1
|
Gao Z, Wang B, Liu T, Zhao Z, Xu J, Zhao X, Zhang Z, Jia Z, Yang Y, Wu S, Chen W, Hou L. Design and construction of a fast synthetic modified vaccinia virus Ankara reverse genetics system for advancing vaccine development. Front Microbiol 2025; 16:1572706. [PMID: 40351316 PMCID: PMC12061933 DOI: 10.3389/fmicb.2025.1572706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 04/07/2025] [Indexed: 05/14/2025] Open
Abstract
The modified vaccinia virus Ankara (MVA) is approved for use as a smallpox and monkeypox virus vaccine and was also designed as a popular recombinant viral vector for vaccine development and gene therapy. However, the extensive genomes of poxviruses present a significant challenge for the development of recombinant viral vaccines; therefore, it is essential to establish a user-friendly in vitro reverse genetic system. We systematically assembled the 180-kb MVA genome into a five-plasmid system, facilitating one-step packaging of the MVA virus. The MVA rescued using this system exhibited similar virological characteristics, including host cell tropism, growth kinetics, plaque size, and viral particles, comparable to those of wild-type MVA. Immunization with rescued MVA intramuscularly or subcutaneously triggered robust-specific immune responses and conferred protection against lethal attacks by the ectromelia virus in mice. We also developed a recombinant MVA-Luc-eGFP virus, which served as a tool for screening antiviral compounds against poxviruses. The synthetic MVA system efficiently generates recombinant vaccines with robust immune responses. These findings provide a novel and fast method for engineering large viral genomes with more specialized structures and lay a foundation for the advancement of more rapid and effective viral vector vaccines.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Lihua Hou
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China
| |
Collapse
|
2
|
Okita G, Suenaga K, Sakaguchi M, Murakami T. A novel oncolytic vaccinia virus with multiple gene modifications involved in viral replication and maturation increases safety for intravenous administration while maintaining proliferative potential in cancer cells. PLoS One 2025; 20:e0312205. [PMID: 40048445 PMCID: PMC11884718 DOI: 10.1371/journal.pone.0312205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 09/06/2024] [Indexed: 03/09/2025] Open
Abstract
To generate a novel oncolytic vaccinia virus with improved safety and productivity, the genome of smallpox vaccine strain LC16m8 was modified by a bacterial artificial chromosome system. By using LC16m8, a replicating virus homologous to the target virus, as a helper virus for the bacterial artificial chromosome system, we successfully recovered genome-edited infectious viruses. Oncolytic viruses with limited growth in normal cells were obtained by deleting the genes for vaccinia virus growth factor (VGF), extracellular signal-regulated kinase-activating protein (O1L), and ribonucleotide reductase (RNR) present in the viral genome. Furthermore, the amino acid residues of seven proteins involved in extracellular enveloped virus virion formation were replaced to the IHD-J strain sequence, which is known to highly express extracellular enveloped virus. In cultured cancer cells (HeLa), these modified viruses showed cytotoxicity and increased productivity, but it was confirmed that the cytotoxicity was suppressed in normal cells (normal human dermal fibroblasts). For in vivo safety evaluation, a modified virus (MD-RVV-ΔRR-EEV6) in which the VGF, O1L, and RNR genes of LC16m8 were deleted and the genes of six extracellular enveloped virus-associated proteins were replaced with sequences derived from IHD-J strain, and another modified virus (MD-RVV) lacking only the VGF and O1L were administered intravenously to severe combined immunodeficiency mice. In the MD-RVV administration, animals in all dose groups died by 40 days after virus administration. On the other hand, after MD-RVV-ΔRR-EEV6 administration, 3 out of 5 animals in the high and medium dose groups and all animals in the low dose group were still alive by day 71, the end of the observation period. These results demonstrate that genome editing of oncolytic vaccinia virus can delete genes involved in viral replication to improve safety in normal cells, while replacing genes involved in maturation improves proliferative potential in cancer cells.
Collapse
Affiliation(s)
- Go Okita
- Research Department, KM Biologics Co., Ltd., Kikuchi, Kumamoto, Japan
| | - Kiyotaka Suenaga
- Research Department, KM Biologics Co., Ltd., Kikuchi, Kumamoto, Japan
| | - Masashi Sakaguchi
- Research Department, KM Biologics Co., Ltd., Kikuchi, Kumamoto, Japan
| | - Toshio Murakami
- Research Department, KM Biologics Co., Ltd., Kikuchi, Kumamoto, Japan
| |
Collapse
|
3
|
Chakravarty N, Hemani D, Paravastu R, Ahmad Z, Palani SN, Arumugaswami V, Kumar A. Mpox Virus and its ocular surface manifestations. Ocul Surf 2024; 34:108-121. [PMID: 38972544 PMCID: PMC11625629 DOI: 10.1016/j.jtos.2024.07.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/28/2024] [Accepted: 07/04/2024] [Indexed: 07/09/2024]
Abstract
The Mpox virus (MPXV) is the causative agent of human Mpox disease - a debilitating rash illness similar to smallpox. Although Clade I MPXV has remained endemic to West and Central Africa, Clade II MPXV has been responsible for many outbreaks worldwide. The most recent outbreak in 2022 resulted from the rapid spread of a new clade of MPXV, classified into Clade IIb - a distinct lineage from the previously circulating viral strains. The rapid spread and increased severity of Mpox disease by the Clade IIb strain have raised the serious public health imperative of better understanding the host and viral determinants during MPXV infection. In addition to typical skin rashes, including in the periorbital area, MPXV causes moderate to severe ophthalmic manifestations - most commonly, ocular surface complications (e.g., keratitis, conjunctivitis, blepharitis). While ocular manifestations of Clade I Mpox within the Congo basin have been well-reported, global incidence trends of ocular Mpox cases by Clade IIb are still emerging. Given the demonstrated ability of all MPXV strains to auto-inoculate ocular tissue, alongside the enhanced transmissibility of the Clade IIb virus, there is an urgent need to elucidate the mechanisms by which MPXV causes ocular anomalies. In this review, we discuss the viral and genomic structures of MPXV, the epidemiology, and pathology of systemic and ocular Mpox, as well as potential prophylactic and therapeutic interventions.
Collapse
Affiliation(s)
- Nikhil Chakravarty
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA; School of Medicine, California University of Science and Medicine, Colton, CA, USA
| | - Darshi Hemani
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University, Detroit, MI, USA
| | - Ramya Paravastu
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Zeeshan Ahmad
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University, Detroit, MI, USA
| | - Sankara Naynar Palani
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Vaithilingaraja Arumugaswami
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, USA.
| | - Ashok Kumar
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University, Detroit, MI, USA.
| |
Collapse
|
4
|
Mazloum A, Karagyaur M, Chernyshev R, van Schalkwyk A, Jun M, Qiang F, Sprygin A. Post-genomic era in agriculture and veterinary science: successful and proposed application of genetic targeting technologies. Front Vet Sci 2023; 10:1180621. [PMID: 37601766 PMCID: PMC10434572 DOI: 10.3389/fvets.2023.1180621] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 07/20/2023] [Indexed: 08/22/2023] Open
Abstract
Gene editing tools have become an indispensable part of research into the fundamental aspects of cell biology. With a vast body of literature having been generated based on next generation sequencing technologies, keeping track of this ever-growing body of information remains challenging. This necessitates the translation of genomic data into tangible applications. In order to address this objective, the generated Next Generation Sequencing (NGS) data forms the basis for targeted genome editing strategies, employing known enzymes of various cellular machinery, in generating organisms with specifically selected phenotypes. This review focuses primarily on CRISPR/Cas9 technology in the context of its advantages over Zinc finger proteins (ZNF) and Transcription activator-like effector nucleases (TALEN) and meganucleases mutagenesis strategies, for use in agricultural and veterinary applications. This review will describe the application of CRISPR/Cas9 in creating modified organisms with custom-made properties, without the undesired non-targeted effects associated with virus vector vaccines and bioactive molecules produced in bacterial systems. Examples of the successful and unsuccessful applications of this technology to plants, animals and microorganisms are provided, as well as an in-depth look into possible future trends and applications in vaccine development, disease resistance and enhanced phenotypic traits will be discussed.
Collapse
Affiliation(s)
- Ali Mazloum
- Federal Center for Animal Health, Vladimir, Russia
| | - Maxim Karagyaur
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia
| | | | - Antoinette van Schalkwyk
- Agricultural Research Council-Onderstepoort Veterinary Institute, Onderstepoort, South Africa
- Department of Biotechnology, University of the Western Cape, Bellville, South Africa
| | - Ma Jun
- School of Life Sciences and Engineering, Foshan University, Foshan, China
| | - Fu Qiang
- School of Life Sciences and Engineering, Foshan University, Foshan, China
| | | |
Collapse
|
5
|
Yoshikawa T, Misu M, Kurosu T, Takamatsu Y, Sugimoto S, Shimojima M, Ebihara H, Saijo M. Markerless bacterial artificial chromosome manipulation method by red proteins of phage λ mediated homologous recombination utilizing fluorescent proteins for both positive and counter selection. Heliyon 2023; 9:e18983. [PMID: 37600421 PMCID: PMC10432722 DOI: 10.1016/j.heliyon.2023.e18983] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 07/25/2023] [Accepted: 08/03/2023] [Indexed: 08/22/2023] Open
Abstract
Manipulating viral genomes is an essential technique in reverse genetics and recombinant vaccine development. A strategy for manipulating large viral genomes involves introducing their entire genome into bacterial artificial chromosomes and employing Escherichia coli genetic tools. For sequence manipulation on bacterial artificial chromosomes (bacterial artificial chromosomes recombineering), a well-established method that relies on the Escherichia coli strain GS1783, and the template plasmid, pEPKan-S, is often used. This method, known as markerless DNA manipulation, allows for the generation of a recombinant bacterial artificial chromosome that does not retain the selection markers used during recombination. Although this method is highly innovative, there remains room for improvement as the plasmid is currently only available for positive selection. Additionally, differentiating true recombinants from false negatives often proves time-consuming. Consequently, an improved method for bacterial artificial chromosomes recombineering, which utilizes fluorescent proteins, has been developed. This method's core comprises three plasmids containing the I-SceI recognition site, antibiotic resistance genes (ampicillin, kanamycin, and zeocin), and fluorescent genes (YPet, mOrange, and mScarlet). The success or failure of Red recombination can be confirmed via fluorescent signals. To validate this method, the Lassa virus genes were introduced into the bacterial artificial chromosomes, containing the entire genome of the vaccinia virus strain LC16m8. Consequently, the expression of fluorescent protein genes contributed to positive selection, such as blue-white screening and counter-selection during the first and second Red recombination.
