1
|
Wang X, Liu K, Wang X, Liu X. Multidimensional Regulatory Mechanisms and Targeting Strategies of the eEF1 Family in RNA Virus Infection. Viruses 2025; 17:682. [PMID: 40431694 PMCID: PMC12115637 DOI: 10.3390/v17050682] [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: 03/31/2025] [Revised: 04/30/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
The eukaryotic translation elongation factor 1 (eEF1) family exhibits critical roles in RNA viral infection beyond its canonical function in protein synthesis. This review analyzes the structural characteristics of eEF1A and the eEF1B complex, and their regulatory mechanisms during viral infection. eEF1A impacts viral replication by stabilizing viral RNA-dependent RNA polymerase (RdRp) complexes, modulating genomic RNA synthesis, and facilitating viral assembly through cytoskeletal regulation. eEF1B subunits contribute through enhancing viral mRNA translation, regulating nuclear transport of viral components, and mediating post-translational modifications. The high conservation of eEF1 proteins across species and their involvement in multiple stages of viral replication establish them as promising broad-spectrum antiviral targets. Current eEF1-targeting compounds like plitidepsin demonstrate efficacy against diverse viral families, though therapeutic development faces challenges in balancing antiviral activity with host toxicity. This review provides a theoretical foundation for developing novel antiviral strategies targeting host-virus interaction interfaces and offers insights into addressing emerging infectious diseases.
Collapse
Affiliation(s)
- Xin Wang
- Key Laboratory of Avian Bioproducts Development, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (X.W.); (X.L.)
| | - Kaituo Liu
- Key Laboratory of Avian Bioproducts Development, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (X.W.); (X.L.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou 225009, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Xiaoquan Wang
- Key Laboratory of Avian Bioproducts Development, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (X.W.); (X.L.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou 225009, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Xiufan Liu
- Key Laboratory of Avian Bioproducts Development, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (X.W.); (X.L.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou 225009, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
2
|
Huang IH, Lai GC, Chao TL, Liu WD, Chang SY, Chang SC. Monkeypox virus H3L protein as the target antigen for developing neutralizing antibody and serological assay. Appl Microbiol Biotechnol 2025; 109:80. [PMID: 40172630 PMCID: PMC11965195 DOI: 10.1007/s00253-025-13466-6] [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: 01/22/2025] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/04/2025]
Abstract
The large number of atypical monkeypox (Mpox) cases caused by emerging monkeypox virus (MPXV) strains was recently found in countries and regions where the Mpox was not reported before. Diagnostic tools and therapeutic agents are important countermeasures for preventing Mpox outbreak. H3L protein is the important surface antigen of MPXV for binding to host cell receptors and mediating viral infection. A broad range of murine anti-MPXV H3L monoclonal antibodies (mAbs) recognizing various binding epitopes have been generated in the study. The rapid test composed of the mAbs 4-2A and 3-3F can specifically detect H3L protein and MPXV virion. The mAb 3-3F exhibited strong MPXV neutralizing activity in a complement-dependent manner. Notably, 3-3F binds to a unique epitope within residues 35-89 of H3L protein. The serum samples collected from Mpox patients barely bound to the N-terminal portion of H3L protein ranging from 2 to 89 residues, indicating that the content of the 3-3F-like antibody is very low in Mpox patient sera. In contrast, the seropositivity was mostly observed using the C-terminal portion of H3L protein ranging from 185 to 282 residues as the target antigen in the immunoblot analysis. Taken together, the anti-MPXV H3L mAb can be developed as the Mpox diagnostic and therapeutic agents. Furthermore, H3L protein is the promising biomarker for serological analysis. KEY POINTS: •Anti-H3L mAbs can cross-react with H3L proteins in MPXV and VACV virions. •The LFIA rapid test using the mAbs 4-2A and 3-3F can specifically detect MPXV. •MPXV was neutralized by mAb 3-3F in a complement-dependent manner.
Collapse
MESH Headings
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/blood
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/isolation & purification
- Animals
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- Humans
- Antigens, Viral/immunology
- Monkeypox virus/immunology
- Mice
- Mpox, Monkeypox/diagnosis
- Mpox, Monkeypox/immunology
- Serologic Tests/methods
- Mice, Inbred BALB C
- Epitopes/immunology
Collapse
Affiliation(s)
- I-Hsiang Huang
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, 106, Taiwan
| | - Guan-Chun Lai
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, 106, Taiwan
| | - Tai-Ling Chao
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Wang-Da Liu
- Department of Internal Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
- Department of Medicine, National Taiwan University Cancer Center, Taipei , 106, Taiwan
| | - Sui-Yuan Chang
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan.
- Department of Laboratory Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, 100, Taiwan.
| | - Shih-Chung Chang
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, 106, Taiwan.
- Center of Biotechnology, National Taiwan University, Taipei, 106, Taiwan.
| |
Collapse
|
3
|
Laidlaw SM, Ulaeto D, Lonsdale S, Clark G, Sumner R, Edwards T, Adams E, Logist AS, Van Holm B, Maluquer de Motes C, Horby P, Maes P, Carroll MW. Detection of mpox and other orthopoxviruses using a lateral flow device as a point-of-care diagnostic. Microbiol Spectr 2025; 13:e0245624. [PMID: 40008874 PMCID: PMC11960085 DOI: 10.1128/spectrum.02456-24] [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: 09/27/2024] [Accepted: 12/23/2024] [Indexed: 02/27/2025] Open
Abstract
In 2022, the World Health Organization declared the worldwide outbreak of mpox to be a public health emergency of international concern. The causative monkeypox virus (MPXV) belonged to clade IIb and is transmitted through sexual contact with a low case fatality rate (0.1%), which, together with under-detection, all contributed to a rapid global spread particularly within the MSM (men who have sex with men) community. As MPXV clade II remains circulating worldwide, a new outbreak of the more fatal clade I disease has been declared in Central and East Africa, and remains uncontrolled in part due to the lack of point-of-care (POC) diagnostics for rapid decisions on treatment and self-isolation. To address the lack of POC solutions for mpox, we have designed and evaluated an orthopoxvirus-specific lateral flow device (LFD) that could be used for the diagnosis of mpox. Using an LFD comprising four monoclonal antibodies against the A27 protein, we demonstrate sensitivity to 3 × 105 pfu/mL. This sensitivity is expected to be sufficient for the detection of MPXV from lesion sites and may also be sufficient for other sample types such as saliva and urine. We found that the presence of guanidinium thiocyanate, a common ingredient in inactivating viral transport media, masked the LFD antigen, resulting in false negatives. POC diagnosis of mpox may be possible using an LFD to reduce delays arising from sample shipment to centralized laboratory testing facilities. In order to achieve this, our work demonstrates that an LFD-optimized buffer is required, as the sample collection buffer may have a detrimental impact on sensitivity for clinical material.IMPORTANCEMpox cases have dramatically increased both in traditionally monkeypox virus endemic countries and also worldwide. This increase comes at a time when immunity derived from smallpox vaccination is no longer available. Diagnosis of mpox is complicated due to both disease presentation and the availability of local diagnostic laboratories. The availability of a point-of-care diagnostic tool such as an lateral flow device (LFD) would play an important role to both diagnose and prevent onward transmission. This manuscript provides developers and assessors with key data for defining true sensitivity and specificity of a successful LFD in addition to buffer conditions for sample collection.
Collapse
Affiliation(s)
- Stephen M. Laidlaw
- Pandemic Sciences Institute (PSI), University of Oxford, Oxford, United Kingdom
- Centre for Human Genetics (CHG), University of Oxford, Oxford, United Kingdom
| | | | | | | | - Rebecca Sumner
- Department of Microbial Sciences, University of Surrey, Guildford, United Kingdom
| | - Thomas Edwards
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Emily Adams
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
- Global Access Diagnostics, Bedford, United Kingdom
| | - Anne-Sophie Logist
- Laboratory of Clinical and Epidemiological Virology (Rega Institute), Ku Leuven, Leuven, Belgium
| | - Bram Van Holm
- Laboratory of Clinical and Epidemiological Virology (Rega Institute), Ku Leuven, Leuven, Belgium
| | | | - Peter Horby
- Pandemic Sciences Institute (PSI), University of Oxford, Oxford, United Kingdom
| | - Piet Maes
- Laboratory of Clinical and Epidemiological Virology (Rega Institute), Ku Leuven, Leuven, Belgium
| | - Miles W. Carroll
- Pandemic Sciences Institute (PSI), University of Oxford, Oxford, United Kingdom
- Centre for Human Genetics (CHG), University of Oxford, Oxford, United Kingdom
| |
Collapse
|
4
|
Teffera M, Boshra H, Bowden TR, Babiuk S. Which Proteins? The Challenge of Identifying the Protective Antigens for Next-Generation Capripoxvirus Vaccines. Vaccines (Basel) 2025; 13:219. [PMID: 40266091 PMCID: PMC11946534 DOI: 10.3390/vaccines13030219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/13/2025] [Accepted: 02/20/2025] [Indexed: 04/24/2025] Open
Abstract
Sheeppox, goatpox, and lumpy skin disease continue to negatively impact the sheep, goat, and cattle industries in countries where these diseases are present and threaten to spread into new regions. Effective vaccines are available for disease control and eradication. However, commercial vaccines are based on live attenuated virus isolates and therefore it is not currently possible to differentiate between infected and vaccinated animals (DIVA), which severely limits the use of these vaccines in countries that are free from disease and at risk of an incursion. The development of next-generation vaccines, including recombinant protein, viral-vectored, and mRNA, has been limited due to the lack of understanding of the protective antigen(s) of capripoxviruses. The complexity of capripoxviruses, with up to 156 open reading frames, makes the identification of protective antigen(s) difficult. This paper identifies the most promising antigens by first considering the membrane-associated proteins and then further selecting proteins based on immunogenicity and their role in immunity by comparing them to known orthopoxvirus homologues. From the 156 potential antigens, 13 have been identified as being the most likely to be protective. Further evaluation of these proteins, as immunogens, would be required to identify the optimal combination of immunodominant antigen(s) for the development of next-generation capripoxvirus vaccines.
Collapse
Affiliation(s)
- Mahder Teffera
- Canadian Food Inspection Agency, National Centre for Foreign Animal Disease, Winnipeg, MB R3E 3M4, Canada;
| | - Hani Boshra
- Department of Pathology, Fundamental and Applied Research for Animals and Health (FARAH), Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium;
| | - Timothy R. Bowden
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Australian Centre for Disease Preparedness (ACDP), East Geelong, VIC 3219, Australia;
| | - Shawn Babiuk
- Canadian Food Inspection Agency, National Centre for Foreign Animal Disease, Winnipeg, MB R3E 3M4, Canada;
- Department of Immunology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| |
Collapse
|
5
|
Duan H, Shi Q, Yue X, Zhang Z, Liu L, Wang Y, Cao Y, Ou Z, Liang L, Hu J, Shi H. Identification of core therapeutic targets for Monkeypox virus and repurposing potential of drugs: A WEB prediction approach. PLoS One 2024; 19:e0303501. [PMID: 39642129 PMCID: PMC11623562 DOI: 10.1371/journal.pone.0303501] [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: 01/29/2024] [Accepted: 10/03/2024] [Indexed: 12/08/2024] Open
Abstract
A new round of monkeypox virus has emerged in the United Kingdom since July 2022 and rapidly swept the world. Currently, despite numerous research groups are studying this virus and seeking effective treatments, the information on the open reading frame, inhibitors, and potential targets of monkeypox has not been updated in time, and the comprehension of monkeypox target ligand interactions remains a key challenge. Here, we first summarized and improved the open reading frame information of monkeypox, constructed the monkeypox inhibitor library and potential targets library by database research as well as literature search, combined with advanced protein modeling technologies (Sequence-based and AI algorithms-based homology modeling). In addition, we build monkeypox virus Docking Server, a web server to predict the binding mode between targets and substrate. The open reading frame information, monkeypox inhibitor library, and monkeypox potential targets library are used as the initial files for server docking, providing free interactive tools for predicting ligand interactions of monkeypox targets, potential drug screening, and potential targets search. In addition, the update of the three databases can also effectively promote the study of monkeypox drug inhibition mechanism and provide theoretical guidance for the development of drugs for monkeypox.
Collapse
Affiliation(s)
- Huaichuan Duan
- Laboratory of Tumor Targeted and Immune Therapy, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Quanshan Shi
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, School of Pharmacy, Chengdu University, Chengdu, China
| | - Xinru Yue
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, School of Pharmacy, Chengdu University, Chengdu, China
| | - Zelan Zhang
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, School of Pharmacy, Chengdu University, Chengdu, China
| | - Ling Liu
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, School of Pharmacy, Chengdu University, Chengdu, China
| | - Yueteng Wang
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, School of Pharmacy, Chengdu University, Chengdu, China
| | - Yujie Cao
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, School of Pharmacy, Chengdu University, Chengdu, China
| | - Zuoxin Ou
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, School of Pharmacy, Chengdu University, Chengdu, China
| | - Li Liang
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, School of Pharmacy, Chengdu University, Chengdu, China
| | - Jianping Hu
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, School of Pharmacy, Chengdu University, Chengdu, China
| | - Hubing Shi
- Laboratory of Tumor Targeted and Immune Therapy, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| |
Collapse
|
6
|
George Pryzdial EL, Perrier JR, Rashid MU, West HE, Sutherland MR. Viral coagulation: pushing the envelope. J Thromb Haemost 2024; 22:3366-3382. [PMID: 39260743 DOI: 10.1016/j.jtha.2024.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 07/11/2024] [Accepted: 08/19/2024] [Indexed: 09/13/2024]
Abstract
Many virus types affect the blood clotting system with correlations to pathology that range widely from thrombosis to hemorrhage linking to inflammation. Here we overview the intricate crosstalk induced by infection between proteins on the virus encoded by either the host or virus genomes, coagulation proteins, platelets, leukocytes, and endothelial cells. For blood-borne viruses with an outer covering acquired from the host cell, the envelope, a key player may be the cell-derived trigger of coagulation on the virus surface, tissue factor (TF). TF is a multifunctional transmembrane cofactor that accelerates factor (F)VIIa-dependent activation of FX to FXa, leading to clot formation. However, the nascent TF/FVIIa/FXa complex also facilitates G protein-coupled modulation of cells via protease-activated receptor 2. As a viral envelope constituent, TF can bypass the physiological modes of regulation, thereby initiating the activation of neighboring platelets, leukocytes, and endothelial cells. A thromboinflammatory environment is predicted due to feedback amplification in response to cellular release of cytokines, procoagulant proteins, neutrophil extracellular traps, and stimulus-induced accessibility of adhesive receptors, resulting in cellular aggregates. The pathobiological effects of thromboinflammation ultimately contribute to innate and adaptive immunity for viral clearance. In contrast, the preceding stages of viral infection may be enhanced via the TF-protease axis.
