1
|
Shi J, Wei X, Jiang F, Zhu J, Shen J, Sun Y. Construction and validation of transcription‑factor‑based prognostic signature for TACE non‑response and characterization of tumor microenvironment infiltration in hepatocellular carcinoma. Oncol Lett 2025; 29:42. [PMID: 39554534 PMCID: PMC11565272 DOI: 10.3892/ol.2024.14788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 10/08/2024] [Indexed: 11/19/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignant tumors worldwide. Despite continuous development of treatment methods, overall survival rate of liver cancer is low. Transcatheter arterial chemoembolization (TACE) is a first-choice treatment for advanced liver cancer. Although it is generally effective, a number of patients do not benefit from it. Therefore, the present study was conducted to assess the response of patients following TACE. RNA-sequencing data and corresponding clinical information were extracted from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus databases. Models were constructed using weighted gene co-expression network analysis and least absolute shrinkage and selection operator-Cox regression analysis based on TCGA-LIHC and GSE104580 cohorts. The receiver operating characteristic curve was used for evaluation. Immunoassay, half-maximal inhibitory concentration analysis of risk groups, genomic enrichment analysis and nomogram construction were also performed. The predictive models were validated at the single-cell level using single-cell databases. Finally, the present study examined the expression of TACE refractoriness-related TFs (TRTs) in TACE-resistant and non-resistant cell lines in vitro. A risk categorization approach was created based on screening of four TRTs. The patients were split into high- and low-risk groups. There were significant variations in immune cell infiltration, medication sensitivity and overall survival (OS) between patients in the high-risk and low-risk groups. Multivariate Cox regression analysis showed that the risk score was an independent prognostic factor for OS. In the single-cell gene set, risk score was a good indicator of tumor microenvironment (TME). Reverse transcription-quantitative PCR revealed that three high-risk TRTs were upregulated in TACE-resistant cells. Prognosis and TME status of liver cancer patients following TACE could be assessed using a predictive model based on transcription factor correlation. This predictive model provided a reliable and simplified method to guide the clinical treatment of HCC.
Collapse
Affiliation(s)
- Jiapeng Shi
- Department of Interventional Medicine, Nantong Traditional Chinese Medicine Hospital, Nantong, Jiangsu 226001, P.R. China
| | - Xintong Wei
- Department of Medical Imaging, Nantong Traditional Chinese Medicine Hospital, Nantong, Jiangsu 226001, P.R. China
| | - Fangmei Jiang
- Department of Oncology, Yancheng Tinghu District People's Hospital, Yancheng, Jiangsu 224000, P.R. China
| | - Jianjun Zhu
- Department of Oncology, Yancheng Tinghu District People's Hospital, Yancheng, Jiangsu 224000, P.R. China
| | - Jiandong Shen
- Department of Invasive Technology, Affiliated Nantong Hospital 3 of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Yanjun Sun
- Department of Oncology, Yancheng Tinghu District People's Hospital, Yancheng, Jiangsu 224000, P.R. China
| |
Collapse
|
2
|
Sinha P, Thio CL, Balagopal A. Intracellular Host Restriction of Hepatitis B Virus Replication. Viruses 2024; 16:764. [PMID: 38793645 PMCID: PMC11125714 DOI: 10.3390/v16050764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/06/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
The hepatitis B virus (HBV) infects hepatocytes and hijacks host cellular mechanisms for its replication. Host proteins can be frontline effectors of the cell's defense and restrict viral replication by impeding multiple steps during its intracellular lifecycle. This review summarizes many of the well-described restriction factors, their mechanisms of restriction, and counteractive measures of HBV, with a special focus on viral transcription. We discuss some of the limitations and knowledge gaps about the restriction factors, highlighting how these factors may be harnessed to facilitate therapeutic strategies against HBV.
Collapse
Affiliation(s)
| | | | - Ashwin Balagopal
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (P.S.); (C.L.T.)
| |
Collapse
|
3
|
Micro-Players of Great Significance-Host microRNA Signature in Viral Infections in Humans and Animals. Int J Mol Sci 2022; 23:ijms231810536. [PMID: 36142450 PMCID: PMC9504570 DOI: 10.3390/ijms231810536] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/04/2022] [Accepted: 09/08/2022] [Indexed: 11/22/2022] Open
Abstract
Over time, more and more is becoming known about micro-players of great significance. This is particularly the case for microRNAs (miRNAs; miR), which have been found to participate in the regulation of many physiological and pathological processes in both humans and animals. One such process is viral infection in humans and animals, in which the host miRNAs—alone or in conjunction with the virus—interact on two levels: viruses may regulate the host’s miRNAs to evade its immune system, while the host miRNAs can play anti- or pro-viral roles. The purpose of this comprehensive review is to present the key miRNAs involved in viral infections in humans and animals. We summarize the data in the available literature, indicating that the signature miRNAs in human viral infections mainly include 12 miRNAs (i.e., miR-155, miR-223, miR-146a, miR-122, miR-125b, miR-132, miR-34a, miR -21, miR-16, miR-181 family, let-7 family, and miR-10a), while 10 miRNAs are commonly found in animals (i.e., miR-155, miR-223, miR-146a, miR-145, miR-21, miR-15a/miR-16 cluster, miR-181 family, let-7 family, and miR-122) in this context. Knowledge of which miRNAs are involved in different viral infections and the biological functions that they play can help in understanding the pathogenesis of viral diseases, facilitating the future development of therapeutic agents for both humans and animals.
