1
|
Rafique S, Rashid F, Wei Y, Zeng T, Xie L, Xie Z. Avian Orthoreoviruses: A Systematic Review of Their Distribution, Dissemination Patterns, and Genotypic Clustering. Viruses 2024; 16:1056. [PMID: 39066218 PMCID: PMC11281703 DOI: 10.3390/v16071056] [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: 03/21/2024] [Revised: 06/14/2024] [Accepted: 06/18/2024] [Indexed: 07/28/2024] Open
Abstract
Avian orthoreviruses have become a global challenge to the poultry industry, causing significant economic impacts on commercial poultry. Avian reoviruses (ARVs) are resistant to heat, proteolytic enzymes, a wide range of pH values, and disinfectants, so keeping chicken farms free of ARV infections is difficult. This review focuses on the global prevalence of ARVs and associated clinical signs and symptoms. The most common signs and symptoms include tenosynovitis/arthritis, malabsorption syndrome, runting-stunting syndrome, and respiratory diseases. Moreover, this review also focused on the characterization of ARVs in genotypic clusters (I-VI) and their relation to tissue tropism or viral distribution. The prevailing strains of ARV in Africa belong to all genotypic clusters (GCs) except for GC VI, whereas all GCs are present in Asia and the Americas. In addition, all ARV strains are associated with or belong to GC I-VI in Europe. Moreover, in Oceania, only GC V and VI are prevalent. This review also showed that, regardless of the genotypic cluster, tenosynovitis/arthritis was the predominant clinical manifestation, indicating its universal occurrence across all clusters. Globally, most avian reovirus infections can be prevented by vaccination against four major strains: S1133, 1733, 2408, and 2177. Nevertheless, these vaccines may not a provide sufficient defense against field isolates. Due to the increase in the number of ARV variants, classical vaccine approaches are being developed depending on the degree of antigenic similarity between the vaccine and field strains, which determines how successful the vaccination will be. Moreover, there is a need to look more closely at the antigenic and pathogenic properties of reported ARV strains. The information acquired will aid in the selection of more effective vaccine strains in combination with biosecurity and farm management methods to prevent ARV infections.
Collapse
Affiliation(s)
- Saba Rafique
- SB Diagnostic Laboratory, Sadiq Poultry Pvt. Ltd., Rawalpindi 46000, Pakistan;
| | - Farooq Rashid
- Department of Biotechnology, Guangxi Veterinary Research Institute, Nanning 530001, China; (F.R.); (Y.W.); (T.Z.); (L.X.)
- Guangxi Key Laboratory of Veterinary Biotechnology, Nanning 530001, China
- Key Laboratory of China (Guangxi)-ASEAN Cross-Border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning 530001, China
| | - You Wei
- Department of Biotechnology, Guangxi Veterinary Research Institute, Nanning 530001, China; (F.R.); (Y.W.); (T.Z.); (L.X.)
- Guangxi Key Laboratory of Veterinary Biotechnology, Nanning 530001, China
- Key Laboratory of China (Guangxi)-ASEAN Cross-Border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning 530001, China
| | - Tingting Zeng
- Department of Biotechnology, Guangxi Veterinary Research Institute, Nanning 530001, China; (F.R.); (Y.W.); (T.Z.); (L.X.)
- Guangxi Key Laboratory of Veterinary Biotechnology, Nanning 530001, China
- Key Laboratory of China (Guangxi)-ASEAN Cross-Border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning 530001, China
| | - Liji Xie
- Department of Biotechnology, Guangxi Veterinary Research Institute, Nanning 530001, China; (F.R.); (Y.W.); (T.Z.); (L.X.)
- Guangxi Key Laboratory of Veterinary Biotechnology, Nanning 530001, China
- Key Laboratory of China (Guangxi)-ASEAN Cross-Border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning 530001, China
| | - Zhixun Xie
- Department of Biotechnology, Guangxi Veterinary Research Institute, Nanning 530001, China; (F.R.); (Y.W.); (T.Z.); (L.X.)
- Guangxi Key Laboratory of Veterinary Biotechnology, Nanning 530001, China
- Key Laboratory of China (Guangxi)-ASEAN Cross-Border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning 530001, China
| |
Collapse
|
2
|
de Matos TRA, Palka APG, de Souza C, Fragoso SP, Pavoni DP. Detection of avian reovirus (ARV) by ELISA based on recombinant σB, σC and σNS full-length proteins and protein fragments. J Med Microbiol 2024; 73. [PMID: 38935078 DOI: 10.1099/jmm.0.001836] [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] [Indexed: 06/28/2024] Open
Abstract
Introduction. Avian reovirus (ARV) is associated with arthritis/tenosynovitis and malabsorption syndrome in chickens. The σC and σB proteins, both exposed to the virus capsid, are highly immunogenic and could form the basis for diagnostic devices designed to assess the immunological status of the flock.Gap Statement. Commercial ARV ELISAs cannot distinguish between vaccinated and infected animals and might not detect circulating ARV strains.Aim. We aimed to develop a customized test to detect the circulating field ARV strains as well as distinguish between vaccinated and unvaccinated animals.Methodology. We developed ELISA assays based on recombinant (r) σB, σC and the nonstructural protein σNS and tested them using antisera of vaccinated and unvaccinated chickens as well as negative controls. Fragments of σB and σC proteins were also used to study regions that could be further exploited in diagnostic tests.Results. Vaccinated and unvaccinated birds were positive by commercial ELISA, with no difference in optical density values. In contrast, samples of unvaccinated animals showed lower absorbance in the rσB and rσC ELISA tests and higher absorbance in the rσNS ELISA test than the vaccinated animals. Negative control samples were negative in all tests. Fragmentation of σB and σC proteins showed that some regions can differentiate between vaccinated and unvaccinated animals. For example, σB amino acids 128-179 (σB-F4) and σC amino acids 121-165 (σC-F4) exhibited 85 and 95% positivity among samples of vaccinated animals but only 5% and zero positivity among samples of unvaccinated animals, respectively.Conclusion. These data suggest that unvaccinated birds might have been exposed to field strains of ARV. The reduction in absorbance in the recombinant tests possibly reflects an increased specificity of our test since unvaccinated samples showed less cross-reactivity with the vaccine proteins immobilized on ELISAs. The discrepant results obtained with the protein fragment tests between vaccinated and unvaccinated animals are discussed in light of the diversity between ARV strains.
Collapse
Affiliation(s)
- Tatiana Reichert Assunção de Matos
- Fundação Oswaldo Cruz, Instituto Carlos Chagas, Fiocruz Paraná, Curitiba/PR, Brazil
- Programa de Pós-graduação em Biociências e Biotecnologia, Fundação Oswaldo Cruz, Instituto Carlos Chagas, Fiocruz Paraná, Curitiba/PR, Brazil
| | - Ana Paula Gori Palka
- Fundação Oswaldo Cruz, Instituto Carlos Chagas, Fiocruz Paraná, Curitiba/PR, Brazil
- Programa de Pós-graduação em Biologia Celular e Molecular, Universidade Federal do Paraná/UFPR, Curitiba/PR, Brazil
- Instituto de Tecnologia do Paraná/Tecpar, Curitiba/PR, Brazil
| | - Claudemir de Souza
- Fundação Oswaldo Cruz, Instituto Carlos Chagas, Fiocruz Paraná, Curitiba/PR, Brazil
- Programa de Pós-graduação em Biologia Celular e Molecular, Universidade Federal do Paraná/UFPR, Curitiba/PR, Brazil
| | - Stenio Perdigão Fragoso
- Fundação Oswaldo Cruz, Instituto Carlos Chagas, Fiocruz Paraná, Curitiba/PR, Brazil
- Programa de Pós-graduação em Biociências e Biotecnologia, Fundação Oswaldo Cruz, Instituto Carlos Chagas, Fiocruz Paraná, Curitiba/PR, Brazil
| | - Daniela Parada Pavoni
- Fundação Oswaldo Cruz, Instituto Carlos Chagas, Fiocruz Paraná, Curitiba/PR, Brazil
- Programa de Pós-graduação em Biociências e Biotecnologia, Fundação Oswaldo Cruz, Instituto Carlos Chagas, Fiocruz Paraná, Curitiba/PR, Brazil
- Programa de Pós-graduação em Biologia Celular e Molecular, Universidade Federal do Paraná/UFPR, Curitiba/PR, Brazil
| |
Collapse
|
3
|
Franzo G, Tucciarone CM, Faustini G, Poletto F, Baston R, Cecchinato M, Legnardi M. Reconstruction of Avian Reovirus History and Dispersal Patterns: A Phylodynamic Study. Viruses 2024; 16:796. [PMID: 38793677 PMCID: PMC11125613 DOI: 10.3390/v16050796] [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: 03/27/2024] [Revised: 05/11/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
Avian reovirus (ARV) infection can cause significant losses to the poultry industry. Disease control has traditionally been attempted mainly through vaccination. However, the increase in clinical outbreaks in the last decades demonstrated the poor effectiveness of current vaccination approaches. The present study reconstructs the evolution and molecular epidemiology of different ARV genotypes using a phylodynamic approach, benefiting from a collection of more than one thousand sigma C (σC) sequences sampled over time at a worldwide level. ARVs' origin was estimated to occur several centuries ago, largely predating the first clinical reports. The origins of all genotypes were inferred at least one century ago, and their emergence and rise reflect the intensification of the poultry industry. The introduction of vaccinations had only limited and transitory effects on viral circulation and further expansion was observed, particularly after the 1990s, likely because of the limited immunity and the suboptimal and patchy vaccination application. In parallel, strong selective pressures acted with different strengths and directionalities among genotypes, leading to the emergence of new variants. While preventing the spread of new variants with different phenotypic features would be pivotal, a phylogeographic analysis revealed an intricate network of viral migrations occurring even over long distances and reflecting well-established socio-economic relationships.
Collapse
Affiliation(s)
- Giovanni Franzo
- Department of Animal Medicine, Production and Health (MAPS), University of Padua, 35020 Legnaro, Italy; (C.M.T.); (G.F.); (F.P.); (R.B.); (M.C.); (M.L.)
| | | | | | | | | | | | | |
Collapse
|
4
|
Zhang X, Chen G, Liu R, Guo J, Mei K, Qin L, Li Z, Yuan S, Huang S, Wen F. Identification, pathological, and genomic characterization of novel goose reovirus associated with liver necrosis in geese, China. Poult Sci 2024; 103:103269. [PMID: 38064883 PMCID: PMC10749903 DOI: 10.1016/j.psj.2023.103269] [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: 09/18/2023] [Revised: 10/31/2023] [Accepted: 11/10/2023] [Indexed: 12/29/2023] Open
Abstract
Since 2021, a novel strain of goose reovirus (GRV) has emerged within the goose farming industry in Guangdong province, China. This particular viral variant is distinguished by the presence of white necrotic foci primarily localized in the liver and spleen, leading to substantial economic losses for the poultry industry. However, the etiology, prevalence and genomic characteristics of the causative agent have not been thoroughly investigated. In this study, we conducted an epidemiological inquiry employing suspected GRV samples collected from May 2021 to September 2022. The macroscopic pathological and histopathological lesions associated with GRV-infected clinical specimens were examined. Moreover, we successfully isolated the GRV strain and elucidated the complete genome sequence of the isolate GD21/88. Through phylogenetic and recombination analysis, we unveiled that the GRV strains represent a novel variant resulting from multiple reassortment events. Specifically, the μNS, λC, and σNS genes of GRV were found to have originated from chicken reovirus, while the σA gene of GRV exhibited a higher degree of similarity with a novel duck reovirus. The remaining genes of GRV were traced back to Muscovy duck reovirus. Collectively, our findings underscore the significance of GRV as a pathogenic agent impacting the goose farming industry. The insights gleaned from this study contribute to a more comprehensive understanding of the epidemiology of GRV in Southern China and shed light on the genetic reassortment events exhibited by the virus.
Collapse
Affiliation(s)
- Xinyu Zhang
- College of Life Science and Engineering, Foshan University, Foshan 528231, Guangdong, China
| | - Gaojie Chen
- College of Life Science and Engineering, Foshan University, Foshan 528231, Guangdong, China
| | - Runzhi Liu
- College of Life Science and Engineering, Foshan University, Foshan 528231, Guangdong, China
| | - Jinyue Guo
- College of Life Science and Engineering, Foshan University, Foshan 528231, Guangdong, China
| | - Kun Mei
- College of Life Science and Engineering, Foshan University, Foshan 528231, Guangdong, China
| | - Limei Qin
- College of Life Science and Engineering, Foshan University, Foshan 528231, Guangdong, China
| | - Zhili Li
- College of Life Science and Engineering, Foshan University, Foshan 528231, Guangdong, China
| | - Sheng Yuan
- College of Life Science and Engineering, Foshan University, Foshan 528231, Guangdong, China
| | - Shujian Huang
- College of Life Science and Engineering, Foshan University, Foshan 528231, Guangdong, China; Guangdong Huasheng Biotechnology Co., Ltd,Guangzhou 511300, Guangdong, China
| | - Feng Wen
- College of Life Science and Engineering, Foshan University, Foshan 528231, Guangdong, China; Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, College of Life Science and Engineering, Foshan University, Foshan 528231, Guangdong, China.
| |
Collapse
|
5
|
Nour I, Alvarez-Narvaez S, Harrell TL, Conrad SJ, Mohanty SK. Whole Genomic Constellation of Avian Reovirus Strains Isolated from Broilers with Arthritis in North Carolina, USA. Viruses 2023; 15:2191. [PMID: 38005869 PMCID: PMC10675200 DOI: 10.3390/v15112191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 10/26/2023] [Accepted: 10/28/2023] [Indexed: 11/26/2023] Open
Abstract
Avian reovirus (ARV) is an emerging pathogen which causes significant economic challenges to the chicken and turkey industry in the USA and globally, yet the molecular characterization of most ARV strains is restricted to a single particular gene, the sigma C gene. The genome of arthrogenic reovirus field isolates (R18-37308 and R18-38167), isolated from broiler chickens in North Carolina (NC), USA in 2018, was sequenced using long-read next-generation sequencing (NGS). The isolates were genotyped based on the amino acid sequence of sigma C (σC) followed by phylogenetic and amino acid analyses of the other 11 genomically encoded proteins for whole genomic constellation and genetic variation detection. The genomic length of the NC field strains was 23,494 bp, with 10 dsRNA segments ranging from 3959 bp (L1) to 1192 bp (S4), and the 5' and 3' untranslated regions (UTRs) of all the segments were found to be conserved. R18-37308 and R18-38167 were found to belong to genotype (G) VI based on the σC analysis and showed nucleotide and amino acid sequence identity ranging from 84.91-98.47% and 83.43-98.46%, respectively, with G VI strains. Phylogenetic analyses of individual genes of the NC strains did not define a single common ancestor among the available completely sequenced ARV strains. Nevertheless, most sequences supported the Chinese strain LY383 as a probable ancestor of these isolates. Moreover, amino acid analysis revealed multiple amino acid substitution events along the entirety of the genes, some of which were unique to each strain, which suggests significant divergence owing to the accumulation of point mutations. All genes from R18-37308 and R18-38167 were found to be clustered within genotypic clusters that included only ARVs of chicken origin, which negates the possibility of genetic pooling or host variation. Collectively, this study revealed sequence divergence between the NC field strains and reference ARV strains, including the currently used vaccine strains could help updating the vaccination regime through the inclusion of these highly divergent circulating indigenous field isolates.
