1
|
Bressanelli S, Fieulaine S, Tubiana T. Structural biology of single-stranded, positive-sense RNA viruses in the age of accurate atomic-scale predictions of protein structures. Virology 2025; 608:110546. [PMID: 40288078 DOI: 10.1016/j.virol.2025.110546] [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: 02/08/2025] [Revised: 04/02/2025] [Accepted: 04/15/2025] [Indexed: 04/29/2025]
Abstract
For decades atomic structures of proteins could only be determined experimentally and at a very slow pace. This was a particular problem for RNA viruses, for which sequences diverge fast and horizontal transfers are common. This made modeling from known structures difficult and uncertain. Only hard experimental structural data could allow accurate atomic descriptions of viral proteins and subsequent analyses, from mutant phenotype prediction to drug design. This has changed. With the advent of AlphaFold, that allows accurate protein structure prediction from sequence only, it is now possible in most cases to have the structure of a new protein of interest in a matter of minutes. In this mini review we focus on important consequences of this new state of affairs. While most of our conclusions are likely relevant to RNA viruses in general, here we focus on single-stranded, positive-sense RNA viruses. Taking as case studies proteins that are studied in our lab, we highlight why these viruses generally encode proteins that are particularly tough cases, being membrane-associated proteins with alternate conformations, structures, and interactions that may not be conserved even between close relatives. For these proteins AlphaFold may even fail or at least mislead, but with a proper approach it may also allow jump-starting the study of difficult or understudied viruses.
Collapse
Affiliation(s)
- Stéphane Bressanelli
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, 91198, France.
| | - Sonia Fieulaine
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, 91198, France.
| | - Thibault Tubiana
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, 91198, France.
| |
Collapse
|
2
|
Zhang F, Xu LD, Wu S, Wang B, Xu P, Huang YW. Deciphering the hepatitis E virus ORF1: Functional domains, protein processing, and patient-derived mutations. Virology 2025; 603:110350. [PMID: 39675187 DOI: 10.1016/j.virol.2024.110350] [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: 10/17/2024] [Revised: 11/25/2024] [Accepted: 12/05/2024] [Indexed: 12/17/2024]
Abstract
Hepatitis E virus (HEV) is a major cause of acute and chronic hepatitis in humans. The HEV open reading frames (ORF1) encodes a large non-structural protein essential for viral replication, which contains several functional domains, including helicase and RNA-dependent RNA polymerase. A confusing aspect is that, while RNA viruses typically encode large polyproteins that rely on their enzymatic activity for processing into functional units, the processing of the ORF1 protein and the mechanisms involved remain unclear. The ORF1 plays a pivotal role in the viral life cycle, thus mutations in this region, especially those occurring under environmental pressures such as during antiviral drug treatment, could significantly affect viral replication and survival. Here, we summarize the recent advances in the functional domains, processing, and mutations of ORF1. Gaining a deeper understanding of HEV biology, particularly focusing on ORF1, could facilitate the development of new strategies to control HEV infections.
Collapse
Affiliation(s)
- Fei Zhang
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China; ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 311215, China; MOE Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China
| | - Ling-Dong Xu
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China; MOE Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China; Laboratory Animal Center, Zhejiang University, Hangzhou, 310058, China
| | - Shiying Wu
- MOE Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China; College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China
| | - Bin Wang
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China; State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, 510642, China
| | - Pinglong Xu
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China; ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 311215, China; MOE Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China; Cancer Center, Zhejiang University, Hangzhou, 310058, China.
| | - Yao-Wei Huang
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China; State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
3
|
Versini R, Sritharan S, Aykac Fas B, Tubiana T, Aimeur SZ, Henri J, Erard M, Nüsse O, Andreani J, Baaden M, Fuchs P, Galochkina T, Chatzigoulas A, Cournia Z, Santuz H, Sacquin-Mora S, Taly A. A Perspective on the Prospective Use of AI in Protein Structure Prediction. J Chem Inf Model 2024; 64:26-41. [PMID: 38124369 DOI: 10.1021/acs.jcim.3c01361] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
AlphaFold2 (AF2) and RoseTTaFold (RF) have revolutionized structural biology, serving as highly reliable and effective methods for predicting protein structures. This article explores their impact and limitations, focusing on their integration into experimental pipelines and their application in diverse protein classes, including membrane proteins, intrinsically disordered proteins (IDPs), and oligomers. In experimental pipelines, AF2 models help X-ray crystallography in resolving the phase problem, while complementarity with mass spectrometry and NMR data enhances structure determination and protein flexibility prediction. Predicting the structure of membrane proteins remains challenging for both AF2 and RF due to difficulties in capturing conformational ensembles and interactions with the membrane. Improvements in incorporating membrane-specific features and predicting the structural effect of mutations are crucial. For intrinsically disordered proteins, AF2's confidence score (pLDDT) serves as a competitive disorder predictor, but integrative approaches including molecular dynamics (MD) simulations or hydrophobic cluster analyses are advocated for accurate dynamics representation. AF2 and RF show promising results for oligomeric models, outperforming traditional docking methods, with AlphaFold-Multimer showing improved performance. However, some caveats remain in particular for membrane proteins. Real-life examples demonstrate AF2's predictive capabilities in unknown protein structures, but models should be evaluated for their agreement with experimental data. Furthermore, AF2 models can be used complementarily with MD simulations. In this Perspective, we propose a "wish list" for improving deep-learning-based protein folding prediction models, including using experimental data as constraints and modifying models with binding partners or post-translational modifications. Additionally, a meta-tool for ranking and suggesting composite models is suggested, driving future advancements in this rapidly evolving field.
