1
|
Lian J, Feng N, Xie M, Zhang H, Li L, Ahmed MMM, Chen Z, Ding Y, Kang X. Hepatitis B Virus Disrupts the Blood-Testis Barrier via the Induction of mTOR-Dependent Autophagy in Sertoli Cells. FASEB J 2025; 39:e70547. [PMID: 40266246 DOI: 10.1096/fj.202403422r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 03/10/2025] [Accepted: 04/07/2025] [Indexed: 04/24/2025]
Abstract
Hepatitis B virus (HBV) is one of the most serious public health threats worldwide. HBV is not only able to pass through the blood-testis barrier (BTB); It can also cause impairment of male fertility. However, the mechanisms involved in this process remain unknown. In this study, we showed that HBV can establish persistent infection in human and mouse testes. Persistent HBV infection triggers inflammatory cell invasion, testes immune homeostasis imbalance, and the disruption of the BTB formed by inter-Sertoli cells. HBV mainly persisted in the Sertoli cells and could induce the autophagy of Sertoli cells by HBV X protein (HBx), a major regulatory protein of HBV. Data indicated that the mTOR signal pathway-mediated autophagy plays a pivotal role in HBV-induced BTB damage. Autophagy inhibitor 3-MA and mTOR activator MHY1485 could ameliorate HBV-induced autophagy and BTB damage. These findings demonstrated that the mTOR-mediated excessive autophagy of Sertoli cells induced by HBx could be one of the pathological mechanisms responsible for the fertility decline caused by HBV infection.
Collapse
Affiliation(s)
- Jingyao Lian
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Na Feng
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Minyu Xie
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hanbin Zhang
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lei Li
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mohamed Morsi M Ahmed
- Faculty of Science, Department of Biological Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Nucleic Acids Research Department, Genetic Engineering, and Biotechnology Research Institute (GEBRI), City for Scientific Research and Technological Applications, Alexandria, Egypt
| | - Zhenguo Chen
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yue Ding
- Department of Pediatrics, NanFang Hospital, Southern Medical University, Guangzhou, China
- Department of Pediatrics, Ganzhou People's Hospital, Ganzhou, China
| | - Xiangjin Kang
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
2
|
Bayat M, Golestani S, Motlaghzadeh S, Bannazadeh Baghi H, Lalehzadeh A, Sadri Nahand J. War or peace: Viruses and metastasis. Biochim Biophys Acta Rev Cancer 2024; 1879:189179. [PMID: 39299491 DOI: 10.1016/j.bbcan.2024.189179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 09/04/2024] [Accepted: 09/07/2024] [Indexed: 09/22/2024]
Abstract
Metastasis, the dissemination of malignant cells from a primary tumor to secondary sites, poses a catastrophic burden to cancer treatment and is the predominant cause of mortality in cancer patients. Metastasis as one of the main aspects of cancer progression could be strongly under the influence of viral infections. In fact, viruses have been central to modern cancer research and are associated with a great number of cancer cases. Viral-encoded elements are involved in modulating essential pathways or specific targets that are implicated in different stages of metastasis. Considering the continuous emergence of new viruses and the establishment of their contribution to cancer progression, the warfare between viruses and cancer appears to be endless. Here we aimed to review the critical mechanism and pathways involved in cancer metastasis and the influence of viral machinery and various routes that viruses adopt to manipulate those pathways for their benefit.
Collapse
Affiliation(s)
- Mobina Bayat
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shahin Golestani
- Department of ophthalmology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeed Motlaghzadeh
- Department of Virology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Bannazadeh Baghi
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aidin Lalehzadeh
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javid Sadri Nahand
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
3
|
Lee J, Kim J, Lee R, Lee E, An H, Kwon Y, Jin H, Pack C, Kim I, Yoon Y, Park G, Jwa E, Kwon JH, Namgoong J, Song G, Hwang S, Tak E, Lee S. SOD1 inhibition enhances sorafenib efficacy in HBV-related hepatocellular carcinoma by modulating PI3K/Akt/mTOR pathway and ROS-mediated cell death. J Cell Mol Med 2024; 28:e18533. [PMID: 39034442 PMCID: PMC11260765 DOI: 10.1111/jcmm.18533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 05/21/2024] [Accepted: 07/05/2024] [Indexed: 07/23/2024] Open
Abstract
Hepatitis B Virus (HBV) infection significantly elevates the risk of hepatocellular carcinoma (HCC), with the HBV X protein (HBx) playing a crucial role in cancer progression. Sorafenib, the primary therapy for advanced HCC, shows limited effectiveness in HBV-infected patients due to HBx-related resistance. Numerous studies have explored combination therapies to overcome this resistance. Sodium diethyldithiocarbamate (DDC), known for its anticancer effects and its inhibition of superoxide dismutase 1 (SOD1), is hypothesized to counteract sorafenib (SF) resistance in HBV-positive HCCs. Our research demonstrates that combining DDC with SF significantly reduces HBx and SOD1 expressions in HBV-positive HCC cells and human tissues. This combination therapy disrupts the PI3K/Akt/mTOR signalling pathway and promotes apoptosis by increasing reactive oxygen species (ROS) levels. These cellular changes lead to reduced tumour viability and enhanced sensitivity to SF, as evidenced by the synergistic suppression of tumour growth in xenograft models. Additionally, DDC-mediated suppression of SOD1 further enhances SF sensitivity in HBV-positive HCC cells and xenografted animals, thereby inhibiting cancer progression more effectively. These findings suggest that the DDC-SF combination could serve as a promising strategy for overcoming SF resistance in HBV-related HCC, potentially optimizing therapy outcomes.
Collapse
Affiliation(s)
- Jooyoung Lee
- Asan Institute for Life Sciences, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulSouth Korea
- Department of Biochemistry and Molecular Biology, AMIST, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulSouth Korea
| | - Jiye Kim
- Asan Institute for Life Sciences, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulSouth Korea
- Department of Biochemistry and Molecular Biology, AMIST, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulSouth Korea
| | - Ryunjin Lee
- Asan Institute for Life Sciences, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulSouth Korea
- Department of Biochemistry and Molecular Biology, AMIST, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulSouth Korea
| | - Eunkyeong Lee
- Asan Institute for Life Sciences, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulSouth Korea
- Department of Biochemistry and Molecular Biology, AMIST, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulSouth Korea
| | - Hye‐In An
- Asan Institute for Life Sciences, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulSouth Korea
- Department of Biochemistry and Molecular Biology, AMIST, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulSouth Korea
| | - Yong‐Jae Kwon
- Department of Surgery, Gangneung Asan HospitalUniversity of Ulsan College of MedicineSeoulSouth Korea
| | - Hana Jin
- Division of Vascular Surgery, Department of Surgery, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulSouth Korea
| | - Chan‐Gi Pack
- Convergence Medicine Research Center (CREDIT)Asan Institute for Life Sciences, ASAN Medical CenterSeoulRepublic of Korea
| | - Inki Kim
- Convergence Medicine Research Center (CREDIT)Asan Institute for Life Sciences, ASAN Medical CenterSeoulRepublic of Korea
| | - Young‐In Yoon
- Division of Hepatobiliary Surgery and Liver Transplantation, Department of Surgery, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulSouth Korea
| | - Gil‐Chun Park
- Division of Hepatobiliary Surgery and Liver Transplantation, Department of Surgery, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulSouth Korea
| | - Eun‐Kyoung Jwa
- Division of Hepatobiliary Surgery and Liver Transplantation, Department of Surgery, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulSouth Korea
| | - Jae Hyun Kwon
- Department of Surgery, Hallym University Sacred Heart HospitalHallym University College of MedicineAnyangSouth Korea
| | - Jung‐Man Namgoong
- Division of Pediatric Surgery, Department of Surgery, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulSouth Korea
| | - Gi‐Won Song
- Division of Hepatobiliary Surgery and Liver Transplantation, Department of Surgery, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulSouth Korea
| | - Shin Hwang
- Division of Hepatobiliary Surgery and Liver Transplantation, Department of Surgery, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulSouth Korea
| | - Eunyoung Tak
- Asan Institute for Life Sciences, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulSouth Korea
- Department of Biochemistry and Molecular Biology, AMIST, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulSouth Korea
| | - Sung‐Gyu Lee
- Division of Hepatobiliary Surgery and Liver Transplantation, Department of Surgery, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulSouth Korea
| |
Collapse
|
4
|
Li Q, Wen W, Wang Y, Gong T, Wang X, Tan Q, Fan B, Xie H, Li Y, Li S, Yang C, Zhou Z, Duan X, Lin W, Chen L. Autophagy-related protein 5 (ATG5) interacts with bone marrow stromal cell antigen 2 (BST2) to stimulate HBV replication through antagonizing the antiviral activity of BST2. J Med Virol 2024; 96:e29659. [PMID: 38747016 DOI: 10.1002/jmv.29659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 04/15/2024] [Accepted: 04/30/2024] [Indexed: 06/05/2024]
Abstract
Hepatitis B virus (HBV) infection is a major global health burden with 820 000 deaths per year. In our previous study, we found that the knockdown of autophagy-related protein 5 (ATG5) significantly upregulated the interferon-stimulated genes (ISGs) expression to exert the anti-HCV effect. However, the regulation of ATG5 on HBV replication and its underlying mechanism remains unclear. In this study, we screened the altered expression of type I interferon (IFN-I) pathway genes using RT² Profiler™ PCR array following ATG5 knock-down and we found the bone marrow stromal cell antigen 2 (BST2) expression was significantly increased. We then verified the upregulation of BST2 by ATG5 knockdown using RT-qPCR and found that the knockdown of ATG5 activated the Janus kinase/signal transducer and activator of transcription (JAK-STAT) signaling pathway. ATG5 knockdown or BST2 overexpression decreased Hepatitis B core Antigen (HBcAg) protein, HBV DNA levels in cells and supernatants of HepAD38 and HBV-infected NTCP-HepG2. Knockdown of BST2 abrogated the anti-HBV effect of ATG5 knockdown. Furthermore, we found that ATG5 interacted with BST2, and further formed a ternary complex together with HBV-X (HBx). In conclusion, our finding indicates that ATG5 promotes HBV replication through decreasing BST2 expression and interacting with it directly to antagonize its antiviral function.
Collapse
Affiliation(s)
- Qingyuan Li
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, Sichuan, China
- Research Platform for Transfusion-transmitted Diseases, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Key Laboratory for Transfusion-transmitted Infectious Diseases of the Health Commission of Sichuan Province, Chengdu, Sichuan, China
| | - Wenxian Wen
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, Sichuan, China
- Research Platform for Transfusion-transmitted Diseases, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Key Laboratory for Transfusion-transmitted Infectious Diseases of the Health Commission of Sichuan Province, Chengdu, Sichuan, China
| | - Yijin Wang
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Tao Gong
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, Sichuan, China
- Research Platform for Transfusion-transmitted Diseases, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Key Laboratory for Transfusion-transmitted Infectious Diseases of the Health Commission of Sichuan Province, Chengdu, Sichuan, China
| | - Xinwei Wang
- Joint Laboratory on Transfusion-transmitted Infectious Diseases between Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Nanning Blood Center, Nanning Blood Center, Key Laboratory for Transfusion-transmitted Infectious Diseases of the Health Commission of Nanning City, Nanning, Guangxi, China
| | - Qi Tan
- Research Platform for Transfusion-transmitted Diseases, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Key Laboratory for Transfusion-transmitted Infectious Diseases of the Health Commission of Sichuan Province, Chengdu, Sichuan, China
| | - Bin Fan
- Research Platform for Transfusion-transmitted Diseases, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Key Laboratory for Transfusion-transmitted Infectious Diseases of the Health Commission of Sichuan Province, Chengdu, Sichuan, China
| | - He Xie
- Department of Clinical Laboratory, The Hospital of Xidian Group, Xian, Shaanxi, China
| | - Yujia Li
- Research Platform for Transfusion-transmitted Diseases, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Key Laboratory for Transfusion-transmitted Infectious Diseases of the Health Commission of Sichuan Province, Chengdu, Sichuan, China
| | - Shilin Li
- Research Platform for Transfusion-transmitted Diseases, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Key Laboratory for Transfusion-transmitted Infectious Diseases of the Health Commission of Sichuan Province, Chengdu, Sichuan, China
| | - Chunhui Yang
- Research Platform for Transfusion-transmitted Diseases, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Key Laboratory for Transfusion-transmitted Infectious Diseases of the Health Commission of Sichuan Province, Chengdu, Sichuan, China
| | - Zhonghui Zhou
- Department of Infectious Diseases, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Xiaoqiong Duan
- Research Platform for Transfusion-transmitted Diseases, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Key Laboratory for Transfusion-transmitted Infectious Diseases of the Health Commission of Sichuan Province, Chengdu, Sichuan, China
| | - Wenyu Lin
- Department of Medicine, Liver Center and Gastrointestinal Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Limin Chen
- Research Platform for Transfusion-transmitted Diseases, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Key Laboratory for Transfusion-transmitted Infectious Diseases of the Health Commission of Sichuan Province, Chengdu, Sichuan, China
- Joint Laboratory on Transfusion-transmitted Infectious Diseases between Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Nanning Blood Center, Nanning Blood Center, Key Laboratory for Transfusion-transmitted Infectious Diseases of the Health Commission of Nanning City, Nanning, Guangxi, China
- Department of Clinical Laboratory, The Hospital of Xidian Group, Xian, Shaanxi, China
| |
Collapse
|
5
|
Agustiningsih A, Rasyak MR, Turyadi, Jayanti S, Sukowati C. The oncogenic role of hepatitis B virus X gene in hepatocarcinogenesis: recent updates. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:120-134. [PMID: 38464387 PMCID: PMC10918233 DOI: 10.37349/etat.2024.00209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/21/2023] [Indexed: 03/12/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the most prevalent form of primary liver cancers with high mortality rate. Among its various etiological factors, one of the major risk factors for HCC is a chronic infection of hepatitis B virus (HBV). HBV X protein (HBx) has been identified to play an important role in the HBV-induced HCC pathogenesis since it may interfere with several key regulators of many cellular processes. HBx localization within the cells may be beneficial to HBx multiple functions at different phases of HBV infection and associated hepatocarcinogenesis. HBx as a regulatory protein modulates cellular transcription, molecular signal transduction, cell cycle, apoptosis, autophagy, protein degradation pathways, and host genetic stability via interaction with various factors, including its association with various non-coding RNAs. A better understanding on the regulatory mechanism of HBx on various characteristics of HCC would provide an overall picture of HBV-associated HCC. This article addresses recent data on HBx role in the HBV-associated hepatocarcinogenesis.
