1
|
Li M, Chen J, Wang F, Kuang J, Peng Y, Asghar S, Zhao W, Yang Y, Shen C. Bispecific antibodies targeting MPXV A29 and B6 demonstrate efficacy against MPXV infection. J Virol 2025; 99:e0232024. [PMID: 40178288 PMCID: PMC12090737 DOI: 10.1128/jvi.02320-24] [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: 01/01/2025] [Accepted: 03/09/2025] [Indexed: 04/05/2025] Open
Abstract
Recently, the monkeypox virus (MPXV) outbreak was once again declared by the World Health Organization as a global health emergency, and currently, there is no specific drug against MPXV. During the replication cycle, MPXV produces two distinct forms of viral particles: extracellular enveloped virus (EEV), released via exocytosis, and intracellular mature virus (IMV), expelled through host cell lysis. A29 and B6 proteins are membrane proteins found on the IMV and EEV viral particles, respectively. This study designed two different bispecific antibodies (bsAbs) targeting specific antigens of the MPXV: the developed bsAb 9F8-3A1 targets two non-competitive binding epitopes on the MPXV protein A29, while bsAb 9F8-7C9 targets different antigen-binding epitopes on both A29 and B6. The in vitro and in vivo characterization assays demonstrated that the bsAbs provided complete protection against three poxvirus strains: vaccinia virus (VACV) Tiantan, VACV Western Reserve (VACV WR), and MPXV, surpassing the efficacy of all the parental monoclonal antibodies. Notably, the bsAb 9F8-7C9 exhibited the most effective antiviral activity. In vivo pharmacokinetic experiments showed that these two bsAbs have long half-lives in rhesus macaques. In conclusion, this study successfully developed two bispecific antibodies that target different epitopes, providing crucial insights for the development of decent antiviral drugs against MPXV and other orthopoxviruses.IMPORTANCEMpox is a viral zoonotic disease caused by MPXV infection. Since 2022, cases of mpox have been reported in non-endemic countries. The number of infections and deaths continues to rise, posing a serious threat to global health and safety. Currently, there are no specific treatments for mpox, making the development of effective therapeutic options urgent. In recent years, antibody-based drugs have been extensively studied for the treatment of various significant human viruses. However, there is a lack of research on therapeutic monoclonal antibodies for mpox, particularly in the development and application of bsAbs. In this context, we have designed effective bsAbs that demonstrate high antiviral activity both in vitro and in vivo. This research provides a theoretical foundation for the development of specific therapeutic agents for mpox and offers new approaches for clinical treatment, which is crucial for controlling the current outbreak.
Collapse
Affiliation(s)
- Mengjun Li
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jiayin Chen
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Fungxiang Wang
- National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, China
| | - Jiahua Kuang
- National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, China
| | - Yun Peng
- National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, China
| | - Sadia Asghar
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Wei Zhao
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yang Yang
- National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, China
| | - Chenguang Shen
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
- Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Guangzhou, China
- State Key Laboratory of Multi-organ Injury Prevention and Treatment, Guangzhou, China
| |
Collapse
|
2
|
Hyun J, Hsieh LTH, Ayala R, Chang W, Wolf M. Methods to Study Poxvirus Structures by Cryo-EM Imaging Modalities. Methods Mol Biol 2025; 2860:191-218. [PMID: 39621269 DOI: 10.1007/978-1-0716-4160-6_13] [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: 03/26/2025]
Abstract
Poxviruses are double-stranded DNA viruses that represent the largest known highly pathogenic viruses infecting humans. They undergo dramatic morphological changes during their maturation process, resulting in structural differences between each virion, and their surface is decorated with more than a dozen randomly distributed surface proteins that facilitate viral entry. These are the main reasons poxviruses have eluded high-resolution structure determination. Over the last three decades, cryo-EM has developed into a mature technology that can increasingly overcome such problems of structural heterogeneity through advances in microscope technology and image processing algorithms. Here, we discuss the essential current modalities in cryo-EM, which promise to solve the structure of poxviruses in parts and as entire virions at near-atomic resolution. With a focus on cryo modalities, we provide an overview of methods, including volume microscopy by plasma ion beam milling, focused ion beam lamella preparation, subtomogram averaging, and single particle averaging. Protocols for poxvirus propagation, purification, and imaging by cryo-EM are presented. This chapter is aimed at experts and nonexpert researchers to help facilitate entry into the structural biology of this critical field in virology.
Collapse
Affiliation(s)
- Jaekyung Hyun
- School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi, Republic of Korea.
| | | | - Rafael Ayala
- Okinawa Institute of Science and Technology Graduate University (OIST), Molecular Cryo-Electron Microscopy Unit, Kunigami, Okinawa, Japan
| | - Wen Chang
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei, Taiwan.
| | - Matthias Wolf
- Okinawa Institute of Science and Technology Graduate University (OIST), Molecular Cryo-Electron Microscopy Unit, Kunigami, Okinawa, Japan.
- Institute of Biological Chemistry, Academia Sinica, Nankang, Taipei, Taiwan.
| |
Collapse
|
3
|
Hicks B, Jones S, Callaby H, Bailey D, Gordon C, Rampling T, Houlihan C, Linley E, Tonge S, Oeser C, Jones R, Pond M, Mehta R, Wright D, Hallis B, Rowe C, Otter A. Evaluation of a multiplexed immunoassay for assessing long-term humoral immunity Orthopoxviruses. Vaccine 2024; 42:126453. [PMID: 39426286 DOI: 10.1016/j.vaccine.2024.126453] [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: 08/17/2024] [Revised: 10/03/2024] [Accepted: 10/10/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND The 2022 Monkeypox virus (MPXV) global outbreak boosted development of multiple serological assays to aid understanding of Mpox immunology. OBJECTIVES The study aimed to assess a multiplexed solid-phase electrochemiluminescence immunoassay (Meso Scale Discovery (MSD)) for simultaneous detection of antibodies against MPXV, including A35, E8 and M1 antigens, along with corresponding Vaccina Virus (VACV) homologues and demonstrate its accuracy in assessing antibody titres post-vaccination and infection. METHODS Assay performance was assessed for simultaneous detection of antibodies against MPXV and corresponding VACV antigens. Sensitivity and specificity were evaluated with paediatric negatives (n = 215), pre- and post-IMVANEX vaccinated (n = 80), and MPXV (Clade IIb, n = 39) infected serum samples. RESULTS The assay demonstrated high specificity (75.68 % (CI: 69.01-81.29) - 95.98 % (CI:92.54-97.87)) and sensitivity (62.11 % (CI:52.06-71.21) - 98.59 % (CI:92.44 %-99.93 %)) depending on the Orthopoxvirus antigen. Preferential binding was observed between MPXV-infected individuals and MPXV antigens, while vaccinated individuals exhibited increased binding to VACV antigens. These results highlight differential binding patterns between antigen homologues in related viruses. CONCLUSION Overall, this assay demonstrates high sensitivities in detecting antibodies for multiple relevant MPXV and VACV antigens post-infection and post-vaccination, indicating its utility in understanding immune responses to Orthopoxviruses in current and future outbreaks and evaluating the immunogenicity of new-generation Mpox-specific vaccinations.
