51
|
NS1 Protein of 2009 Pandemic Influenza A Virus Inhibits Porcine NLRP3 Inflammasome-Mediated Interleukin-1 Beta Production by Suppressing ASC Ubiquitination. J Virol 2018; 92:JVI.00022-18. [PMID: 29386291 DOI: 10.1128/jvi.00022-18] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 01/21/2018] [Indexed: 12/12/2022] Open
Abstract
The inflammasome represents a molecular platform for innate immune regulation and controls proinflammatory cytokine production. The NLRP3 inflammasome is comprised of NLRP3, ASC, and procaspase-1. When the NLRP3 inflammasome is activated, it causes ASC speck formation and caspase-1 activation, resulting in the maturation of interleukin-1β (IL-1β). The NLRP3 inflammasome is regulated at multiple levels, with one level being posttranslational modification. Interestingly, ubiquitination of ASC has been reported to be indispensable for the activation of the NLRP3 inflammasome. Influenza A virus (IAV) infection induces NLRP3 inflammasome-dependent IL-1β secretion, which contributes to the host antiviral defense. However, IAVs have evolved multiple antagonizing mechanisms, one of which is executed by viral NS1 protein to suppress the NLRP3 inflammasome. In this study, we compared IL-1β production in porcine alveolar macrophages in response to IAV infection and found that the 2009 pandemic H1N1 induced less IL-1β than swine influenza viruses (SIVs). Further study revealed that the NS1 C terminus of pandemic H1N1 but not that of SIV was able to significantly inhibit NLRP3 inflammasome-mediated IL-1β production. This inhibitory function was attributed to impaired ASC speck formation and suppression of ASC ubiquitination. Moreover, we identified two target lysine residues, K110 and K140, which are essential for both porcine ASC ubiquitination and NLRP3 inflammasome-mediated IL-1β production. These results revealed a novel mechanism by which the NS1 protein of the 2009 pandemic H1N1 suppresses NLRP3 inflammasome activation.IMPORTANCE Influenza A virus (IAV) infection activates the NLRP3 inflammasome, resulting in the production of IL-1β, which contributes to the host innate immune response. ASC, an adaptor protein of NLRP3, forms specks that are critical for inflammasome activation. Here, we report that the NS1 C terminus of the 2009 pandemic H1N1 has functions to suppress porcine IL-1β production by inhibiting ASC speck formation and ASC ubiquitination. Furthermore, the ubiquitination sites on porcine ASC were identified. The information gained here may contribute to an in-depth understanding of porcine inflammasome activation and regulation in response to different IAVs, helping to further enhance our knowledge of innate immune responses to influenza virus infection in pigs.
Collapse
|
52
|
Kang KI, Linnemann E, Icard AH, Durairaj V, Mundt E, Sellers HS. Chicken astrovirus as an aetiological agent of runting-stunting syndrome in broiler chickens. J Gen Virol 2018; 99:512-524. [PMID: 29458661 DOI: 10.1099/jgv.0.001025] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Despite descriptions of runting-stunting syndrome (RSS) in broiler chickens dating back over 40 years, the aetiology has not yet been described. A novel chicken astrovirus (CkAstV) was isolated in an LMH liver cell line from the intestines of chickens affected with RSS. Clinical RSS is characterized by retarded growth and cystic crypt lesions in the small intestine. In 1-day-old broiler chickens infected with the CkAstV isolate, virus was only detected in the intestinal epithelial cells during the first few days after infection. Notably, the preferred host cells are the crypt epithelial cells following initial replication in the villous epithelial cells, thus implying viral preference for immature intestinal cells. Nevertheless, the CkAstV isolate did not induce remarkable pathological changes, despite the presence of the virus in situ. Serial chicken-to-chicken passages of the virus induced increased virulence, as displayed by decreased weight gain and the presence of cystic lesions in the small intestine reproducing clinical RSS in chickens. The analysis of the full-length genome sequences from the isolated CkAstV and the CkAstV from the bird-to-bird passages showed >99 % similarity. The data obtained in this study suggest that the CkAstV isolate is capable of inducing RSS following serial bird-to-bird passages in broilers and is as an aetiological agent of the disease.
Collapse
Affiliation(s)
- Kyung-Il Kang
- Poultry Diagnostic and Research Center, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA.,Present address: National Poultry Research Center, U.S. Department of Agriculture, Athens, Georgia, USA
| | - Erich Linnemann
- Poultry Diagnostic and Research Center, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Alan H Icard
- Poultry Diagnostic and Research Center, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Vijay Durairaj
- Poultry Diagnostic and Research Center, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA.,Present address: Boehringer Ingelheim Vetmedica, Inc., Ames, Iowa, USA
| | - Egbert Mundt
- Poultry Diagnostic and Research Center, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA.,Present address: Boehringer Ingelheim, Veterinary Research Center, GmbH Co. KG, Bemeroder Straße 31, 30559 Hannover, Germany
| | - Holly S Sellers
- Poultry Diagnostic and Research Center, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
53
|
Rajão DS, Pérez DR. Universal Vaccines and Vaccine Platforms to Protect against Influenza Viruses in Humans and Agriculture. Front Microbiol 2018; 9:123. [PMID: 29467737 PMCID: PMC5808216 DOI: 10.3389/fmicb.2018.00123] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 01/18/2018] [Indexed: 01/22/2023] Open
Abstract
Influenza virus infections pose a significant threat to public health due to annual seasonal epidemics and occasional pandemics. Influenza is also associated with significant economic losses in animal production. The most effective way to prevent influenza infections is through vaccination. Current vaccine programs rely heavily on the vaccine's ability to stimulate neutralizing antibody responses to the hemagglutinin (HA) protein. One of the biggest challenges to an effective vaccination program lies on the fact that influenza viruses are ever-changing, leading to antigenic drift that results in escape from earlier immune responses. Efforts toward overcoming these challenges aim at improving the strength and/or breadth of the immune response. Novel vaccine technologies, the so-called universal vaccines, focus on stimulating better cross-protection against many or all influenza strains. However, vaccine platforms or manufacturing technologies being tested to improve vaccine efficacy are heterogeneous between different species and/or either tailored for epidemic or pandemic influenza. Here, we discuss current vaccines to protect humans and animals against influenza, highlighting challenges faced to effective and uniform novel vaccination strategies and approaches.
Collapse
Affiliation(s)
- Daniela S. Rajão
- Department of Population Health, University of Georgia, Athens, GA, United States
| | | |
Collapse
|
54
|
Khatri M, Richardson LA, Meulia T. Mesenchymal stem cell-derived extracellular vesicles attenuate influenza virus-induced acute lung injury in a pig model. Stem Cell Res Ther 2018; 9:17. [PMID: 29378639 PMCID: PMC5789598 DOI: 10.1186/s13287-018-0774-8] [Citation(s) in RCA: 255] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 12/19/2017] [Accepted: 01/15/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Mesenchymal stem (stromal) cells (MSCs) mediate their immunoregulatory and tissue repair functions by secreting paracrine factors, including extracellular vesicles (EVs). In several animal models of human diseases, MSC-EVs mimic the beneficial effects of MSCs. Influenza viruses cause annual outbreaks of acute respiratory illness resulting in significant mortality and morbidity. Influenza viruses constantly evolve, thus generating drug-resistant strains and rendering current vaccines less effective against the newly generated strains. Therefore, new therapies that can control virus replication and the inflammatory response of the host are needed. The objective of this study was to examine if MSC-EV treatment can attenuate influenza virus-induced acute lung injury in a preclinical model. METHODS We isolated EVs from swine bone marrow-derived MSCs. Morphology of MSC-EVs was determined by electron microscopy and expression of mesenchymal markers was examined by flow cytometry. Next, we examined the anti-influenza activity of MSC-EVs in vitro in lung epithelial cells and anti-viral and immunomodulatory properties in vivo in a pig model of influenza virus. RESULTS MSC-EVs were isolated from MSC-conditioned medium by ultracentrifugation. MSC-EVs were round-shaped and, similarly to MSCs, expressed mesenchymal markers and lacked the expression of swine leukocyte antigens I and II. Incubation of PKH-26-labeled EVs with lung epithelial cells revealed that MSC-EVs incorporated into the epithelial cells. Next, we examined the anti-influenza and anti-inflammatory properties of MSC-EVs. MSC-EVs inhibited the hemagglutination activity of avian, swine, and human influenza viruses at concentrations of 1.25-5 μg/ml. MSC-EVs inhibited influenza virus replication and virus-induced apoptosis in lung epithelial cells. The anti-influenza activity of MSC-EVs was due to transfer of RNAs from EVs to epithelial cells since pre-incubation of MSC-EVs with RNase enzyme abrogated the anti-influenza activity of MSC-EVs. In a pig model of influenza virus, intratracheal administration of MSC-EVs 12 h after influenza virus infection significantly reduced virus shedding in the nasal swabs, influenza virus replication in the lungs, and virus-induced production of proinflammatory cytokines in the lungs of influenza-infected pigs. The histopathological findings revealed that MSC-EVs alleviated influenza virus-induced lung lesions in pigs. CONCLUSIONS Our data demonstrated in a relevant preclinical large animal model of influenza virus that MSC-EVs possessed anti-influenza and anti-inflammatory properties and that EVs may be used as cell-free therapy for influenza in humans.
Collapse
Affiliation(s)
- Mahesh Khatri
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| | - Levi Arthur Richardson
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| | - Tea Meulia
- Molecular and Cellular Imaging Center, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH USA
| |
Collapse
|
55
|
Khatri M, Richardson LA. Therapeutic potential of porcine bronchoalveolar fluid-derived mesenchymal stromal cells in a pig model of LPS-induced ALI. J Cell Physiol 2018; 233:5447-5457. [PMID: 29231967 DOI: 10.1002/jcp.26397] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 12/04/2017] [Indexed: 12/20/2022]
Abstract
In this study, we isolated mesenchymal stromal (stem) cells (MSCs) from broncho-alveolar lavage fluid (BAL) of 2-6-week-old commercial pigs. BAL-MSCs displayed fibroblastic morphology and possessed self-renewal properties. Similar to bone-marrow MSCs, BAL-MSCs expressed mesenchymal markers and both cell types lacked the expression of hematopoetic markers. BAL-MSCs, when cultured in differentiation induction media, differentiated into adipocytes, osteocytes, and chondrocytes. Next, we examined if BAL-MSCs have the ability to treat lipopolysaccharide (LPS)-induced acute lung injury (ALI) in a pig model. Five-week-old commercial pigs were inoculated intra-tracheally with E. coli LPS (1 mg/kg body weight [b.wt.]). Twelve hours after the LPS inoculation, groups of pigs were inoculated intra-tracheally with BM-MSCs or BAL-MSCs (2 × 106 cells/kg b.wt.). Forty eight hours after the cells administration pigs were euthanized and neutrophils in BAL, lung lesions, and cytokines in lung lysates, and engraftment of MSCs in lungs were examined. Engraftment of BAL-MSCs in injured lungs was significantly higher than the BM-MSCs, however, both cell types were equally effective in attenuating LPS-induced ALI as evidenced by decreased inflammation, lung lesions, and proinflammatory cytokines in the lungs of pigs treated with BAL- or BM-MSCs. These data in a preclinical large animal model suggest that BAL-MSCs may be used in clinical settings to treat ALI in humans.
Collapse
Affiliation(s)
- Mahesh Khatri
- Department of Food Animal Health Research Program, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, Ohio
| | - Levi A Richardson
- Department of Food Animal Health Research Program, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, Ohio
| |
Collapse
|
56
|
Lamont EA, Poulin E, Sreevatsan S, Cheeran MCJ. Major histocompatibility complex I of swine respiratory cells presents conserved regions of influenza proteins. J Gen Virol 2018; 99:303-308. [PMID: 29458525 DOI: 10.1099/jgv.0.001008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Influenza A virus in swine (IAV-S) is a prevalent respiratory pathogen in pigs that has deleterious consequences to animal and human health. Pigs represent an important reservoir for influenza and potential mixing vessel for novel gene reassortments. Despite the central role of pigs in recent influenza outbreaks, much remains unknown about the impact of swine immunity on IAV-S transmission, pathogenesis, and evolution. An incomplete understanding of interactions between the porcine immune system and IAV-S has hindered development of new diagnostic tools and vaccines. In order to address this gap in knowledge, we identified swine leukocyte antigen (SLA) restricted IAV-S peptides presented by porcine airway epithelial cells using an immunoproteomics approach. The majority of MHC-associated peptides belonged to matrix 1, nucleoprotein and nonstructural 1 proteins. Future investigation of the potential cross-reactive nature of these peptides is needed to confirm antigen recognition by cytotoxic T lymphocytes and their utility as vaccine candidates.
Collapse
Affiliation(s)
- Elise A Lamont
- Department of Microbiology and Immunology, Medical School, University of Minnesota, Minneapolis, MN 55455, USA
| | - Erin Poulin
- Department of Veterinary Population Medicine, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN 55108, USA
| | - Srinand Sreevatsan
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI 48824, USA
| | - Maxim C-J Cheeran
- Department of Veterinary Population Medicine, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN 55108, USA
| |
Collapse
|
57
|
Siemens N, Oehmcke-Hecht S, Mettenleiter TC, Kreikemeyer B, Valentin-Weigand P, Hammerschmidt S. Port d'Entrée for Respiratory Infections - Does the Influenza A Virus Pave the Way for Bacteria? Front Microbiol 2017; 8:2602. [PMID: 29312268 PMCID: PMC5742597 DOI: 10.3389/fmicb.2017.02602] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 12/13/2017] [Indexed: 12/12/2022] Open
Abstract
Bacterial and viral co-infections of the respiratory tract are life-threatening and present a global burden to the global community. Staphylococcus aureus, Streptococcus pneumoniae, and Streptococcus pyogenes are frequent colonizers of the upper respiratory tract. Imbalances through acquisition of seasonal viruses, e.g., Influenza A virus, can lead to bacterial dissemination to the lower respiratory tract, which in turn can result in severe pneumonia. In this review, we summarize the current knowledge about bacterial and viral co-infections of the respiratory tract and focus on potential experimental models suitable for mimicking this disease. Transmission of IAV and pneumonia is mainly modeled by mouse infection. Few studies utilizing ferrets, rats, guinea pigs, rabbits, and non-human primates are also available. The knowledge gained from these studies led to important discoveries and advances in understanding these infectious diseases. Nevertheless, mouse and other infection models have limitations, especially in translation of the discoveries to humans. Here, we suggest the use of human engineered lung tissue, human ex vivo lung tissue, and porcine models to study respiratory co-infections, which might contribute to a greater translation of the results to humans and improve both, animal and human health.
