1
|
Hales LT, Mountford SJ, Takawy M, Colledge D, Maher B, Shortt J, Thompson PE, Greenall SA, Warner N. VHL-independent degradation of hepatitis B virus e antigen (HBeAg) by VHL-binding chimeric small molecules. RSC Med Chem 2025:d5md00118h. [PMID: 40276592 PMCID: PMC12017376 DOI: 10.1039/d5md00118h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 03/22/2025] [Indexed: 04/26/2025] Open
Abstract
Hepatitis B virus (HBV) is a leading cause of liver cancer worldwide, with current treatment options unable to provide lasting efficacy against chronic infection. A key viral protein, HBV e antigen (HBeAg), plays an important role in suppressing the cellular and humoral immune response during infection and its loss is a precursor to clearance of chronic HBV infection. Its structural similarity to capsid forming HBV core protein antigen (HBcAg) makes it an intriguing, yet understudied target for pharmaceutical intervention. Recently, targeted protein degradation has been successfully applied against several viral proteins. This work investigates the targeting of HBeAg using heterobifunctional degraders derived from reported HBcAg ligands known to interact with HBeAg. Multiple compounds designed to recruit the VHL E3 ligase were found to be capable of reducing recombinant HBeAg protein levels in a HiBiT reporter assay system. Surprisingly, this decrease was found to be independent of VHL recruitment but driven by structural motifs of the VHL recruiting ligand, VH032. Virological assessment of these compounds against wildtype virus revealed an equipotent capability to reduce secreted HBeAg compared to the parental inhibitor, however increased efficacy was observed against an inhibitor resistant strain. Together, this work provides an initial description of the feasibility of converting HBV capsid-targeting ligands into degraders and provides evidence that such degraders may harbour improved activity against mutated forms of target which are resistant to parental compounds.
Collapse
Affiliation(s)
- Liam T Hales
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University Parkville 3052 Australia
| | - Simon J Mountford
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University Parkville 3052 Australia
| | - Mina Takawy
- Victorian Infectious Diseases Reference Laboratory, Royal Melbourne Hospital at The Peter Doherty, Institute for Infection and Immunity Melbourne 3000 Australia
| | - Danni Colledge
- Victorian Infectious Diseases Reference Laboratory, Royal Melbourne Hospital at The Peter Doherty, Institute for Infection and Immunity Melbourne 3000 Australia
| | - Belinda Maher
- Blood Cancer Therapeutics Laboratory, Department of Medicine, School of Clinical Sciences at Monash Health, Monash University Clayton 3168 Australia
- Monash Haematology, Monash Health Clayton 3168 Australia
| | - Jake Shortt
- Blood Cancer Therapeutics Laboratory, Department of Medicine, School of Clinical Sciences at Monash Health, Monash University Clayton 3168 Australia
- Monash Haematology, Monash Health Clayton 3168 Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne Parkville 3010 Australia
| | - Philip E Thompson
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University Parkville 3052 Australia
| | - Sam A Greenall
- Blood Cancer Therapeutics Laboratory, Department of Medicine, School of Clinical Sciences at Monash Health, Monash University Clayton 3168 Australia
| | - Nadia Warner
- Victorian Infectious Diseases Reference Laboratory, Royal Melbourne Hospital at The Peter Doherty, Institute for Infection and Immunity Melbourne 3000 Australia
| |
Collapse
|
2
|
Qiu Y, Tang Q, Liu XQ, Xue YL, Zeng Y, Hu P. Hepatitis B core-related antigen as a promising serological marker for monitoring hepatitis B virus cure. World J Hepatol 2025; 17:98658. [PMID: 39871916 PMCID: PMC11736480 DOI: 10.4254/wjh.v17.i1.98658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 11/25/2024] [Accepted: 12/13/2024] [Indexed: 01/06/2025] Open
Abstract
Hepatitis B virus (HBV) infection is a global health concern. The current sequential endpoints for the treatment of HBV infection include viral suppression, hepatitis B e antigen (HBeAg) seroconversion, functional cure, and covalently closed circular DNA (cccDNA) clearance. Serum hepatitis B core-related antigen (HBcrAg) is an emerging HBV marker comprising three components: HBeAg, hepatitis B core antigen, and p22cr. It responds well to the transcriptional activity of cccDNA in the patient's liver and is a promising alternative marker for serological testing. There is a strong correlation, and a decrease in its level corresponds to sustained viral suppression. In patients with chronic hepatitis B (CHB), serum HBcrAg levels are good predictors of HBeAg seroconversion (both spontaneous and after antiviral therapy), particularly in HBeAg-positive patients. Both low baseline HBcrAg levels and decreasing levels early in antiviral therapy favored HBsAg seroconversion, which may serve as a good surrogate option for treatment endpoints. In this review, we summarize the role of serum HBcrAg in the treatment of CHB. Therefore, long-term continuous monitoring of serum HBcrAg levels contributes to the clinical management of patients with CHB and optimizes the choice of treatment regimen, making it a promising marker for monitoring HBV cure.
