infection remains one of the leading causes of hepatocellular carcinoma (HCC) globally. Over the past few decades, the risk factors of HCC in patients with chronic hepatitis B have been well characterized, and can be divided into host and viral factors. A few groups have also derived and validated HCC prediction scores based on these risk factors. In general, the scores have high negative predictive value in identifying a low risk group who may not need HCC surveillance in the next 3-5 years. The scores have been tested originally in Asian patients, and results on their performance in the Caucasian population are conflicting. Furthermore, new research has identified genetic factors and new virological markers (e.g. hepatitis B surface antigen and core-related antigen levels) for HCC, but they are yet to be applied in routine clinical practice.
Collapse
Affiliation(s)
- Weiqi Xu
- State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong
| | - Jun Yu
- State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong; Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong
| | - Vincent Wai-Sun Wong
- State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong; Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong.
| |
Collapse
|
37
|
EGR-1/ASPP1 inter-regulatory loop promotes apoptosis by inhibiting cyto-protective autophagy. Cell Death Dis 2017; 8:e2869. [PMID: 28594407 PMCID: PMC5520923 DOI: 10.1038/cddis.2017.268] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 05/10/2017] [Accepted: 05/10/2017] [Indexed: 01/07/2023]
Abstract
The decrease of ASPP1 (Apoptosis-Stimulating Protein of p53 1), a known p53 activator, has been linked to carcinogenesis and the cytotoxic resistance in various cancers, yet the underlying mechanisms of ASPP1 expression and its complex functions are not yet clear. Here, we report that ASPP1 forms an inter-regulatory loop with Early Growth Response 1 (EGR-1), and promotes apoptosis via inhibiting cyto-protective autophagy, independent of the well-documented p53-dependent mechanisms. We show that ASPP1 mRNA and protein were remarkably elevated by ectopic EGR-1 expression or endogenous EGR-1 activation, in cells with different tissue origins and p53 status. Conversely, RNAi-mediated EGR-1 knockdown suppressed ASPP1. The further mechanism studies revealed that ASPP1 promoter, mapped to -283/+88, which contained three conserved EGR-1 binding sites, was required for both binding and transactivity of EGR-1. In addition, we demonstrate that ASPP1 promoted EGR-1 in a positive feedback loop by preventing proteasome-mediated EGR-1 degradation or promoting EGR-1 nuclear import in response to anticancer natural compound Quercetin. Furthermore, albeit activating p53 in the nucleus is the well-studied function of ASPP1, we found that ASPP1 was predominately localized in the cytoplasm. Interestingly, the cytoplasmic ASPP1 retained its pro-apoptosis capability. Mechanistically, ASPP1 suppressed Atg5-Atg12 and also bound with Atg5-Atg12 to prevent its further complex formation with Atg16, resulting in the inhibition of cyto-protective autophagy. In conclusion, our results provide new insights into EGR-1/ASPP1 regulatory loop in sensitizing Quercetin-induced apoptosis. EGR-1/ASPP1, therefore, may be potentially used as therapeutic targets to improve cancer's response to pro-apoptosis treatments.
Collapse
|
38
|
Gen Y, Yasui K, Kitaichi T, Iwai N, Terasaki K, Dohi O, Hashimoto H, Fukui H, Inada Y, Fukui A, Jo M, Moriguchi M, Nishikawa T, Umemura A, Yamaguchi K, Konishi H, Naito Y, Itoh Y. ASPP2 suppresses invasion and TGF-β1-induced epithelial-mesenchymal transition by inhibiting Smad7 degradation mediated by E3 ubiquitin ligase ITCH in gastric cancer. Cancer Lett 2017; 398:52-61. [PMID: 28400336 DOI: 10.1016/j.canlet.2017.04.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 03/21/2017] [Accepted: 04/02/2017] [Indexed: 12/12/2022]
Abstract
ASPP2 regulates cell polarity and cell-cell adhesion by binding to, and co-localizing with PAR3 at tight junctions. Here we show a novel role of ASPP2 in suppressing gastric cancer (GC) invasiveness. Immunoprecipitation and immunofluorescence analyses showed that ASPP2 promoted the recruitment of PAR3 to cell-cell junctions in GC cells. Diminished expression of ASPP2 and loss of junctional PAR3 localization were significantly associated with diffuse-type histology, deeper invasion depth, positive peritoneal dissemination and worse prognosis in primary GC. ASPP2 suppressed migration and invasion of GC cells in vitro and peritoneal dissemination of GC cells in vivo in a mouse model. ASPP2 suppressed epithelial-mesenchymal transition (EMT) induced by TGF-β1-Smad2/3 signaling in GC cells through suppression of the degradation of Smad7, a negative regulator of TGF-β1-Smad2/3 signaling, by interacting with the E3 ubiquitin ligase ITCH. In conclusion, ASPP2 suppresses invasion, peritoneal dissemination and TGF-β1-induced EMT by inhibiting Smad7 degradation mediated by ITCH.
