1
|
Ugbaja SC, Mushebenge AGA, Kumalo H, Ngcobo M, Gqaleni N. Potential Benefits of In Silico Methods: A Promising Alternative in Natural Compound's Drug Discovery and Repurposing for HBV Therapy. Pharmaceuticals (Basel) 2025; 18:419. [PMID: 40143195 PMCID: PMC11944881 DOI: 10.3390/ph18030419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 01/30/2025] [Accepted: 03/13/2025] [Indexed: 03/28/2025] Open
Abstract
Hepatitis B virus (HBV) is an important global public health issue. The World Health Organization (WHO) 2024 Global Hepatitis Report estimated that the global prevalence of people living with HBV infection is 254 million, with an estimated prevalence incidence of 1.2 million new HBV infections yearly. Previous studies have shown that natural compounds have antiviral inhibition potentials. In silico methods such as molecular docking, virtual screening, pharmacophore modeling, quantitative structure-activity relationship (QSAR), and molecular dynamic simulations have been successfully applied in identifying bioactive compounds with strong binding energies in HBV treatment targets. The COVID-19 pandemic necessitated the importance of repurposing already approved drugs using in silico methods. This study is aimed at unveiling the benefits of in silico techniques as a potential alternative in natural compounds' drug discovery and repurposing for HBV therapy. Relevant articles from PubMed, Google Scholar, and Web of Science were retrieved and analyzed. Furthermore, this study comprehensively reviewed the literature containing identified bioactive compounds with strong inhibition of essential HBV proteins. Notably, hesperidin, quercetin, kaempferol, myricetin, and flavonoids have shown strong binding energies for hepatitis B surface antigen (HBsAg). The investigation reveals that in silico drug discovery methods offer an understanding of the mechanisms of action, reveal previously overlooked viral targets (including PreS1 Domain of HBsAg and cccDNA (Covalently Closed Circular DNA) regulators, and facilitate the creation of specific inhibitors. The integration of in silico, in vitro, and in vivo techniques is essential for the discovery of new drugs for HBV therapy. The insights further highlight the importance of natural compounds and in silico methods as targets in drug discovery for HBV therapy. Moreover, the combination of natural compounds, an in silico approach, and drug repurposing improves the chances of personalized and precision medicine in HBV treatment. Therefore, we recommend drug repurposing strategies that combine in vitro, in vivo, and in silico approaches to facilitate the discovery of effective HBV drugs.
Collapse
Affiliation(s)
- Samuel Chima Ugbaja
- Discipline of Traditional Medicine, School of Nursing and Public Health, University of KwaZulu Natal, Durban 4000, South Africa;
| | - Aganze Gloire-Aimé Mushebenge
- Department of Pharmacology, University of the Free State, Bloemfontein Campus, Bloemfontein 9301, South Africa;
- Faculty of Pharmaceutical Sciences, University of Lubumbashi, Lubumbashi 1825, Democratic Republic of the Congo
| | - Hezekiel Kumalo
- Drug Research and Innovation Unit, Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban 4000, South Africa;
| | - Mlungisi Ngcobo
- Discipline of Traditional Medicine, School of Nursing and Public Health, University of KwaZulu Natal, Durban 4000, South Africa;
| | - Nceba Gqaleni
- Discipline of Traditional Medicine, School of Nursing and Public Health, University of KwaZulu Natal, Durban 4000, South Africa;
| |
Collapse
|
2
|
Bradley DP, Valkner CJ, Li Q, Gasonoo M, Meyers MJ, Prifti GM, Moianos D, Zoidis G, Zlotnick A, Tavis JE. Discovery of bimodal hepatitis B virus ribonuclease H and capsid assembly inhibitors. PLoS Pathog 2025; 21:e1012920. [PMID: 39928681 PMCID: PMC11828405 DOI: 10.1371/journal.ppat.1012920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 02/14/2025] [Accepted: 01/17/2025] [Indexed: 02/12/2025] Open
Abstract
Hepatitis B virus (HBV) ribonuclease H (RNaseH) inhibitors are a potent class of antivirals that prevent degradation of the viral pregenomic RNA during reverse transcription and block formation of mature HBV DNAs. Development of HBV RNaseH inhibitors is entering advanced preclinical analyses. To ensure the mechanism of action was fully understood, we defined the effects of RNaseH inhibitors on other steps of HBV replication. Some N-hydroxypyridinedione (HPD) HBV RNaseH inhibitors significantly reduced accumulation of capsids in HBV-replicating cells. A representative HPD 1466, with a 50% effective concentration against HBV replication of 0.25 µM, decreased capsid and core protein accumulation by 50-90% in HepDES19 and HepG2.2.15 cells. Surprisingly, 1466 did not affect pregenomic RNA encapsidation, demonstrating a specific effect on empty capsids. HBV genomic replication was not necessary for 1466's inhibitory effect as it decreased capsid accumulation in cells transfected with replication-deficient mutants blocking pgRNA encapsidation (Δ-bulge), DNA synthesis (YMHA), and RNaseH (D702A) activities. 1466 also decreased capsid and core protein accumulation in cells transfected with a core protein expression plasmid, indicating that other HBV products are unneeded. 1466 reduced initial capsid assembly rates in biochemical assembly reactions employing purified core protein (Cp149), demonstrating a specific effect on HBV core protein. We conclude that the bimodal HPD HBV RNaseH inhibitor 1466 is the prototypic member of a new class of capsid assembly modulators (CAM) that inhibits capsid assembly rather than accelerating it, as all other CAM classes do. We propose that this class be called CAM-I, for CAM-inhibitor. These results lay the foundation for identifying bimodal HBV antivirals targeting the RNaseH and capsid assembly.
Collapse
Affiliation(s)
- Daniel P. Bradley
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Saint Louis, Missouri, United States of America
- Saint Louis University Institute for Drug and Biotherapeutic Innovation, Saint Louis, Missouri, United States of America
| | - Caleb J. Valkner
- Department of Molecular & Cellular Biology, Indiana University, Bloomington, Indiana, United States of America
| | - Qilan Li
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Saint Louis, Missouri, United States of America
- Saint Louis University Institute for Drug and Biotherapeutic Innovation, Saint Louis, Missouri, United States of America
| | - Makafui Gasonoo
- Department of Chemistry, Saint Louis University, Saint Louis, Missouri United States of America
| | - Marvin J. Meyers
- Saint Louis University Institute for Drug and Biotherapeutic Innovation, Saint Louis, Missouri, United States of America
- Department of Chemistry, Saint Louis University, Saint Louis, Missouri United States of America
| | - Georgia-Myrto Prifti
- Department of Pharmacy, Division of Pharmaceutical Chemistry, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, Athens, Greece
| | - Dimitrios Moianos
- Department of Pharmacy, Division of Pharmaceutical Chemistry, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, Athens, Greece
| | - Grigoris Zoidis
- Department of Pharmacy, Division of Pharmaceutical Chemistry, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, Athens, Greece
| | - Adam Zlotnick
- Department of Molecular & Cellular Biology, Indiana University, Bloomington, Indiana, United States of America
| | - John E. Tavis
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Saint Louis, Missouri, United States of America
- Saint Louis University Institute for Drug and Biotherapeutic Innovation, Saint Louis, Missouri, United States of America
| |
Collapse
|
3
|
Darvishi M, Amiri R, Ghannad E, Mehrabkhani S, Rastgar N, Razaghi M, Bansal J, Chahar M, Rajput P, Saffarfar H, Ali-Khiavi P, Mobed A, Yazdani Y. Nanodiagnostics in global eradication of hepatitis C virus. Clin Chim Acta 2025; 565:120013. [PMID: 39447823 DOI: 10.1016/j.cca.2024.120013] [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: 09/16/2024] [Revised: 10/20/2024] [Accepted: 10/20/2024] [Indexed: 10/26/2024]
Abstract
Hepatitis C, caused by the hepatitis C virus (HCV), is a prevalent liver disease with severe outcomes, including cirrhosis and hepatocellular carcinoma. Traditional diagnostic methods primarily detect antiviral antibodies (anti-HCV) or viral RNA, but these approaches have limitations. Anti-HCV antibodies may take 2-4 weeks to develop in acute cases and can be absent in some individuals, leading to undiagnosed early-stage infections. This poses significant challenges for public health, particularly in resource-limited settings where early detection is crucial. This article explores the development of biosensors engineered to directly detect HCV surface antigens, such as envelope proteins. These biosensors provide a promising solution for earlier and more accurate diagnosis by identifying viral components at the initial stages of infection. By focusing on direct detection of viral antigens, these innovations could enhance early diagnosis, facilitate timely intervention, and reduce virus transmission. We evaluate the advancements in biosensor technology over the past decade and their potential to improve HCV detection in clinical and field settings, ultimately supporting global efforts to eliminate HCV as a public health threat.
