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] [Download PDF] [Figures] [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
|
Liu L, Wang J, Li R, Wu J, Zhao Y, Yan F, Wang T, Gao Y, Zhao Z, Feng N, Xia X. A Bacterium-like Particle Vaccine Displaying Envelope Proteins of Canine Distemper Virus Can Induce Immune Responses in Mice and Dogs. Viruses 2024; 16:549. [PMID: 38675892 PMCID: PMC11055036 DOI: 10.3390/v16040549] [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: 02/02/2024] [Revised: 03/22/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
Canine distemper virus (CDV) can cause fatal infections in giant pandas. Vaccination is crucial to prevent CDV infection in giant pandas. In this study, two bacterium-like particle vaccines F3-GEM and H4-GEM displaying the trimeric F protein or tetrameric H protein of CDV were constructed based on the Gram-positive enhanced-matrix protein anchor (GEM-PA) surface display system. Electron microscopy and Western blot results revealed that the F or H protein was successfully anchored on the surface of GEM particles. Furthermore, one more bacterium-like particle vaccine F3 and H4-GEM was also designed, a mixture consisting of F3-GEM and H4-GEM at a ratio of 1:1. To evaluate the effect of the three vaccines, mice were immunized with F3-GEM, H4-GEM or F3 and H4-GEM. It was found that the level of IgG-specific antibodies and neutralizing antibodies in the F3 and H4-GEM group was higher than the other two groups. Additionally, F3 and H4-GEM also increased the secretion of Th1-related and Th2-related cytokines. Moreover, F3 and H4-GEM induce IgG and neutralizing antibodies' response in dogs. Conclusions: In summary, F3 and H4-GEM can provoke better immune responses to CDV in mice and dogs. The bacterium-like particle vaccine F3 and H4-GEM might be a potential vaccine candidate for giant pandas against CDV infection.
Collapse
Affiliation(s)
- Lina Liu
- College of Veterinary Medicine, Jilin University, Changchun 130000, China;
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (R.L.); (J.W.); (Y.Z.); (F.Y.); (T.W.); (Y.G.); (Z.Z.)
| | - Jianzhong Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun 130122, China;
| | - Ranran Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (R.L.); (J.W.); (Y.Z.); (F.Y.); (T.W.); (Y.G.); (Z.Z.)
- College of Veterinary Medicine, Jilin Agricultural University, Changchun 130122, China;
| | - Jianzhao Wu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (R.L.); (J.W.); (Y.Z.); (F.Y.); (T.W.); (Y.G.); (Z.Z.)
- College of Veterinary Medicine, Jilin Agricultural University, Changchun 130122, China;
| | - Yongkun Zhao
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (R.L.); (J.W.); (Y.Z.); (F.Y.); (T.W.); (Y.G.); (Z.Z.)
| | - Feihu Yan
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (R.L.); (J.W.); (Y.Z.); (F.Y.); (T.W.); (Y.G.); (Z.Z.)
| | - Tiecheng Wang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (R.L.); (J.W.); (Y.Z.); (F.Y.); (T.W.); (Y.G.); (Z.Z.)
| | - Yuwei Gao
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (R.L.); (J.W.); (Y.Z.); (F.Y.); (T.W.); (Y.G.); (Z.Z.)
| | - Zongzheng Zhao
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (R.L.); (J.W.); (Y.Z.); (F.Y.); (T.W.); (Y.G.); (Z.Z.)
| | - Na Feng
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (R.L.); (J.W.); (Y.Z.); (F.Y.); (T.W.); (Y.G.); (Z.Z.)
- College of Veterinary Medicine, Jilin Agricultural University, Changchun 130122, China;
| | - Xianzhu Xia
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (R.L.); (J.W.); (Y.Z.); (F.Y.); (T.W.); (Y.G.); (Z.Z.)
- College of Veterinary Medicine, Jilin Agricultural University, Changchun 130122, China;
| |
Collapse
|
3
|
Huang J, Cortey M, Darwich L, Griffin J, Obón E, Molina R, Martín M. Study of Canine Distemper Virus Presence in Catalonia's Wild Carnivores through H Gene Amplification and Sequencing. Animals (Basel) 2024; 14:436. [PMID: 38338078 PMCID: PMC10854788 DOI: 10.3390/ani14030436] [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: 12/30/2023] [Revised: 01/22/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
Canine distemper virus (CDV) is recognised worldwide as an important pathogen in both domestic and wild carnivores. Few data are available on its impact and spread on the wildlife/wildlife-domestic animal-environment interface. This study, aimed at developing a conservation-oriented control strategy, analysed 89 sick or deceased animals from 2019 to 2023 at the Wildlife Rehabilitation Centre in Torreferrussa. RT-PCR and sequencing of the partial H gene were used to detect and analyse CDV in tissues. The total positive percentage was 20.22% (18/89), comprising 13 red foxes (44.8%), 4 European badgers (28.6%), and 1 American mink (4.5%), while 24 Eurasian otters tested negative. Phylogenetic analysis indicated that all of the CDV strains belong to the European lineage. Geographically distant individuals and different species shared the same viral strain, suggesting a strong capacity of CDV for interspecies and long-distance transmission. This calls for further research, particularly focusing on potential impacts of CDV on endangered carnivores.
Collapse
Affiliation(s)
- Junhao Huang
- Department of Animal Health and Anatomy, Autonomous University of Barcelona (UAB), 08193 Cerdanyola del Vallès, Spain; (J.H.); (M.C.); (L.D.); (J.G.)
| | - Martí Cortey
- Department of Animal Health and Anatomy, Autonomous University of Barcelona (UAB), 08193 Cerdanyola del Vallès, Spain; (J.H.); (M.C.); (L.D.); (J.G.)
| | - Laila Darwich
- Department of Animal Health and Anatomy, Autonomous University of Barcelona (UAB), 08193 Cerdanyola del Vallès, Spain; (J.H.); (M.C.); (L.D.); (J.G.)
| | - Jenna Griffin
- Department of Animal Health and Anatomy, Autonomous University of Barcelona (UAB), 08193 Cerdanyola del Vallès, Spain; (J.H.); (M.C.); (L.D.); (J.G.)
| | - Elena Obón
- Torreferrussa Wildlife Rehabilitation Centre, Catalan Wildlife Service-Forestal Catalana S.A., 08130 Santa Perpètua de Mogoda, Spain;
| | - Rafael Molina
- Torreferrussa Wildlife Rehabilitation Centre, Catalan Wildlife Service-Forestal Catalana S.A., 08130 Santa Perpètua de Mogoda, Spain;
| | - Margarita Martín
- Department of Animal Health and Anatomy, Autonomous University of Barcelona (UAB), 08193 Cerdanyola del Vallès, Spain; (J.H.); (M.C.); (L.D.); (J.G.)
| |
Collapse
|
4
|
Espinoza I, García Iglesias MJ, Oleaga Á, de Garnica García MG, Balseiro A. Phenotypic Characterization of Encephalitis in the BRAINS of Badgers Naturally Infected with Canine Distemper Virus. Animals (Basel) 2023; 13:3360. [PMID: 37958115 PMCID: PMC10647365 DOI: 10.3390/ani13213360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 10/26/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Canine distemper virus (CDV) affects a huge diversity of domestic and wild carnivores, with increasing numbers of mortality events worldwide. The local cell-mediated immune response elicited against a natural infection is an important factor in determining the outcome of CDV infection. Therefore, the purposes of this study were to describe the local immune response within the central nervous systems (CNSs) of seven badgers naturally infected with CDV in Asturias (Atlantic Spain) and to determine the phenotype and distribution of microglial cells, T and B lymphocytes, and astrocytes in the foci of gliosis located in the thalamus and cerebellum using immunohistochemistry. The immunohistochemical assessment demonstrated the presence of Iba1-positive microglia and GFAP-positive astrocytes in the foci of gliosis, whereas T (CD3-negative) or B (CD20-negative) lymphocytes in those same lesions were absent. Our results also revealed that the badgers with natural CDV encephalitis presented lesions mostly located in the white matter of the thalamus and cerebellum, suggesting a CDV-specific tropism for the white matter of badger brains in those locations. The knowledge gained in the field of the immunopathogenesis of distemper disease affecting the CNSs of badgers could help to clarify CDV disease patterns in this species.
Collapse
Affiliation(s)
- Israel Espinoza
- Departamento de Sanidad Animal, Universidad de León, 24071 León, Spain; (I.E.); (M.J.G.I.); (M.G.d.G.G.)
| | - María José García Iglesias
- Departamento de Sanidad Animal, Universidad de León, 24071 León, Spain; (I.E.); (M.J.G.I.); (M.G.d.G.G.)
- Instituto Universitario (LOU) de Biomedicina (IBIOMED), Universidad de Léon, 24071 León, Spain
| | - Álvaro Oleaga
- Sociedad de Servicios del Principado de Asturias S.A. (SERPA), 33203 Gijón, Spain;
| | - María Gracia de Garnica García
- Departamento de Sanidad Animal, Universidad de León, 24071 León, Spain; (I.E.); (M.J.G.I.); (M.G.d.G.G.)
- Micros Veterinaria, S.L., 24007 León, Spain
| | - Ana Balseiro
- Departamento de Sanidad Animal, Universidad de León, 24071 León, Spain; (I.E.); (M.J.G.I.); (M.G.d.G.G.)