Collapse
Affiliation(s)
| | | | | | | | - Sujit K. Mohanty
- United States Department of Agriculture, Agricultural Research Service (USDA-ARS), US National Poultry Research Center, Athens, GA 30605, USA; (I.N.); (S.A.-N.); (T.L.H.); (S.J.C.)
| |
Collapse
|
6
|
Huang WR, Wu YY, Liao TL, Nielsen BL, Liu HJ. Cell Entry of Avian Reovirus Modulated by Cell-Surface Annexin A2 and Adhesion G Protein-Coupled Receptor Latrophilin-2 Triggers Src and p38 MAPK Signaling Enhancing Caveolin-1- and Dynamin 2-Dependent Endocytosis. Microbiol Spectr 2023; 11:e0000923. [PMID: 37097149 PMCID: PMC10269738 DOI: 10.1128/spectrum.00009-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 04/05/2023] [Indexed: 04/26/2023] Open
Abstract
The specifics of cell receptor-modulated avian reovirus (ARV) entry remain unknown. By using a viral overlay protein-binding assay (VOPBA) and an in-gel digestion coupled with liquid chromatography-tandem mass spectrometry (LC-MS/MS), we determined that cell-surface annexin A2 (AnxA2) and adhesion G protein-coupled receptor Latrophilin-2 (ADGRL2) modulate ARV entry. Direct interaction between the ARV σC protein and AnxA2 and ADGRL2 in Vero and DF-1 cells was demonstrated in situ by proximity ligation assays. By using short hairpin RNAs (shRNAs) to silence the endogenous AnxA2 and ADGRL2 genes, ARV entry could be efficiently blocked. A significant decrease in virus yields and the intracellular specific signal for σC protein was observed in Vero cells preincubated with the specific AnxA2 and ADGRL2 monoclonal antibodies, indicating that AnxA2 and ADGRL2 are involved in modulating ARV entry. Furthermore, we found that cells pretreated with the AnxA2/S100A10 heterotetramer (A2t) inhibitor A2ti-1 suppressed ARV-mediated activation of Src and p38 mitogen-activated protein kinase (MAPK), demonstrating that Src and p38 MAPK serve as downstream molecules of cell-surface AnxA2 signaling. Our results reveal that suppression of cell-surface AnxA2 with the A2ti-1 inhibitor increased Csk-Cbp interaction, suggesting that ARV entry suppresses Cbp-mediated relocation of Csk to the membrane, thereby activating Src. Furthermore, reciprocal coimmunoprecipitation assays revealed that σC can interact with signaling molecules, lipid raft, and vimentin. The current study provides novel insights into cell-surface AnxA2- and ADGRL2-modulated cell entry of ARV which triggers Src and p38 MAPK signaling to enhance caveolin-1-, dynamin 2-, and lipid raft-dependent endocytosis. IMPORTANCE By analyzing results from VOPBA and LC-MS/MS, we have determined that cell-surface AnxA2 and ADGRL2 modulate ARV entry. After ARV binding to receptors, Src and p38 MAPK signaling were triggered and, in turn, increased the phosphorylation of caveolin-1 (Tyr14) and upregulated dynamin 2 expression to facilitate caveolin-1-mediated and dynamin 2-dependent endocytosis. In this work, we demonstrated that ARV triggers Src activation by impeding Cbp-mediated relocation of Csk to the membrane in the early stages of the life cycle. This work provides better insight into cell-surface AnxA2 and ADGRL2, which upregulate Src and p38MAPK signaling pathways to enhance ARV entry and productive infection.
Collapse
Affiliation(s)
- Wei-Ru Huang
- Institute of Molecular Biology, National Chung Hsing University, Taichung, Taiwan
- iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| | - Yi-Ying Wu
- Institute of Molecular Biology, National Chung Hsing University, Taichung, Taiwan
- iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| | - Tsai-Ling Liao
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan
- Ph.D Program in Translational Medicine, National Chung Hsing University, Taichung, Taiwan
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Brent L. Nielsen
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, Utah, USA
| | - Hung-Jen Liu
- Institute of Molecular Biology, National Chung Hsing University, Taichung, Taiwan
- iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan
- Ph.D Program in Translational Medicine, National Chung Hsing University, Taichung, Taiwan
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
7
|
Mosad SM, Elmahallawy EK, Alghamdi AM, El-Khayat F, El-Khadragy MF, Ali LA, Abdo W. Molecular and pathological investigation of avian reovirus (ARV) in Egypt with the assessment of the genetic variability of field strains compared to vaccine strains. Front Microbiol 2023; 14:1156251. [PMID: 37138631 PMCID: PMC10150020 DOI: 10.3389/fmicb.2023.1156251] [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] [Received: 02/01/2023] [Accepted: 03/28/2023] [Indexed: 05/05/2023] Open
Abstract
Avian orthoreovirus (ARV) is among the important viruses that cause drastic economic losses in the Egyptian poultry industry. Despite regular vaccination of breeder birds, a high prevalence of ARV infection in broilers has been noted in recent years. However, no reports have revealed the genetic and antigenic characteristics of Egyptian field ARV and vaccines used against it. Thus, this study was conducted to detect the molecular nature of emerging ARV strains in broiler chickens suffering from arthritis and tenosynovitis in comparison to vaccine strains. Synovial fluid samples (n = 400) were collected from 40 commercial broiler flocks in the Gharbia governorate, Egypt, and then pooled to obtain 40 samples, which were then used to screen ARV using reverse transcriptase polymerase chain reaction (RT-PCR) with the partial amplification of ARV sigma C gene. The obtained RT-PCR products were then sequenced, and their nucleotide and deduced amino acid sequences were analyzed together with other ARV field and vaccine strains from GenBank. RT-PCR successfully amplified the predicted 940 bp PCR products from all tested samples. The phylogenetic tree revealed that the analyzed ARV strains were clustered into six genotypic clusters and six protein clusters, with high antigenic diversity between the genotypic clusters. Surprisingly, our isolates were genetically different from vaccine strains, which aligned in genotypic cluster I/protein cluster I, while our strains were aligned in genotypic cluster V/protein cluster V. More importantly, our strains were highly divergent from vaccine strains used in Egypt, with 55.09-56.23% diversity. Sequence analysis using BioEdit software revealed high genetic and protein diversity between our isolates and vaccine strains (397/797 nucleotide substitutions and 148-149/265 amino acid substitutions). This high genetic diversity explains the vaccination failure and recurrent circulation of ARV in Egypt. The present data highlight the need to formulate a new effective vaccine from locally isolated ARV strains after a thorough screening of the molecular nature of circulating ARV in Egypt.
Collapse
Affiliation(s)
- Samah M. Mosad
- Department of Virology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Ehab Kotb Elmahallawy
- Department of Zoonoses, Faculty of Veterinary Medicine, Sohag University, Sohag, Egypt
- *Correspondence: Ehab Kotb Elmahallawy
| | - Abeer M. Alghamdi
- Department of Biology, Faculty of Science, Al-Baha University, Al-Baha, Saudi Arabia
| | - Fares El-Khayat
- Department of Poultry Diseases, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Manal F. El-Khadragy
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Lobna A. Ali
- Cell Biology and Histochemistry, Zoology Department, Faculty of Science, South Valley University, Qena, Egypt
| | - Walied Abdo
- Department of Pathology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, Egypt
- Walied Abdo
| |
Collapse
|
8
|
Sellers HS. Avian Reoviruses from Clinical Cases of Tenosynovitis: An Overview of Diagnostic Approaches and 10-Year Review of Isolations and Genetic Characterization. Avian Dis 2022; 66:420-426. [PMID: 36715473 DOI: 10.1637/aviandiseases-d-22-99990] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 09/02/2022] [Indexed: 01/24/2023]
Abstract
Reoviral-induced tenosynovitis/viral arthritis is an economically significant disease of poultry. Affected birds present with lameness, unilateral or bilateral swollen hock joints or shanks, and/or reluctance to move. In severe cases, rupture of the gastrocnemius or digital flexor tendons may occur, and significant culling may be necessary. Historically, vaccination with a combination of modified live and inactivated vaccines has successfully controlled disease. Proper vaccination reduced vertical transmission and provided maternal-derived antibodies to progeny to protect against disease, at an age when they were most susceptible. Starting in 2011-2012, an increased incidence of tenosynovitis/viral arthritis was observed in chickens and turkeys. In chickens, progeny from reovirus-vaccinated breeders were affected, suggesting commercial vaccines did not provide adequate protection against disease. In turkeys, clinical disease was primarily in males, although females can also be affected. The most significant signs were observed around 14-16 wks of age and include reluctance to move, lameness, and limping on one or both legs. The incidence of tenosynovitis/viral arthritis presently remains high. Reoviruses isolated from clinical cases are genetically and antigenically characterized as variants, meaning they are different from vaccine strains. Characterization of the field isolates reveals multiple new genotypes and serotypes that are significantly different from commercial vaccines and each other. In 2012, a single prevalent virus was isolated from a majority of the cases submitted to the Poultry Diagnostic and Research Center at the University of Georgia. Genetic characterization of the σC protein revealed the early isolates belonged to genetic cluster (GC) 5. Soon after the initial identification of the GC5 variant reovirus, many broiler companies incorporated these isolates from their farms into their autogenous vaccines and continue to do so today. The incidence of GC5 field isolates has decreased significantly, likely because of the widespread use of the isolates in autogenous vaccines. Unfortunately, variant reoviruses belonging to multiple GCs have emerged, despite inclusion of these isolates in autogenous vaccines. In this review, an overview of nomenclature, sample collection, and diagnostic testing will be covered, and a summary of variant reoviruses isolated from clinical cases of tenosynovitis/viral arthritis over the past 10 yrs will be provided.
Collapse
Affiliation(s)
- Holly S Sellers
- Poultry Diagnostic and Research Center, Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, GA 30602,
| |
Collapse
|
9
|
Dawe WH, Kapczynski DR, Linnemann EG, Gauthiersloan VR, Sellers HS. Analysis of the Immune Response and Identification of Antibody Epitopes Against the Sigma C Protein of Avian Orthoreovirus Following Immunization with Live or Inactivated Vaccines. Avian Dis 2022; 66:465-478. [PMID: 36715481 DOI: 10.1637/aviandiseases-d-22-99992] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 09/01/2022] [Indexed: 01/24/2023]
Abstract
Avian orthoreoviruses are causative agents of tenosynovitis and viral arthritis in both chickens and turkeys. Current commercial reovirus vaccines do not protect against disease caused by emerging variants. Custom-made inactivated reovirus vaccines are commonly utilized to help protect commercial poultry against disease. Antibody epitopes located on the viral attachment protein, σC, involved in virus neutralization, have not been clearly identified. In this study, the S1133 vaccine strain (Genetic Cluster 1 [GC1], a GC1 field isolate (117816), and a GC5 field isolate (94826) were determined to be genetically and serologically unrelated. In addition, chickens were vaccinated with either a commercial S1133 vaccine, 117816 GC1, or 94826 GC5, and sera were used in peptide microarrays to identify linear B-cell epitopes within the σC protein. Specific-pathogen-free (SPF) chickens were vaccinated twice with either: 1) live and live, 2) inactivated and inactivated, or 3) a combination of live and inactivated vaccines. Epitope mapping was performed on individual serum samples from birds in each group using S1133, 117816, and 94826 σC sequences translated into an overlapping peptides and spotted onto microarray chips. Vaccination with a combination of live and inactivated viruses resulted in a greater number of B-cell binding sites on the outer-capsid domains of σC for 117816 and 94826, but not for S1133. In contrast, the S1133-vaccinated birds demonstrated fewer epitopes, and those epitopes were located in the stalk region of the protein. However, within each of the vaccinated groups, the highest virus-neutralization titers were observed in the live/inactivated groups. This study demonstrates differences in antibody binding sites within σC between genetically and antigenically distinct reoviruses and provides initial antigenic characterization of avian orthoreoviruses and insight into the inability of vaccine-induced antibodies to provide adequate protection against variant reovirus-induced disease.
Collapse
Affiliation(s)
- W H Dawe
- Poultry Diagnostic and Research Center, Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, GA 30602
| | - D R Kapczynski
- U.S. National Poultry Research Center, Agricultural Research Services, U.S. Department of Agriculture, Athens, GA 30605
| | - E G Linnemann
- Poultry Diagnostic and Research Center, Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, GA 30602
| | - V R Gauthiersloan
- Poultry Diagnostic and Research Center, Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, GA 30602
| | - H S Sellers
- Poultry Diagnostic and Research Center, Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, GA 30602,
| |
Collapse
|
10
|
Ayalew LE, Ahmed KA, Popowich S, Lockerbie BC, Gupta A, Tikoo SK, Ojkic D, Gomis S. Virulence of Emerging Arthrotropic Avian Reoviruses Correlates With Their Ability to Activate and Traffic Interferon-γ Producing Cytotoxic CD8 + T Cells Into Gastrocnemius Tendon. Front Microbiol 2022; 13:869164. [PMID: 35369435 PMCID: PMC8964311 DOI: 10.3389/fmicb.2022.869164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 02/24/2022] [Indexed: 11/13/2022] Open
Abstract
Newly emerging arthrotropic avian reoviruses (ARVs) are genetically divergent, antigenically heterogeneous, and economically costly. Nevertheless, the mechanism of emerging ARV-induced disease pathogenesis and potential differences in virulence between virus genotypes have not been adequately addressed. In this study, the life cycle of ARV, including the formation of cytoplasmic ARV neo-organelles, paracrystalline structures, and virus release mechanisms, were characterized in the infected host cell by transmission electron microscopy (TEM). In addition, progressive changes in the structure of infected cells were investigated by time-lapse and field emission scanning electron (FE-SE) microscopy. ARVs from the four genotypic cluster groups included in the study caused gross and microscopic lesions in the infected birds. Marked infiltration of γδT cells, CD4+ and CD8+ T lymphocytes were observed in ARV infected tendon tissues starting day 3 post-infection. The ARV variant from genotype cluster-2 triggered significantly high trafficking of IFN-γ producing CD8+ T lymphocytes in tendon tissues and concomitantly showed high morbidity and severe disease manifestations. In contrast, the ARV variant from genotype cluster-4 was less virulent, caused milder disease, and accompanied less infiltration of IFN-γ producing CD8+ T cells. Interestingly, when we blunted antiviral immune responses using clodronate liposomes (which depletes antigen-presenting cells) or cyclosporin (which inhibits cytokine production that regulates T-cell proliferation), significantly lower IFN-γ producing CD8+ T cells infiltrated into tendon tissues, resulting in reduced tendon tissues apoptosis and milder disease manifestations. In summary, these data suggest that the degree of ARV virulence and tenosynovitis/arthritis are potentially directly associated with the ability of the virus to traffic massive infiltration of cytotoxic CD8+ T cells into the infected tissues. Moreover, the ability to traffic cytotoxic CD8+ T cells into infected tendon tissues and the severity of tenosynovitis differ between variants from different ARV genotype cluster groups. However, more than one virus isolate per genotype group needs to be tested to further confirm the association of pathogenicity with genotype. These findings can be used to further examine the interaction of viral and cellular pathways which are essential for the pathogenesis of the disease at the molecular level and to develop effective disease control strategies.
