Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Int J Biol Macromol ; 253(Pt 6): 127276, 2023 Dec 31.
Article in English | MEDLINE | ID: mdl-37804887

ABSTRACT

Porcine Epidemic diarrhea virus (PEDV), which can result in severe vomiting, diarrhea, dehydration and death in newborn piglets, poses a great threat to the pig industry around the world. The S1 subunit of S protein is crucial for triggering neutralizing antibodies binding to the receptor. Based on the advantages of high immunogenicity and precise assembly of nanoparticles, the mi3 nanoparticles and truncated S1 protein were assembled by the SpyTag/SpyCatcher system and then expressed in HEK293F cells, whereafter high-efficiency monoclonal antibodies (mAbs) were produced and identified. The obtained five mAbs can bind to various genotypes of PEDV, including a mAb (12G) which can neutralize G1 and G2 genotypes of PEDV in vitro. By further identification of monoclonal antibody target sequences, 507FNDHSF512 and 553LFYNVTNSYG562 were first identified as B-cell linear epitopes, in which 553LFYNVTNSYG562 was a neutralizing epitope. Alanine scans identified the key amino acid sites of two epitopes. Moreover, the results of multiple sequence alignment analysis showed that these two epitopes were highly conserved in various subtype variants. In brief, these findings can serve as a basis for additional research of PEDV and prospective resources for the creation of later detection and diagnostic techniques.


Subject(s)
Antibodies, Monoclonal , Porcine epidemic diarrhea virus , Animals , Swine , Antibodies, Viral , Porcine epidemic diarrhea virus/genetics , Porcine epidemic diarrhea virus/chemistry , Prospective Studies , Antibodies, Neutralizing , Epitopes, B-Lymphocyte
2.
PLoS One ; 17(2): e0263582, 2022.
Article in English | MEDLINE | ID: mdl-35139120

ABSTRACT

The membrane protein M of the Porcine Epidemic Diarrhea Virus (PEDV) is the most abundant component of the viral envelope. The M protein plays a central role in the morphogenesis and assembly of the virus through protein interactions of the M-M, M-Spike (S) and M-nucleocapsid (N) type. The M protein is known to induce protective antibodies in pigs and to participate in the antagonistic response of the cellular antiviral system coordinated by the type I and type III interferon pathways. The 3D structure of the PEDV M protein is still unknown. The present work exposes a predicted 3D model of the M protein generated using the Robetta protocol. The M protein model is organized into a transmembrane and a globular region. The obtained 3D model of the PEDV M protein was compared with 3D models of the SARS-CoV-2 M protein created using neural networks and with initial machine learning-based models created using trRosetta. The 3D model of the present study predicted four linear B-cell epitopes (RSVNASSGTG and KHGDYSAVSNPSALT peptides are noteworthy), six discontinuous B-cell epitopes, forty weak binding and fourteen strong binding T-cell epitopes in the CV777 M protein. A high degree of conservation of the epitopes predicted in the PEDV M protein was observed among different PEDV strains isolated in different countries. The data suggest that the M protein could be a potential candidate for the development of new treatments or strategies that activate protective cellular mechanisms against viral diseases.


Subject(s)
Coronavirus Infections/virology , Coronavirus M Proteins/chemistry , Porcine epidemic diarrhea virus/chemistry , Swine Diseases/virology , Swine/virology , Amino Acid Sequence , Animals , Coronavirus Infections/immunology , Coronavirus Infections/veterinary , Coronavirus M Proteins/immunology , Epitopes, B-Lymphocyte/chemistry , Epitopes, B-Lymphocyte/immunology , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/immunology , Models, Molecular , Porcine epidemic diarrhea virus/immunology , Protein Conformation , Swine Diseases/immunology
3.
J Virol ; 96(1): e0137221, 2022 01 12.
Article in English | MEDLINE | ID: mdl-34643430

ABSTRACT

Coronaviral papain-like proteases (PLpros) are essential enzymes that mediate not only the proteolytic processes of viral polyproteins during virus replication but also the deubiquitination and deISGylation of cellular proteins that attenuate host innate immune responses. Therefore, PLpros are attractive targets for antiviral drug development. Here, we report the crystal structure of papain-like protease 2 (PLP2) of porcine epidemic diarrhea virus (PEDV) in complex with ubiquitin (Ub). The X-ray structural analyses reveal that PEDV PLP2 interacts with the Ub substrate mainly through the Ub core region and C-terminal tail. Mutations of Ub-interacting residues resulted in a moderately or completely abolished deubiquitinylating function of PEDV PLP2. In addition, our analyses also indicate that 2-residue-extended blocking loop 2 at the S4 subsite contributes to the substrate selectivity and binding affinity of PEDV PLP2. Furthermore, the PEDV PLP2 Glu99 residue, conserved in alphacoronavirus PLpros, was found to govern the preference of a positively charged P4 residue of peptidyl substrates. Collectively, our data provided structure-based information for the substrate binding and selectivity of PEDV PLP2. These findings may help us gain insights into the deubiquitinating (DUB) and proteolytic functions of PEDV PLP2 from a structural perspective. IMPORTANCE Current challenges in coronaviruses (CoVs) include a comprehensive understanding of the mechanistic effects of associated enzymes, including the 3C-like and papain-like proteases. We have previously reported that the PEDV PLP2 exhibits a broader substrate preference, superior DUB function, and inferior peptidase activity. However, the structural basis for these functions remains largely unclear. Here, we show the high-resolution X-ray crystal structure of PEDV PLP2 in complex with Ub. Integrated structural and biochemical analyses revealed that (i) three Ub core-interacting residues are essential for DUB function, (ii) 2-residue-elongated blocking loop 2 regulates substrate selectivity, and (iii) a conserved glutamate residue governs the substrate specificity of PEDV PLP2. Collectively, our findings provide not only structural insights into the catalytic mechanism of PEDV PLP2 but also a model for developing antiviral strategies.