Collapse
|
4
|
Tian X, Dong H, Lai X, Ou G, Cao J, Shi J, Xiang C, Wang L, Zhang X, Zhang K, Song J, Deng J, Deng H, Lu S, Zhuang H, Li T, Xiang K. TRIM56 impairs HBV infection and replication by inhibiting HBV core promoter activity. Antiviral Res 2022; 207:105406. [PMID: 36084850 DOI: 10.1016/j.antiviral.2022.105406] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 08/04/2022] [Accepted: 08/31/2022] [Indexed: 11/02/2022]
Abstract
Members of the tripartite motif (TRIM) protein family strongly induced by interferons (IFNs) are parts of the innate immune system with antiviral activity. However, it is still unclear which TRIMs could play important roles in hepatitis B virus (HBV) inhibition. Here, we identified that TRIM56 expression responded in IFN-treated HepG2-NTCP cells and HBV-infected liver tissues, which was a potent IFN-inducible inhibitor of HBV replication. Mechanistically, TRIM56 suppressed HBV replication via its Ring and C-terminal domain. C-terminal domain was essential for TRIM56 translocating from cytoplasm to nucleus during HBV infection. Further analysis revealed that TRIM56's Ring domain targeted IκBα for ubiquitination. This modification induced phosphorylation of p65, which subsequently inhibited HBV core promoter activity, resulting in the inhibition of HBV replication. The p65 was found to be necessary for NF-κB signal pathway to inhibit HBV replication. We verified our findings using HepG2-NTCP and primary human hepatocytes. Our findings reveal that TRIM56 is a critical antiviral immune effector and exerts an anti-HBV activity via NF-κB signal pathway, which is essential for inhibiting transcription of HBV covalently closed circular DNA.
Collapse
Affiliation(s)
- Xing Tian
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Huijun Dong
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Xinyuan Lai
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Guomin Ou
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Junning Cao
- Faculty of Hepato-Pancreato-Biliary Surgery, Chinese PLA General Hospital, Beijing, 100089, China
| | - Jihang Shi
- Faculty of Hepato-Pancreato-Biliary Surgery, Chinese PLA General Hospital, Beijing, 100089, China
| | - Chengang Xiang
- School of Basic Medical Sciences, State Key Laboratory of Natural and Biomimetic, Drugs, Peking University Health Science Center and the MOE Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua, Center for Life Sciences, Peking University, Beijing, 100191, China; Renal Division, Peking University First Hospital, Beijing, China
| | - Lei Wang
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Xuechao Zhang
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Kai Zhang
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Ji Song
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Juan Deng
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Hongkui Deng
- School of Basic Medical Sciences, State Key Laboratory of Natural and Biomimetic, Drugs, Peking University Health Science Center and the MOE Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua, Center for Life Sciences, Peking University, Beijing, 100191, China
| | - Shichun Lu
- Faculty of Hepato-Pancreato-Biliary Surgery, Chinese PLA General Hospital, Beijing, 100089, China
| | - Hui Zhuang
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
| | - Tong Li
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
| | - Kuanhui Xiang
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
| |
Collapse
|
5
|
Van Damme E, Vanhove J, Severyn B, Verschueren L, Pauwels F. The Hepatitis B Virus Interactome: A Comprehensive Overview. Front Microbiol 2021; 12:724877. [PMID: 34603251 PMCID: PMC8482013 DOI: 10.3389/fmicb.2021.724877] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 08/17/2021] [Indexed: 12/19/2022] Open
Abstract
Despite the availability of a prophylactic vaccine, chronic hepatitis B (CHB) caused by the hepatitis B virus (HBV) is a major health problem affecting an estimated 292 million people globally. Current therapeutic goals are to achieve functional cure characterized by HBsAg seroclearance and the absence of HBV-DNA after treatment cessation. However, at present, functional cure is thought to be complicated due to the presence of covalently closed circular DNA (cccDNA) and integrated HBV-DNA. Even if the episomal cccDNA is silenced or eliminated, it remains unclear how important the high level of HBsAg that is expressed from integrated HBV DNA is for the pathology. To identify therapies that could bring about high rates of functional cure, in-depth knowledge of the virus' biology is imperative to pinpoint mechanisms for novel therapeutic targets. The viral proteins and the episomal cccDNA are considered integral for the control and maintenance of the HBV life cycle and through direct interaction with the host proteome they help create the most optimal environment for the virus whilst avoiding immune detection. New HBV-host protein interactions are continuously being identified. Unfortunately, a compendium of the most recent information is lacking and an interactome is unavailable. This article provides a comprehensive review of the virus-host relationship from viral entry to release, as well as an interactome of cccDNA, HBc, and HBx.