Collapse
Affiliation(s)
- Tomoki Yoshikawa
- Department of Virology 1, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashimurayama-shi, Tokyo, 208-0011, Japan
| | - Masayasu Misu
- Department of Virology 1, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashimurayama-shi, Tokyo, 208-0011, Japan
| | - Takeshi Kurosu
- Department of Virology 1, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashimurayama-shi, Tokyo, 208-0011, Japan
| | - Yuki Takamatsu
- Department of Virology 1, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashimurayama-shi, Tokyo, 208-0011, Japan
| | - Satoko Sugimoto
- Department of Virology 1, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashimurayama-shi, Tokyo, 208-0011, Japan
| | - Masayuki Shimojima
- Department of Virology 1, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashimurayama-shi, Tokyo, 208-0011, Japan
| | - Hideki Ebihara
- Department of Virology 1, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashimurayama-shi, Tokyo, 208-0011, Japan
| | - Masayuki Saijo
- Department of Virology 1, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashimurayama-shi, Tokyo, 208-0011, Japan
| |
Collapse
|
6
|
Drillien R, Pradeau-Aubreton K, Batisse J, Mezher J, Schenckbecher E, Marguin J, Ennifar E, Ruff M. Efficient production of protein complexes in mammalian cells using a poxvirus vector. PLoS One 2022; 17:e0279038. [PMID: 36520869 PMCID: PMC9754296 DOI: 10.1371/journal.pone.0279038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
The production of full length, biologically active proteins in mammalian cells is critical for a wide variety of purposes ranging from structural studies to preparation of subunit vaccines. Prior research has shown that Modified vaccinia virus Ankara encoding the bacteriophage T7 RNA polymerase (MVA-T7) is particularly suitable for high level expression of proteins upon infection of mammalian cells. The expression system is safe for users and 10-50 mg of full length, biologically active proteins may be obtained in their native state, from a few litres of infected cell cultures. Here we report further improvements which allow an increase in the ease and speed of recombinant virus isolation, the scale-up of protein production and the simultaneous synthesis of several polypeptides belonging to a protein complex using a single virus vector. Isolation of MVA-T7 viruses encoding foreign proteins was simplified by combining positive selection for virus recombinants and negative selection against parental virus, a process which eliminated the need for tedious plaque purification. Scale-up of protein production was achieved by infecting a BHK 21 suspension cell line and inducing protein expression with previously infected cells instead of virus, thus saving time and effort in handling virus stocks. Protein complexes were produced from infected cells by concatenating the Tobacco Etch Virus (TEV) N1A protease sequence with each of the genes of the complex into a single ORF, each gene being separated from the other by twin TEV protease cleavage sites. We report the application of these methods to the production of a complex formed on the one hand between the HIV-1 integrase and its cell partner LEDGF and on the other between the HIV-1 VIF protein and its cell partners APOBEC3G, CBFβ, Elo B and Elo C. The strategies developed in this study should be valuable for the overexpression and subsequent purification of numerous protein complexes.
Collapse
Affiliation(s)
- Robert Drillien
- Department of Integrative Structural Biology, IGBMC, University of Strasbourg, CNRS UMR 7104, INSERM U964, Illkirch, France
- * E-mail: (RD); (MR)
| | - Karine Pradeau-Aubreton
- Department of Integrative Structural Biology, IGBMC, University of Strasbourg, CNRS UMR 7104, INSERM U964, Illkirch, France
| | - Julien Batisse
- Department of Integrative Structural Biology, IGBMC, University of Strasbourg, CNRS UMR 7104, INSERM U964, Illkirch, France
| | - Joëlle Mezher
- Structure et Dynamique des Machines Biomoléculaires, Institut de Biologie Moléculaire et Cellulaire, UPR 9002 CNRS/Université de Strasbourg, Strasbourg, France
| | - Emma Schenckbecher
- Structure et Dynamique des Machines Biomoléculaires, Institut de Biologie Moléculaire et Cellulaire, UPR 9002 CNRS/Université de Strasbourg, Strasbourg, France
| | - Justine Marguin
- Structure et Dynamique des Machines Biomoléculaires, Institut de Biologie Moléculaire et Cellulaire, UPR 9002 CNRS/Université de Strasbourg, Strasbourg, France
| | - Eric Ennifar
- Structure et Dynamique des Machines Biomoléculaires, Institut de Biologie Moléculaire et Cellulaire, UPR 9002 CNRS/Université de Strasbourg, Strasbourg, France
| | - Marc Ruff
- Department of Integrative Structural Biology, IGBMC, University of Strasbourg, CNRS UMR 7104, INSERM U964, Illkirch, France
- * E-mail: (RD); (MR)
| |
Collapse
|
7
|
Chiozzini C, Ridolfi B, Federico M. Extracellular Vesicles and Their Use as Vehicles of Immunogens. Methods Mol Biol 2022; 2504:177-198. [PMID: 35467287 DOI: 10.1007/978-1-0716-2341-1_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Healthy cells constitutively release lipid bilayered vesicles of different sizes and recognizing different biogenesis, collectively referred to as extracellular vesicles (EVs). EVs can be distinguished in exosomes and microvesicles. Biological and biomedical research on EVs is an emerging field that is rapidly growing. Many EV features including biogenesis, cell uptake, and functions still require unambiguous elucidation. Nevertheless, it has been well established that EVs are involved in communication among cells, tissues, and organs under both healthy and disease conditions by virtue of their ability to deliver macromolecules to target cells. Here, we summarize most recent findings regarding biogenesis, structure, and functions of both exosomes and microvesicles. In addition, the use of EVs as delivery tools to induce CD8+ T cell immunity is addressed compared to current designs exploiting enveloped viral vectors and virus-like particles. Finally, we describe a both safe and original approach conceived for the induction of strong CTL immunity against antigens uploaded in EVs constitutively released by muscle cells.
Collapse
Affiliation(s)
- Chiara Chiozzini
- National Center for Global Health, Istituto Superiore di Sanità (ISS), Rome, Italy.
| | - Barbara Ridolfi
- National Center for Global Health, Istituto Superiore di Sanità (ISS), Rome, Italy
| | - Maurizio Federico
- National Center for Global Health, Istituto Superiore di Sanità (ISS), Rome, Italy
| |
Collapse
|
8
|
Yoshikawa T. Third-generation smallpox vaccine strain-based recombinant vaccines for viral hemorrhagic fevers. Vaccine 2021; 39:6174-6181. [PMID: 34521550 DOI: 10.1016/j.vaccine.2021.09.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 08/19/2021] [Accepted: 08/30/2021] [Indexed: 12/27/2022]
Abstract
Vaccinia virus has been used as a smallpox vaccine. Now that smallpox has been eradicated, the vaccinia virus is expected to be used as a bioterrorism countermeasure and a recombinant vaccine vector for other infectious diseases, such as viral hemorrhagic fevers. Many vaccinia virus strains were used as smallpox vaccines in the smallpox eradication campaign coordinated by the World Health Organization. These strains can be classified into generations, according to the history of improving production methods and efforts to reduce the adverse reactions. Significantly, the third-generation of smallpox vaccine strains, which include modified vaccinia Ankara (MVA) and LC16m8, are currently popular as recombinant vaccine vectors due to their well-balanced safety and immunogenicity profiles. The present review firstly focuses on the characteristics of the smallpox vaccine generations. The historical background of the development of the third-generation smallpox vaccine strains is detailed, along with the history of the transition of the vaccinia virus generation used as vectors for hemorrhagic fever vaccines to the third generation. Among the vaccinia viruses, MVA is currently the most commonly used vector for developing hemorrhagic fever vaccines, including dengue fever, yellow fever, Ebola viral disease, Lassa fever, Rift Valley fever, and Crimean-Congo hemorrhagic fever. LC16m8 is a vaccine candidate for severe fever with thrombocytopenia syndrome. The current status and recent advances in the development of these hemorrhagic fever vaccines using third-generation vaccinia strains are discussed.
Collapse
Affiliation(s)
- Tomoki Yoshikawa
- Department of Virology 1, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashimurayama-shi, Tokyo 208-0011, Japan.
| |
Collapse
|
9
|
Ma J, Jin C, Čančer M, Wang H, Ramachandran M, Yu D. Concurrent expression of HP-NAP enhances antitumor efficacy of oncolytic vaccinia virus but not for Semliki Forest virus. Mol Ther Oncolytics 2021; 21:356-366. [PMID: 34141872 PMCID: PMC8182386 DOI: 10.1016/j.omto.2021.04.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 04/28/2021] [Indexed: 12/12/2022] Open
Abstract
Oncolytic viruses (OVs) represent promising therapeutic agents for cancer therapy by selective oncolysis and induction of anti-tumor immunity. OVs can be engineered to express tumor-associated antigens and immune-modulating agents to provoke stronger antitumor immunity. Here, we engineered vaccinia virus (VV) and Semliki Forest virus (SFV) to express neuroblastoma-associated antigen disialoganglioside (GD2) and the immune modulator Helicobacter pylori neutrophil-activating protein (NAP) and compared their therapeutic potency. Oncolytic VV did not exhibit any antitumor benefits, whereas SFV was able to delay subcutaneous neuroblastoma (NXS2) tumor growth. Additional expression of the GD2 mimotope (GD2m) by VV-GD2m or SFV-GD2m did not improve their anti-tumor capacity compared to the parent viruses. Further arming these OVs with NAP resulted in contrasting anti-tumor efficacy. VV (VV-GD2m-NAP) significantly improved therapeutic efficacy compared to VV-GD2m, which was also associated with a significantly elevated anti-GD2 antibody, whereas there was no additive antitumor efficacy for SFV-GD2m-NAP compared to SFV-GD2m, nor was the anti-GD2 antibody response improved. Instead, NAP induced higher neutralizing antibodies against SFV. These observations suggest that distinct immune stimulation profiles are elicited when the same immunostimulatory factor is expressed by different OVs. Therefore, careful consideration and detailed characterization are needed when engineering OVs with immune-modulators.