- Departamento de Sanidad Animal, Instituto de Ganadería de Montaña (CSIC—Universidad de León), 24346 León, Spain
| |
Collapse
|
5
|
Guercio A, Mira F, Di Bella S, Gucciardi F, Lastra A, Purpari G, Castronovo C, Pennisi M, Di Marco Lo Presti V, Rizzo M, Giudice E. Biomolecular Analysis of Canine Distemper Virus Strains in Two Domestic Ferrets ( Mustela putorius furo). Vet Sci 2023; 10:375. [PMID: 37368761 DOI: 10.3390/vetsci10060375] [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: 03/23/2023] [Revised: 05/19/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
Canine distemper is a contagious and severe systemic viral disease that affects domestic and wild carnivores worldwide. In this study, two adult female ferrets (Mustela putorius furo) were evaluated for cutaneous lesions. Scab, fur, and swab samples from the external auditory canal, cutaneous lesions, and scrapings were analyzed. Canine distemper virus (CDV)-positive samples underwent RT-PCR/RFLP with the restriction enzyme PsiI, and the hemagglutinin gene sequence was obtained. According to the restriction enzyme and sequence analyses, the viral strains were typed as CDV field strains that are included within the Europe lineage and distinct from those including vaccinal CDV strains. The sequence analysis showed the highest nucleotide identity rates in older Europe lineage CDV strains collected from dogs and a fox in Europe. This study is the first to report on CDV infection in ferrets in southern Italy and contributes to the current knowledge about natural CDV infection in this species. In conclusion, vaccination remains crucial for preventing the disease and counteracting cross-species infection. Molecular biology techniques can enable the monitoring of susceptible wild animals by ensuring the active surveillance of CDV spread.
Collapse
Affiliation(s)
- Annalisa Guercio
- Istituto Zooprofilattico Sperimentale della Sicilia "A. Mirri", Via G. Marinuzzi, 3, 90129 Palermo, Italy
| | - Francesco Mira
- Istituto Zooprofilattico Sperimentale della Sicilia "A. Mirri", Via G. Marinuzzi, 3, 90129 Palermo, Italy
- Department of Veterinary Science, University of Messina, Polo Universitario dell'Annunziata, 98168 Messina, Italy
| | - Santina Di Bella
- Istituto Zooprofilattico Sperimentale della Sicilia "A. Mirri", Via G. Marinuzzi, 3, 90129 Palermo, Italy
| | - Francesca Gucciardi
- Istituto Zooprofilattico Sperimentale della Sicilia "A. Mirri", Via G. Marinuzzi, 3, 90129 Palermo, Italy
| | - Antonio Lastra
- Istituto Zooprofilattico Sperimentale della Sicilia "A. Mirri", Via G. Marinuzzi, 3, 90129 Palermo, Italy
| | - Giuseppa Purpari
- Istituto Zooprofilattico Sperimentale della Sicilia "A. Mirri", Via G. Marinuzzi, 3, 90129 Palermo, Italy
| | - Calogero Castronovo
- Istituto Zooprofilattico Sperimentale della Sicilia "A. Mirri", Via G. Marinuzzi, 3, 90129 Palermo, Italy
| | - Melissa Pennisi
- Department of Veterinary Science, University of Messina, Polo Universitario dell'Annunziata, 98168 Messina, Italy
| | | | - Maria Rizzo
- Department of Veterinary Science, University of Messina, Polo Universitario dell'Annunziata, 98168 Messina, Italy
| | - Elisabetta Giudice
- Department of Veterinary Science, University of Messina, Polo Universitario dell'Annunziata, 98168 Messina, Italy
| |
Collapse
|
6
|
Karki M, Rajak KK, Singh RP. Canine morbillivirus (CDV): a review on current status, emergence and the diagnostics. Virusdisease 2022; 33:309-321. [PMID: 36039286 PMCID: PMC9403230 DOI: 10.1007/s13337-022-00779-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 07/15/2022] [Indexed: 11/12/2022] Open
Abstract
The increasing host range of canine morbillivirus (CDV) affecting important wildlife species such as Lions, Leopard, and Red Pandas has raised the concern. Canine distemper is a pathogen of dogs affecting the respiratory, gastrointestinal, and nervous systems. Seventeen lineages of CDV are reported, and the eighteenth lineage was proposed in 2019 from India. Marked genomic differences in the genome of wild-type virus and vaccine strain are also reported.The variations at the epitope level can be differentiated using specific monoclonal antibodies in neutralization tests. Keeping in mind the current status of the emergence of CDV, genetic and molecular study of circulating strains of the specific geographical region are the essential components of the disease control strategy. New target-based diagnostics and vaccines are in need to counter the effects of the emerging virus population. Control of CDV is necessary to save the endangered, vulnerable, and many other wildlife species to maintain balance in the ecological system. This review provides an overview on emergence reported in CDV, diagnostics developed till today, and a perspective on the disease control strategy, keeping wildlife in consideration.
Collapse
|
7
|
Vrba SM, Kirk NM, Brisse ME, Liang Y, Ly H. Development and Applications of Viral Vectored Vaccines to Combat Zoonotic and Emerging Public Health Threats. Vaccines (Basel) 2020; 8:E680. [PMID: 33202961 PMCID: PMC7712223 DOI: 10.3390/vaccines8040680] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 02/07/2023] Open
Abstract
Vaccination is arguably the most cost-effective preventative measure against infectious diseases. While vaccines have been successfully developed against certain viruses (e.g., yellow fever virus, polio virus, and human papilloma virus HPV), those against a number of other important public health threats, such as HIV-1, hepatitis C, and respiratory syncytial virus (RSV), have so far had very limited success. The global pandemic of COVID-19, caused by the SARS-CoV-2 virus, highlights the urgency of vaccine development against this and other constant threats of zoonotic infection. While some traditional methods of producing vaccines have proven to be successful, new concepts have emerged in recent years to produce more cost-effective and less time-consuming vaccines that rely on viral vectors to deliver the desired immunogens. This review discusses the advantages and disadvantages of different viral vaccine vectors and their general strategies and applications in both human and veterinary medicines. A careful review of these issues is necessary as they can provide important insights into how some of these viral vaccine vectors can induce robust and long-lasting immune responses in order to provide protective efficacy against a variety of infectious disease threats to humans and animals, including those with zoonotic potential to cause global pandemics.
Collapse
Affiliation(s)
- Sophia M. Vrba
- Department of Veterinary & Biomedical Sciences, University of Minnesota, Twin Cities, St. Paul, MN 55108, USA; (S.M.V.); (Y.L.)
| | - Natalie M. Kirk
- Comparative Molecular Biosciences Graduate Program, Department of Veterinary & Biomedical Sciences, University of Minnesota, Twin Cities, St. Paul, MN 55108, USA;
| | - Morgan E. Brisse
- Biochemistry, Molecular Biology and Biophysics Graduate Program, Department of Veterinary & Biomedical Sciences, University of Minnesota, Twin Cities, St. Paul, MN 55108, USA;
| | - Yuying Liang
- Department of Veterinary & Biomedical Sciences, University of Minnesota, Twin Cities, St. Paul, MN 55108, USA; (S.M.V.); (Y.L.)
| | - Hinh Ly
- Department of Veterinary & Biomedical Sciences, University of Minnesota, Twin Cities, St. Paul, MN 55108, USA; (S.M.V.); (Y.L.)
| |
Collapse
|
8
|
Brisse M, Vrba SM, Kirk N, Liang Y, Ly H. Emerging Concepts and Technologies in Vaccine Development. Front Immunol 2020; 11:583077. [PMID: 33101309 PMCID: PMC7554600 DOI: 10.3389/fimmu.2020.583077] [Citation(s) in RCA: 176] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 09/14/2020] [Indexed: 01/05/2023] Open
Abstract
Despite the success of vaccination to greatly mitigate or eliminate threat of diseases caused by pathogens, there are still known diseases and emerging pathogens for which the development of successful vaccines against them is inherently difficult. In addition, vaccine development for people with compromised immunity and other pre-existing medical conditions has remained a major challenge. Besides the traditional inactivated or live attenuated, virus-vectored and subunit vaccines, emerging non-viral vaccine technologies, such as viral-like particle and nanoparticle vaccines, DNA/RNA vaccines, and rational vaccine design, offer innovative approaches to address existing challenges of vaccine development. They have also significantly advanced our understanding of vaccine immunology and can guide future vaccine development for many diseases, including rapidly emerging infectious diseases, such as COVID-19, and diseases that have not traditionally been addressed by vaccination, such as cancers and substance abuse. This review provides an integrative discussion of new non-viral vaccine development technologies and their use to address the most fundamental and ongoing challenges of vaccine development.