Collapse
Affiliation(s)
- Yasuyuki Gen
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan; Department of Molecular Cytogenetics, Medical Research Institute and Graduate School of Medical and Dental Science, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kohichiroh Yasui
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan.
| | - Tomoko Kitaichi
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Naoto Iwai
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kei Terasaki
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Osamu Dohi
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hikaru Hashimoto
- Department of Gastroenterology and Hepatology, North Medical Center Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hayato Fukui
- Department of Gastroenterology and Hepatology, North Medical Center Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yutaka Inada
- Department of Gastroenterology and Hepatology, North Medical Center Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Akifumi Fukui
- Department of Gastroenterology and Hepatology, North Medical Center Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masayasu Jo
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Michihisa Moriguchi
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Taichiro Nishikawa
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Atushi Umemura
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kanji Yamaguchi
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroyuki Konishi
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yuji Naito
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yoshito Itoh
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
39
|
Tao Y, Hu K, Tan F, Zhang S, Zhou M, Luo J, Wang Z. SH3-domain binding protein 1 in the tumor microenvironment promotes hepatocellular carcinoma metastasis through WAVE2 pathway. Oncotarget 2017; 7:18356-70. [PMID: 26933917 PMCID: PMC4951293 DOI: 10.18632/oncotarget.7786] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Accepted: 02/11/2016] [Indexed: 02/06/2023] Open
Abstract
SH3-domain binding protein-1 (SH3BP1) specifically inactivating Rac1 and its target WAVE2 is required for cell motility. The present study shows SH3BP1 expression patterns in human HCC tissues and cell lines were examined. The regulation of SH3BP1 on HCC cell migration and invasion related to Rac1-WAVE2 signaling was characterized using in vitro and in vivo models. SH3BP1 overexpressed in HCC tissues and highly metastatic HCC cells was significantly associated vascular invasion (VI). SH3BP1 promoted VEGF secretion via Rac1-WAVE2 signaling, so as to exert an augmentation on cell invasion and microvessel formation. In three study cohorts with a total of 516 HCC patients, high SH3BP1 expression combined with high microvessel density (MVD) was confirmed as a powerful independent predictor of HCC prognosis in both training cohorts and validation cohort. Being an important angiogenic factor of HCC through Rac1-WAVE2 signaling, SH3BP1 promotes tumor invasion and microvessel formation contributing to HCC metastasis and recurrence. SH3BP1 is a novel WAVE2 regulator, a prognostic marker and a potential therapeutic target of HCC.