Collapse
Affiliation(s)
- Raphaelle Versini
- Laboratoire de Biochimie Théorique, CNRS (UPR9080), Université Paris Cité, F-75005 Paris, France
| | - Sujith Sritharan
- Laboratoire de Biochimie Théorique, CNRS (UPR9080), Université Paris Cité, F-75005 Paris, France
| | - Burcu Aykac Fas
- Laboratoire de Biochimie Théorique, CNRS (UPR9080), Université Paris Cité, F-75005 Paris, France
| | - Thibault Tubiana
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198 Gif-sur-Yvette, France
| | - Sana Zineb Aimeur
- Université Paris-Saclay, CNRS, Institut de Chimie Physique, 91405 Orsay, France
| | - Julien Henri
- Sorbonne Université, CNRS, Laboratoire de Biologie, Computationnelle et Quantitative UMR 7238, Institut de Biologie Paris-Seine, 4 Place Jussieu, F-75005 Paris, France
| | - Marie Erard
- Université Paris-Saclay, CNRS, Institut de Chimie Physique, 91405 Orsay, France
| | - Oliver Nüsse
- Université Paris-Saclay, CNRS, Institut de Chimie Physique, 91405 Orsay, France
| | - Jessica Andreani
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198 Gif-sur-Yvette, France
| | - Marc Baaden
- Laboratoire de Biochimie Théorique, CNRS (UPR9080), Université Paris Cité, F-75005 Paris, France
| | - Patrick Fuchs
- Sorbonne Université, École Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM, 75005 Paris, France
- Université de Paris, UFR Sciences du Vivant, 75013 Paris, France
| | - Tatiana Galochkina
- Université Paris Cité and Université des Antilles and Université de la Réunion, INSERM, BIGR, F-75014 Paris, France
| | - Alexios Chatzigoulas
- Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece
- Department of Informatics and Telecommunications, National and Kapodistrian University of Athens, 15784 Athens, Greece
| | - Zoe Cournia
- Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece
- Department of Informatics and Telecommunications, National and Kapodistrian University of Athens, 15784 Athens, Greece
| | - Hubert Santuz
- Laboratoire de Biochimie Théorique, CNRS (UPR9080), Université Paris Cité, F-75005 Paris, France
| | - Sophie Sacquin-Mora
- Laboratoire de Biochimie Théorique, CNRS (UPR9080), Université Paris Cité, F-75005 Paris, France
| | - Antoine Taly
- Laboratoire de Biochimie Théorique, CNRS (UPR9080), Université Paris Cité, F-75005 Paris, France
| |
Collapse
|
4
|
Glitscher M, Spannaus IM, Behr F, Murra RO, Woytinek K, Bender D, Hildt E. The Protease Domain in HEV pORF1 Mediates the Replicase's Localization to Multivesicular Bodies and Its Exosomal Release. Cell Mol Gastroenterol Hepatol 2024; 17:589-605. [PMID: 38190941 PMCID: PMC10900777 DOI: 10.1016/j.jcmgh.2024.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 01/03/2024] [Accepted: 01/03/2024] [Indexed: 01/10/2024]
Abstract
BACKGROUND A peculiar feature of the hepatitis E virus (HEV) is its reliance on the exosomal route for viral release. Genomic replication is mediated via the viral polyprotein pORF1, yet little is known about its subcellular localization. METHODS Subcellular localization of pORF1 and its subdomains, generated and cloned based on a structural prediciton of the viral replicase, was analyzed via confocal laser scanning microscopy. Exosomes released from cells were isolated via ultracentrifugation and analyzed by isopycnic density gradient centrifugation. This was followed by fluorimetry or Western blot analyses or reverse transcriptase-polymerase chain reaction to analyze separated particles in more detail. RESULTS We found pORF1 to be accumulating within the endosomal system, most dominantly to multivesicular bodies (MVBs). Expression of the polyprotein's 7 subdomains revealed that the papain-like cysteine-protease (PCP) is the only domain localizing like the full-length protein. A PCP-deficient pORF1 mutant lost its association to MVBs. Strikingly, both pORF1 and PCP can be released via exosomes. Similarly, genomic RNA still is released via exosomes in the absence of pORF2/3. CONCLUSIONS Taken together, we found that pORF1 localizes to MVBs in a PCP-dependent manner, which is followed by exosomal release. This reveals new aspects of HEV life cycle, because replication and release could be coupled at the endosomal interface. In addition, this may mediate capsid-independent spread or may facilitate the spread of viral infection, because genomes entering the cell during de novo infection readily encounter exosomally transferred pORF1.
Collapse
Affiliation(s)
- Mirco Glitscher
- Department of Virology, Paul-Ehrlich-Institute, Langen, Germany
| | | | - Fabiane Behr
- Department of Virology, Paul-Ehrlich-Institute, Langen, Germany
| | | | | | - Daniela Bender
- Department of Virology, Paul-Ehrlich-Institute, Langen, Germany
| | - Eberhard Hildt
- Department of Virology, Paul-Ehrlich-Institute, Langen, Germany.