Subject(s)
Coronavirus Papain-Like Proteases/chemistry , Porcine epidemic diarrhea virus/chemistry , Coronavirus/chemistry , Coronavirus/classification , Coronavirus/enzymology , Coronavirus Papain-Like Proteases/genetics , Coronavirus Papain-Like Proteases/metabolism , Crystallography, X-Ray , Mutation , Porcine epidemic diarrhea virus/enzymology , Porcine epidemic diarrhea virus/genetics , Protein Binding , Protein Domains , Structure-Activity Relationship , Substrate Specificity , Ubiquitin/chemistry , Ubiquitin/metabolism
4.
Vet Microbiol ; 254: 108994, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33486326

ABSTRACT

Porcine epidemic diarrhea virus (PEDV) encodes many multifunctional proteins that inhibit host innate immune response during virus infection. As one of important structural proteins, PEDV E protein has been found to block the production of type I interferon (IFN) in virus life cycle, but little is known about this process that E protein subverts host innate immune. Thus, in this present study, we initiated the construction of eukaryotic expression vectors to express PEDV E protein. Subsequently, cellular localization analysis was performed and the results showed that the majority of PEDV E protein distributed at cytoplasm and localized in endoplasmic reticulum (ER). Over-expression of PEDV E protein significantly inhibited poly(I:C)-induced IFN-ß and IFN-stimulated genes (ISGs) productions. We also found that PEDV E protein remarkably suppressed the protein expression of RIG-I signaling-associated molecules, but all their corresponding mRNA levels remained unaffected and unchanged. Furthermore, PEDV E protein obviously interfered with the translocation of IRF3 from cytoplasm to nucleus through direct interaction with IRF3, which is crucial for the IFN-ß production induced by poly(I:C). Taken together, our results suggested that PEDV E protein acts as an IFN-ß antagonist through suppression of the RIG-I-mediated signaling. This study will pave the way for the further investigation into the molecular mechanisms by which PEDV E protein evades host innate immune response.


Subject(s)
DEAD Box Protein 58/metabolism , Host-Pathogen Interactions/immunology , Interferon-beta/immunology , Porcine epidemic diarrhea virus/immunology , Receptors, Immunologic/metabolism , Signal Transduction , Viral Proteins/genetics , Animals , DEAD Box Protein 58/genetics , DEAD Box Protein 58/immunology , HEK293 Cells , HeLa Cells , Host-Pathogen Interactions/genetics , Humans , Immune Evasion , Immunity, Innate , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/immunology , Interferon Regulatory Factor-3/metabolism , Interferon-beta/antagonists & inhibitors , Interferon-beta/biosynthesis , Interferon-beta/genetics , Poly I-C/pharmacology , Porcine epidemic diarrhea virus/chemistry , Porcine epidemic diarrhea virus/drug effects , Porcine epidemic diarrhea virus/genetics , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Swine , Viral Proteins/metabolism
5.
Structure ; 29(4): 385-392.e5, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33378641

ABSTRACT

Porcine epidemic diarrhea virus (PEDV) is an alphacoronavirus responsible for significant morbidity and mortality in pigs. A key determinant of viral tropism and entry, the PEDV spike protein is a key target for the host antibody response and a good candidate for a protein-based vaccine immunogen. We used electron microscopy to evaluate the PEDV spike structure, as well as pig polyclonal antibody responses to viral infection. The structure of the PEDV spike reveals a configuration similar to that of HuCoV-NL63. Several PEDV protein-protein interfaces are mediated by non-protein components, including a glycan at Asn264 and two bound palmitoleic acid molecules. The polyclonal antibody response to PEDV infection shows a dominance of epitopes in the S1 region. This structural and immune characterization provides insights into coronavirus spike stability determinants and explores the immune landscape of viral spike proteins.


Subject(s)
Antibodies, Viral/metabolism , Coronavirus Infections/immunology , Epitopes/immunology , Porcine epidemic diarrhea virus/immunology , Spike Glycoprotein, Coronavirus/chemistry , Animals , Cell Line , Cryoelectron Microscopy , Fatty Acids, Monounsaturated/chemistry , Models, Molecular , Molecular Conformation , Polysaccharides/chemistry , Porcine epidemic diarrhea virus/chemistry , Porcine epidemic diarrhea virus/metabolism , Protein Binding , Sf9 Cells , Spike Glycoprotein, Coronavirus/immunology , Swine
6.
Virol J ; 17(1): 46, 2020 04 03.
Article in English | MEDLINE | ID: mdl-32245493