Collapse
Affiliation(s)
- Nikolai Siemens
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
- Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Sonja Oehmcke-Hecht
- Institute of Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany
| | - Thomas C. Mettenleiter
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institute, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Bernd Kreikemeyer
- Institute of Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany
| | - Peter Valentin-Weigand
- Center for Infection Medicine, Institute for Microbiology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Sven Hammerschmidt
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| |
Collapse
|
58
|
Montoya M, Foni E, Solórzano A, Razzuoli E, Baratelli M, Bilato D, Córdoba L, Del Burgo MAM, Martinez J, Martinez-Orellana P, Chiapponi C, Perlin DS, Del Real G, Amadori M. Expression Dynamics of Innate Immunity in Influenza Virus-Infected Swine. Front Vet Sci 2017; 4:48. [PMID: 28484702 PMCID: PMC5399951 DOI: 10.3389/fvets.2017.00048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 03/22/2017] [Indexed: 12/31/2022] Open
Abstract
The current circulating swine influenza virus (IV) subtypes in Europe (H1N1, H1N2, and H3N2) are associated with clinical outbreaks of disease. However, we showed that pigs could be susceptible to other IV strains that are able to cross the species barrier. In this work, we extended our investigations into whether different IV strains able to cross the species barrier might give rise to different innate immune responses that could be associated with pathological lesions. For this purpose, we used the same samples collected in a previous study of ours, in which healthy pigs had been infected with a H3N2 Swine IV and four different H3N8 IV strains circulating in different animal species. Pigs had been clinically inspected and four subjects/group were sacrificed at 3, 6, and 21 days post infection. In the present study, all groups but mock exhibited antibody responses to IV nucleoprotein protein. Pulmonary lesions and high-titered viral replication were observed in pigs infected with the swine-adapted virus. Interestingly, pigs infected with avian and seal H3N8 strains also showed moderate lesions and viral replication, whereas equine and canine IVs did not cause overt pathological signs, and replication was barely detectable. Swine IV infection induced interferon (IFN)-alpha and interleukin-6 responses in bronchoalveolar fluids (BALF) at day 3 post infection, as opposed to the other non-swine-adapted virus strains. However, IFN-alpha responses to the swine-adapted virus were not associated with an increase of the local, constitutive expression of IFN-alpha genes. Remarkably, the Equine strain gave rise to a Serum Amyloid A response in BALF despite little if any replication. Each virus strain could be associated with expression of cytokine genes and/or proteins after infection. These responses were observed well beyond the period of virus replication, suggesting a prolonged homeostatic imbalance of the innate immune system.
Collapse
Affiliation(s)
- María Montoya
- Centre de Recerca en Sanitat Animal (CReSA), UAB-IRTA, Universitat Autònoma de Barcelona, Barcelona, Spain.,The Pirbright Institute, Woking, UK
| | - Emanuela Foni
- OIE Reference Laboratory for Swine Influenza, Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia-Romagna, Parma, Italy
| | - Alicia Solórzano
- Public Health Research Institute and Regional Biocontainment Laboratory, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Elisabetta Razzuoli
- S.S. Sezione Genova, Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d'Aosta, Genova, Italy
| | - Massimiliano Baratelli
- Centre de Recerca en Sanitat Animal (CReSA), UAB-IRTA, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Dania Bilato
- Laboratory of Cellular Immunology, Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia-Romagna, Brescia, Italy
| | - Lorena Córdoba
- Centre de Recerca en Sanitat Animal (CReSA), UAB-IRTA, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Maria Angeles Martín Del Burgo
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Madrid, Spain
| | - Jorge Martinez
- Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Pamela Martinez-Orellana
- Centre de Recerca en Sanitat Animal (CReSA), UAB-IRTA, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Chiara Chiapponi
- OIE Reference Laboratory for Swine Influenza, Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia-Romagna, Parma, Italy
| | - David S Perlin
- Public Health Research Institute and Regional Biocontainment Laboratory, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Gustavo Del Real
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Madrid, Spain
| | - Massimo Amadori
- Laboratory of Cellular Immunology, Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia-Romagna, Brescia, Italy
| |
Collapse
|
59
|
Dhakal S, Goodman J, Bondra K, Lakshmanappa YS, Hiremath J, Shyu DL, Ouyang K, Kang KI, Krakowka S, Wannemuehler MJ, Won Lee C, Narasimhan B, Renukaradhya GJ. Polyanhydride nanovaccine against swine influenza virus in pigs. Vaccine 2017; 35:1124-1131. [PMID: 28117173 DOI: 10.1016/j.vaccine.2017.01.019] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 01/09/2017] [Accepted: 01/12/2017] [Indexed: 11/25/2022]
|
60
|
Pomorska-Mól M, Dors A, Kwit K, Kowalczyk A, Stasiak E, Pejsak Z. Kinetics of single and dual infection of pigs with swine influenza virus and Actinobacillus pleuropneumoniae. Vet Microbiol 2017; 201:113-120. [PMID: 28284596 DOI: 10.1016/j.vetmic.2017.01.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 01/11/2017] [Accepted: 01/12/2017] [Indexed: 10/20/2022]
Abstract
Porcine respiratory disease complex (PRDC) is a common problem in modern pork production worldwide. Pathogens that are amongst other pathogens frequently involved in PRDC etiology are swine influenza virus (SIV) and A. pleuropneumoniae. The effect of dual infection with mentioned pathogens has not been investigated to date. The aim of the present study was to evaluate the kinetics of single and dual infection of pigs with SIV and A. pleuropneumoniae with regard to clinical course, pathogens shedding, lung lesions and early immune response. The most severe symptoms were observed in co-inoculated piglets. The AUC value for SIV shedding was lower in pigs single inoculated with SIV as compared to co-inoculated animals. In contrast, no significant differences were found between A. pleuropneumoniae shedding in single or dual inoculated pigs. Three out of 5 co-inoculated piglets euthanized at 10 dpi were positive against serotype 2 A. pleuropneumonie. All piglets inoculated with SIV developed specific HI antibodies at 10 dpi. In pigs dual inoculated the specific humoral response against SIV was observed earlier, at 7 dpi. The SIV-like lung lesions were more severe in co-inoculated pigs. In the groups inoculated with A. pleuropneumoniae (single or dual) the acute phase protein response was generally stronger than in SIV-single infected group. Co-infection with SIV and A. pleuropneumoniae potentiated the severity of lung lesions caused by SIV and enhanced virus replication in the lung and nasal SIV shedding. Enhanced SIV replication contributed to a more severe clinical course of the disease as well as earlier and higher magnitude immune response (acute phase proteins, HI antibodies) compared to single inoculated pigs.
Collapse
Affiliation(s)
| | - Arkadiusz Dors
- National Veterinary Research Institute, Department of Swine Diseases, Puławy, Poland
| | - Krzysztof Kwit
- National Veterinary Research Institute, Department of Swine Diseases, Puławy, Poland
| | - Andrzej Kowalczyk
- National Veterinary Research Institute, Department of Swine Diseases, Puławy, Poland
| | - Ewelina Stasiak
- National Veterinary Research Institute, Department of Swine Diseases, Puławy, Poland
| | - Zygmunt Pejsak
- National Veterinary Research Institute, Department of Swine Diseases, Puławy, Poland
| |
Collapse
|
61
|
Dhakal S, Hiremath J, Bondra K, Lakshmanappa YS, Shyu DL, Ouyang K, Kang KI, Binjawadagi B, Goodman J, Tabynov K, Krakowka S, Narasimhan B, Lee CW, Renukaradhya GJ. Biodegradable nanoparticle delivery of inactivated swine influenza virus vaccine provides heterologous cell-mediated immune response in pigs. J Control Release 2017; 247:194-205. [PMID: 28057521 DOI: 10.1016/j.jconrel.2016.12.039] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 12/12/2016] [Accepted: 12/29/2016] [Indexed: 10/20/2022]
Abstract
Swine influenza virus (SwIV) is one of the important zoonotic pathogens. Current flu vaccines have failed to provide cross-protection against evolving viruses in the field. Poly(lactic-co-glycolic acid) (PLGA) is a biodegradable FDA approved polymer and widely used in drug and vaccine delivery. In this study, inactivated SwIV H1N2 antigens (KAg) encapsulated in PLGA nanoparticles (PLGA-KAg) were prepared, which were spherical in shape with 200 to 300nm diameter, and induced maturation of antigen presenting cells in vitro. Pigs vaccinated twice with PLGA-KAg via intranasal route showed increased antigen specific lymphocyte proliferation and enhanced the frequency of T-helper/memory and cytotoxic T cells (CTLs) in peripheral blood mononuclear cells (PBMCs). In PLGA-KAg vaccinated and heterologous SwIV H1N1 challenged pigs, clinical flu symptoms were absent, while the control pigs had fever for four days. Grossly and microscopically, reduced lung pathology and viral antigenic mass in the lung sections with clearance of infectious challenge virus in most of the PLGA-KAg vaccinated pig lung airways were observed. Immunologically, PLGA-KAg vaccine irrespective of not significantly boosting the mucosal antibody response, it augmented the frequency of IFN-γ secreting total T cells, T-helper and CTLs against both H1N2 and H1N1 SwIV. In summary, inactivated influenza virus delivered through PLGA-NPs reduced the clinical disease and induced cross-protective cell-mediated immune response in a pig model. Our data confirmed the utility of a pig model for intranasal particulate flu vaccine delivery platform to control flu in humans.
Collapse
Affiliation(s)
- Santosh Dhakal
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, OH 44691, USA; Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Jagadish Hiremath
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, OH 44691, USA; Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Kathryn Bondra
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, OH 44691, USA; Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Yashavanth S Lakshmanappa
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, OH 44691, USA; Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Duan-Liang Shyu
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, OH 44691, USA; Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Kang Ouyang
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, OH 44691, USA; Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Kyung-Il Kang
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, OH 44691, USA; Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Basavaraj Binjawadagi
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, OH 44691, USA; Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Jonathan Goodman
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011, USA
| | - Kairat Tabynov
- The Research Institute for Biological Safety Problems (RIBSP), Zhambylskaya Oblast, Gvardeiskiy 080409, Kazakhstan
| | - Steven Krakowka
- The Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, 1925 Coffey Road, Columbus, OH, USA
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011, USA
| | - Chang Won Lee
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, OH 44691, USA; Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Gourapura J Renukaradhya
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, OH 44691, USA; Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
62
|
Joseph U, Su YCF, Vijaykrishna D, Smith GJD. The ecology and adaptive evolution of influenza A interspecies transmission. Influenza Other Respir Viruses 2017; 11:74-84. [PMID: 27426214 PMCID: PMC5155642 DOI: 10.1111/irv.12412] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/13/2016] [Indexed: 12/16/2022] Open
Abstract
Since 2013, there have been several alarming influenza-related events; the spread of highly pathogenic avian influenza H5 viruses into North America, the detection of H10N8 and H5N6 zoonotic infections, the ongoing H7N9 infections in China and the continued zoonosis of H5N1 viruses in parts of Asia and the Middle East. The risk of a new influenza pandemic increases with the repeated interspecies transmission events that facilitate reassortment between animal influenza strains; thus, it is of utmost importance to understand the factors involved that promote or become a barrier to cross-species transmission of Influenza A viruses (IAVs). Here, we provide an overview of the ecology and evolutionary adaptations of IAVs, with a focus on a review of the molecular factors that enable interspecies transmission of the various virus gene segments.
Collapse
MESH Headings
- Animals
- Animals, Wild
- Asia/epidemiology
- China/epidemiology
- Disease Reservoirs/virology
- Ducks/virology
- Evolution, Molecular
- Geese/virology
- Humans
- Influenza A Virus, H5N1 Subtype/genetics
- Influenza A Virus, H5N1 Subtype/pathogenicity
- Influenza A Virus, H5N1 Subtype/physiology
- Influenza A Virus, H7N9 Subtype/genetics
- Influenza A Virus, H7N9 Subtype/pathogenicity
- Influenza A Virus, H7N9 Subtype/physiology
- Influenza A virus/genetics
- Influenza A virus/pathogenicity
- Influenza A virus/physiology
- Influenza in Birds/virology
- Influenza, Human/transmission
- Influenza, Human/virology
- Orthomyxoviridae Infections/transmission
- Orthomyxoviridae Infections/virology
- Phylogeny
- Reassortant Viruses/genetics
- Reassortant Viruses/pathogenicity
- Reassortant Viruses/physiology
- Zoonoses
Collapse
Affiliation(s)
| | | | | | - Gavin J. D. Smith
- Duke‐NUS Medical SchoolSingapore
- Duke Global Health InstituteDuke UniversityDurhamNCUSA
| |
Collapse
|
63
|
Abstract
Immunity to targeted infectious diseases may be conferred or enhanced by vaccines, which are manufactured from recombinant forms as well as inactivated or attenuated organisms. These vaccines have to meet requirements for safety, quality, and efficacy. In addition to antigenic components, various adjuvants may be included in vaccines to evoke an effective immune response. To ensure the safety of new vaccines, preclinical toxicology studies are conducted prior to the initiation of, and concurrently with, clinical studies. There are five different types of preclinical toxicology study in the evaluation of vaccine safety: single and/or repeat dose, reproductive and developmental, mutagenicity, carcinogenicity, and safety pharmacology. If any adverse effects are observed in the course of these studies, they should be fully evaluated and a final safety decision made accordingly. Successful preclinical toxicology studies depend on multiple factors including using the appropriate study designs, using the right animal model, and evoking an effective immune response. Additional in vivo and in vitro assays that establish the identity, purity, safety, and potency of the vaccine play a significant role in assessing critical characteristics of vaccine safety.
