1
|
Wu F, Deng Y, Yao X, Li J. Ruminant livestock TR V(D)J genes and CDR3 repertoire. Vet Immunol Immunopathol 2024; 277:110829. [PMID: 39316948 DOI: 10.1016/j.vetimm.2024.110829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 09/26/2024]
Abstract
Ruminant livestock exhibit certain immune characteristics that make them valuable models for studying T cell receptor diversity and immune responses. This resistance is attributed to their well-developed immune system, comprising both innate and adaptive components. In this review, we delve into the intricate workings of the immune system of ruminant livestock, focusing on innate immunity and adaptive immunity. Specifically, we discuss the TR V(D)J genes (including TRB, TRG, and TRA/D chain) and the characteristics of the complementary determining region 3 (CDR3) repertoire in bovine and ovine species, shedding light on the diversity and functionality of the T-cell receptor(TCR) repertoire in these species. Understanding the distinct features of these germline genes and CDR3 repertoires is essential for unraveling the complexities of immune responses in ruminant livestock. Lastly, we outline future prospects in this field, emphasizing the importance of further research to enhance our understanding of ruminant livestock immunity and its potential applications in disease management, vaccine development, and breeding strategies.
Collapse
Affiliation(s)
- Fengli Wu
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi, China; Department of Laboratory, The Affiliated Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Yunlan Deng
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi, China
| | - Xinsheng Yao
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi, China
| | - Jun Li
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi, China.
| |
Collapse
|
2
|
Jiménez-Cabello L, Utrilla-Trigo S, Calvo-Pinilla E, Lorenzo G, Illescas-Amo M, Benavides J, Moreno S, Marín-López A, Nogales A, Ortego J. Co-expression of VP2, NS1 and NS2-Nt proteins by an MVA viral vector induces complete protection against bluetongue virus. Front Immunol 2024; 15:1440407. [PMID: 39072326 PMCID: PMC11272488 DOI: 10.3389/fimmu.2024.1440407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 06/28/2024] [Indexed: 07/30/2024] Open
Abstract
Introduction Bluetongue (BT), caused by bluetongue virus (BTV), is an important arthropod-borne livestock disease listed by the World Organization for Animal Health. Live-attenuated and inactivated vaccines have permitted to control BT but they do not simultaneously protect against the myriad of BTV serotypes. Recently, we identified the highly conserved BTV nonstructural protein NS1 and the N-terminal region of NS2 as antigens capable of conferring multiserotype protection against BTV. Methods Here, we designed Modified Vaccinia Ankara (MVA) viral vectors that expressed BTV-4 proteins VP2 or VP7 along with NS1 and NS2-Nt as well as MVAs that expressed proteins VP2, VP7 or NS1 and NS2-Nt. Results Immunization of IFNAR(-/-) mice with two doses of MVA-NS1-2A-NS2-Nt protected mice from BTV-4M infection by the induction of an antigen-specific T cell immune response. Despite rMVA expressing VP7 alone were not protective in the IFNAR(-/-) mouse model, inclusion of VP7 in the vaccine formulation amplified the cell-mediated response induced by NS1 and NS2-Nt. Expression of VP2 elicited protective non-cross-reactive neutralizing antibodies (nAbs) in immunized animals and improved the protection observed in the MVA-NS1-2A-NS2-Nt immunized mice when these three BTV antigens were co-expressed. Moreover, vaccines candidates co-expressing VP2 or VP7 along with NS1 and NS2-Nt provided multiserotype protection. We assessed protective efficacy of both vaccine candidates in sheep against virulent challenge with BTV-4M. Discussion Immunization with MVA-VP7-NS1-2A-NS2-Nt partially dumped viral replication and clinical disease whereas administration of MVA-VP2-NS1-2A-NS2-Nt promoted a complete protection, preventing viraemia and the pathology produced by BTV infection.
Collapse
Affiliation(s)
- Luis Jiménez-Cabello
- Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Madrid, Spain
| | - Sergio Utrilla-Trigo
- Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Madrid, Spain
| | - Eva Calvo-Pinilla
- Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Madrid, Spain
| | - Gema Lorenzo
- Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Madrid, Spain
| | - Miguel Illescas-Amo
- Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Madrid, Spain
| | - Julio Benavides
- Instituto de Ganadería de Montaña, CSIC-Universidad de León, León, Spain
| | - Sandra Moreno
- Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Madrid, Spain
| | - Alejandro Marín-López
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Aitor Nogales
- Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Madrid, Spain
| | - Javier Ortego
- Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Madrid, Spain
| |
Collapse
|
3
|
Kolla HB, Dutt M, Kumar A, Hebbandi Nanjunadappa R, Karakach T, Singh KP, Kelvin D, Clement Mertens PP, Umeshappa CS. Immuno-informatics study identifies conserved T cell epitopes in non-structural proteins of Bluetongue virus serotypes: formulation of a computationally optimized next-generation broad-spectrum multi-epitope vaccine. Front Immunol 2024; 15:1424307. [PMID: 39011043 PMCID: PMC11246920 DOI: 10.3389/fimmu.2024.1424307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 06/13/2024] [Indexed: 07/17/2024] Open
Abstract
Introduction Bluetongue (BT) poses a significant threat to the livestock industry, affecting various animal species and resulting in substantial economic losses. The existence of numerous BT virus (BTV) serotypes has hindered control efforts, highlighting the need for broad-spectrum vaccines. Methodology In this study, we evaluated the conserved amino acid sequences within key non-structural (NS) proteins of BTV and identified numerous highly conserved murine- and bovine-specific MHC class I-restricted (MHC-I) CD8+ and MHC-II-restricted CD4+ epitopes. We then screened these conserved epitopes for antigenicity, allergenicity, toxicity, and solubility. Using these epitopes, we developed in silico-based broad-spectrum multiepitope vaccines with Toll-like receptor (TLR-4) agonists. The predicted proinflammatory cytokine response was assessed in silico using the C-IMMSIM server. Structural modeling and refinement were achieved using Robetta and GalaxyWEB servers. Finally, we assessed the stability of the docking complexes through extensive 100-nanosecond molecular dynamics simulations before considering the vaccines for codon optimization and in silico cloning. Results We found many epitopes that meet these criteria within NS1 and NS2 proteins and developed in silico broad-spectrum vaccines. The immune simulation studies revealed that these vaccines induce high levels of IFN-γ and IL-2 in the vaccinated groups. Protein-protein docking analysis demonstrated promising epitopes with strong binding affinities to TLR-4. The docked complexes were stable, with minimal Root Mean Square Deviation and Root Mean Square Fluctuation values. Finally, the in silico-cloned plasmids have high % of GC content with > 0.8 codon adaptation index, suggesting they are suitable for expressing the protein vaccines in prokaryotic system. Discussion These next-generation vaccine designs are promising and warrant further investigation in wet lab experiments to assess their immunogenicity, safety, and efficacy for practical application in livestock. Our findings offer a robust framework for developing a comprehensive, broad-spectrum vaccine, potentially revolutionizing BT control and prevention strategies in the livestock industry.
Collapse
Affiliation(s)
- Harish Babu Kolla
- Department of Microbiology, Immunology and Pediatrics, Dalhousie University, Halifax, NS, Canada
- Immunology Division, IWK Health Centre, Halifax, NS, Canada
| | - Mansi Dutt
- Department of Microbiology, Immunology and Pediatrics, Dalhousie University, Halifax, NS, Canada
- Immunology Division, IWK Health Centre, Halifax, NS, Canada
| | - Anuj Kumar
- Department of Microbiology, Immunology and Pediatrics, Dalhousie University, Halifax, NS, Canada
- Immunology Division, IWK Health Centre, Halifax, NS, Canada
| | - Roopa Hebbandi Nanjunadappa
- Department of Microbiology, Immunology and Pediatrics, Dalhousie University, Halifax, NS, Canada
- Immunology Division, IWK Health Centre, Halifax, NS, Canada
| | - Tobias Karakach
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| | - Karam Pal Singh
- Center for Animal Disease Research and Diagnosis, Indian Veterinary Research Institute, Bareilly, India
| | - David Kelvin
- Department of Microbiology, Immunology and Pediatrics, Dalhousie University, Halifax, NS, Canada
- Immunology Division, IWK Health Centre, Halifax, NS, Canada
| | | | - Channakeshava Sokke Umeshappa
- Department of Microbiology, Immunology and Pediatrics, Dalhousie University, Halifax, NS, Canada
- Immunology Division, IWK Health Centre, Halifax, NS, Canada
| |
Collapse
|
4
|
Newbrook K, Khan N, Fisher A, Chong K, Gubbins S, Davies WC, Sanders C, Busquets MG, Cooke L, Corla A, Ashby M, Flannery J, Batten C, Stokes JE, Sanz-Bernardo B, Carpenter S, Moffat K, Darpel KE. Specific T-cell subsets have a role in anti-viral immunity and pathogenesis but not viral dynamics or onwards vector transmission of an important livestock arbovirus. Front Immunol 2024; 15:1328820. [PMID: 38357545 PMCID: PMC10864546 DOI: 10.3389/fimmu.2024.1328820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 01/08/2024] [Indexed: 02/16/2024] Open
Abstract
Introduction Bluetongue virus (BTV) is an arthropod-borne Orbivirus that is almost solely transmitted by Culicoides biting midges and causes a globally important haemorrhagic disease, bluetongue (BT), in susceptible ruminants. Infection with BTV is characterised by immunosuppression and substantial lymphopenia at peak viraemia in the host. Methods In this study, the role of cell-mediated immunity and specific T-cell subsets in BTV pathogenesis, clinical outcome, viral dynamics, immune protection, and onwards transmission to a susceptible Culicoides vector is defined in unprecedented detail for the first time, using an in vivo arboviral infection model system that closely mirrors natural infection and transmission of BTV. Individual circulating CD4+, CD8+, or WC1+ γδ T-cell subsets in sheep were depleted through the administration of specific monoclonal antibodies. Results The absence of cytotoxic CD8+ T cells was consistently associated with less severe clinical signs of BT, whilst the absence of CD4+ and WC1+ γδ T cells both resulted in an increased clinical severity. The absence of CD4+ T cells also impaired both a timely protective neutralising antibody response and the production of IgG antibodies targeting BTV non-structural protein, NS2, highlighting that the CD4+ T-cell subset is important for a timely protective immune response. T cells did not influence viral replication characteristics, including onset/dynamics of viraemia, shedding, or onwards transmission of BTV to Culicoides. We also highlight differences in T-cell dependency for the generation of immunoglobulin subclasses targeting BTV NS2 and the structural protein, VP7. Discussion This study identifies a diverse repertoire of T-cell functions during BTV infection in sheep, particularly in inducing specific anti-viral immune responses and disease manifestation, and will support more effective vaccination strategies.