ABSTRACT

BACKGROUND: Porcine epidemic diarrhea virus (PEDV) infection causes an acute enteric tract infectious disease characterized by vomiting, anorexia, dehydration, weight loss and high mortality in neonatal piglets. During PEDV infection, the spike protein (S) is a major virion structural protein interacting with receptors and inducing neutralizing antibodies. However, the neutralizing B-cell epitopes within PEDV S protein have not been well studied. METHODS: To accurately identify the important immunodominant region of S1, the purified truncated S1 proteins (SA, SB, SC, SD and SE) were used to immunize BALB/c mice to prepare polyclonal antibodies. The antisera titers were determined by indirect ELISA, western blot and IFA after four immunizations to find the important immunodominant region of S1, and then purified the immunodominant region of S1 protein and immunized mice to generate the special antibodies, and then used recombinant peptides to determine the B-cell epitopes of monoclonal antibodies. RESULTS: Five antisera of recombinant proteins of the spike protein region of PEDV were generated and we found that only the polyclonal antibody against part of the S1 region (signed as SE protein, residues 666-789) could recognize the native PEDV. Purified SE protein was used to immunize BALB/c mice and generate mAb 2E10. Pepscan of the SE protein demonstrated that SE16 (722SSTFNSTREL731) is the minimal linear epitope required for reactivity with the mAb 2E10. Further investigation indicated that the epitope SE16 was localized on the surface of PEDV S protein in the 3D structure. CONCLUSIONS: A mAb 2E10 that is specifically bound to PEDV was generated and identified a specific linear B-cell epitope (SE16, 722SSTFNSTREL731) of the mAb. The epitope region of PEDV S1 localized in the different regions in comparison with the earlier identified epitopes. These findings enhance the understanding of the PEDV spike protein structure for vaccine design and provide a potential use for developing diagnostic methods to detect PEDV.


Subject(s)
Epitopes, B-Lymphocyte/immunology , Immunodominant Epitopes/immunology , Porcine epidemic diarrhea virus/immunology , Spike Glycoprotein, Coronavirus/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Chlorocebus aethiops , Female , Mice , Mice, Inbred BALB C , Porcine epidemic diarrhea virus/chemistry , Vero Cells
7.
Res Vet Sci ; 127: 42-46, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31677415

ABSTRACT

The ORF3 protein of porcine epidemic diarrhea virus (PEDV) is found to function as an ion channel which influences virus virulence and production. Taking consideration of the importance of PEDV orf3 gene, we have performed comprehensive analysis to investigate its synonymous codon usage patterns. In this study, the results of base composition analysis showed A/T rich and G/C poor in PEDV orf3 genes, and the most abundant base was nucleotide T. The relative synonymous codon usage value in each codon revealed that codon usage bias existed. The mean ENC value of each gene was 48.75, indicating a low codon usage bias, as well as a relatively instable change in PEDV orf3 genes. The general correlation analysis between base composition and codon usage bias indicated that mutational bias has an impact on the PEDV codon usage bias. Neutral analysis suggested that natural selection pressure takes a more important influence than mutational bias in shaping codon usage bias. Moreover, other factors including hydrophobicity and aromaticity have been also found to influence the codon usage variation among the PEDV orf3 genes. This study not only represents the most systematic analysis of codon usage patterns in PEDV orf3 genes, but also provides a basic shaping mechanism of the codon usage bias.


Subject(s)
Codon Usage , Porcine epidemic diarrhea virus/chemistry , Porcine epidemic diarrhea virus/genetics , Viral Proteins/chemistry , Viral Proteins/genetics , Base Composition , China
8.
Acta Virol ; 63(1): 88-95, 2019.
Article in English | MEDLINE | ID: mdl-30879317

ABSTRACT

The main immunogenic protein of the porcine epidemic diarrhea virus (PEDV) is the spike protein (S protein), which plays an important role in receptor binding, membrane fusion, and viral invasion of the host. In this paper, the linear epitope of the 3F10 non-neutralizing monoclonal antibody that was previously prepared in our laboratory was identified. The expression of truncated forms of the S protein for 3F10 reaction studies proved that the epitope was located between amino acids (aa) 674 and 791. To further locate the core aa of the 3F10 epitope, 12 random peptide libraries were used, and the result showed that the key aa located at aa 685-688 of the S protein (LLAF) were recognized by 3F10. Homology analysis of the regions corresponding to 20 typical strains of different PEDV subtypes showed that the epitope is highly conserved. Identifying the epitope recognized by an antibody helps to improve our understanding of the structure and function of the antigen. Keywords: spike protein; linear epitope; homology analysis.


Subject(s)
Epitopes , Porcine epidemic diarrhea virus , Spike Glycoprotein, Coronavirus , Swine Diseases , Animals , Antibodies/metabolism , Coronavirus Infections/virology , Epitopes/chemistry , Epitopes/genetics , Peptide Library , Porcine epidemic diarrhea virus/chemistry , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/genetics , Swine , Swine Diseases/virology
9.
Int J Biol Macromol ; 117: 704-712, 2018 Oct 01.
Article in English | MEDLINE | ID: mdl-29802920

ABSTRACT

Porcine epidemic diarrhea (PED) has caused huge economic losses to the global pork industry. Infection by its causative agent PED virus (PEDV), an Alpha-coronavirus, was previously proven to be mediated by its spike (S) glycoprotein and a cellular receptor porcine aminopeptidase N (pAPN). Interestingly, some recent studies have indicated that pAPN is not a functional receptor for PEDV. To date, there is a lack of a direct evidence for the interaction between pAPN and PEDV S protein in vitro. Here, we prepared pAPN ectodomain and the truncated variants of PEDV S protein in Drosophila S2 cells. These recombinant proteins were homogeneous after purification by metal-affinity and size-exclusion chromatography. We then assayed the purified target proteins through immunogenicity tests, PEDV binding interference assays, circular dichroism (CD) measurements, pAPN activity assay and structural determination, demonstrating that they were biologically functional. Finally, we characterized their interactions by gel filtration chromatography, native-polyacrylamide gel electrophoresis (PAGE) and surface plasmon resonance (SPR) analyses. The results showed that their affinities were too low to form complexes, which suggest that pAPN may be controversial as the genuine receptor for PEDV. Therefore, further research needs to be carried out to elucidate the interaction between PEDV and its genuine receptor.