Collapse
Affiliation(s)
- Edward Louis George Pryzdial
- Centre for Blood Research, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Division of Medical Affairs and Innovation, Canadian Blood Services, Ottawa, Ontario, Canada.
| | - John Ruggles Perrier
- Centre for Blood Research, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Division of Medical Affairs and Innovation, Canadian Blood Services, Ottawa, Ontario, Canada
| | - Mahamud-Ur Rashid
- Centre for Blood Research, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Division of Medical Affairs and Innovation, Canadian Blood Services, Ottawa, Ontario, Canada
| | - Henry Euan West
- Centre for Blood Research, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Division of Medical Affairs and Innovation, Canadian Blood Services, Ottawa, Ontario, Canada
| | - Michael Ross Sutherland
- Centre for Blood Research, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Division of Medical Affairs and Innovation, Canadian Blood Services, Ottawa, Ontario, Canada
| |
Collapse
|
7
|
Yang X, Fox A, DeCarlo C, Powell RL. Unique Kinetics of the Human Milk Antibody Response to JYNNEOS Vaccine for Prevention of Monkey Pox: A Case Study. Breastfeed Med 2024; 19:974-979. [PMID: 39360771 PMCID: PMC11655390 DOI: 10.1089/bfm.2024.0257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Background: JYNNEOS is a nonreplicating modified vaccinia Ankara vaccine currently licensed to prevent monkeypox infection, and its milk immunogenicity remains unstudied. Objective: Investigate the human milk immunogenicity of the JYNNEOS vaccine in one individual and examine the milk for evidence of vaccine components. Methods: Immunogenicity of milk and plasma samples were tested by Luminex assays against Vaccinia antigens, and vaccine components were tested using PCR and sandwich ELISA. Results: Plasma antibody (Ab) response increased up to 3.7-fold in immunoglobulin G (IgG) titer and 1.4-fold in IgA compared with baseline, confirming vaccine immunogenicity in this participant 2 weeks post dose 2. Specific plasma IgG remained 1.2- to 1.7-fold above baseline 12 weeks post dose 2, while IgA returned to baseline levels. Notably, the milk response exhibited unique kinetics, particularly for IgA. Milk IgA against all three antigens increased 0.9- to 2.2-fold 2 weeks post dose 2, reaching a peak titer increase of 1.1- to 2.7-fold at 12 weeks post dose 2. Secretory (s) Ab levels increased to 1.1- to 2-fold at 2 weeks post dose 2 and reached a peak of 2- to 3.2-fold increase at the 12-week time point. Importantly, IgA and sAb responses in milk exhibited correlation, suggesting most milk IgA was sIgA. Notably, no vaccine components (VACV protein or DNA) were detected in the milk samples. Conclusion: These data suggest that the milk Ab response to this intradermal (ID) VACV-based vaccine is distinct from that observed systemically, indicating a unique mucosal immune response and highlighting its potential to elicit protective long-lasting sIgA. This case report provides strong evidence for inclusion of this vaccine platform in future studies of maternal vaccines aimed to elicit a protective milk Ab response.
Collapse
Affiliation(s)
- Xiaoqi Yang
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Alisa Fox
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Claire DeCarlo
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Rebecca L.R. Powell
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
8
|
Cua S, Tello BA, Farelo MA, Rodriguez E, Escalante GM, Mutsvunguma LZ, Ogembo JG, Reidel IG. A Novel Monoclonal Antibody Against a Modified Vaccinia Ankara (MVA) Envelope Protein as a Tool for MVA Virus Titration by Flow Cytometry. Viruses 2024; 16:1628. [PMID: 39459960 PMCID: PMC11512277 DOI: 10.3390/v16101628] [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: 09/12/2024] [Revised: 10/12/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Modified vaccinia Ankara (MVA) virus is a widely used vaccine platform, making accurate titration essential for vaccination studies. However, the current plaque forming unit (PFU) assay, the standard for MVA titration, is prone to observer bias and other limitations that affect accuracy and precision. To address these challenges, we developed a new flow cytometry-based quantification method using a highly specific monoclonal antibody (mAb) for the detection of MVA-infected cells, as a more accurate titration assay. Through previous work, we serendipitously identified three MVA-specific hybridoma antibody clones, which we characterized through ELISA, immunoblot, and flow cytometry, confirming their specificity for MVA. Sequencing confirmed that each antibody was monoclonal, and mass spectrometry results revealed that all mAbs target the MVA cell surface binding protein (CSBP, MVA105L). We next optimized the titration protocol using the most effective mAb, 33C7 by refining culture conditions and staining protocols to enhance sensitivity and minimize background. Our optimized method demonstrated superior sensitivity, reliability, and reduced processing time when compared with the traditional PFU assay, establishing it as a more accurate and efficient approach for MVA titration.
Collapse
Affiliation(s)
- Simeon Cua
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Brenda A Tello
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Mafalda A Farelo
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Esther Rodriguez
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
- Irell & Manella Graduate School of Biological Sciences, City of Hope, Duarte, CA 91010, USA
| | - Gabriela M Escalante
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Lorraine Z Mutsvunguma
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Javier Gordon Ogembo
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Ivana G Reidel
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
9
|
Huang P, Xia M, Vago FS, Jiang W, Tan M. A Pseudovirus Nanoparticle Displaying the Vaccinia Virus L1 Protein Elicited High Neutralizing Antibody Titers and Provided Complete Protection to Mice against Mortality Caused by a Vaccinia Virus Challenge. Vaccines (Basel) 2024; 12:846. [PMID: 39203972 PMCID: PMC11359793 DOI: 10.3390/vaccines12080846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/16/2024] [Accepted: 07/21/2024] [Indexed: 09/03/2024] Open
Abstract
The recent worldwide incidence of mpox infection and concerns about future emerging variants of mpox viruses highlight the need for the development of a new generation of mpox vaccines. To achieve this goal, we utilized our norovirus S nanoparticle vaccine platform to produce and evaluate two pseudovirus nanoparticles (PVNPs), S-L1 and S-J1. These PVNPs displayed the L1 neutralizing antigen target of the vaccinia virus and a yet-untested J1 antigen of the mpox virus, respectively, with the aim of creating an effective nanoparticle-based mpox vaccine. Each self-assembled PVNP consists of an inner shell resembling the interior layer of the norovirus capsid and multiple L1 or J1 antigens on the surface. The PVNPs improved the antibody responses toward the displayed L1 or J1 antigens in mice, resulting in significantly greater L1/J1-specific IgG and IgA titers than those elicited by the corresponding free L1 or J1 antigens. After immunization with the S-L1 PVNPs, the mouse sera exhibited high neutralizing antibody titers against the vaccinia virus, and the S-L1 PVNPs provided mice with 100% protection against mortality caused by vaccinia virus challenge. In contrast, the S-J1 PVNPs induced low neutralizing antibody titers and conferred mice weak protective immunity. These data confirm that the L1 protein is an excellent vaccine target and that the readily available S-L1 PVNPs are a promising mpox vaccine candidate worthy of further development.
Collapse
Affiliation(s)
- Pengwei Huang
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; (P.H.); (M.X.)
| | - Ming Xia
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; (P.H.); (M.X.)
| | - Frank S. Vago
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA; (F.S.V.); (W.J.)
| | - Wen Jiang
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA; (F.S.V.); (W.J.)
| | - Ming Tan
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; (P.H.); (M.X.)
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| |
Collapse
|
10
|
Liu J, Corroyer-Dulmont S, Pražák V, Khusainov I, Bahrami K, Welsch S, Vasishtan D, Obarska-Kosińska A, Thorkelsson SR, Grünewald K, Quemin ERJ, Turoňová B, Locker JK. The palisade layer of the poxvirus core is composed of flexible A10 trimers. Nat Struct Mol Biol 2024; 31:1105-1113. [PMID: 38316878 PMCID: PMC11257942 DOI: 10.1038/s41594-024-01218-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 01/05/2024] [Indexed: 02/07/2024]
Abstract
Due to its asymmetric shape, size and compactness, the structure of the infectious mature virus (MV) of vaccinia virus (VACV), the best-studied poxvirus, remains poorly understood. Instead, subviral particles, in particular membrane-free viral cores, have been studied with cryo-electron microscopy. Here, we compared viral cores obtained by detergent stripping of MVs with cores in the cellular cytoplasm, early in infection. We focused on the prominent palisade layer on the core surface, combining cryo-electron tomography, subtomogram averaging and AlphaFold2 structure prediction. We showed that the palisade is composed of densely packed trimers of the major core protein A10. Trimers display a random order and their classification indicates structural flexibility. A10 on cytoplasmic cores is organized in a similar manner, indicating that the structures obtained in vitro are physiologically relevant. We discuss our results in the context of the VACV replicative cycle, and the assembly and disassembly of the infectious MV.
Collapse
Affiliation(s)
- Jiasui Liu
- Department of Molecular Sociology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Simon Corroyer-Dulmont
- Centre for Structural Systems Biology, Leibniz Institute of Virology, Hamburg, Germany
- University of Hamburg, Hamburg, Germany
| | - Vojtěch Pražák
- Centre for Structural Systems Biology, Leibniz Institute of Virology, Hamburg, Germany
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Iskander Khusainov
- Department of Molecular Sociology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Karola Bahrami
- Electron Microscopy of Pathogens, Paul Ehrlich Institute, Langen, Germany
- University Clinic Frankfurt, Frankfurt am Main, Germany
| | - Sonja Welsch
- Max Planck Institute of Biophysics, Central Electron Microscopy Facility, Frankfurt am Main, Germany
| | - Daven Vasishtan
- Centre for Structural Systems Biology, Leibniz Institute of Virology, Hamburg, Germany
- Department of Biochemistry, University of Oxford, Oxford, UK
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | | | - Sigurdur R Thorkelsson
- Centre for Structural Systems Biology, Leibniz Institute of Virology, Hamburg, Germany
- MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Kay Grünewald
- Centre for Structural Systems Biology, Leibniz Institute of Virology, Hamburg, Germany.
- University of Hamburg, Hamburg, Germany.
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK.
| | - Emmanuelle R J Quemin
- Centre for Structural Systems Biology, Leibniz Institute of Virology, Hamburg, Germany.
- Department of Virology, Institute for Integrative Biology of the Cell (I2BC), CNRS UMR9198, Université Paris-Saclay, CEA, Gif-sur-Yvette, France.
| | - Beata Turoňová
- Department of Molecular Sociology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany.
| | - Jacomina Krijnse Locker
- Electron Microscopy of Pathogens, Paul Ehrlich Institute, Langen, Germany.
- Justus Liebig University of Giessen, Giessen, Germany.
| |
Collapse
|
11
|
Datler J, Hansen JM, Thader A, Schlögl A, Bauer LW, Hodirnau VV, Schur FKM. Multi-modal cryo-EM reveals trimers of protein A10 to form the palisade layer in poxvirus cores. Nat Struct Mol Biol 2024; 31:1114-1123. [PMID: 38316877 PMCID: PMC11257981 DOI: 10.1038/s41594-023-01201-6] [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: 06/11/2023] [Accepted: 12/06/2023] [Indexed: 02/07/2024]
Abstract
Poxviruses are among the largest double-stranded DNA viruses, with members such as variola virus, monkeypox virus and the vaccination strain vaccinia virus (VACV). Knowledge about the structural proteins that form the viral core has remained sparse. While major core proteins have been annotated via indirect experimental evidence, their structures have remained elusive and they could not be assigned to individual core features. Hence, which proteins constitute which layers of the core, such as the palisade layer and the inner core wall, has remained enigmatic. Here we show, using a multi-modal cryo-electron microscopy (cryo-EM) approach in combination with AlphaFold molecular modeling, that trimers formed by the cleavage product of VACV protein A10 are the key component of the palisade layer. This allows us to place previously obtained descriptions of protein interactions within the core wall into perspective and to provide a detailed model of poxvirus core architecture. Importantly, we show that interactions within A10 trimers are likely generalizable over members of orthopox- and parapoxviruses.
Collapse
Affiliation(s)
- Julia Datler
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| | - Jesse M Hansen
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| | - Andreas Thader
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| | - Alois Schlögl
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| | - Lukas W Bauer
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| | | | - Florian K M Schur
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria.
| |
Collapse
|
12
|
Velez-Brochero M, Behera P, Afreen KS, Odle A, Rajsbaum R. Ubiquitination in viral entry and replication: Mechanisms and implications. Adv Virus Res 2024; 119:1-38. [PMID: 38897707 DOI: 10.1016/bs.aivir.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
The ubiquitination process is a reversible posttranslational modification involved in many essential cellular functions, such as innate immunity, cell signaling, trafficking, protein stability, and protein degradation. Viruses can use the ubiquitin system to efficiently enter host cells, replicate and evade host immunity, ultimately enhancing viral pathogenesis. Emerging evidence indicates that enveloped viruses can carry free (unanchored) ubiquitin or covalently ubiquitinated viral structural proteins that can increase the efficiency of viral entry into host cells. Furthermore, viruses continuously evolve and adapt to take advantage of the host ubiquitin machinery, highlighting its importance during virus infection. This review discusses the battle between viruses and hosts, focusing on how viruses hijack the ubiquitination process at different steps of the replication cycle, with a specific emphasis on viral entry. We discuss how ubiquitination of viral proteins may affect tropism and explore emerging therapeutics strategies targeting the ubiquitin system for antiviral drug discovery.