Collapse
|
64
|
Influenza A Virus Infection in Pigs Attracts Multifunctional and Cross-Reactive T Cells to the Lung. J Virol 2016; 90:9364-82. [PMID: 27512056 PMCID: PMC5044846 DOI: 10.1128/jvi.01211-16] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 08/01/2016] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Pigs are natural hosts for influenza A viruses and play a critical role in influenza epidemiology. However, little is known about their influenza-evoked T-cell response. We performed a thorough analysis of both the local and systemic T-cell response in influenza virus-infected pigs, addressing kinetics and phenotype as well as multifunctionality (gamma interferon [IFN-γ], tumor necrosis factor alpha [TNF-α], and interleukin-2 [IL-2]) and cross-reactivity. A total of 31 pigs were intratracheally infected with an H1N2 swine influenza A virus (FLUAVsw) and consecutively euthanized. Lungs, tracheobronchial lymph nodes, and blood were sampled during the first 15 days postinfection (p.i.) and at 6 weeks p.i. Ex vivo flow cytometry of lung lymphocytes revealed an increase in proliferating (Ki-67(+)) CD8(+) T cells with an early effector phenotype (perforin(+) CD27(+)) at day 6 p.i. Low frequencies of influenza virus-specific IFN-γ-producing CD4(+) and CD8(+) T cells could be detected in the lung as early as 4 days p.i. On consecutive days, influenza virus-specific CD4(+) and CD8(+) T cells produced mainly IFN-γ and/or TNF-α, reaching peak frequencies around day 9 p.i., which were up to 30-fold higher in the lung than in tracheobronchial lymph nodes or blood. At 6 weeks p.i., CD4(+) and CD8(+) memory T cells had accumulated in lung tissue. These cells showed diverse cytokine profiles and in vitro reactivity against heterologous influenza virus strains, all of which supports their potential to combat heterologous influenza virus infections in pigs. IMPORTANCE Pigs not only are a suitable large-animal model for human influenza virus infection and vaccine development but also play a central role in the emergence of new pandemic strains. Although promising candidate universal vaccines are tested in pigs and local T cells are the major correlate of heterologous control, detailed and targeted analyses of T-cell responses at the site of infection are scarce. With the present study, we provide the first detailed characterization of magnitude, kinetics, and phenotype of specific T cells recruited to the lungs of influenza virus-infected pigs, and we could demonstrate multifunctionality, cross-reactivity, and memory formation of these cells. This, and ensuing work in the pig, will strengthen the position of this species as a large-animal model for human influenza virus infection and will immediately benefit vaccine development for improved control of influenza virus infections in pigs.
Collapse
|
65
|
Deblanc C, Delgado-Ortega M, Gorin S, Berri M, Paboeuf F, Berthon P, Herrler G, Meurens F, Simon G. Mycoplasma hyopneumoniae does not affect the interferon-related anti-viral response but predisposes the pig to a higher level of inflammation following swine influenza virus infection. J Gen Virol 2016; 97:2501-2515. [PMID: 27498789 DOI: 10.1099/jgv.0.000573] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
In pigs, influenza A viruses and Mycoplasma hyopneumoniae (Mhp) are major contributors to the porcine respiratory disease complex. Pre-infection with Mhp was previously shown experimentally to exacerbate the clinical outcomes of H1N1 infection during the first week after virus inoculation. In order to better understand the interactions between these pathogens, we aimed to assess very early responses (at 5, 24 and 48 h) after H1N1 infection in pigs pre-infected or not with Mhp. Clinical signs and macroscopic lung lesions were similar in both infected groups at early times post-H1N1 infection; and Mhp pre-infection affected neither the influenza virus replication nor the IFN-induced antiviral responses in the lung. However, it predisposed the animals to a higher inflammatory response to H1N1 infection, as revealed by the massive infiltration of neutrophils and macrophages into the lungs and the increased production of pro-inflammatory cytokines (IL-6, IL-1β and TNF-α). Thus, it seems it is this marked inflammatory state that would play a role in exacerbating the clinical signs subsequent to H1N1 infection.
Collapse
Affiliation(s)
- Céline Deblanc
- Université Bretagne Loire, France.,ANSES, Laboratoire de Ploufragan-Plouzané, Unité Virologie Immunologie Porcines, Ploufragan, France
| | | | - Stéphane Gorin
- ANSES, Laboratoire de Ploufragan-Plouzané, Unité Virologie Immunologie Porcines, Ploufragan, France.,Université Bretagne Loire, France
| | | | - Frédéric Paboeuf
- Université Bretagne Loire, France.,ANSES, Service de Production de Porcs Assainis et d'Expérimentation, Ploufragan, France
| | | | - Georg Herrler
- Institut für Virologie, Tierärztliche Hochschule Hannover, Hannover, Germany
| | | | - Gaëlle Simon
- ANSES, Laboratoire de Ploufragan-Plouzané, Unité Virologie Immunologie Porcines, Ploufragan, France.,Université Bretagne Loire, France
| |
Collapse
|
66
|
Maisonnasse P, Bouguyon E, Piton G, Ezquerra A, Urien C, Deloizy C, Bourge M, Leplat JJ, Simon G, Chevalier C, Vincent-Naulleau S, Crisci E, Montoya M, Schwartz-Cornil I, Bertho N. The respiratory DC/macrophage network at steady-state and upon influenza infection in the swine biomedical model. Mucosal Immunol 2016; 9:835-49. [PMID: 26530136 DOI: 10.1038/mi.2015.105] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 08/12/2015] [Indexed: 02/04/2023]
Abstract
Human and mouse respiratory tracts show anatomical and physiological differences, which will benefit from alternative experimental models for studying many respiratory diseases. Pig has been recognized as a valuable biomedical model, in particular for lung transplantation or pathologies such as cystic fibrosis and influenza infection. However, there is a lack of knowledge about the porcine respiratory immune system. Here we segregated and studied six populations of pig lung dendritic cells (DCs)/macrophages (Mθs) as follows: conventional DCs (cDC) 1 and cDC2, inflammatory monocyte-derived DCs (moDCs), monocyte-derived Mθs, and interstitial and alveolar Mθs. The three DC subsets present migratory and naive T-cell stimulation capacities. As observed in human and mice, porcine cDC1 and cDC2 were able to induce T-helper (Th)1 and Th2 responses, respectively. Interestingly, porcine moDCs increased in the lung upon influenza infection, as observed in the mouse model. Pig cDC2 shared some characteristics observed in human but not in mice, such as the expression of FCɛRIα and Langerin, and an intra-epithelial localization. This work, by unraveling the extended similarities of the porcine and human lung DC/Mθ networks, highlights the relevance of pig, both as an exploratory model of DC/Mθ functions and as a model for human inflammatory lung pathologies.
Collapse
Affiliation(s)
- P Maisonnasse
- Virologie et Immunologie Moléculaires UR892, Institut National de la Recherche Agronomique, Jouy-en-Josas, France
| | - E Bouguyon
- Virologie et Immunologie Moléculaires UR892, Institut National de la Recherche Agronomique, Jouy-en-Josas, France
| | - G Piton
- INRA, UMR Génétique Animale et Biologie Intégrative (GABI), Equipe Génétique Immunité Santé, Jouy-en-Josas, France.,Laboratoire de Radiobiologie et Etude du genome, CEA, Direction des Sciences du Vivant, Institut de Radiobiologie Cellulaire et Moléculaire, Jouy-en-Josas, France
| | - A Ezquerra
- Dpto. de Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Madrid, Spain
| | - C Urien
- Virologie et Immunologie Moléculaires UR892, Institut National de la Recherche Agronomique, Jouy-en-Josas, France
| | - C Deloizy
- Virologie et Immunologie Moléculaires UR892, Institut National de la Recherche Agronomique, Jouy-en-Josas, France
| | - M Bourge
- I2BC, Centre National de la Recherche Scientifique, Gif-sur-Yvette, France
| | - J-J Leplat
- INRA, UMR Génétique Animale et Biologie Intégrative (GABI), Equipe Génétique Immunité Santé, Jouy-en-Josas, France.,Laboratoire de Radiobiologie et Etude du genome, CEA, Direction des Sciences du Vivant, Institut de Radiobiologie Cellulaire et Moléculaire, Jouy-en-Josas, France
| | - G Simon
- Anses, Laboratoire de Ploufragan/Plouzané, Unité Virologie Immunologie Porcines, BP53, Ploufragan, France.,Université Européenne de Bretagne, Rennes, France
| | - C Chevalier
- Virologie et Immunologie Moléculaires UR892, Institut National de la Recherche Agronomique, Jouy-en-Josas, France
| | - S Vincent-Naulleau
- INRA, UMR Génétique Animale et Biologie Intégrative (GABI), Equipe Génétique Immunité Santé, Jouy-en-Josas, France.,Laboratoire de Radiobiologie et Etude du genome, CEA, Direction des Sciences du Vivant, Institut de Radiobiologie Cellulaire et Moléculaire, Jouy-en-Josas, France
| | - E Crisci
- Centre de Recerca en Sanitat Animal (CReSA), UAB-IRTA, Campus de la Universitat Autònoma de Barcelona, Bellaterra (Cerdanyola del Vallès), Spain
| | - M Montoya
- Centre de Recerca en Sanitat Animal (CReSA), UAB-IRTA, Campus de la Universitat Autònoma de Barcelona, Bellaterra (Cerdanyola del Vallès), Spain.,The Pirbright Institute, Surrey, UK
| | - I Schwartz-Cornil
- Virologie et Immunologie Moléculaires UR892, Institut National de la Recherche Agronomique, Jouy-en-Josas, France
| | - N Bertho
- Virologie et Immunologie Moléculaires UR892, Institut National de la Recherche Agronomique, Jouy-en-Josas, France
| |
Collapse
|
67
|
Hiremath J, Kang KI, Xia M, Elaish M, Binjawadagi B, Ouyang K, Dhakal S, Arcos J, Torrelles JB, Jiang X, Lee CW, Renukaradhya GJ. Entrapment of H1N1 Influenza Virus Derived Conserved Peptides in PLGA Nanoparticles Enhances T Cell Response and Vaccine Efficacy in Pigs. PLoS One 2016; 11:e0151922. [PMID: 27093541 PMCID: PMC4836704 DOI: 10.1371/journal.pone.0151922] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 03/07/2016] [Indexed: 11/18/2022] Open
Abstract
Pigs are believed to be one of the important sources of emerging human and swine influenza viruses (SwIV). Influenza virus conserved peptides have the potential to elicit cross-protective immune response, but without the help of potent adjuvant and delivery system they are poorly immunogenic. Biodegradable polylactic-co-glycolic acid (PLGA) nanoparticle (PLGA-NP) based vaccine delivery system enhances cross-presentation of antigens by the professional antigen presenting cells. In this study, Norovirus P particle containing SwIV M2e (extracellular domain of the matrix protein 2) chimera and highly conserved two each of H1N1 peptides of pandemic 2009 and classical human influenza viruses were entrapped in PLGA-NPs. Influenza antibody-free pigs were vaccinated with PLGA-NPs peptides cocktail vaccine twice with or without an adjuvant, Mycobacterium vaccae whole cell lysate, intranasally as mist. Vaccinated pigs were challenged with a virulent heterologous zoonotic SwIV H1N1, and one week later euthanized and the lung samples were analyzed for the specific immune response and viral load. Clinically, pigs vaccinated with PLGA-NP peptides vaccine had no fever and flu symptoms, and the replicating challenged SwIV was undetectable in the bronchoalveolar lavage fluid. Immunologically, PLGA-NP peptides vaccination (without adjuvant) significantly increased the frequency of antigen-specific IFNγ secreting CD4 and CD8 T cells response in the lung lymphocytes, despite not boosting the antibody response both at pre- and post-challenge. In summary, our data indicated that nanoparticle-mediated delivery of conserved H1N1 influenza peptides induced the virus specific T cell response in the lungs and reduced the challenged heterologous virus load in the airways of pigs.
