1
|
Rendon-Marin S, Higuita-Gutiérrez LF, Ruiz-Saenz J. Safety and Immunogenicity of Morbillivirus canis Vaccines for Domestic and Wild Animals: A Scoping Review. Viruses 2024; 16:1078. [PMID: 39066240 PMCID: PMC11281360 DOI: 10.3390/v16071078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/21/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
Morbillivirus canis (canine distemper virus (CDV)) is recognized as a multihost pathogen responsible for a transmissible disease affecting both domestic and wild animals. A considerable portion of wildlife populations remain unvaccinated due to a lack of safety and immunogenicity data on existing vaccines for the prevention of CDV infection in these species. This review aimed to assess the current state of CDV vaccination research for both domestic and wild animals and to explore novel vaccine candidates through in vivo studies. It also sought to synthesize the scattered information from the extensive scientific literature on CDV vaccine research, identify key researchers in the field, and highlight areas where research on CDV vaccination is lacking. A scoping review was conducted across four databases following the PRISMA-ScR protocol, with information analyzed using absolute and relative frequencies and 95% confidence intervals (CIs) for study number proportions. Among the 2321 articles retrieved, 68 met the inclusion criteria and focused on CDV vaccines in various animal species, such as dogs, ferrets, minks, and mice. Most of the scientific community involved in this research was in the USA, Canada, France, and Denmark. Various vaccine types, including MLV CDV, recombinant virus, DNA plasmids, inactivated CDV, and MLV measles virus (MeV), were identified, along with diverse immunization routes and schedules employed in experimental and commercial vaccines. Safety and efficacy data were summarized. Notably, 37 studies reported postimmunization CDV challenge, primarily in dogs, revealing the survival rates of vaccinated animals. In summary, CDV vaccines generally demonstrate an acceptable safety profile in dogs and show promise as a means of controlling CDV. However, significant gaps in vaccine research persist, particularly concerning wildlife reservoirs, indicating the need for further investigation.
Collapse
Affiliation(s)
- Santiago Rendon-Marin
- Grupo de Investigación en Ciencias Animales—GRICA, Facultad de Medicina Veterinaria y Zootecnia, Universidad Cooperativa de Colombia, Bucaramanga 680001, Colombia;
- Grupo de Investigación Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Medellín 050001, Colombia
| | - Luis Felipe Higuita-Gutiérrez
- Facultad de Medicina, Universidad Cooperativa de Colombia, Medellín 050001, Colombia;
- Escuela de Microbiología, Universidad de Antioquia, Medellín 050001, Colombia
| | - Julian Ruiz-Saenz
- Grupo de Investigación en Ciencias Animales—GRICA, Facultad de Medicina Veterinaria y Zootecnia, Universidad Cooperativa de Colombia, Bucaramanga 680001, Colombia;
| |
Collapse
|
2
|
Schmitz KS, Rennick LJ, Tilston-Lunel NL, Comvalius AD, Laksono BM, Geers D, van Run P, de Vries RD, de Swart RL, Duprex WP. Rational attenuation of canine distemper virus (CDV) to develop a morbillivirus animal model that mimics measles in humans. J Virol 2024; 98:e0185023. [PMID: 38415596 PMCID: PMC10949419 DOI: 10.1128/jvi.01850-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 02/05/2024] [Indexed: 02/29/2024] Open
Abstract
Morbilliviruses are members of the family Paramyxoviridae and are known for their ability to cause systemic disease in a variety of mammalian hosts. The prototypic morbillivirus, measles virus (MeV), infects humans and still causes morbidity and mortality in unvaccinated children and young adults. Experimental infection studies in non-human primates have contributed to the understanding of measles pathogenesis. However, ethical restrictions call for the development of new animal models. Canine distemper virus (CDV) infects a wide range of animals, including ferrets, and its pathogenesis shares many features with measles. However, wild-type CDV infection is almost always lethal, while MeV infection is usually self-limiting. Here, we made five recombinant CDVs, predicted to be attenuated, and compared their pathogenesis to the non-attenuated recombinant CDV in a ferret model. Three viruses were insufficiently attenuated based on clinical signs, fatality, and systemic infection, while one virus was too attenuated. The last candidate virus caused a self-limiting infection associated with transient viremia and viral dissemination to all lymphoid tissues, was shed transiently from the upper respiratory tract, and did not result in acute neurological signs. Additionally, an in-depth phenotyping of the infected white blood cells showed lower infection percentages in all lymphocyte subsets when compared to the non-attenuated CDV. In conclusion, infection models using this candidate virus mimic measles and can be used to study pathogenesis-related questions and to test interventions for morbilliviruses in a natural host species.IMPORTANCEMorbilliviruses are transmitted via the respiratory route but cause systemic disease. The viruses use two cellular receptors to infect myeloid, lymphoid, and epithelial cells. Measles virus (MeV) remains an important cause of morbidity and mortality in humans, requiring animal models to study pathogenesis or intervention strategies. Experimental MeV infections in non-human primates are restricted by ethical and practical constraints, and animal morbillivirus infections in natural host species have been considered as alternatives. Inoculation of ferrets with wild-type canine distemper virus (CDV) has been used for this purpose, but in most cases, the virus overwhelms the immune system and causes highly lethal disease. Introduction of an additional transcription unit and an additional attenuating point mutation in the polymerase yielded a candidate virus that caused self-limiting disease with transient viremia and virus shedding. This rationally attenuated CDV strain can be used for experimental morbillivirus infections in ferrets that reflect measles in humans.
Collapse
Affiliation(s)
| | - Linda J. Rennick
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Natasha L. Tilston-Lunel
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | | | | - Daryl Geers
- Department of Viroscience, Erasmus MC, Rotterdam, the Netherlands
| | - Peter van Run
- Department of Viroscience, Erasmus MC, Rotterdam, the Netherlands
| | - Rory D. de Vries
- Department of Viroscience, Erasmus MC, Rotterdam, the Netherlands
| | - Rik L. de Swart
- Department of Viroscience, Erasmus MC, Rotterdam, the Netherlands
| | - W. Paul Duprex
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
3
|
Rennick LJ, Duprex WP, Tilston-Lunel NL. Generation of Defective Interfering Particles of Morbilliviruses Using Reverse Genetics. Methods Mol Biol 2024; 2808:57-70. [PMID: 38743362 DOI: 10.1007/978-1-0716-3870-5_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
RNA viruses generate defective genomes naturally during virus replication. Defective genomes that interfere with the infection dynamics either through resource competition or by interferon stimulation are known as defective interfering (DI) genomes. DI genomes can be successfully packaged into virus-like-particles referred to as defective interfering particles (DIPs). Such DIPs can sustainably coexist with the full-length virus particles and have been shown to negatively impact virus replication in vitro and in vivo. Here, we describe a method to generate a clonal DI genome population by reverse genetics. This method is applicable to other RNA viruses and will enable assessment of DIPs for their antiviral properties.
Collapse
Affiliation(s)
- Linda J Rennick
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - W Paul Duprex
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Natasha L Tilston-Lunel
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
4
|
Canine Distemper Virus in Endangered Species: Species Jump, Clinical Variations, and Vaccination. Pathogens 2022; 12:pathogens12010057. [PMID: 36678405 PMCID: PMC9862170 DOI: 10.3390/pathogens12010057] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/26/2022] [Accepted: 12/27/2022] [Indexed: 12/31/2022] Open
Abstract
Canine morbillivirus (Canine distemper virus, CDV) is the cause of distemper in a large number of different species, some of which are endangered. The clinical outcome associated with infection is variable and based on many factors, including the host species, the immune response of the individual animal to the infection, and variation in virus tropism and virulence. Unfortunately, the viral characteristics associated with virulence versus attenuation are not fully characterized, nor are the specific mutations that allow this virus to easily move and adapt from one species to another. Due to its wide host range, this virus is difficult to manage in ecosystems that are home to endangered species. Vaccination of the domestic dog, historically considered the reservoir species for this virus, at dog-wildlife interfaces has failed to control virus spread. CDV appears to be maintained by a metareservoir rather than a single species, requiring the need to vaccinate the wildlife species at risk. This is controversial, and there is a lack of a safe, effective vaccine for nondomestic species. This review focuses on topics that are paramount to protecting endangered species from a stochastic event, such as a CDV outbreak, that could lead to extinction.
