1
|
Liu H, Pan S, Wang C, Yang W, Wei X, He Y, Xu T, Shi K, Si H. Review of respiratory syndromes in poultry: pathogens, prevention, and control measures. Vet Res 2025; 56:101. [PMID: 40382667 DOI: 10.1186/s13567-025-01506-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 10/22/2024] [Indexed: 05/20/2025] Open
Abstract
Respiratory syndromes (RS) include a variety of diseases that lead to respiratory dysfunction, resulting in significant economic losses for the poultry industry. Infectious agents and unfavourable environmental factors cause these respiratory diseases, and rapid transmission, high morbidity rates, and frequent mixed infections characterise them. The challenge in preventing and treating these diseases arises from the complexity of their triggers and the potential for secondary infections. Current vaccines often do not provide effective prevention, and the overuse of certain medications can lead to increased bacterial resistance, complicating prevention and control efforts. This review article examines the common sources of respiratory infections in poultry flocks, including infectious bronchitis virus, avian influenza virus, Newcastle disease virus, infectious laryngotracheitis virus, avian metapneumovirus, pathogenic Escherichia coli, Haemophilus paragallinarum, Mycoplasma gallisepticum, and Chlamydia. It also considers non-infectious factors such as adverse environmental conditions and management errors. The article provides an updated, comprehensive overview of widespread and economically significant poultry respiratory pathogens. It briefly discusses detection technology and vaccine development based on the transmission characteristics of RS. Furthermore, it explores prevention and control measures such as combination drug strategies and antibiotic alternatives to enhance understanding and implementation of effective disease prevention and control measures.
Collapse
Affiliation(s)
- Huixin Liu
- College of Animal Science and Technology, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi grass station, Guangxi University, Nanning, 530004, China
| | - Sijia Pan
- College of Animal Science and Technology, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi grass station, Guangxi University, Nanning, 530004, China
| | - Chenchen Wang
- College of Animal Science and Technology, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi grass station, Guangxi University, Nanning, 530004, China
| | - Wenwen Yang
- College of Animal Science and Technology, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi grass station, Guangxi University, Nanning, 530004, China
| | - Xiaofang Wei
- College of Animal Science and Technology, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi grass station, Guangxi University, Nanning, 530004, China
| | - Yang He
- College of Animal Science and Technology, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi grass station, Guangxi University, Nanning, 530004, China
| | - Ting Xu
- College of Animal Science and Technology, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi grass station, Guangxi University, Nanning, 530004, China
| | - Kaichuang Shi
- College of Animal Science and Technology, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi grass station, Guangxi University, Nanning, 530004, China
- Guangxi Center for Animal Disease Control and Prevention, Nanning, 530001, China
| | - Hongbin Si
- College of Animal Science and Technology, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi grass station, Guangxi University, Nanning, 530004, China.
| |
Collapse
|
2
|
Yao Z, Li J, Shi Y, Sun T, Yang X, Mao J, Quan R, Wang D, Wang D, Zhou J, Liu J, Chu J, Hou L. Isolation and identification of a subtype C avian metapneumovirus in chickens in Jiangsu, China. Microb Pathog 2025; 202:107394. [PMID: 40021030 DOI: 10.1016/j.micpath.2025.107394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/16/2025] [Accepted: 02/18/2025] [Indexed: 03/03/2025]
Abstract
Avian metapneumovirus (aMPV) is a viral pathogen that mainly causes respiratory signs and drops in egg production in turkeys, chickens, and ducks. Here, an aMPV subgroup C (aMPV/C) strain, designated GX22-01, was isolated and identified from severe respiratory disease in broiler breeder chickens in 2022 in Jiangsu, China, as evidenced by indirect immunofluorescence and western blotting using specific anti-viral protein antibodies and by sequence analysis of viral nucleoprotein (N) gene. N gene sequencing indicated that the GX22-01 strain shares a high identity (94.3%-99.8 %) with aMPV/C isolates, especially with Chinese aMPV/C isolates from ducks and chickens, which are divided into aMPV/C cluster through N gene-based phylogenetic analysis. The aMPV/C GX22-01 strain was continuously passaged in Vero cells and the viral titers approximately reached 106.0 TCID50/0.1 mL. Pathogenic analysis showed that aMPV/C GX22-01 strain inoculation caused respiratory signs in 2-week-old specific-pathogen-free (SPF) chickens and resulted in pathogenic damage in tracheae and lung tissues, accompanied by positive viral signals using indirect immunohistochemistry. These results provide epidemiological and pathogenic data for developing effective measures against aMPV/C infection in China.
Collapse
Affiliation(s)
- Zhiyan Yao
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Jingyi Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Yongyan Shi
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Tong Sun
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Xiaoyu Yang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Jingyu Mao
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Rong Quan
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Dan Wang
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Dedong Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Jianwei Zhou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Jue Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Jun Chu
- College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China.
| | - Lei Hou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China.
| |
Collapse
|
3
|
Sugrue RJ, Tan BH. The link between respiratory syncytial virus (RSV) morphogenesis and virus transmission: Towards a paradigm for understanding RSV transmission in the upper airway. Virology 2025; 604:110413. [PMID: 39869971 DOI: 10.1016/j.virol.2025.110413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/10/2025] [Accepted: 01/16/2025] [Indexed: 01/29/2025]
Abstract
Respiratory syncytial virus (RSV) particle assembly occurs on the surface of infected cells at specialized membrane domain called lipid rafts. The mature RSV particles assemble as filamentous projections called virus filaments, and these structures form on the surface of many permissive cell types indicating that this is a robust feature of the RSV particle assembly. The virus filaments also form on nasal airway organoids systems providing evidence that these structures also have a clinical relevance. Virus filaments also form on cells infected with the closely related human metapneumovirus, suggesting that virus filament formation may be a common feature of assembly process for viruses within the Pneumoviridae family. During RSV infection these virus filaments mediate the localized cell-to-cell spread of virus infection, suggesting that they play an important role in virus transmission. The current understanding of the connection between virus filament formation and virus transmission during RSV infection is presented.
Collapse
Affiliation(s)
- Richard J Sugrue
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Republic of Singapore.
| | - Boon Huan Tan
- LKC School of Medicine, Nanyang Technological University, 11 Mandalay Road, Singapore, 308232, Republic of Singapore
| |
Collapse
|
4
|
Mo J, Mo J. Infectious Laryngotracheitis Virus and Avian Metapneumovirus: A Comprehensive Review. Pathogens 2025; 14:55. [PMID: 39861016 PMCID: PMC11769561 DOI: 10.3390/pathogens14010055] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/08/2025] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
Respiratory avian viral diseases significantly impact the world poultry sector, leading to notable economic losses. The highly contagious DNA virus, infectious laryngotracheitis virus, and the RNA virus, avian metapneumovirus, are well known for their prevalent effects on avian respiratory systems. The infectious laryngotracheitis virus (ILTV), stemming from the Herpesviridae family, manifests as an upper respiratory disease within birds. Characterized by acute respiratory signs, it sporadically emerges worldwide, presenting a persistent threat to poultry health. Avian metapneumovirus (aMPV), belonging to the Pneumoviridae family is identified as the cause behind severe rhinotracheitis in turkeys and swollen head syndrome in chickens. This disease can lead to heightened mortality rates, especially when coupled with secondary bacterial infections. This review offers a comprehensive analysis and understanding of the general properties of these specific avian respiratory viruses, control measures, and their global status.
Collapse
Affiliation(s)
- Jongsuk Mo
- Exotic and Emerging Avian Disease Research Unit, U.S. National Poultry Research Center, Agricultural Research Service, United States Department of Agriculture (USDA), Athens, GA 30605, USA;
| | - Jongseo Mo
- College of Pharmacy, Yeungnam University, Gyeongsan-si 38541, Gyeongsangbuk-do, Republic of Korea
| |
Collapse
|
5
|
Zou Y, Guo Z, Ge XY, Qiu Y. RNA Modifications in Pathogenic Viruses: Existence, Mechanism, and Impacts. Microorganisms 2024; 12:2373. [PMID: 39597761 PMCID: PMC11596894 DOI: 10.3390/microorganisms12112373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/14/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024] Open
Abstract
RNA modification is a key posttranscriptional process playing various biological roles, and one which has been reported to exist extensively in cellular RNAs. Interestingly, recent studies have shown that viral RNAs also contain a variety of RNA modifications, which are regulated dynamically by host modification machinery and play critical roles in different stages of the viral life cycle. In this review, we summarize the reports of four typical modifications reported on viral RNAs, including N6-methyladenosine (m6A), 5-methylcytosine (m5C), N4-acetylcytosine (ac4C), and N1-methyladenosine (m1A), describe the molecular mechanisms of these modification processes, and illustrate their impacts on viral replication, pathogenicity, and innate immune responses. Notably, we find that RNA modifications in different viruses share some common features and mechanisms in their generation, regulation, and function, highlighting the potential for viral RNA modifications and the related host machinery to serve as the targets or bases for the development of antiviral therapeutics and vaccines.
Collapse
Affiliation(s)
| | | | - Xing-Yi Ge
- Hunan Provincial Key Laboratory of Medical Virology, College of Biology, Hunan University, Changsha 410012, China; (Y.Z.); (Z.G.)
| | - Ye Qiu
- Hunan Provincial Key Laboratory of Medical Virology, College of Biology, Hunan University, Changsha 410012, China; (Y.Z.); (Z.G.)
| |
Collapse
|
6
|
Cea-Callejo P, Arca-Lafuente S, Gomez-Lucia E, Doménech A, Biarnés M, Blanco A, Benítez L, Madrid R. An affordable detection system based on RT-LAMP and DNA-nanoprobes for avian metapneumovirus. Appl Microbiol Biotechnol 2024; 108:414. [PMID: 38985204 PMCID: PMC11236856 DOI: 10.1007/s00253-024-13243-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 06/06/2024] [Accepted: 06/20/2024] [Indexed: 07/11/2024]
Abstract
Airborne animal viral pathogens can rapidly spread and become a global threat, resulting in substantial socioeconomic and health consequences. To prevent and control potential epidemic outbreaks, accurate, fast, and affordable point-of-care (POC) tests are essential. As a proof-of-concept, we have developed a molecular system based on the loop-mediated isothermal amplification (LAMP) technique for avian metapneumovirus (aMPV) detection, an airborne communicable agent mainly infecting turkeys and chickens. For this purpose, a colorimetric system was obtained by coupling the LAMP technique with specific DNA-functionalized AuNPs (gold nanoparticles). The system was validated using 50 different samples (pharyngeal swabs and tracheal tissue) collected from aMPV-infected and non-infected chickens and turkeys. Viral detection can be achieved in about 60 min with the naked eye, with 100% specificity and 87.88% sensitivity for aMPV. In summary, this novel molecular detection system allows suitable virus testing in the field, with accuracy and limit of detection (LOD) values highly close to qRT-PCR-based diagnosis. Furthermore, this system can be easily scalable to a platform for the detection of other viruses, addressing the current gap in the availability of POC tests for viral detection in poultry farming. KEY POINTS: •aMPV diagnosis using RT-LAMP is achieved with high sensitivity and specificity. •Fifty field samples have been visualized using DNA-nanoprobe validation. •The developed system is a reliable, fast, and cost-effective option for POCT.
Collapse
Affiliation(s)
- Pablo Cea-Callejo
- BioAssays SL. Parque Científico de Madrid, Madrid, Spain
- Research Group of "Animal Viruses" of Complutense University of Madrid, Madrid, Spain
| | | | - Esperanza Gomez-Lucia
- Research Group of "Animal Viruses" of Complutense University of Madrid, Madrid, Spain
- Deparment of Animal Health, Veterinary Faculty, Complutense University of Madrid (UCM), Madrid, Spain
| | - Ana Doménech
- Deparment of Animal Health, Veterinary Faculty, Complutense University of Madrid (UCM), Madrid, Spain
| | - Mar Biarnés
- Centro de Sanidad Avícola de Cataluña y Aragón (CESAC), Reus, Spain
| | - Angela Blanco
- Centro de Sanidad Avícola de Cataluña y Aragón (CESAC), Reus, Spain
| | - Laura Benítez
- Department of Genetics, Physiology, and Microbiology, School of Biology, Complutense University of Madrid (UCM), Madrid, Spain.
| | - Ricardo Madrid
- BioAssays SL. Parque Científico de Madrid, Madrid, Spain.
- Research Group of "Animal Viruses" of Complutense University of Madrid, Madrid, Spain.
- Department of Genetics, Physiology, and Microbiology, School of Biology, Complutense University of Madrid (UCM), Madrid, Spain.
| |
Collapse
|
7
|
Franzo G, Legnardi M, Faustini G, Baston R, Poletto F, Cecchinato M, Tucciarone CM. Tracing the Flight: Investigating the Introduction of Avian Metapneumovirus (aMPV) A and B. Animals (Basel) 2024; 14:1786. [PMID: 38929405 PMCID: PMC11200574 DOI: 10.3390/ani14121786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/07/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Avian metapneumovirus (aMPV) has been identified as an important cause of respiratory and reproductive disease, leading to significant productive losses worldwide. Different subtypes have been found to circulate in different regions, with aMPV-A and B posing a significant burden especially in the Old World, and aMPV-C in North America, albeit with limited exceptions of marginal economic relevance. Recently, both aMPV-A and aMPV-B have been reported in the U.S.; however, the route of introduction has not been investigated. In the present study, the potential importation pathways have been studied through phylogenetic and phylodynamic analyses based on a broad collection of partial attachment (G) protein sequences collected worldwide. aMPV-B circulating in the U.S. seems the descendant of Eastern Asian strains, which, in turn, are related to European ones. A likely introduction pathway mediated by wild bird migration through the Beringian crucible, where the East Asian and Pacific American flight paths intersect, appears likely and was previously reported for avian influenza. aMPV-A, on the other hand, showed a Mexican origin, involving strains related to Asian ones. Given the low likelihood of trade or illegal importation, the role of wild birds appears probable also in this case, since the region is covered by different flight paths directed in a North-South direction through America. Since the information on the role of wild birds in aMPV epidemiology is still scarce and scattered, considering the significant practical implications for the poultry industry demonstrated by recent U.S. outbreaks, further surveys on wild birds are encouraged.