Collapse
Affiliation(s)
- Jagadish Hiremath
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, Ohio, 44691, United States of America, and Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, 43210, United States of America
| | - Kyung-il Kang
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, Ohio, 44691, United States of America, and Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, 43210, United States of America
| | - Ming Xia
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Mohamed Elaish
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, Ohio, 44691, United States of America, and Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, 43210, United States of America
| | - Basavaraj Binjawadagi
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, Ohio, 44691, United States of America, and Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, 43210, United States of America
| | - Kang Ouyang
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, Ohio, 44691, United States of America, and Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, 43210, United States of America
| | - Santosh Dhakal
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, Ohio, 44691, United States of America, and Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, 43210, United States of America
| | - Jesus Arcos
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, United States of America
| | - Jordi B. Torrelles
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, United States of America
| | - X. Jiang
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Chang Won Lee
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, Ohio, 44691, United States of America, and Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, 43210, United States of America
| | - Gourapura J. Renukaradhya
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, Ohio, 44691, United States of America, and Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, 43210, United States of America
| |
Collapse
|
68
|
Schomberg DT, Tellez A, Meudt JJ, Brady DA, Dillon KN, Arowolo FK, Wicks J, Rousselle SD, Shanmuganayagam D. Miniature Swine for Preclinical Modeling of Complexities of Human Disease for Translational Scientific Discovery and Accelerated Development of Therapies and Medical Devices. Toxicol Pathol 2016; 44:299-314. [PMID: 26839324 DOI: 10.1177/0192623315618292] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Noncommunicable diseases, including cardiovascular disease, diabetes, chronic respiratory disease, and cancer, are the leading cause of death in the world. The cost, both monetary and time, of developing therapies to prevent, treat, or manage these diseases has become unsustainable. A contributing factor is inefficient and ineffective preclinical research, in which the animal models utilized do not replicate the complex physiology that influences disease. An ideal preclinical animal model is one that responds similarly to intrinsic and extrinsic influences, providing high translatability and concordance of preclinical findings to humans. The overwhelming genetic, anatomical, physiological, and pathophysiological similarities to humans make miniature swine an ideal model for preclinical studies of human disease. Additionally, recent development of precision gene-editing tools for creation of novel genetic swine models allows the modeling of highly complex pathophysiology and comorbidities. As such, the utilization of swine models in early research allows for the evaluation of novel drug and technology efficacy while encouraging redesign and refinement before committing to clinical testing. This review highlights the appropriateness of the miniature swine for modeling complex physiologic systems, presenting it as a highly translational preclinical platform to validate efficacy and safety of therapies and devices.
Collapse
Affiliation(s)
- Dominic T Schomberg
- Biomedical & Genomic Research Group, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | | - Jennifer J Meudt
- Biomedical & Genomic Research Group, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | | | | - Folagbayi K Arowolo
- Biomedical & Genomic Research Group, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Joan Wicks
- Alizée Pathology, LLC, Thurmont, Maryland, USA
| | | | | |
Collapse
|
69
|
Abstract
Gnotobiotic (GN) rodent models have provided insight into the contributions of the gut microbiota to host health and preventing disease. However, rodent models are limited by several important physiological and metabolic differences from humans, and many rodent models do not dependably replicate the clinical manifestations of human diseases. Due to the high degree of similarity in anatomy, physiology, immunology and brain growth, the domestic pig (Sus scrofa) is considered a clinically relevant model to study factors influencing human gastrointestinal, immune, and brain development. Gnotobiotic piglet models have been developed and shown to recapitulate key aspects of GN rodent models. Human microbiota-associated (HMA) piglets have been established using inocula from infants, children, and adults. The gut microbiota of recipient HMA piglets was more similar to that of the human donor than that of conventionally reared piglets harboring a pig microbiota. Moreover, Bifidobacterium and Bacteroides, two predominant bacterial groups of infant gut, were successfully established in the HMA piglets. Thus, the HMA pig model has the potential to be a valuable model for investigating how the gut microbiota composition changes in response to environmental factors, such as age, diet, vaccination, antibiotic use and infection. The HMA also represents a robust model for screening the efficacy of pre- and probiotic interventions. Lastly, HMA piglets can be an ideal model with which to elucidate microbe-host interactions in human health and disease due to the similarities to humans in anatomy, physiology, developmental maturity at birth, and the pathophysiology of many human diseases.
Collapse
Affiliation(s)
- Mei Wang
- Mei Wang, PhD, is a research specialist and Sharon M. Donovan, PhD, RD, is a professor in the Department of Food Science & Human Nutrition, University of Illinois, Urbana, Illinois
| | - Sharon M Donovan
- Mei Wang, PhD, is a research specialist and Sharon M. Donovan, PhD, RD, is a professor in the Department of Food Science & Human Nutrition, University of Illinois, Urbana, Illinois
| |
Collapse
|
70
|
Dwivedi V, Manickam C, Dhakal S, Binjawadagi B, Ouyang K, Hiremath J, Khatri M, Hague JG, Lee CW, Renukaradhya GJ. Adjuvant effects of invariant NKT cell ligand potentiates the innate and adaptive immunity to an inactivated H1N1 swine influenza virus vaccine in pigs. Vet Microbiol 2016; 186:157-63. [PMID: 27016770 DOI: 10.1016/j.vetmic.2016.02.028] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 02/27/2016] [Accepted: 02/29/2016] [Indexed: 02/07/2023]
Abstract
Pigs are considered as the source of some of the emerging human flu viruses. Inactivated swine influenza virus (SwIV) vaccine has been in use in the US swine herds, but it failed to control the flu outbreaks. The main reason has been attributed to lack of induction of strong local mucosal immunity in the respiratory tract. Invariant natural killer T (iNKT) cell is a unique T cell subset, and activation of iNKT cell using its ligand α-Galactosylceramide (α-GalCer) has been shown to potentiate the cross-protective immunity to inactivated influenza virus vaccine candidates in mice. Recently, we discovered iNKT cell in pig and demonstrated its activation using α-GalCer. In this study, we evaluated the efficacy of an inactivated H1N1 SwIV coadministered with α-GalCer intranasally against a homologous viral challenge. Our results demonstrated the potent adjuvant effects of α-GalCer in potentiating both innate and adaptive immune responses to SwIV Ags in the lungs of pigs, which resulted in reduction in the lung viral load by 3 logs compared to without adjuvant. Immunologically, in the lungs of pigs vaccinated with α-GalCer an increased virus specific IgA response, IFN-α secretion and NK cell-cytotoxicity was observed. In addition, iNKT cell-stimulation enhanced the secretion of Th1 cytokines (IFN-γ and IL-12) and reduced the production of immunosuppressive cytokines (IL-10 and TGF-β) in the lungs of pigs⋅ In conclusion, we demonstrated for the first time iNKT cell adjuvant effects in pigs to SwIV Ags through augmenting the innate and adaptive immune responses in the respiratory tract.
Collapse
Affiliation(s)
- Varun Dwivedi
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Department of Veterinary Preventive Medicine, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691, United States
| | - Cordelia Manickam
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Department of Veterinary Preventive Medicine, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691, United States
| | - Santosh Dhakal
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Department of Veterinary Preventive Medicine, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691, United States
| | - Basavaraj Binjawadagi
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Department of Veterinary Preventive Medicine, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691, United States
| | - Kang Ouyang
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Department of Veterinary Preventive Medicine, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691, United States; College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Jagadish Hiremath
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Department of Veterinary Preventive Medicine, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691, United States; Indian Council of Agricultural Research, National Institute of Veterinary Epidemiology and Disease Informatics, Bangalore, India
| | - Mahesh Khatri
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Department of Veterinary Preventive Medicine, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691, United States
| | - Jacquelyn Gervay Hague
- Department of Chemistry, University of California Davis, One Shields Avenue, Davis, CA 95616, United States
| | - Chang Won Lee
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Department of Veterinary Preventive Medicine, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691, United States
| | - Gourapura J Renukaradhya
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Department of Veterinary Preventive Medicine, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691, United States.
| |
Collapse
|
71
|
Brogaard L, Heegaard PMH, Larsen LE, Mortensen S, Schlegel M, Dürrwald R, Skovgaard K. Late regulation of immune genes and microRNAs in circulating leukocytes in a pig model of influenza A (H1N2) infection. Sci Rep 2016; 6:21812. [PMID: 26893019 PMCID: PMC4759598 DOI: 10.1038/srep21812] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 02/01/2016] [Indexed: 01/18/2023] Open
Abstract
MicroRNAs (miRNAs) are a class of short regulatory RNA molecules which are implicated in modulating gene expression. Levels of circulating, cell-associated miRNAs in response to influenza A virus (IAV) infection has received limited attention so far. To further understand the temporal dynamics and biological implications of miRNA regulation in circulating leukocytes, we collected blood samples before and after (1, 3, and 14 days) IAV challenge of pigs. Differential expression of miRNAs and innate immune factor mRNA transcripts was analysed using RT-qPCR. A total of 20 miRNAs were regulated after IAV challenge, with the highest number of regulated miRNAs seen on day 14 after infection at which time the infection was cleared. Targets of the regulated miRNAs included genes involved in apoptosis and cell cycle regulation. Significant regulation of both miRNAs and mRNA transcripts at 14 days after challenge points to a protracted effect of IAV infection, potentially affecting the host’s ability to respond to secondary infections. In conclusion, experimental IAV infection of pigs demonstrated the dynamic nature of miRNA and mRNA regulation in circulating leukocytes during and after infection, and revealed the need for further investigation of the potential immunosuppressing effect of miRNA and innate immune signaling after IAV infection.
Collapse
Affiliation(s)
- Louise Brogaard
- Section for Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, 1870 Frederiksberg C, Denmark
| | - Peter M H Heegaard
- Section for Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, 1870 Frederiksberg C, Denmark
| | - Lars E Larsen
- Section for Virology, National Veterinary Institute, Technical University of Denmark, 1870 Frederiksberg C, Denmark
| | - Shila Mortensen
- Section for Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, 1870 Frederiksberg C, Denmark
| | | | | | - Kerstin Skovgaard
- Section for Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, 1870 Frederiksberg C, Denmark
| |
Collapse
|
72
|
Khatri M, Chattha KS. Replication of influenza A virus in swine umbilical cord epithelial stem-like cells. Virulence 2016; 6:40-9. [PMID: 25517546 DOI: 10.4161/21505594.2014.983020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
In this study, we describe the isolation and characterization of epithelial stem-like cells from the swine umbilical cord and their susceptibility to influenza virus infection. Swine umbilical cord epithelial stem cells (SUCECs) expressed stem cell and pluripotency associated markers such as SSEA-1, SSEA-4, TRA 1-60 and TRA 1-81 and Oct4. Morphologically, cells displayed polygonal morphology and were found to express epithelial markers; pancytokeratin, cytokeratin-18 and occludin; mesenchymal cell markers CD44, CD90 and haematopoietic cell marker CD45 were not detected on these cells. The cells had extensive proliferation and self- renewal properties. The cells also possessed immunomodulatory activity and inhibited the proliferation of T cells. Also, higher levels of anti-inflammatory cytokine IL-10 were detected in SUCEC-T cell co-cultures. The cells were multipotent and differentiated into lung epithelial cells when cultured in epithelial differentiation media. We also examined if SUCECs are susceptible to infection with influenza virus. SUCECs expressed sialic acid receptors, used by influenza virus for binding to cells. The 2009 pandemic influenza virus and swine influenza virus replicated in these cells. SUCECs due to their differentiation and immunoregulatory properties will be useful as cellular therapy in a pig model for human diseases. Additionally, our data indicate that influenza virus can infect SUCECs and may transmit influenza virus from mother to fetus through umbilical cord and transplantation of influenza virus-infected stem cells may transmit infection to recipients. Therefore, we propose that umbilical cord cells, in addition to other agents, should also be tested for influenza virus before cryopreservation for future use as a cell therapy for disease conditions.
Collapse
Affiliation(s)
- Mahesh Khatri
- a Food Animal Health Research Program; Ohio Agricultural Research and Development Center; The Ohio State University ; Wooster , OH United States
| | | |
Collapse
|
73
|
Characterization of the Localized Immune Response in the Respiratory Tract of Ferrets following Infection with Influenza A and B Viruses. J Virol 2015; 90:2838-48. [PMID: 26719259 DOI: 10.1128/jvi.02797-15] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 12/20/2015] [Indexed: 01/09/2023] Open
Abstract
UNLABELLED The burden of infection with seasonal influenza viruses is significant. Each year is typically characterized by the dominance of one (sub)type or lineage of influenza A or B virus, respectively. The incidence of disease varies annually, and while this may be attributed to a particular virus strain or subtype, the impacts of prior immunity, population differences, and variations in clinical assessment are also important. To improve our understanding of the impacts of seasonal influenza viruses, we directly compared clinical symptoms, virus shedding, and expression of cytokines, chemokines, and immune mediators in the upper respiratory tract (URT) of ferrets infected with contemporary A(H1N1)pdm09, A(H3N2), or influenza B virus. Gene expression in the lower respiratory tract (LRT) was also assessed. Clinical symptoms were minimal. Overall cytokine/chemokine profiles in the URT were consistent in pattern and magnitude between animals infected with influenza A and B viruses, and peak expression levels of interleukin-1α (IL-1α), IL-1β, IL-6, IL-12p40, alpha interferon (IFN-α), IFN-β, and tumor necrosis factor alpha (TNF-α) mRNAs correlated with peak levels of viral shedding. MCP1 and IFN-γ were expressed after the virus peak. Granzymes A and B and IL-10 reached peak expression as the virus was cleared and seroconversion was detected. Cytokine/chemokine gene expression in the LRT following A(H1N1)pdm09 virus infection reflected the observations seen for the URT but was delayed 2 or 3 days, as was virus replication. These data indicate that disease severities and localized immune responses following infection with seasonal influenza A and B viruses are similar, suggesting that other factors are likely to modulate the incidence and impact of seasonal influenza. IMPORTANCE Both influenza A and B viruses cocirculate in the human population, and annual influenza seasons are typically dominated by an influenza A virus subtype or an influenza B virus lineage. Surveillance data indicate that the burden of disease is higher in some seasons, yet it is unclear whether this is due to specific virus strains or to other factors, such as cross-reactive immunity or clinical definitions of influenza. We directly compared disease severities and localized inflammatory responses to different seasonal influenza virus strains, including the 2009 pandemic strain, in healthy naive ferrets. We found that the disease severities and the cytokine and chemokine responses were similar irrespective of the seasonal strain or the location of the infection in the respiratory tract. This suggests that factors other than the immune response to a particular virus (sub)type contribute to the variable impact of influenza virus infection in a population.
