1
|
Yang P, Wang ZJ, Lu HT, Feng XM, Ye JL, Wang G, Qin CF, Ye Q, Liu ZY. Imaging of viral replication in live cells by using split fluorescent protein-tagged reporter flaviviruses. Virology 2025; 603:110374. [PMID: 39754862 DOI: 10.1016/j.virol.2024.110374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/09/2024] [Accepted: 12/20/2024] [Indexed: 01/06/2025]
Abstract
The knowledge on the life cycle of flaviviruses is still incomplete, and no direct-acting antivirals against their infections are clinically available. Herein, by screening via a Zika virus (ZIKV) replicon assay, we found that the N-terminus of NS2A exhibited great tolerance to the insertions of different split fluorescent proteins (split-FPs). Furthermore, both ZIKV and dengue virus encoding a split-FP-tagged NS2A propagated efficiently, and the split-FP-tagged ZIKVs had good genetic stability. Robust green fluorescence was observed in the reporter cell lines infected with these viruses and the fluorescence responded to anti-flavivirus chemicals with high specificity and sensitivity. Moreover, the sites of viral RNA replication were illuminated in live cells. Interestingly, by blocking viral RNA synthesis with an NS5 inhibitor, we found a correlation between the morphological characteristics of potential replication organelles and RNA amplification, highlighting that the NS2A-tagged viruses are of great value for the in-depth understanding of flavivirus replication mechanisms.
Collapse
Affiliation(s)
- Ping Yang
- The Centre for Infection and Immunity Studies, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Zheng-Jian Wang
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Hai-Tao Lu
- The Centre for Infection and Immunity Studies, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Xu-Meng Feng
- The Centre for Infection and Immunity Studies, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Jing-Long Ye
- The Centre for Infection and Immunity Studies, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Guangchuan Wang
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Cheng-Feng Qin
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Qing Ye
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China.
| | - Zhong-Yu Liu
- The Centre for Infection and Immunity Studies, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China.
| |
Collapse
|
2
|
Ackermann-Gäumann R, Dentand A, Lienhard R, Saeed M, Speiser DE, MacDonald MR, Coste AT, Cagno V. A reporter virus particle seroneutralization assay for tick-borne encephalitis virus overcomes ELISA limitations. J Med Virol 2024; 96:e29843. [PMID: 39092814 DOI: 10.1002/jmv.29843] [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: 03/15/2024] [Revised: 07/12/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024]
Abstract
Tick-borne encephalitis (TBE) virus is the most prevalent tick-transmitted orthoflavivirus in Europe. Due to the nonspecific nature of its symptoms, TBE is primarily diagnosed by ELISA-based detection of specific antibodies in the patient serum. However, cross-reactivity between orthoflaviviruses complicates the diagnosis. Specificity issues may be mitigated by serum neutralization assays (SNT), although the handling of clinically relevant orthoflaviviruses requires biosafety level (BSL) 3 conditions and they have highly divergent viral kinetics and cell tropisms. In the present study, we established a reporter virus particle (RVP)-based SNT in which the infectivity is measured by luminescence and that can be performed under BSL-2 conditions. The RVP-based SNT for TBEV exhibited a highly significant correlation with the traditional virus-based SNT (R2 = 0.8637, p < 0.0001). The RVP-based assay demonstrated a sensitivity of 92.3% (95% CI: 79.7%-97.4%) and specificity of 100% (95% CI: 81.6%-100%). We also tested the cross-reactivity of serum samples in RVP-based assays against other orthoflaviviruses (yellow fever virus, dengue virus type 2, Zika virus, West Nile virus and Japanese encephalitis virus). Interestingly, all serum samples which had tested TBEV-positive by ELISA but negative by RVP-based SNT were reactive for antibodies against other orthoflaviviruses. Thus, the RVP-based seroneutralization assay provides an added value in clinical diagnostics as well as in epidemiological studies.
Collapse
Affiliation(s)
- Rahel Ackermann-Gäumann
- Swiss National Reference Centre for Tick-Transmitted Diseases, Lausanne, Switzerland
- ADMED Microbiologie, La Chaux-de-Fonds, Switzerland
| | - Alexis Dentand
- Institute of Microbiology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Reto Lienhard
- Swiss National Reference Centre for Tick-Transmitted Diseases, Lausanne, Switzerland
- ADMED Microbiologie, La Chaux-de-Fonds, Switzerland
| | - Mohsan Saeed
- Department of Biochemistry & Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts, USA
| | - Daniel E Speiser
- Institute of Microbiology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Margaret R MacDonald
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, New York, USA
| | - Alix T Coste
- Swiss National Reference Centre for Tick-Transmitted Diseases, Lausanne, Switzerland
- Institute of Microbiology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Valeria Cagno
- Swiss National Reference Centre for Tick-Transmitted Diseases, Lausanne, Switzerland
- Institute of Microbiology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
3
|
Nie MS, Li XH, Zhang S, Zeng DD, Cai YR, Peng DX, Jiang T, Shi JP, Li J. Screening for anti-influenza virus compounds from traditional Mongolian medicine by GFP-based reporter virus. Front Cell Infect Microbiol 2024; 14:1431979. [PMID: 39071166 PMCID: PMC11272615 DOI: 10.3389/fcimb.2024.1431979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 06/25/2024] [Indexed: 07/30/2024] Open
Abstract
Introduction Screening for effective antiviral compounds from traditional Mongolian medicine not only aids in the research of antiviral mechanisms of traditional medicines, but is also of significant importance for the development of new antiviral drugs targeting influenza A virus. Our study aimed to establish high-throughput, rapid screening methods for antiviral compounds against influenza A virus from abundant resources of Mongolian medicine. Methods The use of GFP-based reporter viruses plays a pivotal role in antiviral drugs screening by enabling rapid and precise identification of compounds that inhibit viral replication. Herein, a GFP-based reporter influenza A virus was used to identify potent anti-influenza compounds within traditional Mongolian medicine. Results Our study led to the discovery of three active compounds: Cardamonin, Curcumin, and Kaempferide, all of which exhibited significant antiviral properties in vitro. Subsequent analysis confirmed that their effectiveness was largely due to the stimulation of the antiviral signaling pathways of host cells, rather than direct interference with the viral components, such as the viral polymerase. Discussion This study showcased the use of GFP-based reporter viruses in high-throughput screening to unearth antiviral agents from traditional Mongolian medicine, which contains rich antiviral compounds and deserves further exploration. Despite certain limitations, fluorescent reporter viruses present substantial potential for antiviral drug screening research due to their high throughput and efficiency.
Collapse
Affiliation(s)
- Mao-Shun Nie
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Xiao-He Li
- College of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, China
| | - Sen Zhang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Dan-Dan Zeng
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Yu-Rong Cai
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Da-Xin Peng
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Tao Jiang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Jian-Ping Shi
- College of Traditional Chinese Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Jing Li
- College of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, China
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| |
Collapse
|
4
|
King CR, Dodge MJ, MacNeil KM, Tessier TM, Mymryk JS, Mehle A. Expanding the adenovirus toolbox: reporter viruses for studying the dynamics of human adenovirus replication. J Virol 2024; 98:e0020724. [PMID: 38639487 PMCID: PMC11092356 DOI: 10.1128/jvi.00207-24] [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: 01/30/2024] [Accepted: 03/21/2024] [Indexed: 04/20/2024] Open
Abstract
To streamline standard virological assays, we developed a suite of nine fluorescent or bioluminescent replication competent human species C5 adenovirus reporter viruses that mimic their parental wild-type counterpart. These reporter viruses provide a rapid and quantitative readout of various aspects of viral infection and replication based on EGFP, mCherry, or NanoLuc measurement. Moreover, they permit real-time non-invasive measures of viral load, replication dynamics, and infection kinetics over the entire course of infection, allowing measurements that were not previously possible. This suite of replication competent reporter viruses increases the ease, speed, and adaptability of standard assays and has the potential to accelerate multiple areas of human adenovirus research.IMPORTANCEIn this work, we developed a versatile toolbox of nine HAdV-C5 reporter viruses and validated their functions in cell culture. These reporter viruses provide a rapid and quantitative readout of various aspects of viral infection and replication based on EGFP, mCherry, or NanoLuc measurement. The utility of these reporter viruses could also be extended for use in 3D cell culture, organoids, live cell imaging, or animal models, and provides a conceptual framework for the development of new reporter viruses representing other clinically relevant HAdV species.
Collapse
Affiliation(s)
- Cason R. King
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Mackenzie J. Dodge
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Katelyn M. MacNeil
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Tanner M. Tessier
- Division of Protective Immunity, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Joe S. Mymryk
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
- Department of Oncology, University of Western Ontario, London, Ontario, Canada
- Department of Otolaryngology, University of Western Ontario, London, Ontario, Canada
- London Regional Cancer Program, Lawson Health Research Institute, London, Ontario, Canada
| | - Andrew Mehle
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
5
|
Tamura T, Yamamoto H, Ogino S, Morioka Y, Tsujino S, Suzuki R, Hiono T, Suzuki S, Isoda N, Sakoda Y, Fukuhara T. A rapid and versatile reverse genetics approach for generating recombinant positive-strand RNA viruses that use IRES-mediated translation. J Virol 2024; 98:e0163823. [PMID: 38353536 PMCID: PMC10949505 DOI: 10.1128/jvi.01638-23] [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: 10/19/2023] [Accepted: 01/24/2024] [Indexed: 03/20/2024] Open
Abstract
Reverse genetics systems have played a central role in developing recombinant viruses for a wide spectrum of virus research. The circular polymerase extension reaction (CPER) method has been applied to studying positive-strand RNA viruses, allowing researchers to bypass molecular cloning of viral cDNA clones and thus leading to the rapid generation of recombinant viruses. However, thus far, the CPER protocol has only been established using cap-dependent RNA viruses. Here, we demonstrate that a modified version of the CPER method can be successfully applied to positive-strand RNA viruses that use cap-independent, internal ribosomal entry site (IRES)-mediated translation. As a proof-of-concept, we employed mammalian viruses with different types (classes I, II, and III) of IRES to optimize the CPER method. Using the hepatitis C virus (HCV, class III), we found that inclusion in the CPER assembly of an RNA polymerase I promoter and terminator, instead of those from polymerase II, allowed greater viral production. This approach was also successful in generating recombinant bovine viral diarrhea virus (class III) following transfection of MDBK/293T co-cultures to overcome low transfection efficiency. In addition, we successfully generated the recombinant viruses from clinical specimens. Our modified CPER could be used for producing hepatitis A virus (HAV, type I) as well as de novo generation of encephalomyocarditis virus (type II). Finally, we generated recombinant HCV and HAV reporter viruses that exhibited replication comparable to that of the wild-type parental viruses. The recombinant HAV reporter virus helped evaluate antivirals. Taking the findings together, this study offers methodological advances in virology. IMPORTANCE The lack of versatility of reverse genetics systems remains a bottleneck in viral research. Especially when (re-)emerging viruses reach pandemic levels, rapid characterization and establishment of effective countermeasures using recombinant viruses are beneficial in disease control. Indeed, numerous studies have attempted to establish and improve the methods. The circular polymerase extension reaction (CPER) method has overcome major obstacles in generating recombinant viruses. However, this method has not yet been examined for positive-strand RNA viruses that use cap-independent, internal ribosome entry site-mediated translation. Here, we engineered a suitable gene cassette to expand the CPER method for all positive-strand RNA viruses. Furthermore, we overcame the difficulty of generating recombinant viruses because of low transfection efficiency. Using this modified method, we also successfully generated reporter viruses and recombinant viruses from a field sample without virus isolation. Taking these findings together, our adapted methodology is an innovative technology that could help advance virologic research.
Collapse
Affiliation(s)
- Tomokazu Tamura
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
- Institute for Vaccine Research and Development (IVReD), Hokkaido University, Sapporo, Japan
- One Health Research Center, Hokkaido University, Sapporo, Japan
| | - Hirotaka Yamamoto
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Saho Ogino
- Laboratory of Microbiology, Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Yuhei Morioka
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
- Laboratory of Virus Control, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Shuhei Tsujino
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Rigel Suzuki
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
- Institute for Vaccine Research and Development (IVReD), Hokkaido University, Sapporo, Japan
| | - Takahiro Hiono
- Institute for Vaccine Research and Development (IVReD), Hokkaido University, Sapporo, Japan
- One Health Research Center, Hokkaido University, Sapporo, Japan
- Laboratory of Microbiology, Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Saori Suzuki
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
- Institute for Vaccine Research and Development (IVReD), Hokkaido University, Sapporo, Japan
| | - Norikazu Isoda
- Institute for Vaccine Research and Development (IVReD), Hokkaido University, Sapporo, Japan
- One Health Research Center, Hokkaido University, Sapporo, Japan
- Laboratory of Microbiology, Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Yoshihiro Sakoda
- Institute for Vaccine Research and Development (IVReD), Hokkaido University, Sapporo, Japan
- One Health Research Center, Hokkaido University, Sapporo, Japan
- Laboratory of Microbiology, Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Takasuke Fukuhara
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
- Institute for Vaccine Research and Development (IVReD), Hokkaido University, Sapporo, Japan
- One Health Research Center, Hokkaido University, Sapporo, Japan
- Laboratory of Virus Control, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- AMED-CREST, Japan Agency for Medical Research and Development (AMED), Tokyo, Japan
| |
Collapse
|
6
|
Li C, Zhang L, Chen X, Jiang D, Hu J, Guo J, Ding J, Jiao X, Bao W, Li Y. Evaluation of the genotype I Japanese encephalitis virus as a stable viral vector for foreign gene expression. Antiviral Res 2023:105652. [PMID: 37301446 DOI: 10.1016/j.antiviral.2023.105652] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/31/2023] [Accepted: 06/03/2023] [Indexed: 06/12/2023]
Abstract
Manipulation of the flavivirus genome to accommodate and express a heterologous gene of interest has become an attractive approach for gene delivery and the development of viral-vectored vaccines. However, due to the inherent genetic instability of the flavivirus genomes, the construction of recombinant viruses carrying a foreign gene could be problematic and heavily resistant. In this study, the possibility of the Japanese encephalitis virus (JEV) as a stable flavivirus vector for the expression of a foreign gene was assessed using reverse genetics. The full-length cDNA genome of genotype I (GI) JEV inherently possessed excellent stability and manipulability in a bacterial host, while mutations and deletions accumulated in the cDNA genomes of genotype Ⅲ (GⅢ) JEV strains. Using the GI JEV as backbones, we generate a panel of recombinant viruses expressing various foreign genes. All recombinant viruses exhibited excellent genetic stability and efficiently express foreign genes for at least ten serial passages in vitro. In application, a convenient, rapid and reliable image-based assay for neutralizing antibody testing and antiviral drug discovery was established with a mCherry-reporter recombinant virus (rBJ-mCherry). Meanwhile, the recombinant viruses expressing the antigens of the African swine fever virus (ASFV) or Classical swine fever virus (CSFV) could effectively induce antibody responses to the JEV vector and foreign antigens in a mouse vaccination model. Therefore, GI JEV strains could serve as viral vectors accommodating the expression of large foreign genes.