Subject(s)
CD13 Antigens/chemistry , Coronavirus Infections/genetics , Porcine epidemic diarrhea virus/genetics , Spike Glycoprotein, Coronavirus/genetics , Animals , CD13 Antigens/genetics , Chlorocebus aethiops , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Host-Pathogen Interactions/genetics , Porcine epidemic diarrhea virus/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Spike Glycoprotein, Coronavirus/chemistry , Swine , Vero Cells
10.
Microb Cell Fact ; 17(1): 20, 2018 Feb 09.
Article in English | MEDLINE | ID: mdl-29426335

ABSTRACT

BACKGROUND: Porcine epidemic diarrhea caused by porcine epidemic diarrhea virus (PEDV) has led to serious economic losses to the swine industry worldwide. In this study, an oral recombinant Lactobacillus casei vaccine against PEDV infection targeting the intestinal microfold (M) cells and dendritic cells (DCs) for delivering the core neutralizing epitope (COE) of PEDV spike protein was developed with M cell-targeting peptide (Col) and dendritic cell-targeting peptide (DCpep). The immunogenicity of the orally administered recombinant strains was evaluated. RESULTS: After immunization, significantly higher levels of anti-PEDV specific IgG antibodies with PEDV neutralizing activity in the sera and mucosal sIgA antibodies in the tractus genitalis, intestinal mucus, and stools were detected in mice orally administered with the recombinant strain pPG-COE-Col-DCpep/L393, which expressed DCpep and Col targeting ligands fused with the PEDV COE antigen, compared to mice orally immunized with the recombinant strain pPG-COE/L393 without the DCpep and Col targeting ligands. Moreover, in response to restimulation with the PEDV COE antigen in vitro, a significant difference in splenocyte proliferation response and Th2-associated cytokine IL-4 level was observed in the group of mice orally immunized with pPG-COE-Col-DCpep/L393 (p < 0.05) compared to the groups of mice that received pPG-COE-Col/L393 and pPG-COE-DCpep/L393, respectively. CONCLUSIONS: The intestinal M cells- and DCs-targeting oral delivery of genetically engineered Lactobacillus expressing the COE antigen of PEDV can efficiently induce anti-PEDV mucosal, humoral, and cellular immune responses via oral administration, suggesting a promising vaccine strategy against PEDV infection.


Subject(s)
Coronavirus Infections/veterinary , Dendritic Cells/immunology , Intestines/immunology , Lactobacillus/genetics , Porcine epidemic diarrhea virus/immunology , Viral Vaccines/immunology , Administration, Oral , Animals , Antibodies, Viral/blood , Coronavirus Infections/immunology , Coronavirus Infections/prevention & control , Epitopes/chemistry , Epitopes/immunology , Immunoglobulin G/blood , Intestines/cytology , Lactobacillus/immunology , Mice , Porcine epidemic diarrhea virus/chemistry , Porcine epidemic diarrhea virus/genetics , Spike Glycoprotein, Coronavirus/administration & dosage , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology , Swine , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics
11.
J Virol ; 92(5)2018 03 01.
Article in English | MEDLINE | ID: mdl-29237834

ABSTRACT

Porcine epidemic diarrhea virus (PEDV), an enteropathogenic Alphacoronavirus, has caused enormous economic losses in the pork industry. Nonstructural protein 1 (nsp1) is a characteristic feature of alpha- and betacoronaviruses, which exhibits both functional conservation and mechanistic diversity in inhibiting host gene expression and antiviral responses. However, the detailed structure and molecular mechanisms underlying the Alphacoronavirus nsp1 inhibition of host gene expression remain unclear. Here, we report the first full-length crystal structure of Alphacoronavirus nsp1 from PEDV. The structure displays a six-stranded ß-barrel fold in the middle of two α-helices. The core structure of PEDV nsp1 shows high similarity to those of severe acute respiratory syndrome coronavirus (SARS-CoV) nsp1 and transmissible gastroenteritis virus (TGEV) nsp1, despite its low degree of sequence homology. Using ribopuromycylation and Renilla luciferase reporter assays, we showed that PEDV nsp1 can dramatically inhibit general host gene expression. Furthermore, three motifs (amino acids [aa] 67 to 71, 78 to 85, and 103 to 110) of PEDV nsp1 create a stable functional region for inhibiting protein synthesis, differing considerably from Betacoronavirus nsp1. These results elucidate the detailed structural basis through which PEDV nsp1 inhibits host gene expression, providing insight into the development of a new attenuated vaccine with nsp1 modifications.IMPORTANCE Porcine epidemic diarrhea virus (PEDV) has led to tremendous economic losses in the global swine industry. PEDV nsp1 plays a crucial role in inhibiting host gene expression, but its functional mechanism remains unclear. Here, we report the full-length structure of PEDV nsp1, the first among coronaviruses to be reported. The 1.25-Å resolution crystal structure of PEDV nsp1 shows high similarity to severe acute respiratory syndrome coronavirus (SARS-CoV) nsp113-128 and transmissible gastroenteritis virus (TGEV) nsp11-104, despite a lack of sequence homology. Structural and biochemical characterization demonstrated that PEDV nsp1 possesses a stable functional region for inhibition of host protein synthesis, which is formed by loops at residues 67 to 71, 78 to 85, and 103 to 110. The different functional regions in PEDV nsp1 and SARS-CoV nsp1 may explain their distinct mechanisms. Importantly, our structural data are conducive to understanding the mechanism of PEDV nsp1 inhibition of the expression of host genes and may aid in the development of a new attenuated vaccine.