Collapse
|
74
|
Arriaga-Pizano L, Ferat-Osorio E, Rodríguez-Abrego G, Mancilla-Herrera I, Domínguez-Cerezo E, Valero-Pacheco N, Pérez-Toledo M, Lozano-Patiño F, Laredo-Sánchez F, Malagón-Rangel J, Nellen-Hummel H, González-Bonilla C, Arteaga-Troncoso G, Cérbulo-Vázquez A, Pastelin-Palacios R, Klenerman P, Isibasi A, López-Macías C. Differential Immune Profiles in Two Pandemic Influenza A(H1N1)pdm09 Virus Waves at Pandemic Epicenter. Arch Med Res 2015; 46:651-8. [PMID: 26696552 PMCID: PMC4914610 DOI: 10.1016/j.arcmed.2015.12.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 12/01/2015] [Indexed: 11/26/2022]
Abstract
Background and Aims Severe influenza A(H1N1)pdm2009 virus infection cases are characterized by sustained immune activation during influenza pandemics. Seasonal flu data suggest that immune mediators could be modified by wave-related changes. Our aim was to determine the behavior of soluble and cell-related mediators in two waves at the epicenter of the 2009 influenza pandemic. Methods Leukocyte surface activation markers were studied in serum from peripheral blood samples, collected from the 1st (April–May, 2009) and 2nd (October 2009–February 2010) pandemic waves. Patients with confirmed influenza A(H1N1)pdm2009 virus infection (H1N1), influenza-like illness (ILI) or healthy donors (H) were analyzed. Results Serum IL-6, IL-4 and IL-10 levels were elevated in H1N1 patients from the 2nd pandemic wave. Additionally, the frequency of helper and cytotoxic T cells was reduced during the 1st wave, whereas CD69 expression in helper T cells was increased in the 2nd wave for both H1N1 and ILI patients. In contrast, CD62L expression in granulocytes from the ILI group was increased in both waves but in monocytes only in the 2nd wave. Triggering Receptor Expressed on Myeloid cells (TREM)-1 expression was elevated only in H1N1 patients at the 1st wave. Conclusions Our results show that during the 2009 influenza pandemic a T cell activation phenotype is observed in a wave-dependent fashion, with an expanded activation in the 2nd wave, compared to the 1st wave. Conversely, granulocyte and monocyte activation is infection-dependent. This evidence collected at the pandemic epicenter in 2009 could help us understand the differences in the underlying cellular mechanisms that drive the wave-related immune profile behaviors that occur against influenza viruses during pandemics.
Collapse
Affiliation(s)
- Lourdes Arriaga-Pizano
- Medical Research Unit in Immunochemistry, Specialties Hospital, National Medical Center Siglo XXI, IMSS, Mexico City, Mexico
| | - Eduardo Ferat-Osorio
- Medical Research Unit in Immunochemistry, Specialties Hospital, National Medical Center Siglo XXI, IMSS, Mexico City, Mexico; Gastrointestinal Surgery Service, Specialties Hospital, National Medical Center Siglo XXI, IMSS, Mexico City, Mexico
| | | | - Ismael Mancilla-Herrera
- Infectology and Immunology department, National Institute of Perinatology, SSA, Mexico City, Mexico
| | - Esteban Domínguez-Cerezo
- Medical Research Unit in Immunochemistry, Specialties Hospital, National Medical Center Siglo XXI, IMSS, Mexico City, Mexico; Graduate Program on Immunology, ENCB-IPN, Mexico City, Mexico
| | - Nuriban Valero-Pacheco
- Medical Research Unit in Immunochemistry, Specialties Hospital, National Medical Center Siglo XXI, IMSS, Mexico City, Mexico; Graduate Program on Immunology, ENCB-IPN, Mexico City, Mexico
| | - Marisol Pérez-Toledo
- Medical Research Unit in Immunochemistry, Specialties Hospital, National Medical Center Siglo XXI, IMSS, Mexico City, Mexico; Graduate Program on Immunology, ENCB-IPN, Mexico City, Mexico
| | - Fernando Lozano-Patiño
- Internal Medicine Service, Specialties Hospital of the National Medical Center Siglo XXI, IMSS, Mexico City, Mexico
| | - Fernando Laredo-Sánchez
- Internal Medicine Service, Specialties Hospital of the National Medical Center Siglo XXI, IMSS, Mexico City, Mexico
| | - José Malagón-Rangel
- Internal Medicine Service, Specialties Hospital of the National Medical Center Siglo XXI, IMSS, Mexico City, Mexico
| | - Haiko Nellen-Hummel
- Internal Medicine Service, Specialties Hospital of the National Medical Center Siglo XXI, IMSS, Mexico City, Mexico
| | - César González-Bonilla
- Unit for Epidemiological Surveillance, National Medical Center La Raza, IMSS, Mexico City, Mexico
| | | | | | | | - Paul Klenerman
- Oxford Biomedical Research Centre and Oxford Martin School, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Armando Isibasi
- Medical Research Unit in Immunochemistry, Specialties Hospital, National Medical Center Siglo XXI, IMSS, Mexico City, Mexico
| | - Constantino López-Macías
- Medical Research Unit in Immunochemistry, Specialties Hospital, National Medical Center Siglo XXI, IMSS, Mexico City, Mexico; Visiting Professor of Immunology, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
75
|
Renukaradhya GJ, Narasimhan B, Mallapragada SK. Respiratory nanoparticle-based vaccines and challenges associated with animal models and translation. J Control Release 2015; 219:622-631. [PMID: 26410807 PMCID: PMC4760633 DOI: 10.1016/j.jconrel.2015.09.047] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 09/21/2015] [Accepted: 09/23/2015] [Indexed: 12/14/2022]
Abstract
Vaccine development has had a huge impact on human health. However, there is a significant need to develop efficacious vaccines for several existing as well as emerging respiratory infectious diseases. Several challenges need to be overcome to develop efficacious vaccines with translational potential. This review focuses on two aspects to overcome some barriers - 1) the development of nanoparticle-based vaccines, and 2) the choice of suitable animal models for respiratory infectious diseases that will allow for translation. Nanoparticle-based vaccines, including subunit vaccines involving synthetic and/or natural polymeric adjuvants and carriers, as well as those based on virus-like particles offer several key advantages to help overcome the barriers to effective vaccine development. These include the ability to deliver combinations of antigens, target the vaccine formulation to specific immune cells, enable cross-protection against divergent strains, act as adjuvants or immunomodulators, allow for sustained release of antigen, enable single dose delivery, and potentially obviate the cold chain. While mouse models have provided several important insights into the mechanisms of infectious diseases, they are often a limiting step in translation of new vaccines to the clinic. An overview of different animal models involved in vaccine research for respiratory infections, with advantages and disadvantages of each model, is discussed. Taken together, advances in nanotechnology, combined with the right animal models for evaluating vaccine efficacy, has the potential to revolutionize vaccine development for respiratory infections.
Collapse
Affiliation(s)
- Gourapura J Renukaradhya
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, United States
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011, United States
| | - Surya K Mallapragada
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011, United States.
| |
Collapse
|
76
|
Khatri M, O'Brien TD, Chattha KS, Saif LJ. Porcine lung mesenchymal stromal cells possess differentiation and immunoregulatory properties. Stem Cell Res Ther 2015; 6:222. [PMID: 26560714 PMCID: PMC4642738 DOI: 10.1186/s13287-015-0220-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 09/20/2014] [Accepted: 10/29/2015] [Indexed: 01/14/2023] Open
Abstract
Introduction Mesenchymal stem (stromal) cells (MSCs) possess self-renewal, differentiation and immunoregulatory properties, and therefore are being evaluated as cellular therapy for inflammatory and autoimmune diseases, and for tissue repair. MSCs isolated from bone marrow are extensively studied. Besides bone marrow, MSCs have been identified in almost all organs of the body including the lungs. Lung-derived MSCs may be more effective as therapy for lung diseases as compared to bone marrow-derived MSCs. Pigs are similar to humans in anatomy, physiology and immunological responses, and thus may serve as a useful large animal preclinical model to study potential cellular therapy for human diseases. Methods We isolated MSCs from the lungs (L-MSCs) of 4–6-week-old germ-free pigs. We determined the self-renewal, proliferation and differentiation potential of L-MSCs. We also examined the mechanisms of immunoregulation by porcine L-MSCs. Results MSCs isolated from porcine lungs showed spindle-shaped morphology and proliferated actively in culture. Porcine L-MSCs expressed mesenchymal markers CD29, CD44, CD90 and CD105 and lacked the expression of hematopoietic markers CD34 and CD45. These cells were multipotent and differentiated into adipocytes, osteocytes and epithelial cells. Like human MSCs, L-MSCs possessed immunoregulatory properties and inhibited proliferation of T cells and interferon-γ and tumor necrosis factor-α production by T cells and dendritic cells, respectively, and increased the production of T-helper 2 cytokines interleukin (IL)-4 and IL-13 by T cells. L-MSCs induced the production of prostaglandin E2 (PGE2) in MSC–T cell co-cultures and inhibition of PGE2 significantly restored (not completely) the immune modulatory effects of L-MSCs. Conclusions Here, we demonstrate that MSCs can be isolated from porcine lung and that these cells, similar to human lung MSCs, possess in vitro proliferation, differentiation and immunomodulatory functions. Thus, these cells may serve as a model system to evaluate the contribution of lung MSCs in modulating the immune response, interactions with resident epithelial cells and tissue repair in a pig model of human lung diseases.
Collapse
Affiliation(s)
- Mahesh Khatri
- Department of Food Animal Health Research Program, Ohio Agricultural Research and Development Center, The Ohio State University, 1680 Madison Avenue, Wooster, OH, 44691, USA.
| | - Timothy D O'Brien
- Department of Veterinary Population Medicine, College of Veterinary Medicine, University of Minnesota, St. Paul, MN, USA.
| | - Kuldeep S Chattha
- Department of Food Animal Health Research Program, Ohio Agricultural Research and Development Center, The Ohio State University, 1680 Madison Avenue, Wooster, OH, 44691, USA.
| | - Linda J Saif
- Department of Food Animal Health Research Program, Ohio Agricultural Research and Development Center, The Ohio State University, 1680 Madison Avenue, Wooster, OH, 44691, USA.
| |
Collapse
|
77
|
Qiu Y, De Hert K, Van Reeth K. Cross-protection against European swine influenza viruses in the context of infection immunity against the 2009 pandemic H1N1 virus: studies in the pig model of influenza. Vet Res 2015; 46:105. [PMID: 26404790 PMCID: PMC4581489 DOI: 10.1186/s13567-015-0236-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 08/06/2015] [Indexed: 12/31/2022] Open
Abstract
Pigs are natural hosts for the same influenza virus subtypes as humans and are a valuable model for cross-protection studies with influenza. In this study, we have used the pig model to examine the extent of virological protection between a) the 2009 pandemic H1N1 (pH1N1) virus and three different European H1 swine influenza virus (SIV) lineages, and b) these H1 viruses and a European H3N2 SIV. Pigs were inoculated intranasally with representative strains of each virus lineage with 6- and 17-week intervals between H1 inoculations and between H1 and H3 inoculations, respectively. Virus titers in nasal swabs and/or tissues of the respiratory tract were determined after each inoculation. There was substantial though differing cross-protection between pH1N1 and other H1 viruses, which was directly correlated with the relatedness in the viral hemagglutinin (HA) and neuraminidase (NA) proteins. Cross-protection against H3N2 was almost complete in pigs with immunity against H1N2, but was weak in H1N1/pH1N1-immune pigs. In conclusion, infection with a live, wild type influenza virus may offer substantial cross-lineage protection against viruses of the same HA and/or NA subtype. True heterosubtypic protection, in contrast, appears to be minimal in natural influenza virus hosts. We discuss our findings in the light of the zoonotic and pandemic risks of SIVs.
Collapse
Affiliation(s)
- Yu Qiu
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820, Merelbeke, Belgium.
| | - Karl De Hert
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820, Merelbeke, Belgium.
| | - Kristien Van Reeth
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820, Merelbeke, Belgium.
| |
Collapse
|
78
|
Shanmugasundaram R, Kogut MH, Arsenault RJ, Swaggerty CL, Cole K, Reddish JM, Selvaraj RK. Effect of Salmonella infection on cecal tonsil regulatory T cell properties in chickens. Poult Sci 2015; 94:1828-35. [PMID: 26049799 DOI: 10.3382/ps/pev161] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/19/2015] [Indexed: 01/02/2023] Open
Abstract
Two studies were conducted to study regulatory T cell [Treg (CD4⁺CD25⁺)] properties during the establishment of a persistent intestinal infection in broiler chickens. Four-day-old broiler chicks were orally gavaged with 5 × 10⁶ CFU/mL Salmonella enteritidis (S. enteritidis) or sterile PBS (control). Samples were collected at 4, 7, 10, and 14 d postinfection. There was a significant (P < 0.05) increase in the number of CD4⁺CD25⁺ cells by d 4 postinfection that increased steadily throughout the course of the 14-d infection, whereas the number of CD4⁺CD25⁺ cells in the noninfected controls remained steady throughout the study. CD4⁺CD25⁺ cells from cecal tonsils of S. enteritidis-infected birds had a higher (P < 0.05) IL-10 mRNA content than CD4⁺CD25⁺ cells from the noninfected controls at all time-points studied. The amount of IL-2 mRNA content in the cecal tonsil CD4⁺CD25⁻ cells from the infected birds did not differ (P > 0.05) when compared to that of noninfected control birds. At a lower effector/responder cell ratio of 0.25:1, CD4⁺CD25⁺ cells from cecal tonsils of Salmonella-infected birds suppressed T cell proliferation at d 7 and 14 post-S. enteritidis infection, while CD4⁺CD25⁺ cells from noninfected control groups did not suppress T cell proliferation. In the second studu, 1-day-old chickens were orally gavaged with PBS (control) or 1.25 × 10⁸ CFU/bird S. enteritidis. At 7 and 21 d post-Salmonella infection, CD25⁺ cells collected from cecal tonsils of S. enteritidis-infected birds and restimulated in vitro with Salmonella antigen had higher (P < 0.05) IL-10 mRNA content compared to those in the control group. Spleen CD4⁺CD25⁺, CD4⁺, and CD8⁺ cell percentage did not differ (P > 0.05) between the Salmonella-infected and control birds. In conclusion, a persistent intestinal S. enteritidis infection increased the Treg percentage, suppressive properties, and IL-10 mRNA amounts in the cecal tonsils of broiler birds.