Collapse
Affiliation(s)
- Yue Qiu
- Department of Infectious Diseases, Institute for Viral Hepatitis, The Key Laboratory of Molecular Biology for Infectious Diseases, Chinese Ministry of Education, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Qiao Tang
- Department of Infectious Diseases, Institute for Viral Hepatitis, The Key Laboratory of Molecular Biology for Infectious Diseases, Chinese Ministry of Education, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Xiao-Qing Liu
- Department of Infectious Diseases, Institute for Viral Hepatitis, The Key Laboratory of Molecular Biology for Infectious Diseases, Chinese Ministry of Education, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Yun-Ling Xue
- Department of Infectious Diseases, Institute for Viral Hepatitis, The Key Laboratory of Molecular Biology for Infectious Diseases, Chinese Ministry of Education, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Yi Zeng
- Department of Infectious Diseases, Institute for Viral Hepatitis, The Key Laboratory of Molecular Biology for Infectious Diseases, Chinese Ministry of Education, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Peng Hu
- Department of Infectious Diseases, Institute for Viral Hepatitis, The Key Laboratory of Molecular Biology for Infectious Diseases, Chinese Ministry of Education, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| |
Collapse
|
3
|
Hussain T, Zhao Z, Murphy B, Taylor ZE, Gudorf JA, Klein S, Barnes LF, VanNieuwenhze M, Jarrold MF, Zlotnick A. Chemically Tagging Cargo for Specific Packaging inside and on the Surface of Virus-like Particles. ACS NANO 2024; 18:21024-21037. [PMID: 39087909 PMCID: PMC11503556 DOI: 10.1021/acsnano.4c02056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Abstract
Virus-like particles (VLPs) have untapped potential for packaging and delivery of macromolecular cargo. To be a broadly useful platform, there needs to be a strategy for attaching macromolecules to the inside or the outside of the VLP with minimal modification of the platform or cargo. Here, we repurpose antiviral compounds that bind to hepatitis B virus (HBV) capsids to create a chemical tag to noncovalently attach cargo to the VLP. Our tag consists of a capsid assembly modulator, HAP13, connected to a linker terminating in maleimide. Our cargo is a green fluorescent protein (GFP) with a single addressable cysteine, a feature that can be engineered in many proteins. The HAP-GFP construct maintained HAP's intrinsic ability to bind HBV capsids and accelerate assembly. We investigated the capacity of HAP-GFP to coassemble with HBV capsid protein and bind to preassembled capsids. HAP-GFP binding was concentration-dependent, sensitive to capsid stability, and dependent on linker length. Long linkers had the greatest activity to bind capsids, while short linkers impeded assembly and damaged intact capsids. In coassembly reactions, >20 HAP-GFP molecules were presented on the outside and inside of the capsid, concentrating the cargo by more than 100-fold compared to bulk solution. We also tested an HAP-GFP with a cleavable linker so that external GFP molecules could be removed, resulting in exclusive internal packaging. These results demonstrate a generalizable strategy for attaching cargo to a VLP, supporting development of HBV as a modular VLP platform.