Subject(s)
Gene Expression Regulation , Host-Pathogen Interactions , Porcine epidemic diarrhea virus/chemistry , Viral Nonstructural Proteins/chemistry , Amino Acid Sequence , Animals , Cell Line , Coronavirus/genetics , Coronavirus Infections/prevention & control , Coronavirus Infections/virology , Crystallography, X-Ray , HEK293 Cells , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/physiology , Humans , Models, Molecular , Porcine epidemic diarrhea virus/genetics , Protein Folding , Protein Structure, Tertiary , RNA-Dependent RNA Polymerase , Severe acute respiratory syndrome-related coronavirus/chemistry , Severe acute respiratory syndrome-related coronavirus/genetics , Sequence Alignment , Sequence Homology , Swine , Swine Diseases/prevention & control , Swine Diseases/virology , Transmissible gastroenteritis virus/chemistry , Transmissible gastroenteritis virus/genetics , Vaccines, Attenuated , Viral Nonstructural Proteins/classification , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism
12.
J Virol ; 91(12)2017 06 15.
Article in English | MEDLINE | ID: mdl-28381581

ABSTRACT

Porcine epidemic diarrhea virus (PEDV) causes enteric disease in pigs, resulting in significant economic losses to the swine industry worldwide. Current vaccination approaches against this emerging coronavirus are only partially effective, though natural infection protects pigs against reinfection and provides lactogenic immunity to suckling piglets. The viral spike (S) glycoprotein, responsible for receptor binding and cell entry, is the major target for neutralizing antibodies. However, knowledge of antibody epitopes, their nature and location in the spike structure, and the mechanisms by which the antibodies interfere with infection is scarce. Here we describe the generation and characterization of 10 neutralizing and nonneutralizing mouse monoclonal antibodies raised against the S1 receptor binding subunit of the S protein. By expression of different S1 protein fragments, six antibody epitope classes distributed over the five structural domains of the S1 subunit were identified. Characterization of antibodies for cross-reactivity and cross-neutralization revealed antigenic differences among PEDV strains. The epitopes of potent neutralizing antibodies segregated into two epitope classes and mapped within the N-terminal sialic acid binding domain and in the more C-terminal receptor binding domain. Antibody neutralization escape mutants displayed single amino acid substitutions that impaired antibody binding and neutralization and defined the locations of the epitopes. Our observations picture the antibody epitope landscape of the PEDV S1 subunit and reveal that its cell attachment domains are key targets of neutralizing antibodies.IMPORTANCE Porcine epidemic diarrhea virus (PEDV), an emerging porcine coronavirus, causes an economically important enteric disease in pigs. Effective PEDV vaccines for disease control are currently lacking. The spike (S) glycoprotein on the virion surface is the key player in virus cell entry and, therefore, the main target of neutralizing antibodies. To understand the antigenic landscape of the PEDV spike protein, we developed monoclonal antibodies against the spike protein's S1 receptor binding region and characterized their epitopes, neutralizing activity, and cross-reactivity toward multiple PEDV strains. Epitopes of antibodies segregated into six epitope classes dispersed over the multidomain S1 structure. Monoclonal antibodies revealed antigenic variability in B-cell epitopes between PEDV strains. The epitopes of neutralizing antibodies mapped to two distinct domains in S1 that are involved in binding to carbohydrate and proteinaceous cell surface molecules, respectively, indicating the importance of these cell attachment sites on the PEDV spike protein in eliciting a protective humoral immune response.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Porcine epidemic diarrhea virus/chemistry , Porcine epidemic diarrhea virus/immunology , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/immunology , Amino Acid Substitution , Animals , Antibodies, Neutralizing/isolation & purification , Antibodies, Neutralizing/metabolism , Antibody Affinity , Epitopes, B-Lymphocyte/chemistry , Epitopes, B-Lymphocyte/classification , Epitopes, B-Lymphocyte/immunology , Mice , Mutation , Neutralization Tests , Porcine epidemic diarrhea virus/genetics , Porcine epidemic diarrhea virus/physiology , Spike Glycoprotein, Coronavirus/genetics , Swine
13.
J Virol ; 91(10)2017 05 15.
Article in English | MEDLINE | ID: mdl-28250121