Collapse
Affiliation(s)
- Bethany Hicks
- Emerging Pathogen Serology Group, Vaccine Development Evaluation and Preparedness Centre, UK Health Security Agency, Porton Down, Wiltshire, UK.
| | - Scott Jones
- Emerging Pathogen Serology Group, Vaccine Development Evaluation and Preparedness Centre, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Helen Callaby
- Rare and Imported Pathogens Laboratory, UK Health Security Agency, Porton Down, Wiltshire, UK; The Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Daniel Bailey
- Rare and Imported Pathogens Laboratory, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Claire Gordon
- Rare and Imported Pathogens Laboratory, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Tommy Rampling
- Rare and Imported Pathogens Laboratory, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Catherine Houlihan
- Rare and Imported Pathogens Laboratory, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Ezra Linley
- Seroepidemiology Unit, UK Health Security Agency, Manchester, UK
| | - Simon Tonge
- Seroepidemiology Unit, UK Health Security Agency, Manchester, UK
| | - Clarissa Oeser
- Immunisation and Vaccine Preventable Diseases Division, UK Health Security Agency, Colindale, London, UK
| | - Rachael Jones
- Chelsea and Westminster Hospital NHS Foundation Trust, London, UK
| | - Marcus Pond
- Imperial College Healthcare NHS Trust, London, UK
| | - Ravi Mehta
- Imperial College Healthcare NHS Trust, London, UK
| | - Deborah Wright
- Research and Development, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Bassam Hallis
- Emerging Pathogen Serology Group, Vaccine Development Evaluation and Preparedness Centre, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Cathy Rowe
- Emerging Pathogen Serology Group, Vaccine Development Evaluation and Preparedness Centre, UK Health Security Agency, Porton Down, Wiltshire, UK
| | - Ashley Otter
- Emerging Pathogen Serology Group, Vaccine Development Evaluation and Preparedness Centre, UK Health Security Agency, Porton Down, Wiltshire, UK
| |
Collapse
|
4
|
Zhao R, Wu L, Sun J, Liu D, Han P, Gao Y, Zhang Y, Xu Y, Qu X, Wang H, Chai Y, Chen Z, Gao GF, Wang Q. Two noncompeting human neutralizing antibodies targeting MPXV B6 show protective effects against orthopoxvirus infections. Nat Commun 2024; 15:4660. [PMID: 38821921 PMCID: PMC11143242 DOI: 10.1038/s41467-024-48312-2] [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: 10/30/2023] [Accepted: 04/26/2024] [Indexed: 06/02/2024] Open
Abstract
The recent outbreak of mpox epidemic, caused by monkeypox virus (MPXV), poses a new threat to global public health. Here, we initially assessed the preexisting antibody level to the MPXV B6 protein in vaccinia vaccinees born before the end of the immunization program and then identified two monoclonal antibodies (MAbs), hMB621 and hMB668, targeting distinct epitopes on B6, from one vaccinee. Binding assays demonstrate that both MAbs exhibit broad binding abilities to B6 and its orthologs in vaccinia (VACV), variola (VARV) and cowpox viruses (CPXV). Neutralizing assays reveal that the two MAbs showed potent neutralization against VACV. Animal experiments using a BALB/c female mouse model indicate that the two MAbs showed effective protection against VACV via intraperitoneal injection. Additionally, we determined the complex structure of B6 and hMB668, revealing the structural feature of B6 and the epitope of hMB668. Collectively, our study provides two promising antibody candidates for the treatment of orthopoxvirus infections, including mpox.
Collapse
Affiliation(s)
- Runchu Zhao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Lili Wu
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Junqing Sun
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, Shanxi, China
| | - Dezhi Liu
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- Institute of Physical Science and Information, Anhui University, Hefei, Anhui, China
| | - Pu Han
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Yue Gao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- School of Life Sciences, Hebei University, Baoding, Hebei, China
| | - Yi Zhang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- Institute of Physical Science and Information, Anhui University, Hefei, Anhui, China
| | - Yanli Xu
- National Key Laboratory of Intelligent Tracking and Forecasting for infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Xiao Qu
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Han Wang
- College of Future Technology, Peking University, Beijing, China
| | - Yan Chai
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Zhihai Chen
- National Key Laboratory of Intelligent Tracking and Forecasting for infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - George F Gao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, Shanxi, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qihui Wang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.
- Institute of Physical Science and Information, Anhui University, Hefei, Anhui, China.
- University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
5
|
Lee W, Kim YJ, Lee SJ, Ahn DG, Kim SJ. Current Status of Epidemiology, Diagnosis, Therapeutics, and Vaccines for the Re-Emerging Human Monkeypox Virus. J Microbiol Biotechnol 2023; 33:981-991. [PMID: 37519276 PMCID: PMC10468680 DOI: 10.4014/jmb.2306.06033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/18/2023] [Accepted: 07/22/2023] [Indexed: 08/01/2023]
Abstract
Monkeypox (Mpox) virus, a member of the Poxviridae family, causes a severe illness similar to smallpox, which is characterized by symptoms such as high fever, rash, and pustules. Human-to-human transmission cases have been reported but remained low since the first recorded case of human infection occurred in the Congo in 1970. Recently, Mpox has re-emerged, leading to an alarming surge in infections worldwide since 2022, originating in the United Kingdom. Consequently, the World Health Organization (WHO) officially declared the '2022-23 Mpox outbreak'. Currently, no specific therapy or vaccine is available for Mpox. Therefore, patients infected with Mpox are treated using conventional therapies developed for smallpox. However, the vaccines developed for smallpox have demonstrated only partial efficacy against Mpox, allowing viral transmission among humans. In this review, we discuss the current epidemiology of the ongoing Mpox outbreak and provide an update on the progress made in diagnosis, treatment, and development of vaccines for Mpox.
Collapse
Affiliation(s)
- Wooseong Lee
- Department of Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Yu-Jin Kim
- Department of Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Su Jin Lee
- Department of Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Dae-Gyun Ahn
- Department of Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Seong-Jun Kim
- Department of Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| |
Collapse
|
6
|
Patel P, Nandi A, Verma SK, Kaushik N, Suar M, Choi EH, Kaushik NK. Zebrafish-based platform for emerging bio-contaminants and virus inactivation research. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 872:162197. [PMID: 36781138 PMCID: PMC9922160 DOI: 10.1016/j.scitotenv.2023.162197] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/23/2023] [Accepted: 02/08/2023] [Indexed: 05/27/2023]
Abstract
Emerging bio-contaminants such as viruses have affected health and environment settings of every country. Viruses are the minuscule entities resulting in severe contagious diseases like SARS, MERS, Ebola, and avian influenza. Recent epidemic like the SARS-CoV-2, the virus has undergone mutations strengthen them and allowing to escape from the remedies. Comprehensive knowledge of viruses is essential for the development of targeted therapeutic and vaccination treatments. Animal models mimicking human biology like non-human primates, rats, mice, and rabbits offer competitive advantage to assess risk of viral infections, chemical toxins, nanoparticles, and microbes. However, their economic maintenance has always been an issue. Furthermore, the redundancy of experimental results due to aforementioned aspects is also in examine. Hence, exploration for the alternative animal models is crucial for risk assessments. The current review examines zebrafish traits and explores the possibilities to monitor emerging bio-contaminants. Additionally, a comprehensive picture of the bio contaminant and virus particle invasion and abatement mechanisms in zebrafish and human cells is presented. Moreover, a zebrafish model to investigate the emerging viruses such as coronaviridae and poxviridae has been suggested.
