1
|
Wan H, Yang Y, Tu Z, Tang M, Jing B, Feng Y, Xie J, Gao H, Song X, Zhao X. Enhanced mucosal immune response through nanoparticle delivery system based on chitosan-catechol and a recombinant antigen targeted towards M cells. Int J Biol Macromol 2025; 306:141345. [PMID: 40010449 DOI: 10.1016/j.ijbiomac.2025.141345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 02/12/2025] [Accepted: 02/19/2025] [Indexed: 02/28/2025]
Abstract
In mucosal vaccination, the targeted delivery of antigens through M (microfold) cells is essential for initiating a robust antigen-specific immune response. In the present study, we devised a nano-delivery platform to target M cells. This platform involved coating mesoporous silica nanoparticles (MSN) with a mucoadhesive chitosan-catechol (Chic) layer, incorporating a recombinant antigen to form nanoparticles that enhance the immune response. The collagenase equivalent domain (COE) of porcine epidemic diarrhea virus (PEDV) terminated with the M cell-targeting sequence RGD (COER), was initially expressed by Escherichia coli (E. coli) and subsequently conjugated to the surface of MSN-Chic, forming the MSN-Chic-COER nanoparticles. MSN-Chic-COER with strong mucoadhesive properties and a propensity for M cell targeting, demonstrated enhanced uptake by dendritic cells (DCs) and trafficking to lymph nodes, compared to COE/COER after intranasal administration. MSN-Chic-COER recruited more dendritic cells to the antigen-located site via stimulating chemokine CCL20 secretion was evidenced by cell co-culture model. Additionally, it enhanced antigen permeability by disrupting the distribution of the ZO-1 protein in epithelial cells. Notably, MSN-Chic-COER elicited a higher level of cellular immunity, humoral immunity, and PEDV neutralizing antibody production. These findings underscore the potential of MSN-Chic-COER as a promising intranasal vaccine delivery system.
Collapse
Affiliation(s)
- Hongping Wan
- Center for Infectious Diseases Control (CIDC), Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Center for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu 611130, China.
| | - Yunhan Yang
- Center for Infectious Diseases Control (CIDC), Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Center for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu 611130, China
| | - Zhiwen Tu
- Center for Infectious Diseases Control (CIDC), Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Center for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu 611130, China
| | - Mingrun Tang
- Center for Infectious Diseases Control (CIDC), Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Center for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu 611130, China
| | - Bo Jing
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Yumei Feng
- Instrumental analysis center, Sichuan Agricultural University, Chengdu 611130, China
| | - Jiulong Xie
- Instrumental analysis center, Sichuan Agricultural University, Chengdu 611130, China
| | - Huile Gao
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu 610041, China.
| | - Xu Song
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Xinghong Zhao
- Center for Infectious Diseases Control (CIDC), Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Center for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
2
|
Chen R, Zhou G, Yang J, Yuan R, Sun Y, Liang Y, Wu R, Wen Y, Wang Y, Zhao Q, Du S, Yan Q, Cao S, Huang X. A novel neutralizing antibody recognizing a conserved conformational epitope in PDCoV S1 protein and its therapeutic efficacy in piglets. J Virol 2025; 99:e0202524. [PMID: 39840987 PMCID: PMC11853068 DOI: 10.1128/jvi.02025-24] [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: 11/14/2024] [Accepted: 12/11/2024] [Indexed: 01/23/2025] Open
Abstract
Porcine deltacoronavirus (PDCoV) is an enteric pathogen that burdens the global pig industry and is a public health concern. The development of effective antiviral therapies is necessary for the prevention and control of PDCoV, yet to date, there are few studies on the therapeutic potential of PDCoV-neutralizing antibodies. Here, we investigate the therapeutic potential of a novel monoclonal antibody (mAb 4A6) which targets the PDCoV S1 protein and effectively neutralizes PDCoV, both pre- and post-attachment on cells, with IC50 values of 0.537 and 8.487 µg/mL, respectively. A phage-display peptide library was used to determine the epitope recognized by mAb 4A6, and two mimotopes, QYPVSYA (P1) and FPHWPTI (P2), were identified. KLH-P1 reacted with PDCoV-positive sera but failed to induce PDCoV-specific IgG and neutralizing antibodies in mice, suggesting P1 does not fully mimic the conformational epitope. Molecular docking and alanine scanning mutagenesis revealed that S461, P462, T463, E465, and Y467 on the S protein are essential for mAb 4A6 binding. Antibody therapy experiments in PDCoV-infected piglets showed that administering mAb 4A6 once or twice could delay the onset of diarrhea symptoms, reduce the severity of diarrhea, and