Collapse
|
5
|
Wyss M, Gradauskaite V, Ebert N, Thiel V, Zurbriggen A, Plattet P. Efficient Recovery of Attenuated Canine Distemper Virus from cDNA. Virus Res 2022; 316:198796. [PMID: 35568090 DOI: 10.1016/j.virusres.2022.198796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/21/2022] [Accepted: 05/04/2022] [Indexed: 11/28/2022]
Abstract
To provide insights into the biology of the attenuated canine distemper virus (CDV) Onderstepoort (OP) strain (large plaque forming variant), design next-generation multivalent vaccines, or further investigate its promising potential as an oncolytic vector, we employed contemporary modifications to establish an efficient OP-CDV-based reverse genetics platform. Successful viral rescue was obtained however only upon recovery of a completely conserved charged residue (V13E) residing at the N-terminal region of the large protein (L). Although L-V13 and L-V13E did not display drastic differences in cellular localization and physical interaction with P, efficient polymerase complex (P+L) activity was recorded only with L-V13E. Interestingly, grafting mNeonGreen to the viral N protein via a P2A ribosomal skipping sequence (OPneon) and its derivative V-protein-knockout variant (OPneon-Vko) exhibited delayed replication kinetics in cultured cells. Collectively, we established an efficient OP-CDV-based reverse genetics system that enables the design of various strategies potentially contributing to veterinary medicine and research.
Collapse
Affiliation(s)
- Marianne Wyss
- Division of Neurological Sciences, Vetsuisse faculty, University of Bern, Switzerland
| | - Vaiva Gradauskaite
- Division of Neurological Sciences, Vetsuisse faculty, University of Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Nadine Ebert
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland; Institute of Virology and Immunology, Bern and Mittelhäusern, Switzerland
| | - Volker Thiel
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland; Institute of Virology and Immunology, Bern and Mittelhäusern, Switzerland
| | - Andreas Zurbriggen
- Division of Neurological Sciences, Vetsuisse faculty, University of Bern, Switzerland
| | - Philippe Plattet
- Division of Neurological Sciences, Vetsuisse faculty, University of Bern, Switzerland.
| |
Collapse
|
6
|
Wang B, Li S, Qiao Y, Fu Y, Nie J, Jiang S, Yao X, Pan Y, Zhao L, Wu C, Shi Y, Yin Y, Shan Y. Self-assembling ferritin nanoparticles coupled with linear sequences from canine distemper virus haemagglutinin protein elicit robust immune responses. J Nanobiotechnology 2022; 20:32. [PMID: 35012571 PMCID: PMC8744384 DOI: 10.1186/s12951-021-01229-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 12/24/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Canine distemper virus (CDV), which is highly infectious, has caused outbreaks of varying scales in domestic and wild animals worldwide, so the development of a high-efficiency vaccine has broad application prospects. Currently, the commercial vaccine of CDV is an attenuated vaccine, which has the disadvantages of a complex preparation process, high cost and safety risk. It is necessary to develop a safe and effective CDV vaccine that is easy to produce on a large scale. In this study, sequences of CDV haemagglutinin (HA) from the Yanaka strain were aligned, and three potential linear sequences, termed YaH3, YaH4, and YaH5, were collected. To increase the immunogenicity of the epitopes, ferritin was employed as a self-assembling nanoparticle element. The ferritin-coupled forms were termed YaH3F, YaH4F, and YaH5F, respectively. A full-length HA sequence coupled with ferritin was also constructed as a DNA vaccine to compare the immunogenicity of nanoparticles in prokaryotic expression. RESULT The self-assembly morphology of the proteins from prokaryotic expression was verified by transmission electron microscopy. All the proteins self-assembled into nanoparticles. The expression of the DNA vaccine YaHF in HEK-293T cells was also confirmed in vitro. After subcutaneous injection of epitope nanoparticles or intramuscular injection of DNA YaHF, all vaccines induced strong serum titres, and long-term potency of antibodies in serum could be detected after 84 days. Strong anti-CDV neutralizing activities were observed in both the YaH4F group and YaHF group. According to antibody typing and cytokine detection, YaH4F can induce both Th1 and Th2 immune responses. The results of flow cytometry detection indicated that compared with the control group, all the immunogens elicited an increase in CD3. Simultaneously, the serum antibodies induced by YaH4F and YaHF could significantly enhance the ADCC effect compared with the control group, indicating that the antibodies in the serum effectively recognized the antigens on the cell surface and induced NK cells to kill infected cells directly. CONCLUSIONS YaH4F self-assembling nanoparticle obtained by prokaryotic expression has no less of an immune effect than YaHF, and H4 has great potential to become a key target for the easy and rapid preparation of epitope vaccines.
Collapse
Affiliation(s)
- Bo Wang
- School of Chemistry and Life Science, Changchun University of Technology, Changchun, , 130012, Jilin, China
| | - Shuang Li
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, Jilin, China
| | - Yongbo Qiao
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, Jilin, China
| | - Yu Fu
- Changchun Xinuo BioTechnology Co., Ltd, Changchun, 130015, Jilin, China
| | - Jiaojiao Nie
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, Jilin, China
| | - Shun Jiang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, Jilin, China
| | - Xin Yao
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, Jilin, China
| | - Yi Pan
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, Jilin, China
| | - Linye Zhao
- School of Chemistry and Life Science, Changchun University of Technology, Changchun, , 130012, Jilin, China
| | - Congmei Wu
- School of Chemistry and Life Science, Changchun University of Technology, Changchun, , 130012, Jilin, China
| | - Yuhua Shi
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, Jilin, China
| | - Yuhe Yin
- School of Chemistry and Life Science, Changchun University of Technology, Changchun, , 130012, Jilin, China.
| | - Yaming Shan
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, Jilin, China. .,Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, Jilin, China.
| |
Collapse
|
7
|
Dong J, Chen Y, Shi L, Shen B, Sun X, Ruan K, Xia X, Feng H, Feng N. Nanoparticles of conformation-stabilized canine distemper virus hemagglutinin are highly immunogenic and induce robust immunity. Virol J 2021; 18:229. [PMID: 34809642 PMCID: PMC8607554 DOI: 10.1186/s12985-021-01702-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 11/04/2021] [Indexed: 11/13/2022] Open
Abstract
Background Canine distemper virus (CDV) infection of ferrets, dogs, and giant pandas causes an acute systemic disease involving multiple organ systems, including the respiratory tract, lymphoid system, and central nervous system. In this study, we tested a new candidate CDV vaccine-CDV nanoparticles-based on hemagglutinin protein. Methods The nanoparticles were generated from conformation-stabilized CDV hemagglutinin tetramers. Immune responses against CDV were evaluated in mice. Immunization was initiated 6 weeks after birth and boosted two times with 4-week intervals. The blood and mucosal samples were collected 2 weeks after each immunization. Results Vaccination with CDV nanoparticles elicited high levels of IgG antibody titers in mice (approximately sevenfold to eightfold higher than that obtained with soluble CDV H protein) and mucosal immune responses and developed increased CDV-specific neutralizing antibody. The mice that received nanoparticles showed significantly higher IFN-γ- and IL-4-secreting cell population in the spleen and lymph node compared with mice immunized with soluble H protein. The co-stimulatory molecular expression of CD80 and CD86 on the surface of DCs was also upregulated. Conclusion The results demonstrate that self-assembly into nanoparticles can increase the immunogenicity of vaccine antigens, and nanoparticles assembled from conformation-stabilized CDV H protein can serve as a new CDV vaccine.
Collapse
Affiliation(s)
- Jingjian Dong
- Medical School of Jiaxing University, Jiahang Road 118#, Nanhu District, Jiaxing City, Zhejiang Province, 314001, People's Republic of China
| | - Yan Chen
- Medical School of Jiaxing University, Jiahang Road 118#, Nanhu District, Jiaxing City, Zhejiang Province, 314001, People's Republic of China
| | - Lili Shi
- Medical School of Jiaxing University, Jiahang Road 118#, Nanhu District, Jiaxing City, Zhejiang Province, 314001, People's Republic of China
| | - Bing Shen
- Medical School of Jiaxing University, Jiahang Road 118#, Nanhu District, Jiaxing City, Zhejiang Province, 314001, People's Republic of China
| | - Xianliang Sun
- Medical School of Jiaxing University, Jiahang Road 118#, Nanhu District, Jiaxing City, Zhejiang Province, 314001, People's Republic of China
| | - Kaiyi Ruan
- Medical School of Jiaxing University, Jiahang Road 118#, Nanhu District, Jiaxing City, Zhejiang Province, 314001, People's Republic of China
| | - Xianzhu Xia
- Military Veterinary Research Institute of Academy of Military Medical Sciences, Changchun, 130122, People's Republic of China
| | - Hao Feng
- Medical School of Jiaxing University, Jiahang Road 118#, Nanhu District, Jiaxing City, Zhejiang Province, 314001, People's Republic of China.
| | - Na Feng
- Military Veterinary Research Institute of Academy of Military Medical Sciences, Changchun, 130122, People's Republic of China.
| |
Collapse
|
8
|
Heilmann E, Kimpel J, Hofer B, Rössler A, Blaas I, Egerer L, Nolden T, Urbiola C, Kräusslich HG, Wollmann G, von Laer D. Chemogenetic ON and OFF switches for RNA virus replication. Nat Commun 2021; 12:1362. [PMID: 33649317 PMCID: PMC7921684 DOI: 10.1038/s41467-021-21630-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 02/03/2021] [Indexed: 11/09/2022] Open
Abstract
Therapeutic application of RNA viruses as oncolytic agents or gene vectors requires a tight control of virus activity if toxicity is a concern. Here we present a regulator switch for RNA viruses using a conditional protease approach, in which the function of at least one viral protein essential for transcription and replication is linked to autocatalytical, exogenous human immunodeficiency virus (HIV) protease activity. Virus activity can be en- or disabled by various HIV protease inhibitors. Incorporating the HIV protease dimer in the genome of vesicular stomatitis virus (VSV) into the open reading frame of either the P- or L-protein resulted in an ON switch. Here, virus activity depends on co-application of protease inhibitor in a dose-dependent manner. Conversely, an N-terminal VSV polymerase tag with the HIV protease dimer constitutes an OFF switch, as application of protease inhibitor stops virus activity. This technology may also be applicable to other potentially therapeutic RNA viruses.