Collapse
Affiliation(s)
- Giovanni Franzo
- Department of Animal Medicine, Production and Health, University of Padova, 35020 Legnaro, Italy; (M.L.); (G.F.); (R.B.); (F.P.); (M.C.); (C.M.T.)
| | | | | | | | | | | | | |
Collapse
|
8
|
Zhang Q, Ye H, Liu C, Zhou H, He M, Liang X, Zhou Y, Wang K, Qin Y, Li Z, Chen M. PABP-driven secondary condensed phase within RSV inclusion bodies activates viral mRNAs for ribosomal recruitment. Virol Sin 2024; 39:235-250. [PMID: 38072230 PMCID: PMC11074649 DOI: 10.1016/j.virs.2023.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 12/03/2023] [Indexed: 01/12/2024] Open
Abstract
Inclusion bodies (IBs) of respiratory syncytial virus (RSV) are formed by liquid-liquid phase separation (LLPS) and contain internal structures termed "IB-associated granules" (IBAGs), where anti-termination factor M2-1 and viral mRNAs are concentrated. However, the mechanism of IBAG formation and the physiological function of IBAGs are unclear. Here, we found that the internal structures of RSV IBs are actual M2-1-free viral messenger ribonucleoprotein (mRNP) condensates formed by secondary LLPS. Mechanistically, the RSV nucleoprotein (N) and M2-1 interact with and recruit PABP to IBs, promoting PABP to bind viral mRNAs transcribed in IBs by RNA-recognition motif and drive secondary phase separation. Furthermore, PABP-eIF4G1 interaction regulates viral mRNP condensate composition, thereby recruiting specific translation initiation factors (eIF4G1, eIF4E, eIF4A, eIF4B and eIF4H) into the secondary condensed phase to activate viral mRNAs for ribosomal recruitment. Our study proposes a novel LLPS-regulated translation mechanism during viral infection and a novel antiviral strategy via targeting on secondary condensed phase.
Collapse
Affiliation(s)
- Qiang Zhang
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Hanzhe Ye
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Cong Liu
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Haiwu Zhou
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Mingbin He
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Xiaodong Liang
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Yu Zhou
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Kun Wang
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Yali Qin
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, 430072, China.
| | - Zhifei Li
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, 430072, China.
| | - Mingzhou Chen
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, 430072, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430072, China; Hubei Jiangxia Laboratory, Wuhan, 430200, China.
| |
Collapse
|
9
|
Salles GBC, Pilati GVT, Muniz EC, de Lima Neto AJ, Vogt JR, Dahmer M, Savi BP, Padilha DA, Fongaro G. Trends and Challenges in the Surveillance and Control of Avian Metapneumovirus. Viruses 2023; 15:1960. [PMID: 37766366 PMCID: PMC10535940 DOI: 10.3390/v15091960] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/09/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Among the respiratory pathogens of birds, the Avian Metapneumovirus (aMPV) is one of the most relevant, as it is responsible for causing infections of the upper respiratory tract and may induce respiratory syndromes. aMPV is capable of affecting the reproductive system of birds, directly impacting shell quality and decreasing egg production. Consequently, this infection can cause disorders related to animal welfare and zootechnical losses. The first cases of respiratory syndromes caused by aMPV were described in the 1970s, and today six subtypes (A, B, C, D, and two more new subtypes) have been identified and are widespread in all chicken and turkey-producing countries in the world, causing enormous economic losses for the poultry industry. Conventionally, immunological techniques are used to demonstrate aMPV infection in poultry, however, the identification of aMPV through molecular techniques helped in establishing the traceability of the virus. This review compiles data on the main aMPV subtypes present in different countries; aMPV and bacteria co-infection; vaccination against aMPV and viral selective pressure, highlighting the strategies used to prevent and control respiratory disease; and addresses tools for viral diagnosis and virus genome studies aiming at improving and streamlining pathogen detection and corroborating the development of new vaccines that can effectively protect herds, preventing viral escapes.
Collapse
Affiliation(s)
- Gleidson Biasi Carvalho Salles
- Laboratory of Applied Virology, Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianópolis 88040-900, Brazil; (G.B.C.S.); (G.V.T.P.); (M.D.); (B.P.S.); (D.A.P.)
- Zoetis Industry of Veterinary Products LTDA, São Paulo 04709-111, Brazil; (E.C.M.); (J.R.V.)
| | - Giulia Von Tönnemann Pilati
- Laboratory of Applied Virology, Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianópolis 88040-900, Brazil; (G.B.C.S.); (G.V.T.P.); (M.D.); (B.P.S.); (D.A.P.)
| | - Eduardo Correa Muniz
- Zoetis Industry of Veterinary Products LTDA, São Paulo 04709-111, Brazil; (E.C.M.); (J.R.V.)
| | | | - Josias Rodrigo Vogt
- Zoetis Industry of Veterinary Products LTDA, São Paulo 04709-111, Brazil; (E.C.M.); (J.R.V.)
| | - Mariane Dahmer
- Laboratory of Applied Virology, Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianópolis 88040-900, Brazil; (G.B.C.S.); (G.V.T.P.); (M.D.); (B.P.S.); (D.A.P.)
| | - Beatriz Pereira Savi
- Laboratory of Applied Virology, Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianópolis 88040-900, Brazil; (G.B.C.S.); (G.V.T.P.); (M.D.); (B.P.S.); (D.A.P.)
| | - Dayane Azevedo Padilha
- Laboratory of Applied Virology, Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianópolis 88040-900, Brazil; (G.B.C.S.); (G.V.T.P.); (M.D.); (B.P.S.); (D.A.P.)
| | - Gislaine Fongaro
- Laboratory of Applied Virology, Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianópolis 88040-900, Brazil; (G.B.C.S.); (G.V.T.P.); (M.D.); (B.P.S.); (D.A.P.)
| |
Collapse
|
10
|
Graaf A, Hennig C, Jaschniski KL, Koechling M, Stadler J, Boehmer J, Ripp U, Pohlmann A, Schwarz BA, Beer M, Harder T. Emergenceof swine influenza A virus, porcine respirovirus 1 and swine orthopneumovirus in porcine respiratory disease in Germany. Emerg Microbes Infect 2023:2239938. [PMID: 37470510 PMCID: PMC10402848 DOI: 10.1080/22221751.2023.2239938] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/21/2023]
Abstract
Respiratory disease is a significant economic issue in pig farming, with a complex aetiology that includes swine influenza A viruses (swIAV), which are common in European domestic pig populations. The most recent human influenza pandemic in 2009 showed swIAV's zoonotic potential. Monitoring pathogens and disease control are critical from a preventive standpoint, and are based on quick, sensitive, and specific diagnostic assays capable of detecting and distinguishing currently circulating swIAV in clinical samples. For passive surveillance, a set of multiplex quantitative reverse transcription real-time PCRs (mRT-qPCR) and MinION-directed sequencing was updated and deployed. Several lineages and genotypes of swIAV were shown to be dynamically developing, including novel reassortants between human pandemic H1N1 and the avian-derived H1 lineage of swIAV. Despite this, nearly 70% (842/1216) of individual samples from pigs with respiratory symptoms were swIAV-negative, hinting to different aetiologies. The complex and synergistic interactions of swIAV infections with other viral and bacterial infectious agents contribute to the aggravation of pig respiratory diseases. Using a newly developed mRT-qPCR for the combined detection of swIAV and the recently described porcine respirovirus 1 (PRV1) and swine orthopneumovirus (SOV) widespread co-circulation of PRV1 (19.6%, 238/1216 samples) and SOV (14.2%, 173/1216 samples) was evident. Because of the high incidence of PRV1 and SOV infections in pigs with respiratory disease, these viruses may emerge as new allies in the porcine respiratory disease syndrome.
Collapse
Affiliation(s)
- Annika Graaf
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
| | - Christin Hennig
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
| | | | | | - Julia Stadler
- Clinic for Swine at the Centre for Clinical Veterinary Medicine, Ludwig-Maximilians-University Munich, Oberschleissheim, Germany
| | - Jan Boehmer
- IVD Society for Innovative Veterinary Diagnostics mbH, Seelze-Letter, Germany
| | - Ulrike Ripp
- Vaxxinova diagnostics GmbH, Leipzig, Germany
| | - Anne Pohlmann
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
| | | | - Martin Beer
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
| | - Timm Harder
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
| |
Collapse
|
11
|
Kariithi HM, Volkening JD, Alves VV, Reis-Cunha JL, Arantes LCRV, Fernando FS, Filho TF, da Silva Martins NR, Lemiere S, de Freitas Neto OC, Decanini EL, Afonso CL, Suarez DL. Complete Genome Sequences of Avian Metapneumovirus Subtype B Vaccine Strains from Brazil. Microbiol Resour Announc 2023:e0023523. [PMID: 37162354 DOI: 10.1128/mra.00235-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023] Open
Abstract
Avian metapneumovirus (aMPV) causes a highly contagious upper respiratory and reproductive disease in chickens, turkeys, and ducks. Here, complete genome sequences of aMPV-B vaccine strains BR/1890/E1/19 (PL21, Nemovac; Boehringer Ingelheim Animal Health, Brazil) and BR/1891/E2/19 (1062; Hipraviar, France) were sequenced and compared with the pathogenic field strain VCO3/60616.
Collapse
Affiliation(s)
- Henry M Kariithi
- Exotic and Emerging Avian Viral Diseases Research Unit, Southeast Poultry Research Laboratory, U.S. National Poultry Research Center, USDA-ARS, Athens, Georgia, USA
- Biotechnology Research Institute, Kenya Agricultural and Livestock Research Organization, Nairobi, Kenya
| | | | - Victória Veiga Alves
- Department of Preventive Veterinary Medicine, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | | | | | - Tobias Fernandes Filho
- Department of Preventive Veterinary Medicine, Federal University of Minas Gerais, Belo Horizonte, Brazil
- Boehringer Ingelheim Animal Health, São Paulo, Brazil
| | | | | | | | | | | | - David L Suarez
- Exotic and Emerging Avian Viral Diseases Research Unit, Southeast Poultry Research Laboratory, U.S. National Poultry Research Center, USDA-ARS, Athens, Georgia, USA
| |
Collapse
|
12
|
Conan A, Nekouei O, Paudel S, Ching A, Yau D, Pfeiffer D. Serological survey of avian metapneumovirus in vaccinated and unvaccinated broiler chickens in Hong Kong. Trop Anim Health Prod 2023; 55:179. [PMID: 37119359 DOI: 10.1007/s11250-023-03592-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 04/12/2023] [Indexed: 05/01/2023]
Abstract
In chickens, avian metapneumovirus (aMPV) causes the swollen head syndrome, a respiratory disease often associated with a reduction in egg production. The virus' epidemiology in East and Southeast Asia is poorly understood. An aMPV serological survey was conducted on broiler chicken farms of Hong Kong SAR to assess the seroprevalence of aMPV in unvaccinated batches and the serological status of vaccinated batches. Blood samples were collected from 53-93-day-old chickens in 24 chicken farms of Hong Kong SAR and sera were tested for aMPV antibodies by ELISA. Seroprevalence in aMPV unvaccinated birds was 80.6% (95% confidence interval (CI): 78.9-82.2) with a high variation between batches. Batch-level seroprevalence was not significantly different between birds hatched during the rainy season (74.3%, 95% CI: 64.0-84.5) and the ones hatched during the dry season (88.7%, 95% CI: 80.1-97.3, p = 0.5). The high seroprevalence and high antibody titers that are reported in this study indicate repeated exposure of broiler chickens to aMPV in Hong Kong SAR poultry farms. Based on these results, we recommend improving the surveillance of respiratory pathogens and applying appropriate prophylactic measures against aMPV such as vaccination.