decrease virus shedding. Taken together, our findings demonstrate that mAb 4A6 holds promise as a treatment against PDCoV, and the amino acids recognized by mAb 4A6 will be valuable for developing novel epitope-based vaccines or antiviral drugs. IMPORTANCE Porcine deltacoronavirus (PDCoV) is a novel swine enteropathogenic coronavirus that poses a potential threat to public health. Developing effective antiviral therapies is crucial for its prevention and control. Here, we demonstrated that mAb 4A6 shows promise as a treatment against PDCoV. Antibody therapy experiments conducted on PDCoV-infected piglets revealed that administering mAb 4A6 once or twice could delay the onset of diarrhea symptoms, reduce the severity of diarrhea, and decrease virus shedding. Furthermore, we characterized the conformational epitope (S461, P462, T463, E465, and Y467) recognized by mAb 4A6 through an integrated approach involving phage display peptide library, molecular docking, and alanine scanning mutagenesis. More importantly, mAb 4A6 exhibits a broad-spectrum neutralizing activity against different PDCoV strains. These findings indicate that mAb 4A6 has promising therapeutic value for PDCoV-infected piglets, and the identification of mAb 4A6 recognized epitope may provide a new idea for the identification of conformational epitopes.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/therapeutic use
- Swine
- Epitopes/immunology
- Epitopes/chemistry
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Viral/immunology
- Antibodies, Viral/therapeutic use
- Swine Diseases/virology
- Swine Diseases/immunology
- Swine Diseases/therapy
- Swine Diseases/drug therapy
- Mice
- Molecular Docking Simulation
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/chemistry
- Spike Glycoprotein, Coronavirus/genetics
- Deltacoronavirus/immunology
- Peptide Library
- Protein Conformation
- Mice, Inbred BALB C
Collapse
Affiliation(s)
- Rui Chen
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Guiping Zhou
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Junpeng Yang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Rong Yuan
- Chengdu Livestock and Poultry Genetic Resources Protection Center, Chengdu, China
| | - Ying Sun
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yixiao Liang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Rui Wu
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yiping Wen
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yiping Wang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Qin Zhao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Senyan Du
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Qigui Yan
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Sanjie Cao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
| | - Xiaobo Huang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, China
| |
Collapse
|
3
|
Yi J, Luo H, Zhang K, Lv L, Li S, Jiang Y, Zhou Y, Wei Z, Liu C. The Pseudotyped Replication-Deficient VSV with Spike from PEDV Induces Neutralizing Antibody Against PEDV. Vaccines (Basel) 2025; 13:223. [PMID: 40266086 PMCID: PMC11946067 DOI: 10.3390/vaccines13030223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Porcine epidemic diarrhea virus (PEDV) is a significant pathogen in swine, causing substantial economic losses worldwide. Despite the availability of existing vaccines, there is a critical need for novel vaccine platforms that ensure robust protection while maintaining safety. METHODS A recombinant replication-deficient vesicular stomatitis virus (VSV) vaccine, rVSV∆G-PEDV-S, was developed by pseudotyping the virus with the spike (S) protein from PEDV. To achieve high-titer pseudotyped rVSV particles, a stable Huh7 cell line expressing the PEDV S protein (Huh7-PEDV-S) was generated. The infectivity and replication capacity of rVSV∆G-PEDV-S were evaluated in PEDV-susceptible cell lines and Huh7-PEDV-S cells. The vaccine's immunogenicity and safety were assessed in BALB/c mice vaccinated intramuscularly with rVSV∆G-PEDV-S. RESULTS The pseudotyped rVSV∆G-PEDV-S demonstrated infectivity in PEDV-susceptible cell lines and robust replication in Huh7-PEDV-S cells, while remaining replication-deficient in non-complementary cells. In vaccinated BALB/c mice, the vaccine elicited a strong humoral immune response, characterized by high levels of PEDV S1-specific IgG and neutralizing antibodies. No adverse effects, including weight loss or behavioral changes, were observed in the vaccinated mice, confirming the vaccine's safety. CONCLUSIONS The rVSV∆G-PEDV-S vaccine represents a promising platform for controlling PEDV outbreaks. Its replication-deficient design and pseudotyping methodology ensure safety and adaptability to emerging PEDV variants. These findings highlight the potential of rVSV∆G-PEDV-S as a safe and effective solution to the ongoing challenges posed by PEDV.
Collapse
Affiliation(s)
- Jingxuan Yi
- College of Animal Science and Technology, Guangxi University, Nanning 530005, China; (J.Y.); (K.Z.)