Collapse
Affiliation(s)
- Yiming Tao
- Department of Hepatobiliary Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Kuan Hu
- Department of Hepatobiliary Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fengbo Tan
- Department of Hepatobiliary Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Sai Zhang
- Institute of Medical Sciences, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ming Zhou
- Institute of Cancer Research, Central South University, Changsha, Hunan, China
| | - Jia Luo
- Department of Hepatobiliary Surgery, Hunan Provincial Tumor Hospital, Changsha, Hunan, China
| | - Zhiming Wang
- Department of Hepatobiliary Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
40
|
Downregulation of ASPP2 improves hepatocellular carcinoma cells survival via promoting BECN1-dependent autophagy initiation. Cell Death Dis 2016; 7:e2512. [PMID: 27929538 PMCID: PMC5260975 DOI: 10.1038/cddis.2016.407] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 10/01/2016] [Accepted: 10/18/2016] [Indexed: 01/01/2023]
Abstract
Autophagy is an important catabolic process, which sustains intracellular homeostasis and lengthens cell survival under stress. Here we identify the ankyrin-repeat-containing, SH3-domain-containing, and proline-rich region-containing protein 2 (ASPP2), a haploinsufficient tumor suppressor, as a molecular regulator of starvation-induced autophagy in hepatocellular carcinoma (HCC). ASPP2 expression is associated with an autophagic response upon nutrient deprivation and downregulation of ASPP2 facilitates autophagic flux, whereas overexpression of ASPP2 blocks this starvation-induced autophagy in HCC cells. Mechanistically, ASPP2 inhibits autophagy through regulating BECN1 transcription and formation of phosphatidylinositol 3-kinase catalytic subunit type 3 (PIK3C3) complex. Firstly, ASPP2 inhibits p65/RelA-induced transcription of BECN1, directly by an ASPP2-p65/RelA-IκBα complex which inhibits phosphorylation of IκBα and the translocation of p65/RelA into the nucleus. Secondly, ASPP2 binds to BECN1, leading to decreased binding of PIK3C3 and UV radiation resistance-associated gene (UVRAG), and increased binding of Rubicon in PIK3C3 complex. Downregulation of ASPP2 enhances the pro-survival and chemoresistant property via autophagy in HCC cells in vitro and in vivo. Decreased ASPP2 expression was associated with increased BECN1 and poor survival in HCC patients. Therefore, ASPP2 is a key regulator of BECN1-dependent autophagy, and decreased ASPP2 may contribute to tumor progression and chemoresistance via promoting autophagy.
Collapse
|
41
|
Van Hook K, Wang Z, Chen D, Nold C, Zhu Z, Anur P, Lee HJ, Yu Z, Sheppard B, Dai MS, Sears R, Spellman P, Lopez CD. ΔN-ASPP2, a novel isoform of the ASPP2 tumor suppressor, promotes cellular survival. Biochem Biophys Res Commun 2016; 482:1271-1277. [PMID: 27939881 DOI: 10.1016/j.bbrc.2016.12.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 12/05/2016] [Indexed: 12/15/2022]
Abstract
ASPP2 is a tumor suppressor that works, at least in part, through enhancing p53-dependent apoptosis. We now describe a new ASPP2 isoform, ΔN-ASPP2, generated from an internal transcription start site that encodes an N-terminally truncated protein missing a predicted 254 amino acids. ΔN-ASPP2 suppresses p53 target gene transactivation, promoter occupancy, and endogenous p53 target gene expression in response to DNA damage. Moreover, ΔN-ASPP2 promotes progression through the cell cycle, as well as resistance to genotoxic stress-induced growth inhibition and apoptosis. Additionally, we found that ΔN-ASPP2 expression is increased in human breast tumors as compared to adjacent normal breast tissue; in contrast, ASPP2 is suppressed in the majority of these breast tumors. Together, our results provide insight into how this new ASPP2 isoform may play a role in regulating the ASPP2-p53 axis.
Collapse
Affiliation(s)
- Kathryn Van Hook
- Department of Medicine, Division of Hematology and Medical Oncology and the Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Zhiping Wang
- Department of Medicine, Division of Hematology and Medical Oncology and the Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Dexi Chen
- Department of Medicine, Division of Hematology and Medical Oncology and the Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA; Beijing Institute of Hepatology, Beijing You'an Hospital, Capital Medical University, Beijing, 100069, China
| | - Casey Nold
- Department of Medicine, Division of Hematology and Medical Oncology and the Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Zhiyi Zhu
- Department of Medicine, Division of Hematology and Medical Oncology and the Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Pavana Anur
- Department of Molecular and Medical Genetics and the Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Hun-Joo Lee
- Department of Medicine, Division of Hematology and Medical Oncology and the Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Zhiyong Yu
- Shandong Tumor Hospital and Institute, Jinan, 250117, China
| | - Brett Sheppard
- Department of Surgery and the Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Mu-Shui Dai
- Department of Molecular and Medical Genetics and the Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Rosalie Sears
- Department of Molecular and Medical Genetics and the Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Paul Spellman
- Department of Molecular and Medical Genetics and the Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Charles D Lopez
- Department of Medicine, Division of Hematology and Medical Oncology and the Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA.