Collapse
Affiliation(s)
- Chenxi Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China; Comparative Medicine Research Institute, Yangzhou University, Yangzhou, 225009, China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
| | - Linjie Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Xuan Chen
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Daoyuan Jiang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Jingbo Hu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Jinyao Guo
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Jingjing Ding
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Xue Jiao
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Wenbin Bao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Yanhua Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China; Comparative Medicine Research Institute, Yangzhou University, Yangzhou, 225009, China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
| |
Collapse
|
7
|
Kobayashi S, Fukuda Y, Yoshii K, Thammahakin P, Maezono K, Eyer L, Růžek D, Kariwa H. Development of recombinant West Nile virus expressing mCherry reporter protein. J Virol Methods 2023; 317:114744. [PMID: 37119976 DOI: 10.1016/j.jviromet.2023.114744] [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: 01/25/2023] [Revised: 03/30/2023] [Accepted: 04/27/2023] [Indexed: 05/01/2023]
Abstract
West Nile virus (WNV) is transmitted to humans and animals by a mosquito and enters the central nervous system, leading to lethal encephalitis. Reporter viruses expressing fluorescent proteins enable detection of infected cells in vitro and in vivo, facilitating evaluation of the dynamics of viral infection, and the development of diagnostic or therapeutic methods. In this study, we developed a method for production of a recombinant replication-competent WNV expressing mCherry fluorescent protein. The expression of mCherry was observed in viral antigen-positive cells in vitro and in vivo, but the growth of the reporter WNV was reduced as compared to the parental WNV. The expression of mCherry was stable during 5 passages in reporter WNV-infected culture cells. Neurological symptoms were observed in mice inoculated intracranially with the reporter WNV. The reporter WNV expressing mCherry will facilitate research into WNV replication in mouse brains.
Collapse
Affiliation(s)
- Shintaro Kobayashi
- Laboratory of Public Health, Faculty of Veterinary medicine, Hokkaido University, Sapporo, Japan.
| | - Yukine Fukuda
- Laboratory of Public Health, Faculty of Veterinary medicine, Hokkaido University, Sapporo, Japan
| | - Kentaro Yoshii
- National Research Center for the Control and Prevention of Infectious diseases (CCPID), Nagasaki University, Nagasaki, Japan
| | - Passawat Thammahakin
- Laboratory of Public Health, Faculty of Veterinary medicine, Hokkaido University, Sapporo, Japan
| | - Keisuke Maezono
- Laboratory of Public Health, Faculty of Veterinary medicine, Hokkaido University, Sapporo, Japan
| | - Luděk Eyer
- Department of Virology, Veterinary Research Institute, Brno, Czech Republic; Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Ceske Budejovice, Czech Republic; Department of Experimental Biology, Faculty of Science, Masaryk University
| | - Daniel Růžek
- Department of Virology, Veterinary Research Institute, Brno, Czech Republic; Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Ceske Budejovice, Czech Republic; Department of Experimental Biology, Faculty of Science, Masaryk University
| | - Hiroaki Kariwa
- Laboratory of Public Health, Faculty of Veterinary medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
8
|
Holliday M, Corliss L, Lennemann NJ. Construction and Rescue of a DNA-Launched DENV2 Infectious Clone. Viruses 2023; 15:275. [PMID: 36851490 PMCID: PMC9959642 DOI: 10.3390/v15020275] [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: 08/29/2022] [Revised: 12/15/2022] [Accepted: 01/13/2023] [Indexed: 01/20/2023] Open
Abstract
Flaviviruses represent a large group of globally significant, insect-borne pathogens. For many of these viruses, there is a lack of antivirals and vaccines. Thus, there is a need to continue the development of tools to further advance our efforts to combat these pathogens, including reverse genetics techniques. Traditionally, reverse genetics methods for flaviviruses rely on producing infectious RNA from in vitro transcription reactions followed by electroporation or transfection into permissive cell lines. However, the production of Zika virus has been successful from CMV promoter-driven expression plasmids, which provides cost and time advantages. In this report, we describe the design and construction of a DNA-launched infectious clone for dengue virus (DENV) serotype 2 strain 16681. An artificial intron was introduced in the nonstructural protein 1 segment of the viral genome to promote stability in bacteria. We found that rescued viruses maintained the ability to form plaques and replicate efficiently in commonly used cell lines. Thus, we present a rapid and cost-effective method for producing DENV2 strain 16681 from plasmid DNA. This construct will be a useful platform for the continued development of anti-DENV therapeutics and vaccines.
Collapse
Affiliation(s)
| | | | - Nicholas J. Lennemann
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
9
|
A Recombinant Genotype I Japanese Encephalitis Virus Expressing a Gaussia Luciferase Gene for Antiviral Drug Screening Assay and Neutralizing Antibodies Detection. Int J Mol Sci 2022; 23:ijms232415548. [PMID: 36555192 PMCID: PMC9778660 DOI: 10.3390/ijms232415548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/05/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
Japanese encephalitis virus (JEV) is the major cause of viral encephalitis in humans throughout Asia. In the past twenty years, the emergence of the genotype I (GI) JEV as the dominant genotype in Asian countries has raised a significant threat to public health security. However, no clinically approved drug is available for the specific treatment of JEV infection, and the commercial vaccines derived from the genotype III JEV strains merely provided partial protection against the GI JEV. Thus, an easy-to-perform platform in high-throughput is urgently needed for the antiviral drug screening and assessment of neutralizing antibodies specific against the GI JEV. In this study, we established a reverse genetics system for the GI JEV strain (YZ-1) using a homologous recombination strategy. Using this reverse genetic system, a gaussia luciferase (Gluc) expression cassette was inserted into the JEV genome to generate a reporter virus (rGI-Gluc). The reporter virus exhibited similar growth kinetics to the parental virus and remained genetically stable for at least ten passages in vitro. Of note, the bioluminescence signal strength of Gluc in the culture supernatants was well correlated with the viral progenies determined by viral titration. Taking advantage of this reporter virus, we established Gluc readout-based assays for antiviral drug screening and neutralizing antibody detection against the GI JEV. These Gluc readout-based assays exhibited comparable performance to the assays using an actual virus and are less time consuming and are applicable for a high-throughput format. Taken together, we generated a GI JEV reporter virus expressing a Gluc gene that could be a valuable tool for an antiviral drug screening assay and neutralization assay.
Collapse
|
10
|
Reverse genetics in virology: A double edged sword. BIOSAFETY AND HEALTH 2022. [DOI: 10.1016/j.bsheal.2022.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
11
|
Fang E, Liu X, Li M, Liu J, Zhang Z, Liu X, Li X, Li W, Peng Q, Yu Y, Li Y. Construction of a Dengue NanoLuc Reporter Virus for In Vivo Live Imaging in Mice. Viruses 2022; 14:v14061253. [PMID: 35746724 PMCID: PMC9230669 DOI: 10.3390/v14061253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 02/06/2023] Open
Abstract
Since the first isolation in 1943, the dengue virus (DENV) has spread throughout the world, but effective antiviral drugs or vaccines are still not available. To provide a more stable reporter DENV for vaccine development and antiviral drug screening, we constructed a reporter DENV containing the NanoLuc reporter gene, which was inserted into the 5′ untranslated region and capsid junction region, enabling rapid virus rescue by in vitro ligation. In addition, we established a live imaging mouse model and found that the reporter virus maintained the neurovirulence of prototype DENV before engineering. DENV-4 exhibited dramatically increased neurovirulence following a glycosylation site-defective mutation in the envelope protein. Significant mice mortality with neurological onset symptoms was observed after intracranial infection of wild-type (WT) mice, thus providing a visualization tool for DENV virulence assessment. Using this model, DENV was detected in the intestinal tissues of WT mice after infection, suggesting that intestinal lymphoid tissues play an essential role in DENV pathogenesis.
Collapse
Affiliation(s)
- Enyue Fang
- Department of Arbovirus Vaccine, National Institutes for Food and Drug Control, Beijing 102629, China; (E.F.); (X.L.); (M.L.); (J.L.); (Z.Z.); (X.L.); (X.L.); (W.L.); (Q.P.)
- Wuhan Institute of Biological Products, Co., Ltd., Wuhan 430207, China
| | - Xiaohui Liu
- Department of Arbovirus Vaccine, National Institutes for Food and Drug Control, Beijing 102629, China; (E.F.); (X.L.); (M.L.); (J.L.); (Z.Z.); (X.L.); (X.L.); (W.L.); (Q.P.)
| | - Miao Li
- Department of Arbovirus Vaccine, National Institutes for Food and Drug Control, Beijing 102629, China; (E.F.); (X.L.); (M.L.); (J.L.); (Z.Z.); (X.L.); (X.L.); (W.L.); (Q.P.)
| | - Jingjing Liu
- Department of Arbovirus Vaccine, National Institutes for Food and Drug Control, Beijing 102629, China; (E.F.); (X.L.); (M.L.); (J.L.); (Z.Z.); (X.L.); (X.L.); (W.L.); (Q.P.)
| | - Zelun Zhang
- Department of Arbovirus Vaccine, National Institutes for Food and Drug Control, Beijing 102629, China; (E.F.); (X.L.); (M.L.); (J.L.); (Z.Z.); (X.L.); (X.L.); (W.L.); (Q.P.)
| | - Xinyu Liu
- Department of Arbovirus Vaccine, National Institutes for Food and Drug Control, Beijing 102629, China; (E.F.); (X.L.); (M.L.); (J.L.); (Z.Z.); (X.L.); (X.L.); (W.L.); (Q.P.)
| | - Xingxing Li
- Department of Arbovirus Vaccine, National Institutes for Food and Drug Control, Beijing 102629, China; (E.F.); (X.L.); (M.L.); (J.L.); (Z.Z.); (X.L.); (X.L.); (W.L.); (Q.P.)
| | - Wenjuan Li
- Department of Arbovirus Vaccine, National Institutes for Food and Drug Control, Beijing 102629, China; (E.F.); (X.L.); (M.L.); (J.L.); (Z.Z.); (X.L.); (X.L.); (W.L.); (Q.P.)
| | - Qinhua Peng
- Department of Arbovirus Vaccine, National Institutes for Food and Drug Control, Beijing 102629, China; (E.F.); (X.L.); (M.L.); (J.L.); (Z.Z.); (X.L.); (X.L.); (W.L.); (Q.P.)
| | - Yongxin Yu
- Department of Arbovirus Vaccine, National Institutes for Food and Drug Control, Beijing 102629, China; (E.F.); (X.L.); (M.L.); (J.L.); (Z.Z.); (X.L.); (X.L.); (W.L.); (Q.P.)
- Correspondence: (Y.Y.); (Y.L.); Tel.: +86-010-5385-2137 (Y.Y.); +86-010-5385-2128 (Y.L.)
| | - Yuhua Li
- Department of Arbovirus Vaccine, National Institutes for Food and Drug Control, Beijing 102629, China; (E.F.); (X.L.); (M.L.); (J.L.); (Z.Z.); (X.L.); (X.L.); (W.L.); (Q.P.)
- Correspondence: (Y.Y.); (Y.L.); Tel.: +86-010-5385-2137 (Y.Y.); +86-010-5385-2128 (Y.L.)
| |
Collapse
|
12
|
Tracking the Replication-Competent Zika Virus with Tetracysteine-Tagged Capsid Protein in Living Cells. J Virol 2022; 96:e0184621. [PMID: 35285687 PMCID: PMC9006885 DOI: 10.1128/jvi.01846-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Zika virus (ZIKV) is the mosquito-borne enveloped flavivirus that causes microcephaly in neonates. While real-time imaging plays a critical role in dissecting viral biology, no fluorescent, genetically engineered ZIKV for single-particle tracking is currently available.
Collapse
|
13
|
Vrba SM, Hickman HD. Imaging viral infection in vivo to gain unique perspectives on cellular antiviral immunity. Immunol Rev 2022; 306:200-217. [PMID: 34796538 PMCID: PMC9073719 DOI: 10.1111/imr.13037] [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: 10/14/2021] [Accepted: 10/17/2021] [Indexed: 11/29/2022]
Abstract
The past decade has seen near continual global public health crises caused by emerging viral infections. Extraordinary increases in our knowledge of the mechanisms underlying successful antiviral immune responses in animal models and during human infection have accompanied these viral outbreaks. Keeping pace with the rapidly advancing field of viral immunology, innovations in microscopy have afforded a previously unseen view of viral infection occurring in real-time in living animals. Here, we review the contribution of intravital imaging to our understanding of cell-mediated immune responses to viral infections, with a particular focus on studies that visualize the antiviral effector cells responding to infection as well as virus-infected cells. We discuss methods to visualize viral infection in vivo using intravital microscopy (IVM) and significant findings arising through the application of IVM to viral infection. Collectively, these works underscore the importance of developing a comprehensive spatial understanding of the relationships between immune effectors and virus-infected cells and how this has enabled unique discoveries about virus/host interactions and antiviral effector cell biology.