Collapse
Affiliation(s)
- Revathi Shanmugasundaram
- Department of Animal Sciences, Ohio Agricultural Research and Development Center, Wooster, OH 44691
| | - Michael H Kogut
- USDA-Agricultural Research Service (ARS), Plains Area, College Station, TX 77845
| | - Ryan J Arsenault
- USDA-Agricultural Research Service (ARS), Plains Area, College Station, TX 77845
| | | | - Kimberly Cole
- Department of Animal Sciences, Ohio Agricultural Research and Development Center, Wooster, OH 44691
| | - John M Reddish
- Department of Animal Sciences, Ohio Agricultural Research and Development Center, Wooster, OH 44691
| | - Ramesh K Selvaraj
- Department of Animal Sciences, Ohio Agricultural Research and Development Center, Wooster, OH 44691
| |
Collapse
|
79
|
Talker SC, Koinig HC, Stadler M, Graage R, Klingler E, Ladinig A, Mair KH, Hammer SE, Weissenböck H, Dürrwald R, Ritzmann M, Saalmüller A, Gerner W. Magnitude and kinetics of multifunctional CD4+ and CD8β+ T cells in pigs infected with swine influenza A virus. Vet Res 2015; 46:52. [PMID: 25971313 PMCID: PMC4429459 DOI: 10.1186/s13567-015-0182-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 04/14/2015] [Indexed: 11/12/2022] Open
Abstract
Although swine are natural hosts for influenza A viruses, the porcine T-cell response to swine influenza A virus (FLUAVsw) infection has been poorly characterized so far. We have studied Ki-67 expression and FLUAVsw-specific production of IFN-γ, TNF-α and IL-2 in CD4+ and CD8β+ T cells isolated from piglets that had been intratracheally infected with a H1N2 FLUAVsw isolate. IFN-γ+TNF-α+IL-2+ multifunctional CD4+ T cells were present in the blood of all infected animals at one or two weeks after primary infection and their frequency increased in four out of six animals after homologous secondary infection. These cells produced higher amounts of IFN-γ, TNF-α and IL-2 than did CD4+ T cells that only produced a single cytokine. The vast majority of cytokine-producing CD4+ T cells expressed CD8α, a marker associated with activation and memory formation in porcine CD4+ T cells. Analysis of CD27 expression suggested that FLUAVsw-specific CD4+ T cells included both central memory and effector memory populations. Three out of six animals showed a strong increase of Ki-67+perforin+ CD8β+ T cells in blood one week post infection. Blood-derived FLUAVsw-specific CD8β+ T cells could be identified after an in vitro expansion phase and were multifunctional in terms of CD107a expression and co-production of IFN-γ and TNF-α. These data show that multifunctional T cells are generated in response to FLUAVsw infection of pigs, supporting the idea that T cells contribute to the efficient control of infection.
Collapse
Affiliation(s)
- Stephanie C Talker
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria.
| | - Hanna C Koinig
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria. .,University Clinic for Swine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria.
| | - Maria Stadler
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria.
| | - Robert Graage
- University Clinic for Swine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria. .,Present address: Institute of Veterinary Pathology, Vetsuisse-Faculty, University of Zurich, Zurich, Switzerland.
| | - Eva Klingler
- University Clinic for Swine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria.
| | - Andrea Ladinig
- University Clinic for Swine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria.
| | - Kerstin H Mair
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria.
| | - Sabine E Hammer
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria.
| | - Herbert Weissenböck
- Institute of Pathology and Forensic Veterinary Medicine, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria.
| | - Ralf Dürrwald
- Viral Vaccines, Business Unit Animal Health, IDT Biologika GmbH, Dessau-Rosslau, Germany.
| | - Mathias Ritzmann
- University Clinic for Swine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria. .,Present address: Clinic for Swine, Ludwig-Maximilians-University, Munich, Germany.
| | - Armin Saalmüller
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria.
| | - Wilhelm Gerner
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria.
| |
Collapse
|
80
|
De Vleeschauwer A, Qiu Y, Van Reeth K. Vaccination-challenge studies with a Port Chalmers/73 (H3N2)-based swine influenza virus vaccine: Reflections on vaccine strain updates and on the vaccine potency test. Vaccine 2015; 33:2360-6. [PMID: 25804707 DOI: 10.1016/j.vaccine.2015.03.031] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 02/18/2015] [Accepted: 03/11/2015] [Indexed: 11/30/2022]
Abstract
The human A/Port Chalmers/1/73 (H3N2) influenza virus strain, the supposed ancestor of European H3N2 swine influenza viruses (SIVs), was used in most commercial SIV vaccines in Europe until recently. If manufacturers want to update vaccine strains, they have to perform laborious intratracheal (IT) challenge experiments and demonstrate reduced virus titres in the lungs of vaccinated pigs. We aimed to examine (a) the ability of a Port Chalmers/73-based commercial vaccine to induce cross-protection against a contemporary European H3N2 SIV and serologic cross-reaction against H3N2 SIVs from Europe and North America and (b) the validity of intranasal (IN) challenge and virus titrations of nasal swabs as alternatives for IT challenge and titrations of lung tissue in vaccine potency tests. Pigs were vaccinated with Suvaxyn Flu(®) and challenged by the IT or IN route with sw/Gent/172/08. Post-vaccination sera were examined in haemagglutination-inhibition assays against vaccine and challenge strains and additional H3N2 SIVs from Europe and North America, including an H3N2 variant virus. Tissues of the respiratory tract and nasal swabs were collected 3 days post challenge (DPCh) and from 0-7 DPCh, respectively, and examined by virus titration. Two vaccinations consistently induced cross-reactive antibodies against European H3N2 SIVs from 1998-2012, but minimal or undetectable antibody titres against North American viruses. Challenge virus titres in the lungs, trachea and nasal mucosa of the vaccinated pigs were significantly reduced after both IT and IN challenge. Yet the reduction of virus titres and nasal shedding was greater after IT challenge. The Port Chalmers/73-based vaccine still offered protection against a European H3N2 SIV isolated 35 years later and with only 86.9% amino acid homology in its HA1, but it is unlikely to protect against H3N2 SIVs that are endemic in North America. We use our data to reflect on vaccine strain updates and on the vaccine potency test.
Collapse
Affiliation(s)
- Annebel De Vleeschauwer
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820 Merelbeke, Belgium
| | - Yu Qiu
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820 Merelbeke, Belgium
| | - Kristien Van Reeth
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820 Merelbeke, Belgium.
| |
Collapse
|
81
|
Integrative analysis of differentially expressed microRNAs of pulmonary alveolar macrophages from piglets during H1N1 swine influenza A virus infection. Sci Rep 2015; 5:8167. [PMID: 25639204 PMCID: PMC5389138 DOI: 10.1038/srep08167] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 01/08/2015] [Indexed: 12/15/2022] Open
Abstract
H1N1 swine influenza A virus (H1N1 SwIV) is one key subtype of influenza viruses with pandemic potential. MicroRNAs (miRNAs) are endogenous small RNA molecules that regulate gene expression. MiRNAs relevant with H1N1 SwIV have rarely been reported. To understand the biological functions of miRNAs during H1N1 SwIV infection, this study profiled differentially expressed (DE) miRNAs in pulmonary alveolar macrophages from piglets during the H1N1 SwIV infection using a deep sequencing approach, which was validated by quantitative real-time PCR. Compared to control group, 70 and 16 DE miRNAs were respectively identified on post-infection day (PID) 4 and PID 7. 56 DE miRNAs were identified between PID 4 and PID 7. Our results suggest that most host miRNAs are down-regulated to defend the H1N1 SwIV infection during the acute phase of swine influenza whereas their expression levels gradually return to normal during the recovery phase to avoid the occurrence of too severe porcine lung damage. In addition, targets of DE miRNAs were also obtained, for which bioinformatics analyses were performed. Our results would be useful for investigating the functions and regulatory mechanisms of miRNAs in human influenza because pig serves as an excellent animal model to study the pathogenesis of human influenza.
Collapse
|
82
|
Sandbulte MR, Spickler AR, Zaabel PK, Roth JA. Optimal Use of Vaccines for Control of Influenza A Virus in Swine. Vaccines (Basel) 2015; 3:22-73. [PMID: 26344946 PMCID: PMC4494241 DOI: 10.3390/vaccines3010022] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 01/09/2015] [Accepted: 01/19/2015] [Indexed: 12/29/2022] Open
Abstract
Influenza A virus in swine (IAV-S) is one of the most important infectious disease agents of swine in North America. In addition to the economic burden of IAV-S to the swine industry, the zoonotic potential of IAV-S sometimes leads to serious public health concerns. Adjuvanted, inactivated vaccines have been licensed in the United States for over 20 years, and there is also widespread usage of autogenous/custom IAV-S vaccines. Vaccination induces neutralizing antibodies and protection against infection with very similar strains. However, IAV-S strains are so diverse and prone to mutation that these vaccines often have disappointing efficacy in the field. This scientific review was developed to help veterinarians and others to identify the best available IAV-S vaccine for a particular infected herd. We describe key principles of IAV-S structure and replication, protective immunity, currently available vaccines, and vaccine technologies that show promise for the future. We discuss strategies to optimize the use of available IAV-S vaccines, based on information gathered from modern diagnostics and surveillance programs. Improvements in IAV-S immunization strategies, in both the short term and long term, will benefit swine health and productivity and potentially reduce risks to public health.
Collapse
Affiliation(s)
- Matthew R Sandbulte
- Center for Food Security and Public Health, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA.
| | - Anna R Spickler
- Center for Food Security and Public Health, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA.
| | - Pamela K Zaabel
- Center for Food Security and Public Health, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA.
| | - James A Roth
- Center for Food Security and Public Health, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA.
| |
Collapse
|
83
|
Kreikemeier CA, Engle TB, Lucot KL, Kachman SD, Burkey TE, Ciobanu DC. Genome-wide analysis of TNF-alpha response in pigs challenged with porcine circovirus 2b. Anim Genet 2015; 46:205-8. [PMID: 25643812 DOI: 10.1111/age.12262] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2014] [Indexed: 11/30/2022]
Abstract
Tumor necrosis factor alpha (TNF-α) is a pro-inflammatory cytokine with a role in activating adaptive immunity to viral infections. By inhibiting the capacity of plasmacytoid dendritic cells to produce interferon-α and TNF-α, porcine circovirus 2 (PCV2) limits the maturation of myeloid dendritic cells and impairs their ability to recognize viral and bacterial antigens. Previously, we reported QTL for viremia and immune response in PCV2-infected pigs. In this study, we analyzed phenotypic and genetic relationships between TNF-α protein levels, a potential indicator of predisposition to PCV2 co-infection, and PCV2 susceptibility. Following experimental challenge with PCV2b, TNF-α reached the peak at 21 days post-infection (dpi), at which time a difference was observed between pigs that expressed extreme variation in viremia and growth (P < 0.10). A genome-wide association study (n = 297) revealed that genotypes of 56,433 SNPs explained 73.9% of the variation in TNF-α at 21 dpi. Major SNPs were identified on SSC8, SSC10 and SSC14. Haplotypes based on SNPs from a SSC8 (9 Mb) 1-Mb window were associated with variation in TNF-α (P < 0.02), IgG (P = 0.05) and IgM (P < 0.13) levels at 21 dpi. Potential overlap of regulatory mechanisms was supported by the correlations between genomic prediction values of TNF-α and PCV2 antibodies (21 dpi, r > 0.22), viremia (14-21 dpi, P > 0.29) and viral load (r = 0.31, P < 0.0001). Characterization of the QTL regions uncovered genes that could influence variation in TNF-α levels as well as T- and B-cell development, which can affect disease susceptibility.
Collapse
Affiliation(s)
- C A Kreikemeier
- Animal Science Department, University of Nebraska, Lincoln, NE, 68583, USA
| | | | | | | | | | | |
Collapse
|
84
|
Abstract
Influenza A viruses (IAVs) infect a variety of hosts, including humans, swine, and various avian species. The annual influenza disease burden in the human population remains significant even with current vaccine usage, and much about the pathogenesis and transmission of influenza viruses in humans remains unclear. Thus, animal models are a fundamental tool for influenza research to understand mechanisms of virulence and to develop more efficacious vaccines and forms of prevention or treatment. The choice of experimental model to be used should be based on the species characteristics and similarities to humans, and how the limitations of each host interfere the least with the parameters studied. Influenza virus infection in swine has many similarities with that in humans: the same subtypes are endemic in both species, there has been repeated exchange of viruses between these hosts, the clinical manifestation and pathogenesis are similar, and there is a similar distribution of IAV receptors in the respiratory tract. Considering these common characteristics, and the similarities between humans and swine in terms of genetics, anatomy, and physiology, pigs represent an excellent yet often overlooked model for biomedical research and the study of IAV infection.