| |
Collapse
|
42
|
Wang X, Yu M, Zhao K, He M, Ge W, Sun Y, Wang Y, Sun H, Hu Y. Upregulation of MiR-205 under hypoxia promotes epithelial-mesenchymal transition by targeting ASPP2. Cell Death Dis 2016; 7:e2517. [PMID: 27929537 PMCID: PMC5261019 DOI: 10.1038/cddis.2016.412] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 10/24/2016] [Accepted: 11/10/2016] [Indexed: 12/22/2022]
Abstract
The epithelial–mesenchymal transition (EMT) is one of the crucial procedures for cancer invasion and distal metastasis. Despite undergoing intensive studies, the mechanisms underlying EMT remain to be completely elucidated. Here, we identified that apoptosis-stimulating protein of p53-2 (ASPP2) is a novel target of MiR-205 in various cancers. Interestingly, the binding site of MiR-205 at the 3′-untranslated region of ASPP2 was highly conserved among different species. An inverse correlation between MiR-205 and ASPP2 was further observed in vivo in cervical cancers, suggesting MiR-205 may be an important physiological inhibitor of ASPP2. Hypoxia is a hallmark of solid tumor microenvironment and one of such conditions to induce EMT. Notably, MiR-205 was remarkably induced by hypoxia in cervical and lung cancer cells. A marked suppression of ASPP2 was observed simultaneously. Further studies confirmed that hypoxia-induced ASPP2 suppression was mainly attributed to the elevated MiR-205. Interestingly, the alteration of MiR-205/ASPP2 under hypoxia was accompanied with the decreased epithelial marker E-cadherin and increased mesenchymal marker Vimentin, as well as a morphological transition from the typical cobblestone-like appearance to the mesenchymal-like structure. More importantly, MiR-205 mimics or ASPP2 silencing similarly promoted EMT process. By contrast, ASPP2 recovery or MiR-205 inhibitor reversed MiR-205-dependent EMT. Further studies demonstrated that the newly revealed MiR-205/ASPP2 axis promoted cell migration and also increased cell proliferation both in vivo and in vitro. These data together implicated a critical impact of MiR-205/ASPP2 on promoting EMT. MiR-205/ASPP2 may be potential diagnostic and therapeutic biomarkers in cervical and lung cancers.
Collapse
Affiliation(s)
- Xingwen Wang
- School of Life Science and Technology, Harbin Institute of Technology, Shenzhen, China.,Shenzhen Graduate School of Harbin Institute of Technology, Shenzhen, China
| | - Miao Yu
- School of Chemical Engineering and Technology, Harbin Institute of Technology, Harbin, China
| | - Kunming Zhao
- School of Life Science and Technology, Harbin Institute of Technology, Shenzhen, China
| | - Mengmeng He
- School of Life Science and Technology, Harbin Institute of Technology, Shenzhen, China
| | - Wenjie Ge
- School of Life Science and Technology, Harbin Institute of Technology, Shenzhen, China.,Shenzhen Graduate School of Harbin Institute of Technology, Shenzhen, China
| | - Yuhui Sun
- The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yihua Wang
- Biological Sciences, Faculty of Natural and Environmental Sciences, University of Southampton, Southampton, UK
| | - Haizhu Sun
- The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Ying Hu
- School of Life Science and Technology, Harbin Institute of Technology, Shenzhen, China.,Shenzhen Graduate School of Harbin Institute of Technology, Shenzhen, China
| |
Collapse
|
43
|
Hepatitis C virus core protein inhibits E6AP expression via DNA methylation to escape from ubiquitin-dependent proteasomal degradation. Cancer Lett 2016; 380:59-68. [DOI: 10.1016/j.canlet.2016.06.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 06/03/2016] [Accepted: 06/13/2016] [Indexed: 01/16/2023]
|
44
|
Xu L, Tong X, Zhang S, Yin F, Li X, Wei H, Li C, Guo Y, Zhao J. ASPP2 suppresses stem cell-like characteristics and chemoresistance by inhibiting the Src/FAK/Snail axis in hepatocellular carcinoma. Tumour Biol 2016; 37:13669-13677. [PMID: 27473084 DOI: 10.1007/s13277-016-5246-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 07/15/2016] [Indexed: 10/21/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of death in cancer patients worldwide. Understanding the molecular pathogenesis of HCC recurrence and chemoresistance is key to improving patients' prognosis. In this study, we report that downregulation of ASPP2, a member of the ankyrin-repeat-containing, SH3-domain-containing, and proline-rich-region-containing protein (ASPP) family, bestowed HCC cells with stem-like properties and resistance to chemotherapy, including the expansion of side population fractions, formation of hepatospheroids, expression of stem cell-associated genes, loss of chemosensitivity, and increased tumorigenicity in immunodeficient mice. An expression profiling assay revealed that ASPP2 specifically repressed focal adhesion kinase (FAK)/Src/extracellular signal regulated kinase (ERK) signaling. ASPP2 does this by physically interacting with C-terminal Src kinase (CSK) and stimulating its kinase activity, which eventually leads to activator protein 1 (AP1)-mediated downregulation of Snail expression. In addition, pharmacologic inhibition of Src attenuated the effects of ASPP2 deficiency. Our findings present functional and mechanistic insight into the critical role of ASPP2 in the inhibition of HCC stemness and drug resistance and may provide a new strategy for therapeutic combinations to treat HCC.