Collapse
Affiliation(s)
- Ellen Van Damme
- Janssen Research & Development, Janssen Pharmaceutical Companies, Beerse, Belgium
| | - Jolien Vanhove
- Janssen Research & Development, Janssen Pharmaceutical Companies, Beerse, Belgium.,Early Discovery Biology, Charles River Laboratories, Beerse, Belgium
| | - Bryan Severyn
- Janssen Research & Development, Janssen Pharmaceutical Companies, Springhouse, PA, United States
| | - Lore Verschueren
- Janssen Research & Development, Janssen Pharmaceutical Companies, Beerse, Belgium
| | - Frederik Pauwels
- Janssen Research & Development, Janssen Pharmaceutical Companies, Beerse, Belgium
| |
Collapse
|
6
|
Ahluwalia S, Choudhary D, Tyagi P, Kumar V, Vivekanandan P. Vitamin D signaling inhibits HBV activity by directly targeting the HBV core promoter. J Biol Chem 2021; 297:101233. [PMID: 34562448 PMCID: PMC8517215 DOI: 10.1016/j.jbc.2021.101233] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/16/2021] [Accepted: 09/21/2021] [Indexed: 12/12/2022] Open
Abstract
Clinical and epidemiological studies support a role for vitamin D in suppressing hepatitis B virus (HBV). This antiviral role of vitamin D is widely attributed to vitamin D receptor (VDR)/retinoid X receptor-mediated regulation of host immunomodulatory genes through vitamin D response elements (VDREs) in their promoters. Here, we investigated the ability of calcitriol (1α,25-dihydroxyvitamin D3, metabolically activated vitamin D) to directly regulate HBV activity through this signaling pathway. We observed that calcitriol selectively inhibited only the HBV core promoter without affecting the HBV-PreS1, HBV-PreS2/S, or HBx promoters. We then identified a VDRE cluster in the HBV core promoter that is highly conserved across most HBV genotypes. Disruption of this VDRE cluster abrogated calcitriol-mediated suppression of the HBV core promoter. Furthermore, we showed that VDR interacts directly with the VDRE cluster in the HBV core promoter independent of retinoid X receptor. This demonstrates that calcitriol inhibits HBV core promoter activity through a noncanonical calcitriol-activated VDR pathway. Finally, we observed that calcitriol suppressed expression of the canonical HBV core promoter transcripts, pregenomic RNA, and precore RNA in multiple HBV cell culture models. In addition, calcitriol inhibited the secretion of hepatitis B "e" antigen and hepatitis B surface antigen (HBV-encoded proteins linked to poor disease prognosis), without affecting virion secretion. Our findings identify VDR as a novel regulator of HBV core promoter activity and also explain at least in part the correlation of vitamin D levels to HBV activity observed in clinical studies. Furthermore, this study has implications on the potential use of vitamin D along with anti-HBV therapies, and lays the groundwork for studies on vitamin D-mediated regulation of viruses through VDREs in virus promoters.
Collapse
Affiliation(s)
- Shivaksh Ahluwalia
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi, India
| | - Divya Choudhary
- Department of Chemical Engineering, Indian Institute of Technology Delhi, New Delhi, India
| | - Purnima Tyagi
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary sciences, New Delhi, India
| | - Vijay Kumar
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary sciences, New Delhi, India
| | - Perumal Vivekanandan
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi, India.
| |
Collapse
|
7
|
Singh P, Kairuz D, Arbuthnot P, Bloom K. Silencing hepatitis B virus covalently closed circular DNA: The potential of an epigenetic therapy approach. World J Gastroenterol 2021; 27:3182-3207. [PMID: 34163105 PMCID: PMC8218364 DOI: 10.3748/wjg.v27.i23.3182] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [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: 03/23/2021] [Accepted: 05/07/2021] [Indexed: 02/06/2023] Open
Abstract
Global prophylactic vaccination programmes have helped to curb new hepatitis B virus (HBV) infections. However, it is estimated that nearly 300 million people are chronically infected and have a high risk of developing hepatocellular carcinoma. As such, HBV remains a serious health priority and the development of novel curative therapeutics is urgently needed. Chronic HBV infection has been attributed to the persistence of the covalently closed circular DNA (cccDNA) which establishes itself as a minichromosome in the nucleus of hepatocytes. As the viral transcription intermediate, the cccDNA is responsible for producing new virions and perpetuating infection. HBV is dependent on various host factors for cccDNA formation and the minichromosome is amenable to epigenetic modifications. Two HBV proteins, X (HBx) and core (HBc) promote viral replication by modulating the cccDNA epigenome and regulating host cell responses. This includes viral and host gene expression, chromatin remodeling, DNA methylation, the antiviral immune response, apoptosis, and ubiquitination. Elimination of the cccDNA minichromosome would result in a sterilizing cure; however, this may be difficult to achieve. Epigenetic therapies could permanently silence the cccDNA minichromosome and promote a functional cure. This review explores the cccDNA epigenome, how host and viral factors influence transcription, and the recent epigenetic therapies and epigenome engineering approaches that have been described.