Collapse
Affiliation(s)
- Sophia M. Vrba
- Laboratory of Clinical Immunology and Microbiology, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Heather D. Hickman
- Laboratory of Clinical Immunology and Microbiology, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
14
|
Jimenez-Aleman GH, Castro V, Londaitsbehere A, Gutierrez-Rodríguez M, Garaigorta U, Solano R, Gastaminza P. SARS-CoV-2 Fears Green: The Chlorophyll Catabolite Pheophorbide A Is a Potent Antiviral. Pharmaceuticals (Basel) 2021; 14:ph14101048. [PMID: 34681272 PMCID: PMC8538351 DOI: 10.3390/ph14101048] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 10/10/2021] [Accepted: 10/12/2021] [Indexed: 12/20/2022] Open
Abstract
SARS-CoV-2 pandemic is having devastating consequences worldwide. Although vaccination advances at good pace, effectiveness against emerging variants is unpredictable. The virus has displayed a remarkable resistance to treatments and no drugs have been proved fully effective against COVID-19. Thus, despite the international efforts, there is still an urgent need for new potent and safe antivirals against SARS-CoV-2. Here, we exploited the enormous potential of plant metabolism using the bryophyte Marchantia polymorpha L. and identified a potent SARS-CoV-2 antiviral, following a bioactivity-guided fractionation and mass-spectrometry approach. We found that the chlorophyll derivative Pheophorbide a (PheoA), a porphyrin compound similar to animal Protoporphyrin IX, has an extraordinary antiviral activity against SARS-CoV-2, preventing infection of cultured monkey and human cells, without noticeable cytotoxicity. We also show that PheoA targets the viral particle, interfering with its infectivity in a dose- and time-dependent manner. Besides SARS-CoV-2, PheoA also displayed a broad-spectrum antiviral activity against enveloped RNA viral pathogens such as HCV, West Nile, and other coronaviruses. Our results indicate that PheoA displays a remarkable potency and a satisfactory therapeutic index, which together with its previous use in photoactivable cancer therapy in humans, suggest that it may be considered as a potential candidate for antiviral therapy against SARS-CoV-2.
Collapse
Affiliation(s)
- Guillermo H. Jimenez-Aleman
- National Centre for Biotechnology (CNB-CSIC), Department of Plant Molecular Genetics, 28049 Madrid, Spain; (G.H.J.-A.); (A.L.)
| | - Victoria Castro
- National Centre for Biotechnology (CNB-CSIC), Department of Cell & Molecular Biology, 28049 Madrid, Spain; (V.C.); (U.G.)
| | - Addis Londaitsbehere
- National Centre for Biotechnology (CNB-CSIC), Department of Plant Molecular Genetics, 28049 Madrid, Spain; (G.H.J.-A.); (A.L.)
| | - Marta Gutierrez-Rodríguez
- Medicinal Chemistry Institute (IQM-CSIC), Department of Biomimetics for Drug Discovery, 28006 Madrid, Spain;
| | - Urtzi Garaigorta
- National Centre for Biotechnology (CNB-CSIC), Department of Cell & Molecular Biology, 28049 Madrid, Spain; (V.C.); (U.G.)
| | - Roberto Solano
- National Centre for Biotechnology (CNB-CSIC), Department of Plant Molecular Genetics, 28049 Madrid, Spain; (G.H.J.-A.); (A.L.)
- Correspondence: (R.S.); (P.G.)
| | - Pablo Gastaminza
- National Centre for Biotechnology (CNB-CSIC), Department of Cell & Molecular Biology, 28049 Madrid, Spain; (V.C.); (U.G.)
- Correspondence: (R.S.); (P.G.)
| |
Collapse
|
15
|
Cimica V, Saleem S, Matuczinski E, Adams-Fish D, McMahon C, Rashid S, Stedman TT. A Virus-Like Particle-Based Vaccine Candidate against the Tick-Borne Powassan Virus Induces Neutralizing Antibodies in a Mouse Model. Pathogens 2021; 10:pathogens10060680. [PMID: 34072726 PMCID: PMC8229747 DOI: 10.3390/pathogens10060680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/20/2021] [Accepted: 05/25/2021] [Indexed: 12/30/2022] Open
Abstract
Powassan virus (POWV) is a tick-borne flavivirus circulating in North America and the Russian Far East that can cause severe neuroinvasive diseases, including encephalitis, meningitis, and meningoencephalitis. The reported neuroinvasive case fatality is about 10%, and approximately 50% of the survivors from the neuroinfection exhibit long-lasting or permanent neurological sequelae. Currently, treatment of POWV infection is supportive, and no FDA-approved vaccines or specific therapeutics are available. A novel Powassan vaccine candidate was created using virus-like particle technology (POW-VLP) and assembled with the viral structural proteins pre-Membrane (prM) and Envelope (E). Western blot immunoassay demonstrated high antigenicity of POW-VLP structural proteins. Transmission electron microscopy indicated that the POW-VLP exhibited icosahedral morphology typical of flaviviruses. A dose-escalation study in a murine model was performed to test immunogenicity and safety. Serum antibody was tested by ELISA, demonstrating that POW-VLP afforded 100% seroconversion to the E protein. Reporter viral-particle neutralization assay demonstrated high levels of neutralizing antibodies in the serum of immunized mice. Hybridomas expressing monoclonal antibodies were produced following POW-VLP immunization. The POW-VLP vaccine candidate created in this study provides a strategy for inducing protective antibodies against Powassan neuroinvasive infection.
Collapse
|
16
|
Bohning K, Sonnberg S, Chen HL, Zahralban-Steele M, Powell T, Hather G, Patel HK, Dean HJ. A high throughput reporter virus particle microneutralization assay for quantitation of Zika virus neutralizing antibodies in multiple species. PLoS One 2021; 16:e0250516. [PMID: 33891631 PMCID: PMC8064526 DOI: 10.1371/journal.pone.0250516] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 04/06/2021] [Indexed: 12/20/2022] Open
Abstract
Zika virus is a Flavivirus, transmitted via Aedes mosquitos, that causes a range of symptoms including Zika congenital syndrome. Zika has posed a challenging situation for health, public and economic sectors of affected countries. To quantitate Zika virus neutralizing antibody titers in serum samples, we developed a high throughput plate based Zika virus reporter virus particle (RVP) assay that uses an infective, non-replicating particle encoding Zika virus surface proteins and capsid (CprME) and a reporter gene (Renilla luciferase). This is the first characterization of a Zika virus RVP assay in 384-well format using a Dengue replicon Renilla reporter construct. Serially diluted test sera were incubated with RVPs, followed by incubation with Vero cells. RVPs that have not been neutralized by antibodies in the test sera entered the cells and expressed Renilla luciferase. Quantitative measurements of neutralizing activity were determined using a plate-based assay and commercially available substrate. The principle of limiting the infection to a single round increases the precision of the assay measurements. RVP log10EC50 titers correlated closely with titers determined using a plaque reduction neutralization test (PRNT) (R2>95%). The plate-based Zika virus RVP assay also demonstrated high levels of precision, reproducibility and throughput. The assay employs identical reagents for human, rhesus macaque and mouse serum matrices. Spiking studies indicated that the assay performs equally well in different species, producing comparable titers irrespective of the serum species. The assay is conducted in 384-well plates and can be automated to simultaneously achieve high throughput and high reproducibility.
Collapse
Affiliation(s)
- Kelly Bohning
- Takeda Vaccines, Inc., Cambridge, Massachusetts, United States of America
| | - Stephanie Sonnberg
- Takeda Vaccines, Inc., Cambridge, Massachusetts, United States of America
| | - Hui-Ling Chen
- Takeda Vaccines, Inc., Cambridge, Massachusetts, United States of America
| | | | - Timothy Powell
- Takeda Vaccines, Inc., Cambridge, Massachusetts, United States of America
| | - Greg Hather
- Takeda Vaccines, Inc., Cambridge, Massachusetts, United States of America
| | - Hetal K. Patel
- Takeda Vaccines, Inc., Cambridge, Massachusetts, United States of America
| | - Hansi J. Dean
- Takeda Vaccines, Inc., Cambridge, Massachusetts, United States of America
| |
Collapse
|
17
|
Bates TA, Chuong C, Hawks SA, Rai P, Duggal NK, Weger-Lucarelli J. Development and characterization of infectious clones of two strains of Usutu virus. Virology 2020; 554:28-36. [PMID: 33352463 DOI: 10.1016/j.virol.2020.12.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 12/02/2020] [Accepted: 12/07/2020] [Indexed: 12/23/2022]
Abstract
Usutu virus (USUV; genus Flavivirus; family Flaviviridae) is a mosquito-borne, positive-sense RNA virus that is currently causing significant die-offs in numerous bird species throughout Europe and has caused infections in humans. Currently, there are no molecular clones for USUV, hence, hindering studies on the pathogenesis and transmission of USUV. Here, we demonstrate the development and characterization of infectious clones for two modern strains of USUV isolated from Europe and Africa. We show that the infectious clone-derived viruses replicated similarly to the parental strains in mammalian and insect cells. Additionally, we observed similar levels of replication and disease in two mouse models. These clones will aid the study of USUV infection, transmission, diagnostics, and vaccines.
Collapse
Affiliation(s)
- Tyler A Bates
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA
| | - Christina Chuong
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA
| | - Seth A Hawks
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA
| | - Pallavi Rai
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA
| | - Nisha K Duggal
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA
| | - James Weger-Lucarelli
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA.
| |
Collapse
|
18
|
Baker C, Liu Y, Zou J, Muruato A, Xie X, Shi PY. Identifying optimal capsid duplication length for the stability of reporter flaviviruses. Emerg Microbes Infect 2020; 9:2256-2265. [PMID: 32981479 PMCID: PMC7594839 DOI: 10.1080/22221751.2020.1829994] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 09/20/2020] [Accepted: 09/21/2020] [Indexed: 01/04/2023]
Abstract
ABSTRACT Mosquito-transmitted flaviviruses cause widespread disease across the world. To provide better molecular tools for drug screens and pathogenesis studies, we report a new approach to produce stable NanoLuc-tagged flaviviruses, including dengue virus serotypes 1-4, Japanese encephalitis virus, yellow fever virus, West Nile virus, and Zika virus. Since the reporter gene is often engineered at the capsid gene region, the capsid sequence must be duplicated to flank the reporter gene; such capsid duplication is essential for viral replication. The conventional approach for stabilizing reporter flaviviruses has been to shorten or modify the duplicated capsid sequence to minimize homologous recombination. No study has examined the effects of capsid duplication length on reporter virus stability. Here we report an optimal length to stabilize reporter flaviviruses. These viruses were stable after ten rounds of cell culture passaging, and in the case of stable NanoLuc-tagged Zika virus (ZIKV C38), the virus replicated to 107 FFU/ml in cell culture and produced robust luciferase signal after inoculation in mosquitoes. Mechanistically, the optimal length of capsid duplication may contain all the cis-acting RNA elements required for viral RNA replication, thus reducing the selection pressure for recombination. Together, these data describe an improved method of constructing optimal reporter flaviviruses.
Collapse
Affiliation(s)
- Coleman Baker
- Departement of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Yang Liu
- Department of Biology and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Jing Zou
- Department of Biology and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Antonio Muruato
- Departement of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Xuping Xie
- Department of Biology and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Pei-Yong Shi
- Department of Biology and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, USA
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
- Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX, USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, USA
- Sealy Institute for Translational Sciences, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
19
|
Baker C, Shi PY. Construction of Stable Reporter Flaviviruses and Their Applications. Viruses 2020; 12:v12101082. [PMID: 32992987 PMCID: PMC7599567 DOI: 10.3390/v12101082] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 08/28/2020] [Accepted: 09/17/2020] [Indexed: 12/12/2022] Open
Abstract
Flaviviruses are significant human pathogens that cause frequent emerging and reemerging epidemics around the world. Better molecular tools for studying, diagnosing, and treating these diseases are needed. Reporter viruses represent potent tools to fill this gap but have been hindered by genetic instability. Recent advances have overcome these hurdles, opening the way for increased use of stable reporter flaviviruses to diagnose infections, screen and study antiviral compounds, and serve as potential vaccine vectors.
Collapse
Affiliation(s)
- Coleman Baker
- Microbiology and Immunology Department, University of Texas Medical Branch, Galveston, TX 77555, USA;
| | - Pei-Yong Shi
- Biochemistry and Molecular Biology Department, University of Texas Medical Branch, Galveston, TX 77555, USA
- Institute for Human Infections & Immunity, University of Texas Medical Branch, Galveston, TX 77555, USA
- Institute for Translational Science, University of Texas Medical Branch, Galveston, TX 77555, USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX 77555, USA
- Sealy Center for Structural Biology & Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555, USA
- Correspondence:
| |
Collapse
|
20
|
Baker C, Xie X, Zou J, Muruato A, Fink K, Shi PY. Using recombination-dependent lethal mutations to stabilize reporter flaviviruses for rapid serodiagnosis and drug discovery. EBioMedicine 2020; 57:102838. [PMID: 32574959 PMCID: PMC7317239 DOI: 10.1016/j.ebiom.2020.102838] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 05/28/2020] [Accepted: 05/29/2020] [Indexed: 01/11/2023] Open
Abstract
Background Many flaviviruses are significant human pathogens that cause global public health threats. Developing research tools for studying and diagnosing these pathogens is a top priority. Reporter flaviviruses are useful tools for studying viral pathogenesis, diagnosing disease, and screening antiviral compounds. However, the stability of reporter flaviviruses has been challenged by viral RNA recombination, leading to deletion of the engineered reporter gene during viral replication. The instability of reporter viruses has limited their application to research and countermeasure development. Thus, new approaches to overcome the instability of reporter flaviviruses are critically needed to advance the flavivirus field. Methods To create a stable flavivirus bearing a reporter gene, we engineered mutations in the viral capsid gene that are rendered virus-lethal upon recombination. Thus, only non-recombined reporter virus propagates. We tested this strategy using Zika virus (ZIKV) bearing a nano-luciferase (NanoLuc) gene and passaged both virus with capsid mutations and virus without mutations. Findings The recombination-dependent lethal mutations succeeded in stabilizing the NanoLuc ZIKV through ten passages, while WT reporter virus showed instability as early as five passages. The stability of NanoLuc ZIKV was supported by RT-PCR, sequencing, focus forming assay, and luciferase assay. The success of this method was reconfirmed by also establishing a stable NanoLuc Yellow Fever 17D virus, indicating that the recombination-dependent lethal approach can be applied to other flaviviruses. To demonstrate the utility of the stable reporter viruses, we showed that NanoLuc ZIKV and YFV17D could be used to measure neutralizing antibody titers with a turnaround time as short as four hours. Importantly, the neutralizing antibody titers derived from the reporter virus assay were equivalent to those derived from the conventional plaque assay, indicating the new assay maintains the gold standard of serology testing. Furthermore, using a known inhibitor, we showed that the reporter viruses could be reliably used for antiviral evaluation. Interpretation The study has developed a recombination-dependent lethal approach to produce stable reporter flaviviruses that may be used for rapid serodiagnosis, trans-gene delivery, vaccine evaluation, and antiviral discovery. Funding National Institute of Health, Robert J. Kleberg, Jr. and Helen C. Kleberg Foundation; John S. Dunn Foundation; Amon G. Carter Foundation; Gillson Longenbaugh Foundation; Summerfield G. Roberts Foundation.