Collapse
Affiliation(s)
- Kerry Newbrook
- Orbivirus Research, The Pirbright Institute, Woking, United Kingdom
| | - Nakibul Khan
- Orbivirus Research, The Pirbright Institute, Woking, United Kingdom
- Department of Biology, University of York, York, United Kingdom
| | - Aimee Fisher
- Orbivirus Research, The Pirbright Institute, Woking, United Kingdom
- School of Biosciences AND School of Veterinary Medicine, University of Surrey, Guildford, United Kingdom
| | - Karen Chong
- Orbivirus Research, The Pirbright Institute, Woking, United Kingdom
- School of Biosciences AND School of Veterinary Medicine, University of Surrey, Guildford, United Kingdom
| | - Simon Gubbins
- Transmission Biology, The Pirbright Institute, Woking, United Kingdom
| | - William C. Davies
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, United States
| | | | | | - Lyndsay Cooke
- Orbivirus Research, The Pirbright Institute, Woking, United Kingdom
| | - Amanda Corla
- Non Vesicular Reference Laboratory, The Pirbright Institute, Woking, United Kingdom
| | - Martin Ashby
- Non Vesicular Reference Laboratory, The Pirbright Institute, Woking, United Kingdom
| | - John Flannery
- Non Vesicular Reference Laboratory, The Pirbright Institute, Woking, United Kingdom
| | - Carrie Batten
- Non Vesicular Reference Laboratory, The Pirbright Institute, Woking, United Kingdom
| | | | - Beatriz Sanz-Bernardo
- Large Deoxyribonucleic Acid (DNA), Viruses, The Pirbright Institute, Woking, United Kingdom
| | | | - Katy Moffat
- Flow Cytometry, The Pirbright Institute, Woking, United Kingdom
| | - Karin E. Darpel
- Orbivirus Research, The Pirbright Institute, Woking, United Kingdom
- Department of Diagnostics and Development, Institute of Virology and Immunology, Mittelhäusern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
5
|
Rodríguez-Martín D, Louloudes-Lázaro A, Avia M, Martín V, Rojas JM, Sevilla N. The Interplay between Bluetongue Virus Infections and Adaptive Immunity. Viruses 2021; 13:1511. [PMID: 34452376 PMCID: PMC8402766 DOI: 10.3390/v13081511] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 07/19/2021] [Accepted: 07/28/2021] [Indexed: 12/18/2022] Open
Abstract
Viral infections have long provided a platform to understand the workings of immunity. For instance, great strides towards defining basic immunology concepts, such as MHC restriction of antigen presentation or T-cell memory development and maintenance, have been achieved thanks to the study of lymphocytic choriomeningitis virus (LCMV) infections. These studies have also shaped our understanding of antiviral immunity, and in particular T-cell responses. In the present review, we discuss how bluetongue virus (BTV), an economically important arbovirus from the Reoviridae family that affects ruminants, affects adaptive immunity in the natural hosts. During the initial stages of infection, BTV triggers leucopenia in the hosts. The host then mounts an adaptive immune response that controls the disease. In this work, we discuss how BTV triggers CD8+ T-cell expansion and neutralizing antibody responses, yet in some individuals viremia remains detectable after these adaptive immune mechanisms are active. We present some unpublished data showing that BTV infection also affects other T cell populations such as CD4+ T-cells or γδ T-cells, as well as B-cell numbers in the periphery. This review also discusses how BTV evades these adaptive immune mechanisms so that it can be transmitted back to the arthropod host. Understanding the interaction of BTV with immunity could ultimately define the correlates of protection with immune mechanisms that would improve our knowledge of ruminant immunology.
Collapse
Affiliation(s)
| | | | | | | | | | - Noemí Sevilla
- Centro de Investigación en Sanidad Animal, Centro Nacional Instituto de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas (CISA-INIA-CSIC), Valdeolmos, 28130 Madrid, Spain; (D.R.-M.); (A.L.-L.); (M.A.); (V.M.); (J.M.R.)
| |
Collapse
|
6
|
Rojas JM, Barba-Moreno D, Avia M, Sevilla N, Martín V. Vaccination With Recombinant Adenoviruses Expressing the Bluetongue Virus Subunits VP7 and VP2 Provides Protection Against Heterologous Virus Challenge. Front Vet Sci 2021; 8:645561. [PMID: 33778041 PMCID: PMC7987666 DOI: 10.3389/fvets.2021.645561] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 02/05/2021] [Indexed: 11/25/2022] Open
Abstract
Bluetongue virus (BTV) is the causative agent of a disease that affects domestic and wild ruminants and leads to critical economic losses. BTV is an arbovirus from the Reoviridae family that is typically transmitted by the bite of infected Culicoides midges. BTV possesses multiple serotypes (up to 28 have been described), and immunity to one serotype offers little cross-protection to other serotypes. The design of vaccines that provide protection across multiple serotypes is therefore highly desirable to control this disease. We previously reported that a recombinant replication-defective human adenovirus serotype 5 (Ad5) that expresses the VP7 inner core protein of BTV serotype 8 (Ad5VP7-8) induced T-cell responses and provided protection. In the present work, we evaluated as BTV vaccine the combination of Ad5VP7-8 with another recombinant Ad5 that expresses the outer core protein VP2 from BTV-1 (Ad5VP2-1). The combination of Ad5VP2-1 and Ad5VP7-8 protected against homologous BTV challenge (BTV-1 and BTV-8) and partially against heterologous BTV-4 in a murine model. Cross-reactive anti-BTV immunoglobulin G (IgG) were detected in immunized animals, but no significant titers of neutralizing antibodies were elicited. The Ad5VP7-8 immunization induced T-cell responses that recognized all three serotypes tested in this study and primed cytotoxic T lymphocytes specific for VP7. This study further confirms that targeting antigenic determinant shared by several BTV serotypes using cellular immunity could help develop multiserotype BTV vaccines.
Collapse
Affiliation(s)
- José Manuel Rojas
- Centro de Investigación en Sanidad Animal (CISA-INIA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Madrid, Spain
| | - Diego Barba-Moreno
- Centro de Investigación en Sanidad Animal (CISA-INIA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Madrid, Spain
| | - Miguel Avia
- Centro de Investigación en Sanidad Animal (CISA-INIA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Madrid, Spain
| | - Noemí Sevilla
- Centro de Investigación en Sanidad Animal (CISA-INIA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Madrid, Spain
| | - Verónica Martín
- Centro de Investigación en Sanidad Animal (CISA-INIA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Madrid, Spain
| |
Collapse
|
7
|
Activation of OX40 and CD27 Costimulatory Signalling in Sheep through Recombinant Ovine Ligands. Vaccines (Basel) 2020; 8:vaccines8020333. [PMID: 32580486 PMCID: PMC7350415 DOI: 10.3390/vaccines8020333] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 06/08/2020] [Accepted: 06/18/2020] [Indexed: 01/22/2023] Open
Abstract
Members of the tumour necrosis factor (TNF) superfamily OX40L and CD70 and their receptors are costimulating signalling axes critical for adequate T cell activation in humans and mice but characterisation of these molecules in other species including ruminants is lacking. Here we cloned and expressed the predicted ovine orthologues of the receptors OX40 and CD27, as well as soluble recombinant forms of their potential ovine ligands, OaOX40L and OaCD70. Using biochemical and immunofluorescence analyses, we show that both signalling axes are functional in sheep. We show that oligomeric recombinant ligand constructs are able to induce signalling through their receptors on transfected cells. Recombinant defective human adenoviruses were constructed to express the soluble forms of OaOX40L and OaCD70. Both proteins were detected in the supernatant of adenovirus-infected cells and shown to activate NF-κB signalling pathway through their cognate receptor. These adenovirus-secreted OaOX40L and OaCD70 forms could also activate ovine T cell proliferation and enhance IFN-γ production in CD4+ and CD8+ T cells. Altogether, this study provides the first characterisation of the ovine costimulatory OX40L-OX40 and CD70-CD27 signalling axes, and indicates that their activation in vivo may be useful to enhance vaccination-induced immune responses in sheep and other ruminants.