Collapse
Affiliation(s)
- Maria Velez-Brochero
- Center for Virus-Host-Innate Immunity and Department of Medicine, Rutgers Biomedical and Health Sciences, Institute for Infectious and Inflammatory Diseases, Rutgers University, Newark, NJ, United States
| | - Padmanava Behera
- Center for Virus-Host-Innate Immunity and Department of Medicine, Rutgers Biomedical and Health Sciences, Institute for Infectious and Inflammatory Diseases, Rutgers University, Newark, NJ, United States
| | - Kazi Sabrina Afreen
- Center for Virus-Host-Innate Immunity and Department of Medicine, Rutgers Biomedical and Health Sciences, Institute for Infectious and Inflammatory Diseases, Rutgers University, Newark, NJ, United States
| | - Abby Odle
- Center for Virus-Host-Innate Immunity and Department of Medicine, Rutgers Biomedical and Health Sciences, Institute for Infectious and Inflammatory Diseases, Rutgers University, Newark, NJ, United States
| | - Ricardo Rajsbaum
- Center for Virus-Host-Innate Immunity and Department of Medicine, Rutgers Biomedical and Health Sciences, Institute for Infectious and Inflammatory Diseases, Rutgers University, Newark, NJ, United States.
| |
Collapse
|
13
|
Agarwal P, Shukla N, Bhatia A, Mahfooz S, Narayan J. Comparative genome analysis reveals driving forces behind Monkeypox virus evolution and sheds light on the role of ATC trinucleotide motif. Virus Evol 2024; 10:veae043. [PMID: 38827420 PMCID: PMC11141602 DOI: 10.1093/ve/veae043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/25/2024] [Accepted: 05/17/2024] [Indexed: 06/04/2024] Open
Abstract
Monkeypox (MPOX), a zoonotic disease originating in Western and Central Africa in 1970, has seen a recent surge in outbreaks across 100+ countries. A comparative analysis of 404 Monkeypox virus (MPXV) genomes revealed notable changes in microsatellite abundance and density, especially within Clades I, IIa, and IIb. Each clade exhibited unique microsatellite motifs, with twenty-six conserved loci specific to MPXV, suggesting their potential as molecular markers in diagnostics. Additionally, nine genes in the MPXV genome featured ten variable hotspot microsatellite regions associated with surface protein synthesis and host control. Notably, gene OPG153, especially at the SSR locus '(ATC)n', exhibited the most pronounced variations among lineages over time and plays a role in virus pathogenesis within the host cell. These findings not only enhance our understanding of MPXV unique molecular profile but also offer valuable insights into potential pathogenic and evolutionary implications.
Collapse
Affiliation(s)
- Preeti Agarwal
- Bioinformatics and Big Data, CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi 110007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Nityendra Shukla
- Bioinformatics and Big Data, CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi 110007, India
| | - Ajay Bhatia
- Bioinformatics and Big Data, CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi 110007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Sahil Mahfooz
- Department of Industrial Microbiology, Deen Dayal Upadhyaya Gorakhpur University, Civil Lines, Gorakhpur 273009, India
| | - Jitendra Narayan
- Bioinformatics and Big Data, CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi 110007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| |
Collapse
|
14
|
Seo D, Brito Oliveira S, Rex EA, Ye X, Rice LM, da Fonseca FG, Gammon DB. Poxvirus A51R proteins regulate microtubule stability and antagonize a cell-intrinsic antiviral response. Cell Rep 2024; 43:113882. [PMID: 38457341 PMCID: PMC11023057 DOI: 10.1016/j.celrep.2024.113882] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 01/28/2024] [Accepted: 02/13/2024] [Indexed: 03/10/2024] Open
Abstract
Numerous viruses alter host microtubule (MT) networks during infection, but how and why they induce these changes is unclear in many cases. We show that the vaccinia virus (VV)-encoded A51R protein is a MT-associated protein (MAP) that directly binds MTs and stabilizes them by both promoting their growth and preventing their depolymerization. Furthermore, we demonstrate that A51R-MT interactions are conserved across A51R proteins from multiple poxvirus genera, and highly conserved, positively charged residues in A51R proteins mediate these interactions. Strikingly, we find that viruses encoding MT interaction-deficient A51R proteins fail to suppress a reactive oxygen species (ROS)-dependent antiviral response in macrophages that leads to a block in virion morphogenesis. Moreover, A51R-MT interactions are required for VV virulence in mice. Collectively, our data show that poxviral MAP-MT interactions overcome a cell-intrinsic antiviral ROS response in macrophages that would otherwise block virus morphogenesis and replication in animals.
Collapse
Affiliation(s)
- Dahee Seo
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sabrynna Brito Oliveira
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG 31270-901, Brazil
| | - Emily A Rex
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xuecheng Ye
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Luke M Rice
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Flávio Guimarães da Fonseca
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG 31270-901, Brazil
| | - Don B Gammon
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
15
|
Cackett G, Sýkora M, Portugal R, Dulson C, Dixon L, Werner F. Transcription termination and readthrough in African swine fever virus. Front Immunol 2024; 15:1350267. [PMID: 38545109 PMCID: PMC10965686 DOI: 10.3389/fimmu.2024.1350267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 01/30/2024] [Indexed: 04/13/2024] Open
Abstract
Introduction African swine fever virus (ASFV) is a nucleocytoplasmic large DNA virus (NCLDV) that encodes its own host-like RNA polymerase (RNAP) and factors required to produce mature mRNA. The formation of accurate mRNA 3' ends by ASFV RNAP depends on transcription termination, likely enabled by a combination of sequence motifs and transcription factors, although these are poorly understood. The termination of any RNAP is rarely 100% efficient, and the transcriptional "readthrough" at terminators can generate long mRNAs which may interfere with the expression of downstream genes. ASFV transcriptome analyses reveal a landscape of heterogeneous mRNA 3' termini, likely a combination of bona fide termination sites and the result of mRNA degradation and processing. While short-read sequencing (SRS) like 3' RNA-seq indicates an accumulation of mRNA 3' ends at specific sites, it cannot inform about which promoters and transcription start sites (TSSs) directed their synthesis, i.e., information about the complete and unprocessed mRNAs at nucleotide resolution. Methods Here, we report a rigorous analysis of full-length ASFV transcripts using long-read sequencing (LRS). We systematically compared transcription termination sites predicted from SRS 3' RNA-seq with 3' ends mapped by LRS during early and late infection. Results Using in-vitro transcription assays, we show that recombinant ASFV RNAP terminates transcription at polyT stretches in the non-template strand, similar to the archaeal RNAP or eukaryotic RNAPIII, unaided by secondary RNA structures or predicted viral termination factors. Our results cement this T-rich motif (U-rich in the RNA) as a universal transcription termination signal in ASFV. Many genes share the usage of the same terminators, while genes can also use a range of terminators to generate transcript isoforms varying enormously in length. A key factor in the latter phenomenon is the highly abundant terminator readthrough we observed, which is more prevalent during late compared with early infection. Discussion This indicates that ASFV mRNAs under the control of late gene promoters utilize different termination mechanisms and factors to early promoters and/or that cellular factors influence the viral transcriptome landscape differently during the late stages of infection.
Collapse
Affiliation(s)
- Gwenny Cackett
- Institute for Structural and Molecular Biology, University College London, London, United Kingdom
| | - Michal Sýkora
- Institute for Structural and Molecular Biology, University College London, London, United Kingdom
| | | | - Christopher Dulson
- Institute for Structural and Molecular Biology, University College London, London, United Kingdom
| | - Linda Dixon
- Pirbright Institute, Pirbright, Surrey, United Kingdom
| | - Finn Werner
- Institute for Structural and Molecular Biology, University College London, London, United Kingdom
| |
Collapse
|
16
|
Hernandez-Gonzalez M, Calcraft T, Nans A, Rosenthal PB, Way M. Palisade structure in intact vaccinia virions. mBio 2024; 15:e0313423. [PMID: 38171004 PMCID: PMC10865856 DOI: 10.1128/mbio.03134-23] [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] [Received: 11/28/2023] [Accepted: 11/30/2023] [Indexed: 01/05/2024] Open
Abstract
Vaccinia virus assembly in the cytoplasm of infected cells involves the formation of a biconcave viral core inside the maturing viral particle. The boundary of the core is defined by a pseudohexagonal palisade layer, composed of trimers projecting from an inner wall. To understand the assembly of this complex core architecture, we obtained a subnanometer structure of the palisade trimer by cryo-electron tomography and subtomogram averaging of purified intact virions. Using AlphaFold2 structure predictions, we determined that the palisade is formed from trimers of the proteolytically processed form of the viral protein A10. In addition, we found that each A10 protomer associates with an α-helix (residues 24-66) of A4. Cellular localization assays outside the context of infection demonstrate that the A4 N-terminus is necessary and sufficient to interact with A10. The interaction between A4 and A10 provides insights into how the palisade layer might become tightly associated with the viral membrane during virion maturation. Reconstruction of the palisade layer reveals that, despite local hexagonal ordering, the A10/A4 trimers are widely spaced, suggesting that additional components organize the lattice. This spacing would, however, allow the adoption of the characteristic biconcave shape of the viral core. Finally, we also found that the palisade incorporates multiple copies of a hexameric portal structure. We suggest that these portals are formed by E6, a viral protein that is essential for virion assembly and required to release viral mRNA from the core early in infection.IMPORTANCEPoxviruses such as variola virus (smallpox) and monkeypox cause diseases in humans. Other poxviruses, including vaccinia and modified vaccinia Ankara, are used as vaccine vectors. Given their importance, a greater structural understanding of poxvirus virions is needed. We now performed cryo-electron tomography of purified intact vaccinia virions to study the structure of the palisade, a protein lattice that defines the viral core boundary. We identified the main viral proteins that form the palisade and their interaction surfaces and provided new insights into the organization of the viral core.
Collapse
Affiliation(s)
- Miguel Hernandez-Gonzalez
- Cellular Signalling and Cytoskeletal Function Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Thomas Calcraft
- Structural Biology of Cells and Viruses Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Andrea Nans
- Structural Biology Science Technology Platform, The Francis Crick Institute, London, United Kingdom
| | - Peter B. Rosenthal
- Structural Biology of Cells and Viruses Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Michael Way
- Cellular Signalling and Cytoskeletal Function Laboratory, The Francis Crick Institute, London, United Kingdom
- Department of Infectious Disease, Imperial College, London, United Kingdom
| |
Collapse
|
17
|
Schlosser-Perrin L, Holzmuller P, Fernandez B, Miotello G, Dahmani N, Neyret A, Bertagnoli S, Armengaud J, Caufour P. Constitutive proteins of lumpy skin disease virion assessed by next-generation proteomics. J Virol 2023; 97:e0072323. [PMID: 37737587 PMCID: PMC10617387 DOI: 10.1128/jvi.00723-23] [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] [Received: 05/24/2023] [Accepted: 06/10/2023] [Indexed: 09/23/2023] Open
Abstract
IMPORTANCE Lumpy skin disease virus (LSDV) is the causative agent of an economically important cattle disease which is notifiable to the World Organisation for Animal Health. Over the past decades, the disease has spread at an alarming rate throughout the African continent, the Middle East, Eastern Europe, the Russian Federation, and many Asian countries. While multiple LDSV whole genomes have made further genetic comparative analyses possible, knowledge on the protein composition of the LSDV particle remains lacking. This study provides for the first time a comprehensive proteomic analysis of an infectious LSDV particle, prompting new efforts toward further proteomic LSDV strain characterization. Furthermore, this first incursion within the capripoxvirus proteome represents one of very few proteomic studies beyond the sole Orthopoxvirus genus, for which most of the proteomics studies have been performed. Providing new information about other chordopoxviruses may contribute to shedding new light on protein composition within the Poxviridae family.
Collapse
Affiliation(s)
- Léo Schlosser-Perrin
- UMR ASTRE, CIRAD, INRAE, University of Montpellier (I-MUSE), Montpellier, France
| | - Philippe Holzmuller
- UMR ASTRE, CIRAD, INRAE, University of Montpellier (I-MUSE), Montpellier, France
| | - Bernard Fernandez
- UMR ASTRE, CIRAD, INRAE, University of Montpellier (I-MUSE), Montpellier, France
| | - Guylaine Miotello
- Département Médicaments et Technologies pour la Santé, Université Paris Saclay, CEA, INRAE, Bagnols-sur-Cèze, France
| | - Noureddine Dahmani
- UMR ASTRE, CIRAD, INRAE, University of Montpellier (I-MUSE), Montpellier, France
| | - Aymeric Neyret
- CEMIPAI, University of Montpellier, UAR3725 CNRS, Montpellier, France
| | | | - Jean Armengaud
- Département Médicaments et Technologies pour la Santé, Université Paris Saclay, CEA, INRAE, Bagnols-sur-Cèze, France
| | - Philippe Caufour
- UMR ASTRE, CIRAD, INRAE, University of Montpellier (I-MUSE), Montpellier, France
| |
Collapse
|
18
|
Deng L, Liu C, Li L, Hao P, Wang M, Jin N, Yin R, Du S, Li C. Genomic characteristics of an avipoxvirus 282E4 strain. Virus Res 2023; 336:199218. [PMID: 37678517 PMCID: PMC10507152 DOI: 10.1016/j.virusres.2023.199218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 09/02/2023] [Accepted: 09/04/2023] [Indexed: 09/09/2023]
Abstract
Avipoxvirus 282E4 strain was extensively applied into recombinant vaccine vector to prevent other infectious diseases. However, little information on the genomic background, functional and genetic evolutionary of the isolate 282E4 strain was clarified. The results showed that the linear genome of avipoxvirus 282E4 was 308,826 bp, containing 313 open reading frames (ORFs) and 12 new predicted ORFs. The 282E4 strain appears to encode two novel thymidine kinase proteins and two TGF-beta-like proteins that may be associated with the suppression of the host's antiviral response. Avipoxvirus 282E4 also encodes 57 ankyrin repeat proteins and 5 variola B22R-like proteins, which composed 7% of the avipoxvirus 282E4 genome. GO and KEGG analysis further revealed that 12 ORFs participate in viral transcription process, 7 ORFs may function during DNA repair, replication and biological synthesis, and ORF 208 is involved in the process of virus life cycle. Interestingly, phylogenetic analysis based on concatenated sequences p4b and DNA polymerase of avipoxviruses gene demonstrates that avipoxvirus 282E4 strain is divergent from known FWPV isolates and is similar to shearwater poxvirus (SWPV-1) that belongs to the CNPV-like virus. Sequencing avipoxvirus 282E4 is a significant step to judge the genetic position of avipoxviruses within the larger Poxviridae phylogenetic tree and provide a new insight into the genetic background of avipoxvirus 282E4 and interspecies transmission of poxviruses, meanwhile, explanation of gene function provides theoretical foundation for vaccine design with 282E4 strain as skeleton.