Collapse
Affiliation(s)
- Coleman Baker
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Xuping Xie
- Department of Biology and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA.
| | - Jing Zou
- Department of Biology and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Antonio Muruato
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Katja Fink
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore
| | - Pei-Yong Shi
- Department of Biology and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA; Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA; Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX, USA; Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
21
|
Kautz TF, Jaworski E, Routh A, Forrester NL. A Low Fidelity Virus Shows Increased Recombination during the Removal of an Alphavirus Reporter Gene. Viruses 2020; 12:E660. [PMID: 32575413 PMCID: PMC7354468 DOI: 10.3390/v12060660] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/12/2020] [Accepted: 06/16/2020] [Indexed: 01/18/2023] Open
Abstract
Reporter genes for RNA viruses are well-known to be unstable due to putative RNA recombination events that excise inserted nucleic acids. RNA recombination has been demonstrated to be co-regulated with replication fidelity in alphaviruses, but it is unknown how recombination events at the minority variant level act, which is important for vaccine and trans-gene delivery design. Therefore, we sought to characterize the removal of a reporter gene by a low-fidelity alphavirus mutant over multiple replication cycles. To examine this, GFP was inserted into TC-83, a live-attenuated vaccine for the alphavirus Venezuelan equine encephalitis virus, as well as a low-fidelity variant of TC-83, and passaged until fluorescence was no longer observed. Short-read RNA sequencing using ClickSeq was performed to determine which regions of the viral genome underwent recombination and how this changed over multiple replication cycles. A rapid removal of the GFP gene was observed, where minority variants in the virus population accumulated small deletions that increased in size over the course of passaging. Eventually, these small deletions merged to fully remove the GFP gene. The removal was significantly enhanced during the passaging of low-fidelity TC-83, suggesting that increased levels of recombination are a defining characteristic of this mutant.
Collapse
Affiliation(s)
- Tiffany F Kautz
- Institute for Human Infections and Immunity, Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Elizabeth Jaworski
- Department of Biochemistry and Molecular Biology, University of Medical Branch, Galveston, TX 77555-0645, USA
| | - Andrew Routh
- Department of Biochemistry and Molecular Biology, University of Medical Branch, Galveston, TX 77555-0645, USA
| | - Naomi L Forrester
- Institute for Human Infections and Immunity, Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
- School of Life Sciences, University of Keele, Keele ST5 5BG, UK
| |
Collapse
|
22
|
Yun SI, Song BH, Woolley ME, Frank JC, Julander JG, Lee YM. Development, Characterization, and Application of Two Reporter-Expressing Recombinant Zika Viruses. Viruses 2020; 12:v12050572. [PMID: 32456014 PMCID: PMC7290298 DOI: 10.3390/v12050572] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 05/16/2020] [Accepted: 05/18/2020] [Indexed: 12/20/2022] Open
Abstract
Zika virus (ZIKV), a mosquito-borne transplacentally transmissible flavivirus, is an enveloped virus with an ~10.8 kb plus-strand RNA genome that can cause neurological disease. To facilitate the identification of potential antivirals, we developed two reporter-expressing ZIKVs, each capable of expressing an enhanced green fluorescent protein or an improved luminescent NanoLuc luciferase. First, a full-length functional ZIKV cDNA clone was engineered as a bacterial artificial chromosome, with each reporter gene under the cap-independent translational control of a cardiovirus-derived internal ribosome entry site inserted downstream of the single open reading frame of the viral genome. Two reporter-expressing ZIKVs were then generated by transfection of ZIKV-susceptible BHK-21 cells with infectious RNAs derived by in vitro run-off transcription from the respective cDNAs. As compared to the parental virus, the two reporter-expressing ZIKVs grew to lower titers with slower growth kinetics and formed smaller foci; however, they displayed a genome-wide viral protein expression profile identical to that of the parental virus, except for two previously unrecognized larger forms of the C and NS1 proteins. We then used the NanoLuc-expressing ZIKV to assess the in vitro antiviral activity of three inhibitors (T-705, NITD-008, and ribavirin). Altogether, our reporter-expressing ZIKVs represent an excellent molecular tool for the discovery of novel antivirals.
Collapse
Affiliation(s)
- Sang-Im Yun
- Department of Animal, Dairy, and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA; (S.-I.Y.); (B.-H.S.); (M.E.W.); (J.C.F.); (J.G.J.)
| | - Byung-Hak Song
- Department of Animal, Dairy, and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA; (S.-I.Y.); (B.-H.S.); (M.E.W.); (J.C.F.); (J.G.J.)
| | - Michael E. Woolley
- Department of Animal, Dairy, and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA; (S.-I.Y.); (B.-H.S.); (M.E.W.); (J.C.F.); (J.G.J.)
| | - Jordan C. Frank
- Department of Animal, Dairy, and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA; (S.-I.Y.); (B.-H.S.); (M.E.W.); (J.C.F.); (J.G.J.)
| | - Justin G. Julander
- Department of Animal, Dairy, and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA; (S.-I.Y.); (B.-H.S.); (M.E.W.); (J.C.F.); (J.G.J.)
- Institute for Antiviral Research, Utah State University, Logan, UT 84322, USA
| | - Young-Min Lee
- Department of Animal, Dairy, and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA; (S.-I.Y.); (B.-H.S.); (M.E.W.); (J.C.F.); (J.G.J.)
- Veterinary Diagnostics and Infectious Diseases, Utah Science Technology and Research, Utah State University, Logan, UT 84341, USA
- Correspondence: ; Tel.: +1-435-797-9667
| |
Collapse
|
23
|
Zikavirus prME Envelope Pseudotyped Human Immunodeficiency Virus Type-1 as a Novel Tool for Glioblastoma-Directed Virotherapy. Cancers (Basel) 2020; 12:cancers12041000. [PMID: 32325703 PMCID: PMC7226176 DOI: 10.3390/cancers12041000] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/09/2020] [Accepted: 04/15/2020] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma multiforme is the most lethal type of brain tumor that is not yet curable owing to its frequent resurgence after surgery. Resistance is mainly caused by the presence of a subpopulation of tumor cells, the glioma stem cells (GSCs), which are highly resistant to radiation and chemotherapy. In 2015, Zikavirus (ZIKV)-induced microcephaly emerged in newborns, indicating that ZIKV has a specific neurotropism. Accordingly, an oncolytic tropism for infecting GSCs was demonstrated in a murine tumor model. Like other flaviviruses, ZIKV is enveloped by two proteins, prM and E. The pME expression plasmid along with the HIV-1 vector pNL Luc AM generated prME pseudotyped viral particles. Four different prME envelopes, Z1 to Z4, were cloned, and the corresponding pseudotypes, Z1- to Z4-HIVluc, produced by this two-plasmid system, were tested for entry efficiency using Vero-B4 cells. The most efficient pseudotype, Z1-HIVluc, also infected glioma-derived cell lines U87 and 86HG39. The pseudotype system was then extended by using a three-plasmid system including pME-Z1, the HIV-1 packaging plasmid psPAX2, and the lentiviral vector pLenti-luciferase-P2A-Neo. The corresponding pseudotype, designated Z1-LENTIluc, also infected U87 and 86HG39 cells. Altogether, a pseudotyped virus especially targeting glioma-derived cells might be a promising candidate for a prospective glioblastoma-directed virotherapy.
Collapse
|
24
|
Ramanathan HN, Zhang S, Douam F, Mar KB, Chang J, Yang PL, Schoggins JW, Ploss A, Lindenbach BD. A Sensitive Yellow Fever Virus Entry Reporter Identifies Valosin-Containing Protein (VCP/p97) as an Essential Host Factor for Flavivirus Uncoating. mBio 2020; 11:e00467-20. [PMID: 32291299 PMCID: PMC7157815 DOI: 10.1128/mbio.00467-20] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 03/16/2020] [Indexed: 01/17/2023] Open
Abstract
While the basic mechanisms of flavivirus entry and fusion are understood, little is known about the postfusion events that precede RNA replication, such as nucleocapsid disassembly. We describe here a sensitive, conditionally replication-defective yellow fever virus (YFV) entry reporter, YFVΔSK/Nluc, to quantitively monitor the translation of incoming, virus particle-delivered genomes. We validated that YFVΔSK/Nluc gene expression can be neutralized by YFV-specific antisera and requires known flavivirus entry pathways and cellular factors, including clathrin- and dynamin-mediated endocytosis, endosomal acidification, YFV E glycoprotein-mediated fusion, and cellular LY6E and RPLP1 expression. The initial round of YFV translation was shown to require cellular ubiquitylation, consistent with recent findings that dengue virus capsid protein must be ubiquitylated in order for nucleocapsid uncoating to occur. Importantly, translation of incoming YFV genomes also required valosin-containing protein (VCP)/p97, a cellular ATPase that unfolds and extracts ubiquitylated client proteins from large complexes. RNA transfection and washout experiments showed that VCP/p97 functions at a postfusion, pretranslation step in YFV entry. Finally, VCP/p97 activity was required by other flaviviruses in mammalian cells and by YFV in mosquito cells. Together, these data support a critical role for VCP/p97 in the disassembly of incoming flavivirus nucleocapsids during a postfusion step in virus entry.IMPORTANCE Flaviviruses are an important group of RNA viruses that cause significant human disease. The mechanisms by which flavivirus nucleocapsids are disassembled during virus entry remain unclear. Here, we used a yellow fever virus entry reporter, which expresses a sensitive reporter enzyme but does not replicate, to show that nucleocapsid disassembly requires the cellular protein-disaggregating enzyme valosin-containing protein, also known as p97.
Collapse
Affiliation(s)
- Harish N Ramanathan
- Department of Microbial Pathogenesis, Yale University, New Haven, Connecticut, USA
| | - Shuo Zhang
- Department of Microbial Pathogenesis, Yale University, New Haven, Connecticut, USA
| | - Florian Douam
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Katrina B Mar
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jinhong Chang
- Department of Experimental Therapeutics, The Baruch S. Blumberg Institute, Doylestown, Pennsylvania, USA
| | - Priscilla L Yang
- Department of Microbiology and the Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - John W Schoggins
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Alexander Ploss
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Brett D Lindenbach
- Department of Microbial Pathogenesis, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
25
|
Kobayashi S, Yoshii K, Phongphaew W, Muto M, Hirano M, Orba Y, Sawa H, Kariwa H. West Nile virus capsid protein inhibits autophagy by AMP-activated protein kinase degradation in neurological disease development. PLoS Pathog 2020; 16:e1008238. [PMID: 31971978 PMCID: PMC6977728 DOI: 10.1371/journal.ppat.1008238] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 11/25/2019] [Indexed: 12/31/2022] Open
Abstract
West Nile virus (WNV) belongs to the Flaviviridae family and has emerged as a significant cause of viral encephalitis in birds and animals including humans. WNV replication directly induces neuronal injury, followed by neuronal cell death. We previously showed that accumulation of ubiquitinated protein aggregates was involved in neuronal cell death in the WNV-infected mouse brain. In this study, we attempted to elucidate the mechanisms of the accumulation of protein aggregates in the WNV-infected cells. To identify the viral factor inducing the accumulation of ubiquitinated proteins, intracellular accumulation of ubiquitinated proteins was examined in the cells expressing the viral protein. Expression of capsid (C) protein induced the accumulation, while mutations at residues L51 and A52 in C protein abrogated the accumulation. Wild-type (WT) or mutant WNV in which mutations were introduced into the residues was inoculated into human neuroblastoma cells. The expression levels of LC3-II, an autophagy-related protein, and AMP-activated protein kinase (AMPK), an autophagy inducer, were reduced in the cells infected with WT WNV, while the reduction was not observed in the cells infected with WNV with the mutations in C protein. Similarly, ubiquitination and degradation of AMPK were only observed in the cells infected with WT WNV. In the cells expressing C protein, AMPK was co-precipitated with C protein and mutations in L51 and A52 reduced the interaction. Although the viral replication was not affected, the accumulation of ubiquitinated proteins in brain and neurological symptoms were attenuated in the mouse inoculated with WNV with the mutations in C protein as compared with that with WT WNV. Taken together, ubiquitination and degradation of AMPK by C protein resulted in the inhibition of autophagy and the accumulation of protein aggregates, which contributes to the development of neurological disease.