Collapse
Affiliation(s)
- Mohammad Darvishi
- Darvishi M. Associate Professor of Infectious Disease, School of Aerospace and Subaquatic Medicine, Infectious Diseases & Tropical Medicine Research Center (IDTMC), AJA University of Medical Sciences, Tehran, Iran
| | - Reza Amiri
- Rajiv Gandhi University of Health Sciences, Bengaluru, Karnataka, India
| | - Emad Ghannad
- Faculty of Pharmacy, Guilan University of Medical Sciences
| | - Samir Mehrabkhani
- Rajiv Gandhi University of Health Sciences, Bengaluru, Karnataka, India
| | - Nassim Rastgar
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahkameh Razaghi
- Faculty of Pharmacy, Shahid Beheshti University of Medical Sciences
| | - Jaya Bansal
- Chandigarh Pharmacy College, Chandigarh Group of Colleges, Jhanjeri, Mohali 140307, Punjab, India
| | - Mamata Chahar
- Department of Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - Pranchal Rajput
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, India
| | - Hossein Saffarfar
- Cardiovascular Research Center, Tehran, Tehran University of Medical Sciences, Tehran, Iran
| | - Payam Ali-Khiavi
- Medical faculty, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahmad Mobed
- Social Determinants of Health Research Center, Health Management and Safety Promotion Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Yalda Yazdani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
4
|
Ringlander J, Rydell GE, Kann M. From the Cytoplasm into the Nucleus-Hepatitis B Virus Travel and Genome Repair. Microorganisms 2025; 13:157. [PMID: 39858925 PMCID: PMC11767736 DOI: 10.3390/microorganisms13010157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/03/2025] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
Hepatitis B virus (HBV) is a major global health concern, affecting millions of people worldwide. HBV is part of the hepadnaviridae family and one of the primary causes of acute and chronic liver infections, leading to conditions such as cirrhosis and hepatocellular carcinoma (HCC). Understanding the intracellular transport and genome repair mechanisms of HBV is crucial for developing new drugs, which-in combination with immune modulators-may contribute to potential cures. This review will explore the current knowledge of HBV intracytoplasmic and nuclear transport, as well as genome repair processes, while drawing comparisons to other viruses with nuclear replication.
Collapse
Affiliation(s)
- Johan Ringlander
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 41346 Gothenburg, Sweden; (J.R.); (G.E.R.)
- Department of Clinical Microbiology, Region Västra Götaland, Sahlgrenska University Hospital, 41346 Gothenburg, Sweden
| | - Gustaf E. Rydell
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 41346 Gothenburg, Sweden; (J.R.); (G.E.R.)
| | - Michael Kann
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 41346 Gothenburg, Sweden; (J.R.); (G.E.R.)
- Department of Clinical Microbiology, Region Västra Götaland, Sahlgrenska University Hospital, 41346 Gothenburg, Sweden
| |
Collapse
|
5
|
Chuang YC, Ou JHJ. Hepatitis B virus entry, assembly, and egress. Microbiol Mol Biol Rev 2024; 88:e0001424. [PMID: 39440957 DOI: 10.1128/mmbr.00014-24] [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] [Indexed: 10/25/2024] Open
Abstract
SUMMARYHepatitis B virus (HBV) is an important human pathogen that chronically infects approximately 250 million people in the world, resulting in ~1 million deaths annually. This virus is a hepatotropic virus and can cause severe liver diseases including cirrhosis and hepatocellular carcinoma. The entry of HBV into hepatocytes is initiated by the interaction of its envelope proteins with its receptors. This is followed by the delivery of the viral nucleocapsid to the nucleus for the release of its genomic DNA and the transcription of viral RNAs. The assembly of the viral capsid particles may then take place in the nucleus or the cytoplasm and may involve cellular membranes. This is followed by the egress of the virus from infected cells. In recent years, significant research progresses had been made toward understanding the entry, the assembly, and the egress of HBV particles. In this review, we discuss the molecular pathways of these processes and compare them with those used by hepatitis delta virus and hepatitis C virus , two other hepatotropic viruses that are also enveloped. The understanding of these processes will help us to understand how HBV replicates and causes diseases, which will help to improve the treatments for HBV patients.
Collapse
Affiliation(s)
- Yu-Chen Chuang
- Department of Molecular Microbiology and Immunology, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - J-H James Ou
- Department of Molecular Microbiology and Immunology, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| |
Collapse
|
6
|
Leclercq L. Law and Order of Colloidal Tectonics: From Molecules to Self-Assembled Colloids. Molecules 2024; 29:5657. [PMID: 39683815 DOI: 10.3390/molecules29235657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Since biochemists and biologists have progressed in understanding the mechanisms involved in living organisms, biological systems have become a source of inspiration for chemists. In this context, the concept of colloidal tectonics, describing the spontaneous formation of colloidal particles or supracolloidal structures in which the building blocks are called "tectons", has emerged. Therefore, a bottom-up edification of tectons towards (supra) colloidal structures is allowed. Each (supra) colloidal system has at least one of the following properties: amphiphilicity, predictability, versatility, commutability, and reversibility. However, for these systems to perform even more interesting functions, it is necessary for tectons to have very precise chemical and physical properties so that new properties emerge in (supra) colloidal systems. In this way, colloidal tectonics enables engineering at the nano- and micrometric level and contributes to the development of smart bioinspired systems with applications in catalysis, drug delivery, etc. In this review, an overview of the concept of colloidal tectonics is illustrated by some biotic systems. The design of abiotic (supra) colloidal systems and their applications in various fields are also addressed (notably Pickering emulsions for catalysis or drug delivery). Finally, theoretical directions for the design of novel self-assembled (supra) colloidal systems are discussed.
Collapse
Affiliation(s)
- Loïc Leclercq
- Univ. Lille, CNRS, Centrale Lille, Univ. Artois, UMR 8181-UCCS, Unité de Catalyse et Chimie du Solide, Lille 59000, France
| |
Collapse
|
7
|
Dunkoksung W, Udomnilobol U, Ruengsatra T, Chauypen N, Prueksaritanont T. Preclinical characterization of a novel potent core protein assembly modulator for the treatment of chronic hepatitis B viral infection. Eur J Pharm Sci 2024; 200:106834. [PMID: 38906232 DOI: 10.1016/j.ejps.2024.106834] [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: 03/20/2024] [Revised: 05/11/2024] [Accepted: 06/16/2024] [Indexed: 06/23/2024]
Abstract
The hepatitis B virus (HBV) capsid or core protein is a promising drug target currently being investigated for potential curative therapies for chronic HBV infection. In this study, we performed extensive in vitro and in vivo characterization of a novel and potent HBV core protein assembly modulator (CpAM), CU15, for both anti-HBV activity and druggability properties. CU15 potently inhibited HBV DNA replication in in vitro HBV-infected HepG2.2.15 cells (EC50 of 8.6 nM), with a low serum shift. It was also effective in inhibiting HBV DNA and cccDNA formation in de novo HBV-infected primary human hepatocytes. Furthermore, CU15 was active across several HBV genotypes and across clinically relevant core protein variants. After oral administration to an in vivo HBV mouse model, CU15 significantly reduced plasma HBV DNA and RNA levels, at plasma exposure consistent with the estimated in vitro potency. In vitro, CU15 exhibited excellent passive permeability and relatively high metabolic stability in liver preparations across species (human > dog> rat). In vitro human liver microsomal studies suggest that the compound's major metabolic pathway is CYP3A-mediated oxidation. Consistent with the in vitro findings, CU15 is a compound with a low-to-moderate clearance and high oral bioavailability in rats and dogs. Based on the apparent in vitro-in vivo correlation observed, CU15 has the potential to exhibit low clearance and high oral bioavailability in humans. In addition, CU15 also showed low drug-drug interaction liability with an acceptable in vitro safety profile (IC50 > 10 µM).