Collapse
Affiliation(s)
- Lingcong Deng
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Institute of Veterinary Medicine, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Cunxia Liu
- Institute of Poultry Science, Shandong Academy of Agricultural Sciences/Shandong Provincial Key Laboratory of Immunity and Diagnosis of Poultry Diseases, Jinan, 250100, China
| | - Letian Li
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Institute of Veterinary Medicine, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Pengfei Hao
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Institute of Veterinary Medicine, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Maopeng Wang
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Institute of Veterinary Medicine, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Ningyi Jin
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Institute of Veterinary Medicine, Chinese Academy of Agricultural Sciences, Changchun, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, China
| | - Ronglan Yin
- Academy of Animal Science and Veterinary Medicine in Jilin Province, Changchun, 130062, China.
| | - Shouwen Du
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Institute of Veterinary Medicine, Chinese Academy of Agricultural Sciences, Changchun, China.
| | - Chang Li
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Institute of Veterinary Medicine, Chinese Academy of Agricultural Sciences, Changchun, China.
| |
Collapse
|
19
|
Chen S, Yan Y, Gao L, Gao S, Feng K, Li H, Zhang X, Chen W, Chen F, Xie Q. Proteomic profiling of purified avian leukosis virus subgroup J particles. Vet Microbiol 2023; 284:109821. [PMID: 37536160 DOI: 10.1016/j.vetmic.2023.109821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/16/2023] [Accepted: 06/20/2023] [Indexed: 08/05/2023]
Abstract
While the presence of host cell proteins in virions and their role in viral life cycles have been demonstrated in various viruses, such characteristics have remained largely unknown in avian leukosis virus (ALV). To investigate whether this is the case in ALV, we purified high-integrity and high-purity virions from the avian leukosis virus subgroup J (ALV-J) and subjected them to proteome analysis using nano LC-MS/MS. This analysis identified 53 cellular proteins that are incorporated into mature ALV-J virions, and we verified the reliability of the packaged cellular proteins through subtilisin digestion and immunoblot analysis. Functional annotation revealed the potential functions of these proteins in the viral life cycle and tumorigenesis. Overall, our findings have important implications for understanding the interaction between ALV-J and its host, and provide new insights into the cellular requirements that define ALV-J infection.
Collapse
Affiliation(s)
- Sheng Chen
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, PR China; Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, PR China; South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou 510642, PR China; Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangzhou 510642, Guangdong, PR China
| | - Yiming Yan
- Henry Fok School of Biology and Agriculture, Shaoguan University, Shaoguan 510642, PR China
| | - Liguo Gao
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, PR China
| | - Shuang Gao
- Henry Fok School of Biology and Agriculture, Shaoguan University, Shaoguan 510642, PR China
| | - Keyu Feng
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, PR China; Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, PR China; South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou 510642, PR China
| | - Hongxin Li
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, PR China; Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, PR China; South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou 510642, PR China
| | - Xinheng Zhang
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, PR China; Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, PR China; South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou 510642, PR China
| | - Weiguo Chen
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, PR China; Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, PR China; South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou 510642, PR China
| | - Feng Chen
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou 510642, PR China; Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangzhou 510642, Guangdong, PR China
| | - Qingmei Xie
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, PR China; Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, PR China; South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou 510642, PR China; Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangzhou 510642, Guangdong, PR China.
| |
Collapse
|
20
|
Bécares M, Albert M, Tárrega C, Coloma R, Falqui M, Luhmann EK, Radoshevich L, Guerra S. ISG15 Is Required for the Dissemination of Vaccinia Virus Extracellular Virions. Microbiol Spectr 2023; 11:e0450822. [PMID: 37036376 PMCID: PMC10269806 DOI: 10.1128/spectrum.04508-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 03/15/2023] [Indexed: 04/11/2023] Open
Abstract
Viruses have developed many different strategies to counteract immune responses, and Vaccinia virus (VACV) is one of a kind in this aspect. To ensure an efficient infection, VACV undergoes a complex morphogenetic process resulting in the production of two types of infective virions: intracellular mature virus (MV) and extracellular enveloped virus (EV), whose spread depends on different dissemination mechanisms. MVs disseminate after cell lysis, whereas EVs are released or propelled in actin tails from living cells. Here, we show that ISG15 participates in the control of VACV dissemination. Infection of Isg15-/- mouse embryonic fibroblasts with VACV International Health Department-J (IHD-J) strain resulted in decreased EV production, concomitant with reduced induction of actin tails and the abolition of comet-shaped plaque formation, compared to Isg15+/+ cells. Transmission electron microscopy revealed the accumulation of intracellular virus particles and a decrease in extracellular virus particles in the absence of interferon-stimulated gene 15 (ISG15), a finding consistent with altered virus egress. Immunoblot and quantitative proteomic analysis of sucrose gradient-purified virions from both genotypes reported differences in protein levels and composition of viral proteins present on virions, suggesting an ISG15-mediated control of viral proteome. Lastly, the generation of a recombinant IHD-J expressing V5-tagged ISG15 (IHD-J-ISG15) allowed us to identify several viral proteins as potential ISG15 targets, highlighting the proteins A34 and A36, which are essential for EV formation. Altogether, our results indicate that ISG15 is an important host factor in the regulation of VACV dissemination. IMPORTANCE Viral infections are a constant battle between the virus and the host. While the host's only goal is victory, the main purpose of the virus is to spread and conquer new territories at the expense of the host's resources. Along millions of years of incessant encounters, poxviruses have developed a unique strategy consisting in the production two specialized "troops": intracellular mature virions (MVs) and extracellular virions (EVs). MVs mediate transmission between hosts, and EVs ensure advance on the battlefield mediating the long-range dissemination. The mechanism by which the virus "decides" to shed from the primary site of infection and its significant impact in viral transmission is not yet fully established. Here, we demonstrate that this process is finely regulated by ISG15/ISGylation, an interferon-induced ubiquitin-like protein with broad antiviral activity. Studying the mechanism that viruses use during infection could result in new ways of understanding our perpetual war against disease and how we might win the next great battle.
Collapse
Affiliation(s)
- Martina Bécares
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma de Madrid, Madrid, Spain
| | - Manuel Albert
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma de Madrid, Madrid, Spain
| | - Céline Tárrega
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma de Madrid, Madrid, Spain
| | - Rocío Coloma
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma de Madrid, Madrid, Spain
| | - Michela Falqui
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma de Madrid, Madrid, Spain
| | - Emma K. Luhmann
- Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Lilliana Radoshevich
- Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Susana Guerra
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
21
|
Patel CN, Mall R, Bensmail H. AI-driven drug repurposing and binding pose meta dynamics identifies novel targets for Monkeypox virus. J Infect Public Health 2023; 16:799-807. [PMID: 36966703 PMCID: PMC10014505 DOI: 10.1016/j.jiph.2023.03.007] [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: 09/20/2022] [Revised: 02/28/2023] [Accepted: 03/05/2023] [Indexed: 03/17/2023] Open
Abstract
Monkeypox virus (MPXV) was confirmed in May 2022 and designated a global health emergency by WHO in July 2022. MPX virions are big, enclosed, brick-shaped, and contain a linear, double-stranded DNA genome as well as enzymes. MPXV particles bind to the host cell membrane via a variety of viral-host protein interactions. As a result, the wrapped structure is a potential therapeutic target. DeepRepurpose, an artificial intelligence-based compound-viral proteins interaction framework, was used via a transfer learning setting to prioritize a set of FDA approved and investigational drugs which can potentially inhibit MPXV viral proteins. To filter and narrow down the lead compounds from curated collections of pharmaceutical compounds, we used a rigorous computational framework that included homology modeling, molecular docking, dynamic simulations, binding free energy calculations, and binding pose metadynamics. We identified Elvitegravir as a potential inhibitor of MPXV virus using our comprehensive pipeline.
Collapse
Affiliation(s)
- Chirag N. Patel
- Department of Botany, Bioinformatics and Climate Change Impacts Management, School of Science, Gujarat University, Ahmedabad-380009, India,Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institute of Health, Frederick, MD-21702, USA
| | - Raghvendra Mall
- Department of Immunology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee-38105, USA,Biotechnology Research Center, Technology Innovation Institute, Abu Dhabi-9639, United Arab Emirates,Corresponding author at: Department of Immunology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee-38105, USA
| | - Halima Bensmail
- Qatar Computing Research Institute, Hamad Bin Khalifa University, Doha-34110, Qatar,Corresponding author
| |
Collapse
|
22
|
Rampogu S, Kim Y, Kim SW, Lee KW. An overview on monkeypox virus: Pathogenesis, transmission, host interaction and therapeutics. Front Cell Infect Microbiol 2023; 13:1076251. [PMID: 36844409 PMCID: PMC9950268 DOI: 10.3389/fcimb.2023.1076251] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 01/10/2023] [Indexed: 02/12/2023] Open
Abstract
Orthopoxvirus is one of the most notorious genus amongst the Poxviridae family. Monkeypox (MP) is a zoonotic disease that has been spreading throughout Africa. The spread is global, and incidence rates are increasing daily. The spread of the virus is rapid due to human-to-human and animals-to-human transmission. World Health Organization (WHO) has declared monkeypox virus (MPV) as a global health emergency. Since treatment options are limited, it is essential to know the modes of transmission and symptoms to stop disease spread. The information from host-virus interactions revealed significantly expressed genes that are important for the progression of the MP infection. In this review, we highlighted the MP virus structure, transmission modes, and available therapeutic options. Furthermore, this review provides insights for the scientific community to extend their research work in this field.
Collapse
Affiliation(s)
- Shailima Rampogu
- Department of Bio & Medical Big Data (BK4 Program), Division of Life Sciences, Research Institute of Natural Science (RINS), Gyeongsang National University (GNU), Jinju, Republic of Korea
| | - Yongseong Kim
- Department of Pharmaceutical Engineering, Kyungnam University, Changwon, Republic of Korea
| | - Seon-Won Kim
- Division of Applied Life Science (BK21 Four), ABC-RLRC, PMBBRC, Gyeongsang National University, Jinju, Republic of Korea
| | - Keun Woo Lee
- Department of Bio & Medical Big Data (BK4 Program), Division of Life Sciences, Research Institute of Natural Science (RINS), Gyeongsang National University (GNU), Jinju, Republic of Korea
| |
Collapse
|
23
|
Lee N, Jeon YH, Yoo J, Shin SK, Lee S, Park MJ, Jung BJ, Hong YK, Lee DS, Oh K. Generation of novel oncolytic vaccinia virus with improved intravenous efficacy through protection against complement-mediated lysis and evasion of neutralization by vaccinia virus-specific antibodies. J Immunother Cancer 2023; 11:jitc-2022-006024. [PMID: 36717184 PMCID: PMC9887704 DOI: 10.1136/jitc-2022-006024] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/10/2023] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Oncolytic virus immunotherapy has revolutionized cancer immunotherapy by efficiently inducing both oncolysis and systemic immune activation. Locoregional administration has been used for oncolytic virus therapy, but its applications to deep-seated cancers have been limited. Although systemic delivery of the oncolytic virus would maximize viral immunotherapy's potential, this remains a hurdle due to the rapid removal of the administered virus by the complement and innate immune system. Infected cells produce some vaccinia viruses as extracellular enveloped virions, which evade complement attack and achieve longer survival by expressing host complement regulatory proteins (CRPs) on the host-derived envelope. Here, we generated SJ-600 series oncolytic vaccinia viruses that can mimic complement-resistant extracellular enveloped virions by incorporating human CRP CD55 on the intracellular mature virion (IMV) membrane. METHODS The N-terminus of the human CD55 protein was fused to the transmembrane domains of the six type I membrane proteins of the IMV; the resulting recombinant viruses were named SJ-600 series viruses. The SJ-600 series viruses also expressed human granulocyte-macrophage colony-stimulating factor (GM-CSF) to activate dendritic cells. The viral thymidine kinase (J2R) gene was replaced by genes encoding the CD55 fusion proteins and GM-CSF. RESULTS SJ-600 series viruses expressing human CD55 on the IMV membrane showed resistance to serum virus neutralization. SJ-607 virus, which showed the highest CD55 expression and the highest resistance to serum complement-mediated lysis, exhibited superior anticancer activity in three human cancer xenograft models, compared with the control Pexa-Vec (JX-594) virus, after single-dose intravenous administration. The SJ-607 virus administration elicited neutralizing antibody formation in two immunocompetent mouse strains like the control JX-594 virus. Remarkably, we found that the SJ-607 virus evades neutralization by vaccinia virus-specific antibodies. CONCLUSION Our new oncolytic vaccinia virus platform, which expresses human CD55 protein on its membrane, prolonged viral survival by protecting against complement-mediated lysis and by evading neutralization by vaccinia virus-specific antibodies; this may provide a continuous antitumor efficacy until a complete remission has been achieved. Such a platform may expand the target cancer profile to include deep-seated cancers and widespread metastatic cancers.