Collapse
Affiliation(s)
- Agustiningsih Agustiningsih
- Eijkman Research Center for Molecular Biology, National Research and Innovation Agency of Indonesia (BRIN), Jakarta Pusat 10340, Indonesia
| | - Muhammad Rezki Rasyak
- Eijkman Research Center for Molecular Biology, National Research and Innovation Agency of Indonesia (BRIN), Jakarta Pusat 10340, Indonesia
- Post Graduate School, Faculty of Medicine, Universitas Hasanuddin, Makassar 90245, Indonesia
| | - Turyadi
- Eijkman Research Center for Molecular Biology, National Research and Innovation Agency of Indonesia (BRIN), Jakarta Pusat 10340, Indonesia
| | - Sri Jayanti
- Eijkman Research Center for Molecular Biology, National Research and Innovation Agency of Indonesia (BRIN), Jakarta Pusat 10340, Indonesia
| | - Caecilia Sukowati
- Eijkman Research Center for Molecular Biology, National Research and Innovation Agency of Indonesia (BRIN), Jakarta Pusat 10340, Indonesia
- Liver Cancer Unit, Fondazione Italiana Fegato ONLUS, AREA Science Park, Basovizza, 34149 Trieste, Italy
| |
Collapse
|
6
|
Shojaeian A, Nakhaie M, Amjad ZS, Boroujeni AK, Shokri S, Mahmoudvand S. Leveraging metformin to combat hepatocellular carcinoma: its therapeutic promise against hepatitis viral infections. JOURNAL OF CANCER METASTASIS AND TREATMENT 2024. [DOI: 10.20517/2394-4722.2023.147] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Hepatocellular carcinoma (HCC) is categorized among the most common primary malignant liver cancer and a primary global cause of death from cancer. HCC tends to affect males 2-4 times more than females in many nations. The main factors that raise the incidence of HCC are chronic liver diseases, hepatotropic viruses like hepatitis B (HBV) and C (HCV), non-alcoholic fatty liver disease, exposure to toxins like aflatoxin, and non-alcoholic steatohepatitis (NASH). Among these, hepatitis B and C are the most prevalent causes of chronic hepatitis globally. Metformin, which is made from a naturally occurring compound called galegine, derived from the plant Galega officinalis (G. officinalis ), has been found to exhibit antitumor effects in a wide range of malignancies, including HCC. In fact, compared to patients on sulphonylureas or insulin, studies have demonstrated that metformin treatment significantly lowers the risk of HCC in patients with chronic liver disease. This article will first describe the molecular mechanism of hepatitis B and C viruses in the development of HCC. Then, we will provide detailed explanations about metformin, followed by a discussion of the association between metformin and hepatocellular carcinoma caused by the viruses mentioned above.
Collapse
|
7
|
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), as the causative agent of the recent COVID-19 pandemic, continues representing one of the main health concerns worldwide. Autophagy, in addition to its role in cellular homeostasis and metabolism, plays an important part for the host antiviral immunity. However, viruses including SARS-CoV-2 have evolved diverse mechanisms to not only overcome autophagy's antiviral pressure but also manipulate its machinery in order to enhance viral replication and propagation. Here, we discuss our current knowledge on the impact that autophagy exerts on SARS-CoV-2 replication, as well as the different counteracting measures that this virus has developed to manipulate autophagy's complex machinery. Some of the elements regarding this interplay may become future therapeutic targets in the fight against SARS-CoV-2.
Collapse
Affiliation(s)
- Hao Zhou
- Department of Microbiology and Immunology, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhiqiang Hu
- Shandong New Hope Liuhe Agriculture and Animal Husbandry Technology Co., Ltd, Dezhou, China
| | | |
Collapse
|
8
|
Choi YM, Kim DH, Jang J, Choe WH, Kim BJ. rt269L-Type hepatitis B virus (HBV) in genotype C infection leads to improved mitochondrial dynamics via the PERK-eIF2α-ATF4 axis in an HBx protein-dependent manner. Cell Mol Biol Lett 2023; 28:26. [PMID: 36997871 PMCID: PMC10064691 DOI: 10.1186/s11658-023-00440-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 03/15/2023] [Indexed: 04/01/2023] Open
Abstract
BACKGROUND In our previous report, the rt269I type versus the rt269L type in genotype C2 infection led to poor clinical outcomes and enhanced mitochondrial stress in infected hepatocytes. Here, we sought to investigate differences between the rt269L and rt269I types in mitochondrial functionality in hepatitis B virus (HBV) genotype C2 infection, mainly focusing on endoplasmic reticulum (ER) stress-mediated autophagy induction as an upstream signal. METHODS Mitochondrial functionality, ER stress signaling, autophagy induction, and apoptotic cell death between rt269L-type and rt269I-type groups were investigated via in vitro and in vivo experiments. Serum samples were collected from 187 chronic hepatitis patients who visited Konkuk or Seoul National University Hospital. RESULTS Our data revealed that genotype C rt269L versus rt269I infection led to improved mitochondrial dynamics and enhanced autophagic flux, mainly due to the activation of the PERK-eIF2α-ATF4 axis. Furthermore, we demonstrated that the traits found in genotype C rt269L infection were mainly due to increased stability of the HBx protein after deubiquitination. In addition, clinical data using patient sera from two independent Korean cohorts showed that, compared with rt269I, rt269L in infection led to lower 8-OHdG levels, further supporting its improved mitochondrial quality control. CONCLUSION Our data showed that, compared with the rt269I type, the rt269L type, which presented exclusively in HBV genotype C infection, leads to improved mitochondrial dynamics or bioenergetics, mainly due to autophagy induction via activation of the PERK-eIF2α-ATF4 axis in an HBx protein-dependent manner. This suggests that HBx stability and cellular quality control in the rt269L type predominating in genotype C endemic areas could at least partly contribute to some distinctive traits of genotype C infection, such as higher infectivity or longer duration of the hepatitis B e antigen (HBeAg) positive stage.
Collapse
Affiliation(s)
- Yu-Min Choi
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul, 110-799, Republic of Korea
| | - Dong Hyun Kim
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul, 110-799, Republic of Korea
| | - Junghwa Jang
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul, 110-799, Republic of Korea
| | - Won Hyeok Choe
- Department of Internal Medicine, Konkuk University School of Medicine, Seoul, 05030, Republic of Korea
| | - Bum-Joon Kim
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul, 110-799, Republic of Korea.
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea.
- Liver Research Institute, College of Medicine, Seoul National University, Seoul, 03080, Korea.
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul, 03080, Korea.
- Seoul National University Medical Research Center (SNUMRC), Seoul, 03080, Korea.
| |
Collapse
|
9
|
Paskeh MDA, Ghadyani F, Hashemi M, Abbaspour A, Zabolian A, Javanshir S, Razzazan M, Mirzaei S, Entezari M, Goharrizi MASB, Salimimoghadam S, Aref AR, Kalbasi A, Rajabi R, Rashidi M, Taheriazam A, Sethi G. Biological impact and therapeutic perspective of targeting PI3K/Akt signaling in hepatocellular carcinoma: Promises and Challenges. Pharmacol Res 2023; 187:106553. [PMID: 36400343 DOI: 10.1016/j.phrs.2022.106553] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/09/2022] [Accepted: 11/10/2022] [Indexed: 11/17/2022]
Abstract
Cancer progression results from activation of various signaling networks. Among these, PI3K/Akt signaling contributes to proliferation, invasion, and inhibition of apoptosis. Hepatocellular carcinoma (HCC) is a primary liver cancer with high incidence rate, especially in regions with high prevalence of viral hepatitis infection. Autoimmune disorders, diabetes mellitus, obesity, alcohol consumption, and inflammation can also lead to initiation and development of HCC. The treatment of HCC depends on the identification of oncogenic factors that lead tumor cells to develop resistance to therapy. The present review article focuses on the role of PI3K/Akt signaling in HCC progression. Activation of PI3K/Akt signaling promotes glucose uptake, favors glycolysis and increases tumor cell proliferation. It inhibits both apoptosis and autophagy while promoting HCC cell survival. PI3K/Akt stimulates epithelial-to-mesenchymal transition (EMT) and increases matrix-metalloproteinase (MMP) expression during HCC metastasis. In addition to increasing colony formation capacity and facilitating the spread of tumor cells, PI3K/Akt signaling stimulates angiogenesis. Therefore, silencing PI3K/Akt signaling prevents aggressive HCC cell behavior. Activation of PI3K/Akt signaling can confer drug resistance, particularly to sorafenib, and decreases the radio-sensitivity of HCC cells. Anti-cancer agents, like phytochemicals and small molecules can suppress PI3K/Akt signaling by limiting HCC progression. Being upregulated in tumor tissues and clinical samples, PI3K/Akt can also be used as a biomarker to predict patients' response to therapy.
Collapse
Affiliation(s)
- Mahshid Deldar Abad Paskeh
- 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
| | - Fatemeh Ghadyani
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - 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
| | - Alireza Abbaspour
- Cellular and Molecular Research Center,Qazvin University of Medical Sciences, Qazvin, Iran
| | - Amirhossein Zabolian
- Resident of department of Orthopedics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Salar Javanshir
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mehrnaz Razzazan
- Medical Student, Student Research Committee, Golestan University of Medical Sciences, Gorgan, Iran
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, 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
| | | | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Translational Sciences, Xsphera Biosciences Inc. 6, Tide Street, Boston, MA 02210, USA
| | - Alireza Kalbasi
- Department of Pharmacy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Romina Rajabi
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran.
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, 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.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore.
| |
Collapse
|
10
|
He H, Li X, Shen J, Bai S, Li C, Shi H. Bisphenol A exposure causes testicular toxicity by targeting DPY30-mediated post-translational modification of PI3K/AKT signaling in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 243:113996. [PMID: 36030680 DOI: 10.1016/j.ecoenv.2022.113996] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 06/22/2022] [Accepted: 08/16/2022] [Indexed: 06/15/2023]
Abstract
Bisphenol A (BPA), one of the chemicals with the highest volume of production worldwide, has been demonstrated to cause testicular toxicity via different pathways. However, there is little evidence concerning the mechanism of BPA exposure induced histone modification alterations, especially regarding the effect on the histone H3 lysine 4 (H3K4) epigenetic modification. Our results demonstrated a new epigenetic regulation of BPA exposure on testicular damage using both cell culture and mouse models. With BPA treatment, disordered and shrunken seminiferous tubules and poor sperm quality were observed in vivo, and mouse spermatogonial germ cell proliferation was inhibited in vitro. BPA attenuated PI3K expression inducing phospho-AKT inhibition in vivo and in vitro. DPY30 was the only downregulated subunit in BPA and MEK2206 (AKT inhibitor) treated cells, which contributed to reducing H3K4me3 recruitment at the PIK3CA transcriptional start site (TSS) in BPA treated cells. The toxicity caused by BPA exposure was relieved after the transduction of adenoviruses expressing DPY30 transgenes, which resulted in the stimulation of PI3K/AKT with H3K4me3 enriched at the PI3KCA TSS. DPY30 promoted cell glycolysis via AMPK and proliferation through AKT/P21. DPY30 was mainly located in the round and elongated spermatids for energy accumulation in mature sperm in AD-DPY30-treated mice which showed higher sperm quality. Overall, our results indicated that BPA exposure causes testicular toxicity through a DPY30-mediated H3K4me3 epigenetic modification, which serves to regulate the PI3K/AKT/P21 pathway.
Collapse
Affiliation(s)
- Huanshan He
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Xiang Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Jianing Shen
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Shuying Bai
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Cong Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Huaiping Shi
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China.
| |
Collapse
|
11
|
Dong ML, Wen X, He X, Ren JH, Yu HB, Qin YP, Yang Z, Yang ML, Zhou CY, Zhang H, Cheng ST, Chen J. HBx Mediated Increase of DDX17 Contributes to HBV-Related Hepatocellular Carcinoma Tumorigenesis. Front Immunol 2022; 13:871558. [PMID: 35784274 PMCID: PMC9243429 DOI: 10.3389/fimmu.2022.871558] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 05/09/2022] [Indexed: 11/13/2022] Open
Abstract
HBV is strongly associated with HCC development and DEAD-box RNA helicase 17 (DDX17) is a very important member of the DEAD box family that plays key roles in HCC development by promoting cancer metastasis. However, the important role of DDX17 in the pathogenesis of HBV-related HCC remains unclear. In this study, we investigated the role of DDX17 in the replication of HBV and the development of HBV-associated HCC. Based on data from the GEO database and HBV-infected cells, we found that DDX17 was upregulated by the HBV viral protein X (HBx). Mechanistically, increased DDX17 expression promoted HBV replication and transcription by upregulating ZWINT. Further study showed that DDX17 could promote HBx-mediated HCC metastasis. Finally, the promotive effect of DDX17 on HBV and HBV-related HCC was confirmed in vivo. In summary, the results revealed the novel role of DDX17 in the replication of HBV and the metastasis of HBV-associated HCC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Juan Chen
- *Correspondence: Juan Chen, ; Sheng-Tao Cheng,
| |
Collapse
|
12
|
Hepatitis B Viral Protein HBx and the Molecular Mechanisms Modulating the Hallmarks of Hepatocellular Carcinoma: A Comprehensive Review. Cells 2022; 11:cells11040741. [PMID: 35203390 PMCID: PMC8870387 DOI: 10.3390/cells11040741] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/10/2022] [Accepted: 02/16/2022] [Indexed: 02/06/2023] Open
Abstract
With 296 million cases estimated worldwide, chronic hepatitis B virus (HBV) infection is the most common risk factor for hepatocellular carcinoma (HCC). HBV-encoded oncogene X protein (HBx), a key multifunctional regulatory protein, drives viral replication and interferes with several cellular signalling pathways that drive virus-associated hepatocarcinogenesis. This review article provides a comprehensive overview of the role of HBx in modulating the various hallmarks of HCC by supporting tumour initiation, progression, invasion and metastasis. Understanding HBx-mediated dimensions of complexity in driving liver malignancies could provide the key to unlocking novel and repurposed combinatorial therapies to combat HCC.
