1
|
Ander SE, Fish ER, da Silva MOL, Davenport BJ, Parks MG, Morrison TE. Basic patches on the E2 glycoprotein of eastern equine encephalitis virus influence viral vascular clearance and dissemination in mice. J Virol 2025:e0060225. [PMID: 40387358 DOI: 10.1128/jvi.00602-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Accepted: 04/24/2025] [Indexed: 05/20/2025] Open
Abstract
Previously, we found that chimeric Sindbis-eastern equine encephalitis virus (SINV-EEEV) particles can be removed from the murine blood circulation in a phagocyte-dependent manner which can be disrupted by either transient depletion of vascular heparan sulfate (HS) glycosaminoglycans (GAGs), or mutation of the viral E2 glycoprotein (K71/74/77A) associated with decreased GAG binding in vitro. Here, we further investigate the viral determinants of EEEV vascular clearance and evaluate their role in viremia development. We identified two large basic patches on the EEEV E2 glycoprotein which contain two known GAG-binding sites (K71/74/77 and K156/R157) and six additional basic residues (K10, R13, K56, R152, K231, and K232). We find that disruption of either basic patch by single alanine substitutions promotes prolonged retention of SINV-EEEV particles in the murine blood circulation in an experimental viremia model. Furthermore, we observed that the K156/R157A, K10A, and K231A mutations are also associated with similar viral dissemination in a mouse infection model as the attenuated K71/74/77A mutant. Surprisingly, despite known differences in GAG binding and potential alteration in receptor interactions, we find the initial dispersal of wild-type (WT) and mutant SINV-EEEV virions from the inoculation site to the draining lymph node to be equivalent at 1 hour post-subcutaneous inoculation. Moreover, our data suggest the higher viremia associated with mutation of the E2 basic patches may be attributed to evasion of viremic control by blood-filtering phagocytes. Overall, this study defines viral features of the EEEV E2 glycoprotein that influence tissue-specific viral dissemination and highlights the capacity of blood-filtering phagocytes to modulate EEEV viremia.IMPORTANCEVirus-GAG interactions have long been studied in vitro; however, investigating the impact of these interactions in vivo has been challenging. Previously, we showed that blood-filtering phagocytes and vascular HS mediate the removal of enhanced GAG-binding WT SINV-EEEV virions from the blood circulation in a reductionist, experimental viremia model. Here, we demonstrate that single-residue, charge-neutralizing mutations within basic patches of the E2 glycoprotein are sufficient both to promote viral evasion of vascular clearance and viral dissemination in an infection model. We also find that the WT and decreased GAG-binding SINV-EEEV virions traffic similarly from a subcutaneous inoculation until drainage into the bloodstream, upon which the WT virus is selectively depleted. These observations suggest viral dissemination is influenced by tissue-specific, virion-GAG interactions.
Collapse
Affiliation(s)
- Stephanie E Ander
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Microbiology and Immunology, Louisiana State University Health Shreveport, Shreveport, Louisiana, USA
| | - Erin R Fish
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Mariana O L da Silva
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Instituto de Microbiologia Paulo de Goes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bennett J Davenport
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - M Guston Parks
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Thomas E Morrison
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
2
|
Cai R, He Q, Wang Q, Tian L, Chen Z, Wu X, Sun J, Shao Y, Song X, Qi K, Tu J, Wang Z. Development of a reverse genetics system for Getah virus and characterization of rescued strains. Vet Res 2025; 56:80. [PMID: 40221809 PMCID: PMC11992769 DOI: 10.1186/s13567-025-01515-x] [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: 12/08/2024] [Accepted: 03/07/2025] [Indexed: 04/14/2025] Open
Abstract
Getah virus (GETV), a neglected and re-emerging mosquito-borne alphavirus, has become more serious and poses a potential threat to animal safety and public health. Given the lack of antivirals and vaccines against GETV, further development of tools, including reverse genetics techniques, is crucial for combating this pathogen. Herein, we describe the design and construction of a DNA-launched infectious clone for GETV. The full-length genome of the GETV HuN1 strain, flanked by the cytomegalovirus immediate-early (CMV) promoter sequence at the 5' end and the hepatitis delta virus ribozyme along with the bovine growth hormone termination and polyadenylation signal sequences at the 3' end, was packaged in a bacterial artificial chromosome vector to establish the GETV infectious clone pBR322-GETV-HuN1. In parallel, recombinant reporter viruses carrying the reporter gene EGFP between the E1 gene and the 3' UTR were constructed on the basis of the established CMV-driven cDNA clone. Both in vivo and in vitro experiments have shown that the rescued recombinant virus (rGETV-HuN1 and rGETV-EGFP) possesses viral biological activity similar to that of the parental virus. In summary, this study develops a concise and efficient GETV infectious cDNA clone and a recombinant virus carrying an EGFP reporter gene. The availability of GETV infectious clones will facilitate further studies on understanding the molecular mechanisms of GETV biology, virulence determinants, molecular pathogenesis, vaccine development and virus‒host interactions.
Collapse
Affiliation(s)
- Rongxuan Cai
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Veterinary Medicine, Anhui Agricultural University, Hefei, 230036, China
- Anhui Province Engineering Laboratory for Animal Food Quality and Biosafety, College of Veterinary Medicine, Anhui Agricultural University, Hefei, 230036, China
| | - Qi He
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Veterinary Medicine, Anhui Agricultural University, Hefei, 230036, China
- Anhui Province Engineering Laboratory for Animal Food Quality and Biosafety, College of Veterinary Medicine, Anhui Agricultural University, Hefei, 230036, China
| | - Qing Wang
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Veterinary Medicine, Anhui Agricultural University, Hefei, 230036, China
- Anhui Province Engineering Laboratory for Animal Food Quality and Biosafety, College of Veterinary Medicine, Anhui Agricultural University, Hefei, 230036, China
| | - Lan Tian
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Veterinary Medicine, Anhui Agricultural University, Hefei, 230036, China
- Anhui Province Engineering Laboratory for Animal Food Quality and Biosafety, College of Veterinary Medicine, Anhui Agricultural University, Hefei, 230036, China
| | - Zhe Chen
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Veterinary Medicine, Anhui Agricultural University, Hefei, 230036, China
- Anhui Province Engineering Laboratory for Animal Food Quality and Biosafety, College of Veterinary Medicine, Anhui Agricultural University, Hefei, 230036, China
| | - XiaoFeng Wu
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Veterinary Medicine, Anhui Agricultural University, Hefei, 230036, China
- Anhui Province Engineering Laboratory for Animal Food Quality and Biosafety, College of Veterinary Medicine, Anhui Agricultural University, Hefei, 230036, China
| | - Jiumeng Sun
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Veterinary Medicine, Anhui Agricultural University, Hefei, 230036, China
- Anhui Province Engineering Laboratory for Animal Food Quality and Biosafety, College of Veterinary Medicine, Anhui Agricultural University, Hefei, 230036, China
| | - Ying Shao
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Veterinary Medicine, Anhui Agricultural University, Hefei, 230036, China
- Anhui Province Engineering Laboratory for Animal Food Quality and Biosafety, College of Veterinary Medicine, Anhui Agricultural University, Hefei, 230036, China
| | - Xiangjun Song
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Veterinary Medicine, Anhui Agricultural University, Hefei, 230036, China
- Anhui Province Engineering Laboratory for Animal Food Quality and Biosafety, College of Veterinary Medicine, Anhui Agricultural University, Hefei, 230036, China
- Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei, 230036, China
| | - Kezong Qi
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Veterinary Medicine, Anhui Agricultural University, Hefei, 230036, China
- Anhui Province Engineering Laboratory for Animal Food Quality and Biosafety, College of Veterinary Medicine, Anhui Agricultural University, Hefei, 230036, China
- Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei, 230036, China
| | - Jian Tu
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Veterinary Medicine, Anhui Agricultural University, Hefei, 230036, China.
- Anhui Province Engineering Laboratory for Animal Food Quality and Biosafety, College of Veterinary Medicine, Anhui Agricultural University, Hefei, 230036, China.
- Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei, 230036, China.
| | - Zhenyu Wang
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Veterinary Medicine, Anhui Agricultural University, Hefei, 230036, China.
- Anhui Province Engineering Laboratory for Animal Food Quality and Biosafety, College of Veterinary Medicine, Anhui Agricultural University, Hefei, 230036, China.
- Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei, 230036, China.
| |
Collapse
|
3
|
Martin CK, Yin P, Kielian M. The sticky business of Alphavirus capsid-host interactions. Trends Microbiol 2025; 33:321-339. [PMID: 39665907 PMCID: PMC11916923 DOI: 10.1016/j.tim.2024.11.003] [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: 09/30/2024] [Revised: 11/01/2024] [Accepted: 11/06/2024] [Indexed: 12/13/2024]
Abstract
Alphaviruses are a serious threat to global health and can cause lethal encephalitic or arthritogenic disease in humans and animals. As there are no licensed antivirals, it is critical to improve our understanding of alphavirus interactions with the host cell. Here, we focus on the essential alphavirus protein capsid. While its roles in genome packaging and virus assembly have been well-studied, much less is known about capsid's interactions with host proteins and their functional relevance for infection. Recently, several new capsid interactor candidates were identified, collectively emphasising the complexity of capsid-host biology. In this review we summarise these novel interactor candidates, highlight capsid's emerging role in immune evasion, and discuss the challenges and opportunities arising from capsid-host interactions.
Collapse
Affiliation(s)
- Caroline K Martin
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Peiqi Yin
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Margaret Kielian
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
4
|
Pandey B, S S, Chatterjee A, Mangala Prasad V. Role of surface glycans in enveloped RNA virus infections: A structural perspective. Proteins 2025; 93:93-104. [PMID: 37994197 DOI: 10.1002/prot.26636] [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: 08/19/2023] [Revised: 10/16/2023] [Accepted: 10/30/2023] [Indexed: 11/24/2023]
Abstract
Enveloped RNA viruses have been causative agents of major pandemic outbreaks in the recent past. Glycans present on these virus surface proteins are critical for multiple processes during the viral infection cycle. Presence of glycans serves as a key determinant of immunogenicity, but intrinsic heterogeneity, dynamics, and evolutionary shifting of glycans in heavily glycosylated enveloped viruses confounds typical structure-function analysis. Glycosylation sites are also conserved across different viral families, which further emphasizes their functional significance. In this review, we summarize findings regarding structure-function correlation of glycans on enveloped RNA virus proteins.
Collapse
Affiliation(s)
- Bhawna Pandey
- Molecular Biophysics Unit, Indian Institute of Science, Bengaluru, Karnataka, India
| | - Srividhya S
- Molecular Biophysics Unit, Indian Institute of Science, Bengaluru, Karnataka, India
| | - Ananya Chatterjee
- Molecular Biophysics Unit, Indian Institute of Science, Bengaluru, Karnataka, India
| | - Vidya Mangala Prasad
- Molecular Biophysics Unit, Indian Institute of Science, Bengaluru, Karnataka, India
- Center for Infectious Disease Research, Indian Institute of Science, Bengaluru, Karnataka, India
| |
Collapse
|
5
|
Kim YC, Watanabe Y, Lücke AC, Song X, de Oliveira Souza R, Stass R, Azar SR, Rossi SL, Claser C, Kümmerer BM, Crispin M, Bowden TA, Huiskonen JT, Reyes-Sandoval A. Immunogenic recombinant Mayaro virus-like particles present natively assembled glycoprotein. NPJ Vaccines 2024; 9:243. [PMID: 39690153 PMCID: PMC11652679 DOI: 10.1038/s41541-024-01021-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 11/10/2024] [Indexed: 12/19/2024] Open
Abstract
Virus-like particles (VLPs) are an established vaccine platform and can be strong immunogens capable of eliciting both humoral and cellular immune responses against a range of pathogens. Here, we show by cryo-electron microscopy that VLPs of Mayaro virus, which contain envelope glycoproteins E1-E2 and capsid, exhibit an architecture that closely resembles native virus. In contrast to monomeric and soluble envelope 2 (E2) glycoprotein, both VLPs as well as the adenovirus and modified vaccinia virus Ankara (MVA) vaccine platforms expressing the equivalent envelope glycoproteins E1-E2, and capsid induced highly neutralising antibodies after immunisation. The levels of neutralising antibodies elicited by the viral-vectored vaccines of structural proteins and VLPs increased significantly upon boosting. Immunisation of Mayaro virus VLPs in mice with or without an adjuvant (poly:IC) yielded similar levels of neutralising antibodies suggesting that the VLPs may be used for immunisation without the need for an adjuvant. A single or two doses of non-adjuvanted 5 µg of MAYV VLP vaccination provided significant protection against viremia and MAYV-induced foot swelling in the C57BL/6 mouse challenge model. MAYV VLPs represent a non-infectious vaccine candidate, which may constitute a complementary option for future immunisation strategies against this important emerging alphavirus.