Collapse
Affiliation(s)
- Jing Ma
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 75185, Uppsala, Sweden
| | - Chuan Jin
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 75185, Uppsala, Sweden
| | - Matko Čančer
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 75185, Uppsala, Sweden
| | - Hai Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mohanraj Ramachandran
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 75185, Uppsala, Sweden
| | - Di Yu
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 75185, Uppsala, Sweden
| |
Collapse
|
10
|
Latest Advances of Virology Research Using CRISPR/Cas9-Based Gene-Editing Technology and Its Application to Vaccine Development. Viruses 2021; 13:v13050779. [PMID: 33924851 PMCID: PMC8146441 DOI: 10.3390/v13050779] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 02/07/2023] Open
Abstract
In recent years, the CRISPR/Cas9-based gene-editing techniques have been well developed and applied widely in several aspects of research in the biological sciences, in many species, including humans, animals, plants, and even in viruses. Modification of the viral genome is crucial for revealing gene function, virus pathogenesis, gene therapy, genetic engineering, and vaccine development. Herein, we have provided a brief review of the different technologies for the modification of the viral genomes. Particularly, we have focused on the recently developed CRISPR/Cas9-based gene-editing system, detailing its origin, functional principles, and touching on its latest achievements in virology research and applications in vaccine development, especially in large DNA viruses of humans and animals. Future prospects of CRISPR/Cas9-based gene-editing technology in virology research, including the potential shortcomings, are also discussed.
Collapse
|
11
|
Laudermilch E, Chandran K. MAVERICC: Marker-free Vaccinia Virus Engineering of Recombinants through in vitro CRISPR/Cas9 Cleavage. J Mol Biol 2021; 433:166896. [PMID: 33639215 DOI: 10.1016/j.jmb.2021.166896] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/18/2021] [Accepted: 02/19/2021] [Indexed: 12/23/2022]
Abstract
Vaccinia virus (VACV)-based vectors are in extensive use as vaccines and cancer immunotherapies. VACV engineering has traditionally relied on homologous recombination between a parental viral genome and a transgene-bearing transfer plasmid, an inefficient process that necessitates the use of a selection or screening marker to isolate recombinants. Recent extensions of this approach have sought to enhance the recovery of transgene-bearing viruses through the use of CRISPR-Cas9 engineering to cleave the viral genome in infected cells. However, these methods do not completely eliminate the generation of WT viral progeny and thus continue to require multiple rounds of viral propagation and plaque purification. Here, we describe MAVERICC (marker-free vaccinia virus engineering of recombinants through in vitroCRISPR/Cas9 cleavage), a new strategy to engineer recombinant VACVs in a manner that overcomes current limitations. MAVERICC also leverages the CRISPR/Cas9 system but requires no markers and yields essentially pure preparations of the desired recombinants in a single step. We used this approach to introduce point mutations, insertions, and deletions at multiple locations in the VACV genome, both singly and in combination. The efficiency and versatility of MAVERICC make it an ideal choice for generating mutants and mutant libraries at arbitrarily selected locations in the viral genome to build complex VACV vectors, effect vector improvements, and facilitate the study of poxvirus biology.
Collapse
Affiliation(s)
- Ethan Laudermilch
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, United States.
| |
Collapse
|
12
|
Vashee S, Arfi Y, Lartigue C. Budding yeast as a factory to engineer partial and complete microbial genomes. CURRENT OPINION IN SYSTEMS BIOLOGY 2020; 24:1-8. [PMID: 33015421 PMCID: PMC7523139 DOI: 10.1016/j.coisb.2020.09.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Yeast cells have long been used as hosts to propagate exogenous DNA. Recent progress in genome editing opens new avenues in synthetic biology. These developments allow the efficient engineering of microbial genomes in Saccharomyces cerevisiae that can then be rescued to yield modified bacteria/viruses. Recent examples show that the ability to quickly synthesize, assemble, and/or modify viral and bacterial genomes may be a critical factor to respond to emerging pathogens. However, this process has some limitations. DNA molecules much larger than two megabase pairs are complex to clone, bacterial genomes have proven to be difficult to rescue, and the dual-use potential of these technologies must be carefully considered. Regardless, the use of yeast as a factory has enormous appeal for biological applications.
Collapse
Affiliation(s)
| | - Yonathan Arfi
- Univ. Bordeaux, INRAE, Biologie du Fruit et Pathologie, UMR 1332, F-33140, Villenave d'Ornon, France
| | - Carole Lartigue
- Univ. Bordeaux, INRAE, Biologie du Fruit et Pathologie, UMR 1332, F-33140, Villenave d'Ornon, France
| |
Collapse
|
13
|
Matía A, Lorenzo MM, Blasco R. Tools for the targeted genetic modification of poxvirus genomes. Curr Opin Virol 2020; 44:183-190. [PMID: 33242829 DOI: 10.1016/j.coviro.2020.10.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/27/2020] [Accepted: 10/27/2020] [Indexed: 12/14/2022]
Abstract
The potential of viruses as biotechnology platforms is becoming more appealing due to technological advances in synthetic biology techniques and to the increasing accessibility of means to manipulate virus genomes. Among viral systems, poxviruses, and their prototype member Vaccinia Virus, are one of the outstanding choices for different biotechnological and medical applications based on heterologous gene expression, recombinant vaccines or oncolytic viruses. The refinement of genetic engineering methods on Vaccinia Virus over the last decades have contributed to facilitate the manipulation of the genomes of poxviruses, and may aid in the improvement of virus variants designed for different goals through reverse genetic approaches. Targeted genetic changes are usually performed by homologous recombination with the viral genome. In addition to the classic approach, recent methodological advances that may assist new strategies for the mutation or edition of poxvirus genomes are reviewed.
Collapse
Affiliation(s)
- Alejandro Matía
- Departamento de Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (I.N.I.A.), Ctra. La Coruña km 7.5, E-28040 Madrid, Spain
| | - María M Lorenzo
- Departamento de Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (I.N.I.A.), Ctra. La Coruña km 7.5, E-28040 Madrid, Spain
| | - Rafael Blasco
- Departamento de Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (I.N.I.A.), Ctra. La Coruña km 7.5, E-28040 Madrid, Spain.
| |
Collapse
|
14
|
X69R Is a Non-Essential Gene That, When Deleted from African Swine Fever, Does Not Affect Virulence in Swine. Viruses 2020; 12:v12090918. [PMID: 32825617 PMCID: PMC7551905 DOI: 10.3390/v12090918] [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: 06/01/2020] [Accepted: 08/17/2020] [Indexed: 12/21/2022] Open
Abstract
African swine fever virus (ASFV) is currently causing devastating outbreaks in Asia and Europe, and the ASFV strain Georgia (ASFV-G) is responsible for these outbreaks. ASFV-G is highly virulent and continues to be maintained in these outbreak areas, apparently without suffering significant genomic or phenotypic changes. When comparing the genome of ASFV-G to other isolates, a thus-far uncharacterized gene, X69R, is highly conserved and, interestingly, is similar to another ASFV uncharacterized gene, J64R. All sequenced ASFV isolates have one or both of these genes, X69R or J64R, suggesting that the presence of at least one of these genes may be necessary for ASFV replication and or virulence. The X69R gene is present in the ASFV-G genome while J64R is absent. To assess the importance of X69R in ASFV-G functionality, we developed a recombinant virus by deleting the X69R gene from the ASFV-G genome (ASFV-G-ΔX69R). ASFV-G-ΔX69R had the same replication kinetics in primary swine macrophage cultures as the parental ASFV-G, indicating that the X69R gene is not essential for ASFV-G viability or efficient replication in the main target cell during in vivo infection. In addition, swine intramuscularly inoculated with a low dose (102 HAD50) of ASFV-G-ΔX69R developed a clinical disease indistinguishable from that induced by the same dose of the virulent parental ASFV-G isolate. Viremia values of ASFV-G-ΔX69R did not significantly differ from those detected in animals infected with parental virus. Therefore, deletion of the X69R gene from ASFV-G does not affect virus replication or virulence in swine.
Collapse
|
15
|
Moss B. Investigating Viruses During the Transformation of Molecular Biology: Part II. Annu Rev Virol 2020; 7:15-36. [PMID: 32392458 DOI: 10.1146/annurev-virology-021020-100558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
My scientific career started at an extraordinary time, shortly after the discoveries of the helical structure of DNA, the central dogma of DNA to RNA to protein, and the genetic code. Part I of this series emphasizes my education and early studies highlighted by the isolation and characterization of numerous vaccinia virus enzymes, determination of the cap structure of messenger RNA, and development of poxviruses as gene expression vectors for use as recombinant vaccines. Here I describe a shift in my research focus to combine molecular biology and genetics for a comprehensive understanding of poxvirus biology. The dominant paradigm during the early years was to select a function, isolate the responsible proteins, and locate the corresponding gene, whereas later the common paradigm was to select a gene, make a mutation, and determine the altered function. Motivations, behind-the-scenes insights, importance of new technologies, and the vital roles of trainees and coworkers are emphasized.
Collapse
Affiliation(s)
- Bernard Moss
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA;
| |
Collapse
|
16
|
Meyer H, Ehmann R, Smith GL. Smallpox in the Post-Eradication Era. Viruses 2020; 12:E138. [PMID: 31991671 PMCID: PMC7077202 DOI: 10.3390/v12020138] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 01/20/2020] [Accepted: 01/22/2020] [Indexed: 12/19/2022] Open
Abstract
Widespread vaccination programmes led to the global eradication of smallpox, which was certified by the World Health Organisation (WHO), and, since 1978, there has been no case of smallpox anywhere in the world. However, the viable variola virus (VARV), the causative agent of smallpox, is still kept in two maximum security laboratories in Russia and the USA. Despite the eradication of the disease smallpox, clandestine stocks of VARV may exist. In a rapidly changing world, the impact of an intentional VARV release in the human population would nowadays result in a public health emergency of global concern: vaccination programmes were abolished, the percentage of immunosuppressed individuals in the human population is higher, and an increased intercontinental air travel allows for the rapid viral spread of diseases around the world. The WHO has authorised the temporary retention of VARV to enable essential research for public health benefit to take place. This work aims to develop diagnostic tests, antiviral drugs, and safer vaccines. Advances in synthetic biology have made it possible to produce infectious poxvirus particles from chemicals in vitro so that it is now possible to reconstruct VARV. The status of smallpox in the post-eradication era is reviewed.