Collapse
Affiliation(s)
- E Heilmann
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
- Christian Doppler Laboratory for Viral Immunotherapy of Cancer, Medical University of Innsbruck, Innsbruck, Austria
| | - J Kimpel
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - B Hofer
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - A Rössler
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - I Blaas
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - L Egerer
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
- ViraTherapeutics GmbH, Innsbruck, Austria
| | - T Nolden
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
- ViraTherapeutics GmbH, Innsbruck, Austria
| | - C Urbiola
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
- Christian Doppler Laboratory for Viral Immunotherapy of Cancer, Medical University of Innsbruck, Innsbruck, Austria
- ViraTherapeutics GmbH, Innsbruck, Austria
| | - H G Kräusslich
- Department of Infectious Diseases, Virology, University Hospital Heidelberg, Heidelberg, Germany
- German Center for Infectious Disease Research, partner site Heidelberg, Heidelberg, Germany
| | - G Wollmann
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria.
- Christian Doppler Laboratory for Viral Immunotherapy of Cancer, Medical University of Innsbruck, Innsbruck, Austria.
| | - D von Laer
- Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
9
|
Stable Attenuation of Human Respiratory Syncytial Virus for Live Vaccines by Deletion and Insertion of Amino Acids in the Hinge Region between the mRNA Capping and Methyltransferase Domains of the Large Polymerase Protein. J Virol 2020; 94:JVI.01831-20. [PMID: 32999025 DOI: 10.1128/jvi.01831-20] [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] [Received: 09/17/2020] [Accepted: 09/17/2020] [Indexed: 01/02/2023] Open
Abstract
Human respiratory syncytial virus (RSV) is the leading viral cause of lower respiratory tract disease in infants and children worldwide. Currently, there are no FDA-approved vaccines to combat this virus. The large (L) polymerase protein of RSV replicates the viral genome and transcribes viral mRNAs. The L protein is organized as a core ring-like domain containing the RNA-dependent RNA polymerase and an appendage of globular domains containing an mRNA capping region and a cap methyltransferase region, which are linked by a flexible hinge region. Here, we found that the flexible hinge region of RSV L protein is tolerant to amino acid deletion or insertion. Recombinant RSVs carrying a single or double deletion or a single alanine insertion were genetically stable, highly attenuated in immortalized cells, had defects in replication and spread, and had a delay in innate immune cytokine responses in primary, well-differentiated, human bronchial epithelial (HBE) cultures. The replication of these recombinant viruses was highly attenuated in the upper and lower respiratory tracts of cotton rats. Importantly, these recombinant viruses elicited high levels of neutralizing antibody and provided complete protection against RSV replication. Taken together, amino acid deletions or insertions in the hinge region of the L protein can serve as a novel approach to rationally design genetically stable, highly attenuated, and immunogenic live virus vaccine candidates for RSV.IMPORTANCE Despite tremendous efforts, there are no FDA-approved vaccines for human respiratory syncytial virus (RSV). A live attenuated RSV vaccine is one of the most promising vaccine strategies for RSV. However, it has been a challenge to identify an RSV vaccine strain that has an optimal balance between attenuation and immunogenicity. In this study, we generated a panel of recombinant RSVs carrying a single and double deletion or a single alanine insertion in the large (L) polymerase protein that are genetically stable, sufficiently attenuated, and grow to high titer in cultured cells, while retaining high immunogenicity. Thus, these recombinant viruses may be promising vaccine candidates for RSV.
Collapse
|
10
|
Viral Pathogenesis, Recombinant Vaccines, and Oncolytic Virotherapy: Applications of the Canine Distemper Virus Reverse Genetics System. Viruses 2020; 12:v12030339. [PMID: 32244946 PMCID: PMC7150803 DOI: 10.3390/v12030339] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/17/2020] [Accepted: 03/18/2020] [Indexed: 12/26/2022] Open
Abstract
Canine distemper virus (CDV) is a highly contagious pathogen transmissible to a broad range of terrestrial and aquatic carnivores. Despite the availability of attenuated vaccines against CDV, the virus remains responsible for outbreaks of canine distemper (CD) with significant morbidity and mortality in domesticated and wild carnivores worldwide. CDV uses the signaling lymphocytic activation molecule (SLAM, or CD150) and nectin-4 (PVRL4) as entry receptors, well-known tumor-associated markers for several lymphadenomas and adenocarcinomas, which are also responsible for the lysis of tumor cells and apparent tumor regression. Thus, CDV vaccine strains have emerged as a promising platform of oncolytic viruses for use in animal cancer therapy. Recent advances have revealed that use of the CDV reverse genetic system (RGS) has helped increase the understanding of viral pathogenesis and explore the development of recombinant CDV vaccines. In addition, genetic engineering of CDV based on RGS approaches also has the potential of enhancing oncolytic activity and selectively targeting tumors. Here, we reviewed the host tropism and pathogenesis of CDV, and current development of recombinant CDV-based vaccines as well as their use as oncolytic viruses against cancers.
Collapse
|
11
|
Heilmann E, Kimpel J, Geley S, Naschberger A, Urbiola C, Nolden T, von Laer D, Wollmann G. The Methyltransferase Region of Vesicular Stomatitis Virus L Polymerase Is a Target Site for Functional Intramolecular Insertion. Viruses 2019; 11:v11110989. [PMID: 31717818 PMCID: PMC6893670 DOI: 10.3390/v11110989] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 10/20/2019] [Accepted: 10/23/2019] [Indexed: 11/16/2022] Open
Abstract
The L-protein of vesicular stomatitis virus (VSV) is a single-chain multi-domain RNA-dependent RNA polymerase. Previously reported attempts of intramolecular insertions of fluorescent proteins into the L-protein resulted in temperature-sensitive and highly attenuated polymerase activity. Here, we describe a novel insertion site that was selected based on in silico prediction. Of five preselected locations, insertion of the fluorescent protein mCherry in the VSV polymerase between amino acids 1620 and 1621 preserved polymerase function even after extended passaging and showed only mild attenuation compared to wildtype VSV polymerase. High magnification fluorescence imaging revealed a corpuscular cytosolic pattern for the L-protein. To confirm that the insertion site tolerates inclusion of proteins others than mCherry, we cloned mWasabi into the same position in L, generating a VSV-LmWasabi, which was also functional. We also generated a functional dual-color-dual-insertion VSV construct with intramolecularly labeled P and L-proteins. Together, our data present an approach to tag VSV polymerase intramolecularly without perturbing enzymatic activity. This L fusion protein might enable future tracing studies to monitor intracellular location of the VSV transcription and replication machinery in real-time life-imaging studies.
Collapse
Affiliation(s)
- Emmanuel Heilmann
- Division of Virology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (E.H.); (J.K.); (C.U.)
- Christian Doppler Laboratory for Viral Immunotherapy of Cancer, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Janine Kimpel
- Division of Virology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (E.H.); (J.K.); (C.U.)
| | - Stephan Geley
- Division of Molecular Pathophysiology, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| | - Andreas Naschberger
- Division of Genetic Epidemiology, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| | - Carles Urbiola
- Division of Virology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (E.H.); (J.K.); (C.U.)
- Christian Doppler Laboratory for Viral Immunotherapy of Cancer, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | | | - Dorotheé von Laer
- Division of Virology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (E.H.); (J.K.); (C.U.)
- Correspondence: (D.v.L.); (G.W.); Tel.: +43-512-9003-71701 (D.v.L.); +43-512-9003-71742 (G.W.)
| | - Guido Wollmann
- Division of Virology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (E.H.); (J.K.); (C.U.)
