1
|
Hishiki T, Morita T, Akazawa D, Ohashi H, Park ES, Kataoka M, Mifune J, Shionoya K, Tsuchimoto K, Ojima S, Azam AH, Nakajima S, Kawahara M, Yoshikawa T, Shimojima M, Kiga K, Maeda K, Suzuki T, Ebihara H, Takahashi Y, Watashi K. Identification of IMP Dehydrogenase as a Potential Target for Anti-Mpox Virus Agents. Microbiol Spectr 2023; 11:e0056623. [PMID: 37409948 PMCID: PMC10434032 DOI: 10.1128/spectrum.00566-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 06/11/2023] [Indexed: 07/07/2023] Open
Abstract
Mpox virus (formerly monkeypox virus [MPXV]) is a neglected zoonotic pathogen that caused a worldwide outbreak in May 2022. Given the lack of an established therapy, the development of an anti-MPXV strategy is of vital importance. To identify drug targets for the development of anti-MPXV agents, we screened a chemical library using an MPXV infection cell assay and found that gemcitabine, trifluridine, and mycophenolic acid (MPA) inhibited MPXV propagation. These compounds showed broad-spectrum anti-orthopoxvirus activities and presented lower 90% inhibitory concentrations (0.026 to 0.89 μM) than brincidofovir, an approved anti-smallpox agent. These three compounds have been suggested to target the postentry step to reduce the intracellular production of virions. Knockdown of IMP dehydrogenase (IMPDH), the rate-limiting enzyme of guanosine biosynthesis and a target of MPA, dramatically reduced MPXV DNA production. Moreover, supplementation with guanosine recovered the anti-MPXV effect of MPA, suggesting that IMPDH and its guanosine biosynthetic pathway regulate MPXV replication. By targeting IMPDH, we identified a series of compounds with stronger anti-MPXV activity than MPA. This evidence shows that IMPDH is a potential target for the development of anti-MPXV agents. IMPORTANCE Mpox is a zoonotic disease caused by infection with the mpox virus, and a worldwide outbreak occurred in May 2022. The smallpox vaccine has recently been approved for clinical use against mpox in the United States. Although brincidofovir and tecovirimat are drugs approved for the treatment of smallpox by the U.S. Food and Drug Administration, their efficacy against mpox has not been established. Moreover, these drugs may present negative side effects. Therefore, new anti-mpox virus agents are needed. This study revealed that gemcitabine, trifluridine, and mycophenolic acid inhibited mpox virus propagation and exhibited broad-spectrum anti-orthopoxvirus activities. We also suggested IMP dehydrogenase as a potential target for the development of anti-mpox virus agents. By targeting this molecule, we identified a series of compounds with stronger anti-mpox virus activity than mycophenolic acid.
Collapse
Affiliation(s)
- Takayuki Hishiki
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan
| | - Takeshi Morita
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan
| | - Daisuke Akazawa
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan
| | - Hirofumi Ohashi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan
| | - Eun-Sil Park
- Department of Veterinary Science, National Institute of Infectious Diseases, Tokyo, Japan
| | - Michiyo Kataoka
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Junki Mifune
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan
| | - Kaho Shionoya
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
- Department of Applied Biological Science, Tokyo University of Science, Noda, Japan
| | - Kana Tsuchimoto
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan
| | - Shinjiro Ojima
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan
| | - Aa Haeruman Azam
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan
| | - Shogo Nakajima
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
| | - Madoka Kawahara
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Tomoki Yoshikawa
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Masayuki Shimojima
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Kotaro Kiga
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan
| | - Ken Maeda
- Department of Veterinary Science, National Institute of Infectious Diseases, Tokyo, Japan
| | - Tadaki Suzuki
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Hideki Ebihara
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yoshimasa Takahashi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan
| | - Koichi Watashi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
- Department of Applied Biological Science, Tokyo University of Science, Noda, Japan
- MIRAI, Japan Science and Technology Agency (JST), Saitama, Japan
| |
Collapse
|
2
|
A recombinant capripoxvirus expressing the F protein of peste des petits ruminants virus and the P12A3C of foot-and-mouth disease virus. BMC Vet Res 2023; 19:18. [PMID: 36670401 PMCID: PMC9863095 DOI: 10.1186/s12917-022-03529-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 11/24/2022] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Peste des petits ruminants (PPR), foot-and-mouth disease (FMD) and sheep pox and goat pox are three important infectious diseases that infect goats, sheep and other small ruminants. It is well-known that the prevention of three diseases rely mainly on their individual vaccines. However, the vaccines have a variety of different disadvantages, such as short duration of immunity, increasing the number of vaccinations, and poor thermal stability. The purpose of this study is to construct a recombinant goat pox virus (rGPV) capable of expressing the F gene of PPRV and the P12A3C gene of FMDV as a live vector vaccine. RESULTS The IRES, FMDV P12A3C and PPRV F genes into the multi-cloning site of the universal transfer plasmid pTKfpgigp to construct a recombinant transfer plasmid pTKfpgigpFiP12A3C, and transfected GPV-infected lamb testis (LT) cells with liposomes and produced by homologous recombination Recombinant GPV (rGPV/PPRVF-FMDVP12A3C, rGPV). The rGPV was screened and purified by green florescence protein (GFP) and xanthine-guanine-phosphoribosyltransferase gene (gpt) of Escherichia coli as selective markers, and the expression of rGPV in LT cells was detected by RT-PCR and immunofluorescence techniques. The results showed that the virus strain rGPV/PPRVF-FMDVP12A3C containing FMDV P12A3C and PPRV F genes was obtained. The exogenous genes FMDV P12A3C and PPRV F contained in rGPV were normally transcribed and translated in LT cells, and the expression products could specifically react with PPRV and FMDV antiserum. Then, the rGPV was intradermally inoculated with goats, the animal experiments showed that rGPV/PPRVF-FMDVP12A3C could induce high levels of specific antibodies against GPV, PPRV and FMDV. CONCLUSIONS The constructed rGPV induced high levels of specific antibodies against GPV, PPRV and FMDV. The study provides a reference for " one vaccine with multiple uses " of GPV live vector vaccine.
Collapse
|
3
|
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
|
4
|
Iyer RF, Edwards DM, Kolb P, Raué HP, Nelson CA, Epperson ML, Slifka MK, Nolz JC, Hengel H, Fremont DH, Früh K. The secreted protein Cowpox Virus 14 contributes to viral virulence and immune evasion by engaging Fc-gamma-receptors. PLoS Pathog 2022; 18:e1010783. [PMID: 36121874 PMCID: PMC9521928 DOI: 10.1371/journal.ppat.1010783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 09/29/2022] [Accepted: 07/29/2022] [Indexed: 11/19/2022] Open
Abstract
The genome of cowpoxvirus (CPXV) could be considered prototypical for orthopoxviridae (OXPV) since it contains many open reading frames (ORFs) absent or lost in other OPXV, including vaccinia virus (VACV). These additional ORFs are non-essential for growth in vitro but are expected to contribute to the broad host range, virulence and immune evasion characteristics of CPXV. For instance, unlike VACV, CPXV encodes proteins that interfere with T cell stimulation, either directly or by preventing antigen presentation or co-stimulation. When studying the priming of naïve T cells, we discovered that CPXV, but not VACV, encodes a secreted factor that interferes with activation and proliferation of naïve CD8+ and CD4+ T cells, respectively, in response to anti-CD3 antibodies, but not to other stimuli. Deletion mapping revealed that the inhibitory protein is encoded by CPXV14, a small secreted glycoprotein belonging to the poxvirus immune evasion (PIE) family and containing a smallpoxvirus encoded chemokine receptor (SECRET) domain that mediates binding to chemokines. We demonstrate that CPXV14 inhibition of antibody-mediated T cell activation depends on the presence of Fc-gamma receptors (FcγRs) on bystander cells. In vitro, CPXV14 inhibits FcγR-activation by antigen/antibody complexes by binding to FcγRs with high affinity and immobilized CPXV14 can trigger signaling through FcγRs, particularly the inhibitory FcγRIIB. In vivo, CPXV14-deleted virus showed reduced viremia and virulence resulting in reduced weight loss and death compared to wildtype virus whereas both antibody and CD8+ T cell responses were increased in the absence of CPXV14. Furthermore, no impact of CPXV14-deletion on virulence was observed in mice lacking the inhibitory FcγRIIB. Taken together our results suggest that CPXV14 contributes to virulence and immune evasion by binding to host FcγRs.
Collapse
Affiliation(s)
- Ravi F. Iyer
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - David M. Edwards
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Philipp Kolb
- Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hans-Peter Raué
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Chris A. Nelson
- Department of Pathology & Immunology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, United States of America
| | - Megan L. Epperson
- Department of Pathology & Immunology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, United States of America
| | - Mark K. Slifka
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Jeffrey C. Nolz
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Hartmut Hengel
- Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Daved H. Fremont
- Department of Pathology & Immunology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, United States of America
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine in St. Louis, St. Louis, Missouri, United States of America
- Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, United States of America
| | - Klaus Früh
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| |
Collapse
|
5
|
Hood AJM, Sumner RP, Maluquer de Motes C. Disruption of the cGAS/STING axis does not impair sensing of MVA in BHK21 cells. J Gen Virol 2022; 103. [PMID: 35584007 DOI: 10.1099/jgv.0.001755] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Modified vaccinia Ankara (MVA) is an attenuated strain of vaccinia virus (VACV), a dsDNA virus that replicates its genome in the cytoplasm and as a result is canonically sensed by the cyclic GMP-AMP synthase (cGAS) and its downstream stimulator of interferon genes (STING). MVA has a highly restricted host range due to major deletions in its genome including inactivation of immunomodulatory genes, only being able to grow in avian cells and the hamster cell line BHK21. Here we studied the interplay between MVA and the cGAS/STING DNA in this permissive cell line and determined whether manipulation of this axis could impact MVA replication and cell responses. We demonstrate that BHK21 cells retain a functional cGAS/STING axis that responds to canonical DNA sensing agonists, upregulating interferon stimulated genes (ISGs). BHK21 cells also respond to MVA, but with a distinct ISG profile. This profile remains unaltered after CRISPR/Cas9 knock-out editing of STING and ablation of cytosolic DNA responses, indicating that MVA responses are independent of the cGAS/STING axis. Furthermore, infection by MVA diminishes the ability of BHK21 cells to respond to exogenous DNA suggesting that MVA still encodes uncharacterised inhibitors of DNA sensing. This suggests that using attenuated strains in permissive cell lines may assist in identification of novel host-virus interactions that may be of relevance to disease or the therapeutic applications of poxviruses.
Collapse
Affiliation(s)
- Alasdair J M Hood
- Department of Microbial Sciences, University of Surrey, Guildford, UK
| | - Rebecca P Sumner
- Department of Microbial Sciences, University of Surrey, Guildford, UK
| | | |
Collapse
|
6
|
Comparison of Recombinant MVA Selection Methods Based on F13L, D4R and K1L Genes. Viruses 2022; 14:v14030528. [PMID: 35336935 PMCID: PMC8954814 DOI: 10.3390/v14030528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/13/2022] [Accepted: 03/01/2022] [Indexed: 02/04/2023] Open
Abstract
Modified vaccinia Ankara (MVA) is a promising vaccine vector due to its highly attenuated phenotype and good immunogenicity. However, obtaining a new recombinant MVA remains a tedious and laborious procedure involving many rounds of plaque purification. Recombinant MVA generation can be greatly improved and facilitated by different selection techniques. Here, we describe a comparison between techniques based on K1L, F13L and D4R genes.