Collapse
Affiliation(s)
- Lisanework E Ayalew
- Department of Veterinary Pathology, Western College of Veterinary Medicine (WCVM), University of Saskatchewan, Saskatoon, SK, Canada
| | - Khawaja Ashfaque Ahmed
- Department of Veterinary Pathology, Western College of Veterinary Medicine (WCVM), University of Saskatchewan, Saskatoon, SK, Canada
| | - Shelly Popowich
- Department of Veterinary Pathology, Western College of Veterinary Medicine (WCVM), University of Saskatchewan, Saskatoon, SK, Canada
| | - Betty-Chow Lockerbie
- Department of Veterinary Pathology, Western College of Veterinary Medicine (WCVM), University of Saskatchewan, Saskatoon, SK, Canada
| | - Ashish Gupta
- Department of Veterinary Pathology, Western College of Veterinary Medicine (WCVM), University of Saskatchewan, Saskatoon, SK, Canada
| | - Suresh K Tikoo
- Vaccinology and Immunotherapeutics Program, School of Public Health, University of Saskatchewan, Saskatoon, SK, Canada
| | - Davor Ojkic
- Animal Health Laboratory, Laboratory Services Division, University of Guelph, Guelph, ON, Canada
| | - Susantha Gomis
- Department of Veterinary Pathology, Western College of Veterinary Medicine (WCVM), University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
11
|
Huang W, Li JY, Wu YY, Liao TL, Nielsen BL, Liu HJ. p17-Modulated Hsp90/Cdc37 Complex Governs Oncolytic Avian Reovirus Replication by Chaperoning p17, Which Promotes Viral Protein Synthesis and Accumulation of Viral Proteins σC and σA in Viral Factories. J Virol 2022; 96:e0007422. [PMID: 35107368 PMCID: PMC8941905 DOI: 10.1128/jvi.00074-22] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 01/25/2022] [Indexed: 11/20/2022] Open
Abstract
In this work we have determined that heat shock protein 90 (Hsp90) is essential for avian reovirus (ARV) replication by chaperoning the ARV p17 protein. p17 modulates the formation of the Hsp90/Cdc37 complex by phosphorylation of Cdc37, and this chaperone machinery protects p17 from ubiquitin-proteasome degradation. Inhibition of the Hsp90/Cdc37 complex by inhibitors (17-N-allylamino-17-demethoxygeldanamycin 17-AGG, and celastrol) or short hairpin RNAs (shRNAs) significantly reduced expression levels of viral proteins and virus yield, suggesting that the Hsp90/Cdc37 chaperone complex functions in virus replication. The expression levels of p17 were decreased at the examined time points (2 to 7 h and 7 to 16 h) in 17-AAG-treated cells in a dose-dependent manner while the expression levels of viral proteins σA, σC, and σNS were decreased at the examined time point (7 to 16 h). Interestingly, the expression levels of σC, σA, and σNS proteins increased along with coexpression of p17 protein. p17 together with the Hsp90/Cdc37 complex does not increase viral genome replication but enhances viral protein stability, maturation, and virus production. Virus factories of ARV are composed of nonstructural proteins σNS and μNS. We found that the Hsp90/Cdc37 chaperone complex plays an important role in accumulation of the outer-capsid protein σC, inner core protein σA, and nonstructural protein σNS of ARV in viral factories. Depletion of Hsp90 inhibited σA, σC, and p17 proteins colocalized with σNS in viral factories. This study provides novel insights into p17-modulated formation of the Hsp90/Cdc37 chaperone complex governing virus replication via stabilization and maturation of viral proteins and accumulation of viral proteins in viral factories for virus assembly. IMPORTANCE Molecular mechanisms that control stabilization of ARV proteins and the intermolecular interactions among inclusion components remain largely unknown. Here, we show that the ARV p17 is an Hsp90 client protein. The Hsp90/Cdc37 chaperone complex is essential for ARV replication by protecting p17 chaperone from ubiquitin-proteasome degradation. p17 modulates the formation of Hsp90/Cdc37 complex by phosphorylation of Cdc37, and this chaperone machinery protects p17 from ubiquitin-proteasome degradation, suggesting a feedback loop between p17 and the Hsp90/Cdc37 chaperone complex. p17 together with the Hsp90/Cdc37 complex does not increase viral genome replication but enhances viral protein stability and virus production. Depletion of Hsp90 prevented viral proteins σA, σC, and p17 from colocalizing with σNS in viral factories. Our findings elucidate that the Hsp90/Cdc37 complex chaperones p17, which, in turn, promotes the synthesis of viral proteins σA, σC, and σNS and facilitates accumulation of the outer-capsid protein σC and inner core protein σA in viral factories for virus assembly.
Collapse
Affiliation(s)
- Wei‐Ru Huang
- Institute of Molecular Biology, National Chung Hsing University, Taichung, Taiwan
- The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| | - Jyun-Yi Li
- Institute of Molecular Biology, National Chung Hsing University, Taichung, Taiwan
- The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| | - Yi-Ying Wu
- Institute of Molecular Biology, National Chung Hsing University, Taichung, Taiwan
- The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| | - Tsai-Ling Liao
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan
- Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Brent L. Nielsen
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, Utah, USA
| | - Hung-Jen Liu
- Institute of Molecular Biology, National Chung Hsing University, Taichung, Taiwan
- The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan
- Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung, Taiwan
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
12
|
Kim SW, Choi YR, Park JY, Wei B, Shang K, Zhang JF, Jang HK, Cha SY, Kang M. Isolation and Genomic Characterization of Avian Reovirus From Wild Birds in South Korea. Front Vet Sci 2022; 9:794934. [PMID: 35155656 PMCID: PMC8831841 DOI: 10.3389/fvets.2022.794934] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 01/06/2022] [Indexed: 11/16/2022] Open
Abstract
Avian reoviruses (ARVs) cause severe arthritis, tenosynovitis, pericarditis, and depressed growth in chickens, and these conditions have become increasingly frequent in recent years. Studies on the role of wild birds in the epidemiology of ARVs are insufficient. This study provides information about currently circulating ARVs in wild birds by gene detection using diagnostic RT-PCR, virus isolation, and genomic characterization. In this study, we isolated and identified 10 ARV isolates from 7,390 wild birds' fecal samples, including migratory bird species (bean goose, Eurasian teal, Indian spot-billed duck, and mallard duck) from 2015 to 2019 in South Korea. On comparing the amino acid sequences of the σC-encoding gene, most isolates, except A18-13, shared higher sequence similarity with the commercial vaccine isolate S1133 and Chinese isolates. However, the A18-13 isolate is similar to live attenuated vaccine av-S1133 and vaccine break isolates (SD09-1, LN09-1, and GX110116). For the p10- and p17-encoding genes, all isolates have identical fusion associated small transmembrane (FAST) protein and nuclear localization signal (SNL) motif to chicken-origin ARVs. Phylogenetic analysis of the amino acid sequences of the σC-encoding gene revealed that all isolates were belonged to genotypic cluster I. For the p10- and p17-encoding genes, the nucleotide sequences of all isolates indicated close relationship with commercial vaccine isolate S1133 and Chinese isolates. For the σNS-encoding gene, the nucleotide sequences of all isolates indicated close relationship with the Californian chicken-origin isolate K1600657 and belonged to chicken-origin ARV cluster. Our data indicates that wild birds ARVs were derived from the chicken farms. This finding suggests that wild birds serve as natural carriers of such viruses for domestic poultry.
Collapse
|
13
|
Zhang J, Li T, Wang W, Xie Q, Wan Z, Qin A, Ye J, Shao H. Isolation and Molecular Characteristics of a Novel Recombinant Avian Orthoreovirus From Chickens in China. Front Vet Sci 2021; 8:771755. [PMID: 34950724 PMCID: PMC8688761 DOI: 10.3389/fvets.2021.771755] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 10/31/2021] [Indexed: 11/13/2022] Open
Abstract
In recent years, the emergence of avian orthoreovirus (ARV) has caused significant losses to the poultry industry worldwide. In this study, a novel ARV isolate, designated as AHZJ19, was isolated and identified from domestic chicken with viral arthritis syndrome in China. AHZJ19 can cause typical syncytial cytopathic effect in the chicken hepatocellular carcinoma cell line, LMH. High-throughput sequencing using Illumina technology revealed that the genome size of AHZJ19 is about 23,230 bp, which codes 12 major proteins. Phylogenetic tree analysis found that AHZJ19 was possibly originated from a recombination among Hungarian strains, North American strains, and Chinese strains based on the sequences of the 12 proteins. Notably, the σC protein of AHZJ19 shared only about 50% homology with that of the vaccine strains S1133 and 1733, which also significantly differed from other reported Chinese ARV strains. The isolation and molecular characteristics of AHZJ19 provided novel insights into the molecular epidemiology of ARV and laid the foundation for developing efficient strategies for control of ARV in China.
Collapse
Affiliation(s)
- Jun Zhang
- Ministry of Education Key Laboratory for Avian Preventive Medicine, Key Laboratory of Jiangsu Preventive Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Tuofan Li
- Ministry of Education Key Laboratory for Avian Preventive Medicine, Key Laboratory of Jiangsu Preventive Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Weikang Wang
- Ministry of Education Key Laboratory for Avian Preventive Medicine, Key Laboratory of Jiangsu Preventive Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Quan Xie
- Ministry of Education Key Laboratory for Avian Preventive Medicine, Key Laboratory of Jiangsu Preventive Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Zhimin Wan
- Ministry of Education Key Laboratory for Avian Preventive Medicine, Key Laboratory of Jiangsu Preventive Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Aijian Qin
- Ministry of Education Key Laboratory for Avian Preventive Medicine, Key Laboratory of Jiangsu Preventive Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Jianqiang Ye
- Ministry of Education Key Laboratory for Avian Preventive Medicine, Key Laboratory of Jiangsu Preventive Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Hongxia Shao
- Ministry of Education Key Laboratory for Avian Preventive Medicine, Key Laboratory of Jiangsu Preventive Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China
| |
Collapse
|
14
|
Huang WR, Li JY, Liao TL, Yeh CM, Wang CY, Wen HW, Hu NJ, Wu YY, Hsu CY, Chang YK, Chang CD, Nielsen BL, Liu HJ. Molecular chaperone TRiC governs avian reovirus replication by protecting outer-capsid protein σC and inner core protein σA and non-structural protein σNS from ubiquitin- proteasome degradation. Vet Microbiol 2021; 264:109277. [PMID: 34826648 DOI: 10.1016/j.vetmic.2021.109277] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/27/2021] [Accepted: 11/07/2021] [Indexed: 01/15/2023]
Abstract
Avian reoviruses (ARVs) are important pathogens that cause considerable economic losses in poultry farming. To date, host factors that control stabilization of ARV proteins remain largely unknown. In this work we determined that the eukaryotic chaperonin T-complex protein-1 (TCP-1) ring complex (TRiC) is essential for avian reovirus (ARV) replication by stabilizing outer-capsid protein σC, inner core protein σA, and the non-structural protein σNS of ARV. TriC serves as a chaperone of viral proteins and prevent their degradation via the ubiquitin-proteasome pathway. Furthermore, reciprocal co-immunoprecipitation assays confirmed the association of viral proteins (σA, σC, and σNS) with TRiC. Immunofluorescence staining indicated that the TRiC chaperonins (CCT2 and CCT5) are colocalized with viral proteins σC, σA, and σNS of ARV. In this study, inhibition of TRiC chaperonins (CCT2 and CCT5) by the inhibitor HSF1A or shRNAs significantly reduced expression levels of the σC, σA, and σNS proteins of ARV as well as virus yield, suggesting that the TRiC complex functions in stabilization of viral proteins and virus replication. This study provides novel insights into TRiC chaperonin governing virus replication via stabilization of outer-capsid protein σC, inner core protein σA, and the non-structural protein σNS of ARV.
Collapse
Affiliation(s)
- Wei-Ru Huang
- Institute of Molecular Biology, National Chung Hsing University, Taichung, Taiwan; The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| | - Jyun-Yi Li
- Institute of Molecular Biology, National Chung Hsing University, Taichung, Taiwan; The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| | - Tsai-Ling Liao
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan; Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan; Ph.D Program in Translational Medicine, National Chung Hsing University, Taichung, 402, Taiwan
| | - Chuan-Ming Yeh
- Institute of Molecular Biology, National Chung Hsing University, Taichung, Taiwan; Bioproduction Reearch Institute, National Institute of Advanced Industrial Science and Technology, Tsukaba, Japan
| | - Chi-Young Wang
- The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan; Department of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Hsiao-Wei Wen
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung, Taiwan
| | - Nien-Jen Hu
- Institute of Biochemistry, National Chung Hsing University, Taichung, Taiwan
| | - Yi-Ying Wu
- Institute of Molecular Biology, National Chung Hsing University, Taichung, Taiwan; The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| | - Chao-Yu Hsu
- Ph.D Program in Translational Medicine, National Chung Hsing University, Taichung, 402, Taiwan; Division of Urology, Department of Surgery, Tung's Taichung MetroHarbor Hospital, Taichung, Taiwan
| | - Yu-Kang Chang
- Department of Medical Research, Tung's Taichung MetroHarbor Hospital, Taichung, Taiwan; Depertment of Nursing, Jen-Teh Junior College of Medicine and Management, Taiwan
| | - Ching-Dong Chang
- Department of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Brent L Nielsen
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, USA
| | - Hung-Jen Liu
- Institute of Molecular Biology, National Chung Hsing University, Taichung, Taiwan; The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan; Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan; Ph.D Program in Translational Medicine, National Chung Hsing University, Taichung, 402, Taiwan; Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan.
| |
Collapse
|
15
|
De la Torre D, Astolfi-Ferreira CS, Chacón RD, Puga B, Piantino Ferreira AJ. Emerging new avian reovirus variants from cases of enteric disorders and arthritis/tenosynovitis in Brazilian poultry flocks. Br Poult Sci 2021; 62:361-372. [PMID: 33448227 DOI: 10.1080/00071668.2020.1864808] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
1. The objective of this study was to characterise circulating Brazilian avian reovirus (ARV) strains by genetic analysis of the σC protein encoded by segment 1 of the viral genome and compare these with those of viral strains used for immunising commercial poultry.2. The analysis detected the presence of ARV genomes by quantitative reverse transcriptase PCR (RT-qPCR) in the enteric samples and the joint tissues (JT) of birds with signs of viral arthritis/tenosynovitis. Nucleotide sequencing used 16 strains (three commercial vaccines, 10 from enteric tissues and three from JT). The results indicated high variability in the amino acid sequences of 13 wild strains, showing between 40% and 75% similarity compared with the vaccine strains (S1133 and 2177).3. The sequences were grouped into three well-defined clusters in a phylogenetic tree, two of these clusters together with previous Brazilian σC ARV sequences, and one cluster (VII) that was novel for Brazilian strains. Antigenic analysis showed that there were amino acids within putative epitopes located on the surface of the receptor-binding region of the σC protein with a high degree of variability.4. The study confirmed the presence of ARV genetic variants circulating in commercial birds in Brazil, and according to the antigenic prediction, the possibility of antigenic variants appears to be high.
Collapse
Affiliation(s)
- D De la Torre
- School of Veterinary Medicine, Institute for Research in Biomedicine, Central University of Ecuador, Quito, CP, Ecuador.,School of Veterinary Medicine, University of São Paulo, São Paulo, SP, Brazil
| | | | - R D Chacón
- School of Veterinary Medicine, University of São Paulo, São Paulo, SP, Brazil
| | - B Puga
- School of Veterinary Medicine, Institute for Research in Biomedicine, Central University of Ecuador, Quito, CP, Ecuador
| | | |
Collapse
|
16
|
Development of a Recombinant Pichinde Virus-Vectored Vaccine against Turkey Arthritis Reovirus and Its Immunological Response Characterization in Vaccinated Animals. Pathogens 2021; 10:pathogens10020197. [PMID: 33668435 PMCID: PMC7918942 DOI: 10.3390/pathogens10020197] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/10/2021] [Accepted: 02/11/2021] [Indexed: 12/15/2022] Open
Abstract
Vaccination may be an effective way to reduce turkey arthritis reovirus (TARV)-induced lameness in turkey flocks. However, there are currently no commercial vaccines available against TARV infection. Here, we describe the use of reverse genetics technology to generate a recombinant Pichinde virus (PICV) that expresses the Sigma C and/or Sigma B proteins of TARV as antigens. Nine recombinant PICV-based TARV vaccines were developed carrying the wild-type S1 (Sigma C) and/or S3 (Sigma B) genes from three different TARV strains. In addition, three recombinant PICV-based TARV vaccines were produced carrying codon-optimized S1 and/or S3 genes of a TARV strain. The S1 and S3 genes and antigens were found to be expressed in virus-infected cells via reverse transcriptase polymerase chain reaction (RT-PCR) and the direct fluorescent antibody (DFA) technique, respectively. Turkey poults inoculated with the recombinant PICV-based TARV vaccine expressing the bivalent TARV S1 and S3 antigens developed high anti-TARV antibody titers, indicating the immunogenicity (and safety) of this vaccine. Future in vivo challenge studies using a turkey reovirus infection model will determine the optimum dose and protective efficacy of this recombinant virus-vectored candidate vaccine.