Collapse
Affiliation(s)
- Morgan Brisse
- Biochemistry, Molecular Biology, and Biophysics Graduate Program, University of Minnesota Twin Cities, St. Paul, MN, United States
- Department of Veterinary & Biomedical Sciences, University of Minnesota Twin Cities, St. Paul, MN, United States
| | - Sophia M. Vrba
- Department of Veterinary & Biomedical Sciences, University of Minnesota Twin Cities, St. Paul, MN, United States
| | - Natalie Kirk
- Department of Veterinary & Biomedical Sciences, University of Minnesota Twin Cities, St. Paul, MN, United States
- Comparative Molecular Biosciences Graduate Program, University of Minnesota Twin Cities, St. Paul, MN, United States
| | - Yuying Liang
- Department of Veterinary & Biomedical Sciences, University of Minnesota Twin Cities, St. Paul, MN, United States
| | - Hinh Ly
- Department of Veterinary & Biomedical Sciences, University of Minnesota Twin Cities, St. Paul, MN, United States
| |
Collapse
|
9
|
Chen C, Liang H, Hu B, Ning B, Lai H, He Y, Guo G, Zhong S, Li L. Determination of parvovirus antigen in the vaccine using time-resolved fluorescence immunoassay. Biotechnol Appl Biochem 2020; 68:597-602. [PMID: 32533780 DOI: 10.1002/bab.1967] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 06/06/2020] [Indexed: 12/16/2022]
Abstract
As a highly contagious and potentially fatal disease of dogs, canine parvovirus type 2 (CPV-2) usually causes severe myocarditis and gastroenteritis, while vaccine injection has greatly reduced the incidence of CPV-2 diseases. However, there is currently a lack of simple and effective method for quantitative detection of CPV-2 in vaccine. Therefore, this study aims to prepare an accurate method to determine the CPV-2 antigen (CPV-2-Ag) in vaccine. Here, a sandwich time-resolved fluorescence immunoassay (TRFIA) was established and optimized. Anti-CPV-2 antibodies were immobilized on 96-well plates to capture CPV-2-Ag, and then bound together with the detection antibodies labeled with Europium(III) (Eu3+ ) chelates; finally, time-resolved fluorometry was employed to measure the fluorescence intensity. Vaccination was performed to evaluate the relationship between CPV-2-Ag concentration and antibody titer. The sensitivity is 1.15 mEU/mL (LogY = 1.524 + 0.8667 × LogX, R2 = 0.9933), and the average recovery is among 91.00% to 106.39% without cross-reactions with the other canine viral antigen. The correlation between ELISA assay and this method is up to 0.9861. And, there is high correlation between the CPV-2-Ag concentration and antibody titers (R2 = 0.9234). This immunoassay established has high sensitivity, accuracy, and specificity, which indicate that this method could be suitable for quantitative detection of CPV-2-Ag in vaccine evaluation.
Collapse
Affiliation(s)
- Cuicui Chen
- Guangzhou Youdi Bio-Technology Co., Ltd., Guangzhou, 510663, China
| | - Huankun Liang
- Guangzhou Youdi Bio-Technology Co., Ltd., Guangzhou, 510663, China
| | - Baoyu Hu
- Guangzhou Youdi Bio-Technology Co., Ltd., Guangzhou, 510663, China
| | - Bo Ning
- Guangzhou Youdi Bio-Technology Co., Ltd., Guangzhou, 510663, China
| | - Hongrui Lai
- Guangzhou Youdi Bio-Technology Co., Ltd., Guangzhou, 510663, China
| | - Ying He
- Guangzhou Youdi Bio-Technology Co., Ltd., Guangzhou, 510663, China
| | - Guiling Guo
- Guangzhou Youdi Bio-Technology Co., Ltd., Guangzhou, 510663, China
| | - Shuhai Zhong
- Guangzhou Youdi Bio-Technology Co., Ltd., Guangzhou, 510663, China
| | - Laiqing Li
- Guangzhou Youdi Bio-Technology Co., Ltd., Guangzhou, 510663, China
| |
Collapse
|
10
|
Willis E, Pardi N, Parkhouse K, Mui BL, Tam YK, Weissman D, Hensley SE. Nucleoside-modified mRNA vaccination partially overcomes maternal antibody inhibition of de novo immune responses in mice. Sci Transl Med 2020; 12:eaav5701. [PMID: 31915303 PMCID: PMC7339908 DOI: 10.1126/scitranslmed.aav5701] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 06/21/2019] [Accepted: 09/23/2019] [Indexed: 12/12/2022]
Abstract
Maternal antibodies provide short-term protection to infants against many infections. However, they can inhibit de novo antibody responses in infants elicited by infections or vaccination, leading to increased long-term susceptibility to infectious diseases. Thus, there is a need to develop vaccines that are able to elicit protective immune responses in the presence of antigen-specific maternal antibodies. Here, we used a mouse model to demonstrate that influenza virus-specific maternal antibodies inhibited de novo antibody responses in mouse pups elicited by influenza virus infection or administration of conventional influenza vaccines. We found that a recently developed influenza vaccine, nucleoside-modified mRNA encapsulated in lipid nanoparticles (mRNA-LNP), partially overcame this inhibition by maternal antibodies. The mRNA-LNP influenza vaccine established long-lived germinal centers in the mouse pups and elicited stronger antibody responses than did a conventional influenza vaccine approved for use in humans. Vaccination with mRNA-LNP vaccines may offer a promising strategy for generating robust immune responses in infants in the presence of maternal antibodies.
Collapse
Affiliation(s)
- Elinor Willis
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Norbert Pardi
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kaela Parkhouse
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Ying K Tam
- Acuitas Therapeutics, Vancouver, BC V6T 1Z3, Canada
| | - Drew Weissman
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Scott E Hensley
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
11
|
Chen C, Guo X, Liang H, Ning B, Li J, Zhong S, Liu X, Li L. Determination of parvovirus antibodies in canine serum using magnetic bead-based chemiluminescence immunoassay. Biotechnol Appl Biochem 2019; 66:586-590. [PMID: 31039295 PMCID: PMC7161764 DOI: 10.1002/bab.1758] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 03/16/2019] [Indexed: 02/02/2023]
Abstract
Canine parvovirus type 2 (CPV-2), as a highly contagious and potentially fatal disease of dogs and many other carnivores, usually causes severe gastroenteritis and myocarditis. Therefore, it is very necessary and urgent to have an accurate method to determine the CPV-2 antibodies (CPV-2-Ab) in canine samples. Here, a magnetic bead-based chemiluminescence immunoassay was established and optimized to detect the concentration of CPV-2-Ab in serum. And a commercial assay was also used to evaluate the consistency with our method. After optimization of the detective system, the CPV-2-Ab was captured by CPV-antigen-magnetic bead (8.3 µg/mL); then combined with the conjugation of anti-canine IgG antibody-acridinium ester (0.36 µg/mL). Finally, collected the signal (read the luminosity) after 1 H reaction time. The linear correlation coefficient (R2 ) is 0.9924. The limit of detection (sensitivity) is 0.36 ng/mL (the linear dynamic range: 1.32-93.75 ng/mL), and the average recovery is 100.89% without cross-reactions with other canine viral antibodies. The results' correlation between commercial assays and this method is 0.9888. This immunoassay establishes that it has high sensitivity, accuracy, and specificity in clinical analysis, indicating that this method could be suitable for quantitative detection of CPV-2-Ab and evaluation of vaccination effect.
Collapse
Affiliation(s)
- Cuicui Chen
- Guangzhou Youdi Bio-technology Co., Ltd., Guangzhou, 510663, China
| | - Xiaoxiao Guo
- Guangzhou Youdi Bio-technology Co., Ltd., Guangzhou, 510663, China
| | - Huankun Liang
- Guangzhou Youdi Bio-technology Co., Ltd., Guangzhou, 510663, China
| | - Bo Ning
- Guangzhou Youdi Bio-technology Co., Ltd., Guangzhou, 510663, China
| | - Jiexing Li
- Guangzhou Youdi Bio-technology Co., Ltd., Guangzhou, 510663, China
| | - Shuhai Zhong
- Guangzhou Youdi Bio-technology Co., Ltd., Guangzhou, 510663, China
| | - Xipan Liu
- Guangzhou Youdi Bio-technology Co., Ltd., Guangzhou, 510663, China
| | - Laiqing Li
- Guangzhou Youdi Bio-technology Co., Ltd., Guangzhou, 510663, China
| |
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
|
Inactivated Recombinant Rabies Viruses Displaying Canine Distemper Virus Glycoproteins Induce Protective Immunity against Both Pathogens. J Virol 2017; 91:JVI.02077-16. [PMID: 28148801 DOI: 10.1128/jvi.02077-16] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 01/25/2017] [Indexed: 12/25/2022] Open
Abstract
The development of multivalent vaccines is an attractive methodology for the simultaneous prevention of several infectious diseases in vulnerable populations. Both canine distemper virus (CDV) and rabies virus (RABV) cause lethal disease in wild and domestic carnivores. While RABV vaccines are inactivated, the live-attenuated CDV vaccines retain residual virulence for highly susceptible wildlife species. In this study, we developed recombinant bivalent vaccine candidates based on recombinant vaccine strain rabies virus particles, which concurrently display the protective CDV and RABV glycoprotein antigens. The recombinant viruses replicated to near-wild-type titers, and the heterologous glycoproteins were efficiently expressed and incorporated in the viral particles. Immunization of ferrets with beta-propiolactone-inactivated recombinant virus particles elicited protective RABV antibody titers, and animals immunized with a combination of CDV attachment protein- and fusion protein-expressing recombinant viruses were protected from lethal CDV challenge. However, animals that were immunized with only a RABV expressing the attachment protein of CDV vaccine strain Onderstepoort succumbed to infection with a more recent wild-type strain, indicating that immune responses to the more conserved fusion protein contribute to protection against heterologous CDV strains.IMPORTANCE Rabies virus and canine distemper virus (CDV) cause high mortality rates and death in many carnivores. While rabies vaccines are inactivated and thus have an excellent safety profile and high stability, live-attenuated CDV vaccines can retain residual virulence in highly susceptible species. Here we generated recombinant inactivated rabies viruses that carry one of the CDV glycoproteins on their surface. Ferrets immunized twice with a mix of recombinant rabies viruses carrying the CDV fusion and attachment glycoproteins were protected from lethal CDV challenge, whereas all animals that received recombinant rabies viruses carrying only the CDV attachment protein according to the same immunization scheme died. Irrespective of the CDV antigens used, all animals developed protective titers against rabies virus, illustrating that a bivalent rabies virus-based vaccine against CDV induces protective immune responses against both pathogens.