- Christian Doppler Laboratory for Viral Immunotherapy of Cancer, Medical University of Innsbruck, 6020 Innsbruck, Austria
- Correspondence: (D.v.L.); (G.W.); Tel.: +43-512-9003-71701 (D.v.L.); +43-512-9003-71742 (G.W.)
| |
Collapse
|
12
|
Chen C, Zhou M, Yan XG, Chen YX, Cui M, Chen HC, Fu ZF, Zhao L. A recombinant canine distemper virus expressing interleukin-7 enhances humoral immunity. J Gen Virol 2019; 100:602-615. [DOI: 10.1099/jgv.0.001247] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Chen Chen
- 1State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, PR China
- 2Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agriculture University, Wuhan, PR China
- 3College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, PR China
| | - Ming Zhou
- 1State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, PR China
- 2Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agriculture University, Wuhan, PR China
- 3College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, PR China
| | - Xiao-geng Yan
- 1State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, PR China
- 2Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agriculture University, Wuhan, PR China
- 3College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, PR China
| | - Yi-xi Chen
- 1State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, PR China
- 2Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agriculture University, Wuhan, PR China
- 3College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, PR China
| | - Min Cui
- 1State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, PR China
- 2Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agriculture University, Wuhan, PR China
- 3College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, PR China
| | - Huan-chun Chen
- 1State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, PR China
- 2Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agriculture University, Wuhan, PR China
- 3College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, PR China
| | - Zhen-fang Fu
- 4Department of Pathology, University of Georgia, Athens, GA, USA
- 3College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, PR China
- 1State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, PR China
- 2Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agriculture University, Wuhan, PR China
| | - Ling Zhao
- 3College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, PR China
- 1State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, PR China
- 2Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agriculture University, Wuhan, PR China
| |
Collapse
|
13
|
Schmidt ML, Hoenen T. Characterization of the catalytic center of the Ebola virus L polymerase. PLoS Negl Trop Dis 2017; 11:e0005996. [PMID: 28991917 PMCID: PMC5648267 DOI: 10.1371/journal.pntd.0005996] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 10/19/2017] [Accepted: 09/27/2017] [Indexed: 01/02/2023] Open
Abstract
Background Ebola virus (EBOV) causes a severe hemorrhagic fever in humans and non-human primates. While no licensed therapeutics are available, recently there has been tremendous progress in developing antivirals. Targeting the ribonucleoprotein complex (RNP) proteins, which facilitate genome replication and transcription, and particularly the polymerase L, is a promising antiviral approach since these processes are essential for the virus life cycle. However, until now little is known about L in terms of its structure and function, and in particular the catalytic center of the RNA-dependent RNA polymerase (RdRp) of L, which is one of the most promising molecular targets, has never been experimentally characterized. Methodology/Principal findings Using multiple sequence alignments with other negative sense single-stranded RNA viruses we identified the putative catalytic center of the EBOV RdRp. An L protein with mutations in this center was then generated and characterized using various life cycle modelling systems. These systems are based on minigenomes, i.e. miniature versions of the viral genome, in which the viral genes are exchanged against a reporter gene. When such minigenomes are coexpressed with RNP proteins in mammalian cells, the RNP proteins recognize them as authentic templates for replication and transcription, resulting in reporter activity reflecting these processes. Replication-competent minigenome systems indicated that our L catalytic domain mutant was impaired in genome replication and/or transcription, and by using replication-deficient minigenome systems, as well as a novel RT-qPCR-based genome replication assay, we showed that it indeed no longer supported either of these processes. However, it still showed similar expression to wild-type L, and retained its ability to be incorporated into inclusion bodies, which are the sites of EBOV genome replication. Conclusions/Significance We have experimentally defined the catalytic center of the EBOV RdRp, and thus a promising antiviral target regulating an essential aspect of the EBOV life cycle. Ebola viruses cause severe hemorrhagic fevers, and were responsible for the devastating Ebola virus disease epidemic in West Africa from 2013 to 2016. While a number of experimental therapeutics against these viruses target the viral polymerase, there are still significant gaps in our knowledge regarding this essential viral protein. In particular, until now no experimental evidence has been provided identifying the catalytic center of the viral RNA-dependent RNA polymerase, which is absolutely essential for the virus life cycle due to its role in replicating and transcribing the viral negative-sense RNA genome. Based on a comparison to related negative-sense RNA viruses from other virus families we identified a putative catalytic center within the Ebola virus polymerase, and provide the experimental evidence that the Ebola virus polymerase indeed utilizes a classical GDNQ motif for both genome replication and transcription. This finding not only increases our knowledge regarding the molecular biology of Ebola viruses, but also defines a molecular target for the development of antivirals against this deadly virus.
Collapse
Affiliation(s)
- Marie Luisa Schmidt
- Institute for Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald–Insel Riems, Germany
| | - Thomas Hoenen
- Institute for Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald–Insel Riems, Germany
- * E-mail:
| |
Collapse
|
14
|
Pan Z, Liu J, Ma J, Jin Q, Yao H, Osterrieder N. The recombinant EHV-1 vector producing CDV hemagglutinin as potential vaccine against canine distemper. Microb Pathog 2017; 111:388-394. [DOI: 10.1016/j.micpath.2017.09.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 09/02/2017] [Accepted: 09/05/2017] [Indexed: 10/18/2022]
|
15
|
Morbillivirus Experimental Animal Models: Measles Virus Pathogenesis Insights from Canine Distemper Virus. Viruses 2016; 8:v8100274. [PMID: 27727184 PMCID: PMC5086610 DOI: 10.3390/v8100274] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 09/29/2016] [Accepted: 09/30/2016] [Indexed: 12/19/2022] Open
Abstract
Morbilliviruses share considerable structural and functional similarities. Even though disease severity varies among the respective host species, the underlying pathogenesis and the clinical signs are comparable. Thus, insights gained with one morbillivirus often apply to the other members of the genus. Since the Canine distemper virus (CDV) causes severe and often lethal disease in dogs and ferrets, it is an attractive model to characterize morbillivirus pathogenesis mechanisms and to evaluate the efficacy of new prophylactic and therapeutic approaches. This review compares the cellular tropism, pathogenesis, mechanisms of persistence and immunosuppression of the Measles virus (MeV) and CDV. It then summarizes the contributions made by studies on the CDV in dogs and ferrets to our understanding of MeV pathogenesis and to vaccine and drugs development.
Collapse
|
16
|
[The multifunctional RNA polymerase L protein of non-segmented negative strand RNA viruses catalyzes unique mRNA capping]. Uirusu 2016; 64:165-78. [PMID: 26437839 DOI: 10.2222/jsv.64.165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Non-segmented negative strand RNA viruses belonging to the Mononegavirales order possess RNA-dependent RNA polymerase L proteins within viral particles. The L protein is a multifunctional enzyme catalyzing viral RNA synthesis and processing (i.e., mRNA capping, cap methylation, and polyadenylation). Using vesicular stomatitis virus (VSV) as a prototypic model virus, we have shown that the L protein catalyzes the unconventional mRNA capping reaction, which is strikingly different from the eukaryotic reaction. Furthermore, co-transcriptional pre-mRNA capping with the VSV L protein was found to be required for accurate stop?start transcription to synthesize full-length mRNAs in vitro and virus propagation in host cells. This article provides a review of historical and present studies leading to the elucidation of the molecular mechanism of VSV mRNA capping.
Collapse
|
17
|
Bloyet LM, Welsch J, Enchery F, Mathieu C, de Breyne S, Horvat B, Grigorov B, Gerlier D. HSP90 Chaperoning in Addition to Phosphoprotein Required for Folding but Not for Supporting Enzymatic Activities of Measles and Nipah Virus L Polymerases. J Virol 2016; 90:6642-6656. [PMID: 27170753 PMCID: PMC4944277 DOI: 10.1128/jvi.00602-16] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 05/03/2016] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED Nonsegmented negative-stranded RNA viruses, or members of the order Mononegavirales, share a conserved gene order and the use of elaborate transcription and replication machinery made up of at least four molecular partners. These partners have coevolved with the acquisition of the permanent encapsidation of the entire genome by the nucleoprotein (N) and the use of this N-RNA complex as a template for the viral polymerase composed of the phosphoprotein (P) and the large enzymatic protein (L). Not only is P required for polymerase function, but it also stabilizes the L protein through an unknown underlying molecular mechanism. By using NVP-AUY922 and/or 17-dimethylaminoethylamino-17-demethoxygeldanamycin as specific inhibitors of cellular heat shock protein 90 (HSP90), we found that efficient chaperoning of L by HSP90 requires P in the measles, Nipah, and vesicular stomatitis viruses. While the production of P remains unchanged in the presence of HSP90 inhibitors, the production of soluble and functional L requires both P and HSP90 activity. Measles virus P can bind the N terminus of L in the absence of HSP90 activity. Both HSP90 and P are required for the folding of L, as evidenced by a luciferase reporter insert fused within measles virus L. HSP90 acts as a true chaperon; its activity is transient and dispensable for the activity of measles and Nipah virus polymerases of virion origin. That the cellular chaperoning of a viral polymerase into a soluble functional enzyme requires the assistance of another viral protein constitutes a new paradigm that seems to be conserved within the Mononegavirales order. IMPORTANCE Viruses are obligate intracellular parasites that require a cellular environment for their replication. Some viruses particularly depend on the cellular chaperoning apparatus. We report here that for measles virus, successful chaperoning of the viral L polymerase mediated by heat shock protein 90 (HSP90) requires the presence of the viral phosphoprotein (P). Indeed, while P protein binds to the N terminus of L independently of HSP90 activity, both HSP90 and P are required to produce stable, soluble, folded, and functional L proteins. Once formed, the mature P+L complex no longer requires HSP90 to exert its polymerase functions. Such a new paradigm for the maturation of a viral polymerase appears to be conserved in several members of the Mononegavirales order, including the Nipah and vesicular stomatitis viruses.