| |
Collapse
|
5
|
Fieulaine S, Tubiana T, Bressanelli S. Hepatitis E virus RNA replication polyprotein: taking structural biology seriously. Front Microbiol 2023; 14:1254741. [PMID: 37601361 PMCID: PMC10436323 DOI: 10.3389/fmicb.2023.1254741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 07/25/2023] [Indexed: 08/22/2023] Open
Affiliation(s)
- Sonia Fieulaine
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | | | - Stéphane Bressanelli
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| |
Collapse
|
6
|
Pierce DM, Buchanan FJT, Macrae FL, Mills JT, Cox A, Abualsaoud KM, Ward JC, Ariëns RAS, Harris M, Stonehouse NJ, Herod MR. Thrombin cleavage of the hepatitis E virus polyprotein at multiple conserved locations is required for genome replication. PLoS Pathog 2023; 19:e1011529. [PMID: 37478143 PMCID: PMC10395923 DOI: 10.1371/journal.ppat.1011529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 07/03/2023] [Indexed: 07/23/2023] Open
Abstract
The genomes of positive-sense RNA viruses encode polyproteins that are essential for mediating viral replication. These viral polyproteins must undergo proteolysis (also termed polyprotein processing) to generate functional protein units. This proteolysis can be performed by virally-encoded proteases as well as host cellular proteases, and is generally believed to be a key step in regulating viral replication. Hepatitis E virus (HEV) is a leading cause of acute viral hepatitis. The positive-sense RNA genome is translated to generate a polyprotein, termed pORF1, which is necessary and sufficient for viral genome replication. However, the mechanism of polyprotein processing in HEV remains to be determined. In this study, we aimed to understand processing of this polyprotein and its role in viral replication using a combination of in vitro translation experiments and HEV sub-genomic replicons. Our data suggest no evidence for a virally-encoded protease or auto-proteolytic activity, as in vitro translation predominantly generates unprocessed viral polyprotein precursors. However, seven cleavage sites within the polyprotein (suggested by bioinformatic analysis) are susceptible to the host cellular protease, thrombin. Using two sub-genomic replicon systems, we demonstrate that mutagenesis of these sites prevents replication, as does pharmacological inhibition of serine proteases including thrombin. Overall, our data supports a model where HEV uses host proteases to support replication and could have evolved to be independent of a virally-encoded protease for polyprotein processing.
Collapse
Affiliation(s)
- Danielle M Pierce
- School of Molecular and Cellular Biology, Faculty of Biological Sciences and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom
| | - Frazer J T Buchanan
- School of Molecular and Cellular Biology, Faculty of Biological Sciences and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom
| | - Fraser L Macrae
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Jake T Mills
- School of Molecular and Cellular Biology, Faculty of Biological Sciences and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom
| | - Abigail Cox
- School of Molecular and Cellular Biology, Faculty of Biological Sciences and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom
| | - Khadijah M Abualsaoud
- School of Molecular and Cellular Biology, Faculty of Biological Sciences and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom
- Department of Laboratory and Blood Bank, Al Mikhwah General Hospital, Al Mikhwah, Saudi Arabia
| | - Joseph C Ward
- School of Molecular and Cellular Biology, Faculty of Biological Sciences and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom
| | - Robert A S Ariëns
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Mark Harris
- School of Molecular and Cellular Biology, Faculty of Biological Sciences and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom
| | - Nicola J Stonehouse
- School of Molecular and Cellular Biology, Faculty of Biological Sciences and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom
| | - Morgan R Herod
- School of Molecular and Cellular Biology, Faculty of Biological Sciences and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
7
|
LeDesma R, Heller B, Biswas A, Maya S, Gili S, Higgins J, Ploss A. Structural features stabilized by divalent cation coordination within hepatitis E virus ORF1 are critical for viral replication. eLife 2023; 12:e80529. [PMID: 36852909 PMCID: PMC9977285 DOI: 10.7554/elife.80529] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 02/12/2023] [Indexed: 03/01/2023] Open
Abstract
Hepatitis E virus (HEV) is an RNA virus responsible for over 20 million infections annually. HEV's open reading frame (ORF)1 polyprotein is essential for genome replication, though it is unknown how the different subdomains function within a structural context. Our data show that ORF1 operates as a multifunctional protein, which is not subject to proteolytic processing. Supporting this model, scanning mutagenesis performed on the putative papain-like cysteine protease (pPCP) domain revealed six cysteines essential for viral replication. Our data are consistent with their role in divalent metal ion coordination, which governs local and interdomain interactions that are critical for the overall structure of ORF1; furthermore, the 'pPCP' domain can only rescue viral genome replication in trans when expressed in the context of the full-length ORF1 protein but not as an individual subdomain. Taken together, our work provides a comprehensive model of the structure and function of HEV ORF1.
Collapse
Affiliation(s)
- Robert LeDesma
- Department of Molecular Biology, Lewis Thomas Laboratory, Princeton UniversityPrincetonUnited States
| | - Brigitte Heller
- Department of Molecular Biology, Lewis Thomas Laboratory, Princeton UniversityPrincetonUnited States
| | - Abhishek Biswas
- Department of Molecular Biology, Lewis Thomas Laboratory, Princeton UniversityPrincetonUnited States
| | - Stephanie Maya
- Department of Molecular Biology, Lewis Thomas Laboratory, Princeton UniversityPrincetonUnited States
| | - Stefania Gili
- Department of Geosciences, Princeton UniversityPrincetonUnited States
| | - John Higgins
- Department of Geosciences, Princeton UniversityPrincetonUnited States
| | - Alexander Ploss
- Department of Molecular Biology, Lewis Thomas Laboratory, Princeton UniversityPrincetonUnited States
| |
Collapse
|
8
|
Fieulaine S, Tubiana T, Bressanelli S. De novo modelling of HEV replication polyprotein: Five-domain breakdown and involvement of flexibility in functional regulation. Virology 2023; 578:128-140. [PMID: 36527931 DOI: 10.1016/j.virol.2022.12.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/01/2022] [Accepted: 12/02/2022] [Indexed: 12/14/2022]
Abstract
Hepatitis E virus (HEV), a major cause of acute viral hepatitis, is a single-stranded, positive-sense RNA virus. As such, it encodes a 1700-residue replication polyprotein pORF1 that directs synthesis of new viral RNA in infected cells. Here we report extensive modeling with AlphaFold2 of the full-length pORF1, and its production by in vitro translation. From this, we give a detailed update on the breakdown into domains of HEV pORF1. We also provide evidence that pORF1's N-terminal domain is likely to oligomerize to form a dodecameric pore, homologously to what has been described for Chikungunya virus. Beyond providing accurate folds for its five domains, our work highlights that there is no canonical protease encoded in pORF1 and that flexibility in several functionally important regions rather than proteolytic processing may serve to regulate HEV RNA synthesis.
