1
|
Ying Q, Zhang X, Wang S, Gu T, Zhang J, Feng W, Li D, Dong Y, Wu X, Wang F. A Novel HTNV Budding Inhibitor Interferes the Interaction Between Viral Glycoprotein and Host ESCRT Accessory Protein ALIX. J Med Virol 2025; 97:e70182. [PMID: 39868900 DOI: 10.1002/jmv.70182] [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: 07/04/2024] [Revised: 10/21/2024] [Accepted: 11/06/2024] [Indexed: 01/28/2025]
Abstract
Virus budding is a critical step in the replication cycle of enveloped viruses, closely linked to viral spread, disease progression, and clinical outcomes. The budding of many enveloped RNA viruses is facilitated by the hijacking of the host endosomal sorting complex required for transport (ESCRT) proteins through viral late domains. These late domains are essential for progeny virus production and are highly conserved, making the interaction between late domains and host ESCRT proteins a potential target for the development of antiviral therapeutics. In this study, we elucidated the functional role of the conserved YRTL motif within the glycoprotein Gn cytoplasmic tail of Orthohantavirus hantanense (Hantaan virus, HTNV), demonstrating that HTNV production is regulated by the interaction between YRTL and the ESCRT accessory protein ALIX (ALG-2 interacting protein X). Through virtual molecule docking screening, followed by in vitro and in vivo assays, we discovered a novel compound, AN-329, which disrupts the YRTL-ALIX interaction and effectively inhibits infectious HTNV production, as well as Crimean-Congo hemorrhagic fever virus (CCHFV) and Rift Valley fever virus (RVFV) VLP release. This makes AN-329 a promising therapeutic candidate for reducing viral dissemination. Given that YRTL is conserved across many hantaviruses, our findings may serve as a prototype for the development of broad-spectrum antiviral drugs.
Collapse
Affiliation(s)
- Qikang Ying
- Department of Microbiology, School of Basic Medicine, Air Force Military Medical University, Xi'an, China
| | - Xiaoxiao Zhang
- Department of Microbiology, School of Basic Medicine, Air Force Military Medical University, Xi'an, China
| | - Shengzheng Wang
- Department of Medicinal Chemistry and Pharmaceutical Analysis, School of Pharmacy, Air Force Military Medical University, Xi'an, China
| | - Tianle Gu
- Department of Pathogen Biology, College of Basic Medical Science, Chongqing Medical University, Chongqing, China
| | - Junmei Zhang
- College of Life Sciences, Yan'an University, Yan'an, China
| | - Wenjie Feng
- College of Life Sciences, Yan'an University, Yan'an, China
| | - Dongjing Li
- College of Life Sciences, Yan'an University, Yan'an, China
| | - Yuhang Dong
- Department of Microbiology, School of Basic Medicine, Air Force Military Medical University, Xi'an, China
| | - Xingan Wu
- Department of Microbiology, School of Basic Medicine, Air Force Military Medical University, Xi'an, China
| | - Fang Wang
- Department of Microbiology, School of Basic Medicine, Air Force Military Medical University, Xi'an, China
| |
Collapse
|
2
|
Shkair L, Sharma D, Hamza S, Garanina E, Shakirova V, Khaertynova I, Markelova M, Pavelkina V, Rizvanov A, Khaiboullina S, Baranwal M, Martynova E. Cross-reactivity of hantavirus antibodies after immunization with PUUV antigens. Biotechnol Appl Biochem 2024; 71:1139-1153. [PMID: 38779849 DOI: 10.1002/bab.2604] [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: 11/29/2023] [Accepted: 05/05/2024] [Indexed: 05/25/2024]
Abstract
Nephropathia epidemica (NE), caused by Puumala (PUUV) orthohantavirus, is endemic in the Republic of Tatarstan (RT). There are limited options for NE prevention in RT. Currently, available vaccines are made using Haantan (HNTV) orthohantavirus antigens. In this study, the efficacy of microvesicles (MVs) loaded with PUUV antigens to induce the humoral immune response in small mammals was analyzed. Additionally, the cross-reactivity of serum from immunized small mammals and NE patients with HNTV, Dobrava, and Andes orthohantaviruses was investigated using nucleocapsid (N) protein peptide libraries. Finally, the selected peptides were analyzed for allergenicity, their ability to induce an autoimmune response, and their interaction with Class II HLA. Several N protein peptides were found to be cross-reactive with serum from MVs immunized small mammals. These cross-reactive epitopes were located in oligomerization perinuclear targeting and Daxx-interacting domains. Most cross-reactive peptides lack allergenic and autoimmune reactivity. Molecular docking revealed two cross-reacting peptides, N6 and N19, to have good binding with three Class II HLA alleles. These peptides could be candidates for developing vaccines and therapeutics for NE.
Collapse
Affiliation(s)
- Layaly Shkair
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Diksha Sharma
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, India
| | - Shaimaa Hamza
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Ekaterina Garanina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Venara Shakirova
- Department of Infectious Diseases, Kazan State Medical Academy, Kazan, Russia
| | - Ilsiyar Khaertynova
- Department of Infectious Diseases, Kazan State Medical Academy, Kazan, Russia
| | - Maria Markelova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Vera Pavelkina
- Infectious Diseases Department, National Research Ogarev Mordovia State University, Saransk, Russia
| | - Albert Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Svetlana Khaiboullina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Manoj Baranwal
- Infectious Diseases Department, National Research Ogarev Mordovia State University, Saransk, Russia
| | - Ekaterina Martynova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| |
Collapse
|
3
|
Kuzmin IV, Soto Acosta R, Pruitt L, Wasdin PT, Kedarinath K, Hernandez KR, Gonzales KA, Hill K, Weidner NG, Mire C, Engdahl TB, Moon WJ, Popov V, Crowe JE, Georgiev IS, Garcia-Blanco MA, Abbott RK, Bukreyev A. Comparison of uridine and N1-methylpseudouridine mRNA platforms in development of an Andes virus vaccine. Nat Commun 2024; 15:6421. [PMID: 39080316 PMCID: PMC11289437 DOI: 10.1038/s41467-024-50774-3] [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: 07/29/2023] [Accepted: 07/19/2024] [Indexed: 08/02/2024] Open
Abstract
The rodent-borne Andes virus (ANDV) causes a severe disease in humans. We developed an ANDV mRNA vaccine based on the M segment of the viral genome, either with regular uridine (U-mRNA) or N1-methylpseudouridine (m1Ψ-mRNA). Female mice immunized by m1Ψ-mRNA developed slightly greater germinal center (GC) responses than U-mRNA-immunized mice. Single cell RNA and BCR sequencing of the GC B cells revealed similar levels of activation, except an additional cluster of cells exhibiting interferon response in animals vaccinated with U-mRNA but not m1Ψ-mRNA. Similar immunoglobulin class-switching and somatic hypermutations were observed in response to the vaccines. Female Syrian hamsters were immunized via a prime-boost regimen with two doses of each vaccine. The titers of glycoprotein-binding antibodies were greater for U-mRNA construct than for m1Ψ-mRNA construct; however, the titers of ANDV-neutralizing antibodies were similar. Vaccinated animals were challenged with a lethal dose of ANDV, along with a naïve control group. All control animals and two animals vaccinated with a lower dose of m1Ψ-mRNA succumbed to infection whereas other vaccinated animals survived without evidence of virus replication. The data demonstrate the development of a protective vaccine against ANDV and the lack of a substantial effect of m1Ψ modification on immunogenicity and protection in rodents.
Collapse
MESH Headings
- Animals
- Female
- Mice
- Mesocricetus
- Uridine
- Viral Vaccines/immunology
- Viral Vaccines/administration & dosage
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Messenger/immunology
- Antibodies, Viral/immunology
- Orthohantavirus/immunology
- Orthohantavirus/genetics
- Antibodies, Neutralizing/immunology
- Germinal Center/immunology
- Pseudouridine/immunology
- Cricetinae
- mRNA Vaccines
- Hemorrhagic Fever, American/prevention & control
- Hemorrhagic Fever, American/immunology
- Hemorrhagic Fever, American/virology
- RNA, Viral/genetics
- RNA, Viral/immunology
- B-Lymphocytes/immunology
- Humans
- Vaccine Development
Collapse
Affiliation(s)
- Ivan V Kuzmin
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Galveston National Laboratory, Galveston, TX, USA
| | - Ruben Soto Acosta
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Galveston National Laboratory, Galveston, TX, USA
| | - Layne Pruitt
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Perry T Wasdin
- Vanderbilt University Medical Center, Vanderbilt Vaccine Center, Nashville, TN, USA
| | - Kritika Kedarinath
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Galveston National Laboratory, Galveston, TX, USA
| | - Keziah R Hernandez
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Galveston National Laboratory, Galveston, TX, USA
| | - Kristyn A Gonzales
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Kharighan Hill
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Nicole G Weidner
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Chad Mire
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Galveston National Laboratory, Galveston, TX, USA
| | - Taylor B Engdahl
- Vanderbilt University Medical Center, Vanderbilt Vaccine Center, Nashville, TN, USA
| | | | - Vsevolod Popov
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - James E Crowe
- Vanderbilt University Medical Center, Vanderbilt Vaccine Center, Nashville, TN, USA
| | - Ivelin S Georgiev
- Vanderbilt University Medical Center, Vanderbilt Vaccine Center, Nashville, TN, USA
| | - Mariano A Garcia-Blanco
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Robert K Abbott
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA.
| | - Alexander Bukreyev
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA.
- Galveston National Laboratory, Galveston, TX, USA.
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
4
|
Meola A, Guardado-Calvo P. Production and Purification of Hantavirus Glycoproteins in Drosophila melanogaster S2 Cells. Methods Mol Biol 2024; 2762:3-16. [PMID: 38315356 DOI: 10.1007/978-1-0716-3666-4_1] [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: 02/07/2024]
Abstract
Hantaviruses, are rodent-borne viruses found worldwide that are transmitted to humans through inhalation of contaminated excreta. They can cause a renal or a pulmonary syndrome, depending on the virus, and no effective treatment is currently available for either of these diseases. Hantaviral particles are covered by a protein lattice composed of two glycoproteins (Gn and Gc) that mediate adsorption to target cells and fusion with endosomal membranes, making them prime targets for neutralizing antibodies. Here we present the methodology to produce soluble recombinant glycoproteins in different conformations, either alone or as a stabilized Gn/Gc complex, using stably transfected Drosophila S2 cells.