Collapse
Affiliation(s)
- Anne Conan
- Centre for Applied One Health Research and Policy Advice, City University of Hong Kong, Hong Kong SAR, China.
| | - Omid Nekouei
- Centre for Applied One Health Research and Policy Advice, City University of Hong Kong, Hong Kong SAR, China
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong SAR, China
| | - Surya Paudel
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong SAR, China
| | - Arthur Ching
- CityU Veterinary Diagnostic Laboratory, City University of Hong Kong, Hong Kong SAR, China
| | - Denis Yau
- Centre for Applied One Health Research and Policy Advice, City University of Hong Kong, Hong Kong SAR, China
| | - Dirk Pfeiffer
- Centre for Applied One Health Research and Policy Advice, City University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
13
|
Avian Metapneumovirus Subgroup C Phosphoprotein Suppresses Type I Interferon Production by Blocking Interferon Regulatory Factor 3 Nuclear Translocation. Microbiol Spectr 2023; 11:e0341322. [PMID: 36537793 PMCID: PMC9927154 DOI: 10.1128/spectrum.03413-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023] Open
Abstract
Avian metapneumovirus subgroup C (aMPV/C) is an important pathogen that causes upper respiratory symptoms and egg production decline in turkeys and chickens. aMPV/C infection leads to inhibition of the host antiviral immune response. However, our understanding of the molecular mechanisms underlying host immune response antagonized by aMPV/C infection is limited. In this study, we demonstrated that the aMPV/C phosphoprotein (P) inhibits the IFN antiviral signaling pathway triggered by melanoma differentiation gene 5 (MDA5) and reduces interferon β (IFN-β) production and IFN-stimulated genes (ISGs) by targeting IFN regulatory factor 7 (IRF7) but not nuclear factor κB (NF-κB) in DF-1 cells. Moreover, we found that aMPV/C P protein only blocks the nuclear translocation of IRF3 by interacting with IRF3 in HEK-293T cells, instead of affecting IRF3 phosphorylation and inducing IRF3 degradation, which suppresses IRF3 signaling activation and results in a decrease in IFN-β production. Collectively, these results reveal a novel mechanism by which aMPV/C infection disrupts IFN-β production in the host. IMPORTANCE The innate immune response is the first defense line of host cells and organisms against viral infections. When RNA viruses infect cells, viral RNA induces activation of retinoic acid-induced gene I and melanoma differentiation gene 5, which initiates downstream molecules and finally produces type I interferon (IFN-I) to regulate antiviral immune responses. The mechanism for avian metapneumovirus (aMPV) modulating IFN-I production to benefit its replication remains unknown. Here, we demonstrate that phosphoprotein of aMPV subgroup C (aMPV/C) selectively inhibits the nuclear translocation of interferon regulatory 3 (IRF3), instead of affecting the expression and phosphorylation of IRF3, which finally downregulates IFN-I production. This study showed a novel mechanism for aMPV/C infection antagonizing the host IFN response.
Collapse
|
14
|
Lemaitre E, Bougeard S, Allée C, Eterradossi N, Courtillon C, Brown PA. Avian metapneumovirus: A five-plex digital droplet RT-PCR method for identification of subgroups A, B, C, and D. Front Vet Sci 2022; 9:1058294. [PMID: 36458056 PMCID: PMC9705331 DOI: 10.3389/fvets.2022.1058294] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 10/24/2022] [Indexed: 09/29/2023] Open
Abstract
End-point and real-time avian metapneumovirus (AMPV) RT-PCRs have been developed to detect one or two of the four recognized subgroups (A,B,C, and D) simultaneously or for broad range AMPV detection. Current subgroup specific tests target variable areas of the genome which makes these PCRs sensitive to specificity defects as recently documented. In the current study, a single five-plex digital droplet RT-PCR targeting the conserved viral polymerase gene of AMPV, which is less prone to genetic drift, has been designed. This digital droplet RT-PCR was capable of identifying each of the four AMPV subgroups. Each subgroup was identified according to a specifically assigned fluorescent amplitude. Specificity, which was tested including 31 AMPV strains, non-AMPV avian viruses and closely related human respiratory viruses, was 100%. The specific limit of detection for extracted viral RNA was estimated between 1 and 3 copies/μl. This tool simplifies the number of tests required for AMPV genotype diagnostics and should be theoretically less effected by viral genome evolution due to its target region. Ultimately, application of this test will contribute to an improved understanding of the global geographic distribution and subgroup host range of field strains.
Collapse
Affiliation(s)
- Evelyne Lemaitre
- Ploufragan-Plouzané-Niort Laboratory, French Agency for Food, Environmental and Occupational Health & Safety (Anses), VIPAC Unit, WOAH Reference Laboratory for Avian Metapneumovirus Infections, Ploufragan, France
| | - Stéphanie Bougeard
- Ploufragan-Plouzané-Niort Laboratory, French Agency for Food, Environmental and Occupational Health & Safety (Anses), EPISABE Unit, Ploufragan, France
| | - Chantal Allée
- Ploufragan-Plouzané-Niort Laboratory, French Agency for Food, Environmental and Occupational Health & Safety (Anses), VIPAC Unit, WOAH Reference Laboratory for Avian Metapneumovirus Infections, Ploufragan, France
| | - Nicolas Eterradossi
- Ploufragan-Plouzané-Niort Laboratory, French Agency for Food, Environmental and Occupational Health & Safety (Anses), VIPAC Unit, WOAH Reference Laboratory for Avian Metapneumovirus Infections, Ploufragan, France
| | - Céline Courtillon
- Ploufragan-Plouzané-Niort Laboratory, French Agency for Food, Environmental and Occupational Health & Safety (Anses), VIPAC Unit, WOAH Reference Laboratory for Avian Metapneumovirus Infections, Ploufragan, France
| | - Paul Alun Brown
- Ploufragan-Plouzané-Niort Laboratory, French Agency for Food, Environmental and Occupational Health & Safety (Anses), VIPAC Unit, WOAH Reference Laboratory for Avian Metapneumovirus Infections, Ploufragan, France
| |
Collapse
|
15
|
Kariithi HM, Christy N, Decanini EL, Lemiere S, Volkening JD, Afonso CL, Suarez DL. Detection and Genome Sequence Analysis of Avian Metapneumovirus Subtype A Viruses Circulating in Commercial Chicken Flocks in Mexico. Vet Sci 2022; 9:vetsci9100579. [PMID: 36288192 PMCID: PMC9612082 DOI: 10.3390/vetsci9100579] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/14/2022] [Accepted: 10/17/2022] [Indexed: 11/11/2022] Open
Abstract
Avian metapneumoviruses (aMPV subtypes A-D) are respiratory and reproductive pathogens of poultry. Since aMPV-A was initially reported in Mexico in 2014, there have been no additional reports of its detection in the country. Using nontargeted next-generation sequencing (NGS) of FTA card-spotted respiratory samples from commercial chickens in Mexico, seven full genome sequences of aMPV-A (lengths of 13,288-13,381 nucleotides) were de novo assembled. Additionally, complete coding sequences of genes N (n = 2), P and M (n = 7 each), F and L (n = 1 each), M2 (n = 6), SH (n = 5) and G (n = 2) were reference-based assembled from another seven samples. The Mexican isolates phylogenetically group with, but in a distinct clade separate from, other aMPV-A strains. The genome and G-gene nt sequences of the Mexican aMPVs are closest to strain UK/8544/06 (97.22-97.47% and 95.07-95.83%, respectively). Various amino acid variations distinguish the Mexican isolates from each other, and other aMPV-A strains, most of which are in the G (n = 38), F (n = 12), and L (n = 19) proteins. Using our sequence data and publicly available aMPV-A data, we revised a previously published rRT-PCR test, which resulted in different cycling and amplification conditions for aMPV-A to make it more compatible with other commonly used rRT-PCR diagnostic cycling conditions. This is the first comprehensive sequence analysis of aMPVs in Mexico and demonstrates the value of nontargeted NGS to identify pathogens where targeted virus surveillance is likely not routinely performed.
Collapse
Affiliation(s)
- Henry M. Kariithi
- Exotic and Emerging Avian Viral Diseases Research Unit, Southeast Poultry Research Laboratory, U.S. National Poultry Research Center, USDA-ARS, Athens, GA 30605, USA
- Biotechnology Research Institute, Kenya Agricultural and Livestock Research Organization, Kaptagat Rd, Nairobi P.O. Box 57811-00200, Kenya
- Correspondence: (H.M.K.); (D.L.S.); Tel.: +1-(706)-546-3479 (D.L.S.)
| | - Nancy Christy
- Boehringer Ingelheim Animal Health, Guadalajara 44940, Mexico
| | - Eduardo L. Decanini
- Boehringer Ingelheim Animal Health IMETA, Dubai P.O. Box 507066, United Arab Emirates
| | | | | | | | - David L. Suarez
- Exotic and Emerging Avian Viral Diseases Research Unit, Southeast Poultry Research Laboratory, U.S. National Poultry Research Center, USDA-ARS, Athens, GA 30605, USA
- Correspondence: (H.M.K.); (D.L.S.); Tel.: +1-(706)-546-3479 (D.L.S.)
| |
Collapse
|
16
|
Li H, Ma L, Li W, Zheng B, Wang J, Chen S, Wang Y, Ge F, Qin B, Zheng X, Deng Y, Zeng R. Proline metabolism reprogramming of trained macrophages induced by early respiratory infection combined with allergen sensitization contributes to development of allergic asthma in childhood of mice. Front Immunol 2022; 13:977235. [PMID: 36211408 PMCID: PMC9533174 DOI: 10.3389/fimmu.2022.977235] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 09/02/2022] [Indexed: 11/13/2022] Open
Abstract
Background Infants with respiratory syncytial virus (RSV)-associated bronchiolitis are at increased risk of childhood asthma. Recent studies demonstrated that certain infections induce innate immune memory (also termed trained immunity), especially in macrophages, to respond more strongly to future stimuli with broad specificity, involving in human inflammatory diseases. Metabolic reprogramming increases the capacity of the innate immune cells to respond to a secondary stimulation, is a crucial step for the induction of trained immunity. We hypothesize that specific metabolic reprogramming of lung trained macrophages induced by neonatal respiratory infection is crucial for childhood allergic asthma. Objective To address the role of metabolic reprogramming in lung trained macrophages induced by respiratory virus infection in allergic asthma. Methods Neonatal mice were infected and sensitized by the natural rodent pathogen Pneumonia virus of mice (PVM), a mouse equivalent strain of human RSV, combined with ovalbumin (OVA). Lung CD11b+ macrophages in the memory phase were re-stimulated to investigate trained immunity and metabonomics. Adoptive transfer, metabolic inhibitor and restore experiments were used to explore the role of specific metabolic reprogramming in childhood allergic asthma. Results PVM infection combined with OVA sensitization in neonatal mice resulted in non-Th2 (Th1/Th17) type allergic asthma following OVA challenge in childhood of mice. Lung CD11b+ macrophages in the memory phage increased, and showed enhanced inflammatory responses following re-stimulation, suggesting trained macrophages. Adoptive transfer of the trained macrophages mediated the allergic asthma in childhood. The trained macrophages showed metabolic reprogramming after re-stimulation. Notably, proline biosynthesis remarkably increased. Inhibition of proline biosynthesis suppressed the development of the trained macrophages as well as the Th1/Th17 type allergic asthma, while supplement of proline recovered the trained macrophages as well as the allergic asthma. Conclusion Proline metabolism reprogramming of trained macrophages induced by early respiratory infection combined with allergen sensitization contributes to development of allergic asthma in childhood. Proline metabolism could be a well target for prevention of allergic asthma in childhood.
Collapse
Affiliation(s)
- Hanglin Li
- Department of Immunology, Hebei Medical University, Shijiazhuang, China
| | - Linyan Ma
- Department of Immunology, Hebei Medical University, Shijiazhuang, China
| | - Wenjian Li
- Department of Immunology, Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Immune Mechanism and Intervention on Serious Disease in Hebei Province, Hebei Medical University, Shijiazhuang, China
| | - Boyang Zheng
- Department of Immunology, Hebei Medical University, Shijiazhuang, China
- The Fourth Affiliated Hospital of Hebei Medical University, Shijiazhuang, China
| | - Junhai Wang
- Department of Immunology, Hebei Medical University, Shijiazhuang, China
| | - Shunyan Chen
- Department of Immunology, Hebei Medical University, Shijiazhuang, China
| | - Yang Wang
- Department of Immunology, Hebei Medical University, Shijiazhuang, China
| | - Fei Ge
- Department of Immunology, Hebei Medical University, Shijiazhuang, China
| | - Beibei Qin
- Department of Immunology, Hebei Medical University, Shijiazhuang, China
- Clinical Lab, Hebei Provincial People’s Hospital, Shijiazhuang, China
| | - Xiaoqing Zheng
- Department of Immunology, Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Immune Mechanism and Intervention on Serious Disease in Hebei Province, Hebei Medical University, Shijiazhuang, China
| | - Yuqing Deng
- Department of Immunology, Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Immune Mechanism and Intervention on Serious Disease in Hebei Province, Hebei Medical University, Shijiazhuang, China
| | - Ruihong Zeng
- Department of Immunology, Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Immune Mechanism and Intervention on Serious Disease in Hebei Province, Hebei Medical University, Shijiazhuang, China
- *Correspondence: Ruihong Zeng,
| |
Collapse
|
17
|
Respiratory Syncytial virus NS1 protein targets the transactivator binding domain of MED25. J Mol Biol 2022; 434:167763. [PMID: 35907573 DOI: 10.1016/j.jmb.2022.167763] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 06/28/2022] [Accepted: 07/21/2022] [Indexed: 11/20/2022]
Abstract
Human RSV is the leading cause of infantile bronchiolitis in the world and one of the major causes of childhood deaths in resource-poor settings. It is a major unmet target for vaccines and anti-viral drugs. Respiratory syncytial virus has evolved a unique strategy to evade host immune response by coding for two non-structural proteins NS1 and NS2. Recently it was shown that in infected cells, nuclear NS1 could be involved in transcription regulation of host genes linked to innate immune response, via interactions with chromatin and the Mediator complex. Here we identified the MED25 Mediator subunit as an NS1 interactor in a yeast two-hybrid screen. We demonstrate that NS1 directly interacts with MED25 in vitro and in cellula, and that this interaction involves the MED25 transactivator binding ACID domain on the one hand, and the C-terminal α3 helix of NS1, with an additional contribution of the globular domain of NS1, on the other hand. By NMR we show that the NS1 α3 sequence primarily binds to the MED25 ACID H2 face, similarly to the α-helical transactivation domains (TADs) of transcription regulators such as Herpex simplex VP16 and ATF6α, a master regulator of ER stress response activated upon viral infection. Moreover, we found out that the NS1 could compete with ATF6α TAD for binding to MED25. These findings point to a mechanism of NS1 interfering with innate immune response by impairing recruitment by cellular TADs of the Mediator via MED25 and hence transcription of specific genes by RNA polymerase II.