Collapse
Affiliation(s)
- Prashika Singh
- Wits/SAMRC Antiviral Gene Therapy Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg 2050, Gauteng, South Africa
| | - Dylan Kairuz
- Wits/SAMRC Antiviral Gene Therapy Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg 2050, Gauteng, South Africa
| | - Patrick Arbuthnot
- Wits/SAMRC Antiviral Gene Therapy Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg 2050, Gauteng, South Africa
| | - Kristie Bloom
- Wits/SAMRC Antiviral Gene Therapy Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg 2050, Gauteng, South Africa
| |
Collapse
|
8
|
Transcription Repressor Protein ZBTB25 Associates with HDAC1-Sin3a Complex in Mycobacterium tuberculosis-Infected Macrophages, and Its Inhibition Clears Pathogen by Autophagy. mSphere 2021; 6:6/1/e00036-21. [PMID: 33627504 PMCID: PMC8544881 DOI: 10.1128/msphere.00036-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Downregulation of host gene expression is a key strategy employed by intracellular pathogens for their survival in macrophages and subsequent pathogenesis. In a previous study, we have shown that histone deacetylase 1 (HDAC1) levels go up in macrophages infected with Mycobacterium tuberculosis, and it hypoacetylates histone H3 at the promoter of IL-12B gene, leading to its downregulation. We now show that after infection with M. tuberculosis, HDAC1 is phosphorylated, and the levels of phosphorylated HDAC1 (pHDAC1) increase significantly in macrophages. We found that transcriptional repressor protein zinc finger and BTB domain 25 (ZBTB25) and transcriptional corepressor Sin3a associate with the HDAC1 silencing complex, which is recruited to the promoter of IL-12B to downregulate its expression in infected macrophages. Knocking down of ZBTB25 enhanced release of IL-12p40 from infected macrophages. Inhibition of HDAC1 and ZBTB25 promoted colocalization of M. tuberculosis and LC3 (microtubule-associated protein 1A/1B-light chain 3) in autophagosomes. Induction of autophagy resulted in the killing of intracellular M. tuberculosis. Enhanced phosphorylation of JAK2 and STAT4 was observed in macrophages upon treatment with HDAC1 and ZBTB inhibitors, and inhibition of JAK2/STAT4 negated the killing of the intracellular pathogen, suggesting their role in the autophagy-mediated killing of intracellular M. tuberculosis. In view of the emergence of drug resistance in M. tuberculosis, host-directed therapy is an attractive alternative strategy to combat tuberculosis (TB). HDACs have been proposed to be host targets for TB treatment. Our study indicates that ZBTB25, a functional subunit of the HDAC1/Sin3a repressor complex involved in IL-12B suppression, could be an alternative target for host-directed anti-TB therapy. IMPORTANCE Following infection with M. tuberculosis, levels of HDAC1 go up in macrophages, and it is recruited to the promoter of IL-12B where it hypoacetylates histone H3, leading to the downregulation of the gene. Here, we show that host transcriptional repressor protein ZBTB25 and transcriptional corepressor Sin3a associate with HDAC1 in the silencing complex. Knocking down of ZBTB25 prevented the recruitment of the complex to the promoter and consequently enhanced the gene expression and the release of IL-12p40 from infected macrophages. Pharmacological inhibition of ZBTB25 in infected macrophages resulted in the induction of autophagy and killing of intracellular M. tuberculosis. Drug-resistant TB is a serious challenge to TB control programs all over the world which calls for finding alternative therapeutic methods. Host-directed therapy is gaining significant momentum in treating infectious diseases. We propose that ZBTB25 is a potential target for host-directed treatment of TB.
Collapse
|
9
|
Minor MM, Hollinger FB, McNees AL, Jung SY, Jain A, Hyser JM, Bissig KD, Slagle BL. Hepatitis B Virus HBx Protein Mediates the Degradation of Host Restriction Factors through the Cullin 4 DDB1 E3 Ubiquitin Ligase Complex. Cells 2020; 9:E834. [PMID: 32235678 PMCID: PMC7226812 DOI: 10.3390/cells9040834] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 02/06/2023] Open
Abstract
The hepatitis B virus (HBV) regulatory HBx protein is required for infection, and its binding to cellular damaged DNA binding protein 1 (DDB1) is critical for this function. DDB1 is an adaptor protein for the cullin 4A Really Interesting New Gene (RING) E3 ubiquitin ligase (CRL4) complex and functions by binding cellular DDB1 cullin associated factor (DCAF) receptor proteins that recruit substrates for ubiquitination and degradation. We compared the proteins found in the CRL4 complex immunoprecipitated from uninfected versus HBV-infected hepatocytes from human liver chimeric mice for insight into mechanisms by which HBV and the cell interact within the CRL4 complex. Consistent with its role as a viral DCAF, HBx was found in the HBV CRL4 complexes. In tissue culture transfection experiments, we showed that HBx expression led to decreased levels of known restriction factor structural maintenance of chromosomes protein 6 (SMC6) and putative restriction factors stromal interaction molecule 1 (STIM1, zinc finger E-box binding homeobox 2 (ZEB2), and proteasome activator subunit 4 (PSME4). Moreover, silencing of these proteins led to increased HBV replication in the HepG2-sodium taurocholate cotransporting polypeptide (NTCP) infection model. We also identified cellular DCAF receptors in CRL4 complexes from humanized mice. Increasing amounts of HBx did not reveal competitive DCAF binding to cullin4 (CUL4)-DDB1 in plasmid-transfected cells. Our results suggest a model in which HBx benefits virus replication by directly or indirectly degrading multiple cellular restriction factors.