Collapse
Affiliation(s)
- Shintaro Kobayashi
- Laboratory of Public Health, Faculty of Veterinary Medicine, Hokkaido University, Kita-ku, Sapporo, Japan
- * E-mail:
| | - Kentaro Yoshii
- Laboratory of Public Health, Faculty of Veterinary Medicine, Hokkaido University, Kita-ku, Sapporo, Japan
| | - Wallaya Phongphaew
- Division of Molecular Pathobiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Memi Muto
- Laboratory of Public Health, Faculty of Veterinary Medicine, Hokkaido University, Kita-ku, Sapporo, Japan
| | - Minato Hirano
- Laboratory of Public Health, Faculty of Veterinary Medicine, Hokkaido University, Kita-ku, Sapporo, Japan
| | - Yasuko Orba
- Division of Molecular Pathobiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Hirofumi Sawa
- Division of Molecular Pathobiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo, Japan
- Global Virus Network, Baltimore, Maryland, United States of America
| | - Hiroaki Kariwa
- Laboratory of Public Health, Faculty of Veterinary Medicine, Hokkaido University, Kita-ku, Sapporo, Japan
| |
Collapse
|
26
|
Novel Approach for Insertion of Heterologous Sequences into Full-Length ZIKV Genome Results in Superior Level of Gene Expression and Insert Stability. Viruses 2020; 12:v12010061. [PMID: 31947825 PMCID: PMC7019263 DOI: 10.3390/v12010061] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 12/27/2019] [Accepted: 12/31/2019] [Indexed: 12/15/2022] Open
Abstract
Zika virus (ZIKV) emerged in the Americas in 2015, presenting unique challenges to public health. Unlike other arboviruses of the Flaviviridae family, it is transmissible by sexual contact, which facilitates the spread of the virus into new geographic areas. Additionally, ZIKV can be transmitted from mother to fetus, causing microcephaly and other severe developmental abnormalities. Reliable and easy-to-work-with clones of ZIKV expressing heterologous genes will significantly facilitate studies aimed at understanding the virus pathogenesis and tissue tropism. Here, we developed and characterized two novel approaches for expression of heterologous genes of interest in the context of full-length ZIKV genome and compared them to two previously published strategies for ZIKV-mediated gene expression. We demonstrated that among the four tested viruses expressing nLuc gene, the virus constructed using a newly developed approach of partial capsid gene duplication (PCGD) attained the highest titer in Vero cells and resulted in the highest level of nLuc expression. Suitability of the PCGD approach for expression of different genes of interest was validated by replacing nLuc sequence with that of eGFP gene. The generated constructs were further characterized in cell culture. Potential applications of ZIKV clones stably expressing heterologous genes include development of detection assays, antivirals, therapeutics, live imaging systems, and vaccines.
Collapse
|
27
|
Abstract
Viruses are widely used as vectors for heterologous gene expression in cultured cells or natural hosts, and therefore a large number of viruses with exogenous sequences inserted into their genomes have been engineered. Many of these engineered viruses are viable and express heterologous proteins at high levels, but the inserted sequences often prove to be unstable over time and are rapidly lost, limiting heterologous protein expression. Although virologists are aware that inserted sequences can be unstable, processes leading to insert instability are rarely considered from an evolutionary perspective. Here, we review experimental work on the stability of inserted sequences over a broad range of viruses, and we present some theoretical considerations concerning insert stability. Different virus genome organizations strongly impact insert stability, and factors such as the position of insertion can have a strong effect. In addition, we argue that insert stability not only depends on the characteristics of a particular genome, but that it will also depend on the host environment and the demography of a virus population. The interplay between all factors affecting stability is complex, which makes it challenging to develop a general model to predict the stability of genomic insertions. We highlight key questions and future directions, finding that insert stability is a surprisingly complex problem and that there is need for mechanism-based, predictive models. Combining theoretical models with experimental tests for stability under varying conditions can lead to improved engineering of viral modified genomes, which is a valuable tool for understanding genome evolution as well as for biotechnological applications, such as gene therapy.
Collapse
Affiliation(s)
- Anouk Willemsen
- Laboratory MIVEGEC (UMR CNRS IRD University of Montpellier), Centre National de la Recherche Scientifique (CNRS), 911 Avenue Agropolis, BP 64501, 34394 Montpellier cedex 5, France
| | - Mark P Zwart
- Netherlands Institute of Ecology (NIOO-KNAW), Postbus 50, 6700 AB, Wageningen, The Netherlands
| |
Collapse
|
28
|
Riedel C, Lamp B, Chen HW, Heimann M, Rümenapf T. Fluorophore labelled BVDV: a novel tool for the analysis of infection dynamics. Sci Rep 2019; 9:5972. [PMID: 30979966 PMCID: PMC6461705 DOI: 10.1038/s41598-019-42540-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 04/03/2019] [Indexed: 11/10/2022] Open
Abstract
Genetic labelling of viruses with a fluorophore allows to study their life cycle in real time, without the need for fixation or staining techniques. Within the family Flaviviridae, options for genetic labelling of non-structural proteins exist. Yet, no system to genetically label structural proteins has been put forward to date. Taking advantage of a previously described site within the structural protein E2, a fluorophore was introduced into a cytopathogenic (cpe) BVDV-1 virus (BVDVE2_fluo). This insertion was well tolerated, resulting in a 2-fold drop in titer compared to the parental virus, and remained stably integrated into the genome for more than 10 passages. The fluorophore E2 fusion protein was readily detectable in purified virus particles by Western blot and fluorescence microscopy and the particle integrity and morphology was confirmed by cryo electron microscopy. The same integration site could also be used to label the related Classical swine fever virus. Also, BVDVE2_fluo particles bound to fluorophore labelled CD46 expressing cells could be resolved in fluorescence microscopy. This underlines the applicability of BVDVE2_fluo as a tool to study the dynamics of the whole life cycle of BVDV in real time.
Collapse
Affiliation(s)
- Christiane Riedel
- Institute of Virology, Department of Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria.
| | - Benjamin Lamp
- Institute of Virology, Department of Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Hann-Wei Chen
- Institute of Virology, Department of Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Manuela Heimann
- Institute of Anatomy, Faculty of Veterinary Medicine, Justus-Liebig University, Giessen, Germany
| | - Till Rümenapf
- Institute of Virology, Department of Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| |
Collapse
|
29
|
Burgado J, Greenberg L, Niezgoda M, Kumar A, Olson V, Wu X, Satheshkumar PS. A high throughput neutralization test based on GFP expression by recombinant rabies virus. PLoS Negl Trop Dis 2018; 12:e0007011. [PMID: 30550592 PMCID: PMC6310286 DOI: 10.1371/journal.pntd.0007011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 12/28/2018] [Accepted: 11/19/2018] [Indexed: 12/25/2022] Open
Abstract
The effectiveness of rabies vaccination in both humans and animals is determined by the presence of virus neutralizing antibodies (VNAs). The Rapid Fluorescent Focus Inhibition Test (RFFIT) is the method traditionally used for detection and quantification of VNAs. It is a functional in vitro test for assessing the ability of antibodies in serum to bind and prevent infection of cultured cells with rabies virus (RABV). The RFFIT is a labor intensive, low throughput and semi-quantitative assay performed by trained laboratorians. It requires staining of RABV-infected cells by rabies specific fluorescent antibodies and manual quantification of fluorescent fields for titer determination. Although the quantification of fluorescent fields observed in each sample is recorded, the corresponding images are not stored or captured to be used for future analysis. To circumvent several of these disadvantages, we have developed an alternative, automated high throughput neutralization test (HTNT) for determination of rabies VNAs based on green fluorescent protein (GFP) expression by a recombinant RABV and compared with the RFFIT. The HTNT assay utilizes the recombinant RABV ERA variant expressing GFP with a nuclear localization signal (NLS) for efficient quantification. The HTNT is a quantitative method where the number of RABV-infected cells are determined and the images are stored for future analysis. Both RFFIT and HTNT results correlated 100% for a panel of human and animal positive and negative rabies serum samples. Although, the VNA titer values are generally agreeable, HTNT titers tend to be lower than that of RFFIT, probably due to the differences in quantification methods. Our data demonstrates the potential for HTNT assays in determination of rabies VNA titers. The potency of rabies vaccine is demonstrated by the presence of virus neutralizing antibodies (VNAs) in serum. It is critical to evaluate immunologic status of individuals who work directly with rabies virus (RABV) (laboratorians) or at high risk of infection due to interaction with animals (veterinarians and animal control workers). In addition, rabies vaccination records and demonstration of VNAs in animals are mandatory before initiating pet travel to rabies-free counties or regions. Rabies VNAs are currently determined by the rapid fluorescent focus inhibition test (RFFIT) and the fluorescent antibody virus neutralization (FAVN) test, which measure the ability of antibodies to bind and prevent infection of RABV in vitro. Both assays require staining of infected cells using anti-rabies antibodies and manual observation of infected cells by a fluorescent microscope to determine VNA titers. In this study, we have developed a GFP reporter-based high throughput neutralization test (HTNT) for automated quantification of infected cells. This method has the advantages of allowing investigators to analyze and store the results, and can accommodate large sample sizes. Overall, the results from HTNT exhibited 100% correlation with that of RFFIT, albeit with differences in rabies VNA titer values due to quantification methods.
Collapse
Affiliation(s)
- Jillybeth Burgado
- Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Lauren Greenberg
- Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Mike Niezgoda
- Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Amrita Kumar
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Victoria Olson
- Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Xianfu Wu
- Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | | |
Collapse
|
30
|
Han M, Rajput C, Hinde JL, Wu Q, Lei J, Ishikawa T, Bentley JK, Hershenson MB. Construction of a recombinant rhinovirus accommodating fluorescent marker expression. Influenza Other Respir Viruses 2018; 12:717-727. [PMID: 30120824 PMCID: PMC6185886 DOI: 10.1111/irv.12602] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 07/02/2018] [Accepted: 08/15/2018] [Indexed: 12/28/2022] Open
Abstract
Background Rhinovirus (RV) causes the common cold and asthma exacerbations. The RV genome is a 7.3 kb single‐strand positive‐sense RNA. Objective Using minor group RV1A as a backbone, we sought to design and generate a recombinant RV1A accommodating fluorescent marker expression, thereby allowing tracking of viral infection. Method Recombinant RV1A infectious cDNA clones harboring the coding sequence of green fluorescent protein (GFP), Renilla luciferase, or iLOV (for light, oxygen, or voltage sensing) were engineered and constructed. RV‐infected cells were determined by flow cytometry, immunohistochemistry, and immunofluorescence microscopy. Results RV1A‐GFP showed a cytopathic effect in HeLa cells but failed to express GFP or Renilla luciferase due to deletion. The smaller fluorescent protein construct, RV1A‐iLOV, was stably expressed in infected cells. RV1A‐iLOV expression was used to examine the antiviral effect of bafilomycin in HeLa cells. Compared to parental virus, RV1A‐iLOV infection of BALB/c mice yielded a similar viral load and level of cytokine mRNA expression. However, imaging of fixed lung tissue failed to reveal a fluorescent signal, likely due to the oxidation and bleaching of iLOV‐bound flavin mononucleotide. We therefore employed an anti‐iLOV antibody for immunohistochemical and immunofluorescence imaging. The iLOV signal was identified in airway epithelial cells and CD45+ CD11b+ lung macrophages. Conclusions These results suggest that RV1A‐iLOV is a useful molecular tool for studying RV pathogenesis. The construction strategy for RV1A‐iLOV could be applied to other RV serotypes. However, the detection of iLOV‐expressing RV in fixed tissue required the use of an anti‐iLOV antibody, limiting the value of this construct.
Collapse
Affiliation(s)
- Mingyuan Han
- Department of Pediatrics & Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan
| | - Charu Rajput
- Department of Pediatrics & Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan
| | - Joanna L Hinde
- Department of Pediatrics & Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan
| | - Qian Wu
- Department of Pediatrics & Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan
| | - Jing Lei
- Department of Pediatrics & Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan
| | - Tomoko Ishikawa
- Department of Pediatrics & Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan
| | - J Kelley Bentley
- Department of Pediatrics & Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan
| | - Marc B Hershenson
- Department of Pediatrics & Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan.,Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
31
|
Chen S, He Y, Zhang R, Liu P, Yang C, Wu Z, Zhang J, Wang M, Jia R, Zhu D, Liu M, Yang Q, Wu Y, Cheng A. Establishment of a reverse genetics system for duck Tembusu virus to study virulence and screen antiviral genes. Antiviral Res 2018; 157:120-127. [PMID: 30057296 DOI: 10.1016/j.antiviral.2018.06.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 06/06/2018] [Accepted: 06/30/2018] [Indexed: 10/28/2022]
Abstract
Recently, a newly emerged avian flavivirus, duck Tembusu virus (TMUV), was identified as the causative agent of a serious duck viral disease in Asia. Its rapid spread and expanded host range have raised substantial concerns regarding its potential threat to non-avian hosts, including humans. In this study, we report an infectious cDNA clone for a clinical strain CQW1 isolated from Southwest China, which is representative of the disease outbreak in the Chinese mainland. We generated a full-length cDNA clone pACYC FL-TMUV, which is infectious, and this cDNA clone-derived recombinant TMUV (rTMUV) showed comparative growth kinetics in both BHK21 cells and DEF cells compared with parental TMUV (pTMUV). In addition, rTMUV also showed the same high virulence in 9-day-old duck embryos as that in pTMUV, suggesting that rTMUV possessed similar properties to the natural virus both in vitro and in vivo. Based on the cDNA-clone, we first generated a reporter TMUV (TMUV-RLuc) carrying a Renilla luciferase (RLuc) gene. The luciferase kinetics of TMUV-RLuc were determined both in BHK21 and DEF cells. It seems that TMUV-RLuc grew well in vitro; however, the insertion of the RLuc gene attenuated viral replication in vitro. The higher viral titres of TMUV-RLuc were observed in BHK21 compared with that in DEF cells. The antiviral effects of exogenous-expressed duck RIG-I, MDA5, STING, MAVS, TBK1, IFNα and IFNγ were studied in vitro by using TMUV-RLuc. Our reverse genetics system will provide a multicomponent platform for the pathogenesis study of duck TMUV and the development of molecular countermeasures against duck TMUV infection.
Collapse
Affiliation(s)
- Shun Chen
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China.
| | - Yu He
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Rujuan Zhang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Peng Liu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Chao Yang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Zhen Wu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Jinyue Zhang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Mingshu Wang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Renyong Jia
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Dekang Zhu
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Mafeng Liu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Qiao Yang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Ying Wu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Anchun Cheng
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China.
| |
Collapse
|
32
|
Suphatrakul A, Duangchinda T, Jupatanakul N, Prasittisa K, Onnome S, Pengon J, Siridechadilok B. Multi-color fluorescent reporter dengue viruses with improved stability for analysis of a multi-virus infection. PLoS One 2018; 13:e0194399. [PMID: 29547653 PMCID: PMC5856417 DOI: 10.1371/journal.pone.0194399] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 03/03/2018] [Indexed: 01/08/2023] Open
Abstract
Reporter virus is a versatile tool to visualize and to analyze virus infections. However, for flaviviruses, it is difficult to maintain the inserted reporter genes on the viral genome, limiting its use in several studies that require homogeneous virus particles and several rounds of virus replication. Here, we showed that flanking inserted GFP genes on both sides with ribosome-skipping 2A sequences improved the stability and the consistency of their fluorescent signals for dengue-virus-serotype 2 (DENV2) reporter viruses. The reporter viruses can infect known susceptible mammalian cell lines and primary CD14+ human monocytes. This design can accommodate several fluorescent protein genes, enabling the generation of multi-color DENV2-16681 reporter viruses with comparable replication capabilities, as demonstrated by their abilities to maintain their fluorescent intensities during co-infections and to exclude superinfections regardless of the fluorescent tags. The reported design of multi-color DENV2 should be useful for high-throughput analyses, single-cell analysis, and characterizations of interference and superinfection in animal models.