Collapse
Affiliation(s)
- Namhee Lee
- Research Center, SillaJen, Inc, Seongnam, Gyeonggi-do, Republic of Korea
| | - Yun-Hui Jeon
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea,Wide River Institute of Immunology, Seoul National University, Gangwon, Republic of Korea
| | - Jiyoon Yoo
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea,Wide River Institute of Immunology, Seoul National University, Gangwon, Republic of Korea
| | - Suk-kyung Shin
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea,Wide River Institute of Immunology, Seoul National University, Gangwon, Republic of Korea
| | - Songyi Lee
- Research Center, SillaJen, Inc, Seongnam, Gyeonggi-do, Republic of Korea
| | - Mi-Ju Park
- Research Center, SillaJen, Inc, Seongnam, Gyeonggi-do, Republic of Korea
| | - Byung-Jin Jung
- Research Center, SillaJen, Inc, Seongnam, Gyeonggi-do, Republic of Korea
| | - Yun-Kyoung Hong
- Research Center, SillaJen, Inc, Seongnam, Gyeonggi-do, Republic of Korea
| | - Dong-Sup Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea,Wide River Institute of Immunology, Seoul National University, Gangwon, Republic of Korea
| | - Keunhee Oh
- Research Center, SillaJen, Inc, Seongnam, Gyeonggi-do, Republic of Korea
| |
Collapse
|
24
|
Basu R, Moles CM. Rational selection of an ideal oncolytic virus to address current limitations in clinical translation. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023. [PMID: 37541726 DOI: 10.1016/bs.ircmb.2023.03.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
Abstract
Oncolytic virus therapy (OVT) is a promising modality that leverages the propensity of natural or engineered viruses to selectively replicate in and kill cancer cells. Over the past decade, (pre)clinical studies have focused on the development and testing of adenovirus, herpes simplex virus, and vaccinia virus-based vectors. These studies have identified barriers to success confronting the field. Here, we propose a set of selection criteria or ideal properties of a successful oncolytic virus, which include lack of pathogenicity, low seroprevalence, selectivity (infection and replication), transgene carrying capacity, and genome stability. We use these requirements to analyze the oncolytic virus landscape, and then identify a potentially optimal species for platform development - vesicular stomatitis virus.
Collapse
|
25
|
Poxviral ANKR/F-box Proteins: Substrate Adapters for Ubiquitylation and More. Pathogens 2022; 11:pathogens11080875. [PMID: 36014996 PMCID: PMC9414399 DOI: 10.3390/pathogens11080875] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 02/01/2023] Open
Abstract
Poxviruses are double-stranded DNA viruses that infect insects and a variety of vertebrate species. The large genomes of poxviruses contain numerous genes that allow these viruses to successfully establish infection, including those that help evade the host immune response and prevent cell death. Ankyrin-repeat (ANKR)/F-box proteins are almost exclusively found in poxviruses, and they function as substrate adapters for Skp1-Cullin-1-F-box protein (SCF) multi-subunit E3 ubiquitin (Ub)-ligases. In this regard, they use their C-terminal F-box domain to bind Skp1, Cullin-1, and Roc1 to recruit cellular E2 enzymes to facilitate the ubiquitylation, and subsequent proteasomal degradation, of proteins bound to their N-terminal ANKRs. However, these proteins do not just function as substrate adapters as they also have Ub-independent activities. In this review, we examine both Ub-dependent and -independent activities of ANKR/F-box proteins and discuss how poxviruses use these proteins to counteract the host innate immune response, uncoat their genome, replicate, block cell death, and influence transcription. Finally, we consider important outstanding questions that need to be answered in order to better understand the function of this versatile protein family.
Collapse
|
26
|
Bidgood SR, Samolej J, Novy K, Collopy A, Albrecht D, Krause M, Burden JJ, Wollscheid B, Mercer J. Poxviruses package viral redox proteins in lateral bodies and modulate the host oxidative response. PLoS Pathog 2022; 18:e1010614. [PMID: 35834477 PMCID: PMC9282662 DOI: 10.1371/journal.ppat.1010614] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 05/24/2022] [Indexed: 01/23/2023] Open
Abstract
All poxviruses contain a set of proteinaceous structures termed lateral bodies (LB) that deliver viral effector proteins into the host cytosol during virus entry. To date, the spatial proteotype of LBs remains unknown. Using the prototypic poxvirus, vaccinia virus (VACV), we employed a quantitative comparative mass spectrometry strategy to determine the poxvirus LB proteome. We identified a large population of candidate cellular proteins, the majority being mitochondrial, and 15 candidate viral LB proteins. Strikingly, one-third of these are VACV redox proteins whose LB residency could be confirmed using super-resolution microscopy. We show that VACV infection exerts an anti-oxidative effect on host cells and that artificial induction of oxidative stress impacts early and late gene expression as well as virion production. Using targeted repression and/or deletion viruses we found that deletion of individual LB-redox proteins was insufficient for host redox modulation suggesting there may be functional redundancy. In addition to defining the spatial proteotype of VACV LBs, these findings implicate poxvirus redox proteins as potential modulators of host oxidative anti-viral responses and provide a solid starting point for future investigations into the role of LB resident proteins in host immunomodulation.
Collapse
Affiliation(s)
- Susanna R. Bidgood
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Jerzy Samolej
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, United Kingdom
| | - Karel Novy
- Swiss Federal Institute of Technology (ETH Zürich), Department of Health Sciences and Technology (D-HEST), Institute of Translational Medicine (ITM), Zürich, Switzerland
| | - Abigail Collopy
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - David Albrecht
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Melanie Krause
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Jemima J. Burden
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Bernd Wollscheid
- Swiss Federal Institute of Technology (ETH Zürich), Department of Health Sciences and Technology (D-HEST), Institute of Translational Medicine (ITM), Zürich, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Jason Mercer
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
27
|
Depierreux DM, Altenburg AF, Soday L, Fletcher-Etherington A, Antrobus R, Ferguson BJ, Weekes MP, Smith GL. Selective modulation of cell surface proteins during vaccinia infection: A resource for identifying viral immune evasion strategies. PLoS Pathog 2022; 18:e1010612. [PMID: 35727847 PMCID: PMC9307158 DOI: 10.1371/journal.ppat.1010612] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 07/22/2022] [Accepted: 05/23/2022] [Indexed: 11/24/2022] Open
Abstract
The interaction between immune cells and virus-infected targets involves multiple plasma membrane (PM) proteins. A systematic study of PM protein modulation by vaccinia virus (VACV), the paradigm of host regulation, has the potential to reveal not only novel viral immune evasion mechanisms, but also novel factors critical in host immunity. Here, >1000 PM proteins were quantified throughout VACV infection, revealing selective downregulation of known T and NK cell ligands including HLA-C, downregulation of cytokine receptors including IFNAR2, IL-6ST and IL-10RB, and rapid inhibition of expression of certain protocadherins and ephrins, candidate activating immune ligands. Downregulation of most PM proteins occurred via a proteasome-independent mechanism. Upregulated proteins included a decoy receptor for TRAIL. Twenty VACV-encoded PM proteins were identified, of which five were not recognised previously as such. Collectively, this dataset constitutes a valuable resource for future studies on antiviral immunity, host-pathogen interaction, poxvirus biology, vector-based vaccine design and oncolytic therapy. Vaccinia virus (VACV) is the vaccine used to eradicate smallpox and an excellent model for studying host-pathogen interactions. Many VACV-mediated immune evasion strategies are known, however how immune cells recognise VACV-infected cells is incompletely understood because of the complexity of surface proteins regulating such interactions. Here, a systematic study of proteins on the cell surface at different times during infection with VACV is presented. This shows not only the precise nature and kinetics of appearance of VACV proteins, but also the selective alteration of cellular surface proteins. The latter thereby identified potential novel immune evasion strategies and host proteins regulating immune activation. Comprehensive comparisons with published datasets provided further insight into mechanisms used to regulate surface protein expression. Such comparisons also identified proteins that are targeted by both VACV and human cytomegalovirus (HCMV), and which are therefore likely to represent host proteins regulating immune recognition and activation. Collectively, this work provides a valuable resource for studying viral immune evasion mechanisms and novel host proteins critical in host immunity.
Collapse
Affiliation(s)
| | | | - Lior Soday
- Cambridge Institute for Medical Research, University of Cambridge, United Kingdom
| | | | - Robin Antrobus
- Cambridge Institute for Medical Research, University of Cambridge, United Kingdom
| | | | - Michael P. Weekes
- Cambridge Institute for Medical Research, University of Cambridge, United Kingdom
- * E-mail: (MPW); (GLS)
| | - Geoffrey L. Smith
- Department of Pathology, University of Cambridge, United Kingdom
- * E-mail: (MPW); (GLS)
| |
Collapse
|
28
|
Fischer U, Bartuli J, Grimm C. Structure and function of the poxvirus transcription machinery. Enzymes 2021; 50:1-20. [PMID: 34861934 DOI: 10.1016/bs.enz.2021.06.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Members of the Poxviridae family are large double-stranded DNA viruses that replicate exclusively in the cytoplasm of their hosts. This goes in hand with a high level of independence from the host cell, which supports transcription and replication events only in the nucleus or in DNA-containing organelles. Consequently, virus specific, rather than cellular enzymes mediate most processes involving DNA replication and mRNA synthesis. Recent technological advances allowed a detailed functional and structural investigation of the transcription machinery of the prototypic poxvirus vaccinia. The DNA-dependent RNA polymerase (RNAP) at its core displays distinct similarities to eukaryotic RNAPs. Strong idiosyncrasies, however, are apparent for viral factors that are associated with the viral RNAP during mRNA production. We expect that future studies will unravel more key aspects of poxvirus gene expression, helping also the understanding of nuclear transcription mechanisms.
Collapse
Affiliation(s)
- Utz Fischer
- Department of Biochemistry and Cancer Therapy Research Center (CTRC), Theodor Boveri-Institute, University of Würzburg, Würzburg, Germany; Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Würzburg, Germany
| | - Julia Bartuli
- Department of Biochemistry and Cancer Therapy Research Center (CTRC), Theodor Boveri-Institute, University of Würzburg, Würzburg, Germany
| | - Clemens Grimm
- Department of Biochemistry and Cancer Therapy Research Center (CTRC), Theodor Boveri-Institute, University of Würzburg, Würzburg, Germany.
| |
Collapse
|
29
|
Abstract
The chikungunya virus has spread globally with a remarkably high attack rate. Infection causes arthralgic sequelae that can last for years. Nevertheless, there are no specific drugs or vaccines to contain the virus. Understanding the biology of the virus, such as its replication cycle, is a powerful tool to identify new drugs and comprehend virus-host interactions. Even though the chikungunya virus has been known for a long time (first described in 1952), many aspects of the replication cycle remain unclear. Furthermore, part of the cycle is based on observations of other alphaviruses. In this study, we used electron and scanning microscopy, as well as biological assays, to analyze and investigate the stages of the chikungunya virus replication cycle. Based on our data, we found other infection cellular activities than those usually described for the chikungunya virus replication cycle, i.e. we show particles enveloping intracellularly without budding in a membrane-delimited morphogenesis area; and we also observed virion release by membrane protrusions. Our work provides novel details regarding the biology of chikungunya virus and fills gaps in our knowledge of its replication cycle. These findings may contribute to a better understanding of virus-host interactions and support the development of antivirals. IMPORTANCE The understanding of virus biology is essential to containing virus dissemination, and exploring the virus replication cycle is a powerful tool to do this. There are many points in the biology of the chikungunya virus that need to be clarified, especially regarding its replication cycle. Our incomplete understanding of chikungunya virus infection stages is based on studies with other alphaviruses. We systematized the chikungunya virus replication cycle using microscopic imaging in the order of infection stages: entry, replication, protein synthesis, assembly/morphogenesis, and release. The imaging evidence shows novel points in the replication cycle of enveloping without budding, as well as particle release by cell membrane protrusion.
Collapse
|
30
|
Lant S, Maluquer de Motes C. Poxvirus Interactions with the Host Ubiquitin System. Pathogens 2021; 10:pathogens10081034. [PMID: 34451498 PMCID: PMC8399815 DOI: 10.3390/pathogens10081034] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/12/2021] [Accepted: 08/13/2021] [Indexed: 12/16/2022] Open
Abstract
The ubiquitin system has emerged as a master regulator of many, if not all, cellular functions. With its large repertoire of conjugating and ligating enzymes, the ubiquitin system holds a unique mechanism to provide selectivity and specificity in manipulating protein function. As intracellular parasites viruses have evolved to modulate the cellular environment to facilitate replication and subvert antiviral responses. Poxviruses are a large family of dsDNA viruses with large coding capacity that is used to synthetise proteins and enzymes needed for replication and morphogenesis as well as suppression of host responses. This review summarises our current knowledge on how poxvirus functions rely on the cellular ubiquitin system, and how poxviruses exploit this system to their own advantage, either facilitating uncoating and genome release and replication or rewiring ubiquitin ligases to downregulate critical antiviral factors. Whilst much remains to be known about the intricate interactions established between poxviruses and the host ubiquitin system, our knowledge has revealed crucial viral processes and important restriction factors that open novel avenues for antiviral treatment and provide fundamental insights on the biology of poxviruses and other virus families.
Collapse
|
31
|
Wood JJ, White IJ, Samolej J, Mercer J. Acrylamide inhibits vaccinia virus through vimentin-independent anti-viral granule formation. Cell Microbiol 2021; 23:e13334. [PMID: 33792166 PMCID: PMC11478914 DOI: 10.1111/cmi.13334] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 03/23/2021] [Accepted: 03/29/2021] [Indexed: 11/28/2022]
Abstract
The replication and assembly of vaccinia virus (VACV), the prototypic poxvirus, occurs exclusively in the cytoplasm of host cells. While the role of cellular cytoskeletal components in these processes remains poorly understood, vimentin-a type III intermediate filament-has been shown to associate with viral replication sites and to be incorporated into mature VACV virions. Here, we employed chemical and genetic approaches to further investigate the role of vimentin during the VACV lifecycle. The collapse of vimentin filaments, using acrylamide, was found to inhibit VACV infection at the level of genome replication, intermediate- and late-gene expression. However, we found that CRISPR-mediated knockout of vimentin did not impact VACV replication. Combining these tools, we demonstrate that acrylamide treatment results in the formation of anti-viral granules (AVGs) known to mediate translational inhibition of many viruses. We conclude that vimentin is dispensable for poxvirus replication and assembly and that acrylamide, as a potent inducer of AVGs during VACV infection, serves to bolster cell's anti-viral response to poxvirus infection.