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; (H.L.); (L.L.); (S.L.); (Y.J.); (Y.Z.)
| | - Huaye Luo
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; (H.L.); (L.L.); (S.L.); (Y.J.); (Y.Z.)
| | - Kang Zhang
- College of Animal Science and Technology, Guangxi University, Nanning 530005, China; (J.Y.); (K.Z.)
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; (H.L.); (L.L.); (S.L.); (Y.J.); (Y.Z.)
| | - Lilei Lv
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; (H.L.); (L.L.); (S.L.); (Y.J.); (Y.Z.)
| | - Siqi Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; (H.L.); (L.L.); (S.L.); (Y.J.); (Y.Z.)
| | - Yifeng Jiang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; (H.L.); (L.L.); (S.L.); (Y.J.); (Y.Z.)
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou 225009, China
| | - Yanjun Zhou
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; (H.L.); (L.L.); (S.L.); (Y.J.); (Y.Z.)
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou 225009, China
| | - Zuzhang Wei
- College of Animal Science and Technology, Guangxi University, Nanning 530005, China; (J.Y.); (K.Z.)
| | - Changlong Liu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; (H.L.); (L.L.); (S.L.); (Y.J.); (Y.Z.)
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
4
|
Egelkrout E, Maj M, Manjarin R, Fake G, Watanabe M, Williams J, Blanchard N, Walker J, Hayden C, Howard J. Effect of Fusion to the LTB Carrier Protein on Coronavirus Spike Protein Vaccine Candidates Produced in Maize. Viruses 2024; 17:7. [PMID: 39861796 PMCID: PMC11768699 DOI: 10.3390/v17010007] [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: 11/01/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 01/27/2025] Open
Abstract
Coronaviruses continue to disrupt health and economic productivity worldwide. Porcine epidemic diarrhea virus (PEDV) is a devastating swine disease and SARS-CoV-2 is the latest coronavirus to infect the human population. Both viruses display a similar spike protein on the surface that is a target of vaccine development. Despite the availability of commercial vaccines for both viruses, there is still a high occurrence of infections and a great need for enhanced efficacy and lower costs. We previously produced the PEDV spike protein (S) using transgenic maize, enabling a low-cost supply of the vaccine candidate. In this study, we (1) test orally delivered PEDV vaccine candidates in pigs to optimize the mucosal immune response; (2) generate the SARS-CoV-2 S1 protein in maize; and (3) perform structural characterization of the S1 protein for PEDV and SARS-CoV-2. We demonstrated high expression levels in maize of the S1 subunit of the SARS-CoV-2 spike protein, both with and without fusion to the heat-labile enterotoxin B (LTB) subunit. We found that the LTB fusion protein from both coronaviruses preferentially assembles into higher molecular weight multimers, consistent with the formation of trimers. For PEDV, administering the spike protein fused to LTB to young pigs elicited a higher level of mucosal IgAs compared to maize grain containing the S1 protein alone or controls. This suggests that fusing the coronavirus spike protein with LTB may provide better protection.
Collapse
Affiliation(s)
- Erin Egelkrout
- Applied Biotechnology Institute, California Polytechnic Tech Park, San Luis Obispo, CA 93407, USA; (E.E.); (G.F.); (M.W.)
| | - Magdalena Maj
- Department of Biological Sciences, California Polytechnic State University, San Luis Obispo, CA 93407, USA; (M.M.)
| | - Rodrigo Manjarin
- Department of Animal Sciences, California Polytechnic State University, San Luis Obispo, CA 93407, USA;
| | - Gina Fake
- Applied Biotechnology Institute, California Polytechnic Tech Park, San Luis Obispo, CA 93407, USA; (E.E.); (G.F.); (M.W.)
| | - Muneaki Watanabe
- Applied Biotechnology Institute, California Polytechnic Tech Park, San Luis Obispo, CA 93407, USA; (E.E.); (G.F.); (M.W.)
| | - Jenna Williams
- Department of Biological Sciences, California Polytechnic State University, San Luis Obispo, CA 93407, USA; (M.M.)
| | - Nate Blanchard
- Department of Biological Sciences, California Polytechnic State University, San Luis Obispo, CA 93407, USA; (M.M.)
| | - John Walker
- Department of Statistics, California Polytechnic State University, San Luis Obispo, CA 93407, USA
| | - Celine Hayden
- Applied Biotechnology Institute, California Polytechnic Tech Park, San Luis Obispo, CA 93407, USA; (E.E.); (G.F.); (M.W.)
| | - John Howard
- Applied Biotechnology Institute, California Polytechnic Tech Park, San Luis Obispo, CA 93407, USA; (E.E.); (G.F.); (M.W.)