Collapse
Affiliation(s)
- Sonia Fieulaine
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France.
| | - Thibault Tubiana
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France.
| | - Stéphane Bressanelli
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France.
| |
Collapse
|
9
|
Goulet A, Cambillau C, Roussel A, Imbert I. Structure Prediction and Analysis of Hepatitis E Virus Non-Structural Proteins from the Replication and Transcription Machinery by AlphaFold2. Viruses 2022; 14:1537. [PMID: 35891516 PMCID: PMC9316534 DOI: 10.3390/v14071537] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 02/04/2023] Open
Abstract
Hepatitis E virus (HEV) is a major cause of acute viral hepatitis in humans globally. Considered for a long while a public health issue only in developing countries, the HEV infection is now a global public health concern. Most human infections are caused by the HEV genotypes 1, 2, 3 and 4 (HEV-1 to HEV-4). Although HEV-3 and HEV-4 can evolve to chronicity in immunocompromised patients, HEV-1 and HEV-2 lead to self-limited infections. HEV has a positive-sense single-stranded RNA genome of ~7.2 kb that is translated into a large pORF1 replicative polyprotein, essential for the viral RNA genome replication and transcription. Unfortunately, the composition and structure of these replicases are still unknown. The recent release of the powerful machine-learning protein structure prediction software AlphaFold2 (AF2) allows us to accurately predict the structure of proteins and their complexes. Here, we used AF2 with the replicase encoded by the polyprotein pORF1 of the human-infecting HEV-3. The boundaries and structures reveal five domains or nonstructural proteins (nsPs): the methyltransferase, Zn-binding domain, macro, helicase, and RNA-dependent RNA polymerase, reliably predicted. Their substrate-binding sites are similar to those observed experimentally for other related viral proteins. Precisely knowing enzyme boundaries and structures is highly valuable to recombinantly produce stable and active proteins and perform structural, functional and inhibition studies.
Collapse
Affiliation(s)
- Adeline Goulet
- Aix-Marseille Université, Centre National de la Recherche Scientifique, UMR 7255, LISM, 31 Chemin Joseph Aiguier, 13009 Marseille, France; (A.G.); (A.R.)
| | - Christian Cambillau
- School of Microbiology, University College Cork, T12 YT20 Cork, Ireland;
- AlphaGraphix, 24 Carrer d’Amont, 66210 Formiguères, France
| | - Alain Roussel
- Aix-Marseille Université, Centre National de la Recherche Scientifique, UMR 7255, LISM, 31 Chemin Joseph Aiguier, 13009 Marseille, France; (A.G.); (A.R.)
| | - Isabelle Imbert
- Aix-Marseille Université, Centre National de la Recherche Scientifique, UMR 7255, LISM, 31 Chemin Joseph Aiguier, 13009 Marseille, France; (A.G.); (A.R.)
| |
Collapse
|
10
|
Metzger K, Bentaleb C, Hervouet K, Alexandre V, Montpellier C, Saliou JM, Ferrié M, Camuzet C, Rouillé Y, Lecoeur C, Dubuisson J, Cocquerel L, Aliouat-Denis CM. Processing and Subcellular Localization of the Hepatitis E Virus Replicase: Identification of Candidate Viral Factories. Front Microbiol 2022; 13:828636. [PMID: 35283856 PMCID: PMC8908324 DOI: 10.3389/fmicb.2022.828636] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 02/01/2022] [Indexed: 01/26/2023] Open
Abstract
Hepatitis E virus (HEV) is the major cause of acute hepatitis worldwide. HEV is a positive-sense RNA virus expressing three open reading frames (ORFs). ORF1 encodes the ORF1 non-structural polyprotein, the viral replicase which transcribes the full-length genome and a subgenomic RNA that encodes the structural ORF2 and ORF3 proteins. The present study is focused on the replication step with the aim to determine whether the ORF1 polyprotein is processed during the HEV lifecycle and to identify where the replication takes place inside the host cell. As no commercial antibody recognizes ORF1 in HEV-replicating cells, we aimed at inserting epitope tags within the ORF1 protein without impacting the virus replication efficacy. Two insertion sites located in the hypervariable region were thus selected to tolerate the V5 epitope while preserving HEV replication efficacy. Once integrated into the infectious full-length Kernow C-1 p6 strain, the V5 epitopes did neither impact the replication of genomic nor the production of subgenomic RNA. Also, the V5-tagged viral particles remained as infectious as the wildtype particles to Huh-7.5 cells. Next, the expression pattern of the V5-tagged ORF1 was compared in heterologous expression and replicative HEV systems. A high molecular weight protein (180 kDa) that was expressed in all three systems and that likely corresponds to the unprocessed form of ORF1 was detected up to 25 days after electroporation in the p6 cell culture system. Additionally, less abundant products of lower molecular weights were detected in both in cytoplasmic and nuclear compartments. Concurrently, the V5-tagged ORF1 was localized by confocal microscopy inside the cell nucleus but also as compact perinuclear substructures in which ORF2 and ORF3 proteins were detected. Importantly, using in situ hybridization (RNAScope ®), positive and negative-strand HEV RNAs were localized in the perinuclear substructures of HEV-producing cells. Finally, by simultaneous detection of HEV genomic RNAs and viral proteins in these substructures, we identified candidate HEV factories.