Collapse
|
8
|
Yang JL, Yen LHC, Yen WCW, Wang FI. A SUBCLINICAL BLUETONGUE VIRUS INFECTION IN RUMINANTS WITH THREE UNIQUE AMINO ACID VARIATIONS ON VP7 CORE PROTEIN OF TAIWAN ISOLATES. ACTA ACUST UNITED AC 2019. [DOI: 10.1142/s168264851950001x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Bluetongue is an arthropod-borne disease in domestic and wild ruminants caused by bluetongue virus (BTV), and it leads to great economic loss worldwide. Previous studies showed that BTV in ruminants in Taiwan was often subclinical infection. The aim of this study was to determine the current status (years 2016–2017) of BTV infection in ruminants in Taiwan, to compare it to the results of a large-scale study conducted in the year 2003, and to investigate whether new viral strains exist. Competitive ELISA tests of serum samples for anti-BTV-VP7 group-specific antibody revealed seropositive rates of 26.7% in cattle by head, similar to 32.7% in the year 2003, suggestive of a BTV-vector-host (cattle) dynamic balance. In goats, the seropositive rate was 18.6%, slightly increased from 8.2% in the year 2003, suggestive of a slow but active infection taking place. This notion was supported by the detection of VP1 gene nucleic acid from whole blood in six out of 29 seropositive goats by reverse transcription–polymerase chain reaction. However, no new virus strain was isolated from embryonating chicken embryos (ECEs) inoculation. Alignment of VP7 amino acid sequences revealed that Taiwan and Japan isolates possessed three specific amino acids on sites No. 82 (arginine), No. 328 (aspartate), and No. 336 (glutamine), which are different from many countries. In a three-dimensional model, these amino acids were located closely on the middle lateral surface of VP7 trimers. Since VP7 is a major outer protein engaged in entry into insect cells and a strong T cell response inducer, these differences likely indicate the result of positive selection of local vectors and hosts in Taiwan.
Collapse
Affiliation(s)
- Jia-Ling Yang
- School of Veterinary Medicine, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 10617, Taiwan, R. O. C
| | - Lenny Hao-Che Yen
- School of Veterinary Medicine, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 10617, Taiwan, R. O. C
| | - Well Chia-Wei Yen
- School of Veterinary Medicine, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 10617, Taiwan, R. O. C
| | - Fun-In Wang
- School of Veterinary Medicine, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 10617, Taiwan, R. O. C
| |
Collapse
|
9
|
Martinelle L, Dal Pozzo F, Thys C, De Leeuw I, Van Campe W, De Clercq K, Thiry E, Saegerman C. Assessment of cross-protection induced by a bluetongue virus (BTV) serotype 8 vaccine towards other BTV serotypes in experimental conditions. Vet Res 2018; 49:63. [PMID: 30012223 PMCID: PMC6048908 DOI: 10.1186/s13567-018-0556-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 06/25/2018] [Indexed: 02/06/2023] Open
Abstract
Bluetongue disease is caused by bluetongue virus (BTV) and BTV serotype 8 (BTV8) caused great economic damage in Europe during the last decade. From 1998 to 2007, in addition to BTV8, Europe had to face the emergence of BTV1, 2, 4, 9, and 16, spreading in countries where the virus has never been detected before. These unprecedented outbreaks trigger the need to evaluate and compare the clinical, virological and serological features of the European BTV serotypes in the local epidemiological context. In this study groups of calves were infected with one of the following European BTV serotypes, namely BTV1, 2, 4, 9 and 16. For each tested serotype, two groups of three male Holstein calves were used: one group vaccinated against BTV8, the other non-vaccinated. Clinical signs were quantified, viral RNA was detected in blood and organs and serological relationship was assessed. Calves were euthanized 35 days post-infection and necropsied. Most of the infected animals showed mild clinical signs. A partial serological cross reactivity has been reported between BTV8 and BTV4, and between BTV1 and BTV8. BTV2 and BTV4 viral RNA only reached low levels in blood, when compared to other serotypes, whereas in vitro growth assays could not highlight significant differences. Altogether the results of this study support the hypothesis of higher adaptation of some BTV strains to specific hosts, in this case calves. Furthermore, cross-protection resulting from a prior vaccination with BTV8 was highlighted based on cross-neutralization. However, the development of neutralizing antibodies is probably not totally explaining the mild protection induced by the heterologous vaccination.
Collapse
Affiliation(s)
- Ludovic Martinelle
- Faculty of Veterinary Medicine, Research Unit in Epidemiology and Risk Analysis Applied to Veterinary Sciences (UREAR-ULg), Fundamental and Applied Research for Animal and Health (FARAH) Center, University of Liege, Liege, Belgium
| | - Fabiana Dal Pozzo
- Faculty of Veterinary Medicine, Research Unit in Epidemiology and Risk Analysis Applied to Veterinary Sciences (UREAR-ULg), Fundamental and Applied Research for Animal and Health (FARAH) Center, University of Liege, Liege, Belgium
| | - Christine Thys
- Faculty of Veterinary Medicine, Research Unit in Epidemiology and Risk Analysis Applied to Veterinary Sciences (UREAR-ULg), Fundamental and Applied Research for Animal and Health (FARAH) Center, University of Liege, Liege, Belgium
| | | | | | | | - Etienne Thiry
- Faculty of Veterinary Medicine, Fundamental and Applied Research for Animal and Health (FARAH) Center, Veterinary Virology and Animal Viral Diseases, University of Liege, Liege, Belgium
| | - Claude Saegerman
- Faculty of Veterinary Medicine, Research Unit in Epidemiology and Risk Analysis Applied to Veterinary Sciences (UREAR-ULg), Fundamental and Applied Research for Animal and Health (FARAH) Center, University of Liege, Liege, Belgium.
| |
Collapse
|
10
|
Macchi F, Rojas JM, Verna AE, Sevilla N, Franceschi V, Tebaldi G, Cavirani S, Martín V, Donofrio G. Bovine Herpesvirus-4-Based Vector Delivering Peste des Petits Ruminants Virus Hemagglutinin ORF Induces both Neutralizing Antibodies and Cytotoxic T Cell Responses. Front Immunol 2018; 9:421. [PMID: 29556236 PMCID: PMC5845008 DOI: 10.3389/fimmu.2018.00421] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 02/15/2018] [Indexed: 12/12/2022] Open
Abstract
Peste des Petits Ruminants Virus (PPRV) is an extremely infective morbillivirus that primarily affects goats and sheep. In underdeveloped countries where livestock are the main economical resource, PPRV causes considerable economic losses. Protective live attenuated vaccines are currently available but they induce antibody responses similar to those produced in PPRV naturally infected animals. Effective vaccines able to distinguish between vaccinated and naturally infected animals are required to PPRV control and eradication programs. Hemagglutinin (H) is a highly immunogenic PPRV envelope glycoprotein displaying both hemagglutinin and neuraminidase activities, playing a crucial role in virus attachment and penetration. In this study, a recombinant Bovine Herpesvirus-4 (BoHV-4)-based vector delivering an optimized PPRV-Hemagglutinin expression cassette, BoHV-4-A-PPRV-H-ΔTK, was assessed in immunocompetent C57BL/6 mice. BoHV-4-A-PPRV-H-ΔTK-immunization elicited both cellular and humoral immune responses with specific T cell, cytotoxic T lymphocyte, and sero-neutralizing antibody against PPRV. These data suggest recombinant BoHV-4-A-PPRV-H-ΔTK as an effective vaccine candidate to protect against PPRV herd infection and potentially applicable for eradication programs.
Collapse
Affiliation(s)
- Francesca Macchi
- Department of Medical Veterinary Science, University of Parma, Parma, Italy
| | - José Manuel Rojas
- Centro de Investigación en Sanidad Animal (CISA-INIA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Madrid, Spain
| | | | - Noemí Sevilla
- Centro de Investigación en Sanidad Animal (CISA-INIA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Madrid, Spain
| | | | - Giulia Tebaldi
- Department of Medical Veterinary Science, University of Parma, Parma, Italy
| | - Sandro Cavirani
- Department of Medical Veterinary Science, University of Parma, Parma, Italy
| | - Verónica Martín
- Centro de Investigación en Sanidad Animal (CISA-INIA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Madrid, Spain
| | - Gaetano Donofrio
- Department of Medical Veterinary Science, University of Parma, Parma, Italy
| |
Collapse
|
11
|
Russell BL, Parbhoo N, Gildenhuys S. Analysis of Conserved, Computationally Predicted Epitope Regions for VP5 and VP7 Across three Orbiviruses. Bioinform Biol Insights 2018; 12:1177932218755348. [PMID: 29434468 PMCID: PMC5802602 DOI: 10.1177/1177932218755348] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 01/04/2018] [Indexed: 12/15/2022] Open
Abstract
Orbiviruses are double-stranded RNA viruses that have profound economic and veterinary significance, 3 of the most important being African horse sickness virus (AHSV), bluetongue virus (BTV), and epizootic hemorrhagic disease virus (EHDV). Currently, vaccination and vector control are used as preventative measures; however, there are several problems with the current vaccines. Comparing viral amino acid sequences, we obtained an AHSV-BTV-EHDV consensus sequence for VP5 (viral protein 5) and for VP7 (viral protein 7) and generated homology models for these proteins. The structures and sequences were analyzed for amino acid sequence conservation, entropy, surface accessibility, and epitope propensity, to computationally determine whether consensus sequences still possess potential epitope regions. In total, 5 potential linear epitope regions on VP5 and 11 on VP7, as well as potential discontinuous B-cell epitopes, were identified and mapped onto the homology models created. Regions identified for VP5 and VP7 could be important in vaccine design against orbiviruses.