Collapse
Affiliation(s)
- Lu Xu
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
| | - Xin Tong
- PLA General Hospital Cancer Center Key Lab, PLA Postgraduate School of Medicine, 28 Fuxing Road, Beijing, China.,Beijing Key Laboratory of Cell Engineering and Antibody, Beijing, China
| | - Sujie Zhang
- PLA General Hospital Cancer Center Key Lab, PLA Postgraduate School of Medicine, 28 Fuxing Road, Beijing, China.,Beijing Key Laboratory of Cell Engineering and Antibody, Beijing, China
| | - Fan Yin
- PLA General Hospital Cancer Center Key Lab, PLA Postgraduate School of Medicine, 28 Fuxing Road, Beijing, China.,Beijing Key Laboratory of Cell Engineering and Antibody, Beijing, China
| | - Xiaoyan Li
- PLA General Hospital Cancer Center Key Lab, PLA Postgraduate School of Medicine, 28 Fuxing Road, Beijing, China.,Beijing Key Laboratory of Cell Engineering and Antibody, Beijing, China
| | - Huafeng Wei
- PLA General Hospital Cancer Center Key Lab, PLA Postgraduate School of Medicine, 28 Fuxing Road, Beijing, China.,Beijing Key Laboratory of Cell Engineering and Antibody, Beijing, China
| | - Cheng Li
- PLA General Hospital Cancer Center Key Lab, PLA Postgraduate School of Medicine, 28 Fuxing Road, Beijing, China.,Beijing Key Laboratory of Cell Engineering and Antibody, Beijing, China
| | - Yajun Guo
- State Key Laboratory of Antibody Medicine and Targeting Therapy, Shanghai, China
| | - Jian Zhao
- PLA General Hospital Cancer Center Key Lab, PLA Postgraduate School of Medicine, 28 Fuxing Road, Beijing, China. .,Beijing Key Laboratory of Cell Engineering and Antibody, Beijing, China. .,International Joint Cancer Institute, The Second Military Medical University, 800 Xiang Yin Road, New Building 9th Floor, Shanghai, China.
| |
Collapse
|
45
|
Levrero M, Zucman-Rossi J. Mechanisms of HBV-induced hepatocellular carcinoma. J Hepatol 2016; 64:S84-S101. [PMID: 27084040 DOI: 10.1016/j.jhep.2016.02.021] [Citation(s) in RCA: 630] [Impact Index Per Article: 78.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 02/17/2016] [Accepted: 02/17/2016] [Indexed: 02/06/2023]
Abstract
Hepatitis B virus (HBV) contributes to hepatocellular carcinoma (HCC) development through direct and indirect mechanisms. HBV DNA integration into the host genome occurs at early steps of clonal tumor expansion and induces both genomic instability and direct insertional mutagenesis of diverse cancer-related genes. Prolonged expression of the viral regulatory protein HBx and/or altered versions of the preS/S envelope proteins dysregulates cell transcription and proliferation control and sensitizes liver cells to carcinogenic factors. Accumulation of preS1 large envelope proteins and/or preS2/S mutant proteins activates the unfold proteins response, that can contribute to hepatocyte transformation. Epigenetic changes targeting the expression of tumor suppressor genes occur early in the development of HCC. A major role is played by the HBV protein, HBx, which is recruited on cellular chromatin and modulates chromatin dynamics at specific gene loci. Compared with tumors associated with other risk factors, HBV-related tumors have a higher rate of chromosomal alterations, p53 inactivation by mutations and overexpression of fetal liver/hepatic progenitor cells genes. The WNT/β-catenin pathway is also often activated but HBV-related tumors display a low rate of activating β-catenin mutations. HBV-related HCCs may arise on non-cirrhotic livers, further supporting the notion that HBV plays a direct role in liver transformation by triggering both common and etiology specific oncogenic pathways in addition to stimulating the host immune response and driving liver chronic necro-inflammation.