Collapse
Affiliation(s)
- Karoline Metzger
- CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 – UMR 9017 – Center for Infection and Immunity of Lille (CIIL), Université de Lille, Lille, France
| | - Cyrine Bentaleb
- CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 – UMR 9017 – Center for Infection and Immunity of Lille (CIIL), Université de Lille, Lille, France
| | - Kévin Hervouet
- CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 – UMR 9017 – Center for Infection and Immunity of Lille (CIIL), Université de Lille, Lille, France
| | - Virginie Alexandre
- CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 – UMR 9017 – Center for Infection and Immunity of Lille (CIIL), Université de Lille, Lille, France
| | - Claire Montpellier
- CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 – UMR 9017 – Center for Infection and Immunity of Lille (CIIL), Université de Lille, Lille, France
| | - Jean-Michel Saliou
- CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR2014 – US41 – Plateformes Lilloises de Biologie and Santé (PLBS), Université de Lille, Lille, France
| | - Martin Ferrié
- CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 – UMR 9017 – Center for Infection and Immunity of Lille (CIIL), Université de Lille, Lille, France
| | - Charline Camuzet
- CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 – UMR 9017 – Center for Infection and Immunity of Lille (CIIL), Université de Lille, Lille, France
| | - Yves Rouillé
- CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 – UMR 9017 – Center for Infection and Immunity of Lille (CIIL), Université de Lille, Lille, France
| | - Cécile Lecoeur
- CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 – UMR 9017 – Center for Infection and Immunity of Lille (CIIL), Université de Lille, Lille, France
| | - Jean Dubuisson
- CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 – UMR 9017 – Center for Infection and Immunity of Lille (CIIL), Université de Lille, Lille, France
| | - Laurence Cocquerel
- CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 – UMR 9017 – Center for Infection and Immunity of Lille (CIIL), Université de Lille, Lille, France
| | - Cécile-Marie Aliouat-Denis
- CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 – UMR 9017 – Center for Infection and Immunity of Lille (CIIL), Université de Lille, Lille, France
| |
Collapse
|
11
|
Oechslin N, Da Silva N, Szkolnicka D, Cantrelle F, Hanoulle X, Moradpour D, Gouttenoire J. Hepatitis E virus RNA-dependent RNA polymerase is involved in RNA replication and infectious particle production. Hepatology 2022; 75:170-181. [PMID: 34387882 PMCID: PMC9300124 DOI: 10.1002/hep.32100] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/09/2021] [Accepted: 07/31/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS Hepatitis E virus (HEV) is one of the most common causes of acute hepatitis worldwide. Its positive-strand RNA genome encodes three open reading frames (ORF). ORF1 is translated into a large protein composed of multiple domains and is known as the viral replicase. The RNA-dependent RNA polymerase (RDRP) domain is responsible for the synthesis of viral RNA. APPROACH AND RESULTS Here, we identified a highly conserved α-helix located in the RDRP thumb subdomain. Nuclear magnetic resonance demonstrated an amphipathic α-helix extending from amino acids 1628 to 1644 of the ORF1 protein. Functional analyses revealed a dual role of this helix in HEV RNA replication and virus production, including assembly and release. Mutations on the hydrophobic side of the amphipathic α-helix impaired RNA replication and resulted in the selection of a second-site compensatory change in the RDRP palm subdomain. Other mutations enhanced RNA replication but impaired virus assembly and/or release. CONCLUSIONS Structure-function analyses identified a conserved amphipathic α-helix in the thumb subdomain of the HEV RDRP with a dual role in viral RNA replication and infectious particle production. This study provides structural insights into a key segment of the ORF1 protein and describes the successful use of reverse genetics in HEV, revealing functional interactions between the RDRP thumb and palm subdomains. On a broader scale, it demonstrates that the HEV replicase, similar to those of other positive-strand RNA viruses, is also involved in virus production.