Collapse
Affiliation(s)
- Paritosh Patel
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, 01897 Seoul, South Korea
| | - Aditya Nandi
- School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Suresh K Verma
- School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India; Condensed Matter Theory Group, Materials Theory Division, Department of Physics and Astronomy, Uppsala University, Box 516, SE-751 20 Uppsala, Sweden
| | - Neha Kaushik
- Department of Biotechnology, College of Engineering, The University of Suwon, 18323 Hwaseong, Republic of Korea
| | - Mrutyunjay Suar
- School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Eun Ha Choi
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, 01897 Seoul, South Korea.
| | - Nagendra Kumar Kaushik
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, 01897 Seoul, South Korea.
| |
Collapse
|
7
|
Saud Z, Hitchings MD, Butt TM. Nanopore sequencing and de novo assembly of a misidentified Camelpox vaccine reveals putative epigenetic modifications and alternate protein signal peptides. Sci Rep 2021; 11:17758. [PMID: 34493784 PMCID: PMC8423768 DOI: 10.1038/s41598-021-97158-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 08/19/2021] [Indexed: 11/25/2022] Open
Abstract
DNA viruses can exploit host cellular epigenetic processes to their advantage; however, the epigenome status of most DNA viruses remains undetermined. Third generation sequencing technologies allow for the identification of modified nucleotides from sequencing experiments without specialized sample preparation, permitting the detection of non-canonical epigenetic modifications that may distinguish viral nucleic acid from that of their host, thus identifying attractive targets for advanced therapeutics and diagnostics. We present a novel nanopore de novo assembly pipeline used to assemble a misidentified Camelpox vaccine. Two confirmed deletions of this vaccine strain in comparison to the closely related Vaccinia virus strain modified vaccinia Ankara make it one of the smallest non-vector derived orthopoxvirus genomes to be reported. Annotation of the assembly revealed a previously unreported signal peptide at the start of protein A38 and several predicted signal peptides that were found to differ from those previously described. Putative epigenetic modifications around various motifs have been identified and the assembly confirmed previous work showing the vaccine genome to most closely resemble that of Vaccinia virus strain Modified Vaccinia Ankara. The pipeline may be used for other DNA viruses, increasing the understanding of DNA virus evolution, virulence, host preference, and epigenomics.
Collapse
Affiliation(s)
- Zack Saud
- Department of Biosciences, College of Science, Swansea University, Singleton Park, Swansea, SA2 8PP, Wales, UK.
| | - Matthew D Hitchings
- Swansea University Medical School, Swansea University, Singleton Park, Swansea, Sa2 8PP, Wales, UK
| | - Tariq M Butt
- Department of Biosciences, College of Science, Swansea University, Singleton Park, Swansea, SA2 8PP, Wales, UK
| |
Collapse
|
8
|
Connor AJ, Zha RH, Koffas M. Bioproduction of biomacromolecules for antiviral applications. Curr Opin Biotechnol 2021; 69:263-272. [PMID: 33667798 DOI: 10.1016/j.copbio.2021.01.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 01/14/2021] [Accepted: 01/25/2021] [Indexed: 02/07/2023]
Abstract
The societal damage brought on by viral epidemics indicates that next-generation antiviral treatments must be developed and deployed. Biomacromolecules are a diverse class of compounds that can potentially exhibit potent antiviral activity. Their efficacy and mechanisms of action are dependent upon multiple structural factors, including molecular weight, degree and position of sulfation, and backbone stereochemistry. Extracting biomacromolecules from animals and plants for healthcare applications is undesirable, as these methods are unable to yield products with well-defined chemical structures. Modern advances utilizing recombinant microbes and metabolic pathway engineering can be a key step towards large-scale bioproduction of tailored biomacromolecules for targeted antiviral applications.
Collapse
Affiliation(s)
- Alexander J Connor
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Runye H Zha
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Mattheos Koffas
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA.
| |
Collapse
|
9
|
Realegeno S, Priyamvada L, Kumar A, Blackburn JB, Hartloge C, Puschnik AS, Sambhara S, Olson VA, Carette JE, Lupashin V, Satheshkumar PS. Conserved Oligomeric Golgi (COG) Complex Proteins Facilitate Orthopoxvirus Entry, Fusion and Spread. Viruses 2020; 12:v12070707. [PMID: 32629851 PMCID: PMC7411930 DOI: 10.3390/v12070707] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 06/25/2020] [Indexed: 02/07/2023] Open
Abstract
Although orthopoxviruses (OPXV) are known to encode a majority of the genes required for replication in host cells, genome-wide genetic screens have revealed that several host pathways are indispensable for OPXV infection. Through a haploid genetic screen, we previously identified several host genes required for monkeypox virus (MPXV) infection, including the individual genes that form the conserved oligomeric Golgi (COG) complex. The COG complex is an eight-protein (COG1-COG8) vesicle tethering complex important for regulating membrane trafficking, glycosylation enzymes, and maintaining Golgi structure. In this study, we investigated the role of the COG complex in OPXV infection using cell lines with individual COG gene knockout (KO) mutations. COG KO cells infected with MPXV and vaccinia virus (VACV) produced small plaques and a lower virus yield compared to wild type (WT) cells. In cells where the KO phenotype was reversed using a rescue plasmid, the size of virus plaques increased demonstrating a direct link between the decrease in viral spread and the KO of COG genes. KO cells infected with VACV displayed lower levels of viral fusion and entry compared to WT suggesting that the COG complex is important for early events in OPXV infection. Additionally, fewer actin tails were observed in VACV-infected KO cells compared to WT. Since COG complex proteins are required for cellular trafficking of glycosylated membrane proteins, the disruption of this process due to lack of individual COG complex proteins may potentially impair the virus-cell interactions required for viral entry and egress. These data validate that the COG complex previously identified in our genetic screens plays a role in OPXV infection.
Collapse
Affiliation(s)
- Susan Realegeno
- Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, Atlanta, GA 3033, USA; (S.R.); (L.P.); (C.H.); (V.A.O.)
| | - Lalita Priyamvada
- Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, Atlanta, GA 3033, USA; (S.R.); (L.P.); (C.H.); (V.A.O.)
| | - Amrita Kumar
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA 3033, USA; (A.K.); (S.S.)
| | - Jessica B. Blackburn
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (J.B.B.); (V.L.)
| | - Claire Hartloge
- Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, Atlanta, GA 3033, USA; (S.R.); (L.P.); (C.H.); (V.A.O.)
| | - Andreas S. Puschnik
- Department of Microbiology and Immunology, Stanford University, Stanford, CA 94035, USA; (A.S.P.); (J.E.C.)
| | - Suryaprakash Sambhara
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA 3033, USA; (A.K.); (S.S.)
| | - Victoria A. Olson
- Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, Atlanta, GA 3033, USA; (S.R.); (L.P.); (C.H.); (V.A.O.)
| | - Jan E. Carette
- Department of Microbiology and Immunology, Stanford University, Stanford, CA 94035, USA; (A.S.P.); (J.E.C.)
| | - Vladimir Lupashin
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (J.B.B.); (V.L.)
| | - Panayampalli Subbian Satheshkumar
- Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, Atlanta, GA 3033, USA; (S.R.); (L.P.); (C.H.); (V.A.O.)