Collapse
Affiliation(s)
- Annalisa Meola
- G5 Structural Biology of Infectious Diseases, Institut Pasteur, Université Paris Cité, Paris, France
| | - Pablo Guardado-Calvo
- G5 Structural Biology of Infectious Diseases, Institut Pasteur, Université Paris Cité, Paris, France.
| |
Collapse
|
5
|
Afzal S, Ali L, Batool A, Afzal M, Kanwal N, Hassan M, Safdar M, Ahmad A, Yang J. Hantavirus: an overview and advancements in therapeutic approaches for infection. Front Microbiol 2023; 14:1233433. [PMID: 37901807 PMCID: PMC10601933 DOI: 10.3389/fmicb.2023.1233433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 09/25/2023] [Indexed: 10/31/2023] Open
Abstract
Hantaviruses are a significant and emerging global public health threat, impacting more than 200,000 individuals worldwide each year. The single-stranded RNA viruses belong to the Hantaviridae family and are responsible for causing two acute febrile diseases in humans: Hantavirus pulmonary syndrome (HPS) and hemorrhagic fever with renal syndrome (HFRS). Currently, there are no licensed treatments or vaccines available globally for HTNV infection. Various candidate drugs have shown efficacy in increasing survival rates during the early stages of HTNV infection. Some of these drugs include lactoferrin, ribavirin, ETAR, favipiravir and vandetanib. Immunotherapy utilizing neutralizing antibodies (NAbs) generated from Hantavirus convalescent patients show efficacy against HTNV. Monoclonal antibodies such as MIB22 and JL16 have demonstrated effectiveness in protecting against HTNV infection. The development of vaccines and antivirals, used independently and/or in combination, is critical for elucidating hantaviral infections and the impact on public health. RNA interference (RNAi) arised as an emerging antiviral therapy, is a highly specific degrades RNA, with post-transcriptional mechanism using eukaryotic cells platform. That has demonstrated efficacy against a wide range of viruses, both in vitro and in vivo. Recent antiviral methods involve using small interfering RNA (siRNA) and other, immune-based therapies to target specific gene segments (S, M, or L) of the Hantavirus. This therapeutic approach enhances viral RNA clearance through the RNA interference process in Vero E6 cells or human lung microvascular endothelial cells. However, the use of siRNAs faces challenges due to their low biological stability and limited in vivo targeting ability. Despite their successful inhibition of Hantavirus replication in host cells, their antiviral efficacy may be hindered. In the current review, we focus on advances in therapeutic strategies, as antiviral medications, immune-based therapies and vaccine candidates aimed at enhancing the body's ability to control the progression of Hantavirus infections, with the potential to reduce the risk of severe disease.
Collapse
Affiliation(s)
- Samia Afzal
- CEMB, University of the Punjab, Lahore, Pakistan
| | - Liaqat Ali
- Department of Biological Sciences, National University of Medical Sciences (NUMS), Rawalpindi, Pakistan
| | - Anum Batool
- Department of Biological Sciences, National University of Medical Sciences (NUMS), Rawalpindi, Pakistan
| | - Momina Afzal
- CEMB, University of the Punjab, Lahore, Pakistan
| | - Nida Kanwal
- Department of Biological Sciences, National University of Medical Sciences (NUMS), Rawalpindi, Pakistan
| | | | | | - Atif Ahmad
- CEMB, University of the Punjab, Lahore, Pakistan
| | - Jing Yang
- Wuhan Institute of Biological Products Co., Ltd., Wuhan, Hubei, China
| |
Collapse
|
6
|
LaPointe A, Gale M, Kell AM. Orthohantavirus Replication in the Context of Innate Immunity. Viruses 2023; 15:1130. [PMID: 37243216 PMCID: PMC10220641 DOI: 10.3390/v15051130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/05/2023] [Accepted: 05/06/2023] [Indexed: 05/28/2023] Open
Abstract
Orthohantaviruses are rodent-borne, negative-sense RNA viruses that are capable of causing severe vascular disease in humans. Over the course of viral evolution, these viruses have tailored their replication cycles in such a way as to avoid and/or antagonize host innate immune responses. In the rodent reservoir, this results in life long asymptomatic infections. However, in hosts other than its co-evolved reservoir, the mechanisms for subduing the innate immune response may be less efficient or absent, potentially leading to disease and/or viral clearance. In the case of human orthohantavirus infection, the interaction of the innate immune response with viral replication is thought to give rise to severe vascular disease. The orthohantavirus field has made significant advancements in understanding how these viruses replicate and interact with host innate immune responses since their identification by Dr. Ho Wang Lee and colleagues in 1976. Therefore, the purpose of this review, as part of this special issue dedicated to Dr. Lee, was to summarize the current knowledge of orthohantavirus replication, how viral replication activates innate immunity, and how the host antiviral response, in turn, impacts viral replication.
Collapse
Affiliation(s)
- Autumn LaPointe
- Department of Molecular Genetics and Microbiology, University of New Mexico, 915 Camino de Salud NE, Albuquerque, NM 87131, USA
| | - Michael Gale
- Department of Immunology, Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA 98109, USA
| | - Alison M. Kell
- Department of Molecular Genetics and Microbiology, University of New Mexico, 915 Camino de Salud NE, Albuquerque, NM 87131, USA
| |
Collapse
|
7
|
Abstract
Unlike many segmented negative-sense RNA viruses, most members of the Bunyavirales bud at Golgi membranes, as opposed to the plasma membrane. Central players in this assembly process are the envelope glycoproteins, Gn and Gc, which upon translation undergo proteolytic processing, glycosylation and trafficking to the Golgi, where they interact with ribonucleoprotein genome segments and bud into Golgi-derived compartments. The processes involved in genome packaging during virion assembly can lead to the generation of reassorted viruses, if a cell is co-infected with two different bunyaviruses, due to mismatching of viral genome segment packaging. This can lead to viruses with high pathogenic potential, as demonstrated by the emergence of Schmallenberg virus. This review focuses on the assembly pathways of tri-segmented bunyaviruses, highlighting some areas in need of further research to understand these important pathogens with zoonotic potential.
Collapse
Affiliation(s)
- Jake Barker
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Luis L P daSilva
- Departamento de Biologia Celular e Molecular, Centro de Pesquisa em Virologia, Faculdade de Medicina de Ribeirão Preto, University of São Paulo, State of São Paulo, Brazil
| | - Colin M Crump
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| |
Collapse
|
8
|
Sehgal A, Mehta S, Sahay K, Martynova E, Rizvanov A, Baranwal M, Chandy S, Khaiboullina S, Kabwe E, Davidyuk Y. Hemorrhagic Fever with Renal Syndrome in Asia: History, Pathogenesis, Diagnosis, Treatment, and Prevention. Viruses 2023; 15:v15020561. [PMID: 36851775 PMCID: PMC9966805 DOI: 10.3390/v15020561] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/30/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
Hemorrhagic Fever with Renal Syndrome (HFRS) is the most frequently diagnosed zoonosis in Asia. This zoonotic infection is the result of exposure to the virus-contaminated aerosols. Orthohantavirus infection may cause Hemorrhagic Fever with Renal Syndrome (HRFS), a disease that is characterized by acute kidney injury and increased vascular permeability. Several species of orthohantaviruses were identified as causing infection, where Hantaan, Puumala, and Seoul viruses are most common. Orthohantaviruses are endemic to several Asian countries, such as China, South Korea, and Japan. Along with those countries, HFRS tops the list of zoonotic infections in the Far Eastern Federal District of Russia. Recently, orthohantavirus circulation was demonstrated in small mammals in Thailand and India, where orthohantavirus was not believed to be endemic. In this review, we summarized the current data on orthohantaviruses in Asia. We gave the synopsis of the history and diversity of orthohantaviruses in Asia. We also described the clinical presentation and current understanding of the pathogenesis of orthohantavirus infection. Additionally, conventional and novel approaches for preventing and treating orthohantavirus infection are discussed.
Collapse
Affiliation(s)
- Ayushi Sehgal
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala 147004, India
| | - Sanya Mehta
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala 147004, India
| | - Kritika Sahay
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala 147004, India
| | - Ekaterina Martynova
- OpenLab “Gene and Cell Technologies”, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia
| | - Albert Rizvanov
- OpenLab “Gene and Cell Technologies”, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia
| | - Manoj Baranwal
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala 147004, India
| | - Sara Chandy
- Childs Trust Medical Research Foundation, Kanchi Kamakoti Childs Trust Hospital, Chennai 600034, India
| | - Svetlana Khaiboullina
- OpenLab “Gene and Cell Technologies”, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia
| | - Emmanuel Kabwe
- OpenLab “Gene and Cell Technologies”, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia
- Kazan Research Institute of Epidemiology and Microbiology, Kazan 420012, Russia
| | - Yuriy Davidyuk
- OpenLab “Gene and Cell Technologies”, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia
- Correspondence:
| |
Collapse
|
9
|
Barbosa NS, Concha JO, daSilva LLP, Crump CM, Graham SC. Oropouche Virus Glycoprotein Topology and Cellular Requirements for Glycoprotein Secretion. J Virol 2023; 97:e0133122. [PMID: 36475765 PMCID: PMC9888203 DOI: 10.1128/jvi.01331-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 11/19/2022] [Indexed: 12/13/2022] Open
Abstract
Oropouche virus (OROV; genus Orthobunyavirus) is the etiological agent of Oropouche fever, a debilitating febrile illness common in South America. We used recombinant expression of the OROV M polyprotein, which encodes the surface glycoproteins Gn and Gc plus the nonstructural protein NSm, to probe the cellular determinants for OROV assembly and budding. Gn and Gc self-assemble and are secreted independently of NSm. Mature OROV Gn has two predicted transmembrane domains that are crucial for glycoprotein translocation to the Golgi complex and glycoprotein secretion, and unlike related orthobunyaviruses, both transmembrane domains are retained during Gn maturation. Disruption of Golgi function using the drugs brefeldin A and monensin inhibits glycoprotein secretion. Infection studies have previously shown that the cellular endosomal sorting complexes required for transport (ESCRT) machinery is recruited to Golgi membranes during OROV assembly and that ESCRT activity is required for virus secretion. A dominant-negative form of the ESCRT-associated ATPase VPS4 significantly reduces recombinant OROV glycoprotein secretion and blocks virus release from infected cells, and VPS4 partly colocalizes with OROV glycoproteins and membranes costained with Golgi markers. Furthermore, immunoprecipitation and fluorescence microscopy experiments demonstrate that OROV glycoproteins interact with the ESCRT-III component CHMP6, with overexpression of a dominant-negative form of CHMP6 significantly reducing OROV glycoprotein secretion. Taken together, our data highlight differences in M polyprotein processing across orthobunyaviruses, indicate that Golgi and ESCRT function are required for glycoprotein secretion, and identify CHMP6 as an ESCRT-III component that interacts with OROV glycoproteins. IMPORTANCE Oropouche virus causes Oropouche fever, a debilitating illness common in South America that is characterized by high fever, headache, myalgia, and vomiting. The tripartite genome of this zoonotic virus is capable of reassortment, and there have been multiple epidemics of Oropouche fever in South America over the last 50 years, making Oropouche virus infection a significant threat to public health. However, the molecular characteristics of this arbovirus are poorly understood. We developed a recombinant protein expression system to investigate the cellular determinants of OROV glycoprotein maturation and secretion. We show that the proteolytic processing of the M polypeptide, which encodes the surface glycoproteins (Gn and Gc) plus a nonstructural protein (NSm), differs between OROV and its close relative Bunyamwera virus. Furthermore, we demonstrate that OROV M glycoprotein secretion requires the cellular endosomal sorting complexes required for transport (ESCRT) membrane-remodeling machinery and identify that the OROV glycoproteins interact with the ESCRT protein CHMP6.