Collapse
|
13
|
Bai X, Ran J, Zhao X, Liang Y, Yang X, Xi Y. The S100A10-AnxA2 complex is associated with the exocytosis of hepatitis B virus in intrauterine infection. J Transl Med 2022; 102:57-68. [PMID: 34645932 PMCID: PMC8512653 DOI: 10.1038/s41374-021-00681-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 09/22/2021] [Accepted: 09/24/2021] [Indexed: 12/21/2022] Open
Abstract
Mother-to-child transmission (MTCT) is the major cause of chronic infection of hepatitis B virus (HBV) in patients. However, whether and how HBV crosses the placenta to cause infection in utero remains unclear. In this study, we investigate the mechanism as to how the HBV virions pass through layers of the trophoblast. Our data demonstrate the exocytosis of virions from the trophoblast after exposure to HBV where the endocytosed HBV virions co-localized with an S100A10/AnxA2 complex and LC3, an autophagosome membrane marker. Knockdown of either AnxA2 or S100A10 in trophoblast cells led to a reduction of the amount of exo-virus in Transwell assay. Immunohistochemistry also showed a high expression of AnxA2 and S100A10 in the placental tissue samples of HBV-infected mothers with congenital HBV-positive infants (HBV+/+). We conclude that in HBV intrauterine infection and mother-to-child transmission, a proportion of HBV hijacks autophagic protein secretion pathway and translocate across the trophoblast via S100A10/AnxA2 complex and multivesicular body (MVB)-mediated exocytosis. Our study provides a potential target for the interference of the mechanisms of HBV intrauterine infection and mother-to-child transmission.
Collapse
Grants
- National Natural Science Foundation of China (National Science Foundation of China)
- China’s National Key R&D Programs (NKPs) are a new category of projects created after the 2014 reform of the national STI funding system. They have incorporated numerous previously-existing programmes such as MOST’s “863 Programme” for R&D, “Programme 973” for basic research, Key Technologies R&D Programme, and International S&T Cooperation Programme; and NDRC and MIIT’s Industrial Technology R&D Fund. China’s National Key R&D Programmes support R&D in areas of social welfare and people’s livelihood, such as agriculture, energy and resources, environment, and health. They focus in particular on key and strategic technologies, featuring several well-targeted and defined objectives and deliverables to be achieved in a period ranging from three to five years, and reflecting a top-down and industry-university-research cooperation design which integrates basic research, technology application, demonstration and commercialisation.
Collapse
Affiliation(s)
- Xiaoxia Bai
- The Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Shangcheng District, Hangzhou, Zhejiang, 310001, China.
| | - Jinshi Ran
- The Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Shangcheng District, Hangzhou, Zhejiang, 310001, China
- Institute of Genetics and Department of Human Genetics, Zhejiang University School of Medicine; Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, No. 866, Yuhangtang Road, Hangzhou, Zhejiang, 310058, China
| | - Xianlei Zhao
- The Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Shangcheng District, Hangzhou, Zhejiang, 310001, China
- Institute of Genetics and Department of Human Genetics, Zhejiang University School of Medicine; Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, No. 866, Yuhangtang Road, Hangzhou, Zhejiang, 310058, China
| | - Yun Liang
- The Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Shangcheng District, Hangzhou, Zhejiang, 310001, China
| | - Xiaohang Yang
- The Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Shangcheng District, Hangzhou, Zhejiang, 310001, China
- Institute of Genetics and Department of Human Genetics, Zhejiang University School of Medicine; Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, No. 866, Yuhangtang Road, Hangzhou, Zhejiang, 310058, China
- Joint Institute of Genetics and Genomic Medicine between Zhejiang University and University of Toronto, Zhejiang University, No. 866, Yuhangtang Road, Hangzhou, Zhejiang, 310058, China
| | - Yongmei Xi
- The Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Shangcheng District, Hangzhou, Zhejiang, 310001, China.
- Institute of Genetics and Department of Human Genetics, Zhejiang University School of Medicine; Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, No. 866, Yuhangtang Road, Hangzhou, Zhejiang, 310058, China.
| |
Collapse
|
14
|
Yassin NYS, AbouZid SF, El-Kalaawy AM, Ali TM, Almehmadi MM, Ahmed OM. Silybum marianum total extract, silymarin and silibinin abate hepatocarcinogenesis and hepatocellular carcinoma growth via modulation of the HGF/c-Met, Wnt/β-catenin, and PI3K/Akt/mTOR signaling pathways. Biomed Pharmacother 2022; 145:112409. [PMID: 34781148 DOI: 10.1016/j.biopha.2021.112409] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/30/2021] [Accepted: 11/03/2021] [Indexed: 12/19/2022] Open
Abstract
Hepatocellular carcinoma (HCC) has been identified as one of the most deadly malignancies with limited therapeutic efficacy worldwide. However, understanding the molecular mechanisms of crosstalk between signaling pathways in HCC and predicting cancer cell responses to targeted therapeutic interventions remain to be challenge. Thus, in this study, we aimed to evaluate the anticancerous efficacy of Silybum marianum total extract (STE), silymarin (Sm), and silibinin (Sb) against experimentally-induced HCC in rats. In vitro investigations were also performed and the anticancer effects against HCC cell lines (HepG2 and Huh7) were confirmed. Wistar rats were given diethylnitrosamine (DEN)/2-acetylaminofluorene (AAF)/carbon tetrachloride (CCl4) and were orally treated with STE (200 mg/kg body weight (bw)), Sm (150 mg/kg bw), and Sb (5 mg/kg bw) every other day from the 1st or 16th week to the 25th week of DEN/AAF/CCl4 injection. Treatment with STE, Sm, and Sb inhibited the growth of cancerous lesions in DEN/AAF/CCl4-treated rats. This inhibition was associated with inhibition of Ki-67 expression and repression of HGF/cMet, Wnt/β-catenin, and PI3K/Akt/mTOR signaling pathways. STE, Sm, and Sb improved liver function biomarkers and tumor markers (AFP, CEA, and CA19.9) and increased total protein and albumin levels in serum. STE, Sm, and Sb treatment was also noted to reduce the hepatic production of lipid peroxides, increase hepatic glutathione content, and induce the activities of hepatic antioxidant enzymes in DEN/AAF/CCl4-treated rats. These results indicate that STE, Sm, and Sb exert anti-HCC effects through multiple pathways, including suppression of Ki-67 expression and HGF/cMet, Wnt/β-catenin, and PI3K/Akt/mTOR pathways and enhancement of antioxidant defense mechanisms.
Collapse
Affiliation(s)
- Nour Y S Yassin
- Physiology Division, Department of Zoology, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| | - Sameh F AbouZid
- Department of Pharmacognosy, Faculty of Pharmacy, Heliopolis University for Sustainable Development, 3 Cairo-Belbeis Desert Road, P.O. Box 3020 El Salam, 11785 Cairo, Egypt
| | - Asmaa M El-Kalaawy
- Department of Pharmacology, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Tarek M Ali
- Department of Physiology, College of Medicine, Taif University, P. O. Box 11099, Taif 21944, Saudi Arabia
| | - Mazen M Almehmadi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P. O. Box 11099, Taif 21944, Saudi Arabia
| | - Osama M Ahmed
- Physiology Division, Department of Zoology, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt.
| |
Collapse
|
15
|
Zhang X, Ming Y, Fu X, Niu Y, Lin Q, Liang H, Luo X, Liu L, Li N. PI3K/AKT/p53 pathway inhibits infectious spleen and kidney necrosis virus infection by regulating autophagy and immune responses. FISH & SHELLFISH IMMUNOLOGY 2022; 120:648-657. [PMID: 34968710 DOI: 10.1016/j.fsi.2021.12.046] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/20/2021] [Accepted: 12/26/2021] [Indexed: 06/14/2023]
Abstract
The PI3K/AKT/p53 signaling pathway is activated by various types of cellular stimuli or pathogenic infection, and then regulates fundamental cellular functions to combat these stimulations. Here, we studied the meaningful roles of PI3K/AKT/p53 in regulating cellular machine such as autophagy, immune responses, as well as antiviral activity in Chinese perch brain (CPB) cells infected by infectious spleen and kidney necrosis virus (ISKNV), which is an agent caused devastating losses in mandarin fish (Siniperca chuatsi) industry. We found that ISKNV infection induced up-regulation of host PI3K/AKT/p53 axis, but inhibited autophagy in CPB cells. Interestingly, activation of PI3K/AKT/p53 axis factors trough agonists or overexpression dramatically decreased host autophagy level, inhibited ISKNV replication, and elevated the expression of immune-related genes in CPB cells. In contrast, suppression of PI3K/AKT/p53 pathway by inhibitors or small interfering RNA (siRNA)-mediated gene silence increased the autophagy and ISKNV replication, but down-regulated immune responses in CPB cells. All these results indicate that PI3K/AKT/p53 pathway plays an important role in anti-ISKNV infection and can be used as a new target for controlling ISKNV disease.
Collapse
Affiliation(s)
- Xiaoting Zhang
- Pearl River Fishery Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of Fishery Drug Development, Ministry of Agriculture and Rural Affairs, Guangdong Province Key Laboratory of Aquatic Animal Immune Technology, Guangzhou, 510380, China
| | - Yue Ming
- Pearl River Fishery Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of Fishery Drug Development, Ministry of Agriculture and Rural Affairs, Guangdong Province Key Laboratory of Aquatic Animal Immune Technology, Guangzhou, 510380, China
| | - Xiaozhe Fu
- Pearl River Fishery Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of Fishery Drug Development, Ministry of Agriculture and Rural Affairs, Guangdong Province Key Laboratory of Aquatic Animal Immune Technology, Guangzhou, 510380, China
| | - Yinjie Niu
- Pearl River Fishery Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of Fishery Drug Development, Ministry of Agriculture and Rural Affairs, Guangdong Province Key Laboratory of Aquatic Animal Immune Technology, Guangzhou, 510380, China
| | - Qiang Lin
- Pearl River Fishery Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of Fishery Drug Development, Ministry of Agriculture and Rural Affairs, Guangdong Province Key Laboratory of Aquatic Animal Immune Technology, Guangzhou, 510380, China
| | - Hongru Liang
- Pearl River Fishery Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of Fishery Drug Development, Ministry of Agriculture and Rural Affairs, Guangdong Province Key Laboratory of Aquatic Animal Immune Technology, Guangzhou, 510380, China
| | - Xia Luo
- Pearl River Fishery Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of Fishery Drug Development, Ministry of Agriculture and Rural Affairs, Guangdong Province Key Laboratory of Aquatic Animal Immune Technology, Guangzhou, 510380, China
| | - Lihui Liu
- Pearl River Fishery Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of Fishery Drug Development, Ministry of Agriculture and Rural Affairs, Guangdong Province Key Laboratory of Aquatic Animal Immune Technology, Guangzhou, 510380, China
| | - Ningqiu Li
- Pearl River Fishery Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of Fishery Drug Development, Ministry of Agriculture and Rural Affairs, Guangdong Province Key Laboratory of Aquatic Animal Immune Technology, Guangzhou, 510380, China.
| |
Collapse
|
16
|
Sadri Nahand J, Salmaninejad A, Mollazadeh S, Tamehri Zadeh SS, Rezaee M, Sheida AH, Sadoughi F, Dana PM, Rafiyan M, Zamani M, Taghavi SP, Dashti F, Mirazimi SMA, Bannazadeh Baghi H, Moghoofei M, Karimzadeh M, Vosough M, Mirzaei H. Virus, Exosome, and MicroRNA: New Insights into Autophagy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1401:97-162. [DOI: 10.1007/5584_2022_715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
17
|
Son J, Kim MJ, Lee JS, Kim JY, Chun E, Lee KY. Hepatitis B virus X Protein Promotes Liver Cancer Progression through Autophagy Induction in Response to TLR4 Stimulation. Immune Netw 2021; 21:e37. [PMID: 34796041 PMCID: PMC8568915 DOI: 10.4110/in.2021.21.e37] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/28/2021] [Accepted: 10/20/2021] [Indexed: 12/14/2022] Open
Abstract
Hepatitis B virus X (HBx) protein has been reported as a key protein regulating the pathogenesis of HBV-induced hepatocellular carcinoma (HCC). Recent evidence has shown that HBx is implicated in the activation of autophagy in hepatic cells. Nevertheless, the precise molecular and cellular mechanism by which HBx induces autophagy is still controversial. Herein, we investigated the molecular and cellular mechanism by which HBx is involved in the TRAF6-BECN1-Bcl-2 signaling for the regulation of autophagy in response to TLR4 stimulation, therefore influencing the HCC progression. HBx interacts with BECN1 (Beclin 1) and inhibits the association of the BECN1-Bcl-2 complex, which is known to prevent the assembly of the pre-autophagosomal structure. Furthermore, HBx enhances the interaction between VPS34 and TRAF6-BECN1 complex, increases the ubiquitination of BECN1, and subsequently enhances autophagy induction in response to LPS stimulation. To verify the functional role of HBx in liver cancer progression, we utilized different HCC cell lines, HepG2, SK-Hep-1, and SNU-761. HBx-expressing HepG2 cells exhibited enhanced cell migration, invasion, and cell mobility in response to LPS stimulation compared to those of control HepG2 cells. These results were consistently observed in HBx-expressed SK-Hep-1 and HBx-expressed SNU-761 cells. Taken together, our findings suggest that HBx positively regulates the induction of autophagy through the inhibition of the BECN1-Bcl-2 complex and enhancement of the TRAF6-BECN1-VPS34 complex, leading to enhance liver cancer migration and invasion.