Collapse
|
7
|
Bréard E, Turpaud M, Beaud G, Postic L, Fablet A, Beer M, Sailleau C, Caignard G, Viarouge C, Hoffmann B, Vitour D, Zientara S. Development and Validation of an ELISA for the Detection of Bluetongue Virus Serotype 4-Specific Antibodies. Viruses 2021; 13:v13091741. [PMID: 34578322 PMCID: PMC8473233 DOI: 10.3390/v13091741] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/21/2021] [Accepted: 08/24/2021] [Indexed: 12/14/2022] Open
Abstract
In this article, we describe the development and evaluation of a double antigen sandwich enzyme-linked immunosorbent assay (ELISA) able to detect serotype 4-specific antibodies from BTV-4 infected or vaccinated animals using a recombinant BTV-4 VP2 protein. The coding sequence of VP2 was inserted into a pVote plasmid by recombination in the Gateway® cloning system. Vaccinia virus (VacV) was used as a vector for the expression of the recombinant VP2. After production in BSR cells, recombinant VP2 was purified by immunoprecipitation using a FLAG tag and then used both as the coated ELISA antigen and as the HRP-tagged conjugate. The performance of the ELISA was evaluated with 1186 samples collected from BTV negative, infected or vaccinated animals. The specificity and sensitivity of the BTV-4 ELISA were above the expected standards for the detection of anti-BTV-4 VP2 antibodies in animals reared in Europe or in the Mediterranean basin. Cross-reactions were observed with reference sera for serotypes 10 and 20, and to a lesser extent with serotypes 12, 17 and 24, due to their genetic proximity to serotype 4. Nevertheless, these serotypes have never been detected in Europe and the Mediterranean area. This ELISA, which requires only the production of a recombinant protein, can be used to detect BTV serotype 4-specific antibodies and is therefore an attractive alternative diagnostic method to serum neutralization.
Collapse
Affiliation(s)
- Emmanuel Bréard
- UMR 1161 Virologie, Laboratory for Animal Health, INRAE, Department of Animal Health, Ecole Nationale Vétérinaire d’Alfort, ANSES, Université Paris-Est, 94700 Maisons-Alfort, France; (M.T.); (G.B.); (L.P.); (A.F.); (C.S.); (G.C.); (C.V.); (D.V.); (S.Z.)
- Correspondence:
| | - Mathilde Turpaud
- UMR 1161 Virologie, Laboratory for Animal Health, INRAE, Department of Animal Health, Ecole Nationale Vétérinaire d’Alfort, ANSES, Université Paris-Est, 94700 Maisons-Alfort, France; (M.T.); (G.B.); (L.P.); (A.F.); (C.S.); (G.C.); (C.V.); (D.V.); (S.Z.)
| | - Georges Beaud
- UMR 1161 Virologie, Laboratory for Animal Health, INRAE, Department of Animal Health, Ecole Nationale Vétérinaire d’Alfort, ANSES, Université Paris-Est, 94700 Maisons-Alfort, France; (M.T.); (G.B.); (L.P.); (A.F.); (C.S.); (G.C.); (C.V.); (D.V.); (S.Z.)
| | - Lydie Postic
- UMR 1161 Virologie, Laboratory for Animal Health, INRAE, Department of Animal Health, Ecole Nationale Vétérinaire d’Alfort, ANSES, Université Paris-Est, 94700 Maisons-Alfort, France; (M.T.); (G.B.); (L.P.); (A.F.); (C.S.); (G.C.); (C.V.); (D.V.); (S.Z.)
| | - Aurore Fablet
- UMR 1161 Virologie, Laboratory for Animal Health, INRAE, Department of Animal Health, Ecole Nationale Vétérinaire d’Alfort, ANSES, Université Paris-Est, 94700 Maisons-Alfort, France; (M.T.); (G.B.); (L.P.); (A.F.); (C.S.); (G.C.); (C.V.); (D.V.); (S.Z.)
| | - Martin Beer
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany; (M.B.); (B.H.)
| | - Corinne Sailleau
- UMR 1161 Virologie, Laboratory for Animal Health, INRAE, Department of Animal Health, Ecole Nationale Vétérinaire d’Alfort, ANSES, Université Paris-Est, 94700 Maisons-Alfort, France; (M.T.); (G.B.); (L.P.); (A.F.); (C.S.); (G.C.); (C.V.); (D.V.); (S.Z.)
| | - Grégory Caignard
- UMR 1161 Virologie, Laboratory for Animal Health, INRAE, Department of Animal Health, Ecole Nationale Vétérinaire d’Alfort, ANSES, Université Paris-Est, 94700 Maisons-Alfort, France; (M.T.); (G.B.); (L.P.); (A.F.); (C.S.); (G.C.); (C.V.); (D.V.); (S.Z.)
| | - Cyril Viarouge
- UMR 1161 Virologie, Laboratory for Animal Health, INRAE, Department of Animal Health, Ecole Nationale Vétérinaire d’Alfort, ANSES, Université Paris-Est, 94700 Maisons-Alfort, France; (M.T.); (G.B.); (L.P.); (A.F.); (C.S.); (G.C.); (C.V.); (D.V.); (S.Z.)
| | - Bernd Hoffmann
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany; (M.B.); (B.H.)
| | - Damien Vitour
- UMR 1161 Virologie, Laboratory for Animal Health, INRAE, Department of Animal Health, Ecole Nationale Vétérinaire d’Alfort, ANSES, Université Paris-Est, 94700 Maisons-Alfort, France; (M.T.); (G.B.); (L.P.); (A.F.); (C.S.); (G.C.); (C.V.); (D.V.); (S.Z.)
| | - Stéphan Zientara
- UMR 1161 Virologie, Laboratory for Animal Health, INRAE, Department of Animal Health, Ecole Nationale Vétérinaire d’Alfort, ANSES, Université Paris-Est, 94700 Maisons-Alfort, France; (M.T.); (G.B.); (L.P.); (A.F.); (C.S.); (G.C.); (C.V.); (D.V.); (S.Z.)
| |
Collapse
|
8
|
Chervyakova O, Tailakova E, Kozhabergenov N, Sadikaliyeva S, Sultankulova K, Zakarya K, Maksyutov RA, Strochkov V, Sandybayev N. Engineering of Recombinant Sheep Pox Viruses Expressing Foreign Antigens. Microorganisms 2021; 9:microorganisms9051005. [PMID: 34067124 PMCID: PMC8150597 DOI: 10.3390/microorganisms9051005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/29/2021] [Accepted: 05/05/2021] [Indexed: 01/15/2023] Open
Abstract
Capripoxviruses with a host range limited to ruminants have the great potential to be used as vaccine vectors. The aim of this work was to evaluate attenuated sheep pox virus (SPPV) vaccine strain NISKHI as a vector expressing several genes. Open reading frames SPPV020 (ribonucleotide kinase) and SPPV066 (thymidine kinase) were selected as sites for the insertion of foreign genes. Two integration plasmids with expression cassette were designed and constructed. Recombinant SPPVs expressing an enhanced green fluorescent protein (EGFP) (rSPPV(RRΔ)EGFP and rSPPV(TKΔ)EGFP), Foot-and-mouth disease virus capsid protein (VP1), and Brucella spp. outer membrane protein 25 (OMP25) (rSPPV(RRΔ)VP1A-(TKΔ)OMP25) were generated under the transient dominant selection method. The insertion of foreign genes into the SPPV020 and SPPV066 open reading frames did not influence the replication of the recombinant viruses in the cells. Successful foreign gene expression in vitro was assessed by luminescent microscopy (EGFP) and Western blot (VP1 and OMP25). Our results have shown that foreign genes were expressed by rSPPV both in permissive (lamb testicles) and non-permissive (bovine kidney, saiga kidney, porcine kidney) cells. Mice immunized with rSPPV(RRΔ)VP1A-(TKΔ)OMP25 elicited specific antibodies to both SPPV and foreign genes VP1 and OMP25. Thus, SPPV NISKHI may be used as a potential safe immunogenic viral vector for the development of polyvalent vaccines.
Collapse
Affiliation(s)
- Olga Chervyakova
- Research Institute for Biological Safety Problems, RK ME&S–Science Committee, Gvardeiskiy 080409, Kazakhstan; (E.T.); (N.K.); (S.S.); (K.S.); (K.Z.); (V.S.); (N.S.)
- Correspondence: ; Tel.: +7-72636-7-22-28
| | - Elmira Tailakova
- Research Institute for Biological Safety Problems, RK ME&S–Science Committee, Gvardeiskiy 080409, Kazakhstan; (E.T.); (N.K.); (S.S.); (K.S.); (K.Z.); (V.S.); (N.S.)
| | - Nurlan Kozhabergenov
- Research Institute for Biological Safety Problems, RK ME&S–Science Committee, Gvardeiskiy 080409, Kazakhstan; (E.T.); (N.K.); (S.S.); (K.S.); (K.Z.); (V.S.); (N.S.)
| | - Sandugash Sadikaliyeva
- Research Institute for Biological Safety Problems, RK ME&S–Science Committee, Gvardeiskiy 080409, Kazakhstan; (E.T.); (N.K.); (S.S.); (K.S.); (K.Z.); (V.S.); (N.S.)
| | - Kulyaisan Sultankulova
- Research Institute for Biological Safety Problems, RK ME&S–Science Committee, Gvardeiskiy 080409, Kazakhstan; (E.T.); (N.K.); (S.S.); (K.S.); (K.Z.); (V.S.); (N.S.)
| | - Kunsulu Zakarya
- Research Institute for Biological Safety Problems, RK ME&S–Science Committee, Gvardeiskiy 080409, Kazakhstan; (E.T.); (N.K.); (S.S.); (K.S.); (K.Z.); (V.S.); (N.S.)
| | - Rinat A. Maksyutov
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, 630559 Novosibirsk Region, Russia;
| | - Vitaliy Strochkov
- Research Institute for Biological Safety Problems, RK ME&S–Science Committee, Gvardeiskiy 080409, Kazakhstan; (E.T.); (N.K.); (S.S.); (K.S.); (K.Z.); (V.S.); (N.S.)
| | - Nurlan Sandybayev
- Research Institute for Biological Safety Problems, RK ME&S–Science Committee, Gvardeiskiy 080409, Kazakhstan; (E.T.); (N.K.); (S.S.); (K.S.); (K.Z.); (V.S.); (N.S.)
| |
Collapse
|
9
|
Aggio JB, Krmeská V, Ferguson BJ, Wowk PF, Rothfuchs AG. Vaccinia Virus Infection Inhibits Skin Dendritic Cell Migration to the Draining Lymph Node. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 206:776-784. [PMID: 33419767 PMCID: PMC7851745 DOI: 10.4049/jimmunol.2000928] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 12/08/2020] [Indexed: 12/17/2022]
Abstract
There is a paucity of information on dendritic cell (DC) responses to vaccinia virus (VACV), including the traffic of DCs to the draining lymph node (dLN). In this study, using a mouse model of infection, we studied skin DC migration in response to VACV and compared it with the tuberculosis vaccine Mycobacterium bovis bacille Calmette-Guérin (BCG), another live attenuated vaccine administered via the skin. In stark contrast to BCG, skin DCs did not relocate to the dLN in response to VACV. Infection with UV-inactivated VACV or modified VACV Ankara promoted DC movement to the dLN, indicating that interference with skin DC migration requires replication-competent VACV. This suppressive effect of VACV was capable of mitigating responses to a secondary challenge with BCG in the skin, ablating DC migration, reducing BCG transport, and delaying CD4+ T cell priming in the dLN. Expression of inflammatory mediators associated with BCG-triggered DC migration were absent from virus-injected skin, suggesting that other pathways invoke DC movement in response to replication-deficient VACV. Despite adamant suppression of DC migration, VACV was still detected early in the dLN and primed Ag-specific CD4+ T cells. In summary, VACV blocks skin DC mobilization from the site of infection while retaining the ability to access the dLN to prime CD4+ T cells.