Collapse
|
18
|
PRRSV Infection Induces Gasdermin D-Driven Pyroptosis of Porcine Alveolar Macrophages through NLRP3 Inflammasome Activation. J Virol 2022; 96:e0212721. [PMID: 35758658 PMCID: PMC9327688 DOI: 10.1128/jvi.02127-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
For more than 3 decades, mounting evidence has associated porcine reproductive and respiratory syndrome virus (PRRSV) infection with late-term abortions and stillbirths in sows and respiratory disease in piglets, causing enormous economic losses to the global swine industry. However, to date, the underlying mechanisms of PRRSV-triggered cell death have not been well clarified, especially in the pulmonary inflammatory injury characterized by the massive release of pro-inflammatory factors. Here, we demonstrated that PRRSV infection triggered gasdermin D-mediated host pyroptosis in vitro and in vivo. Mechanistically, PRRSV infection triggered disassembly of the trans-Golgi network (TGN); the dispersed TGN then acted as a scaffold for NLRP3 activation through phosphatidylinositol-4-phosphate. In addition, PRRSV replication-transcription complex (RTC) formation stimulated TGN dispersion and pyroptotic cell death. Furthermore, our results indicated that TMEM41B, an endoplasmic reticulum (ER)-resident host protein, functioned as a crucial host factor in the formation of PRRSV RTC, which is surrounded by the intermediate filament network. Collectively, these findings uncover new insights into clinical features as previously unrecognized mechanisms for PRRSV-induced pathological effects, which may be conducive to providing treatment options for PRRSV-associated diseases and may be conserved during infection by other highly pathogenic viruses. IMPORTANCE Porcine reproductive and respiratory syndrome virus (PRRSV) is one of the pathogens responsible for major economic losses in the global swine industry. Characterizing the detailed process by which PRRSV induces cell death pathways will help us better understand viral pathogenesis and provide implications for therapeutic intervention against PRRSV. Here, we showed that PRRSV infection induces GSDMD-driven host pyroptosis and IL-1β secretion through NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome activation in vitro and in vivo. Furthermore, the molecular mechanisms of PRRSV-induced NLRP3 inflammasome activation and pyroptosis are elucidated here. The dispersed trans-Golgi network (TGN) induced by PRRSV serves as a scaffold for NLRP3 aggregation into multiple puncta via phosphatidylinositol 4-phosphate (PtdIns4P). Moreover, the formation of PRRSV replication-transcription complex is essential for TGN dispersion and host pyroptosis. This research advances our understanding of the PRRSV-mediated inflammatory response and cell death pathways, paving the way for the development of effective treatments for PRRSV diseases.
Collapse
|
19
|
Wang D, Hou L, Zhu N, Yang X, Zhou J, Cui Y, Guo J, Feng X, Liu J. Interaction of Nucleolin with the Fusion Protein of Avian Metapneumovirus Subgroup C Contributes to Viral Replication. Viruses 2022; 14:v14071402. [PMID: 35891383 PMCID: PMC9317408 DOI: 10.3390/v14071402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 06/23/2022] [Accepted: 06/24/2022] [Indexed: 12/02/2022] Open
Abstract
Avian metapneumovirus subgroup C (aMPV/C) is highly pathogenic to various avian species with acute respiratory tract clinicopathology and/or drops in egg production. Nucleolin (NCL), an important nucleolar protein, has been shown to regulate multiple viral replication and serve as a functional receptor for viral entry and internalization. Whether NCL is involved in aMPV/C pathogenesis is not known. In this study, we found that aMPV/C infection altered the subcellular localization of NCL in cultured cells. siRNA-targeted NCL resulted in a remarkable decline in aMPV/C replication in Vero cells. DF-1 cells showed a similar response after CRISPR/Cas9-mediated knock out of NCL during aMPV/C infection. Conversely, NCL overexpression significantly increased aMPV/C replication. Pretreatment with AS1411-a aptamer, a guanine (G)-rich oligonucleotide that forms four-stranded structures and competitively binding to NCL, decreased aMPV/C replication and viral titers in cultured cells. Additionally, we found that the aMPV/C fusion (F) protein specifically interacts with NCL through its central domain and that AS1411 disrupts this interaction, thus inhibiting viral replication. Taken together, these results reveal that the aMPV/C F protein interacts with NCL, which is employed by aMPV/C for efficient replication, thereby highlighting the strategic potential for control and therapy of aMPV/C infection.
Collapse
Affiliation(s)
- Dedong Wang
- College of Veterimary Medicine, Yangzhou University, Yangzhou 225009, China; (D.W.); (L.H.); (N.Z.); (X.Y.); (J.Z.); (Y.C.); (J.G.); (X.F.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Lei Hou
- College of Veterimary Medicine, Yangzhou University, Yangzhou 225009, China; (D.W.); (L.H.); (N.Z.); (X.Y.); (J.Z.); (Y.C.); (J.G.); (X.F.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Ning Zhu
- College of Veterimary Medicine, Yangzhou University, Yangzhou 225009, China; (D.W.); (L.H.); (N.Z.); (X.Y.); (J.Z.); (Y.C.); (J.G.); (X.F.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Xiaoyu Yang
- College of Veterimary Medicine, Yangzhou University, Yangzhou 225009, China; (D.W.); (L.H.); (N.Z.); (X.Y.); (J.Z.); (Y.C.); (J.G.); (X.F.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Jianwei Zhou
- College of Veterimary Medicine, Yangzhou University, Yangzhou 225009, China; (D.W.); (L.H.); (N.Z.); (X.Y.); (J.Z.); (Y.C.); (J.G.); (X.F.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Yongqiu Cui
- College of Veterimary Medicine, Yangzhou University, Yangzhou 225009, China; (D.W.); (L.H.); (N.Z.); (X.Y.); (J.Z.); (Y.C.); (J.G.); (X.F.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Jinshuo Guo
- College of Veterimary Medicine, Yangzhou University, Yangzhou 225009, China; (D.W.); (L.H.); (N.Z.); (X.Y.); (J.Z.); (Y.C.); (J.G.); (X.F.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Xufei Feng
- College of Veterimary Medicine, Yangzhou University, Yangzhou 225009, China; (D.W.); (L.H.); (N.Z.); (X.Y.); (J.Z.); (Y.C.); (J.G.); (X.F.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Jue Liu
- College of Veterimary Medicine, Yangzhou University, Yangzhou 225009, China; (D.W.); (L.H.); (N.Z.); (X.Y.); (J.Z.); (Y.C.); (J.G.); (X.F.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
- Correspondence:
| |
Collapse
|
20
|
Manna S, McAuley J, Jacobson J, Nguyen CD, Ullah MA, Sebina I, Williamson V, Mulholland EK, Wijburg O, Phipps S, Satzke C. Synergism and Antagonism of Bacterial-Viral Coinfection in the Upper Respiratory Tract. mSphere 2022; 7:e0098421. [PMID: 35044807 PMCID: PMC8769199 DOI: 10.1128/msphere.00984-21] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 12/28/2021] [Indexed: 01/03/2023] Open
Abstract
Streptococcus pneumoniae (the pneumococcus) is a leading cause of pneumonia in children under 5 years of age. Coinfection by pneumococci and respiratory viruses enhances disease severity. Little is known about pneumococcal coinfections with respiratory syncytial virus (RSV). Here, we developed a novel infant mouse model of coinfection using pneumonia virus of mice (PVM), a murine analogue of RSV, to examine the dynamics of coinfection in the upper respiratory tract, an anatomical niche that is essential for host-to-host transmission and progression to disease. Coinfection increased damage to the nasal tissue and increased production of the chemokine CCL3. Nasopharyngeal pneumococcal density and shedding in nasal secretions were increased by coinfection. In contrast, coinfection reduced PVM loads in the nasopharynx, an effect that was independent of pneumococcal strain and the order of infection. We showed that this "antagonistic" effect was absent using either ethanol-killed pneumococci or a pneumococcal mutant deficient in capsule production and incapable of nasopharyngeal carriage. Colonization with a pneumococcal strain naturally unable to produce capsule also reduced viral loads. The pneumococcus-mediated reduction in PVM loads was caused by accelerated viral clearance from the nasopharynx. Although these synergistic and antagonistic effects occurred with both wild-type pneumococcal strains used in this study, the magnitude of the effects was strain dependent. Lastly, we showed that pneumococci can also antagonize influenza virus. Taken together, our study has uncovered multiple novel facets of bacterial-viral coinfection. Our findings have important public health implications, including for bacterial and viral vaccination strategies in young children. IMPORTANCE Respiratory bacterial-viral coinfections (such as pneumococci and influenza virus) are often synergistic, resulting in enhanced disease severity. Although colonization of the nasopharynx is the precursor to disease and transmission, little is known about bacterial-viral interactions that occur within this niche. In this study, we developed a novel mouse model to examine pneumococcal-viral interactions in the nasopharynx with pneumonia virus of mice (PVM) and influenza. We found that PVM infection benefits pneumococci by increasing their numbers in the nasopharynx and shedding of these bacteria in respiratory secretions. In contrast, we discovered that pneumococci decrease PVM numbers by accelerating viral clearance. We also report a similar effect of pneumococci on influenza. By showing that coinfections lead to both synergistic and antagonistic outcomes, our findings challenge the existing dogma in the field. Our work has important applications and implications for bacterial and viral vaccines that target these microbes.
Collapse
Affiliation(s)
- Sam Manna
- Infection and Immunity, Murdoch Children’s Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Julie McAuley
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Jonathan Jacobson
- Infection and Immunity, Murdoch Children’s Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Cattram D. Nguyen
- Infection and Immunity, Murdoch Children’s Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| | - Md. Ashik Ullah
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Ismail Sebina
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Victoria Williamson
- Infection and Immunity, Murdoch Children’s Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - E. Kim Mulholland
- Infection and Immunity, Murdoch Children’s Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Odilia Wijburg
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Simon Phipps
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Catherine Satzke
- Infection and Immunity, Murdoch Children’s Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
21
|
Interactions between the Nucleoprotein and the Phosphoprotein of Pneumoviruses: Structural Insight for Rational Design of Antivirals. Viruses 2021; 13:v13122449. [PMID: 34960719 PMCID: PMC8706346 DOI: 10.3390/v13122449] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/30/2021] [Accepted: 12/01/2021] [Indexed: 11/17/2022] Open
Abstract
Pneumoviruses include pathogenic human and animal viruses, the most known and studied being the human respiratory syncytial virus (hRSV) and the metapneumovirus (hMPV), which are the major cause of severe acute respiratory tract illness in young children worldwide, and main pathogens infecting elderly and immune-compromised people. The transcription and replication of these viruses take place in specific cytoplasmic inclusions called inclusion bodies (IBs). These activities depend on viral polymerase L, associated with its cofactor phosphoprotein P, for the recognition of the viral RNA genome encapsidated by the nucleoprotein N, forming the nucleocapsid (NC). The polymerase activities rely on diverse transient protein-protein interactions orchestrated by P playing the hub role. Among these interactions, P interacts with the NC to recruit L to the genome. The P protein also plays the role of chaperone to maintain the neosynthesized N monomeric and RNA-free (called N0) before specific encapsidation of the viral genome and antigenome. This review aims at giving an overview of recent structural information obtained for hRSV and hMPV P, N, and more specifically for P-NC and N0-P complexes that pave the way for the rational design of new antivirals against those viruses.
Collapse
|
22
|
Kaboudi K, Lachheb J. Avian metapneumovirus infection in turkeys: a review on turkey rhinotracheitis. J APPL POULTRY RES 2021. [DOI: 10.1016/j.japr.2021.100211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
23
|
Rüger N, Sid H, Meens J, Szostak MP, Baumgärtner W, Bexter F, Rautenschlein S. New Insights into the Host-Pathogen Interaction of Mycoplasma gallisepticum and Avian Metapneumovirus in Tracheal Organ Cultures of Chicken. Microorganisms 2021; 9:microorganisms9112407. [PMID: 34835532 PMCID: PMC8618481 DOI: 10.3390/microorganisms9112407] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/10/2021] [Accepted: 11/16/2021] [Indexed: 01/04/2023] Open
Abstract
Respiratory pathogens are a health threat for poultry. Co-infections lead to the exacerbation of clinical symptoms and lesions. Mycoplasma gallisepticum (M. gallispeticum) and Avian Metapneumovirus (AMPV) are two avian respiratory pathogens that co-circulate worldwide. The knowledge about the host-pathogen interaction of M. gallispeticum and AMPV in the chicken respiratory tract is limited. We aimed to investigate how co-infections affect the pathogenesis of the respiratory disease and whether the order of invading pathogens leads to changes in host-pathogen interaction. We used chicken tracheal organ cultures (TOC) to investigate pathogen invasion and replication, lesion development, and selected innate immune responses, such as interferon (IFN) α, inducible nitric oxide synthase (iNOS) and IFNλ mRNA expression levels. We performed mono-inoculations (AMPV or M. gallispeticum) or dual-inoculations in two orders with a 24-h interval between the first and second pathogen. Dual-inoculations compared to mono-inoculations resulted in more severe host reactions. Pre-infection with AMPV followed by M. gallispeticum resulted in prolonged viral replication, more significant innate immune responses, and lesions (p < 0.05). AMPV as the secondary pathogen impaired the bacterial attachment process. Consequently, the M. gallispeticum replication was delayed, the innate immune response was less pronounced, and lesions appeared later. Our results suggest a competing process in co-infections and offer new insights in disease processes.