Collapse
Affiliation(s)
- Louis-Marie Bloyet
- CIRI, International Center for Infectiology Research, Université de Lyon, Lyon, France
- INSERM, U1111, Lyon, France
- CNRS, UMR5308, Lyon, France
- Université Claude Bernard Lyon 1; Centre International de Recherche en Infectiologie, Lyon, France
- ENS Lyon, Lyon, France
| | - Jérémy Welsch
- CIRI, International Center for Infectiology Research, Université de Lyon, Lyon, France
- INSERM, U1111, Lyon, France
- CNRS, UMR5308, Lyon, France
- Université Claude Bernard Lyon 1; Centre International de Recherche en Infectiologie, Lyon, France
- ENS Lyon, Lyon, France
- Laboratoire d'Excellence Ecofect, Lyon, France
| | - François Enchery
- CIRI, International Center for Infectiology Research, Université de Lyon, Lyon, France
- INSERM, U1111, Lyon, France
- CNRS, UMR5308, Lyon, France
- Université Claude Bernard Lyon 1; Centre International de Recherche en Infectiologie, Lyon, France
- ENS Lyon, Lyon, France
| | - Cyrille Mathieu
- CIRI, International Center for Infectiology Research, Université de Lyon, Lyon, France
- INSERM, U1111, Lyon, France
- CNRS, UMR5308, Lyon, France
- Université Claude Bernard Lyon 1; Centre International de Recherche en Infectiologie, Lyon, France
- ENS Lyon, Lyon, France
- Laboratoire d'Excellence Ecofect, Lyon, France
| | - Sylvain de Breyne
- CIRI, International Center for Infectiology Research, Université de Lyon, Lyon, France
- INSERM, U1111, Lyon, France
- CNRS, UMR5308, Lyon, France
- Université Claude Bernard Lyon 1; Centre International de Recherche en Infectiologie, Lyon, France
- ENS Lyon, Lyon, France
| | - Branka Horvat
- CIRI, International Center for Infectiology Research, Université de Lyon, Lyon, France
- INSERM, U1111, Lyon, France
- CNRS, UMR5308, Lyon, France
- Université Claude Bernard Lyon 1; Centre International de Recherche en Infectiologie, Lyon, France
- ENS Lyon, Lyon, France
- Laboratoire d'Excellence Ecofect, Lyon, France
| | - Boyan Grigorov
- CIRI, International Center for Infectiology Research, Université de Lyon, Lyon, France
- INSERM, U1111, Lyon, France
- CNRS, UMR5308, Lyon, France
- Université Claude Bernard Lyon 1; Centre International de Recherche en Infectiologie, Lyon, France
- ENS Lyon, Lyon, France
- Cancer Research Center of Lyon (CRCL), INSERM, U1052, Université Claude Bernard Lyon 1, Lyon, France
| | - Denis Gerlier
- CIRI, International Center for Infectiology Research, Université de Lyon, Lyon, France
- INSERM, U1111, Lyon, France
- CNRS, UMR5308, Lyon, France
- Université Claude Bernard Lyon 1; Centre International de Recherche en Infectiologie, Lyon, France
- ENS Lyon, Lyon, France
- Laboratoire d'Excellence Ecofect, Lyon, France
| |
Collapse
|
18
|
Yi L, Cheng S. Preparation and Identification of a Single-chain Variable Fragment Antibody Against Canine Distemper Virus. Monoclon Antib Immunodiagn Immunother 2015; 34:228-32. [PMID: 26301925 DOI: 10.1089/mab.2015.0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The variable regions of the heavy chain (VH) and light chain (VL) were amplified by RT-PCR from the hybridoma 1N8, which secretes the monoclonal antibody against CDV N protein (aa 277-471). The VL and VH amplicons were combined using SOE-PCR by a 12 amino acid flexible linker (SSGGGGSGGGGS), which produced the scFv gene (named scFv/1N8). After sequence analysis, the scFv/1N8 gene was cloned into the prokaryotic expression vector PET32a with a His-tag. The recombinant scFv/1N8 protein was successfully expressed in recombinant Escherichia coli by IPTG induction. Moreover, the binding activity and specificity of the scFv were determined by indirect ELISA (His-tag) and competitive ELISA. The recombinant scFv/1N8 protein reported here will provide some basis for further antiviral drug research based on the scFv molecule.
Collapse
Affiliation(s)
- Li Yi
- Institute of Special Wild Economic Animal and Plant Science , Chinese Academy of Agricultural Sciences, Changchun, Jilin Province, P.R. China
| | - Shipeng Cheng
- Institute of Special Wild Economic Animal and Plant Science , Chinese Academy of Agricultural Sciences, Changchun, Jilin Province, P.R. China
| |
Collapse
|
19
|
Development of a reverse genetics system to generate a recombinant Ebola virus Makona expressing a green fluorescent protein. Virology 2015; 484:259-264. [PMID: 26122472 DOI: 10.1016/j.virol.2015.06.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 04/21/2015] [Accepted: 06/10/2015] [Indexed: 10/23/2022]
Abstract
Previous studies have demonstrated the potential application of reverse genetics technology in studying a broad range of aspects of viral biology, including gene regulation, protein function, cell entry, and pathogenesis. Here, we describe a highly efficient reverse genetics system used to generate recombinant Ebola virus (EBOV) based on a recent isolate from a human patient infected during the 2014-2015 outbreak in Western Africa. We also rescued a recombinant EBOV expressing a fluorescent reporter protein from a cleaved VP40 protein fusion. Using this virus and an inexpensive method to quantitate the expression of the foreign gene, we demonstrate its potential usefulness as a tool for screening antiviral compounds and measuring neutralizing antibodies.
Collapse
|
20
|
Ge J, Wang X, Tian M, Gao Y, Wen Z, Yu G, Zhou W, Zu S, Bu Z. Recombinant Newcastle disease viral vector expressing hemagglutinin or fusion of canine distemper virus is safe and immunogenic in minks. Vaccine 2015; 33:2457-62. [PMID: 25865465 DOI: 10.1016/j.vaccine.2015.03.091] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 03/22/2015] [Accepted: 03/27/2015] [Indexed: 11/26/2022]
Abstract
Canine Distemper Virus (CDV) infects many carnivores and cause several high-mortality disease outbreaks. The current CDV live vaccine cannot be safely used in some exotic species, such as mink and ferret. Here, we generated recombinant lentogenic Newcastle disease virus (NDV) LaSota expressing either envelope glycoproyein, heamagglutinine (H) or fusion protein (F), named as rLa-CDVH and rLa-CDVF, respectively. The feasibility of these recombinant NDVs to serve as live virus-vectored CD vaccine was evaluated in minks. rLa-CDVH induced significant neutralization antibodies (NA) to CDV and provided solid protection against virulent CDV challenge. On the contrast, rLa-CDVF induced much lower NA to CDV and fail to protected mink from virulent CDV challenge. Results suggest that recombinant NDV expressing CDV H is safe and efficient candidate vaccine against CDV in mink, and maybe other host species.
Collapse
Affiliation(s)
- Jinying Ge
- Key Laboratory of Veterinary Public Health of Ministry of Agriculture, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China.
| | - Xijun Wang
- Key Laboratory of Veterinary Public Health of Ministry of Agriculture, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Meijie Tian
- Key Laboratory of Veterinary Public Health of Ministry of Agriculture, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Yuwei Gao
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun 130122, China
| | - Zhiyuan Wen
- Key Laboratory of Veterinary Public Health of Ministry of Agriculture, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Guimei Yu
- Key Laboratory of Veterinary Public Health of Ministry of Agriculture, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Weiwei Zhou
- Key Laboratory of Veterinary Public Health of Ministry of Agriculture, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Shulong Zu
- Key Laboratory of Veterinary Public Health of Ministry of Agriculture, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Zhigao Bu
- Key Laboratory of Veterinary Public Health of Ministry of Agriculture, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China.