Collapse
Affiliation(s)
- Bonnie L Russell
- Department of Life and Consumer Sciences, College of Agriculture and Environmental Sciences, University of South Africa, Roodepoort, South Africa
| | - Nishal Parbhoo
- Department of Life and Consumer Sciences, College of Agriculture and Environmental Sciences, University of South Africa, Roodepoort, South Africa
| | - Samantha Gildenhuys
- Department of Life and Consumer Sciences, College of Agriculture and Environmental Sciences, University of South Africa, Roodepoort, South Africa
| |
Collapse
|
12
|
Rojas JM, Avia M, Pascual E, Sevilla N, Martín V. Vaccination with recombinant adenovirus expressing peste des petits ruminants virus-F or -H proteins elicits T cell responses to epitopes that arises during PPRV infection. Vet Res 2017; 48:79. [PMID: 29157291 PMCID: PMC5697415 DOI: 10.1186/s13567-017-0482-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 10/26/2017] [Indexed: 12/22/2022] Open
Abstract
Peste des petits ruminants virus (PPRV) causes an economically important disease that limits productivity in small domestic ruminants and often affects the livestock of the poorest populations in developing countries. Animals that survive PPRV develop strong cellular and humoral responses, which are probably necessary for protection. Vaccination should thus aim at mimicking these natural responses. Immunization strategies against this morbillivirus using recombinant adenoviruses expressing PPRV-F or -H proteins can protect PPRV-challenged animals and permit differentiation of infected from vaccinated animals. Little is known of the T cell repertoire these recombinant vaccines induce. In the present work, we identified several CD4+ and CD8+ T cell epitopes in sheep infected with PPRV. We also show that recombinant adenovirus vaccination induced T cell responses to the same epitopes, and led to memory T cell differentiation. T cells primed by these recombinant adenovirus vaccines expanded after PPRV challenge and probably contributed to protection. These data validate the use of recombinant adenovirus expressing PPRV genes as DIVA strategies to control this highly contagious disease.
Collapse
Affiliation(s)
- José Manuel Rojas
- Centro de Investigación en Sanidad Animal (CISA-INIA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Valdeolmos, Madrid, Spain
| | - Miguel Avia
- Centro de Investigación en Sanidad Animal (CISA-INIA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Valdeolmos, Madrid, Spain
| | - Elena Pascual
- Centro de Investigación en Sanidad Animal (CISA-INIA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Valdeolmos, Madrid, Spain
| | - Noemí Sevilla
- Centro de Investigación en Sanidad Animal (CISA-INIA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Valdeolmos, Madrid, Spain
| | - Verónica Martín
- Centro de Investigación en Sanidad Animal (CISA-INIA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Valdeolmos, Madrid, Spain.
| |
Collapse
|
13
|
Rojas JM, Rodríguez-Calvo T, Sevilla N. Recall T cell responses to bluetongue virus produce a narrowing of the T cell repertoire. Vet Res 2017; 48:38. [PMID: 28662714 PMCID: PMC5492282 DOI: 10.1186/s13567-017-0444-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Accepted: 06/26/2017] [Indexed: 11/12/2022] Open
Abstract
In most viral infections, recall T cell responses are critical for protection. The magnitude of these secondary responses can also affect the CD8 and CD4 epitope repertoire diversity. Bluetongue virus (BTV) infection in sheep elicits a T cell response that contributes to viremia control and could be relevant for cross-protection between BTV serotypes. Here, we characterized CD4+ and CD8+ T cell responses during primary and recall responses. During primary immune responses, both CD4+ and CD8+ T cell populations expanded by 14 days post-infection (dpi). CD4+ T cell populations showed a lower peak of expansion and prolonged contraction phase compared to CD8+ T cell populations. Recall responses to BTV challenge led to BTV-specific expansion and activation of CD8+ but not of CD4+ T cells. The evolution of the BTV-specific TCR repertoire was also characterized in response to VP7 peptide stimulation. Striking differences in repertoire development were noted over the time-course of infection. During primary responses, a broader repertoire was induced for MHC-I and MHC-II epitopes. However, during memory responses, a narrowed repertoire was activated towards a dominant motif in VP7 comprising amino acids 139–291. Monocytes were also examined, and expanded during acute infection resolution. In addition, pro-inflammatory cytokine levels increased after BTV inoculation and persisted throughout the experiment, indicative of a prolonged inflammatory state during BTV infections. These findings could have implications for vaccine design as the narrowing memory T cell repertoire induced after BTV re-infection could lead to the development of protective immunodominant TCR repertoires that differs between individual sheep.
Collapse
Affiliation(s)
- José-Manuel Rojas
- Centro de Investigación en Sanidad Animal (CISA-INIA), Instituto Nacional de Investigación Agraria y Alimentaria, Ctra Algete a El Casar km 8, Valdeolmos, 28130, Madrid, Spain
| | - Teresa Rodríguez-Calvo
- Centro de Investigación en Sanidad Animal (CISA-INIA), Instituto Nacional de Investigación Agraria y Alimentaria, Ctra Algete a El Casar km 8, Valdeolmos, 28130, Madrid, Spain.,Institute of Diabetes Research, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Neuherberg, Germany
| | - Noemí Sevilla
- Centro de Investigación en Sanidad Animal (CISA-INIA), Instituto Nacional de Investigación Agraria y Alimentaria, Ctra Algete a El Casar km 8, Valdeolmos, 28130, Madrid, Spain.
| |
Collapse
|
14
|
Marín-López A, Calvo-Pinilla E, Barriales D, Lorenzo G, Benavente J, Brun A, Martínez-Costas JM, Ortego J. Microspheres-prime/rMVA-boost vaccination enhances humoral and cellular immune response in IFNAR(-/-) mice conferring protection against serotypes 1 and 4 of bluetongue virus. Antiviral Res 2017; 142:55-62. [PMID: 28322923 DOI: 10.1016/j.antiviral.2017.03.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 03/15/2017] [Accepted: 03/15/2017] [Indexed: 02/05/2023]
Abstract
Bluetongue virus (BTV) is the causative agent of bluetongue disease (BT), which affects domestic and wild ruminants. At the present, 27 different serotypes have been documented. Vaccination has been demonstrated as one of the most effective methods to avoid viral dissemination. To overcome the drawbacks associated with the use of inactivated and attenuated vaccines we engineered a new recombinant BTV vaccine candidate based on proteins VP2, VP7, and NS1 of BTV-4 that were incorporated into avian reovirus muNS-Mi microspheres (MS-VP2/VP7/NS1) and recombinant modified vaccinia virus Ankara (rMVA). The combination of these two antigen delivery systems in a heterologous prime-boost vaccination strategy generated significant levels of neutralizing antibodies in IFNAR(-/-) mice. Furthermore, this immunization strategy increased the ratio of IgG2a/IgG1 in sera, indicating an induction of a Th1 response, and elicited a CD8 T cell response. Immunized mice were protected against lethal challenges with the homologous serotype 4 and the heterologous serotype 1 of BTV. All these results support the strategy based on microspheres in combination with rMVAs as a promising multiserotype vaccine candidate against BTV.
Collapse
Affiliation(s)
| | - Eva Calvo-Pinilla
- Centro de Investigación en Sanidad Animal, INIA-CISA, Valdeolmos, Madrid, Spain
| | - Diego Barriales
- Centro de Investigación en Sanidad Animal, INIA-CISA, Valdeolmos, Madrid, Spain
| | - Gema Lorenzo
- Centro de Investigación en Sanidad Animal, INIA-CISA, Valdeolmos, Madrid, Spain
| | - Javier Benavente
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS) and Departamento de Bioquímica e Bioloxía Molecular, Universidade de Santiago de Compostela, Spain
| | - Alejandro Brun
- Centro de Investigación en Sanidad Animal, INIA-CISA, Valdeolmos, Madrid, Spain
| | - Jose Manuel Martínez-Costas
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS) and Departamento de Bioquímica e Bioloxía Molecular, Universidade de Santiago de Compostela, Spain
| | - Javier Ortego
- Centro de Investigación en Sanidad Animal, INIA-CISA, Valdeolmos, Madrid, Spain.
| |
Collapse
|
15
|
Analysis of the three-dimensional structure of the African horse sickness virus VP7 trimer by homology modelling. Virus Res 2017; 232:80-95. [DOI: 10.1016/j.virusres.2017.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Revised: 01/27/2017] [Accepted: 02/02/2017] [Indexed: 01/21/2023]
|
16
|
Marín-López A, Bermúdez R, Calvo-Pinilla E, Moreno S, Brun A, Ortego J. Pathological Characterization Of IFNAR(-/-) Mice Infected With Bluetongue Virus Serotype 4. Int J Biol Sci 2016; 12:1448-1460. [PMID: 27994510 PMCID: PMC5166487 DOI: 10.7150/ijbs.14967] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 06/11/2016] [Indexed: 01/11/2023] Open
Abstract
Bluetongue virus (BTV) replicates in lymphoid tissues where infected mononuclear leukocytes secrete proinflammatory and vasoactive mediators that can contribute to bluetongue (BT) pathogenesis. Using the well-characterized IFNAR(-/-) mice animal model, we have now studied the histopathology and dynamics of leukocyte populations in different target tissues (spleen, thymus, and lung) during BTV-4 infection by histological and immunohistochemical techniques. The spleen and thymus of BTV-4 infected mice showed severe lymphoid depletion on H&E stained sections. This finding was confirmed by IHC, showing moderate decreased immunopositivity against CD3 in the thymus, and scarce immunoreactivity against CD3 and CD79 in the rest of the white pulp in the spleen, together with an increase in MAC387 immunostaining. BTV-4 infection also induced the expression of active caspase-3 in the spleen, where apoptotic debris was observed by H&E. A dramatic increase in iNOS immunoreactivity associated to necrotic areas of the white pulp was observed, being less noticeable in the thymus and the lung. The induction of pro-inflammatory cytokines in tissues where BTV replicates was evaluated by measuring transcript levels by RT-qPCR. BTV-4 infection led to enhance transcription of IFN-γ, TNF, IL-6, IL-12-p40, and IL-1β mRNA in the thymus, spleen and lung, correlating with the level of virus replication in these tissues. Disease progression and pathogenesis in IFNAR(-/-) mice closely mimics hallmarks of bluetongue disease in ruminants. IFNAR(-/-) mice are a good choice to facilitate a faster advance in the field of orbiviruses.