Collapse
Affiliation(s)
- Amporn Suphatrakul
- National Center for Genetic Engineering and Biotechnology, Klong Luang, Pathumthani, Thailand
| | - Thaneeya Duangchinda
- National Center for Genetic Engineering and Biotechnology, Klong Luang, Pathumthani, Thailand
| | - Natapong Jupatanakul
- National Center for Genetic Engineering and Biotechnology, Klong Luang, Pathumthani, Thailand
| | - Kanjanawadee Prasittisa
- National Center for Genetic Engineering and Biotechnology, Klong Luang, Pathumthani, Thailand
| | - Suppachoke Onnome
- National Center for Genetic Engineering and Biotechnology, Klong Luang, Pathumthani, Thailand
| | - Jutharat Pengon
- National Center for Genetic Engineering and Biotechnology, Klong Luang, Pathumthani, Thailand
| | - Bunpote Siridechadilok
- National Center for Genetic Engineering and Biotechnology, Klong Luang, Pathumthani, Thailand
- * E-mail:
| |
Collapse
|
33
|
Characterization of Recombinant Flaviviridae Viruses Possessing a Small Reporter Tag. J Virol 2018; 92:JVI.01582-17. [PMID: 29093094 DOI: 10.1128/jvi.01582-17] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 10/19/2017] [Indexed: 01/13/2023] Open
Abstract
The family Flaviviridae consists of four genera, Flavivirus, Pestivirus, Pegivirus, and Hepacivirus, and comprises important pathogens of human and animals. Although the construction of recombinant viruses carrying reporter genes encoding fluorescent and bioluminescent proteins has been reported, the stable insertion of foreign genes into viral genomes retaining infectivity remains difficult. Here, we applied the 11-amino-acid subunit derived from NanoLuc luciferase to the engineering of the Flaviviridae viruses and then examined the biological characteristics of the viruses. We successfully generated recombinant viruses carrying the split-luciferase gene, including dengue virus, Japanese encephalitis virus, hepatitis C virus (HCV), and bovine viral diarrhea virus. The stability of the viruses was confirmed by five rounds of serial passages in the respective susceptible cell lines. The propagation of the recombinant luciferase viruses in each cell line was comparable to that of the parental viruses. By using a purified counterpart luciferase protein, this split-luciferase assay can be applicable in various cell lines, even when it is difficult to transduce the counterpart gene. The efficacy of antiviral reagents against the recombinant viruses could be monitored by the reduction of luciferase expression, which was correlated with that of viral RNA, and the recombinant HCV was also useful to examine viral dynamics in vivo Taken together, our findings indicate that the recombinant Flaviviridae viruses possessing the split NanoLuc luciferase gene generated here provide powerful tools to understand viral life cycle and pathogenesis and a robust platform to develop novel antivirals against Flaviviridae viruses.IMPORTANCE The construction of reporter viruses possessing a stable transgene capable of expressing specific signals is crucial to investigations of viral life cycle and pathogenesis and the development of antivirals. However, it is difficult to maintain the stability of a large foreign gene, such as those for fluorescence and bioluminescence, after insertion into a viral genome. Here, we successfully generated recombinant Flaviviridae viruses carrying the 11-amino-acid subunit derived from NanoLuc luciferase and demonstrated that these viruses are applicable to in vitro and in vivo experiments, suggesting that these recombinant Flaviviridae viruses are powerful tools for increasing our understanding of viral life cycle and pathogenesis and that these recombinant viruses will provide a robust platform to develop antivirals against Flaviviridae viruses.
Collapse
|
34
|
New reverse genetics and transfection methods to rescue arboviruses in mosquito cells. Sci Rep 2017; 7:13983. [PMID: 29070887 PMCID: PMC5656662 DOI: 10.1038/s41598-017-14522-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 10/11/2017] [Indexed: 01/04/2023] Open
Abstract
Reverse genetics is a critical tool to decrypt the biological properties of arboviruses. However, whilst reverse genetics methods have been usually applied to vertebrate cells, their use in insect cells remains uncommon due to the conjunction of laborious molecular biology techniques and of specific difficulties surrounding the transfection of such cells. To leverage reverse genetics studies in both vertebrate and mosquito cells, we designed an improved DNA transfection protocol for insect cells and then demonstrated that the simple and flexible ISA (Infectious Subgenomic Amplicons) reverse-genetics method can be efficiently applied to both mammalian and mosquito cells to generate in days recombinant infectious positive-stranded RNA viruses belonging to genera Flavivirus (Japanese encephalitis, Yellow fever, West Nile and Zika viruses) and Alphavirus (Chikungunya virus). This method represents an effective option to potentially overcome technological issues related to the study of arboviruses.
Collapse
|
35
|
Tartour K, Nguyen XN, Appourchaux R, Assil S, Barateau V, Bloyet LM, Burlaud Gaillard J, Confort MP, Escudero-Perez B, Gruffat H, Hong SS, Moroso M, Reynard O, Reynard S, Decembre E, Ftaich N, Rossi A, Wu N, Arnaud F, Baize S, Dreux M, Gerlier D, Paranhos-Baccala G, Volchkov V, Roingeard P, Cimarelli A. Interference with the production of infectious viral particles and bimodal inhibition of replication are broadly conserved antiviral properties of IFITMs. PLoS Pathog 2017; 13:e1006610. [PMID: 28957419 PMCID: PMC5619827 DOI: 10.1371/journal.ppat.1006610] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 08/28/2017] [Indexed: 12/31/2022] Open
Abstract
IFITMs are broad antiviral factors that block incoming virions in endosomal vesicles, protecting target cells from infection. In the case of HIV-1, we and others reported the existence of an additional antiviral mechanism through which IFITMs lead to the production of virions of reduced infectivity. However, whether this second mechanism of inhibition is unique to HIV or extends to other viruses is currently unknown. To address this question, we have analyzed the susceptibility of a broad spectrum of viruses to the negative imprinting of the virion particles infectivity by IFITMs. The results we have gathered indicate that this second antiviral property of IFITMs extends well beyond HIV and we were able to identify viruses susceptible to the three IFITMs altogether (HIV-1, SIV, MLV, MPMV, VSV, MeV, EBOV, WNV), as well as viruses that displayed a member-specific susceptibility (EBV, DUGV), or were resistant to all IFITMs (HCV, RVFV, MOPV, AAV). The swapping of genetic elements between resistant and susceptible viruses allowed us to point to specificities in the viral mode of assembly, rather than glycoproteins as dominant factors of susceptibility. However, we also show that, contrarily to X4-, R5-tropic HIV-1 envelopes confer resistance against IFITM3, suggesting that viral receptors add an additional layer of complexity in the IFITMs-HIV interplay. Lastly, we show that the overall antiviral effects ascribed to IFITMs during spreading infections, are the result of a bimodal inhibition in which IFITMs act both by protecting target cells from incoming viruses and in driving the production of virions of reduced infectivity. Overall, our study reports for the first time that the negative imprinting of the virion particles infectivity is a conserved antiviral property of IFITMs and establishes IFITMs as a paradigm of restriction factor capable of interfering with two distinct phases of a virus life cycle. IFITMs are interferon-regulated proteins that inhibit a broad range of viruses. Until recently, IFITMs had been described to arrest incoming viral particles in target cells, by inducing their retention in endosomal vesicles. More recently in the case of HIV-1, ours and other laboratories have highlighted the existence of an additional antiviral mechanism with which IFITMs could act in virus-producing cells, leading to the production of virion particles of reduced infectivity. In the present study, we assessed whether the negative imprinting of the virion particles infectivity was a conserved antiviral property of IFITMs by examining a panel of fourteen different DNA or RNA viruses. Our results indicate that a wide spectrum of viruses is susceptible to this antiviral mechanism of inhibition, although some are able to resist it. Swapping of elements between susceptible and resistant viruses strongly suggests that specificities in the mode of virion assembly and not the viral glycoprotein are the dominant factor in the susceptibility of a given virus to this inhibition. However, we also show that HIV-1 strains that engage the CCR5 co-receptor display a notable resistance towards IFITM3, indicating that at least in the case of HIV-1, co-receptor usage is likely to add an additional layer of complexity in the relationship established between IFITMs and the virus, that may or may not extend to other viral families as well. In the context of spreading infections, the results of this study highlight that the overall antiviral effect of IFITMs is mechanistically caused by a previously unappreciated dual mode of action in which they act both in target cells and in virus-producing cells, by respectively forcing endosome trapping of incoming viruses and by commandeering the formation of new virion particles of reduced infectivity. Overall, the results presented here indicate that the negative imprinting of viral particles is a largely conserved antiviral feature of IFITMs and point to IFITMs as a novel paradigm of innate defense proteins capable of interfering with viral replication at two distinct steps of a virus life cycle.
Collapse
Affiliation(s)
- Kevin Tartour
- CIRI, Centre International de Recherche en Infectiologie, Lyon, France
- INSERM, U1111, Lyon, France
- Université Claude Bernard Lyon1, Lyon, France
- CNRS, UMR5308, Lyon, France
- Ecole Normale Supérieure de Lyon, Lyon, France
- Univ Lyon, Lyon, France, Lyon, France
| | - Xuan-Nhi Nguyen
- CIRI, Centre International de Recherche en Infectiologie, Lyon, France
- INSERM, U1111, Lyon, France
- Université Claude Bernard Lyon1, Lyon, France
- CNRS, UMR5308, Lyon, France
- Ecole Normale Supérieure de Lyon, Lyon, France
- Univ Lyon, Lyon, France, Lyon, France
| | - Romain Appourchaux
- CIRI, Centre International de Recherche en Infectiologie, Lyon, France
- INSERM, U1111, Lyon, France
- Université Claude Bernard Lyon1, Lyon, France
- CNRS, UMR5308, Lyon, France
- Ecole Normale Supérieure de Lyon, Lyon, France
- Univ Lyon, Lyon, France, Lyon, France
| | - Sonia Assil
- CIRI, Centre International de Recherche en Infectiologie, Lyon, France
- INSERM, U1111, Lyon, France
- Université Claude Bernard Lyon1, Lyon, France
- CNRS, UMR5308, Lyon, France
- Ecole Normale Supérieure de Lyon, Lyon, France
- Univ Lyon, Lyon, France, Lyon, France
| | - Véronique Barateau
- CIRI, Centre International de Recherche en Infectiologie, Lyon, France
- INSERM, U1111, Lyon, France
- Université Claude Bernard Lyon1, Lyon, France
- CNRS, UMR5308, Lyon, France
- Ecole Normale Supérieure de Lyon, Lyon, France
- Univ Lyon, Lyon, France, Lyon, France
| | - Louis-Marie Bloyet
- CIRI, Centre International de Recherche en Infectiologie, Lyon, France
- INSERM, U1111, Lyon, France
- Université Claude Bernard Lyon1, Lyon, France
- CNRS, UMR5308, Lyon, France
- Ecole Normale Supérieure de Lyon, Lyon, France
- Univ Lyon, Lyon, France, Lyon, France
| | - Julien Burlaud Gaillard
- Plateforme IBiSA de Microscopie Electronique, Université F. Rabelais et CHRU de Tours, Tours, France
- INSERM U966, Université F. Rabelais et CHRU de Tours, Tours, France
| | - Marie-Pierre Confort
- IVPC UMR754, INRA, Univ Lyon, Université Claude Bernard Lyon1, EPHE, Lyon, France
| | - Beatriz Escudero-Perez
- CIRI, Centre International de Recherche en Infectiologie, Lyon, France
- INSERM, U1111, Lyon, France
- Université Claude Bernard Lyon1, Lyon, France
- CNRS, UMR5308, Lyon, France
- Ecole Normale Supérieure de Lyon, Lyon, France
- Univ Lyon, Lyon, France, Lyon, France
| | - Henri Gruffat
- CIRI, Centre International de Recherche en Infectiologie, Lyon, France
- INSERM, U1111, Lyon, France
- Université Claude Bernard Lyon1, Lyon, France
- CNRS, UMR5308, Lyon, France
- Ecole Normale Supérieure de Lyon, Lyon, France
- Univ Lyon, Lyon, France, Lyon, France
| | - Saw See Hong
- IVPC UMR754, INRA, Univ Lyon, Université Claude Bernard Lyon1, EPHE, Lyon, France
| | - Marie Moroso
- CIRI, Centre International de Recherche en Infectiologie, Lyon, France
- INSERM, U1111, Lyon, France
- Université Claude Bernard Lyon1, Lyon, France
- CNRS, UMR5308, Lyon, France
- Ecole Normale Supérieure de Lyon, Lyon, France
- Univ Lyon, Lyon, France, Lyon, France
- Fondation Mérieux, Lyon, France
| | - Olivier Reynard
- CIRI, Centre International de Recherche en Infectiologie, Lyon, France
- INSERM, U1111, Lyon, France
- Université Claude Bernard Lyon1, Lyon, France
- CNRS, UMR5308, Lyon, France
- Ecole Normale Supérieure de Lyon, Lyon, France
- Univ Lyon, Lyon, France, Lyon, France
| | - Stéphanie Reynard
- CIRI, Centre International de Recherche en Infectiologie, Lyon, France
- INSERM, U1111, Lyon, France
- Université Claude Bernard Lyon1, Lyon, France
- CNRS, UMR5308, Lyon, France
- Ecole Normale Supérieure de Lyon, Lyon, France
- Univ Lyon, Lyon, France, Lyon, France
- Institut Pasteur, Lyon, France
| | - Elodie Decembre
- CIRI, Centre International de Recherche en Infectiologie, Lyon, France
- INSERM, U1111, Lyon, France
- Université Claude Bernard Lyon1, Lyon, France
- CNRS, UMR5308, Lyon, France
- Ecole Normale Supérieure de Lyon, Lyon, France
- Univ Lyon, Lyon, France, Lyon, France
| | - Najate Ftaich
- IVPC UMR754, INRA, Univ Lyon, Université Claude Bernard Lyon1, EPHE, Lyon, France
| | - Axel Rossi
- CIRI, Centre International de Recherche en Infectiologie, Lyon, France
- INSERM, U1111, Lyon, France
- Université Claude Bernard Lyon1, Lyon, France
- CNRS, UMR5308, Lyon, France
- Ecole Normale Supérieure de Lyon, Lyon, France
- Univ Lyon, Lyon, France, Lyon, France
| | - Nannan Wu
- CIRI, Centre International de Recherche en Infectiologie, Lyon, France