Collapse
Affiliation(s)
- Juhee Son
- Department of Immunology and Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Mi-Jeong Kim
- Department of Immunology and Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Ji Su Lee
- Department of Immunology and Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Ji Young Kim
- Department of Immunology and Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Korea
| | | | - Ki-Young Lee
- Department of Immunology and Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Korea.,Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Samsung Medical Center, Sungkyunkwan University, Seoul, Korea.,Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, Korea
| |
Collapse
|
18
|
Bracic Tomazic S, Schatz C, Haybaeck J. Translational Regulation in Hepatocellular Carcinogenesis. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:4359-4369. [PMID: 34703211 PMCID: PMC8523516 DOI: 10.2147/dddt.s255582] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 10/04/2021] [Indexed: 12/17/2022]
Abstract
The mortality of hepatocellular carcinoma (HCC) is distributed unevenly worldwide. One of the major causes is hepatitis B or hepatitis C virus infection and the development and progression of liver cirrhosis. The carcinogenesis of HCC is among others regulated via the mTOR (mechanistic target of rapamycin) signaling pathway and represents a possible method of targeted treatment. The aim of our article was to address the most recent clinical advances and findings of basic studies on the mTOR signaling pathway and the involved factors. Risk factors play a key role in dysregulation of the signaling pathway, where both mTORCs are upregulated and protein synthesis is altered. eIFs and, to a lesser extent, eEFs play an essential role in this process. Whether the factor will be upregulated or downregulated, among others, depends on hepatitis B/C virus infection. The amount of a particular factor in a patient sample lets us know whether HCC recurrence will occur, what is the likelihood of chemoresistance, and what outcome is predicted for patients with an increased value. Our analysis shows that in addition to mTOR, eIF3, eIF4, and eIF5 play an important role, as they can serve as biomarkers for non- and virus-related HCC.
Collapse
Affiliation(s)
- Suzana Bracic Tomazic
- Department of Pathology, Hospital Graz II, Graz, 8020, Austria.,Faculty of Medicine, University of Maribor, Maribor, 2000, Slovenia
| | - Christoph Schatz
- Institute of Pathology, Neuropathology and Molecular Pathology, Medical University of Innsbruck, Innsbruck, 6020, Austria
| | - Johannes Haybaeck
- Institute of Pathology, Neuropathology and Molecular Pathology, Medical University of Innsbruck, Innsbruck, 6020, Austria.,Diagnostic & Research Center for Molecular BioMedicine, Institute of Pathology, Medical University Graz, Graz, 8010, Austria
| |
Collapse
|
19
|
Wang X, Wei Z, Jiang Y, Meng Z, Lu M. mTOR Signaling: The Interface Linking Cellular Metabolism and Hepatitis B Virus Replication. Virol Sin 2021; 36:1303-1314. [PMID: 34580816 PMCID: PMC8692646 DOI: 10.1007/s12250-021-00450-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 08/24/2021] [Indexed: 01/05/2023] Open
Abstract
Mammalian target of rapamycin (mTOR) is a conserved Ser/Thr kinase that includes mTOR complex (mTORC) 1 and mTORC2. The mTOR pathway is activated in viral hepatitis, including hepatitis B virus (HBV) infection-induced hepatitis. Currently, chronic HBV infection remains one of the most serious public health issues worldwide. The unavailability of effective therapeutic strategies for HBV suggests that clarification of the pathogenesis of HBV infection is urgently required. Increasing evidence has shown that HBV infection can activate the mTOR pathway, indicating that HBV utilizes or hijacks the mTOR pathway to benefit its own replication. Therefore, the mTOR signaling pathway might be a crucial target for controlling HBV infection. Here, we summarize and discuss the latest findings from model biology research regarding the interaction between the mTOR signaling pathway and HBV replication.
Collapse
Affiliation(s)
- Xueyu Wang
- Department of Infectious Diseases, The Second Xiangya Hospital, Central South University, Changsha, 410011, China.,Institute of Virology, University Hospital Essen, University of Duisburg-Essen, 45122, Essen, Germany
| | - Zhiqiang Wei
- Institute of Biomedical Research, Hubei Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China
| | - Yongfang Jiang
- Department of Infectious Diseases, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Zhongji Meng
- Institute of Biomedical Research, Hubei Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China. .,Department of Infectious Diseases, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China.
| | - Mengji Lu
- Institute of Virology, University Hospital Essen, University of Duisburg-Essen, 45122, Essen, Germany.
| |
Collapse
|
20
|
Helmy MA, Abdalla HA, Abd El Rahman HA, Ahmed DAM. Hepatotoxic effect of tramadol and O-desmethyltramadol in HepG2 cells and potential role of PI3K/AKT/mTOR. Xenobiotica 2021; 51:1029-1037. [PMID: 34319855 DOI: 10.1080/00498254.2021.1961919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
1. The aim of this study was to compare the in vitro cytotoxic effect of tramadol and M1 metabolite in HepG2 cell line, the underlying mechanism, and PI3K/AKT/mTOR as potential target.2. Concentrations representing therapeutic level for tramadol (2 µM) and M1 metabolite (0.5 µM) were used. In addition, other increasing concentrations representing higher toxic levels were used (6, 10 µM for tramadol and 1.5, 2.5 µM for M1 metabolites). Cytotoxicity was assessed at 24, 48 and 72 h.3. Both tramadol and M1 metabolites were able to produce cytotoxicity in a dose and time dependent manner. Insignificant difference was detected between cells exposed to tramadol and M1 metabolite at therapeutic concentrations. Tramadol-induced apoptotic and autophagic cell death while M1 metabolite-induced apoptosis only. For PI3K/AKT/mTOR pathway, the therapeutic concentration of tramadol was only able to increase phosphorylation of AKT while higher toxic concentrations were able to increase phosphorylation of whole pathway; Meanwhile, M1 metabolite was able to increase the phosphorylation of the whole pathway significantly in therapeutic and toxic concentrations.4. In conclusion, both tramadol and M1 are equally cytotoxic. Apoptosis and autophagy both mediate hepatic cell death. PI3K/AKT pathway is involved in apoptosis induction while autophagy is regulated through mTOR independent pathway.
Collapse
Affiliation(s)
- Manar A Helmy
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Hussein Abdelaziz Abdalla
- Department of Medical Biochemistry, Faculty of Medicine, Mansoura University, Mansoura, Egypt.,Department of Clinical Biochemistry and Molecular Medicine, Faculty of Medicine, Taibah University, Medina, Saudi Arabia
| | - Heba Allah Abd El Rahman
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Dalia Alsaied Moustafa Ahmed
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
21
|
de Mattos ÂZ, Debes JD, Boonstra A, Yang JD, Balderramo DC, Sartori GDP, de Mattos AA. Current impact of viral hepatitis on liver cancer development: The challenge remains. World J Gastroenterol 2021; 27:3556-3567. [PMID: 34239269 PMCID: PMC8240060 DOI: 10.3748/wjg.v27.i24.3556] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/11/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023] Open
Abstract
Chronic infections due to hepatitis B and hepatitis C viruses are responsible for most cases of hepatocellular carcinoma (HCC) worldwide, and this association is likely to remain during the next decade. Moreover, viral hepatitis-related HCC imposes an important burden on public health in terms of disability-adjusted life years. In order to reduce such a burden, some major challenges must be faced. Universal vaccination against hepatitis B virus, especially in the neonatal period, is probably the most relevant primary preventive measure against the development of HCC. Moreover, considering the large adult population already infected with hepatitis B and C viruses, it is also imperative to identify these individuals to ensure their access to treatment. Both hepatitis B and C currently have highly effective therapies, which are able to diminish the risk of development of liver cancer. Finally, it is essential for individuals at high-risk of HCC to be included in surveillance programs, so that tumors are detected at an early stage. Patients with hepatitis B or C and advanced liver fibrosis or cirrhosis benefit from being followed in a surveillance program. As hepatitis B virus is oncogenic and capable of leading to liver cancer even in individuals with early stages of liver fibrosis, other high-risk groups of patients with hepatitis B are also candidates for surveillance. Considerable effort is required concerning these strategies in order to decrease the incidence and the mortality of viral hepatitis-related HCC.
Collapse
MESH Headings
- Adult
- Carcinoma, Hepatocellular/epidemiology
- Carcinoma, Hepatocellular/etiology
- Carcinoma, Hepatocellular/prevention & control
- Hepatitis B/complications
- Hepatitis B/epidemiology
- Hepatitis B/prevention & control
- Hepatitis B virus
- Hepatitis B, Chronic/complications
- Hepatitis B, Chronic/epidemiology
- Hepatitis, Viral, Human/complications
- Hepatitis, Viral, Human/epidemiology
- Humans
- Infant, Newborn
- Liver Neoplasms/epidemiology
- Liver Neoplasms/prevention & control
- Risk Factors
Collapse
Affiliation(s)
- Ângelo Zambam de Mattos
- Department of Gastroenterology and Hepatology, Federal University of Health Sciences of Porto Alegre, Porto Alegre 90020-090, Brazil
- Gastroenterology and Hepatology Unit, Irmandade Santa Casa de Misericórdia de Porto Alegre, Porto Alegre 90050-170, Brazil
| | - Jose D Debes
- Department of Medicine, Division of Gastroenterology and Infectious Diseases, University of Minnesota, Minneapolis, MN 55455, United States
- Department of Gastroenterology and Hepatology, Erasmus Medical Center, Rotterdam NL-3015, Netherlands
| | - Andre Boonstra
- Department of Gastroenterology and Hepatology, Erasmus Medical Center, Rotterdam NL-3015, Netherlands
| | - Ju-Dong Yang
- Division of Digestive and Liver Diseases, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States
| | - Domingo C Balderramo
- Department of Gastroenterology, Hospital Privado Universitario de Córdoba, Córdoba 5016, Argentina
- Department of Medicine, Instituto Universitario de Ciencias Biomédicas de Córdoba, Córdoba 5016, Argentina
| | - Giovana D P Sartori
- Department of Internal Medicine, Hospital Nossa Senhora da Conceição, Porto Alegre 91350-200, Brazil
| | - Angelo Alves de Mattos
- Department of Gastroenterology and Hepatology, Federal University of Health Sciences of Porto Alegre, Porto Alegre 90020-090, Brazil
- Gastroenterology and Hepatology Unit, Irmandade Santa Casa de Misericórdia de Porto Alegre, Porto Alegre 90050-170, Brazil
| |
Collapse
|
22
|
Guo H, Fan Q. Identification of the HMMR Gene as a Diagnostic and Prognostic Biomarker in Hepatocellular Carcinoma Based on Integrated Bioinformatics Analysis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:5970085. [PMID: 34221079 PMCID: PMC8221880 DOI: 10.1155/2021/5970085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/02/2021] [Accepted: 06/08/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND We aimed to investigate the expression of the hyaluronan-mediated motility receptor (HMMR) gene in hepatocellular carcinoma (HCC) and nonneoplastic tissues and to investigate the diagnostic and prognostic value of HMMR. METHOD With the reuse of the publicly available The Cancer Genome Atlas (TCGA) data, 374 HCC patients and 50 nonneoplastic tissues were used to investigate the diagnostic and prognostic values of HMMR genes by receiver operating characteristic (ROC) curve analysis and survival analysis. All patients were divided into low- and high-expression groups based on the median value of HMMR expression level. Univariate and multivariate Cox regression analysis were used to identify prognostic factors. Gene set enrichment analysis (GSEA) was performed to explore the potential mechanism of the HMMR genes involved in HCC. The diagnostic and prognostic values were further validated in an external cohort from the International Cancer Genome Consortium (ICGC). RESULTS HMMR mRNA expression was significantly elevated in HCC tissues compared with that in normal tissues from both TCGA and the ICGC cohorts (all P values <0.001). Increased HMMR expression was significantly associated with histologic grade, pathological stage, and survival status (all P values <0.05). The area under the ROC curve for HMMR expression in HCC and normal tissues was 0.969 (95% CI: 0.948-0.983) in the TCGA cohort and 0.956 (95% CI: 0.932-0.973) in the ICGC cohort. Patients with high HMMR expression had a poor prognosis than patients with low expression group in both cohorts (all P < 0.001). Univariate and multivariate analysis also showed that HMMR is an independent predictor factor associated with overall survival in both cohorts (all P values <0.001). GSEA showed that genes upregulated in the high-HMMR HCC subgroup were mainly significantly enriched in the cell cycle pathway, pathways in cancer, and P53 signaling pathway. CONCLUSION HMMR is expressed at high levels in HCC. HMMR overexpression may be an unfavorable prognostic factor for HCC.
Collapse
Affiliation(s)
- Honglan Guo
- Department of Gastroenterology, The First People's Hospital of Chenzhou City, Chenzhou 423000, Hunan, China
| | - Qinqiao Fan
- Department of Hepatobiliary, The First People's Hospital of Chenzhou City, Chenzhou 423000, Hunan, China
| |
Collapse
|
23
|
Lei Y, Xu X, Liu H, Chen L, Zhou H, Jiang J, Yang Y, Wu B. HBx induces hepatocellular carcinogenesis through ARRB1-mediated autophagy to drive the G 1/S cycle. Autophagy 2021; 17:4423-4441. [PMID: 33866937 DOI: 10.1080/15548627.2021.1917948] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The hepatitis B virus X protein (HBx) is involved in the process of hepatocellular carcinoma via the activation of various oncogenes. Our previous study indicated that ARBB1 (arrestin beta 1) promotes hepatocellular carcinogenesis (HCC). However, the role of ARRB1 in HBx-related HCC remains unclear. Herein, we identified that ARRB1 was upregulated by HBx in vivo and in vitro. Arrb1 deficiency suppressed HBx-induced hepatocellular carcinogenesis in several mouse models. Furthermore, knockdown of ARRB1 blocked HBx-induced macroautophagic/autophagic flux and disrupted the formation of autophagosomes. ARRB1 interacted with HBx, and the autophagic core protein MAP1LC3/LC3, a scaffolding protein, was essential for complete autophagy. Inhibition of autophagy by 3-methyladenine or interference of ATG5 or ATG7 attenuated HBx-induced cell cycle acceleration and the subsequent proliferative response via the induction of G1/S arrest. The absence of autophagy abolished the phosphorylation of CDK2 and the activity of the CDK2-CCNE1 complex. Our results demonstrate that ARRB1 plays a critical role in HBV-related HCC via modulating autophagy and the CDKN1B-CDK2-CCNE1-E2F1 axis and indicate that ARRB1 may be a potential therapeutic target for HCC.