Collapse
Affiliation(s)
| | - Roberto Bermúdez
- Departamento de Ciencias Clínicas Veterinarias, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain
| | | | - Sandra Moreno
- INIA-CISA, Ctra. Algete-El Casar, Valdeolmos, 28130 Madrid, Spain
| | - Alejandro Brun
- INIA-CISA, Ctra. Algete-El Casar, Valdeolmos, 28130 Madrid, Spain
| | - Javier Ortego
- INIA-CISA, Ctra. Algete-El Casar, Valdeolmos, 28130 Madrid, Spain
| |
Collapse
|
17
|
Marín-López A, Barriales D, Moreno S, Ortego J, Calvo-Pinilla E. Defeating Bluetongue virus: new approaches in the development of multiserotype vaccines. Future Virol 2016. [DOI: 10.2217/fvl-2016-0061] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Bluetongue virus (BTV) is a global threat to domestic and wild ruminants, causing massive economic losses throughout the world. New serotypes of the virus are rapidly emerging in different continents, unfortunately there is little cross-protection between BTV serotypes. The eradication of the virus from a region is particularly complicated in areas where multiple serotypes circulate for a long time. The present review summarizes the actual concerns about the spread of the virus and relevant approaches to develop efficient vaccines against BTV, in particular those focused on a multiserotype design.
Collapse
Affiliation(s)
| | - Diego Barriales
- Centro de Investigación en Sanidad Animal, INIA-CISA, Valdeolmos-Madrid, Spain
| | - Sandra Moreno
- Centro de Investigación en Sanidad Animal, INIA-CISA, Valdeolmos-Madrid, Spain
| | - Javier Ortego
- Centro de Investigación en Sanidad Animal, INIA-CISA, Valdeolmos-Madrid, Spain
| | - Eva Calvo-Pinilla
- Centro de Investigación en Sanidad Animal, INIA-CISA, Valdeolmos-Madrid, Spain
| |
Collapse
|
18
|
Faber F, van Kleef M, Tshilwane S, Pretorius A. African horse sickness virus serotype 4 antigens, VP1-1, VP2-2, VP4, VP7 and NS3, induce cytotoxic T cell responses in vitro. Virus Res 2016; 220:12-20. [DOI: 10.1016/j.virusres.2016.04.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 04/06/2016] [Accepted: 04/06/2016] [Indexed: 02/03/2023]
|
19
|
Martín V, Pascual E, Avia M, Peña L, Valcárcel F, Sevilla N. Protective Efficacy in Sheep of Adenovirus-Vectored Vaccines against Bluetongue Virus Is Associated with Specific T Cell Responses. PLoS One 2015; 10:e0143273. [PMID: 26619062 PMCID: PMC4664254 DOI: 10.1371/journal.pone.0143273] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 11/03/2015] [Indexed: 12/31/2022] Open
Abstract
Bluetongue virus (BTV) is an economically important Orbivirus of the Reoviridae family that causes a hemorrhagic disease in ruminants. Its control has been achieved by inactivated-vaccines that have proven to protect against homologous BTV challenge although unable to induce long-term immunity. Therefore, a more efficient control strategy needs to be developed. Recombinant adenovirus vectors are lead vaccine candidates for protection of several diseases, mainly because of their potency to induce potent T cell immunity. Here we report the induction of humoral and T-cell mediated responses able to protect animals against BTV challenge by recombinant replication-defective human adenovirus serotype 5 (Ad5) expressing either VP7, VP2 or NS3 BTV proteins. First we used the IFNAR(-/-) mouse model system to establish a proof of principle, and afterwards we assayed the protective efficacy in sheep, the natural host of BTV. Mice were completely protected against BTV challenge, developing humoral and BTV-specific CD8+- and CD4+-T cell responses by vaccination with the different rAd5. Sheep vaccinated with Ad5-BTV-VP2 and Ad5-BTV-VP7 or only with Ad5-BTV-VP7 and challenged with BTV showed mild disease symptoms and reduced viremia. This partial protection was achieved in the absence of neutralizing antibodies but strong BTV-specific CD8+ T cell responses in those sheep vaccinated with Ad5-BTV-VP7. These data indicate that rAd5 is a suitable vaccine vector to induce T cell immunity during BTV vaccination and provide new data regarding the relevance of T cell responses in protection during BTV infection.
Collapse
Affiliation(s)
- Verónica Martín
- Centro de Investigación en Sanidad Animal (CISA-INIA), Instituto Nacional de Investigación Agraria y Alimentaria, Valdeolmos, Madrid, Spain
| | - Elena Pascual
- Centro de Investigación en Sanidad Animal (CISA-INIA), Instituto Nacional de Investigación Agraria y Alimentaria, Valdeolmos, Madrid, Spain
| | - Miguel Avia
- Centro de Investigación en Sanidad Animal (CISA-INIA), Instituto Nacional de Investigación Agraria y Alimentaria, Valdeolmos, Madrid, Spain
| | - Lourdes Peña
- Centro de Investigación en Sanidad Animal (CISA-INIA), Instituto Nacional de Investigación Agraria y Alimentaria, Valdeolmos, Madrid, Spain
| | - Félix Valcárcel
- Centro de Investigación en Sanidad Animal (CISA-INIA), Instituto Nacional de Investigación Agraria y Alimentaria, Valdeolmos, Madrid, Spain
| | - Noemí Sevilla
- Centro de Investigación en Sanidad Animal (CISA-INIA), Instituto Nacional de Investigación Agraria y Alimentaria, Valdeolmos, Madrid, Spain
- * E-mail:
| |
Collapse
|
20
|
Sánchez-Cordón PJ, Pérez de Diego AC, Gómez-Villamandos JC, Sánchez-Vizcaíno JM, Pleguezuelos FJ, Garfia B, del Carmen P, Pedrera M. Comparative analysis of cellular immune responses and cytokine levels in sheep experimentally infected with bluetongue virus serotype 1 and 8. Vet Microbiol 2015; 177:95-105. [PMID: 25769647 DOI: 10.1016/j.vetmic.2015.02.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 02/18/2015] [Accepted: 02/19/2015] [Indexed: 11/24/2022]
Abstract
Protective immunity in sheep with bluetongue virus (BTV) infection as well as the role of BTV-induced cytokines during immune response remains unclear. Understanding the basis immunological mechanisms in sheep experimentally infected with serotypes 1 and 8 (BTV-1 and -8) was the aim of this study. A time-course study was carried out in order to evaluate cell-mediated immune response and serum concentrations of cytokines (IL-1β, TNFα, IL-12, IFNγ, IL-4 and IL-10) with inflammatory and immunological functions. Depletion of T cell subsets (mainly CD4(+), γδ and CD25(+)) together with the absence of cytokines (IFNγ and IL-12) involved in the regulation of cell-mediated antiviral immunity at the first stage of the disease suggested that both BTV-1 and BTV-8 might impair host's capability against primary infections which would favor viral replication and spreading. However, cellular immune response and cytokines elicited an immune response in sheep that efficiently reduced viremia in the final stage of the experiment. Recovery of T cell subsets (CD4(+) and CD25(+)) together with a significant increase of CD8(+) T lymphocytes in both infected groups were observed in parallel with the decrease of viremia. Additionally, the recovery of CD4(+) T lymphocytes together with the significant increase of IL-4 serum levels at the final stage of the experiment might contribute to humoral immune response activation and neutralizing antibodies production against BTV previously described in the course of this experiment. These results suggested that both cellular and humoral immune response may contribute to protective immunity against BTV-1 and BTV-8 in sheep. The possible role played by IL-10 and CD25(+) cells in controlling inflammatory and immune response in the final stage of the experiment has also been suggested.