- INSERM, U1111, Lyon, France
- Université Claude Bernard Lyon1, Lyon, France
- CNRS, UMR5308, Lyon, France
- Ecole Normale Supérieure de Lyon, Lyon, France
- Univ Lyon, Lyon, France, Lyon, France
- Institute of BioMedical Science (IBMS), East China Normal University (ECNU), Shanghai, China
| | - Frédérick Arnaud
- IVPC UMR754, INRA, Univ Lyon, Université Claude Bernard Lyon1, EPHE, Lyon, France
| | - Sylvain Baize
- CIRI, Centre International de Recherche en Infectiologie, Lyon, France
- INSERM, U1111, Lyon, France
- Université Claude Bernard Lyon1, Lyon, France
- CNRS, UMR5308, Lyon, France
- Ecole Normale Supérieure de Lyon, Lyon, France
- Univ Lyon, Lyon, France, Lyon, France
- Institut Pasteur, Lyon, France
| | - Marlène Dreux
- CIRI, Centre International de Recherche en Infectiologie, Lyon, France
- INSERM, U1111, Lyon, France
- Université Claude Bernard Lyon1, Lyon, France
- CNRS, UMR5308, Lyon, France
- Ecole Normale Supérieure de Lyon, Lyon, France
- Univ Lyon, Lyon, France, Lyon, France
| | - Denis Gerlier
- CIRI, Centre International de Recherche en Infectiologie, Lyon, France
- INSERM, U1111, Lyon, France
- Université Claude Bernard Lyon1, Lyon, France
- CNRS, UMR5308, Lyon, France
- Ecole Normale Supérieure de Lyon, Lyon, France
- Univ Lyon, Lyon, France, Lyon, France
| | - Glaucia Paranhos-Baccala
- CIRI, Centre International de Recherche en Infectiologie, Lyon, France
- INSERM, U1111, Lyon, France
- Université Claude Bernard Lyon1, Lyon, France
- CNRS, UMR5308, Lyon, France
- Ecole Normale Supérieure de Lyon, Lyon, France
- Univ Lyon, Lyon, France, Lyon, France
- Fondation Mérieux, Lyon, France
| | - Viktor Volchkov
- CIRI, Centre International de Recherche en Infectiologie, Lyon, France
- INSERM, U1111, Lyon, France
- Université Claude Bernard Lyon1, Lyon, France
- CNRS, UMR5308, Lyon, France
- Ecole Normale Supérieure de Lyon, Lyon, France
- Univ Lyon, Lyon, France, Lyon, France
| | - Philippe Roingeard
- Plateforme IBiSA de Microscopie Electronique, Université F. Rabelais et CHRU de Tours, Tours, France
- INSERM U966, Université F. Rabelais et CHRU de Tours, Tours, France
| | - Andrea Cimarelli
- CIRI, Centre International de Recherche en Infectiologie, Lyon, France
- INSERM, U1111, Lyon, France
- Université Claude Bernard Lyon1, Lyon, France
- CNRS, UMR5308, Lyon, France
- Ecole Normale Supérieure de Lyon, Lyon, France
- Univ Lyon, Lyon, France, Lyon, France
- * E-mail:
| |
Collapse
|
36
|
Mercier-Delarue S, Durier C, Colin de Verdière N, Poveda JD, Meiffrédy V, Fernandez Garcia MD, Lastère S, Césaire R, Manuggera JC, Molina JM, Amara A, Simon F. Screening test for neutralizing antibodies against yellow fever virus, based on a flavivirus pseudotype. PLoS One 2017; 12:e0177882. [PMID: 28562615 PMCID: PMC5451040 DOI: 10.1371/journal.pone.0177882] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 05/04/2017] [Indexed: 11/26/2022] Open
Abstract
Given the possibility of yellow fever virus reintroduction in epidemiologically receptive geographic areas, the risk of vaccine supply disruption is a serious issue. New strategies to reduce the doses of injected vaccines should be evaluated very carefully in terms of immunogenicity. The plaque reduction test for the determination of neutralizing antibodies (PRNT) is particularly time-consuming and requires the use of a confinement laboratory. We have developed a new test based on the use of a non-infectious pseudovirus (WN/YF17D). The presence of a reporter gene allows sensitive determination of neutralizing antibodies by flow cytometry. This WN/YF17D test was as sensitive as PRNT for the follow-up of yellow fever vaccinees. Both tests lacked specificity with sera from patients hospitalized for acute Dengue virus infection. Conversely, both assays were strictly negative in adults never exposed to flavivirus infection or vaccination, and in patients sampled some time after acute Dengue infection. This WN/YF17D test will be particularly useful for large epidemiological studies and for screening for neutralizing antibodies against yellow fever virus.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Stéphane Lastère
- Department of Medical Biology, Centre Hospitalier de Polynésie Française, Papeete–Tahiti, French Polynesia
| | - Raymond Césaire
- Department of Viro-Immunology, University Hospital of Fort de France, Fort de France- Martinique, French West Indies
| | - Jean-Claude Manuggera
- Institut Pasteur, Environment and Infectious Risks Research and Expertise Unit, Laboratory for Urgent Response to Biological Threats, Paris, France
| | - Jean-Michel Molina
- Department of Infectious Diseases, Saint Louis University Hospital, Paris, France
| | - Ali Amara
- INSERM U944 -UMR 7212, Saint Louis University Hospital, Paris, France
| | - François Simon
- Department of Microbiology, Saint Louis University Hospital, Paris, France
- * E-mail:
| |
Collapse
|
37
|
Kobayashi S, Yoshii K, Hirano M, Muto M, Kariwa H. A novel reverse genetics system for production of infectious West Nile virus using homologous recombination in mammalian cells. J Virol Methods 2016; 240:14-20. [PMID: 27865748 DOI: 10.1016/j.jviromet.2016.11.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 11/11/2016] [Accepted: 11/11/2016] [Indexed: 11/16/2022]
Abstract
Reverse genetics systems facilitate investigation of many aspects of the life cycle and pathogenesis of viruses. However, genetic instability in Escherichia coli has hampered development of a reverse genetics system for West Nile virus (WNV). In this study, we developed a novel reverse genetics system for WNV based on homologous recombination in mammalian cells. Introduction of the DNA fragment coding for the WNV structural protein together with a DNA-based replicon resulted in the release of infectious WNV. The growth rate and plaque size of the recombinant virus were almost identical to those of the parent WNV. Furthermore, chimeric WNV was produced by introducing the DNA fragment coding for the structural protein and replicon plasmid derived from various strains. Here, we report development of a novel system that will facilitate research into WNV infection.
Collapse
Affiliation(s)
- Shintaro Kobayashi
- Laboratory of Public Health, Graduate School of Veterinary Medicine, Hokkaido University, N18, W9, Kita-ku, Sapporo 060-0818, Japan.
| | - Kentaro Yoshii
- Laboratory of Public Health, Graduate School of Veterinary Medicine, Hokkaido University, N18, W9, Kita-ku, Sapporo 060-0818, Japan
| | - Minato Hirano
- Laboratory of Public Health, Graduate School of Veterinary Medicine, Hokkaido University, N18, W9, Kita-ku, Sapporo 060-0818, Japan
| | - Memi Muto
- Laboratory of Public Health, Graduate School of Veterinary Medicine, Hokkaido University, N18, W9, Kita-ku, Sapporo 060-0818, Japan
| | - Hiroaki Kariwa
- Laboratory of Public Health, Graduate School of Veterinary Medicine, Hokkaido University, N18, W9, Kita-ku, Sapporo 060-0818, Japan
| |
Collapse
|
38
|
Blitvich BJ, Wang T, Saxena V, Zeng S, Harmon KM, Raymond MD, Goins KM, Reed CR, Mullins RF, Greiner MA. West Nile Virus Infection in Human and Mouse Cornea Tissue. Am J Trop Med Hyg 2016; 95:1185-1191. [PMID: 27672204 DOI: 10.4269/ajtmh.16-0256] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 08/01/2016] [Indexed: 12/13/2022] Open
Abstract
The purpose of this study was to determine the in vitro and ex vivo susceptibility of human corneal cells to West Nile virus (WNV) infection and evaluate the ability of the virus to disseminate to the corneas of infected mice. Human corneal epithelial cells were challenged with WNV, incubated for 1-6 days, and tested for evidence of WNV infection. Viral RNA and antigen were detected at every time point, and the virus reached a peak titer of 2.5 × 107 plaque-forming units (pfu)/mL at 3 days postinoculation (PI). Corneas procured from donors were incubated in culture dishes containing WNV for 1-5 days and tested for evidence of WNV. Viral RNA and antigen were detected, and the virus reached a mean peak titer of 4.9 × 104 pfu/mL at 5 days PI. Mice were inoculated intraperitoneally with WNV, and their eyes were harvested at 2, 5, and 8 days PI and tested for evidence of WNV. Viral RNA was detected in corneas of four of nine systemically infected mice as early as 2 days PI. We conclude that human corneal cells support WNV replication in vitro and ex vivo, and WNV may disseminate into the corneas of experimentally infected mice. These findings indicate that corneal transmission cannot be ruled out as a novel mode of human-to-human WNV transmission and additional experiments should be conducted to assess this risk further.
Collapse
Affiliation(s)
- Bradley J Blitvich
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, Iowa
| | - Tian Wang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas.,Department of Pathology, University of Texas Medical Branch, Galveston, Texas.,Sealy Center for Vaccine Development, University of Texas Medical Branch, Galveston, Texas
| | - Vandana Saxena
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| | - Shemin Zeng
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, Iowa.,Stephen A. Wynn Institute for Vision Research, University of Iowa, Iowa City, Iowa
| | - Karen M Harmon
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, Iowa
| | - Matthew D Raymond
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, Iowa
| | - Kenneth M Goins
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, Iowa.,Iowa Lions Eye Bank, Coralville, Iowa
| | | | - Robert F Mullins
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, Iowa.,Stephen A. Wynn Institute for Vision Research, University of Iowa, Iowa City, Iowa
| | - Mark A Greiner
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, Iowa. .,Stephen A. Wynn Institute for Vision Research, University of Iowa, Iowa City, Iowa.,Iowa Lions Eye Bank, Coralville, Iowa
| |
Collapse
|
39
|
A robust method for the rapid generation of recombinant Zika virus expressing the GFP reporter gene. Virology 2016; 497:157-162. [PMID: 27471954 DOI: 10.1016/j.virol.2016.07.015] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 07/13/2016] [Accepted: 07/18/2016] [Indexed: 02/07/2023]
Abstract
Zika virus (ZIKV) infection is a major public health problem with severe human congenital and neurological anomalies. The screening of anti-ZIKV compounds and neutralizing antibodies needs reliable and rapid virus-based assays. Here, we described a convenient method leading to the rapid production of molecular clones of ZIKV. To generate a molecular clone of ZIKV strain MR766(NIID), the viral genome was directly assembled into Vero cells after introduction of four overlapping synthetic fragments that cover the full-length genomic RNA sequence. Such strategy has allowed the production of a recombinant ZIKV expressing the GFP reporter gene that is stable over two culturing rounds on Vero cells. Our data demonstrate that the ZIKV reporter virus is a very reliable GFP-based tool for analyzing viral growth and measuring the neutralizing antibody as well as rapid screening of antiviral effect of different classes of inhibitors.
Collapse
|
40
|
Jia F, Zhu X, Xu F. A single adaptive point mutation in Japanese encephalitis virus capsid is sufficient to render the virus as a stable vector for gene delivery. Virology 2016; 490:109-18. [DOI: 10.1016/j.virol.2016.01.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Revised: 01/06/2016] [Accepted: 01/07/2016] [Indexed: 01/01/2023]
|
41
|
Vaccine and Wild-Type Strains of Yellow Fever Virus Engage Distinct Entry Mechanisms and Differentially Stimulate Antiviral Immune Responses. mBio 2016; 7:e01956-15. [PMID: 26861019 PMCID: PMC4752603 DOI: 10.1128/mbio.01956-15] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The live attenuated yellow fever virus (YFV) vaccine 17D stands as a “gold standard” for a successful vaccine. 17D was developed empirically by passaging the wild-type Asibi strain in mouse and chicken embryo tissues. Despite its immense success, the molecular determinants for virulence attenuation and immunogenicity of the 17D vaccine are poorly understood. 17D evolved several mutations in its genome, most of which lie within the envelope (E) protein. Given the major role played by the YFV E protein during virus entry, it has been hypothesized that the residues that diverge between the Asibi and 17D E proteins may be key determinants of attenuation. In this study, we define the process of YFV entry into target cells and investigate its implication in the activation of the antiviral cytokine response. We found that Asibi infects host cells exclusively via the classical clathrin-mediated endocytosis, while 17D exploits a clathrin-independent pathway for infectious entry. We demonstrate that the mutations in the 17D E protein acquired during the attenuation process are sufficient to explain the differential entry of Asibi versus 17D. Interestingly, we show that 17D binds to and infects host cells more efficiently than Asibi, which culminates in increased delivery of viral RNA into the cytosol and robust activation of the cytokine-mediated antiviral response. Overall, our study reveals that 17D vaccine and Asibi enter target cells through distinct mechanisms and highlights a link between 17D attenuation, virus entry, and immune activation. The yellow fever virus (YFV) vaccine 17D is one of the safest and most effective live virus vaccines ever developed. The molecular determinants for virulence attenuation and immunogenicity of 17D are poorly understood. 17D was generated by serially passaging the virulent Asibi strain in vertebrate tissues. Here we examined the entry mechanisms engaged by YFV Asibi and the 17D vaccine. We found the two viruses use different entry pathways. We show that the mutations differentiating the Asibi envelope (E) protein from the 17D E protein, which arose during attenuation, are key determinants for the use of these distinct entry routes. Finally, we demonstrate that 17D binds and enters host cells more efficiently than Asibi. This results in a higher uptake of viral RNA into the cytoplasm and consequently a greater cytokine-mediated antiviral response. Overall, our data provide new insights into the biology of YFV infection and the mechanisms of viral attenuation.