Collapse
Affiliation(s)
- Daniela S Rajao
- Daniela S. Rajao, DVM, PhD, is a visiting research microbiologist post-doctoral associate, and Amy L. Vincent, DVM, PhD, is a research veterinary medical officer in the Virus and Prion Research Unit at the National Animal Disease Center of the Agricultural Research Service (ARS) of the United States Department of Agriculture (USDA) in Ames, Iowa
| | - Amy L Vincent
- Daniela S. Rajao, DVM, PhD, is a visiting research microbiologist post-doctoral associate, and Amy L. Vincent, DVM, PhD, is a research veterinary medical officer in the Virus and Prion Research Unit at the National Animal Disease Center of the Agricultural Research Service (ARS) of the United States Department of Agriculture (USDA) in Ames, Iowa
| |
Collapse
|
85
|
Thomas M, Wang Z, Sreenivasan CC, Hause BM, Gourapura J Renukaradhya, Li F, Francis DH, Kaushik RS, Khatri M. Poly I:C adjuvanted inactivated swine influenza vaccine induces heterologous protective immunity in pigs. Vaccine 2014; 33:542-8. [PMID: 25437101 PMCID: PMC7115561 DOI: 10.1016/j.vaccine.2014.11.034] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Revised: 11/14/2014] [Accepted: 11/19/2014] [Indexed: 11/28/2022]
Abstract
Intranasal administration of Poly I:C adjuvanted bivalent swine influenza vaccine induced challenge virus-specific HI antibodies. Poly I:C adjuvanted vaccine also induced IgA and IgG antibodies in the lungs. Poly I:C adjuvanted vaccine provided protection against antigenic variant and heterologous swine influenza viruses.
Swine influenza is widely prevalent in swine herds in North America and Europe causing enormous economic losses and a public health threat. Pigs can be infected by both avian and mammalian influenza viruses and are sources of generation of reassortant influenza viruses capable of causing pandemics in humans. Current commercial vaccines provide satisfactory immunity against homologous viruses; however, protection against heterologous viruses is not adequate. In this study, we evaluated the protective efficacy of an intranasal Poly I:C adjuvanted UV inactivated bivalent swine influenza vaccine consisting of Swine/OH/24366/07 H1N1 and Swine/CO/99 H3N2, referred as PAV, in maternal antibody positive pigs against an antigenic variant and a heterologous swine influenza virus challenge. Groups of three-week-old commercial-grade pigs were immunized intranasally with PAV or a commercial vaccine (CV) twice at 2 weeks intervals. Three weeks after the second immunization, pigs were challenged with the antigenic variant Swine/MN/08 H1N1 (MN08) and the heterologous Swine/NC/10 H1N2 (NC10) influenza virus. Antibodies in serum and respiratory tract, lung lesions, virus shedding in nasal secretions and virus load in lungs were assessed. Intranasal administration of PAV induced challenge viruses specific-hemagglutination inhibition- and IgG antibodies in the serum and IgA and IgG antibodies in the respiratory tract. Importantly, intranasal administration of PAV provided protection against the antigenic variant MN08 and the heterologous NC10 swine influenza viruses as evidenced by significant reductions in lung virus load, gross lung lesions and significantly reduced shedding of challenge viruses in nasal secretions. These results indicate that Poly I:C or its homologues may be effective as vaccine adjuvants capable of generating cross-protective immunity against antigenic variants/heterologous swine influenza viruses in pigs.
Collapse
Affiliation(s)
- Milton Thomas
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD, USA
| | - Zhao Wang
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD, USA
| | - Chithra C Sreenivasan
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD, USA
| | - Ben M Hause
- Department of Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, KS, USA
| | - Gourapura J Renukaradhya
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691, USA
| | - Feng Li
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, USA; Department of Biology and Microbiology, South Dakota State University, Brookings, SD, USA
| | - David H Francis
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, USA
| | - Radhey S Kaushik
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, USA; Department of Biology and Microbiology, South Dakota State University, Brookings, SD, USA
| | - Mahesh Khatri
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691, USA.
| |
Collapse
|
86
|
Identification of Niche Conditions Supporting Short-term Culture of Spermatogonial Stem Cells Derived from Porcine Neonatal Testis. JOURNAL OF ANIMAL REPRODUCTION AND BIOTECHNOLOGY 2014. [DOI: 10.12750/jet.2014.29.3.221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
87
|
Forberg H, Hauge AG, Valheim M, Garcon F, Nunez A, Gerner W, Mair KH, Graham SP, Brookes SM, Storset AK. Early responses of natural killer cells in pigs experimentally infected with 2009 pandemic H1N1 influenza A virus. PLoS One 2014; 9:e100619. [PMID: 24955764 PMCID: PMC4067341 DOI: 10.1371/journal.pone.0100619] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 05/29/2014] [Indexed: 12/21/2022] Open
Abstract
Natural killer (NK) cells are important players in the innate immune response against influenza A virus and the activating receptor NKp46, which binds hemagglutinin on the surface of infected cells, has been assigned a role in this context. As pigs are natural hosts for influenza A viruses and pigs possess both NKp46− and NKp46+ NK cells, they represent a good animal model for studying the role of the NKp46 receptor during influenza. We explored the role of NK cells in piglets experimentally infected with 2009 pandemic H1N1 influenza virus by flow cytometric analyses of cells isolated from blood and lung tissue and by immunostaining of lung tissue sections. The number of NKp46+ NK cells was reduced while NKp46− NK cells remained unaltered in the blood 1–3 days after infection. In the lungs, the intensity of NKp46 expression on NK cells was increased during the first 3 days, and areas where influenza virus nucleoprotein was detected were associated with increased numbers of NKp46+ NK cells when compared to uninfected areas. NKp46+ NK cells in the lung were neither found to be infected with influenza virus nor to be undergoing apoptosis. The binding of porcine NKp46 to influenza virus infected cells was verified in an in vitro assay. These data support the involvement of porcine NKp46+ NK cells in the local immune response against influenza virus.
Collapse
Affiliation(s)
- Hilde Forberg
- Department of Laboratory Services, Norwegian Veterinary Institute, Oslo, Norway
- * E-mail:
| | - Anna G. Hauge
- Department of Laboratory Services, Norwegian Veterinary Institute, Oslo, Norway
| | - Mette Valheim
- Department of Laboratory Services, Norwegian Veterinary Institute, Oslo, Norway
| | - Fanny Garcon
- Virology Department, Animal Health and Veterinary Laboratories Agency, Addlestone, United Kingdom
| | - Alejandro Nunez
- Pathology Department, Animal Health and Veterinary Laboratories Agency, Addlestone, United Kingdom
| | - Wilhelm Gerner
- Department of Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Kerstin H. Mair
- Department of Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Simon P. Graham
- Virology Department, Animal Health and Veterinary Laboratories Agency, Addlestone, United Kingdom
| | - Sharon M. Brookes
- Virology Department, Animal Health and Veterinary Laboratories Agency, Addlestone, United Kingdom
| | - Anne K. Storset
- Department of Food Safety and Infection Biology, Norwegian University of Life Sciences, Oslo, Norway
| |
Collapse
|
88
|
Pomorska-Mól M, Kwit K, Markowska-Daniel I, Kowalski C, Pejsak Z. Local and systemic immune response in pigs during subclinical and clinical swine influenza infection. Res Vet Sci 2014; 97:412-21. [PMID: 25000875 DOI: 10.1016/j.rvsc.2014.06.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Revised: 05/26/2014] [Accepted: 06/08/2014] [Indexed: 11/18/2022]
Abstract
Local and systemic immune responses in pigs intranasally (IN) and intratracheally (IT) inoculated with swine influenza virus (SIV) were studied. No clinical signs were observed in IN-inoculated pigs, while IT-inoculated pigs developed typical signs of influenza. Significantly higher titres of specific antibodies and changes of haematological parameters were found only in IT-inoculated pigs. Because positive correlations between viral titre, local cytokine concentration, and lung pathology have been observed, we hypothesise that both viral load and the local secretion of cytokines play a role in the induction of lung lesions. It could be that a higher replication of SIV stimulates immune cells to secrete higher amounts of cytokines. The results of the present study indicate that pathogenesis of SIV is dependent on both, the damage caused to the lung parenchyma directly by virus, and the effects on the cells of the host's immune system.
Collapse
Affiliation(s)
- M Pomorska-Mól
- Department of Swine Diseases, National Veterinary Research Institute, 24-100 Pulawy, Poland.
| | - K Kwit
- Department of Swine Diseases, National Veterinary Research Institute, 24-100 Pulawy, Poland
| | - I Markowska-Daniel
- Department of Swine Diseases, National Veterinary Research Institute, 24-100 Pulawy, Poland
| | - C Kowalski
- Department of Pharmacology, Faculty of Veterinary Medicine, University of Life Sciences, 20-033 Lublin, Poland
| | - Z Pejsak
- Department of Swine Diseases, National Veterinary Research Institute, 24-100 Pulawy, Poland
| |
Collapse
|
89
|
Low infectivity of a novel avian-origin H7N9 influenza virus in pigs. Arch Virol 2014; 159:2745-9. [PMID: 24906526 DOI: 10.1007/s00705-014-2143-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 05/31/2014] [Indexed: 10/25/2022]
Abstract
We studied the pathogenesis and transmissibility of a novel avian-origin H7N9 influenza virus in pigs. When pigs were infected with H7N9 influenza virus, they did not show any clear clinical signs (such as sneezing, fever and loss of body weight), and they shed viruses through their noses for 2 days after infection. No transmission occurred between infected and naïve pigs. Pigs suffered from mild pneumonia, which was accompanied by the induction of inflammatory cytokines and chemokines such as IL-8 and CCL1. Taken together, our results suggest that pigs may not play an active role in transmitting H7N9 influenza virus to mammals.
Collapse
|
90
|
Pomorska-Mól M, Markowska-Daniel I, Kwit K, Czyżewska E, Dors A, Rachubik J, Pejsak Z. Immune and inflammatory response in pigs during acute influenza caused by H1N1 swine influenza virus. Arch Virol 2014; 159:2605-14. [PMID: 24846450 PMCID: PMC4173111 DOI: 10.1007/s00705-014-2116-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 05/08/2014] [Indexed: 01/05/2023]
Abstract
Swine influenza (SI) is an acute respiratory disease of pigs, caused by swine influenza virus (SIV). Little is known about the inflammatory response in the lung during acute SI and its correlation with clinical signs or lung pathology. Moreover, until now there has been a limited amount of data available on the relationship between the concentrations of pro- and anti-inflammatory cytokines in the lungs and the serum concentration of acute-phase proteins (APPs) in SIV-infected pigs. In the present study, the porcine inflammatory and immune responses during acute influenza caused by H1N1 SIV (SwH1N1) were studied. Nine pigs were infected intratracheally, and five served as controls. Antibodies against SIV were measured by haemagglutination inhibition assay, and the influenza-virus-specific T-cell response was measured using a proliferation assay. C-reactive protein (CRP), haptoglobin (Hp), serum amyloid A (SAA), and pig major acute-phase protein (Pig-MAP) the concentrations in serum and concentration of IL-1β, IL-6, IL-8, IL-10, TNF-α and IFN-γ in lung tissues were measured using commercial ELISAs.
Collapse
Affiliation(s)
- Małgorzata Pomorska-Mól
- Department of Swine Diseases, National Veterinary Research Institute, Partyzantów 57, 24-100, Pulawy, Poland,
| | | | | | | | | | | | | |
Collapse
|
91
|
Pretreatment of epithelial cells with live Streptococcus pneumoniae has no detectable effect on influenza A virus replication in vitro. PLoS One 2014; 9:e90066. [PMID: 24594847 PMCID: PMC3940721 DOI: 10.1371/journal.pone.0090066] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 01/29/2014] [Indexed: 12/16/2022] Open
Abstract
Influenza A virus (IAV) and Streptococcus pneumoniae (pneumococcus) are two major upper respiratory tract pathogens responsible for exacerbated disease in coinfected individuals. Despite several studies showing increased susceptibility to secondary bacterial infections following IAV infection, information on the direct effect of S. pneumoniae on IAV in vitro is unknown. This is an important area of investigation as S. pneumoniae is a common commensal of the human upper respiratory tract, present as an important coinfecting pathogen with IAV infection. A recent study showed that S. pneumoniae enhances human metapneumovirus infection in polarized bronchial epithelial cells in vitro. The aim of the current study was to determine whether treatment of epithelial cells with S. pneumoniae affects IAV replication using a standard immunofluorescence assay (IFA). For this study we used four IAV permissive epithelial cell lines including two human-derived cell lines, 12 pneumococcal strains including recent human clinical isolates which represent different genetic backgrounds and serotypes, and six IAV strains of varying genetic nature and pathogenic potential including the pandemic 2009 H1N1 virus. Our results suggested that pretreatment of MDCK cells with 7.5×106 colony-forming units (CFUs) of live S. pneumoniae resulted in gradual cell-death in a time-dependent manner (0.5 to 4 hr). But, pretreatment of cell lines with 7.5×105 and lower CFUs of S. pneumoniae had no detectable effect on either the morphology of cells or on the IAV replication. However, unlike in epithelial cell lines, due to influence of secreted host factors the effect of pneumococci on IAV replication may be different during coinfections in vivo in the human upper respiratory tract, and in vitro with primary human polarized bronchial epithelial cells.