Collapse
Affiliation(s)
- Hermann Meyer
- Bundeswehr Institute of Microbiology, 80937 Munich, Germany
| | - Rosina Ehmann
- Bundeswehr Institute of Microbiology, 80937 Munich, Germany
| | - Geoffrey L. Smith
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK;
| |
Collapse
|
17
|
Tournier JN. [The eradication of infectious viral diseases endangered by advances in synthetic biology]. Med Sci (Paris) 2019; 35:181-186. [PMID: 30774089 DOI: 10.1051/medsci/2019005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The eradication of infectious diseases is one of the oldest dreams of mankind. It has been materialized only once in History with smallpox in 1980. Considerable efforts are being developed against poliomyelitis viruses since 1988, but the ultimate goal of eradication is not yet achieved. Paradoxically, while the objective of having eradicated these two viral diseases is approaching, synthetic biology multiplies the prowesses of virus "neosynthesis", imperiling at least virtually the durability of these advances. This article emphasizes the potential of a new biology on one side, and the difficult reality of the fight against infections on the other.
Collapse
Affiliation(s)
- Jean-Nicolas Tournier
- Institut de recherche biomédicale des armées, Unité Biothérapies anti-infectieuses et immunité, Département Microbiologie et maladies infectieuses, 1, place général Valérie André, 91220 Brétigny-sur-Orge, France - Institut Pasteur, unité génomique virale et vaccination, CNRS UMR-3569, 25, rue du Docteur Roux, 75015 Paris, France - École du Val-de-Grâce, 74, boulevard de Port-Royal, 75005 Paris, France
| |
Collapse
|
18
|
Affiliation(s)
- Volker Thiel
- Department of Infectious Diseases and Pathobiology, University of Bern, Bern, Switzerland
- Federal Department of Home Affairs, Institute of Virology and Immunology, Bern and Mittelhäusern, Switzerland
| |
Collapse
|
19
|
Affiliation(s)
- Ryan S. Noyce
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, Alberta, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| | - David H. Evans
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, Alberta, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
- * E-mail:
| |
Collapse
|
20
|
Luteijn RD, Drexler I, Smith GL, Lebbink RJ, Wiertz EJHJ. Mutagenic repair of double-stranded DNA breaks in vaccinia virus genomes requires cellular DNA ligase IV activity in the cytosol. J Gen Virol 2018; 99:790-804. [PMID: 29676720 PMCID: PMC7614823 DOI: 10.1099/jgv.0.001034] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Poxviruses comprise a group of large dsDNA viruses that include members relevant to human and animal health, such as variola virus, monkeypox virus, cowpox virus and vaccinia virus (VACV). Poxviruses are remarkable for their unique replication cycle, which is restricted to the cytoplasm of infected cells. The independence from the host nucleus requires poxviruses to encode most of the enzymes involved in DNA replication, transcription and processing. Here, we use the CRISPR/Cas9 genome engineering system to induce DNA damage to VACV (strain Western Reserve) genomes. We show that targeting CRISPR/Cas9 to essential viral genes limits virus replication efficiently. Although VACV is a strictly cytoplasmic pathogen, we observed extensive viral genome editing at the target site; this is reminiscent of a non-homologous end-joining DNA repair mechanism. This pathway was not dependent on the viral DNA ligase, but critically involved the cellular DNA ligase IV. Our data show that DNA ligase IV can act outside of the nucleus to allow repair of dsDNA breaks in poxvirus genomes. This pathway might contribute to the introduction of mutations within the genome of poxviruses and may thereby promote the evolution of these viruses.
Collapse
Affiliation(s)
- Rutger David Luteijn
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands.,Present address: Department of Molecular and Cell Biology, University of California, Berkeley, USA
| | - Ingo Drexler
- Institute for Virology, University Hospital Düsseldorf, Düsseldorf, Germany
| | | | - Robert Jan Lebbink
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Emmanuel J H J Wiertz
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
21
|
Ramírez M, Santos S, Martínez O, Rodríguez R, Miranda E, Ramos-Perez WD, Otero M. Characterization of the immune response elicited by the vaccinia virus L3 protein delivered as naked DNA. Vaccine 2018. [PMID: 29525282 PMCID: PMC6065253 DOI: 10.1016/j.vaccine.2018.02.033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Poxviruses are complex dsDNA viruses with over 200 genes, many of them with unknown role in the stimulation of immune responses. Among these, the vaccinia virus (VACV) L3L ORF encodes an essential protein for the transcription of the VACV early genes. To the best of our knowledge, the immune response elicited by L3 has not been characterized. In this regard, our data describes a DNA L3-coding plasmid (pL3L) that stimulates both, humoral- and cell-mediated immune responses in a mouse model. Cell-mediated immune responses were measured by IFN-γ and IL-4 ELISPOT assays. We performed CD8+ cells depletion and flow cytometry analysis to account for the contribution of cytotoxic T lymphocytes in the IFN-γ production. Moreover, results from ELISPOT were confirmed by measuring the concentration of IL-4 and IFN-γ in supernatant of antigen-stimulated splenocytes by cytokine ELISA. Additionally, dominant antigenic regions of L3 protein were identified by epitope mapping analysis. Humoral immune responses were assessed by ELISA. Specifically, the production of total IgG, IgG1 (TH-2) and IgG2a (TH-1) were determined one week after the final immunization. Our ELISPOT data shows pL3L-immunized animals to produce significantly higher frequencies of IFN-γ Spot-Forming Cells (SFC) versus controls. IL-4 levels remained unchanged in all three groups, demonstrating the increase in antigen-specific IFN-γ releasing cells. Flow cytometry assay results showed that CD8+ T cells are a major contributor to the production of IFN-γ. Moreover, our formulation enhances the production of total IgG, predominantly IgG2a isotype. Immunization with pL3L promotes a robust cytotoxic immune response, crucial against viral pathogens. In addition, our vaccine candidate promotes an increase in IgG levels, especially IgG2a (TH-1 type). Our data encourages further studies of L3 as a novel antigen in vaccine development against poxviruses.
Collapse
Affiliation(s)
- Maite Ramírez
- Department of Microbiology, University of Puerto Rico, Medical Sciences Campus, San Juan, PR, USA.
| | - Saritza Santos
- Department of Microbiology, University of Puerto Rico, Medical Sciences Campus, San Juan, PR, USA.
| | - Osmarie Martínez
- Department of Microbiology, University of Puerto Rico, Medical Sciences Campus, San Juan, PR, USA.
| | - Ricardo Rodríguez
- University of Puerto Rico, Medical Sciences Campus, School of Medicine, San Juan, PR, USA.
| | - Eric Miranda
- Department of Microbiology, University of Puerto Rico, Medical Sciences Campus, San Juan, PR, USA.
| | - Willy D Ramos-Perez
- Department of Microbiology, University of Puerto Rico, Medical Sciences Campus, San Juan, PR, USA.
| | - Miguel Otero
- Department of Microbiology, University of Puerto Rico, Medical Sciences Campus, San Juan, PR, USA.
| |
Collapse
|
22
|
Albarnaz JD, Torres AA, Smith GL. Modulating Vaccinia Virus Immunomodulators to Improve Immunological Memory. Viruses 2018; 10:E101. [PMID: 29495547 PMCID: PMC5869494 DOI: 10.3390/v10030101] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 02/21/2018] [Accepted: 02/22/2018] [Indexed: 12/14/2022] Open
Abstract
The increasing frequency of monkeypox virus infections, new outbreaks of other zoonotic orthopoxviruses and concern about the re-emergence of smallpox have prompted research into developing antiviral drugs and better vaccines against these viruses. This article considers the genetic engineering of vaccinia virus (VACV) to enhance vaccine immunogenicity and safety. The virulence, immunogenicity and protective efficacy of VACV strains engineered to lack specific immunomodulatory or host range proteins are described. The ultimate goal is to develop safer and more immunogenic VACV vaccines that induce long-lasting immunological memory.
Collapse
Affiliation(s)
- Jonas D Albarnaz
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK.
| | - Alice A Torres
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK.
| | - Geoffrey L Smith
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK.
| |
Collapse
|
23
|
Construction and characterization of bacterial artificial chromosomes harboring the full-length genome of a highly attenuated vaccinia virus LC16m8. PLoS One 2018; 13:e0192725. [PMID: 29474493 PMCID: PMC5825015 DOI: 10.1371/journal.pone.0192725] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 01/29/2018] [Indexed: 01/01/2023] Open
Abstract
LC16m8 (m8), a highly attenuated vaccinia virus (VAC) strain, was developed as a smallpox vaccine, and its safety and immunogenicity have been confirmed. Here, we aimed to develop a system that recovers infectious m8 from a bacterial artificial chromosome (BAC) that retains the full-length viral genomic DNA (m8-BAC system). The infectious virus was successfully recovered from a VAC-BAC plasmid, named pLC16m8-BAC. Furthermore, the bacterial replicon-free virus was generated by intramolecular homologous recombination and was successfully recovered from a modified VAC-BAC plasmid, named pLC16m8.8S-BAC. Also, the growth of the recovered virus was indistinguishable from that of authentic m8. The full genome sequence of the plasmid, which harbors identical inverted terminal repeats (ITR) to that of authentic m8, was determined by long-read next-generation sequencing (NGS). The ITR contains x 18 to 32 of the 70 and x 30 to 45 of 54 base pair tandem repeats, and the number of tandem repeats was different between the ITR left and right. Since the virus recovered from pLC16m8.8S-BAC was expected to retain the identical viral genome to that of m8, including the ITR, a reference-based alignment following a short-read NGS was performed to validate the sequence of the recovered virus. Based on the pattern of coverage depth in the ITR, no remarkable differences were observed between the virus and m8, and the other region was confirmed to be identical as well. In summary, this new system can recover the virus, which is geno- and phenotypically indistinguishable from authentic m8.