Collapse
|
17
|
Yang Y, Gaspard G, McMullen N, Duncan R. Polycistronic Genome Segment Evolution and Gain and Loss of FAST Protein Function during Fusogenic Orthoreovirus Speciation. Viruses 2020; 12:v12070702. [PMID: 32610593 PMCID: PMC7412057 DOI: 10.3390/v12070702] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 06/03/2020] [Accepted: 06/25/2020] [Indexed: 12/29/2022] Open
Abstract
The Reoviridae family is the only non-enveloped virus family with members that use syncytium formation to promote cell–cell virus transmission. Syncytiogenesis is mediated by a fusion-associated small transmembrane (FAST) protein, a novel family of viral membrane fusion proteins. Previous evidence suggested the fusogenic reoviruses arose from an ancestral non-fusogenic virus, with the preponderance of fusogenic species suggesting positive evolutionary pressure to acquire and maintain the fusion phenotype. New phylogenetic analyses that included the atypical waterfowl subgroup of avian reoviruses and recently identified new orthoreovirus species indicate a more complex relationship between reovirus speciation and fusogenic capacity, with numerous predicted internal indels and 5’-terminal extensions driving the evolution of the orthoreovirus’ polycistronic genome segments and their encoded FAST and fiber proteins. These inferred recombination events generated bi- and tricistronic genome segments with diverse gene constellations, they occurred pre- and post-orthoreovirus speciation, and they directly contributed to the evolution of the four extant orthoreovirus FAST proteins by driving both the gain and loss of fusion capability. We further show that two distinct post-speciation genetic events led to the loss of fusion in the waterfowl isolates of avian reovirus, a recombination event that replaced the p10 FAST protein with a heterologous, non-fusogenic protein and point substitutions in a conserved motif that destroyed the p10 assembly into multimeric fusion platforms.
Collapse
Affiliation(s)
- Yiming Yang
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (Y.Y.); (G.G.); (N.M.)
| | - Gerard Gaspard
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (Y.Y.); (G.G.); (N.M.)
| | - Nichole McMullen
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (Y.Y.); (G.G.); (N.M.)
| | - Roy Duncan
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (Y.Y.); (G.G.); (N.M.)
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Correspondence:
| |
Collapse
|
18
|
Ayalew LE, Ahmed KA, Mekuria ZH, Lockerbie B, Popowich S, Tikoo SK, Ojkic D, Gomis S. The dynamics of molecular evolution of emerging avian reoviruses through accumulation of point mutations and genetic re-assortment. Virus Evol 2020; 6:veaa025. [PMID: 32411390 PMCID: PMC7211400 DOI: 10.1093/ve/veaa025] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
In the last decade, the emergence of variant strains of avian reovirus (ARV) has caused enormous economic impact in the poultry industry across Canada and USA. ARVs are non-enveloped viruses with ten segments of double-stranded RNA genome. So far, only six genotyping cluster groups are identified worldwide based on sequence analysis of the σC protein encoded by the S1 segment. In this study, we performed deep next generation whole-genome sequencing and analysis of twelve purified ARVs isolated from Saskatchewan, Canada. The viruses represent different genotyping cluster. A genome-wide sequence divergence of up to 25 per cent was observed between the virus isolates with a comparable and contrasting evolutionary history. The proportion of synonymous single-nucleotide variations (sSNVs) was higher than the non-synonymous (ns) SNVs across all the genomic segments. Genomic segment S1 was the most variable as compared with the other genes followed by segment M2. Evidence of positive episodic/diversifying selection was observed at different codon positions in the σC protein sequence, which is the genetic marker for the classification of ARV genotypes. In addition, the N-terminus of σC protein had a persuasive diversifying selection, which was not detected in other genomic segments. We identified only four ARV genotypes based on the most variable σC gene sequence. However, a different pattern of phylogenetic clustering was observed with concatenated whole-genome sequences. Together with the accumulation of point mutations, multiple re-assortment events appeared as mechanisms of ARV evolution. For the first time, we determined the mean rate of molecular evolution of ARVs, which was computed as 2.3 × 10-3 substitution/site/year. In addition, widespread geographic intermixing of ARVs was observed between Canada and USA, and between different countries of the world. In conclusion, the study provides a comprehensive analysis of the complete genome of different genotyping clusters of ARVs including their molecular rate of evolution and spatial distribution. The new findings in this study can be utilized for the development of effective vaccines and other control strategies against ARV-induced arthritis/tenosynovitis in the poultry industry worldwide.
Collapse
Affiliation(s)
- Lisanework E Ayalew
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, Saskatchewan, S7N 5B4, Canada
| | - Khawaja Ashfaque Ahmed
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, Saskatchewan, S7N 5B4, Canada
| | - Zelalem H Mekuria
- College of Veterinary Medicine and Global One Health Initiative, Infectious Disease Molecular Epidemiology Laboratory, The Ohio State University, 1900 Coffey Road, Columbus, Ohio, 43210, USA
| | - Betty Lockerbie
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, Saskatchewan, S7N 5B4, Canada
| | - Shelly Popowich
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, Saskatchewan, S7N 5B4, Canada
| | - Suresh K Tikoo
- Vaccinology & Immunotherapeutic Program, School of Public Health, University of Saskatchewan, 104 Clinic Place, Saskatoon, Saskatchewan, S7N 5E3, Canada
| | - Davor Ojkic
- Animal Health Laboratory, Laboratory Services Division, University of Guelph, 419 Gordon St., Guelph, Ontario, N1H 6R8, Canada
| | - Susantha Gomis
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, Saskatchewan, S7N 5B4, Canada
| |
Collapse
|
19
|
Abstract
With no limiting membrane surrounding virions, nonenveloped viruses have no need for membrane fusion to gain access to intracellular replication compartments. Consequently, nonenveloped viruses do not encode membrane fusion proteins. The only exception to this dogma is the fusogenic reoviruses that encode fusion-associated small transmembrane (FAST) proteins that induce syncytium formation. FAST proteins are the smallest viral membrane fusion proteins and, unlike their enveloped virus counterparts, are nonstructural proteins that evolved specifically to induce cell-to-cell, not virus-cell, membrane fusion. This distinct evolutionary imperative is reflected in structural and functional features that distinguish this singular family of viral fusogens from all other protein fusogens. These rudimentary fusogens comprise specific combinations of different membrane effector motifs assembled into small, modular membrane fusogens. FAST proteins offer a minimalist model to better understand the ubiquitous process of protein-mediated membrane fusion and to reveal novel mechanisms of nonenveloped virus dissemination that contribute to virulence.
Collapse
Affiliation(s)
- Roy Duncan
- Department of Microbiology & Immunology, Department of Biochemistry & Molecular Biology, and Department of Pediatrics, Dalhousie University, Halifax, Nova Scotia, Canada, B3H 4R2;
| |
Collapse
|
20
|
Singh AK, Nguyen TH, Vidovszky MZ, Harrach B, Benkő M, Kirwan A, Joshi L, Kilcoyne M, Berbis MÁ, Cañada FJ, Jiménez-Barbero J, Menéndez M, Wilson SS, Bromme BA, Smith JG, van Raaij MJ. Structure and N-acetylglucosamine binding of the distal domain of mouse adenovirus 2 fibre. J Gen Virol 2018; 99:1494-1508. [PMID: 30277856 DOI: 10.1099/jgv.0.001145] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Murine adenovirus 2 (MAdV-2) infects cells of the mouse gastrointestinal tract. Like human adenoviruses, it is a member of the genus Mastadenovirus, family Adenoviridae. The MAdV-2 genome has a single fibre gene that expresses a 787 residue-long protein. Through analogy to other adenovirus fibre proteins, it is expected that the carboxy-terminal virus-distal head domain of the fibre is responsible for binding to the host cell, although the natural receptor is unknown. The putative head domain has little sequence identity to adenovirus fibres of known structure. In this report, we present high-resolution crystal structures of the carboxy-terminal part of the MAdV-2 fibre. The structures reveal a domain with the typical adenovirus fibre head topology and a domain containing two triple β-spiral repeats of the shaft domain. Through glycan microarray profiling, saturation transfer difference nuclear magnetic resonance spectroscopy, isothermal titration calorimetry and site-directed mutagenesis, we show that the fibre specifically binds to the monosaccharide N-acetylglucosamine (GlcNAc). The crystal structure of the complex reveals that GlcNAc binds between the AB and CD loops at the top of each of the three monomers of the MAdV-2 fibre head. However, infection competition assays show that soluble GlcNAc monosaccharide and natural GlcNAc-containing polymers do not inhibit infection by MAdV-2. Furthermore, site-directed mutation of the GlcNAc-binding residues does not prevent the inhibition of infection by soluble fibre protein. On the other hand, we show that the MAdV-2 fibre protein binds GlcNAc-containing mucin glycans, which suggests that the MAdV-2 fibre protein may play a role in viral mucin penetration in the mouse gut.
Collapse
Affiliation(s)
- Abhimanyu K Singh
- 1Departamento de Estructura de Macromoléculas, Centro Nacional de Biotecnologia (CNB-CSIC), Calle Darwin 3, 28049 Madrid, Spain.,†Present address: School of Biosciences, Stacey Building, University of Kent, Canterbury CT2 7NJ, UK
| | - Thanh H Nguyen
- 1Departamento de Estructura de Macromoléculas, Centro Nacional de Biotecnologia (CNB-CSIC), Calle Darwin 3, 28049 Madrid, Spain.,‡Present address: Genetic Engineering Laboratory, Institute of Biotechnology (IBT-VAST), 18 Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam
| | - Márton Z Vidovszky
- 2Institute for Veterinary Medical Research, Centre for Agricultural Research, Hungarian Academy of Sciences, Budapest, Hungary
| | - Balázs Harrach
- 2Institute for Veterinary Medical Research, Centre for Agricultural Research, Hungarian Academy of Sciences, Budapest, Hungary
| | - Mária Benkő
- 2Institute for Veterinary Medical Research, Centre for Agricultural Research, Hungarian Academy of Sciences, Budapest, Hungary
| | - Alan Kirwan
- 3Glycoscience Group, National Centre for Biomedical Engineering Science, National University of Ireland, Galway, Ireland
| | - Lokesh Joshi
- 3Glycoscience Group, National Centre for Biomedical Engineering Science, National University of Ireland, Galway, Ireland
| | - Michelle Kilcoyne
- 4Carbohydrate Signalling Group, Microbiology, School of Natural Sciences, National University of Ireland, Galway, Ireland
| | - M Álvaro Berbis
- 5Departamento de Biología Estructural y Química, Centro de Investigaciones Biológicas (CIB-CSIC), Madrid, Spain
| | - F Javier Cañada
- 5Departamento de Biología Estructural y Química, Centro de Investigaciones Biológicas (CIB-CSIC), Madrid, Spain
| | - Jesús Jiménez-Barbero
- 5Departamento de Biología Estructural y Química, Centro de Investigaciones Biológicas (CIB-CSIC), Madrid, Spain.,§Present address: Molecular Recognition and Host-Pathogen Interactions Unit, CIC bioGUNE, Bizkaia Technology Park, Building 801A, 48170 Derio, Spain.,¶Present address: Ikerbasque, Basque Foundation for Science, Maria Diaz de Haro 13, 48009 Bilbao, Spain
| | - Margarita Menéndez
- 6Departamento de Química Física-Biológica, Instituto de Química Física Rocasolano (IQFR-CSIC), Madrid, Spain.,7CIBER of Respiratory Diseases (CIBERES-ISCIII), Madrid, Spain
| | - Sarah S Wilson
- 8Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Beth A Bromme
- 8Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Jason G Smith
- 8Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Mark J van Raaij
- 1Departamento de Estructura de Macromoléculas, Centro Nacional de Biotecnologia (CNB-CSIC), Calle Darwin 3, 28049 Madrid, Spain
| |
Collapse
|
21
|
Structural and Functional Features of the Reovirus σ1 Tail. J Virol 2018; 92:JVI.00336-18. [PMID: 29695426 DOI: 10.1128/jvi.00336-18] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 04/12/2018] [Indexed: 12/11/2022] Open
Abstract
Mammalian orthoreovirus attachment to target cells is mediated by the outer capsid protein σ1, which projects from the virion surface. The σ1 protein is a homotrimer consisting of a filamentous tail, which is partly inserted into the virion; a body domain constructed from β-spiral repeats; and a globular head with receptor-binding properties. The σ1 tail is predicted to form an α-helical coiled coil. Although σ1 undergoes a conformational change during cell entry, the nature of this change and its contributions to viral replication are unknown. Electron micrographs of σ1 molecules released from virions identified three regions of flexibility, including one at the midpoint of the molecule, that may be involved in its structural rearrangement. To enable a detailed understanding of essential σ1 tail organization and properties, we determined high-resolution structures of the reovirus type 1 Lang (T1L) and type 3 Dearing (T3D) σ1 tail domains. Both molecules feature extended α-helical coiled coils, with T1L σ1 harboring central chloride ions. Each molecule displays a discontinuity (stutter) within the coiled coil and an unexpectedly seamless transition to the body domain. The transition region features conserved interdomain interactions and appears rigid rather than highly flexible. Functional analyses of reoviruses containing engineered σ1 mutations suggest that conserved residues predicted to stabilize the coiled-coil-to-body junction are essential for σ1 folding and encapsidation, whereas central chloride ion coordination and the stutter are dispensable for efficient replication. Together, these findings enable modeling of full-length reovirus σ1 and provide insight into the stabilization of a multidomain virus attachment protein.IMPORTANCE While it is established that different conformational states of attachment proteins of enveloped viruses mediate receptor binding and membrane fusion, less is understood about how such proteins mediate attachment and entry of nonenveloped viruses. The filamentous reovirus attachment protein σ1 binds cellular receptors; contains regions of predicted flexibility, including one at the fiber midpoint; and undergoes a conformational change during cell entry. Neither the nature of the structural change nor its contribution to viral infection is understood. We determined crystal structures of large σ1 fragments for two different reovirus serotypes. We observed an unexpectedly tight transition between two domains spanning the fiber midpoint, which allows for little flexibility. Studies of reoviruses with engineered changes near the σ1 midpoint suggest that the stabilization of this region is critical for function. Together with a previously determined structure, we now have a complete model of the full-length, elongated reovirus σ1 attachment protein.
Collapse
|
22
|
Ayalew LE, Gupta A, Fricke J, Ahmed KA, Popowich S, Lockerbie B, Tikoo SK, Ojkic D, Gomis S. Phenotypic, genotypic and antigenic characterization of emerging avian reoviruses isolated from clinical cases of arthritis in broilers in Saskatchewan, Canada. Sci Rep 2017; 7:3565. [PMID: 28620186 PMCID: PMC5472580 DOI: 10.1038/s41598-017-02743-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 04/19/2017] [Indexed: 11/08/2022] Open
Abstract
In recent years, emerging strains of pathogenic arthrogenic avian reovirus (ARV) have become a challenge to the chicken industry across USA and Canada causing significant economic impact. In this study, we characterized emerging variant ARV strains and examined their genetic and antigenic relationship with reference strains. We isolated 37 emerging variant ARV strains from tendons of broiler chickens with clinical cases of arthritis/tenosynovitis at commercial farms in Saskatchewan, Canada. Viral characterization using immunocytochemistry, gold-immunolabeling and electron microscopy revealed distinct features characteristic of ARV. Polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) analyses of the viral Sigma C gene revealed genetic heterogeneity between the field isolates. On phylogenetic analyses, the Sigma C amino acid sequences of the isolates were clustered into four distinct genotypic groups. These ARV field strains were genetically diverse and quite distant from the vaccine and vaccine related field strains. Antibodies produced against a commercial Reo 2177 ® vaccine did not neutralize these variants. Moreover, structure based analysis of the Sigma C protein revealed significant antigenic variability between the cluster groups and the vaccine strains. To the best of our knowledge, this is the first report on the genetic, phenotypic and antigenic characterization of emerging ARVs in Canada.
Collapse
Affiliation(s)
- Lisanework E Ayalew
- Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Ashish Gupta
- Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Jenny Fricke
- Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Khawaja Ashfaque Ahmed
- Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Shelly Popowich
- Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Betty Lockerbie
- Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Suresh K Tikoo
- Vaccinology & Immunotherapeutic Program, School of Public Health, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Davor Ojkic
- Animal Health Laboratory, Laboratory Services Division, University of Guelph, Guelph, Ontario, Canada
| | - Susantha Gomis
- Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.
| |
Collapse
|
23
|
Grass carp reovirus-GD108 fiber protein is involved in cell attachment. Virus Genes 2017; 53:613-622. [PMID: 28550501 DOI: 10.1007/s11262-017-1467-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 05/15/2017] [Indexed: 10/19/2022]
Abstract
Viral attachment to specific host receptors is the first step in viral infection and serves an essential function in the selection of target cells. In this study, structure analysis, neutralization assays, and cell attachment assays were carried out to evaluate the cell attachment functions of the outer capsid fiber protein of grass carp reovirus GD108 strain (GCRV-GD108). The GCRV-GD108 fiber protein contained 512 amino acids encoded by S7 segment and shared sequence similarities with mammalian reovirus cell attachment protein σ1 and adenovirus fiber. Structural analyses predicted the presence of a coiled-coil tail domain, three adenoviral shafts in the body domain, and a globular head domain, similar to other fiber proteins. Neutralization assays showed that polyclonal antibodies against the fiber protein could prevent viral infection in both fish and grass carp snout fibroblast cells (PSF), suggesting that the recombinant fiber protein could induce neutralized antibodies against GCRV-GD108. Cell attachment assays showed that recombinant fiber protein could bind to PSF cells, demonstrating that the fiber protein functioned as the cell attachment protein in GCRV-GD108. These results provided the basis for further studies of the pathogenesis of grass carp reovirus.