Collapse
Affiliation(s)
- Marissa M. Minor
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; (M.M.M.); (F.B.H.); (A.L.M.); (J.M.H.)
| | - F. Blaine Hollinger
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; (M.M.M.); (F.B.H.); (A.L.M.); (J.M.H.)
| | - Adrienne L. McNees
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; (M.M.M.); (F.B.H.); (A.L.M.); (J.M.H.)
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Sung Yun Jung
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA;
- Department of Biochemistry, Baylor College of Medicine, Houston, TX 77030, USA
| | - Antrix Jain
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Joseph M. Hyser
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; (M.M.M.); (F.B.H.); (A.L.M.); (J.M.H.)
| | - Karl-Dimiter Bissig
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Betty L. Slagle
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; (M.M.M.); (F.B.H.); (A.L.M.); (J.M.H.)
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA;
| |
Collapse
|
10
|
SOX9 represses hepatitis B virus replication through binding to HBV EnhII/Cp and inhibiting the promoter activity. Antiviral Res 2020; 177:104761. [PMID: 32147495 DOI: 10.1016/j.antiviral.2020.104761] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 02/26/2020] [Accepted: 02/28/2020] [Indexed: 12/13/2022]
Abstract
Hepatitis B virus (HBV) infection affects 364 million people worldwide and causes a serious global public health problem. The SRY-related high mobility group-box 9 (SOX9) is a risk of developing cirrhosis in patients with chronic hepatitis B and a cancer stem cell marker. However, the role of SOX9 in HBV replication has not been reported. This study revealed a distinct mechanism underling the regulation of HBV replication mediated by SOX9. HBV induces SOX9 mRNA and protein expression in human hepatoma cells, including HepG2.2.15, HepG2, Huh7, and HepG2-NTCP cells. Further study demonstrated that HBV activates SOX9 expression at the transcriptional level through inducing SOX9 promoter activity and HBc could induce the activity of SOX9 promoter. Interestingly, SOX9 in turn represses HBV replication in human hepatoma cells. More importantly, SOX9 inhibits HBV infection in HepG2-NTCP cells and C57/BL6 mice. Detailed study revealed that SOX9 suppresses HBV replication through directly binding to HBV EnhII/Cp (HBV 1667-1672 nt) to inhibit EnhII/Cp activation. Results from deletion mutant analysis, ChIP assay, nuclear and cytoplasmic extraction analysis, and immunofluorescence demonstrated that SOX9 high mobility group (HMG) domain is required for SOX9 anti-HBV activity. Moreover, we demonstrated that SOX9 and hepatocyte nuclear factor 4 alpha (HNF4α) can bind to HBV EnhII/Cp (HBV 1667-1672 nt) individually and simultaneously to regulate the promoter activity. Collectively, the results revealed a distinct negative feedback mechanism underlying HBV replication and SOX9 expression, and identified SOX9 as a new host restriction factor in HBV replication and infection. IMPORTANCE: HBV infection is a global public health problem by causing serious liver diseases, but the mechanisms underlying HBV pathogenesis remain largely unknown. SOX9 is a risk of developing cirrhosis and a cancer stem cell marker, however, the role of SOX9 in HBV infection has not been reported. The authors revealed a distinct mechanism underling the regulation of HBV replication and SOX9 expression. On the one hand, HBV induces SOX9 expression in human hepatoma cells through activating SOX9 promoter. On the other hand, SOX9 in turn represses HBV replication in human hepatoma cells by binding to and inhibiting HBV EnhII/Cp through its HMG domain. More importantly, SOX9 inhibits HBV infection in HepG2-NTCP cells and C57/BL6 mice. Therefore, this study identifies SOX9 as a novel and potential therapeutic reagent for the prevention and treatment of HBV-associated diseases.
Collapse
|
11
|
Yang H, Mo J, Xiang Q, Zhao P, Song Y, Yang G, Wu K, Liu Y, Liu W, Wu J. SOX2 Represses Hepatitis B Virus Replication by Binding to the Viral EnhII/Cp and Inhibiting the Promoter Activation. Viruses 2020; 12:v12030273. [PMID: 32121397 PMCID: PMC7150879 DOI: 10.3390/v12030273] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/15/2020] [Accepted: 02/25/2020] [Indexed: 12/21/2022] Open
Abstract
Hepatitis B virus (HBV) replication is controlled by four promoters (preS1, preS2, Cp, and Xp) and two enhancers (EnhI and EnhII). EnhII stimulates Cp activity to regulate the transcriptions of precore, core, polymerase, and pregenomic RNAs, and therefore, EnhII/Cp is essential for the regulation of HBV replication. This study revealed a distinct mechanism underlying the suppression of EnhII/Cp activation and HBV replication. On the one hand, the sex determining region Y box2 (SOX2), a transcription factor, is induced by HBV. On the other hand, SOX2, in turn, represses the expression levels of HBV RNAs, HBV core-associated DNA, hepatitis B surface antigen (HBsAg), and hepatitis B e antigen (HBeAg), thereby playing an inhibitory role during HBV replication. Further studies indicated that SOX2 bound to the EnhII/Cp DNA and repressed the promoter activation. With the deletion of the high mobility group (HMG) domain, SOX2 loses the ability to repress EnhII/Cp activation, viral RNA transcription, HBV core-associated DNA replication, HBsAg and HBeAg production, as well as fails to enter the nucleus, demonstrating that the HMG domain is required for the SOX2-mediated repression of HBV replication. Moreover, SOX2 represses HBsAg and HBeAg secretion in BALB/c mice sera, and attenuates HBV 3.5 kb RNA transcription and hepatitis B virus core protein (HBc) production in the liver tissues, demonstrating that SOX2 suppresses HBV replication in mice. Furthermore, the results revealed that the HMG domain was required for SOX2-mediated repression of HBV replication in the mice. Taken together, the above facts indicate that SOX2 acts as a new host restriction factor to repress HBV replication by binding to the viral EnhII/Cp and inhibiting the promoter activation through the HMG domain.