Collapse
Affiliation(s)
- Young Chan Kim
- Department of Paediatrics, Oxford Vaccine Group, University of Oxford, Oxford, UK.
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK.
| | - Yasunori Watanabe
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Arlen-Celina Lücke
- Institute of Virology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Xiyong Song
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, Helsinki, Finland
| | - Raquel de Oliveira Souza
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Robert Stass
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Sasha R Azar
- Department of Pathology and the Institute for Human Infection and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - Shannan L Rossi
- Department of Pathology and the Institute for Human Infection and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - Carla Claser
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Beate Mareike Kümmerer
- Institute of Virology, Medical Faculty, University of Bonn, Bonn, Germany
- German Centre for Infection Research (DZIF), Partner Site-Bonn-Cologne, Bonn, Germany
| | - Max Crispin
- School of Biological Sciences, University of Southampton, Southampton, UK
| | - Thomas A Bowden
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Juha T Huiskonen
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, Helsinki, Finland
| | - Arturo Reyes-Sandoval
- Instituto Politécnico Nacional, IPN. Av. Luis Enrique Erro s/n. Unidad Adolfo López Mateos, Mexico City, Mexico
| |
Collapse
|
6
|
Comas-Garcia M. How structural biology has changed our understanding of icosahedral viruses. J Virol 2024; 98:e0111123. [PMID: 39291975 PMCID: PMC11495149 DOI: 10.1128/jvi.01111-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024] Open
Abstract
Cryo-electron microscopy and tomography have allowed us to unveil the remarkable structure of icosahedral viruses. However, in the past few years, the idea that these viruses must have perfectly symmetric virions, but in some cases, it might not be true. This has opened the door to challenging paradigms in structural virology and raised new questions about the biological implications of "unusual" or "defective" symmetries and structures. Also, the continual improvement of these technologies, coupled with more rigorous sample purification protocols, improvements in data processing, and the use of artificial intelligence, has allowed solving the structure of sub-viral particles in highly heterogeneous samples and finding novel symmetries or structural defects. In this review, I initially analyzed the case of the symmetry and composition of hepatitis B virus-produced spherical sub-viral particles. Then, I focused on Alphaviruses as an example of "imperfect" icosahedrons and analyzed how structural biology has changed our understanding of the Alphavirus assembly and some biological implications arising from these discoveries.
Collapse
Affiliation(s)
- Mauricio Comas-Garcia
- Science Department, Autonomous University of San Luis Potosi, San Luis Potosí, Mexico
- High-Resolution Microscopy Section, Research Center for Health Sciences and Biomedicine, Autonomous University of San Luis Potosi, San Luis Potosi, Mexico
- Translational and Molecular Medicine Section, Research Center for Health Sciences and Biomedicine, Autonomous University of San Luis Potosi, San Luis Potosí, Mexico
| |
Collapse
|
7
|
Bell MG, Parry RH, Lee TSE, Habarugira G, McMahon IE, Thompson MA, Modhiran N, Watterson D, Batovska J, Lynch SE, Hall RA, Harrison JJ, Hobson-Peters J. Synthetic recovery of Yada Yada virus expands insect-specific alphavirus knowledge and facilitates production of chimeric viruses. NPJ VIRUSES 2024; 2:45. [PMID: 40295870 PMCID: PMC11721364 DOI: 10.1038/s44298-024-00052-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 08/12/2024] [Indexed: 04/30/2025]
Abstract
Few insect-specific alphaviruses (ISA) have been discovered, with even fewer culturable to facilitate full characterisation. Here, we report the recovery of an infectious clone of Yada Yada virus (YYV)-a virus previously only detected by metagenomic sequencing of mosquito homogenates. Using the infectious clone, we confirmed the inability of YYV to replicate in vertebrate cells in vitro, with replication limited to only Aedes mosquito-derived cell lines. We further produced and characterised the first monoclonal antibodies (mAbs) to ISAs. Through successful replacement of the structural proteins of YYV with those of other ISAs, Eilat virus, Agua Salud (ASALV), Taï Forest (TALV) and Mwinilunga alphaviruses (MWAV), we established that a replication block for in vitro culture of TALV and MWAV in mosquito cells does not exist at virus entry. Unexpectedly, ASALV structural proteins were recognised by cross-reactive mAbs made to chikungunya (CHIKV) and Ross River viruses (RRV), suggesting a potential antigenic link between ASALV and pathogenic alphaviruses. The YYV genetic backbone was also investigated to generate chimeras displaying the structural proteins of various pathogenic vertebrate-infecting alphaviruses including CHIKV, RRV, Barmah Forest, Sindbis and Mayaro viruses. These chimeras retained the antigenic properties of the parental viruses and did not replicate in vertebrate cells, demonstrating the potential of the YYV platform for vaccine and diagnostic antigen production.
Collapse
Affiliation(s)
- Mikaela G Bell
- School of Chemistry and Molecular Biosciences, The University of Queensland St Lucia, Brisbane, Australia
| | - Rhys H Parry
- School of Chemistry and Molecular Biosciences, The University of Queensland St Lucia, Brisbane, Australia
| | - Tricia S E Lee
- School of Chemistry and Molecular Biosciences, The University of Queensland St Lucia, Brisbane, Australia
| | - Gervais Habarugira
- School of Chemistry and Molecular Biosciences, The University of Queensland St Lucia, Brisbane, Australia
| | - Isabella E McMahon
- School of Chemistry and Molecular Biosciences, The University of Queensland St Lucia, Brisbane, Australia
| | - Madeline A Thompson
- School of Chemistry and Molecular Biosciences, The University of Queensland St Lucia, Brisbane, Australia
| | - Naphak Modhiran
- School of Chemistry and Molecular Biosciences, The University of Queensland St Lucia, Brisbane, Australia
- Australian Infectious Diseases Research Centre, Brisbane, Australia
| | - Daniel Watterson
- School of Chemistry and Molecular Biosciences, The University of Queensland St Lucia, Brisbane, Australia
- Australian Infectious Diseases Research Centre, Brisbane, Australia
| | - Jana Batovska
- Agriculture Victoria Research, AgriBio Centre for AgriBioscience, Bundoora, VIC, Australia
| | - Stacey E Lynch
- Agriculture Victoria Research, AgriBio Centre for AgriBioscience, Bundoora, VIC, Australia
| | - Roy A Hall
- School of Chemistry and Molecular Biosciences, The University of Queensland St Lucia, Brisbane, Australia
- Australian Infectious Diseases Research Centre, Brisbane, Australia
| | - Jessica J Harrison
- School of Chemistry and Molecular Biosciences, The University of Queensland St Lucia, Brisbane, Australia.
| | - Jody Hobson-Peters
- School of Chemistry and Molecular Biosciences, The University of Queensland St Lucia, Brisbane, Australia.
- Australian Infectious Diseases Research Centre, Brisbane, Australia.
| |
Collapse
|
8
|
Cao D, Ma B, Cao Z, Xu X, Zhang X, Xiang Y. The receptor VLDLR binds Eastern Equine Encephalitis virus through multiple distinct modes. Nat Commun 2024; 15:6866. [PMID: 39127734 PMCID: PMC11316819 DOI: 10.1038/s41467-024-51293-x] [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: 12/13/2023] [Accepted: 08/01/2024] [Indexed: 08/12/2024] Open
Abstract
Eastern Equine Encephalitis virus (EEEV) is an alphavirus that can cause severe diseases in infected humans. The very low-density lipoprotein receptor (VLDLR) was recently identified as a receptor of EEEV. Herein, we performed cryo-electron microscopy structural and biochemistry studies on the specific interactions between EEEV and VLDLR. Our results show that VLDLR binds EEEV at three different sites A, B and C through its membrane-distal LDLR class A (LA) repeats. Site A is located in the cleft in between the E1-E2 heterodimers. Site B is located near the connecting β ribbon of E2 and is in proximity to site A, while site C is on the domain B of E2. The binding of VLDLR LAs to EEEV is in complex modes, including the LA1-2 and LA3-5 mediated two major modes. Disruption of the LA1-2 mediated binding significantly affect the cell attachment of EEEV. However, the mutation W132G of VLDLR impairs the binding of LA3, drives the switch of the binding modes, and significantly enhances the attachment of EEEV to the cell. The W132G variant of VLDLR could be identified in human genome and SNP sequences, implying that people with similar mutations in VLDLR may be highly susceptible to EEEV infection.
Collapse
Affiliation(s)
- Duanfang Cao
- National Laboratory of Biomacromolecules, Key Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences (CAS), Beijing, 100101, P.R. China
| | - Bingting Ma
- Beijing Frontier Research Center for Biological Structure, Center for Infectious Disease Research, School of Basic Medical Sciences, Tsinghua University, Beijing, 100084, P.R. China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Taiyuan, 030001, P.R. China
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084, P.R. China
| | - Ziyi Cao
- National Laboratory of Biomacromolecules, Key Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences (CAS), Beijing, 100101, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
| | - Xiaoyu Xu
- Beijing Frontier Research Center for Biological Structure, Center for Infectious Disease Research, School of Basic Medical Sciences, Tsinghua University, Beijing, 100084, P.R. China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Taiyuan, 030001, P.R. China
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084, P.R. China
| | - Xinzheng Zhang
- National Laboratory of Biomacromolecules, Key Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences (CAS), Beijing, 100101, P.R. China.
- University of Chinese Academy of Sciences, Beijing, 100049, P.R. China.
| | - Ye Xiang
- Beijing Frontier Research Center for Biological Structure, Center for Infectious Disease Research, School of Basic Medical Sciences, Tsinghua University, Beijing, 100084, P.R. China.
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Taiyuan, 030001, P.R. China.
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084, P.R. China.
| |
Collapse
|
9
|
Yang P, Li W, Fan X, Pan J, Mann CJ, Varnum H, Clark LE, Clark SA, Coscia A, Basu H, Smith KN, Brusic V, Abraham J. Structural basis for VLDLR recognition by eastern equine encephalitis virus. Nat Commun 2024; 15:6548. [PMID: 39095394 PMCID: PMC11297306 DOI: 10.1038/s41467-024-50887-9] [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: 11/07/2023] [Accepted: 07/23/2024] [Indexed: 08/04/2024] Open
Abstract
Eastern equine encephalitis virus (EEEV) is the most virulent alphavirus that infects humans, and many survivors develop neurological sequelae, including paralysis and intellectual disability. Alphavirus spike proteins comprise trimers of heterodimers of glycoproteins E2 and E1 that mediate binding to cellular receptors and fusion of virus and host cell membranes during entry. We recently identified very-low density lipoprotein receptor (VLDLR) and apolipoprotein E receptor 2 (ApoER2) as cellular receptors for EEEV and a distantly related alphavirus, Semliki Forest virus (SFV). Here, we use single-particle cryo-electron microscopy (cryo-EM) to determine structures of the EEEV and SFV spike glycoproteins bound to the VLDLR ligand-binding domain and found that EEEV and SFV interact with the same cellular receptor through divergent binding modes. Our studies suggest that the ability of LDLR-related proteins to interact with viral spike proteins through very small footprints with flexible binding modes results in a low evolutionary barrier to the acquisition of LDLR-related proteins as cellular receptors for diverse sets of viruses.
Collapse
Affiliation(s)
- Pan Yang
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Wanyu Li
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Xiaoyi Fan
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Junhua Pan
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Biomedical Research Institute and School of Life and Health Sciences, Hubei University of Technology, Wuhan, Hubei, China
| | - Colin J Mann
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Haley Varnum
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Lars E Clark
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Sarah A Clark
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Adrian Coscia
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Himanish Basu
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Katherine Nabel Smith
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Vesna Brusic
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Jonathan Abraham
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
- Department of Medicine, Division of Infectious Diseases, Brigham & Women's Hospital, Boston, MA, USA.