Collapse
Affiliation(s)
- Wilasinee Dunkoksung
- Chulalongkorn University Drug Discovery and Drug Development Research Center (Chula4DR), Chulalongkorn University, Bangkok, Thailand
| | - Udomsak Udomnilobol
- Chulalongkorn University Drug Discovery and Drug Development Research Center (Chula4DR), Chulalongkorn University, Bangkok, Thailand
| | - Tanachote Ruengsatra
- Chulalongkorn University Drug Discovery and Drug Development Research Center (Chula4DR), Chulalongkorn University, Bangkok, Thailand
| | - Natthaya Chauypen
- Center of Excellence in Hepatitis and Liver Cancer, Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Thomayant Prueksaritanont
- Chulalongkorn University Drug Discovery and Drug Development Research Center (Chula4DR), Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
8
|
Bendahmane S, Follo M, Zhang F, Linhardt RJ. Towards Cell-Permeable Hepatitis B Virus Core Protein Variants as Potential Antiviral Agents. Microorganisms 2024; 12:1776. [PMID: 39338451 PMCID: PMC11434381 DOI: 10.3390/microorganisms12091776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/17/2024] [Accepted: 08/01/2024] [Indexed: 09/30/2024] Open
Abstract
Hepatitis B virus (HBV) infection remains a major health threat with limited treatment options. One of various new antiviral strategies is based on a fusion of Staphylococcus aureus nuclease (SN) with the capsid-forming HBV core protein (HBc), termed coreSN. Through co-assembly with wild-type HBc-subunits, the fusion protein is incorporated into HBV nucleocapsids, targeting the nuclease to the encapsidated viral genome. However, coreSN expression was based on transfection of a plasmid vector. Here, we explored whether introducing protein transduction domains (PTDs) into a fluorescent coreSN model could confer cell-penetrating properties for direct protein delivery into cells. Four PTDs were inserted into two different positions of the HBc sequence, comprising the amphiphilic translocation motif (TLM) derived from the HBV surface protein PreS2 domain and three basic PTDs derived from the Tat protein of human immunodeficiency virus-1 (HIV-1), namely Tat4, NP, and NS. To directly monitor the interaction with cells, the SN in coreSN was replaced with the green fluorescent protein (GFP). The fusion proteins were expressed in E. coli, and binding to and potential uptake by human cells was examined through flow cytometry and fluorescence microscopy. The data indicate PTD-dependent interactions with the cells, with evidence of uptake in particular for the basic PTDs. Uptake was enhanced by a triplicated Simian virus 40 (SV40) large T antigen nuclear localization signal (NLS). Interestingly, the basic C terminal domain of the HBV core protein was found to function as a novel PTD. Hence, further developing cell-permeable viral capsid protein fusions appears worthwhile.
Collapse
Affiliation(s)
- Sanaa Bendahmane
- Private Faculty of Health Professions and Technologies, Private University of Marrakech, Marrakech 42312, Morocco
| | - Marie Follo
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79085 Freiburg, Germany
| | - Fuming Zhang
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Robert J Linhardt
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| |
Collapse
|
9
|
Inoue T, Yagi S, Tanaka Y. Two Concepts of Hepatitis B Core-Related Antigen Assay: A Highly Sensitive and Rapid Assay or an Effective Tool for Widespread Screening. Viruses 2024; 16:848. [PMID: 38932141 PMCID: PMC11209401 DOI: 10.3390/v16060848] [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: 04/10/2024] [Revised: 05/20/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
Hepatitis B core-related antigen (HBcrAg) reflects the activity of intrahepatic covalently closed circular DNA. HBcrAg can be detected even in chronic hepatitis B patients in whom serum HBV DNA or hepatitis B surface antigen is undetectable. The HBcrAg measurement system was developed based on two concepts. One is a fully-automated and highly-sensitive HBcrAg assay (iTACT-HBcrAg) and the other is a point-of-care testing (POCT) that can be used in in resource-limited areas. iTACT-HBcrAg is an alternative to HBV DNA for monitoring HBV reactivation and predicting the development of hepatocellular carcinoma. This validated biomarker is available in routine clinical practice in Japan. Currently, international guidelines for the prevention of mother-to-child transmission recommend anti-HBV prophylaxis for pregnant women with high viral loads. However, over 95% of HBV-infected individuals live in countries where HBV DNA quantification is widely unavailable. Given this situation, a rapid and simple HBcrAg assay for POCT would be highly effective. Long-term anti-HBV therapy may have potential side effects and appropriate treatment should be provided to eligible patients. Therefore, a simple method of determining the indication for anti-HBV treatment would be ideal. This review provides up-to-date information regarding the clinical value of HBcrAg in HBV management, based on iTACT-HBcrAg or POCT.
Collapse
Affiliation(s)
- Takako Inoue
- Department of Clinical Laboratory Medicine, Nagoya City University Hospital, Nagoya 467-8602, Japan;
| | - Shintaro Yagi
- Research and Development Department, Advanced Life Science Institute, Inc., Hachioji 192-0031, Japan;
| | - Yasuhito Tanaka
- Department of Gastroenterology and Hepatology, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| |
Collapse
|
10
|
Dörnbrack K, Beck J, Nassal M. Structural Analysis of the Hepatitis B Virus RNA Encapsidation Signal ε by Selective 2'-Hydroxyl Acylation Analyzed by Primer Extension (SHAPE). Methods Mol Biol 2024; 2837:67-87. [PMID: 39044076 DOI: 10.1007/978-1-0716-4027-2_7] [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] [Indexed: 07/25/2024]
Abstract
RNA structure is crucial for RNA function, including in viral cis-elements such as the hepatitis B virus (HBV) RNA encapsidation signal ε. Interacting with the viral polymerase ε mediates packaging of the pregenomic (pg) RNA into capsids, initiation of reverse transcription, and it affects the mRNA functions of pgRNA. As free RNA, the 61-nucleotide (nt) ε sequence adopts a bipartite stem-loop structure with a central bulge and an apical loop. Due to stable Watson-Crick base pairing, this was already predicted by early RNA folding programs and confirmed by classical enzymatic and chemical structure probing. A newer, high-resolution probing technique exploits the selective acylation of solvent-accessible 2'-hydroxyls in the RNA backbone by electrophilic compounds such as 2-methylnicotinic acid imidazolide (NAI), followed by mapping of the modified sites by primer extension. This SHAPE principle has meanwhile been extended to numerous applications. Here we provide a basic protocol for NAI-based SHAPE of isolated HBV ε RNA which already provided insights into the impact of mutations, and preliminarily, of polymerase binding on the RNA structural dynamics. While the focus is on NAI modification, we also briefly cover target RNA preparation by in vitro transcription, primer extension using a radiolabeled primer, and analysis of the resulting cDNAs by denaturing polyacrylamide gelelectrophoresis (PAGE). Given the high tolerance of SHAPE chemistry to different conditions, including applicability in live cells, we expect this technique to greatly facilitate deciphering the conformational dynamics underlying the various functions of the ε element, especially in concert with the recently solved three-dimensional structure of the free RNA.
Collapse
Affiliation(s)
- Katharina Dörnbrack
- Clinical Trials Unit of the Medical Center, University of Freiburg, Freiburg, Germany
| | - Jürgen Beck
- Internal Medicine II/Molecular Biology, University Hospital Freiburg, Freiburg, Germany
| | - Michael Nassal
- Internal Medicine II/Molecular Biology, University Hospital Freiburg, Freiburg, Germany.
| |
Collapse
|
11
|
Wang M, Zhang J, Dou Y, Liang M, Xie Y, Xue P, Liu L, Li C, Wang Y, Tao F, Zhang X, Hu H, Feng K, Zhang L, Wu Z, Chen Y, Zhan P, Jia H. Design, Synthesis, and Biological Evaluation of Novel Thioureidobenzamide (TBA) Derivatives as HBV Capsid Assembly Modulators. J Med Chem 2023; 66:13968-13990. [PMID: 37839070 DOI: 10.1021/acs.jmedchem.3c01022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
Hepatitis B virus (HBV) capsid assembly modulators (CAMs) represent a promising therapeutic approach for the treatment of HBV infection. In this study, we designed and synthesized five series of benzamide derivatives based on a multisite-binding strategy at the tolerant region and diversity modification in the solvent-exposed region. Among them, thioureidobenzamide compound 17i exhibited significantly increased anti-HBV activity in HepAD38 (EC50 = 0.012 μM) and HBV-infected HLCZ01 cells (EC50 = 0.033 μM). Moreover, 17i displayed a better inhibitory effect on the assembly of HBV capsid protein compared with NVR 3-778 and a inhibitory effect similar to the clinical drug GLS4. In addition, 17i showed moderate metabolic stability in human microsomes, had excellent oral bioavailability in Sprague-Dawley (SD) rats, and inhibited HBV replication in the HBV carrier mice model, which could be considered as a promising candidate drug for further development.