Collapse
|
24
|
Okoli A, Okeke MI, Tryland M, Moens U. CRISPR/Cas9-Advancing Orthopoxvirus Genome Editing for Vaccine and Vector Development. Viruses 2018; 10:E50. [PMID: 29361752 PMCID: PMC5795463 DOI: 10.3390/v10010050] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 01/17/2018] [Accepted: 01/21/2018] [Indexed: 12/17/2022] Open
Abstract
The clustered regularly interspaced short palindromic repeat (CRISPR)/associated protein 9 (Cas9) technology is revolutionizing genome editing approaches. Its high efficiency, specificity, versatility, flexibility, simplicity and low cost have made the CRISPR/Cas9 system preferable to other guided site-specific nuclease-based systems such as TALENs (Transcription Activator-like Effector Nucleases) and ZFNs (Zinc Finger Nucleases) in genome editing of viruses. CRISPR/Cas9 is presently being applied in constructing viral mutants, preventing virus infections, eradicating proviral DNA, and inhibiting viral replication in infected cells. The successful adaptation of CRISPR/Cas9 to editing the genome of Vaccinia virus paves the way for its application in editing other vaccine/vector-relevant orthopoxvirus (OPXV) strains. Thus, CRISPR/Cas9 can be used to resolve some of the major hindrances to the development of OPXV-based recombinant vaccines and vectors, including sub-optimal immunogenicity; transgene and genome instability; reversion of attenuation; potential of spread of transgenes to wildtype strains and close contacts, which are important biosafety and risk assessment considerations. In this article, we review the published literature on the application of CRISPR/Cas9 in virus genome editing and discuss the potentials of CRISPR/Cas9 in advancing OPXV-based recombinant vaccines and vectors. We also discuss the application of CRISPR/Cas9 in combating viruses of clinical relevance, the limitations of CRISPR/Cas9 and the current strategies to overcome them.
Collapse
Affiliation(s)
- Arinze Okoli
- Biosafety of Genome Editing Research Group, GenØk-Centre for Biosafety, Siva Innovation Centre, N-9294 Tromsø, Norway.
| | - Malachy I Okeke
- Biosafety of Genome Editing Research Group, GenØk-Centre for Biosafety, Siva Innovation Centre, N-9294 Tromsø, Norway.
| | - Morten Tryland
- Biosafety of Genome Editing Research Group, GenØk-Centre for Biosafety, Siva Innovation Centre, N-9294 Tromsø, Norway.
- Artic Infection Biology, Department of Artic and Marine Biology, The Artic University of Norway, N-9037 Tromsø, Norway.
| | - Ugo Moens
- Molecular Inflammation Research Group, Institute of Medical Biology, The Arctic University of Norway, N-9037 Tromsø, Norway.
| |
Collapse
|
25
|
Exploiting 2A peptides to elicit potent neutralizing antibodies by a multi-subunit herpesvirus glycoprotein complex. J Virol Methods 2017; 251:30-37. [PMID: 28989096 DOI: 10.1016/j.jviromet.2017.10.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 09/22/2017] [Accepted: 10/05/2017] [Indexed: 02/06/2023]
Abstract
Neutralizing antibodies (NAb) interfering with glycoprotein complex-mediated virus entry into host cells are thought to contribute to the protection against herpesvirus infection. However, using herpesvirus glycoprotein complexes as vaccine antigens can be complicated by the necessity of expressing multiple subunits simultaneously to allow efficient complex assembly and formation of conformational NAb epitopes. By using a novel bacterial artificial chromosome (BAC) clone of the clinically deployable Modified Vaccinia Ankara (MVA) vector and exploiting ribosomal skipping mediated by 2A peptides, MVA vectors were generated that expressed self-processing subunits of the human cytomegalovirus (HCMV) pentamer complex (PC) composed of gH, gL, UL128, UL130, and UL131A. These MVA vectors expressed 2A-linked HCMV PC subunits that were efficiently cleaved and transported to the cell surface as protein complexes forming conformational neutralizing epitopes. In addition, vaccination of mice by only two immunizations with these MVA vectors resulted in potent HCMV NAb responses that remained stable over a period of at least six months. This method of eliciting NAb by 2A-linked, self-processing HCMV PC subunits could contribute to develop a HCMV vaccine candidate and may serve as a template to facilitate the development of subunit vaccine strategies against other herpesviruses.
Collapse
|
26
|
Volz A, Sutter G. Modified Vaccinia Virus Ankara: History, Value in Basic Research, and Current Perspectives for Vaccine Development. Adv Virus Res 2016; 97:187-243. [PMID: 28057259 PMCID: PMC7112317 DOI: 10.1016/bs.aivir.2016.07.001] [Citation(s) in RCA: 222] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Safety tested Modified Vaccinia virus Ankara (MVA) is licensed as third-generation vaccine against smallpox and serves as a potent vector system for development of new candidate vaccines against infectious diseases and cancer. Historically, MVA was developed by serial tissue culture passage in primary chicken cells of vaccinia virus strain Ankara, and clinically used to avoid the undesirable side effects of conventional smallpox vaccination. Adapted to growth in avian cells MVA lost the ability to replicate in mammalian hosts and lacks many of the genes orthopoxviruses use to conquer their host (cell) environment. As a biologically well-characterized mutant virus, MVA facilitates fundamental research to elucidate the functions of poxvirus host-interaction factors. As extremely safe viral vectors MVA vaccines have been found immunogenic and protective in various preclinical infection models. Multiple recombinant MVA currently undergo clinical testing for vaccination against human immunodeficiency viruses, Mycobacterium tuberculosis or Plasmodium falciparum. The versatility of the MVA vector vaccine platform is readily demonstrated by the swift development of experimental vaccines for immunization against emerging infections such as the Middle East Respiratory Syndrome. Recent advances include promising results from the clinical testing of recombinant MVA-producing antigens of highly pathogenic avian influenza virus H5N1 or Ebola virus. This review summarizes our current knowledge about MVA as a unique strain of vaccinia virus, and discusses the prospects of exploiting this virus as research tool in poxvirus biology or as safe viral vector vaccine to challenge existing and future bottlenecks in vaccinology.
Collapse
Affiliation(s)
- A Volz
- German Center for Infection Research (DZIF), Institute for Infectious Diseases and Zoonoses, LMU University of Munich, Munich, Germany
| | - G Sutter
- German Center for Infection Research (DZIF), Institute for Infectious Diseases and Zoonoses, LMU University of Munich, Munich, Germany.
| |
Collapse
|
27
|
Stellberger T, Stockmar I, Haase M, Meyer H, Zoeller G, Pavlovic M, Büttner M, Konrad R, Lang H, Tischer K, Kaufer BB, Busch U, Baiker A. Multiplex Real-Time PCR Assay for the Detection and Differentiation of Poxviruses and Poxvirus Vectors. APPLIED BIOSAFETY 2015. [DOI: 10.1177/153567601502000405] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
| | - Iris Stockmar
- Bavarian Health and Food Safety Authority, Oberschleissheim, Bavaria, Germany
| | - Maren Haase
- Bavarian Health and Food Safety Authority, Oberschleissheim, Bavaria, Germany
| | - Hermann Meyer
- Bundeswehr Institute of Microbiology, Munich, Germany
| | | | - Melanie Pavlovic
- Bavarian Health and Food Safety Authority, Oberschleissheim, Bavaria, Germany
| | - Mathias Büttner
- Bavarian Health and Food Safety Authority, Oberschleissheim, Bavaria, Germany
| | - Regina Konrad
- Bavarian Health and Food Safety Authority, Oberschleissheim, Bavaria, Germany
| | - Heike Lang
- Bavarian Health and Food Safety Authority, Oberschleissheim, Bavaria, Germany
| | | | | | - Ulrich Busch
- Bavarian Health and Food Safety Authority, Oberschleissheim, Bavaria, Germany
| | - Armin Baiker
- Bavarian Health and Food Safety Authority, Oberschleissheim, Bavaria, Germany
| |
Collapse
|
28
|
Suenaga T, Kohyama M, Hirayasu K, Arase H. Engineering large viral DNA genomes using the CRISPR-Cas9 system. Microbiol Immunol 2015; 58:513-22. [PMID: 25040500 PMCID: PMC7168497 DOI: 10.1111/1348-0421.12180] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 07/08/2014] [Accepted: 07/14/2014] [Indexed: 12/11/2022]
Abstract
Manipulation of viral genomes is essential for studying viral gene function and utilizing viruses for therapy. Several techniques for viral genome engineering have been developed. Homologous recombination in virus-infected cells has traditionally been used to edit viral genomes; however, the frequency of the expected recombination is quite low. Alternatively, large viral genomes have been edited using a bacterial artificial chromosome (BAC) plasmid system. However, cloning of large viral genomes into BAC plasmids is both laborious and time-consuming. In addition, because it is possible for insertion into the viral genome of drug selection markers or parts of BAC plasmids to affect viral function, artificial genes sometimes need to be removed from edited viruses. Herpes simplex virus (HSV), a common DNA virus with a genome length of 152 kbp, causes labialis, genital herpes and encephalitis. Mutant HSV is a candidate for oncotherapy, in which HSV is used to kill tumor cells. In this study, the clustered regularly interspaced short palindromic repeat-Cas9 system was used to very efficiently engineer HSV without inserting artificial genes into viral genomes. Not only gene-ablated HSV but also gene knock-in HSV were generated using this method. Furthermore, selection with phenotypes of edited genes promotes the isolation efficiencies of expectedly mutated viral clones. Because our method can be applied to other DNA viruses such as Epstein-Barr virus, cytomegaloviruses, vaccinia virus and baculovirus, our system will be useful for studying various types of viruses, including clinical isolates.
Collapse
Affiliation(s)
- Tadahiro Suenaga
- Department of Immunochemistry, Research Institute for Microbial Diseases; Laboratory of Immunochemistry, WPI Immunology Frontier Research Center, Osaka University, 3-1, Yamada-oka, Suita, Osaka, 565-0871
| | | | | | | |
Collapse
|
29
|
Efficiently editing the vaccinia virus genome by using the CRISPR-Cas9 system. J Virol 2015; 89:5176-9. [PMID: 25741005 DOI: 10.1128/jvi.00339-15] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 02/17/2015] [Indexed: 02/06/2023] Open
Abstract
Vaccinia virus (VACV) continues to be used in immunotherapy for the prevention of infectious diseases and treatment of cancer since its use for the eradication of smallpox. However, the current method of editing the VACV genome is not efficient. Here, we demonstrate that the CRISPR-Cas9 system can be used to edit the VACV genome rapidly and efficiently. Additionally, a set of 8,964 computationally designed unique guide RNAs (gRNAs) targeting all VACV genes will be valuable for the study of VACV gene functions.