- Center for Integrated Solutions in Infectious Diseases, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
10
|
Zhai X, Li X, Veit M, Wang N, Wang Y, Merits A, Jiang Z, Qin Y, Zhang X, Qi K, Jiao H, He WT, Chen Y, Mao Y, Su S. LDLR is used as a cell entry receptor by multiple alphaviruses. Nat Commun 2024; 15:622. [PMID: 38245515 PMCID: PMC10799924 DOI: 10.1038/s41467-024-44872-5] [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: 06/05/2023] [Accepted: 01/04/2024] [Indexed: 01/22/2024] Open
Abstract
Alphaviruses are arboviruses transmitted by mosquitoes and are pathogenic to humans and livestock, causing a substantial public health burden. So far, several receptors have been identified for alphavirus entry; however, they cannot explain the broad host range and tissue tropism of certain alphaviruses, such as Getah virus (GETV), indicating the existence of additional receptors. Here we identify the evolutionarily conserved low-density lipoprotein receptor (LDLR) as a new cell entry factor for GETV, Semliki Forest virus (SFV), Ross River virus (RRV) and Bebaru virus (BEBV). Ectopic expression of LDLR facilitates cellular binding and internalization of GETV, which is mediated by the interaction between the E2-E1 spike of GETV and the ligand-binding domain (LBD) of LDLR. Antibodies against LBD block GETV infection in cultured cells. In addition, the GST-LBD fusion protein inhibits GETV infection both in vitro and in vivo. Notably, we identify the key amino acids in LDLR-LBD that played a crucial role in viral entry; specific mutations in the CR4 and CR5 domain of LDLR-LBD reduce viral entry to cells by more than 20-fold. These findings suggest that targeting the LDLR-LBD could be a potential strategy for the development of antivirals against multiple alphaviruses.
Collapse
Affiliation(s)
- Xiaofeng Zhai
- Academy for Advanced Interdisciplinary Studies, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xiaoling Li
- Academy for Advanced Interdisciplinary Studies, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Michael Veit
- Institute for Virology, Center for Infection Medicine, Veterinary Faculty, Free University Berlin, Berlin, Germany
| | - Ningning Wang
- Academy for Advanced Interdisciplinary Studies, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yu Wang
- Academy for Advanced Interdisciplinary Studies, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Andres Merits
- Institute of Bioengineering, University of Tartu, Nooruse Street 1, 50411, Tartu, Estonia
| | - Zhiwen Jiang
- Academy for Advanced Interdisciplinary Studies, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yan Qin
- Academy for Advanced Interdisciplinary Studies, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xiaoguang Zhang
- Academy for Advanced Interdisciplinary Studies, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Kaili Qi
- Academy for Advanced Interdisciplinary Studies, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Houqi Jiao
- Academy for Advanced Interdisciplinary Studies, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Wan-Ting He
- Academy for Advanced Interdisciplinary Studies, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Ye Chen
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Yang Mao
- School of Pharmaceutical Sciences and National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Sun Yat-sen University, Guangzhou, China.
| | - Shuo Su
- Academy for Advanced Interdisciplinary Studies, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.
| |
Collapse
|
11
|
Adams LJ, Raju S, Ma H, Gilliland T, Reed DS, Klimstra WB, Fremont DH, Diamond MS. Structural and functional basis of VLDLR usage by Eastern equine encephalitis virus. Cell 2024; 187:360-374.e19. [PMID: 38176410 PMCID: PMC10843625 DOI: 10.1016/j.cell.2023.11.031] [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: 05/01/2023] [Revised: 10/06/2023] [Accepted: 11/28/2023] [Indexed: 01/06/2024]
Abstract
The very-low-density lipoprotein receptor (VLDLR) comprises eight LDLR type A (LA) domains and supports entry of distantly related alphaviruses, including Eastern equine encephalitis virus (EEEV) and Semliki Forest virus (SFV). Here, by resolving multiple cryo-electron microscopy structures of EEEV-VLDLR complexes and performing mutagenesis and functional studies, we show that EEEV uses multiple sites (E1/E2 cleft and E2 A domain) to engage more than one LA domain simultaneously. However, no single LA domain is necessary or sufficient to support efficient EEEV infection. Whereas all EEEV strains show conservation of two VLDLR-binding sites, the EEEV PE-6 strain and a few other EEE complex members feature a single amino acid substitution that enables binding of LA domains to an additional site on the E2 B domain. These structural and functional analyses informed the design of a minimal VLDLR decoy receptor that neutralizes EEEV infection and protects mice from lethal challenge.
Collapse
Affiliation(s)
- Lucas J Adams
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Saravanan Raju
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hongming Ma
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Theron Gilliland
- The Center for Vaccine Research and Department of Immunology, The University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Douglas S Reed
- The Center for Vaccine Research and Department of Immunology, The University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - William B Klimstra
- The Center for Vaccine Research and Department of Immunology, The University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Daved H Fremont
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Michael S Diamond
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO 63110, USA; Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
12
|
Newby ML, Allen JD, Crispin M. Influence of glycosylation on the immunogenicity and antigenicity of viral immunogens. Biotechnol Adv 2024; 70:108283. [PMID: 37972669 PMCID: PMC10867814 DOI: 10.1016/j.biotechadv.2023.108283] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 10/04/2023] [Accepted: 11/09/2023] [Indexed: 11/19/2023]
Abstract
A key aspect of successful viral vaccine design is the elicitation of neutralizing antibodies targeting viral attachment and fusion glycoproteins that embellish viral particles. This observation has catalyzed the development of numerous viral glycoprotein mimetics as vaccines. Glycans can dominate the surface of viral glycoproteins and as such, the viral glycome can influence the antigenicity and immunogenicity of a candidate vaccine. In one extreme, glycans can form an integral part of epitopes targeted by neutralizing antibodies and are therefore considered to be an important feature of key immunogens within an immunization regimen. In the other extreme, the existence of peptide and bacterially expressed protein vaccines shows that viral glycosylation can be dispensable in some cases. However, native-like glycosylation can indicate native-like protein folding and the presence of conformational epitopes. Furthermore, going beyond native glycan mimicry, in either occupancy of glycosylation sites or the glycan processing state, may offer opportunities for enhancing the immunogenicity and associated protection elicited by an immunogen. Here, we review key determinants of viral glycosylation and how recombinant immunogens can recapitulate these signatures across a range of enveloped viruses, including HIV-1, Ebola virus, SARS-CoV-2, Influenza and Lassa virus. The emerging understanding of immunogen glycosylation and its control will help guide the development of future vaccines in both recombinant protein- and nucleic acid-based vaccine technologies.
Collapse
Affiliation(s)
- Maddy L Newby
- School of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Joel D Allen
- School of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, UK.
| | - Max Crispin
- School of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, UK.
| |
Collapse
|
13
|
Adams LJ, Raju S, Ma H, Gilliland T, Reed DS, Klimstra WB, Fremont DH, Diamond MS. Structural and functional basis of VLDLR receptor usage by Eastern equine encephalitis virus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.15.567188. [PMID: 38014196 PMCID: PMC10680733 DOI: 10.1101/2023.11.15.567188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
The very low-density lipoprotein receptor (VLDLR) is comprised of eight LDLR type A (LA) domains and supports entry of distantly related Eastern equine encephalitis (EEEV) and Semliki Forest (SFV) alphaviruses. Here, by resolving multiple cryo-electron microscopy structures of EEEV-VLDLR complexes and performing mutagenesis and functional studies, we show that EEEV uses multiple sites (E1/E2 cleft and E2 A domain) to engage different LA domains simultaneously. However, no single LA domain is necessary or sufficient to support efficient EEEV infection, highlighting complexity in domain usage. Whereas all EEEV strains show conservation of two VLDLR binding sites, the EEEV PE-6 strain and other EEE complex members feature a single amino acid substitution that mediates binding of LA domains to an additional site on the E2 B domain. These structural and functional analyses informed the design of a minimal VLDLR decoy receptor that neutralizes EEEV infection and protects mice from lethal challenge.
Collapse
Affiliation(s)
- Lucas J. Adams
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Saravanan Raju
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hongming Ma
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Theron Gilliland
- The Center for Vaccine Research and Department of Immunology, The University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Douglas S. Reed
- The Center for Vaccine Research and Department of Immunology, The University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - William B. Klimstra
- The Center for Vaccine Research and Department of Immunology, The University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Daved H. Fremont
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Michael S. Diamond
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, Saint Louis, MO 63110, USA
| |
Collapse
|
14
|
Song D, Jia X, Gao Y, Xiao T, Dan J, Shen R, Cai J, Liang J, Zhu W, Hu J, Yan G, Zhang Q, Lin Y. STT3A-mediated viral N-glycosylation underlies the tumor selectivity of oncolytic virus M1. Oncogene 2023; 42:3575-3588. [PMID: 37864032 DOI: 10.1038/s41388-023-02872-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/04/2023] [Accepted: 10/12/2023] [Indexed: 10/22/2023]
Abstract
Oncolytic viruses are emerging as promising anticancer agents. Although the essential biological function of N-glycosylation on viruses are widely accepted, roles of N-glycan and glycan-processing enzyme in oncolytic viral therapy are remain elusive. Here, via cryo-EM analysis, we identified three distinct N-glycans on the envelope of oncolytic virus M1 (OVM) as being necessary for efficient receptor binding. E1-N141-glycan has immediate impact on the binding of MXRA8 receptor, E2-N200-glycan mediates the maturation of E2 from its precursor PE2 which is unable to bind with MXRA8, and E2-N262-glycan slightly promotes receptor binding. The necessity of OVM N-glycans in receptor binding make them indispensable for oncolysis in vitro and in vivo. Further investigations identified STT3A, a key catalytic subunit of oligosaccharyltransferase (OST), as the determinant of OVM N-glycosylation, and STT3A expression in tumor cells is positively correlated with OVM-induced oncolysis. Increased STT3A expression was observed in various solid tumors, pointing to a broad-spectrum anticancer potential of OVM. Collectively, our research supports the importance of STT3A-mediated N-glycosylation in receptor binding and oncolysis of OVM, thus providing a novel predictive biomarker for OVM.
Collapse
Affiliation(s)
- Deli Song
- State Key Lab for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xudong Jia
- State Key Lab for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yuanzhu Gao
- State Key Lab for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Tong Xiao
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jia Dan
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Runling Shen
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jing Cai
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jiankai Liang
- Department of Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Wenbo Zhu
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jun Hu
- Department of Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Guangmei Yan
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Qinfen Zhang
- State Key Lab for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China.
| | - Yuan Lin
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
- Advanced Medical Technology Center, The First Affiliated Hospital-Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
15
|
Kim AS, Diamond MS. A molecular understanding of alphavirus entry and antibody protection. Nat Rev Microbiol 2023; 21:396-407. [PMID: 36474012 PMCID: PMC9734810 DOI: 10.1038/s41579-022-00825-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/01/2022] [Indexed: 12/12/2022]
Abstract
Alphaviruses are arthropod-transmitted RNA viruses that cause epidemics of human infection and disease on a global scale. These viruses are classified as either arthritogenic or encephalitic based on their genetic relatedness and the clinical syndromes they cause. Although there are currently no approved therapeutics or vaccines against alphaviruses, passive transfer of monoclonal antibodies confers protection in animal models. This Review highlights recent advances in our understanding of the host factors required for alphavirus entry, the mechanisms of action by which protective antibodies inhibit different steps in the alphavirus infection cycle and candidate alphavirus vaccines currently under clinical evaluation that focus on humoral immunity. A comprehensive understanding of alphavirus entry and antibody-mediated protection may inform the development of new classes of countermeasures for these emerging viruses.
Collapse
Affiliation(s)
- Arthur S Kim
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA.
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA.
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO, USA.
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
16
|
Li M, Pedersen LC, Xu D. Targeting heparan sulfate-protein interactions with oligosaccharides and monoclonal antibodies. Front Mol Biosci 2023; 10:1194293. [PMID: 37275960 PMCID: PMC10235622 DOI: 10.3389/fmolb.2023.1194293] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 05/10/2023] [Indexed: 06/07/2023] Open
Abstract
Heparan sulfate-binding proteins (HSBPs) are structurally diverse extracellular and membrane attached proteins that interact with HS under normal physiological conditions. Interactions with HS offer an additional level of control over the localization and function of HSBPs, which enables them to behave in a more refined manner. Because all cell signaling events start at the cell membrane, and cell-cell communication relies on translocation of soluble factors across the extracellular matrix, HS occupies an apical position in cellular signal transduction by interacting with hundreds of growth factors, cytokines, chemokines, enzymes, enzyme inhibitors, receptors and adhesion molecules. These extracellular and membrane proteins can play important roles in physiological and pathological conditions. For most HS-binding proteins, the interaction with HS represents an essential element in regulating their normal physiological functions. Such dependence on HS suggests that manipulating HS-protein interactions could be explored as a therapeutic strategy to selectively antagonize/activate HS-binding proteins. In this review, we will discuss current understanding of the diverse nature of HS-HSBP interactions, and the latest advancements in targeting the HS-binding site of HSBPs using structurally-defined HS oligosaccharides and monoclonal antibodies.