Collapse
Affiliation(s)
- Noémie Oechslin
- Division of Gastroenterology and HepatologyLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Nathalie Da Silva
- Division of Gastroenterology and HepatologyLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Dagmara Szkolnicka
- Division of Gastroenterology and HepatologyLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - François‐Xavier Cantrelle
- UMR1167University of LilleINSERMLille University HospitalInstitut Pasteur de LilleLilleFrance,CNRS, ERL9002 ‐ Integrative Structural BiologyLilleFrance
| | - Xavier Hanoulle
- UMR1167University of LilleINSERMLille University HospitalInstitut Pasteur de LilleLilleFrance,CNRS, ERL9002 ‐ Integrative Structural BiologyLilleFrance
| | - Darius Moradpour
- Division of Gastroenterology and HepatologyLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Jérôme Gouttenoire
- Division of Gastroenterology and HepatologyLausanne University Hospital and University of LausanneLausanneSwitzerland
| |
Collapse
|
12
|
Cancela F, Noceti O, Arbiza J, Mirazo S. Structural aspects of hepatitis E virus. Arch Virol 2022; 167:2457-2481. [PMID: 36098802 PMCID: PMC9469829 DOI: 10.1007/s00705-022-05575-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/04/2022] [Indexed: 12/14/2022]
Abstract
Hepatitis E virus (HEV) is a leading cause of acute hepatitis worldwide. Hepatitis E is an enterically transmitted zoonotic disease that causes large waterborne epidemic outbreaks in developing countries and has become an increasing public-health concern in industrialized countries. In this setting, the infection is usually acute and self-limiting in immunocompetent individuals, although chronic cases in immunocompromised patients have been reported, frequently associated with several extrahepatic manifestations. Moreover, extrahepatic manifestations have also been reported in immunocompetent individuals with acute HEV infection. HEV belongs to the alphavirus-like supergroup III of single-stranded positive-sense RNA viruses, and its genome contains three partially overlapping open reading frames (ORFs). ORF1 encodes a nonstructural protein with eight domains, most of which have not been extensively characterized: methyltransferase, Y domain, papain-like cysteine protease, hypervariable region, proline-rich region, X domain, Hel domain, and RNA-dependent RNA polymerase. ORF2 and ORF3 encode the capsid protein and a multifunctional protein believed to be involved in virion release, respectively. The novel ORF4 is only expressed in HEV genotype 1 under endoplasmic reticulum stress conditions, and its exact function has not yet been elucidated. Despite important advances in recent years, the biological and molecular processes underlying HEV replication remain poorly understood, primarily due to a lack of detailed information about the functions of the viral proteins and the mechanisms involved in host-pathogen interactions. This review summarizes the current knowledge concerning HEV proteins and their biological properties, providing updated detailed data describing their function and focusing in detail on their structural characteristics. Furthermore, we review some unclear aspects of the four proteins encoded by the ORFs, highlighting the current key information gaps and discussing potential novel experimental strategies for shedding light on those issues.
Collapse
Affiliation(s)
- Florencia Cancela
- grid.11630.350000000121657640Sección Virología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Ofelia Noceti
- grid.414402.70000 0004 0469 0889Programa Nacional de Trasplante Hepático y Unidad Docente Asistencial Centro Nacional de Tratamiento Hepatobiliopancreatico. Hospital Central de las Fuerzas Armadas, Montevideo, Uruguay
| | - Juan Arbiza
- grid.11630.350000000121657640Sección Virología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Santiago Mirazo
- grid.11630.350000000121657640Sección Virología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay ,grid.11630.350000000121657640Departamento de Bacteriología y Virología, Instituto de Higiene, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay ,Av. Alfredo Navarro 3051, PC 11600 Montevideo, Uruguay
| |
Collapse
|
13
|
Tietcheu Galani BR, Ayissi Owona VB, Guemmogne Temdie RJ, Metzger K, Atsama Amougou M, Djamen Chuisseu PD, Fondjo Kouam A, Ngounoue Djuidje M, Aliouat-Denis CM, Cocquerel L, Fewou Moundipa P. In silico and in vitro screening of licensed antimalarial drugs for repurposing as inhibitors of hepatitis E virus. In Silico Pharmacol 2021; 9:35. [PMID: 33959472 PMCID: PMC8093904 DOI: 10.1007/s40203-021-00093-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 04/16/2021] [Indexed: 01/15/2023] Open
Abstract
ABSTRACT Hepatitis E virus (HEV) infection is emerging in Cameroon and represents one of the most common causes of acute hepatitis and jaundice. Moreover, earlier reports showed evidence of falciparum malaria/HEVcoexistence. Although the Sofosbuvir/Ribavirin combination was recently proposed in the treatment of HEV-infected patients, no specific antiviral drug has been approved so far, thereby urging the search for new therapies. Fortunately, drug repurposing offers a good alternative to this end. In this study, we report the in silico and in vitro activities of 8 licensed antimalarial drugs and two anti-hepatitis C virus agents used as references (Sofosbuvir, and Ribavirin), for repurposing as antiviral inhibitors against HEV. Compounds were docked against five HEV-specific targets including the Zinc-binding non-structural protein (6NU9), RNA-dependent RNA polymerase (RdRp), cryoEM structure of HEV VLP, genotype 1 (6LAT), capsid protein ORF-2, genotype 3 (2ZTN), and the E2s domain of genotype 1 (3GGQ) using the iGEMDOCK software and their pharmacokinetic profiles and toxicities were predicted using ADMETlab2.0. Their in vitro effects were also assessed on a gt 3 p6Gluc replicon system using the luciferase reporter assay. The docking results showed that Sofosbuvir had the best binding affinities with 6NU9 (- 98.22 kcal/mol), RdRp (- 113.86 kcal/mol), 2ZTN (- 106.96 kcal/mol), while Ribavirin better collided with 6LAT (- 99.33 kcal/mol). Interestingly, Lumefantrine showed the best affinity with 3GGQ (-106.05 kcal/mol). N-desethylamodiaquine and Amodiaquine presented higher binding scores with 6NU9 (- 93.5 and - 89.9 kcal/mol respectively vs - 80.83 kcal/mol), while Lumefantrine had the greatest energies with RdRp (- 102 vs - 84.58), and Pyrimethamine and N-desethylamodiaquine had stronger affinities with 2ZTN compared to Ribavirin (- 105.17 and - 102.65 kcal/mol vs - 96.04 kcal/mol). The biological screening demonstrated a significant (P < 0.001) antiviral effect on replication with 1 µM N-desethylamodiaquine, the major metabolite of Amodiaquine. However, Lumefantrine showed no effect at the tested concentrations (1, 5, and 10 µM). The biocomputational analysis of the pharmacokinetic profile of both drugs revealed a low permeability of Lumefantrine and a specific inactivation by CYP3A2 which might partly contribute to the short half-time of this drug. In conclusion, Amodiaquine and Lumefantrine may be good antimalarial drug candidates for repurposing against HEV. Further in vitro and in vivo experiments are necessary to validate these predictions. GRAPHIC ABSTRACT SUPPLEMENTARY INFORMATION The online version contains supplementary material available at 10.1007/s40203-021-00093-y.