Collapse
|
30
|
Drug resistance of clinical varicella-zoster virus strains confirmed by recombinant thymidine kinase expression and by targeted resistance mutagenesis of a cloned wild-type isolate. Antimicrob Agents Chemother 2015; 59:2726-34. [PMID: 25712361 DOI: 10.1128/aac.05115-14] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 02/16/2015] [Indexed: 01/02/2023] Open
Abstract
In this study, approaches were developed to examine the phenotypes of nonviable clinical varicella-zoster virus (VZV) strains with amino acid substitutions in the thymidine kinase (TK) (open reading frame 36 [ORF36]) and/or DNA polymerase (Pol) (ORF28) suspected to cause resistance to antivirals. Initially, recombinant TK proteins containing amino acid substitutions described as known or suspected causes of antiviral resistance were analyzed by measuring the TK activity by applying a modified commercial enzyme immunoassay. To examine the effects of these TK and Pol substitutions on the replication of recombinant virus strains, the method of en passant mutagenesis was used. Targeted mutations within ORF36 and/or ORF28 and an autonomously expressed gene of the monomeric red fluorescent protein for plaque identification were introduced into the European wild-type VZV strain HJO. Plaque reduction assays revealed that the amino acid substitutions with unknown functions in TK, Q303stop, N334stop, A163stop, and the deletion of amino acids 7 to 74 aa (Δaa 7 to 74), were associated with resistance against acyclovir (ACV), penciclovir, or brivudine, whereas the L73I substitution and the Pol substitutions T237K and A955T revealed sensitive viral phenotypes. The results were confirmed by quantitative PCR by measuring the viral load under increasing ACV concentrations. In conclusion, analyzing the enzymatic activities of recombinant TK proteins represent a useful tool for evaluating the significance of amino acid substitutions in the antiviral resistance of clinical VZV strains. However, direct testing of replication-competent viruses by the introduction of nonsynonymous mutations in a VZV bacterial artificial chromosome using en passant mutagenesis led to reliable phenotypic characterization results.
Collapse
|
31
|
Tobler K, Fraefel C. Infectious delivery of alphaherpesvirus bacterial artificial chromosomes. Methods Mol Biol 2015; 1227:217-230. [PMID: 25239748 DOI: 10.1007/978-1-4939-1652-8_10] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Bacterial artificial chromosomes (BACs) can accommodate and stably propagate the genomes of large DNA viruses in E. coli. As DNA virus genomes are often per se infectious upon transfection into mammalian cells, their cloning in BACs and easy modification by homologous recombination in bacteria has become an important strategy to investigate the functions of individual virus genes. This chapter describes a strategy to clone the genomes of viruses of the Alphaherpesvirinae subfamily within the family of the Herpesviridae, which is a group of large DNA viruses that can establish both lytic and latent infections in most animal species including humans. The cloning strategy includes the following steps: (1) Construction of a transfer plasmid that contains the BAC backbone with selection and screening markers, and targeting sequences which support homologous recombination between the transfer plasmid and the alphaherpesvirus genome. (2) Introduction of the transfer plasmid sequences into the alphaherpesvirus genome via homologous recombination in mammalian cells. (3) Isolation of recombinant virus genomes containing the BAC backbone sequences from infected mammalian cells and electroporation into E. coli. (4) Preparation of infectious BAC DNA from bacterial cultures and transfection into mammalian cells. (5) Isolation and characterization of progeny virus.
Collapse
Affiliation(s)
- Kurt Tobler
- Vetsuisse Faculty, Institute for Virology, University of Zurich, Winterthurerstrasse 266a, Zurich, CH-8057, Switzerland
| | | |
Collapse
|
32
|
|
33
|
Human cytomegalovirus vaccine based on the envelope gH/gL pentamer complex. PLoS Pathog 2014; 10:e1004524. [PMID: 25412505 PMCID: PMC4239111 DOI: 10.1371/journal.ppat.1004524] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 10/16/2014] [Indexed: 12/22/2022] Open
Abstract
Human Cytomegalovirus (HCMV) utilizes two different pathways for host cell entry. HCMV entry into fibroblasts requires glycoproteins gB and gH/gL, whereas HCMV entry into epithelial and endothelial cells (EC) requires an additional complex composed of gH, gL, UL128, UL130, and UL131A, referred to as the gH/gL-pentamer complex (gH/gL-PC). While there are no established correlates of protection against HCMV, antibodies are thought to be important in controlling infection. Neutralizing antibodies (NAb) that prevent gH/gL-PC mediated entry into EC are candidates to be assessed for in vivo protective function. However, these potent NAb are predominantly directed against conformational epitopes derived from the assembled gH/gL-PC. To address these concerns, we constructed Modified Vaccinia Ankara (MVA) viruses co-expressing all five gH/gL-PC subunits (MVA-gH/gL-PC), subsets of gH/gL-PC subunits (gH/gL or UL128/UL130/UL131A), or the gB subunit from HCMV strain TB40/E. We provide evidence for cell surface expression and assembly of complexes expressing full-length gH or gB, or their secretion when the corresponding transmembrane domains are deleted. Mice or rhesus macaques (RM) were vaccinated three times with MVA recombinants and serum NAb titers that prevented 50% infection of human EC or fibroblasts by HCMV TB40/E were determined. NAb responses induced by MVA-gH/gL-PC blocked HCMV infection of EC with potencies that were two orders of magnitude greater than those induced by MVA expressing gH/gL, UL128-UL131A, or gB. In addition, MVA-gH/gL-PC induced NAb responses that were durable and efficacious to prevent HCMV infection of Hofbauer macrophages, a fetal-derived cell localized within the placenta. NAb were also detectable in saliva of vaccinated RM and reached serum peak levels comparable to NAb titers found in HCMV hyperimmune globulins. This vaccine based on a translational poxvirus platform co-delivers all five HCMV gH/gL-PC subunits to achieve robust humoral responses that neutralize HCMV infection of EC, placental macrophages and fibroblasts, properties of potential value in a prophylactic vaccine. Human cytomegalovirus (HCMV) fetal infection during pregnancy and infection of immunocompromised patients are both clinical problems considered extremely important by the Institute of Medicine. Limited efficacy against primary HCMV infection was found using a subunit vaccine based on glycoprotein B, an important neutralizing antibody determinant blocking HCMV entry into fibroblasts. The HCMV field has been transformed by the discovery that a five-member (pentamer) protein complex is a required factor for epithelial and endothelial cell entry and indispensable for transmission as shown in non-human primates. Targeting HCMV with antibodies specific to the pentamer may interrupt horizontal and vertical transmission. We describe an innovative vaccine strategy to induce serum neutralizing antibodies of impressive magnitude against HCMV in two animal models. Using an attenuated poxvirus vector system, we demonstrate that co-expression of all five pentamer components is significantly more potent to induce serum neutralizing antibodies than subunit subsets of the complex or glycoprotein B, reaching peak levels comparable to HCMV hyperimmune globulin. A vaccine that elicits systemic and mucosal antibody responses that prevents infection of multiple cell types crucial to natural history of HCMV infection could play a role in preventing congenital HCMV infection and control of infection in immunocompromised patients.
Collapse
|
34
|
Stellberger T, Haase M, Guertler P, Stockmar I, Busch U, Baiker A. Characterization of Recombinant Vaccinia Viruses by MLPA Technology. APPLIED BIOSAFETY 2014. [DOI: 10.1177/153567601401900304] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
| | - Maren Haase
- Bavarian Health and Food Safety Authority, Oberschleißheim, Bavaria, Germany
| | - Patrick Guertler
- Bavarian Health and Food Safety Authority, Oberschleißheim, Bavaria, Germany
| | - Iris Stockmar
- Bavarian Health and Food Safety Authority, Oberschleißheim, Bavaria, Germany
| | - Ulrich Busch
- Bavarian Health and Food Safety Authority, Oberschleißheim, Bavaria, Germany
| | - Armin Baiker
- Bavarian Health and Food Safety Authority, Oberschleißheim, Bavaria, Germany
| |
Collapse
|
35
|
Kan S, Jia P, Sun L, Hu N, Li C, Lu H, Tian M, Qi Y, Jin N, Li X. Generation of an attenuated Tiantan vaccinia virus by deletion of the ribonucleotide reductase large subunit. Arch Virol 2014; 159:2223-31. [PMID: 24677065 DOI: 10.1007/s00705-014-2057-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Accepted: 02/28/2014] [Indexed: 10/25/2022]
Abstract
Attenuation of the virulence of vaccinia Tiantan virus (VTT) underlies the strategy adopted for mass vaccination campaigns. This strategy provides advantages of safety and efficacy over traditional vaccines and is aimed at minimization of adverse health effects. In this study, a mutant form of the virus, MVTT was derived from VTT by deletion of the ribonucleotide reductase large subunit (R1) (TI4L). Compared to wild-type parental (VTT) and revertant (VTT-rev) viruses, virulence of the mutant MVTT was reduced by 100-fold based on body weight reduction and by 3,200-fold based on determination of the intracranial 50% lethal infectious dose. However, the immunogenicity of MVTT was equivalent to that of the parental VTT. We also demonstrated that the TI4L gene is not required for efficient replication. These data support the conclusion that MVTT can be used as a replicating virus vector or as a platform for the development of vaccines against infectious diseases and for cancer therapy.
Collapse
Affiliation(s)
- Shifu Kan
- Institute of Military Veterinary Medicine, Academy of Military Medical Sciences of PLA, Jilin, People's Republic of China,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Skinner MA, Laidlaw SM, Eldaghayes I, Kaiser P, Cottingham MG. Fowlpox virus as a recombinant vaccine vector for use in mammals and poultry. Expert Rev Vaccines 2014; 4:63-76. [PMID: 15757474 DOI: 10.1586/14760584.4.1.63] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Live vaccines against fowlpox virus, which causes moderate pathology in poultry and is the type species of the Avipoxvirus genus, were developed in the 1920s. Development of recombinant fowlpox virus vector vaccines began in the 1980s, for use not only in poultry, but also in mammals including humans. In common with other avipoxviruses, such as canarypox virus, fowlpox virus enters mammalian cells and expresses proteins, but replicates abortively. The use of fowlpox virus as a safe vehicle for expression of foreign antigens and host immunomodulators, is being evaluated in numerous clinical trials of vaccines against cancer, malaria, tuberculosis and AIDS, notably in heterologous prime-boost regimens. In this article, technical approaches to, and issues surrounding, the use of fowlpox virus as a recombinant vaccine vector in poultry and mammals are reviewed.