Collapse
Affiliation(s)
- Miaomiao Li
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, the State University of New York, Buffalo, NY, United States
| | - Lars C. Pedersen
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - Ding Xu
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, the State University of New York, Buffalo, NY, United States
| |
Collapse
|
17
|
Williamson LE, Bandyopadhyay A, Bailey K, Sirohi D, Klose T, Julander JG, Kuhn RJ, Crowe JE. Structural constraints link differences in neutralization potency of human anti-Eastern equine encephalitis virus monoclonal antibodies. Proc Natl Acad Sci U S A 2023; 120:e2213690120. [PMID: 36961925 PMCID: PMC10068833 DOI: 10.1073/pnas.2213690120] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 02/10/2023] [Indexed: 03/26/2023] Open
Abstract
Selection and development of monoclonal antibody (mAb) therapeutics against pathogenic viruses depends on certain functional characteristics. Neutralization potency, or the half-maximal inhibitory concentration (IC50) values, is an important characteristic of candidate therapeutic antibodies. Structural insights into the bases of neutralization potency differences between antiviral neutralizing mAbs are lacking. In this report, we present cryo-electron microscopy (EM) reconstructions of three anti-Eastern equine encephalitis virus (EEEV) neutralizing human mAbs targeting overlapping epitopes on the E2 protein, with greater than 20-fold differences in their respective IC50 values. From our structural and biophysical analyses, we identify several constraints that contribute to the observed differences in the neutralization potencies. Cryo-EM reconstructions of EEEV in complex with these Fab fragments reveal structural constraints that dictate intravirion or intervirion cross-linking of glycoprotein spikes by their IgG counterparts as a mechanism of neutralization. Additionally, we describe critical features for the recognition of EEEV by these mAbs including the epitope-paratope interaction surface, occupancy, and kinetic differences in on-rate for binding to the E2 protein. Each constraint contributes to the extent of EEEV inhibition for blockade of virus entry, fusion, and/or egress. These findings provide structural and biophysical insights into the differences in mechanism and neutralization potencies of these antibodies, which help inform rational design principles for candidate vaccines and therapeutic antibodies for all icosahedral viruses.
Collapse
Affiliation(s)
- Lauren E. Williamson
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN37232
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN37232
| | - Abhishek Bandyopadhyay
- Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN47907
| | - Kevin Bailey
- Institute for Antiviral Research, Utah State University, Logan, UT84335
| | - Devika Sirohi
- Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN47907
| | - Thomas Klose
- Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN47907
| | | | - Richard J. Kuhn
- Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN47907
| | - James E. Crowe
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN37232
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN37232
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN37232
| |
Collapse
|
18
|
Alphavirus Particles Can Assemble with an Alternate Triangulation Number. Viruses 2022; 14:v14122650. [PMID: 36560655 PMCID: PMC9780915 DOI: 10.3390/v14122650] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/24/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022] Open
Abstract
Alphaviruses are spherical, enveloped RNA viruses primarily transmitted by mosquitoes, and cause significant arthritogenic and neurotropic disease in humans and livestock. Previous reports have shown that-in contrast to prototypical icosahedral viruses-alphaviruses incorporate frequent defects, and these may serve important functions in the viral life cycle. We confirm the genus-wide pleomorphism in live viral particles and extend our understanding of alphavirus assembly through the discovery of an alternate architecture of Eastern equine encephalitis virus (EEEV) particles. The alternate T = 3 icosahedral architecture differs in triangulation number from the classic T = 4 icosahedral organization that typifies alphaviruses, but the alternate architecture maintains the quasi-equivalence relationship of asymmetric units. The fusion spike glycoproteins are more loosely apposed in the T = 3 form with corresponding changes in the underlying capsid protein lattice. This alternate architecture could potentially be exploited in engineering alphavirus-based particles for delivery of alphaviral or other RNA.
Collapse
|
19
|
Elmasri Z, Negi V, Kuhn RJ, Jose J. Requirement of a functional ion channel for Sindbis virus glycoprotein transport, CPV-II formation, and efficient virus budding. PLoS Pathog 2022; 18:e1010892. [PMID: 36191050 PMCID: PMC9560593 DOI: 10.1371/journal.ppat.1010892] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 10/13/2022] [Accepted: 09/21/2022] [Indexed: 12/14/2022] Open
Abstract
Many viruses encode ion channel proteins that oligomerize to form hydrophilic pores in membranes of virus-infected cells and the viral membrane in some enveloped viruses. Alphavirus 6K, human immunodeficiency virus type 1 Vpu (HIV-Vpu), influenza A virus M2 (IAV-M2), and hepatitis C virus P7 (HCV-P7) are transmembrane ion channel proteins that play essential roles in virus assembly, budding, and entry. While the oligomeric structures and mechanisms of ion channel activity are well-established for M2 and P7, these remain unknown for 6K. Here we investigated the functional role of the ion channel activity of 6K in alphavirus assembly by utilizing a series of Sindbis virus (SINV) ion channel chimeras expressing the ion channel helix from Vpu or M2 or substituting the entire 6K protein with full-length P7, in cis. We demonstrate that the Vpu helix efficiently complements 6K, whereas M2 and P7 are less efficient. Our results indicate that while SINV is primarily insensitive to the M2 ion channel inhibitor amantadine, the Vpu inhibitor 5-N, N-Hexamethylene amiloride (HMA), significantly reduces SINV release, suggesting that the ion channel activity of 6K similar to Vpu, promotes virus budding. Using live-cell imaging of SINV with a miniSOG-tagged 6K and mCherry-tagged E2, we further demonstrate that 6K and E2 colocalize with the Golgi apparatus in the secretory pathway. To contextualize the localization of 6K in the Golgi, we analyzed cells infected with SINV and SINV-ion channel chimeras using transmission electron microscopy. Our results provide evidence for the first time for the functional role of 6K in type II cytopathic vacuoles (CPV-II) formation. We demonstrate that in the absence of 6K, CPV-II, which originates from the Golgi apparatus, is not detected in infected cells, with a concomitant reduction in the glycoprotein transport to the plasma membrane. Substituting a functional ion channel, M2 or Vpu localizing to Golgi, restores CPV-II production, whereas P7, retained in the ER, is inadequate to induce CPV-II formation. Altogether our results indicate that ion channel activity of 6K is required for the formation of CPV-II from the Golgi apparatus, promoting glycoprotein spike transport to the plasma membrane and efficient virus budding.
Collapse
Affiliation(s)
- Zeinab Elmasri
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Vashi Negi
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana, United States of America
| | - Richard J. Kuhn
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana, United States of America
- Markey Center for Structural Biology and Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, Indiana, United States of America
| | - Joyce Jose
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| |
Collapse
|
20
|
Chen CL, Klose T, Sun C, Kim AS, Buda G, Rossmann MG, Diamond MS, Klimstra WB, Kuhn RJ. Cryo-EM structures of alphavirus conformational intermediates in low pH-triggered prefusion states. Proc Natl Acad Sci U S A 2022; 119:e2114119119. [PMID: 35867819 PMCID: PMC9335222 DOI: 10.1073/pnas.2114119119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 06/03/2022] [Indexed: 01/24/2023] Open
Abstract
Alphaviruses can cause severe human arthritis and encephalitis. During virus infection, structural changes of viral glycoproteins in the acidified endosome trigger virus-host membrane fusion for delivery of the capsid core and RNA genome into the cytosol to initiate virus translation and replication. However, mechanisms by which E1 and E2 glycoproteins rearrange in this process remain unknown. Here, we investigate prefusion cryoelectron microscopy (cryo-EM) structures of eastern equine encephalitis virus (EEEV) under acidic conditions. With models fitted into the low-pH cryo-EM maps, we suggest that E2 dissociates from E1, accompanied by a rotation (∼60°) of the E2-B domain (E2-B) to expose E1 fusion loops. Cryo-EM reconstructions of EEEV bound to a protective antibody at acidic and neutral pH suggest that stabilization of E2-B prevents dissociation of E2 from E1. These findings reveal conformational changes of the glycoprotein spikes in the acidified host endosome. Stabilization of E2-B may provide a strategy for antiviral agent development.
Collapse
Affiliation(s)
- Chun-Liang Chen
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
| | - Thomas Klose
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
| | - Chengqun Sun
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 15261
| | - Arthur S. Kim
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Geeta Buda
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
| | - Michael G. Rossmann
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
| | - Michael S. Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110
| | - William B. Klimstra
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 15261
| | - Richard J. Kuhn
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
| |
Collapse
|
21
|
Abstract
Alphaviruses are enveloped viruses transmitted by arthropod vectors to vertebrate hosts. The surface of the virion contains 80 glycoprotein spikes embedded in the membrane, and these spikes mediate attachment to the host cell and initiate viral fusion. Each spike consists of a trimer of E2-E1 heterodimers. These heterodimers interact at the following two interfaces: (i) the intradimer interactions between E2 and E1 of the same heterodimer and (ii) the interdimer interactions between E2 of one heterodimer and E1 of the adjacent heterodimer (E1'). We hypothesized that the interdimer interactions are essential for trimerization of the E2-E1 heterodimers into a functional spike. In this work, we made a mutant virus (chikungunya piggyback [CPB]) where we replaced six interdimeric residues in the E2 protein of Sindbis virus (wild-type [WT] SINV) with those from the E2 protein from chikungunya virus and studied its effect in both mammalian and mosquito cell lines. CPB produced fewer infectious particles in mammalian cells than in mosquito cells, relative to WT SINV. When CPB virus was purified from mammalian cells, particles showed reduced amounts of glycoproteins relative to the capsid protein and contained defects in particle morphology compared with virus derived from mosquito cells. Using cryo-electron microscopy (cryo-EM), we determined that the spikes of CPB had a different conformation than WT SINV. Last, we identified two revertants, E2-H333N and E1-S247L, that restored particle growth and assembly to different degrees. We conclude the interdimer interface is critical for spike trimerization and is a novel target for potential antiviral drug design. IMPORTANCE Alphaviruses, which can cause disease when spread to humans by mosquitoes, have been classified as emerging pathogens, with infections occurring worldwide. The spikes on the surface of the alphavirus particle are absolutely required for the virus to enter a new host cell and initiate an infection. Using a structure-guided approach, we made a mutant virus that alters spike assembly in mammalian cells but not mosquito cells. This finding is important because it identifies a region in the spike that could be a target for antiviral drug design.
Collapse
|
22
|
Structure of infective Getah virus at 2.8 Å resolution determined by cryo-electron microscopy. Cell Discov 2022; 8:12. [PMID: 35149682 PMCID: PMC8832435 DOI: 10.1038/s41421-022-00374-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 01/03/2022] [Indexed: 11/30/2022] Open
Abstract
Getah virus (GETV), a member of the genus alphavirus, is a mosquito-borne pathogen that can cause pyrexia and reproductive losses in animals. Although antibodies to GETV have been found in over 10% of healthy people, there are no reports of clinical symptoms associated with GETV. The biological and pathological properties of GETV are largely unknown and antiviral or vaccine treatments against GETV are still unavailable due to a lack of knowledge of the structure of the GETV virion. Here, we present the structure of infective GETV at a resolution of 2.8 Å with the atomic models of the capsid protein and the envelope glycoproteins E1 and E2. We have identified numerous glycosylation and S-acylation sites in E1 and E2. The surface-exposed glycans indicate a possible impact on viral immune evasion and host cell invasion. The S-acylation sites might be involved in stabilizing the transmembrane assembly of E1 and E2. In addition, a cholesterol and a phospholipid molecule are observed in a transmembrane hydrophobic pocket, together with two more cholesterols surrounding the pocket. The cholesterol and phospholipid stabilize the hydrophobic pocket in the viral envelope membrane. The structural information will assist structure-based antiviral and vaccine screening, design, and optimization.