Collapse
|
14
|
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
|
15
|
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
|
16
|
Sánchez-Sampedro L, Perdiguero B, Mejías-Pérez E, García-Arriaza J, Di Pilato M, Esteban M. The evolution of poxvirus vaccines. Viruses 2015; 7:1726-803. [PMID: 25853483 PMCID: PMC4411676 DOI: 10.3390/v7041726] [Citation(s) in RCA: 149] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 03/16/2015] [Accepted: 03/27/2015] [Indexed: 02/07/2023] Open
Abstract
After Edward Jenner established human vaccination over 200 years ago, attenuated poxviruses became key players to contain the deadliest virus of its own family: Variola virus (VARV), the causative agent of smallpox. Cowpox virus (CPXV) and horsepox virus (HSPV) were extensively used to this end, passaged in cattle and humans until the appearance of vaccinia virus (VACV), which was used in the final campaigns aimed to eradicate the disease, an endeavor that was accomplished by the World Health Organization (WHO) in 1980. Ever since, naturally evolved strains used for vaccination were introduced into research laboratories where VACV and other poxviruses with improved safety profiles were generated. Recombinant DNA technology along with the DNA genome features of this virus family allowed the generation of vaccines against heterologous diseases, and the specific insertion and deletion of poxvirus genes generated an even broader spectrum of modified viruses with new properties that increase their immunogenicity and safety profile as vaccine vectors. In this review, we highlight the evolution of poxvirus vaccines, from first generation to the current status, pointing out how different vaccines have emerged and approaches that are being followed up in the development of more rational vaccines against a wide range of diseases.
Collapse
MESH Headings
- Animals
- History, 18th Century
- History, 19th Century
- History, 20th Century
- History, 21st Century
- Humans
- Poxviridae/immunology
- Poxviridae/isolation & purification
- Smallpox/prevention & control
- Smallpox Vaccine/history
- Smallpox Vaccine/immunology
- Smallpox Vaccine/isolation & purification
- Vaccines, Attenuated/history
- Vaccines, Attenuated/immunology
- Vaccines, Attenuated/isolation & purification
- Vaccines, Synthetic/history
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/isolation & purification
Collapse
Affiliation(s)
- Lucas Sánchez-Sampedro
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid-28049, Spain.
| | - Beatriz Perdiguero
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid-28049, Spain.
| | - Ernesto Mejías-Pérez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid-28049, Spain
| | - Juan García-Arriaza
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid-28049, Spain
| | - Mauro Di Pilato
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid-28049, Spain.
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid-28049, Spain.
| |
Collapse
|
17
|
Enkirch T, von Messling V. Ferret models of viral pathogenesis. Virology 2015; 479-480:259-70. [PMID: 25816764 PMCID: PMC7111696 DOI: 10.1016/j.virol.2015.03.017] [Citation(s) in RCA: 126] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 01/28/2015] [Accepted: 03/02/2015] [Indexed: 11/26/2022]
Abstract
Emerging and well-known viral diseases remain one the most important global public health threats. A better understanding of their pathogenesis and mechanisms of transmission requires animal models that accurately reproduce these aspects of the disease. Here we review the role of ferrets as an animal model for the pathogenesis of different respiratory viruses with an emphasis on influenza and paramyxoviruses. We will describe the anatomic and physiologic characteristics that contribute to the natural susceptibility of ferrets to these viruses, and provide an overview of the approaches available to analyze their immune responses. Recent insights gained using this model will be highlighted, including the development of new prophylactic and therapeutic approaches. To provide decision criteria for the use of this animal model, its strengths and limitations will be discussed. Ferrets as models for respiratory virus pathogenesis. Ferrets as models for vaccine and drug efficacy assessment. Immunological tools for ferrets. Housing and handling of ferrets.
Collapse
Affiliation(s)
- T Enkirch
- Veterinary Medicine Division, Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Paul-Ehrlich-Straße 51-59, 63225 Langen, Germany
| | - V von Messling
- Veterinary Medicine Division, Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Paul-Ehrlich-Straße 51-59, 63225 Langen, Germany.
| |
Collapse
|
18
|
Canine distemper virus DNA vaccination of mink can overcome interference by maternal antibodies. Vaccine 2015; 33:1375-81. [PMID: 25637861 DOI: 10.1016/j.vaccine.2015.01.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 11/22/2014] [Accepted: 01/09/2015] [Indexed: 11/24/2022]
Abstract
Canine distemper virus (CDV) is highly contagious and can cause severe disease against which conventional live vaccines are ineffective in the presence of maternal antibodies. Vaccination in the presences of maternal antibodies was challenged by vaccination of 5 days old and 3 weeks old mink kits with CDV DNA vaccines. Virus neutralising (VN) antibody responses were induced in mink kits vaccinated with a plasmid encoding the haemaglutinin protein (H) of CDV (n=5, pCDV-H) or a combination of the H, fusion (F) and nucleoprotein (N) of CDV (n=5, pCDV-HFN). These DNA vaccinated kits were protected against virulent experimental infection with field strains of CDV. The pCDV-H was more efficient in inducing protective immunity in the presence of maternal antibodies compared to the pCDV-HFN. The results show that DNA vaccination with the pCDV-H or pCDV-HFN (n=4) only given once at 5 days of age induces virus specific immune response in neonatal mink and protection against virulent CDV exposure later in life.
Collapse
|
19
|
Niewiesk S. Maternal antibodies: clinical significance, mechanism of interference with immune responses, and possible vaccination strategies. Front Immunol 2014; 5:446. [PMID: 25278941 PMCID: PMC4165321 DOI: 10.3389/fimmu.2014.00446] [Citation(s) in RCA: 343] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 09/01/2014] [Indexed: 01/28/2023] Open
Abstract
Neonates have an immature immune system, which cannot adequately protect against infectious diseases. Early in life, immune protection is accomplished by maternal antibodies transferred from mother to offspring. However, decaying maternal antibodies inhibit vaccination as is exemplified by the inhibition of seroconversion after measles vaccination. This phenomenon has been described in both human and veterinary medicine and is independent of the type of vaccine being used. This review will discuss the use of animal models for vaccine research. I will review clinical solutions for inhibition of vaccination by maternal antibodies, and the testing and development of potentially effective vaccines. These are based on new mechanistic insight about the inhibitory mechanism of maternal antibodies. Maternal antibodies inhibit the generation of antibodies whereas the T cell response is usually unaffected. B cell inhibition is mediated through a cross-link between B cell receptor (BCR) with the Fcγ-receptor IIB by a vaccine-antibody complex. In animal experiments, this inhibition can be partially overcome by injection of a vaccine-specific monoclonal IgM antibody. IgM stimulates the B cell directly through cross-linking the BCR via complement protein C3d and antigen to the complement receptor 2 (CR2) signaling complex. In addition, it was shown that interferon alpha binds to the CD21 chain of CR2 as well as the interferon receptor and that this dual receptor usage drives B cell responses in the presence of maternal antibodies. In lieu of immunizing the infant, the concept of maternal immunization as a strategy to protect neonates has been proposed. This approach would still not solve the question of how to immunize in the presence of maternal antibodies but would defer the time of infection to an age where infection might not have such a detrimental outcome as in neonates. I will review successful examples and potential challenges of implementing this concept.
Collapse
Affiliation(s)
- Stefan Niewiesk
- Department of Veterinary Biosciences, The Ohio State University , Columbus, OH , USA
| |
Collapse
|
20
|
Abstract
Vaccines remain one of the practitioner's greatest tools in preventing disease and maintaining individual and population health. This article is an update to "Current Vaccination Strategies in Puppies and Kittens" published in Veterinary Clinics of North America, Small Animal Practitioner, in May 2006. There are now comprehensive guidelines readily available for small animal practitioners regarding canine and feline pediatric (and adult) vaccination recommendations. Perhaps more importantly, there is an increased dialogue regarding all aspects of preventive medicine, of which vaccination is only a small, yet significant portion; and an increased drive to provide scientific evidence for developing vaccination recommendations.
Collapse
Affiliation(s)
- Gina M Davis-Wurzler
- Outpatient Medicine, Department of Veterinary Medicine and Epidemiology, School of Veterinary Medicine, University of California, One Garrod Drive, Davis, Davis, CA 95616, USA; Small Animal Outpatient Medicine Service, William R. Pritchard Veterinary Medical Teaching Hospital, Davis, CA 95616, USA.
| |
Collapse
|
21
|
Azizi A, Edamura KN, Leung G, Gisonni-Lex L, Mallet L. Short communication: evaluating the level of expressed HIV type 1 gp120 and gag proteins in the vCP1521 vector by two immunoplaque methods. AIDS Res Hum Retroviruses 2013; 29:397-9. [PMID: 22992109 DOI: 10.1089/aid.2012.0212] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Over the past few years, several recombinant ALVAC constructs have been used as delivery systems in various vaccine research studies and trials. The ALVAC-HIV vCP1521 vector has been used as a vaccine delivery system in the RV144 study, a phase III HIV study that displayed over 31% protective efficacy. One of the important parameters for evaluating the potency of an ALVAC construct is the stable expression of proteins encoded by the inserted genes. Herein, the expression of inserted gp120 and gag genes in two manufactured ALVAC-HIV vCP1521 lots have been determined by two immunoplaque methods (dish and plaque lift). Both methods were specific and robust and demonstrated that the ALVAC-HIV vCP1521 lots were able to express gp120 and gag proteins in over 99% of the infectious plaques.