Collapse
Affiliation(s)
- Mei Wang
- School of Pharmacy, Weifang Medical University, Weifang 261053, Shandong, P. R. China
| | - Jian Zhang
- School of Pharmacy, Weifang Medical University, Weifang 261053, Shandong, P. R. China
| | - Yutong Dou
- Key Laboratory for Experimental Teratology of Ministry of Education, Key Laboratory of Infection and Immunity of Shandong Province and Dept. Immunology, School of Basic Medical Sciences, Qilu Hospital, Cheeloo Medical College, Shandong University, Jinan 250012, Shandong, P. R. China
| | - Minghui Liang
- School of Pharmacy, Weifang Medical University, Weifang 261053, Shandong, P. R. China
| | - Yong Xie
- State Key Laboratory of Anti-Infective Drug Development (NO. 2015DQ780357), Sunshine Lake Pharma Co., Ltd, Dongguan 523871, P. R. China
| | - Peng Xue
- School of Public Health, Weifang Medical University, Weifang 261053, Shandong, P. R. China
| | - Linyue Liu
- School of Pharmacy, Weifang Medical University, Weifang 261053, Shandong, P. R. China
| | - Chuanju Li
- Department of Pharmacy, Affiliated Hospital of Jining Medical University, Jining 272000, Shandong, P. R. China
| | - Yuanze Wang
- Guangzhou Laboratory, Guangzhou International Bio Island, Guangzhou 510000, Guangdong, P. R. China
| | - Feiyan Tao
- School of Public Health, Weifang Medical University, Weifang 261053, Shandong, P. R. China
| | - Xiaohui Zhang
- Key Laboratory of Experimental Teratology, Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Qilu Hospital, Cheeloo Medical College, Shandong University, Jinan 250012, P. R. China
- The Research Center of Stem Cell and Regenerative Medicine, Department of Systems Biomedicine, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, P. R. China
| | - Huili Hu
- Key Laboratory of Experimental Teratology, Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Qilu Hospital, Cheeloo Medical College, Shandong University, Jinan 250012, P. R. China
- The Research Center of Stem Cell and Regenerative Medicine, Department of Systems Biomedicine, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, P. R. China
| | - Kairui Feng
- School of Pharmacy, Weifang Medical University, Weifang 261053, Shandong, P. R. China
| | - Lei Zhang
- School of Pharmacy, Weifang Medical University, Weifang 261053, Shandong, P. R. China
| | - Zhuanchang Wu
- Key Laboratory for Experimental Teratology of Ministry of Education, Key Laboratory of Infection and Immunity of Shandong Province and Dept. Immunology, School of Basic Medical Sciences, Qilu Hospital, Cheeloo Medical College, Shandong University, Jinan 250012, Shandong, P. R. China
| | - Yunfu Chen
- State Key Laboratory of Anti-Infective Drug Development (NO. 2015DQ780357), Sunshine Lake Pharma Co., Ltd, Dongguan 523871, P. R. China
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, P. R. China
| | - Haiyong Jia
- School of Pharmacy, Weifang Medical University, Weifang 261053, Shandong, P. R. China
| |
Collapse
|
12
|
Olenginski LT, Attionu SK, Henninger EN, LeBlanc RM, Longhini AP, Dayie TK. Hepatitis B Virus Epsilon (ε) RNA Element: Dynamic Regulator of Viral Replication and Attractive Therapeutic Target. Viruses 2023; 15:1913. [PMID: 37766319 PMCID: PMC10534774 DOI: 10.3390/v15091913] [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: 07/01/2023] [Revised: 09/01/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
Hepatitis B virus (HBV) chronically infects millions of people worldwide, which underscores the importance of discovering and designing novel anti-HBV therapeutics to complement current treatment strategies. An underexploited but attractive therapeutic target is ε, a cis-acting regulatory stem-loop RNA situated within the HBV pregenomic RNA (pgRNA). The binding of ε to the viral polymerase protein (P) is pivotal, as it triggers the packaging of pgRNA and P, as well as the reverse transcription of the viral genome. Consequently, small molecules capable of disrupting this interaction hold the potential to inhibit the early stages of HBV replication. The rational design of such ligands necessitates high-resolution structural information for the ε-P complex or its individual components. While these data are currently unavailable for P, our recent structural elucidation of ε through solution nuclear magnetic resonance spectroscopy marks a significant advancement in this area. In this review, we provide a brief overview of HBV replication and some of the therapeutic strategies to combat chronic HBV infection. These descriptions are intended to contextualize our recent experimental efforts to characterize ε and identify ε-targeting ligands, with the ultimate goal of developing novel anti-HBV therapeutics.
Collapse
Affiliation(s)
- Lukasz T. Olenginski
- Center for Biomolecular Structure and Organization, Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742, USA (R.M.L.)
- Department of Biochemistry, University of Colorado, Boulder, CO 80309, USA
| | - Solomon K. Attionu
- Center for Biomolecular Structure and Organization, Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742, USA (R.M.L.)
| | - Erica N. Henninger
- Center for Biomolecular Structure and Organization, Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742, USA (R.M.L.)
| | - Regan M. LeBlanc
- Center for Biomolecular Structure and Organization, Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742, USA (R.M.L.)
| | - Andrew P. Longhini
- Center for Biomolecular Structure and Organization, Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742, USA (R.M.L.)
- Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
- Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | - Theodore K. Dayie
- Center for Biomolecular Structure and Organization, Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742, USA (R.M.L.)
| |
Collapse
|
13
|
McFadden WM, Sarafianos SG. Biology of the hepatitis B virus (HBV) core and capsid assembly modulators (CAMs) for chronic hepatitis B (CHB) cure. Glob Health Med 2023; 5:199-207. [PMID: 37655181 PMCID: PMC10461335 DOI: 10.35772/ghm.2023.01065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/03/2023] [Accepted: 06/30/2023] [Indexed: 09/02/2023]
Abstract
Hepatitis B virus (HBV) is a hepadnavirus, a small DNA virus that infects liver tissue, with some unusual replication steps that share similarities to retroviruses. HBV infection can lead to chronic hepatitis B (CHB), a life-long infection associated with significant risks of liver disease, especially if untreated. HBV is a significant global health problem, with hundreds of millions currently living with CHB. Currently approved strategies to prevent or inhibit HBV are highly effective, however, a cure for CHB has remained elusive. To achieve a cure, elimination of the functionally integrated HBV covalently closed chromosomal DNA (cccDNA) genome is required. The capsid core is an essential component of HBV replication, serving roles when establishing infection and in creating new virions. Over the last two and a half decades, significant efforts have been made to find and characterize antivirals that target the capsid, specifically the HBV core protein (Cp). The antivirals that interfere with the kinetics and morphology of the capsid, termed capsid assembly modulators (CAMs), are extremely potent, and clinical investigations indicate they are well tolerated and highly effective. Several CAMs offer the potential to cure CHB by decreasing the cccDNA pools. Here, we review the biology of the HBV capsid, focused on Cp, and the development of inhibitors that target it.
Collapse
Affiliation(s)
- William M. McFadden
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Stefan G. Sarafianos
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Children's Healthcare of Atlanta, Atlanta, GA, USA
| |
Collapse
|
14
|
Basit L, Amblard F, Patel DJ, Biteau N, Chen Z, Kasthuri M, Zhou S, Schinazi RF. The premise of capsid assembly modulators towards eliminating HBV persistence. Expert Opin Drug Discov 2023; 18:1031-1041. [PMID: 37477111 PMCID: PMC10530454 DOI: 10.1080/17460441.2023.2239701] [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: 04/04/2023] [Accepted: 07/19/2023] [Indexed: 07/22/2023]
Abstract
INTRODUCTION The burden of chronic hepatitis B virus (HBV) results in almost a million deaths per year. The most common treatment for chronic hepatitis B infection is long-term nucleoside analogs (NUC) or one-year interferon-alpha (pegylated or non-pegylated) therapy before or after NUC therapy. Unfortunately, these therapies rarely result in HBV functional cure because they do not eradicate HBV from the nucleus of the hepatocytes, where the covalently closed circular DNA (cccDNA) is formed and/or where the integrated HBV DNA persists in the host genome. Hence, the search continues for novel antiviral therapies that target different steps of the HBV replication cycle to cure chronically infected HBV individuals and eliminate HBV from the liver reservoirs. AREAS COVERED The authors focus on capsid assembly modulators (CAMs). These molecules are unique because they impact not only one but several steps of HBV viral replication, including capsid assembly, capsid trafficking into the nucleus, reverse transcription, pre-genomic RNA (pgRNA), and polymerase protein co-packaging. EXPERT OPINION Mono- or combination therapy, including CAMs with other HBV drugs, may potentially eliminate hepatitis B infections. Nevertheless, more data on their potential effect on HBV elimination is needed, especially when used daily for 6-12 months.