Collapse
|
23
|
Structural Insights into Alphavirus Assembly Revealed by the Cryo-EM Structure of Getah Virus. Viruses 2022; 14:v14020327. [PMID: 35215918 PMCID: PMC8876998 DOI: 10.3390/v14020327] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 01/28/2022] [Accepted: 02/03/2022] [Indexed: 11/16/2022] Open
Abstract
Getah virus (GETV) is a member of the alphavirus genus, and it infects a variety of animal species, including horses, pigs, cattle, and foxes. Human infection with this virus has also been reported. The structure of GETV has not yet been determined. In this study, we report the cryo-EM structure of GETV at a resolution of 3.5 Å. This structure reveals conformational polymorphism of the envelope glycoproteins E1 and E2 at icosahedral 3-fold and quasi-3-fold axes, which is believed to be a necessary organization in forming a curvature surface of virions. In our density map, three extra densities are identified, one of which is believed a “pocket factor”; the other two are located by domain D of E2, and they may maintain the stability of E1/E2 heterodimers. We also identify three N-glycosylations at E1 N141, E2 N200, and E2 N262, which might be associated with receptor binding and membrane fusion. The resolving of the structure of GETV provides new insights into the structure and assembly of alphaviruses and lays a basis for studying the differences of biology and pathogenicity between arthritogenic and encephalitic alphaviruses.
Collapse
|
24
|
Phelps AL, O’Brien LM, Ulaeto DO, Holtsberg FW, Liao GC, Douglas R, Aman MJ, Glass PJ, Moyer CL, Ennis J, Zeitlin L, Nagata LP, Hu WG. Cross-Strain Neutralizing and Protective Monoclonal Antibodies against EEEV or WEEV. Viruses 2021; 13:2231. [PMID: 34835037 PMCID: PMC8621548 DOI: 10.3390/v13112231] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 10/28/2021] [Accepted: 11/01/2021] [Indexed: 11/17/2022] Open
Abstract
The three encephalitic alphaviruses, namely, the Venezuelan, eastern, and western equine encephalitis viruses (VEEV, EEEV, and WEEV), are classified by the Centers for Disease Control and Prevention (CDC) as biothreat agents. Currently, no licensed medical countermeasures (MCMs) against these viruses are available for humans. Neutralizing antibodies (NAbs) are fast-acting and highly effective MCMs for use in both pre- and post-exposure settings against biothreat agents. While significant work has been done to identify anti-VEEV NAbs, less has been done to identify NAbs against EEEV and WEEV. In order to develop anti-EEEV or -WEEV NAbs, mice were immunized using complementary strategies with a variety of different EEEV or WEEV immunogens to maximize the generation of NAbs to each of these viruses. Of the hybridomas generated, three anti-EEEV and seven anti-WEEV monoclonal antibodies were identified with in vitro neutralization activity. The most potent neutralizers (two anti-EEEV NAbs and three anti-WEEV NAbs) were further evaluated for neutralization activity against additional strains of EEEV, a single strain of Madariaga virus (formerly South American EEEV), or WEEV. Of these, G1-2-H4 and G1-4-C3 neutralized all three EEEV strains and the Madariaga virus strain, whereas G8-2-H9 and 12 WA neutralized six out of eight WEEV strains. To determine the protective efficacy of these NAbs, the five most potent neutralizers were evaluated in respective mouse aerosol challenge models. All five NAbs demonstrated various levels of protection when administered at doses of 2.5 mg/kg or 10 mg/kg 24 h before the respective virus exposure via the aerosol route. Of these, anti-EEEV NAb G1-4-C3 and anti-WEEV NAb 8C2 provided 100% protection at both doses and all surviving mice were free of clinical signs throughout the study. Additionally, no virus was detected in the brain 14 days post virus exposure. Taken together, efficacious NAbs were developed that demonstrate the potential for the development of cross-strain antibody-based MCMs against EEEV and WEEV infections.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/immunology
- Antibodies, Neutralizing/administration & dosage
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/administration & dosage
- Antibodies, Viral/immunology
- Cross Protection
- Disease Models, Animal
- Encephalitis Virus, Eastern Equine/immunology
- Encephalitis Virus, Western Equine/immunology
- Encephalomyelitis, Equine/prevention & control
- Immunization
- Mice
- Neutralization Tests
Collapse
Affiliation(s)
- Amanda L. Phelps
- Defence Science and Technology Laboratory, Porton Down, Salisbury SP4 0JQ, UK; (A.L.P.); (L.M.O.); (D.O.U.)
| | - Lyn M. O’Brien
- Defence Science and Technology Laboratory, Porton Down, Salisbury SP4 0JQ, UK; (A.L.P.); (L.M.O.); (D.O.U.)
| | - David O. Ulaeto
- Defence Science and Technology Laboratory, Porton Down, Salisbury SP4 0JQ, UK; (A.L.P.); (L.M.O.); (D.O.U.)
| | - Frederick W. Holtsberg
- Integrated BioTherapeutics, Inc., Rockville, MD 20850, USA; (F.W.H.); (G.C.L.); (R.D.); (M.J.A.)
| | - Grant C. Liao
- Integrated BioTherapeutics, Inc., Rockville, MD 20850, USA; (F.W.H.); (G.C.L.); (R.D.); (M.J.A.)
| | - Robin Douglas
- Integrated BioTherapeutics, Inc., Rockville, MD 20850, USA; (F.W.H.); (G.C.L.); (R.D.); (M.J.A.)
| | - M. Javad Aman
- Integrated BioTherapeutics, Inc., Rockville, MD 20850, USA; (F.W.H.); (G.C.L.); (R.D.); (M.J.A.)
| | - Pamela J. Glass
- US Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA;
| | - Crystal L. Moyer
- Mapp Biopharmaceutical, Inc., San Diego, CA 92121, USA; (C.L.M.); (J.E.); (L.Z.)
| | - Jane Ennis
- Mapp Biopharmaceutical, Inc., San Diego, CA 92121, USA; (C.L.M.); (J.E.); (L.Z.)
| | - Larry Zeitlin
- Mapp Biopharmaceutical, Inc., San Diego, CA 92121, USA; (C.L.M.); (J.E.); (L.Z.)
| | - Les P. Nagata
- Defence Research and Development Canada, Suffield Research Centre, Medicine Hat, AB T1A 8K6, Canada;
| | - Wei-Gang Hu
- Defence Research and Development Canada, Suffield Research Centre, Medicine Hat, AB T1A 8K6, Canada;
| |
Collapse
|
25
|
Capsid-E2 Interactions Rescue Core Assembly in Viruses That Cannot Form Cytoplasmic Nucleocapsid Cores. J Virol 2021; 95:e0106221. [PMID: 34495691 DOI: 10.1128/jvi.01062-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Alphavirus capsid proteins (CPs) have two domains: the N-terminal domain (NTD), which interacts with the viral RNA, and the C-terminal domain (CTD), which forms CP-CP interactions and interacts with the cytoplasmic domain of the E2 spike protein (cdE2). In this study, we examine how mutations in the CP NTD affect CP CTD interactions with cdE2. We changed the length and/or charge of the NTD of Ross River virus CP and found that changing the charge of the NTD has a greater impact on core and virion assembly than changing the length of the NTD. The NTD CP insertion mutants are unable to form cytoplasmic cores during infection, but they do form cores or core-like structures in virions. Our results are consistent with cdE2 having a role in core maturation during virion assembly and rescuing core formation when cytoplasmic cores are not assembled. We go on to find that the isolated cores from some mutant virions are now assembly competent in that they can be disassembled and reassembled back into cores. These results show how the two domains of CP may have distinct yet coordinated roles. IMPORTANCE Structural viral proteins have multiple roles during entry and assembly. The capsid protein (CP) of alphaviruses has one domain that interacts with the viral genome and another domain that interacts with the E2 spike protein. In this work, we determined that the length and/or charge of the CP affects cytoplasmic core formation. However, defects in cytoplasmic core formation can be overcome by E2-CP interactions, thus assembling a core or core-like complex in the virion. In the absence of both cytoplasmic cores and CP-E2 interactions, CP is not even packaged in the released virions, but some infectious particles are still released, presumably as RNA packaged in a glycoprotein-containing membrane shell. This suggests that the virus has multiple mechanisms in place to ensure the viral genome is surrounded by a capsid core during its life cycle.
Collapse
|
26
|
Structure of Venezuelan equine encephalitis virus with its receptor LDLRAD3. Nature 2021; 598:677-681. [PMID: 34646021 DOI: 10.1038/s41586-021-03909-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 08/13/2021] [Indexed: 11/08/2022]
Abstract
Venezuelan equine encephalitis virus (VEEV) is an enveloped RNA virus that causes encephalitis and potentially mortality in infected humans and equines1. At present, no vaccines or drugs are available that prevent or cure diseases caused by VEEV. Low-density lipoprotein receptor class A domain-containing 3 (LDLRAD3) was recently identified as a receptor for the entry of VEEV into host cells2. Here we present the cryo-electron microscopy structure of the LDLRAD3 extracellular domain 1 (LDLRAD3-D1) in complex with VEEV virus-like particles at a resolution of 3.0 Å. LDLRAD3-D1 has a cork-like structure and is inserted into clefts formed between adjacent VEEV E2-E1 heterodimers in the viral-surface trimer spikes through hydrophobic and polar contacts. Mutagenesis studies of LDLRAD3-D1 identified residues that are involved in the key interactions with VEEV. Of note, some of the LDLRAD3-D1 mutants showed a significantly increased binding affinity for VEEV, suggesting that LDLRAD3-D1 may serve as a potential scaffold for the development of inhibitors of VEEV entry. Our structures provide insights into alphavirus assembly and the binding of receptors to alphaviruses, which may guide the development of therapeutic countermeasures against alphaviruses.
Collapse
|
27
|
Williamson LE, Reeder KM, Bailey K, Tran MH, Roy V, Fouch ME, Kose N, Trivette A, Nargi RS, Winkler ES, Kim AS, Gainza C, Rodriguez J, Armstrong E, Sutton RE, Reidy J, Carnahan RH, McDonald WH, Schoeder CT, Klimstra WB, Davidson E, Doranz BJ, Alter G, Meiler J, Schey KL, Julander JG, Diamond MS, Crowe JE. Therapeutic alphavirus cross-reactive E1 human antibodies inhibit viral egress. Cell 2021; 184:4430-4446.e22. [PMID: 34416147 PMCID: PMC8418820 DOI: 10.1016/j.cell.2021.07.033] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 04/11/2021] [Accepted: 07/26/2021] [Indexed: 12/11/2022]
Abstract
Alphaviruses cause severe arthritogenic or encephalitic disease. The E1 structural glycoprotein is highly conserved in these viruses and mediates viral fusion with host cells. However, the role of antibody responses to the E1 protein in immunity is poorly understood. We isolated E1-specific human monoclonal antibodies (mAbs) with diverse patterns of recognition for alphaviruses (ranging from Eastern equine encephalitis virus [EEEV]-specific to alphavirus cross-reactive) from survivors of natural EEEV infection. Antibody binding patterns and epitope mapping experiments identified differences in E1 reactivity based on exposure of epitopes on the glycoprotein through pH-dependent mechanisms or presentation on the cell surface prior to virus egress. Therapeutic efficacy in vivo of these mAbs corresponded with potency of virus egress inhibition in vitro and did not require Fc-mediated effector functions for treatment against subcutaneous EEEV challenge. These studies reveal the molecular basis for broad and protective antibody responses to alphavirus E1 proteins.