Collapse
Affiliation(s)
- Hua Yang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China; (H.Y.); (J.M.); (Q.X.); (P.Z.); (Y.S.); (G.Y.); (K.W.); (Y.L.)
| | - Jiayin Mo
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China; (H.Y.); (J.M.); (Q.X.); (P.Z.); (Y.S.); (G.Y.); (K.W.); (Y.L.)
| | - Qi Xiang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China; (H.Y.); (J.M.); (Q.X.); (P.Z.); (Y.S.); (G.Y.); (K.W.); (Y.L.)
| | - Peiyi Zhao
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China; (H.Y.); (J.M.); (Q.X.); (P.Z.); (Y.S.); (G.Y.); (K.W.); (Y.L.)
| | - Yunting Song
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China; (H.Y.); (J.M.); (Q.X.); (P.Z.); (Y.S.); (G.Y.); (K.W.); (Y.L.)
| | - Ge Yang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China; (H.Y.); (J.M.); (Q.X.); (P.Z.); (Y.S.); (G.Y.); (K.W.); (Y.L.)
| | - Kailang Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China; (H.Y.); (J.M.); (Q.X.); (P.Z.); (Y.S.); (G.Y.); (K.W.); (Y.L.)
| | - Yingle Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China; (H.Y.); (J.M.); (Q.X.); (P.Z.); (Y.S.); (G.Y.); (K.W.); (Y.L.)
- Guangdong Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou 510632, China
| | - Weiyong Liu
- Department of Clinical Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Correspondence: (W.L.); (J.W.); Tel.: +86-27-68754979 (J.W.)
| | - Jianguo Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China; (H.Y.); (J.M.); (Q.X.); (P.Z.); (Y.S.); (G.Y.); (K.W.); (Y.L.)
- Guangdong Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou 510632, China
- Correspondence: (W.L.); (J.W.); Tel.: +86-27-68754979 (J.W.)
| |
Collapse
|
12
|
Host Transcription Factors in Hepatitis B Virus RNA Synthesis. Viruses 2020; 12:v12020160. [PMID: 32019103 PMCID: PMC7077322 DOI: 10.3390/v12020160] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 01/27/2020] [Accepted: 01/28/2020] [Indexed: 02/06/2023] Open
Abstract
The hepatitis B virus (HBV) chronically infects over 250 million people worldwide and is one of the leading causes of liver cancer and hepatocellular carcinoma. HBV persistence is due in part to the highly stable HBV minichromosome or HBV covalently closed circular DNA (cccDNA) that resides in the nucleus. As HBV replication requires the help of host transcription factors to replicate, focusing on host protein–HBV genome interactions may reveal insights into new drug targets against cccDNA. The structural details on such complexes, however, remain poorly defined. In this review, the current literature regarding host transcription factors’ interactions with HBV cccDNA is discussed.
Collapse
|
13
|
Ye Y, Yang J, Hu Q, Mao J, Yang Q, Chen H, Li D, Li P, Duan L, Wang B, Chen J, Chen W. SIP1 serves a role in HBx‑induced liver cancer growth and metastasis. Int J Oncol 2019; 55:1019-1032. [PMID: 31793654 PMCID: PMC6776188 DOI: 10.3892/ijo.2019.4884] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 07/30/2019] [Indexed: 12/24/2022] Open
Abstract
Hepatitis B virus (HBV) has been revealed to be involved in the development of hepatocellular carcinoma. However, the mechanism remains to be fully elucidated. Smad‑interacting protein 1 (SIP1) is a transcriptional repressor, which serves a pivotal role in cell metastasis. In the present study, the role of SIP1 in HBx‑induced hepatocyte EMT and cancer aggressiveness was examined. It was found that HBV X protein (HBx) increased the expression of SIP1 and recruited it to the promoter of E‑cadherin, resulting in depression of the transcription of E‑cadherin. Histone deacetylase 1 was also found to be involved in the repressive complex formation. Furthermore, in an orthotopic tumor transplantation model in vivo, HBx promoted tumor growth and metastasis, whereas the knockdown of SIP1 attenuated the effect of HBx. These results indicate a novel mechanism for the development of HBV‑related liver cancer.