Collapse
Affiliation(s)
- Jennifer J. Wood
- MRC Laboratory for Molecular Cell Biology, University College LondonLondonUK
| | - Ian J. White
- MRC Laboratory for Molecular Cell Biology, University College LondonLondonUK
| | - Jerzy Samolej
- Institute of Microbiology and Infection, University of BirminghamBirminghamUK
| | - Jason Mercer
- MRC Laboratory for Molecular Cell Biology, University College LondonLondonUK
- Institute of Microbiology and Infection, University of BirminghamBirminghamUK
| |
Collapse
|
32
|
Narita F, Wang Z, Kurita H, Li Z, Shi Y, Jia Y, Soutis C. A Review of Piezoelectric and Magnetostrictive Biosensor Materials for Detection of COVID-19 and Other Viruses. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2005448. [PMID: 33230875 PMCID: PMC7744850 DOI: 10.1002/adma.202005448] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/19/2020] [Indexed: 05/19/2023]
Abstract
The spread of the severe acute respiratory syndrome coronavirus has changed the lives of people around the world with a huge impact on economies and societies. The development of wearable sensors that can continuously monitor the environment for viruses may become an important research area. Here, the state of the art of research on biosensor materials for virus detection is reviewed. A general description of the principles for virus detection is included, along with a critique of the experimental work dedicated to various virus sensors, and a summary of their detection limitations. The piezoelectric sensors used for the detection of human papilloma, vaccinia, dengue, Ebola, influenza A, human immunodeficiency, and hepatitis B viruses are examined in the first section; then the second part deals with magnetostrictive sensors for the detection of bacterial spores, proteins, and classical swine fever. In addition, progress related to early detection of COVID-19 (coronavirus disease 2019) is discussed in the final section, where remaining challenges in the field are also identified. It is believed that this review will guide material researchers in their future work of developing smart biosensors, which can further improve detection sensitivity in monitoring currently known and future virus threats.
Collapse
Affiliation(s)
- Fumio Narita
- Department of Frontier Sciences for Advanced EnvironmentGraduate School of Environmental StudiesTohoku UniversityAoba‐yama 6‐6‐02Sendai980‐8579Japan
| | - Zhenjin Wang
- Department of Materials ProcessingGraduate School of EngineeringTohoku UniversityAoba‐yama 6‐6‐02Sendai980‐8579Japan
| | - Hiroki Kurita
- Department of Frontier Sciences for Advanced EnvironmentGraduate School of Environmental StudiesTohoku UniversityAoba‐yama 6‐6‐02Sendai980‐8579Japan
| | - Zhen Li
- College of Automation EngineeringNanjing University of Aeronautics and Astronautics29 Jiangjun AvenueNanjing211106China
| | - Yu Shi
- Department of Mechanical EngineeringUniversity of ChesterThornton Science Park, Pool LaneChesterCH2 4NUUK
| | - Yu Jia
- School of Engineering and Applied ScienceAston UniversityBirminghamB4 7ETUK
| | - Constantinos Soutis
- Aerospace Research InstituteThe University of ManchesterOxford RoadManchesterM13 9PLUK
| |
Collapse
|
33
|
El-Jesr M, Teir M, Maluquer de Motes C. Vaccinia Virus Activation and Antagonism of Cytosolic DNA Sensing. Front Immunol 2020; 11:568412. [PMID: 33117352 PMCID: PMC7559579 DOI: 10.3389/fimmu.2020.568412] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 08/24/2020] [Indexed: 12/16/2022] Open
Abstract
Cells express multiple molecules aimed at detecting incoming virus and infection. Recognition of virus infection leads to the production of cytokines, chemokines and restriction factors that limit virus replication and activate an adaptive immune response offering long-term protection. Recognition of cytosolic DNA has become a central immune sensing mechanism involved in infection, autoinflammation, and cancer immunotherapy. Vaccinia virus (VACV) is the prototypic member of the family Poxviridae and the vaccine used to eradicate smallpox. VACV harbors enormous potential as a vaccine vector and several attenuated strains are currently being developed against infectious diseases. In addition, VACV has emerged as a popular oncolytic agent due to its cytotoxic capacity even in hypoxic environments. As a poxvirus, VACV is an unusual virus that replicates its large DNA genome exclusively in the cytoplasm of infected cells. Despite producing large amounts of cytosolic DNA, VACV efficiently suppresses the subsequent innate immune response by deploying an arsenal of proteins with capacity to disable host antiviral signaling, some of which specifically target cytosolic DNA sensing pathways. Some of these strategies are conserved amongst orthopoxviruses, whereas others are seemingly unique to VACV. In this review we provide an overview of the VACV replicative cycle and discuss the recent advances on our understanding of how VACV induces and antagonizes innate immune activation via cytosolic DNA sensing pathways. The implications of these findings in the rational design of vaccines and oncolytics based on VACV are also discussed.
Collapse
Affiliation(s)
- Misbah El-Jesr
- Department of Microbial Sciences, University of Surrey, Guildford, United Kingdom
| | - Muad Teir
- Department of Microbial Sciences, University of Surrey, Guildford, United Kingdom
| | | |
Collapse
|
34
|
Croft S, Wong YC, Smith SA, Flesch IEA, Tscharke DC. Surprisingly Effective Priming of CD8 + T Cells by Heat-Inactivated Vaccinia Virus Virions. J Virol 2020; 94:e01486-20. [PMID: 32759313 PMCID: PMC7527048 DOI: 10.1128/jvi.01486-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 07/27/2020] [Indexed: 12/29/2022] Open
Abstract
Robust priming of CD8+ T cells by viruses is considered to require infection and de novo expression of viral antigens. A corollary of this is that inactivated viruses are thought of as being inevitably poor vaccines for eliciting these responses. In contrast to this dogma, we found that some antigens present in vaccinia virus (VACV) virions prime strong CD8+ T cell responses when the virus was rendered noninfectious by heat. More surprisingly, in some cases these responses were similar in magnitude to those primed by infectious virus administered at an equivalent dose. Next, we tested whether this was a special property of particular antigens and their epitopes and found that foreign epitopes tagged onto three different VACV virion proteins were able to elicit CD8+ T cell responses irrespective of whether the virus was viable or heat killed. Further, the polyfunctionality and cytotoxic ability of the CD8+ T cells primed by these VACVs was equivalent irrespective of whether they were administered to mice as inactivated or live viruses. Finally, we used these VACVs in prime-boost combinations of inactivated and live virus and found that priming with dead virus before a live booster was the most immunogenic regime. We conclude that VACV virions can be efficient vectors for targeting antigens to dendritic cells for effective priming of CD8+ T cells, even when rendered noninfectious and speculate that this might also be the case for other viruses.IMPORTANCE The design of viral vectored vaccines is often considered to require a trade-off between efficacy and safety. This is especially the case for vaccines that aim to induce killer (CD8+) T cells, where there is a well-established dogma that links infection in vaccinated individuals with effective induction of immunity. However, we found that some proteins of vaccinia virus generate strong CD8+ T cell responses even when the virus preparation was inactivated by heat prior to administration as a vaccine. We took advantage of this finding by engineering a new vaccine vector virus that could be used as an inactivated vaccine. These results suggest that vaccinia virus may be a more versatile vaccine vector than previously appreciated and that in some instances safety can be prioritized by the complete elimination of viral replication without a proportional loss of immunogenicity.
Collapse
Affiliation(s)
- Sarah Croft
- John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Yik Chun Wong
- John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Stewart A Smith
- John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Inge E A Flesch
- John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - David C Tscharke
- John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| |
Collapse
|
35
|
Flores EB, Bartee MY, Bartee E. Reduced cellular binding affinity has profoundly different impacts on the spread of distinct poxviruses. PLoS One 2020; 15:e0231977. [PMID: 32352982 PMCID: PMC7192435 DOI: 10.1371/journal.pone.0231977] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 04/03/2020] [Indexed: 01/11/2023] Open
Abstract
Poxviruses are large enveloped viruses that replicate exclusively in the cytoplasm. Like all viruses, their replication cycle begins with virion adsorption to the cell surface. Unlike most other viral families, however, no unique poxviral receptor has ever been identified. In the absence of a unique receptor, poxviruses are instead thought to adhere to the cell surface primarily through electrostatic interactions between the positively charged viral envelope proteins and the negatively charged sulfate groups on cellular glycosaminoglycans (GAGs). While these negatively charged GAGs are an integral part of all eukaryotic membranes, their specific expression and sulfation patterns differ between cell types. Critically, while poxviral binding has been extensively studied using virally centered genetic strategies, the impact of cell-intrinsic changes to GAG charge has never been examined. Here we show that loss of heparin sulfation, accomplished by deleting the enzyme N-Deacetylase and N-Sulfotransferase-1 (NDST1) which is essential for GAG sulfation, significantly reduces the binding affinity of both vaccinia and myxoma viruses to the cell surface. Strikingly, however, while this lowered binding affinity inhibits the subsequent spread of myxoma virus, it actually enhances the overall spread of vaccinia by generating more diffuse regions of infection. These data indicate that cell-intrinsic GAG sulfation plays a major role in poxviral infection, however, this role varies significantly between different members of the poxviridae.
Collapse
Affiliation(s)
- Erica B. Flores
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, United States of America
| | - Mee Y. Bartee
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, United States of America
| | - Eric Bartee
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, United States of America
| |
Collapse
|
36
|
O’Connell CM, Jasperse B, Hagen CJ, Titong A, Verardi PH. Replication-inducible vaccinia virus vectors with enhanced safety in vivo. PLoS One 2020; 15:e0230711. [PMID: 32240193 PMCID: PMC7117657 DOI: 10.1371/journal.pone.0230711] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 03/06/2020] [Indexed: 11/18/2022] Open
Abstract
Vaccinia virus (VACV) has been used extensively as the vaccine against smallpox and as a viral vector for the development of recombinant vaccines and cancer therapies. Replication-competent, non-attenuated VACVs induce strong, long-lived humoral and cell-mediated immune responses and can be effective oncolytic vectors. However, complications from uncontrolled VACV replication in vaccinees and their close contacts can be severe, particularly in individuals with predisposing conditions. In an effort to develop replication-competent VACV vectors with improved safety, we placed VACV late genes encoding core or virion morphogenesis proteins under the control of tet operon elements to regulate their expression with tetracycline antibiotics. These replication-inducible VACVs would only express the selected genes in the presence of tetracyclines. VACVs inducibly expressing the A3L or A6L genes replicated indistinguishably from wild-type VACV in the presence of tetracyclines, whereas there was no evidence of replication in the absence of antibiotics. These outcomes were reflected in mice, where the VACV inducibly expressing the A6L gene caused weight loss and mortality equivalent to wild-type VACV in the presence of tetracyclines. In the absence of tetracyclines, mice were protected from weight loss and mortality, and viral replication was not detected. These findings indicate that replication-inducible VACVs based on the conditional expression of the A3L or A6L genes can be used for the development of safer, next-generation live VACV vectors and vaccines. The design allows for administration of replication-inducible VACV in the absence of tetracyclines (as a replication-defective vector) or in the presence of tetracyclines (as a replication-competent vector) with enhanced safety.
Collapse
Affiliation(s)
- Caitlin M. O’Connell
- Department of Pathobiology and Veterinary Science and Center of Excellence for Vaccine Research, College of Agriculture, Health and Natural Resources, University of Connecticut, Storrs, Connecticut, United States of America
| | - Brittany Jasperse
- Department of Pathobiology and Veterinary Science and Center of Excellence for Vaccine Research, College of Agriculture, Health and Natural Resources, University of Connecticut, Storrs, Connecticut, United States of America
| | - Caitlin J. Hagen
- Department of Pathobiology and Veterinary Science and Center of Excellence for Vaccine Research, College of Agriculture, Health and Natural Resources, University of Connecticut, Storrs, Connecticut, United States of America
| | - Allison Titong
- Department of Pathobiology and Veterinary Science and Center of Excellence for Vaccine Research, College of Agriculture, Health and Natural Resources, University of Connecticut, Storrs, Connecticut, United States of America
| | - Paulo H. Verardi
- Department of Pathobiology and Veterinary Science and Center of Excellence for Vaccine Research, College of Agriculture, Health and Natural Resources, University of Connecticut, Storrs, Connecticut, United States of America
| |
Collapse
|
37
|
Repair of a previously uncharacterized second host-range gene contributes to full replication of modified vaccinia virus Ankara (MVA) in human cells. Proc Natl Acad Sci U S A 2020; 117:3759-3767. [PMID: 32019881 DOI: 10.1073/pnas.1921098117] [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] [Indexed: 12/19/2022] Open
Abstract
Modified vaccinia virus Ankara (MVA), a widely used vaccine vector for expression of genes of unrelated pathogens, is safe, immunogenic, and can incorporate large amounts of added DNA. MVA was derived by extensively passaging the chorioallantois vaccinia virus Ankara (CVA) vaccine strain in chicken embryo fibroblasts during which numerous mutations and deletions occurred with loss of replicative ability in most mammalian cells. Restoration of the deleted C12L gene, encoding serine protease inhibitor 1, enhances replication of MVA in human MRC-5 cells but only slightly in other human cells. Here we show that repair of the inactivated C16L/B22R gene of MVA enhances replication in numerous human cell lines. This previously uncharacterized gene is present at both ends of the genome of many orthopoxviruses and is highly conserved in most, including smallpox and monkeypox viruses. The C16L/B22R gene is expressed early in infection from the second methionine of the previously annotated Copenhagen strain open reading frame (ORF) as a 17.4-kDa protein. The C16/B22 and C12 proteins together promote MVA replication in human cells to levels that are comparable to titers in chicken embryo fibroblasts. Both proteins enhance virion assembly, but C16/B22 increases proteolytic processing of core proteins in A549 cells consistent with higher infectious virus titers. Furthermore, human A549 cells expressing codon-optimized C16L/B22R and C12L genes support higher levels of MVA replication than cell lines expressing neither or either alone. Identification of the genes responsible for the host-range defect of MVA may allow more rational engineering of vaccines for efficacy, safety, and propagation.