Collapse
Affiliation(s)
- Yiming Lei
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China.,Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, Guangdong Province, China
| | - Xuan Xu
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China.,Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, Guangdong Province, China
| | - Huiling Liu
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China.,Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, Guangdong Province, China
| | - Lingjun Chen
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China.,Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, Guangdong Province, China
| | - Haoxiong Zhou
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China.,Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, Guangdong Province, China
| | - Jie Jiang
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China.,Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, Guangdong Province, China
| | - Yidong Yang
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China.,Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, Guangdong Province, China
| | - Bin Wu
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China.,Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, Guangdong Province, China
| |
Collapse
|
24
|
Li H, Huang H, Li S, Mei H, Cao T, Lu Q. Long non-coding RNA ADAMTS9-AS2 inhibits liver cancer cell proliferation, migration and invasion. Exp Ther Med 2021; 21:559. [PMID: 33850531 PMCID: PMC8027749 DOI: 10.3892/etm.2021.9991] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 07/21/2020] [Indexed: 12/14/2022] Open
Abstract
Long non-coding RNA (lncRNA) ADAM metallopeptidase with thrombospondin type 1 motif 9 antisense RNA 2 (ADAMTS9-AS2) is involved in various types of cancer, such as ovarian cancer, lung cancer and clear cell renal cell carcinoma. However, the roles of ADAMTS9-AS2 in liver cancer are not completely understood. The present study aimed to determine the functional role of ADAMTS9-AS2 in human liver cancer and investigate the potential underlying molecular mechanisms. The expression levels of ADAMTS9-AS2 and ADAMTS9 were determined following ADAMTS9-AS2 overexpression and knockdown. The results indicated that ADAMTS9-AS2 overexpression and knockdown increased and decreased ADAMTS9 mRNA and protein expression levels, respectively, indicating that alterations in ADAMTS9 expression corresponded with ADAMTS9-AS2 expression. Subsequently, the effects of ADAMTS9-AS2 on liver cancer cell proliferation, migration and invasion were analyzed by performing Cell Counting Kit-8, wound healing and Transwell assays, respectively. The results demonstrated that ADAMTS9-AS2 inhibited liver cancer cell proliferation, migration and invasion. Finally, the effect of ADAMTS9 on PI3K/AKT/mTOR signaling pathway-associated proteins [AKT, phosphorylated-AKT, phosphatidylinositol-4, 5-bisphosphate 3-kinase catalytic subunit β (PIK3CB), mTOR and phosphorylated-mTOR], several key autophagy-related proteins [light chain 3-I/II (LC3-I/II), beclin 1 (BECN1) and sequestosome 1 (SQSTM1)] and apoptosis-related proteins (Bax and Bcl-2) was detected via western blotting. The results suggested that ADAMTS9-AS2 downregulated the phosphorylation of AKT and mTOR, the protein expression level of PIK3CB, as well as the expression levels of autophagy protein SQSTM1 and antiapoptotic protein Bcl-2. By contrast, ADAMTS9-AS2 upregulated the expression levels of autophagy proteins LC3-II and BECN1, and the proapoptotic protein Bax. Collectively, ADAMTS9-AS2 inhibited liver cancer cell proliferation, migration and invasion via inhibiting the PI3K/AKT/mTOR signaling pathway. The present study provided a novel insight into the role of ADAMTS9-AS2 in liver cancer.
Collapse
Affiliation(s)
- Hanjun Li
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China.,Department of Pancreatic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Hu Huang
- Cancer Center, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing 400042, P.R. China.,Department of Oncology, The People's Liberation Army No. 161 Hospital, Wuhan, Hubei 430010, P.R. China
| | - Sha Li
- Department of Anesthesiology, General Hospital of Central Theater Command of The People's Liberation Army, Wuhan, Hubei 430070, P.R. China
| | - Hongliang Mei
- Department of General Surgery, General Hospital of Central Theater Command of The People's Liberation Army, Wuhan, Hubei 430070, P.R. China
| | - Tingjia Cao
- Department of General Surgery, General Hospital of Central Theater Command of The People's Liberation Army, Wuhan, Hubei 430070, P.R. China
| | - Qiping Lu
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China.,Department of General Surgery, General Hospital of Central Theater Command of The People's Liberation Army, Wuhan, Hubei 430070, P.R. China
| |
Collapse
|
25
|
Yang G, Xia X, Zhong H, Shen J, Li S. Protective Effect of Tangeretin and 5-Hydroxy-6,7,8,3',4'-Pentamethoxyflavone on Collagen-Induced Arthritis by Inhibiting Autophagy via Activation of the ROS-AKT/mTOR Signaling Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:259-266. [PMID: 33372513 DOI: 10.1021/acs.jafc.0c06801] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by long duration and repeated relapse. This study explored the preventive effect of tangeretin (TAN) and 5-hydroxy-6,7,8,3',4'-pentamethoxyflavone (5-HPMF) on RA, and the underlying molecular mechanism based on a rat model stimulated by bovine type II collagen (BIIC). After the intervention of TAN or 5-HPMF (TAN/5-HPMF) for 5 weeks, the RA lesions and autophagy levels of the synovial tissue were significantly reduced, and the ROS content and HO-1 expression level were down-regulated simultaneously. The relative expression levels of p-AKT and p-mTOR were down-regulated after TAN/5-HPMF feeding. Meanwhile, the relative expression level of p62 increased by more than two-fold for TAN/5-HPMF treated rats at 200 mg/kg BW comparing with those in BIIC group. Results of immunofluorescence staining and Western blotting further confirmed that TAN/5-HPMF treatment reduced BIIC-induced conversion from LC3I to LC3II. Observations under transmission electron microscope also demonstrated that the autophagy level was reduced upon TAN/5-HPMF intervention. Collectively, these results revealed that TAN and 5-HPMF prevented the pathological process of BIIC-stimulated arthritis through inhibiting the autophagy of synovial cells, achieved via the ROS-AKT/mTOR signal axis. Thus, our findings confirmed the protective potential of TAN and 5-HPMF for RA disease.
Collapse
Affiliation(s)
- Guliang Yang
- National Engineering Laboratory for Rice and Byproducts Processing, Food Science and Engineering College, Central South University of Forestry and Technology, Changsha, Hunan 410004, P. R. China
| | - Xinxin Xia
- National Engineering Laboratory for Rice and Byproducts Processing, Food Science and Engineering College, Central South University of Forestry and Technology, Changsha, Hunan 410004, P. R. China
| | - Haiyan Zhong
- National Engineering Laboratory for Rice and Byproducts Processing, Food Science and Engineering College, Central South University of Forestry and Technology, Changsha, Hunan 410004, P. R. China
| | - Junfeng Shen
- Hubei Key Laboratory of EFGIR, Huanggang Normal University, Huanggang, Hubei 438000, P. R. China
| | - Shiming Li
- Hubei Key Laboratory of EFGIR, Huanggang Normal University, Huanggang, Hubei 438000, P. R. China
| |
Collapse
|
26
|
Niture S, Gyamfi MA, Lin M, Chimeh U, Dong X, Zheng W, Moore J, Kumar D. TNFAIP8 regulates autophagy, cell steatosis, and promotes hepatocellular carcinoma cell proliferation. Cell Death Dis 2020; 11:178. [PMID: 32152268 PMCID: PMC7062894 DOI: 10.1038/s41419-020-2369-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 02/07/2023]
Abstract
Tumor necrosis factor-α-induced protein 8 (TNFAIP8) expression has been linked to tumor progression in various cancer types, but the detailed mechanisms of TNFAIP8 are not fully elucidated. Here we define the role of TNFAIP8 in early events associated with development of hepatocellular carcinoma (HCC). Increased TNFAIP8 levels in HCC cells enhanced cell survival by blocking apoptosis, rendering HCC cells more resistant to the anticancer drugs, sorafenib and regorafenib. TNFAIP8 also induced autophagy and steatosis in liver cancer cells. Consistent with these observations, TNFAIP8 blocked AKT/mTOR signaling and showed direct interaction with ATG3-ATG7 proteins. TNFAIP8 also exhibited binding with fatty acids and modulated expression of lipid/fatty-acid metabolizing enzymes. Chronic feeding of mice with alcohol increased hepatic levels of TNFAIP8, autophagy, and steatosis but not in high-fat-fed obese mice. Similarly, higher TNFAIP8 expression was associated with steatotic livers of human patients with a history of alcohol use but not in steatotic patients with no history of alcohol use. Our data indicate a novel role of TNFAIP8 in modulation of drug resistance, autophagy, and hepatic steatosis, all key early events in HCC progression.
Collapse
Affiliation(s)
- Suryakant Niture
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, Durham, NC, 27707, USA
| | - Maxwell A Gyamfi
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, Durham, NC, 27707, USA
| | - Minghui Lin
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, Durham, NC, 27707, USA
- Ningxia Medical University, Ningxia Hui Autonomous Region, Yinchuan, 750004, China
| | - Uchechukwu Chimeh
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, Durham, NC, 27707, USA
| | - Xialan Dong
- Department of Pharmaceutical Sciences, Bio-manufacturing Research Institute and Technology Enterprise (BRITE), North Carolina Central University Durham, Durham, NC, 27707, USA
| | - Weifan Zheng
- Department of Pharmaceutical Sciences, Bio-manufacturing Research Institute and Technology Enterprise (BRITE), North Carolina Central University Durham, Durham, NC, 27707, USA
| | - John Moore
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, Durham, NC, 27707, USA
| | - Deepak Kumar
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, Durham, NC, 27707, USA.
- Department of Pharmaceutical Sciences, North Carolina Central University Durham, Durham, NC, 27707, USA.
| |
Collapse
|
27
|
Protective effects of morin against acrylamide-induced hepatotoxicity and nephrotoxicity: A multi-biomarker approach. Food Chem Toxicol 2020; 138:111190. [PMID: 32068001 DOI: 10.1016/j.fct.2020.111190] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 02/07/2020] [Accepted: 02/11/2020] [Indexed: 01/02/2023]
Abstract
Acrylamide (ACR) is a heat-induced carcinogen substance that is found in some foods due to cooking or other thermal processes. The aim of present study was to assess the probable protective effects of morin against ACR-induced hepatorenal toxicity in rats. The rats were treated with ACR (38.27 mg/kg b.w., p.o.) alone or with morin (50 and 100 mg/kg b.w., p.o.) for 10 consecutive days. Morin treatment attenuated the ACR-induced liver and kidney tissue injury by diminishing the serum AST, ALP, ALT, urea and creatinine levels. Morin increased activities of SOD, CAT and GPx and levels of GSH, and suppressed lipid peroxidation in ACR induced tissues. Histopathological changes and immunohistochemical expressions of p53, EGFR, nephrin and AQP2 in the ACR-induced liver and kidney tissues were decreased after administration of morin. In addition, morin reversed the changes in levels of apoptotic, autophagic and inflammatory parameters such as caspase-3, bax, bcl-2, cytochrome c, beclin-1, LC3A, LC3B, p38α MAPK, NF-κB, IL-1β, IL-6, TNF-α and COX-2 in the ACR-induced toxicity. Morin also affected the protein levels by regulating the PI3K/Akt/mTOR signaling pathway and thus alleviated ACR-induced apoptosis and autophagy. Overall, these findings may shed some lights on new approaches for the treatment of ACR-induced hepatotoxicity and nephrotoxicity.
Collapse
|
28
|
He P, Lei J, Miao JN, Wu D, Wang C. Cordyceps sinensis attenuates HBx‑induced cell apoptosis in HK‑2 cells through suppressing the PI3K/Akt pathway. Int J Mol Med 2020; 45:1261-1269. [PMID: 32124952 DOI: 10.3892/ijmm.2020.4503] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 01/16/2020] [Indexed: 11/06/2022] Open
Abstract
The authors' previous studies demonstrated that the major renal damage from hepatitis B virus infection is HBx‑induced apoptosis of renal tubular epithelial cells. Cordyceps sinensis is one of the most valuable of traditional Chinese medicines and is extensively used to treat chronic renal diseases. However, there is no research on the potential renal protective effect of C. sinensis on HBx‑induced apoptosis of renal tubular cells. The protective effect and underlying mechanism of C. sinensis were examined using a renal tubular epithelial cell line stably overexpressing HBx. HK‑2 cells were stably transfected with pCMV‑HBx to establish HBx‑overexpression in an in vitro cell model and HK‑2 cells transfected with an empty vector were generated as a control. The effect of C. sinensis on cell proliferation and apoptosis, the phosphatidylinositol‑3‑kinase (PI3K)/protein kinase B (Akt) signaling pathway, and the enzyme activity of caspase‑3 and caspase‑9 was measured. The present study demonstrated that HBx transfection inhibited cell proliferation; increased apoptosis, caspase‑3 and caspase‑9 activity; and increased the activity of the PI3K/Akt pathway. Treatment with C. sinensis attenuated all of these HBx‑induced responses. HBx triggered apoptosis and activated the PI3K/Akt signaling pathway in HK‑2 cells. C. sinensis treatment significantly attenuated the effect of HBx, at least in part by suppressing the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Ping He
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Jing Lei
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Jia-Ning Miao
- Medical Research Center, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Di Wu
- Medical Research Center, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Cheng Wang
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| |
Collapse
|
29
|
Hepatitis Delta Virus Alters the Autophagy Process To Promote Its Genome Replication. J Virol 2020; 94:JVI.01936-19. [PMID: 31748400 DOI: 10.1128/jvi.01936-19] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 11/15/2019] [Indexed: 02/07/2023] Open
Abstract
A substantial number of viruses have been demonstrated to subvert autophagy to promote their own replication. Recent publications have reported the proviral effect of autophagy induction on hepatitis B virus (HBV) replication. Hepatitis delta virus (HDV) is a defective virus and an occasional obligate satellite of HBV. However, no previous work has studied the relationship between autophagy and HDV. In this article, we analyze the impact of HBV and HDV replication on autophagy as well as the involvement of the autophagy machinery in the HDV life cycle when produced alone and in combination with HBV. We prove that HBxAg and HBsAg can induce early steps of autophagy but ultimately block flux. It is worth noting that the two isoforms of the HDV protein, the small HDAg (S-HDAg) and large HDAg (L-HDAg) isoforms, can also efficiently promote autophagosome accumulation and disturb autophagic flux. Using CRISPR-Cas9 technology to generate specific knockouts, we demonstrate that the autophagy machinery, specifically the proteins implicated in the elongation step (ATG7, ATG5, and LC3), is important for the release of HBV without affecting the level of intracellular HBV genomes. Surprisingly, the knockout of ATG5 and ATG7 decreased the intracellular HDV RNA level in both Huh7 and HepG2.2.15 cells without an additional effect on HDV secretion. Therefore, we conclude that HBV and HDV have evolved to utilize the autophagy machinery so as to assist at different steps of their life cycle.IMPORTANCE Hepatitis delta virus is a defective RNA virus that requires hepatitis B virus envelope proteins (HBsAg) to fulfill its life cycle. Thus, HDV can only infect individuals at the same time as HBV (coinfection) or superinfect individuals who are already chronic carriers of HBV. The presence of HDV in the liver accelerates the progression of infection to fibrosis and to hepatic cancer. Since current treatments against HBV are ineffective against HDV, it is of paramount importance to study the interaction between HBV, HDV, and host factors. This will help unravel new targets whereby a therapy that is capable of simultaneously impeding both viruses could be developed. In this research paper, we evidence that the autophagy machinery promotes the replication of HBV and HDV at different steps of their life cycle. Notwithstanding their contribution to HBV release, autophagy proteins seem to assist HDV intracellular replication but not its secretion.