Collapse
Affiliation(s)
- Ali Azizi
- Microbiology and Virology Platform, Department of Analytical Research and Development North America, Sanofi Pasteur, Toronto, Ontario, Canada
| | - Kerrie Nichol Edamura
- Microbiology and Virology Platform, Department of Analytical Research and Development North America, Sanofi Pasteur, Toronto, Ontario, Canada
| | - Glenda Leung
- Microbiology and Virology Platform, Department of Analytical Research and Development North America, Sanofi Pasteur, Toronto, Ontario, Canada
| | - Lucy Gisonni-Lex
- Microbiology and Virology Platform, Department of Analytical Research and Development North America, Sanofi Pasteur, Toronto, Ontario, Canada
| | - Laurent Mallet
- Microbiology and Virology Platform, Department of Analytical Research and Development North America, Sanofi Pasteur, Toronto, Ontario, Canada
| |
Collapse
|
22
|
|
23
|
Barron HW, Rosenthal KL. Respiratory Diseases. FERRETS, RABBITS, AND RODENTS 2012. [PMCID: PMC7152247 DOI: 10.1016/b978-1-4160-6621-7.00006-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
24
|
Abstract
Canine distemper virus (CDV) causes a major disease of domestic dogs that develops as a serious systemic infection in unvaccinated or improperly vaccinated dogs. Domesticated dogs are the main reservoir of CDV, a multihost pathogen. This virus of the genus Morbillivirus in the family Paramyxoviridae occurs in other carnivorous species including all members of the Canidae and Mustelidae families and in some members of the Procyonidae, Hyaenidae, Ursidae, and Viverridae families. Canine distemper also has been reported in the Felidae family and marine mammals. The spread and incidences of CDV epidemics in dogs and wildlife here and worldwide are increasing.
Collapse
Affiliation(s)
- Sanjay Kapil
- Department of Veterinary Pathobiology, Oklahoma Animal Disease Diagnostic Laboratory, Center for Veterinary Health Sciences, Farm and Ridge Road, Stillwater, OK 74078, USA.
| | | |
Collapse
|
25
|
Sato H, Yoneda M, Honda T, Kai C. Recombinant vaccines against the mononegaviruses--what we have learned from animal disease controls. Virus Res 2011; 162:63-71. [PMID: 21982973 PMCID: PMC7114506 DOI: 10.1016/j.virusres.2011.09.038] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Accepted: 09/28/2011] [Indexed: 11/30/2022]
Abstract
The mononegaviruses include a number of highly contagious and severe disease-causing viruses of both animals and humans. For the control of these viral diseases, development of vaccines, either with classical methods or with recombinant DNA virus vectors, has been attempted over the years. Recently reverse genetics of mononegaviruses has been developed and used to generate infectious viruses possessing genomes derived from cloned cDNA in order to study the consequent effects of viral gene manipulations on phenotype. This technology allows us to develop novel candidate vaccines. In particular, a variety of different attenuation strategies to produce a range of attenuated mononegavirus vaccines have been studied. In addition, because of their ideal nature as live vaccines, recombinant mononegaviruses expressing foreign proteins have also been produced with the aim of developing multivalent vaccines against more than one pathogen. These recombinant mononegaviruses are currently under evaluation as new viral vectors for vaccination. Reverse genetics could have great potential for the preparation of vaccines against many mononegaviruses.
Collapse
Affiliation(s)
- Hiroki Sato
- Laboratory Animal Research Center/International Research Center for Infectious Diseases, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan.
| | | | | | | |
Collapse
|
26
|
Early life DNA vaccination with the H gene of Canine distemper virus induces robust protection against distemper. Vaccine 2009; 27:5178-83. [DOI: 10.1016/j.vaccine.2009.06.077] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2009] [Revised: 06/09/2009] [Accepted: 06/22/2009] [Indexed: 11/18/2022]
|
27
|
Nielsen L, Søgaard M, Karlskov-Mortensen P, Jensen TH, Jensen TD, Aasted B, Blixenkrone-Møller M. Humoral and cell-mediated immune responses in DNA immunized mink challenged with wild-type canine distemper virus. Vaccine 2009; 27:4791-7. [DOI: 10.1016/j.vaccine.2009.05.090] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2009] [Revised: 05/29/2009] [Accepted: 05/31/2009] [Indexed: 11/28/2022]
|
28
|
Antigen delivery systems for veterinary vaccine development. Viral-vector based delivery systems. Vaccine 2009; 26:6508-28. [PMID: 18838097 PMCID: PMC7131726 DOI: 10.1016/j.vaccine.2008.09.044] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2008] [Revised: 08/21/2008] [Accepted: 09/16/2008] [Indexed: 11/30/2022]
Abstract
The recent advances in molecular genetics, pathogenesis and immunology have provided an optimal framework for developing novel approaches in the rational design of vaccines effective against viral epizootic diseases. This paper reviews most of the viral-vector based antigen delivery systems (ADSs) recently developed for vaccine testing in veterinary species, including attenuated virus and DNA and RNA viral vectors. Besides their usefulness in vaccinology, these ADSs constitute invaluable tools to researchers for understanding the nature of protective responses in different species, opening the possibility of modulating or potentiating relevant immune mechanisms involved in protection.
Collapse
|
29
|
Hu N, Yu R, Shikuma C, Shiramizu B, Ostrwoski MA, Yu Q. Role of cell signaling in poxvirus-mediated foreign gene expression in mammalian cells. Vaccine 2009; 27:2994-3006. [PMID: 19428911 DOI: 10.1016/j.vaccine.2009.02.103] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2008] [Revised: 02/15/2009] [Accepted: 02/24/2009] [Indexed: 10/21/2022]
Abstract
Poxviruses have been extensively used as a promising vehicle to efficiently deliver a variety of antigens in mammalian hosts to induce immune responses against infectious diseases and cancer. Using recombinant vaccinia virus (VV) and canarypox virus (ALVAC) expressing enhanced green fluorescent protein (EGFP) or multiple HIV-1 gene products, we studied the role of four cellular signaling pathways, the phosphoinositide-3-OH kinase (PI3K), extracellular signal-regulated kinase (ERK), p38 mitogen-activated protein kinase (p38 MAPK), and c-Jun N-terminal kinase (JNK), in poxvirus-mediated foreign gene expression in mammalian cells. In nonpermissive infection (human monocytes), activation of PI3K, ERK, p38 MAPK, and JNK was observed in both VV and ALVAC and blocking PI3K, p38 MAKP, and JNK pathways with their specific inhibitors significantly reduced viral and vaccine antigen gene expression. Whereas, blocking the ERK pathway had no significant effect. Among these cellular signaling pathways studied, PI3K was the most critical pathway involved in gene expression by VV- or ALVAC-infected monocytes. The important role of PI3K in poxvirus-mediated gene expression was further confirmed in mouse epidermal cells stably transfected with dominant-negative PI3K mutant, as poxvirus-mediated targeted gene expression was significantly decreased in these cells when compared with their parental cells. Signaling pathway activation influenced gene expression at the mRNA level rather than virus binding. In permissive mammalian cells, however, VV DNA copies were also significantly decreased in the absence of normal function of the PI3K pathway. Poxvirus-triggered activation of PI3K pathway could be completely abolished by atazanavir, a new generation of antiretroviral protease inhibitors (PIs). As a consequence, ALVAC-mediated EGFP or HIV-1 gag gene expression in infected primary human monocytes was significantly reduced in the presence of atazanavir. These findings implicate that antiretroviral therapy (ART), also known as highly active antiretroviral therapy (HAART), may negatively impact the efficacy of live poxvirus vector-based vaccines and should be carefully considered when administering such live vaccines to individuals on ART.
Collapse
Affiliation(s)
- Ningjie Hu
- Hawaii AIDS Clinical Research Program, University of Hawaii at Manoa, Leahi Hospital, Honolulu, HI 96816, USA
| | | | | | | | | | | |
Collapse
|
30
|
Zehnder AM, Hawkins MG, Koski MA, Luff JA, Benak J, Lowenstine LJ, White SD. An unusual presentation of canine distemper virus infection in a domestic ferret (Mustela putorius furo). Vet Dermatol 2009; 19:232-8. [PMID: 18547381 DOI: 10.1111/j.1365-3164.2008.00677.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A 4.5-year-old, male castrated ferret was examined with a 27-day history of severe pruritus, generalized erythema and scaling. Skin scrapings and a trichogram were negative for mites and dermatophyte organisms. A fungal culture of hair samples was negative. The ferret was treated presumptively for scabies and secondary bacterial and yeast infection with selamectin, enrofloxacin, fluconazole, diphenhydramine and a miconazole-chlorhexidine shampoo. The ferret showed mild improvement in clinical signs over the subsequent 3 weeks, but was inappetent and required supportive feeding and subcutaneous fluids by the owner. The ferret was then examined on an emergency basis at the end of 3 weeks (53 days following initial signs of illness) for severe blood loss from a haematoma over the interscapular region, hypotension and shock. The owners elected euthanasia due to a poor prognosis and deteriorating condition. On post-mortem examination intraepithelial canine distemper viral inclusions were identified systemically, and abundant canine distemper virus antigen was identified with immunohistochemical staining. It is important to note the prolonged course of disease along with the absence of respiratory and neurological signs because this differs from the classic presentation of canine distemper virus infection in ferrets. Canine distemper virus should remain a clinical suspicion for ferrets with skin lesions that do not respond to appropriate therapy, even in animals that were previously vaccinated.