| |
Collapse
|
5
|
Yang D, Wang X, Yang X, Qi S, Zhao F, Guo D, Li C, Zhu Q, Xing X, Cao Y, Sun D. Construction and immune effect evaluation of the S protein heptad repeat-based nanoparticle vaccine against porcine epidemic diarrhea virus. Virology 2024; 596:110113. [PMID: 38801794 DOI: 10.1016/j.virol.2024.110113] [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: 04/05/2024] [Revised: 04/26/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024]
Abstract
Porcine epidemic diarrhea virus (PEDV), a highly virulent enteropathogenic coronavirus, is a significant threat to the pig industry. High frequency mutations in the PEDV genome have limited the effectiveness of current vaccines in providing immune protection. Developing efficient vaccines that can quickly adapt to mutant strains is a challenging but crucial task. In this study, we chose the pivotal protein heptad repeat (HR) responsible for coronavirus entry into host cells, as the vaccine antigen. HR-Fer nanoparticles prepared using ferritin were evaluated them as PEDV vaccine candidates. Nanoparticle vaccines elicited stronger neutralizing antibody responses in mice compared to monomer vaccines. Additionally, HR protein delivered via nanoparticles increased antigen uptake by antigen-presenting cells in vitro by 2.75-fold. The collective results suggest that HR can be used as antigens for vaccines, and the HR vaccine based on ferritin nanoparticles significantly enhances immunogenicity.
Collapse
Affiliation(s)
- Dan Yang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, No. 5 Xinfeng Road, Sartu District, Daqing, 163319, China
| | - Xinglin Wang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, No. 5 Xinfeng Road, Sartu District, Daqing, 163319, China
| | - Xu Yang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, No. 5 Xinfeng Road, Sartu District, Daqing, 163319, China
| | - Shanshan Qi
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, No. 5 Xinfeng Road, Sartu District, Daqing, 163319, China
| | - Feiyu Zhao
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, No. 5 Xinfeng Road, Sartu District, Daqing, 163319, China
| | - Donghua Guo
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, No. 5 Xinfeng Road, Sartu District, Daqing, 163319, China
| | - Chunqiu Li
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, No. 5 Xinfeng Road, Sartu District, Daqing, 163319, China
| | - Qinghe Zhu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, No. 5 Xinfeng Road, Sartu District, Daqing, 163319, China
| | - Xiaoxu Xing
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, No. 5 Xinfeng Road, Sartu District, Daqing, 163319, China
| | - Yang Cao
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, No. 5 Xinfeng Road, Sartu District, Daqing, 163319, China.
| | - Dongbo Sun
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, No. 5 Xinfeng Road, Sartu District, Daqing, 163319, China.
| |
Collapse
|
6
|
Deng K, Huang Z, Jing B, Zhu L, Feng Y, Jiang Q, Xu Z, Wan H, Zhao X. Mucoadhesive chitosan-catechol as an efficient vaccine delivery system for intranasal immunization. Int J Biol Macromol 2024; 273:133008. [PMID: 38852736 DOI: 10.1016/j.ijbiomac.2024.133008] [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: 12/19/2023] [Revised: 05/30/2024] [Accepted: 06/06/2024] [Indexed: 06/11/2024]
Abstract
The mucosal barrier and scavenging effect of the mucosal layer are two main obstacles in inducing mucosal immunization. To overcome these obstacles, we synthesized a bio-inspired mucoadhesive material, chitosan-catechol (ChiC), for surface modification of inactive porcine epidemic diarrhea virus (PEDV). Studies have revealed that PEDV particles can be facilely and mildly modified by Chi-C forming Chi-C-PEDV nanoparticles (Chic-Ps) through the covalent and electrostatic bond, which effectively prolongs the retention time of PEDV in the nasal mucosa. The cell co-culture model demonstrated that Chic-Ps exhibit enhanced recruitment of dendritic cells via the secretion of stimulating chemokine CCL20 and improving antigen permeability by disruption the distribution of ZO-1 protein in epithelial cells. Additionally, the flow cytometry (FCM) analysis revealed that Chic-Ps facilitate trafficking to lymph nodes and induce stronger cellular and humoral immune responses compared to unmodified PEDV. Notably, Chic-Ps induced a higher level of PEDV neutralizing antibody was induced by Chic-Ps in the nasal washes, as confirmed by a plaque reduction neutralization test. These results demonstrate that Chi-C is a promising nasal delivery system for vaccines. Proof of principle was obtained for inactivated PEDV, but similar delivery mechanisms could be applied in other vaccines when intranasal administration is needed.