Collapse
Affiliation(s)
- Tariq Hussain
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Zhongchao Zhao
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Brennan Murphy
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Zachary E Taylor
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Jessica A Gudorf
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Shelby Klein
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Lauren F Barnes
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Michael VanNieuwenhze
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Martin F Jarrold
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Adam Zlotnick
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, Indiana 47405, United States
| |
Collapse
|
4
|
Li S, Zandi R. Biophysical Modeling of SARS-CoV-2 Assembly: Genome Condensation and Budding. Viruses 2022; 14:2089. [PMID: 36298645 PMCID: PMC9611094 DOI: 10.3390/v14102089] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/12/2022] [Accepted: 09/16/2022] [Indexed: 11/18/2022] Open
Abstract
The COVID-19 pandemic caused by the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has spurred unprecedented and concerted worldwide research to curtail and eradicate this pathogen. SARS-CoV-2 has four structural proteins: Envelope (E), Membrane (M), Nucleocapsid (N), and Spike (S), which self-assemble along with its RNA into the infectious virus by budding from intracellular lipid membranes. In this paper, we develop a model to explore the mechanisms of RNA condensation by structural proteins, protein oligomerization and cellular membrane-protein interactions that control the budding process and the ultimate virus structure. Using molecular dynamics simulations, we have deciphered how the positively charged N proteins interact and condense the very long genomic RNA resulting in its packaging by a lipid envelope decorated with structural proteins inside a host cell. Furthermore, considering the length of RNA and the size of the virus, we find that the intrinsic curvature of M proteins is essential for virus budding. While most current research has focused on the S protein, which is responsible for viral entry, and it has been motivated by the need to develop efficacious vaccines, the development of resistance through mutations in this crucial protein makes it essential to elucidate the details of the viral life cycle to identify other drug targets for future therapy. Our simulations will provide insight into the viral life cycle through the assembly of viral particles de novo and potentially identify therapeutic targets for future drug development.
Collapse
Affiliation(s)
- Siyu Li
- Songshan Lake Materials Laboratory, Dongguan 523808, China
| | - Roya Zandi
- Department of Physics and Astronomy, University of California Riverside, Riverside, CA 92521, USA
| |
Collapse
|
5
|
Ji X, Jiang X, Kobayashi C, Ren Y, Hu L, Gao Z, Kang D, Jia R, Zhang X, Zhao S, Watashi K, Liu X, Zhan P. Design, Synthesis, and Evaluation of a Set of Carboxylic Acid and Phosphate Prodrugs Derived from HBV Capsid Protein Allosteric Modulator NVR 3-778. Molecules 2022; 27:molecules27185987. [PMID: 36144715 PMCID: PMC9505734 DOI: 10.3390/molecules27185987] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/04/2022] [Accepted: 09/10/2022] [Indexed: 11/22/2022] Open
Abstract
Hepatitis B virus (HBV) capsid protein (Cp) is necessary for viral replication and the maintenance of viral persistence, having become an attractive target of anti-HBV drugs. To improve the water solubility of HBV capsid protein allosteric modulator (CpAM) NVR 3-778, a series of novel carboxylic acid and phosphate prodrugs were designed and synthesized using a prodrug strategy. In vitro HBV replication assay showed that these prodrugs maintained favorable antiviral potency (EC50 = 0.28−0.42 µM), which was comparable to that of NVR 3-778 (EC50 = 0.38 µM). More importantly, the cytotoxicity of prodrug N8 (CC50 > 256 µM) was significantly reduced compared to NVR 3-778 (CC50 = 13.65 ± 0.21 µM). In addition, the water solubility of prodrug N6 was hundreds of times better than that of NVR 3-778 in three phosphate buffers with various pH levels (2.0, 7.0, 7.4). In addition, N6 demonstrated excellent plasma and blood stability in vitro and good pharmacokinetic properties in rats. Finally, the hemisuccinate prodrug N6 significantly improved the candidate drug NVR 3-778’s water solubility and increased metabolic stability while maintaining its antiviral efficacy.
Collapse
Affiliation(s)
- Xiangkai Ji
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012, China
| | - Xiangyi Jiang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012, China
| | - Chisa Kobayashi
- Department of Virology II, National Institute of Infectious Diseases, Tokyo 163-8001, Japan
- Department of Applied Biological Science, Tokyo University of Science, Noda 278-8510, Japan
| | - Yujie Ren
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012, China
| | - Lide Hu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012, China
| | - Zhen Gao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012, China
| | - Dongwei Kang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012, China
| | - Ruifang Jia
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012, China
| | - Xujie Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012, China
| | - Shujie Zhao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012, China
| | - Koichi Watashi
- Department of Virology II, National Institute of Infectious Diseases, Tokyo 163-8001, Japan
- Department of Applied Biological Science, Tokyo University of Science, Noda 278-8510, Japan
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 163-8001, Japan
- Correspondence: (K.W.); (X.L.); (P.Z.)
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012, China
- Correspondence: (K.W.); (X.L.); (P.Z.)