Collapse
MESH Headings
- Alphavirus/immunology
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/isolation & purification
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/immunology
- Antigens, Viral/immunology
- Cell Line
- Chikungunya virus/immunology
- Cross Reactions/immunology
- Encephalitis Virus, Eastern Equine/immunology
- Encephalomyelitis, Equine/immunology
- Encephalomyelitis, Equine/virology
- Epitope Mapping
- Female
- Horses
- Humans
- Hydrogen-Ion Concentration
- Joints/pathology
- Male
- Mice, Inbred C57BL
- Models, Biological
- Protein Binding
- RNA, Viral/metabolism
- Receptors, Fc/metabolism
- Temperature
- Viral Proteins/immunology
- Virion/metabolism
- Virus Internalization
- Virus Release/physiology
- Mice
Collapse
Affiliation(s)
- Lauren E Williamson
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN 37232, USA; The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Kristen M Reeder
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Kevin Bailey
- Institute for Antiviral Research, Utah State University, Logan, UT 84335, USA
| | - Minh H Tran
- Chemical and Physical Biology Program, Vanderbilt University, Nashville, TN, USA; Center of Structural Biology, Vanderbilt University, Nashville, TN, USA; Department of Biochemistry and Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN, USA
| | - Vicky Roy
- Ragon Institute of MGH, MIT, and Harvard University, Cambridge, MA 02139, USA
| | | | - Nurgun Kose
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Andrew Trivette
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Rachel S Nargi
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Emma S Winkler
- Department of Medicine, Washington University, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University, St. Louis, MO 63110, USA
| | - Arthur S Kim
- Department of Medicine, Washington University, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University, St. Louis, MO 63110, USA
| | - Christopher Gainza
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jessica Rodriguez
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Erica Armstrong
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Rachel E Sutton
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Joseph Reidy
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Robert H Carnahan
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - W Hayes McDonald
- Department of Biochemistry and Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN, USA
| | - Clara T Schoeder
- Center of Structural Biology, Vanderbilt University, Nashville, TN, USA; Department of Chemistry, Vanderbilt University, Nashville, TN, USA
| | - William B Klimstra
- The Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 165261, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA 165261, USA
| | | | | | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard University, Cambridge, MA 02139, USA
| | - Jens Meiler
- Center of Structural Biology, Vanderbilt University, Nashville, TN, USA; Department of Chemistry, Vanderbilt University, Nashville, TN, USA; Institute for Drug Discovery, Leipzig University Medical School, Leipzig, Germany
| | - Kevin L Schey
- Department of Biochemistry and Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN, USA
| | - Justin G Julander
- Institute for Antiviral Research, Utah State University, Logan, UT 84335, USA
| | - Michael S Diamond
- Department of Medicine, Washington University, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University, St. Louis, MO 63110, USA
| | - James E Crowe
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN 37232, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA; The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
28
|
Kim AS, Kafai NM, Winkler ES, Gilliland TC, Cottle EL, Earnest JT, Jethva PN, Kaplonek P, Shah AP, Fong RH, Davidson E, Malonis RJ, Quiroz JA, Williamson LE, Vang L, Mack M, Crowe JE, Doranz BJ, Lai JR, Alter G, Gross ML, Klimstra WB, Fremont DH, Diamond MS. Pan-protective anti-alphavirus human antibodies target a conserved E1 protein epitope. Cell 2021; 184:4414-4429.e19. [PMID: 34416146 PMCID: PMC8382027 DOI: 10.1016/j.cell.2021.07.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 04/01/2021] [Accepted: 07/02/2021] [Indexed: 12/13/2022]
Abstract
Alphaviruses are emerging, mosquito-transmitted pathogens that cause musculoskeletal and neurological disease in humans. Although neutralizing antibodies that inhibit individual alphaviruses have been described, broadly reactive antibodies that protect against both arthritogenic and encephalitic alphaviruses have not been reported. Here, we identify DC2.112 and DC2.315, two pan-protective yet poorly neutralizing human monoclonal antibodies (mAbs) that avidly bind to viral antigen on the surface of cells infected with arthritogenic and encephalitic alphaviruses. These mAbs engage a conserved epitope in domain II of the E1 protein proximal to and within the fusion peptide. Treatment with DC2.112 or DC2.315 protects mice against infection by both arthritogenic (chikungunya and Mayaro) and encephalitic (Venezuelan, Eastern, and Western equine encephalitis) alphaviruses through multiple mechanisms, including inhibition of viral egress and monocyte-dependent Fc effector functions. These findings define a conserved epitope recognized by weakly neutralizing yet protective antibodies that could be targeted for pan-alphavirus immunotherapy and vaccine design.
Collapse
Affiliation(s)
- Arthur S Kim
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Natasha M Kafai
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Emma S Winkler
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Theron C Gilliland
- Center for Vaccine Research and Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Emily L Cottle
- Center for Vaccine Research and Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - James T Earnest
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Prashant N Jethva
- Department of Chemistry, Washington University in St. Louis, Saint Louis, MO 63130, USA
| | - Paulina Kaplonek
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Aadit P Shah
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Rachel H Fong
- Integral Molecular, Inc., Philadelphia, PA 19104, USA
| | | | - Ryan J Malonis
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jose A Quiroz
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Lauren E Williamson
- Vanderbilt Vaccine Center and Departments of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Lo Vang
- Emergent BioSolutions, Gaithersburg, MD 20879, USA
| | - Matthias Mack
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - James E Crowe
- Vanderbilt Vaccine Center and Departments of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | - Jonathan R Lai
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Michael L Gross
- Department of Chemistry, Washington University in St. Louis, Saint Louis, MO 63130, USA
| | - William B Klimstra
- Center for Vaccine Research and Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Daved H Fremont
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, Saint Louis, MO 63110, USA; Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| |
Collapse
|
29
|
Hasan SS, Dey D, Singh S, Martin M. The Structural Biology of Eastern Equine Encephalitis Virus, an Emerging Viral Threat. Pathogens 2021; 10:pathogens10080973. [PMID: 34451437 PMCID: PMC8400090 DOI: 10.3390/pathogens10080973] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/21/2021] [Accepted: 07/28/2021] [Indexed: 11/16/2022] Open
Abstract
Alphaviruses are arboviruses that cause arthritis and encephalitis in humans. Eastern Equine Encephalitis Virus (EEEV) is a mosquito-transmitted alphavirus that is implicated in severe encephalitis in humans with high mortality. However, limited insights are available into the fundamental biology of EEEV and residue-level details of its interactions with host proteins. In recent years, outbreaks of EEEV have been reported mainly in the United States, raising concerns about public safety. This review article summarizes recent advances in the structural biology of EEEV based mainly on single-particle cryogenic electron microscopy (cryoEM) structures. Together with functional analyses of EEEV and related alphaviruses, these structural investigations provide clues to how EEEV interacts with host proteins, which may open avenues for the development of therapeutics.
Collapse
Affiliation(s)
- S. Saif Hasan
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 N. Greene Street, Baltimore, MD 21201, USA; (D.D.); (S.S.); (M.M.)
- Center for Biomolecular Therapeutics, University of Maryland School of Medicine, 9600 Gudelsky Drive, Rockville, MD 20850, USA
- University of Maryland Marlene and Stewart Greenebaum Cancer Center, University of Maryland Medical Center, 22. S. Greene St., Baltimore, MD 21201, USA
- Correspondence:
| | - Debajit Dey
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 N. Greene Street, Baltimore, MD 21201, USA; (D.D.); (S.S.); (M.M.)
| | - Suruchi Singh
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 N. Greene Street, Baltimore, MD 21201, USA; (D.D.); (S.S.); (M.M.)
| | - Matthew Martin
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 N. Greene Street, Baltimore, MD 21201, USA; (D.D.); (S.S.); (M.M.)
| |
Collapse
|
30
|
Hughes HR, Velez JO, Davis EH, Laven J, Gould CV, Panella AJ, Lambert AJ, Staples JE, Brault AC. Fatal Human Infection with Evidence of Intrahost Variation of Eastern Equine Encephalitis Virus, Alabama, USA, 2019. Emerg Infect Dis 2021; 27:1886-1892. [PMID: 34152960 PMCID: PMC8237905 DOI: 10.3201/eid2707.210315] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Eastern equine encephalitis virus (EEEV) is an arbovirus in the family Togaviridae, genus Alphavirus, found in North America and associated with freshwater/hardwood swamps in the Atlantic, Gulf Coast, and Great Lakes regions. EEEV disease in humans is rare but causes substantial illness and death. To investigate the molecular epidemiology and microevolution of EEEV from a fatal case in Alabama, USA, in 2019, we used next-generation sequencing of serum and cerebrospinal fluid (CSF). Phylogenetic inference indicated that the infecting strain may be closely related to isolates from Florida detected during 2010-2014, suggesting potential seeding from Florida. EEEV detected in serum displayed a higher degree of variability with more single-nucleotide variants than that detected in the CSF. These data refine our knowledge of EEEV molecular epidemiologic dynamics in the Gulf Coast region and demonstrate potential quasispecies bottlenecking within the central nervous system of a human host.
Collapse
|
31
|
Cryo-EM structure of the mature and infective Mayaro virus at 4.4 Å resolution reveals features of arthritogenic alphaviruses. Nat Commun 2021; 12:3038. [PMID: 34031424 PMCID: PMC8144435 DOI: 10.1038/s41467-021-23400-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 04/27/2021] [Indexed: 12/19/2022] Open
Abstract
Mayaro virus (MAYV) is an emerging arbovirus of the Americas that may cause a debilitating arthritogenic disease. The biology of MAYV is not fully understood and largely inferred from related arthritogenic alphaviruses. Here, we present the structure of MAYV at 4.4 Å resolution, obtained from a preparation of mature, infective virions. MAYV presents typical alphavirus features and organization. Interactions between viral proteins that lead to particle formation are described together with a hydrophobic pocket formed between E1 and E2 spike proteins and conformational epitopes specific of MAYV. We also describe MAYV glycosylation residues in E1 and E2 that may affect MXRA8 host receptor binding, and a molecular “handshake” between MAYV spikes formed by N262 glycosylation in adjacent E2 proteins. The structure of MAYV is suggestive of structural and functional complexity among alphaviruses, which may be targeted for specificity or antiviral activity. Mayaro virus (MAYV) is an emerging arbovirus in Central and South America that is transmitted by mosquitoes and causes arthritogenic disease. Here, the authors present the 4.4 Å resolution cryo-EM structure of MAYV and describe specific features of the virus, which could be exploited for the design of MAYV-specific diagnostics and therapeutics.
Collapse
|
32
|
Hulswit RJG, Paesen GC, Bowden TA, Shi X. Recent Advances in Bunyavirus Glycoprotein Research: Precursor Processing, Receptor Binding and Structure. Viruses 2021; 13:353. [PMID: 33672327 PMCID: PMC7926653 DOI: 10.3390/v13020353] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/13/2021] [Accepted: 02/15/2021] [Indexed: 01/04/2023] Open
Abstract
The Bunyavirales order accommodates related viruses (bunyaviruses) with segmented, linear, single-stranded, negative- or ambi-sense RNA genomes. Their glycoproteins form capsomeric projections or spikes on the virion surface and play a crucial role in virus entry, assembly, morphogenesis. Bunyavirus glycoproteins are encoded by a single RNA segment as a polyprotein precursor that is co- and post-translationally cleaved by host cell enzymes to yield two mature glycoproteins, Gn and Gc (or GP1 and GP2 in arenaviruses). These glycoproteins undergo extensive N-linked glycosylation and despite their cleavage, remain associated to the virion to form an integral transmembrane glycoprotein complex. This review summarizes recent advances in our understanding of the molecular biology of bunyavirus glycoproteins, including their processing, structure, and known interactions with host factors that facilitate cell entry.
Collapse
Affiliation(s)
- Ruben J. G. Hulswit
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; (R.J.G.H.); (G.C.P.)
| | - Guido C. Paesen
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; (R.J.G.H.); (G.C.P.)
| | - Thomas A. Bowden
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; (R.J.G.H.); (G.C.P.)
| | - Xiaohong Shi
- MRC-University of Glasgow Centre for Virus Research, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G61 1QH, UK
| |
Collapse
|
33
|
Henss L, Yue C, Von Rhein C, Tschismarov R, Lewis-Ximenez LL, Dölle A, Baylis SA, Schnierle BS. Analysis of Humoral Immune Responses in Chikungunya Virus (CHIKV)-Infected Patients and Individuals Vaccinated With a Candidate CHIKV Vaccine. J Infect Dis 2021; 221:1713-1723. [PMID: 31828322 DOI: 10.1093/infdis/jiz658] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 12/11/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Chikungunya virus (CHIKV) is a mosquito-transmitted alphavirus that causes severe flu-like symptoms. The acute symptoms disappear after 1 week, but chronic arthralgia can persist for years. In this study, humoral immune responses in CHIKV-infected patients and vaccinees were analyzed. METHODS Alphavirus neutralization activity was analyzed with pseudotyped lentiviral vectors, and antibody epitope mapping was performed with a peptide array. RESULTS The greatest CHIKV neutralization activity was observed 60-92 days after onset of symptoms. The amount of CHIKV-specific antibodies and their binding avidity and cross-reactivity with other alphaviruses increased over time. Chikungunya virus and o'nyong-nyong virus (ONNV) were both neutralized to a similar extent. Linear antibody binding epitopes were mainly found in E2 domain B and the acid-sensitive regions (ASRs). In addition, serum samples from healthy volunteers vaccinated with a measles-vectored chikungunya vaccine candidate, MV-CHIK, were analyzed. Neutralization activity in the samples from the vaccine cohort was 2- to 6-fold lower than in samples from CHIKV-infected patients. In contrast to infection, vaccination only induced cross-neutralization with ONNV, and the E2 ASR1 was the major antibody target. CONCLUSIONS These data could assist vaccine design and enable the identification of correlates of protection necessary for vaccine efficacy.