Collapse
Affiliation(s)
- Juliana Bernardi Aggio
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
- Instituto Carlos Chagas, FIOCRUZ, Curitiba PR 81310-020, Brazil; and
| | - Veronika Krmeská
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Brian J Ferguson
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, United Kingdom
| | - Pryscilla Fanini Wowk
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
- Instituto Carlos Chagas, FIOCRUZ, Curitiba PR 81310-020, Brazil; and
| | | |
Collapse
|
10
|
Capripoxvirus vectors for vaccine development. GENE REPORTS 2020. [DOI: 10.1016/j.genrep.2020.100890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
11
|
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: 1.5] [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
|
12
|
Zhao Y, Han Z, Zhang X, Zhang X, Sun J, Ma D, Liu S. Construction and immune protection evaluation of recombinant virus expressing Newcastle disease virus F protein by the largest intergenic region of fowlpox virus NX10. Virus Genes 2020; 56:734-748. [PMID: 33009986 DOI: 10.1007/s11262-020-01799-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 09/26/2020] [Indexed: 01/27/2023]
Abstract
Fowlpox virus (FPV) is used as a vaccine vector to prevent diseases in poultry and mammals. The insertion site is considered as one of the main factors influencing foreign gene expression. Therefore, the identification of insertion sites that can stably and efficiently express foreign genes is crucial for the construction of recombinant vaccines. In this study, we found that the insertion of foreign genes into ORF054 and the ORF161/ORF162 intergenic region of the FPV genome did not affect replication, and that the foreign genes inserted into the intergenic region were more efficiently expressed than when they were inserted into a gene. Based on these results, the recombinant virus rFPVNX10-NDV F-E was constructed and immune protection against virulent FPV and Newcastle disease virus (NDV) was evaluated. Tests for anti-FPV antibodies in the vaccinated chickens were positive within 14 days post-vaccination. After challenge with FPV102, no clinical signs of FP were observed in vaccinated chickens, as compared to that in the control group (unvaccinated), which showed 100% morbidity. Low levels of NDV-specific neutralizing antibodies were detected in vaccinated chickens before challenge. After challenge with NDV ck/CH/LHLJ/01/06, all control chickens died within 4 days post-challenge, whereas 5/15 vaccinated chickens died between 4 and 12 days post-challenge. Vaccination provided an immune protection rate of 66.7%, whereas the control group showed 100% mortality. These results indicate that the ORF161/ORF162 intergenic region of FPVNX10 can be used as a recombination site for foreign gene expression in vivo and in vitro.
Collapse
Affiliation(s)
- Yan Zhao
- College of Animal Science and Technology, Northeast Agricultural University, No. 600 Changjiang Road, Xiangfang District, Harbin, 150030, China.,Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Xiangfang District, Harbin, 150069, China
| | - Zongxi Han
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Xiangfang District, Harbin, 150069, China
| | - Xiaocai Zhang
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Xiangfang District, Harbin, 150069, China
| | - Xuemei Zhang
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Xiangfang District, Harbin, 150069, China
| | - Junfeng Sun
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Xiangfang District, Harbin, 150069, China
| | - Deying Ma
- College of Animal Science and Technology, Northeast Agricultural University, No. 600 Changjiang Road, Xiangfang District, Harbin, 150030, China.
| | - Shengwang Liu
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Xiangfang District, Harbin, 150069, China.
| |
Collapse
|
13
|
Monticelli SR, Bryk P, Ward BM. The Molluscum Contagiosum Gene MC021L Partially Compensates for the Loss of Its Vaccinia Virus Homolog, F13L. J Virol 2020; 94:e01496-20. [PMID: 32727873 PMCID: PMC7527044 DOI: 10.1128/jvi.01496-20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 07/23/2020] [Indexed: 11/20/2022] Open
Abstract
Orthopoxviruses produce two antigenically distinct infectious enveloped virions termed intracellular mature virions and extracellular virions (EV). EV have an additional membrane compared to intracellular mature virions due to a wrapping process at the trans-Golgi network and are required for cell-to-cell spread and pathogenesis. Specific to the EV membrane are a number of proteins highly conserved among orthopoxviruses, including F13, which is required for the efficient wrapping of intracellular mature virions to produce EV and which plays a role in EV entry. The distantly related molluscipoxvirus, molluscum contagiosum virus, is predicted to encode several vaccinia virus homologs of EV-specific proteins, including the homolog of F13L, MC021L. To study the function of MC021, we replaced the F13L open reading frame in vaccinia virus with an epitope-tagged version of MC021L. The resulting virus (vMC021L-HA) had a small-plaque phenotype compared to vF13L-HA but larger than vΔF13L. The localization of MC021-HA was markedly different from that of F13-HA in infected cells, but MC021-HA was still incorporated in the EV membrane. Similar to F13-HA, MC021-HA was capable of interacting with both A33 and B5. Although MC021-HA expression did not fully restore plaque size, vMC021L-HA produced amounts of EV similar to those produced by vF13L-HA, suggesting that MC021 retained some of the functionality of F13. Further analysis revealed that EV produced from vMC021L-HA exhibit a marked reduction in target cell binding and an increase in dissolution, both of which correlated with a small-plaque phenotype.IMPORTANCE The vaccinia virus extracellular virion protein F13 is required for the production and release of infectious extracellular virus, which in turn is essential for the subsequent spread and pathogenesis of orthopoxviruses. Molluscum contagiosum virus infects millions of people worldwide each year, but it is unknown whether EV are produced during infection for spread. Molluscum contagiosum virus contains a homolog of F13L termed MC021L. To study the potential function of this homolog during infection, we utilized vaccinia virus as a surrogate and showed that a vaccinia virus expressing MC021L-HA in place of F13L-HA exhibits a small-plaque phenotype but produces similar levels of EV. These results suggest that MC021-HA can compensate for the loss of F13-HA by facilitating wrapping to produce EV and further delineates the dual role of F13 during infection.
Collapse
Affiliation(s)
- Stephanie R Monticelli
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Peter Bryk
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Brian M Ward
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
14
|
O’Connell CM, Jasperse B, Hagen CJ, Titong A, Verardi PH. Replication-inducible vaccinia virus vectors with enhanced safety in vivo. PLoS One 2020; 15:e0230711. [PMID: 32240193 PMCID: PMC7117657 DOI: 10.1371/journal.pone.0230711] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 03/06/2020] [Indexed: 11/18/2022] Open
Abstract
Vaccinia virus (VACV) has been used extensively as the vaccine against smallpox and as a viral vector for the development of recombinant vaccines and cancer therapies. Replication-competent, non-attenuated VACVs induce strong, long-lived humoral and cell-mediated immune responses and can be effective oncolytic vectors. However, complications from uncontrolled VACV replication in vaccinees and their close contacts can be severe, particularly in individuals with predisposing conditions. In an effort to develop replication-competent VACV vectors with improved safety, we placed VACV late genes encoding core or virion morphogenesis proteins under the control of tet operon elements to regulate their expression with tetracycline antibiotics. These replication-inducible VACVs would only express the selected genes in the presence of tetracyclines. VACVs inducibly expressing the A3L or A6L genes replicated indistinguishably from wild-type VACV in the presence of tetracyclines, whereas there was no evidence of replication in the absence of antibiotics. These outcomes were reflected in mice, where the VACV inducibly expressing the A6L gene caused weight loss and mortality equivalent to wild-type VACV in the presence of tetracyclines. In the absence of tetracyclines, mice were protected from weight loss and mortality, and viral replication was not detected. These findings indicate that replication-inducible VACVs based on the conditional expression of the A3L or A6L genes can be used for the development of safer, next-generation live VACV vectors and vaccines. The design allows for administration of replication-inducible VACV in the absence of tetracyclines (as a replication-defective vector) or in the presence of tetracyclines (as a replication-competent vector) with enhanced safety.
Collapse
Affiliation(s)
- Caitlin M. O’Connell
- Department of Pathobiology and Veterinary Science and Center of Excellence for Vaccine Research, College of Agriculture, Health and Natural Resources, University of Connecticut, Storrs, Connecticut, United States of America
| | - Brittany Jasperse
- Department of Pathobiology and Veterinary Science and Center of Excellence for Vaccine Research, College of Agriculture, Health and Natural Resources, University of Connecticut, Storrs, Connecticut, United States of America
| | - Caitlin J. Hagen
- Department of Pathobiology and Veterinary Science and Center of Excellence for Vaccine Research, College of Agriculture, Health and Natural Resources, University of Connecticut, Storrs, Connecticut, United States of America
| | - Allison Titong
- Department of Pathobiology and Veterinary Science and Center of Excellence for Vaccine Research, College of Agriculture, Health and Natural Resources, University of Connecticut, Storrs, Connecticut, United States of America
| | - Paulo H. Verardi
- Department of Pathobiology and Veterinary Science and Center of Excellence for Vaccine Research, College of Agriculture, Health and Natural Resources, University of Connecticut, Storrs, Connecticut, United States of America
| |
Collapse
|
15
|
Davola ME, Vito A, Wei J, El-Sayes N, Workenhe S, Mossman KL. Genetic modification of oncolytic viruses to enhance antitumor immunity. Methods Enzymol 2019; 635:231-250. [PMID: 32122548 DOI: 10.1016/bs.mie.2019.05.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Among the many immunotherapies being developed and tested both preclinically and clinically, oncolytic viruses (OVs) are gaining traction as a forerunner in the search for potent new therapeutic agents, with a genetically engineered herpes simplex virus type 1 (HSV-1) recently approved by the FDA for the treatment of melanoma. The great potential of OVs to fight cancer is driving different approaches to improve OV-based therapy, with genetic modification of OVs to enhance host antitumor immunity being one of the most promising approaches. In this chapter we describe possible modifications in the OV genome that could increase its antitumor activity and immunostimulatory capacity, together with different methods to achieve these goals. Finally, we present different analyses to verify the desired genetic modification and evaluate its impact on host antitumor immunity in preliminary stages.
Collapse
Affiliation(s)
- Maria Eugenia Davola
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Michael DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Alyssa Vito
- Department of Biochemistry and Biomedical Science, McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Jiarun Wei
- Department of Biochemistry and Biomedical Science, McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Nader El-Sayes
- Department of Biochemistry and Biomedical Science, McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Samuel Workenhe
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Michael DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Karen Louise Mossman
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Michael DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
16
|
Sun Z, Liu L, Zhang H, Li Y, Wei F, Li Z, Wang P, Fu Q, Ren Y, Zhang Y, Guo Z, Chen C. Expression and functional analysis of Brucella outer membrane protein 25 in recombinant goat pox virus. Mol Med Rep 2019; 19:2323-2329. [PMID: 30664205 DOI: 10.3892/mmr.2019.9868] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 05/22/2017] [Indexed: 11/06/2022] Open
Abstract
The Capripoxvirus (CaPV) has a large double‑stranded DNA genome and a restricted host‑range. At present, it is being investigated as an ideal vaccine vector. In the present study, a novel recombinant goat pox virus (rGTPV) was constructed to express Brucella outer membrane protein (OMP)25, and was validated by in vitro and in vivo immunization assays. A novel rGTPV vector was created, in which the thymidine kinase gene was used as a flanking sequence, I1L was inserted as a promoter element to enhance Brucella OMP25 expression, and p7.5 as another promoter element was used to regulate guanine phosphoribosyl‑transferase as a selection maker. The rGTPV vector was transfected into sheep fetal fibroblast/lamb testis cells pre‑infected with GTPV G14‑STV44‑55 to recombine. Brucella OMP25 protein was expressed in cells by rGTPV, and activated immune reactivity to Brucella OMP25 protein, as detected by western blotting. Furthermore, rGTPV elicited, anti‑Brucella‑specific immunoglobulin G responses, as determined by ELISA. Mice vaccinated with rGTPV did not exhibit pathology alterations in the kidney and liver. These results suggested that the novel rGTPV was able to efficiently drive Brucella OMP25 protein expression and activate immune reactivity, and may have applications in CaPV live vector vaccines and associated research.
Collapse
Affiliation(s)
- Zhihua Sun
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832000, P.R. China
| | - Laizhen Liu
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832000, P.R. China
| | - Hui Zhang
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832000, P.R. China
| | - Youwen Li
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832000, P.R. China
| | - Feng Wei
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832000, P.R. China
| | - Zhiqiang Li
- College of Biology and Food, Shangqiu Normal University, Shangqiu, Henan 476000, P.R. China
| | - Pengyan Wang
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832000, P.R. China
| | - Qiang Fu
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832000, P.R. China
| | - Yan Ren
- College of Medicine, Shihezi University, Shihezi, Xinjiang 832000, P.R. China
| | - Yu Zhang
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832000, P.R. China
| | - Zhiru Guo
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832000, P.R. China
| | - Chuangfu Chen
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832000, P.R. China
| |
Collapse
|
17
|
Conrad SJ, Liu J. Poxviruses as Gene Therapy Vectors: Generating Poxviral Vectors Expressing Therapeutic Transgenes. Methods Mol Biol 2019; 1937:189-209. [PMID: 30706397 DOI: 10.1007/978-1-4939-9065-8_11] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Treatments with poxvirus vectors can have long-lasting immunological impact in the host, and thus they have been extensively studied to treat diseases and for vaccine development. More importantly, the oncolytic properties of poxviruses have led to their development as cancer therapeutics. Two poxviruses, vaccinia virus (VACV) and myxoma virus (MYXV), have been extensively studied as virotherapeutics with promising results. Vaccinia virus vectors have advanced to the clinic and have been tested as oncolytic therapeutics for several cancer types with successes in phase I/II clinical trials. In addition to oncolytic applications, MYXV has been explored for additional applications including immunotherapeutics, purging of cancer progenitor cells, and treatments for graft-versus-host diseases. These novel therapeutic applications have encouraged its advancement into clinical trials. To meet the demands of different treatment needs, VACV and MYXV can be genetically engineered to express therapeutic transgenes. The engineering process used in poxvirus vectors can be very different from that of other DNA virus vectors (e.g., the herpesviruses). This chapter is intended to serve as a guide to those wishing to engineer poxvirus vectors for therapeutic transgene expression and to produce viral preparations for preclinical studies.