Collapse
|
24
|
Nguyen TH, Ballmann MZ, Do HT, Truong HN, Benkő M, Harrach B, van Raaij MJ. Crystal structure of raptor adenovirus 1 fibre head and role of the beta-hairpin in siadenovirus fibre head domains. Virol J 2016; 13:106. [PMID: 27334597 PMCID: PMC4918002 DOI: 10.1186/s12985-016-0558-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 06/08/2016] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Most adenoviruses recognize their host cells via an interaction of their fibre head domains with a primary receptor. The structural framework of adenovirus fibre heads is conserved between the different adenovirus genera for which crystal structures have been determined (Mastadenovirus, Aviadenovirus, Atadenovirus and Siadenovirus), but genus-specific differences have also been observed. The only known siadenovirus fibre head structure, that of turkey adenovirus 3 (TAdV-3), revealed a twisted beta-sandwich resembling the reovirus fibre head architecture more than that of other adenovirus fibre heads, plus a unique beta-hairpin embracing a neighbouring monomer. The TAdV-3 fibre head was shown to bind sialyllactose. METHODS Raptor adenovirus 1 (RAdV-1) fibre head was expressed, crystallized and its structure was solved and refined at 1.5 Å resolution. The structure could be solved by molecular replacement using the TAdV-3 fibre head structure as a search model, despite them sharing a sequence identity of only 19 %. Versions of both the RAdV-1 and TAdV-3 fibre heads with their beta-hairpin arm deleted were prepared and their stabilities were compared with the non-mutated proteins by a thermal unfolding assay. RESULTS The structure of the RAdV-1 fibre head contains the same twisted ABCJ-GHID beta-sandwich and beta-hairpin arm as the TAdV-3 fibre head. However, while the predicted electro-potential surface charge of the TAdV-3 fibre head is mainly positive, the RAdV-1 fibre head shows positively and negatively charged patches and does not appear to bind sialyllactose. Deletion of the beta-hairpin arm does not affect the structure of the raptor adenovirus 1 fibre head and only affects the stability of the RAdV-1 and TAdV-3 fibre heads slightly. CONCLUSIONS The high-resolution structure of RAdV-1 fibre head is the second known structure of a siadenovirus fibre head domain. The structure shows that the siadenovirus fibre head structure is conserved, but differences in the predicted surface charge suggest that RAdV-1 uses a different natural receptor for cell attachment than TAdV-3. Deletion of the beta-hairpin arm shows little impact on the structure and stability of the siadenovirus fibre heads.
Collapse
Affiliation(s)
- Thanh H Nguyen
- Departamento de Estructura de Macromoléculas, Centro Nacional de Biotecnología (CNB-CSIC), Calle Darwin 3, E-28049, Madrid, Spain.,Genetic Engineering Laboratory, Institute of Biotechnology (IBT-VAST), 18 Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam
| | - Mónika Z Ballmann
- Institute for Veterinary Medical Research, Centre for Agricultural Research, Hungarian Academy of Sciences, Budapest, Hungary
| | - Huyen T Do
- Genetic Engineering Laboratory, Institute of Biotechnology (IBT-VAST), 18 Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam
| | - Hai N Truong
- Genetic Engineering Laboratory, Institute of Biotechnology (IBT-VAST), 18 Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam
| | - Mária Benkő
- Institute for Veterinary Medical Research, Centre for Agricultural Research, Hungarian Academy of Sciences, Budapest, Hungary
| | - Balázs Harrach
- Institute for Veterinary Medical Research, Centre for Agricultural Research, Hungarian Academy of Sciences, Budapest, Hungary
| | - Mark J van Raaij
- Departamento de Estructura de Macromoléculas, Centro Nacional de Biotecnología (CNB-CSIC), Calle Darwin 3, E-28049, Madrid, Spain.
| |
Collapse
|
25
|
Goldenberg D, Lublin A, Rosenbluth E, Heller ED, Pitcovski J. Optimized polypeptide for a subunit vaccine against avian reovirus. Vaccine 2016; 34:3178-3183. [PMID: 27155492 DOI: 10.1016/j.vaccine.2016.04.036] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Revised: 04/10/2016] [Accepted: 04/12/2016] [Indexed: 11/29/2022]
Abstract
Avian reovirus (ARV) is a disease-causing agent. The disease is prevented by vaccination with a genotype-specific vaccine while many variants of ARV exist in the field worldwide. Production of new attenuated vaccines is a long-term process and in the case of fast-mutating viruses, an impractical one. In the era of molecular biology, vaccines may be produced by using only the relevant protein for induction of neutralizing antibodies, enabling fast adjustment to the emergence of new genetic strains. Sigma C (SC) protein of ARV is a homotrimer that facilitates host-cell attachment and induce the production and secretion of neutralizing antibodies. The aim of this study was to identify the region of SC that will elicit a protective immune response. Full-length (residues 1-326) and two partial fragments of SC (residues 122-326 and 192-326) were produced in Escherichia coli. The SC fragment of residues 122-326 include the globular head, shaft and hinge domains, while eliminating intra-capsular region. This fragment induces significantly higher levels of anti-ARV antibodies than the shorter fragment or full length SC, which neutralized embryos infection by the virulent strain to a higher extent compared with the antibodies produced in response to the whole virus vaccine. Residues 122-326 fragment is assumed to be folded correctly, exposing linear as well as conformational epitopes that are identical to those of the native protein, while possibly excluding suppressor sequences. The results of this study may serve for the development of a recombinant subunit vaccine for ARV.
Collapse
Affiliation(s)
- Dana Goldenberg
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel; Migal - Galilee Technology Center, Kiryat Shmona, Israel
| | - Avishai Lublin
- Division of Avian and Fish Diseases, Kimron Veterinary Institute, Bet Dagan, Israel
| | - Ezra Rosenbluth
- Division of Avian and Fish Diseases, Kimron Veterinary Institute, Bet Dagan, Israel
| | - E Dan Heller
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Jacob Pitcovski
- Migal - Galilee Technology Center, Kiryat Shmona, Israel; Department of Biotechnology, Tel-Hai Academic College, Israel.
| |
Collapse
|
26
|
Reverse Genetics for Fusogenic Bat-Borne Orthoreovirus Associated with Acute Respiratory Tract Infections in Humans: Role of Outer Capsid Protein σC in Viral Replication and Pathogenesis. PLoS Pathog 2016; 12:e1005455. [PMID: 26901882 PMCID: PMC4762779 DOI: 10.1371/journal.ppat.1005455] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 01/24/2016] [Indexed: 12/26/2022] Open
Abstract
Nelson Bay orthoreoviruses (NBVs) are members of the fusogenic orthoreoviruses and possess 10-segmented double-stranded RNA genomes. NBV was first isolated from a fruit bat in Australia more than 40 years ago, but it was not associated with any disease. However, several NBV strains have been recently identified as causative agents for respiratory tract infections in humans. Isolation of these pathogenic bat reoviruses from patients suggests that NBVs have evolved to propagate in humans in the form of zoonosis. To date, no strategy has been developed to rescue infectious viruses from cloned cDNA for any member of the fusogenic orthoreoviruses. In this study, we report the development of a plasmid-based reverse genetics system free of helper viruses and independent of any selection for NBV isolated from humans with acute respiratory infection. cDNAs corresponding to each of the 10 full-length RNA gene segments of NBV were cotransfected into culture cells expressing T7 RNA polymerase, and viable NBV was isolated using a plaque assay. The growth kinetics and cell-to-cell fusion activity of recombinant strains, rescued using the reverse genetics system, were indistinguishable from those of native strains. We used the reverse genetics system to generate viruses deficient in the cell attachment protein σC to define the biological function of this protein in the viral life cycle. Our results with σC-deficient viruses demonstrated that σC is dispensable for cell attachment in several cell lines, including murine fibroblast L929 cells but not in human lung epithelial A549 cells, and plays a critical role in viral pathogenesis. We also used the system to rescue a virus that expresses a yellow fluorescent protein. The reverse genetics system developed in this study can be applied to study the propagation and pathogenesis of pathogenic NBVs and in the generation of recombinant NBVs for future vaccines and therapeutics. Nelson Bay orthoreoviruses (NBVs) are members of the fusogenic orthoreoviruses that have various host species, including reptiles, birds, and mammals. Recently, several NBV strains have been isolated from patients with acute respiratory tract infections. Isolation of these pathogenic reoviruses raises concerns about the potential emerging infections of bat-borne orthoreoviruses in humans. The development of an entirely plasmid-based reverse genetics system for double-stranded RNA viruses has trailed other systems of major animal RNA virus groups because of the technical complexities involved in the manipulation of genomes composed of 10 or more segments. In this study, we developed a plasmid-based reverse genetics system for a pathogenic NBV strain. We used this system to generate viruses incapable of expressing the cell attachment protein σC and to rescue a replication-competent virus that expresses a yellow fluorescent protein. Our studies using σC-deficient viruses suggest that NBVs may engage multiple independent viral ligands and cellular receptors for efficient cell attachment and viral pathogenesis, thus providing new insight into the biology of orthoreoviruses. The reverse genetics approach described in this study can be exploited for fusogenic orthoreovirus biology and used to develop vaccines, diagnostics, and therapeutics.
Collapse
|
27
|
Kryshtafovych A, Moult J, Baslé A, Burgin A, Craig TK, Edwards RA, Fass D, Hartmann MD, Korycinski M, Lewis RJ, Lorimer D, Lupas AN, Newman J, Peat TS, Piepenbrink KH, Prahlad J, van Raaij MJ, Rohwer F, Segall AM, Seguritan V, Sundberg EJ, Singh AK, Wilson MA, Schwede T. Some of the most interesting CASP11 targets through the eyes of their authors. Proteins 2015; 84 Suppl 1:34-50. [PMID: 26473983 PMCID: PMC4834066 DOI: 10.1002/prot.24942] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 09/17/2015] [Accepted: 10/11/2015] [Indexed: 11/17/2022]
Abstract
The Critical Assessment of protein Structure Prediction (CASP) experiment would not have been possible without the prediction targets provided by the experimental structural biology community. In this article, selected crystallographers providing targets for the CASP11 experiment discuss the functional and biological significance of the target proteins, highlight their most interesting structural features, and assess whether these features were correctly reproduced in the predictions submitted to CASP11. Proteins 2016; 84(Suppl 1):34–50. © 2015 The Authors. Proteins: Structure, Function, and Bioinformatics Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | - John Moult
- Department of Cell Biology and Molecular Genetics, Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, Maryland, 20850
| | - Arnaud Baslé
- Institute for Cell and Molecular Biosciences, University of Newcastle, Newcastle upon Tyne, NE2 4HH, United Kingdom
| | - Alex Burgin
- Broad Institute, Cambridge, Massachusetts, 02142
| | | | - Robert A Edwards
- Department of Biology, San Diego State University, San Diego, California, 92182.,Department of Computer Science, San Diego State University, San Diego, California, 92182
| | - Deborah Fass
- Department of Structural Biology, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Marcus D Hartmann
- Department of Protein Evolution, Max Planck Institute for Developmental Biology, Tübingen, 72076, Germany
| | - Mateusz Korycinski
- Department of Protein Evolution, Max Planck Institute for Developmental Biology, Tübingen, 72076, Germany
| | - Richard J Lewis
- Institute for Cell and Molecular Biosciences, University of Newcastle, Newcastle upon Tyne, NE2 4HH, United Kingdom
| | | | - Andrei N Lupas
- Department of Protein Evolution, Max Planck Institute for Developmental Biology, Tübingen, 72076, Germany
| | - Janet Newman
- Biomedical Manufacturing Program, CSIRO, Parkville, VIC, Australia
| | - Thomas S Peat
- Biomedical Manufacturing Program, CSIRO, Parkville, VIC, Australia
| | - Kurt H Piepenbrink
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland, 21201
| | - Janani Prahlad
- Department of Biochemistry and Redox Biology Center, University of Nebraska-Lincoln, Lincoln, Nebraska, 68588
| | - Mark J van Raaij
- Centro Nactional De Biotecnologia (CNB-CSIC), Madrid, E-28049, Spain
| | - Forest Rohwer
- Department of Biology and Viral Information Institute, San Diego State University, San Diego, California, 92182
| | - Anca M Segall
- Department of Biology and Viral Information Institute, San Diego State University, San Diego, California, 92182
| | | | - Eric J Sundberg
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland, 21201.,Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, 21201.,Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, 21201
| | - Abhimanyu K Singh
- School of Biosciences, University of Kent, Canterbury, Kent, United Kingdom
| | - Mark A Wilson
- Department of Biochemistry and Redox Biology Center, University of Nebraska-Lincoln, Lincoln, Nebraska, 68588
| | - Torsten Schwede
- Biozentrum, University of Basel, Basel, 4056, Switzerland. .,SIB Swiss Institute of Bioinformatics, Basel, 4056, Switzerland.
| |
Collapse
|
28
|
Singh AK, Berbís MÁ, Ballmann MZ, Kilcoyne M, Menéndez M, Nguyen TH, Joshi L, Cañada FJ, Jiménez-Barbero J, Benkő M, Harrach B, van Raaij MJ. Structure and Sialyllactose Binding of the Carboxy-Terminal Head Domain of the Fibre from a Siadenovirus, Turkey Adenovirus 3. PLoS One 2015; 10:e0139339. [PMID: 26418008 PMCID: PMC4587935 DOI: 10.1371/journal.pone.0139339] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 09/11/2015] [Indexed: 01/16/2023] Open
Abstract
The virulent form of turkey adenovirus 3 (TAdV-3), also known as turkey hemorrhagic enteritis virus (THEV), is an economically important poultry pathogen, while the avirulent form is used as a vaccine. TAdV-3 belongs to the genus Siadenovirus. The carboxy-terminal region of its fibre does not have significant sequence similarity to any other adenovirus fibre heads of known structure. Two amino acid sequence differences between virulent and avirulent TAdV-3 map on the fibre head: where virulent TAdV-3 contains Ile354 and Thr376, avirulent TAdV-3 contains Met354 and Met376. We determined the crystal structures of the trimeric virulent and avirulent TAdV-3 fibre head domains at 2.2 Å resolution. Each monomer contains a beta-sandwich, which, surprisingly, resembles reovirus fibre head more than other adenovirus fibres, although the ABCJ-GHID topology is conserved in all. A beta-hairpin insertion in the C-strand of each trimer subunit embraces its neighbouring monomer. The avirulent and virulent TAdV-3 fibre heads are identical apart from the exact orientation of the beta-hairpin insertion. In vitro, sialyllactose was identified as a ligand by glycan microarray analysis, nuclear magnetic resonance spectroscopy, and crystallography. Its dissociation constant was measured to be in the mM range by isothermal titration calorimetry. The ligand binds to the side of the fibre head, involving amino acids Glu392, Thr419, Val420, Lys421, Asn422, and Gly423 binding to the sialic acid group. It binds slightly more strongly to the avirulent form. We propose that, in vivo, the TAdV-3 fibre may bind a sialic acid-containing cell surface component.