Collapse
Affiliation(s)
- Leda Basit
- Center for ViroScience and Cure, Laboratory of Biochemical
Pharmacology, Department of Pediatrics, Emory University School of Medicine and
Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322,
USA
| | - Franck Amblard
- Center for ViroScience and Cure, Laboratory of Biochemical
Pharmacology, Department of Pediatrics, Emory University School of Medicine and
Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322,
USA
| | - Dharmeshkumar J. Patel
- Center for ViroScience and Cure, Laboratory of Biochemical
Pharmacology, Department of Pediatrics, Emory University School of Medicine and
Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322,
USA
| | - Nicolas Biteau
- Center for ViroScience and Cure, Laboratory of Biochemical
Pharmacology, Department of Pediatrics, Emory University School of Medicine and
Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322,
USA
| | - Zhe Chen
- Center for ViroScience and Cure, Laboratory of Biochemical
Pharmacology, Department of Pediatrics, Emory University School of Medicine and
Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322,
USA
| | - Mahesh Kasthuri
- Center for ViroScience and Cure, Laboratory of Biochemical
Pharmacology, Department of Pediatrics, Emory University School of Medicine and
Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322,
USA
| | - Shaoman Zhou
- Center for ViroScience and Cure, Laboratory of Biochemical
Pharmacology, Department of Pediatrics, Emory University School of Medicine and
Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322,
USA
| | - Raymond F. Schinazi
- Center for ViroScience and Cure, Laboratory of Biochemical
Pharmacology, Department of Pediatrics, Emory University School of Medicine and
Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322,
USA
| |
Collapse
|
15
|
Hofmann S, Plank V, Groitl P, Skvorc N, Hofmann K, Luther J, Ko C, Zimmerman P, Bruss V, Stadler D, Carpentier A, Rezk S, Nassal M, Protzer U, Schreiner S. SUMO Modification of Hepatitis B Virus Core Mediates Nuclear Entry, Promyelocytic Leukemia Nuclear Body Association, and Efficient Formation of Covalently Closed Circular DNA. Microbiol Spectr 2023; 11:e0044623. [PMID: 37199632 PMCID: PMC10269885 DOI: 10.1128/spectrum.00446-23] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 04/24/2023] [Indexed: 05/19/2023] Open
Abstract
Persistence of hepatitis B virus (HBV) infection is due to a nuclear covalently closed circular DNA (cccDNA), generated from the virion-borne relaxed circular DNA (rcDNA) genome in a process likely involving numerous cell factors from the host DNA damage response (DDR). The HBV core protein mediates rcDNA transport to the nucleus and likely affects stability and transcriptional activity of cccDNA. Our study aimed at investigating the role of HBV core protein and its posttranslational modification (PTM) with SUMO (small ubiquitin-like modifiers) during the establishment of cccDNA. HBV core protein SUMO PTM was analyzed in His-SUMO-overexpressing cell lines. The impact of HBV core SUMOylation on association with cellular interaction partners and on the HBV life cycle was determined using SUMOylation-deficient mutants of the HBV core protein. Here, we show that the HBV core protein is posttranslationally modified by the addition of SUMO and that this modification impacts nuclear import of rcDNA. By using SUMOylation-deficient HBV core mutants, we show that SUMO modification is a prerequisite for the association with specific promyelocytic leukemia nuclear bodies (PML-NBs) and regulates the conversion of rcDNA to cccDNA. By in vitro SUMOylation of HBV core, we obtained evidence that SUMOylation triggers nucleocapsid disassembly, providing novel insights into the nuclear import process of rcDNA. HBV core protein SUMOylation and subsequent association with PML bodies in the nucleus constitute a key step in the conversion of HBV rcDNA to cccDNA and therefore a promising target for inhibiting formation of the HBV persistence reservoir. IMPORTANCE HBV cccDNA is formed from the incomplete rcDNA involving several host DDR proteins. The exact process and the site of cccDNA formation are poorly understood. Here, we show that HBV core protein SUMO modification is a novel PTM regulating the function of HBV core. A minor specific fraction of the HBV core protein resides with PML-NBs in the nuclear matrix. SUMO modification of HBV core protein mediates its recruitment to specific PML-NBs within the host cell. Within HBV nucleocapsids, SUMOylation of HBV core induces HBV capsid disassembly and is a prerequisite for nuclear entry of HBV core. SUMO HBV core protein association with PML-NBs is crucial for efficient conversion of rcDNA to cccDNA and for the establishment of the viral persistence reservoir. HBV core protein SUMO modification and the subsequent association with PML-NBs might constitute a potential novel target in the development of drugs targeting the cccDNA.
Collapse
Affiliation(s)
- Samuel Hofmann
- Institute of Virology, School of Medicine, Technical University of Munich, Germany
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Verena Plank
- Institute of Virology, School of Medicine, Technical University of Munich, Germany
| | - Peter Groitl
- Institute of Virology, School of Medicine, Technical University of Munich, Germany
| | - Nathalie Skvorc
- Institute of Virology, School of Medicine, Technical University of Munich, Germany
| | - Katharina Hofmann
- Institute of Virology, School of Medicine, Technical University of Munich, Germany
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Julius Luther
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Chunkyu Ko
- Institute of Virology, School of Medicine, Technical University of Munich, Germany
- Institute of Virology, Helmholtz Zentrum München, Munich, Germany
| | - Peter Zimmerman
- Department of Internal Medicine II/Molecular Biology, University Hospital Freiburg, Freiburg, Germany
| | - Volker Bruss
- Institute of Virology, Helmholtz Zentrum München, Munich, Germany
| | - Daniela Stadler
- Institute of Virology, School of Medicine, Technical University of Munich, Germany
- Institute of Virology, Helmholtz Zentrum München, Munich, Germany
| | | | - Shahinda Rezk
- Institute of Virology, School of Medicine, Technical University of Munich, Germany
- Medical Research Institute, Department of Molecular and Diagnostic Microbiology, Alexandria University, Alexandria, Egypt
| | - Michael Nassal
- Department of Internal Medicine II/Molecular Biology, University Hospital Freiburg, Freiburg, Germany
| | - Ulrike Protzer
- Institute of Virology, School of Medicine, Technical University of Munich, Germany
- Institute of Virology, Helmholtz Zentrum München, Munich, Germany
- German Center for Infection Research, Munich, Germany
| | - Sabrina Schreiner
- Institute of Virology, School of Medicine, Technical University of Munich, Germany
- Institute of Virology, Hannover Medical School, Hannover, Germany
- Institute of Virology, Helmholtz Zentrum München, Munich, Germany
- German Center for Infection Research, Munich, Germany
- Cluster of Excellence RESIST (Resolving Infection Susceptibility; EXC 2155), Hannover Medical School, Hannover, Germany
| |
Collapse
|
16
|
Nassal M. How many steps to a feasible mouse model of hepatitis B virus infection? Hepatology 2023; 77:1090-1094. [PMID: 35871466 DOI: 10.1002/hep.32684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 07/20/2022] [Indexed: 12/08/2022]
Affiliation(s)
- Michael Nassal
- Department of Internal Medicine 2/Molecular Biology , University Hospital Freiburg , Freiburg , Germany
| |
Collapse
|
17
|
Starr CA, Nair S, Huang SY, Hagan MF, Jacobson SC, Zlotnick A. Engineering Metastability into a Virus-like Particle to Enable Triggered Dissociation. J Am Chem Soc 2023; 145:2322-2331. [PMID: 36651799 PMCID: PMC10018796 DOI: 10.1021/jacs.2c10937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
For a virus-like particle (VLP) to serve as a delivery platform, the VLP must be able to release its cargo in response to a trigger. Here, we use a chemical biology approach to destabilize a self-assembling capsid for a subsequent triggered disassembly. We redesigned the dimeric hepatitis B virus (HBV) capsid protein (Cp) with two differentially addressable cysteines, C150 for reversibly crosslinking the capsid and C124 to react with a destabilizing moiety. The resulting construct, Cp150-V124C, assembles into icosahedral, 120-dimer VLPs that spontaneously crosslink via the C-terminal C150, leaving C124 buried at a dimer-dimer interface. The VLP is driven into a metastable state when C124 is reacted with the bulky fluorophore, maleimidyl BoDIPY-FL. The resulting VLP is stable until exposed to modest, physiologically relevant concentrations of reducing agent. We observe dissociation with FRET relaxation of polarization, size exclusion chromatography, and resistive-pulse sensing. Dissociation is slow, minutes to hours, with a characteristic lag phase. Mathematical modeling based on the presence of a nucleation step predicts disassembly dynamics that are consistent with experimental observations. VLPs transfected into hepatoma cells show similar dissociation behavior. These results suggest a generalizable strategy for designing a VLP that can release its contents in an environmentally responsive reaction.