Collapse
Affiliation(s)
- Steven J Conrad
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences (UAMS), Little Rock, AR, USA
| | - Jia Liu
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences (UAMS), Little Rock, AR, USA. .,The Center for Microbial Pathogenesis and Host Inflammatory Responses, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| |
Collapse
|
18
|
A virus-encoded type I interferon decoy receptor enables evasion of host immunity through cell-surface binding. Nat Commun 2018; 9:5440. [PMID: 30575728 PMCID: PMC6303335 DOI: 10.1038/s41467-018-07772-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 11/21/2018] [Indexed: 12/12/2022] Open
Abstract
Soluble cytokine decoy receptors are potent immune modulatory reagents with therapeutic applications. Some virus-encoded secreted cytokine receptors interact with glycosaminoglycans expressed at the cell surface, but the biological significance of this activity in vivo is poorly understood. Here, we show the type I interferon binding protein (IFNα/βBP) encoded by vaccinia and ectromelia viruses requires of this cell binding activity to confer full virulence to these viruses and to retain immunomodulatory activity. Expression of a variant form of the IFNα/βBP that inhibits IFN activity, but does not interact with cell surface glycosaminoglycans, results in highly attenuated viruses with a virulence similar to that of the IFNα/βBP deletion mutant viruses. Transcriptomics analysis and infection of IFN receptor-deficient mice confirmed that the control of IFN activity is the main function of the IFNα/βBP in vivo. We propose that retention of secreted cytokine receptors at the cell surface may largely enhance their immunomodulatory activity.
Collapse
|
19
|
Lynn H, Howell LM, Diefenbach RJ, Newsome TP. Phototracking Vaccinia Virus Transport Reveals Dynamics of Cytoplasmic Dispersal and a Requirement for A36R and F12L for Exit from the Site of Wrapping. Viruses 2018; 10:v10080390. [PMID: 30042325 PMCID: PMC6115981 DOI: 10.3390/v10080390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 07/18/2018] [Accepted: 07/18/2018] [Indexed: 11/16/2022] Open
Abstract
The microtubule cytoskeleton is a primary organizer of viral infections for delivering virus particles to their sites of replication, establishing and maintaining subcellular compartments where distinct steps of viral morphogenesis take place, and ultimately dispersing viral progeny. One of the best characterized examples of virus motility is the anterograde transport of the wrapped virus form of vaccinia virus (VACV) from the trans-Golgi network (TGN) to the cell periphery by kinesin-1. Yet many aspects of this transport event are elusive due to the speed of motility and the challenges of imaging this stage at high resolution over extended time periods. We have established a novel imaging technology to track virus transport that uses photoconvertible fluorescent recombinant viruses to track subsets of virus particles from their site of origin and determine their destination. Here we image virus exit from the TGN and their rate of egress to the cell periphery. We demonstrate a role for kinesin-1 engagement in regulating virus exit from the TGN by removing A36 and F12 function, critical viral mediators of kinesin-1 recruitment to virus particles. Phototracking viral particles and components during infection is a powerful new imaging approach to elucidate mechanisms of virus replication.
Collapse
Affiliation(s)
- Helena Lynn
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia.
| | - Liam M Howell
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia.
| | - Russell J Diefenbach
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia.
| | - Timothy P Newsome
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
20
|
Barbieri A, Panigada M, Soprana E, Di Mario G, Gubinelli F, Bernasconi V, Recagni M, Donatelli I, Castrucci MR, Siccardi AG. Strategies to obtain multiple recombinant modified vaccinia Ankara vectors. Applications to influenza vaccines. J Virol Methods 2017; 251:7-14. [PMID: 28987424 DOI: 10.1016/j.jviromet.2017.10.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 10/02/2017] [Accepted: 10/02/2017] [Indexed: 11/27/2022]
Abstract
As a vaccination vector, MVA has been widely investigated both in animal models and humans. The construction of recombinant MVA (rMVA) relies on homologous recombination between an acceptor virus and a donor plasmid in infected/transfected permissive cells. Our construction strategy "Red-to-Green gene swapping" - based on the exchange of two fluorescent markers within the flanking regions of MVA deletion ΔIII, coupled to fluorescence activated cell sorting - is here extended to a second insertion site, within the flanking regions of MVA deletion ΔVI. Exploiting this strategy, both double and triple rMVA were constructed, expressing as transgenes the influenza A proteins HA, NP, M1, and PB1. Upon validation of the harbored transgenes co-expression, double and triple recombinants rMVA(ΔIII)-NP-P2A-M1 and rMVA(ΔIII)-NP-P2A-M1-(ΔVI)-PB1 were assayed for in vivo immunogenicity and protection against lethal challenge. In vivo responses were identical to those obtained with the reported combinations of single recombinants, supporting the feasibility and reliability of the present improvement and the extension of Red-to-Green gene swapping to insertion sites other than ΔIII.
Collapse
Affiliation(s)
- Andrea Barbieri
- Molecular Immunology Unit, San Raffaele Research Institute, Via Olgettina 58, 20132, Milan, Italy; Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Vanvitelli, 32, 20129, Milan, Italy
| | - Maddalena Panigada
- Molecular Immunology Unit, San Raffaele Research Institute, Via Olgettina 58, 20132, Milan, Italy
| | - Elisa Soprana
- Molecular Immunology Unit, San Raffaele Research Institute, Via Olgettina 58, 20132, Milan, Italy
| | - Giuseppina Di Mario
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Francesco Gubinelli
- Molecular Immunology Unit, San Raffaele Research Institute, Via Olgettina 58, 20132, Milan, Italy
| | - Valentina Bernasconi
- Molecular Immunology Unit, San Raffaele Research Institute, Via Olgettina 58, 20132, Milan, Italy
| | - Marta Recagni
- Molecular Immunology Unit, San Raffaele Research Institute, Via Olgettina 58, 20132, Milan, Italy
| | - Isabella Donatelli
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Maria R Castrucci
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Antonio G Siccardi
- Molecular Immunology Unit, San Raffaele Research Institute, Via Olgettina 58, 20132, Milan, Italy.
| |
Collapse
|
21
|
Guo ZS, Liu Z, Sathaiah M, Wang J, Ravindranathan R, Kim E, Huang S, Kenniston TW, Bell JC, Zeh HJ, Butterfield LH, Gambotto A, Bartlett DL. Rapid Generation of Multiple Loci-Engineered Marker-free Poxvirus and Characterization of a Clinical-Grade Oncolytic Vaccinia Virus. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017; 7:112-122. [PMID: 29085848 PMCID: PMC5651493 DOI: 10.1016/j.omtm.2017.09.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 09/27/2017] [Indexed: 12/13/2022]
Abstract
Recombinant poxviruses, utilized as vaccine vectors and oncolytic viruses, often require manipulation at multiple genetic loci in the viral genome. It is essential for viral vectors to possess no adventitious mutations and no (antibiotic) selection marker in the final product for human patients in order to comply with the guidance from the regulatory agencies. Rintoul et al. have previously developed a selectable and excisable marker (SEM) system for the rapid generation of recombinant vaccinia virus. In the current study, we describe an improved methodology for rapid creation and selection of recombinant poxviruses with multiple genetic manipulations solely based on expression of a fluorescent protein and with no requirement for drug selection that can lead to cellular stress and the risk of adventitious mutations throughout the viral genome. Using this improved procedure combined with the SEM system, we have constructed multiple marker-free oncolytic poxviruses expressing different cytokines and other therapeutic genes. The high fidelity of inserted DNA sequences validates the utility of this improved procedure for generation of therapeutic viruses for human patients. We have created an oncolytic poxvirus expressing human chemokine CCL5, designated as vvDD-A34R-hCCL5, with manipulations at two genetic loci in a single virus. Finally, we have produced and purified this virus in clinical grade for its use in a phase I clinical trial and presented data on initial in vitro characterization of the virus.
Collapse
Affiliation(s)
- Zong Sheng Guo
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Corresponding author: Zong Sheng Guo, PhD, UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Zuqiang Liu
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Magesh Sathaiah
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jiahu Wang
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Roshni Ravindranathan
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Eun Kim
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Shaohua Huang
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Thomas W. Kenniston
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - John C. Bell
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Herbert J. Zeh
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Lisa H. Butterfield
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Departments of Medicine and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Andrea Gambotto
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - David L. Bartlett
- UPMC Hillman Cancer Center and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
22
|
Earl PL, Moss B, Wyatt LS. Generation of Recombinant Vaccinia Viruses. CURRENT PROTOCOLS IN PROTEIN SCIENCE 2017; 89:5.13.1-5.13.18. [PMID: 28762491 PMCID: PMC5765993 DOI: 10.1002/cpps.33] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
This unit describes how to infect cells with vaccinia virus and then transfect them with a plasmid-transfer vector or PCR fragment to generate a recombinant virus. Selection and screening methods used to isolate recombinant viruses and a method for the amplification of recombinant viruses are described. Finally, a method for live immunostaining that has been used primarily for detection of recombinant modified vaccinia virus Ankara (MVA) is presented. © 2017 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Patricia L. Earl
- National Institutes of Health, National Institute of Allergy and Infectious Diseases, 33 North Drive, Bethesda, MD 20892-3210
| | - Bernard Moss
- National Institutes of Health, National Institute of Allergy and Infectious Diseases, 33 North Drive, Bethesda, MD 20892-3210
| | - Linda S. Wyatt
- National Institutes of Health, National Institute of Allergy and Infectious Diseases, 33 North Drive, Bethesda, MD 20892-3210
| |
Collapse
|
23
|
Eldi P, Cooper TH, Liu L, Prow NA, Diener KR, Howley PM, Suhrbier A, Hayball JD. Production of a Chikungunya Vaccine Using a CHO Cell and Attenuated Viral-Based Platform Technology. Mol Ther 2017; 25:2332-2344. [PMID: 28720468 PMCID: PMC5628773 DOI: 10.1016/j.ymthe.2017.06.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 06/03/2017] [Accepted: 06/18/2017] [Indexed: 02/06/2023] Open
Abstract
Vaccinia-based systems have been extensively explored for the development of recombinant vaccines. Herein we describe an innovative vaccinia virus (VACV)-derived vaccine platform technology termed Sementis Copenhagen Vector (SCV), which was rendered multiplication-defective by targeted deletion of the essential viral assembly gene D13L. A SCV cell substrate line was developed for SCV vaccine production by engineering CHO cells to express D13 and the VACV host-range factor CP77, because CHO cells are routinely used for manufacture of biologics. To illustrate the utility of the platform technology, a SCV vaccine against chikungunya virus (SCV-CHIK) was developed and shown to be multiplication-defective in a range of human cell lines and in immunocompromised mice. A single vaccination of mice with SCV-CHIK induced antibody responses specific for chikungunya virus (CHIKV) that were similar to those raised following vaccination with a replication-competent VACV-CHIK and able to neutralize CHIKV. Vaccination also provided protection against CHIKV challenge, preventing both viremia and arthritis. Moreover, SCV retained capacity as an effective mouse smallpox vaccine. In summary, SCV represents a new and safe vaccine platform technology that can be manufactured in modified CHO cells, with pre-clinical evaluation illustrating utility for CHIKV vaccine design and construction.