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012, China
- Correspondence: (K.W.); (X.L.); (P.Z.)
| |
Collapse
|
6
|
Buzón P, Maity S, Christodoulis P, Wiertsema MJ, Dunkelbarger S, Kim C, Wuite GJ, Zlotnick A, Roos WH. Virus self-assembly proceeds through contact-rich energy minima. SCIENCE ADVANCES 2021; 7:eabg0811. [PMID: 34730996 PMCID: PMC8565845 DOI: 10.1126/sciadv.abg0811] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Self-assembly of supramolecular complexes such as viral capsids occurs prominently in nature. Nonetheless, the mechanisms underlying these processes remain poorly understood. Here, we uncover the assembly pathway of hepatitis B virus (HBV), applying fluorescence optical tweezers and high-speed atomic force microscopy. This allows tracking the assembly process in real time with single-molecule resolution. Our results identify a specific, contact-rich pentameric arrangement of HBV capsid proteins as a key on-path assembly intermediate and reveal the energy balance of the self-assembly process. Real-time nucleic acid packaging experiments show that a free energy change of ~1.4 kBT per condensed nucleotide is used to drive protein oligomerization. The finding that HBV assembly occurs via contact-rich energy minima has implications for our understanding of the assembly of HBV and other viruses and also for the development of new antiviral strategies and the rational design of self-assembling nanomaterials.
Collapse
Affiliation(s)
- Pedro Buzón
- Moleculaire Biofysica, Zernike Instituut, Rijksuniversiteit Groningen, Groningen, Netherlands
| | - Sourav Maity
- Moleculaire Biofysica, Zernike Instituut, Rijksuniversiteit Groningen, Groningen, Netherlands
| | | | - Monique J. Wiertsema
- Moleculaire Biofysica, Zernike Instituut, Rijksuniversiteit Groningen, Groningen, Netherlands
| | - Steven Dunkelbarger
- Molecular and Cellular Biochemistry Department, Indiana University, Bloomington, IN, USA
| | - Christine Kim
- Molecular and Cellular Biochemistry Department, Indiana University, Bloomington, IN, USA
| | - Gijs J.L. Wuite
- Physics of Living Systems, Vrije Universiteit, Amsterdam, Netherlands
| | - Adam Zlotnick
- Molecular and Cellular Biochemistry Department, Indiana University, Bloomington, IN, USA
| | - Wouter H. Roos
- Moleculaire Biofysica, Zernike Instituut, Rijksuniversiteit Groningen, Groningen, Netherlands
- Corresponding author.
| |
Collapse
|
7
|
Abstract
Hepatitis B virus (HBV) core protein (Cp) can be found in the nucleus and cytoplasm of infected hepatocytes; however, it preferentially segregates to a specific compartment correlating with disease status. Regulation of this intracellular partitioning of Cp remains obscure. In this paper, we report that cellular compartments are filled and vacated by Cp in a time- and concentration-dependent manner in both transfections and infections. At early times after transfection, Cp, in a dimeric state, preferentially localizes to the nucleolus. Later, the nucleolar compartment is emptied and Cp progresses to being predominantly nuclear, with a large fraction of the protein in an assembled state. Nuclear localization is followed by cell-wide distribution, and then Cp becomes exclusively cytoplasmic. The same trend in Cp movement is seen during an infection. Putative nucleolar retention signals have been identified and appear to be structure dependent. Export of Cp from the nucleus involves the CRM1 exportin. Time-dependent flux can be recapitulated by modifying Cp concentration, suggesting transitions are regulated by reaching a threshold concentration.
Collapse
|
8
|
Ren Y, Ma Y, Cherukupalli S, Tavis JE, Menéndez-Arias L, Liu X, Zhan P. Discovery and optimization of benzenesulfonamides-based hepatitis B virus capsid modulators via contemporary medicinal chemistry strategies. Eur J Med Chem 2020; 206:112714. [PMID: 32949990 DOI: 10.1016/j.ejmech.2020.112714] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 07/29/2020] [Accepted: 07/29/2020] [Indexed: 12/18/2022]
Abstract
Hepatitis B is a vaccine-preventable, but potentially life-threatening liver infection caused by the Hepatitis B virus (HBV). It represents an important health burden, with 257 million active cases globally. Current HBV treatments using nucleos(t)ide analogs and pegylated interferons cannot alleviate the situation completely since they are unable to cure the infection or reduce the amount of viral covalently closed circular DNA (cccDNA). The HBV core protein is a small protein of 183 amino acids that participates in multiple essential functions in the HBV replicative cycle. Capsid assembly modulators that target the core protein are being developed. Sulfonamides are synthetic functional groups, found in several drugs. Herein, we provide a concise report focusing on the sulfamoylbenzamides as HBV capsid modulators, and medicinal chemistry strategies used in their design and development.