Collapse
Affiliation(s)
- Abhimanyu K. Singh
- Departamento de Estructura de Macromoléculas, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - M. Álvaro Berbís
- Departamento de Biología Física-Química, Centro de Investigaciones Biológicas (CIB-CSIC), Madrid, Spain
| | - Mónika Z. Ballmann
- Institute for Veterinary Medical Research, Centre for Agricultural Research, Hungarian Academy of Sciences, Budapest, Hungary
| | - Michelle Kilcoyne
- Glycoscience Group, National Centre for Biomedical Engineering Science, National University of Ireland Galway, Galway, Ireland
- Microbiology, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - Margarita Menéndez
- Departamento de Química Física-Biológica, Instituto de Química Física Rocasolano (IQFR-CSIC) and CIBER de Enfermedades Respiratorias (CIBERES), calle Serrano 119, E-28006 Madrid, Spain
| | - Thanh H. Nguyen
- Departamento de Estructura de Macromoléculas, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Lokesh Joshi
- Glycoscience Group, National Centre for Biomedical Engineering Science, National University of Ireland Galway, Galway, Ireland
| | - F. Javier Cañada
- Departamento de Biología Física-Química, Centro de Investigaciones Biológicas (CIB-CSIC), Madrid, Spain
| | - Jesús Jiménez-Barbero
- Departamento de Biología Física-Química, Centro de Investigaciones Biológicas (CIB-CSIC), Madrid, Spain
- Centro de Investigación Cooperativa en Biociencias (CIC bioGUNE), Parque Tecnológico de Bizkaia, Derio, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Mária Benkő
- Institute for Veterinary Medical Research, Centre for Agricultural Research, Hungarian Academy of Sciences, Budapest, Hungary
| | - Balázs Harrach
- Institute for Veterinary Medical Research, Centre for Agricultural Research, Hungarian Academy of Sciences, Budapest, Hungary
| | - Mark J. van Raaij
- Departamento de Estructura de Macromoléculas, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
- * E-mail:
| |
Collapse
|
29
|
Nguyen TH, Vidovszky MZ, Ballmann MZ, Sanz-Gaitero M, Singh AK, Harrach B, Benkő M, van Raaij MJ. Crystal structure of the fibre head domain of bovine adenovirus 4, a ruminant atadenovirus. Virol J 2015; 12:81. [PMID: 25994880 PMCID: PMC4451742 DOI: 10.1186/s12985-015-0309-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 05/11/2015] [Indexed: 01/20/2023] Open
Abstract
Background In adenoviruses, primary host cell recognition is generally performed by the head domains of their homo-trimeric fibre proteins. This first interaction is reversible. A secondary, irreversible interaction subsequently takes place via other adenovirus capsid proteins and leads to a productive infection. Although many fibre head structures are known for human mastadenoviruses, not many animal adenovirus fibre head structures have been determined, especially not from those belonging to adenovirus genera other than Mastadenovirus. Methods We constructed an expression vector for the fibre head domain from a ruminant atadenovirus, bovine adenovirus 4 (BAdV-4), consisting of amino acids 414–535, expressed the protein in Escherichia coli, purified it by metal affinity and cation exchange chromatography and crystallized it. The structure was solved using single isomorphous replacement plus anomalous dispersion of a mercury derivative and refined against native data that extended to 1.2 Å resolution. Results Like in other adenoviruses, the BAdV-4 fibre head monomer contains a beta-sandwich consisting of ABCJ and GHID sheets. The topology is identical to the fibre head of the other studied atadenovirus, snake adenovirus 1 (SnAdV-1), including the alpha-helix in the DG-loop, despite of them having a sequence identity of only 15 %. There are also differences which may have implications for ligand binding. Beta-strands G and H are longer and differences in several surface-loops and surface charge are observed. Conclusions Chimeric adenovirus fibres have been used to retarget adenovirus-based anti-cancer and gene therapy vectors. Ovine adenovirus 7 (OAdV-7), another ruminant atadenovirus, is intensively tested as a basis for such a vector. Here, we present the high-resolution atomic structure of the BAdV-4 fibre head domain, the second atadenovirus fibre head structure known and the first of an atadenovirus that infects a mammalian host. Future research should focus on the receptor-binding properties of these fibre head domains.
Collapse
Affiliation(s)
- Thanh H Nguyen
- Departamento de Estructura de Macromoleculas, Centro Nacional de Biotecnologia (CNB-CSIC), calle Darwin 3, 28049, Madrid, Spain.
| | - Márton Z Vidovszky
- Institute for Veterinary Medical Research, Centre for Agricultural Research, Hungarian Academy of Sciences, Budapest, Hungary.
| | - Mónika Z Ballmann
- Institute for Veterinary Medical Research, Centre for Agricultural Research, Hungarian Academy of Sciences, Budapest, Hungary.
| | - Marta Sanz-Gaitero
- Departamento de Estructura de Macromoleculas, Centro Nacional de Biotecnologia (CNB-CSIC), calle Darwin 3, 28049, Madrid, Spain. .,Department of Biological Sciences, Cork Institute of Technology, Bishopstown, Cork, Ireland.
| | - Abhimanyu K Singh
- Departamento de Estructura de Macromoleculas, Centro Nacional de Biotecnologia (CNB-CSIC), calle Darwin 3, 28049, Madrid, Spain. .,Current address: School of Biosciences, Stacey Building, University of Kent, Canterbury, Kent, CT2 7NJ, United Kingdom.
| | - Balázs Harrach
- Institute for Veterinary Medical Research, Centre for Agricultural Research, Hungarian Academy of Sciences, Budapest, Hungary.
| | - Mária Benkő
- Institute for Veterinary Medical Research, Centre for Agricultural Research, Hungarian Academy of Sciences, Budapest, Hungary.
| | - Mark J van Raaij
- Departamento de Estructura de Macromoleculas, Centro Nacional de Biotecnologia (CNB-CSIC), calle Darwin 3, 28049, Madrid, Spain.
| |
Collapse
|
30
|
Crystal structure of the fibre head domain of the Atadenovirus Snake Adenovirus 1. PLoS One 2014; 9:e114373. [PMID: 25486282 PMCID: PMC4259310 DOI: 10.1371/journal.pone.0114373] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 11/06/2014] [Indexed: 02/05/2023] Open
Abstract
Adenoviruses are non-enveloped icosahedral viruses with trimeric fibre proteins protruding from their vertices. There are five known genera, from which only Mastadenoviruses have been widely studied. Apart from studying adenovirus as a biological model system and with a view to prevent or combat viral infection, there is a major interest in using adenovirus for vaccination, cancer therapy and gene therapy purposes. Adenoviruses from the Atadenovirus genus have been isolated from squamate reptile hosts, ruminants and birds and have a characteristic gene organization and capsid morphology. The carboxy-terminal virus-distal fibre head domains are likely responsible for primary receptor recognition. We determined the high-resolution crystal structure of the Snake Adenovirus 1 (SnAdV-1) fibre head using the multi-wavelength anomalous dispersion (MAD) method. Despite the absence of significant sequence homology, this Atadenovirus fibre head has the same beta-sandwich propeller topology as other adenovirus fibre heads. However, it is about half the size, mainly due to much shorter loops connecting the beta-strands. The detailed structure of the SnAdV-1 fibre head and other animal adenovirus fibre heads, together with the future identification of their natural receptors, may lead to the development of new strategies to target adenovirus vectors to cells of interest.
Collapse
|
31
|
Tu JM, Chang MC, Huang LLH, Chang CD, Huang HJ, Lee RH, Chang CC. The blue fluorescent protein from Vibrio vulnificus CKM-1 is a useful reporter for plant research. BOTANICAL STUDIES 2014; 55:79. [PMID: 28510958 PMCID: PMC5432841 DOI: 10.1186/s40529-014-0079-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 12/04/2014] [Indexed: 06/07/2023]
Abstract
BACKGROUND The mBFP is an improved variant of NADPH-dependent blue fluorescent protein that was originally identified from the non-bioluminescent pathogenic bacteria Vibrio vulnificus CKM-1. To explore the application of mBFP in plants, the mBFP gene expression was driven by one of the three promoters, namely, leaf-specific (RbcS), hypoxia-inducible (Adh) or auxin-inducible (DR5) promoters, in different plant tissues such as leaves, roots and flowers under diverse treatments. In addition, the expressed mBFP protein was targeted to five subcellular compartments such as cytosol, endoplasmic reticulum, apoplast, chloroplast and mitochondria, respectively, in plant cells. RESULTS When the mBFP was transiently expressed in the tobacco leaves and floral tissues of moth orchid, the cytosol and apoplast exhibited brighter blue fluorescence than other compartments. The recombinant mBFP-mS1C fusion protein exhibited enhanced fluorescence intensity that was correlated with more abundant RNA transcripts (1.8 fold) as compared with a control. In the root tips of horizontally grown transgenic Arabidopsis, mBFP could be induced as a reporter under hypoxia condition. Furthermore, the mBFP was localized to the expected subcellular compartments, except that dual targeting was found when the mBFP was fused with the mitochondria-targeting signal peptide. Additionally, the brightness of mBFP blue fluorescence was correlated with NADPH concentration. CONCLUSION The NADPH-dependent blue fluorescent protein could serve as a useful reporter in plants under aerobic or hypoxic condition. However, to avoid masking the mitochondrial targeting signal, fusing mBFP as a fusion tag in the C-terminal will be better when the mBFP is applied in mitochondria trafficking study. Furthermore, mBFP might have the potential to be further adopted as a NADPH biosensor in plant cells. Future codon optimization of mBFP for plants could significantly enhance its brightness and expand its potential applications.
Collapse
Affiliation(s)
- Jin-Min Tu
- Institute of Tropical Plant Sciences, National Cheng Kung University, 1 University Rd, Tainan, 701 Taiwan
| | - Ming-Chung Chang
- Department of Nutrition, Hung Kuang University, Taichung, 433 Taiwan
| | - Lynn LH Huang
- Institute of Biotechnology, National Cheng Kung University, Tainan, 701 Taiwan
| | - Ching-Dong Chang
- Department of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, 912 Taiwan
| | - Hao-Jen Huang
- Department of Life Sciences, National Cheng Kung University, Tainan, 701 Taiwan
| | - Ruey-Hua Lee
- Institute of Tropical Plant Sciences, National Cheng Kung University, 1 University Rd, Tainan, 701 Taiwan
| | - Ching-Chun Chang
- Institute of Tropical Plant Sciences, National Cheng Kung University, 1 University Rd, Tainan, 701 Taiwan
- Institute of Biotechnology, National Cheng Kung University, Tainan, 701 Taiwan
| |
Collapse
|
32
|
Chen WT, Wu YL, Chen T, Cheng CS, Chan HL, Chou HC, Chen YW, Yin HS. Proteomics analysis of the DF-1 chicken fibroblasts infected with avian reovirus strain S1133. PLoS One 2014; 9:e92154. [PMID: 24667214 PMCID: PMC3965424 DOI: 10.1371/journal.pone.0092154] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Accepted: 02/18/2014] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Avian reovirus (ARV) is a member of the Orthoreovirus genus in the Reoviridae family. It is the etiological agent of several diseases, among which viral arthritis and malabsorption syndrome are the most commercially important, causing considerable economic losses in the poultry industry. Although a small but increasing number of reports have characterized some aspects of ARV infection, global changes in protein expression in ARV-infected host cells have not been examined. The current study used a proteomics approach to obtain a comprehensive view of changes in protein levels in host cells upon infection by ARV. METHODOLOGY AND PRINCIPAL FINDINGS The proteomics profiles of DF-1 chicken fibroblast cells infected with ARV strain S1133 were analyzed by two-dimensional differential-image gel electrophoresis. The majority of protein expression changes (≥ 1.5 fold, p<0.05) occurred at 72 h post-infection. Matrix-assisted laser desorption ionization time-of-flight mass spectrometry identified 51 proteins with differential expression levels, including 25 that were upregulated during ARV infection and 26 that were downregulated. These proteins were divided into eight groups according to biological function: signal transduction, stress response, RNA processing, the ubiquitin-proteasome pathway, lipid metabolism, carbohydrate metabolism, energy metabolism, and cytoskeleton organization. They were further examined by immunoblotting to validate the observed alterations in protein expression. CONCLUSION/SIGNIFICANCE This is the first report of a time-course proteomic analysis of ARV-infected host cells. Notably, all identified proteins involved in signal transduction, RNA processing, and the ubiquitin-proteasome pathway were downregulated in infected cells, whereas proteins involved in DNA synthesis, apoptosis, and energy production pathways were upregulated. In addition, other differentially expressed proteins were linked with the cytoskeleton, metabolism, redox regulation, and stress response. These proteomics data provide valuable information about host cell responses to ARV infection and will facilitate further studies of the molecular mechanisms underlying ARV pathogenesis.
Collapse
Affiliation(s)
- Wen-Ting Chen
- Institute of Bioinformatics and Structural Biology and College of Life Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Yi-Le Wu
- Institute of Bioinformatics and Structural Biology and College of Life Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Ting Chen
- Institute of Bioinformatics and Structural Biology and College of Life Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Chao-Sheng Cheng
- Institute of Bioinformatics and Structural Biology and College of Life Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Hong-Lin Chan
- Institute of Bioinformatics and Structural Biology and College of Life Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Hsiu-Chuan Chou
- Department of Applied Science, National Hsinchu University of Education, Hsinchu, Taiwan
| | - Yi-Wen Chen
- Institute of Bioinformatics and Structural Biology and College of Life Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Hsien-Sheng Yin
- Institute of Bioinformatics and Structural Biology and College of Life Sciences, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
33
|
Rosa ACG, Ferreira HL, Gomes DE, Táparo CV, Cardoso TC. Isolation and molecular characterization of Brazilian turkey reovirus from immunosuppressed young poults. Arch Virol 2013; 159:1453-7. [PMID: 24327096 PMCID: PMC7086608 DOI: 10.1007/s00705-013-1947-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Accepted: 11/01/2013] [Indexed: 11/27/2022]
Abstract
In this study, we investigated turkey reovirus (TReoV) in tissue samples from young birds, aged 15 days. RT-PCR for TReoV detected 3.3 % positive samples and TReoV was successfully isolated in Vero cells. Histological analysis of positive bursa of Fabricius (BF) revealed atrophied follicles and lymphocyte depletion. The number of CD8+, CD4+ and IgM+ cells was lower in infected BF. Phylogenetic analysis based on S3 gene showed that the Brazilian TReoV isolates clustered in a single group with 98-100 % similarity to TReoV strains circulating in the United States. This is the first indication that TReoV infection may be a contributing factor to immunosuppression in young birds.
Collapse
Affiliation(s)
- Ana Carolina G. Rosa
- Laboratório de Virologia, Faculdade de Medicina Veterinária, University of São Paulo State, Rua Clóvis Pestana, 793, Araçatuba, SP CEP 16050-680 Brazil
| | - Helena Lage Ferreira
- Departamento de Medicina Veterinária, FZEA-USP, Av. Duque de Caxias Norte, 225, Pirassununga, SP CEP 13635-900 Brazil
| | - Deriane Elias Gomes
- Laboratório de Virologia, Faculdade de Medicina Veterinária, University of São Paulo State, Rua Clóvis Pestana, 793, Araçatuba, SP CEP 16050-680 Brazil
| | - Cilene Vidovix Táparo
- Laboratório de Virologia, Faculdade de Medicina Veterinária, University of São Paulo State, Rua Clóvis Pestana, 793, Araçatuba, SP CEP 16050-680 Brazil
| | - Tereza Cristina Cardoso
- Laboratório de Virologia, Faculdade de Medicina Veterinária, University of São Paulo State, Rua Clóvis Pestana, 793, Araçatuba, SP CEP 16050-680 Brazil
- Departamento de Apoio, Produção e Saúde Animal, Curso de Medicina Veterinária, Rua Clóvis Pestana, 793, Araçatuba, SP CEP 16050-680 Brazil
| |
Collapse
|
34
|
Yin CH, Qin LT, Sun MY, Gao YL, Qi XL, Gao HL, Wang YQ, Jang LL, Wang XM. Identification of a linear B-Cell epitope on avian reovirus protein sigmaC. Virus Res 2013; 178:530-4. [PMID: 24076298 DOI: 10.1016/j.virusres.2013.09.028] [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: 07/30/2013] [Revised: 09/03/2013] [Accepted: 09/16/2013] [Indexed: 11/24/2022]
Abstract
SigmaC (σC) protein, which mediates virus attachment to target cells, is the most variable proteins of avian reovirus (ARV). It is responsible for inducing protective antibody immune responses in animals. To understand the antigenic determinants of σC protein, a set of partially overlapping and consecutive peptides spanning σC were expressed and then screened with the monoclonal antibody (mAb) 2B5 directed against σC. The mAb 2B5 recognized peptides with the σC motif (45)ELLHRSISDISTTV(58). Further identification of the displayed B-cell epitope was conducted with a set of truncated peptides expressed as GST fusion proteins. The Western blot and ELISA results indicated that (45)ELLHRSISDI(54) was the minimal determinant of the linear B-cell epitope. Using sequences analysis, we found that this epitope was not a common motif shared among the other members of the ARV and DRV groups. Furthermore, cross reactivity analysis showed that the associated coding motif of other ARV and DRV groups was not recognized by 2B5. These data suggested that (45)ELLHRSISDI(54) was a type-specific linear B-cell epitope of avian reovirus. The results in this study may have potential applications in the development of diagnostic techniques and epitope-based marker vaccines against ARV, which is prevalent in China.