Collapse
Affiliation(s)
- Nancy Rüger
- Clinic for Poultry, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; (N.R.); (F.B.)
| | - Hicham Sid
- Reproductive Biotechnology, TUM School of Life Sciences Weihenstephan, Technical University of Munich, 85354 Munich, Germany;
| | - Jochen Meens
- Institute for Microbiology, Centre for Infection Medicine, University of Veterinary Medicine Hannover, 30559 Hannover, Germany;
| | - Michael P. Szostak
- Institute of Microbiology, Department of Pathobiology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria;
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine Hannover, 30559 Hannover, Germany;
| | - Frederik Bexter
- Clinic for Poultry, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; (N.R.); (F.B.)
| | - Silke Rautenschlein
- Clinic for Poultry, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; (N.R.); (F.B.)
- Correspondence: ; Tel.: +49-511-953-8779
| |
Collapse
|
24
|
Avian Metapneumovirus Subgroup C Induces Mitochondrial Antiviral Signaling Protein Degradation through the Ubiquitin-Proteasome Pathway. Viruses 2021; 13:v13101990. [PMID: 34696420 PMCID: PMC8537000 DOI: 10.3390/v13101990] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 09/30/2021] [Accepted: 10/01/2021] [Indexed: 12/12/2022] Open
Abstract
The mitochondrial antiviral signaling (MAVS) protein, a critical adapter, links the upstream recognition of viral RNA to downstream antiviral signal transduction. However, the interaction mechanism between avian metapneumovirus subgroup C (aMPV/C) infection and MAVS remains unclear. Here, we confirmed that aMPV/C infection induced a reduction in MAVS expression in Vero cells in a dose-dependent manner, and active aMPV/C replication was required for MAVS decrease. We also found that the reduction in MAVS occurred at the post-translational level rather than at the transcriptional level. Different inhibitors were used to examine the effect of proteasome or autophagy on the regulation of MAVS. Treatment with a proteasome inhibitor MG132 effectively blocked MAVS degradation. Moreover, we demonstrated that MAVS mainly underwent K48-linked ubiquitination in the presence of MG132 in aMPV/C-infected cells, with amino acids 363, 462, and 501 of MAVS being pivotal sites in the formation of polyubiquitin chains. Finally, E3 ubiquitin ligases for MAVS degradation were screened and identified and RNF5 targeting MAVS at Lysine 363 and 462 was shown to involve in MAVS degradation in aMPV/C-infected Vero cells. Overall, these results reveal the molecular mechanism underlying aMPV/C infection-induced MAVS degradation by the ubiquitin-proteasome pathway.
Collapse
|
25
|
Saied AA, Metwally AA, Mohamed HMA, Haridy MAM. The contribution of bovines to human health against viral infections. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:46999-47023. [PMID: 34272669 PMCID: PMC8284698 DOI: 10.1007/s11356-021-14941-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 06/12/2021] [Indexed: 04/12/2023]
Abstract
In the last 40 years, novel viruses have evolved at a much faster pace than other pathogens. Viral diseases pose a significant threat to public health around the world. Bovines have a longstanding history of significant contributions to human nutrition, agricultural, industrial purposes, medical research, drug and vaccine development, and livelihood. The life cycle, genomic structures, viral proteins, and pathophysiology of bovine viruses studied in vitro paved the way for understanding the human counterparts. Calf model has been used for testing vaccines against RSV, papillomavirus vaccines and anti-HCV agents were principally developed after using the BPV and BVDV model, respectively. Some bovine viruses-based vaccines (BPIV-3 and bovine rotaviruses) were successfully developed, clinically tried, and commercially produced. Cows, immunized with HIV envelope glycoprotein, produced effective broadly neutralizing antibodies in their serum and colostrum against HIV. Here, we have summarized a few examples of human viral infections for which the use of bovines has contributed to the acquisition of new knowledge to improve human health against viral infections covering the convergence between some human and bovine viruses and using bovines as disease models. Additionally, the production of vaccines and drugs, bovine-based products were covered, and the precautions in dealing with bovines and bovine-based materials.
Collapse
Affiliation(s)
- AbdulRahman A Saied
- Department of Food Establishments Licensing (Aswan Branch), National Food Safety Authority (NFSA), Aswan, 81511, Egypt.
- Touristic Activities and Interior Offices Sector (Aswan Office), Ministry of Tourism and Antiquities, Aswan, 81511, Egypt.
| | - Asmaa A Metwally
- Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Aswan University, Aswan, 81511, Egypt
| | - Hams M A Mohamed
- Department of Microbiology, Faculty of Veterinary Medicine, South Valley University, Qena, 83523, Egypt
| | - Mohie A M Haridy
- Department of Pathology and Clinical Pathology, Faculty of Veterinary Medicine, South Valley University, Qena, 83523, Egypt.
| |
Collapse
|
26
|
Soto JA, Gálvez NMS, Pacheco GA, Canedo-Marroquín G, Bueno SM, Kalergis AM. Induction of Protective Immunity by a Single Low Dose of a Master Cell Bank cGMP-rBCG-P Vaccine Against the Human Metapneumovirus in Mice. Front Cell Infect Microbiol 2021; 11:662714. [PMID: 34268134 PMCID: PMC8276701 DOI: 10.3389/fcimb.2021.662714] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 05/24/2021] [Indexed: 12/29/2022] Open
Abstract
Human metapneumovirus (hMPV) is an emergent virus, which mainly infects the upper and lower respiratory tract epithelium. This pathogen is responsible for a significant portion of hospitalizations due to bronchitis and pneumonia in infants and the elderly worldwide. hMPV infection induces a pro-inflammatory immune response upon infection of the host, which is not adequate for the clearance of this pathogen. The lack of knowledge regarding the different molecular mechanisms of infection of this virus has delayed the licensing of effective treatments or vaccines. As part of this work, we evaluated whether a single and low dose of a recombinant Mycobacterium bovis Bacillus Calmette-Guérin (BCG) expressing the phosphoprotein of hMPV (rBCG-P) can induce a protective immune response in mice. Immunization with the rBCG-P significantly decreased neutrophil counts and viral loads in the lungs of infected mice at different time points. This immune response was also associated with a modulated infiltration of innate cells into the lungs, such as interstitial macrophages (IM) and alveolar macrophages (AM), activated CD4+ and CD8+ T cells, and changes in the population of differentiated subsets of B cells, such as marginal zone B cells and plasma cells. The humoral immune response induced by the rBCG-P led to an early and robust IgA response and a late and constant IgG response. Finally, we determined that the transfer of cells or sera from immunized and infected mice to naïve mice promoted an efficient viral clearance. Therefore, a single and low dose of rBCG-P can protect mice from the disease caused by hMPV, and this vaccine could be a promising candidate for future clinical trials.
Collapse
Affiliation(s)
- Jorge A. Soto
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nicolás M. S. Gálvez
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Gaspar A. Pacheco
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Gisela Canedo-Marroquín
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M. Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
27
|
Rhinovirus Reduces the Severity of Subsequent Respiratory Viral Infections by Interferon-Dependent and -Independent Mechanisms. mSphere 2021; 6:e0047921. [PMID: 34160242 PMCID: PMC8265665 DOI: 10.1128/msphere.00479-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Coinfection by heterologous viruses in the respiratory tract is common and can alter disease severity compared to infection by individual virus strains. We previously found that inoculation of mice with rhinovirus (RV) 2 days before inoculation with a lethal dose of influenza A virus [A/Puerto Rico/8/34 (H1N1) (PR8)] provides complete protection against mortality. Here, we extended that finding to a second lethal respiratory virus, pneumonia virus of mice (PVM), and analyzed potential mechanisms of RV-induced protection. RV completely prevented mortality and weight loss associated with PVM infection. Major changes in host gene expression upon PVM infection were delayed compared to PR8. RV induced earlier recruitment of inflammatory cells, which were reduced at later times in RV-inoculated mice. Findings common to both virus pairs included the upregulated expression of mucin-associated genes and dampening of inflammation-related genes in mice that were inoculated with RV before lethal virus infection. However, type I interferon (IFN) signaling was required for RV-mediated protection against PR8 but not PVM. IFN signaling had minor effects on PR8 replication and contributed to controlling neutrophilic inflammation and hemorrhagic lung pathology in RV/PR8-infected mice. These findings, combined with differences in virus replication levels and disease severity, suggest that the suppression of inflammation in RV/PVM-infected mice may be due to early, IFN-independent suppression of viral replication, while that in RV/PR8-infected mice may be due to IFN-dependent modulation of immune responses. Thus, a mild upper respiratory viral infection can reduce the severity of a subsequent severe viral infection in the lungs through virus-dependent mechanisms. IMPORTANCE Respiratory viruses from diverse families cocirculate in human populations and are frequently detected within the same host. Although clinical studies suggest that infection by multiple different respiratory viruses may alter disease severity, animal models in which we can control the doses, timing, and strains of coinfecting viruses are critical to understanding how coinfection affects disease severity. Here, we compared gene expression and immune cell recruitment between two pairs of viruses (RV/PR8 and RV/PVM) inoculated sequentially in mice, both of which result in reduced severity compared to lethal infection by PR8 or PVM alone. Reduced disease severity was associated with suppression of inflammatory responses in the lungs. However, differences in disease kinetics and host and viral gene expression suggest that protection by coinfection with RV may be due to distinct molecular mechanisms. Indeed, we found that antiviral cytokine signaling was required for RV-mediated protection against lethal infection by PR8 but not PVM.
Collapse
|
28
|
Rodriguez PE, Frutos MC, Adamo MP, Cuffini C, Cámara JA, Paglini MG, Moreno L, Cámara A. Human Metapneumovirus: Epidemiology and genotype diversity in children and adult patients with respiratory infection in Córdoba, Argentina. PLoS One 2020; 15:e0244093. [PMID: 33370354 PMCID: PMC7769284 DOI: 10.1371/journal.pone.0244093] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 12/02/2020] [Indexed: 11/19/2022] Open
Abstract
Human Metapneumovirus (hMPV) is responsible for acute respiratory infections in humans, with clinical and epidemiological relevance in pediatric, elderly, and immunocompromised populations. These features are largely unknown in Córdoba, Argentina and in adults in general. Hence, our goal was to broadly characterize hMPV infection in patients of all ages hospitalized with acute respiratory infections in Córdoba, Argentina, including epidemiology, clinical features and genetic diversity. Nasopharyngeal secretions were obtained from 795 patients during 2011-2013, 621 patients were 0-25 years old and 174 were 26-85 years old. HMPV was assayed by RT-PCR and other respiratory viruses by indirect immunofluorescence. Local strains were identified by sequence analysis. Human Metapneumovirus was detected in 20.3% (161/795) patients, 13.1% as single infections and 7.2% in co-infections, more frequently with Respiratory Syncytial Virus. HMPV circulated during late winter and spring in all age patients, but mainly in children under 4 years old in 71.4% (115/161) and adults between 26 and 59 years old in 12.4% (20/161). The most prevalent diagnosis was mild acute respiratory infection in 59.6% (96/161) and bronchiolitis in 9.3% (15/161). Local strains were clustered within A2 subtype; they presented 73-100% identities among them, showing a high degree of homology compared to isolations from neighboring countries. We demonstrate that hMPV circulated among all age patients with respiratory infection during 2011-2013 in Córdoba, contributing to the understanding of this virus, its diagnosis and patient handling in local health-care centers.
Collapse
Affiliation(s)
- Pamela Elizabeth Rodriguez
- Instituto de Virología “Dr. J. M. Vanella”, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba Capital, Córdoba, Argentina
- * E-mail:
| | - María Celia Frutos
- Instituto de Virología “Dr. J. M. Vanella”, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba Capital, Córdoba, Argentina
| | - María Pilar Adamo
- Instituto de Virología “Dr. J. M. Vanella”, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba Capital, Córdoba, Argentina
| | - Cecilia Cuffini
- Instituto de Virología “Dr. J. M. Vanella”, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba Capital, Córdoba, Argentina
| | - Jorge Augusto Cámara
- Instituto de Virología “Dr. J. M. Vanella”, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba Capital, Córdoba, Argentina
| | - María Gabriela Paglini
- Instituto de Virología “Dr. J. M. Vanella”, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba Capital, Córdoba, Argentina
- Instituto de Investigaciones Médicas Mercedes y Martín Ferreyra, INIMEC- CONICET, Universidad Nacional de Córdoba, Córdoba Capital, Córdoba, Argentina
| | - Laura Moreno
- Cátedra de Clínica Pediátrica, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Hospital de Niños “Santísima Trinidad de Córdoba”, Córdoba Capital, Córdoba, Argentina
| | - Alicia Cámara
- Instituto de Virología “Dr. J. M. Vanella”, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba Capital, Córdoba, Argentina
| |
Collapse
|
29
|
Tegegne D, Deneke Y, Sori T, Abdurahaman M, Kebede N, Cecchinato M, Franzo G. Molecular Epidemiology and Genotyping of Infectious Bronchitis Virus and Avian Metapneumovirus in Backyard and Commercial Chickens in Jimma Zone, Southwestern Ethiopia. Vet Sci 2020; 7:vetsci7040187. [PMID: 33255570 PMCID: PMC7711717 DOI: 10.3390/vetsci7040187] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/18/2020] [Accepted: 11/21/2020] [Indexed: 01/26/2023] Open
Abstract
Poultry production plays a relevant role in the Ethiopian economy and represents a source of poverty alleviation for several social classes. Infectious diseases can therefore significantly impact the economy and welfare. Despite infectious bronchitis virus (IBV) and avian metapneumovirus (aMPV) being present, the knowledge of their epidemiology and impact is extremely limited. In the present work, a cross-sectional study based on 500 tracheal swabs collected from 50 intensive and backyard unvaccinated flocks of the Jimma Zone was performed to investigate the circulation of these viruses and molecularly characterize them. IBV and aMPV presence was tested by molecular assays, and genotyping was carried out on positive samples. Accordingly, 6% (95% CI 2.06% to 16.22%) and 8% (95% CI 3.15% to 18.84%) of flocks tested IBV and aMPV positive, respectively. Particularly, IBV 793B (GI-13) strains were detected in backyard flocks only, and identical or closely related sequences (p-distance <2%) were detected in distantly spaced flocks, suggesting relevant viral circulation. On the contrary, both backyard and intensive flocks were affected by aMPV subtype B. Potential epidemiological links associated to the importation of parental birds from foreign countries could be established. These results highlight non-negligible circulation of these viruses, warranting further epidemiological studies and the evaluation of control measure implementation.