- Correspondence:
| |
Collapse
|
10
|
Nakatake M, Kurosaki H, Kuwano N, Horita K, Ito M, Kono H, Okamura T, Hasegawa K, Yasutomi Y, Nakamura T. Partial Deletion of Glycoprotein B5R Enhances Vaccinia Virus Neutralization Escape while Preserving Oncolytic Function. MOLECULAR THERAPY-ONCOLYTICS 2019; 14:159-171. [PMID: 31236440 PMCID: PMC6580015 DOI: 10.1016/j.omto.2019.05.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 05/09/2019] [Indexed: 11/12/2022]
Abstract
Vaccinia virus (VV) has been utilized in oncolytic virotherapy, but it risks a host antiviral immune response. VV has an extracellular enveloped virus (EEV) form consisting of a normal virion covered with a host-derived outer membrane that enables its spread via circulation while evading host immune mechanisms. However, the immune resistance of EEV is only partial, owing to expression of the surface protein B5R, which has four short consensus repeat (SCR) domains that are targeted by host immune factors. To engineer a more effective virus for oncolytic virotherapy, we developed an enhanced immune-evading oncolytic VV by removing the SCRs from the attenuated strain LC16mO. Although deletion of only the SCRs preserved viral replication, progeny production, and oncolytic activity, deletion of whole B5R led to attenuation of the virus. Importantly, SCR-deleted EEV had higher neutralization resistance than did B5R-wild-type EEV against VV-immunized animal serum; moreover, it retained oncolytic function, thereby prolonging the survival of tumor-bearing mice treated with anti-VV antibody. These results demonstrate that partial SCR deletion increases neutralization escape without affecting the oncolytic potency of VV, making it useful for the treatment of tumors under the anti-virus antibody existence.
Collapse
Affiliation(s)
- Motomu Nakatake
- Division of Molecular Medicine, Department of Biomedical Science, Graduate School of Medical Sciences, Tottori University, 86 Nishi-cho, Yonago 683-8503, Japan
| | - Hajime Kurosaki
- Division of Molecular Medicine, Department of Biomedical Science, Graduate School of Medical Sciences, Tottori University, 86 Nishi-cho, Yonago 683-8503, Japan
| | - Nozomi Kuwano
- Division of Molecular Medicine, Department of Biomedical Science, Graduate School of Medical Sciences, Tottori University, 86 Nishi-cho, Yonago 683-8503, Japan
| | - Kosuke Horita
- Division of Molecular Medicine, Department of Biomedical Science, Graduate School of Medical Sciences, Tottori University, 86 Nishi-cho, Yonago 683-8503, Japan
| | - Mai Ito
- Division of Molecular Medicine, Department of Biomedical Science, Graduate School of Medical Sciences, Tottori University, 86 Nishi-cho, Yonago 683-8503, Japan
| | - Hiromichi Kono
- Division of Molecular Medicine, Department of Biomedical Science, Graduate School of Medical Sciences, Tottori University, 86 Nishi-cho, Yonago 683-8503, Japan
| | - Tomotaka Okamura
- Laboratory of Immunoregulation and Vaccine Research, Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, Tsukuba, Ibaraki 305-0843, Japan
| | - Kosei Hasegawa
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, 1397-1, Yamane, Hidaka-City, Saitama 350-1298, Japan
| | - Yasuhiro Yasutomi
- Laboratory of Immunoregulation and Vaccine Research, Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, Tsukuba, Ibaraki 305-0843, Japan
| | - Takafumi Nakamura
- Division of Molecular Medicine, Department of Biomedical Science, Graduate School of Medical Sciences, Tottori University, 86 Nishi-cho, Yonago 683-8503, Japan
| |
Collapse
|
11
|
The Ectodomain of the Vaccinia Virus Glycoprotein A34 Is Required for Cell Binding by Extracellular Virions and Contains a Large Region Capable of Interaction with Glycoprotein B5. J Virol 2019; 93:JVI.01343-18. [PMID: 30463966 DOI: 10.1128/jvi.01343-18] [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] [Received: 08/16/2018] [Accepted: 11/12/2018] [Indexed: 12/17/2022] Open
Abstract
An interaction between the orthopoxvirus glycoproteins A34 and B5 has been reported. The transmembrane and ectodomain of A34 are sufficient for interaction with B5, localization of B5 to the site of intracellular wrapping, and subsequent incorporation into the envelope of released extracellular virions. Several mutagenic approaches were undertaken to better define the B5 interaction domain on A34. A set of C-terminal truncations in A34 identified residues 1 to 80 as sufficient for interaction with B5. Additional truncations identified residues 80 to 130 of A34 as sufficient for interaction with B5. To better understand the function of this region, a set of recombinant viruses expressing A34 with the full, partial, or no B5 interaction site (residues 1 to 130, 1 to 100, and 1 to 70, respectively) was constructed. All the recombinants expressing truncations of A34 incorporated B5 into extracellular virions but had a small-plaque phenotype similar to that of a virus with the A34R gene deleted (vΔA34R). Further characterization indicated that the small-plaque phenotype exhibited by these viruses is due to a combination of abrogated actin tail formation, reduced cell binding, and a defect in polyanion-induced nonfusogenic dissolution. Taken together, these results suggest that residues 80 to 130 of A34 are not necessary for the proper localization and incorporation of B5 into extracellular virions and, furthermore, that the C-terminal residues of A34 are involved in cell binding and dissolution.IMPORTANCE Previous studies have shown that the vaccinia virus glycoproteins A34 and B5 interact, and in the absence of A34, B5 is mislocalized and not incorporated into extracellular virions. Here, using a transient-transfection assay, residues 80 to 130 of the ectodomain of A34 were determined to be sufficient for interaction with B5. Recombinant viruses expressing A34 with a full, partial, or no B5 interaction site were constructed and characterized. All of the A34 truncations interacted with B5 as predicted by the transient-transfection studies but had a small-plaque phenotype. Further analysis revealed that all of the recombinants incorporated detectable levels of B5 into released virions but were defective in cell binding and extracellular virion (EV) dissolution. This study is the first to directly demonstrate that A34 is involved in cell binding and implicate the ectodomain in this role.
Collapse
|
12
|
Abstract
Interferons (IFNs) are secreted glycoproteins that are produced by cells in response to virus infection and other stimuli and induce an antiviral state in cells bearing IFN receptors. In this way, IFNs restrict virus replication and spread before an adaptive immune response is developed. Viruses are very sensitive to the effects of IFNs and consequently have evolved many strategies to interfere with interferon. This is particularly well illustrated by poxviruses, which have large dsDNA genomes and encode hundreds of proteins. Vaccinia virus is the prototypic poxvirus and expresses many proteins that interfere with IFN and are considered in this review. These proteins act either inside or outside the cell and within the cytoplasm or nucleus. They function by restricting the production of IFN by blocking the signaling pathways leading to transcription of IFN genes, stopping IFNs binding to their receptors, blocking IFN-induced signal transduction leading to expression of interferon-stimulated genes (ISGs), or inhibiting the antiviral activity of ISG products.