Collapse
Affiliation(s)
- Chun-hong Yin
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Nibert ML, Duncan R. Bioinformatics of recent aqua- and orthoreovirus isolates from fish: evolutionary gain or loss of FAST and fiber proteins and taxonomic implications. PLoS One 2013; 8:e68607. [PMID: 23861926 PMCID: PMC3701659 DOI: 10.1371/journal.pone.0068607] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 05/30/2013] [Indexed: 01/07/2023] Open
Abstract
Family Reoviridae, subfamily Spinareovirinae, includes nine current genera. Two of these genera, Aquareovirus and Orthoreovirus, comprise members that are closely related and consistently share nine homologous proteins. Orthoreoviruses have 10 dsRNA genome segments and infect reptiles, birds, and mammals, whereas aquareoviruses have 11 dsRNA genome segments and infect fish. Recently, the first 10-segmented fish reovirus, piscine reovirus (PRV), has been identified and shown to be phylogenetically divergent from the 11-segmented viruses constituting genus Aquareovirus. We have recently extended results for PRV by showing that it does not encode a fusion-associated small transmembrane (FAST) protein, but does encode an outer-fiber protein containing a long N-terminal region of predicted α-helical coiled coil. Three recently characterized 11-segmented fish reoviruses, obtained from grass carp in China and sequenced in full, are also divergent from the viruses now constituting genus Aquareovirus, though not to the same extent as PRV. In the current study, we reexamined the sequences of these three recent isolates of grass carp reovirus (GCRV)–HZ08, GD108, and 104–for further clues to their evolution relative to other aqua- and orthoreoviruses. Structure-based fiber motifs in their encoded outer-fiber proteins were characterized, and other bioinformatics analyses provided evidence against the presence of a FAST protein among their encoded nonstructural proteins. Phylogenetic comparisons showed the combination of more distally branching, approved Aquareovirus and Orthoreovirus members, plus more basally branching isolates GCRV104, GCRV-HZ08/GD108, and PRV, constituting a larger, monophyletic taxon not suitably recognized by the current taxonomic hierarchy. Phylogenetics also suggested that the last common ancestor of all these viruses was a fiber-encoding, nonfusogenic virus and that the FAST protein family arose from at least two separate gain-of-function events. In addition, an apparent evolutionary correlation was found between the gain or loss of NS-FAST and outer-fiber proteins among more distally branching members of this taxon.
Collapse
Affiliation(s)
- Max L. Nibert
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail: E-mails: (MLN); (RD)
| | - Roy Duncan
- Department of Microbiology and Immunology, Department of Biochemistry and Molecular Biology, and Department of Pediatrics, Dalhousie University, Halifax, Nova Scotia, Canada
- * E-mail: E-mails: (MLN); (RD)
| |
Collapse
|
36
|
Danthi P, Holm GH, Stehle T, Dermody TS. Reovirus receptors, cell entry, and proapoptotic signaling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 790:42-71. [PMID: 23884585 DOI: 10.1007/978-1-4614-7651-1_3] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Mammalian orthoreoviruses (reoviruses) are members of the Reoviridae. Reoviruses contain 10 double-stranded (ds) RNA gene segments enclosed in two concentric protein shells, called outer capsid and core. These viruses serve as a versatile experimental system for studies of viral replication events at the virus-cell interface, including engagement of cell-surface receptors, internalization and disassembly, and activation of the innate immune response, including NF-κB-dependent cellular signaling pathways. Reoviruses also provide a model system for studies of virus-induced apoptosis and organ-specific disease in vivo.Reoviruses attach to host cells via the filamentous attachment protein, σ1. The σ1 protein of all reovirus serotypes engages junctional adhesion molecule-A (JAM-A), an integral component of intercellular tight junctions. The σ1 protein also binds to cell-surface carbohydrate, with the type of carbohydrate bound varying by serotype. Following attachment to JAM-A and carbohydrate, reovirus internalization is mediated by β1 integrins, most likely via clathrin-dependent endocytosis. In the endocytic compartment, reovirus outer-capsid protein σ3 is removed by acid-dependent cysteine proteases in most cell types. Removal of σ3 results in the exposure of a hydrophobic conformer of the viral membrane-penetration protein, μ1, which pierces the endosomal membrane and delivers transcriptionally active reovirus core particles into the cytoplasm.Reoviruses induce apoptosis in both cultured cells and infected mice. Perturbation of reovirus disassembly using inhibitors of endosomal acidification or protease activity abrogates apoptosis. The μ1-encoding M2 gene is genetically linked to strain-specific differences in apoptosis-inducing capacity, suggesting a function for μ1 in induction of death signaling. Reovirus disassembly leads to activation of transcription factor NF-κB, which modulates apoptotic signaling in numerous types of cells. Inhibition of NF-κB nuclear translocation using either pharmacologic agents or expression of transdominant forms of IκB blocks reovirus-induced apoptosis, suggesting an essential role for NF-κB activation in the death response. Multiple effector pathway s downstream of NF-κB-directed gene expression execute reovirus-induced cell death. This chapter will focus on the mechanisms by which reovirus attachment and disassembly activate NF-κB and stimulate the cellular proapoptotic machinery.
Collapse
Affiliation(s)
- Pranav Danthi
- Department of Biology, Indiana University, Bloomington, IN, USA
| | | | | | | |
Collapse
|
37
|
Structure of the receptor-binding carboxy-terminal domain of bacteriophage T7 tail fibers. Proc Natl Acad Sci U S A 2012; 109:9390-5. [PMID: 22645347 DOI: 10.1073/pnas.1119719109] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The six bacteriophage T7 tail fibers, homo-trimers of gene product 17, are thought to be responsible for the first specific, albeit reversible, attachment to Escherichia coli lipopolysaccharide. The protein trimer forms kinked fibers comprised of an amino-terminal tail-attachment domain, a slender shaft, and a carboxyl-terminal domain composed of several nodules. Previously, we expressed, purified, and crystallized a carboxyl-terminal fragment comprising residues 371-553. Here, we report the structure of this protein trimer, solved using anomalous diffraction and refined at 2 Å resolution. Amino acids 371-447 form a tapered pyramid with a triangular cross-section composed of interlocked β-sheets from each of the three chains. The triangular pyramid domain has three α-helices at its narrow end, which are connected to a carboxyl-terminal three-blade β-propeller tip domain by flexible loops. The monomers of this tip domain each contain an eight-stranded β-sandwich. The exact topology of the β-sandwich fold is novel, but similar to that of knob domains of other viral fibers and the phage Sf6 needle. Several host-range change mutants have been mapped to loops located on the top of this tip domain, suggesting that this surface of the tip domain interacts with receptors on the cell surface.
Collapse
|
38
|
Lin KH, Hsu AP, Shien JH, Chang TJ, Liao JW, Chen JR, Lin CF, Hsu WL. Avian reovirus sigma C enhances the mucosal and systemic immune responses elicited by antigen-conjugated lactic acid bacteria. Vaccine 2012; 30:5019-29. [PMID: 22531554 PMCID: PMC7115360 DOI: 10.1016/j.vaccine.2012.04.043] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Revised: 03/29/2012] [Accepted: 04/10/2012] [Indexed: 11/16/2022]
Abstract
Mucosal surfaces are common sites of pathogen colonization/entry. Effective mucosal immunity by vaccination should provide protection at this primary infection site. Our aim was to develop a new vaccination strategy that elicits a mucosal immune response. A new strain of Enterococcus faecium, a non pathogenic lactic acid bacteria (LAB) with strong cell adhesion ability, was identified and used as a vaccine vector to deliver two model antigens. Specifically, sigma (σ) C protein of avian reovirus (ARV), a functional homolog of mammalian reovirus σ1 protein and responsible for M-cell targeting, was administered together with a subfragment of the spike protein of infectious bronchitis virus (IBV). Next, the effect of immunization route on the immune response was assessed by delivering the antigens via the LAB strain. Intranasal (IN) immunization induced stronger humoral responses than intragastic (IG) immunization. IN immunization produced antigen specific IgA both systemically and in the lungs. A higher IgA titer was induced by the LAB with ARV σC protein attached. Moreover, the serum of mice immunized with LAB displaying divalent antigens had much stronger immune reactivity against ARV σC protein compared to IBV-S1. Our results indicate that ARV σC protein delivered by LAB via the IN route elicits strong mucosal immunity. A needle-free delivery approach is a convenient and cost effective method of vaccine administration, especially for respiratory infections in economic animals. Furthermore, ARV σC, a strong immunogen of ARV, may be able to serve as an immunoenhancer for other vaccines, especially avian vaccines.
Collapse
Affiliation(s)
- Kuan-Hsun Lin
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung Hsing University, 250 Kuo Kuang Road, Taichung 402, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Lublin A, Goldenberg D, Rosenbluth E, Heller ED, Pitcovski J. Wide-range protection against avian reovirus conferred by vaccination with representatives of four defined genotypes. Vaccine 2011; 29:8683-8. [PMID: 21911023 DOI: 10.1016/j.vaccine.2011.08.114] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Revised: 08/24/2011] [Accepted: 08/26/2011] [Indexed: 10/17/2022]
Abstract
Many isolates of the contagious avian reovirus have been characterized, mainly based on the sequence of their sigma C protein. These isolates have been classified into four genotypes. Currently available vaccines are of limited effectiveness, likely due to the existence of many variants. The aim of this study was to test the efficacy of a vaccine consisting of a mixture of prototypes (representatives) of the four defined genotypic groups of avian reovirus. The prototypes were selected based on their distance from the isolates within each genotype. All prototypes were found to be virulent. Antibodies produced against each of the prototypes neutralized all members of its genotype. Birds were then vaccinated with a mixture of the four prototypes. Results suggest that the 4-valent vaccine can prevent disease and confer broad protection against field isolates of avian reovirus.
Collapse
Affiliation(s)
- Avishai Lublin
- Division of Avian and Fish Diseases, Kimron Veterinary Institute, Bet Dagan, Israel
| | | | | | | | | |
Collapse
|
40
|
Lu SW, Wang KC, Liu HJ, Chang CD, Huang HJ, Chang CC. Expression of avian reovirus minor capsid protein in plants. J Virol Methods 2011; 173:287-93. [PMID: 21354211 DOI: 10.1016/j.jviromet.2011.02.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2010] [Revised: 09/17/2010] [Accepted: 02/17/2011] [Indexed: 11/27/2022]
Abstract
The minor coat protein of the avian reovirus (ARV), σC, encoded by the S1 genome segment, is one of the major candidates for the development of a subunit vaccine against ARV infection. To develop a plant-based vaccine to immunize poultry against ARV infection, we constructed 4 plant nuclear expression vectors with or without codon modification of the S1 gene, and their expression was driven by a CaMV 35S promoter or rice actin1 promoter. In addition, the expressed σC proteins were targeted subcellularly to cytosol or chloroplasts, respectively. Agrobacterium containing the S1 expression constructs was used to transform tobacco leaf disks, and transformants were selected with kanamycin (100 μg/ml). The integration of the S1 transgene into the tobacco chromosome was confirmed by PCR and Southern blot analysis. Western blot analysis with antiserum against σC was performed to determine the expression of σC protein in transgenic tobacco plants. The highest expression levels of σC protein in the cellular extracts of selected p35S1, pActS1 and p35UmS1 transgenic lines were 0.013%, 0.021% and 0.0013% of the total soluble protein, respectively, but the protein was barely detectable in p35TmS1 transgenic lines. However, the level of σC protein expression was not associated with the level of corresponding RNA transcripts in selected transgenic lines. Taken together, the results suggest that the major limiting factor for the expression of σC protein in plants might be at the post-transcriptional level.
Collapse
MESH Headings
- Antigens, Viral/biosynthesis
- Antigens, Viral/genetics
- Blotting, Southern
- Blotting, Western
- Capsid Proteins/biosynthesis
- Capsid Proteins/genetics
- Genetic Vectors
- Mutagenesis, Insertional
- Orthoreovirus, Avian/genetics
- Plants, Genetically Modified/genetics
- Plants, Genetically Modified/metabolism
- Polymerase Chain Reaction
- Recombinant Proteins/biosynthesis
- Recombinant Proteins/genetics
- Recombination, Genetic
- Rhizobium/genetics
- Nicotiana/genetics
- Nicotiana/metabolism
- Transformation, Genetic
- Viral Vaccines/biosynthesis
- Viral Vaccines/genetics
Collapse
Affiliation(s)
- Shih-Wei Lu
- Institute of Biotechnology, National Cheng Kung University, Tainan 701, Taiwan
| | | | | | | | | | | |
Collapse
|
41
|
Danthi P, Guglielmi KM, Kirchner E, Mainou B, Stehle T, Dermody TS. From touchdown to transcription: the reovirus cell entry pathway. Curr Top Microbiol Immunol 2011; 343:91-119. [PMID: 20397070 PMCID: PMC4714703 DOI: 10.1007/82_2010_32] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
Mammalian orthoreoviruses (reoviruses) are prototype members of the Reoviridae family of nonenveloped viruses. Reoviruses contain ten double-stranded RNA gene segments enclosed in two concentric protein shells, outer capsid and core. These viruses serve as a versatile experimental system for studies of virus cell entry, innate immunity, and organ-specific disease. Reoviruses engage cells by binding to cell-surface carbohydrates and the immunoglobulin superfamily member, junctional adhesion molecule-A (JAM-A). JAM-A is a homodimer formed by extensive contacts between its N-terminal immunoglobulin-like domains. Reovirus attachment protein σ1 disrupts the JAM-A dimer, engaging a single JAM-A molecule by virtually the same interface used for JAM-A homodimerization. Following attachment to JAM-A and carbohydrate, reovirus internalization is promoted by β1 integrins, most likely via clathrin-dependent endocytosis. In the endocytic compartment, reovirus outer-capsid protein σ3 is removed by cathepsin proteases, which exposes the viral membrane-penetration protein, μ1. Proteolytic processing and conformational rearrangements of μ1 mediate endosomal membrane rupture and delivery of transcriptionally active reovirus core particles into the host cell cytoplasm. These events also allow the φ cleavage fragment of μ1 to escape into the cytoplasm where it activates NF-κB and elicits apoptosis. This review will focus on mechanisms of reovirus cell entry and activation of innate immune response signaling pathways.
Collapse
Affiliation(s)
- Pranav Danthi
- Department of Biology, Indiana University, Bloomington, IN, 47405, USA
| | | | | | | | | | | |
Collapse
|
42
|
Goldenberg D, Pasmanik-Chor M, Pirak M, Kass N, Lublin A, Yeheskel A, Heller D, Pitcovski J. Genetic and antigenic characterization of sigma C protein from avian reovirus. Avian Pathol 2010; 39:189-99. [PMID: 20544425 DOI: 10.1080/03079457.2010.480969] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Avian reovirus (ARV) causes viral arthritis, tenosynovitis, liver infection and immunosuppression in birds. Live-attenuated and inactivated vaccines for ARV are available, but do not efficiently protect against recent variants. Sigma C, which mediates virus attachment to target cells, is the most variable protein in ARV. Antibodies to this protein neutralize viral infection. The purpose of the present study was to characterize sigma C in isolates of ARV from infected birds, as compared with the vaccine strain. Amino acids 27 to 293 of sigma C from 28 Israeli isolates were compared, classified and analysed using bioinformatics tools. Large variations were found among the isolates, and the vaccine strain was shown to differ from most of the studied strains, which could explain the failure of commonly used vaccinations in protecting birds against ARV infection. Based on sigma C protein sequences from all over the world, ARV can be divided into four groups. Isolates from all groups were found in the field simultaneously, possibly explaining the insufficient protection achieved by the vaccine strain, which is represented in one of the groups. The results point out the need and the difficulty in producing a wide-ranging vaccine. Several conserved regions among all reported ARV sigma C proteins were identified. These peptides were further studied for structural and functional properties, and for antigenic characterization. The results of this study shed light on peptide selection for a broad and efficient vaccine.