Collapse
Affiliation(s)
- Dechassa Tegegne
- School of Veterinary Medicine, Jimma University College of Agriculture and Veterinary Medicine, P.O. Box 307 Jimma, Ethiopia; (D.T.); (Y.D.); (T.S.); (M.A.)
| | - Yosef Deneke
- School of Veterinary Medicine, Jimma University College of Agriculture and Veterinary Medicine, P.O. Box 307 Jimma, Ethiopia; (D.T.); (Y.D.); (T.S.); (M.A.)
| | - Takele Sori
- School of Veterinary Medicine, Jimma University College of Agriculture and Veterinary Medicine, P.O. Box 307 Jimma, Ethiopia; (D.T.); (Y.D.); (T.S.); (M.A.)
| | - Mukarim Abdurahaman
- School of Veterinary Medicine, Jimma University College of Agriculture and Veterinary Medicine, P.O. Box 307 Jimma, Ethiopia; (D.T.); (Y.D.); (T.S.); (M.A.)
| | - Nigatu Kebede
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, P.O. Box 1176 Addis Ababa, Ethiopia;
| | - Mattia Cecchinato
- Department of Animal Medicine, Production and Health (MAPS), University of Padua, 35020 Legnaro (PD), Italy;
| | - Giovanni Franzo
- Department of Animal Medicine, Production and Health (MAPS), University of Padua, 35020 Legnaro (PD), Italy;
- Correspondence:
| |
Collapse
|
30
|
Features of the Course of Metapneumoviral Infection in Adults. Fam Med 2019. [DOI: 10.30841/2307-5112.5-6.2019.193437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
31
|
Innate immune protection from pneumonia virus of mice induced by a novel immunomodulator is prolonged by dual treatment and mediated by macrophages. Antiviral Res 2019; 171:104594. [PMID: 31470041 DOI: 10.1016/j.antiviral.2019.104594] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 08/20/2019] [Accepted: 08/26/2019] [Indexed: 12/20/2022]
Abstract
Respiratory syncytial virus (RSV) is responsible for a large proportion of acute lower respiratory tract infections, specifically in children. Pneumonia virus of mice (PVM) causes similar lung pathology and clinical disease in rodents, and is therefore an appropriate model of RSV infection. Previously, we demonstrated that a single intranasal dose of P-I-P, a novel immunomodulator composed of the toll-like receptor 3 agonist poly(I:C), an innate defense regulator peptide and a polyphosphazene, confers protection in Balb/c mice for up to 3 days from lethal PVM-15 infection. In the present study a dual intranasal treatment with P-I-P was shown to extend the duration of the protection conferred by P-I-P from PVM-15 challenge. Balb/c mice treated twice with P-I-P showed higher survival rates and milder clinical signs when compared to animals that received a single P-I-P dose. While the mice treated with two consecutive doses of P-I-P experienced some weight loss, they all recovered. The dual P-I-P treatment mediated infiltration of several innate immune cells into the BALF and lung, including alveolar macrophages, neutrophils, and γδ T cells. Partial depletion of alveolar macrophages decreased survival rates and exacerbated clinical signs of mice subjected to the P-I-P dual treatment regime followed by PVM-15 challenge. This suggests that the alveolar macrophage is at least partially responsible for the protection elicited by this novel prophylactic treatment strategy.
Collapse
|
32
|
Altamirano-Lagos MJ, Díaz FE, Mansilla MA, Rivera-Pérez D, Soto D, McGill JL, Vasquez AE, Kalergis AM. Current Animal Models for Understanding the Pathology Caused by the Respiratory Syncytial Virus. Front Microbiol 2019; 10:873. [PMID: 31130923 PMCID: PMC6510261 DOI: 10.3389/fmicb.2019.00873] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 04/04/2019] [Indexed: 12/14/2022] Open
Abstract
The human respiratory syncytial virus (hRSV) is the main etiologic agent of severe lower respiratory tract infections that affect young children throughout the world, associated with significant morbidity and mortality, becoming a serious public health problem globally. Up to date, no licensed vaccines are available to prevent severe hRSV-induced disease, and the generation of safe-effective vaccines has been a challenging task, requiring constant biomedical research aimed to overcome this ailment. Among the difficulties presented by the study of this pathogen, it arises the fact that there is no single animal model that resembles all aspects of the human pathology, which is due to the specificity that this pathogen has for the human host. Thus, for the study of hRSV, different animal models might be employed, depending on the goal of the study. Of all the existing models, the murine model has been the most frequent model of choice for biomedical studies worldwide and has been of great importance at contributing to the development and understanding of vaccines and therapies against hRSV. The most notable use of the murine model is that it is very useful as a first approach in the development of vaccines or therapies such as monoclonal antibodies, suggesting in this way the direction that research could have in other preclinical models that have higher maintenance costs and more complex requirements in its management. However, several additional different models for studying hRSV, such as other rodents, mustelids, ruminants, and non-human primates, have been explored, offering advantages over the murine model. In this review, we discuss the various applications of animal models to the study of hRSV-induced disease and the advantages and disadvantages of each model, highlighting the potential of each model to elucidate different features of the pathology caused by the hRSV infection.
Collapse
Affiliation(s)
- María José Altamirano-Lagos
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Fabián E. Díaz
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Miguel Andrés Mansilla
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Daniela Rivera-Pérez
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Daniel Soto
- Sección Biotecnología, Instituto de Salud Pública de Chile, Santiago, Chile
| | - Jodi L. McGill
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, United States
| | - Abel E. Vasquez
- Sección Biotecnología, Instituto de Salud Pública de Chile, Santiago, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia, Santiago, Chile
| | - Alexis M. Kalergis
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
33
|
Kennedy DA, Read AF. Why the evolution of vaccine resistance is less of a concern than the evolution of drug resistance. Proc Natl Acad Sci U S A 2018; 115:12878-12886. [PMID: 30559199 PMCID: PMC6304978 DOI: 10.1073/pnas.1717159115] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Vaccines and antimicrobial drugs both impose strong selection for resistance. Yet only drug resistance is a major challenge for 21st century medicine. Why is drug resistance ubiquitous and not vaccine resistance? Part of the answer is that vaccine resistance is far less likely to evolve than drug resistance. But what happens when vaccine resistance does evolve? We review six putative cases. We find that in contrast to drug resistance, vaccine resistance is harder to detect and harder to confirm and that the mechanistic basis is less well understood. Nevertheless, in the cases we examined, the pronounced health benefits associated with vaccination have largely been sustained. Thus, we contend that vaccine resistance is less of a concern than drug resistance because it is less likely to evolve and when it does, it is less harmful to human and animal health and well-being. Studies of pathogen strains that evolve the capacity to replicate and transmit from vaccinated hosts will enhance our ability to develop next-generation vaccines that minimize the risk of harmful pathogen evolution.
Collapse
Affiliation(s)
- David A Kennedy
- Center for Infectious Disease Dynamics, Departments of Biology and Entomology, The Pennsylvania State University, University Park, PA 16802
| | - Andrew F Read
- Center for Infectious Disease Dynamics, Departments of Biology and Entomology, The Pennsylvania State University, University Park, PA 16802
| |
Collapse
|
34
|
Bayraktar E, Umar S, Yilmaz A, Turan N, Franzo G, Tucciarone CM, Cecchinato M, Cakan B, Iqbal M, Yilmaz H. First Molecular Characterization of Avian Metapneumovirus (aMPV) in Turkish Broiler Flocks. Avian Dis 2018; 62:425-430. [PMID: 31119927 DOI: 10.1637/11915-061818-resnote.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Indexed: 11/05/2022]
Abstract
Viral respiratory diseases, including avian metapneumovirus (aMPV), have a significant economic impact on poultry industries. The frequency and genotype diversity of aMPV in Turkish broiler flocks is not known at present. The aim of this study was to report the first molecular identification and phylogeny of aMPV, which is circulating in Turkish broiler flocks. Trachea tissue samples and tracheal swabs were collected from 110 broiler flocks distributed in different geographical regions in Turkey between March 2017 and March 2018. Detection of aMPV was confirmed with the use of universal reverse transcriptase (RT) PCR, and eight (7.2%) broiler farms were positive for aMPV. Sequence analysis of the G gene revealed the exclusive presence of subtype B viruses. Three field isolates clustered closely with a 2002 Israel isolate, indicating a potential transmission route between these two countries and through the Middle East. The remaining five field isolates were closely related to a vaccine strain, even though broiler flocks in Turkey are not routinely vaccinated against aMPV. Therefore, we speculate these five isolates could have originated from nearby vaccinated turkey farms. Additionally, the presence of some nucleotide substitutions compared to the reference vaccine sequence suggests prolonged circulation and evolution of the original vaccine virus or a vaccine subpopulation was selected under field conditions. This evidence emphasizes the need for further detailed and more systemic approaches to evaluate aMPV spread and evolution in order to design effective control strategies.
Collapse
Affiliation(s)
- E Bayraktar
- CEVA Animal Health, Poultry Section, Istanbul, Turkey
| | - S Umar
- Veterinary Faculty, Department of Virology, University of Istanbul-Cerrahpas_a, Avcilar, 34320, Istanbul, Turkey
| | - A Yilmaz
- Veterinary Faculty, Department of Virology, University of Istanbul-Cerrahpas_a, Avcilar, 34320, Istanbul, Turkey
| | - N Turan
- Veterinary Faculty, Department of Virology, University of Istanbul-Cerrahpas_a, Avcilar, 34320, Istanbul, Turkey
| | - G Franzo
- Department of Animal Medicine, Production and Health (MAPS), University of Padua, Viale dell'Università 16, 35020 Legnaro (PD), Italy
| | - C M Tucciarone
- Department of Animal Medicine, Production and Health (MAPS), University of Padua, Viale dell'Università 16, 35020 Legnaro (PD), Italy
| | - M Cecchinato
- Department of Animal Medicine, Production and Health (MAPS), University of Padua, Viale dell'Università 16, 35020 Legnaro (PD), Italy
| | - B Cakan
- CEVA Animal Health, Poultry Section, Istanbul, Turkey
| | - M Iqbal
- The Pirbright Institute, United Kingdom
| | - H Yilmaz
- Veterinary Faculty, Department of Virology, University of Istanbul-Cerrahpas_a, Avcilar, 34320, Istanbul, Turkey,
| |
Collapse
|
35
|
Uche IK, Guerrero-Plata A. Interferon-Mediated Response to Human Metapneumovirus Infection. Viruses 2018; 10:v10090505. [PMID: 30231515 PMCID: PMC6163993 DOI: 10.3390/v10090505] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 09/12/2018] [Accepted: 09/15/2018] [Indexed: 12/13/2022] Open
Abstract
Human metapneumovirus (HMPV) is one of the leading causes of respiratory diseases in infants and children worldwide. Although this pathogen infects mainly young children, elderly and immunocompromised people can be also seriously affected. To date, there is no commercial vaccine available against it. Upon HMPV infection, the host innate arm of defense produces interferons (IFNs), which are critical for limiting HMPV replication. In this review, we offer an updated landscape of the HMPV mediated-IFN response in different models as well as some of the defense tactics employed by the virus to circumvent IFN response.
Collapse
Affiliation(s)
- Ifeanyi K Uche
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA.
| | - Antonieta Guerrero-Plata
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA.