Collapse
Affiliation(s)
- Massimo Levrero
- Cancer Research Center of Lyon (CRCL) - INSERM U1052, Lyon, France; IIT Centre for Life Nanoscience (CLNS), Rome, Italy; Dept of Internal Medicine (DMISM), Sapienza University, Rome, Italy.
| | - Jessica Zucman-Rossi
- Inserm, UMR-1162, Génomique Fonctionnelle des Tumeurs Solides, Equipe Labellisée Ligue Contre le Cancer, Institut Universitaire d'Hematologie, Paris, France; Université Paris Descartes, Labex Immuno-Oncology, Sorbonne Paris Cité, Faculté de Médecine, Paris, France; Université Paris 13, Sorbonne Paris Cité, Unité de Formation et de Recherche Santé, Médecine, Biologie Humaine, Bobigny, France; Université Paris Diderot, Paris, France.
| |
Collapse
|
46
|
Liang HW, Wang N, Wang Y, Wang F, Fu Z, Yan X, Zhu H, Diao W, Ding Y, Chen X, Zhang CY, Zen K. Hepatitis B virus-human chimeric transcript HBx-LINE1 promotes hepatic injury via sequestering cellular microRNA-122. J Hepatol 2016; 64:278-291. [PMID: 26409216 DOI: 10.1016/j.jhep.2015.09.013] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 09/04/2015] [Accepted: 09/13/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Chronic hepatitis B virus (HBV) carriers have a high risk to develop hepatocellular carcinoma (HCC) but the underlying mechanism remains unclear. Recent studies suggest that viral-human hybrid RNA transcripts, which play a critical role in promoting HCC progression, may be the molecules responsible for the development of HCC in HBV infected patients. Here we determine whether HBx-LINE1, a hybrid RNA transcript of the human LINE1 and the HBV-encoded X gene generated in tumor cells of HBV-positive HCC, can serve as a molecular sponge for sequestering miR-122 and promoting liver cell abnormal mitosis and mouse hepatic injury. METHODS Paired tumor and distal normal liver tissue specimens, as well as HBx-LINE1 overexpressing hepatic cells, were used to test the relationship between HBx-LINE1 and miR-122. Levels of HBx-LINE1 and miR-122 were assayed by qRT-PCR and Northern blot. HBx-LINE1-miR-122 binding was analyzed by luciferase reporter assay. Mouse hepatic injury was monitored by tissue staining and serum aspartate transaminase, alanine aminotransferase and total bilirubin measurement. RESULTS HBx-LINE1 in HBV-positive HCC tissues was inversely correlated with miR-122. Each HBx-LINE1 consists of six miR-122-binding sites, and forced expression of HBx-LINE1 effectively depleted cellular miR-122, promoting hepatic cell epithelial-mesenchymal transition (EMT)-like changes, including β-catenin signaling activation, E-cadherin reduction and cell migration enhancement. Mice administered with HBx-LINE1 display a significant mouse liver cell abnormal mitosis and hepatic injury. However, all these effects of HBx-LINE1 are completely abolished by miR-122. CONCLUSIONS Our finding illustrates a previously uncharacterized miR-122-sequestering mechanism by which HBx-LINE1 promotes hepatic cell EMT-like changes and mouse liver injury.
Collapse
Affiliation(s)
- Hong-Wei Liang
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University Advanced Institute of Life Sciences, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Nan Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University Advanced Institute of Life Sciences, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Yanbo Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University Advanced Institute of Life Sciences, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Feng Wang
- Department of General Surgery, the Affiliated Gulou Hospital of Nanjing University, Nanjing, Jiangsu 210093, China
| | - Zheng Fu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University Advanced Institute of Life Sciences, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Xin Yan
- Comprehensive Cancer Center, the Affiliated Gulou Hospital of Nanjing University, Nanjing, Jiangsu 210093, China
| | - Hao Zhu
- Department of Gastroenterology, the Affiliated Gulou Hospital of Nanjing University, Nanjing, Jiangsu 210093, China
| | - Wenli Diao
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University Advanced Institute of Life Sciences, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Yitao Ding
- Department of Hepatobiliary Surgery, the Affiliated Gulou Hospital of Nanjing University, Nanjing, Jiangsu 210093, China.