Collapse
Affiliation(s)
- Lisa Henss
- Paul-Ehrlich-Institut, Department of Virology, Langen, Germany
| | - Constanze Yue
- Paul-Ehrlich-Institut, Department of Virology, Langen, Germany
| | | | | | | | | | - Sally A Baylis
- Paul-Ehrlich-Institut, Department of Virology, Langen, Germany
| | | |
Collapse
|
34
|
|
35
|
Williamson LE, Gilliland T, Yadav PK, Binshtein E, Bombardi R, Kose N, Nargi RS, Sutton RE, Durie CL, Armstrong E, Carnahan RH, Walker LM, Kim AS, Fox JM, Diamond MS, Ohi MD, Klimstra WB, Crowe JE. Human Antibodies Protect against Aerosolized Eastern Equine Encephalitis Virus Infection. Cell 2020; 183:1884-1900.e23. [PMID: 33301709 DOI: 10.1016/j.cell.2020.11.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/23/2020] [Accepted: 11/06/2020] [Indexed: 12/19/2022]
Abstract
Eastern equine encephalitis virus (EEEV) is one of the most virulent viruses endemic to North America. No licensed vaccines or antiviral therapeutics are available to combat this infection, which has recently shown an increase in human cases. Here, we characterize human monoclonal antibodies (mAbs) isolated from a survivor of natural EEEV infection with potent (<20 pM) inhibitory activity of EEEV. Cryo-electron microscopy reconstructions of two highly neutralizing mAbs, EEEV-33 and EEEV-143, were solved in complex with chimeric Sindbis/EEEV virions to 7.2 Å and 8.3 Å, respectively. The mAbs recognize two distinct antigenic sites that are critical for inhibiting viral entry into cells. EEEV-33 and EEEV-143 protect against disease following stringent lethal aerosol challenge of mice with highly pathogenic EEEV. These studies provide insight into the molecular basis for the neutralizing human antibody response against EEEV and can facilitate development of vaccines and candidate antibody therapeutics.
Collapse
Affiliation(s)
- Lauren E Williamson
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN 37232, USA
| | - Theron Gilliland
- The Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 165261, USA
| | - Pramod K Yadav
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Elad Binshtein
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Robin Bombardi
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Nurgun Kose
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Rachel S Nargi
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Rachel E Sutton
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Clarissa L Durie
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Erica Armstrong
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Robert H Carnahan
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Lauren M Walker
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN 37232, USA
| | - Arthur S Kim
- Department of Medicine, Washington University, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University, St. Louis, MO 63110, USA
| | - Julie M Fox
- Department of Medicine, Washington University, St. Louis, MO 63110, USA
| | - Michael S Diamond
- Department of Medicine, Washington University, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University, St. Louis, MO 63110, USA
| | - Melanie D Ohi
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - William B Klimstra
- The Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 165261, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA 165261, USA
| | - James E Crowe
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN 37232, USA; The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
36
|
Zengel J, Carette JE. Cracking the cell access code for the deadly virus VEEV. Nature 2020; 588:223-224. [PMID: 33208905 DOI: 10.1038/d41586-020-03192-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
37
|
Abstract
Alphaviruses cause severe human illnesses including persistent arthritis and fatal encephalitis. As alphavirus entry into target cells is the first step in infection, intensive research efforts have focused on elucidating aspects of this pathway, including attachment, internalization, and fusion. Herein, we review recent developments in the molecular understanding of alphavirus entry both in vitro and in vivo and how these advances might enable the design of therapeutics targeting this critical step in the alphavirus life cycle.
Collapse
|
38
|
Aksnes I, Markussen T, Braaen S, Rimstad E. Mutation of N-glycosylation Sites in Salmonid Alphavirus (SAV) Envelope Proteins Attenuate the Virus in Cell Culture. Viruses 2020; 12:v12101071. [PMID: 32987930 PMCID: PMC7650630 DOI: 10.3390/v12101071] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/18/2020] [Accepted: 09/21/2020] [Indexed: 12/16/2022] Open
Abstract
Salmonid alphavirus (SAV) is the cause of pancreas disease and sleeping disease in farmed salmonid fish in Europe. The spread of these diseases has been difficult to control with biosecurity and current vaccination strategies, and increased understanding of the viral pathogenesis could be beneficial for the development of novel vaccine strategies. N-glycosylation of viral envelope proteins may be crucial for viral virulence and a possible target for its purposed attenuation. In this study, we mutated the N-glycosylation consensus motifs of the E1 and E2 glycoproteins of a SAV3 infectious clone using site-directed mutagenesis. Mutation of the glycosylation motif in E1 gave a complete inactivation of the virus as no viral replication could be detected in cell culture and infectious particles could not be rescued. In contrast, infectious virus particles could be recovered from the SAV3 E2 mutants (E2319Q, E2319A), but not if they were accompanied by lack of N-glycosylation in E1. Compared to the non-mutated infectious clone, the SAV3-E2319Q and SAV3-E2319A recombinant viruses produced less cytopathic effects in cell culture and lower amounts of infectious viral particles. In conclusion, the substitution in the N-linked glycosylation site in E2 attenuated SAV3 in cell culture. The findings could be useful for immunization strategies using live attenuated vaccines and testing in fish will be desirable to study the clone’s properties in vivo.
Collapse
|
39
|
Brown RS, Anastasakis DG, Hafner M, Kielian M. Multiple capsid protein binding sites mediate selective packaging of the alphavirus genomic RNA. Nat Commun 2020; 11:4693. [PMID: 32943634 PMCID: PMC7499256 DOI: 10.1038/s41467-020-18447-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 08/24/2020] [Indexed: 12/16/2022] Open
Abstract
The alphavirus capsid protein (Cp) selectively packages genomic RNA (gRNA) into the viral nucleocapsid to produce infectious virus. Using photoactivatable ribonucleoside crosslinking and an innovative biotinylated Cp retrieval method, here we comprehensively define binding sites for Semliki Forest virus (SFV) Cp on the gRNA. While data in infected cells demonstrate Cp binding to the proposed genome packaging signal (PS), mutagenesis experiments show that PS is not required for production of infectious SFV or Chikungunya virus. Instead, we identify multiple Cp binding sites that are enriched on gRNA-specific regions and promote infectious SFV production and gRNA packaging. Comparisons of binding sites in cytoplasmic vs. viral nucleocapsids demonstrate that budding causes discrete changes in Cp-gRNA interactions. Notably, Cp’s top binding site is maintained throughout virus assembly, and specifically binds and assembles with Cp into core-like particles in vitro. Together our data suggest a model for selective alphavirus genome recognition and assembly. Alphaviruses need to selectively package genomic viral RNA for transmission, but the packaging mechanism remains unclear. Here, Brown et al. combine PAR-CLIP with biotinylated capsid protein (Cp) retrieval and identify multiple Cp binding sites on genomic viral RNA that promote virion formation.
Collapse
Affiliation(s)
- Rebecca S Brown
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Dimitrios G Anastasakis
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Disease, Bethesda, MD, 20892, USA
| | - Markus Hafner
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Disease, Bethesda, MD, 20892, USA
| | - Margaret Kielian
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| |
Collapse
|
40
|
Gale P. How virus size and attachment parameters affect the temperature sensitivity of virus binding to host cells: Predictions of a thermodynamic model for arboviruses and HIV. MICROBIAL RISK ANALYSIS 2020; 15:100104. [PMID: 32292808 PMCID: PMC7110232 DOI: 10.1016/j.mran.2020.100104] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 03/05/2020] [Accepted: 03/06/2020] [Indexed: 05/14/2023]
Abstract
Virus binding to host cells involves specific interactions between viral (glyco)proteins (GP) and host cell surface receptors (Cr) (protein or sialic acid (SA)). The magnitude of the enthalpy of association changes with temperature according to the change in heat capacity (ΔCp) on GP/Cr binding, being little affected for avian influenza virus (AIV) haemagglutinin (HA) binding to SA (ΔCp = 0 kJ/mol/K) but greatly affected for HIV gp120 binding to CD4 receptor (ΔCp = -5.0 kJ/mol/K). A thermodynamic model developed here predicts that values of ΔCp from 0 to ~-2.0 kJ/mol/K have relatively little impact on the temperature sensitivity of the number of mosquito midgut cells with bound arbovirus, while intermediate values of ΔCp of ~-3.0 kJ/mol/K give a peak binding at a temperature of ~20 °C as observed experimentally for Western equine encephalitis virus. More negative values of ΔCp greatly decrease arbovirus binding at temperatures below ~20 °C. Thus to promote transmission at low temperatures, arboviruses may benefit from ΔCp ~ 0 kJ/mol/K as for HA/SA and it is interesting that bluetongue virus binds to SA in midge midguts. Large negative values of ΔCp as for HIV gp120:CD4 diminish binding at 37 °C. Of greater importance, however, is the decrease in entropy of the whole virus (ΔSa_immob) on its immobilisation on the host cell surface. ΔSa_immob presents a repulsive force which the enthalpy-driven GP/Cr interactions weakened at higher temperatures struggle to overcome. ΔSa_immob is more negative (less favourable) for larger diameter viruses which therefore show diminished binding at higher temperatures than smaller viruses. It is proposed that small size phenotype through a less negative ΔSa_immob is selected for viruses infecting warmer hosts thus explaining the observation that virion volume decreases with increasing host temperature from 0 °C to 40 °C in the case of dsDNA viruses. Compared to arboviruses which also infect warm-blooded vertebrates, HIV is large at 134 nm diameter and thus would have a large negative ΔSa_immob which would diminish its binding at human body temperature. It is proposed that prior non-specific binding of HIV through attachment factors takes much of the entropy loss for ΔSa_immob so enhancing subsequent specific gp120:CD4 binding at 37 °C. This is consistent with the observation that HIV attachment factors are not essential but augment infection. Antiviral therapies should focus on increasing virion size, for example through binding of zinc oxide nanoparticles to herpes simplex virus, hence making ΔSa_immob more negative, and thus reducing binding affinity at 37 °C.
Collapse
Key Words
- AIV, avian influenza virus
- Antivirals
- BBF, brush border fragments from midgut
- BTV, bluetongue virus
- C.VT, number of host cells with bound virus at temperature T
- CD4, host cell receptor for HIV
- Cp, heat capacity at constant pressure
- Cr, host cell receptor
- Ctotal, number of host cells which can bind virus in a given volume of host fluid (midgut or blood)
- DENV, Dengue virus
- EA, activation energy
- EBOV, Zaire ebolavirus
- EM, electron microscopy
- Entropy
- Env, HIV gp120 trimer envelope protein which binds to a single CD4 molecule
- FcT, fraction of arthropod midgut cells with bound virus at temperature T
- GP, viral (glyco)protein on virus surface that binds to Cr
- HA, haemagglutinin
- HIV, human immunodeficiency virus
- HSV-2, herpes simplex virus type 2
- Heat capacity
- Ka_virus_T, association constant for binding of virus to host cells at temperature T
- Kd_receptor_T, dissociation constant for GP from Cr at temperature T
- Kd_virus, dissociation constant for virus from host cell
- M, molar (moles dm-3)
- R, ideal gas constant
- SA, sialic acid
- SIV, simian immunodeficiency virus
- Temperature
- Vfree, virus not bound to cells
- Virus size
- Vtotal, virus challenge dose in volume of host fluid
- WEEV, Western equine encephalitis virus
- WNV, West Nile virus
- ZnOT, zinc oxide tetrapod
- n, number of GP/Cr contacts made on virus binding to cell
- pcompleteT, probability given a virion has bound to the surface of a midgut cell that that midgut cell becomes infected and that its progeny viruses go on to infect the salivary gland so completing the arthropod infection process within the life time of the arthropod at temperature T
- ptransmissionT, probability of successful infection of the arthropod salivary glands after oral exposure at temperature T
- ΔCp, change in heat capacity
- ΔGa_virus_T, change in Gibbs free energy on association of virus and host cell at temperature T
- ΔHa_receptor_T, change in enthalpy for binding of virus GP to host Cr receptor at a temperature T
- ΔHa_virus_T, change in enthalpy for binding of virus to host cell at temperature T
- ΔSa_immob, change in entropy on immobilization of whole virus to cell surface
- ΔSa_non_specific, change in entropy on immobilization of virus to cell surface through non-specific binding
- ΔSa_receptor_T, change in entropy for binding of virus GP to host Cr receptor
- ΔSa_specific, change in entropy on immobilization of virus to cell surface through specific GP/Cr-driven binding
- ΔSa_virus_T, change in entropy for binding of virus to host cell at temperature T
Collapse
Affiliation(s)
- Paul Gale
- Independent Scientist, 15 Weare Close, Portland, Dorset, DT5 1JP, United Kingdom
| |
Collapse
|
41
|
Button JM, Qazi SA, Wang JCY, Mukhopadhyay S. Revisiting an old friend: new findings in alphavirus structure and assembly. Curr Opin Virol 2020; 45:25-33. [PMID: 32683295 DOI: 10.1016/j.coviro.2020.06.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 06/04/2020] [Accepted: 06/16/2020] [Indexed: 01/17/2023]
Abstract
Alphaviruses are transmitted by an arthropod vector to a vertebrate host. The disease pathologies, cellular environments, immune responses, and host factors are very different in these organisms. Yet, the virus is able to infect, replicate, and assemble into new particles in these two animals using one set of genetic instructions. The balance between conserved mechanisms and unique strategies during virus assembly is critical for fitness of the virus. In this review, we discuss new findings in receptor binding, polyprotein topology, nucleocapsid core formation, and particle budding that have emerged in the last five years and share opinions on how these new findings might answer some questions regarding alphavirus structure and assembly.