Collapse
Affiliation(s)
- Michael A Skinner
- Institute for Animal Health, Compton, Newbury, Berkshire, RG20 7NN, UK.
| | | | | | | | | |
Collapse
|
37
|
Generation of a complete single-gene knockout bacterial artificial chromosome library of cowpox virus and identification of its essential genes. J Virol 2013; 88:490-502. [PMID: 24155400 DOI: 10.1128/jvi.02385-13] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cowpox virus (CPXV) belongs to the genus Orthopoxvirus in the Poxviridae family. It infects a broad range of vertebrates and can cause zoonotic infections. CPXV has the largest genome among the orthopoxviruses and is therefore considered to have the most complete set of genes of all members of the genus. Since CPXV has also become a model for studying poxvirus genetics and pathogenesis, we created and characterized a complete set of single gene knockout bacterial artificial chromosome (BAC) clones of the CPXV strain Brighton Red. These mutants allow a systematic assessment of the contribution of single CPXV genes to the outcome of virus infection and replication, as well as to the virus host range. A full-length BAC clone of CPXV strain Brighton Red (pBRF) harboring the gene expressing the enhanced green fluorescent protein under the control of a viral late promoter was modified by introducing the mrfp1 gene encoding the monomeric red fluorescent protein driven by a synthetic early vaccinia virus promoter. Based on the modified BAC (pBRFseR), a library of targeted knockout mutants for each single viral open reading frame (ORF) was generated. Reconstitution of infectious virus was successful for 109 of the 183 mutant BAC clones, indicating that the deleted genes are not essential for virus replication. In contrast, 74 ORFs were identified as essential because no virus progeny was obtained upon transfection of the mutant BAC clones and in the presence of a helper virus. More than 70% of all late CPXV genes belonged to this latter group of essential genes.
Collapse
|
38
|
A vaccine based on the rhesus cytomegalovirus UL128 complex induces broadly neutralizing antibodies in rhesus macaques. J Virol 2012; 87:1322-32. [PMID: 23152525 DOI: 10.1128/jvi.01669-12] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Neutralizing antibodies (NAb) are important for interfering with horizontal transmission of human cytomegalovirus (HCMV) leading to primary and congenital HCMV infection. Recent findings have shown that a pentameric virion complex formed by the glycoproteins gH/gL, UL128, UL130, and UL131A (UL128C) is required for HCMV entry into epithelial/endothelial cells (Epi/EC) and is the target of potent NAb in HCMV-seropositive individuals. Using bacterial artificial chromosome technology, we have generated a modified vaccinia Ankara virus (MVA) that stably coexpresses all 5 rhesus CMV (RhCMV) proteins homologous to HCMV UL128C, termed MVA-RhUL128C. Coimmunoprecipitation confirmed the interaction of RhgH with the other 4 RhCMV subunits of the pentameric complex. All 8 RhCMV-naïve rhesus macaques (RM) vaccinated with MVA-RhUL128C developed NAb that blocked infection of monkey kidney epithelial cells (MKE) and rhesus fibroblasts. NAb titers induced by MVA-RhUL128C measured on both cell types at 2 to 6 weeks postvaccination were comparable to levels observed in naturally infected RM. In contrast, MVA expressing a subset of RhUL128C proteins or RhgB glycoprotein only minimally stimulated NAb that inhibited infection of MKE. In addition, following subcutaneous RhCMV challenge at 8 weeks postvaccination, animals vaccinated with MVA-RhUL128C showed reduced plasma viral loads. These results indicate that MVA expressing the RhUL128C induces NAb inhibiting RhCMV entry into both Epi/EC and fibroblasts and limits RhCMV replication in RM. This novel approach is the first step in developing a prophylactic HCMV vaccine designed to interfere with virus entry into major cell types permissive for viral replication, a required property of an effective vaccine.
Collapse
|
39
|
Orubu T, Alharbi NK, Lambe T, Gilbert SC, Cottingham MG. Expression and cellular immunogenicity of a transgenic antigen driven by endogenous poxviral early promoters at their authentic loci in MVA. PLoS One 2012; 7:e40167. [PMID: 22761956 PMCID: PMC3384612 DOI: 10.1371/journal.pone.0040167] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Accepted: 06/02/2012] [Indexed: 01/12/2023] Open
Abstract
CD8(+) T cell responses to vaccinia virus are directed almost exclusively against early gene products. The attenuated strain modified vaccinia virus Ankara (MVA) is under evaluation in clinical trials of new vaccines designed to elicit cellular immune responses against pathogens including Plasmodium spp., M. tuberculosis and HIV-1. All of these recombinant MVAs (rMVA) utilize the well-established method of linking the gene of interest to a cloned poxviral promoter prior to insertion into the viral genome at a suitable locus by homologous recombination in infected cells. Using BAC recombineering, we show that potent early promoters that drive expression of non-functional or non-essential MVA open reading frames (ORFs) can be harnessed for immunogenic expression of recombinant antigen. Precise replacement of the MVA orthologs of C11R, F11L, A44L and B8R with a model antigen positioned to use the same translation initiation codon allowed early transgene expression similar to or slightly greater than that achieved by the commonly-used p7.5 or short synthetic promoters. The frequency of antigen-specific CD8(+) T cells induced in mice by single shot or adenovirus-prime, rMVA-boost vaccination were similarly equal or marginally enhanced using endogenous promoters at their authentic genomic loci compared to the traditional constructs. The enhancement in immunogenicity observed using the C11R or F11L promoters compared with p7.5 was similar to that obtained with the mH5 promoter compared with p7.5. Furthermore, the growth rates of the viruses were unimpaired and the insertions were genetically stable. Insertion of a transgenic ORF in place of a viral ORF by BAC recombineering can thus provide not only a potent promoter, but also, concomitantly, a suitable insertion site, potentially facilitating development of MVA vaccines expressing multiple recombinant antigens.
Collapse
Affiliation(s)
- Toritse Orubu
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - Teresa Lambe
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Sarah C. Gilbert
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | |
Collapse
|
40
|
Schuenadel L, Tischer BK, Nitsche A. Generation and characterization of a Cowpox virus mutant lacking host range factor CP77. Virus Res 2012; 168:23-32. [PMID: 22705200 DOI: 10.1016/j.virusres.2012.06.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Revised: 06/05/2012] [Accepted: 06/05/2012] [Indexed: 11/27/2022]
Abstract
Cowpox virus (CPXV) host range factor CP77 was identified to be required for virus replication in Chinese hamster ovary (CHO) cells, but the underlying molecular mechanism by which CP77 modulates host range has remained unclear. Therefore, a CPXVΔCP77 deletion mutant was constructed by applying bacterial artificial chromosome (BAC) technology. Integrity of BAC-derived viral DNA was confirmed by whole genome sequencing. In vitro growth characteristics of CPXV wild type (WT), BAC-derived vCPXV WT and vCPXVΔCP77 were virtually indistinguishable in HEK293T cells, whereas in CHO-K1 cells replication of virus lacking CP77 was unambiguously attenuated. This block of viral replication was confirmed by lack of late viral protein expression. The replication defect of various Orthopoxviruses lacking CP77 in CHO cells could be restored by recombinant expression of CP77. Thus, for the first time, the described CP77-dependent host range effect in CHO cells was shown in the background of CPXV as well as Camelpox virus. To further characterize the mutant virus, cells of several different species were comparably infected with vCPXV WT and vCPXVΔCP77, respectively. Interestingly, except for CHO-K1 cells, vCPXV WT and vCPXVΔCP77 showed no significant difference in terms of morphology of cytopathic effects, expression of a late transcribed virus-encoded green fluorescent protein and virus reproduction, even in other hamster-derived cells. Additionally, in ovo inoculation with either virus revealed the same red-pock phenotype on chicken egg chorioallantoic membranes. Since the data presented indicate a CP77-dependent host range effect only for CHO cells, we conclude that the protein might mediate additional functions not identified yet. The vCPXVΔCP77 deletion mutant generated can now be applied as a useful tool to investigate the function of the putative host range protein CP77.
Collapse
Affiliation(s)
- Livia Schuenadel
- Robert Koch-Institut, Zentrum für Biologische Sicherheit 1, Nordufer 20, 13353 Berlin, Germany
| | | | | |
Collapse
|
41
|
Viral bacterial artificial chromosomes: generation, mutagenesis, and removal of mini-F sequences. J Biomed Biotechnol 2012; 2012:472537. [PMID: 22496607 PMCID: PMC3303620 DOI: 10.1155/2012/472537] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Revised: 10/21/2011] [Accepted: 10/27/2011] [Indexed: 12/29/2022] Open
Abstract
Maintenance and manipulation of large DNA and RNA virus genomes had presented an obstacle for virological research. BAC vectors provided a solution to both problems as they can harbor large DNA sequences and can efficiently be modified using well-established mutagenesis techniques in Escherichia coli. Numerous DNA virus genomes of herpesvirus and pox virus were cloned into mini-F vectors. In addition, several reverse genetic systems for RNA viruses such as members of Coronaviridae and Flaviviridae could be established based on BAC constructs. Transfection into susceptible eukaryotic cells of virus DNA cloned as a BAC allows reconstitution of recombinant viruses. In this paper, we provide an overview on the strategies that can be used for the generation of virus BAC vectors and also on systems that are currently available for various virus species. Furthermore, we address common mutagenesis techniques that allow modification of BACs from single-nucleotide substitutions to deletion of viral genes or insertion of foreign sequences. Finally, we review the reconstitution of viruses from BAC vectors and the removal of the bacterial sequences from the virus genome during this process.
Collapse
|
42
|
Kan S, Wang Y, Sun L, Jia P, Qi Y, Su J, Liu L, Yang G, Liu L, Wang Z, Wang J, Liu G, Jin N, Li X, Ding Z. Attenuation of vaccinia Tian Tan strain by removal of viral TC7L-TK2L and TA35R genes. PLoS One 2012; 7:e31979. [PMID: 22363781 PMCID: PMC3283712 DOI: 10.1371/journal.pone.0031979] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2011] [Accepted: 01/16/2012] [Indexed: 12/19/2022] Open
Abstract
Vaccinia Tian Tan (VTT) was attenuated by deletion of the TC7L-TK2L and TA35R genes to generate MVTT3. The mutant was generated by replacing the open reading frames by a gene encoding enhanced green fluorescent protein (EGFP) flanked by loxP sites. Viruses expressing EGFP were then screened for and purified by serial plaque formation. In a second step the marker EGFP gene was removed by transfecting cells with a plasmid encoding cre recombinase and selecting for viruses that had lost the EGFP phenotype. The MVTT3 mutant was shown to be avirulent and immunogenic. These results support the conclusion that TC7L-TK2L and TA35R deletion mutants can be used as safe viral vectors or as platform for vaccines.