Collapse
Affiliation(s)
| | | | - Yongxu Lu
- University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
13
|
Khanna M, Ranasinghe C, Jackson R, Parish CR. Heparan sulfate as a receptor for poxvirus infections and as a target for antiviral agents. J Gen Virol 2017; 98:2556-2568. [PMID: 28933686 DOI: 10.1099/jgv.0.000921] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
To establish the importance of virus-heparan sulfate (HS) interactions in virus infectivity, the poxvirus vaccinia virus (VACV) was used, as it binds HS and has both enveloped virus (EV) and non-enveloped mature virus (MV) forms. Initial studies showed that heparin inhibited plaque formation by both MV-rich WR and EV-rich IHD-J strains of VACV, with the EV-rich strain also losing trademark 'comet'-shaped plaques. However, using GFP-tagged EV and MV forms of VACV, based on IC50 values, heparin was 16-fold more effective at inhibiting the infectivity of the EV form compared to the MV form. Furthermore, 6-O and N-sulfation of the glucosamine residues of heparin was essential for inhibition of the infectivity of both VACV forms. Several low-molecular-weight HS mimetics were also shown to have substantial antiviral activity, with glycosidic linkages, chain length and monosaccharide backbone being important contributors towards anti-VACV activity. In fact, the d-mannose-based sulfated oligosaccharide mixture, PI-88 (Muparfostat), was four-fold more active than heparin at inhibiting MV infections. Paradoxically, despite heparin and HS mimetics being potent inhibitors of VACV infections, removal of HS from cell surfaces by enzymatic or genetic means resulted in only a modest reduction in infectivity. It is unlikely that this paradox can be explained by steric hindrance, due to the low molecular weight of the HS mimetics (~1-2.5 kDa), with a more likely explanation being that binding of heparin/HS mimetics to free VACV initiates an abortive viral infection. Based on this explanation, HS mimetics have considerable potential as antivirals against HS-binding viruses.
Collapse
Affiliation(s)
- Mayank Khanna
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology and Infectious Diseases, The John Curtin School of Medical Research, The Australian National University (ANU), Canberra ACT 2601, Australia
| | - Charani Ranasinghe
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology and Infectious Diseases, The John Curtin School of Medical Research, The Australian National University (ANU), Canberra ACT 2601, Australia
| | - Ronald Jackson
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology and Infectious Diseases, The John Curtin School of Medical Research, The Australian National University (ANU), Canberra ACT 2601, Australia
| | - Christopher Richard Parish
- Cancer and Vascular Biology Group, ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra ACT 2601, Australia
| |
Collapse
|
14
|
Ziem B, Thien H, Achazi K, Yue C, Stern D, Silberreis K, Gholami MF, Beckert F, Gröger D, Mülhaupt R, Rabe JP, Nitsche A, Haag R. Highly Efficient Multivalent 2D Nanosystems for Inhibition of Orthopoxvirus Particles. Adv Healthc Mater 2016; 5:2922-2930. [PMID: 27581958 DOI: 10.1002/adhm.201600812] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Indexed: 12/15/2022]
Abstract
Efficient inhibition of cell-pathogen interaction to prevent subsequent infection is an urgent but yet unsolved problem. In this study, the synthesis and functionalization of novel multivalent 2D carbon nanosystems as well as their antiviral efficacy in vitro are shown. For this reason, a new multivalent 2D flexible carbon architecture is developed in this study, functionalized with sulfated dendritic polyglycerol, to enable virus interaction. A simple "graft from" approach enhances the solubility of thermally reduced graphene oxide and provides a suitable 2D surface for multivalent ligand presentation. Polysulfation is used to mimic the heparan sulfate-containing surface of cells and to compete with this natural binding site of viruses. In correlation with the degree of sulfation and the grafted polymer density, the interaction efficiency of these systems can be varied. In here, orthopoxvirus strains are used as model viruses as they use heparan sulfate for cell entry as other viruses, e.g., herpes simplex virus, dengue virus, or cytomegalovirus. The characterization results of the newly designed graphene derivatives demonstrate excellent binding as well as efficient inhibition of orthopoxvirus infection. Overall, these new multivalent 2D polymer nanosystems are promising candidates to develop potent inhibitors for viruses, which possess a heparan sulfate-dependent cell entry mechanism.
Collapse
Affiliation(s)
- Benjamin Ziem
- Institute of Chemistry and Biochemistry; Freie Universität; 14195 Berlin Germany
| | - Hendrik Thien
- Institute of Virology; University of Leipzig; 04103 Leipzig Germany
- Robert Koch Institute; Center for Biological Threats and Special Pathogens; 13353 Berlin Germany
| | - Katharina Achazi
- Institute of Chemistry and Biochemistry; Freie Universität; 14195 Berlin Germany
| | - Constanze Yue
- Robert Koch Institute; Center for Biological Threats and Special Pathogens; 13353 Berlin Germany
| | - Daniel Stern
- Robert Koch Institute; Center for Biological Threats and Special Pathogens; 13353 Berlin Germany
| | - Kim Silberreis
- Robert Koch Institute; Center for Biological Threats and Special Pathogens; 13353 Berlin Germany
| | | | - Fabian Beckert
- Institute for Macromolecular Chemistry; University of Freiburg; 79104 Freiburg Germany
| | - Dominic Gröger
- Institute of Chemistry and Biochemistry; Freie Universität; 14195 Berlin Germany
| | - Rolf Mülhaupt
- Institute for Macromolecular Chemistry; University of Freiburg; 79104 Freiburg Germany
| | - Jürgen P. Rabe
- Institute for Physics and IRIS Adlershof; Humboldt-Universität Berlin; 12489 Berlin Germany
| | - Andreas Nitsche
- Robert Koch Institute; Center for Biological Threats and Special Pathogens; 13353 Berlin Germany
| | - Rainer Haag
- Institute of Chemistry and Biochemistry; Freie Universität; 14195 Berlin Germany
| |
Collapse
|
15
|
Moss B. Membrane fusion during poxvirus entry. Semin Cell Dev Biol 2016; 60:89-96. [PMID: 27423915 DOI: 10.1016/j.semcdb.2016.07.015] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 07/11/2016] [Accepted: 07/12/2016] [Indexed: 12/23/2022]
Abstract
Poxviruses comprise a large family of enveloped DNA viruses that infect vertebrates and invertebrates. Poxviruses, unlike most DNA viruses, replicate in the cytoplasm and encode enzymes and other proteins that enable entry, gene expression, genome replication, virion assembly and resistance to host defenses. Entry of vaccinia virus, the prototype member of the family, can occur at the plasma membrane or following endocytosis. Whereas many viruses encode one or two proteins for attachment and membrane fusion, vaccinia virus encodes four proteins for attachment and eleven more for membrane fusion and core entry. The entry-fusion proteins are conserved in all poxviruses and form a complex, known as the Entry Fusion Complex (EFC), which is embedded in the membrane of the mature virion. An additional membrane that encloses the mature virion and is discarded prior to entry is present on an extracellular form of the virus. The EFC is held together by multiple interactions that depend on nine of the eleven proteins. The entry process can be divided into attachment, hemifusion and core entry. All eleven EFC proteins are required for core entry and at least eight for hemifusion. To mediate fusion the virus particle is activated by low pH, which removes one or more fusion repressors that interact with EFC components. Additional EFC-interacting fusion repressors insert into cell membranes and prevent secondary infection. The absence of detailed structural information, except for two attachment proteins and one EFC protein, is delaying efforts to determine the fusion mechanism.