Collapse
Affiliation(s)
- Caleb A. Starr
- Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN 47405 USA
| | - Smita Nair
- Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN 47405 USA
- current address: Door Pharmaceuticals, Bloomington, IN 47401 USA
| | - Sheng-Yuan Huang
- Department of Chemistry, Indiana University, Bloomington, IN 47405 USA
| | - Michael F. Hagan
- Martin Fisher School of Physics, Brandeis University, Waltham, MA 02454 USA
| | | | - Adam Zlotnick
- Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN 47405 USA
| |
Collapse
|
18
|
Molecular elucidation of drug-induced abnormal assemblies of the hepatitis B virus capsid protein by solid-state NMR. Nat Commun 2023; 14:471. [PMID: 36709212 PMCID: PMC9884277 DOI: 10.1038/s41467-023-36219-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 01/18/2023] [Indexed: 01/29/2023] Open
Abstract
Hepatitis B virus (HBV) capsid assembly modulators (CAMs) represent a recent class of anti-HBV antivirals. CAMs disturb proper nucleocapsid assembly, by inducing formation of either aberrant assemblies (CAM-A) or of apparently normal but genome-less empty capsids (CAM-E). Classical structural approaches have revealed the CAM binding sites on the capsid protein (Cp), but conformational information on the CAM-induced off-path aberrant assemblies is lacking. Here we show that solid-state NMR can provide such information, including for wild-type full-length Cp183, and we find that in these assemblies, the asymmetric unit comprises a single Cp molecule rather than the four quasi-equivalent conformers typical for the icosahedral T = 4 symmetry of the normal HBV capsids. Furthermore, while in contrast to truncated Cp149, full-length Cp183 assemblies appear, on the mesoscopic level, unaffected by CAM-A, NMR reveals that on the molecular level, Cp183 assemblies are equally aberrant. Finally, we use a eukaryotic cell-free system to reveal how CAMs modulate capsid-RNA interactions and capsid phosphorylation. Our results establish a structural view on assembly modulation of the HBV capsid, and they provide a rationale for recently observed differences between in-cell versus in vitro capsid assembly modulation.
Collapse
|
19
|
Zoulim F, Zlotnick A, Buchholz S, Donaldson E, Fry J, Gaggar A, Hu J, Kann M, Lenz O, Lin K, Mani N, Nassal M, Delaney W, Wang S, Westman G, Miller V, Janssen HLA. Nomenclature of HBV core protein-targeting antivirals. Nat Rev Gastroenterol Hepatol 2022; 19:748-750. [PMID: 36207612 PMCID: PMC10442071 DOI: 10.1038/s41575-022-00700-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/08/2022]
Affiliation(s)
- Fabien Zoulim
- Hepatology Department, INSERM Unit 1052, Hospices Civils de Lyon, Lyon University, Lyon, France.
| | - Adam Zlotnick
- Molecular and Cellular Biochemistry Department, Indiana University, Bloomington, IN, USA
| | | | | | - John Fry
- Aligos Therapeutics, San Francisco, CA, USA
| | | | - Jianming Hu
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Michael Kann
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Microbiology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | | | - Kai Lin
- Aerium Therapeutics, Boston, MA, USA
| | - Nagraj Mani
- Arbutus Biopharma Inc., Department of Biology, Warminster, PA, USA
| | - Michael Nassal
- Department of Medicine II, Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Su Wang
- Cooperman Barnabas Medical Center, RWJBarnabas-Rutgers Medical Group, Livingston, NJ, USA
| | - Gabriel Westman
- Swedish Medical Products Agency, Uppsala University, Uppsala, Sweden
| | | | - Harry L A Janssen
- Department of Gastroenterology & Hepatology, Erasmus Medical Center, Rotterdam, Netherlands
- Toronto Centre for Liver Disease, University of Toronto, Ontario, Canada
| |
Collapse
|
20
|
Prange R. Hepatitis B virus movement through the hepatocyte: An update. Biol Cell 2022; 114:325-348. [PMID: 35984727 DOI: 10.1111/boc.202200060] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/26/2022] [Accepted: 08/12/2022] [Indexed: 11/29/2022]
Abstract
Viruses are obligate intracellular pathogens that utilize cellular machinery for many aspects of their propagation and effective egress of virus particles from host cells is one important determinant of virus infectivity. Hijacking host cell processes applies in particular to the hepatitis B virus (HBV), as its DNA genome with about 3 kb in size is one of the smallest viral genomes known. HBV is a leading cause of liver disease and still displays one of the most successful pathogens in human populations worldwide. The extremely successful spread of this virus is explained by its efficient transmission strategies and its versatile particle types, including virions, empty envelopes, naked capsids and others. HBV exploits distinct host trafficking machineries to assemble and release its particle types including nucleocytoplasmic shuttling transport, secretory and exocytic pathways, the Endosomal Sorting Complexes Required for Transport pathway, and the autophagy pathway. Understanding how HBV uses and subverts host membrane trafficking systems offers the chance of obtaining new mechanistic insights into the regulation and function of this essential cellular processes. It can also help to identify potential targets for antiviral interventions. Here, I will provide an overview of HBV maturation, assembly, and budding, with a focus on recent advances, and will point out areas where questions remain that can benefit from future studies. Unless otherwise indicated, almost all presented knowledge was gained from cell culture-based, HBV in vitro -replication and in vitro -infection systems. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Reinhild Prange
- Department of Virology, University Medical Center of the Johannes Gutenberg University Mainz, Augustusplatz, Mainz, D-55131, Germany
| |
Collapse
|
21
|
Callon M, Malär AA, Lecoq L, Dujardin M, Fogeron M, Wang S, Schledorn M, Bauer T, Nassal M, Böckmann A, Meier BH. Fast Magic-Angle-Spinning NMR Reveals the Evasive Hepatitis B Virus Capsid C-Terminal Domain. Angew Chem Int Ed Engl 2022; 61:e202201083. [PMID: 35653505 PMCID: PMC9400876 DOI: 10.1002/anie.202201083] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Indexed: 11/08/2022]
Abstract
Experimentally determined protein structures often feature missing domains. One example is the C-terminal domain (CTD) of the hepatitis B virus capsid protein, a functionally central part of this assembly, crucial in regulating nucleic-acid interactions, cellular trafficking, nuclear import, particle assembly and maturation. However, its structure remained elusive to all current techniques, including NMR. Here we show that the recently developed proton-detected fast magic-angle-spinning solid-state NMR at >100 kHz MAS allows one to detect this domain and unveil its structural and dynamic behavior. We describe the experimental framework used and compare the domain's behavior in different capsid states. The developed approaches extend solid-state NMR observations to residues characterized by large-amplitude motion on the microsecond timescale, and shall allow one to shed light on other flexible protein domains still lacking their structural and dynamic characterization.
Collapse
Affiliation(s)
| | | | - Lauriane Lecoq
- Molecular Microbiology and Structural Biochemistry (MMSB) UMR 5086 CNRS/Université de Lyon, Labex Ecofect7 passage du Vercors69367LyonFrance
| | - Marie Dujardin
- Molecular Microbiology and Structural Biochemistry (MMSB) UMR 5086 CNRS/Université de Lyon, Labex Ecofect7 passage du Vercors69367LyonFrance
| | - Marie‐Laure Fogeron
- Molecular Microbiology and Structural Biochemistry (MMSB) UMR 5086 CNRS/Université de Lyon, Labex Ecofect7 passage du Vercors69367LyonFrance
| | - Shishan Wang
- Molecular Microbiology and Structural Biochemistry (MMSB) UMR 5086 CNRS/Université de Lyon, Labex Ecofect7 passage du Vercors69367LyonFrance
| | | | - Thomas Bauer
- Physical Chemistry, ETH Zürich8093ZürichSwitzerland
| | - Michael Nassal
- University Hospital FreiburgDept. of Medicine II/Molecular BiologyMedical CenterUniversity of FreiburgFreiburgGermany
| | - Anja Böckmann
- Molecular Microbiology and Structural Biochemistry (MMSB) UMR 5086 CNRS/Université de Lyon, Labex Ecofect7 passage du Vercors69367LyonFrance
| | | |
Collapse
|
22
|
Ivanova Bencheva L, Donnici L, Ferrante L, Prandi A, Sinisi R, De Matteo M, Randazzo P, Conti M, Di Lucia P, Bono E, Giustini L, Vittoria Orsale M, Patsilinakos A, Monteagudo E, Iannacone M, Summa V, Guidotti LG, De Francesco R, Di Fabio R. Discovery and Antiviral Profile of New Sulfamoylbenzamide Derivatives as HBV Capsid Assembly Modulators. Bioorg Med Chem Lett 2022; 73:128904. [PMID: 35868496 DOI: 10.1016/j.bmcl.2022.128904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/12/2022] [Accepted: 07/15/2022] [Indexed: 11/02/2022]
Abstract
Chronic hepatitis B (CHB) is a major worldwide public health problem and novel anti-HBV therapies preventing liver disease progression to cirrhosis and hepatocellular carcinoma are urgently needed. Over the last several years, capsid assembly modulators (CAM) have emerged as clinically effective anti-HBV agents which can inhibit HBV replication in CHB patients. As part of a drug discovery program aimed at obtaining novel CAM endowed with high in vitro and in vivo antiviral activity, we identified a novel series of sulfamoylbenzamide (SBA) derivatives. Compound 10, one of the most in vitro potent SBA-derived CAM discovered to date, showed excellent pharmacokinetics in mice suitable for oral dosing. When studied in a transgenic mouse model of hepatic HBV replication, it was considerably more potent than NVR 3-778, the first sulfamoylbenzamide (SBA) CAM that entered clinical trials for CHB, at reducing viral replication in a dose-dependent fashion. We present herein the discovery process, the SAR analysis and the pre-clinical profile of this novel SBA CAM.