Collapse
Affiliation(s)
- Natalia S. Barbosa
- Center for Virus Research, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Juan O. Concha
- Center for Virus Research, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Luis L. P. daSilva
- Center for Virus Research, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Colin M. Crump
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Stephen C. Graham
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
10
|
Menke L, Sperber HS, Aji AK, Chiantia S, Schwarzer R, Sieben C. Advances in fluorescence microscopy for orthohantavirus research. Microscopy (Oxf) 2023:6987530. [PMID: 36639937 DOI: 10.1093/jmicro/dfac075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 11/30/2022] [Accepted: 01/12/2023] [Indexed: 01/15/2023] Open
Abstract
Orthohantaviruses are important zoonotic pathogens responsible for a considerable disease burden globally. Partly due to our incomplete understanding of orthohantavirus replication, there is currently no effective antiviral treatment available. Recently, novel microscopy techniques and cutting-edge, automated image analysis algorithms have emerged, enabling to study cellular, subcellular and even molecular processes in unprecedented detail and depth. To date, fluorescence light microscopy allows us to visualize viral and cellular components and macromolecular complexes in live cells which in turn enables the study of specific steps of the viral replication cycle such as particle entry or protein trafficking at high temporal and spatial resolution. In this review, we highlight how fluorescence microscopy has provided new insights and improved our understanding of orthohantavirus biology. We discuss technical challenges such as studying live infected cells, give alternatives with recombinant protein expression and highlight future opportunities for example the application of super-resolution microscopy techniques, which has shown great potential in studies of different cellular processes and viral pathogens.
Collapse
Affiliation(s)
- Laura Menke
- Nanoscale Infection Biology Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Hannah S Sperber
- Institute for Translational HIV Research, University Hospital Essen, Essen, Germany
| | - Amit Koikkarah Aji
- University of Potsdam, Institute of Biochemistry and Biology, Department of Physical Biochemistry, Potsdam, Germany
| | - Salvatore Chiantia
- University of Potsdam, Institute of Biochemistry and Biology, Department of Physical Biochemistry, Potsdam, Germany
| | - Roland Schwarzer
- Institute for Translational HIV Research, University Hospital Essen, Essen, Germany
| | - Christian Sieben
- Nanoscale Infection Biology Helmholtz Centre for Infection Research, Braunschweig, Germany.,Institute of Genetics, Technische Universität Braunschweig, Braunschweig, Germany
| |
Collapse
|
11
|
Generation of Multiple Arbovirus-like Particles Using a Rapid Recombinant Vaccinia Virus Expression Platform. Pathogens 2022; 11:pathogens11121505. [PMID: 36558839 PMCID: PMC9785247 DOI: 10.3390/pathogens11121505] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/06/2022] [Accepted: 12/07/2022] [Indexed: 12/13/2022] Open
Abstract
As demonstrated by the 2015 Zika virus outbreak in the Americas, emerging and re-emerging arboviruses are public health threats that warrant research investment for the development of effective prophylactics and therapeutics. Many arboviral diseases are underreported, neglected, or of low prevalence, yet they all have the potential to cause outbreaks of local and international concern. Here, we show the production of virus-like particles (VLPs) using a rapid and efficient recombinant vaccinia virus (VACV) expression system for five tick- and mosquito-borne arboviruses: Powassan virus (POWV), Heartland virus (HRTV), severe fever with thrombocytopenia syndrome virus (SFTSV), Bourbon virus (BRBV) and Mayaro virus (MAYV). We detected the expression of arbovirus genes of interest by Western blot and observed the expression of VLPs that resemble native virions under transmission electron microscopy. We were also able to improve the secretion of POWV VLPs by modifying the signal sequence within the capsid gene. This study describes the use of a rapid VACV platform for the production and purification of arbovirus VLPs that can be used as subunit or vectored vaccines, and provides insights into the selection of arbovirus genes for VLP formation and genetic modifications to improve VLP secretion and yield.
Collapse
|
12
|
Rissanen I, Krumm SA, Stass R, Whitaker A, Voss JE, Bruce EA, Rothenberger S, Kunz S, Burton DR, Huiskonen JT, Botten JW, Bowden TA, Doores KJ. Structural Basis for a Neutralizing Antibody Response Elicited by a Recombinant Hantaan Virus Gn Immunogen. mBio 2021; 12:e0253120. [PMID: 34225492 PMCID: PMC8406324 DOI: 10.1128/mbio.02531-20] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Hantaviruses are a group of emerging pathogens capable of causing severe disease upon zoonotic transmission to humans. The mature hantavirus surface presents higher-order tetrameric assemblies of two glycoproteins, Gn and Gc, which are responsible for negotiating host cell entry and constitute key therapeutic targets. Here, we demonstrate that recombinantly derived Gn from Hantaan virus (HTNV) elicits a neutralizing antibody response (serum dilution that inhibits 50% infection [ID50], 1:200 to 1:850) in an animal model. Using antigen-specific B cell sorting, we isolated monoclonal antibodies (mAbs) exhibiting neutralizing and non-neutralizing activity, termed mAb HTN-Gn1 and mAb nnHTN-Gn2, respectively. Crystallographic analysis reveals that these mAbs target spatially distinct epitopes at disparate sites of the N-terminal region of the HTNV Gn ectodomain. Epitope mapping onto a model of the higher order (Gn-Gc)4 spike supports the immune accessibility of the mAb HTN-Gn1 epitope, a hypothesis confirmed by electron cryo-tomography of the antibody with virus-like particles. These data define natively exposed regions of the hantaviral Gn that can be targeted in immunogen design. IMPORTANCE The spillover of pathogenic hantaviruses from rodent reservoirs into the human population poses a continued threat to human health. Here, we show that a recombinant form of the Hantaan virus (HTNV) surface-displayed glycoprotein, Gn, elicits a neutralizing antibody response in rabbits. We isolated a neutralizing (HTN-Gn1) and a non-neutralizing (nnHTN-Gn2) monoclonal antibody and provide the first molecular-level insights into how the Gn glycoprotein may be targeted by the antibody-mediated immune response. These findings may guide rational vaccine design approaches focused on targeting the hantavirus glycoprotein envelope.
Collapse
Affiliation(s)
- Ilona Rissanen
- Division of Structural Biology, Wellcome Centre for Human Genetics, grid.4991.5University of Oxford, Oxford, United Kingdom
- Institute of Biotechnology and Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Stefanie A. Krumm
- Department of Infectious Diseases, King's College London, London, United Kingdom
| | - Robert Stass
- Division of Structural Biology, Wellcome Centre for Human Genetics, grid.4991.5University of Oxford, Oxford, United Kingdom
| | - Annalis Whitaker
- Division of Immunobiology, Department of Medicine, Larner College of Medicine, grid.59062.38University of Vermont, Burlington, Vermont, USA
- Cellular, Molecular, and Biomedical Sciences Graduate Program, grid.59062.38University of Vermont, Burlington, Vermont, USA
| | - James E. Voss
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
| | - Emily A. Bruce
- Division of Immunobiology, Department of Medicine, Larner College of Medicine, grid.59062.38University of Vermont, Burlington, Vermont, USA
- Department of Microbiology and Molecular Genetics, Larner College of Medicine, grid.59062.38University of Vermont, Burlington, Vermont, USA
| | - Sylvia Rothenberger
- Institute of Microbiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Stefan Kunz
- Institute of Microbiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Dennis R. Burton
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, Massachusetts, USA
| | - Juha T. Huiskonen
- Division of Structural Biology, Wellcome Centre for Human Genetics, grid.4991.5University of Oxford, Oxford, United Kingdom
- Institute of Biotechnology and Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Jason W. Botten
- Division of Immunobiology, Department of Medicine, Larner College of Medicine, grid.59062.38University of Vermont, Burlington, Vermont, USA
- Department of Microbiology and Molecular Genetics, Larner College of Medicine, grid.59062.38University of Vermont, Burlington, Vermont, USA
| | - Thomas A. Bowden
- Division of Structural Biology, Wellcome Centre for Human Genetics, grid.4991.5University of Oxford, Oxford, United Kingdom
| | - Katie J. Doores
- Department of Infectious Diseases, King's College London, London, United Kingdom
| |
Collapse
|
13
|
Guardado-Calvo P, Rey FA. The surface glycoproteins of hantaviruses. Curr Opin Virol 2021; 50:87-94. [PMID: 34418649 DOI: 10.1016/j.coviro.2021.07.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/22/2021] [Accepted: 07/26/2021] [Indexed: 11/30/2022]
Abstract
Hantaviruses are rodent-borne viruses distributed worldwide, transmitted through the air and with the ability to spread from person to person. They maintain a non-symptomatic persistent infection in their rodent hosts, but their spillover to humans produces a renal or pulmonary syndrome associated with high fatality rates. Hantavirus particles are lipid-enveloped and display a characteristic surface lattice built up of tetragonal spikes composed of two glycoproteins, Gn and Gc. The pleomorphism of these particles has hindered cryo-EM efforts to obtain detailed structural information and only by using a combination of X-ray crystallography and cryo-electron tomography it was possible to build an atomic model of the surface lattice. Here we review these structural efforts and the unanticipated evolutionary relations between hantaviruses and alphaviruses highlighted by these studies.