| |
Collapse
|
21
|
Wang FX, Zhang SQ, Zhu HW, Yang Y, Sun N, Tan B, Li ZG, Cheng SP, Fu ZF, Wen YJ. Recombinant rabies virus expressing the H protein of canine distemper virus protects dogs from the lethal distemper challenge. Vet Microbiol 2014; 174:362-371. [DOI: 10.1016/j.vetmic.2014.10.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 10/15/2014] [Accepted: 10/27/2014] [Indexed: 11/16/2022]
|
22
|
Putative domain-domain interactions in the vesicular stomatitis virus L polymerase protein appendage region. J Virol 2014; 88:14458-66. [PMID: 25297996 DOI: 10.1128/jvi.02267-14] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED The multidomain polymerase protein (L) of nonsegmented negative-strand (NNS) RNA viruses catalyzes transcription and replication of the virus genome. The N-terminal half of the protein forms a ring-like polymerase structure, while the C-terminal half encoding viral mRNA transcript modifications consists of a flexible appendage with three distinct globular domains. To gain insight into putative transient interactions between L domains during viral RNA synthesis, we exchanged each of the four distinct regions encompassing the appendage region of vesicular stomatitis virus (VSV) Indiana serotype L protein with their counterparts from VSV New Jersey and analyzed effects on virus polymerase activity in a minigenome system. The methyltransferase domain exchange yielded a fully active polymerase protein, which functioned as well as wild-type L in the context of a recombinant virus. Exchange of the downstream C-terminal nonconserved region abolished activity, but coexchanging it with the methyltransferase domain generated a polymerase favoring replicase over transcriptase activity, providing strong evidence of interaction between these two regions. Exchange of the capping enzyme domain or the adjacent nonconserved region thought to function as an "unstructured" linker also abrogated polymerase activity even when either domain was coexchanged with other appendage domains. Further probing of the putative linker segment using in-frame enhanced green fluorescent protein (EGFP) insertions similarly abrogated activity. We discuss the implications of these findings with regard to L protein appendage domain structure and putative domain-domain interactions required for polymerase function. IMPORTANCE NNS viruses include many well-known human pathogens (e.g., rabies, measles, and Ebola viruses), as well as emerging viral threats (e.g., Nipah and Hendra viruses). These viruses all encode a large L polymerase protein similarly organized into multiple domains that work in concert to enable virus genome transcription and replication. But how the unique L protein carries out the multiplicity of individual steps in these two distinct processes is poorly understood. Using two different approaches, i.e., exchanging individual domains in the C-terminal appendage region of the protein between two closely related VSV serotypes and inserting unrelated protein domains, we shed light on requirements for domain-domain interactions and domain contiguity in polymerase function. These findings further our understanding of the conformational dynamics of NNS L polymerase proteins, which play an essential role in the pathogenic properties of these viruses and represent attractive targets for the development of antiviral measures.
Collapse
|
23
|
In vitro anti-canine distemper virus activity of fucoidan extracted from the brown alga Cladosiphon okamuranus. Virusdisease 2014; 25:474-80. [PMID: 25674625 DOI: 10.1007/s13337-014-0228-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 09/10/2014] [Indexed: 10/24/2022] Open
Abstract
Canine distemper virus (CDV) is a morbillivirus related to measles virus that infects dogs and other carnivores. CDV has a significant global impact on animal health; however, there is no current antiviral treatment for CDV infection. In recent years, it has been demonstrated that sulfated polysaccharides exhibit antiviral properties both in vivo and in vitro, despite their low cytotoxicity to host cells. Fucoidan is a sulfated polysaccharide found in the cell wall matrix of brown algae. In this study, we evaluated in vitro anti-CDV activity of fucoidan, which was derived from Cladosiphon okamuranus. Fucoidan actively inhibited CDV replication in Vero cells at a 50 % inhibitory concentration (IC50) of 0.1 µg/ml. The derived selectivity index (SI50) was >20,000. This polysaccharide likely inhibits viral infection by interference in the early steps and by inhibiting CDV-mediated cell fusion. Fucoidan may be useful in development of pharmacological strategies to treat and control CDV infection.
Collapse
|
24
|
Buczkowski H, Muniraju M, Parida S, Banyard AC. Morbillivirus vaccines: recent successes and future hopes. Vaccine 2014; 32:3155-61. [PMID: 24703852 PMCID: PMC7115685 DOI: 10.1016/j.vaccine.2014.03.053] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 03/05/2014] [Accepted: 03/13/2014] [Indexed: 01/21/2023]
Abstract
Morbilliviruses cause severe disease in both human and animal populations. Morbilliviruses are recognised targets for eradication. Live attenuated vaccines are available for some morbilliviruses. DIVA vaccines may be important for future morbillivirus eradication attempts.
The impact of morbilliviruses on both human and animal populations is well documented in the history of mankind. Indeed, prior to the development of vaccines for these diseases, morbilliviruses plagued both humans and their livestock that were heavily relied upon for food and motor power within communities. Measles virus (MeV) was responsible for the death of millions of people annually across the world and those fortunate enough to escape the disease often faced starvation where their livestock had died following infection with rinderpest virus (RPV) or peste des petits ruminants virus (PPRV). Canine distemper virus has affected dog populations for centuries and in the past few decades appears to have jumped species, now causing disease in a number of non-canid species, some of which are been pushed to the brink of extinction by the virus. During the age of vaccination, the introduction and successful application of vaccines against rinderpest and measles has led to the eradication of the former and the greater control of the latter. Vaccines against PPR and canine distemper have also been generated; however, the diseases still pose a threat to susceptible species. Here we review the currently available vaccines against these four morbilliviruses and discuss the prospects for the development of new generation vaccines.
Collapse
Affiliation(s)
- Hubert Buczkowski
- Animal Health and Veterinary Laboratories Agency, Woodham Lane, Weybridge, Surrey, KT15 3NB, United Kingdom
| | - Murali Muniraju
- The Pirbright Institute, Ash Road, Pirbright, Surrey, GU24 0NF, United Kingdom
| | - Satya Parida
- The Pirbright Institute, Ash Road, Pirbright, Surrey, GU24 0NF, United Kingdom
| | - Ashley C Banyard
- Animal Health and Veterinary Laboratories Agency, Woodham Lane, Weybridge, Surrey, KT15 3NB, United Kingdom.
| |
Collapse
|
25
|
Falzarano D, Groseth A, Hoenen T. Development and application of reporter-expressing mononegaviruses: current challenges and perspectives. Antiviral Res 2014; 103:78-87. [PMID: 24462694 DOI: 10.1016/j.antiviral.2014.01.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 01/06/2014] [Accepted: 01/12/2014] [Indexed: 12/19/2022]
Abstract
Reverse genetics allows the generation of recombinant viruses entirely from cDNA. One application of this technology is the creation of reporter-expressing viruses, which greatly increase the detail and ease with which these viruses can be studied. However, there are a number of challenges when working with reporter-expressing viruses. Both the reporter protein itself as well as the genetic manipulations within the viral genome required for expression of this reporter can result in altered biological properties of the recombinant virus, and lead to attenuation in vitro and/or in vivo. Further, instability of reporter expression and purging of the genetic information encoding for the reporter from the viral genome can be an issue. Finally, a practical challenge for in vivo studies lies in the attenuation of light signals when traversing tissues. Novel expression strategies and the continued development of brighter, red and far-red shifted reporters and the increased use of bioluminescent reporters for in vivo applications promise to overcome some of these limitations in future. However, a "one size fits all" approach to the design of reporter-expressing viruses has thus far not been possible. Rather, a reporter suited to the intended application must be selected and an appropriate expression strategy and location for the reporter in the viral genome chosen. Still, attenuating effects of the reporter on viral fitness are difficult to predict and have to be carefully assessed with respect to the intended application. Despite these limitations the generation of suitable reporter-expressing viruses will become more common as technology and our understanding of the intricacies of viral gene expression and regulation improves, allowing deeper insight into virus biology both in living cells and in animals.
Collapse
Affiliation(s)
- Darryl Falzarano
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Allison Groseth
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Thomas Hoenen
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA.
| |
Collapse
|
26
|
Tien Nguyen D, Boes J, van Amerongen G, van Remmerden Y, Yüksel S, Guichelaar T, Osterhaus ADME, de Swart RL. Infection-enhancing lipopeptides do not improve intranasal immunization of cotton rats with a delta-G candidate live-attenuated human respiratory syncytial virus vaccine. Hum Vaccin Immunother 2013; 9:2578-83. [PMID: 23955280 DOI: 10.4161/hv.26096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Development of live-attenuated human respiratory syncytial virus (HRSV) vaccines has proven to be difficult. Several vaccine candidates were found to be over-attenuated and displayed limited immunogenicity. Recently, we identified three synthetic cationic lipopeptides that enhanced paramyxovirus infections in vitro. The infection enhancement proved to be mediated by enhanced virus binding to target cells. We hypothesized that these lipopeptides can be used as adjuvants to promote immune responses induced by live-attenuated paramyxovirus vaccines. This hypothesis was tested in a vaccination and challenge model in cotton rats, using a previously described recombinant live-attenuated candidate HRSV vaccine lacking the gene encoding the G glycoprotein (rHRSVΔG). Surprisingly, intranasal vaccination of cotton rats with rHRSVΔG formulated in infection-enhancing lipopeptides resulted in reduced virus loads in nasopharyngeal lavages, reduced seroconversion levels and reduced protection from wild-type HRSV challenge. In conclusion, we were unable to demonstrate the feasibility of lipopeptides as adjuvants for a candidate live-attenuated HRSV vaccine in the cotton rat model.