Collapse
|
30
|
Wang Q, Cheng ST, Chen J. HBx mediated Increase of SIRT1 Contributes to HBV-related Hepatocellular Carcinoma Tumorigenesis. Int J Med Sci 2020; 17:1783-1794. [PMID: 32714081 PMCID: PMC7378664 DOI: 10.7150/ijms.43491] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 05/26/2020] [Indexed: 12/16/2022] Open
Abstract
Objective: Hepatocellular carcinoma (HCC) is one of the main causes of cancer-related deaths worldwide, and chronic hepatitis B virus (HBV) infection is strongly associated with HCC development, but the pathogenesis of HBV-related HCC remains obscure. Sirtuin 1 (SIRT1) has been implicated to enhance the replication of HBV and to promote the tumorigenesis of HCC. In this study, we aim to investigate the functional role of SIRT1 on HBV viral protein and HBV-induced HCC. Methods: Tumorous liver tissues from patient diagnosed with HBV-related HCC were collected and further divided into two groups (with or without metastasis). Then, the mRNA and protein level of SIRT1 in those tissues were detected by real time PCR and Western blot, respectively. Meanwhile, the protein level of epithelial-mesenchymal transition (EMT) relative markers in those tissues was determined by Western blot. Furthermore, the expression of SIRT1 in HBV-expressing HCC cells was examined. Next, the relationship between viral proteins and SIRT1 expression were determined by real time PCR and Western blot. In addition, the potential role of HBx-upregulated SIRT1 in HCC proliferation, migration and invasion were analyzed by cell viability assays, cell proliferation assay, wound healing assay, transwell assay and Western blot. Results: In this study, we found that the expression of SIRT1 was obviously increased in patients with metastasis compared to the patients without metastasis. Consistently, the expression of SIRT1 was also upregulated in HBV-expressing HCC cells compared to the controls. Further investigation showed that viral protein HBx was responsible for the elevated SIRT1 in HBV-expressing HCC cells. Meanwhile, the expression of HBx could be upregulated by SIRT1. Additionally, functional studies showed that HBx-elevated SIRT1 could promote HCC cell proliferation, migration and invasion. Importantly, HBx induced HCC proliferation and migration could be suppressed by Nicotinamide in a dose dependent manner. Conclusions: Our findings uncovered the positive role of SIRT1 in HBx-mediated tumorigenesis which implicated the potential role of SIRT1 in HBV-related HCC treatment.
Collapse
Affiliation(s)
- Qing Wang
- Key Laboratory of Molecular Biology for Infectious Diseases, Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| | - Sheng-Tao Cheng
- Key Laboratory of Molecular Biology for Infectious Diseases, Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| | - Juan Chen
- Key Laboratory of Molecular Biology for Infectious Diseases, Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| |
Collapse
|
31
|
Wang H, Liu Y, Wang D, Xu Y, Dong R, Yang Y, Lv Q, Chen X, Zhang Z. The Upstream Pathway of mTOR-Mediated Autophagy in Liver Diseases. Cells 2019; 8:E1597. [PMID: 31835352 PMCID: PMC6953127 DOI: 10.3390/cells8121597] [Citation(s) in RCA: 177] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 11/29/2019] [Accepted: 12/03/2019] [Indexed: 12/11/2022] Open
Abstract
Autophagy, originally found in liver experiments, is a cellular process that degrades damaged organelle or protein aggregation. This process frees cells from various stress states is a cell survival mechanism under stress stimulation. It is now known that dysregulation of autophagy can cause many liver diseases. Therefore, how to properly regulate autophagy is the key to the treatment of liver injury. mechanistic target of rapamycin (mTOR)is the core hub regulating autophagy, which is subject to different upstream signaling pathways to regulate autophagy. This review summarizes three upstream pathways of mTOR: the phosphoinositide 3-kinase (PI3K)/protein kinase (AKT) signaling pathway, the adenosine monophosphate-activated protein kinase (AMPK) signaling pathway, and the rat sarcoma (Ras)/rapidly accelerated fibrosarcoma (Raf)/mitogen-extracellular activated protein kinase kinase (MEK)/ extracellular-signal-regulated kinase (ERK) signaling pathway, specifically explored their role in liver fibrosis, hepatitis B, non-alcoholic fatty liver, liver cancer, hepatic ischemia reperfusion and other liver diseases through the regulation of mTOR-mediated autophagy. Moreover, we also analyzed the crosstalk between these three pathways, aiming to find new targets for the treatment of human liver disease based on autophagy.
Collapse
Affiliation(s)
- Haojie Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; (H.W.); (Y.X.); (R.D.); (Y.Y.); (Q.L.); (X.C.)
| | - Yumei Liu
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; (H.W.); (Y.X.); (R.D.); (Y.Y.); (Q.L.); (X.C.)
| | - Dongmei Wang
- College of Medical, Henan University of Science and Technology, Luoyang 471000, China;
| | - Yaolu Xu
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; (H.W.); (Y.X.); (R.D.); (Y.Y.); (Q.L.); (X.C.)
| | - Ruiqi Dong
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; (H.W.); (Y.X.); (R.D.); (Y.Y.); (Q.L.); (X.C.)
| | - Yuxiang Yang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; (H.W.); (Y.X.); (R.D.); (Y.Y.); (Q.L.); (X.C.)
| | - Qiongxia Lv
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; (H.W.); (Y.X.); (R.D.); (Y.Y.); (Q.L.); (X.C.)
| | - Xiaoguang Chen
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; (H.W.); (Y.X.); (R.D.); (Y.Y.); (Q.L.); (X.C.)
| | - Ziqiang Zhang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; (H.W.); (Y.X.); (R.D.); (Y.Y.); (Q.L.); (X.C.)
| |
Collapse
|
32
|
Abdoli A, Nakhaie M, Feizi N, Salimi Jeda A, Ramezani A. Harmonized Autophagy Versus Full-Fledged Hepatitis B Virus: Victorious or Defeated. Viral Immunol 2019; 32:322-334. [PMID: 31483214 DOI: 10.1089/vim.2019.0042] [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] [Indexed: 12/17/2022] Open
Abstract
Autophagy is a finely tuned process in the regulation of innate immunity to avoid excessive inflammatory responses and inflammasome signaling. In contrast, the results of recent studies have shown that autophagy may disease-dependently contribute to the pathogenesis of liver diseases, such as fibrosis, cirrhosis, and hepatocellular carcinoma (HCC) during hepatitis B virus (HBV) infection. HBV has learned to subvert the cell's autophagic machinery to promote its replication. Given the great impact of the autophagy mechanism on the HBV infection and HCC, recognizing these factors may be offered new hope for human intervention and treatment of chronic HBV. This review focuses on recent findings viewing the dual role of autophagy plays in the pathogenesis of HBV infected hepatocytes.
Collapse
Affiliation(s)
- Asghar Abdoli
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Mohsen Nakhaie
- Department of Virology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Neda Feizi
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
| | - Ali Salimi Jeda
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amitis Ramezani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
33
|
Cui C, Li Z, Wu D. The long non-coding RNA H19 induces hypoxia/reoxygenation injury by up-regulating autophagy in the hepatoma carcinoma cells. Biol Res 2019; 52:32. [PMID: 31196153 PMCID: PMC6567522 DOI: 10.1186/s40659-019-0239-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 05/25/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Long non-coding RNA H19 (H19) plays an important role by regulating protein expression in different tissues and organs of the body. However, whether H19 induces hypoxia/reoxygenation (h/R) injury via increase of autophagy in the hepatoma carcinoma cells is unknown. RESULTS H19 was expressed in the hepatoma carcinoma cells (Hep G2 and HCCLM3 cells) and its expression was most in 8 h/24R. The knockdown of H19 and 3-MA (an autophagy inhibitor) protected against h/R-induced apoptosis, cell damage, the expression of cleaved caspase-3 and cleaved caspase-9, the release of cytochrome c (Cyt c). The knockdown of H19 and 3-MA also decreased the autophagic vesicles (AVs) and the expression of Beclin-1 and the ration of LC3-II/LC3-I, and increased cell viability, the expression of Bcl-2 and P62 and the phosphorylation of PI3K, Akt and mTOR. In addition, chloroquine (CQ, an inhibitor of autophagy flux) markedly decreased formation of autophagy flux (the ration of LC3-II/LC3-I). The results of the knockdown of H19 group were similar to those of the 3-MA (or CQ) group. Rapamycin (a mTOR inhibitor, an autophagy activator) further down-regulated h/R-induced decrease of the phosphorylated PI3K, Akt and mTOR. The knockdown of H19 cancelled the effect of rapamycin. The overexpression of H19 further expanded h/R-induced increase of the ration of LC3-II/LC3-I and decrease of the phosphorylated PI3K, Akt and mTOR. CONCLUSIONS Our results suggest that the long non-coding RNA H19 induces h/R injury by up-regulation of autophagy via activation of PI3K-Akt-mTOR pathway in the hepatoma carcinoma cells.
Collapse
Affiliation(s)
- Chao Cui
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Baojian Road, Harbin, 150086, China
| | - Zhiyu Li
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Baojian Road, Harbin, 150086, China
| | - Dequan Wu
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Baojian Road, Harbin, 150086, China.
| |
Collapse
|
34
|
Dhar SK, Batinic-Haberle I, St Clair DK. UVB-induced inactivation of manganese-containing superoxide dismutase promotes mitophagy via ROS-mediated mTORC2 pathway activation. J Biol Chem 2019; 294:6831-6842. [PMID: 30858178 DOI: 10.1074/jbc.ra118.006595] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 03/05/2019] [Indexed: 12/23/2022] Open
Abstract
Mitochondria are major sites of energy metabolism that influence numerous cellular events, including immunity and cancer development. Previously, we reported that the mitochondrion-specific antioxidant enzyme, manganese-containing superoxide dismutase (MnSOD), has dual roles in early- and late-carcinogenesis stages. However, how defective MnSOD impacts the chain of events that lead to cell transformation in pathologically normal epidermal cells that have been exposed to carcinogens is unknown. Here, we show that UVB radiation causes nitration and inactivation of MnSOD leading to mitochondrial injury and mitophagy. In keratinocytes, exposure to UVB radiation decreased mitochondrial oxidative phosphorylation, increased glycolysis and the expression of autophagy-related genes, and enhanced AKT Ser/Thr kinase (AKT) phosphorylation and cell growth. Interestingly, UVB initiated a prosurvival mitophagy response by mitochondria-mediated reactive oxygen species (ROS) signaling via the mammalian target of the mTOR complex 2 (mTORC2) pathway. Knockdown of rictor but not raptor abrogated UVB-induced mitophagy responses. Furthermore, fractionation and proximity-ligation assays reveal that ROS-mediated mTOC2 activation in mitochondria is necessary for UVB-induced mitophagy. Importantly, pretreatment with the MnSOD mimic MnTnBuOE-2-PyP5+ (MnP) attenuates mTORC2 activation and suppresses UVB-induced mitophagy. UVB radiation exposure also increased cell growth as assessed by soft-agar colony survival and cell growth assays, and pretreatment with MnP or the known autophagy inhibitor 3-methyladenine abrogated UVB-induced cell growth. These results indicate that MnSOD is a major redox regulator that maintains mitochondrial health and show that UVB-mediated MnSOD inactivation promotes mitophagy and thereby prevents accumulation of damaged mitochondria.
Collapse
Affiliation(s)
- Sanjit K Dhar
- From the Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky 40536 and
| | - Ines Batinic-Haberle
- the Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina 27710
| | - Daret K St Clair
- From the Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky 40536 and
| |
Collapse
|
35
|
Di Yang M, Shen XB, Hu YS, Chen YY, Liu XH. Novel naphthalene-enoates: Design and anticancer activity through regulation cell autophagy. Biomed Pharmacother 2019; 113:108747. [PMID: 30849638 DOI: 10.1016/j.biopha.2019.108747] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 02/27/2019] [Indexed: 10/27/2022] Open
Abstract
Eleven dihydroxy-2-(1-hydroxy-4-methylpent-3-enyl)naphthalene derivatives as anticancer agents through regulating cell autophagy were designed and synthesized. The anticancer activity results indicated that most compounds manifested obvious un-toxic effect on GES-1 and L-02 with IC50 from 0.58 to 1.41 mM. Among them, (S,Z)-1-(5,8-dihydroxy-1,4-dioxo-1,4-dihydronaphthalen-2-yl)-4-methylpent-3-enyl 4-(3,4- dihydroisoquinolin-2(1 H)-yl)-4-oxobut-2-enoate (compound 4i) could induce cancer cells apoptosis. Further experiments showed that autophagy played an important role in the pro-apoptotic effect of this compound. Preliminary mechanism indicated that this compound could inhibit phosphoinositide 3-kinase/protein kinase B and the mammalian target of rapamycin (PI3K/AKT/mTOR) pathway by mediating apoptosis in an autophagy-dependent manner.