Collapse
Affiliation(s)
- Kai Deng
- Center for Infectious Diseases Control (CIDC), Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Center for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu 611130, China
| | - Zhengqun Huang
- Center for Infectious Diseases Control (CIDC), Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Center for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu 611130, China
| | - Bo Jing
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Lin Zhu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Yumei Feng
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University at Wenjiang, Chengdu 611130, China
| | - Qin Jiang
- College of Environmental Science, Sichuan Agricultural University, Chengdu 611130, China
| | - Zhiwen Xu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Hongping Wan
- Center for Infectious Diseases Control (CIDC), Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Center for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu 611130, China.
| | - Xinghong Zhao
- Center for Infectious Diseases Control (CIDC), Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Center for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
7
|
Wei MZ, Chen L, Zhang R, Chen Z, Shen YJ, Zhou BJ, Wang KG, Shan CL, Zhu EP, Cheng ZT. Overview of the recent advances in porcine epidemic diarrhea vaccines. Vet J 2024; 304:106097. [PMID: 38479492 DOI: 10.1016/j.tvjl.2024.106097] [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: 09/27/2023] [Revised: 02/04/2024] [Accepted: 03/04/2024] [Indexed: 03/18/2024]
Abstract
Vaccination is the most effective means of preventing and controlling porcine epidemic diarrhea (PED). Conventional vaccines developed from porcine epidemic diarrhea virus (PEDV) GI-a subtypes (CV777 and SM98) have played a vital role in preventing classical PED. However, with the emergence of PEDV mutants in 2010, conventional PEDV GI-a subtype-targeting vaccines no longer provide adequate protection against PEDV GII mutants, thereby making novel-type PED vaccine development an urgent concern to be addressed. Novel vaccines, including nucleic acid vaccines, genetically engineered subunit vaccines, and live vector vaccines, are associated with several advantages, such as high safety and stability, clear targeting, high yield, low cost, and convenient usage. These vaccines can be combined with corresponding ELISA kits to differentiate infected from vaccinated animals, which is beneficial for disease confirmation. This review provides a detailed overview of the recent advancements in PED vaccines, emphasizing on the research and application evaluation of novel PED vaccines. It also considers the future directions and challenges in advancing these vaccines to widespread use in clinics.
Collapse
Affiliation(s)
- Miao-Zhan Wei
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Lan Chen
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Rong Zhang
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Ze Chen
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Yan-Juan Shen
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Bi-Jun Zhou
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang 550025, China; Key Laboratory of Animal Diseases and Veterinary Public Health of Guizhou Province, College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Kai-Gong Wang
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang 550025, China; Key Laboratory of Animal Diseases and Veterinary Public Health of Guizhou Province, College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Chun-Lan Shan
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang 550025, China; Key Laboratory of Animal Diseases and Veterinary Public Health of Guizhou Province, College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Er-Peng Zhu
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang 550025, China; Key Laboratory of Animal Diseases and Veterinary Public Health of Guizhou Province, College of Animal Science, Guizhou University, Guiyang 550025, China.
| | - Zhen-Tao Cheng
- Department of Veterinary Medicine, College of Animal Science, Guizhou University, Guiyang 550025, China; Key Laboratory of Animal Diseases and Veterinary Public Health of Guizhou Province, College of Animal Science, Guizhou University, Guiyang 550025, China.
| |
Collapse
|
8
|
Park JE. Porcine Epidemic Diarrhea: Insights and Progress on Vaccines. Vaccines (Basel) 2024; 12:212. [PMID: 38400195 PMCID: PMC10892315 DOI: 10.3390/vaccines12020212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/16/2024] [Accepted: 02/17/2024] [Indexed: 02/25/2024] Open
Abstract
Porcine epidemic diarrhea (PED) is a swine-wasting disease caused by coronavirus infection. It causes great economic damage to the swine industry worldwide. Despite the continued use of vaccines, PED outbreaks continue, highlighting the need to review the effectiveness of current vaccines and develop additional vaccines based on new platforms. Here, we review existing vaccine technologies for preventing PED and highlight promising technologies that may help control PED virus in the future.