Collapse
Affiliation(s)
- Yujie Ren
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, China
| | - Yue Ma
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, China
| | - Srinivasulu Cherukupalli
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, China
| | - John E Tavis
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO, USA; Saint Louis University Liver Center, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Luis Menéndez-Arias
- Centro de Biología Molecular "Severo Ochoa" (Consejo Superior de Investigaciones Científicas & Universidad Autónoma de Madrid), Madrid, Spain.
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, China.
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, China.
| |
Collapse
|
9
|
Buzón P, Maity S, Roos WH. Physical virology: From virus self-assembly to particle mechanics. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 12:e1613. [PMID: 31960585 PMCID: PMC7317356 DOI: 10.1002/wnan.1613] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 10/01/2019] [Accepted: 12/11/2019] [Indexed: 12/19/2022]
Abstract
Viruses are highly ordered supramolecular complexes that have evolved to propagate by hijacking the host cell's machinery. Although viruses are very diverse, spreading through cells of all kingdoms of life, they share common functions and properties. Next to the general interest in virology, fundamental viral mechanisms are of growing importance in other disciplines such as biomedicine and (bio)nanotechnology. However, in order to optimally make use of viruses and virus-like particles, for instance as vehicle for targeted drug delivery or as building blocks in electronics, it is essential to understand their basic chemical and physical properties and characteristics. In this context, the number of studies addressing the mechanisms governing viral properties and processes has recently grown drastically. This review summarizes a specific part of these scientific achievements, particularly addressing physical virology approaches aimed to understand the self-assembly of viruses and the mechanical properties of viral particles. Using a physicochemical perspective, we have focused on fundamental studies providing an overview of the molecular basis governing these key aspects of viral systems. This article is categorized under: Biology-Inspired Nanomaterials > Protein and Virus-Based Structures Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
- Pedro Buzón
- Moleculaire Biofysica, Zernike Instituut, Rijksuniversiteit Groningen, Groningen, The Netherlands
| | - Sourav Maity
- Moleculaire Biofysica, Zernike Instituut, Rijksuniversiteit Groningen, Groningen, The Netherlands
| | - Wouter H Roos
- Moleculaire Biofysica, Zernike Instituut, Rijksuniversiteit Groningen, Groningen, The Netherlands
| |
Collapse
|
10
|
Deffieu MS, Gaudin R. Imaging the Hepatitis B Virus: Broadcasting Live. Trends Microbiol 2019; 27:810-813. [PMID: 31421968 DOI: 10.1016/j.tim.2019.07.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 07/16/2019] [Accepted: 07/22/2019] [Indexed: 01/05/2023]
Abstract
Although important breakthroughs in our understanding of the hepatitis B virus (HBV) life cycle have been made since the discovery of its main entry factor, the spatiotemporal dynamics of HBV-host interactions remains understudied. Here, we discuss recent advances and continuing challenges to image the HBV life cycle in live cells.
Collapse
Affiliation(s)
- Maika S Deffieu
- Institut de Recherche en Infectiologie de Montpellier, CNRS, Univ Montpellier, 34293 Montpellier, France
| | - Raphael Gaudin
- Institut de Recherche en Infectiologie de Montpellier, CNRS, Univ Montpellier, 34293 Montpellier, France.