Collapse
Affiliation(s)
- Meng Di Yang
- School of Pharmacy, Anhui Medical University, Hefei, 230032, PR China
| | - Xiao Bao Shen
- School of Pharmacy, Anhui Medical University, Hefei, 230032, PR China
| | - Yang Sheng Hu
- School of Pharmacy, Anhui Medical University, Hefei, 230032, PR China
| | - Yan Yan Chen
- School of Pharmacy, Anhui Medical University, Hefei, 230032, PR China
| | - Xin Hua Liu
- School of Pharmacy, Anhui Medical University, Hefei, 230032, PR China; School of Material Science Chemical Engineering, ChuZhou University, ChuZhou, 239000, PR China.
| |
Collapse
|
36
|
Ke PY. Diverse Functions of Autophagy in Liver Physiology and Liver Diseases. Int J Mol Sci 2019; 20:E300. [PMID: 30642133 PMCID: PMC6358975 DOI: 10.3390/ijms20020300] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 01/05/2019] [Accepted: 01/08/2019] [Indexed: 01/09/2023] Open
Abstract
Autophagy is a catabolic process by which eukaryotic cells eliminate cytosolic materials through vacuole-mediated sequestration and subsequent delivery to lysosomes for degradation, thus maintaining cellular homeostasis and the integrity of organelles. Autophagy has emerged as playing a critical role in the regulation of liver physiology and the balancing of liver metabolism. Conversely, numerous recent studies have indicated that autophagy may disease-dependently participate in the pathogenesis of liver diseases, such as liver hepatitis, steatosis, fibrosis, cirrhosis, and hepatocellular carcinoma. This review summarizes the current knowledge on the functions of autophagy in hepatic metabolism and the contribution of autophagy to the pathophysiology of liver-related diseases. Moreover, the impacts of autophagy modulation on the amelioration of the development and progression of liver diseases are also discussed.
Collapse
Affiliation(s)
- Po-Yuan Ke
- Department of Biochemistry & Molecular Biology and Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan.
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan.
- Division of Allergy, Immunology, and Rheumatology, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan.
| |
Collapse
|
37
|
Wang X, Huo B, Liu J, Huang X, Zhang S, Feng T. Hepatitis B virus X reduces hepatocyte apoptosis and promotes cell cycle progression through the Akt/mTOR pathway in vivo. Gene 2019; 691:87-95. [PMID: 30630095 DOI: 10.1016/j.gene.2018.12.054] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 11/19/2018] [Accepted: 12/27/2018] [Indexed: 02/06/2023]
Abstract
Hepatitis B virus X (HBx), a viral onco-protein encoded by HBV, can promote oncogenesis of HCC. However, the mechanism of HBx in hepatocarcinogenesis is still unclear. In this study, we establish a new mouse model with normal immune system to investigate the role of HBx and its functional mechanisms under normal immune function. The animal model was established by injecting HBx-EGFP-14-19 cells into the hepatic portal vein of KM mice. To verify the mouse model, the expression of HBx in the liver tissue of mice was detected by qRT-PCR, western blotting and immunohistochemistry. The apoptosis index was calculated using the terminal deoxynucleotidyl transferase-dUTP nick-end labeling (TUNEL) assay, and the expression levels of apoptosis-related and cell cycle-related factors were measured. Moreover, expression of proteins in the protein kinase B/mammalian target of rapamycin (Akt/mTOR) signaling pathway was detected in HBx-EGFP-14-19 mice with and without use of an Akt inhibitor. The results showed the HBx was successfully overexpressed in liver of KM mice. After overexpressing HBx, the apoptosis index was downregulated in HBx-EGFP-14-19 liver tissue, and the expression levels of caspase-9 and Bad were reduced, but Bcl-xl was increased in HBx-EGFP-14-19 liver tissue. Overexpression of HBx increased the expression of the cyclin-dependent kinase 2 (CDK2), cyclinD1 and cyclinE. Moreover, compared with the low-level HBx group, p-Akt and p-mTOR were increased in the livers of mice with high levels of HBx. However, inactivation of apoptosis by overexpression of HBx was abolished by the treatment with an Akt inhibitor. These results indicate that HBx can induce anti-apoptosis mechanisms in hepatocytes in vivo, which is mediated by the Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Xue Wang
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China; Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing 400016, China
| | - Bennian Huo
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China; School of Pharmaceutical Science, Chongqing Medical University, Chongqing 400016, China
| | - Jie Liu
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China; School of Pharmaceutical Science, Chongqing Medical University, Chongqing 400016, China
| | - Xin Huang
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China; Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing 400016, China
| | - Siyao Zhang
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China; School of Pharmaceutical Science, Chongqing Medical University, Chongqing 400016, China
| | - Tao Feng
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China; School of Pharmaceutical Science, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
38
|
Niture S, Gyamfi MA, Kedir H, Arthur E, Ressom H, Deep G, Kumar D. Serotonin induced hepatic steatosis is associated with modulation of autophagy and notch signaling pathway. Cell Commun Signal 2018; 16:78. [PMID: 30409162 PMCID: PMC6225666 DOI: 10.1186/s12964-018-0282-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 10/09/2018] [Indexed: 01/08/2023] Open
Abstract
Background Besides its neurotransmitter and vasoconstriction functions, serotonin is an important mediator of numerous biological processes in peripheral tissues including cell proliferation, steatosis, and fibrogenesis. Recent reports indicate that serotonin may promote tumor growth in liver cancer, however, the molecular mechanisms remain elusive. n this study, we investigated the role and molecular signaling mechanisms mediated by serotonin in liver cancer cell survival, drug resistance, and steatosis. Methods Effect of serotonin on modulation of cell survival/proliferation was determined by MTT/WST1 assay. Effect of serotonin on the regulation of autophagy biomarkers and lipid/fatty acid proteins expression, AKT/mTOR and Notch signaling was evaluated by immunoblotting. The role of serotonin in normal human hepatocytes and liver cancer cell steatosis was analyzed by Oil Red O staining. The mRNA expression levels of lipid/fatty acid proteins and serotonin receptors were validated by qRT-PCR. The important roles of autophagy, Notch signaling, serotonin receptors and serotonin re-uptake proteins on serotonin-mediated cell steatosis were investigated by using selective inhibitors or antagonists. The association of peripheral serotonin, autophagy, and hepatic steatosis was also investigated using chronic EtOH fed mouse model. Results Exposure of liver cancer cells to serotonin induced Notch signaling and autophagy, independent of AKT/mTOR pathway. Also, serotonin enhanced cancer cell proliferation/survival and drug resistance. Furthermore, serotonin treatment up-regulated the expression of lipogenic proteins and increased steatosis in liver cancer cells. Inhibition of autophagy or Notch signaling reduced serotonin-mediated cell steatosis. Treatment with serotonin receptor antagonists 5-HTr1B and 5-HTr2B reduced serotonin-mediated cell steatosis; in contrast, treatment with selective serotonin reuptake inhibitors (SSRIs) increased steatosis. In addition, mice fed with chronic EtOH resulted in increased serum serotonin levels which were associated with the induction of hepatic steatosis and autophagy. Conclusions Serotonin regulates liver cancer cell steatosis, cells survival, and may promote liver carcinogenesis by activation of Notch signaling and autophagy. Electronic supplementary material The online version of this article (10.1186/s12964-018-0282-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Suryakant Niture
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, 1801 Fayetteville St, Durham, NC, 27707, USA
| | - Maxwell A Gyamfi
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, 1801 Fayetteville St, Durham, NC, 27707, USA
| | - Habib Kedir
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, 1801 Fayetteville St, Durham, NC, 27707, USA
| | - Elena Arthur
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, 1801 Fayetteville St, Durham, NC, 27707, USA
| | - Habtom Ressom
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, 20008, USA
| | - Gagan Deep
- Wake Forest Baptist Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC, 27109, USA
| | - Deepak Kumar
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, 1801 Fayetteville St, Durham, NC, 27707, USA. .,Department of Pharmaceutical Sciences, North Carolina Central University, Durham, NC, 27707, USA. .,Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, 20008, USA.
| |
Collapse
|
39
|
Sun W, Zhang J, Chen J. MicroRNA-520a suppresses HBV replication in HepG2.2.15 cells by inactivating AKT. J Int Med Res 2018; 46:4693-4704. [PMID: 30191752 PMCID: PMC6259377 DOI: 10.1177/0300060518792780] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Objective To investigate whether the mechanism by which a microRNA, miR-520a, suppresses the replication of hepatitis B virus (HBV) involves the regulation of the serine/threonine kinase (AKT) gene. Methods The effects of miR-520a on the proliferation, mitotic index and apoptosis of the HBV-replicating human hepatocellular carcinoma cell line HepG2.2.15 were measured using standard laboratory methods including flow cytometry. The effects of miR-520a on HBV transcription and replication were assessed using methods including immunoassays and reverse transcription–polymerase chain reaction. The effect of small interfering RNA (siRNA) to AKT on the levels of AKT mRNA and protein were also evaluated. Results In HepG2.2.15 cells, miRNA-520a reduced HBV transcription and replication by reducing AKT levels. MiRNA-520a decreased cell proliferation and mitosis entry of cells and increased apoptosis in HepG2.2.15 cells. AKT levels were reduced significantly by its siRNA, which resulted in suppression of HBV replication in HepG2.2.15 cells. Conclusions MiRNA-520a inhibited AKT gene expression and suppressed HBV transcription and replication. These findings suggest that miRNA-520a may be a novel target for the treatment of HBV infection because miRNA-520a reduced HepG2.2.15 cell survival and inhibited HBV replication associated with the AKT signalling pathway.
Collapse
Affiliation(s)
- Wei Sun
- 1 Department of Oncology, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Jinqian Zhang
- 2 Department of Laboratory Medicine, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong Province, China
| | - Jinglong Chen
- 1 Department of Oncology, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
40
|
Dhar SK, Bakthavatchalu V, Dhar B, Chen J, Tadahide I, Zhu H, Gao T, St Clair DK. DNA polymerase gamma (Polγ) deficiency triggers a selective mTORC2 prosurvival autophagy response via mitochondria-mediated ROS signaling. Oncogene 2018; 37:6225-6242. [PMID: 30038268 PMCID: PMC6265263 DOI: 10.1038/s41388-018-0404-z] [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: 11/06/2017] [Revised: 04/30/2018] [Accepted: 06/11/2018] [Indexed: 11/24/2022]
Abstract
Autophagy is a highly regulated evolutionarily conserved metabolic process induced by stress and energy deprivation. Here, we show that DNA polymerase gamma (Polγ) deficiency activates a selective prosurvival autophagic response via mitochondria-mediated reactive oxygen species (ROS) signaling and the mammalian target of rapamycin complex 2 (mTORC2) activities. In keratinocytes, Polγ deficiency causes metabolic adaptation that triggers cytosolic sensing of energy demand for survival. Knockdown of Polγ causes mitochondrial stress, decreases mitochondrial energy production, increases glycolysis, increases the expression of autophagy-associated genes, and enhances AKT phosphorylation and cell proliferation. Deficiency of Polγ preferentially activates mTORC2 formation to increase autophagy and cell proliferation, and knocking down Rictor abrogates these responses. Overexpression of Rictor, but not Raptor, reactivates autophagy in Polγ-deficient cells. Importantly, inhibition of ROS by a mitochondria-selective ROS scavenger abolishes autophagy and cell proliferation. These results identify Rictor as a critical link between mitochondrial stress, ROS, and autophagy. They represent a major shift in our understanding of the prosurvival role of the mTOR complexes and highlight mitochondria-mediated ROS as a prosurvival autophagy regulator during cancer development.
Collapse
Affiliation(s)
- Sanjit K Dhar
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, 40536, USA
| | - Vasudevan Bakthavatchalu
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Bithika Dhar
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, 40536, USA
| | - Jing Chen
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, 40536, USA
| | - Izumi Tadahide
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, 40536, USA
| | - Haining Zhu
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, 40536, USA
| | - Tianyan Gao
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, 40536, USA
| | - Daret K St Clair
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, 40536, USA.
| |
Collapse
|
41
|
The role of HBV-induced autophagy in HBV replication and HBV related-HCC. Life Sci 2018; 205:107-112. [DOI: 10.1016/j.lfs.2018.04.051] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 04/24/2018] [Accepted: 04/26/2018] [Indexed: 02/06/2023]
|
42
|
Li W, Gao C, Zhao L, Yuan Z, Chen Y, Jiang Y. Phthalimide conjugations for the degradation of oncogenic PI3K. Eur J Med Chem 2018; 151:237-247. [DOI: 10.1016/j.ejmech.2018.03.066] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Revised: 03/21/2018] [Accepted: 03/22/2018] [Indexed: 12/11/2022]
|
43
|
Wang X, Hassan W, Jabeen Q, Khan GJ, Iqbal F. Interdependent and independent multidimensional role of tumor microenvironment on hepatocellular carcinoma. Cytokine 2017; 103:150-159. [PMID: 29029799 DOI: 10.1016/j.cyto.2017.09.026] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 09/03/2017] [Accepted: 09/22/2017] [Indexed: 02/07/2023]
Abstract
The novelty of an effective therapeutic targeting for hepatocellular carcinoma (HCC) is based on improved understanding of each component of tumor microenvironment (TME) and its correspondent interactions at biological and molecular levels. In this context, new expansions for the treatment against TME and its communication with HCC are under exploration. Despite of the fact that blockage of growth factor receptors has become a treatment of choice in late phases of HCC in clinical practice, still a precise targeted treatment should address all the components of TME. Targeting one specific element out of cellular (cancer associated fibroblasts, endothelial cells, hepatic stellate cells, Kupffer cells and lymphocytes) or non-cellular (extracellular matrix, growth factors, inflammatory cytokines, proteolytic enzymes) parts of TME may not be a successful remedy for the disease because of well-designed hindrances of each component and their functional alternativeness. Meanwhile there are some elements of TME like epithelial-mesenchymal transition and CAF, which are considerably important and need thorough investigations. Ascertaining the potential role of these elements, and a single or combinational drug therapy targeting these elements of TME simultaneously, may provide the appreciable considerations to eventually improve in clinical practices and may also minimize the chances of reoccurrence of HCC.