Collapse
Affiliation(s)
- Bernard Moss
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
16
|
Grélard A, Guichard P, Bonnafous P, Marco S, Lambert O, Manin C, Ronzon F, Dufourc EJ. Hepatitis B subvirus particles display both a fluid bilayer membrane and a strong resistance to freeze drying: a study by solid‐state NMR, light scattering, and cryo‐electron microscopy/tomography. FASEB J 2013; 27:4316-26. [DOI: 10.1096/fj.13-232843] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Axelle Grélard
- Institute of Chemistry and Biology of Membranes and NanoobjectsUniversity of BordeauxChemistry and Biology of Membranes and Nanoobjects (CBMN)Unité Mixte de Recherche (UMR) 5248PessacFrance
- Institute of Chemistry and Biology of Membranes and NanoobjectsCentre National de la Recherche Scientifique (CNRS)CBMNUMR5248PessacFrance
| | - Paul Guichard
- Institut CurieCentre de RechercheOrsayFrance
- Institut National de la Santé et de la Recherche Médicale (INSERM)U759OrsayFrance
| | - Pierre Bonnafous
- Institute of Chemistry and Biology of Membranes and NanoobjectsUniversity of BordeauxChemistry and Biology of Membranes and Nanoobjects (CBMN)Unité Mixte de Recherche (UMR) 5248PessacFrance
- Institute of Chemistry and Biology of Membranes and NanoobjectsCentre National de la Recherche Scientifique (CNRS)CBMNUMR5248PessacFrance
| | - Sergio Marco
- Institut CurieCentre de RechercheOrsayFrance
- Institut National de la Santé et de la Recherche Médicale (INSERM)U759OrsayFrance
| | - Olivier Lambert
- Institute of Chemistry and Biology of Membranes and NanoobjectsUniversity of BordeauxChemistry and Biology of Membranes and Nanoobjects (CBMN)Unité Mixte de Recherche (UMR) 5248PessacFrance
- Institute of Chemistry and Biology of Membranes and NanoobjectsCentre National de la Recherche Scientifique (CNRS)CBMNUMR5248PessacFrance
| | | | | | - Erick J. Dufourc
- Institute of Chemistry and Biology of Membranes and NanoobjectsUniversity of BordeauxChemistry and Biology of Membranes and Nanoobjects (CBMN)Unité Mixte de Recherche (UMR) 5248PessacFrance
- Institut CurieCentre de RechercheOrsayFrance
| |
Collapse
|
17
|
Kamhi E, Joo EJ, Dordick JS, Linhardt RJ. Glycosaminoglycans in infectious disease. Biol Rev Camb Philos Soc 2013; 88:928-43. [DOI: 10.1111/brv.12034] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Revised: 02/07/2013] [Accepted: 02/27/2013] [Indexed: 12/14/2022]
Affiliation(s)
- Eyal Kamhi
- Department of Chemistry and Chemical Biology; Rensselaer Polytechnic Institute; Troy New York 12180-3590 U.S.A
- Drughoming Ltd; Rehovot Israel
| | - Eun Ji Joo
- Department of Chemistry and Chemical Biology; Rensselaer Polytechnic Institute; Troy New York 12180-3590 U.S.A
| | - Jonathan S. Dordick
- Department of Biology; Rensselaer Polytechnic Institute; Troy New York 12180-3590 U.S.A
- Department of Chemical and Biological Engineering; Rensselaer Polytechnic Institute; Troy New York 12180-3590 U.S.A
- Department of Biomedical Engineering; Center for Biotechnology & Interdisciplinary Studies; Rensselaer Polytechnic Institute; Troy New York 12180-3590 U.S.A
| | - Robert J. Linhardt
- Department of Chemistry and Chemical Biology; Rensselaer Polytechnic Institute; Troy New York 12180-3590 U.S.A
- Department of Biology; Rensselaer Polytechnic Institute; Troy New York 12180-3590 U.S.A
- Department of Chemical and Biological Engineering; Rensselaer Polytechnic Institute; Troy New York 12180-3590 U.S.A
- Department of Biomedical Engineering; Center for Biotechnology & Interdisciplinary Studies; Rensselaer Polytechnic Institute; Troy New York 12180-3590 U.S.A
| |
Collapse
|
18
|
Breiman A, Carpentier DCJ, Ewles HA, Smith GL. Transport and stability of the vaccinia virus A34 protein is affected by the A33 protein. J Gen Virol 2012; 94:720-725. [PMID: 23255618 PMCID: PMC3709683 DOI: 10.1099/vir.0.049486-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Vaccinia virus (VACV) has two infectious forms called intracellular mature virus and extracellular enveloped virus (EEV). Two of the seven viral proteins in the EEV outer envelope, A33 and A34, are type II membrane glycoproteins that each interact with another EEV protein called B5; however, evidence for direct A33–A34 interaction is lacking. The localization and stability of A34 is affected by B5 and here data are presented showing that A34 is also affected by A33. In the absence of A33, just as without B5, the level, localization and glycosylation profile of A34 was altered. However, the glycosylation profile of A34 without A33 is different to that observed in the absence of B5, and A34 accumulates in the Golgi apparatus rather than in the endoplasmic reticulum. Thus, A34 requires more than one other EEV protein for its processing and cellular transport.
Collapse
Affiliation(s)
- Adrien Breiman
- Department of Virology, Faculty of Medicine, Imperial College London, Norfolk Place, London W2 1PG, UK
| | - David C J Carpentier
- Department of Virology, Faculty of Medicine, Imperial College London, Norfolk Place, London W2 1PG, UK
| | - Helen A Ewles
- Department of Virology, Faculty of Medicine, Imperial College London, Norfolk Place, London W2 1PG, UK
| | - Geoffrey L Smith
- Department of Virology, Faculty of Medicine, Imperial College London, Norfolk Place, London W2 1PG, UK
| |
Collapse
|
19
|
Increased interaction between vaccinia virus proteins A33 and B5 is detrimental to infectious extracellular enveloped virion production. J Virol 2012; 86:8232-44. [PMID: 22623782 DOI: 10.1128/jvi.00253-12] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Two mechanisms exist for the incorporation of B5 into extracellular virions, one of which is dependent on A33. In the companion to this paper (W. M. Chan and B. M. Ward, J. Virol. 86:8210-8220, 2012), we show that the lumenal domain of A33 is sufficient for interaction with the coiled-coil domain of B5 and capable of directing B5-green fluorescent protein (GFP) into extracellular virions. Here, we have created a panel of charge-to-alanine mutations in the lumenal domain of A33 to map the B5 interaction site. While none of these mutations abolished the interaction with B5, a subset displayed an increased interaction with both B5 and B5-GFP. Both B5 and B5-GFP recombinant viruses expressing these mutant proteins in place of normal A33 had a small-plaque phenotype. The increased interaction of the mutant proteins was detected during infection, suggesting that normally the interaction is either weak or transient. In addition, the increased A33-B5 interaction was detected on virions produced by recombinant viruses and correlated with reduced target cell binding. Taken together, these results show that both B5 and B5-GFP interact with A33 during infection and that the duration of this interaction needs to be regulated for the production of fully infectious extracellular virions.
Collapse
|
20
|
The A33-dependent incorporation of B5 into extracellular enveloped vaccinia virions is mediated through an interaction between their lumenal domains. J Virol 2012; 86:8210-20. [PMID: 22623777 DOI: 10.1128/jvi.00249-12] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
There are two mechanisms for the incorporation of B5 into the envelope of extracellular virions produced by orthopoxviruses, one that requires A33 and one that does not. We have hypothesized that the A33-dependent mechanism requires a direct interaction between A33 and B5. In this study, chimeric constructs of A33 and B5/B5-green fluorescent protein (GFP) were used to show that the two proteins interact through their lumenal domains and that the coiled-coil domain of B5 is sufficient for an interaction with A33. Furthermore, our experiments reveal that a transmembrane domain, not necessarily its own, is requisite for the lumenal domain of B5 to interact with A33. In contrast, the lumenal domain of A33 is sufficient for interaction with B5. Furthermore, the lumenal domain of A33 is sufficient to restore the proper localization of B5-GFP in infected cells. Taken together, our results demonstrate that the lumenal domains of A33 and B5 interact and that the interaction is required for the incorporation of B5-GFP into extracellular virions, whereas the incorporation of A33 is independent of B5. These results suggest that viral protein incorporation into extracellular virions is an active process requiring specific protein-protein interactions.