Collapse
|
38
|
Ni JS, Li Y, Yue W, Liu B, Li K. Nanoparticle-based Cell Trackers for Biomedical Applications. Theranostics 2020; 10:1923-1947. [PMID: 32042345 PMCID: PMC6993224 DOI: 10.7150/thno.39915] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 11/12/2019] [Indexed: 12/11/2022] Open
Abstract
The continuous or real-time tracking of biological processes using biocompatible contrast agents over a certain period of time is vital for precise diagnosis and treatment, such as monitoring tissue regeneration after stem cell transplantation, understanding the genesis, development, invasion and metastasis of cancer and so on. The rationally designed nanoparticles, including aggregation-induced emission (AIE) dots, inorganic quantum dots (QDs), nanodiamonds, superparamagnetic iron oxide nanoparticles (SPIONs), and semiconducting polymer nanoparticles (SPNs), have been explored to meet this urgent need. In this review, the development and application of these nanoparticle-based cell trackers for a variety of imaging technologies, including fluorescence imaging, photoacoustic imaging, magnetic resonance imaging, magnetic particle imaging, positron emission tomography and single photon emission computing tomography are discussed in detail. Moreover, the further therapeutic treatments using multi-functional trackers endowed with photodynamic and photothermal modalities are also introduced to provide a comprehensive perspective in this promising research field.
Collapse
Affiliation(s)
- Jen-Shyang Ni
- Department of Biomedical Engineering, Academy for Advanced Interdisciplinary Studies, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
- HKUST-Shenzhen Research Institute, Shenzhen 518057, China
| | - Yaxi Li
- Department of Biomedical Engineering, Academy for Advanced Interdisciplinary Studies, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Wentong Yue
- Department of Biomedical Engineering, Academy for Advanced Interdisciplinary Studies, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Bin Liu
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Singapore
| | - Kai Li
- Department of Biomedical Engineering, Academy for Advanced Interdisciplinary Studies, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| |
Collapse
|
39
|
Shchelkunov SN, Shchelkunova GA. Genes that Control Vaccinia Virus Immunogenicity. Acta Naturae 2020; 12:33-41. [PMID: 32477596 PMCID: PMC7245956 DOI: 10.32607/actanaturae.10935] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 01/13/2020] [Indexed: 12/23/2022] Open
Abstract
The live smallpox vaccine was a historical first and highly effective vaccine. However, along with high immunogenicity, the vaccinia virus (VACV) caused serious side effects in vaccinees, sometimes with lethal outcomes. Therefore, after global eradication of smallpox, VACV vaccination was stopped. For this reason, most of the human population worldwide lacks specific immunity against not only smallpox, but also other zoonotic orthopoxviruses. Outbreaks of diseases caused by these viruses have increasingly occurred in humans on different continents. However, use of the classical live VACV vaccine for prevention against these diseases is unacceptable because of potential serious side effects, especially in individuals with suppressed immunity or immunodeficiency (e.g., HIV-infected patients). Therefore, highly attenuated VACV variants that preserve their immunogenicity are needed. This review discusses current ideas about the development of a humoral and cellular immune response to orthopoxvirus infection/vaccination and describes genetic engineering approaches that could be utilized to generate safe and highly immunogenic live VACV vaccines.
Collapse
Affiliation(s)
- S. N. Shchelkunov
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, Novosibirsk region, Koltsovo, 630559 Russia
- The Federal Research Center Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences, Novosibirsk, 630090 Russia
- Novosibirsk State University, Novosibirsk, 630090 Russia
| | - G. A. Shchelkunova
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, Novosibirsk region, Koltsovo, 630559 Russia
| |
Collapse
|
40
|
Jordan I, Horn D, Thiele K, Haag L, Fiddeke K, Sandig V. A Deleted Deletion Site in a New Vector Strain and Exceptional Genomic Stability of Plaque-Purified Modified Vaccinia Ankara (MVA). Virol Sin 2019; 35:212-226. [PMID: 31833037 PMCID: PMC7198643 DOI: 10.1007/s12250-019-00176-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 09/18/2019] [Indexed: 12/29/2022] Open
Abstract
Vectored vaccines based on highly attenuated modified vaccinia Ankara (MVA) are reported to be immunogenic, tolerant to pre-existing immunity, and able to accommodate and stably maintain very large transgenes. MVA is usually produced on primary chicken embryo fibroblasts, but production processes based on continuous cell lines emerge as increasingly robust and cost-effective alternatives. An isolate of a hitherto undescribed genotype was recovered by passage of a non-plaque-purified preparation of MVA in a continuous anatine suspension cell line (CR.pIX) in chemically defined medium. The novel isolate (MVA-CR19) replicated to higher infectious titers in the extracellular volume of suspension cultures and induced fewer syncytia in adherent cultures. We now extend previous studies with the investigation of the point mutations in structural genes of MVA-CR19 and describe an additional point mutation in a regulatory gene. We furthermore map and discuss an extensive rearrangement of the left telomer of MVA-CR19 that appears to have occurred by duplication of the right telomer. This event caused deletions and duplications of genes that may modulate immunologic properties of MVA-CR19 as a vaccine vector. Our characterizations also highlight the exceptional genetic stability of plaque-purified MVA: although the phenotype of MVA-CR19 appears to be advantageous for replication, we found that all genetic markers that differentiate wildtype and MVA-CR19 are stably maintained in passages of recombinant viruses based on either wildtype or MVA-CR.
Collapse
Affiliation(s)
- Ingo Jordan
- ProBioGen AG, Herbert-Bayer-Straße 8, 13086, Berlin, Germany.
| | - Deborah Horn
- ProBioGen AG, Herbert-Bayer-Straße 8, 13086, Berlin, Germany
| | - Kristin Thiele
- ProBioGen AG, Herbert-Bayer-Straße 8, 13086, Berlin, Germany.,Sartorius Stedim Cellca GmbH, Erwin-Rentschler-Str 21, 88471, Laupheim, Germany
| | - Lars Haag
- Vironova AB, Gävlegatan 22, 113 30, Stockholm, Sweden.,Department of Laboratory Medicine, Karolinska Universitetsjukhuset i Huddinge, 14152, Huddinge, Sweden
| | | | - Volker Sandig
- ProBioGen AG, Herbert-Bayer-Straße 8, 13086, Berlin, Germany
| |
Collapse
|
41
|
Jayawardena N, Burga LN, Poirier JT, Bostina M. Virus-Receptor Interactions: Structural Insights For Oncolytic Virus Development. Oncolytic Virother 2019; 8:39-56. [PMID: 31754615 PMCID: PMC6825474 DOI: 10.2147/ov.s218494] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 10/02/2019] [Indexed: 12/11/2022] Open
Abstract
Recent advancements in oncolytic virotherapy commend a special attention to developing new strategies for targeting cancer cells with oncolytic viruses (OVs). Modifications of the viral envelope or coat proteins serve as a logical mean of repurposing viruses for cancer treatment. In this review, we discuss how detailed structural knowledge of the interactions between OVs and their natural receptors provide valuable insights into tumor specificity of some viruses and re-targeting of alternate receptors for broad tumor tropism or improved tumor selectivity.
Collapse
Affiliation(s)
- Nadishka Jayawardena
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Laura N Burga
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - John T Poirier
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Mihnea Bostina
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
- Otago Micro and Nano Imaging, University of Otago, Dunedin, New Zealand
| |
Collapse
|
42
|
Mary B, Maurya S, Arumugam S, Kumar V, Jayandharan GR. Post-translational modifications in capsid proteins of recombinant adeno-associated virus (AAV) 1-rh10 serotypes. FEBS J 2019; 286:4964-4981. [PMID: 31330090 PMCID: PMC7496479 DOI: 10.1111/febs.15013] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 05/13/2019] [Accepted: 07/19/2019] [Indexed: 01/27/2023]
Abstract
Post‐translational modifications in viral capsids are known to fine‐tune and regulate several aspects of the infective life cycle of several viruses in the host. Recombinant viruses that are generated in a specific producer cell line are likely to inherit unique post‐translational modifications during intra‐cellular maturation of its capsid proteins. Data on such post‐translational modifications in the capsid of recombinant adeno‐associated virus serotypes (AAV1‐rh10) is limited. We have employed liquid chromatography and mass spectrometry analysis to characterize post‐translational modifications in AAV1‐rh10 capsid protein. Our analysis revealed a total of 52 post‐translational modifications in AAV2‐AAVrh10 capsids, including ubiquitination (17%), glycosylation (36%), phosphorylation (21%), SUMOylation (13%) and acetylation (11%). While AAV1 had no detectable post‐translational modification, at least four AAV serotypes had >7 post‐translational modifications in their capsid protein. About 82% of these post‐translational modifications are novel. A limited validation of AAV2 capsids by MALDI‐TOF and western blot analysis demonstrated minimal glycosylation and ubiquitination of AAV2 capsids. To further validate this, we disrupted a glycosylation site identified in AAV2 capsid (AAV2‐N253Q), which severely compromised its packaging efficiency (~ 100‐fold vs. AAV2 wild‐type vectors). In order to confirm other post‐translational modifications detected such as SUMOylation, mutagenesis of a SUMOylation site(K258Q) in AAV2 was performed. This mutant vector demonstrated reduced levels of SUMO‐1/2/3 proteins and negligible transduction, 2 weeks after ocular gene transfer. Our study underscores the heterogeneity of post‐translational modifications in AAV vectors. The data presented here, should facilitate further studies to understand the biological relevance of post‐translational modifications in AAV life cycle and the development of novel bioengineered AAV vectors for gene therapy applications. Enzymes Trypsin, EC 3.4.21.4
Collapse
Affiliation(s)
- Bertin Mary
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, India
| | - Shubham Maurya
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, India
| | - Sathyathithan Arumugam
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, India.,SASTRA University, Thanjavur, India
| | - Vikas Kumar
- Mass Spectrometry and Proteomics Core Facility, University of Nebraska Medical Center, Omaha, NE, USA
| | - Giridhara R Jayandharan
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, India.,Department of Haematology and Centre for Stem Cell Research, Vellore, India
| |
Collapse
|
43
|
Pant A, Cao S, Yang Z. Asparagine Is a Critical Limiting Metabolite for Vaccinia Virus Protein Synthesis during Glutamine Deprivation. J Virol 2019; 93:e01834-18. [PMID: 30996100 PMCID: PMC6580962 DOI: 10.1128/jvi.01834-18] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 04/07/2019] [Indexed: 12/13/2022] Open
Abstract
Viruses actively interact with host metabolism because viral replication relies on host cells to provide nutrients and energy. Vaccinia virus (VACV; the prototype poxvirus) prefers glutamine to glucose for efficient replication to the extent that VACV replication is hindered in glutamine-free medium. Remarkably, our data show that VACV replication can be fully rescued from glutamine depletion by asparagine supplementation. By global metabolic profiling, as well as genetic and chemical manipulation of the asparagine supply, we provide evidence demonstrating that the production of asparagine, which exclusively requires glutamine for biosynthesis, accounts for VACV's preference of glutamine to glucose rather than glutamine's superiority over glucose in feeding the tricarboxylic acid (TCA) cycle. Furthermore, we show that sufficient asparagine supply is required for efficient VACV protein synthesis. Our study highlights that the asparagine supply, the regulation of which has been evolutionarily tailored in mammalian cells, presents a critical barrier to VACV replication due to a high asparagine content of viral proteins and a rapid demand of viral protein synthesis. The identification of asparagine availability as a critical limiting factor for efficient VACV replication suggests a new direction of antiviral strategy development.IMPORTANCE Viruses rely on their infected host cells to provide nutrients and energy for replication. Vaccinia virus, the prototypic member of the poxviruses, which comprise many significant human and animal pathogens, prefers glutamine to glucose for efficient replication. Here, we show that the preference is not because glutamine is superior to glucose as the carbon source to fuel the tricarboxylic acid cycle for vaccinia virus replication. Rather interestingly, the preference is because the asparagine supply for efficient viral protein synthesis becomes limited in the absence of glutamine, which is necessary for asparagine biosynthesis. We provide further genetic and chemical evidence to demonstrate that asparagine availability plays a critical role in efficient vaccinia virus replication. This discovery identifies a weakness of vaccinia virus and suggests a possible direction to intervene in poxvirus infection.
Collapse
Affiliation(s)
- Anil Pant
- Division of Biology, Kansas State University, Manhattan, Kansas, USA
| | - Shuai Cao
- Division of Biology, Kansas State University, Manhattan, Kansas, USA
| | - Zhilong Yang
- Division of Biology, Kansas State University, Manhattan, Kansas, USA
| |
Collapse
|
44
|
Peroxiredoxin 1, a Novel HBx-Interacting Protein, Interacts with Exosome Component 5 and Negatively Regulates Hepatitis B Virus (HBV) Propagation through Degradation of HBV RNA. J Virol 2019; 93:JVI.02203-18. [PMID: 30567989 DOI: 10.1128/jvi.02203-18] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 12/12/2018] [Indexed: 12/20/2022] Open
Abstract
Hepatitis B virus (HBV) infection is a major risk factor for the development of chronic liver diseases, including cirrhosis and hepatocellular carcinoma (HCC). A growing body of evidence suggests that HBV X protein (HBx) plays a crucial role in viral replication and HCC development. Here, we identified peroxiredoxin 1 (Prdx1), a cellular hydrogen peroxide scavenger, as a novel HBx-interacting protein. Coimmunoprecipitation analysis coupled with site-directed mutagenesis revealed that the region from amino acids 17 to 20 of the HBx, particularly HBx Cys17, is responsible for the interaction with Prdx1. Knockdown of Prdx1 by siRNA significantly increased the levels of intracellular HBV RNA, HBV antigens, and extracellular HBV DNA, whereas knockdown of Prdx1 did not increase the activities of HBV core, enhancer I (Enh1)/X, preS1, and preS2/S promoters. Kinetic analysis of HBV RNA showed that knockdown of Prdx1 inhibited HBV RNA decay, suggesting that Prdx1 reduces HBV RNA levels posttranscriptionally. The RNA coimmunoprecipitation assay revealed that Prdx1 interacted with HBV RNA. The exosome component 5 (Exosc5), a member of the RNA exosome complexes, was coimmunoprecipitated with Prdx1, suggesting its role in regulation of HBV RNA stability. Taken together, these results suggest that Prdx1 and Exosc5 play crucial roles in host defense mechanisms against HBV infection.IMPORTANCE Hepatitis B virus (HBV) infection is a major global health problem. HBx plays important roles in HBV replication and viral carcinogenesis through its interaction with host factors. In this study, we identified Prdx1 as a novel HBx-binding protein. We provide evidence suggesting that Prdx1 promotes HBV RNA decay through interaction with HBV RNA and Exosc5, leading to downregulation of HBV RNA. These results suggest that Prdx1 negatively regulates HBV propagation. Our findings may shed new light on the roles of Prdx1 and Exosc5 in host defense mechanisms in HBV infection.