Collapse
Affiliation(s)
- Xue Wang
- Jiangnan University, Wuxi Medical School, Wuxi 214122, China; China Pharmaceutical University, Department of Pharmacology, Nanjing 210009, China.
| | - Waseem Hassan
- Department of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan; Department of Pharmacy, The University of Lahore, Pakistan.
| | - Qaiser Jabeen
- Department of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan.
| | - Ghulam Jilany Khan
- China Pharmaceutical University, Department of Pharmacology, Nanjing 210009, China.
| | - Furqan Iqbal
- Department of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan.
| |
Collapse
|
44
|
X protein variants of the autochthonous Latin American hepatitis B virus F genotype promotes human hepatocyte death by the induction of apoptosis and autophagy. Virus Res 2017; 242:156-165. [PMID: 28986109 PMCID: PMC7114566 DOI: 10.1016/j.virusres.2017.09.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 09/29/2017] [Accepted: 09/30/2017] [Indexed: 02/07/2023]
Abstract
The impact of BCP mutations on HBV-X biologic activity was analyzed. Genotype F wild type and mutant HBV-X induce apoptosis of human hepatocytes. HBV-X variants modulate the expression of Bcl-2 family proteins. Subgenotypes F1b and F4 HBV-X and variants induce autophagy of human hepatocytes.
The hepatitis B virus X protein (HBV-X) is a multifunctional regulatory protein associated with the pathogenesis of liver disease in chronic HBV infection. Basal core promoter mutations (BCP), associated with the clinical course of chronic HBV infection, affect HBV-X at 130–131 positions. The role of these mutations on HBV-X biological activity remains largely unknown. The aim of this study was to analyze the impact of the presence of different amino acids at 130–131 positions of HBV-X on the biological activity of the protein. Transient expression of wild type and mutant F1b and F4 HBV-X increased cell mortality by the induction of apoptosis in human hepatoma cells. The wild type and mutant HBV-X differentially modulate the expression of pro-apoptotic (Bax) and anti-apoptotic (Bcl-2 and Bcl-X) regulatory proteins of the Bcl-2 family. Furthermore, the expression of HBV-X variants of both subgenotypes induced autophagy of human tumoral hepatocytes. In conclusion, HBV-X variants of the Latin American HBV F genotype promotes human hepatocytes death by the induction of apoptosis and autophagy. The results of this work describe some of the molecular mechanisms by which HBV-X variants contribute to the pathogenesis of liver diseases in the infected liver and help to the biological characterization of genotype F, responsible of the majority of HBV infections in Argentina.
Collapse
|
45
|
Gu H, Li L, Cui C, Zhao Z, Song G. Overexpression of let-7a increases neurotoxicity in a PC12 cell model of Alzheimer's disease via regulating autophagy. Exp Ther Med 2017; 14:3688-3698. [PMID: 29042965 DOI: 10.3892/etm.2017.4977] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 06/16/2017] [Indexed: 12/12/2022] Open
Abstract
Increased deposition of β-amyloid (Aβ) protein is one of the typical characteristics of Alzheimer's disease (AD). Recent evidence has demonstrated that the microRNA let-7 family, which is highly expressed in the central nervous system, participates in the regulation of pathologic processes of AD. In the present study, the effect of let-7a overexpression on Aβ1-40-induced neurotoxicity was evaluated in PC12 and SK-N-SH cells. The results indicated that overexpression of let-7a enhanced the neurotoxicity induced by Aβ1-40 in PC12 and SK-N-SH cells. In addition, the apoptosis induced by Aβ1-40 in PC12 and SK-N-SH cells was increased by let-7a overexpression. Furthermore, Aβ1-40 treatment increased the protein levels of microtubule-associated protein 1A/1B-light chain 3 (LC3) and beclin-1 and increased the LC3 II/I ratio. The mRNA expression levels of beclin-1, autophagy protein 5 (Atg-5) and Atg-7 were also increased by Aβ1-40 treatment in PC12 cells. Let-7a overexpression further upregulated the above autophagy-related markers. Furthermore, the protein level of p62 was increased by Aβ1-40 treatment, and this was further enhanced by let-7a overexpression. Finally, the present results demonstrated that the phosphoinositide-3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) signaling pathway was involved in the autophagy regulation by let-7a. In conclusion, the present study demonstrates that the neurotoxicity induced by Aβ1-40 is augmented by let-7a overexpression via regulation of autophagy, and the PI3K/Akt/mTOR signaling pathway also serves a function in this process.
Collapse
Affiliation(s)
- Huizi Gu
- First Department of Neurology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| | - Lan Li
- First Department of Neurology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| | - Chen Cui
- First Department of Neurology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| | - Zihui Zhao
- First Department of Neurology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| | - Guijun Song
- First Department of Neurology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| |
Collapse
|
46
|
Kindrachuk J. Selective inhibition of host cell signaling for rotavirus antivirals: PI3K/Akt/mTOR-mediated rotavirus pathogenesis. Virulence 2017; 9:5-8. [PMID: 28723236 PMCID: PMC5955445 DOI: 10.1080/21505594.2017.1356539] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Jason Kindrachuk
- a Laboratory of Emerging and Re-Emerging Viruses , Department of Medical Microbiology, University of Manitoba , Winnipeg , MB , Canada
| |
Collapse
|
47
|
Li Y, Lu L, Luo N, Wang YQ, Gao HM. Inhibition of PI3K/AKt/mTOR signaling pathway protects against d-galactosamine/lipopolysaccharide-induced acute liver failure by chaperone-mediated autophagy in rats. Biomed Pharmacother 2017; 92:544-553. [PMID: 28577493 DOI: 10.1016/j.biopha.2017.05.037] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 05/05/2017] [Accepted: 05/08/2017] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE This study aims to investigate the effects of PI3K/AKt/mTOR signaling pathway on the proliferation and apoptosis in acute liver failure (ALF) by chaperone mediated autophagy (CMA). METHODS The hepatocytes extracted from both normal rats and rats with ALF were assigned to control, acute injury, P13K agonist, and P13K inhibitor groups. Quantitative real-time polymerase chain reaction (qRT-PCR) and western blotting were used as part of this investigation to detect the expression of PI3K/AKt/mTOR signaling pathway related-proteins (PI3K, AKt, mTOR), apoptosis related-proteins (Fas, Bax, Bcl-2), chaperone-mediated autophagy (CMA) marker proteins (LAMP-2A, HSC 70), p-PI3K, p-AKt, p-4E-BPI, and p-S6K. An MTT assay was used for analysis of cell proliferation after transfection. Flow cytometry is performed to detect the cell apoptosis. RESULTS In comparison to the normal group, the model group showed enhanced positive rate of PI3K, AKt, mTOR, increased expression levels of PI3K, AKt, mTOR, Fas, Bax, p-PI3K, p-AKt, p-4E-BPI and p-S6K, reduced expression levels of Bcl-2, LAMP-2A and HSC 70. The results in vitro experiment: compared with the acute injury group, the PI3K agonist group showed elevated expression levels of PI3K, AKt, mTOR, Fas, Bax, p-PI3K, p-AKt, p-4E-BPI and p-S6K, decreased expression levels of Bcl-2, LAMP-2A and HSC 70, inhibited cell proliferation, more arrested cells in G1 stage, and promoted cell apoptosis. Opposing this, the P13K inhibitor group exhibited an opposite trend. CONCLUSION In conclusion, inhibition of the PI3K/AKt/mTOR signaling pathway plays a protective role in ALF by promoting CMA expression, which could arrest cell proliferation and promote cell apoptosis.
Collapse
Affiliation(s)
- Yin Li
- Intensive Care Unit, Emergency Medical Research Institute, Tianjin First Center Hospital, Tianjin 300192, PR China
| | - Ling Lu
- Intensive Care Unit, Emergency Medical Research Institute, Tianjin First Center Hospital, Tianjin 300192, PR China
| | - Ning Luo
- Intensive Care Unit, Emergency Medical Research Institute, Tianjin First Center Hospital, Tianjin 300192, PR China
| | - Yong-Qiang Wang
- Intensive Care Unit, Emergency Medical Research Institute, Tianjin First Center Hospital, Tianjin 300192, PR China
| | - Hong-Mei Gao
- Intensive Care Unit, Emergency Medical Research Institute, Tianjin First Center Hospital, Tianjin 300192, PR China.
| |
Collapse
|
48
|
Reactive Oxygen Species-Mediated c-Jun NH 2-Terminal Kinase Activation Contributes to Hepatitis B Virus X Protein-Induced Autophagy via Regulation of the Beclin-1/Bcl-2 Interaction. J Virol 2017; 91:JVI.00001-17. [PMID: 28515304 DOI: 10.1128/jvi.00001-17] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Accepted: 05/07/2017] [Indexed: 02/08/2023] Open
Abstract
Autophagy is closely associated with the regulation of hepatitis B virus (HBV) replication. HBV X protein (HBx), a multifunctional regulator in HBV-associated biological processes, has been demonstrated to be crucial for autophagy induction by HBV. However, the molecular mechanisms of autophagy induction by HBx, especially the signaling pathways involved, remain elusive. In the present investigation, we demonstrated that HBx induced autophagosome formation independently of the class I phosphatidylinositol 3-kinase (PI3K)/AKT/mTOR signaling pathway. In contrast, the class III PI3K(VPS34)/beclin-1 pathway was revealed to be critical for HBx-induced autophagosome formation. Further study showed that HBx did not affect the level of VPS34 and beclin-1 expression but inhibited beclin-1/Bcl-2 association, and c-Jun NH2-terminal kinase (JNK) signaling was found to be important for this process. Moreover, it was found that HBx treatment led to the generation of reactive oxygen species (ROS), and inhibition of ROS activity abrogated both JNK activation and autophagosome formation. Of importance, ROS-JNK signaling was also revealed to play an important role in HBV-induced autophagosome formation and subsequent HBV replication. These data may provide deeper insight into the mechanisms of autophagy induction by HBx and help in the design of new therapeutic strategies against HBV infection.IMPORTANCE HBx plays a key role in diverse HBV-associated biological processes, including autophagy induction. However, the molecular mechanisms of autophagy induction by HBx, especially the signaling pathways involved, remain elusive. In the present investigation, we found that HBx induced autophagy independently of the class I PI3K/AKT/mTOR signaling pathway, while the class III PI3K(VPS34)/beclin-1 pathway was revealed to be crucial for this process. Further data showed that ROS-JNK activation by HBx resulted in the release of beclin-1 from its association with Bcl-2 to form a complex with VPS34, thus enhancing autophagosome formation. Of importance, ROS-JNK signaling was also demonstrated to be critical for HBV replication via regulation of autophagy induction. These data help to elucidate the molecular mechanisms of autophagy induction by HBx/HBV and might be useful for designing novel therapeutic approaches to HBV infection.
Collapse
|
49
|
Yin Y, Dang W, Zhou X, Xu L, Wang W, Cao W, Chen S, Su J, Cai X, Xiao S, Peppelenbosch MP, Pan Q. PI3K-Akt-mTOR axis sustains rotavirus infection via the 4E-BP1 mediated autophagy pathway and represents an antiviral target. Virulence 2017; 9:83-98. [PMID: 28475412 PMCID: PMC5955461 DOI: 10.1080/21505594.2017.1326443] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Rotavirus infection is a major cause of severe dehydrating diarrhea in infants younger than 5 y old and in particular cases of immunocompromised patients irrespective to the age of the patients. Although vaccines have been developed, antiviral therapy is an important complement that cannot be substituted. Because of the lack of specific approved treatment, it is urgent to facilitate the cascade of further understanding of the infection biology, identification of druggable targets and the final development of effective antiviral therapies. PI3K-Akt-mTOR signaling pathway plays a vital role in regulating the infection course of many viruses. In this study, we have dissected the effects of PI3K-Akt-mTOR signaling pathway on rotavirus infection using both conventional cell culture models and a 3D model of human primary intestinal organoids. We found that PI3K-Akt-mTOR signaling is essential in sustaining rotavirus infection. Thus, blocking the key elements of this pathway, including PI3K, mTOR and 4E-BP1, has resulted in potent anti-rotavirus activity. Importantly, a clinically used mTOR inhibitor, rapamycin, potently inhibited both experimental and patient-derived rotavirus strains. This effect involves 4E-BP1 mediated induction of autophagy, which in turn exerts anti-rotavirus effects. These results revealed new insights on rotavirus-host interactions and provided new avenues for antiviral drug development.
Collapse
Affiliation(s)
- Yuebang Yin
- a Department of Gastroenterology and Hepatology , Erasmus MC-University Medical Center , Rotterdam , The Netherlands
| | - Wen Dang
- a Department of Gastroenterology and Hepatology , Erasmus MC-University Medical Center , Rotterdam , The Netherlands
| | - Xinying Zhou
- a Department of Gastroenterology and Hepatology , Erasmus MC-University Medical Center , Rotterdam , The Netherlands
| | - Lei Xu
- a Department of Gastroenterology and Hepatology , Erasmus MC-University Medical Center , Rotterdam , The Netherlands
| | - Wenshi Wang
- a Department of Gastroenterology and Hepatology , Erasmus MC-University Medical Center , Rotterdam , The Netherlands
| | - Wanlu Cao
- a Department of Gastroenterology and Hepatology , Erasmus MC-University Medical Center , Rotterdam , The Netherlands
| | - Sunrui Chen
- a Department of Gastroenterology and Hepatology , Erasmus MC-University Medical Center , Rotterdam , The Netherlands
| | - Junhong Su
- b Medical Faculty, Kunming University of Science and Technology , Kunming , P. R. China
| | - Xuepeng Cai
- c State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS) , Lanzhou , P. R. China
| | - Shaobo Xiao
- d State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University , Wuhan , P. R. China
| | - Maikel P Peppelenbosch
- a Department of Gastroenterology and Hepatology , Erasmus MC-University Medical Center , Rotterdam , The Netherlands
| | - Qiuwei Pan
- a Department of Gastroenterology and Hepatology , Erasmus MC-University Medical Center , Rotterdam , The Netherlands
| |
Collapse
|
50
|
Gu YP, Yang XM, Luo P, Li YQ, Tao YX, Duan ZH, Xiao W, Zhang DY, Liu HZ. Inhibition of acrolein-induced autophagy and apoptosis by a glycosaminoglycan from Sepia esculenta ink in mouse Leydig cells. Carbohydr Polym 2017; 163:270-279. [DOI: 10.1016/j.carbpol.2017.01.081] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 01/16/2017] [Accepted: 01/21/2017] [Indexed: 01/06/2023]
|