Collapse
Affiliation(s)
- Preethi Eldi
- Experimental Therapeutics Laboratory, Hanson Institute and Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Tamara H Cooper
- Experimental Therapeutics Laboratory, Hanson Institute and Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Liang Liu
- Experimental Therapeutics Laboratory, Hanson Institute and Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Natalie A Prow
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| | - Kerrilyn R Diener
- Experimental Therapeutics Laboratory, Hanson Institute and Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA 5000, Australia; Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Paul M Howley
- Experimental Therapeutics Laboratory, Hanson Institute and Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA 5000, Australia; Sementis Ltd., Melbourne, VIC 3000, Australia.
| | - Andreas Suhrbier
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| | - John D Hayball
- Experimental Therapeutics Laboratory, Hanson Institute and Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA 5000, Australia; Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia.
| |
Collapse
|
24
|
Novel Nonreplicating Vaccinia Virus Vector Enhances Expression of Heterologous Genes and Suppresses Synthesis of Endogenous Viral Proteins. mBio 2017; 8:mBio.00790-17. [PMID: 28588133 PMCID: PMC5461411 DOI: 10.1128/mbio.00790-17] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Viruses are used as expression vectors for protein synthesis, immunology research, vaccines, and therapeutics. Advantages of poxvirus vectors include the accommodation of large amounts of heterologous DNA, the presence of a cytoplasmic site of transcription, and high expression levels. On the other hand, competition of approximately 200 viral genes with the target gene for expression and immune recognition may be disadvantageous. We describe a vaccinia virus (VACV) vector that uses an early promoter to express the bacteriophage T7 RNA polymerase; has the A23R intermediate transcription factor gene deleted, thereby restricting virus replication to complementing cells; and has a heterologous gene regulated by a T7 promoter. In noncomplementing cells, viral early gene expression and DNA replication occurred normally but synthesis of intermediate and late proteins was prevented. Nevertheless, the progeny viral DNA provided templates for abundant expression of heterologous genes regulated by a T7 promoter. Selective expression of the Escherichia coli lac repressor gene from an intermediate promoter reduced transcription of the heterologous gene specifically in complementing cells, where large amounts might adversely impact VACV replication. Expression of heterologous proteins mediated by the A23R deletion vector equaled that of a replicating VACV, was higher than that of a nonreplicating modified vaccinia virus Ankara (MVA) vector used for candidate vaccines in vitro and in vivo, and was similarly immunogenic in mice. Unlike the MVA vector, the A23R deletion vector still expresses numerous early genes that can restrict immunogenicity as demonstrated here by the failure of the prototype vector to induce interferon alpha. By deleting immunomodulatory genes, we anticipate further improvements in the system. Vaccines provide an efficient and effective way of preventing infectious diseases. Nevertheless, new and better vaccines are needed. Vaccinia virus, which was used successfully as a live vaccine to eradicate smallpox, has been further attenuated and adapted as a recombinant vector for immunization against other pathogens. However, since the initial description of this vector system, only incremental improvements largely related to safety have been implemented. Here we described novel modifications of the platform that increased expression of the heterologous target gene and decreased expression of endogenous vaccinia virus genes while providing safety by preventing replication of the candidate vaccine except in complementing cells used for vector propagation.
Collapse
|
25
|
Teferi WM, Desaulniers MA, Noyce RS, Shenouda M, Umer B, Evans DH. The vaccinia virus K7 protein promotes histone methylation associated with heterochromatin formation. PLoS One 2017; 12:e0173056. [PMID: 28257484 PMCID: PMC5336242 DOI: 10.1371/journal.pone.0173056] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 02/14/2017] [Indexed: 12/12/2022] Open
Abstract
It has been well established that many vaccinia virus proteins suppress host antiviral pathways by targeting the transcription of antiviral proteins, thus evading the host innate immune system. However, whether viral proteins have an effect on the host’s overall cellular transcription is less understood. In this study we investigated the regulation of heterochromatin during vaccinia virus infection. Heterochromatin is a highly condensed form of chromatin that is less transcriptionally active and characterized by methylation of histone proteins. We examined the change in methylation of two histone proteins, H3 and H4, which are major markers of heterochromatin, during the course of viral infection. Using immunofluorescence microscopy and flow cytometry we were able to track the overall change in the methylated levels of H3K9 and H4K20. Our results suggest that there is significant increase in methylation of H3K9 and H4K20 during Orthopoxviruses infection compared to mock-infected cells. However, this effect was not seen when we infected cells with Leporipoxviruses. We further screened several vaccinia virus single and multi-gene deletion mutant and identified the vaccinia virus gene K7R as a contributor to the increase in cellular histone methylation during infection.
Collapse
Affiliation(s)
- Wondimagegnehu M. Teferi
- Department of Medical Microbiology & Immunology, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| | - Megan A. Desaulniers
- Department of Medical Microbiology & Immunology, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| | - Ryan S. Noyce
- Department of Medical Microbiology & Immunology, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| | - Mira Shenouda
- Department of Medical Microbiology & Immunology, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| | - Brittany Umer
- Department of Medical Microbiology & Immunology, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| | - David H. Evans
- Department of Medical Microbiology & Immunology, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
- * E-mail:
| |
Collapse
|
26
|
Wyatt LS, Earl PL, Moss B. Generation of Recombinant Vaccinia Viruses. ACTA ACUST UNITED AC 2017; 117:16.17.1-16.17.18. [PMID: 28060405 DOI: 10.1002/cpmb.32] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
This unit describes how to infect cells with vaccinia virus and then transfect them with a plasmid-transfer vector or PCR fragment to generate a recombinant virus. Selection and screening methods used to isolate recombinant viruses and a method for the amplification of recombinant viruses are described. Finally, a method for live immunostaining that has been used primarily for detection of recombinant modified vaccinia virus Ankara (MVA) is presented. © 2017 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Linda S Wyatt
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Patricia L Earl
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Bernard Moss
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
27
|
Kilinc MO, Ehrig K, Pessian M, Minev BR, Szalay AA. Colonization of xenograft tumors by oncolytic vaccinia virus (VACV) results in enhanced tumor killing due to the involvement of myeloid cells. J Transl Med 2016; 14:340. [PMID: 27993141 PMCID: PMC5168712 DOI: 10.1186/s12967-016-1096-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 11/24/2016] [Indexed: 12/25/2022] Open
Abstract
Background The mechanisms by which vaccinia virus (VACV) interacts with the innate immune components are complex and involve different mechanisms. iNOS-mediated NO production by myeloid cells is one of the central antiviral mechanisms and this study aims to investigate specifically whether iNOS-mediated NO production by myeloid cells, is involved in tumor eradication following the virus treatment. Methods Human colon adenocarcinoma (HCT-116) xenograft tumors were infected by VACV. Infiltration of iNOS+ myeloid cell population into the tumor, and virus titer was monitored following the treatment. Single-cell suspensions were stained for qualitative and quantitative flow analysis. The effect of different myeloid cell subsets on tumor growth and colonization were investigated by depletion studies. Finally, in vitro culture experiments were carried out to study NO production and tumor cell killing. Student’s t test was used for comparison between groups in all of the experiments. Results Infection of human colon adenocarcinoma (HCT-116) xenograft tumors by VACV has led to recruitment of many CD11b+ ly6G+ myeloid-derived suppressor cells (MDSCs), with enhanced iNOS expression in the tumors, and to an increased intratumoral virus titer between days 7 and 10 post-VACV therapy. In parallel, both single and multiple rounds of iNOS-producing cell depletions caused very rapid tumor growth within the same period after virus injection, indicating that VACV-induced iNOS+ MDSCs could be an important antitumor effector component. A continuous blockade of iNOS by its specific inhibitor, L-NIL, showed similar tumor growth enhancement 7–10 days post-infection. Finally, spleen-derived iNOS+ MDSCs isolated from virus-injected tumor bearing mice produced higher amounts of NO and effectively killed HCT-116 cells in in vitro transwell experiments. Conclusions We initially hypothesized that NO could be one of the factors that limits active spreading of the virus in the cancerous tissue. In contrast to our initial hypothesis, we observed that PMN-MDSCs were the main producer of NO through iNOS and NO provided a beneficial antitumor effect, The results strongly support an important novel role for VACV infection in the tumor microenvironment. VACV convert tumor-promoting MDSCs into tumor-killing cells by inducing higher NO production.
Collapse
Affiliation(s)
- Mehmet Okyay Kilinc
- Department of Biochemistry, Biocenter, University of Würzburg, Am Hubland, 97074, Würzburg, Germany.,San Diego Science Center, Genelux Corporation, San Diego, CA, USA
| | - Klaas Ehrig
- Department of Biochemistry, Biocenter, University of Würzburg, Am Hubland, 97074, Würzburg, Germany
| | - Maysam Pessian
- San Diego Science Center, Genelux Corporation, San Diego, CA, USA
| | - Boris R Minev
- San Diego Science Center, Genelux Corporation, San Diego, CA, USA.,Department of Radiation Medicine and Applied Sciences, Rebecca & John Moores Comprehensive Cancer Center, University of California, San Diego, CA, USA
| | - Aladar A Szalay
- Department of Biochemistry, Biocenter, University of Würzburg, Am Hubland, 97074, Würzburg, Germany. .,San Diego Science Center, Genelux Corporation, San Diego, CA, USA. .,Department of Radiation Medicine and Applied Sciences, Rebecca & John Moores Comprehensive Cancer Center, University of California, San Diego, CA, USA.
| |
Collapse
|
28
|
Increased attenuation but decreased immunogenicity by deletion of multiple vaccinia virus immunomodulators. Vaccine 2016; 34:4827-34. [PMID: 27544585 PMCID: PMC5022402 DOI: 10.1016/j.vaccine.2016.08.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 07/30/2016] [Accepted: 08/01/2016] [Indexed: 12/18/2022]
Abstract
Vaccinia virus-derived vaccine vectors are being engineered to improve immunogenicity. Deleting genes with immunomodulatory function can increase immunogenicity and decrease virulence. Deletion of N1L, C6L or K7R individually improves immunogenicity, but not in combination. A virus lacking all three genes induces poorer CD8+ T cell and neutralising antibody responses.
Vaccinia virus (VACV)-derived vectors are popular candidates for vaccination against diseases such as HIV-1, malaria and tuberculosis. However, their genomes encode a multitude of proteins with immunomodulatory functions, several of which reduce the immunogenicity of these vectors. Hitherto only limited studies have investigated whether the removal of these immunomodulatory genes in combination can increase vaccine efficacy further. To this end we constructed viruses based on VACV strain Western Reserve (WR) lacking up to three intracellular innate immunomodulators (N1, C6 and K7). These genes were selected because the encoded proteins had known functions in innate immunity and the deletion of each gene individually had caused a decrease in virus virulence in the murine intranasal and intradermal models of infection and an increase in immunogenicity. Data presented here demonstrate that deletion of two, or three of these genes in combination attenuated the virus further in an incremental manner. However, when vaccinated mice were challenged with VACV WR the double and triple gene deletion viruses provided weaker protection against challenge. This was accompanied by inferior memory CD8+ T cell responses and lower neutralising antibody titres. This study indicates that, at least for the three genes studied in the context of VACV WR, the single gene deletion viruses are the best vaccine vectors, and that increased attenuation induced by deletion of additional genes decreased immunogenicity. These data highlight the fine balance and complex relationship between viral attenuation and immunogenicity. Given that the proteins encoded by the genes examined in this study are known to affect specific aspects of innate immunity, the set of viruses constructed here are interesting tools to probe the role of the innate immune response in influencing immune memory and vaccine efficacy.