Collapse
Affiliation(s)
- Ashley M Zehnder
- Veterinary Medical Teaching Hospital, School of Veterinary Medicine, University of California, Davis, California 95614, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
Poxviruses including canarypox (ALVAC) and vaccinia viruses have, in recent years, received considerable attention as live vectors for the development of vaccines against infectious diseases such as AIDS, malaria, and tuberculosis. However, the cellular targets for viral infection within the human immune system and the consequences of infection for cells involved in the generation of immune responses have not been clearly delineated. Using recombinant enhanced green fluorescence protein (EGFP)-expressing ALVAC and vaccinia viruses, we have focused here on a side-by-side comparison of ALVAC and vaccinia virus tropism for cells from human peripheral blood and bone marrow. Both ALVAC and vaccinia viruses showed a strong bias toward monocyte infection. ALVAC minimally infected CD19(+) B cells and was unable to infect ex vivo NK cells and T lymphocytes, whereas vaccinia virus could infect B lymphocytes and NK cell populations. Vaccinia virus was also able to infect T lymphocytes at low but detectable levels which could be enhanced upon their activation. Both ALVAC and vaccinia viruses could infect immature monocyte-derived dendritic cells (MDDCs), but only ALVAC infection induced their subsequent maturation. Infection in human bone marrow cells showed that ALVAC infection was restricted to a myelomonocytoid cell-specific CD33(+) cell population, while vaccinia virus showed a strong, but not exclusive, preference for these cells.
Collapse
Affiliation(s)
- Qigui Yu
- University of Hawaii at Manoa, Honolulu, Hawaii, USA.
| | | | | |
Collapse
|
32
|
Griffin DE, Oldstone MBA, von Messling V. Ferrets as a model for morbillivirus pathogenesis, complications, and vaccines. Curr Top Microbiol Immunol 2009; 330:73-87. [PMID: 19203105 PMCID: PMC7121116 DOI: 10.1007/978-3-540-70617-5_4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The ferret is a standard laboratory animal that can be accommodated in most animal facilities. While not susceptible to measles, ferrets are a natural host of canine distemper virus (CDV), the closely related carnivore morbillivirus. CDV infection in ferrets reproduces all clinical signs associated with measles in humans, including the typical rash, fever, general immunosuppression, gastrointestinal and respiratory involvement, and neurological complications. Due to this similarity, experimental CDV infection of ferrets is frequently used to assess the efficacy of novel vaccines, and to characterize pathogenesis mechanisms. In addition, direct intracranial inoculation of measles isolates from subacute sclerosing panencephalitis (SSPE) patients results in an SSPE-like disease in animals that survive the acute phase. Since the advent of reverse genetics systems that allow the targeted manipulation of viral genomes, the model has been used to evaluate the contribution of the accessory proteins C and V, and signalling lymphocyte activation molecule (SLAM)-binding to immunosuppression and overall pathogenesis. Similarly produced green fluorescent protein-expressing derivatives that maintain parental virulence have been instrumental in the direct visualization of systemic dissemination and neuroinvasion. As more immunological tools become available for this model, its contribution to our understanding of morbillivirus-host interactions is expected to increase.
Collapse
Affiliation(s)
- Diane E. Griffin
- grid.21107.350000000121719311Department of Molecular Microbiology, Johns Hopkins University School of Hygiene and Public Health, 615 N. Wolfe Street, Baltimore, MD 21205 USA
| | - Michael B. A. Oldstone
- grid.214007.00000000122199231Department of Immunology and Microbial Science, The Scripps Research Institute, 10550 N. Torrey Pines, La Jolla, CA 92037 USA
| | | |
Collapse
|
33
|
Abstract
This chapter familiarizes veterinarians with basic knowledge of ferret biology, medicine, surgery, and care. Ferrets have a long, slender body with short muscular legs, a long thin tail, small eyes, and short ears. The life span of the ferret is 5 to 8 years. Ferrets may be housed singly or in groups, inside or outside of a house. When kept outdoors, however, they must be protected from extreme weather. Ferrets have difficulty tolerating temperatures above 90°F or below 20°F, and appropriate precautions must be taken to prevent their exposure to these extremes. Ferrets are carnivorous and require a suitable diet. A diet that is high in good-quality animal protein and fat and low in complex carbohydrates and fiber is recommended. Ferrets are routinely immunized against canine distemper virus (CDV) and rabies virus. Ferrets are quite susceptible to CDV, and there is a 100% mortality rate in unvaccinated ferrets infected with CDV. When a new ferret is brought into the household, a quarantine period is recommended before introducing it to other animals, particularly other ferrets. The purpose of the quarantine period is to identify and prevent transmission of infectious disease potentially carried by the new ferret. The duration of this period allows for the development of any clinical signs in a seemingly healthy ferret following entrance into the new household.
Collapse
|
34
|
Perpiñán D, Ramis A, Tomás A, Carpintero E, Bargalló F. Outbreak of canine distemper in domestic ferrets (Mustela putorius furo). Vet Rec 2008; 163:246-50. [PMID: 18723866 DOI: 10.1136/vr.163.8.246] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
In 2006 an outbreak of canine distemper affected 14 young domestic ferrets in Barcelona, Spain. Their clinical signs included a reduced appetite, lethargy, dyspnoea, coughing, sneezing, mucopurulent ocular and nasal discharges, facial and perineal dermatitis, diarrhoea, splenomegaly and fever. Late in the course of the disease, general desquamation and pruritus, and hyperkeratotic/crusting dermatitis of the lips, eyes, nose, footpads, and perineal area were observed. None of the ferrets developed neurological signs. Non-regenerative anaemia and high serum concentrations of alpha- and beta-globulins were the most common laboratory findings. Most of the animals died or were euthanased because of respiratory complications. Postmortem there were no signs of lung collapse. Distemper was diagnosed by direct immunofluorescence of conjunctival swabs or pcr of several organs, and histology revealed the characteristic eosinophilic intracytoplasmic and intranuclear inclusion bodies of canine distemper virus in several organs. The minimum incubation periods calculated for six of the ferrets were 11 to 56 days, and in 13 of the ferrets the signs of disease lasted 14 to 34 days. Inclusion bodies compatible with infection by herpesvirus were found in the lungs of one of the ferrets.
Collapse
Affiliation(s)
- D Perpiñán
- Maragall Exotics Centre Veterinari, Xiprer 53, 08041 Barcelona, Spain
| | | | | | | | | |
Collapse
|
35
|
Beineke A, Puff C, Seehusen F, Baumgärtner W. Pathogenesis and immunopathology of systemic and nervous canine distemper. Vet Immunol Immunopathol 2008; 127:1-18. [PMID: 19019458 DOI: 10.1016/j.vetimm.2008.09.023] [Citation(s) in RCA: 203] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2008] [Revised: 09/17/2008] [Accepted: 09/18/2008] [Indexed: 10/21/2022]
Abstract
Canine distemper is a worldwide occurring infectious disease of dogs, caused by a morbillivirus, closely related to measles and rinderpest virus. The natural host range comprises predominantly carnivores. Canine distemper virus (CDV), an enveloped, negative-sense RNA virus, infects different cell types, including epithelial, mesenchymal, neuroendocrine and hematopoietic cells of various organs and tissues. CDV infection of dogs is characterized by a systemic and/or nervous clinical course and viral persistence in selected organs including the central nervous system (CNS) and lymphoid tissue. Main manifestations include respiratory and gastrointestinal signs, immunosuppression and demyelinating leukoencephalomyelitis (DL). Impaired immune function, associated with depletion of lymphoid organs, consists of a viremia-associated loss of lymphocytes, especially of CD4+ T cells, due to lymphoid cell apoptosis in the early phase. After clearance of the virus from the peripheral blood an assumed diminished antigen presentation and altered lymphocyte maturation cause an ongoing immunosuppression despite repopulation of lymphoid organs. The early phase of DL is a sequel of a direct virus-mediated damage and infiltrating CD8+ cytotoxic T cells associated with an up-regulation of pro-inflammatory cytokines such as interleukin (IL)-6, IL-8, tumor necrosis factor (TNF)-alpha and IL-12 and a lacking response of immunomodulatory cytokines such as IL-10 and transforming growth factor (TGF)-beta. A CD4+-mediated delayed type hypersensitivity and cytotoxic CD8+ T cells contribute to myelin loss in the chronic phase. Additionally, up-regulation of interferon-gamma and IL-1 may occur in advanced lesions. Moreover, an altered balance between matrix metalloproteinases and their inhibitors seems to play a pivotal role for the pathogenesis of DL. Summarized, DL represents a biphasic disease process consisting of an initial direct virus-mediated process and immune-mediated plaque progression. Immunosuppression is due to early virus-mediated lymphocytolysis followed by still poorly understood mechanisms affecting antigen presentation and lymphocyte maturation.