Collapse
Affiliation(s)
- Jung-Eun Park
- Laboratory of Veterinary Public Health, College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| |
Collapse
|
9
|
Guo W, Wang C, Song X, Xu H, Zhao S, Gu J, Zou Z, Li J, Qian J, Zhang X, Guo R, Li J, Li L, Hu Z, Ren L, Fan B, Li B. Immunogenicity and protective efficacy of a trimeric full-length S protein subunit vaccine for porcine epidemic diarrhea virus. Vaccine 2024; 42:828-839. [PMID: 38220489 DOI: 10.1016/j.vaccine.2024.01.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/31/2023] [Accepted: 01/04/2024] [Indexed: 01/16/2024]
Abstract
Porcine epidemic diarrhea virus (PEDV) has caused serious economic losses to the pig husbandry worldwide, and the effects of existing commercialized vaccines are suboptimal. Therefore, research to develop an efficacious vaccine for prevention and control of PEDV is essential. In this study, we designed and produced trimerized proteins of full-length PEDV spike (S) protein, S1 subunit, and a tandem of multiple epitopes of S protein using an efficient mammalian expression vector system in HEK 293F cells. The immunogenicity of two commercial adjuvants, M401 and M103, was also evaluated in mice. Enzyme-linked immunosorbent assays demonstrated that all immunized mice generated highly systemic PEDV S-specific IgG and IgA antibodies. Mice in S/M103-immunized group generated the highest neutralizing antibody titer with 1:96. Compared with control group, the subunit vaccines elicited multifunctional CD3+CD4+ and CD3+CD8+ T cells, B220+CD19+ B cells, and CD3-CD49b+ natural killer cells in the spleen. PEDV S/M103 vaccine, which had the best immune effect, was selected for further evaluation in piglets. Immunization with S/M103 vaccine induced high levels of S-specific IgG, IgA, and neutralizing antibodies, and increased the proliferation of peripheral blood mononuclear cells and the expression levels of interferon-γ and interleukin-4 in peripheral blood of piglets. Virus challenge test results showed significantly lower diarrheal index scores and fecal viral loads, and less pathological damage to the intestines in S/M103-immunized piglets than in controls, indicating that S/M103 provides good protection against the virulent virus challenge. Our findings suggest that trimeric PEDV S/M103 has potential as a clinical vaccine candidate.
Collapse
Affiliation(s)
- Weilu Guo
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing 210014, China; School of Pharmacy, Nanjing Tech University, 5th Mofan Road, Nanjing 210009, Jiangsu, China; Taizhou Polytechnic College, Taizhou 225300, Jiangsu, China
| | - Chuanhong Wang
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing 210014, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; School of Life Sciences, Jiangsu University, Zhenjiang 212013, China
| | - Xu Song
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing 210014, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Hong Xu
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing 210014, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Shuqing Zhao
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing 210014, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Jun Gu
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing 210014, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Zhikun Zou
- Chengdu Yisikang Biotechnology LLC, Chendou 610095, China
| | - Jing Li
- Chengdu Yisikang Biotechnology LLC, Chendou 610095, China
| | - Jiali Qian
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing 210014, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Xue Zhang
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing 210014, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Rongli Guo
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing 210014, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Jizong Li
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing 210014, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Li Li
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing 210014, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Zhaoyang Hu
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China
| | - Lili Ren
- School of Pharmacy, Nanjing Tech University, 5th Mofan Road, Nanjing 210009, Jiangsu, China.
| | - Baochao Fan
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing 210014, China; College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China.
| | - Bin Li
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing 210014, China; College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, China; GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou 225300, China.
| |
Collapse
|
10
|
Li X, Wu Y, Yan Z, Li G, Luo J, Huang S, Guo X. A Comprehensive View on the Protein Functions of Porcine Epidemic Diarrhea Virus. Genes (Basel) 2024; 15:165. [PMID: 38397155 PMCID: PMC10887554 DOI: 10.3390/genes15020165] [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: 12/28/2023] [Revised: 01/24/2024] [Accepted: 01/24/2024] [Indexed: 02/25/2024] Open
Abstract
Porcine epidemic diarrhea (PED) virus (PEDV) is one of the main pathogens causing diarrhea in piglets and fattening pigs. The clinical signs of PED are vomiting, acute diarrhea, dehydration, and mortality resulting in significant economic losses and becoming a major challenge in the pig industry. PEDV possesses various crucial structural and functional proteins, which play important roles in viral structure, infection, replication, assembly, and release, as well as in escaping host innate immunity. Over the past few years, there has been progress in the study of PEDV pathogenesis, revealing the crucial role of the interaction between PEDV viral proteins and host cytokines in PEDV infection. At present, the main control measure against PEDV is vaccine immunization of sows, but the protective effect for emerging virus strains is still insufficient, and there is no ideal safe and efficient vaccine. Although scientists have persistently delved their research into the intricate structure and functionalities of the PEDV genome and viral proteins for years, the pathogenic mechanism of PEDV remains incompletely elucidated. Here, we focus on reviewing the research progress of PEDV structural and nonstructural proteins to facilitate the understanding of biological processes such as PEDV infection and pathogenesis.