Collapse
Affiliation(s)
- Julie M Button
- Department of Molecular and Cellular Biochemistry, Indiana University, 212 S. Hawthorne Drive, Bloomington, IN 47405, United States
| | - Shefah A Qazi
- Department of Biology, Indiana University, 212 S. Hawthorne Drive, Bloomington, IN 47405, United States
| | - Joseph Che-Yen Wang
- Department of Microbiology and Immunology, Penn State College of Medicine, 700 HMC Crescent Road, Hershey, PA 17033, United States
| | - Suchetana Mukhopadhyay
- Department of Biology, Indiana University, 212 S. Hawthorne Drive, Bloomington, IN 47405, United States.
| |
Collapse
|
42
|
Roe K. A proposed treatment for pathogenic enveloped viruses having high rates of mutation or replication. Scand J Immunol 2020; 92:e12928. [PMID: 32640050 PMCID: PMC7361161 DOI: 10.1111/sji.12928] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/24/2020] [Accepted: 07/02/2020] [Indexed: 01/12/2023]
Abstract
Several enveloped viruses, particularly some RNA viruses, have high rates of mutation or replication, which can make them virulent pathogens in humans and other mammals. A proposed treatment could use synthesized proteins to mask pathogenic viral surface proteins to quickly induce an immune attack on specific enveloped viruses by using existing immune cells. One treatment could inject dual‐protein ligand masks into patients' bloodstreams to mask pathogenic surface proteins used to infect mammalian cells. The mammalian immune system already uses an analogous, more complex structure called a pentraxin to neutralize some pathogens by connecting their surface proteins to immune cells. And several types of antiviral peptides have already experimentally demonstrated effectiveness in blocking various viral pathogen infections. These treatments offer advantages, especially for currently untreatable viral pathogens. Furthermore, using dual‐protein ligands and the antigenic memory of some sub‐populations of NK cells would also allow the creation of defacto vaccines based on a host's NK cells, instead of vaccines utilizing CD4 and CD8 α:β T cells, which are limited by the requirement of MHC presentation of the target antigens to α:β T cells. Targeted NK cell vaccines could attack host cells latently or actively infected by intracellular pathogens, even host cells having pathogen downregulated MHC antigen presentation. Eight postulates concerning the effects of pathogen mutation, or change in phenotype from genetic recombination or rearrangement, and replication rates on pathogen vs host dominance are also listed, which should be applicable to viral and non‐viral pathogens.
Collapse
|
43
|
Abstract
Alphaviruses are enveloped positive-sense RNA viruses that can cause serious human illnesses such as polyarthritis and encephalitis. Despite their widespread distribution and medical importance, there are no licensed vaccines or antivirals to combat alphavirus infections. Berberine chloride (BBC) is a pan-alphavirus inhibitor that was previously identified in a replicon-based small-molecule screen. This work showed that BBC inhibits alphavirus replication but also suggested that BBC might have additional effects later in the viral life cycle. Here, we show that BBC has late effects that target the virus nucleocapsid (NC) core. Infected cells treated with BBC late in infection were unable to form stable cytoplasmic NCs or assembly intermediates, as assayed by gradient sedimentation. In vitro studies with recombinant capsid protein (Cp) and purified genomic RNA (gRNA) showed that BBC perturbs core-like particle formation and potentially traps the assembly process in intermediate states. Particles produced from BBC-treated cells were less infectious, despite efficient particle production and only minor decreases in genome packaging. In addition, BBC treatment of free virus particles strongly decreased alphavirus infectivity. In contrast, the infectivity of the negative-sense RNA virus vesicular stomatitis virus was resistant to BBC treatment of infected cells or free virus. Together, our data indicate that BBC alters alphavirus Cp-gRNA interactions and oligomerization and suggest that this may cause defects in NC assembly and in disassembly during subsequent virus entry. Thus, BBC may be considered a novel alphavirus NC assembly inhibitor.IMPORTANCE The alphavirus chikungunya virus (CHIKV) is an example of an emerging human pathogen with increased and rapid global spread. Although an acute CHIKV infection is rarely fatal, many patients suffer from debilitating chronic arthralgia for years. Antivirals against chikungunya and other alphaviruses have been identified in vitro, but to date none have been shown to be efficacious and have been licensed for human use. Here, we investigated a small molecule, berberine chloride (BBC), and showed that it inhibited infectious virus production by several alphaviruses including CHIKV. BBC acted on a late step in the alphavirus exit pathway, namely the formation of the nucleocapsid containing the infectious viral RNA. Better understanding of nucleocapsid formation and its inhibition by BBC will provide important information on the mechanisms of infectious alphavirus production and may enable their future targeting in antiviral strategies.
Collapse
|
44
|
Cryo-EM structure of eastern equine encephalitis virus in complex with heparan sulfate analogues. Proc Natl Acad Sci U S A 2020; 117:8890-8899. [PMID: 32245806 DOI: 10.1073/pnas.1910670117] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Eastern equine encephalitis virus (EEEV), a mosquito-borne icosahedral alphavirus found mainly in North America, causes human and equine neurotropic infections. EEEV neurovirulence is influenced by the interaction of the viral envelope protein E2 with heparan sulfate (HS) proteoglycans from the host's plasma membrane during virus entry. Here, we present a 5.8-Å cryoelectron microscopy (cryo-EM) structure of EEEV complexed with the HS analog heparin. "Peripheral" HS binding sites were found to be associated with the base of each of the E2 glycoproteins that form the 60 quasi-threefold spikes (q3) and the 20 sites associated with the icosahedral threefold axes (i3). In addition, there is one HS site at the vertex of each q3 and i3 spike (the "axial" sites). Both the axial and peripheral sites are surrounded by basic residues, suggesting an electrostatic mechanism for HS binding. These residues are highly conserved among EEEV strains, and therefore a change in these residues might be linked to EEEV neurovirulence.
Collapse
|
45
|
Stass R, Ng WM, Kim YC, Huiskonen JT. Structures of enveloped virions determined by cryogenic electron microscopy and tomography. Adv Virus Res 2019; 105:35-71. [PMID: 31522708 PMCID: PMC7112279 DOI: 10.1016/bs.aivir.2019.07.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Enveloped viruses enclose their genomes inside a lipid bilayer which is decorated by membrane proteins that mediate virus entry. These viruses display a wide range of sizes, morphologies and symmetries. Spherical viruses are often isometric and their envelope proteins follow icosahedral symmetry. Filamentous and pleomorphic viruses lack such global symmetry but their surface proteins may display locally ordered assemblies. Determining the structures of enveloped viruses, including the envelope proteins and their protein-protein interactions on the viral surface, is of paramount importance. These structures can reveal how the virions are assembled and released by budding from the infected host cell, how the progeny virions infect new cells by membrane fusion, and how antibodies bind surface epitopes to block infection. In this chapter, we discuss the uses of cryogenic electron microscopy (cryo-EM) in elucidating structures of enveloped virions. Starting from a detailed outline of data collection and processing strategies, we highlight how cryo-EM has been successfully utilized to provide unique insights into enveloped virus entry, assembly, and neutralization.
Collapse
Affiliation(s)
- Robert Stass
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Weng M Ng
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Young Chan Kim
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Juha T Huiskonen
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom; Helsinki Institute of Life Science HiLIFE and Research Programme in Molecular and Integrative Biosciences, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
46
|
Cross RW, Hastie KM, Mire CE, Robinson JE, Geisbert TW, Branco LM, Ollmann Saphire E, Garry RF. Antibody therapy for Lassa fever. Curr Opin Virol 2019; 37:97-104. [PMID: 31401518 DOI: 10.1016/j.coviro.2019.07.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Revised: 07/01/2019] [Accepted: 07/03/2019] [Indexed: 02/06/2023]
Abstract
Serum from convalescent Lassa fever patients was previously shown to be ineffective as a source of protective antibodies in some early studies. Subsequently, monoclonal antibodies (MAbs) to the Lassa virus (LASV) glycoprotein produced by memory B cells of West African patients who survived Lassa fever were identified. Development of MAbs as potential Lassa immunotherapeutics was facilitated by structural studies and mutational analyses that identified protective epitopes on the prefusion form of the LASV glycoprotein. Human mAbs were screened for reactivity to different neutralizing epitopes, potency, and broad reactivity against multiple lineages of LASV. MAbs were downselected in a guinea pig model of Lassa fever. A cocktail of three human MAbs designated Arevirumab-3 rescued 100% of Cynomolgus macaques at advanced stages of disease more than a week post-infection. Antibody therapeutics may be further developed in clinical trials in endemic areas potentially offering a key treatment option for Lassa fever.
Collapse
Affiliation(s)
- Robert W Cross
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA; Galveston National Laboratory, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | | | - Chad E Mire
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA; Galveston National Laboratory, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | - James E Robinson
- Department of Pediatrics, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70112, USA
| | - Thomas W Geisbert
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA; Galveston National Laboratory, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | | | | | - Robert F Garry
- Zalgen Labs, LLC, Germantown, MD 20876, USA; Department of Microbiology and Immunology, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70112, USA.
| |
Collapse
|
47
|
Johnson JE. Michael G. Rossmann (1930–2019): Leadership in structural biology for 60 years. Protein Sci 2019. [DOI: 10.1002/pro.3671] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- John E. Johnson
- Department of Integrative Structural and Computational BiologyThe Scripps Research Institute La Jolla California 92037
| |
Collapse
|
48
|
Cryo-EM Structure of Chikungunya Virus in Complex with the Mxra8 Receptor. Cell 2019; 177:1725-1737.e16. [PMID: 31080061 DOI: 10.1016/j.cell.2019.04.006] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 01/22/2019] [Accepted: 04/01/2019] [Indexed: 11/21/2022]
Abstract
Mxra8 is a receptor for multiple arthritogenic alphaviruses that cause debilitating acute and chronic musculoskeletal disease in humans. Herein, we present a 2.2 Å resolution X-ray crystal structure of Mxra8 and 4 to 5 Å resolution cryo-electron microscopy reconstructions of Mxra8 bound to chikungunya (CHIKV) virus-like particles and infectious virus. The Mxra8 ectodomain contains two strand-swapped Ig-like domains oriented in a unique disulfide-linked head-to-head arrangement. Mxra8 binds by wedging into a cleft created by two adjacent CHIKV E2-E1 heterodimers in one trimeric spike and engaging a neighboring spike. Two binding modes are observed with the fully mature VLP, with one Mxra8 binding with unique contacts. Only the high-affinity binding mode was observed in the complex with infectious CHIKV, as viral maturation and E3 occupancy appear to influence receptor binding-site usage. Our studies provide insight into how Mxra8 binds CHIKV and creates a path for developing alphavirus entry inhibitors.
Collapse
|