Collapse
Affiliation(s)
- P J Sánchez-Cordón
- Department of Comparative Pathology, Veterinary Faculty, University of Córdoba-Agrifood Campus of International Excellence (ceiA3), Edificio Sanidad Animal, Campus de Rabanales, 14014 Córdoba, Spain.
| | - A C Pérez de Diego
- VISAVET Center and Animal Health Department, Veterinary Faculty, University Complutense of Madrid, Av. Puerta de Hierro s/n, 28040 Madrid, Spain
| | - J C Gómez-Villamandos
- Department of Comparative Pathology, Veterinary Faculty, University of Córdoba-Agrifood Campus of International Excellence (ceiA3), Edificio Sanidad Animal, Campus de Rabanales, 14014 Córdoba, Spain
| | - J M Sánchez-Vizcaíno
- VISAVET Center and Animal Health Department, Veterinary Faculty, University Complutense of Madrid, Av. Puerta de Hierro s/n, 28040 Madrid, Spain
| | - F J Pleguezuelos
- Department of Comparative Pathology, Veterinary Faculty, University of Córdoba-Agrifood Campus of International Excellence (ceiA3), Edificio Sanidad Animal, Campus de Rabanales, 14014 Córdoba, Spain
| | - B Garfia
- Garfia Veterinary Medicine Laboratory S.L., Polígono Industrial Tecnocórdoba, C/Varsovia, 53, 14014 Córdoba, Spain
| | - P del Carmen
- VISAVET Center and Animal Health Department, Veterinary Faculty, University Complutense of Madrid, Av. Puerta de Hierro s/n, 28040 Madrid, Spain
| | - M Pedrera
- Department of Comparative Pathology, Veterinary Faculty, University of Córdoba-Agrifood Campus of International Excellence (ceiA3), Edificio Sanidad Animal, Campus de Rabanales, 14014 Córdoba, Spain
| |
Collapse
|
21
|
Bouet-Cararo C, Contreras V, Caruso A, Top S, Szelechowski M, Bergeron C, Viarouge C, Desprat A, Relmy A, Guibert JM, Dubois E, Thiery R, Bréard E, Bertagnoli S, Richardson J, Foucras G, Meyer G, Schwartz-Cornil I, Zientara S, Klonjkowski B. Expression of VP7, a Bluetongue virus group specific antigen by viral vectors: analysis of the induced immune responses and evaluation of protective potential in sheep. PLoS One 2014; 9:e111605. [PMID: 25364822 PMCID: PMC4218782 DOI: 10.1371/journal.pone.0111605] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Accepted: 10/06/2014] [Indexed: 11/29/2022] Open
Abstract
Bluetongue virus (BTV) is an economically important Orbivirus transmitted by biting midges to domestic and wild ruminants. The need for new vaccines has been highlighted by the occurrence of repeated outbreaks caused by different BTV serotypes since 1998. The major group-reactive antigen of BTV, VP7, is conserved in the 26 serotypes described so far, and its role in the induction of protective immunity has been proposed. Viral-based vectors as antigen delivery systems display considerable promise as veterinary vaccine candidates. In this paper we have evaluated the capacity of the BTV-2 serotype VP7 core protein expressed by either a non-replicative canine adenovirus type 2 (Cav-VP7 R0) or a leporipoxvirus (SG33-VP7), to induce immune responses in sheep. Humoral responses were elicited against VP7 in almost all animals that received the recombinant vectors. Both Cav-VP7 R0 and SG33-VP7 stimulated an antigen-specific CD4+ response and Cav-VP7 R0 stimulated substantial proliferation of antigen-specific CD8+ lymphocytes. Encouraged by the results obtained with the Cav-VP7 R0 vaccine vector, immunized animals were challenged with either the homologous BTV-2 or the heterologous BTV-8 serotype and viral burden in plasma was followed by real-time RT-PCR. The immune responses triggered by Cav-VP7 R0 were insufficient to afford protective immunity against BTV infection, despite partial protection obtained against homologous challenge. This work underscores the need to further characterize the role of BTV proteins in cross-protective immunity.
Collapse
Affiliation(s)
| | - Vanessa Contreras
- Virologie et Immunologie Moléculaires, UR 892 INRA, Jouy-en-Josas, France
| | - Agathe Caruso
- INRA, UMR1225, IHAP, Université de Toulouse, INP, ENVT, Toulouse, France
| | - Sokunthea Top
- INRA, UMR1225, IHAP, Université de Toulouse, INP, ENVT, Toulouse, France
| | - Marion Szelechowski
- Centre de Physiopathologie de Toulouse Purpan, INSERM U1043, CNRS U5282, Université Paul-Sabatier, Toulouse, France
| | - Corinne Bergeron
- UPE, ANSES, INRA, ENVA, UMR 1161 ANSES/INRA/ENVA, Maisons-Alfort, France
| | - Cyril Viarouge
- UPE, ANSES, INRA, ENVA, UMR 1161 ANSES/INRA/ENVA, Maisons-Alfort, France
| | - Alexandra Desprat
- UPE, ANSES, INRA, ENVA, UMR 1161 ANSES/INRA/ENVA, Maisons-Alfort, France
| | - Anthony Relmy
- UPE, ANSES, INRA, ENVA, UMR 1161 ANSES/INRA/ENVA, Maisons-Alfort, France
| | | | - Eric Dubois
- Unité de pathologie des petits ruminants, ANSES, Sophia-Antipolis, France
| | - Richard Thiery
- Unité de pathologie des petits ruminants, ANSES, Sophia-Antipolis, France
| | - Emmanuel Bréard
- UPE, ANSES, INRA, ENVA, UMR 1161 ANSES/INRA/ENVA, Maisons-Alfort, France
| | | | | | - Gilles Foucras
- INRA, UMR1225, IHAP, Université de Toulouse, INP, ENVT, Toulouse, France
| | - Gilles Meyer
- INRA, UMR1225, IHAP, Université de Toulouse, INP, ENVT, Toulouse, France
| | | | - Stephan Zientara
- UPE, ANSES, INRA, ENVA, UMR 1161 ANSES/INRA/ENVA, Maisons-Alfort, France
| | - Bernard Klonjkowski
- UPE, ANSES, INRA, ENVA, UMR 1161 ANSES/INRA/ENVA, Maisons-Alfort, France
- * E-mail:
| |
Collapse
|
22
|
Marín-López A, Otero-Romero I, de la Poza F, Menaya-Vargas R, Calvo-Pinilla E, Benavente J, Martínez-Costas JM, Ortego J. VP2, VP7, and NS1 proteins of bluetongue virus targeted in avian reovirus muNS-Mi microspheres elicit a protective immune response in IFNAR(-/-) mice. Antiviral Res 2014; 110:42-51. [PMID: 25057758 DOI: 10.1016/j.antiviral.2014.07.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 07/04/2014] [Accepted: 07/14/2014] [Indexed: 11/17/2022]
Abstract
Vaccination is critical for controlling the spread of bluetongue virus (BTV). The inactivated BTV vaccines that are now being used in Europe are effective in preventing outbreaks of BTV but secondary effects associated to repetitive inoculation of aluminum-containing adjuvants and the need to develop safer, cross-reactive, and more efficacious vaccines with differential diagnostic capability have re-stimulated the interest in developing improved vaccination strategies against BTV. We have engineered a subunit BTV vaccine candidate based on proteins VP2, VP7, and NS1 of BTV-4 incorporated into avian reovirus (ARV) muNS-Mi microspheres (MS-VP2/MS-VP7/MS-NS1). IFNAR(-/-) mice immunized with MS-VP2/MS-VP7/MS-NS1 without adjuvant generated significant levels of neutralizing antibodies specific to BTV-4. In addition, vaccination stimulated specific T cell responses, predominantly CD4+, against the virus. Immunized mice were fully protected against a homologous challenge with a lethal dose of BTV-4 and partially cross-protected against a heterologous challenge with a lethal dose of BTV-1. These results support MS-VP2/MS-VP7/MS-NS1 as a promising subunit vaccine candidate against multiple serotypes of BTV as well as the use of microspheres as an alternative delivery method with potent intrinsic adjuvant activity.
Collapse
Affiliation(s)
- Alejandro Marín-López
- Centro de Investigación en Sanidad Animal, INIA-CISA, Valdeolmos, 28130 Madrid, Spain
| | - Iria Otero-Romero
- Centro Singular de Investigación en Química Biológica y Materiales Moleculares (CIQUS), Universidad de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Francisco de la Poza
- Centro de Investigación en Sanidad Animal, INIA-CISA, Valdeolmos, 28130 Madrid, Spain
| | - Rebeca Menaya-Vargas
- Centro Singular de Investigación en Química Biológica y Materiales Moleculares (CIQUS), Universidad de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Eva Calvo-Pinilla
- Centro de Investigación en Sanidad Animal, INIA-CISA, Valdeolmos, 28130 Madrid, Spain
| | - Javier Benavente
- Centro Singular de Investigación en Química Biológica y Materiales Moleculares (CIQUS), Universidad de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - José M Martínez-Costas
- Centro Singular de Investigación en Química Biológica y Materiales Moleculares (CIQUS), Universidad de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Javier Ortego
- Centro de Investigación en Sanidad Animal, INIA-CISA, Valdeolmos, 28130 Madrid, Spain.
| |
Collapse
|
23
|
Rojas JM, Peña L, Martín V, Sevilla N. Ovine and murine T cell epitopes from the non-structural protein 1 (NS1) of bluetongue virus serotype 8 (BTV-8) are shared among viral serotypes. Vet Res 2014; 45:30. [PMID: 24621015 PMCID: PMC3995764 DOI: 10.1186/1297-9716-45-30] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 02/27/2014] [Indexed: 11/24/2022] Open
Abstract
Bluetongue virus (BTV) is a non-enveloped dsRNA virus that causes a haemorrhagic disease mainly in sheep. It is an economically important Orbivirus of the Reoviridae family. In order to estimate the importance of T cell responses during BTV infection, it is essential to identify the epitopes targeted by the immune system. In the present work, we selected potential T cell epitopes (3 MHC-class II-binding and 8 MHC-class I binding peptides) for the C57BL/6 mouse strain from the BTV-8 non-structural protein NS1, using H2b-binding predictive algorithms. Peptide binding assays confirmed all MHC-class I predicted peptides bound MHC-class I molecules. The immunogenicity of these 11 predicted peptides was then determined using splenocytes from BTV-8-inoculated C57BL/6 mice. Four MHC-class I binding peptides elicited specific IFN-γ production and generated cytotoxic T lymphocytes (CTL) in BTV-8 infected mice. CTL specific for 2 of these peptides were also able to recognise target cells infected with different BTV serotypes. Similarly, using a combination of IFN-γ ELISPOT, intracellular cytokine staining and proliferation assays, two MHC-class II peptides were identified as CD4+ T cell epitopes in BTV-8 infected mice. Importantly, two peptides were also consistently immunogenic in sheep infected with BTV-8 using IFN-γ ELISPOT assays. Both of these peptides stimulated CD4+ T cells that cross-reacted with other BTV serotypes. The characterisation of these T cell epitopes can help develop vaccines protecting against a broad spectrum of BTV serotypes and differentiate infected from vaccinated animals.