Collapse
|
92
|
Kowalczyk A, Pomorska-Mól M, Kwit K, Pejsak Z, Rachubik J, Markowska-Daniel I. Cytokine and chemokine mRNA expression profiles in BALF cells isolated from pigs single infected or co-infected with swine influenza virus and Bordetella bronchiseptica. Vet Microbiol 2014; 170:206-12. [PMID: 24629899 DOI: 10.1016/j.vetmic.2014.02.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Revised: 07/04/2013] [Accepted: 02/05/2014] [Indexed: 11/30/2022]
Abstract
Pigs serve as a valuable animal experimental model for several respiratory pathogens, including Swine Influenza Virus (SIV) and Bordetella bronchiseptica (Bbr). To investigate the effect of SIV and Bbr coinfection on cytokine and viral RNA expression, we performed a study in which pigs were inoculated with SIV, Bbr or both pathogens (SIV/Bbr). Our results indicate that Bbr infection alters SIV clearance. Pulmonary lesions in the SIV/Bbr group were more severe when compared to SIV or Bbr groups and Bbr did not cause significant lesions. Broncho-alveolar lavage fluid (BALF) was examined for inflammatory mediators by qPCR. Interferon (IFN)-α, interleukin IL-8, IL-1 peaked in BALF at 2 DPI, while the virus titres and severity of clinical signs were maximal at the same time. Despite its increased expression in co-infected pigs, interferon-α did not enhance SIV clearance, since the viral replication was detected at the same day as the highest IFN levels. The mRNA levels for IFN-α, IL-1β and IL-8 were significantly higher in BALF of co-infected pigs and correlated with enhanced viral RNA titers in lungs, trachea and nasal swabs. Transcription of mRNA for IL-1β was stable in SIV and SIV/Bbr groups throughout all the study. In Bbr group, the levels of mRNAs for IL-1β were significantly higher at 2, 4 and 9 DPI. The mean levels of mRNAs for TNF-α were lower than the levels of other chemokines and cytokines in all infected groups. Transcript levels of IL-10 and IL-4 did not increase at each time points. Overall, SIV replication was increased by Bbr presence and the enhanced production of pro-inflammatory mediators could contribute to the exacerbated pulmonary lesions.
Collapse
Affiliation(s)
- Andrzej Kowalczyk
- The National Veterinary Research Institute, 57 al Partyzantow, 24-100 Puławy, Poland.
| | | | - Krzysztof Kwit
- The National Veterinary Research Institute, 57 al Partyzantow, 24-100 Puławy, Poland
| | - Zygmunt Pejsak
- The National Veterinary Research Institute, 57 al Partyzantow, 24-100 Puławy, Poland
| | - Jarosław Rachubik
- The National Veterinary Research Institute, 57 al Partyzantow, 24-100 Puławy, Poland
| | | |
Collapse
|
93
|
Wang Z, Burwinkel M, Chai W, Lange E, Blohm U, Breithaupt A, Hoffmann B, Twardziok S, Rieger J, Janczyk P, Pieper R, Osterrieder N. Dietary Enterococcus faecium NCIMB 10415 and zinc oxide stimulate immune reactions to trivalent influenza vaccination in pigs but do not affect virological response upon challenge infection. PLoS One 2014; 9:e87007. [PMID: 24489827 PMCID: PMC3904981 DOI: 10.1371/journal.pone.0087007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 12/16/2013] [Indexed: 11/18/2022] Open
Abstract
Swine influenza viruses (SIV) regularly cause significant disease in pigs worldwide. Since there is no causative treatment of SIV, we tested if probiotic Enterococcus (E.) faecium NCIMB 10415 or zinc (Zn) oxide as feed supplements provide beneficial effects upon SIV infection in piglets. Seventy-two weaned piglets were fed three different diets containing either E. faecium or different levels of Zn (2500 ppm, Znhigh; 50 ppm, Znlow). Half of the piglets were vaccinated intramuscularly (VAC) twice with an inactivated trivalent SIV vaccine, while all piglets were then infected intranasally with H3N2 SIV. Significantly higher weekly weight gains were observed in the E. faecium group before virus infection, and piglets in Znhigh and E. faecium groups gained weight after infection while those in the control group (Znlow) lost weight. Using ELISA, we found significantly higher H3N2-specific antibody levels in the E. faecium+VAC group 2 days before and at the day of challenge infection as well as at 4 and 6 days after challenge infection. Higher hemagglutination inhibition (HI) titers were also observed in the Znhigh+VAC and E. faecium+VAC groups at 0, 1 and 4 days after infection. However, there were no significant differences in virus shedding and lung lesions between the dietary groups. Using flow cytometry analysis significantly higher activated T helper cells and cytotoxic T lymphocyte percentages in the PBMCs were detected in the Znhigh and E. faecium groups at single time points after infection compared to the Znlow control group, but no prolonged effect was found. In the BAL cells no influence of dietary supplementation on immune cell percentages could be detected. Our results suggest that feeding high doses of zinc oxide and particularly E. faecium could beneficially influence humoral immune responses after vaccination and recovery from SIV infection, but not affect virus shedding and lung pathology.
Collapse
Affiliation(s)
- Zhenya Wang
- Institut für Virologie, Freie Universität Berlin, Berlin, Germany
| | - Michael Burwinkel
- Institut für Virologie, Freie Universität Berlin, Berlin, Germany
- * E-mail:
| | - Weidong Chai
- Institut für Virologie, Freie Universität Berlin, Berlin, Germany
| | - Elke Lange
- Abteilung für experimentelle Tierhaltung und Biosicherheit, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
| | - Ulrike Blohm
- Institut für Immunologie, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
| | - Angele Breithaupt
- Abteilung für experimentelle Tierhaltung und Biosicherheit, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
- Institut für Veterinärpathologie, Freie Universität Berlin, Berlin, Germany
| | - Bernd Hoffmann
- Institut für Virusdiagnostik, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
| | - Sven Twardziok
- Molekularbiologie und Bioinformatik, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Juliane Rieger
- Institut für Veterinäranatomie, Freie Universität Berlin, Berlin, Germany
| | - Pawel Janczyk
- Bundesinstitut für Risikobewertung, Abteilung für Biologische Sicherheit, Fachgruppe für Molekulare Diagnostik und Genetik, Berlin, Germany
| | - Robert Pieper
- Institut für Tierernährung, Freie Universität Berlin, Berlin, Germany
| | | |
Collapse
|
94
|
Zhao Y, Qin G, Han R, Wang J, Zhang X, Liu D. β-Conglycinin reduces the tight junction occludin and ZO-1 expression in IPEC-J2. Int J Mol Sci 2014; 15:1915-26. [PMID: 24473141 PMCID: PMC3958829 DOI: 10.3390/ijms15021915] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2013] [Revised: 01/12/2014] [Accepted: 01/20/2014] [Indexed: 12/21/2022] Open
Abstract
Soybean allergy presents a health threat to humans and animals. The mechanism by which food/feed allergen β-conglycinin injures the intestinal barrier has not been well understood. In this study, the changes of epithelial permeability, integrity, metabolic activity, the tight junction (TJ) distribution and expression induced by β-conglycinin were evaluated using IPEC-J2 model. The results showed a significant decrease of trans-epithelial electrical resistance (TEER) (p < 0.001) and metabolic activity (p < 0.001) and a remarkable increase of alkaline phosphatase (AP) activity (p < 0.001) in a dose-dependent manner. The expression levels of tight junction occludin and ZO-1 were decreased (p < 0.05). The reduced fluorescence of targets and change of cellular morphology were recorded. The tight junction occludin and ZO-1 mRNA expression linearly declined with increasing β-conglycinin (p < 0.001).
Collapse
Affiliation(s)
- Yuan Zhao
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China.
| | - Guixin Qin
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China.
| | - Rui Han
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China.
| | - Jun Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China.
| | - Xiaodong Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Dandan Liu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China.
| |
Collapse
|
95
|
Abstract
Swine influenza is an acute respiratory disease of pigs caused by influenza A virus (IAV) and characterized by fever followed by lethargy, anorexia, and serous nasal discharge. The disease progresses rapidly and may be complicated when associated with other respiratory pathogens. IAV is one of the most prevalent respiratory pathogens of swine, resulting in substantial economic burden to pork producers. In the past 10-15 years, a dramatic evolution of the IAV in U.S. swine has occurred, resulting in the co-circulation of many antigenically distinct IAV strains, derived from 13 phylogenetically distinct hemagglutinin clusters of H1 and H3 viruses. Vaccination is the most common strategy to prevent influenza in pigs, however, the current diverse IAV epidemiology poses a challenge for the production of efficacious and protective vaccines. A concern regarding the use of traditional inactivated vaccines is the possibility of inducing vaccine-associated enhanced respiratory disease (VAERD) when vaccine virus strains are mismatched with the infecting strain. In this review, we discuss the current epidemiology and pathogenesis of swine influenza in the United States, different vaccines platforms with potential to control influenza in pigs, and the factors associated with vaccine-associated disease enhancement.
Collapse
|
96
|
Vandecasteele T, Vandevelde K, Doom M, Van Mulken E, Simoens P, Cornillie P. The Pulmonary Veins of the Pig as an Anatomical Model for the Development of a New Treatment for Atrial Fibrillation. Anat Histol Embryol 2013; 44:1-12. [PMID: 24372538 DOI: 10.1111/ahe.12099] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Accepted: 11/29/2013] [Indexed: 11/30/2022]
Affiliation(s)
- T. Vandecasteele
- Department of Morphology; Faculty of Veterinary Medicine; Ghent University; Salisburylaan 133 9820 Merelbeke Belgium
| | - K. Vandevelde
- Department of Morphology; Faculty of Veterinary Medicine; Ghent University; Salisburylaan 133 9820 Merelbeke Belgium
| | - M. Doom
- Department of Morphology; Faculty of Veterinary Medicine; Ghent University; Salisburylaan 133 9820 Merelbeke Belgium
| | - E. Van Mulken
- Department of Morphology; Faculty of Veterinary Medicine; Ghent University; Salisburylaan 133 9820 Merelbeke Belgium
| | - P. Simoens
- Department of Morphology; Faculty of Veterinary Medicine; Ghent University; Salisburylaan 133 9820 Merelbeke Belgium
| | - P. Cornillie
- Department of Morphology; Faculty of Veterinary Medicine; Ghent University; Salisburylaan 133 9820 Merelbeke Belgium
| |
Collapse
|
97
|
Abstract
Influenza has been recognized as a respiratory disease in swine since its first appearance concurrent with the 1918 "Spanish flu" human pandemic. All influenza viruses of significance in swine are type A, subtype H1N1, H1N2, or H3N2 viruses. Influenza viruses infect epithelial cells lining the surface of the respiratory tract, inducing prominent necrotizing bronchitis and bronchiolitis and variable interstitial pneumonia. Cell death is due to direct virus infection and to insult directed by leukocytes and cytokines of the innate immune system. The most virulent viruses consistently express the following characteristics of infection: (1) higher or more prolonged virus replication, (2) excessive cytokine induction, and (3) replication in the lower respiratory tract. Nearly all the viral proteins contribute to virulence. Pigs are susceptible to infection with both human and avian viruses, which often results in gene reassortment between these viruses and endemic swine viruses. The receptors on the epithelial cells lining the respiratory tract are major determinants of infection by influenza viruses from other hosts. The polymerases, especially PB2, also influence cross-species infection. Methods of diagnosis and characterization of influenza viruses that infect swine have improved over the years, driven both by the availability of new technologies and by the necessity of keeping up with changes in the virus. Testing of oral fluids from pigs for virus and antibody is a recent development that allows efficient sampling of large numbers of animals.
Collapse
Affiliation(s)
- B H Janke
- DVM, PhD, Veterinary Diagnostic Laboratory, Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA.
| |
Collapse
|
98
|
Crisci E, Mussá T, Fraile L, Montoya M. Review: Influenza virus in pigs. Mol Immunol 2013; 55:200-11. [PMID: 23523121 DOI: 10.1016/j.molimm.2013.02.008] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2012] [Revised: 02/23/2013] [Accepted: 02/25/2013] [Indexed: 12/19/2022]
|
99
|
EFSA Panel on Animal Health and Welfare (AHAW), European Centre for Disease Prevention and Control, European Medicines Agency. Scientific opinion on the possible risks posed by the influenza A (H3N2v) virus for animal health and its potential spread and implications for animal and human health. EFSA J 2013. [DOI: 10.2903/j.efsa.2013.3383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
100
|
Skovgaard K, Cirera S, Vasby D, Podolska A, Breum SØ, Dürrwald R, Schlegel M, Heegaard PMH. Expression of innate immune genes, proteins and microRNAs in lung tissue of pigs infected experimentally with influenza virus (H1N2). Innate Immun 2013; 19:531-44. [PMID: 23405029 DOI: 10.1177/1753425912473668] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
This study aimed at providing a better understanding of the involvement of innate immune factors, including miRNA, in the local host response to influenza virus infection. Twenty pigs were challenged by influenza A virus subtype H1N2. Expression of microRNA (miRNA), mRNA and proteins were quantified in lung tissue at different time points after challenge (24 h, 72 h and 14 d post-infection (p.i.). Several groups of genes were significantly regulated according to time point and infection status including pattern recognition receptors (TLR2, TLR3, TLR7, retinoic acid-inducible gene I, melanoma differentiation associated protein-5), IFN and IFN-induced genes (IFN-β, IFN-γ, IRF7, STAT1, ISG15 and OASL), cytokines (IL-1 β, IL-1RN, IL-6, IL-7, IL-10, IL-12A, TNF-α, CCL2, CCL3 and CXCL10) and several acute phase proteins. Likewise, the following miRNAs were differentially expressed in one or more time groups compared with the control pigs: miR-15a, miR-21, miR-146, miR-206, miR-223 and miR-451. At d 1 p.i. lung tissue protein levels of IL-6, IL-12 and IFN-α were significantly increased compared with the control group, and haptoglobin and C-reactive protein were significantly increased at d 3 p.i. Our results suggest that, in addition to a wide range of innate immune factors, miRNAs may also be involved in controlling acute influenza infection in pigs.
Collapse
Affiliation(s)
- Kerstin Skovgaard
- 1Innate Immunology Group, National Veterinary Institute, Technical University of Denmark, Copenhagen, Denmark
| | | | | | | | | | | | | | | |
Collapse
|