Collapse
Affiliation(s)
- Borris Rosnay Tietcheu Galani
- Laboratory of Applied Biochemistry, Department of Biological Sciences, Faculty of Science, University of Ngaoundere, P.O. Box 454, Ngaoundere, Cameroon
- Laboratory of Pharmacology and Toxicology, Department of Biochemistry, Faculty ofScience, University of Yaounde I, P.O. Box 812, Yaounde, Cameroon
| | - Vincent Brice Ayissi Owona
- Laboratory of Pharmacology and Toxicology, Department of Biochemistry, Faculty ofScience, University of Yaounde I, P.O. Box 812, Yaounde, Cameroon
| | - Romeo Joel Guemmogne Temdie
- Laboratory of Medicinal Plants, Health, and Galenic Formulation, Department of Biological Sciences, Faculty of Science, University of Ngaoundere, P.O. Box 454, Ngaoundere, Cameroon
| | - Karoline Metzger
- University of Lille, CNRS, INSERM, CHU Lille, Pasteur Institute of Lille, U1019-UMR9017-CIIL-Center for Infection and Immunity of Lille, 59000 Lille, France
| | - Marie Atsama Amougou
- Laboratory of Pharmacology and Toxicology, Department of Biochemistry, Faculty ofScience, University of Yaounde I, P.O. Box 812, Yaounde, Cameroon
- Research Center for Emerging and Reemerging Infectious Diseases (CREMER-IMPM), Virology Unit, P.O. Box 906, Yaounde, Cameroon
| | - Pascal Dieudonné Djamen Chuisseu
- Department of Medicine, Medical and Biomedical Sciences, Higher Institute of Health Sciences, Université Des Montagnes, P.O. Box 208, Bangangte, Cameroon
| | - Arnaud Fondjo Kouam
- Laboratory of Pharmacology and Toxicology, Department of Biochemistry, Faculty ofScience, University of Yaounde I, P.O. Box 812, Yaounde, Cameroon
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Buea, P.O Box 63, Buea, South West Region Cameroon
| | - Marceline Ngounoue Djuidje
- Laboratory of Pharmacology and Toxicology, Department of Biochemistry, Faculty ofScience, University of Yaounde I, P.O. Box 812, Yaounde, Cameroon
| | - Cécile-Marie Aliouat-Denis
- University of Lille, CNRS, INSERM, CHU Lille, Pasteur Institute of Lille, U1019-UMR9017-CIIL-Center for Infection and Immunity of Lille, 59000 Lille, France
| | - Laurence Cocquerel
- University of Lille, CNRS, INSERM, CHU Lille, Pasteur Institute of Lille, U1019-UMR9017-CIIL-Center for Infection and Immunity of Lille, 59000 Lille, France
| | - Paul Fewou Moundipa
- Laboratory of Pharmacology and Toxicology, Department of Biochemistry, Faculty ofScience, University of Yaounde I, P.O. Box 812, Yaounde, Cameroon
| |
Collapse
|
14
|
Lhomme S, Migueres M, Abravanel F, Marion O, Kamar N, Izopet J. Hepatitis E Virus: How It Escapes Host Innate Immunity. Vaccines (Basel) 2020; 8:E422. [PMID: 32731452 PMCID: PMC7564545 DOI: 10.3390/vaccines8030422] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 07/23/2020] [Accepted: 07/24/2020] [Indexed: 12/12/2022] Open
Abstract
Hepatitis E virus (HEV) is a leading cause of viral hepatitis in the world. It is usually responsible for acute hepatitis, but can lead to a chronic infection in immunocompromised patients. The host's innate immune response is the first line of defense against a virus infection; there is growing evidence that HEV RNA is recognized by toll-like receptors (TLRs) and retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs), leading to interferon (IFN) production. The IFNs activate interferon-stimulated genes (ISGs) to limit HEV replication and spread. HEV has developed strategies to counteract this antiviral response, by limiting IFN induction and signaling. This review summarizes the advances in our knowledge of intracellular pathogen recognition, interferon and inflammatory response, and the role of virus protein in immune evasion.
Collapse
Affiliation(s)
- Sébastien Lhomme
- National Reference Center for Hepatitis E Virus, Toulouse Purpan University Hospital, 31300 Toulouse, France; (M.M.); (F.A.); (J.I.)
- INSERM UMR1043, CNRS UMR5282, Center for Pathophysiology of Toulouse Purpan, 31300 Toulouse, France; (O.M.); (N.K.)
- Université Toulouse III Paul Sabatier, 31330 Toulouse, France
| | - Marion Migueres
- National Reference Center for Hepatitis E Virus, Toulouse Purpan University Hospital, 31300 Toulouse, France; (M.M.); (F.A.); (J.I.)
- INSERM UMR1043, CNRS UMR5282, Center for Pathophysiology of Toulouse Purpan, 31300 Toulouse, France; (O.M.); (N.K.)
- Université Toulouse III Paul Sabatier, 31330 Toulouse, France
| | - Florence Abravanel
- National Reference Center for Hepatitis E Virus, Toulouse Purpan University Hospital, 31300 Toulouse, France; (M.M.); (F.A.); (J.I.)