Collapse
|
29
|
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: 205] [Impact Index Per Article: 25.6] [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
|
30
|
Targeted Mutagenesis of Guinea Pig Cytomegalovirus Using CRISPR/Cas9-Mediated Gene Editing. J Virol 2016; 90:6989-6998. [PMID: 27226370 DOI: 10.1128/jvi.00139-16] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 05/17/2016] [Indexed: 12/20/2022] Open
Abstract
UNLABELLED The cytomegaloviruses (CMVs) are among the most genetically complex mammalian viruses, with viral genomes that often exceed 230 kbp. Manipulation of cytomegalovirus genomes is largely performed using infectious bacterial artificial chromosomes (BACs), which necessitates the maintenance of the viral genome in Escherichia coli and successful reconstitution of virus from permissive cells after transfection of the BAC. Here we describe an alternative strategy for the mutagenesis of guinea pig cytomegalovirus that utilizes clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9)-mediated genome editing to introduce targeted mutations to the viral genome. Transient transfection and drug selection were used to restrict lytic replication of guinea pig cytomegalovirus to cells that express Cas9 and virus-specific guide RNA. The result was highly efficient editing of the viral genome that introduced targeted insertion or deletion mutations to nonessential viral genes. Cotransfection of multiple virus-specific guide RNAs or a homology repair template was used for targeted, markerless deletions of viral sequence or to introduce exogenous sequence by homology-driven repair. As CRISPR/Cas9 mutagenesis occurs directly in infected cells, this methodology avoids selective pressures that may occur during propagation of the viral genome in bacteria and may facilitate genetic manipulation of low-passage or clinical CMV isolates. IMPORTANCE The cytomegalovirus genome is complex, and viral adaptations to cell culture have complicated the study of infection in vivo Recombineering of viral bacterial artificial chromosomes enabled the study of recombinant cytomegaloviruses. Here we report the development of an alternative approach using CRISPR/Cas9-based mutagenesis in guinea pig cytomegalovirus, a small-animal model of congenital cytomegalovirus disease. CRISPR/Cas9 mutagenesis can introduce the same types of mutations to the viral genome as bacterial artificial chromosome recombineering but does so directly in virus-infected cells. CRISPR/Cas9 mutagenesis is not dependent on a bacterial intermediate, and defined viral mutants can be recovered after a limited number of viral genome replications, minimizing the risk of spontaneous mutation.
Collapse
|
31
|
Use of Reporter Genes in the Generation of Vaccinia Virus-Derived Vectors. Viruses 2016; 8:v8050134. [PMID: 27213433 PMCID: PMC4885089 DOI: 10.3390/v8050134] [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] [Received: 03/18/2016] [Revised: 05/10/2016] [Accepted: 05/12/2016] [Indexed: 12/14/2022] Open
Abstract
Vaccinia virus (VACV) is one of the most extensively-studied viruses of the Poxviridae family. It is easy to genetically modify, so it has become a key tool for many applications. In this context, reporter genes facilitate the study of the role of foreign genes introduced into the genome of VACV. In this review, we describe the type of reporter genes that have been used to generate reporter-expressing VACV and the applications of the recombinant viruses obtained. Reporter-expressing VACV are currently employed in basic and immunology research, in the development of vaccines and cancer treatment.
Collapse
|
32
|
Wyatt LS, Earl PL, Moss B. Generation of Recombinant Vaccinia Viruses. CURRENT PROTOCOLS IN MICROBIOLOGY 2015; 39:14A.4.1-14A.4.18. [PMID: 26528782 PMCID: PMC5123791 DOI: 10.1002/9780471729259.mc14a04s39] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
This unit describes how to infect cells with vaccinia virus and then transfect them with a plasmid-transfer vector or PCR fragment to generate a recombinant virus. Selection and screening methods used to isolate recombinant viruses and a method for the amplification of recombinant viruses are described. Finally, a method for live immunostaining that has been used primarily for detection of recombinant modified vaccinia virus Ankara (MVA) is presented.
Collapse
Affiliation(s)
- Linda S Wyatt
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Patricia L Earl
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Bernard Moss
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
33
|
A marker-free system for highly efficient construction of vaccinia virus vectors using CRISPR Cas9. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2015; 2:15035. [PMID: 26417609 PMCID: PMC4571730 DOI: 10.1038/mtm.2015.35] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 08/05/2015] [Indexed: 02/07/2023]
Abstract
The current method for creation of vaccinia virus (VACV) vectors involves using a selection and purification marker, however inclusion of a gene without therapeutic value in the resulting vector is not desirable for clinical use. The Cre-LoxP system has been used to make marker-free Poxviruses, but the efficiency was very low. To obtain a marker-free VACV vector, we developed marker gene excision systems to modify the thymidine kinase (TK) region and N1L regions using Cre-Loxp and Flp-FRET systems respectively. CRISPR-Cas9 system significantly resulted in a high efficiency (~90%) in generation of marker gene-positive TK-mutant VACV vector. The marker gene (RFP) could be excised from the recombinant virus using Cre recombinase. To make a marker-free VV vector with double gene deletions targeting the TK and N1L gene, we constructed a donor repair vector targeting the N1L gene, which can carry a therapeutic gene and the marker (RFP) that could be excised from the recombinant virus using Flp recombinase. The marker-free system developed here can be used to efficiently construct VACV vectors armed with any therapeutic genes in the TK region or N1L region without marker genes. Our marker-free system platform has significant potential for development of new marker-free VACV vectors for clinical application.
Collapse
|
34
|
Strnadova P, Ren H, Valentine R, Mazzon M, Sweeney TR, Brierley I, Smith GL. Inhibition of Translation Initiation by Protein 169: A Vaccinia Virus Strategy to Suppress Innate and Adaptive Immunity and Alter Virus Virulence. PLoS Pathog 2015; 11:e1005151. [PMID: 26334635 PMCID: PMC4559412 DOI: 10.1371/journal.ppat.1005151] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Accepted: 08/13/2015] [Indexed: 12/19/2022] Open
Abstract
Vaccinia virus (VACV) is the prototypic orthopoxvirus and the vaccine used to eradicate smallpox. Here we show that VACV strain Western Reserve protein 169 is a cytoplasmic polypeptide expressed early during infection that is excluded from virus factories and inhibits the initiation of cap-dependent and cap-independent translation. Ectopic expression of protein 169 causes the accumulation of 80S ribosomes, a reduction of polysomes, and inhibition of protein expression deriving from activation of multiple innate immune signaling pathways. A virus lacking 169 (vΔ169) replicates and spreads normally in cell culture but is more virulent than parental and revertant control viruses in intranasal and intradermal murine models of infection. Intranasal infection by vΔ169 caused increased pro-inflammatory cytokines and chemokines, infiltration of pulmonary leukocytes, and lung weight. These alterations in innate immunity resulted in a stronger CD8+ T-cell memory response and better protection against virus challenge. This work illustrates how inhibition of host protein synthesis can be a strategy for virus suppression of innate and adaptive immunity. Long after smallpox was eradicated by vaccination with vaccinia virus, the study of this virus continues to reveal novel aspects of the interactions between a virus and the host in which it replicates. In this work we investigated the function of a previously uncharacterized VACV protein, called 169. The results show that protein 169 inhibits the synthesis of host proteins in cells and thereby provides a broad inhibition of the host innate immune response to infection. Unlike several other virus inhibitors of host protein synthesis, protein 169 acts by inhibiting the initiation of protein synthesis by both cap-dependent and cap-independent pathways. Also unlike several other virus protein synthesis inhibitors, the loss of protein 169 does not affect virus replication or spread, but the virus virulence was increased. This more severe infection is, however, cleared more rapidly and results in a stronger immunological memory response that is mediated by T-cells and provides better protection against re-infection. This work illustrates how shutting down host protein synthesis can be a strategy to block the host immune response to infection rather than a means to manufacture more virus particles.
Collapse
Affiliation(s)
- Pavla Strnadova
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
- Department of Virology, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Hongwei Ren
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
- Department of Virology, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Robert Valentine
- Department of Virology, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Michela Mazzon
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
- Department of Virology, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Trevor R. Sweeney
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Ian Brierley
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Geoffrey L. Smith
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
- Department of Virology, Faculty of Medicine, Imperial College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
35
|
Lakis NS, Li Y, Abraham JL, Upton C, Blair DC, Smith S, Zhao H, Damon IK. Novel Poxvirus Infection in an Immune Suppressed Patient. Clin Infect Dis 2015; 61:1543-8. [PMID: 26243783 DOI: 10.1093/cid/civ643] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 07/19/2015] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Human and animal poxvirus infections are being reported with increasing frequency. We describe a challenging case history and treatment of a previously unknown poxvirus rash illness in a renal transplant patient. METHODS A combination of classical microbiology techniques, including viral culture and electron microscopy, were used to provide initial clinical diagnosis. Subsequent standard polymerase chain reaction assays available in 2001 were noncontributory. Next generation sequencing was used to provide definitive diagnosis. RESULTS Retrospectively, next generation sequencing methods were used to ultimately provide the definitive diagnosis of a novel poxvirus infection initially identified by electron microscopy. The closest relative of this poxvirus, identified in North America, is a poxvirus collected from a mosquito pool from Central Africa in 1972. CONCLUSIONS This diagnostic quandary was ultimately solved using next generation DNA sequencing. This article describes the use of classical and next generation diagnostic strategies to identify etiologic agents of emerging infectious diseases and once again demonstrates the susceptibility of immunossupressed patients to novel pathogens. The virus identified is closely related to Yoka virus; these viruses appear to have independently diverged from a common ancestor of all known orthopoxviruses.
Collapse
Affiliation(s)
- Nelli S Lakis
- Department of Pathology, SUNY Upstate Medical University, Syracuse, New York
| | - Yu Li
- Division of High Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Jerrold L Abraham
- Department of Pathology, SUNY Upstate Medical University, Syracuse, New York
| | - Chris Upton
- Department of Biochemistry and Microbiology, University of Victoria, British Columbia, Canada
| | - Donald C Blair
- Department of Medicine, SUNY Upstate Medical University, Syracuse, New York
| | - Scott Smith
- Division of High Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Hui Zhao
- Division of High Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Inger K Damon
- Division of High Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| |
Collapse
|
36
|
Kirscher L, Deán-Ben XL, Scadeng M, Zaremba A, Zhang Q, Kober C, Fehm TF, Razansky D, Ntziachristos V, Stritzker J, Szalay AA. Doxycycline Inducible Melanogenic Vaccinia Virus as Theranostic Anti-Cancer Agent. Theranostics 2015; 5:1045-57. [PMID: 26199644 PMCID: PMC4508495 DOI: 10.7150/thno.12533] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 05/14/2015] [Indexed: 12/02/2022] Open
Abstract
We reported earlier the diagnostic potential of a melanogenic vaccinia virus based system in magnetic resonance (MRI) and optoacoustic deep tissue imaging (MSOT). Since melanin overproduction lead to attenuated virus replication, we constructed a novel recombinant vaccinia virus strain (rVACV), GLV-1h462, which expressed the key enzyme of melanogenesis (tyrosinase) under the control of an inducible promoter-system. In this study melanin production was detected after exogenous addition of doxycycline in two different tumor xenograft mouse models. Furthermore, it was confirmed that this novel vaccinia virus strain still facilitated signal enhancement as detected by MRI and optoacoustic tomography. At the same time we demonstrated an enhanced oncolytic potential compared to the constitutively melanin synthesizing rVACV system.
Collapse
Affiliation(s)
- Lorenz Kirscher
- 1. University of Würzburg, Department of Biochemistry, Am Hubland, 97074 Würzburg, Germany
| | - Xosé Luis Deán-Ben
- 4. Helmholtz Institute, IBMI, Ingolstädter Landstraße 1, 85764 Oberschleißheim, Germany
| | - Miriam Scadeng
- 3. University of San Diego, Center of Functional MRI, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Angelika Zaremba
- 4. Helmholtz Institute, IBMI, Ingolstädter Landstraße 1, 85764 Oberschleißheim, Germany
| | - Qian Zhang
- 2. Genelux Cooperation, San Diego Science Center, 3030 Bunker Hill St, San Diego, CA 92109, USA
| | - Christina Kober
- 1. University of Würzburg, Department of Biochemistry, Am Hubland, 97074 Würzburg, Germany
| | - Thomas Felix Fehm
- 4. Helmholtz Institute, IBMI, Ingolstädter Landstraße 1, 85764 Oberschleißheim, Germany
| | - Daniel Razansky
- 4. Helmholtz Institute, IBMI, Ingolstädter Landstraße 1, 85764 Oberschleißheim, Germany
| | - Vasilis Ntziachristos
- 4. Helmholtz Institute, IBMI, Ingolstädter Landstraße 1, 85764 Oberschleißheim, Germany
| | - Jochen Stritzker
- 1. University of Würzburg, Department of Biochemistry, Am Hubland, 97074 Würzburg, Germany
- 2. Genelux Cooperation, San Diego Science Center, 3030 Bunker Hill St, San Diego, CA 92109, USA
| | - Aladar A. Szalay
- 1. University of Würzburg, Department of Biochemistry, Am Hubland, 97074 Würzburg, Germany
- 2. Genelux Cooperation, San Diego Science Center, 3030 Bunker Hill St, San Diego, CA 92109, USA
- 5. Department of Radiation Oncology, Moores Cancer Center, University of California, La Jolla, CA 92093, USA
| |
Collapse
|
37
|
Almazán F, Sola I, Zuñiga S, Marquez-Jurado S, Morales L, Becares M, Enjuanes L. Reprint of: Coronavirus reverse genetic systems: infectious clones and replicons. Virus Res 2014; 194:67-75. [PMID: 25261606 PMCID: PMC7114485 DOI: 10.1016/j.virusres.2014.09.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Coronaviruses (CoVs) infect humans and many animal species, and are associated with respiratory, enteric, hepatic, and central nervous system diseases. The large size of the CoV genome and the instability of some CoV replicase gene sequences during its propagation in bacteria, represent serious obstacles for the development of reverse genetic systems similar to those used for smaller positive sense RNA viruses. To overcome these limitations, several alternatives to more conventional plasmid-based approaches have been established in the last 13 years. In this report, we briefly review and discuss the different reverse genetic systems developed for CoVs, paying special attention to the severe acute respiratory syndrome CoV (SARS-CoV).