Collapse
Affiliation(s)
- Yuanyuan Ye
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Jun Yang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Qin Hu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Jinju Mao
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Qianfan Yang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Hong Chen
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Dandan Li
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Pu Li
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Liang Duan
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Bo Wang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Juan Chen
- Key Laboratory of Molecular Biology of Infectious Diseases Designated by The Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Weixian Chen
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| |
Collapse
|
14
|
He Q, Song X, Huang Y, Huang W, Ye B, Luo H, Luo H, Wu L, Wang Z, Chen W, Zhang L. Dexamethasone Stimulates Hepatitis B Virus (HBV) Replication Through Autophagy. Med Sci Monit 2018; 24:4617-4624. [PMID: 29972684 PMCID: PMC6064191 DOI: 10.12659/msm.906250] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background Reactivation of hepatitis B virus (HBV) is a fatal complication of chemotherapy. Occult HBV infection might be reactivated in patients undergoing chemotherapy or immunosuppression. However, the mechanism of HBV reactivation induced by chemotherapy or immunosuppression remains unclear. Material/Methods HepG2.2.15 cells were treated with an autophagy inducer (rapamycin), an inhibitor (3-methyladenine, 3-MA), and dexamethasone. Autophagosomes were observed by a transmission electron microscope (TEM). LC3-I, LC3-II, and P62 were analyzed by western blot. HBV replicative intermediates were detected by southern blot. HBV DNA expression was quantitated with real-time polymerase chain reaction (PCR). The level of HBV surface antigen (HBsAg) in culture medium was examined by ELISA. Results In this study, we find that dexamethasone stimulates HBV replication and protein expression by inducing autophagy in HepG2.2.15 cells. In contrast, autophagy inhibitor (3-MA) abrogates HBsAg secretion stimulated by dexamethasone. Conclusions Our results suggest that dexamethasone stimulates HBV replication through autophagy. This might provide a novel insight into the mechanism of glucocorticoid-mediated HBV reactivation through autophagy, which might be a new therapeutic target.
Collapse
Affiliation(s)
- Qiao He
- Department of Clinical Laboratory, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China (mainland).,Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China (mainland)
| | - Xiaoyu Song
- Department of Clinical Laboratory, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China (mainland)
| | - Yecai Huang
- Department of Radiation Oncology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China (mainland)
| | - Wenjuan Huang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China (mainland)
| | - Bo Ye
- Department of Clinical Laboratory, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China (mainland)
| | - Huaichao Luo
- Department of Clinical Laboratory, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China (mainland)
| | - Hao Luo
- Department of Clinical Laboratory, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China (mainland)
| | - Lichun Wu
- Department of Clinical Laboratory, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China (mainland)
| | - Zuo Wang
- Department of Clinical Laboratory, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China (mainland)
| | - Weixian Chen
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China (mainland)
| | - Li Zhang
- Department of Clinical Laboratory, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China (mainland)
| |
Collapse
|
15
|
miR-146 promotes HBV replication and expression by targeting ZEB2. Biomed Pharmacother 2018; 99:576-582. [PMID: 29902868 DOI: 10.1016/j.biopha.2018.01.097] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 01/17/2018] [Accepted: 01/24/2018] [Indexed: 12/18/2022] Open
Abstract
Hepatitis B virus (HBV) is associated with the development of a wide spectrum of liver diseases. The involvement of miRNAs in HBV replication is being gradually identified. Among these miRNAs, miR-146a expression was found to be positively correlated with HBV replication levels. However, the regulatory relationship between miR-146a and HBV replication is still unclear. In the present study, miR-146a was upregulated in HBV-expressing HepG2.2.15 cells compared with HepG2 cells. Overexpression of miR-146a or knockdown of Zinc finger E-box-binding homeobox 2 (ZEB2) promoted HBV replication and expression, while downregulation of miR-146a or overexpression of ZEB2 suppressed HBV replication and expression. In addition, miR-146a was demonstrated to directly target ZEB2. Furthermore, ZEB2 silencing abated anti-miR-146a-induced inhibition on HBV replication and expression. These findings suggested that miR-146a promoted HBV replication by targeting ZEB2, providing a new antiviral strategy for HBV infection.
Collapse
|
16
|
Role of HBx in hepatitis B virus persistence and its therapeutic implications. Curr Opin Virol 2018; 30:32-38. [PMID: 29454995 DOI: 10.1016/j.coviro.2018.01.007] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 01/17/2018] [Accepted: 01/24/2018] [Indexed: 12/12/2022]
Abstract
Chronic hepatitis B virus infection is a significant risk factor for cirrhosis and hepatocellular carcinoma. The HBx protein is required for virus replication, but the lack of robust infection models has hindered our understanding of HBx functions that could be targeted for antiviral purposes. We briefly review three properties of HBx: its binding to DDB1 and its regulation of cell survival and metabolism, to illustrate how a single viral protein can have multiple effects in a cell. We propose that different functions of HBx are needed, depending on the changing hepatocyte environment encountered during a chronic virus infection, and that these functions might serve as novel therapeutic targets for inhibiting hepatitis B virus replication and the development of associated diseases.