ABSTRACT

Porcine deltacoronavirus (PDCoV) is an emerging swine enteropathogenic coronavirus. The first outbreak of PDCoV was announced from the United States in 2014, followed by reports in Asia. The nonstructural protein nsp5 is a 3C-like protease of coronavirus, and our previous study showed that PDCoV nsp5 inhibits type I interferon (IFN) production. In this study, we found that PDCoV nsp5 significantly inhibited IFN-stimulated response element (ISRE) promoter activity and transcription of IFN-stimulated genes (ISGs), suggesting that PDCoV nsp5 also suppresses IFN signaling. Detailed analysis showed that nsp5 cleaved signal transducer and activator of transcription 2 (STAT2) but not Janus kinase 1 (JAK1), tyrosine kinase 2 (TYK2), STAT1, and interferon regulatory factor 9 (IRF9), key molecules of the JAK-STAT pathway. STAT2 cleavage was dependent on the protease activity of nsp5. Interestingly, nsp5 cleaved STAT2 at two sites, glutamine 685 (Q685) and Q758, and similar cleavage was observed in PDCoV-infected cells. As expected, cleaved STAT2 impaired the ability to induce ISGs, demonstrating that STAT2 cleavage is an important mechanism utilized by PDCoV nsp5 to antagonize IFN signaling. We also discussed the substrate selection and binding mode of PDCoV nsp5 by homologous modeling of PDCoV nsp5 with the two cleaved peptide substrates. The results of our study demonstrate that PDCoV nsp5 antagonizes type I IFN signaling by cleaving STAT2 and provides structural insights for comprehending the cleavage mechanism of PDCoV nsp5, revealing a potential new function for PDCoV nsp5 in type I IFN signaling.IMPORTANCE The 3C-like protease encoded by nsp5 is a major protease of coronaviruses; thus, it is an attractive target for development of anticoronavirus drugs. Previous studies have revealed that the 3C-like protease of coronaviruses, including PDCoV and porcine epidemic diarrhea virus (PEDV), antagonizes type I IFN production by targeting the NF-κB essential modulator (NEMO). Here, for the first time, we demonstrate that overexpression of PDCoV nsp5 also antagonizes IFN signaling by cleaving STAT2, an essential component of transcription factor complex ISGF3, and that PDCoV infection reduces the levels of STAT2, which may affect the innate immune response.


Subject(s)
Coronavirus/chemistry , Interferon Type I/metabolism , Porcine epidemic diarrhea virus/chemistry , STAT2 Transcription Factor/metabolism , Signal Transduction , Viral Proteins/metabolism , Animals , Coronavirus/genetics , Coronavirus/physiology , Coronavirus Infections , HEK293 Cells , Host-Pathogen Interactions , Humans , Immunity, Innate , Porcine epidemic diarrhea virus/genetics , Porcine epidemic diarrhea virus/physiology , Sequence Alignment , Swine , Viral Proteins/genetics , Viral Proteins/isolation & purification
14.
Protein Expr Purif ; 126: 77-83, 2016 10.
Article in English | MEDLINE | ID: mdl-27260969

ABSTRACT

The emergence of highly pathogenic variant porcine epidemic diarrhea virus (PEDV) strains, from 2013 to 2014, in North American and Asian countries have greatly threatened global swine industry. Therefore, development of effective vaccines against PEDV variant strains is urgently needed. Recently, it has been reported that the N-terminal domain (NTD) of S1 domain of PEDV spike protein is responsible for binding to the 5-N-acetylneuraminic acid (Neu5Ac), a possible sugar co-receptor. Therefore, the NTD of S1 domain could be an attractive target for the development of subunit vaccines. In this study, the NTD spanning amino acid residues 25-229 (S25-229) of S1 domain of PEDV variant strain was expressed in Escherichia coli BL21 (DE3) in the form of inclusion bodies (IBs). S25-229 IBs were solubilized in 20 mM sodium acetate (pH 4.5) buffer containing 8 M urea and 1 mM dithiothreitol with 95% yield. Solubilized S25-229 IBs were refolded by 10-fold flash dilution and purified by one-step cation exchange chromatography with >95% purity and 20% yield. The CD spectrum of S25-229 showed the characteristic pattern of alpha helical structure. In an indirect ELISA, purified S25-229 showed strong reactivity with mouse anti-PEDV sera. In addition, immunization of mice with 20 µg of purified S25-229 elicited highly potent serum IgG titers. Finally, mouse antisera against S25-229 showed immune reactivity with native PEDV S protein in an immunofluorescence assay. These results suggest that purified S25-229 may have potential to be used as a subunit vaccine against PEDV variant strains.


Subject(s)
Inclusion Bodies , Porcine epidemic diarrhea virus , Spike Glycoprotein, Coronavirus , Viral Vaccines , Animals , Chlorocebus aethiops , Immunization , Inclusion Bodies/chemistry , Inclusion Bodies/genetics , Inclusion Bodies/immunology , Inclusion Bodies/metabolism , Mice , Porcine epidemic diarrhea virus/chemistry , Porcine epidemic diarrhea virus/genetics , Porcine epidemic diarrhea virus/immunology , Solubility , Spike Glycoprotein, Coronavirus/biosynthesis , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/isolation & purification , Swine , Vero Cells , Viral Vaccines/biosynthesis , Viral Vaccines/genetics , Viral Vaccines/immunology , Viral Vaccines/isolation & purification
15.
Sci Rep ; 6: 25961, 2016 05 13.
Article in English | MEDLINE | ID: mdl-27173881

ABSTRACT

Porcine epidemic diarrhea virus (PEDV) is a coronavirus that infects pigs and can have mortality rates approaching 100% in piglets, causing serious economic impact. The 3C-like protease (3CL(pro)) is essential for the coronaviral life cycle and is an appealing target for the development of therapeutics. We report the expression, purification, crystallization and 2.10 Å X-ray structure of 3CL(pro) from PEDV. Analysis of the PEDV 3CL(pro) structure and comparison to other coronaviral 3CL(pro)'s from the same alpha-coronavirus phylogeny shows that the overall structures and active site architectures across 3CL(pro)'s are conserved, with the exception of a loop that comprises the protease S2 pocket. We found a known inhibitor of severe acute respiratory syndrome coronavirus (SARS-CoV) 3CL(pro), (R)-16, to have inhibitor activity against PEDV 3CL(pro), despite that SARS-3CL(pro) and PEDV 3CL(pro) share only 45.4% sequence identity. Structural comparison reveals that the majority of residues involved in (R)-16 binding to SARS-3CL(pro) are conserved in PEDV-3CL(pro); however, the sequence variation and positional difference in the loop forming the S2 pocket may account for large observed difference in IC50 values. This work advances our understanding of the subtle, but important, differences in coronaviral 3CL(pro) architecture and contributes to the broader structural knowledge of coronaviral 3CL(pro)'s.