Collapse
Affiliation(s)
| | - Félix A Rey
- Institut Pasteur, Structural Virology Unit, and CNRS UMR 3569, Paris, France
| |
Collapse
|
14
|
Meier K, Thorkelsson SR, Quemin ERJ, Rosenthal M. Hantavirus Replication Cycle-An Updated Structural Virology Perspective. Viruses 2021; 13:1561. [PMID: 34452426 PMCID: PMC8402763 DOI: 10.3390/v13081561] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/31/2021] [Accepted: 08/02/2021] [Indexed: 11/17/2022] Open
Abstract
Hantaviruses infect a wide range of hosts including insectivores and rodents and can also cause zoonotic infections in humans, which can lead to severe disease with possible fatal outcomes. Hantavirus outbreaks are usually linked to the population dynamics of the host animals and their habitats being in close proximity to humans, which is becoming increasingly important in a globalized world. Currently there is neither an approved vaccine nor a specific and effective antiviral treatment available for use in humans. Hantaviruses belong to the order Bunyavirales with a tri-segmented negative-sense RNA genome. They encode only five viral proteins and replicate and transcribe their genome in the cytoplasm of infected cells. However, many details of the viral amplification cycle are still unknown. In recent years, structural biology methods such as cryo-electron tomography, cryo-electron microscopy, and crystallography have contributed essentially to our understanding of virus entry by membrane fusion as well as genome encapsidation by the nucleoprotein. In this review, we provide an update on the hantavirus replication cycle with a special focus on structural virology aspects.
Collapse
Affiliation(s)
- Kristina Meier
- Department of Virology, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany;
| | - Sigurdur R. Thorkelsson
- Centre for Structural Systems Biology, Leibniz Institute for Experimental Virology, University of Hamburg, 22607 Hamburg, Germany;
| | - Emmanuelle R. J. Quemin
- Centre for Structural Systems Biology, Leibniz Institute for Experimental Virology, University of Hamburg, 22607 Hamburg, Germany;
| | - Maria Rosenthal
- Department of Virology, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany;
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 22525 Hamburg, Germany
| |
Collapse
|
15
|
Minakshi P, Ghosh M, Kumar R, Brar B, Lambe UP, Banerjee S, Ranjan K, Kumar B, Goel P, Malik YS, Prasad G. An Insight into Nanomedicinal Approaches to Combat Viral Zoonoses. Curr Top Med Chem 2021; 20:915-962. [PMID: 32209041 DOI: 10.2174/1568026620666200325114400] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 12/31/2019] [Accepted: 12/31/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Emerging viral zoonotic diseases are one of the major obstacles to secure the "One Health" concept under the current scenario. Current prophylactic, diagnostic and therapeutic approaches often associated with certain limitations and thus proved to be insufficient for customizing rapid and efficient combating strategy against the highly transmissible pathogenic infectious agents leading to the disastrous socio-economic outcome. Moreover, most of the viral zoonoses originate from the wildlife and poor knowledge about the global virome database renders it difficult to predict future outbreaks. Thus, alternative management strategy in terms of improved prophylactic vaccines and their delivery systems; rapid and efficient diagnostics and effective targeted therapeutics are the need of the hour. METHODS Structured literature search has been performed with specific keywords in bibliographic databases for the accumulation of information regarding current nanomedicine interventions along with standard books for basic virology inputs. RESULTS Multi-arrayed applications of nanomedicine have proved to be an effective alternative in all the aspects regarding the prevention, diagnosis, and control of zoonotic viral diseases. The current review is focused to outline the applications of nanomaterials as anti-viral vaccines or vaccine/drug delivery systems, diagnostics and directly acting therapeutic agents in combating the important zoonotic viral diseases in the recent scenario along with their potential benefits, challenges and prospects to design successful control strategies. CONCLUSION This review provides significant introspection towards the multi-arrayed applications of nanomedicine to combat several important zoonotic viral diseases.
Collapse
Affiliation(s)
- Prasad Minakshi
- Department of Animal Biotechnology, LLR University of Veterinary and Animal Sciences, Hisar-125001, Haryana, 125004, India
| | - Mayukh Ghosh
- Department of Veterinary Physiology and Biochemistry, RGSC, Banaras Hindu University, Mirzapur (UP) - 231001, India
| | - Rajesh Kumar
- Department of Veterinary Physiology and Biochemistry, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar-125001, Haryana, 125004, India
| | - Basanti Brar
- Department of Animal Biotechnology, LLR University of Veterinary and Animal Sciences, Hisar-125001, Haryana, 125004, India
| | - Upendra P Lambe
- Department of Animal Biotechnology, LLR University of Veterinary and Animal Sciences, Hisar-125001, Haryana, 125004, India
| | - Somesh Banerjee
- Department of Veterinary Microbiology, Immunology Section, LUVAS, Hisar-125004, India
| | - Koushlesh Ranjan
- Department of Veterinary Physiology and Biochemistry, Sardar Vallabhbhai Patel University of Agriculture and Technology, Meerut, 250110, India
| | | | - Parveen Goel
- Department of Veterinary Medicine, LLR University of Veterinary and Animal Sciences, Hisar, Haryana, 125004, India
| | - Yashpal S Malik
- Division of Standardisation, Indian Veterinary Research Institute Izatnagar - Bareilly (UP) - 243122, India
| | - Gaya Prasad
- Sardar Vallabhbhai Patel University of Agriculture and Technology, Meerut, UP, 250110, India
| |
Collapse
|
16
|
Munir N, Jahangeer M, Hussain S, Mahmood Z, Ashiq M, Ehsan F, Akram M, Ali Shah SM, Riaz M, Sana A. Hantavirus diseases pathophysiology, their diagnostic strategies and therapeutic approaches: A review. Clin Exp Pharmacol Physiol 2021; 48:20-34. [PMID: 32894790 DOI: 10.1111/1440-1681.13403] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 08/24/2020] [Accepted: 08/25/2020] [Indexed: 12/20/2022]
Abstract
Hantaviruses are enveloped negative (-) single-stranded RNA viruses belongs to Hantaviridae family, hosted by small rodents and entering into the human body through inhalation, causing haemorrhagic fever with renal syndrome (HFRS) and hantavirus pulmonary syndrome (HPS) also known as hantavirus cardiopulmonary syndrome (HCPS). Hantaviruses infect approximately more than 200 000 people annually all around the world and its mortality rate is about 35%-40%. Hantaviruses play significant role in affecting the target cells as these inhibit the apoptotic factor in these cells. These viruses impair the integrity of endothelial barrier due to an excessive innate immune response that is proposed to be central in the pathogenesis and is a hallmark of hantavirus disease. A wide range of different diagnostic tools including polymerase chain reaction (PCR), focus reduction neutralization test (FRNT), enzyme-linked immunosorbent assay (ELISA), immunoblot assay (IBA), immunofluorescence assay (IFA), and other molecular techniques are used as detection tools for hantavirus in the human body. Now the availability of therapeutic modalities is the major challenge to control this deadly virus because still no FDA approved drug or vaccine is available. Antiviral agents, DNA-based vaccines, polyclonal and monoclonal antibodies neutralized the viruses so these techniques are considered as the hope for the treatment of hantavirus disease. This review has been compiled to provide a comprehensive overview of hantaviruses disease, its pathophysiology, diagnostic tools and the treatment approaches to control the hantavirus infection.
Collapse
Affiliation(s)
- Naveed Munir
- Department of Biochemistry, Government College University Faisalabad, Faisalabad, Pakistan
| | - Muhammad Jahangeer
- Department of Biochemistry, Government College University Faisalabad, Faisalabad, Pakistan
| | - Shoukat Hussain
- Department of Biochemistry, Government College University Faisalabad, Faisalabad, Pakistan
| | - Zahed Mahmood
- Department of Biochemistry, Government College University Faisalabad, Faisalabad, Pakistan
| | - Mehvish Ashiq
- Department of Chemistry, The Women University Multan, Multan, Pakistan
| | - Fatima Ehsan
- Department of Biochemistry, Government College University Faisalabad, Faisalabad, Pakistan
| | - Muhammad Akram
- Department of Eastern Medicine, Directorate of Medical Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| | - Syed Muhammad Ali Shah
- Department of Eastern Medicine, Directorate of Medical Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| | - Muhammad Riaz
- Department of Allied Health Sciences, Sargodha Medical College, University of Sargodha, Sargodha, Pakistan
| | - Aneezah Sana
- Department of Biochemistry, Government College University Faisalabad, Faisalabad, Pakistan
| |
Collapse
|
17
|
Rissanen I, Stass R, Krumm SA, Seow J, Hulswit RJG, Paesen GC, Hepojoki J, Vapalahti O, Lundkvist Å, Reynard O, Volchkov V, Doores KJ, Huiskonen JT, Bowden TA. Molecular rationale for antibody-mediated targeting of the hantavirus fusion glycoprotein. eLife 2020; 9:e58242. [PMID: 33349334 PMCID: PMC7755396 DOI: 10.7554/elife.58242] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 11/26/2020] [Indexed: 01/22/2023] Open
Abstract
The intricate lattice of Gn and Gc glycoprotein spike complexes on the hantavirus envelope facilitates host-cell entry and is the primary target of the neutralizing antibody-mediated immune response. Through study of a neutralizing monoclonal antibody termed mAb P-4G2, which neutralizes the zoonotic pathogen Puumala virus (PUUV), we provide a molecular-level basis for antibody-mediated targeting of the hantaviral glycoprotein lattice. Crystallographic analysis demonstrates that P-4G2 binds to a multi-domain site on PUUV Gc and may preclude fusogenic rearrangements of the glycoprotein that are required for host-cell entry. Furthermore, cryo-electron microscopy of PUUV-like particles in the presence of P-4G2 reveals a lattice-independent configuration of the Gc, demonstrating that P-4G2 perturbs the (Gn-Gc)4 lattice. This work provides a structure-based blueprint for rationalizing antibody-mediated targeting of hantaviruses.