Collapse
|
42
|
Abstract
WNV infectious clones are valuable tools for elucidating WNV biology. Nevertheless, relatively few infectious WNV clones have been generated because their construction is hampered by the instability of flaviviral genomes. More recently, advances in cloning techniques as well as the development of several two-plasmid WNV infectious clone systems have facilitated the generation of WNV infectious clones. Here we described a protocol for recovering WNV from a two-plasmid system. In this approach, large quantities of these constructs are digested with restriction enzymes to produce complementary restriction sites at the 3' end of the upstream fragment and the 5' end of the downstream fragment. These fragments are then annealed to produce linear template for in vitro transcription to synthesize infectious RNA. The resulting RNA is transfected into cells and after several days WNV is recovered in the culture supernatant. This method can be used to generate virus from infectious clones encoding high- and low-pathogenicity strains of WNV, as well as chimeric virues.
Collapse
|
43
|
Zhang PT, Shan C, Li XD, Liu SQ, Deng CL, Ye HQ, Shang BD, Shi PY, Lv M, Shen BF, Qin CF, Zhang B. Generation of a recombinant West Nile virus stably expressing the Gaussia luciferase for neutralization assay. Virus Res 2016; 211:17-24. [DOI: 10.1016/j.virusres.2015.09.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 09/22/2015] [Accepted: 09/23/2015] [Indexed: 10/23/2022]
|
44
|
Gullberg RC, Jordan Steel J, Moon SL, Soltani E, Geiss BJ. Oxidative stress influences positive strand RNA virus genome synthesis and capping. Virology 2014; 475:219-29. [PMID: 25514423 PMCID: PMC4332586 DOI: 10.1016/j.virol.2014.10.037] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 10/21/2014] [Indexed: 12/24/2022]
Abstract
Flaviviruses are 5′ capped positive-stranded RNA viruses that replicate their genomes within endoplasmic reticulum-derived vesicles. Flaviviruses are well known to induce oxidative stress late in infection but it is unknown if oxidative stress plays a positive role in the viral RNA replication cycle. We therefore examined how oxidation affects flavivirus RNA replication. We found that antioxidant treatment reduced virus production, reduced the viral positive-to-negative strand RNA ratio, and resulted in the accumulation of uncapped positive-sense viral RNAs. Treatment of the NS5 RNA capping enzyme in vitro with oxidizing agents enhanced guanylyltransferase activity, indicating that the guanylyltransferase function of the flavivirus NS5 RNA capping enzyme is activated by oxidative conditions. Antioxidant treatment also reduced alphavirus RNA replication and protein expression while enhancing nsP1 capping activity. These findings suggest that RNA viruses may utilize oxidative stress induced during infection to help temporally control genome RNA capping and genome replication.
Collapse
Affiliation(s)
- Rebekah C Gullberg
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - J Jordan Steel
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Stephanie L Moon
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Elnaz Soltani
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Brian J Geiss
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA; Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
45
|
Flavivirus reverse genetic systems, construction techniques and applications: a historical perspective. Antiviral Res 2014; 114:67-85. [PMID: 25512228 PMCID: PMC7173292 DOI: 10.1016/j.antiviral.2014.12.007] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 11/26/2014] [Accepted: 12/03/2014] [Indexed: 12/20/2022]
Abstract
The study of flaviviruses, which cause some of the most important emerging tropical and sub-tropical human arbovirus diseases, has greatly benefited from the use of reverse genetic systems since its first development for yellow fever virus in 1989. Reverse genetics technology has completely revolutionized the study of these viruses, making it possible to manipulate their genomes and evaluate the direct effects of these changes on their biology and pathogenesis. The most commonly used reverse genetics system is the infectious clone technology. Whilst flavivirus infectious clones provide a powerful tool, their construction as full-length cDNA molecules in bacterial vectors can be problematic, laborious and time consuming, because they are often unstable, contain unwanted induced substitutions and may be toxic for bacteria due to viral protein expression. The incredible technological advances that have been made during the past 30years, such as the use of PCR or new sequencing methods, have allowed the development of new approaches to improve preexisting systems or elaborate new strategies that overcome these problems. This review summarizes the evolution and major technical breakthroughs in the development of flavivirus reverse genetics technologies and their application to the further understanding and control of these viruses and their diseases.
Collapse
|
46
|
Sensing of immature particles produced by dengue virus infected cells induces an antiviral response by plasmacytoid dendritic cells. PLoS Pathog 2014; 10:e1004434. [PMID: 25340500 PMCID: PMC4207819 DOI: 10.1371/journal.ppat.1004434] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 08/29/2014] [Indexed: 11/19/2022] Open
Abstract
Dengue virus (DENV) is the leading cause of mosquito-borne viral illness and death in humans. Like many viruses, DENV has evolved potent mechanisms that abolish the antiviral response within infected cells. Nevertheless, several in vivo studies have demonstrated a key role of the innate immune response in controlling DENV infection and disease progression. Here, we report that sensing of DENV infected cells by plasmacytoid dendritic cells (pDCs) triggers a robust TLR7-dependent production of IFNα, concomitant with additional antiviral responses, including inflammatory cytokine secretion and pDC maturation. We demonstrate that unlike the efficient cell-free transmission of viral infectivity, pDC activation depends on cell-to-cell contact, a feature observed for various cell types and primary cells infected by DENV, as well as West Nile virus, another member of the Flavivirus genus. We show that the sensing of DENV infected cells by pDCs requires viral envelope protein-dependent secretion and transmission of viral RNA. Consistently with the cell-to-cell sensing-dependent pDC activation, we found that DENV structural components are clustered at the interface between pDCs and infected cells. The actin cytoskeleton is pivotal for both this clustering at the contacts and pDC activation, suggesting that this structural network likely contributes to the transmission of viral components to the pDCs. Due to an evolutionarily conserved suboptimal cleavage of the precursor membrane protein (prM), DENV infected cells release uncleaved prM containing-immature particles, which are deficient for membrane fusion function. We demonstrate that cells releasing immature particles trigger pDC IFN response more potently than cells producing fusion-competent mature virus. Altogether, our results imply that immature particles, as a carrier to endolysosome-localized TLR7 sensor, may contribute to regulate the progression of dengue disease by eliciting a strong innate response.
Collapse
|
47
|
Pu SY, Wu RH, Tsai MH, Yang CC, Chang CM, Yueh A. A novel approach to propagate flavivirus infectious cDNA clones in bacteria by introducing tandem repeat sequences upstream of virus genome. J Gen Virol 2014; 95:1493-1503. [DOI: 10.1099/vir.0.064915-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Despite tremendous efforts to improve the methodology for constructing flavivirus infectious cDNAs, the manipulation of flavivirus cDNAs remains a difficult task in bacteria. Here, we successfully propagated DNA-launched type 2 dengue virus (DENV2) and Japanese encephalitis virus (JEV) infectious cDNAs by introducing seven repeats of the tetracycline-response element (7×TRE) and a minimal cytomegalovirus (CMVmin) promoter upstream of the viral genome. Insertion of the 7×TRE-CMVmin sequence upstream of the DENV2 or JEV genome decreased the cryptic E. coli promoter (ECP) activity of the viral genome in bacteria, as measured using fusion constructs containing DENV2 or JEV segments and the reporter gene Renilla luciferase in an empty vector. The growth kinetics of recombinant viruses derived from DNA-launched DENV2 and JEV infectious cDNAs were similar to those of parental viruses. Similarly, RNA-launched DENV2 infectious cDNAs were generated by inserting 7×TRE-CMVmin, five repeats of the GAL4 upstream activating sequence, or five repeats of BamHI linkers upstream of the DENV2 genome. All three tandem repeat sequences decreased the ECP activity of the DENV2 genome in bacteria. Notably, 7×TRE-CMVmin stabilized RNA-launched JEV infectious cDNAs and reduced the ECP activity of the JEV genome in bacteria. The growth kinetics of recombinant viruses derived from RNA-launched DENV2 and JEV infectious cDNAs displayed patterns similar to those of the parental viruses. These results support a novel methodology for constructing flavivirus infectious cDNAs, which will facilitate research in virology, viral pathogenesis and vaccine development of flaviviruses and other RNA viruses.
Collapse
Affiliation(s)
- Szu-Yuan Pu
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, R.O.C
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 350, Taiwan, R.O.C
| | - Ren-Huang Wu
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, R.O.C
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 350, Taiwan, R.O.C
| | - Ming-Han Tsai
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 350, Taiwan, R.O.C
| | - Chi-Chen Yang
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, R.O.C
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 350, Taiwan, R.O.C
| | - Chung-Ming Chang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 350, Taiwan, R.O.C
| | - Andrew Yueh
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, R.O.C
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 350, Taiwan, R.O.C
| |
Collapse
|
48
|
Generation of West Nile virus infectious clones containing amino acid insertions between capsid and capsid anchor. Viruses 2014; 6:1637-53. [PMID: 24721788 PMCID: PMC4014714 DOI: 10.3390/v6041637] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 03/19/2014] [Accepted: 03/21/2014] [Indexed: 12/13/2022] Open
Abstract
West Nile virus (WNV) is a positive-sense RNA arbovirus responsible for recent outbreaks of severe neurological disease within the US and Europe. Large-scale analyses of antiviral compounds that inhibit virus replication have been limited due to the lack of an adequate WN reporter virus. Previous attempts to insert a reporter into the 3' untranslated region of WNV generated unstable viruses, suggesting that this region does not accommodate additional nucleotides. Here, we engineered two WNV infectious clones containing insertions at the Capsid (C)/Capsid Anchor (CA) junction of the viral polyprotein. Recombinant viruses containing a TAT(1-67) or Gaussia Luciferase (GLuc) gene at this location were successfully recovered. However, rapid loss of most, if not all, of the reporter sequence occurred for both viruses, indicating that the reporter viruses were not stable. While the GLuc viruses predominantly reverted back to wild-type WNV length, the TAT viruses retained up to 75 additional nucleotides of the reporter sequence. These additional nucleotides were stable over at least five passages and did not significantly alter WNV fitness. Thus, the C/CA junction of WNV can tolerate additional nucleotides, though insertions are subject to certain constraints.
Collapse
|
49
|
Bonaldo MC, Sequeira PC, Galler R. The yellow fever 17D virus as a platform for new live attenuated vaccines. Hum Vaccin Immunother 2014; 10:1256-65. [PMID: 24553128 DOI: 10.4161/hv.28117] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The live-attenuated yellow fever 17D virus is one of the most outstanding human vaccines ever developed. It induces efficacious immune responses at a low production cost with a well-established manufacture process. These advantages make the YF17D virus attractive as a vector for the development of new vaccines. At the beginning of vector development studies, YF17D was genetically manipulated to express other flavivirus prM and E proteins, components of the viral envelope. While these 17D recombinants are based on the substitution of equivalent YF17D genes, other antigens from unrelated pathogens have also been successfully expressed and delivered by recombinant YF17D viruses employing alternative strategies for genetic manipulation of the YF17D genome. Herein, we discuss these strategies in terms of possibilities of single epitope or larger sequence expression and the main properties of these replication-competent viral platforms.
Collapse
Affiliation(s)
- Myrna C Bonaldo
- Laboratório de Biologia Molecular de Flavivírus, IOC, Fiocruz; Rio de Janeiro, Brazil
| | - Patrícia C Sequeira
- Laboratório de Biologia Molecular de Flavivírus, IOC, Fiocruz; Rio de Janeiro, Brazil
| | - Ricardo Galler
- Instituto de Tecnologia em Imunobiológicos, Fiocruz, Rio de Janeiro, Brazil
| |
Collapse
|
50
|
Song KY, Zhao H, Jiang ZY, Li XF, Deng YQ, Jiang T, Zhu SY, Shi PY, Zhang B, Zhang FC, Qin ED, Qin CF. A novel reporter system for neutralizing and enhancing antibody assay against dengue virus. BMC Microbiol 2014; 14:44. [PMID: 24548533 PMCID: PMC3930823 DOI: 10.1186/1471-2180-14-44] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 02/12/2014] [Indexed: 12/21/2022] Open
Abstract
Background Dengue virus (DENV) still poses a global public health threat, and no vaccine or antiviral therapy is currently available. Antibody plays distinct roles in controlling DENV infections. Neutralizing antibody is protective against DENV infection, whereas sub-neutralizing concentration of antibody can increase DENV infection, termed antibody-dependent enhancement (ADE). Plaque-based assay represents the most widely accepted method measuring neutralizing or enhancing antibodies. Results In this study, a novel reporter virus-based system was developed for measuring neutralization and ADE activity. A stable Renilla luciferase reporter DENV (Luc-DENV) that can produce robust luciferase signals in BHK-21 and K562 cells were used to establish the assay and validated against traditional plaque-based assay. Luciferase value analysis using various known DENV-specific monoclonal antibodies showed good repeatability and a well linear correlation with conventional plaque-based assays. The newly developed assay was finally validated with clinical samples from infected animals and individuals. Conclusions This reporter virus-based assay for neutralizing and enhancing antibody evaluation is rapid, lower cost, and high throughput, and will be helpful for laboratory detection and epidemiological investigation for DENV antibodies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Cheng-Feng Qin
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China.
| |
Collapse
|