Collapse
Affiliation(s)
- Fernando Almazán
- Department of Molecular and Cell Biology. Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, C/ Darwin 3, Cantoblanco, 28049 Madrid, Spain
| | - Isabel Sola
- Department of Molecular and Cell Biology. Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, C/ Darwin 3, Cantoblanco, 28049 Madrid, Spain
| | - Sonia Zuñiga
- Department of Molecular and Cell Biology. Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, C/ Darwin 3, Cantoblanco, 28049 Madrid, Spain
| | - Silvia Marquez-Jurado
- Department of Molecular and Cell Biology. Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, C/ Darwin 3, Cantoblanco, 28049 Madrid, Spain
| | - Lucia Morales
- Department of Molecular and Cell Biology. Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, C/ Darwin 3, Cantoblanco, 28049 Madrid, Spain
| | - Martina Becares
- Department of Molecular and Cell Biology. Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, C/ Darwin 3, Cantoblanco, 28049 Madrid, Spain
| | - Luis Enjuanes
- Department of Molecular and Cell Biology. Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, C/ Darwin 3, Cantoblanco, 28049 Madrid, Spain.
| |
Collapse
|
38
|
Almazán F, Sola I, Zuñiga S, Marquez-Jurado S, Morales L, Becares M, Enjuanes L. Coronavirus reverse genetic systems: infectious clones and replicons. Virus Res 2014; 189:262-70. [PMID: 24930446 PMCID: PMC4727449 DOI: 10.1016/j.virusres.2014.05.026] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 05/27/2014] [Accepted: 05/28/2014] [Indexed: 01/09/2023]
Abstract
Coronaviruses (CoVs) infect humans and many animal species, and are associated with respiratory, enteric, hepatic, and central nervous system diseases. The large size of the CoV genome and the instability of some CoV replicase gene sequences during its propagation in bacteria, represent serious obstacles for the development of reverse genetic systems similar to those used for smaller positive sense RNA viruses. To overcome these limitations, several alternatives to more conventional plasmid-based approaches have been established in the last 13 years. In this report, we briefly review and discuss the different reverse genetic systems developed for CoVs, paying special attention to the severe acute respiratory syndrome CoV (SARS-CoV).
Collapse
Affiliation(s)
- Fernando Almazán
- Department of Molecular and Cell Biology. Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, C/ Darwin 3, Cantoblanco, 28049 Madrid, Spain
| | - Isabel Sola
- Department of Molecular and Cell Biology. Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, C/ Darwin 3, Cantoblanco, 28049 Madrid, Spain
| | - Sonia Zuñiga
- Department of Molecular and Cell Biology. Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, C/ Darwin 3, Cantoblanco, 28049 Madrid, Spain
| | - Silvia Marquez-Jurado
- Department of Molecular and Cell Biology. Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, C/ Darwin 3, Cantoblanco, 28049 Madrid, Spain
| | - Lucia Morales
- Department of Molecular and Cell Biology. Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, C/ Darwin 3, Cantoblanco, 28049 Madrid, Spain
| | - Martina Becares
- Department of Molecular and Cell Biology. Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, C/ Darwin 3, Cantoblanco, 28049 Madrid, Spain
| | - Luis Enjuanes
- Department of Molecular and Cell Biology. Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, C/ Darwin 3, Cantoblanco, 28049 Madrid, Spain.
| |
Collapse
|
39
|
Ma W, Wei J, Wei Y, Guo H, Jin Y, Xue Y, Wang Y, Yi Z, Liu L, Huang J, Wang L. Immunogenicity of the capsid precursor and a nine-amino-acid site-directed mutant of the 3C protease of foot-and-mouth disease virus coexpressed by a recombinant goatpox virus. Arch Virol 2014; 159:1715-22. [PMID: 24473707 DOI: 10.1007/s00705-014-1984-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2013] [Accepted: 01/11/2014] [Indexed: 11/29/2022]
Abstract
The myristoylated capsid precursor mP1-2A of foot-and-mouth disease virus (FMDV), when expressed in mammalian cells and processed by the FMDV 3C protease, can self-assemble into virus-like particles (VLPs). In the present study, nine amino acids of the 3C protease were replaced by site-directed mutagenesis to create a mutant 3C protease, 9m3C. To coexpress mP1-2A and 9m3C and test the resulting proteolytic processing and VLP assembly, two recombinant goatpox viruses (rGTPVs) were constructed by the insertion of two coding regions, one for mP1-2A and the other for either 9m3C (rGTPV-mP1-2A-9m3C) or Theileria protective antigen (TPA) as a control (rGTPV-mP1-2A-TPA). The two exogenous genes were inserted into an intergenic region between loci gp_24 and gp_24.5 of the rGTPV genome. Western blotting of cells infected with rGTPV-mP1-2A-9m3C showed that proteins VP0, VP1, and VP3 from the mP1-2A processed by the 9m3C protease could be detected by polyclonal FMDV sera. As observed by electron microscopy, the infected cells produced VLPs with a diameter of about 25 ± 2 nm. Titers of neutralizing antibody against FMDV were significantly higher in mice inoculated with rGTPV-mP1-2A-9m3C, which expresses the 9m3C protease together with mP1-2A, than mice inoculated with the control rGTPV-mP1-2A-TPA, which does not express the protease. An ovine immunization test determined that sheep inoculated intramuscularly with rGTPV-mP1-2A-9m3C produced FMDV-specific neutralizing antibody, but its titers did not meet the requirement of the World Organization for Animal Health. The result indicates that further modifications of rGTPV-mP1-2A-9m3C are necessary to produce an effective vaccine.
Collapse
Affiliation(s)
- Wenge Ma
- Institute of Veterinary Medicine, Xinjiang Academy of Animal Science, 151 Eastern Kelamayi Street, Ürümqi, 830000, People's Republic of China,
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Recombinant Ag85B vaccine by taking advantage of characteristics of human parainfluenza type 2 virus vector showed Mycobacteria-specific immune responses by intranasal immunization. Vaccine 2014; 32:1727-35. [DOI: 10.1016/j.vaccine.2013.11.108] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Revised: 11/25/2013] [Accepted: 11/29/2013] [Indexed: 01/01/2023]
|
41
|
Verardi PH, Legrand FA, Chan KS, Peng Y, Jones LA, Yilma TD. IL-18 expression results in a recombinant vaccinia virus that is highly attenuated and immunogenic. J Interferon Cytokine Res 2014; 34:169-78. [PMID: 24168450 PMCID: PMC3942681 DOI: 10.1089/jir.2013.0052] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 08/19/2013] [Indexed: 01/06/2023] Open
Abstract
Interferon-γ (IFN-γ) is an attenuating factor for vaccinia virus (VACV), decreasing its virulence in vivo by more than a million fold. It is also a highly effective adjuvant when administered at the time of immunization with protein antigens. However, recombinant VACV (rVACV) vaccines expressing IFN-γ do not induce enhanced immune responses. It is possible that the IFN-γ expressed by rVACVs induces both an antiviral state and increased immunological clearance, thus resulting in decreased levels of antigen expression due to reduced viral replication and spread. We conjectured that delaying expression of IFN-γ would result in enhanced production of antigens by rVACVs thus resulting in increased immune responses to foreign antigens. Interleukin (IL)-18, also known as IFN-γ inducing factor, is a cytokine that induces T and NK cells to produce IFN-γ. In this study, we demonstrated that an rVACV expressing bioactive murine IL-18 replicated to low but detectable levels in vivo, unlike an rVACV expressing IFN-γ. Moreover, the rVACV expressing IL-18 was significantly attenuated in both immunocompromised and immunocompetent mice. This attenuation was dependent on IFN-γ, as IL-18 expression failed to attenuate VACV in IFN-γ knock-out mice. Cytotoxic T-cell (CTL) and anamnestic antibody responses were slightly increased in animals vaccinated with the rVACV expressing IL-18. Thus, induction of IFN-γ because of IL-18 expression resulted in an rVACV that replicated to low but detectable levels in vivo, yet elicited slightly better CTL and anamnestic humoral immune responses.
Collapse
Affiliation(s)
- Paulo H. Verardi
- Department of Pathobiology and Veterinary Science and Center of Excellence for Vaccine Research, College of Agriculture and Natural Resources, University of Connecticut, Storrs, Connecticut
| | - Fatema A. Legrand
- International Laboratory of Molecular Biology for Tropical Disease Agents, School of Veterinary Medicine, University of California, Davis, California
| | - Kenneth S. Chan
- International Laboratory of Molecular Biology for Tropical Disease Agents, School of Veterinary Medicine, University of California, Davis, California
| | - Yue Peng
- International Laboratory of Molecular Biology for Tropical Disease Agents, School of Veterinary Medicine, University of California, Davis, California
| | - Leslie A. Jones
- International Laboratory of Molecular Biology for Tropical Disease Agents, School of Veterinary Medicine, University of California, Davis, California
| | - Tilahun D. Yilma
- International Laboratory of Molecular Biology for Tropical Disease Agents, School of Veterinary Medicine, University of California, Davis, California
- Department of Medical Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, California
| |
Collapse
|
42
|
Marzook NB, Procter DJ, Lynn H, Yamamoto Y, Horsington J, Newsome TP. Methodology for the efficient generation of fluorescently tagged vaccinia virus proteins. J Vis Exp 2014:e51151. [PMID: 24473272 PMCID: PMC4089431 DOI: 10.3791/51151] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Tagging of viral proteins with fluorescent proteins has proven an indispensable approach to furthering our understanding of virus-host interactions. Vaccinia virus (VACV), the live vaccine used in the eradication of smallpox, is particularly amenable to fluorescent live-cell microscopy owing to its large virion size and the ease with which it can be engineered at the genome level. We report here an optimized protocol for generating recombinant viruses. The minimal requirements for targeted homologous recombination during vaccinia replication were determined, which allows the simplification of construct generation. This enabled the alliance of transient dominant selection (TDS) with a fluorescent reporter and metabolic selection to provide a rapid and modular approach to fluorescently label viral proteins. By streamlining the generation of fluorescent recombinant viruses, we are able to facilitate downstream applications such as advanced imaging analysis of many aspects of the virus-host interplay that occurs during virus replication.
Collapse
|
43
|
Hagen CJ, Titong A, Sarnoski EA, Verardi PH. Antibiotic-dependent expression of early transcription factor subunits leads to stringent control of vaccinia virus replication. Virus Res 2014; 181:43-52. [PMID: 24394294 DOI: 10.1016/j.virusres.2013.12.033] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2013] [Revised: 12/12/2013] [Accepted: 12/17/2013] [Indexed: 12/31/2022]
Abstract
The use of vaccinia virus (VACV) as the vaccine against variola virus resulted in the eradication of smallpox. VACV has since been used in the development of recombinant vaccine and therapeutic vectors, but complications associated with uncontrolled viral replication have constrained its use as a live viral vector. We propose to improve the safety of VACV as a live-replicating vector by using elements of the tet operon to control the transcription of genes that are essential for viral growth. Poxviruses encode all enzymes and factors necessary for their replication within the host cell cytoplasm. One essential VACV factor is the vaccinia early transcription factor (VETF) packaged into the viral core. This heterodimeric protein is required for expression of early VACV genes. VETF is composed of a large subunit encoded by the A7L gene and a small subunit encoded by the D6R gene. Two recombinant VACVs were generated in which either the A7L or D6R gene was placed under the control of tet operon elements to allow their transcription, and therefore viral replication, to be dependent on tetracycline antibiotics such as doxycycline. In the absence of inducers, no plaques were produced but abortively infected cells could be identified by expression of a reporter gene. In the presence of doxycycline, both recombinant viruses replicated indistinguishably from the wild-type strain. This stringent control of VACV replication can be used for the development of safer, next-generation VACV vaccines and therapeutic vectors. Such replication-inducible VACVs would only replicate when administered with tetracycline antibiotics, and if adverse events were to occur, treatment would be as simple as antibiotic cessation.