Subject(s)
Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/metabolism , Porcine epidemic diarrhea virus/enzymology , Viral Proteins/chemistry , Viral Proteins/metabolism , Animals , Catalytic Domain , Crystallography, X-Ray , Models, Molecular , Porcine epidemic diarrhea virus/chemistry , Protein Conformation , Structural Homology, Protein , Swine
16.
Arch Virol ; 161(8): 2189-95, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27224981

ABSTRACT

Porcine epidemic diarrhea virus (PEDV) is the etiological agent of porcine epidemic diarrhea (PED), which is threatening the swine industry all over the world. In Japan, although there were no reported PED cases from 2007 to 2012, a large-scale PED outbreak started in 2013, causing severe economic losses. Although several PEDV studies have been conducted in Japan, more PEDV isolates and sequence information are needed to understand the molecular biology and epidemiology of PEDV. Here, we isolated seven Japanese PEDV strains from intestinal tissue samples collected in 2014 and determined the spike gene sequences of 13 Japanese PEDV strains, including the above seven isolates. Phylogenetic analysis shows that all of the strains are genetically distinct from classical Japanese PEDV strains isolated prior to 2013 and can be classified into two different genotypes: 12 strains belong to the North American clade composed of recent highly pathogenic PEDV strains, and the remaining one strain belongs to the so-called insertion deletion (INDEL) clade. These data suggest multiple PEDV invasions from abroad to Japan. Notably, compared to classical Japanese strains, all of the recent Japanese strains have two amino acid substitutions in a known neutralizing epitope. In addition, one of the strains acquired an additional mutation in another neutralizing epitope that is highly conserved among PEDVs, including the classical and recent isolates. Our isolates and findings will be useful for future investigations aimed at understanding, controlling, and preventing PED.


Subject(s)
Coronavirus Infections/veterinary , Porcine epidemic diarrhea virus/genetics , Porcine epidemic diarrhea virus/isolation & purification , Swine Diseases/virology , Amino Acid Sequence , Animals , Coronavirus Infections/epidemiology , Coronavirus Infections/virology , Japan/epidemiology , Molecular Sequence Data , Phylogeny , Porcine epidemic diarrhea virus/chemistry , Porcine epidemic diarrhea virus/classification , Sequence Alignment , Swine/virology , Swine Diseases/epidemiology , Viral Proteins/chemistry , Viral Proteins/genetics
17.
J Virol ; 89(15): 8077-81, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25972540

ABSTRACT

The emerging porcine epidemic diarrhea virus (PEDV) requires trypsin supplementation to activate its S protein for membrane fusion and virus propagation in cell culture. By substitution of a single amino acid in the S protein, we created a recombinant PEDV with an artificial furin protease cleavage site N terminal of the putative fusion peptide (PEDV-SFCS). PEDV-SFCS exhibited trypsin-independent cell-cell fusion and was able to replicate in culture cells independently of trypsin, though to low titer.


Subject(s)
Coronavirus Infections/veterinary , Furin/metabolism , Point Mutation , Porcine epidemic diarrhea virus/genetics , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/metabolism , Swine Diseases/enzymology , Trypsin/metabolism , Virus Internalization , Amino Acid Motifs , Amino Acid Substitution , Animals , Coronavirus Infections/enzymology , Coronavirus Infections/virology , Porcine epidemic diarrhea virus/chemistry , Porcine epidemic diarrhea virus/physiology , Protein Processing, Post-Translational , Spike Glycoprotein, Coronavirus/chemistry , Swine , Swine Diseases/virology
18.
J Virol ; 89(6): 3332-42, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25589635

ABSTRACT

UNLABELLED: Porcine epidemic diarrhea virus (PEDV) and transmissible gastroenteritis virus (TGEV) are economically important swine enteropathogenic coronaviruses. These two viruses belong to two distinct species of the Alphacoronavirus genus within Coronaviridae and induce similar clinical signs and pathological lesions in newborn piglets, but they are presumed to be antigenically distinct. In the present study, two-way antigenic cross-reactivity examinations between the prototype PEDV CV777 strain, three distinct U.S. PEDV strains (the original highly virulent PC22A, S indel Iowa106, and S 197del PC177), and two representative U.S. TGEV strains (Miller and Purdue) were conducted by cell culture immunofluorescent (CCIF) and viral neutralization (VN) assays. None of the pig TGEV antisera neutralized PEDV and vice versa. One-way cross-reactions were observed by CCIF between TGEV Miller hyperimmune pig antisera and all PEDV strains. Enzyme-linked immunosorbent assays, immunoblotting using monoclonal antibodies and Escherichia coli-expressed recombinant PEDV and TGEV nucleocapsid (N) proteins, and sequence analysis suggested at least one epitope on the N-terminal region of PEDV/TGEV N protein that contributed to this cross-reactivity. Biologically, PEDV strain CV777 induced greater cell fusion in Vero cells than did U.S. PEDV strains. Consistent with the reported genetic differences, the results of CCIF and VN assays also revealed higher antigenic variation between PEDV CV777 and U.S. strains. IMPORTANCE: Evidence of antigenic cross-reactivity between porcine enteric coronaviruses, PEDV and TGEV, in CCIF assays supports the idea that these two species are evolutionarily related, but they are distinct species defined by VN assays. Identification of PEDV- or TGEV-specific antigenic regions allows the development of more specific immunoassays for each virus. Antigenic and biologic variations between the prototype and current PEDV strains could explain, at least partially, the recurrence of PEDV epidemics. Information on the conserved antigenicity among PEDV strains is important for the development of PEDV vaccines to protect swine from current highly virulent PEDV infections.