- Center for Experimental Infectious Disease Research, Louisiana State University, Baton Rouge, LA 70803, USA.
| |
Collapse
|
36
|
Santos da Silva GN, Monti Atik D, Antunes Fernandes JL, de Freitas do Nascimento D, Fazolo T, Duarte de Souza AP, Baggio Gnoatto SC. Synthesis of three triterpene series and their activity against respiratory syncytial virus. Arch Pharm (Weinheim) 2018; 351:e1800108. [PMID: 29999539 DOI: 10.1002/ardp.201800108] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 06/23/2018] [Accepted: 06/29/2018] [Indexed: 01/03/2023]
Abstract
The human respiratory syncytial virus (hRSV) is a leading cause of hospitalization due to acute lower respiratory infection especially in infants and young children, sometimes causing fatal cases. The monoclonal antibody palivizumab is one of the available options for preventing this virus, and at the moment there are several hRSV vaccine trials underway. Unfortunately, the only drug option to treat hRSV infection is ribavirin, which can be used in severe high-risk cases. For this reason, new medicines are needed and, in this context, the triterpenes and their derivatives are promising alternatives, since many of them have shown important antiviral activity, such as bevirimat. Therefore, we report three series of triterpene (betulin (BE), betulinic acid (BA), and ursolic acid (UA)) derivatives tested against hRSV. The derivatives were synthesized by using commercial anhydrides in an easy and inexpensive step reaction. For the antiviral assay, A549 cells were infected by hRSV and after 96 h of compound or ribavirin (positive control) treatment, the cell viability was tested by MTT assay. DMSO, non-infected cells and infected cells without treatment were used as negative control. The triterpene esterification at the hydroxyl group resulted in 17 derivatives. The 3,28-di-O-acetylbetulin derivative (1a) showed the best results for cell viability, and real-time PCR amplification was performed for 1a treatment. Remarkably, one new anti-hRSV prototype was obtained through an easy synthesis of BE, which shall represent an alternative for a new lead compound for anti-hRSV therapy.
Collapse
Affiliation(s)
- Gloria N Santos da Silva
- Phytochemistry and Organic Synthesis Laboratory, School of Pharmacy, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Diana Monti Atik
- Clinical and Immunology Laboratory, Biomedical Research Institute, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Jheini L Antunes Fernandes
- Clinical and Immunology Laboratory, Biomedical Research Institute, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Deise de Freitas do Nascimento
- Clinical and Immunology Laboratory, Biomedical Research Institute, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Tiago Fazolo
- Clinical and Immunology Laboratory, Biomedical Research Institute, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Ana Paula Duarte de Souza
- Clinical and Immunology Laboratory, Biomedical Research Institute, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Simone C Baggio Gnoatto
- Phytochemistry and Organic Synthesis Laboratory, School of Pharmacy, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
37
|
Abstract
Human metapneumovirus (HMPV) is a leading cause of acute respiratory infection, particularly in children, immunocompromised patients, and the elderly. HMPV, which is closely related to avian metapneumovirus subtype C, has circulated for at least 65 years, and nearly every child will be infected with HMPV by the age of 5. However, immunity is incomplete, and re-infections occur throughout adult life. Symptoms are similar to those of other respiratory viral infections, ranging from mild (cough, rhinorrhea, and fever) to more severe (bronchiolitis and pneumonia). The preferred method for diagnosis is reverse transcription-polymerase chain reaction as HMPV is difficult to culture. Although there have been many advances made in the past 16 years since its discovery, there are still no US Food and Drug Administration-approved antivirals or vaccines available to treat HMPV. Both small animal and non-human primate models have been established for the study of HMPV. This review will focus on the epidemiology, transmission, and clinical manifestations in humans as well as the animal models of HMPV pathogenesis and host immune response.
Collapse
Affiliation(s)
- Nazly Shafagati
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - John Williams
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
38
|
Ribaudo M, Barik S. The nonstructural proteins of Pneumoviruses are remarkably distinct in substrate diversity and specificity. Virol J 2017; 14:215. [PMID: 29110727 PMCID: PMC5674761 DOI: 10.1186/s12985-017-0881-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 10/27/2017] [Indexed: 11/23/2022] Open
Abstract
Background Interferon (IFN) inhibits viruses by inducing several hundred cellular genes, aptly named ‘interferon (IFN)-stimulated genes’ (ISGs). The only two RNA viruses of the Pneumovirus genus of the Paramyxoviridae family, namely Respiratory Syncytial Virus (RSV) and Pneumonia Virus of Mice (PVM), each encode two nonstructural (NS) proteins that share no sequence similarity but yet suppress IFN. Since suppression of IFN underlies the ability of these viruses to replicate in the host cells, the mechanism of such suppression has become an important area of research. This Short Report is an important extension of our previous efforts in defining this mechanism. Results We show that, like their PVM counterparts, the RSV NS proteins also target multiple members of the ISG family. While significantly extending the substrate repertoire of the RSV NS proteins, these results, unexpectedly, also reveal that the target preferences of the NS proteins of the two viruses are entirely different. This is surprising since the two Pneumoviruses are phylogenetically close with similar genome organization and gene function, and the NS proteins of both also serve as suppressors of host IFN response. Conclusion The finding that the NS proteins of the two highly similar viruses suppress entirely different members of the ISG family raises intriguing questions of pneumoviral NS evolution and mechanism of action.
Collapse
Affiliation(s)
- Michael Ribaudo
- Department of Biological, Geological and Environmental Sciences, and Center for Gene Regulation in Health and Disease, Cleveland State University, 2121 Euclid Avenue, Cleveland, OH, 44115, USA
| | - Sailen Barik
- Department of Biological, Geological and Environmental Sciences, and Center for Gene Regulation in Health and Disease, Cleveland State University, 2121 Euclid Avenue, Cleveland, OH, 44115, USA. .,3780 Pelham Drive, Mobile, AL, 36619, USA.
| |
Collapse
|
39
|
Hou L, Wei L, Zhu S, Wang J, Quan R, Li Z, Liu J. Avian metapneumovirus subgroup C induces autophagy through the ATF6 UPR pathway. Autophagy 2017; 13:1709-1721. [PMID: 28949785 PMCID: PMC5640183 DOI: 10.1080/15548627.2017.1356950] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 06/16/2017] [Accepted: 07/12/2017] [Indexed: 12/12/2022] Open
Abstract
An increasing number of studies have demonstrated that macroautophagy/autophagy plays an important role in the infectious processes of diverse pathogens. However, it remains unknown whether autophagy is induced in avian metapneumovirus (aMPV)-infected host cells, and, if so, how this occurs. Here, we report that aMPV subgroup C (aMPV/C) induces autophagy in cultured cells. We demonstrated this relationship by detecting classical autophagic features, including the formation of autophagsomes, the presence of GFP-LC3 puncta and the conversation of LC3-I into LC3-II. Also, we used pharmacological regulators and siRNAs targeting ATG7 or LC3 to examine the role of autophagy in aMPV/C replication. The results showed that autophagy is required for efficient replication of aMPV/C. Moreover, infection with aMPV/C promotes autophagosome maturation and induces a complete autophagic process. Finally, the ATF6 pathway, of which one component is the unfolded protein response (UPR), becomes activated in aMPV/C-infected cells. Knockdown of ATF6 inhibited aMPV/C-induced autophagy and viral replication. Collectively, these results not only show that autophagy promotes aMPV/C replication in the cultured cells, but also reveal that the molecular mechanisms underlying aMPV/C-induced autophagy depends on regulation of the ER stress-related UPR pathway.
Collapse
Affiliation(s)
- Lei Hou
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Li Wei
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Shanshan Zhu
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Jing Wang
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Rong Quan
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Zixuan Li
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Jue Liu
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| |
Collapse
|
40
|
Epitope mapping and kinetics of CD4 T cell immunity to pneumonia virus of mice in the C57BL/6 strain. Sci Rep 2017; 7:3472. [PMID: 28615708 PMCID: PMC5471230 DOI: 10.1038/s41598-017-03042-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 04/19/2017] [Indexed: 11/08/2022] Open
Abstract
Pneumonia virus of mice (PVM) infection has been widely used as a rodent model to study the closely related human respiratory syncytial virus (hRSV). While T cells are indispensable for viral clearance, they also contribute to immunopathology. To gain more insight into mechanistic details, novel tools are needed that allow to study virus-specific T cells in C57BL/6 mice as the majority of transgenic mice are only available on this background. While PVM-specific CD8 T cell epitopes were recently described, so far no PVM-specific CD4 T cell epitopes have been identified within the C57BL/6 strain. Therefore, we set out to map H2-IAb-restricted epitopes along the PVM proteome. By means of in silico prediction and subsequent functional validation, we were able to identify a MHCII-restricted CD4 T cell epitope, corresponding to amino acids 37–47 in the PVM matrix protein (M37–47). Using this newly identified MHCII-restricted M37–47 epitope and a previously described MHCI-restricted N339–347 epitope, we generated peptide-loaded MHCII and MHCI tetramers and characterized the dynamics of virus-specific CD4 and CD8 T cell responses in vivo. The findings of this study can provide a basis for detailed investigation of T cell-mediated immune responses to PVM in a variety of genetically modified C57BL/6 mice.
Collapse
|
41
|
Zhao Y, Jamaluddin M, Zhang Y, Sun H, Ivanciuc T, Garofalo RP, Brasier AR. Systematic Analysis of Cell-Type Differences in the Epithelial Secretome Reveals Insights into the Pathogenesis of Respiratory Syncytial Virus-Induced Lower Respiratory Tract Infections. THE JOURNAL OF IMMUNOLOGY 2017; 198:3345-3364. [PMID: 28258195 DOI: 10.4049/jimmunol.1601291] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 01/23/2017] [Indexed: 11/19/2022]
Abstract
Lower respiratory tract infections from respiratory syncytial virus (RSV) are due, in part, to secreted signals from lower airway cells that modify the immune response and trigger airway remodeling. To understand this process, we applied an unbiased quantitative proteomics analysis of the RSV-induced epithelial secretory response in cells representative of the trachea versus small airway bronchiolar cells. A workflow was established using telomerase-immortalized human epithelial cells that revealed highly reproducible cell type-specific differences in secreted proteins and nanoparticles (exosomes). Approximately one third of secretome proteins are exosomal; the remainder are from lysosomal and vacuolar compartments. We applied this workflow to three independently derived primary human cultures from trachea versus bronchioles. A total of 577 differentially expressed proteins from control supernatants and 966 differentially expressed proteins from RSV-infected cell supernatants were identified at a 1% false discovery rate. Fifteen proteins unique to RSV-infected primary human cultures from trachea were regulated by epithelial-specific ets homologous factor. A total of 106 proteins unique to RSV-infected human small airway epithelial cells was regulated by the transcription factor NF-κB. In this latter group, we validated the differential expression of CCL20/macrophage-inducible protein 3α, thymic stromal lymphopoietin, and CCL3-like 1 because of their roles in Th2 polarization. CCL20/macrophage-inducible protein 3α was the most active mucin-inducing factor in the RSV-infected human small airway epithelial cell secretome and was differentially expressed in smaller airways in a mouse model of RSV infection. These studies provide insights into the complexity of innate responses and regional differences in the epithelial secretome participating in RSV lower respiratory tract infection-induced airway remodeling.
Collapse
Affiliation(s)
- Yingxin Zhao
- Institute for Translational Sciences, University of Texas Medical Branch, Galveston, TX 77555.,Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555.,Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, TX 77555; and
| | - Mohammad Jamaluddin
- Institute for Translational Sciences, University of Texas Medical Branch, Galveston, TX 77555.,Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555
| | - Yueqing Zhang
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555
| | - Hong Sun
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555
| | - Teodora Ivanciuc
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX 77555
| | - Roberto P Garofalo
- Institute for Translational Sciences, University of Texas Medical Branch, Galveston, TX 77555.,Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, TX 77555; and.,Department of Pediatrics, University of Texas Medical Branch, Galveston, TX 77555
| | - Allan R Brasier
- Institute for Translational Sciences, University of Texas Medical Branch, Galveston, TX 77555; .,Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555.,Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, TX 77555; and
| |
Collapse
|
42
|
A Reverse Genetics Approach for the Design of Methyltransferase-Defective Live Attenuated Avian Metapneumovirus Vaccines. Methods Mol Biol 2016. [PMID: 27076293 DOI: 10.1007/978-1-4939-3389-1_7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Avian metapneumovirus (aMPV), also known as avian pneumovirus or turkey rhinotracheitis virus, is the causative agent of turkey rhinotracheitis and is associated with swollen head syndrome in chickens. aMPV belongs to the family Paramyxoviridae which includes many important human pathogens such as human respiratory syncytial virus (RSV), human metapneumovirus (hMPV), and human parainfluenza virus type 3 (PIV3). The family also includes highly lethal emerging pathogens such as Nipah virus and Hendra virus, as well as agriculturally important viruses such as Newcastle disease virus (NDV). For many of these viruses, there is no effective vaccine. Here, we describe a reverse genetics approach to develop live attenuated aMPV vaccines by inhibiting the viral mRNA cap methyltransferase. The viral mRNA cap methyltransferase is an excellent target for the attenuation of paramyxoviruses because it plays essential roles in mRNA stability, efficient viral protein translation and innate immunity. We have described in detail the materials and methods used to generate recombinant aMPVs that lack viral mRNA cap methyltransferase activity. We have also provided methods to evaluate the genetic stability, pathogenesis, and immunogenicity of live aMPV vaccine candidates in turkeys.
Collapse
|
43
|
Dhar J, Barik S. Unique nonstructural proteins of Pneumonia Virus of Mice (PVM) promote degradation of interferon (IFN) pathway components and IFN-stimulated gene proteins. Sci Rep 2016; 6:38139. [PMID: 27905537 PMCID: PMC5131486 DOI: 10.1038/srep38139] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 11/04/2016] [Indexed: 12/25/2022] Open
Abstract
Pneumonia Virus of Mice (PVM) is the only virus that shares the Pneumovirus genus of the Paramyxoviridae family with Respiratory Syncytial Virus (RSV). A deadly mouse pathogen, PVM has the potential to serve as a robust animal model of RSV infection, since human RSV does not fully replicate the human pathology in mice. Like RSV, PVM also encodes two nonstructural proteins that have been implicated to suppress the IFN pathway, but surprisingly, they exhibit no sequence similarity with their RSV equivalents. The molecular mechanism of PVM NS function, therefore, remains unknown. Here, we show that recombinant PVM NS proteins degrade the mouse counterparts of the IFN pathway components. Proteasomal degradation appears to be mediated by ubiquitination promoted by PVM NS proteins. Interestingly, NS proteins of PVM lowered the levels of several ISG (IFN-stimulated gene) proteins as well. These results provide a molecular foundation for the mechanisms by which PVM efficiently subverts the IFN response of the murine cell. They also reveal that in spite of their high sequence dissimilarity, the two pneumoviral NS proteins are functionally and mechanistically similar.