Collapse
Affiliation(s)
- Leda Ivanova Bencheva
- Promidis, Via Olgettina 60, 20132 Milano, Italy; INGM, National Institute of Molecular Genetics "Romeo ed Enrica Invernizzi", Via Francesco Sforza, 35, 20122 Milan, Italy
| | | | | | | | | | | | - Pietro Randazzo
- INGM, National Institute of Molecular Genetics "Romeo ed Enrica Invernizzi", Via Francesco Sforza, 35, 20122 Milan, Italy
| | - Matteo Conti
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Pietro Di Lucia
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Elisa Bono
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | | | | | | | - Edith Monteagudo
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy; Vita-Salute San Raffaele University, Via Olgettina 60, 20132 Milan, Italy; Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Matteo Iannacone
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Vial Domenico Montesano 49, 80131, Naples, Italy
| | - Vincenzo Summa
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy; Vita-Salute San Raffaele University, Via Olgettina 60, 20132 Milan, Italy
| | - Luca G Guidotti
- INGM, National Institute of Molecular Genetics "Romeo ed Enrica Invernizzi", Via Francesco Sforza, 35, 20122 Milan, Italy; Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti, 9, 20133 Milan, Italy
| | - Raffaele De Francesco
- Promidis, Via Olgettina 60, 20132 Milano, Italy; IRBM Science Park, Via Pontina Km 30.600, 00070 Pomezia, Italy
| | | |
Collapse
|
23
|
Callon M, Malär AA, Lecoq L, Dujardin M, Fogeron ML, Wang S, Schledorn M, Bauer T, Nassal M, Böckmann A, Meier BH. Fast Magic‐Angle‐Spinning NMR Reveals the Evasive Hepatitis B Virus Capsid C‐Terminal Domain. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202201083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Morgane Callon
- ETH Zurich D-CHAB: Eidgenossische Technische Hochschule Zurich Departement Chemie und Angewandte Biowissenschaften D-CHAB SWITZERLAND
| | - Alexander A. Malär
- ETH Zurich D-CHAB: Eidgenossische Technische Hochschule Zurich Departement Chemie und Angewandte Biowissenschaften D-CHAB SWITZERLAND
| | | | | | | | | | - Maarten Schledorn
- ETH Zurich D-CHAB: Eidgenossische Technische Hochschule Zurich Departement Chemie und Angewandte Biowissenschaften D-CHAB SWITZERLAND
| | - Thomas Bauer
- ETH Zurich D-CHAB: Eidgenossische Technische Hochschule Zurich Departement Chemie und Angewandte Biowissenschaften D-CHAB SWITZERLAND
| | - Michael Nassal
- University of Freiburg Hospital: Universitatsklinikum Freiburg Molecular Biology GERMANY
| | | | - Beat H Meier
- ETH Zurich D-CHAB: Eidgenossische Technische Hochschule Zurich Departement Chemie und Angewandte Biowissenschaften Department of Physical Chemistry Wolfgang-Pauli-Strasse 10 8093 Zürich SWITZERLAND
| |
Collapse
|
24
|
Xi J, Cui X, Liu K, Liu H, Wang J, Hu J. Region-Specific Hepatitis B Virus Genome Exposure from Nucleocapsid Modulated by Capsid Linker Sequence and Inhibitor: Implications for Uncoating. J Virol 2022; 96:e0039922. [PMID: 35389266 PMCID: PMC9044944 DOI: 10.1128/jvi.00399-22] [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: 03/07/2022] [Accepted: 03/09/2022] [Indexed: 11/20/2022] Open
Abstract
Hepatitis B virus (HBV) contains a partially double-stranded, relaxed circular (RC) DNA genome synthesized within a nucleocapsid (NC) in the host cell cytoplasm. The release of RC DNA from the NC, in an ill-defined process called uncoating, to the nucleus is required for its conversion to the covalently closed circular (CCC) DNA, the viral episome serving as the transcriptional template for all viral RNAs necessary for replication and, thus, essential for establishing and sustaining viral infection. In efforts to better understand uncoating, we analyzed HBV core (HBc) mutants that show various levels of nuclear CCC DNA but little to no cytoplasmic RC DNA. We found that RC DNA could be synthesized by these mutants outside the cell, but in contrast to the wild type (wt), the mutant NCs were unable to protect RC DNA from digestion by the endogenous nuclease(s) in cellular lysates or exogenous DNase. Subcellular fractionation suggested that the major RC DNA-degrading activity was membrane associated. Digestion with sequence-specific and nonspecific DNases revealed the exposure of specific regions of RC DNA from the mutant NC. Similarly, treatment of wt NCs with a core inhibitor known to increase CCC DNA by affecting uncoating also led to region-specific exposure of RC DNA. Furthermore, a subpopulation of untreated wild type (wt) mature NCs showed site-specific exposure of RC DNA as well. Competition between RC DNA degradation and its conversion to CCC DNA during NC uncoating thus likely plays an important role in the establishment and persistence of HBV infection and has implications for the development of capsid-targeted antivirals. IMPORTANCE Disassembly of the hepatitis B virus (HBV) nucleocapsid (NC) to release its genomic DNA, in an ill-understood process called uncoating, is required to form the viral nuclear episome in the host cell nucleus, a viral DNA essential for establishing and sustaining HBV infection. The elimination of the HBV nuclear episome remains the holy grail for the development of an HBV cure. We report here that the HBV genomic DNA is exposed in a region-specific manner during uncoating, which is enhanced by mutations of the capsid protein and a capsid-targeted antiviral compound. The exposure of the viral genome can result in its rapid degradation or, alternatively, can enhance the formation of the nuclear episome, thus having a major impact on HBV infection and persistence. These results are thus important for understanding fundamental mechanisms of HBV replication and persistence and for the ongoing pursuit of an HBV cure.
Collapse
Affiliation(s)
- Ji Xi
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Xiuji Cui
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Kuancheng Liu
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Haitao Liu
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Joseph Wang
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Jianming Hu
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| |
Collapse
|
25
|
Zi J, Gao X, Du J, Xu H, Niu J, Chi X. Multiple Regions Drive Hepatitis Delta Virus Proliferation and Are Therapeutic Targets. Front Microbiol 2022; 13:838382. [PMID: 35464929 PMCID: PMC9022428 DOI: 10.3389/fmicb.2022.838382] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 03/11/2022] [Indexed: 12/02/2022] Open
Abstract
Hepatitis Delta Virus (HDV) is the smallest mammalian single-stranded RNA virus. It requires host cells and hepatitis B virus (HBV) to complete its unique life cycle. The present review summarizes the specific regions on hepatitis D antigen (HDAg) and hepatitis B surface antigen (HBsAg) that drive HDV to utilize host cell machinery system to produce three types of RNA and two forms of HDAg, and hijack HBsAg for its secretion and de novo entry. Previously, interferon-α was the only recommended therapy for HDV infection. In recent years, some new therapies targeting these regions, such as Bulevirtide, Lonafarnib, Nucleic acid polymers have appeared, with better curative effects and fewer adverse reactions.