Collapse
|
45
|
Lorente E, Martín-Galiano AJ, Barnea E, Barriga A, Palomo C, García-Arriaza J, Mir C, Lauzurica P, Esteban M, Admon A, López D. Proteomics Analysis Reveals That Structural Proteins of the Virion Core and Involved in Gene Expression Are the Main Source for HLA Class II Ligands in Vaccinia Virus-Infected Cells. J Proteome Res 2019; 18:900-911. [PMID: 30629447 DOI: 10.1021/acs.jproteome.8b00595] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Protective cellular and humoral immune responses require previous recognition of viral antigenic peptides complexed with human leukocyte antigen (HLA) class II molecules on the surface of the antigen presenting cells. The HLA class II-restricted immune response is important for the control and the clearance of poxvirus infection including vaccinia virus (VACV), the vaccine used in the worldwide eradication of smallpox. In this study, a mass spectrometry analysis was used to identify VACV ligands bound to HLA-DR and -DP class II molecules present on the surface of VACV-infected cells. Twenty-six naturally processed viral ligands among the tens of thousands of cell peptides bound to HLA class II proteins were identified. These viral ligands arose from 19 parental VACV proteins: A4, A5, A18, A35, A38, B5, B13, D1, D5, D7, D12, D13, E3, E8, H5, I2, I3, J2, and K2. The majority of these VACV proteins yielded one HLA ligand and were generated mainly, but not exclusively, by the classical HLA class II antigen processing pathway. Medium-sized and abundant proteins from the virion core and/or involved in the viral gene expression were the major source of VACV ligands bound to HLA-DR and -DP class II molecules. These findings will help to understand the effectiveness of current poxvirus-based vaccines and will be important in the design of new ones.
Collapse
Affiliation(s)
| | | | - Eilon Barnea
- Department of Biology , Technion-Israel Institute of Technology , 32000 Haifa , Israel
| | | | | | - Juan García-Arriaza
- Department of Molecular and Cellular Biology , Centro Nacional de Biotecnología , Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid , Spain
| | | | | | - Mariano Esteban
- Department of Molecular and Cellular Biology , Centro Nacional de Biotecnología , Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid , Spain
| | - Arie Admon
- Department of Biology , Technion-Israel Institute of Technology , 32000 Haifa , Israel
| | | |
Collapse
|
46
|
Burnie J, Guzzo C. The Incorporation of Host Proteins into the External HIV-1 Envelope. Viruses 2019; 11:v11010085. [PMID: 30669528 PMCID: PMC6356245 DOI: 10.3390/v11010085] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 01/15/2019] [Accepted: 01/16/2019] [Indexed: 02/07/2023] Open
Abstract
The incorporation of biologically active host proteins into HIV-1 is a well-established phenomenon, particularly due to the budding mechanism of viral egress in which viruses acquire their external lipid membrane directly from the host cell. While this mechanism might seemingly imply that host protein incorporation is a passive uptake of all cellular antigens associated with the plasma membrane at the site of budding, this is not the case. Herein, we review the evidence indicating that host protein incorporation can be a selective and conserved process. We discuss how HIV-1 virions displaying host proteins on their surface can exhibit a myriad of altered phenotypes, with notable impacts on infectivity, homing, neutralization, and pathogenesis. This review describes the canonical and emerging methods to detect host protein incorporation, highlights the well-established host proteins that have been identified on HIV-1 virions, and reflects on the role of these incorporated proteins in viral pathogenesis and therapeutic targeting. Despite many advances in HIV treatment and prevention, there remains a global effort to develop increasingly effective anti-HIV therapies. Given the broad range of biologically active host proteins acquired on the surface of HIV-1, additional studies on the mechanisms and impacts of these incorporated host proteins may inform the development of novel treatments and vaccine designs.
Collapse
Affiliation(s)
- Jonathan Burnie
- Department of Cell and Systems Biology, University of Toronto, 25 Harbord Street, Toronto, ON M5S 3G5, Canada.
- Department of Biological Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, ON M1C 1A4, Canada.
| | - Christina Guzzo
- Department of Cell and Systems Biology, University of Toronto, 25 Harbord Street, Toronto, ON M5S 3G5, Canada.
- Department of Biological Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, ON M1C 1A4, Canada.
| |
Collapse
|
47
|
Mirzakhanyan Y, Gershon P. The Vaccinia virion: Filling the gap between atomic and ultrastructure. PLoS Pathog 2019; 15:e1007508. [PMID: 30615658 PMCID: PMC6336343 DOI: 10.1371/journal.ppat.1007508] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 01/17/2019] [Accepted: 12/06/2018] [Indexed: 01/19/2023] Open
Abstract
We have investigated the molecular-level structure of the Vaccinia virion in situ by protein-protein chemical crosslinking, identifying 4609 unique-mass crosslink ions at an effective FDR of 0.33%, covering 2534 unique pairs of crosslinked protein positions, 625 of which were inter-protein. The data were statistically non-random and rational in the context of known structures, and showed biological rationality. Crosslink density strongly tracked the individual proteolytic maturation products of p4a and p4b, the two major virion structural proteins, and supported the prediction of transmembrane domains within membrane proteins. A clear sub-network of four virion structural proteins provided structural insights into the virion core wall, and proteins VP8 and A12 formed a strongly-detected crosslinked pair with an apparent structural role. A strongly-detected sub-network of membrane proteins A17, H3, A27 and A26 represented an apparent interface of the early-forming virion envelope with structures added later during virion morphogenesis. Protein H3 seemed to be the central hub not only for this sub-network but also for an 'attachment protein' sub-network comprising membrane proteins H3, ATI, CAHH(D8), A26, A27 and G9. Crosslinking data lent support to a number of known interactions and interactions within known complexes. Evidence is provided for the membrane targeting of genome telomeres. In covering several orders of magnitude in protein abundance, this study may have come close to the bottom of the protein-protein crosslinkome of an intact organism, namely a complex animal virus.
Collapse
Affiliation(s)
- Yeva Mirzakhanyan
- Department of Molecular Biology & Biochemistry, UC-Irvine, Irvine, California, United States of America
| | - Paul Gershon
- Department of Molecular Biology & Biochemistry, UC-Irvine, Irvine, California, United States of America
| |
Collapse
|
48
|
A Proteomic Atlas of the African Swine Fever Virus Particle. J Virol 2018; 92:JVI.01293-18. [PMID: 30185597 DOI: 10.1128/jvi.01293-18] [Citation(s) in RCA: 299] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 08/30/2018] [Indexed: 12/11/2022] Open
Abstract
African swine fever virus (ASFV) is a large and complex DNA virus that causes a highly lethal swine disease for which there is no vaccine available. The ASFV particle, with an icosahedral multilayered structure, contains multiple polypeptides whose identity is largely unknown. Here, we analyzed by mass spectroscopy the protein composition of highly purified extracellular ASFV particles and performed immunoelectron microscopy to localize several of the detected proteins. The proteomic analysis identified 68 viral proteins, which account for 39% of the genome coding capacity. The ASFV proteome includes essentially all the previously described virion proteins and, interestingly, 44 newly identified virus-packaged polypeptides, half of which have an unknown function. A great proportion of the virion proteins are committed to the virus architecture, including two newly identified structural proteins, p5 and p8, which are derived from the core polyproteins pp220 and pp62, respectively. In addition, the virion contains a full complement of enzymes and factors involved in viral transcription, various enzymes implicated in DNA repair and protein modification, and some proteins concerned with virus entry and host defense evasion. Finally, 21 host proteins, many of them localized at the cell surface and related to the cortical actin cytoskeleton, were reproducibly detected in the ASFV particle. Immunoelectron microscopy strongly supports the suggestion that these host membrane-associated proteins are recruited during virus budding at actin-dependent membrane protrusions. Altogether, the results of this study provide a comprehensive model of the ASFV architecture that integrates both compositional and structural information.IMPORTANCE African swine fever virus causes a highly contagious and lethal disease of swine that currently affects many countries of sub-Saharan Africa, the Caucasus, the Russian Federation, and Eastern Europe and has very recently spread to China. Despite extensive research, effective vaccines or antiviral strategies are still lacking, and many basic questions on the molecular mechanisms underlying the infective cycle remain. One such gap regards the composition and structure of the infectious virus particle. In the study described in this report, we identified the set of viral and host proteins that compose the virion and determined or inferred the localization of many of them. This information significantly increases our understanding of the biological and structural features of an infectious African swine fever virus particle and will help direct future research efforts.
Collapse
|
49
|
Gale TV, Horton TM, Hoffmann AR, Branco LM, Garry RF. Host Proteins Identified in Extracellular Viral Particles as Targets for Broad-Spectrum Antiviral Inhibitors. J Proteome Res 2018; 18:7-17. [PMID: 30351952 DOI: 10.1021/acs.jproteome.8b00204] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Liquid chromatography mass spectrometry (LCMS) proteomic analyses have revealed that host proteins are often captured in extracellular virions. These proteins may play a role in viral replication or infectivity and can represent targets for broad-spectrum antiviral agent development. We utilized LCMS to determine the host protein composition of Lassa virus-like particles (LASV VLPs). Multiple host proteins incorporated in LASV VLPs are also incorporated in unrelated viruses, notably ribosomal proteins. We assembled a data set of host proteins incorporated into extracellular viral particles. The frequent incorporation of specific host proteins into viruses of diverse families suggests that interactions of these proteins with viral factors may be important for effective viral replication. Drugs that target virion-associated host proteins could affect the protein in the extracellular virion or the host cell. Compounds that target proteins incorporated into virions with high frequency, but with no known antiviral activity, were assayed in a scalable viral screening platform, and hits were tested in competent viral systems. One of these molecules, GAPDH modulating small molecule CGP 3466B maleate (Omigapil), exhibited a dose-dependent inhibition of HIV, dengue virus, and Zika virus.
Collapse
Affiliation(s)
- Trevor V Gale
- Department of Microbiology and Immunology , Tulane University , New Orleans , Louisiana 70112 , United States
| | - Timothy M Horton
- Department of Microbiology and Immunology , Tulane University , New Orleans , Louisiana 70112 , United States
| | - Andrew R Hoffmann
- Department of Microbiology and Immunology , Tulane University , New Orleans , Louisiana 70112 , United States
| | - Luis M Branco
- Zalgen Laboratories, LLC , Germantown , Maryland 20876 , United States
| | - Robert F Garry
- Department of Microbiology and Immunology , Tulane University , New Orleans , Louisiana 70112 , United States.,Zalgen Laboratories, LLC , Germantown , Maryland 20876 , United States
| |
Collapse
|
50
|
Sviben D, Forcic D, Halassy B, Allmaier G, Marchetti-Deschmann M, Brgles M. Mass spectrometry-based investigation of measles and mumps virus proteome. Virol J 2018; 15:160. [PMID: 30326905 PMCID: PMC6192076 DOI: 10.1186/s12985-018-1073-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Accepted: 10/02/2018] [Indexed: 02/08/2023] Open
Abstract
Background Measles (MEV) and mumps virus (MUV) are enveloped, non-segmented, negative single stranded RNA viruses of the family Paramyxoviridae, and are the cause of measles and mumps, respectively, both preventable by vaccination. Aside from proteins coded by the viral genome, viruses are considered to contain host cell proteins (HCPs). The presence of extracellular vesicles (ECVs), which are often co-purified with viruses due to their similarity in size, density and composition, also contributes to HCPs detected in virus preparations, and this has often been neglected. The aim was to identify which virus-coded proteins are present in MEV and MUV virions, and to try to detect which HCPs, if any, are incorporated inside the virions or adsorbed on their outer surface, and which are more likely to be a contamination from co-purified ECVs. Methods MUV, MEV and ECVs were purified by ultracentrifugation, hydrophobic interaction chromatography and immunoaffinity chromatography, proteins in the samples were resolved by SDS-PAGE and subjected to identification by MALDI-TOF/TOF-MS. A comparative analysis of HCPs present in all samples was carried out. Results By proteomics approach, it was verified that almost all virus-coded proteins are present in MEV and MUV particles. Protein C in MEV which was until now considered to be non-structural viral protein, was found to be present inside the MeV virions. Results on the presence of HCPs in differently purified virus preparations imply that actin, annexins, cyclophilin A, moesin and integrin β1 are part of the virions. Conclusions All HCPs detected in the viruses are present in ECVs as well, indicating their possible function in vesicle formation, or that most of them are only present in ECVs. Only five HCPs were constantly present in purified virus preparations, regardless of the purification method used, implying they are likely the integral part of the virions. The approach described here is helpful for further investigation of HCPs in other virus preparations. Electronic supplementary material The online version of this article (10.1186/s12985-018-1073-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Dora Sviben
- Centre for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Rockefellerova 10, HR-10 000, Zagreb, Croatia. .,Centre of Excellence for Viral Immunology and Vaccines, CERVirVac, Zagreb, Croatia.
| | - Dubravko Forcic
- Centre for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Rockefellerova 10, HR-10 000, Zagreb, Croatia.,Centre of Excellence for Viral Immunology and Vaccines, CERVirVac, Zagreb, Croatia
| | - Beata Halassy
- Centre for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Rockefellerova 10, HR-10 000, Zagreb, Croatia.,Centre of Excellence for Viral Immunology and Vaccines, CERVirVac, Zagreb, Croatia
| | - Günter Allmaier
- Institute of Chemical Technologies and Analytics, TU Wien, Getreidemarkt 9, AT-1060, Vienna, Austria
| | | | - Marija Brgles
- Centre for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Rockefellerova 10, HR-10 000, Zagreb, Croatia.,Centre of Excellence for Viral Immunology and Vaccines, CERVirVac, Zagreb, Croatia
| |
Collapse
|