Collapse
|
21
|
Doceul V, Hollinshead M, Breiman A, Laval K, Smith GL. Protein B5 is required on extracellular enveloped vaccinia virus for repulsion of superinfecting virions. J Gen Virol 2012; 93:1876-1886. [PMID: 22622330 PMCID: PMC3709573 DOI: 10.1099/vir.0.043943-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Vaccinia virus (VACV) spreads across cell monolayers fourfold faster than predicted from its replication kinetics. Early after infection, infected cells repulse some superinfecting extracellular enveloped virus (EEV) particles by the formation of actin tails from the cell surface, thereby causing accelerated spread to uninfected cells. This strategy requires the expression of two viral proteins, A33 and A36, on the surface of infected cells and upon contact with EEV this complex induces actin polymerization. Here we have studied this phenomenon further and investigated whether A33 and A36 expression in cell lines causes an increase in VACV plaque size, whether these proteins are able to block superinfection by EEV, and which protein(s) on the EEV surface are required to initiate the formation of actin tails from infected cells. Data presented show that VACV plaque size was not increased by expression of A33 and A36, and these proteins did not block entry of the majority of EEV binding to these cells. In contrast, expression of proteins A56 and K2 inhibited entry of both EEV and intracellular mature virus. Lastly, VACV protein B5 was required on EEV to induce the formation of actin tails at the surface of cells expressing A33 and A36, and B5 short consensus repeat 4 is critical for this induction.
Collapse
|
22
|
Kerr PJ. Myxomatosis in Australia and Europe: a model for emerging infectious diseases. Antiviral Res 2012; 93:387-415. [PMID: 22333483 DOI: 10.1016/j.antiviral.2012.01.009] [Citation(s) in RCA: 125] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Revised: 01/20/2012] [Accepted: 01/26/2012] [Indexed: 11/18/2022]
Abstract
Myxoma virus is a poxvirus naturally found in two American leporid (rabbit) species (Sylvilagus brasiliensis and Sylvilagus bachmani) in which it causes an innocuous localised cutaneous fibroma. However, in European rabbits (Oryctolagus cuniculus) the same virus causes the lethal disseminated disease myxomatosis. The introduction of myxoma virus into the European rabbit population in Australia in 1950 initiated the best known example of what happens when a novel pathogen jumps into a completely naïve new mammalian host species. The short generation time of the rabbit and their vast numbers in Australia meant evolution could be studied in real time. The carefully documented emergence of attenuated strains of virus that were more effectively transmitted by the mosquito vector and the subsequent selection of rabbits with genetic resistance to myxomatosis is the paradigm for pathogen virulence and host-pathogen coevolution. This natural experiment was repeated with the release of a separate strain of myxoma virus in France in 1952. The subsequent spread of the virus throughout Europe and its coevolution with the rabbit essentially paralleled what occurred in Australia. Detailed molecular studies on myxoma virus have dissected the role of virulence genes in the pathogenesis of myxomatosis and when combined with genomic data and reverse genetics should in future enable the understanding of the molecular evolution of the virus as it adapted to its new host. This review describes the natural history and evolution of myxoma virus together with the molecular biology and experimental pathogenesis studies that are informing our understanding of evolution of emerging diseases.
Collapse
Affiliation(s)
- Peter J Kerr
- CSIRO Ecosystem Sciences, GPO Box 1700, Canberra, ACT 2601, Australia.
| |
Collapse
|
23
|
Schmidt FI, Bleck CKE, Mercer J. Poxvirus host cell entry. Curr Opin Virol 2012; 2:20-7. [DOI: 10.1016/j.coviro.2011.11.007] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Accepted: 11/10/2011] [Indexed: 12/20/2022]
|
24
|
Lorenzo MM, Sánchez-Puig JM, Blasco R. Mutagenesis of the palmitoylation site in vaccinia virus envelope glycoprotein B5. J Gen Virol 2012; 93:733-743. [PMID: 22238237 DOI: 10.1099/vir.0.039016-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The outer envelope of vaccinia virus extracellular virions is derived from intracellular membranes that, at late times in infection, are enriched in several virus-encoded proteins. Although palmitoylation is common in vaccinia virus envelope proteins, little is known about the role of palmitoylation in the biogenesis of the enveloped virus. We have studied the palmitoylation of B5, a 42 kDa type I transmembrane glycoprotein comprising a large ectodomain and a short (17 aa) cytoplasmic tail. Mutation of two cysteine residues located in the cytoplasmic tail in close proximity to the transmembrane domain abrogated palmitoylation of the protein. Virus mutants expressing non-palmitoylated versions of B5 and/or lacking most of the cytoplasmic tail were isolated and characterized. Cell-to-cell virus transmission and extracellular virus formation were only slightly affected by those mutations. Notably, B5 versions lacking palmitate showed decreased interactions with proteins A33 and F13, but were still incorporated into the virus envelope. Expression of mutated B5 by transfection into uninfected cells showed that both the cytoplasmic tail and palmitate have a role in the intracellular transport of B5. These results indicate that the C-terminal portion of protein B5, while involved in protein transport and in protein-protein interactions, is broadly dispensable for the formation and egress of infectious extracellular virus and for virus transmission.
Collapse
Affiliation(s)
- María M Lorenzo
- Departamento de Biotecnología - I.N.I.A. Ctra, La Coruña km 7, E-28040 Madrid, Spain
| | - Juana M Sánchez-Puig
- Departamento de Biotecnología - I.N.I.A. Ctra, La Coruña km 7, E-28040 Madrid, Spain
| | - Rafael Blasco
- Departamento de Biotecnología - I.N.I.A. Ctra, La Coruña km 7, E-28040 Madrid, Spain
| |
Collapse
|
25
|
Vaccinia extracellular virions enter cells by macropinocytosis and acid-activated membrane rupture. EMBO J 2011; 30:3647-61. [PMID: 21792173 PMCID: PMC3181475 DOI: 10.1038/emboj.2011.245] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2011] [Accepted: 06/29/2011] [Indexed: 11/12/2022] Open
Abstract
Double membrane-bound vaccinia extracellular virions actively promote their own macropinocytosis. This, followed by acid-induced rupture of the outer membrane in endocytic vesicles, exposes the inner membrane for fusion with the endocytic membrane and release into the cytosol. Vaccinia virus (VACV), the model poxvirus, produces two types of infectious particles: mature virions (MVs) and extracellular virions (EVs). EV particles possess two membranes and therefore require an unusual cellular entry mechanism. By a combination of fluorescence and electron microscopy as well as flow cytometry, we investigated the cellular processes that EVs required to infect HeLa cells. We found that EV particles were endocytosed, and that internalization and infection depended on actin rearrangements, activity of Na+/H+ exchangers, and signalling events typical for the macropinocytic mechanism of endocytosis. To promote their internalization, EVs were capable of actively triggering macropinocytosis. EV infection also required vacuolar acidification, and acid exposure in endocytic vacuoles was needed to disrupt the outer EV membrane. Once exposed, the underlying MV-like particle presumably fused its single membrane with the limiting vacuolar membrane. Release of the viral core into the host cell cytosol allowed for productive infection.