Collapse
Affiliation(s)
- Caitlin J Hagen
- Department of Pathobiology and Veterinary Science and Center of Excellence for Vaccine Research, College of Agriculture and Natural Resources, University of Connecticut, Storrs, CT 06269, United States
| | - Allison Titong
- Department of Pathobiology and Veterinary Science and Center of Excellence for Vaccine Research, College of Agriculture and Natural Resources, University of Connecticut, Storrs, CT 06269, United States
| | - Ethan A Sarnoski
- Department of Pathobiology and Veterinary Science and Center of Excellence for Vaccine Research, College of Agriculture and Natural Resources, University of Connecticut, Storrs, CT 06269, United States
| | - Paulo H Verardi
- Department of Pathobiology and Veterinary Science and Center of Excellence for Vaccine Research, College of Agriculture and Natural Resources, University of Connecticut, Storrs, CT 06269, United States.
| |
Collapse
|
44
|
Rubio D, Xu RH, Remakus S, Krouse TE, Truckenmiller ME, Thapa RJ, Balachandran S, Alcamí A, Norbury CC, Sigal LJ. Crosstalk between the type 1 interferon and nuclear factor kappa B pathways confers resistance to a lethal virus infection. Cell Host Microbe 2013; 13:701-10. [PMID: 23768494 DOI: 10.1016/j.chom.2013.04.015] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Revised: 02/28/2013] [Accepted: 04/08/2013] [Indexed: 01/09/2023]
Abstract
Nuclear factor kappa B (NF-κB) and type 1 interferon (T1-IFN) signaling are innate immune mechanisms activated upon viral infection. However, the role of NF-κB and its interplay with T1-IFN in antiviral immunity is poorly understood. We show that NF-κB is essential for resistance to ectromelia virus (ECTV), a mouse orthopoxvirus related to the virus causing human smallpox. Additionally, an ECTV mutant lacking an NF-κB inhibitor activates NF-κB more effectively in vivo, resulting in increased proinflammatory molecule transcription in uninfected cells and organs and decreased viral replication. Unexpectedly, NF-κB activation compensates for genetic defects in the T1-IFN pathway, such as a deficiency in the IRF7 transcription factor, resulting in virus control. Thus, overlap between the T1-IFN and NF-κB pathways allows the host to overcome genetic or pathogen-induced deficiencies in T1-IFN and survive an otherwise lethal poxvirus infection. These findings may also explain why some pathogens target both pathways to cause disease.
Collapse
Affiliation(s)
- Daniel Rubio
- Immune Cell Development and Host Defense Program, Research Institute of Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
The transcription factor nuclear factor kappa light-chain enhancer of activated B cells (NF-κB) plays a critical role in host defense against viral infection by inducing the production of proinflammatory mediators and type I interferon. Consequently, viruses have evolved many mechanisms to block its activation. The poxvirus vaccinia virus (VACV) encodes numerous inhibitors of NF-κB activation that target multiple points in the signaling pathway. A derivative of VACV strain Copenhagen, called vv811, lacking 55 open reading frames in the left and right terminal regions of the genome was reported to still inhibit NF-κB activation downstream of tumor necrosis factor alpha (TNF-α) and interleukin-1β (IL-1β), suggesting the presence of one or more additional inhibitors. In this study, we constructed a recombinant vv811 lacking the recently described NF-κB inhibitor A49 (vv811ΔA49), yielding a virus that lacked all currently described inhibitors downstream of TNF-α and IL-1β. Unlike vv811, vv811ΔA49 no longer inhibited degradation of the phosphorylated inhibitor of κBα and p65 translocated into the nucleus. However, despite this translocation, vv811ΔA49 still inhibited TNF-α- and IL-1β-induced NF-κB-dependent reporter gene expression and the transcription and production of cytokines induced by these agonists. This inhibition did not require late viral gene expression. These findings indicate the presence of another inhibitor of NF-κB that is expressed early during infection and acts by a novel mechanism downstream of p65 translocation into the nucleus.
Collapse
|
46
|
Abstract
Poxvirus expression vectors were described in 1982 and quickly became widely used for vaccine development as well as research in numerous fields. Advantages of the vectors include simple construction, ability to accommodate large amounts of foreign DNA and high expression levels. Numerous poxvirus-based veterinary vaccines are currently in use and many others are in human clinical trials. The early reports of poxvirus vectors paved the way for and stimulated the development of other viral vectors and recombinant DNA vaccines.
Collapse
Affiliation(s)
- Bernard Moss
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
47
|
Vaccinia virus-mediated melanin production allows MR and optoacoustic deep tissue imaging and laser-induced thermotherapy of cancer. Proc Natl Acad Sci U S A 2013; 110:3316-20. [PMID: 23401518 DOI: 10.1073/pnas.1216916110] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
We reported earlier the delivery of antiangiogenic single chain antibodies by using oncolytic vaccinia virus strains to enhance their therapeutic efficacy. Here, we provide evidence that gene-evoked production of melanin can be used as a therapeutic and diagnostic mediator, as exemplified by insertion of only one or two genes into the genome of an oncolytic vaccinia virus strain. We found that produced melanin is an excellent reporter for optical imaging without addition of substrate. Melanin production also facilitated deep tissue optoacoustic imaging as well as MRI. In addition, melanin was shown to be a suitable target for laser-induced thermotherapy and enhanced oncolytic viral therapy. In conclusion, melanin as a mediator for thermotherapy and reporter for different imaging modalities may soon become a versatile alternative to replace fluorescent proteins also in other biological systems. After ongoing extensive preclinical studies, melanin overproducing oncolytic virus strains might be used in clinical trials in patients with cancer.
Collapse
|
48
|
Characterization of ectromelia virus deficient in EVM036, the homolog of vaccinia virus F13L, and its application for rapid generation of recombinant viruses. J Virol 2012; 86:13501-7. [PMID: 23035222 DOI: 10.1128/jvi.01732-12] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The orthopoxvirus (OPV) vaccinia virus (VACV) requires an intact F13L gene to produce enveloped virions (EV) and to form plaques in cell monolayers. Simultaneous introduction of an exogenous gene and F13L into F13L-deficient VACV results in expression of the foreign gene and restoration of plaque size. This is used as a method to rapidly generate VACV recombinants without the need for drug selection. However, whether other OPVs require the orthologs of F13L to generate EV and form plaques, whether F13L orthologs and EV are important for OPV pathogenesis in natural hosts, and whether a system based on F13L ortholog deficiency can be used to generate recombinant OPVs other than VACV have not been reported. The F13L ortholog in ectromelia virus (ECTV), the agent of mousepox, is EVM036. We show that ECTV lacking EVM036 formed small plaques and was highly attenuated in vivo but still induced strong antibody responses. Reintroduction of EVM036 in tandem with the DsRed gene resulted in a virus that expressed DsRed in infected cells but was indistinguishable from wild-type ECTV in terms of plaque size and in vivo virulence. Thus, our data show that, like F13L in VACV, EVM036 is required for ECTV plaque formation and that EVM036 and EV are important for ECTV virulence. Our experiments also suggest that OPVs deficient in F13L orthologs could serve as safer anti-OPV vaccines. Further, our results demonstrate that ECTV deficient in EVM036 can be exploited for the rapid generation of fully virulent ECTV expressing foreign genes of interest.
Collapse
|
49
|
Ayari-Fakhfakh E, do Valle TZ, Guillemot L, Panthier JJ, Bouloy M, Ghram A, Albina E, Cêtre-Sossah C. MBT/Pas mouse: a relevant model for the evaluation of Rift Valley fever vaccines. J Gen Virol 2012; 93:1456-1464. [DOI: 10.1099/vir.0.042754-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Currently, there are no worldwide licensed vaccines for Rift Valley fever (RVF) that are both safe and effective. Development and evaluation of vaccines, diagnostics and treatments depend on the availability of appropriate animal models. Animal models are also necessary to understand the basic pathobiology of infection. Here, we report the use of an inbred MBT/Pas mouse model that consistently reproduces RVF disease and serves our purpose for testing the efficacy of vaccine candidates; an attenuated Rift Valley fever virus (RVFV) and a recombinant RVFV–capripoxvirus. We show that this model is relevant for vaccine testing.
Collapse
Affiliation(s)
- Emna Ayari-Fakhfakh
- Institut Pasteur de Tunis, Université de Tunis El Manar, 1002 Tunis Belvédère, Tunisia
| | - Tânia Zaverucha do Valle
- Centre National de la Recherche Scientifique, Unité de Recherche Associée 2578, Paris, France
- Mouse Functional Genetics, Institut Pasteur, Paris, France
| | - Laurent Guillemot
- Centre National de la Recherche Scientifique, Unité de Recherche Associée 2578, Paris, France
- Mouse Functional Genetics, Institut Pasteur, Paris, France
| | - Jean-Jacques Panthier
- Centre National de la Recherche Scientifique, Unité de Recherche Associée 2578, Paris, France
- Mouse Functional Genetics, Institut Pasteur, Paris, France
| | - Michèle Bouloy
- Molecular Genetics of Bunyaviruses, Institut Pasteur, Paris, France
| | - Abdeljelil Ghram
- Institut Pasteur de Tunis, Université de Tunis El Manar, 1002 Tunis Belvédère, Tunisia
| | - Emmanuel Albina
- CIRAD, UMR Contrôle des Maladies, Montpellier, F-34398, France
| | | |
Collapse
|
50
|
Hudson PN, Self J, Weiss S, Braden Z, Xiao Y, Girgis NM, Emerson G, Hughes C, Sammons SA, Isaacs SN, Damon IK, Olson VA. Elucidating the role of the complement control protein in monkeypox pathogenicity. PLoS One 2012; 7:e35086. [PMID: 22496894 PMCID: PMC3322148 DOI: 10.1371/journal.pone.0035086] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Accepted: 03/12/2012] [Indexed: 11/19/2022] Open
Abstract
Monkeypox virus (MPXV) causes a smallpox-like disease in humans. Clinical and epidemiological studies provide evidence of pathogenicity differences between two geographically distinct monkeypox virus clades: the West African and Congo Basin. Genomic analysis of strains from both clades identified a ∼10 kbp deletion in the less virulent West African isolates sequenced to date. One absent open reading frame encodes the monkeypox virus homologue of the complement control protein (CCP). This modulatory protein prevents the initiation of both the classical and alternative pathways of complement activation. In monkeypox virus, CCP, also known as MOPICE, is a ∼24 kDa secretory protein with sequence homology to this superfamily of proteins. Here we investigate CCP expression and its role in monkeypox virulence and pathogenesis. CCP was incorporated into the West African strain and removed from the Congo Basin strain by homologous recombination. CCP expression phenotypes were confirmed for both wild type and recombinant monkeypox viruses and CCP activity was confirmed using a C4b binding assay. To characterize the disease, prairie dogs were intranasally infected and disease progression was monitored for 30 days. Removal of CCP from the Congo Basin strain reduced monkeypox disease morbidity and mortality, but did not significantly decrease viral load. The inclusion of CCP in the West African strain produced changes in disease manifestation, but had no apparent effect on disease-associated mortality. This study identifies CCP as an important immuno-modulatory protein in monkeypox pathogenesis but not solely responsible for the increased virulence seen within the Congo Basin clade of monkeypox virus.
Collapse
Affiliation(s)
- Paul N Hudson
- Poxvirus and Rabies Branch, Division of High Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases and Biotechnology Core Facility Branch, Division of Safety Research, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|