Collapse
Affiliation(s)
- Xin Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (X.L.); (Y.W.); (Z.Y.); (G.L.); (J.L.)
- Zhaoqing Branch Center of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing 526238, China
| | - Yiwan Wu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (X.L.); (Y.W.); (Z.Y.); (G.L.); (J.L.)
- Zhaoqing Branch Center of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing 526238, China
| | - Zhibin Yan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (X.L.); (Y.W.); (Z.Y.); (G.L.); (J.L.)
- Zhaoqing Branch Center of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing 526238, China
| | - Gen Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (X.L.); (Y.W.); (Z.Y.); (G.L.); (J.L.)
| | - Jun Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (X.L.); (Y.W.); (Z.Y.); (G.L.); (J.L.)
| | - Shile Huang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, USA
- Department of Hematology and Oncology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, USA
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, USA
| | - Xiaofeng Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (X.L.); (Y.W.); (Z.Y.); (G.L.); (J.L.)
- Zhaoqing Branch Center of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing 526238, China
| |
Collapse
|
11
|
Su K, Wang Y, Yuan C, Zhang Y, Li Y, Li T, Song Q. Intranasally inoculated bacterium-like particles displaying porcine epidemic diarrhea virus S1 protein induced intestinal mucosal immune response in mice. Front Immunol 2023; 14:1269409. [PMID: 37790942 PMCID: PMC10544335 DOI: 10.3389/fimmu.2023.1269409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 08/31/2023] [Indexed: 10/05/2023] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) causes acute watery diarrhea and high mortality in newborn piglets. Activation of intestinal mucosal immunity is crucial to anti-PEDV infection. To develop a vaccine capable of stimulating intestinal mucosal immunity, we prepared a bacterium (Lactococcus lactis)-like particle (BLP) vaccine (S1-BLPs) displaying the S1 protein, a domain of PEDV spike protein (S), based on gram-positive enhancer matrix (GEM) particle display technology. We further compared the effects of different vaccination routes on mucosal immune responses in mice induced by S1-BLPs. The specific IgG titer in serum of intramuscularly immunized mice with S1-BLPs was significantly higher than that of the intranasally administered. The specific IgA antibody was found in the serum and intestinal lavage fluid of mice vaccinated intranasally, but not intramuscularly. Moreover, the intranasally inoculated S1-BLPs induced higher levels of IFN-γ and IL-4 in serum than the intramuscularly inoculated. In addition, the ratio of serum IgG2a/IgG1 of mice inoculated intramuscularly was significantly higher with S1-BLPs compared to that of with S1 protein, suggesting that the immune responses induced by S1-BLPs was characterized by helper T (Th) cell type 1 immunity. The results indicated that S1-BLPs induced systemic and local immunity, and the immunization routes significantly affected the specific antibody classes and Th immune response types. The intranasally administered S1-BLPs could effectively stimulate intestinal mucosal specific secretory IgA response. S1-BLPs have the potential to be developed as PEDV mucosal vaccine.
Collapse
Affiliation(s)
- Kai Su
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, Hebei, China
- National Center of Technology Innovation for Pigs, Chongqing, China
- Hebei Veterinary Biotechnology Innovation Center, Baoding, Hebei, China
| | - Yawen Wang
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, Hebei, China
- Hebei Veterinary Biotechnology Innovation Center, Baoding, Hebei, China
| | - Chen Yuan
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, Hebei, China
- National Center of Technology Innovation for Pigs, Chongqing, China
- Hebei Veterinary Biotechnology Innovation Center, Baoding, Hebei, China
| | - Yanan Zhang
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, Hebei, China
- Hebei Veterinary Biotechnology Innovation Center, Baoding, Hebei, China
| | - Yanrui Li
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, Hebei, China
- Hebei Veterinary Biotechnology Innovation Center, Baoding, Hebei, China
| | - Tanqing Li
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, Hebei, China
- Hebei Veterinary Biotechnology Innovation Center, Baoding, Hebei, China
| | - Qinye Song
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, Hebei, China
- National Center of Technology Innovation for Pigs, Chongqing, China
- Hebei Veterinary Biotechnology Innovation Center, Baoding, Hebei, China
| |
Collapse
|
12
|
Huang CY, Draczkowski P, Wang YS, Chang CY, Chien YC, Cheng YH, Wu YM, Wang CH, Chang YC, Chang YC, Yang TJ, Tsai YX, Khoo KH, Chang HW, Hsu STD. In situ structure and dynamics of an alphacoronavirus spike protein by cryo-ET and cryo-EM. Nat Commun 2022; 13:4877. [PMID: 35986008 PMCID: PMC9388967 DOI: 10.1038/s41467-022-32588-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 08/04/2022] [Indexed: 11/21/2022] Open