Collapse
Affiliation(s)
- Ilona Rissanen
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of OxfordOxfordUnited Kingdom
- Helsinki Institute of Life Science HiLIFE, University of HelsinkiHelsinkiFinland
- Molecular and Integrative Biosciences Research Programme, The Faculty of Biological and Environmental Sciences, University of HelsinkiHelsinkiFinland
| | - Robert Stass
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of OxfordOxfordUnited Kingdom
| | - Stefanie A Krumm
- Department of Infectious Diseases, King's College London, Guy's HospitalLondonUnited Kingdom
| | - Jeffrey Seow
- Department of Infectious Diseases, King's College London, Guy's HospitalLondonUnited Kingdom
| | - Ruben JG Hulswit
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of OxfordOxfordUnited Kingdom
| | - Guido C Paesen
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of OxfordOxfordUnited Kingdom
| | - Jussi Hepojoki
- Institute of Veterinary Pathology, Vetsuisse Faculty, University of ZürichZürichSwitzerland
- Department of Virology, Medicum, Faculty of Medicine, University of HelsinkiHelsinkiFinland
| | - Olli Vapalahti
- Departments of Virology and Veterinary Biosciences, University of Helsinki and HUSLAB, Helsinki University HospitalHelsinkiFinland
| | - Åke Lundkvist
- Zoonosis Science Center, Department of Medical Biochemistry and Microbiology, Uppsala UniversityUppsalaSweden
| | - Olivier Reynard
- CIRI, Centre International de Recherche en Infectiologie, INSERM U1111, CNRS UMR5308, Université LyonLyonFrance
| | - Viktor Volchkov
- CIRI, Centre International de Recherche en Infectiologie, INSERM U1111, CNRS UMR5308, Université LyonLyonFrance
| | - Katie J Doores
- Department of Infectious Diseases, King's College London, Guy's HospitalLondonUnited Kingdom
| | - Juha T Huiskonen
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of OxfordOxfordUnited Kingdom
- Helsinki Institute of Life Science HiLIFE, University of HelsinkiHelsinkiFinland
- Molecular and Integrative Biosciences Research Programme, The Faculty of Biological and Environmental Sciences, University of HelsinkiHelsinkiFinland
| | - Thomas A Bowden
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of OxfordOxfordUnited Kingdom
| |
Collapse
|
18
|
Mittler E, Dieterle ME, Kleinfelter LM, Slough MM, Chandran K, Jangra RK. Hantavirus entry: Perspectives and recent advances. Adv Virus Res 2019; 104:185-224. [PMID: 31439149 DOI: 10.1016/bs.aivir.2019.07.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Hantaviruses are important zoonotic pathogens of public health importance that are found on all continents except Antarctica and are associated with hemorrhagic fever with renal syndrome (HFRS) in the Old World and hantavirus pulmonary syndrome (HPS) in the New World. Despite the significant disease burden they cause, no FDA-approved specific therapeutics or vaccines exist against these lethal viruses. The lack of available interventions is largely due to an incomplete understanding of hantavirus pathogenesis and molecular mechanisms of virus replication, including cellular entry. Hantavirus Gn/Gc glycoproteins are the only viral proteins exposed on the surface of virions and are necessary and sufficient to orchestrate virus attachment and entry. In vitro studies have implicated integrins (β1-3), DAF/CD55, and gC1qR as candidate receptors that mediate viral attachment for both Old World and New World hantaviruses. Recently, protocadherin-1 (PCDH1) was demonstrated as a requirement for cellular attachment and entry of New World hantaviruses in vitro and lethal HPS in vivo, making it the first clade-specific host factor to be identified. Attachment of hantavirus particles to cellular receptors induces their internalization by clathrin-mediated, dynamin-independent, or macropinocytosis-like mechanisms, followed by particle trafficking to an endosomal compartment where the fusion of viral and endosomal membranes can occur. Following membrane fusion, which requires cholesterol and acid pH, viral nucleocapsids escape into the cytoplasm and launch genome replication. In this review, we discuss the current mechanistic understanding of hantavirus entry, highlight gaps in our existing knowledge, and suggest areas for future inquiry.
Collapse
Affiliation(s)
- Eva Mittler
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Maria Eugenia Dieterle
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Lara M Kleinfelter
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Megan M Slough
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States.
| | - Rohit K Jangra
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States.
| |
Collapse
|
19
|
Progress on the Prevention and Treatment of Hantavirus Disease. Viruses 2019; 11:v11070610. [PMID: 31277410 PMCID: PMC6669544 DOI: 10.3390/v11070610] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 06/28/2019] [Accepted: 06/29/2019] [Indexed: 12/22/2022] Open
Abstract
Hantaviruses, members of the order Bunyavirales, family Hantaviridae, have a world-wide distribution and are responsible for greater than 150,000 cases of disease per year. The spectrum of disease associated with hantavirus infection include hemorrhagic fever with renal syndrome (HFRS) and hantavirus pulmonary syndrome (HPS) also known as hantavirus cardiopulmonary syndrome (HCPS). There are currently no FDA-approved vaccines or treatments for these hantavirus diseases. This review provides a summary of the status of vaccine and antiviral treatment efforts including those tested in animal models or human clinical trials.
Collapse
|
20
|
Bignon EA, Albornoz A, Guardado-Calvo P, Rey FA, Tischler ND. Molecular organization and dynamics of the fusion protein Gc at the hantavirus surface. eLife 2019; 8:46028. [PMID: 31180319 PMCID: PMC6609335 DOI: 10.7554/elife.46028] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 06/10/2019] [Indexed: 01/01/2023] Open
Abstract
The hantavirus envelope glycoproteins Gn and Gc mediate virion assembly and cell entry, with Gc driving fusion of viral and endosomal membranes. Although the X-ray structures and overall arrangement of Gn and Gc on the hantavirus spikes are known, their detailed interactions are not. Here we show that the lateral contacts between spikes are mediated by the same 2-fold contacts observed in Gc crystals at neutral pH, allowing the engineering of disulfide bonds to cross-link spikes. Disrupting the observed dimer interface affects particle assembly and overall spike stability. We further show that the spikes display a temperature-dependent dynamic behavior at neutral pH, alternating between ‘open’ and ‘closed’ forms. We show that the open form exposes the Gc fusion loops but is off-pathway for productive Gc-induced membrane fusion and cell entry. These data also provide crucial new insights for the design of optimized Gn/Gc immunogens to elicit protective immune responses. Hantaviruses infect rodents and other small mammals, but do not harm them. When transmitted to humans, often through rodent urine, feces or saliva, they can cause serious and even fatal diseases. Currently, there are no known methods that effectively prevent hantavirus infections or treat the diseases that they cause. During an infection, viruses invade the cells of their host. A hantavirus interacts with target cells through proteins on its surface called Gn and Gc glycoproteins. Previous work has shown that these glycoproteins are organized in bundles of four Gn and four Gc proteins, termed spikes, which project from the membrane that surrounds the virus. The Gc protein changes shape when it is activated and exposes a hidden region that can insert into the membrane of the target cell. The Gc proteins then change shape again to force the cell to fuse with the viral membrane. This process allows the virus to be taken up into the cell, where it can replicate. While the structures of each viral glycoprotein have been determined in isolation, it was not known how they interact within the Gn/Gc spike. Such information is crucial to understand how the viruses infect cells and which areas are exposed to the immune system of the host – and so could be targeted by antiviral treatments. Bignon et al. have now identified the molecular contacts that occur between spikes and interconnect them into a grid-like lattice on the surface of the virus. Genetically altering specific sections of the Gc glycoprotein strengthened or weakened these contacts, which correspondingly increased or decreased how stable the spike was. Preventing the contacts from forming resulted in cells releasing fewer virus-like particles. Bignon et al. also show that at the body temperature of mammals, the shape of the spike fluctuates between an ‘open’ form that exposes the region of Gc that inserts into the cell membrane, and a ‘closed’ form that hides this region. However, when Gc is activated, the open form becomes unable to cause the viral and cell membranes to fuse together. Together, the results presented by Bignon et al. help us to understand how changes to the hantavirus surface enable the virus to infect cells. This knowledge will help researchers to design vaccines that protect against hantavirus infections.
Collapse
Affiliation(s)
- Eduardo A Bignon
- Laboratorio de Virología Molecular, Fundación Ciencia & Vida, Santiago, Chile
| | - Amelina Albornoz
- Laboratorio de Virología Molecular, Fundación Ciencia & Vida, Santiago, Chile
| | - Pablo Guardado-Calvo
- Structural Virology Unit, Virology Department, Institut Pasteur, CNRS UMR 3569, Paris, France
| | - Félix A Rey
- Structural Virology Unit, Virology Department, Institut Pasteur, CNRS UMR 3569, Paris, France
| | - Nicole D Tischler
- Laboratorio de Virología Molecular, Fundación Ciencia & Vida, Santiago, Chile
| |
Collapse
|
21
|
Sperber HS, Welke RW, Petazzi RA, Bergmann R, Schade M, Shai Y, Chiantia S, Herrmann A, Schwarzer R. Self-association and subcellular localization of Puumala hantavirus envelope proteins. Sci Rep 2019; 9:707. [PMID: 30679542 PMCID: PMC6345964 DOI: 10.1038/s41598-018-36879-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 11/28/2018] [Indexed: 01/08/2023] Open
Abstract
Hantavirus assembly and budding are governed by the surface glycoproteins Gn and Gc. In this study, we investigated the glycoproteins of Puumala, the most abundant Hantavirus species in Europe, using fluorescently labeled wild-type constructs and cytoplasmic tail (CT) mutants. We analyzed their intracellular distribution, co-localization and oligomerization, applying comprehensive live, single-cell fluorescence techniques, including confocal microscopy, imaging flow cytometry, anisotropy imaging and Number&Brightness analysis. We demonstrate that Gc is significantly enriched in the Golgi apparatus in absence of other viral components, while Gn is mainly restricted to the endoplasmic reticulum (ER). Importantly, upon co-expression both glycoproteins were found in the Golgi apparatus. Furthermore, we show that an intact CT of Gc is necessary for efficient Golgi localization, while the CT of Gn influences protein stability. Finally, we found that Gn assembles into higher-order homo-oligomers, mainly dimers and tetramers, in the ER while Gc was present as mixture of monomers and dimers within the Golgi apparatus. Our findings suggest that PUUV Gc is the driving factor of the targeting of Gc and Gn to the Golgi region, while Gn possesses a significantly stronger self-association potential.