Collapse
Affiliation(s)
- D Tien Nguyen
- Department of Viroscience; Erasmus MC; Rotterdam, The Netherlands
| | - Jolande Boes
- National Institute of Public Health and the Environment; Bilthoven, The Netherlands
| | - Geert van Amerongen
- Department of Viroscience; Erasmus MC; Rotterdam, The Netherlands; National Institute of Public Health and the Environment; Bilthoven, The Netherlands
| | - Yvonne van Remmerden
- National Institute of Public Health and the Environment; Bilthoven, The Netherlands
| | - Selma Yüksel
- Department of Viroscience; Erasmus MC; Rotterdam, The Netherlands
| | - Teun Guichelaar
- National Institute of Public Health and the Environment; Bilthoven, The Netherlands
| | | | - Rik L de Swart
- Department of Viroscience; Erasmus MC; Rotterdam, The Netherlands
| |
Collapse
|
27
|
Wang X, Feng N, Ge J, Shuai L, Peng L, Gao Y, Yang S, Xia X, Bu Z. Recombinant canine distemper virus serves as bivalent live vaccine against rabies and canine distemper. Vaccine 2012; 30:5067-72. [DOI: 10.1016/j.vaccine.2012.06.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Revised: 05/26/2012] [Accepted: 06/01/2012] [Indexed: 12/25/2022]
|
28
|
Ludlow M, Nguyen DT, Silin D, Lyubomska O, de Vries RD, von Messling V, McQuaid S, De Swart RL, Duprex WP. Recombinant canine distemper virus strain Snyder Hill expressing green or red fluorescent proteins causes meningoencephalitis in the ferret. J Virol 2012; 86:7508-19. [PMID: 22553334 PMCID: PMC3416283 DOI: 10.1128/jvi.06725-11] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Accepted: 04/25/2012] [Indexed: 12/17/2022] Open
Abstract
The propensity of canine distemper virus (CDV) to spread to the central nervous system is one of the primary features of distemper. Therefore, we developed a reverse genetics system based on the neurovirulent Snyder Hill (SH) strain of CDV (CDV(SH)) and show that this virus rapidly circumvents the blood-brain and blood-cerebrospinal fluid (CSF) barriers to spread into the subarachnoid space to induce dramatic viral meningoencephalitis. The use of recombinant CDV(SH) (rCDV(SH)) expressing enhanced green fluorescent protein (EGFP) or red fluorescent protein (dTomato) facilitated the sensitive pathological assessment of routes of virus spread in vivo. Infection of ferrets with these viruses led to the full spectrum of clinical signs typically associated with distemper in dogs during a rapid, fatal disease course of approximately 2 weeks. Comparison with the ferret-adapted CDV(5804P) and the prototypic wild-type CDV(R252) showed that hematogenous infection of the choroid plexus is not a significant route of virus spread into the CSF. Instead, viral spread into the subarachnoid space in rCDV(SH)-infected animals was triggered by infection of vascular endothelial cells and the hematogenous spread of virus-infected leukocytes from meningeal blood vessels into the subarachnoid space. This resulted in widespread infection of cells of the pia and arachnoid mater of the leptomeninges over large areas of the cerebral hemispheres. The ability to sensitively assess the in vivo spread of a neurovirulent strain of CDV provides a novel model system to study the mechanisms of virus spread into the CSF and the pathogenesis of acute viral meningitis.
Collapse
Affiliation(s)
- M. Ludlow
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - D. T. Nguyen
- Department of Virology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - D. Silin
- School of Medicine, Dentistry and Biomedical Sciences, The Queen's University of Belfast, Belfast, Northern Ireland, United Kingdom
| | - O. Lyubomska
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - R. D. de Vries
- Department of Virology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - V. von Messling
- INRS—Institut Armand-Frappier, University of Quebec, Laval, QC, Canada
| | - S. McQuaid
- School of Medicine, Dentistry and Biomedical Sciences, The Queen's University of Belfast, Belfast, Northern Ireland, United Kingdom
- Tissue Pathology Laboratories, Belfast Health and Social Care Trust, Belfast, Northern Ireland, United Kingdom
| | - R. L. De Swart
- Department of Virology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - W. P. Duprex
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, USA
- School of Medicine, Dentistry and Biomedical Sciences, The Queen's University of Belfast, Belfast, Northern Ireland, United Kingdom
| |
Collapse
|
29
|
Nguyen DT, Ludlow M, van Amerongen G, de Vries RD, Yüksel S, Verburgh RJ, Osterhaus ADME, Duprex WP, de Swart RL. Evaluation of synthetic infection-enhancing lipopeptides as adjuvants for a live-attenuated canine distemper virus vaccine administered intra-nasally to ferrets. Vaccine 2012; 30:5073-80. [PMID: 22705079 DOI: 10.1016/j.vaccine.2012.05.079] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2012] [Revised: 05/11/2012] [Accepted: 05/30/2012] [Indexed: 11/19/2022]
Abstract
BACKGROUND Inactivated paramyxovirus vaccines have been associated with hypersensitivity responses upon challenge infection. For measles and canine distemper virus (CDV) safe and effective live-attenuated virus vaccines are available, but for human respiratory syncytial virus and human metapneumovirus development of such vaccines has proven difficult. We recently identified three synthetic bacterial lipopeptides that enhance paramyxovirus infections in vitro, and hypothesized these could be used as adjuvants to promote immune responses induced by live-attenuated paramyxovirus vaccines. METHODS Here, we tested this hypothesis using a CDV vaccination and challenge model in ferrets. Three groups of six animals were intra-nasally vaccinated with recombinant (r) CDV(5804P)L(CCEGFPC) in the presence or absence of the infection-enhancing lipopeptides Pam3CSK4 or PHCSK4. The recombinant CDV vaccine virus had previously been described to be over-attenuated in ferrets. A group of six animals was mock-vaccinated as control. Six weeks after vaccination all animals were challenged with a lethal dose of rCDV strain Snyder-Hill expressing the red fluorescent protein dTomato. RESULTS Unexpectedly, intra-nasal vaccination of ferrets with rCDV(5804P)L(CCEGFPC) in the absence of lipopeptides resulted in good immune responses and protection against lethal challenge infection. However, in animals vaccinated with lipopeptide-adjuvanted virus significantly higher vaccine virus loads were detected in nasopharyngeal lavages and peripheral blood mononuclear cells. In addition, these animals developed significantly higher CDV neutralizing antibody titers compared to animals vaccinated with non-adjuvanted vaccine. CONCLUSIONS This study demonstrates that the synthetic cationic lipopeptides Pam3CSK4 and PHCSK4 not only enhance paramyxovirus infection in vitro, but also in vivo. Given the observed enhancement of immunogenicity their potential as adjuvants for other live-attenuated paramyxovirus vaccines should be considered.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Administration, Intranasal
- Animals
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Chlorocebus aethiops
- Distemper/immunology
- Distemper/prevention & control
- Distemper Virus, Canine/immunology
- Distemper Virus, Canine/pathogenicity
- Drug Evaluation, Preclinical
- Female
- Ferrets/immunology
- Ferrets/virology
- Lipopeptides/administration & dosage
- Lymphocytes/immunology
- Lymphocytes/virology
- Neutralization Tests/methods
- Transfection
- Vaccines, Attenuated/administration & dosage
- Vaccines, Attenuated/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/immunology
- Vero Cells
- Viral Load
- Viral Vaccines/administration & dosage
- Viral Vaccines/immunology
Collapse
Affiliation(s)
- D Tien Nguyen
- Department of Virology, Erasmus MC, University Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
The 200-kDa L protein of arenaviruses plays a central role in viral genome replication and transcription. This study aimed at providing evidence for the domain structure of L protein by combining bioinformatics with a stepwise mutagenesis approach using the Lassa virus minireplicon system. Potential interdomain linkers were predicted using various algorithms. The prediction was challenged by insertion of flexible sequences into the predicted linkers. Insertion of 5 or 10 amino acid residues was tolerated at seven sites (S407, G446, G467, G774, G939, S1952, and V2074 in Lassa virus AV). At two of these sites, G467 and G939, L protein could be split into an N-terminal and a C-terminal part, which were able to trans-complement each other and reconstitute a functional complex upon coexpression. Coimmunoprecipitation studies revealed physical interaction between the N- and C-terminal domains, irrespective of whether L protein was split at G467 or G939. In confocal immunofluorescence microscopy, the N-terminal domains showed a dot-like, sometimes perinuclear, cytoplasmic distribution similar to that of full-length L protein, while the C-terminal domains were homogenously distributed in cytoplasm. The latter were redistributed into the dot-like structures upon coexpression with the corresponding N-terminal domain. In conclusion, this study demonstrates two interdomain linkers in Lassa virus L protein, at G467 and G939, suggesting that L protein is composed of at least three structural domains spanning residues 1 to 467, 467 to 939, and 939 to 2220. The first domain seems to mediate accumulation of L protein into cytoplasmic dot-like structures.