Collapse
Affiliation(s)
- Shifu Kan
- College of Animal Science and Veterinary Medicine, Jilin University, Jilin, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Golden JW, Hooper JW. The strategic use of novel smallpox vaccines in the post-eradication world. Expert Rev Vaccines 2012; 10:1021-35. [PMID: 21806397 PMCID: PMC9491137 DOI: 10.1586/erv.11.46] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
We still face a threat of orthopoxviruses in the form of biological weapons and emerging zoonoses. Therefore, there is a need to maintain a comprehensive defense strategy to counter the low-probability, high-impact threat of smallpox, as well as the ongoing threat of naturally occurring orthopoxvirus disease. The currently licensed live-virus smallpox vaccine ACAM2000 is effective, but associated with serious and even life-threatening adverse events. The health threat posed by this vaccine, and other previously licensed vaccines, has prevented many first responders, and even many in the military, from receiving a vaccine against smallpox. At the same time, global immunity produced during the smallpox eradication campaign is waning. Here, we review novel subunit/component vaccines and how they might play roles in unconventional strategies to defend against emerging orthopoxvirus diseases throughout the world and against smallpox used as a weapon of mass destruction.
Collapse
Affiliation(s)
- Joseph W Golden
- Department of Molecular Virology, Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | | |
Collapse
|
44
|
Back to BAC: the use of infectious clone technologies for viral mutagenesis. Viruses 2012; 4:211-35. [PMID: 22470833 PMCID: PMC3315213 DOI: 10.3390/v4020211] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Revised: 01/26/2012] [Accepted: 01/30/2012] [Indexed: 12/18/2022] Open
Abstract
Bacterial artificial chromosome (BAC) vectors were first developed to facilitate the propagation and manipulation of large DNA fragments in molecular biology studies for uses such as genome sequencing projects and genetic disease models. To facilitate these studies, methodologies have been developed to introduce specific mutations that can be directly applied to the mutagenesis of infectious clones (icBAC) using BAC technologies. This has resulted in rapid identification of gene function and expression at unprecedented rates. Here we review the major developments in BAC mutagenesis in vitro. This review summarises the technologies used to construct and introduce mutations into herpesvirus icBAC. It also explores developing technologies likely to provide the next leap in understanding these important viruses.
Collapse
|
45
|
Employing Live Microbes for Vaccine Delivery. DEVELOPMENT OF NOVEL VACCINES 2012. [PMCID: PMC7123214 DOI: 10.1007/978-3-7091-0709-6_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/29/2022]
|
46
|
Cottingham MG. Genetic manipulation of poxviruses using bacterial artificial chromosome recombineering. Methods Mol Biol 2012; 890:37-57. [PMID: 22688760 DOI: 10.1007/978-1-61779-876-4_3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Traditional methods for genetic manipulation of poxviruses rely on low-frequency natural recombination in virus-infected cells. Although these powerful systems represent the technical foundation of current knowledge and applications of poxviruses, they require long (≥ 500 bp) flanking sequences for homologous recombination, an efficient viral selection method, and burdensome, time-consuming plaque purification. The beginning of the twenty-first century has seen the application of bacterial artificial chromosome (BAC) technology to poxviruses as an alternative method for their genetic manipulation, following the invention of a long-sought-after method for deriving a BAC clone of vaccinia virus (VAC-BAC) by Arban Domi and Bernard Moss. The key advantages of the BAC system are the ease and versatility of performing genetic manipulation using bacteriophage λ Red recombination (recombineering), which requires only ∼50 bp homology arms that can be easily created by PCR, and which allows seamless mutations lacking any marker gene without having to perform transient-dominant selection. On the other hand, there are disadvantages, including the significant setup time, the risk of contamination of the cloned genome with bacterial insertion sequences, and the nontrivial issue of removal of the BAC cassette from derived viruses. These must be carefully weighed to decide whether the use of BACs will be advantageous for a particular application, making pox-BAC systems likely to complement, rather than supplant, traditional methods in most laboratories.
Collapse
|
47
|
Ricci PS, Schäfer B, Kreil TR, Falkner FG, Holzer GW. Selection of recombinant MVA by rescue of the essential D4R gene. Virol J 2011; 8:529. [PMID: 22152060 PMCID: PMC3293099 DOI: 10.1186/1743-422x-8-529] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Accepted: 12/12/2011] [Indexed: 11/25/2022] Open
Abstract
Modified vaccinia virus Ankara (MVA) has become a promising vaccine vector due to its immunogenicity and its proven safety in humans. As a general approach for stringent and rapid selection of recombinant MVA, we assessed marker rescue of the essential viral D4R gene in an engineered deletion mutant that is fully replication defective in wild-type cells. Recombinant, replicating virus was obtained by re-introduction of the deleted viral gene as a dominant selection marker into the deletion mutant.
Collapse
Affiliation(s)
- Patricia S Ricci
- Baxter BioScience, Biomedical Research Center, Uferstrasse 15, 2304 Orth an der Donau, Austria
| | - Birgit Schäfer
- Baxter BioScience, Biomedical Research Center, Uferstrasse 15, 2304 Orth an der Donau, Austria
| | | | - Falko G Falkner
- Baxter BioScience, Biomedical Research Center, Uferstrasse 15, 2304 Orth an der Donau, Austria
| | - Georg W Holzer
- Baxter BioScience, Biomedical Research Center, Uferstrasse 15, 2304 Orth an der Donau, Austria
| |
Collapse
|
48
|
Zmorzynska A, Suk JE, Biederbick W, Maidhof H, Sasse J, Semenza JC, Hunger I. Unfinished business: efforts to define dual-use research of bioterrorism concern. Biosecur Bioterror 2011; 9:372-8. [PMID: 22060036 DOI: 10.1089/bsp.2011.0021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Biotechnological research poses a special security problem because of the duality between beneficial use and misuse. In order to find a balance between regulating potentially dangerous research and assuring scientific advancement, a number of assessments have tried to define which types of research are especially open to misuse and should therefore be considered dual-use research of special concern requiring rigorous oversight. So far, there has been no common understanding of what such activities are. Here we present a review of 27 assessments focusing on biological dual-use issues published between 1997 and 2008. Dual-use research activities identified by these assessments as being of special concern were compiled and compared. Moreover, from these 27 assessments, the primary research publications explicitly identified as examples of concerning research activities were extracted and analyzed. We extracted a core list of 11 activities of special concern and show that this list does not match with the reasons why primary research publications were identified as being of special concern. Additionally, we note that the 11 activities identified are not easily conducted or replicated, and therefore the likelihood of their being used in a high-tech mass casualty bioterrorism event should be reevaluated.
Collapse
Affiliation(s)
- Anna Zmorzynska
- Research Group for Biological Arms Control, Weizsäcker Centre for Science and Peace Research, University of Hamburg, Hamburg, Germany.
| | | | | | | | | | | | | |
Collapse
|
49
|
Li Y, Wang S, Zhu H, Zheng C. Cloning of the herpes simplex virus type 1 genome as a novel luciferase-tagged infectious bacterial artificial chromosome. Arch Virol 2011; 156:2267-72. [PMID: 21894520 DOI: 10.1007/s00705-011-1094-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Accepted: 08/19/2011] [Indexed: 01/26/2023]
Abstract
Herpes simplex virus type 1 (HSV-1) is a ubiquitous human pathogen of skin and mucous membranes. In the present study, the genome of the HSV-1 F strain was cloned as an infectious bacterial artificial chromosome (BAC) clone without any deletions of the viral genes. Additionally, a firefly luciferase cassette was inserted to generate a novel luciferase-expressing HSV-1 BAC. Importantly, the resulting recombinant HSV-1 BAC Luc behaved indistinguishably from the wild-type virus in Vero cells, and the luciferase activity could be easily quantified in vitro. Thus, this novel HSV-1 BAC system would serve as a powerful tool for gene function profiling.
Collapse
Affiliation(s)
- You Li
- Molecular Virology and Viral Immunology Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuchang, People's Republic of China
| | | | | | | |
Collapse
|
50
|
Roth SJ, Höper D, Beer M, Feineis S, Tischer BK, Osterrieder N. Recovery of infectious virus from full-length cowpox virus (CPXV) DNA cloned as a bacterial artificial chromosome (BAC). Vet Res 2011; 42:3. [PMID: 21314965 PMCID: PMC3031225 DOI: 10.1186/1297-9716-42-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2010] [Accepted: 09/13/2010] [Indexed: 11/16/2022] Open
Abstract
Transmission from pet rats and cats to humans as well as severe infection in felids and other animal species have recently drawn increasing attention to cowpox virus (CPXV). We report the cloning of the entire genome of cowpox virus strain Brighton Red (BR) as a bacterial artificial chromosome (BAC) in Escherichia coli and the recovery of infectious virus from cloned DNA. Generation of a full-length CPXV DNA clone was achieved by first introducing a mini-F vector, which allows maintenance of large circular DNA in E. coli, into the thymidine kinase locus of CPXV by homologous recombination. Circular replication intermediates were then electroporated into E. coli DH10B cells. Upon successful establishment of the infectious BR clone, we modified the full-length clone such that recombination-mediated excision of bacterial sequences can occur upon transfection in eukaryotic cells. This self-excision of the bacterial replicon is made possible by a sequence duplication within mini-F sequences and allows recovery of recombinant virus progeny without remaining marker or vector sequences. The in vitro growth properties of viruses derived from both BAC clones were determined and found to be virtually indistinguishable from those of parental, wild-type BR. Finally, the complete genomic sequence of the infectious clone was determined and the cloned viral genome was shown to be identical to that of the parental virus. In summary, the generated infectious clone will greatly facilitate studies on individual genes and pathogenesis of CPXV. Moreover, the vector potential of CPXV can now be more systematically explored using this newly generated tool.
Collapse
Affiliation(s)
- Swaantje J Roth
- Institut für Virologie, Freie Universität Berlin; Philippstrasse 13, Haus 18; 10115 Berlin, Germany.
| | | | | | | | | | | |
Collapse
|