Collapse
Affiliation(s)
- Hannah Sabeth Sperber
- Institute for Biology, IRI Life Science, Humboldt-Universität zu Berlin, Invalidenstr. 42, 10115, Berlin, Germany.,Vitalant Research Institute, 270 Masonic Ave, San Francisco, CA, 94118, USA
| | - Robert-William Welke
- Institute for Biology, IRI Life Science, Humboldt-Universität zu Berlin, Invalidenstr. 42, 10115, Berlin, Germany
| | - Roberto Arturo Petazzi
- University of Potsdam, Institute of Biochemistry and Biology, Karl-Liebknecht-Str. 24-25, 14476, Potsdam, Germany
| | - Ronny Bergmann
- Institute for Biology, IRI Life Science, Humboldt-Universität zu Berlin, Invalidenstr. 42, 10115, Berlin, Germany
| | - Matthias Schade
- Institute for Biology, IRI Life Science, Humboldt-Universität zu Berlin, Invalidenstr. 42, 10115, Berlin, Germany
| | - Yechiel Shai
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Salvatore Chiantia
- University of Potsdam, Institute of Biochemistry and Biology, Karl-Liebknecht-Str. 24-25, 14476, Potsdam, Germany
| | - Andreas Herrmann
- Institute for Biology, IRI Life Science, Humboldt-Universität zu Berlin, Invalidenstr. 42, 10115, Berlin, Germany.
| | - Roland Schwarzer
- Institute for Biology, IRI Life Science, Humboldt-Universität zu Berlin, Invalidenstr. 42, 10115, Berlin, Germany. .,Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel. .,Gladstone Institute of Virology and Immunology, 1650 Owens Street, San Francisco, CA, 95158, USA.
| |
Collapse
|
22
|
Two Point Mutations in Old World Hantavirus Glycoproteins Afford the Generation of Highly Infectious Recombinant Vesicular Stomatitis Virus Vectors. mBio 2019; 10:mBio.02372-18. [PMID: 30622188 PMCID: PMC6325249 DOI: 10.1128/mbio.02372-18] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human hantavirus infections cause hantavirus pulmonary syndrome in the Americas and hemorrhagic fever with renal syndrome (HFRS) in Eurasia. No FDA-approved vaccines and therapeutics exist for these deadly viruses, and their development is limited by the requirement for high biocontainment. In this study, we identified and characterized key amino acid changes in the surface glycoproteins of HFRS-causing Hantaan virus that enhance their incorporation into recombinant vesicular stomatitis virus (rVSV) particles. The replication-competent rVSVs encoding Hantaan virus and Dobrava-Belgrade virus glycoproteins described in this work provide a powerful and facile system to study hantavirus entry under lower biocontainment and may have utility as hantavirus vaccines. Rodent-to-human transmission of hantaviruses is associated with severe disease. Currently, no FDA-approved, specific antivirals or vaccines are available, and the requirement for high biocontainment (biosafety level 3 [BSL-3]) laboratories limits hantavirus research. To study hantavirus entry in a BSL-2 laboratory, we set out to generate replication-competent, recombinant vesicular stomatitis viruses (rVSVs) bearing the Gn and Gc (Gn/Gc) entry glycoproteins. As previously reported, rVSVs bearing New World hantavirus Gn/Gc were readily rescued from cDNAs, but their counterparts bearing Gn/Gc from the Old World hantaviruses, Hantaan virus (HTNV) or Dobrava-Belgrade virus (DOBV), were refractory to rescue. However, serial passage of the rescued rVSV-HTNV Gn/Gc virus markedly increased its infectivity and capacity for cell-to-cell spread. This gain in viral fitness was associated with the acquisition of two point mutations: I532K in the cytoplasmic tail of Gn and S1094L in the membrane-proximal stem of Gc. Follow-up experiments with rVSVs and single-cycle VSV pseudotypes confirmed these results. Mechanistic studies revealed that both mutations were determinative and contributed to viral infectivity in a synergistic manner. Our findings indicate that the primary mode of action of these mutations is to relocalize HTNV Gn/Gc from the Golgi complex to the cell surface, thereby affording significantly enhanced Gn/Gc incorporation into budding VSV particles. Finally, I532K/S1094L mutations in DOBV Gn/Gc permitted the rescue of rVSV-DOBV Gn/Gc, demonstrating that incorporation of cognate mutations into other hantaviral Gn/Gc proteins could afford the generation of rVSVs that are otherwise challenging to rescue. The robust replication-competent rVSVs, bearing HTNV and DOBV Gn/Gc, reported herein may also have utility as vaccines.
Collapse
|
23
|
Protocadherin-1 is essential for cell entry by New World hantaviruses. Nature 2018; 563:559-563. [PMID: 30464266 DOI: 10.1038/s41586-018-0702-1] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 09/20/2018] [Indexed: 01/26/2023]
Abstract
The zoonotic transmission of hantaviruses from their rodent hosts to humans in North and South America is associated with a severe and frequently fatal respiratory disease, hantavirus pulmonary syndrome (HPS)1,2. No specific antiviral treatments for HPS are available, and no molecular determinants of in vivo susceptibility to hantavirus infection and HPS are known. Here we identify the human asthma-associated gene protocadherin-1 (PCDH1)3-6 as an essential determinant of entry and infection in pulmonary endothelial cells by two hantaviruses that cause HPS, Andes virus (ANDV) and Sin Nombre virus (SNV). In vitro, we show that the surface glycoproteins of ANDV and SNV directly recognize the outermost extracellular repeat domain of PCDH1-a member of the cadherin superfamily7,8-to exploit PCDH1 for entry. In vivo, genetic ablation of PCDH1 renders Syrian golden hamsters highly resistant to a usually lethal ANDV challenge. Targeting PCDH1 could provide strategies to reduce infection and disease caused by New World hantaviruses.
Collapse
|
24
|
Ying Q, Ma T, Cheng L, Zhang X, Truax AD, Ma R, Liu Z, Lei Y, Zhang L, Ye W, Zhang F, Xu Z, Shang L, Liu R, Wang F, Wu X. Construction and immunological characterization of CD40L or GM-CSF incorporated Hantaan virus like particle. Oncotarget 2018; 7:63488-63503. [PMID: 27542281 PMCID: PMC5325379 DOI: 10.18632/oncotarget.11329] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 07/10/2016] [Indexed: 12/22/2022] Open
Abstract
Infection of Hantaan virus (HTNV) usually causes hemorrhagic fever with renal syndrome (HFRS). China has the worst epidemic incidence of HFRS as well as high fatality. Inactivated whole virus has been used for HFRS vaccination, however there are still problems such as safety concerns. CD40 ligand (CD40L) and granulocyte macrophage colony-stimulating factor (GM-CSF) are well-known immune stimulating molecules that can enhance antigen presenting, lymphocytes activation and maturation, incorporation of CD40L and GM-CSF to the surface of virus like particles (VLPs) can greatly improve the vaccination effect. We constructed eukaryotic vectors expressing HTNV M segment and S segment, as well as vectors expressing HTNV M segment with CD40L or GM-CSF, our results showed successful production of CD40L or GM-CSF incorporated HTNV VLPs. In vitro stimulation with CD40L or GM-CSF anchored HTNV VLP showed enhanced activation of macrophages and DCs. CD40L/GM-CSF incorporated VLP can induce higher level of HTNV specific antibody and neutralizing antibody in mice. Immunized mice splenocytes showed higher ability of secreting IFN-γ and IL-2, as well as enhancing CTL activity. These results suggest CD40L/GM-CSF incorporated VLP can serve as prospective vaccine candidate.