Collapse
|
31
|
Banyard AC, Simpson J, Monaghan P, Barrett T. Rinderpest virus expressing enhanced green fluorescent protein as a separate transcription unit retains pathogenicity for cattle. J Gen Virol 2010; 91:2918-27. [DOI: 10.1099/vir.0.023598-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
32
|
Insertion of enhanced green fluorescent protein in a hinge region of vesicular stomatitis virus L polymerase protein creates a temperature-sensitive virus that displays no virion-associated polymerase activity in vitro. J Virol 2009; 83:12241-52. [PMID: 19793815 DOI: 10.1128/jvi.01273-09] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The RNA-dependent RNA polymerase of viruses belonging to the order Mononegavirales is part of a large multifunctional L protein that also catalyzes viral mRNA capping and cap methylation. The L protein of this diverse group of agents displays six blocks of conserved sequences. The precise relationship between these conserved regions and individual functions is largely unknown, except for "domain" VI that clearly encodes a viral mRNA cap methylase. The L protein of morbilliviruses (family Paramyxoviridae) was reported to tolerate insertion of the enhanced green fluorescent protein (EGFP) in a region just upstream of domain VI. Recombinant viruses with this insertion grow well in cell culture but are highly attenuated in animal hosts. We show here that the L protein of vesicular stomatitis virus (VSV), the prototype of the Rhabdoviridae family, also tolerates insertion of EGFP at a similar site. The modified protein (L(EGFP)) and the resultant recombinant virus both demonstrated a sharp temperature-sensitive phenotype for polymerase activity, with reduced activity at 37 degrees C and no activity at 37.5 degrees C. Neither translation nor methylation of mutant virus transcripts was affected at 37 degrees C. Curiously, mutant virus grown at permissive temperature contained about threefold-less L protein than the wild-type virus did and displayed no virion-associated polymerase activity in vitro. These findings support the notion that a flexible "hinge" region separates the cap methylase domain of L proteins from upstream functions and open up a number of avenues for studies of L-protein function in the more-tractable VSV model system.
Collapse
|
33
|
Young DF, Galiano MC, Lemon K, Chen YH, Andrejeva J, Duprex WP, Rima BK, Randall RE. Mumps virus Enders strain is sensitive to interferon (IFN) despite encoding a functional IFN antagonist. J Gen Virol 2009; 90:2731-2738. [PMID: 19625458 PMCID: PMC2885035 DOI: 10.1099/vir.0.013722-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Although the Enders strain of mumps virus (MuV) encodes a functional V protein that acts as an interferon (IFN) antagonist, in multi-cycle growth assays MuV Enders grew poorly in naïve ('IFN-competent' Hep2) cells but grew to high titres in 'IFN-compromised' Hep2 cells. Even so, the growth rate of MuV Enders was significantly slower in 'IFN-compromised' Hep2 cells when compared with its replication rate in Vero cells and with the replication rate of parainfluenza virus type 5 (a closely related paramyxovirus) in both naïve and 'IFN-compromised' Hep2 cells. This suggests that a consequence of slower growth is that the IFN system of naïve Hep2 cells can respond quickly enough to control the growth of MuV Enders. This is supported by the finding that rapidly growing variants of MuV Enders that were selected on 'IFN-compromised' Hep2 cells (i.e. in the absence of any selection pressure exerted by the IFN response) also grew to high titres on naïve Hep2 cells. Sequencing of the complete genome of one of these variants identified a single point mutation that resulted in a substitution of a conserved asparagine by histidine at position 498 of the haemagglutinin-neuraminidase protein, although this mutation was not present in all rapidly growing variants. These results support the concept that there is a race between the ability of a cell to detect and respond to virus infection and the ability of a virus to block the IFN response. Importantly, this emphasizes that factors other than viral IFN antagonists influence the sensitivity of viruses to IFN.
Collapse
Affiliation(s)
- D F Young
- Centre for Biomolecular Sciences, University of St Andrews, St Andrews, Fife KY16 9ST, UK
| | - M C Galiano
- Centre for Biomolecular Sciences, University of St Andrews, St Andrews, Fife KY16 9ST, UK
| | - K Lemon
- Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Sciences, The Queen's University of Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Y-H Chen
- Centre for Biomolecular Sciences, University of St Andrews, St Andrews, Fife KY16 9ST, UK
| | - J Andrejeva
- Centre for Biomolecular Sciences, University of St Andrews, St Andrews, Fife KY16 9ST, UK
| | - W P Duprex
- Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Sciences, The Queen's University of Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - B K Rima
- Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Sciences, The Queen's University of Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - R E Randall
- Centre for Biomolecular Sciences, University of St Andrews, St Andrews, Fife KY16 9ST, UK
| |
Collapse
|
34
|
Shi X, Elliott RM. Generation and analysis of recombinant Bunyamwera orthobunyaviruses expressing V5 epitope-tagged L proteins. J Gen Virol 2009; 90:297-306. [PMID: 19141438 PMCID: PMC2885054 DOI: 10.1099/vir.0.007567-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The L protein of Bunyamwera virus (BUNV; family Bunyaviridae) is an RNA-dependent RNA polymerase, 2238 aa in length, that catalyses transcription and replication of the negative-sense, tripartite RNA genome. To learn more about the molecular interactions of the L protein and to monitor its intracellular distribution we inserted a 14 aa V5 epitope derived from parainfluenza virus type 5, against which high-affinity antibodies are available, into different regions of the protein. Insertion of the epitope at positions 1935 or 2046 resulted in recombinant L proteins that retained functionality in a minireplicon assay. Two viable recombinant viruses, rBUNL4V5 and rBUNL5V5, expressing the tagged L protein were rescued by reverse genetics, and characterized with respect to their plaque size, growth kinetics and protein synthesis profile. The recombinant viruses behaved similarly to wild-type (wt) BUNV in BHK-21 cells, but formed smaller plaques and grew to lower titres in Vero E6 cells compared with wt BUNV. Immunofluorescent staining of infected cells showed the L protein to have a punctate to reticular distribution in the cytoplasm, and cell fractionation studies indicated that the L protein was present in both soluble and microsomal fractions. Co-immunoprecipitation and confocal microscopic assays confirmed an interaction between BUNV L and N proteins. The recombinant viruses expressing tagged L protein will be highly valuable reagents for the detailed dissection of the role of the BUNV L protein in virus replication.
Collapse
Affiliation(s)
- Xiaohong Shi
- Centre for Biomolecular Sciences, School of Biology, University of St Andrews, North Haugh, St Andrews, Scotland KY16 9ST, UK
| | - Richard M Elliott
- Centre for Biomolecular Sciences, School of Biology, University of St Andrews, North Haugh, St Andrews, Scotland KY16 9ST, UK
| |
Collapse
|
35
|
Capraro GA, Johnson JB, Kock ND, Parks GD. Virus growth and antibody responses following respiratory tract infection of ferrets and mice with WT and P/V mutants of the paramyxovirus Simian Virus 5. Virology 2008; 376:416-28. [PMID: 18456301 PMCID: PMC2574746 DOI: 10.1016/j.virol.2008.03.034] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2008] [Revised: 03/19/2008] [Accepted: 03/27/2008] [Indexed: 11/29/2022]
Abstract
P/V gene substitutions convert the non-cytopathic paramyxovirus Simian Virus 5 (SV5), which is a poor inducer of host cell responses in human tissue culture cells, into a mutant (P/V-CPI−) that induces high levels of apoptosis, interferon (IFN)-beta, and proinflammatory cytokines. However, the effect of SV5-P/V gene mutations on virus growth and adaptive immune responses in animals has not been determined. Here, we used two distinct animal model systems to test the hypothesis that SV5-P/V mutants which are more potent activators of innate responses in tissue culture will also elicit higher antiviral antibody responses. In mouse cells, in vitro studies identified a panel of SV5-P/V mutants that ranged in their ability to limit IFN responses. Intranasal infection of mice with these WT and P/V mutant viruses elicited equivalent anti-SV5 IgG responses at all doses tested, and viral titers recovered from the respiratory tract were indistinguishable. In primary cultures of ferret lung fibroblasts, WT rSV5 and P/V-CPI− viruses had phenotypes similar to those established in human cell lines, including differential induction of IFN secretion, IFN signaling and apoptosis. Intranasal infection of ferrets with a low dose of WT rSV5 elicited ~ 500 fold higher anti-SV5 serum IgG responses compared to the P/V-CPI− mutant, and this correlated with overall higher viral titers for the WT virus in tracheal tissues. There was a dose-dependent increase in antibody response to infection of ferrets with P/V-CPI−, but not with WT rSV5. Together our data indicate that WT rSV5 and P/V mutants can elicit distinct innate and adaptive immunity phenotypes in the ferret animal model system, but not in the mouse system. We present a model for the effect of P/V gene substitutions on SV5 growth and immune responses in vivo.
Collapse
Affiliation(s)
- Gerald A Capraro
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157-1064, USA
| | | | | | | |
Collapse
|
36
|
Chapter 4 Receptor Interactions, Tropism, and Mechanisms Involved in Morbillivirus‐Induced Immunomodulation. Adv Virus Res 2008; 71:173-205. [DOI: 10.1016/s0065-3527(08)00004-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|