- INSERM UMR1043, CNRS UMR5282, Center for Pathophysiology of Toulouse Purpan, 31300 Toulouse, France; (O.M.); (N.K.)
- Université Toulouse III Paul Sabatier, 31330 Toulouse, France
| | - Olivier Marion
- INSERM UMR1043, CNRS UMR5282, Center for Pathophysiology of Toulouse Purpan, 31300 Toulouse, France; (O.M.); (N.K.)
- Université Toulouse III Paul Sabatier, 31330 Toulouse, France
- Department of Nephrology and Organs Transplantation, Toulouse Rangueil University Hospital, 31400 Toulouse, France
| | - Nassim Kamar
- INSERM UMR1043, CNRS UMR5282, Center for Pathophysiology of Toulouse Purpan, 31300 Toulouse, France; (O.M.); (N.K.)
- Université Toulouse III Paul Sabatier, 31330 Toulouse, France
- Department of Nephrology and Organs Transplantation, Toulouse Rangueil University Hospital, 31400 Toulouse, France
| | - Jacques Izopet
- National Reference Center for Hepatitis E Virus, Toulouse Purpan University Hospital, 31300 Toulouse, France; (M.M.); (F.A.); (J.I.)
- INSERM UMR1043, CNRS UMR5282, Center for Pathophysiology of Toulouse Purpan, 31300 Toulouse, France; (O.M.); (N.K.)
- Université Toulouse III Paul Sabatier, 31330 Toulouse, France
| |
Collapse
|
15
|
Himoto T, Masaki T. Current Trends of Essential Trace Elements in Patients with Chronic Liver Diseases. Nutrients 2020; 12:nu12072084. [PMID: 32674425 PMCID: PMC7400835 DOI: 10.3390/nu12072084] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/16/2020] [Accepted: 06/29/2020] [Indexed: 02/06/2023] Open
Abstract
Essential trace elements play crucial roles in the maintenance of health, since they are involved in many metabolic pathways. A deficiency or an excess of some trace elements, including zinc, selenium, iron, and copper, frequently causes these metabolic disorders such as impaired glucose tolerance and dyslipidemia. The liver largely regulates most of the metabolism of trace elements, and accordingly, an impairment of liver functions can result in numerous metabolic disorders. The administration or depletion of these trace elements can improve such metabolic disorders and liver dysfunction. Recent advances in molecular biological techniques have helped to elucidate the putative mechanisms by which liver disorders evoke metabolic abnormalities that are due to deficiencies or excesses of these trace elements. A genome-wide association study revealed that a genetic polymorphism affected the metabolism of a specific trace element. Gut dysbiosis was also responsible for impairment of the metabolism of a trace element. This review focuses on the current trends of four trace elements in chronic liver diseases, including chronic hepatitis, liver cirrhosis, nonalcoholic fatty liver disease, and autoimmune liver diseases. The novel mechanisms by which the trace elements participated in the pathogenesis of the chronic liver diseases are also mentioned.
Collapse
Affiliation(s)
- Takashi Himoto
- Department of Medical Technology, Kagawa Prefectural University of Health Sciences, 281-1, Hara, Mure-Cho, Takamatsu, Kagawa 761-0123, Japan
- Correspondence: ; Tel.: +81-87-870-1240; Fax: +81-87-870-1202
| | - Tsutomu Masaki
- Department of Gastroenterology and Neurology, Kagawa University School of Medicine, Kagawa 761-0123, Japan;
| |
Collapse
|
16
|
First Crystal Structure of a Nonstructural Hepatitis E Viral Protein Identifies a Putative Novel Zinc-Binding Protein. J Virol 2019; 93:JVI.00170-19. [PMID: 31019049 DOI: 10.1128/jvi.00170-19] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 04/17/2019] [Indexed: 12/14/2022] Open
Abstract
Hepatitis E virus (HEV) is a 7.2-kb positive-sense, single-stranded RNA virus containing three partially overlapping reading frames, ORF1 to ORF3. All nonstructural proteins required for viral replication are encoded by ORF1 and are transcribed as a single transcript. Computational analysis of the complete ORF1 polyprotein identified a previously uncharacterized region of predicted secondary structure bordered by two disordered regions coinciding partially with a region predicted as a putative cysteine protease. Following successful cloning, expression, and purification of this region, the crystal structure of the identified protein was determined and identified to have considerable structural homology to a fatty acid binding domain. Further analysis of the structure revealed a metal binding site, shown unambiguously to specifically bind zinc via a nonclassical, potentially catalytic zinc-binding motif. Based on the structural homology of the HEV protein with known structures, along with the presence of a catalytic zinc-binding motif, it is possible that the identified protein corresponds to the HEV protease, which could require activation or repression through the binding of a fatty acid. This represents a significant step forward in the characterization and the understanding of the molecular mechanisms of the HEV genome. We present analysis for the first time of this identified nonstructural protein, expanding the knowledge and understanding of the complex mechanisms of HEV biology.IMPORTANCE Hepatitis E virus (HEV) is an emerging virus found predominately in developing countries; it causes an estimated 20 million infections, which result in approximately 57,000 deaths a year. Although it is known that the nonstructural proteins of HEV ORF1 are expressed as a single transcript, there is debate as to whether ORF1 functions as a single polyprotein or if it is processed into separate domains via a viral or endogenous cellular protease. Here we present the first structural and biophysical characterization of an HEV nonstructural protein using a construct that has partially overlapping boundaries with the predicted putative cysteine protease.
Collapse
|