Collapse
Affiliation(s)
- A Beineke
- Department of Pathology, University of Veterinary Medicine, Hannover, Germany
| | | | | | | |
Collapse
|
36
|
Abstract
Poxviruses identified in skin lesions of domestic, pet or wild birds are assigned largely by default to the Avipoxvirus genus within the subfamily Chordopoxvirinae of the family Poxviridae. Avipoxviruses have been identified as the causative agent of disease in at least 232 species in 23 orders of birds. Vaccines based upon attenuated avipoxvirus strains provide good disease control in production poultry, although with the large and intensive production systems there are suggestions and real risks of emergence of strains against which current vaccines might be ineffective. Sequence analysis of the whole genome has revealed overall genome structure and function resemblance to the Chordopoxvirinae; however, avipoxvirus genomes exhibit large-scale genomic rearrangements with more extensive gene families and novel host range gene in comparison with the other Chordopoxvirinae. Phylogenetic analysis places the avipoxviruses externally to the Chorodopoxvirinae to such an extent that in the future it might be appropriate to consider the Avipoxviruses as a separate subfamily within the Poxviridae. A unique relationship exists between Fowlpox virus (FWPV) and reticuloendothelosis viruses. All FWPV strains carry a remnant long terminal repeat, while field strains carry a near full-length provirus integrated at the same location in the FWPV genome. With the development of techniques to construct poxviruses expressing foreign vaccine antigens, the avipoxviruses have gone from neglected obscurity to important vaccine vectors in the past 20 years. The seminal observation of their utility for delivery of vaccine antigens to non-avian species has driven much of the interest in this group of viruses. In the veterinary area, several recombinant avipoxviruses are commercially licensed vaccines. The most successful have been those expressing glycoprotein antigens of enveloped viruses, e.g. avian influenza, Newcastle diseases and West Nile viruses. Several recombinants have undergone extensive human clinical trials as experimental vaccines against HIV/AIDS and malaria or as treatment regimens in cancer patients. The safety profile of avipoxvirus recombinants for use as veterinary and human vaccines or therapeutics is now well established.
Collapse
Affiliation(s)
- Andrew A. Mercer
- Department of Microbiology, University of Otago, 56, 700 Cumberland Street, Dunedin, New Zealand
| | - Axel Schmidt
- Faculty of Medicine, University Witten/Herdecke, Alfred-Herrhausen-Str. 50, 58448 Witten, Germany
| | - Olaf Weber
- BAYER HEALTHCARE AG, Product-related Research, 42096 Wuppertal, Germany
| |
Collapse
|
37
|
Yu Q, Jones B, Hu N, Chang H, Ahmad S, Liu J, Parrington M, Ostrowski M. Comparative analysis of tropism between canarypox (ALVAC) and vaccinia viruses reveals a more restricted and preferential tropism of ALVAC for human cells of the monocytic lineage. Vaccine 2006; 24:6376-91. [PMID: 16859816 DOI: 10.1016/j.vaccine.2006.06.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2006] [Revised: 06/07/2006] [Accepted: 06/11/2006] [Indexed: 10/24/2022]
Abstract
The poxviruses including canarypox (ALVAC) and vaccinia viruses are promising vaccine vectors in humans, but little is known about their biology in human cells. Using recombinant enhanced green fluorescence protein (EGFP)-expressing ALVAC and vaccinia viruses, we have focused here on a side-by-side comparison of ALVAC and vaccinia virus tropism for cells from human peripheral blood and bone marrow. Both ALVAC and vaccinia viruses showed a strong bias towards monocyte infection. ALVAC minimally infected CD19+ B cells and was unable to infect ex vivo NK cells and T lymphocytes, whereas vaccinia virus could infect B lymphocytes and NK cell populations. Vaccinia virus was also able to infect T lymphocytes at low, but detectable levels that could be enhanced upon their activation. The observed preferential infection of ALVAC or vaccinia virus to monocytes was the result of preferential binding to this population, rather than lineage-specific differences in the expression of viral genes. Moreover, the level of CD14 expression on monocytes correlated with their preference to be infected with ALVAC or vaccinia virus. Both ALVAC and vaccinia viruses could infect immature monocyte derived dendritic cells (MDDCs), but only ALVAC infection induced their subsequent maturation. Vaccinia virus, however, showed greater tropism for mature MDDCs compared to ALVAC. Infection in human bone marrow cultures showed that ALVAC infection was restricted to a myelomonocytoid cell-specific CD33(+) cell population, while vaccinia virus showed a strong, but not exclusive, preference for these cells. These findings have implications in terms of choosing optimal pox virus derived vectors as vaccines in terms of reducing clinical reactogenicity and inducing dendritic cell (DC) maturation.
Collapse
Affiliation(s)
- Qigui Yu
- Hawaii AIDS Clinical Research Program, Department of Medicine, University of Hawaii at Manoa, Honolulu, HI, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
Motivation in writing this article stems from many things: a lack of time spent in the veterinary curriculum discussing vaccines, a growing concern(by the general public and the veterinary community) regarding adverse reactions associated with vaccines, and a desire to prevent a recurrence of preventable infectious diseases resulting from a fear-driven cessation of vaccine administration. The objectives of this article are to present a basic review of immunology as related to vaccines, to discuss general guidelines for pediatric vaccines in canine and feline patients,and to offer suggestions as to how we can most positively influence our patients' health from the first visit.
Collapse
Affiliation(s)
- Gina M Davis-Wurzler
- Department of Veterinary Medicine and Epidemiology, School of Veterinary Medicine, University of California at Davis, Davis, CA 95616, USA.
| |
Collapse
|
39
|
Gerdts V, Mutwiri GK, Tikoo SK, Babiuk LA. Mucosal delivery of vaccines in domestic animals. Vet Res 2006; 37:487-510. [PMID: 16611560 DOI: 10.1051/vetres:2006012] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2005] [Accepted: 10/11/2005] [Indexed: 12/29/2022] Open
Abstract
Mucosal vaccination is proving to be one of the greatest challenges in modern vaccine development. Although highly beneficial for achieving protective immunity, the induction of mucosal immunity, especially in the gastro-intestinal tract, still remains a difficult task. As a result, only very few mucosal vaccines are commercially available for domestic animals. Here, we critically review various strategies for mucosal delivery of vaccines in domestic animals. This includes live bacterial and viral vectors, particulate delivery-systems such as polymers, alginate, polyphosphazenes, immune stimulating complex and liposomes, and receptor mediated-targeting strategies to the mucosal tissues. The most commonly used routes of immunization, strategies for delivering the antigen to the mucosal surfaces, and future prospects in the development of mucosal vaccines are discussed.
Collapse
Affiliation(s)
- Volker Gerdts
- Vaccine and Infectious Disease Organization, VIDO, University of Saskatchewan, 120 Veterinary Rd., Saskatoon, S7N 5E3, Canada.
| | | | | | | |
Collapse
|
40
|
Siger L, Bowen RA, Karaca K, Murray MJ, Gordy PW, Loosmore SM, Audonnet JCF, Nordgren RM, Minke JM. Assessment of the efficacy of a single dose of a recombinant vaccine against West Nile virus in response to natural challenge with West Nile virus-infected mosquitoes in horses. Am J Vet Res 2004; 65:1459-62. [PMID: 15566080 DOI: 10.2460/ajvr.2004.65.1459] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To determine the onset of immunity after IM administration of a single dose of a recombinant canarypox virus vaccine against West Nile virus (WNV) in horses in a blind challenge trial. ANIMALS 20 mixed-breed horses. PROCEDURE Horses with no prior exposure to WNV were randomly assigned to 1 of 2 groups (10 horses/group). In 1 group, a recombinant canarypox virus vaccine against WNV was administered to each horse once (day 0). The other 10 control horses were untreated. On day 26, 9 treated and 10 control horses were challenged via the bites of mosquitoes (Aedes albopictus) infected with WNV. Clinical responses and WNV isolation were monitored for 14 days after challenge exposure; antibody responses against WNV after administration of the vaccine and challenge were also assessed in both groups. RESULTS Following challenge via WNV-infected mosquitoes, 1 of 9 treated horses developed viremia. In contrast, 8 of 10 control horses developed viremia after challenge exposure to WNV-infected mosquitoes. All horses seroconverted after WNV challenge; compared with control horses, antibody responses in the horses that received the vaccine were detected earlier. CONCLUSIONS AND CLINICAL RELEVANCE In horses, a single dose of the recombinant canarypox virus-WNV vaccine appears to provide early protection against development of viremia after challenge with WNV-infected mosquitoes, even in the absence of measurable antibody titers in some horses. This vaccine may provide veterinarians with an important tool in controlling WNV infection during a natural outbreak or under conditions in which a rapid onset of protection is required.
Collapse
|
41
|
Dahl L, Jensen TH, Gottschalck E, Karlskov-Mortensen P, Jensen TD, Nielsen L, Andersen MK, Buckland R, Wild TF, Blixenkrone-Møller M. Immunization with plasmid DNA encoding the hemagglutinin and the nucleoprotein confers robust protection against a lethal canine distemper virus challenge. Vaccine 2004; 22:3642-8. [PMID: 15315843 DOI: 10.1016/j.vaccine.2004.03.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2003] [Revised: 03/04/2004] [Accepted: 03/15/2004] [Indexed: 10/26/2022]
Abstract
We have investigated the protective effect of immunization of a highly susceptible natural host of canine distemper virus (CDV) with DNA plasmids encoding the viral nucleoprotein (N) and hemagglutinin (H). The combined intradermal and intramuscular routes of immunization elicited high virus-neutralizing serum antibody titres in mink (Mustela vison). To mimic natural exposure, we also conducted challenge infection by horizontal transmission from infected contact animals. Other groups received a lethal challenge infection by administration to the mucosae of the respiratory tract and into the muscle. One of the mink vaccinated with N plasmid alone developed severe disease after challenge. In contrast, vaccination with the H plasmid together with the N plasmid conferred solid protection against disease and we were unable to detect CDV infection in PBMCs or in different tissues after challenge. Our findings show that DNA immunization by the combined intradermal and intramuscular routes can confer solid protective immunity against naturally transmitted morbillivirus infection and disease.