Subject(s)
Antibodies, Viral/immunology , Coronavirus Infections/veterinary , Porcine epidemic diarrhea virus/immunology , Swine Diseases/immunology , Transmissible gastroenteritis virus/immunology , Amino Acid Sequence , Animals , Antigenic Variation , Coronavirus Infections/immunology , Coronavirus Infections/virology , Cross Reactions , Gastroenteritis, Transmissible, of Swine/immunology , Gastroenteritis, Transmissible, of Swine/virology , Molecular Sequence Data , Porcine epidemic diarrhea virus/chemistry , Porcine epidemic diarrhea virus/classification , Porcine epidemic diarrhea virus/genetics , Sequence Alignment , Swine , Swine Diseases/virology , Transmissible gastroenteritis virus/chemistry , Transmissible gastroenteritis virus/classification , Transmissible gastroenteritis virus/genetics
19.
Arch Virol ; 159(11): 2977-87, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25008896

ABSTRACT

Porcine epidemic diarrhea virus (PEDV) is a highly contagious enteric pathogen of swine. Acute PEDV outbreaks have continually emerged in most swine-producing Asian countries and, recently, in the United States, causing significant economic losses in the pig industry. The spike (S) protein of PEDV is a type 1 transmembrane envelope glycoprotein and consists of the S1 and S2 domains, which are responsible for virus binding and fusion, respectively. Since the S1 domain is involved in a specific high-affinity interaction with the cellular receptor and induction of neutralizing antibody in the natural host, it is a primary target for the development of effective vaccines against PEDV. In this study, a codon-optimized PEDV S1 gene containing amino acid residues 25-738 was synthesized based on a multiple alignment of the S amino acid sequences of PEDV field isolates and used to establish a stable porcine cell line constitutively expressing the PEDV S1 protein. The purified recombinant S1 protein was found to mediate highly potent antibody responses in immunized rabbits. The antibodies strongly recognized the recombinant S1 protein from cell lysates and supernatants of S1-expressing cells, whereas they bound weakly to the authentic S protein of PEDV vaccine strain SM98-1. Furthermore, a serum neutralization test revealed that the rabbit antisera completely inhibit infection of the PEDV vaccine strain at a serum dilution of 1:16. We then tested the ability of vaccination with the recombinant S1 protein to protect piglets against PEDV. Late-term pregnant sows were inoculated intramuscularly with the purified S1 protein, and the outcome was investigated in passively immunized suckling piglets after a virulent PEDV challenge. The results showed that vaccination with S1 protein efficiently protected neonatal piglets against PEDV. Our data suggest that the recombinant S1 protein shows potential as an effective and safe subunit vaccine for PED prevention.


Subject(s)
Coronavirus Infections/veterinary , Porcine epidemic diarrhea virus/immunology , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/immunology , Swine Diseases/prevention & control , Animals , Antibodies, Viral/immunology , Coronavirus Infections/immunology , Coronavirus Infections/prevention & control , Coronavirus Infections/virology , Porcine epidemic diarrhea virus/chemistry , Porcine epidemic diarrhea virus/genetics , Protein Structure, Tertiary , Rabbits , Spike Glycoprotein, Coronavirus/administration & dosage , Spike Glycoprotein, Coronavirus/genetics , Swine , Swine Diseases/immunology , Swine Diseases/virology , Viral Vaccines/administration & dosage , Viral Vaccines/chemistry , Viral Vaccines/genetics , Viral Vaccines/immunology
20.
Article in English | MEDLINE | ID: mdl-23600505

ABSTRACT

The variable regions of the heavy chain (VH) and light chain (VL) were amplified by RT-PCR from the hybridoma 6E6, which secretes the monoclonal antibody against PEDV S protein. The VL and VH amplicons were combined using SOE-PCR by a 12 amino acid flexible linker (SSGGGGSGGGGS), which produced the scFv gene (named scFv/6E6). After sequence analysis, the scFv/6E6 gene was cloned into the prokaryotic expression vector pGEX-6p-1 with a GST-tag. The recombinant scFv/6E6 protein was successfully expressed in recombinant Escherichia coli by IPTG induction. Moreover, the recombinant scFv/6E6 protein was purified from the inclusion body form by the gel-cutting measure followed by electroelution and dialysis. The recombinant scFv/6E6 protein reported here will provide some basis for further antiviral drug research based on the scFv molecule.


Subject(s)
Antibodies, Monoclonal/immunology , Hybridomas/immunology , Membrane Glycoproteins/immunology , Porcine epidemic diarrhea virus/chemistry , Single-Chain Antibodies/metabolism , Viral Envelope Proteins/immunology , Blotting, Western , DNA Primers/genetics , Isopropyl Thiogalactoside , Oligopeptides/immunology , Porcine epidemic diarrhea virus/immunology , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Single-Chain Antibodies/genetics , Spike Glycoprotein, Coronavirus
SELECTION OF CITATIONS
SEARCH DETAIL
...