Collapse
|
26
|
Breiman A, Smith GL. Vaccinia virus B5 protein affects the glycosylation, localization and stability of the A34 protein. J Gen Virol 2010; 91:1823-7. [PMID: 20200189 PMCID: PMC3052527 DOI: 10.1099/vir.0.020677-0] [Citation(s) in RCA: 15] [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: 02/02/2010] [Accepted: 03/02/2010] [Indexed: 11/18/2022] Open
Abstract
Vaccinia virus has two infectious forms, the intracellular mature virus, which has a single envelope, and the extracellular enveloped virus (EEV), which is surrounded by two lipid bilayers. The outer membrane of the EEV contains at least six viral proteins. Among them A34, a type II membrane glycoprotein, and B5, a type I membrane glycoprotein, form a complex and are involved in processes such as morphogenesis and EEV entry. A34 is required for normal incorporation of B5 into the EEV membrane. Here, we used a virus lacking B5 and viruses with mutations in the B5 membrane-proximal stalk region and looked at the effect of those modifications on A34. Data presented show that B5 is required for the correct glycosylation, trafficking and stability of A34, emphasizing the complex interactions and mutual dependence of these vaccinia EEV proteins.
Collapse
Affiliation(s)
- Adrien Breiman
- Department of Virology, Faculty of Medicine, Imperial College London, St Mary's Campus, Norfolk Place, London W2 1PG, UK
| | | |
Collapse
|
27
|
Chan WM, Ward BM. There is an A33-dependent mechanism for the incorporation of B5-GFP into vaccinia virus extracellular enveloped virions. Virology 2010; 402:83-93. [PMID: 20378144 PMCID: PMC2872037 DOI: 10.1016/j.virol.2010.03.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2009] [Revised: 02/03/2010] [Accepted: 03/10/2010] [Indexed: 11/23/2022]
Abstract
Orthopoxviruses produce two, antigenically distinct, infectious virions, intracellular mature virions and extracellular virions (EV). A33 and B5 are found on EV but not on intracellular mature virions. To investigate the function of A33, a recombinant virus that has A33R deleted and expresses B5R-GFP (vB5R-GFP/DeltaA33R) was generated. A comparison of vB5R-GFP/DeltaA33R to an analogous virus (vDeltaA33R) revealed an additional defect in infectious EV production that was not apparent when A33R was present. Characterization of these recombinants revealed that EV produced in the absence of A33 had undetectable levels of B5-GFP. Both recombinants released similar amounts of EV but there were differences in their infectivity. Approximately equal numbers of virions produced by these recombinants were able to bind cells even though EV produced by vB5R-GFP/DeltaA33R do not contain B5. These results suggest that in the absence of A33, the cytoplasmic tail of B5 contributes to its incorporation into the envelope of progeny virions.
Collapse
Affiliation(s)
- Winnie M. Chan
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York 14642, USA
| | - Brian M. Ward
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York 14642, USA
| |
Collapse
|
28
|
Sandgren KJ, Wilkinson J, Miranda-Saksena M, McInerney GM, Byth-Wilson K, Robinson PJ, Cunningham AL. A differential role for macropinocytosis in mediating entry of the two forms of vaccinia virus into dendritic cells. PLoS Pathog 2010; 6:e1000866. [PMID: 20421949 PMCID: PMC2858709 DOI: 10.1371/journal.ppat.1000866] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2009] [Accepted: 03/22/2010] [Indexed: 11/18/2022] Open
Abstract
Vaccinia virus (VACV) is being developed as a recombinant viral vaccine vector for several key pathogens. Dendritic cells (DCs) are specialised antigen presenting cells that are crucial for the initiation of primary immune responses; however, the mechanisms of uptake of VACV by these cells are unclear. Therefore we examined the binding and entry of both the intracellular mature virus (MV) and extracellular enveloped virus (EV) forms of VACV into vesicular compartments of monocyte-derived DCs. Using a panel of inhibitors, flow cytometry and confocal microscopy we have shown that neither MV nor EV binds to the highly expressed C-type lectin receptors on DCs that are responsible for capturing many other viruses. We also found that both forms of VACV enter DCs via a clathrin-, caveolin-, flotillin- and dynamin-independent pathway that is dependent on actin, intracellular calcium and host-cell cholesterol. Both MV and EV entry were inhibited by the macropinocytosis inhibitors rottlerin and dimethyl amiloride and depended on phosphotidylinositol-3-kinase (PI(3)K), and both colocalised with dextran but not transferrin. VACV was not delivered to the classical endolysosomal pathway, failing to colocalise with EEA1 or Lamp2. Finally, expression of early viral genes was not affected by bafilomycin A, indicating that the virus does not depend on low pH to deliver cores to the cytoplasm. From these collective results we conclude that VACV enters DCs via macropinocytosis. However, MV was consistently less sensitive to inhibition and is likely to utilise at least one other entry pathway. Definition and future manipulation of these pathways may assist in enhancing the activity of recombinant vaccinia vectors through effects on antigen presentation. Vaccinia virus (VACV) is a relative of the smallpox virus and was used for many decades as a successful vaccine that contributed to the eradication of smallpox. Today, through genetic recombination technology, VACV shows potential as a modern vaccine for many unconquered diseases including HIV and cancer. Dendritic cells (DCs) are a specialised subset of immune cells that initiate adaptive immune responses and exploiting the interaction between VACV and DCs, which has not been well studied, may be a key to improving the efficacy of these vaccines. In this study we investigated the mechanisms by which VACV binds to and enters DCs. Here, we examined both the abundant mature virus form of VACV as well as the less common, poorly studied extracellular form. We found that VACV does not bind to the common pathogen-uptake C-type lectin receptors expressed on DCs and that the virus enters DCs via macropinocytosis—a fluid-phase uptake process. Furthermore, the virus is not delivered to the conventional endolysosomal antigen processing pathway in these cells. Our study provides new insights into VACV biology and into possible mechanisms of action of VACV as a recombinant viral vaccine vector which may assist in their rational design in the future.
Collapse
Affiliation(s)
- Kerrie J. Sandgren
- Centre for Virus Research, Westmead Millennium Institute, Sydney, New South Wales, Australia
- Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - John Wilkinson
- Centre for Virus Research, Westmead Millennium Institute, Sydney, New South Wales, Australia
| | - Monica Miranda-Saksena
- Centre for Virus Research, Westmead Millennium Institute, Sydney, New South Wales, Australia
| | - Gerald M. McInerney
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Karen Byth-Wilson
- Centre for Virus Research, Westmead Millennium Institute, Sydney, New South Wales, Australia
| | - Phillip J. Robinson
- Children's Medical Research Institute, Westmead, Sydney, New South Wales, Australia
| | - Anthony L. Cunningham
- Centre for Virus Research, Westmead Millennium Institute, Sydney, New South Wales, Australia
- * E-mail:
| |
Collapse
|
29
|
Lipid membranes in poxvirus replication. Viruses 2010; 2:972-986. [PMID: 21994664 PMCID: PMC3185658 DOI: 10.3390/v2040972] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2010] [Revised: 03/26/2010] [Accepted: 03/30/2010] [Indexed: 01/25/2023] Open
Abstract
Poxviruses replicate in the cytoplasm, where they acquire multiple lipoprotein membranes. Although a proposal that the initial membrane arises de novo has not been substantiated, there is no accepted explanation for its formation from cellular membranes. A subsequent membrane-wrapping step involving modified trans-Golgi or endosomal cisternae results in a particle with three membranes. These wrapped virions traverse the cytoplasm on microtubules; the outermost membrane is lost during exocytosis, the middle one is lost just prior to cell entry, and the remaining membrane fuses with the cell to allow the virus core to enter the cytoplasm and initiate a new infection.
Collapse
|