Collapse
|
43
|
Guardado-Calvo P, Fox GC, Llamas-Saiz AL, van Raaij MJ. Crystallographic structure of the -helical triple coiled-coil domain of avian reovirus S1133 fibre. J Gen Virol 2009; 90:672-677. [DOI: 10.1099/vir.0.008276-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
44
|
Kirchner E, Guglielmi KM, Strauss HM, Dermody TS, Stehle T. Structure of reovirus sigma1 in complex with its receptor junctional adhesion molecule-A. PLoS Pathog 2008; 4:e1000235. [PMID: 19079583 PMCID: PMC2588538 DOI: 10.1371/journal.ppat.1000235] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2008] [Accepted: 11/11/2008] [Indexed: 01/12/2023] Open
Abstract
Viral attachment to specific host receptors is the first step in viral infection and serves an essential function in the selection of target cells. Mammalian reoviruses are highly useful experimental models for studies of viral pathogenesis and show promise as vectors for oncolytics and vaccines. Reoviruses engage cells by binding to carbohydrates and the immunoglobulin superfamily member, junctional adhesion molecule-A (JAM-A). JAM-A exists at the cell surface as a homodimer formed by extensive contacts between its N-terminal immunoglobulin-like domains. We report the crystal structure of reovirus attachment protein σ1 in complex with a soluble form of JAM-A. The σ1 protein disrupts the JAM-A dimer, engaging a single JAM-A molecule via virtually the same interface that is used for JAM-A homodimerization. Thus, reovirus takes advantage of the adhesive nature of an immunoglobulin-superfamily receptor by usurping the ligand-binding site of this molecule to attach to the cell surface. The dissociation constant (KD) of the interaction between σ1 and JAM-A is 1,000-fold lower than that of the homophilic interaction between JAM-A molecules, indicating that JAM-A strongly prefers σ1 as a ligand. Analysis of reovirus mutants engineered by plasmid-based reverse genetics revealed residues in σ1 required for binding to JAM-A and infectivity of cultured cells. These studies define biophysical mechanisms of reovirus cell attachment and provide a platform for manipulating reovirus tropism to enhance vector targeting. Mammalian orthoreoviruses (reoviruses) are useful models for studies of virus–receptor interactions and viral pathogenesis. They are closely related in structure to adenoviruses and share similar mechanisms of cell attachment and entry. The receptor for reovirus, junctional adhesion molecule-A (JAM-A), is a component of cellular junctions and also used as a receptor by feline calicivirus. To better understand how viruses engage cellular receptors, we determined the structure of reovirus attachment protein σ1 bound to JAM-A. The structure provides an understanding of the biological function of the interaction and yields information that may enable targeting of reovirus to alternate receptors. Since the repertoire of receptors bound by a virus contributes importantly to determining which types of cells are infected, such targeting plays an essential role in gene delivery for vaccine or therapeutic applications. New cancer therapy approaches include the use of viruses, including reovirus, to lyse tumor cells. New knowledge about reovirus attachment to cellular receptors at an atomic level will help to harness the therapeutic potential of this virus.
Collapse
Affiliation(s)
- Eva Kirchner
- Interfaculty Institute for Biochemistry, University of Tuebingen, Tuebingen, Germany
| | - Kristen M. Guglielmi
- Department of Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- Elizabeth B. Lamb Center for Pediatric Research, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | | | - Terence S. Dermody
- Department of Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- Elizabeth B. Lamb Center for Pediatric Research, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- * E-mail: (TSD); (TS)
| | - Thilo Stehle
- Interfaculty Institute for Biochemistry, University of Tuebingen, Tuebingen, Germany
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- * E-mail: (TSD); (TS)
| |
Collapse
|
45
|
Abstract
Avian reovirus, an important avian pathogen, expresses eight structural and four nonstructural proteins. The structural sigmaA protein is a major component of the inner capsid, clamping together lambdaA building blocks. sigmaA has also been implicated in the resistance of avian reovirus to the antiviral action of interferon by strongly binding double-stranded RNA in the host cell cytoplasm and thus inhibiting activation of the double-stranded RNA-dependent protein kinase. We have solved the structure of bacterially expressed sigmaA by molecular replacement and refined it using data to 2.3-A resolution. Twelve sigmaA molecules are present in the P1 unit cell, arranged as two short double helical hexamers. A positively charged patch is apparent on the surface of sigmaA on the inside of this helix and mutation of either of two key arginine residues (Arg155 and Arg273) within this patch abolishes double-stranded RNA binding. The structural data, together with gel shift assay, electron microscopy, and sedimentation velocity centrifugation results, provide evidence for cooperative binding of sigmaA to double-stranded RNA. The minimal length of double-stranded RNA required for sigmaA binding was observed to be 14 to 18 bp.
Collapse
|
46
|
Day JM, Pantin-Jackwood MJ, Spackman E. Sequence and phylogenetic analysis of the S1 genome segment of turkey-origin reoviruses. Virus Genes 2007; 35:235-42. [PMID: 17265142 DOI: 10.1007/s11262-006-0044-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2006] [Accepted: 09/20/2006] [Indexed: 10/23/2022]
Abstract
Based on previous reports characterizing the turkey-origin avian reovirus (TRV) sigmaB (sigma2) major outer capsid protein gene, the TRVs may represent a new group within the fusogenic orthoreoviruses. However, no sequence data from other TRV genes or genome segments has been reported. The sigmaC protein encoded by the avian reovirus S1 genome segment is the cell attachment protein and a major antigenic determinant for avian reovirus. The chicken reovirus S1 genome segment is well characterized and is well conserved in viruses from that species. This report details the amplification, cloning and sequencing of the entire S1 genome segment from two and the entire coding sequences of the sigmaC, p10 and p17 genes from an additional five TRVs. Sequence analysis reveals that of the three proteins encoded by the TRV S1 genome segment, sigmaC shares at most 57% amino acid identity with sigmaC from the chicken reovirus reference strain S1133, while the most similar p10 and p17 proteins share 72% and 61% identity, respectively, with the corresponding S1133 proteins. The most closely related mammalian reovirus, the fusogenic Nelson Bay reovirus, encodes a sigmaC protein that shares from 25% to 28% amino acid identity with the TRV sigmaC proteins. This report supports the earlier suggestion that the TRVs are a separate virus species within the Orthoreovirus genus, and may provide some insight into TRV host specificity and pathogenesis.
Collapse
Affiliation(s)
- J Michael Day
- Southeast Poultry Research Laboratory, USDA-ARS, 934 College Station Road, Athens, GA 30605, USA
| | | | | |
Collapse
|
47
|
Racine T, Barry C, Roy K, Dawe SJ, Shmulevitz M, Duncan R. Leaky scanning and scanning-independent ribosome migration on the tricistronic S1 mRNA of avian reovirus. J Biol Chem 2007; 282:25613-22. [PMID: 17604272 DOI: 10.1074/jbc.m703708200] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The S1 genome segments of avian and Nelson Bay reovirus encode tricistronic mRNAs containing three sequential partially overlapping open reading frames (ORFs). The translation start site of the 3'-proximal ORF encoding the sigmaC protein lies downstream of two ORFs encoding the unrelated p10 and p17 proteins and more than 600 nucleotides distal from the 5'-end of the mRNA. It is unclear how translation of this remarkable tricistronic mRNA is regulated. We now show that the p10 and p17 ORFs are coordinately expressed by leaky scanning. Translation initiation events at these 5'-proximal ORFs, however, have little to no effect on translation of the 3'-proximal sigmaC ORF. Northern blotting, insertion of upstream stop codons or optimized translation start sites, 5'-truncation analysis, and poliovirus 2A protease-mediated cleavage of eIF4G indicated sigmaC translation derives from a full-length tricistronic mRNA using a mechanism that is eIF4G-dependent but leaky scanning- and translation reinitiation-independent. Further analysis of artificial bicistronic mRNAs failed to provide any evidence that sigmaC translation derives from an internal ribosome entry site. Additional features of the S1 mRNA and the mechanism of sigmaC translation also differ from current models of ribosomal shunting. Translation of the tricistronic reovirus S1 mRNA, therefore, is dependent both on leaky scanning and on a novel scanning-independent mechanism that allows translation initiation complexes to efficiently bypass two functional upstream ORFs.
Collapse
Affiliation(s)
- Trina Racine
- Department of Microbiology and Immunology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia B3H 1X5, Canada
| | | | | | | | | | | |
Collapse
|
48
|
Hermo-Parrado XL, Guardado-Calvo P, Llamas-Saiz AL, Fox GC, Vazquez-Iglesias L, Martínez-Costas J, Benavente J, van Raaij MJ. Crystallization of the avian reovirus double-stranded RNA-binding and core protein sigmaA. Acta Crystallogr Sect F Struct Biol Cryst Commun 2007; 63:426-9. [PMID: 17565188 PMCID: PMC2335010 DOI: 10.1107/s1744309107017988] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2007] [Accepted: 04/10/2007] [Indexed: 11/10/2022]
Abstract
The avian reovirus protein sigmaA plays a dual role: it is a structural protein forming part of the transcriptionally active core, but it has also been implicated in the resistance of the virus to interferon by strongly binding double-stranded RNA and thus inhibiting the double-stranded RNA-dependent protein kinase. The sigmaA protein has been crystallized from solutions containing ammonium sulfate at pH values around 6. Crystals belonging to space group P1, with unit-cell parameters a = 103.2, b = 129.9, c = 144.0 A, alpha = 93.8, beta = 105.1, gamma = 98.2 degrees were grown and a complete data set has been collected to 2.3 A resolution. The self-rotation function suggests that sigmaA may form symmetric arrangements in the crystals.
Collapse
Affiliation(s)
- X. Lois Hermo-Parrado
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Santiago de Compostela, Campus Sur, E-15782 Santiago de Compostela, Spain
| | - Pablo Guardado-Calvo
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Santiago de Compostela, Campus Sur, E-15782 Santiago de Compostela, Spain
| | - Antonio L. Llamas-Saiz
- Unidad de Difracción de Rayos X, Laboratorio Integral de Dinámica y Estructura de Biomoléculas José R. Carracido, Edificio CACTUS, Universidad de Santiago de Compostela, Campus Sur, E-15782 Santiago de Compostela, Spain
| | - Gavin C. Fox
- Spanish CRG Beamline BM16, European Synchrotron Radiation Facility (ESRF), 6 Rue Jules Horowitz, BP 220, F-38043 Grenoble, France
| | - Lorena Vazquez-Iglesias
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Santiago de Compostela, Campus Sur, E-15782 Santiago de Compostela, Spain
| | - José Martínez-Costas
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Santiago de Compostela, Campus Sur, E-15782 Santiago de Compostela, Spain
| | - Javier Benavente
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Santiago de Compostela, Campus Sur, E-15782 Santiago de Compostela, Spain
| | - Mark J. van Raaij
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Santiago de Compostela, Campus Sur, E-15782 Santiago de Compostela, Spain
- Unidad de Difracción de Rayos X, Laboratorio Integral de Dinámica y Estructura de Biomoléculas José R. Carracido, Edificio CACTUS, Universidad de Santiago de Compostela, Campus Sur, E-15782 Santiago de Compostela, Spain
| |
Collapse
|
49
|
Benavente J, Martínez-Costas J. Avian reovirus: Structure and biology. Virus Res 2007; 123:105-19. [PMID: 17018239 DOI: 10.1016/j.virusres.2006.09.005] [Citation(s) in RCA: 168] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2006] [Revised: 09/06/2006] [Accepted: 09/07/2006] [Indexed: 11/28/2022]
Abstract
Avian reoviruses are important pathogens that cause considerable losses to the poultry industry, but they have been poorly characterized at the molecular level in the past, mostly because they have been considered to be very similar to the well-studied mammalian reoviruses. Studies performed over the last 20 years have revealed that avian reoviruses have unique properties and activities, different to those displayed by their mammalian counterparts, and of considerable interest to molecular virologists. Notably, the avian reovirus S1 gene is unique, in that it is a functional tricistronic gene that possesses three out-of-phase and partially overlapping open reading frames; the identification of the mechanisms that govern the initiation of translation of the three S1 cistrons, and the study of the properties and activities displayed by their encoded proteins, are particularly interesting areas of research. For instance, avian reoviruses are one of the few nonenveloped viruses that cause cell-cell fusion, and their fusogenic phenotype has been associated with a nonstructural 10 kDa transmembrane protein, which is expressed by the second cistron of the S1 gene; the small size of this atypical fusion protein offers an interesting model for studying the mechanisms of cell-cell fusion and for identifying fusogenic domains. Finally, avian reoviruses are highly resistant to interferon, and therefore they may be useful for investigating the mechanisms and strategies that viruses utilize to counteract the antiviral actions of interferons.
Collapse
Affiliation(s)
- Javier Benavente
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| | | |
Collapse
|
50
|
Guardado Calvo P, Llamas-Saiz AL, Langlois P, van Raaij MJ. Crystallization of the C-terminal head domain of the avian adenovirus CELO long fibre. Acta Crystallogr Sect F Struct Biol Cryst Commun 2006; 62:449-52. [PMID: 16682773 PMCID: PMC2219974 DOI: 10.1107/s1744309106012024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2006] [Accepted: 04/03/2006] [Indexed: 11/11/2022]
Abstract
Avian adenovirus CELO contains two different fibres: fibre 1, the long fibre, and fibre 2, the short fibre. The short fibre is responsible for binding to an unknown avian receptor and is essential for infection of birds. The long fibre is not essential, but is known to bind the coxsackievirus and adenovirus receptor protein. Both trimeric fibres are attached to the same penton base, of which each icosahedral virus contains 12 copies. The short fibre extends straight outwards, while the long fibre emerges at an angle. The carboxy-terminal amino acids 579-793 of the avian adenovirus long fibre have been expressed with an amino-terminal hexahistidine tag and the expressed trimeric protein has been purified by nickel-affinity chromatography and crystallized. Crystals were grown at low pH using PEG 10,000 as precipitant and belonged to space group C2. The crystals diffracted rotating-anode Cu Kalpha radiation to at least 1.9 angstroms resolution and a complete data set was collected from a single crystal to 2.2 angstroms resolution. Unit-cell parameters were a = 216.5, b = 59.2, c = 57.5 angstroms, beta = 101.3 degrees, suggesting one trimer per asymmetric unit and a solvent content of 46%. The long fibre head does not have significant sequence homology to any other protein of known structure and molecular-replacement attempts with known fibre-head structures were unsuccessful. However, a map calculated using SIRAS phasing shows a clear trimer with a shape similar to known adenovirus fibre-head structures. Structure solution is in progress.
Collapse
Affiliation(s)
- Pablo Guardado Calvo
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Santiago de Compostela, Campus Sur, E-15782 Santiago de Compostela, Spain
| | - Antonio L. Llamas-Saiz
- Unidad de Difracción de Rayos X, Laboratorio Integral de Dinámica y Estructura de Biomoléculas José R. Carracido, Edificio CACTUS, Universidad de Santiago de Compostela, Campus Sur, E-15782 Santiago de Compostela, Spain
| | - Patrick Langlois
- Agence Francaise de Securité Sanitaire des Aliments, Unité Génétique Virale et Biosecurité, Site Les Croix, BP 53, F-22440 Ploufragan, France
| | - Mark J. van Raaij
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Santiago de Compostela, Campus Sur, E-15782 Santiago de Compostela, Spain
- Unidad de Difracción de Rayos X, Laboratorio Integral de Dinámica y Estructura de Biomoléculas José R. Carracido, Edificio CACTUS, Universidad de Santiago de Compostela, Campus Sur, E-15782 Santiago de Compostela, Spain
- Correspondence e-mail:
| |
Collapse
|