| |
Collapse
|
11
|
Rat V, Seigneuret F, Burlaud-Gaillard J, Lemoine R, Hourioux C, Zoulim F, Testoni B, Meunier JC, Tauber C, Roingeard P, de Rocquigny H. BAY 41-4109-mediated aggregation of assembled and misassembled HBV capsids in cells revealed by electron microscopy. Antiviral Res 2019; 169:104557. [PMID: 31302151 DOI: 10.1016/j.antiviral.2019.104557] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 07/05/2019] [Accepted: 07/10/2019] [Indexed: 02/07/2023]
Abstract
HBc is a small protein essential for the formation of the icosahedral HBV capsid. Its multiple roles in the replication cycle make this protein a promising target for the development of antiviral molecules. Based on the structure of HBc, a series of HBV assembly inhibitors, also known as capsid assembly modulators, were identified. We investigated the effect of BAY 41-4109, a heteroaryldihydropyrimidine derivative that promotes the assembly of a non-capsid polymer. We showed, by confocal microscopy, that BAY 41-4109 mediated HBc aggregation, mostly in the cytoplasm of Huh7 cells. Image analysis revealed that aggregate size depended on BAY 41-4109 concentration and treatment duration. Large aggregates in the vicinity of the nucleus were enclosed by invaginations of the nuclear envelope. This deformation of the nuclear envelope was confirmed by transmission electron microscopy (TEM) and immuno-TEM. These two techniques also revealed that the HBc aggregates were accumulations of capsid-like shells with an electron-dense material consisting of HBV core fragments. These findings, shedding light on the ultrastructural organization of HBc aggregates, provide insight into the mechanisms of action of BAY 41-4109 against HBV and will serve as a basis for comparison with other HBV capsid assembly inhibitors.
Collapse
Affiliation(s)
- Virgile Rat
- Morphogenèse et Antigénicité Du VIH et des Virus des Hépatites, Inserm - U1259 MAVIVH, Université de Tours et CHRU de Tours, 10 Boulevard Tonnellé - BP 3223, 37032, Tours Cedex 1, France
| | - Florian Seigneuret
- Morphogenèse et Antigénicité Du VIH et des Virus des Hépatites, Inserm - U1259 MAVIVH, Université de Tours et CHRU de Tours, 10 Boulevard Tonnellé - BP 3223, 37032, Tours Cedex 1, France
| | - Julien Burlaud-Gaillard
- Plate-Forme IBiSA des Microscopies, PPF ASB, Université de Tours and CHRU de Tours, 10 Boulevard Tonnellé - BP 3223, 37032, Tours Cedex 1, France
| | - Roxane Lemoine
- B-Cell Resources Platform, EA4245 "Transplantation, Immunologie et Inflammation", Université de Tours, 10 Boulevard Tonnellé, 37032, Tours Cedex 1, France
| | - Christophe Hourioux
- Morphogenèse et Antigénicité Du VIH et des Virus des Hépatites, Inserm - U1259 MAVIVH, Université de Tours et CHRU de Tours, 10 Boulevard Tonnellé - BP 3223, 37032, Tours Cedex 1, France; Plate-Forme IBiSA des Microscopies, PPF ASB, Université de Tours and CHRU de Tours, 10 Boulevard Tonnellé - BP 3223, 37032, Tours Cedex 1, France
| | - Fabien Zoulim
- INSERM U1052-Cancer Research Center of Lyon (CRCL), 69008, Lyon, France; University of Lyon, UMR_S1052, CRCL, 69008, Lyon, France; Department of Hepatology, Croix Rousse Hospital, Hospices Civils de Lyon, France
| | - Barbara Testoni
- INSERM U1052-Cancer Research Center of Lyon (CRCL), 69008, Lyon, France; University of Lyon, UMR_S1052, CRCL, 69008, Lyon, France; Department of Hepatology, Croix Rousse Hospital, Hospices Civils de Lyon, France
| | - Jean-Christophe Meunier
- Morphogenèse et Antigénicité Du VIH et des Virus des Hépatites, Inserm - U1259 MAVIVH, Université de Tours et CHRU de Tours, 10 Boulevard Tonnellé - BP 3223, 37032, Tours Cedex 1, France
| | - Clovis Tauber
- UMRS Inserm U1253 - Université de Tours, 10 Boulevard Tonnellé - BP 3223, 37032, Tours Cedex 1, France
| | - Philippe Roingeard
- Morphogenèse et Antigénicité Du VIH et des Virus des Hépatites, Inserm - U1259 MAVIVH, Université de Tours et CHRU de Tours, 10 Boulevard Tonnellé - BP 3223, 37032, Tours Cedex 1, France; Plate-Forme IBiSA des Microscopies, PPF ASB, Université de Tours and CHRU de Tours, 10 Boulevard Tonnellé - BP 3223, 37032, Tours Cedex 1, France
| | - Hugues de Rocquigny
- Morphogenèse et Antigénicité Du VIH et des Virus des Hépatites, Inserm - U1259 MAVIVH, Université de Tours et CHRU de Tours, 10 Boulevard Tonnellé - BP 3223, 37032, Tours Cedex 1, France.
| |
Collapse
|