Collapse
|
17
|
Zinc Finger-Containing Cellular Transcription Corepressor ZBTB25 Promotes Influenza Virus RNA Transcription and Is a Target for Zinc Ejector Drugs. J Virol 2017; 91:JVI.00842-17. [PMID: 28768860 DOI: 10.1128/jvi.00842-17] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 07/24/2017] [Indexed: 02/03/2023] Open
Abstract
Influenza A virus (IAV) replication relies on an intricate interaction between virus and host cells. How the cellular proteins are usurped for IAV replication remains largely obscure. The aim of this study was to search for novel and potential cellular factors that participate in IAV replication. ZBTB25, a transcription repressor of a variety of cellular genes, was identified by an RNA interference (RNAi) genomic library screen. Depletion of ZBTB25 significantly reduced IAV production. Conversely, overexpression of ZBTB25 enhanced it. ZBTB25 interacted with the viral RNA-dependent RNA polymerase (RdRp) protein and modulated its transcription activity. In addition, ZBTB25 also functioned as a viral RNA (vRNA)-binding protein, binding preferentially to the U-rich sequence within the 5' untranslated region (UTR) of vRNA. Both protein-protein and protein-RNA interactions involving ZBTB25 facilitated viral RNA transcription and replication. In addition, ZBTB25 suppressed interferon production, further enhancing viral replication. ZBTB25-associated functions required an intact zinc finger domain and posttranslational SUMO-1 modification of ZBTB25. Furthermore, treatment with disulfiram (a zinc ejector) of ZBTB25-overexpressing cells showed significantly reduced IAV production as a result of reduced RNA synthesis. Our findings indicate that IAV usurps ZBTB25 for IAV RNA synthesis and serves as a novel and potential therapeutic antiviral target.IMPORTANCE IAV-induced seasonal influenza causes severe illness and death in high-risk populations. However, IAV has developed resistance to current antiviral drugs due to its high mutation rate. Therefore, development of drugs targeting cellular factors required for IAV replication is an attractive alternative for IAV therapy. Here, we discovered a cellular protein, ZBTB25, that enhances viral RdRp activity by binding to both viral RdRp and viral RNA to stimulate viral RNA synthesis. A unique feature of ZBTB25 in the regulation of viral replication is its dual transcription functions, namely, promoting viral RNA transcription through binding to the U-rich region of vRNA and suppressing cellular interferon production. ZBTB25 contains a zinc finger domain that is required for RNA-inhibitory activity by chelating zinc ions. Disulfiram treatment disrupts the zinc finger functions, effectively repressing IAV replication. Based on our findings, we demonstrate that ZBTB25 regulates IAV RNA transcription and replication and serves as a promising antiviral target for IAV treatment.
Collapse
|
18
|
Huang H, Zhou W, Zhu H, Zhou P, Shi X. Baicalin benefits the anti-HBV therapy via inhibiting HBV viral RNAs. Toxicol Appl Pharmacol 2017; 323:36-43. [PMID: 28322895 DOI: 10.1016/j.taap.2017.03.016] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 03/14/2017] [Accepted: 03/15/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Although current antiviral treatments (nucleoside analogs, NAs) for chronic hepatitis B virus (HBV) infection are effective in suppressing HBV-DNA replication, their clinical outcomes can be compromised by the increasing drug resistance and the inefficiency in promoting HBsAg/HBeAg seroconversion. OBJECTIVES In this study, we will explore possible effects and mechanism of a natural product baicalin (BA) with the anti-HBV efficacy of entecavir (ETV), a first-line anti-HBV drug, in HBV-DNA, HBsAg/HBeAg seroconversion and drug-resistance. METHODS The co-effects of BA and ETV were conducted in wild-type/NA-resistance mutant HBV cell lines and DHBV-infected duckling models. HBV-DNA/RNAs, HBsAg/HBeAg, host factors (hepatocyte nuclear factors) were explored for possible anti-HBV mechanism. RESULTS AND DISCUSSION BA could significantly enhance and reduced HBsAg and HBeAg in hepG2.2.15, a wild-type HBV cell line. Co-treatment of BA and ETV had a more dramatic effect in NA-resistant HBVrtM204V/rtLl80M transfected hepG2 cells. Our study further revealed that BA mainly inhibited the production of HBV RNAs (3.5, 2.4, 2.1kb), the templates for viral proteins and HBV-DNA synthesis. BA blocked HBV RNAs transcription possibly by down-regulating transcription and expression of HBV replication dependent hepatocyte nuclear factors (HNF1α and HNF4α). Thus, BA may benefit the anti-HBV therapy via inhibiting HBV viral RNAs.
Collapse
Affiliation(s)
- Hai Huang
- Department of Microbiology and Biopharmacy, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China.
| | - Wei Zhou
- Department of Chemistry, Fudan University, 220 Han Dan Road, Shanghai 200433, China.
| | - Haiyan Zhu
- Department of Microbiology and Biopharmacy, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China.
| | - Pei Zhou
- Department of Microbiology and Biopharmacy, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China.
| | - Xunlong Shi
- Department of Microbiology and Biopharmacy, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China.
| |
Collapse
|