Collapse
MESH Headings
- Animals
- Antibodies, Viral/analysis
- Antibodies, Viral/biosynthesis
- Antigens, Viral/immunology
- Distemper/immunology
- Distemper/prevention & control
- Distemper/virology
- Distemper Virus, Canine/immunology
- Dogs
- Female
- Genes, Viral/genetics
- Genes, Viral/immunology
- Hemagglutinins/immunology
- Injections, Intradermal
- Injections, Intramuscular
- Mink/immunology
- Neutralization Tests
- Nucleoproteins/immunology
- Reverse Transcriptase Polymerase Chain Reaction
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/immunology
- Viral Vaccines/administration & dosage
- Viral Vaccines/immunology
Collapse
Affiliation(s)
- Lotte Dahl
- Laboratory of Virology and Immunology, Department of Veterinary Microbiology, The Royal Veterinary and Agricultural University, Stigbøjlen 7, DK 1870 Frederiksberg C, Denmark
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
The discovery of Jenner in 1798 founded the science of immunology and eventually led to smallpox eradication from the earth in 1980 after a world-wide vaccination campaign with vaccinia virus (another poxvirus) and paradoxically, despite the eradication of smallpox, there has been an explosion of interest in vaccinia virus in the eighties. This interest has stemmed in part from the application of molecular genetics to clone and express foreign genes from recombinant vaccinia viruses. Vaccinia is also gaining renewed interest due to bioterrorism. These recombinant viruses have multiple applications in research and vaccinology and led to the development of vectored vaccines, such as the recombinant vaccinia rabies vaccine used to eliminate rabies in Western Europe and, more recently, in the United States. Secondly, alternative poxvirus vectors, such as avipox viruses, were proved to be even safer and efficacious non-replicating vectors (suiciole vectors) when used in non-avian species.
Collapse
Affiliation(s)
- P-P Pastoret
- Compton Laboratory, Institute for Animal Health, Compton, Newbury, RG20 7NN, Berks, UK.
| | | |
Collapse
|
43
|
Greenacre CB. Incidence of adverse events in ferrets vaccinated with distemper or rabies vaccine: 143 cases (1995-2001). J Am Vet Med Assoc 2003; 223:663-5. [PMID: 12959385 DOI: 10.2460/javma.2003.223.663] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To determine the incidence of adverse events in ferrets vaccinated with a modified-live avian cell culture canine distemper virus vaccine licensed for use in ferrets, an inactivated rabies vaccine licensed for use in ferrets, or both. DESIGN Retrospective study. ANIMALS 143 ferrets. PROCEDURE Medical records were reviewed to identify ferrets that had an adverse event after vaccination. RESULTS Adverse events developed within 25 minutes after vaccination in 13 ferrets. One ferret developed an adverse event after receiving a distemper and a rabies vaccine simultaneously and developed a second adverse event the following year after receiving the rabies vaccine alone. Therefore, a total of 14 adverse events were identified. All adverse events were an anaphylactic reaction characterized by generalized hyperemia, hypersalivation, and vomiting. Ten of the 14 anaphylactic reactions occurred after ferrets received both vaccines, 3 occurred after ferrets received the distemper vaccine alone, and 1 occurred after a ferret received the rabies vaccine alone. Incidences of adverse events after administration of both vaccines, the distemper vaccine alone, and the rabies vaccine alone were 5.6, 5.9, and 5.6%, respectively. Ferrets that had an anaphylactic reaction were significantly older at the time of vaccination than were ferrets that did not. CONCLUSIONS AND CLINICAL RELEVANCE Results suggest that there may be a high incidence of anaphylactic reactions after vaccination of domestic ferrets. Ferrets should be observed for at least 25 minutes after vaccination, and veterinarians who vaccinate ferrets should be prepared to treat anaphylactic reactions.
Collapse
Affiliation(s)
- Cheryl B Greenacre
- Department of Small Animal Medicine, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
44
|
Aspden K, Passmore JA, Tiedt F, Williamson AL. Evaluation of lumpy skin disease virus, a capripoxvirus, as a replication-deficient vaccine vector. J Gen Virol 2003; 84:1985-1996. [PMID: 12867628 DOI: 10.1099/vir.0.19116-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Lumpy skin disease virus (LSDV), a capripoxvirus with a host range limited to ruminants, was evaluated as a replication-deficient vaccine vector for use in non-ruminant hosts. By using the rabies virus glycoprotein (RG) as a model antigen, it was demonstrated that recombinant LSDV encoding the rabies glycoprotein (rLSDV-RG) was able to express RG in both permissive (ruminant) and non-permissive (non-ruminant) cells. The recombinant LSDV, however, replicated to maturity only in permissive but not in non-permissive cells. Recombinant LSDV-RG was assessed for its ability to generate immunity against RG in non-ruminant hosts (rabbits and mice). Rabbits inoculated with rLSDV-RG produced rabies virus (RV) neutralizing antibodies at levels twofold higher than those reported by the WHO to be protective. BALB/c mice immunized with rLSDV-RG elicited levels of RV-specific cellular immunity (T-cell proliferation) comparable with those of mice immunized with a commercial inactivated rabies vaccine (Verorab; Pasteur Merieux). Most importantly, mice immunized with rLSDV-RG were protected from an aggressive intracranial rabies virus challenge.
Collapse
Affiliation(s)
- Kate Aspden
- Division of Medical Virology, Department of Clinical Laboratory Science & Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory 7925, Cape Town, South Africa
| | - Jo-Ann Passmore
- Division of Medical Virology, Department of Clinical Laboratory Science & Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory 7925, Cape Town, South Africa
| | - Friedrich Tiedt
- Division of Medical Virology, Department of Clinical Laboratory Science & Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory 7925, Cape Town, South Africa
| | - Anna-Lise Williamson
- National Health Laboratory Service, University of Cape Town, Observatory 7925, Cape Town, South Africa
- Division of Medical Virology, Department of Clinical Laboratory Science & Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory 7925, Cape Town, South Africa
| |
Collapse
|
45
|
von Messling V, Zimmer G, Herrler G, Haas L, Cattaneo R. The hemagglutinin of canine distemper virus determines tropism and cytopathogenicity. J Virol 2001; 75:6418-27. [PMID: 11413309 PMCID: PMC114365 DOI: 10.1128/jvi.75.14.6418-6427.2001] [Citation(s) in RCA: 146] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Canine distemper virus (CDV) and measles virus (MV) cause severe illnesses in their respective hosts. The viruses display a characteristic cytopathic effect by forming syncytia in susceptible cells. For CDV, the proficiency of syncytium formation varies among different strains and correlates with the degree of viral attenuation. In this study, we examined the determinants for the differential fusogenicity of the wild-type CDV isolate 5804Han89 (CDV(5804)), the small- and large-plaque-forming variants of the CDV vaccine strain Onderstepoort (CDV(OS) and CDV(OL), respectively), and the MV vaccine strain Edmonston B (MV(Edm)). The cotransfection of different combinations of fusion (F) and hemagglutinin (H) genes in Vero cells indicated that the H protein is the main determinant of fusion efficiency. To verify the significance of this observation in the viral context, a reverse genetic system to generate recombinant CDVs was established. This system is based on a plasmid containing the full-length antigenomic sequence of CDV(OS). The coding regions of the H proteins of all CDV strains and MV(Edm) were introduced into the CDV and MV genetic backgrounds, and recombinant viruses rCDV-H(5804), rCDV-H(OL), rCDV-H(Edm), rMV-H(5804), rMV-H(OL), and rMV-H(OS) were recovered. Thus, the H proteins of the two morbilliviruses are interchangeable and fully functional in a heterologous complex. This is in contrast with the glycoproteins of other members of the family Paramyxoviridae, which do not function efficiently with heterologous partners. The fusogenicity, growth characteristics, and tropism of the recombinant viruses were examined and compared with those of the parental strains. All these characteristics were found to be predominantly mediated by the H protein regardless of the viral backbone used.
Collapse
Affiliation(s)
- V von Messling
- Molecular Medicine Program, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | | | | | | | | |
Collapse
|
46
|
El Kasmi KC, Muller CP. New strategies for closing the gap of measles susceptibility in infants: towards vaccines compatible with current vaccination schedules. Vaccine 2001; 19:2238-44. [PMID: 11257340 DOI: 10.1016/s0264-410x(00)00452-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Peptides representing epitopes of the measles virus glycoproteins have been designed to induce neutralizing and protective antibodies. Those that escape recognition by passively acquired anti-whole virus antibodies could potentially be used as components of a 'pre-vaccine' that could be given during early childhood irrespective of persisting maternal antibodies. Unlike vaccines based on recombinant proteins, epitope-based vaccines can be designed to be compatible with a subsequent boost with the standard life attenuated vaccine. Although synthetic peptides may induce only short-term immunity they have the potential to close in young infants the gap of vulnerability until the standard live attenuated vaccine can be given at 9 or 15 months of age.
Collapse
Affiliation(s)
- K C El Kasmi
- Medizinische Fakultät Universität Tübingen, D-72076, Tübingen, Germany
| | | |
Collapse
|