Collapse
Affiliation(s)
- Jun Zi
- Gene Therapy Laboratory, Center for Pathogen Biology and Infectious Diseases, First Hospital of Jilin University, Changchun, China
| | - Xiuzhu Gao
- Department of Hepatology, Center for Pathogen Biology and Infectious Diseases, First Hospital of Jilin University, Changchun, China
| | - Juan Du
- Institute of Virology and AIDS Research, First Hospital of Jilin University, Changchun, China
| | - Hongqin Xu
- Department of Hepatology, Center for Pathogen Biology and Infectious Diseases, First Hospital of Jilin University, Changchun, China
| | - Junqi Niu
- Department of Hepatology, Center for Pathogen Biology and Infectious Diseases, First Hospital of Jilin University, Changchun, China
| | - Xiumei Chi
- Gene Therapy Laboratory, Center for Pathogen Biology and Infectious Diseases, First Hospital of Jilin University, Changchun, China
| |
Collapse
|
26
|
Dörnbrack K, Beck J, Nassal M. Relaxing the restricted structural dynamics in the human hepatitis B virus RNA encapsidation signal enables replication initiation in vitro. PLoS Pathog 2022; 18:e1010362. [PMID: 35259189 PMCID: PMC8903280 DOI: 10.1371/journal.ppat.1010362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/10/2022] [Indexed: 11/18/2022] Open
Abstract
Hepadnaviruses, including hepatitis B virus (HBV) as a major human pathogen, replicate their tiny 3 kb DNA genomes by capsid-internal protein-primed reverse transcription of a pregenomic (pg) RNA. Initiation requires productive binding of the viral polymerase, P protein, to a 5´ proximal bipartite stem-loop, the RNA encapsidation signal ε. Then a residue in the central ε bulge directs the covalent linkage of a complementary dNMP to a Tyr sidechain in P protein´s Terminal Protein (TP) domain. After elongation by two or three nucleotides (nt) the TP-linked DNA oligo is transferred to a 3´ proximal acceptor, enabling full-length minus-strand DNA synthesis. No direct structural data are available on hepadnaviral initiation complexes but their cell-free reconstitution with P protein and ε RNA (Dε) from duck HBV (DHBV) provided crucial mechanistic insights, including on a major conformational rearrangement in the apical Dε part. Analogous cell-free systems for human HBV led at most to P—ε binding but no detectable priming. Here we demonstrate that local relaxation of the highly basepaired ε upper stem, by mutation or via synthetic split RNAs, enables ε-dependent in vitro priming with full-length P protein from eukaryotic translation extract yet also, and without additional macromolecules, with truncated HBV miniP proteins expressed in bacteria. Using selective 2-hydroxyl acylation analyzed by primer extension (SHAPE) we confirm that upper stem destabilization correlates with in vitro priming competence and show that the supposed bulge-closing basepairs are largely unpaired even in wild-type ε. We define the two 3´ proximal nt of this extended bulge as main initiation sites and provide evidence for a Dε-like opening of the apical ε part upon P protein binding. Beyond new HBV-specific basic aspects our novel in vitro priming systems should facilitate the development of high-throughput screens for priming inhibitors targeting this highly virus-specific process. Chronic hepatitis B virus (HBV) infection puts >250 million people at an increased risk for severe liver disease. Current treatments can control but rarely cure infection. HBV features a 3,200 bp DNA genome, generated by reverse transcription of a pregenomic (pg) RNA. To initiate DNA synthesis the viral polymerase, P protein, employs a stem-loop on pgRNA, ε, to covalently link a defined first nucleotide to its Terminal Protein (TP) domain. This protein-priming is highly virus-specific yet poorly understood. More is known for duck HBV (DHBV) where, different from HBV, protein-priming was successfully reconstituted in vitro years ago. One insight was that gaining priming-competence involves opening of the apical stem in DHBV ε RNA (Dε); in HBV ε the more extensive basepairing might restrict such dynamics. Here we relaxed these constraints by identifying functional but less stably folded, including split, HBV ε variants. Several such variants supported in vitro priming, including in a simple two-component-system employing a shortened recombinant P protein. Amongst other data the new cell-free systems yielded a first view on a major conformational change in HBV ε RNA bound to P protein, highlighting the importance of RNA dynamics for the human virus. Beyond furthering basic understanding our data should facilitate screening for protein-priming inhibitors as new anti-HBV agents.
Collapse
Affiliation(s)
- Katharina Dörnbrack
- Department of Internal Medicine II/Molecular Biology, University Hospital Freiburg, Freiburg, Germany
| | - Jürgen Beck
- Department of Internal Medicine II/Molecular Biology, University Hospital Freiburg, Freiburg, Germany
- * E-mail: (JB); , (MN)
| | - Michael Nassal
- Department of Internal Medicine II/Molecular Biology, University Hospital Freiburg, Freiburg, Germany
- * E-mail: (JB); , (MN)
| |
Collapse
|
27
|
Briday M, Hallé F, Lecoq L, Radix S, Martin J, Montserret R, Dujardin M, Fogeron ML, Nassal M, Meier BH, Lomberget T, Böckmann A. Pharmacomodulation of a ligand targeting the HBV capsid hydrophobic pocket. Chem Sci 2022; 13:8840-8847. [PMID: 36042894 PMCID: PMC9358932 DOI: 10.1039/d2sc02420a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 07/06/2022] [Indexed: 11/21/2022] Open
Abstract
Hepatitis B virus (HBV) is a small enveloped retrotranscribing DNA virus and an important human pathogen. Its capsid-forming core protein (Cp) features a hydrophobic pocket proposed to be central notably in capsid envelopment. Indeed, mutations in and around this pocket can profoundly modulate, and even abolish, secretion of enveloped virions. We have recently shown that Triton X-100, a detergent used during Cp purification, binds to the hydrophobic pocket with micromolar affinity. We here performed pharmacomodulation of pocket binders through systematic modifications of the three distinct chemical moieties composing the Triton X-100 molecule. Using NMR and ITC, we found that the flat aromatic moiety is essential for binding, while the number of atoms of the aliphatic chain modulates binding affinity. The hydrophilic tail, in contrast, is highly tolerant to changes in both length and type. Our data provide essential information for designing a new class of HBV antivirals targeting capsid–envelope interactions. Small-molecule binding to the Hepatitis B virus core protein hydrophobic pocket, a possible strategy for targeting viral particle assembly.![]()
Collapse
Affiliation(s)
- Mathilde Briday
- Molecular Microbiology and Structural Biochemistry (MMSB) UMR 5086 CNRS/Université de Lyon, Labex Ecofect, 7 Passage du Vercors, 69367 Lyon, France
| | - François Hallé
- Université de Lyon, Université Lyon 1, CNRS UMR 5246 Institut de Chimie et Biochimie Moléculaires et Supramoléculaires (ICBMS), Faculté de Pharmacie-ISPB, 8 Avenue Rockefeller, FR-69373 Lyon Cedex 08, France
| | - Lauriane Lecoq
- Molecular Microbiology and Structural Biochemistry (MMSB) UMR 5086 CNRS/Université de Lyon, Labex Ecofect, 7 Passage du Vercors, 69367 Lyon, France
| | - Sylvie Radix
- Université de Lyon, Université Lyon 1, CNRS UMR 5246 Institut de Chimie et Biochimie Moléculaires et Supramoléculaires (ICBMS), Faculté de Pharmacie-ISPB, 8 Avenue Rockefeller, FR-69373 Lyon Cedex 08, France
| | - Juliette Martin
- Molecular Microbiology and Structural Biochemistry (MMSB) UMR 5086 CNRS/Université de Lyon, Labex Ecofect, 7 Passage du Vercors, 69367 Lyon, France
| | - Roland Montserret
- Molecular Microbiology and Structural Biochemistry (MMSB) UMR 5086 CNRS/Université de Lyon, Labex Ecofect, 7 Passage du Vercors, 69367 Lyon, France
| | - Marie Dujardin
- Molecular Microbiology and Structural Biochemistry (MMSB) UMR 5086 CNRS/Université de Lyon, Labex Ecofect, 7 Passage du Vercors, 69367 Lyon, France
| | - Marie-Laure Fogeron
- Molecular Microbiology and Structural Biochemistry (MMSB) UMR 5086 CNRS/Université de Lyon, Labex Ecofect, 7 Passage du Vercors, 69367 Lyon, France
| | - Michael Nassal
- Department of Medicine II/Molecular Biology, University Hospital Freiburg, Medical Center, University of Freiburg, Freiburg 79106, Germany
| | - Beat H. Meier
- Physical Chemistry, ETH Zürich, 8093 Zürich, Switzerland
| | - Thierry Lomberget
- Université de Lyon, Université Lyon 1, CNRS UMR 5246 Institut de Chimie et Biochimie Moléculaires et Supramoléculaires (ICBMS), Faculté de Pharmacie-ISPB, 8 Avenue Rockefeller, FR-69373 Lyon Cedex 08, France
| | - Anja Böckmann
- Molecular Microbiology and Structural Biochemistry (MMSB) UMR 5086 CNRS/Université de Lyon, Labex Ecofect, 7 Passage du Vercors, 69367 Lyon, France
| |
Collapse
|
28
|
Raimondo G, Rebello-Pinho JR, Panduro A. Hepatitis B and C viruses and hepatocellular carcinoma. Ann Hepatol 2022; 27 Suppl 1:100650. [PMID: 34864201 DOI: 10.1016/j.aohep.2021.100650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 11/24/2021] [Indexed: 02/04/2023]
Affiliation(s)
- Giovanni Raimondo
- Division of Medicine and Hepatology, Department of Clinical and Experimental Medicine, University Hospital of Messina, Messina, Italy
| | - Joao R Rebello-Pinho
- LIM-07, Institute of Tropical Medicine, Department of Gastroenterology, University of São Paulo School of Medicine, São Paulo, Brazil; Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Arturo Panduro
- Department of Genomic Medicine in Hepatology, Civil Hospital of Guadalajara, Fray Antonio Alcalde, Guadalajara, Jalisco, Mexico; Health Sciences Center, University of Guadalajara, Guadalajara, Jalisco, Mexico.
| |
Collapse
|