Collapse
Affiliation(s)
| | | | | | - Noemí Sevilla
- Centro de Investigación en Sanidad Animal (CISA-INIA), Instituto Nacional de Investigación Agraria y Alimentaria, Valdeolmos, Madrid, Spain.
| |
Collapse
|
24
|
Coetzee P, van Vuuren M, Venter EH, Stokstad M. A review of experimental infections with bluetongue virus in the mammalian host. Virus Res 2014; 182:21-34. [PMID: 24462840 PMCID: PMC7132480 DOI: 10.1016/j.virusres.2013.12.044] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Revised: 12/27/2013] [Accepted: 12/31/2013] [Indexed: 11/23/2022]
Abstract
Experimental infection studies with bluetongue virus (BTV) in the mammalian host have a history that stretches back to the late 18th century. Studies in a wide range of ruminant and camelid species as well as mice have been instrumental in understanding BTV transmission, bluetongue (BT) pathogenicity/pathogenesis, viral virulence, the induced immune response, as well as reproductive failures associated with BTV infection. These studies have in many cases been complemented by in vitro studies with BTV in different cell types in tissue culture. Together these studies have formed the basis for the understanding of BTV-host interaction and have contributed to the design of successful control strategies, including the development of effective vaccines. This review describes some of the fundamental and contemporary infection studies that have been conducted with BTV in the mammalian host and provides an overview of the principal animal welfare issues that should be considered when designing experimental infection studies with BTV in in vivo infection models. Examples are provided from the authors' own laboratory where the three Rs (replacement, reduction and refinement) have been implemented in the design of experimental infection studies with BTV in mice and goats. The use of the ARRIVE guidelines for the reporting of data from animal infection studies is emphasized.
Collapse
Affiliation(s)
- Peter Coetzee
- Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Private Bag X04, Onderstepoort, Pretoria 0110, South Africa; Department of Production Animal Clinical Sciences, Norwegian School of Veterinary Science, P. O. Box 8146 Dep., N-0033 Oslo, Norway.
| | - Moritz van Vuuren
- Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Private Bag X04, Onderstepoort, Pretoria 0110, South Africa.
| | - Estelle H Venter
- Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Private Bag X04, Onderstepoort, Pretoria 0110, South Africa.
| | - Maria Stokstad
- Department of Production Animal Clinical Sciences, Norwegian School of Veterinary Science, P. O. Box 8146 Dep., N-0033 Oslo, Norway.
| |
Collapse
|
25
|
Calvo-Pinilla E, Castillo-Olivares J, Jabbar T, Ortego J, de la Poza F, Marín-López A. Recombinant vaccines against bluetongue virus. Virus Res 2013; 182:78-86. [PMID: 24287057 DOI: 10.1016/j.virusres.2013.11.013] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 11/18/2013] [Accepted: 11/18/2013] [Indexed: 11/28/2022]
Abstract
Bluetongue (BT) is a hemorrhagic disease of ruminants caused by bluetongue virus (BTV), the prototype member of the genus Orbivirus within the family Reoviridae and is transmitted via biting midges of the genus Culicoides. BTV can be found on all continents except Antarctica, and up to 26 immunologically distinct BTV serotypes have been identified. Live attenuated and inactivated BTV vaccines have been used over the years with different degrees of success. The multiple outbreaks of BTV in Mediterranean Europe in the last two decades and the incursion of BTV-8 in Northern Europe in 2008 has re-stimulated the interest to develop improved vaccination strategies against BTV. In particular, safer, cross-reactive, more efficacious vaccines with differential diagnostic capability have been pursued by multiple BTV research groups and vaccine manufacturers. A wide variety of recombinant BTV vaccine prototypes have been investigated, ranging from baculovirus-expressed sub-unit vaccines to the use of live viral vectors. This article gives a brief overview of all these modern approaches to develop vaccines against BTV including some recent unpublished data.
Collapse
Affiliation(s)
| | | | - Tamara Jabbar
- The Pirbright Institute, Pirbright, Surrey, United Kingdom
| | - Javier Ortego
- Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación Agraria (INIA), Valdeolmos, Madrid, Spain
| | - Francisco de la Poza
- Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación Agraria (INIA), Valdeolmos, Madrid, Spain
| | - Alejandro Marín-López
- Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación Agraria (INIA), Valdeolmos, Madrid, Spain
| |
Collapse
|
26
|
Rojas JM, Moreno H, García A, Ramírez JC, Sevilla N, Martín V. Two replication-defective adenoviral vaccine vectors for the induction of immune responses to PPRV. Vaccine 2013; 32:393-400. [PMID: 24269622 DOI: 10.1016/j.vaccine.2013.11.033] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 10/30/2013] [Accepted: 11/06/2013] [Indexed: 11/20/2022]
Abstract
Peste des petits ruminants is a highly contagious disease of small ruminants caused by a Morbillivirus, peste des petits ruminants virus (PPRV). Two recombinant replication-defective human adenovirus serotype 5 (Ad5) containing the highly immunogenic fusion protein (F) and hemaglutinine protein (H) genes from PPRV were constructed. HEK293A cells infected with either virus (Ad5-PPRV-F or -H) express F and H proteins respectively. These viruses were used to vaccinate mice by intramuscular inoculation. Both viruses elicited PPRV-specific B- and T-cell responses. Thus, after two immunizations, sera from immunized mice elicited neutralizing antibody response, indicating that this approach has the potential to confer protective immunity. In addition, we detected a significant antigen specific CD4(+) and CD8(+) T-cell response in mice vaccinated with either virus. These results indicate that these adenovirus constructs offer a promising alternative to current vaccine strategies for the development of PPRV DIVA vaccines.
Collapse
Affiliation(s)
- José M Rojas
- Centro de Investigación en Sanidad Animal (CISA-INIA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, 28130 Valdeolmos, Madrid, Spain.
| | - Héctor Moreno
- Centro de Investigación en Sanidad Animal (CISA-INIA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, 28130 Valdeolmos, Madrid, Spain.
| | - Aída García
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), C/Melchor Fernández Almagro no 3, 28029 Madrid, Spain.
| | - Juan C Ramírez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), C/Melchor Fernández Almagro no 3, 28029 Madrid, Spain.
| | - Noemí Sevilla
- Centro de Investigación en Sanidad Animal (CISA-INIA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, 28130 Valdeolmos, Madrid, Spain.
| | - Verónica Martín
- Centro de Investigación en Sanidad Animal (CISA-INIA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, 28130 Valdeolmos, Madrid, Spain.
| |
Collapse
|
27
|
Type I interferon limits the capacity of bluetongue virus to infect hematopoietic precursors and dendritic cells in vitro and in vivo. J Virol 2013; 88:859-67. [PMID: 24173228 DOI: 10.1128/jvi.02697-13] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Hematopoietic stem cells (HSCs) give rise to progenitors with potential to produce multiple cell types, including dendritic cells (DCs). DCs are the principal antigen-presenting cells and represent the crucial link between innate and adaptive immune responses. Bluetongue virus (BTV), an economically important Orbivirus of the Reoviridae family, causes a hemorrhagic disease mainly in sheep and occasionally in other species of ruminants. BTV is transmitted between its mammalian hosts by certain species of biting midges (Culicoides spp.) and is a potent alpha interferon (IFN-α) inducer. In the present report, we show that BTV infects cells of hematopoietic origin but not HSCs in immunocompetent sheep. However, BTV infects HSCs in the absence of type I IFN (IFN-I) signaling in vitro and in vivo. Infection of HSCs in vitro results in cellular death by apoptosis. Furthermore, BTV infects bone marrow-derived DCs (BM-DCs), interfering with their development to mature DCs in the absence of type I IFN signaling. Costimulatory molecules CD80 and CD86 and costimulatory molecules CD40 and major histocompatibility complex class II (MHC-II) are affected by BTV infection, suggesting that BTV interferes with DC antigen-presenting capacity. In vivo, different DC populations are also affected during the course of infection, probably as a result of a direct effect of BTV replication in DCs and the production of infectious virus. These new findings suggest that BTV infection of HSCs and DCs can impair the immune response, leading to persistence or animal death, and that this relies on IFN-I.