Collapse
Affiliation(s)
- Jayeeta Dhar
- Department of Biological, Geological and Environmental Sciences, and Centre for Gene Regulation in Health and Disease, Cleveland State University, 2121 Euclid Avenue, Cleveland, Ohio 44115, USA
| | - Sailen Barik
- Department of Biological, Geological and Environmental Sciences, and Centre for Gene Regulation in Health and Disease, Cleveland State University, 2121 Euclid Avenue, Cleveland, Ohio 44115, USA
| |
Collapse
|
44
|
Martinez EC, Garg R, Shrivastava P, Gomis S, van Drunen Littel-van den Hurk S. Intranasal treatment with a novel immunomodulator mediates innate immune protection against lethal pneumonia virus of mice. Antiviral Res 2016; 135:108-119. [PMID: 27771388 PMCID: PMC7126411 DOI: 10.1016/j.antiviral.2016.10.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 10/07/2016] [Accepted: 10/18/2016] [Indexed: 12/26/2022]
Abstract
Respiratory syncytial virus (RSV) is the leading cause of acute lower respiratory tract infections in infants and young children. There are no licensed RSV vaccines available, and the few treatment options for high-risk individuals are either extremely costly or cause severe side effects and toxicity. Immunomodulation mediated by a novel formulation consisting of the toll-like receptor 3 agonist poly(I:C), an innate defense regulator peptide and a polyphosphazene (P-I-P) was evaluated in the context of lethal infection with pneumonia virus of mice (PVM). Intranasal delivery of a single dose of P-I-P protected adult mice against PVM when given 24 h prior to challenge. These animals experienced minimal weight loss, no clinical disease, 100% survival, and reduced lung pathology. Similar clinical outcomes were observed in mice treated up to 3 days prior to infection. P-I-P pre-treatment induced early mRNA and protein expression of key chemokine and cytokine genes, reduced the recruitment of neutrophils and eosinophils, decreased virus titers in the lungs, and modulated the delayed exacerbated nature of PVM disease without any short-term side effects. On day 14 post-infection, P-I-P-treated mice were confirmed to be PVM-free. These results demonstrate the capacity of this formulation to prevent PVM and possibly other viral respiratory infections. P-I-P pre-treatment, consisting of poly(I:C), IDR peptide and PCEP, was tested in the context of PVM infection in mice. P-I-P confers complete protection against lethal PVM infection by reducing clinical signs and immunopathology. P-I-P minimizes viral titers in the lungs reduces the influx of neutrophils and eosinophils into the tissue. P-I-P induces early upregulation of genes involved in host defense without any observable adverse effects. Survivor mice were PVM negative, suggesting that P-I-P mediates the successfully clearance of the virus in vivo.
Collapse
Affiliation(s)
- Elisa C Martinez
- Department of Microbiology and Immunology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, 107 Wiggins Road, S7N 5E5, Canada; Vaccine and Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, Saskatoon, Saskatchewan, 120 Veterinary Road, S7N 5E3, Canada
| | - Ravendra Garg
- Vaccine and Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, Saskatoon, Saskatchewan, 120 Veterinary Road, S7N 5E3, Canada
| | - Pratima Shrivastava
- Vaccine and Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, Saskatoon, Saskatchewan, 120 Veterinary Road, S7N 5E3, Canada
| | - Susantha Gomis
- Department of Veterinary Pathology, Western College of Veterinary Medicine (WCVM), University of Saskatchewan, Saskatoon, Saskatchewan, 52 Campus Drive, S7N 5B4, Canada
| | - Sylvia van Drunen Littel-van den Hurk
- Department of Microbiology and Immunology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, 107 Wiggins Road, S7N 5E5, Canada; Vaccine and Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, Saskatoon, Saskatchewan, 120 Veterinary Road, S7N 5E3, Canada.
| |
Collapse
|
45
|
Paudel S, Easwaran M, Jang H, Jung HK, Kim JH, Shin HJ. Immunization with avian metapneumovirus harboring chicken Fc induces higher immune responses. Virus Res 2016; 220:129-35. [PMID: 27130629 DOI: 10.1016/j.virusres.2016.04.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 04/12/2016] [Accepted: 04/18/2016] [Indexed: 10/21/2022]
Abstract
In this study, we evaluated the immune responses of avian metapneumovirus harboring chicken Fc molecule. Stable Vero cells expressing chicken Fc chimera on its surface (Vero-cFc) were established, and we confirmed that aMPV grown in Vero-cFc incorporated host derived chimera Fc into the aMPV virions. Immunization of chicken with aMPV-cFc induced higher level of antibodies and inflammatory cytokines; (Interferon (IFN)-γ and Interleukin (IL)-1β) compared to those of aMPV. The increased levels of antibodies and inflammatory cytokines in chicken immunized with aMPV-cFc were statistically significantly (p<0.05) to that of aMPV and control. The aMPV-cFc group also generated the highest neutralizing antibody response. After challenges, chickens immunized with aMPV-cFc showed much less pathological signs in nasal turbinates and trachea so that we could confirm aMPV-cFc induced higher protection than that of aMPV. The greater ability of aMPV harboring chicken Fc to that of aMPV presented it as a possible vaccine candidate.
Collapse
Affiliation(s)
- Sarita Paudel
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Maheswaran Easwaran
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Hyun Jang
- Komipharm Co., Ltd., Republic of Korea
| | | | - Joo-Hun Kim
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea; Research Institute of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea; Komipharm Co., Ltd., Republic of Korea
| | - Hyun-Jin Shin
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea; Research Institute of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea.
| |
Collapse
|
46
|
Immune Response to Human Metapneumovirus Infection: What We Have Learned from the Mouse Model. Pathogens 2015; 4:682-96. [PMID: 26393657 PMCID: PMC4584281 DOI: 10.3390/pathogens4030682] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 09/09/2015] [Accepted: 09/15/2015] [Indexed: 01/17/2023] Open
Abstract
Human Metapneumovirus (hMPV) is a leading respiratory viral pathogen associated with bronchiolitis, pneumonia, and asthma exacerbation in young children, the elderly and immunocompromised individuals. The development of a potential vaccine against hMPV requires detailed understanding of the host immune system, which plays a significant role in hMPV pathogenesis, susceptibility and vaccine efficacy. As a result, animal models have been developed to better understand the mechanisms by which hMPV causes disease. Several animal models have been evaluated and established so far to study the host immune responses and pathophysiology of hMPV infection. However, inbred laboratory mouse strains have been one of the most used animal species for experimental modeling and therefore used for the studies of immunity and immunopathogenesis to hMPV. This review summarizes the contributions of the mouse model to our understanding of the immune response against hMPV infection.
Collapse
|
47
|
Paudel S, Shin HJ. Role of trypsin in the replication of Avian metapneumovirus subtype C (strain MN-2a) and its entry into the Vero cells. Mol Cell Probes 2015; 29:485-491. [PMID: 26028611 DOI: 10.1016/j.mcp.2015.05.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 05/12/2015] [Accepted: 05/27/2015] [Indexed: 01/12/2023]
Abstract
To understand the molecular mechanisms of Avian metapneumovirus (aMPV) and the requirements involved in the infection and fusion, trypsin treatment was done in the different stages of virus; before infection, during entry and after virus infection followed by aMPV infection. The growth kinetics of aMPV was compared in time dependent manner. The effect of trypsin was found in the later stage of aMPV infection increasing the numbers of infected cells with the significant higher titer of infectious virions to that of trypsin treated before infection, during entry and aMPV. A serine protease inhibitor reduced aMPV replication in a significant way, whereas cysteine peptidase (E-64), aspartic protease (pepstatin A), and metalloprotease (phosphoramidon) inhibitors had no effect on aMPV replication. Inoculation of aMPV on Vero cells expressing the membrane-associated protease TMPRSS2 resulted in higher virus titers than that inoculated on normal Vero cells and is statistically significant (p < 0.05). Also, an inhibitor of clathrin/caveolae-mediated endocytosis had no effect on virus progeny, indicating that aMPV does not use the endocytic pathway for entry but undergoes direct fusion. The effect of lysosomotropic agents was not significant, suggesting that aMPV does not require low-pH environment in endosomes to fuse its envelope with the plasma membrane.
Collapse
Affiliation(s)
- Sarita Paudel
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Chungnam National University, Daejeon 305-764, Republic of Korea
| | - Hyun-Jin Shin
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Chungnam National University, Daejeon 305-764, Republic of Korea; Research Institute of Veterinary Medicine, Chungnam National University, Daejeon 305-764, Republic of Korea.
| |
Collapse
|
48
|
Phylogenetic analysis of human metapneumovirus detected in hospitalized patients in Kuwait during the years 2009-2011. J Infect Public Health 2015; 8:448-57. [PMID: 25773766 DOI: 10.1016/j.jiph.2015.01.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2014] [Revised: 12/30/2014] [Accepted: 01/23/2015] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Human metapneumovirus (hMPV) is an important cause of both upper and lower respiratory tract infections (RTIs) in all age groups. Children, elderly, and immunocompromised individuals are the most affected groups. HMPV infection accounts for 5% of hospitalized patients with respiratory tract infections in Kuwait. It is mostly detected among infants and elderly age groups, and both hMPV genotypes A and B circulate in Kuwait. METHODS In this study, the genetic diversity of detected hMPV was evaluated, and a phylogenetic analysis based on partial nucleotide and amino acid sequences of the G gene was performed for hMPV detected among hospitalized patients with RTIs. RESULTS Our results showed that 62% of hMPV sequences belonged to the A genotype and 38% to the B genotype. A2b and B2 subtypes were detected and circulated during the study period, whereas A1 and B1 subtypes were not detected. Based on nucleotide sequences of the G gene, most of hMPV strains (57%) were clustered with Indian strains, followed by Greek strains (24%) and Canadian strains (14%). One strain (5%) clustered within the B genotype but had different branches than B1 and B2 branches. CONCLUSION Our data showed the co-circulation of hMPV genotypes A2b and B2 in Kuwait with genetic diversity suggestive of evolution through negative selection.
Collapse
|
49
|
Madbouly HM, Tamam SM, Hussein AS, Mady W, Arafa AS. Immunomodular effect of fusion gene DNA vaccine of avian metapneumoviruses. J APPL POULTRY RES 2014. [DOI: 10.3382/japr.2014-00963] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
50
|
Methyltransferase-defective avian metapneumovirus vaccines provide complete protection against challenge with the homologous Colorado strain and the heterologous Minnesota strain. J Virol 2014; 88:12348-63. [PMID: 25122790 DOI: 10.1128/jvi.01095-14] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
UNLABELLED Avian metapneumovirus (aMPV), also known as avian pneumovirus or turkey rhinotracheitis virus, is the causative agent of turkey rhinotracheitis and is associated with swollen head syndrome in chickens. Since its discovery in the 1970s, aMPV has been recognized as an economically important pathogen in the poultry industry worldwide. The conserved region VI (CR VI) of the large (L) polymerase proteins of paramyxoviruses catalyzes methyltransferase (MTase) activities that typically methylate viral mRNAs at guanine N-7 (G-N-7) and ribose 2'-O positions. In this study, we generated a panel of recombinant aMPV (raMPV) Colorado strains carrying mutations in the S-adenosyl methionine (SAM) binding site in the CR VI of L protein. These recombinant viruses were specifically defective in ribose 2'-O, but not G-N-7 methylation and were genetically stable and highly attenuated in cell culture and viral replication in the upper and lower respiratory tracts of specific-pathogen-free (SPF) young turkeys. Importantly, turkeys vaccinated with these MTase-defective raMPVs triggered a high level of neutralizing antibody and were completely protected from challenge with homologous aMPV Colorado strain and heterologous aMPV Minnesota strain. Collectively, our results indicate (i) that aMPV lacking 2'-O methylation is highly attenuated in vitro and in vivo and (ii) that inhibition of mRNA cap MTase can serve as a novel target to rationally design live attenuated vaccines for aMPV and perhaps other paramyxoviruses. IMPORTANCE Paramyxoviruses include many economically and agriculturally important viruses such as avian metapneumovirus (aMPV), and Newcastle disease virus (NDV), human pathogens such as human respiratory syncytial virus, human metapneumovirus, human parainfluenza virus type 3, and measles virus, and highly lethal emerging pathogens such as Nipah virus and Hendra virus. For many of them, there is no effective vaccine or antiviral drug. These viruses share common strategies for viral gene expression and replication. During transcription, paramyxoviruses produce capped, methylated, and polyadenylated mRNAs. Using aMPV as a model, we found that viral ribose 2'-O methyltransferase (MTase) is a novel approach to rationally attenuate the virus for vaccine purpose. Recombinant aMPV (raMPV) lacking 2'-O MTase were not only highly attenuated in turkeys but also provided complete protection against the challenge of homologous and heterologous aMPV strains. This novel approach can be applicable to other animal and human paramyxoviruses for rationally designing live attenuated vaccines.
Collapse
|