Collapse
Affiliation(s)
- Qikang Ying
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Tiejun Ma
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Linfeng Cheng
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Xiaoxiao Zhang
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Agnieszka D Truax
- The Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7295, USA
| | - Ruixue Ma
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Ziyu Liu
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Yingfeng Lei
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Liang Zhang
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Wei Ye
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Fanglin Zhang
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Zhikai Xu
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Lei Shang
- Department of Statistics, Fourth Military Medical University, Xi'an, 710032, China
| | - Rongrong Liu
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Fang Wang
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Xingan Wu
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| |
Collapse
|
25
|
Crystal Structure of Glycoprotein C from a Hantavirus in the Post-fusion Conformation. PLoS Pathog 2016; 12:e1005948. [PMID: 27783673 PMCID: PMC5081248 DOI: 10.1371/journal.ppat.1005948] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 09/22/2016] [Indexed: 01/02/2023] Open
Abstract
Hantaviruses are important emerging human pathogens and are the causative agents of serious diseases in humans with high mortality rates. Like other members in the Bunyaviridae family their M segment encodes two glycoproteins, GN and GC, which are responsible for the early events of infection. Hantaviruses deliver their tripartite genome into the cytoplasm by fusion of the viral and endosomal membranes in response to the reduced pH of the endosome. Unlike phleboviruses (e.g. Rift valley fever virus), that have an icosahedral glycoprotein envelope, hantaviruses display a pleomorphic virion morphology as GN and GC assemble into spikes with apparent four-fold symmetry organized in a grid-like pattern on the viral membrane. Here we present the crystal structure of glycoprotein C (GC) from Puumala virus (PUUV), a representative member of the Hantavirus genus. The crystal structure shows GC as the membrane fusion effector of PUUV and it presents a class II membrane fusion protein fold. Furthermore, GC was crystallized in its post-fusion trimeric conformation that until now had been observed only in Flavi- and Togaviridae family members. The PUUV GC structure together with our functional data provides intriguing evolutionary and mechanistic insights into class II membrane fusion proteins and reveals new targets for membrane fusion inhibitors against these important pathogens. Hantaviruses (family: Bunyaviridae) encompass pathogens responsible to serious human diseases and economic burden worldwide. Following endocytosis, these enveloped RNA viruses are directed to an endosomal compartment where a sequence of pH-dependent conformational changes of the viral envelope glycoproteins mediates the fusion between the viral and endosomal membranes. The lack of high-resolution structural information for the entry of hantaviruses impair our ability to rationalize new treatments and prevention strategies. We determined the three-dimensional structure of a glycoprotein C from Puumala virus (PUUV) using X-ray crystallography. The two structures (at pH 6.0 and 8.0) were determined to 1.8 Å and 2.3 Å resolutions, respectively. Both structures reveal a class II membrane fusion protein in its post-fusion trimeric conformation with novel structural features in the trimer assembly and stabilization. Our structures suggest that neutralizing antibodies against GC target its conformational changes as inhibition mechanism and highlight new molecular targets for hantavirus-specific membrane fusion inhibitors. Furthermore, combined with the available structures of other class II proteins, we remodeled the evolutionary relationships between virus families encompassing these proteins.
Collapse
|
26
|
Mechanistic Insight into Bunyavirus-Induced Membrane Fusion from Structure-Function Analyses of the Hantavirus Envelope Glycoprotein Gc. PLoS Pathog 2016; 12:e1005813. [PMID: 27783711 PMCID: PMC5082683 DOI: 10.1371/journal.ppat.1005813] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 07/17/2016] [Indexed: 01/03/2023] Open
Abstract
Hantaviruses are zoonotic viruses transmitted to humans by persistently infected rodents, giving rise to serious outbreaks of hemorrhagic fever with renal syndrome (HFRS) or of hantavirus pulmonary syndrome (HPS), depending on the virus, which are associated with high case fatality rates. There is only limited knowledge about the organization of the viral particles and in particular, about the hantavirus membrane fusion glycoprotein Gc, the function of which is essential for virus entry. We describe here the X-ray structures of Gc from Hantaan virus, the type species hantavirus and responsible for HFRS, both in its neutral pH, monomeric pre-fusion conformation, and in its acidic pH, trimeric post-fusion form. The structures confirm the prediction that Gc is a class II fusion protein, containing the characteristic β-sheet rich domains termed I, II and III as initially identified in the fusion proteins of arboviruses such as alpha- and flaviviruses. The structures also show a number of features of Gc that are distinct from arbovirus class II proteins. In particular, hantavirus Gc inserts residues from three different loops into the target membrane to drive fusion, as confirmed functionally by structure-guided mutagenesis on the HPS-inducing Andes virus, instead of having a single "fusion loop". We further show that the membrane interacting region of Gc becomes structured only at acidic pH via a set of polar and electrostatic interactions. Furthermore, the structure reveals that hantavirus Gc has an additional N-terminal "tail" that is crucial in stabilizing the post-fusion trimer, accompanying the swapping of domain III in the quaternary arrangement of the trimer as compared to the standard class II fusion proteins. The mechanistic understandings derived from these data are likely to provide a unique handle for devising treatments against these human pathogens.
Collapse
|
27
|
Barriga GP, Villalón-Letelier F, Márquez CL, Bignon EA, Acuña R, Ross BH, Monasterio O, Mardones GA, Vidal SE, Tischler ND. Inhibition of the Hantavirus Fusion Process by Predicted Domain III and Stem Peptides from Glycoprotein Gc. PLoS Negl Trop Dis 2016; 10:e0004799. [PMID: 27414047 PMCID: PMC4945073 DOI: 10.1371/journal.pntd.0004799] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 06/02/2016] [Indexed: 12/17/2022] Open
Abstract
Hantaviruses can cause hantavirus pulmonary syndrome or hemorrhagic fever with renal syndrome in humans. To enter cells, hantaviruses fuse their envelope membrane with host cell membranes. Previously, we have shown that the Gc envelope glycoprotein is the viral fusion protein sharing characteristics with class II fusion proteins. The ectodomain of class II fusion proteins is composed of three domains connected by a stem region to a transmembrane anchor in the viral envelope. These fusion proteins can be inhibited through exogenous fusion protein fragments spanning domain III (DIII) and the stem region. Such fragments are thought to interact with the core of the fusion protein trimer during the transition from its pre-fusion to its post-fusion conformation. Based on our previous homology model structure for Gc from Andes hantavirus (ANDV), here we predicted and generated recombinant DIII and stem peptides to test whether these fragments inhibit hantavirus membrane fusion and cell entry. Recombinant ANDV DIII was soluble, presented disulfide bridges and beta-sheet secondary structure, supporting the in silico model. Using DIII and the C-terminal part of the stem region, the infection of cells by ANDV was blocked up to 60% when fusion of ANDV occurred within the endosomal route, and up to 95% when fusion occurred with the plasma membrane. Furthermore, the fragments impaired ANDV glycoprotein-mediated cell-cell fusion, and cross-inhibited the fusion mediated by the glycoproteins from Puumala virus (PUUV). The Gc fragments interfered in ANDV cell entry by preventing membrane hemifusion and pore formation, retaining Gc in a non-resistant homotrimer stage, as described for DIII and stem peptide inhibitors of class II fusion proteins. Collectively, our results demonstrate that hantavirus Gc shares not only structural, but also mechanistic similarity with class II viral fusion proteins, and will hopefully help in developing novel therapeutic strategies against hantaviruses. The infection of cells by enveloped viruses involves the fusion of membranes between viruses and cells. This process is mediated by viral fusion proteins that have been grouped into at least three structural classes. Membrane-enveloped hantaviruses are worldwide spread pathogens that can cause human disease with mortality rates reaching up to 50%, however, neither a therapeutic drug nor preventive measures are currently available. Here we show that the entrance of Andes hantavirus into target cells can be blocked by fragments derived from the Gc fusion protein that are analogous to inhibitory fragments of class II fusion proteins. The Gc fragments acted directly over the viral fusion process, preventing its late stages. Together, our data demonstrate that the hantavirus Gc protein shares not only structural, but also mechanistic similarity with class II fusion proteins, suggesting its evolution from a common or related ancestral fusion protein. Furthermore, the results outline novel approaches for therapeutic intervention.
Collapse
Affiliation(s)
- Gonzalo P. Barriga
- Molecular Virology Laboratory, Fundación Ciencia & Vida, Santiago, Chile
| | | | - Chantal L. Márquez
- Molecular Virology Laboratory, Fundación Ciencia & Vida, Santiago, Chile
| | - Eduardo A. Bignon
- Molecular Virology Laboratory, Fundación Ciencia & Vida, Santiago, Chile
| | - Rodrigo Acuña
- Molecular Virology Laboratory, Fundación Ciencia & Vida, Santiago, Chile
| | - Breyan H. Ross
- Laboratory of Structural Cell Biology, Department of Physiology, and Center for Interdisciplinary Studies of the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
| | - Octavio Monasterio
- Laboratorio de Biología Estructural y Molecular, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Gonzalo A. Mardones
- Laboratory of Structural Cell Biology, Department of Physiology, and Center for Interdisciplinary Studies of the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
| | - Simon E. Vidal
- Molecular Virology Laboratory, Fundación Ciencia & Vida, Santiago, Chile
| | - Nicole D. Tischler
- Molecular Virology Laboratory, Fundación Ciencia & Vida, Santiago, Chile
- * E-mail:
| |
Collapse
|
28
|
Liu J, Dai S, Wang M, Hu Z, Wang H, Deng F. Virus like particle-based vaccines against emerging infectious disease viruses. Virol Sin 2016; 31:279-87. [PMID: 27405928 PMCID: PMC7090901 DOI: 10.1007/s12250-016-3756-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 06/25/2016] [Indexed: 12/24/2022] Open
Abstract
Emerging infectious diseases are major threats to human health. Most severe viral disease outbreaks occur in developing regions where health conditions are poor. With increased international travel and business, the possibility of eventually transmitting infectious viruses between different countries is increasing. The most effective approach in preventing viral diseases is vaccination. However, vaccines are not currently available for numerous viral diseases. Virus-like particles (VLPs) are engineered vaccine candidates that have been studied for decades. VLPs are constructed by viral protein expression in various expression systems that promote the selfassembly of proteins into structures resembling virus particles. VLPs have antigenicity similar to that of the native virus, but are non-infectious as they lack key viral genetic material. VLP vaccines have attracted considerable research interest because they offer several advantages over traditional vaccines. Studies have shown that VLP vaccines can stimulate both humoral and cellular immune responses, which may offer effective antiviral protection. Here we review recent developments with VLP-based vaccines for several highly virulent emerging or re-emerging infectious diseases. The infectious agents discussed include RNA viruses from different virus families, such as the Arenaviridae, Bunyaviridae, Caliciviridae, Coronaviridae, Filoviridae, Flaviviridae, Orthomyxoviridae, Paramyxoviridae, and Togaviridae families.