| | - Xi Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University Advanced Institute of Life Sciences, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, Nanjing University, Nanjing, Jiangsu 210093, China.
| | - Chen-Yu Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University Advanced Institute of Life Sciences, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, Nanjing University, Nanjing, Jiangsu 210093, China.
| | - Ke Zen
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University Advanced Institute of Life Sciences, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, Nanjing University, Nanjing, Jiangsu 210093, China.
| |
Collapse
|
47
|
Idrissi ME, Hachem H, Koering C, Merle P, Thénoz M, Mortreux F, Wattel E. HBx triggers either cellular senescence or cell proliferation depending on cellular phenotype. J Viral Hepat 2016; 23:130-8. [PMID: 26316075 DOI: 10.1111/jvh.12450] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 07/16/2015] [Indexed: 01/04/2023]
Abstract
Replicative senescence is a hallmark of chronic liver diseases including chronic hepatitis B virus (HBV) infection, whereas HBV-encoded oncoproteins HBx and preS2 have been found to overcome senescence. HBx possesses a C-terminal truncation mainly in hepatocellular carcinomas but also in noncancerous liver tissues. Here, by cell counting, BrdU incorporation, MTT proliferation assay, cell cycle analysis, SA-βgal staining and Western blotting in primary and malignant cells, we investigated the effect of HBx C-terminal mutants on cellular senescence. HBx C-terminal mutants were found to trigger cellular senescence in primary MRC5 cells, and malignant liver cells Huh7, and SK-Hep1. In contrast, these mutants promoted the proliferation of HepG2 malignant liver cells. The pro-senescent effect of HBx relied on an increased p16(INK4a) and p21(Waf1/Cip1) expression, and a decreased phosphorylation of Rb. Together, these results suggest that the two main variants of HBx present in HBV-infected liver possess opposite effects on cellular senescence that depend on the phenotype of infected cells.
Collapse
Affiliation(s)
- M E Idrissi
- Université Lyon-1, CNRS UMR5239, Oncovirologie et Biothérapies, Lyon, France
| | - H Hachem
- Université Lyon-1, CNRS UMR5239, Oncovirologie et Biothérapies, Lyon, France
| | - C Koering
- Université Lyon-1, CNRS UMR5239, Oncovirologie et Biothérapies, Lyon, France
| | - P Merle
- INSERM U1052, CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France.,Université Lyon-1, Villeurbanne, France.,Hospices Civils de Lyon, Service d'Hépatologie et de Gastroentérologie, Groupement Hospitalier Lyon Nord, Lyon, France
| | - M Thénoz
- Université Lyon-1, CNRS UMR5239, Oncovirologie et Biothérapies, Lyon, France
| | - F Mortreux
- Université Lyon-1, CNRS UMR5239, Oncovirologie et Biothérapies, Lyon, France
| | - E Wattel
- Université Lyon-1, CNRS UMR5239, Oncovirologie et Biothérapies, Lyon, France.,Université Lyon-1, Service d'Hématologie, Pavillon Marcel Bérard, Centre Hospitalier Lyon-Sud, Pierre Bénite, France
| |
Collapse
|
48
|
Guerrieri F, Belloni L, Pediconi N, Levrero M. Pathobiology of Hepatitis B Virus-Induced Carcinogenesis. ACTA ACUST UNITED AC 2016. [DOI: 10.1007/978-3-319-22330-8_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
49
|
Liu J, Su S. Relationship between long non-coding RNAs and liver-related diseases and injuries. Shijie Huaren Xiaohua Zazhi 2015; 23:5784-5789. [DOI: 10.11569/wcjd.v23.i36.5784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) are a class of non-coding RNAs>200 bp in length. In recent years, it was discovered that lncRNAs play an important role in many physiological processes, such as transcription activation, transcriptional interference, X chromosome silencing, genomic imprinting and chromatin modification, transport and so on. More and more studies show alterations of lncRNA expression in primary liver cancer, hepatitis and other liver diseases. This paper reviews the relationship between long non-coding RNAs and liver-related diseases and injuries.
Collapse
|
50
|
Minarovits J, Demcsák A, Banati F, Niller HH. Epigenetic Dysregulation in Virus-Associated Neoplasms. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 879:71-90. [DOI: 10.1007/978-3-319-24738-0_4] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|