Collapse
|
28
|
Ortego J, de la Poza F, Marín-López A. Interferon α/β receptor knockout mice as a model to study bluetongue virus infection. Virus Res 2013; 182:35-42. [PMID: 24100234 DOI: 10.1016/j.virusres.2013.09.038] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 09/24/2013] [Accepted: 09/25/2013] [Indexed: 12/21/2022]
Abstract
Bluetongue is an arthropod-borne disease caused by a virus of the genus Orbivirus, the bluetongue virus (BTV), which affects ruminant livestock such as cattle, sheep, and goats and wild ruminants such as deer, and camelids. Recently, adult mice with gene knockouts of the interferon α/β receptor (IFNAR-/-) have been described as a model of lethal BTV infection. IFNAR(-/-) mice are highly susceptible to BTV-1, BTV-4 and BTV-8 infection when the virus is administered intravenously or subcutaneosuly. Disease progression and pathogenesis closely mimics signs of bluetongue disease in ruminants. In the present paper we review the studies where IFNAR(-/-) mice have been used as an animal model to study BTV transmission, pathogenesis, virulence, and protective efficacy of inactivated and new recombinant marker BTV vaccines. Furthermore, we report new data on protective efficacy of different strategies of BTV vaccination and also on induction of interferon α/β and proinflammatory immune responses in IFNAR(-/-) mice infected with BTV.
Collapse
Affiliation(s)
- Javier Ortego
- Centro de Investigación en Sanidad Animal, CISA-INIA, Valdeolmos, Madrid, Spain.
| | | | | |
Collapse
|
29
|
Huang XJ, Lü X, Lei YF, Yang J, Yao M, Lan HY, Zhang JM, Jia ZS, Yin W, Xu ZK. Cellular immunogenicity of a multi-epitope peptide vaccine candidate based on hepatitis C virus NS5A, NS4B and core proteins in HHD-2 mice. J Virol Methods 2013; 189:47-52. [PMID: 23333413 DOI: 10.1016/j.jviromet.2013.01.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Revised: 12/17/2012] [Accepted: 01/08/2013] [Indexed: 12/19/2022]
Abstract
To develop a vaccine against hepatitis C virus (HCV), a multi-epitope peptide was synthesized from nonstructural proteins containing HLA-A2 epitopes inducing mainly responses in natural infection. The engineered vaccine candidate, VAL-44, consists of multiple epitopes from the HCV NS5A, NS4B and core proteins. Immunization with the VAL-44 peptide induced higher CTL responses than those by the smaller VL-20 peptide. VAL-44 induced antigen-specific IFN-γ-producing CD4+ T cells and CD8+ T cells. VAL-44 elicited a Th1-biased immune response with secretion of high amounts of IFN-γ and IL-2, compared with VL-20. These results suggest that VAL-44 can elicit strong cellular immune responses. The VAL-44 peptide stimulated IFN-γ production from viral-specific peripheral blood mononuclear cells (PBMCs) of patients infected with HCV. These results suggest that VAL-44 could be developed as a potential HCV multi-epitope peptide vaccine.
Collapse
Affiliation(s)
- Xiao-Jun Huang
- Center of Teaching Experiment, Fourth Military Medical University, Xi'an, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
El Garch H, Crafford JE, Amouyal P, Durand PY, Edlund Toulemonde C, Lemaitre L, Cozette V, Guthrie A, Minke JM. An African horse sickness virus serotype 4 recombinant canarypox virus vaccine elicits specific cell-mediated immune responses in horses. Vet Immunol Immunopathol 2012; 149:76-85. [PMID: 22763149 DOI: 10.1016/j.vetimm.2012.06.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2011] [Revised: 05/30/2012] [Accepted: 06/06/2012] [Indexed: 12/24/2022]
Abstract
A recombinant canarypox virus vectored vaccine co-expressing synthetic genes encoding outer capsid proteins, VP2 and VP5, of African horse sickness virus (AHSV) serotype 4 (ALVAC(®)-AHSV4) has been demonstrated to fully protect horses against homologous challenge with virulent field virus. Guthrie et al. (2009) detected weak and variable titres of neutralizing antibody (ranging from <10 to 40) 8 weeks after vaccination leading us to hypothesize that there could be a participation of cell mediated immunity (CMI) in protection against AHSV4. The present study aimed at characterizing the CMI induced by the experimental ALVAC(®)-AHSV4 vaccine. Six horses received two vaccinations twenty-eight days apart and three horses remained unvaccinated. The detection of VP2/VP5 specific IFN-γ responses was assessed by enzyme linked immune spot (ELISpot) assay and clearly demonstrated that all ALVAC(®)-AHSV4 vaccinated horses developed significant IFN-γ production compared to unvaccinated horses. More detailed immune responses obtained by flow cytometry demonstrated that ALVAC(®)-AHSV4 vaccinations induced immune cells, mainly CD8(+) T cells, able to recognize multiple T-epitopes through all VP2 and only the N-terminus sequence of VP5. Neither VP2 nor VP5 specific IFN-γ responses were detected in unvaccinated horses. Overall, our data demonstrated that an experimental recombinant canarypox based vaccine induced significant CMI specific for both VP2 and VP5 proteins of AHSV4.
Collapse
|
31
|
Eschbaumer M, Wernike K, Batten CA, Savini G, Edwards L, Di Gennaro A, Teodori L, Oura CAL, Beer M, Hoffmann B. Epizootic hemorrhagic disease virus serotype 7 in European cattle and sheep: diagnostic considerations and effect of previous BTV exposure. Vet Microbiol 2012; 159:298-306. [PMID: 22560764 DOI: 10.1016/j.vetmic.2012.04.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2012] [Revised: 03/30/2012] [Accepted: 04/12/2012] [Indexed: 10/28/2022]
Abstract
Epizootic hemorrhagic disease virus (EHDV), an arthropod-borne orbivirus (family Reoviridae), is an emerging pathogen of wild and domestic ruminants that is closely related to bluetongue virus (BTV). The present study examines the outcome of an experimental EHDV-7 infection of Holstein cattle and East Frisian sheep. Apart from naïve animals that had not been exposed to BTV, it included animals that had been experimentally infected with either BTV-6 or BTV-8 two months earlier. In addition, EHDV-infected cattle were subsequently challenged with BTV-8. Samples were tested with commercially available ELISA and real-time RT-PCR kits and a custom NS3-specific real-time RT-PCR assay. Virus isolation was attempted in Vero, C6/36 and KC cells (from Culicoides variipennis), embryonated chicken eggs and type I interferon receptor-deficient IFNAR(-/-) mice. EHDV-7 productively infected Holstein cattle, but caused no clinical signs. The inoculation of East Frisian sheep, on the other hand, apparently did not lead to a productive infection. The commercial diagnostic kits performed adequately. KC cells proved to be the most sensitive means of virus isolation, but viremia was shorter than 2 weeks in most animals. No interference between EHDV and BTV infection was observed; therefore the pre-existing immunity to some BTV serotypes in Europe is not expected to protect against a possible introduction of EHDV, in spite of the close relation between the viruses.
Collapse
Affiliation(s)
- Michael Eschbaumer
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Suedufer 10, 17493 Greifswald - Insel Riems, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Stewart M, Dovas CI, Chatzinasiou E, Athmaram TN, Papanastassopoulou M, Papadopoulos O, Roy P. Protective efficacy of Bluetongue virus-like and subvirus-like particles in sheep: presence of the serotype-specific VP2, independent of its geographic lineage, is essential for protection. Vaccine 2012; 30:2131-9. [PMID: 22285887 DOI: 10.1016/j.vaccine.2012.01.042] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Revised: 01/10/2012] [Accepted: 01/13/2012] [Indexed: 12/31/2022]
Abstract
There have been multiple separate outbreaks of Bluetongue (BT) disease of ruminants in Europe since 1998, often entering via the Mediterranean countries of Italy, Spain and Greece. BT is caused by an orbivirus, Bluetongue virus (BTV), a member of the family Reoviridae. BTV is a non-enveloped double-capsid virus, which encodes 7 structural proteins (VP1-VP7) and several non-structural proteins (NS1, NS2, NS3/3a and NS4) from ten double-stranded RNA segments of the genome. In this report, we have prepared BTV virus-like particles (VLPs, composed of VP2, VP3, VP5 and VP7) and sub-viral, inner core-like particles (CLPs, VP3 and VP7) using a recombinant baculovirus expression system. We compared the protective efficacy of VLPs and CLPs in sheep and investigated the importance of geographical lineages of BTV in the development of vaccines. The Greek crossbred Karagouniko sheep, which display mild to sub-clinical BT, were vaccinated with VLPs or CLPs of BTV-1, derived from western lineage and were challenged with virulent BTV-1 from an eastern lineage. All VLP-vaccinated animals developed a neutralising antibody response to BTV-1 from both lineages prior to challenge. Moreover, post-challenged animals had no clinical manifestation or viraemia and the challenged virus replication was completely inhibited. In contrast, CLP-vaccinated animals did not induce any neutralising antibody response but developed the group specific VP7 antibodies. CLPs also failed to prevent the clinical manifestation and virus replication, but in comparison to controls, the severity of disease manifestation and viraemia was mitigated. The data demonstrated that the outer capsid was essential for complete protection, while the geographical origin of the BTV was not critical for development of a serotype specific vaccine.
Collapse
Affiliation(s)
- M Stewart
- Department of Infectious Diseases, London School of Hygiene and Tropical Medicine, United Kingdom
| | | | | | | | | | | | | |
Collapse
|