Collapse
Affiliation(s)
- Jinliang Liu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Shiyu Dai
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Manli Wang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Zhihong Hu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Hualin Wang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Fei Deng
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.
| |
Collapse
|
29
|
Paneth Iheozor-Ejiofor R, Levanov L, Hepojoki J, Strandin T, Lundkvist Å, Plyusnin A, Vapalahti O. Vaccinia virus-free rescue of fluorescent replication-defective vesicular stomatitis virus and pseudotyping with Puumala virus glycoproteins for use in neutralization tests. J Gen Virol 2016; 97:1052-1059. [PMID: 26916544 DOI: 10.1099/jgv.0.000437] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Puumala virus (PUUV) grows slowly in cell culture. To study antigenic properties of PUUV, an amenable method for their expression would be beneficial. To achieve this, a replication-defective recombinant vesicular stomatitis virus, rVSVΔG*EGFP, was rescued using BSRT7/5 and encephalomyocarditis virus (EMCV) internal ribosomal entry site (IRES)-enabled rescue plasmids. Using these particles, pseudotypes bearing PUUV Sotkamo strain glycoproteins were produced, with titres in the range 105-108, and were used in pseudotype focus reduction neutralization tests (pFRNTs) with neutralizing monoclonal antibodies and patient sera. The results were compared with those from orthodox focus reduction neutralization tests (oFRNTs) using native PUUV with the same samples and showed a strong positive correlation (rs = 0.82) between the methods. While developing the system we identified three amino acids which were mutated in the Vero E6 cell culture adapted PUUV prototype Sotkamo strain sequence, and changing these residues was critical for expression and neutralizing antibody binding of PUUV glycoproteins.
Collapse
Affiliation(s)
| | - Lev Levanov
- Department of Virology, Medicum, Helsinki, Finland
| | | | | | - Åke Lundkvist
- Department of Medical Biochemistry and Microbiology, Microbiology-Immunology, Uppsala University, Sweden
| | - Alexander Plyusnin
- Department of Virology, Medicum, Helsinki, Finland.,Department of Medical Biochemistry and Microbiology, Microbiology-Immunology, Uppsala University, Sweden
| | - Olli Vapalahti
- Department of Virology, Medicum, Helsinki, Finland.,Department of Virology and Immunology, HUSLAB, Helsinki University Hospital, Helsinki, Finland.,Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
| |
Collapse
|
30
|
The application of virus-like particles as vaccines and biological vehicles. Appl Microbiol Biotechnol 2015; 99:10415-32. [PMID: 26454868 PMCID: PMC7080154 DOI: 10.1007/s00253-015-7000-8] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 09/01/2015] [Accepted: 09/04/2015] [Indexed: 01/04/2023]
Abstract
Virus-like particles (VLPs) can be spontaneously self-assembled by viral structural proteins under appropriate conditions in vitro while excluding the genetic material and potential replication probability. In addition, VLPs possess several features including can be rapidly produced in large quantities through existing expression systems, highly resembling native viruses in terms of conformation and appearance, and displaying repeated cluster of epitopes. Their capsids can be modified via genetic insertion or chemical conjugation which facilitating the multivalent display of a homologous or heterogeneous epitope antigen. Therefore, VLPs are considered as a safe and effective candidate of prophylactic and therapeutic vaccines. VLPs, with a diameter of approximately 20 to 150 nm, also have the characteristics of nanometer materials, such as large surface area, surface-accessible amino acids with reactive moieties (e.g., lysine and glutamic acid residues), inerratic spatial structure, and good biocompatibility. Therefore, assembled VLPs have great potential as a delivery system for specifically carrying a variety of materials. This review summarized recent researches on VLP development as vaccines and biological vehicles, which demonstrated the advantages and potential of VLPs in disease control and prevention and diagnosis. Then, the prospect of VLP biology application in the future is discussed as well.
Collapse
|
31
|
Beltrán-Ortiz CE, Starck-Mendez MF, Fernández Y, Farnós O, González EE, Rivas CI, Camacho F, Zuñiga FA, Toledo JR, Sánchez O. Expression and purification of the surface proteins from Andes virus. Protein Expr Purif 2015; 139:63-70. [PMID: 26374989 DOI: 10.1016/j.pep.2015.09.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 09/08/2015] [Accepted: 09/09/2015] [Indexed: 11/17/2022]
Abstract
Andes virus is the main causative agent of Hantavirus cardiopulmonary syndrome in South America. There are currently no vaccines or treatments against Andes virus. However, there are several evidences suggesting that antibodies against Andes virus envelope glycoproteins may be enough to confer full protection against Hantavirus cardiopulmonary syndrome. The goal of the present work was to express, purify and characterize the extracellular domains of Andes virus glycoproteins Gn and Gc. We generated two adenoviral vectors encoding the extracellular domains of Andes virus glycoproteins Gn and Gc. Both molecules were expressed by adenoviral transduction in SiHa cells. We found that sGc ectodomain was mainly secreted into the culture medium, whereas sGn was predominantly retained inside the cells. Both molecules were expressed at very low concentrations (below 1 μg/mL). Treatment with the proteasome inhibitor ALLN raised sGc concentration in the cell culture medium, but did not affect expression levels of sGn. Both ectodomains were purified by immobilized metal ion affinity chromatography, and were recognized by sera from persons previously exposed to Andes virus. To our knowledge, this is the first work that addresses the expression and purification of Andes virus glycoproteins Gn and Gc. Our results demonstrate that sGn and sGc maintain epitopes that are exposed on the surface of the viral envelope. However, our work also highlights the need to explore new strategies to achieve high-level expression of these proteins for development of a vaccine candidate against Andes virus.
Collapse
Affiliation(s)
- Camila E Beltrán-Ortiz
- Department of Pharmacology, School of Biological Sciences, University of Concepcion, Chile
| | - Maria F Starck-Mendez
- Department of Pharmacology, School of Biological Sciences, University of Concepcion, Chile
| | - Yaiza Fernández
- Department of Pharmacology, School of Biological Sciences, University of Concepcion, Chile
| | - Omar Farnós
- Department of Pharmacology, School of Biological Sciences, University of Concepcion, Chile
| | - Eddy E González
- Department of Physiopathology, School of Biological Sciences, University of Concepcion, Chile
| | - Coralia I Rivas
- Department of Physiopathology, School of Biological Sciences, University of Concepcion, Chile
| | - F Camacho
- Department of Pharmacology, School of Biological Sciences, University of Concepcion, Chile
| | - Felipe A Zuñiga
- Department of Clinical Biochemistry and Immunology, School of Pharmacia, University of Concepcion, Chile
| | - Jorge R Toledo
- Department of Physiopathology, School of Biological Sciences, University of Concepcion, Chile; Center for Biotechnology and Biomedicine Spa., Chile
| | - Oliberto Sánchez
- Department of Pharmacology, School of Biological Sciences, University of Concepcion, Chile; Center for Biotechnology and Biomedicine Spa., Chile.
| |
Collapse
|
32
|
Acuña R, Bignon EA, Mancini R, Lozach PY, Tischler ND. Acidification triggers Andes hantavirus membrane fusion and rearrangement of Gc into a stable post-fusion homotrimer. J Gen Virol 2015; 96:3192-3197. [PMID: 26310672 DOI: 10.1099/jgv.0.000269] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The hantavirus membrane fusion process is mediated by the Gc envelope glycoprotein from within endosomes. However, little is known about the specific mechanism that triggers Gc fusion activation, and its pre- and post-fusion conformations. We established cell-free in vitro systems to characterize hantavirus fusion activation. Low pH was sufficient to trigger the interaction of virus-like particles with liposomes. This interaction was dependent on a pre-fusion glycoprotein arrangement. Further, low pH induced Gc multimerization changes leading to non-reversible Gc homotrimers. These trimers were resistant to detergent, heat and protease digestion, suggesting characteristics of a stable post-fusion structure. No acid-dependent oligomerization rearrangement was detected for the trypsin-sensitive Gn envelope glycoprotein. Finally, acidification induced fusion of glycoprotein-expressing effector cells with non-susceptible CHO cells. Together, the data provide novel information on the Gc fusion trigger and its non-reversible activation involving lipid interaction, multimerization changes and membrane fusion which ultimately allow hantavirus entry into cells.
Collapse
Affiliation(s)
- Rodrigo Acuña
- Molecular Virology Laboratory, Fundación Ciencia & Vida, Av. Zanartu 1482, Santiago, Chile
| | - Eduardo A Bignon
- Molecular Virology Laboratory, Fundación Ciencia & Vida, Av. Zanartu 1482, Santiago, Chile
| | - Roberta Mancini
- Institute of Biochemistry, ETH Zurich, Schafmattstrasse 18, 8093 Zurich, Switzerland
| | - Pierre-Yves Lozach
- Department of Infectious Diseases, Virology, University Hospital Heidelberg, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - Nicole D Tischler
- Facultad de Ciencias Biologicas, Universidad Andres Bello, República 275, Santiago, Chile.,Molecular Virology Laboratory, Fundación Ciencia & Vida, Av. Zanartu 1482, Santiago, Chile
| |
Collapse
|
33
|
Pijlman GP. Enveloped virus-like particles as vaccines against pathogenic arboviruses. Biotechnol J 2015; 10:659-70. [DOI: 10.1002/biot.201400427] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 11/27/2014] [Accepted: 12/22/2014] [Indexed: 12/26/2022]
|
34
|
Hantavirus Gn and Gc envelope glycoproteins: key structural units for virus cell entry and virus assembly. Viruses 2014; 6:1801-22. [PMID: 24755564 PMCID: PMC4014721 DOI: 10.3390/v6041801] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 03/20/2014] [Accepted: 03/31/2014] [Indexed: 01/24/2023] Open
Abstract
In recent years, ultrastructural studies of viral surface spikes from three different genera within the Bunyaviridae family have revealed a remarkable diversity in their spike organization. Despite this structural heterogeneity, in every case the spikes seem to be composed of heterodimers formed by Gn and Gc envelope glycoproteins. In this review, current knowledge of the Gn and Gc structures and their functions in virus cell entry and exit is summarized. During virus cell entry, the role of Gn and Gc in receptor binding has not yet been determined. Nevertheless, biochemical studies suggest that the subsequent virus-membrane fusion activity is accomplished by Gc. Further, a class II fusion protein conformation has been predicted for Gc of hantaviruses, and novel crystallographic data confirmed such a fold for the Rift Valley fever virus (RVFV) Gc protein. During virus cell exit, the assembly of different viral components seems to be established by interaction of Gn and Gc cytoplasmic tails (CT) with internal viral ribonucleocapsids. Moreover, recent findings show that hantavirus glycoproteins accomplish important roles during virus budding since they self-assemble into virus-like particles. Collectively, these novel insights provide essential information for gaining a more detailed understanding of Gn and Gc functions in the early and late steps of the hantavirus infection cycle.
Collapse
|