Abstract
Porcine epidemic diarrhea (PED) is a highly contagious swine disease caused by porcine epidemic diarrhea virus (PEDV). PED causes enteric disorders with an exceptionally high fatality in neonates, bringing substantial economic losses in the pork industry. The trimeric spike (S) glycoprotein of PEDV is responsible for virus-host recognition, membrane fusion, and is the main target for vaccine development and antigenic analysis. The atomic structures of the recombinant PEDV S proteins of two different strains have been reported, but they reveal distinct N-terminal domain 0 (D0) architectures that may correspond to different functional states. The existence of the D0 is a unique feature of alphacoronavirus. Here we combined cryo-electron tomography (cryo-ET) and cryo-electron microscopy (cryo-EM) to demonstrate in situ the asynchronous S protein D0 motions on intact viral particles of a highly virulent PEDV Pintung 52 strain. We further determined the cryo-EM structure of the recombinant S protein derived from a porcine cell line, which revealed additional domain motions likely associated with receptor binding. By integrating mass spectrometry and cryo-EM, we delineated the complex compositions and spatial distribution of the PEDV S protein N-glycans, and demonstrated the functional role of a key N-glycan in modulating the D0 conformation. Hsu and co-workers integrate cryo-electron tomography, cryo-electron microscopy and mass spectrometry to reveal the structural polymorphism of a pig coronavirus spike protein within intact viral particles, and how glycosylation modulates the conformational changes pertinent to host recognition.
Collapse
|
13
|
Wang R, Suzuki S, Guest JD, Heller B, Almeda M, Andrianov AK, Marin A, Mariuzza RA, Keck ZY, Foung SKH, Yunus AS, Pierce BG, Toth EA, Ploss A, Fuerst TR. Induction of broadly neutralizing antibodies using a secreted form of the hepatitis C virus E1E2 heterodimer as a vaccine candidate. Proc Natl Acad Sci U S A 2022; 119:e2112008119. [PMID: 35263223 PMCID: PMC8931252 DOI: 10.1073/pnas.2112008119] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 01/19/2022] [Indexed: 11/26/2022] Open
Abstract
SignificanceHepatitis C virus chronically infects approximately 1% of the world's population, making an effective vaccine for hepatitis C virus a major unmet public health need. The membrane-associated E1E2 envelope glycoprotein has been used in clinical studies as a vaccine candidate. However, limited neutralization breadth and difficulty in producing large amounts of homogeneous membrane-associated E1E2 have hampered efforts to develop an E1E2-based vaccine. Our previous work described the design and biochemical validation of a native-like soluble secreted form of E1E2 (sE1E2). Here, we describe the immunogenic characterization of the sE1E2 complex. sE1E2 elicited broadly neutralizing antibodies in immunized mice, with increased neutralization breadth relative to the membrane-associated E1E2, thereby validating this platform as a promising model system for vaccine development.
Collapse
Affiliation(s)
- Ruixue Wang
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850
| | - Saori Suzuki
- Department of Molecular Biology, Princeton University, Princeton, NJ 08540
| | - Johnathan D. Guest
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742
| | - Brigitte Heller
- Department of Molecular Biology, Princeton University, Princeton, NJ 08540
| | - Maricar Almeda
- Department of Molecular Biology, Princeton University, Princeton, NJ 08540
| | - Alexander K. Andrianov
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850
| | - Alexander Marin
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850
| | - Roy A. Mariuzza
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742
| | - Zhen-Yong Keck
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305
| | - Steven K. H. Foung
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305
| | - Abdul S. Yunus
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850
| | - Brian G. Pierce
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742
| | - Eric A. Toth
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850
| | - Alexander Ploss
- Department of Molecular Biology, Princeton University, Princeton, NJ 08540
| | - Thomas R. Fuerst
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742
| |
Collapse
|
14
|
Chang CY, Wang YS, Wu JF, Yang TJ, Chang YC, Chae C, Chang HW, Hsu STD. Generation and Characterization of a Spike Glycoprotein Domain A-Specific Neutralizing Single-Chain Variable Fragment against Porcine Epidemic Diarrhea Virus. Vaccines (Basel) 2021; 9:vaccines9080833. [PMID: 34451958 PMCID: PMC8402611 DOI: 10.3390/vaccines9080833] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 12/26/2022] Open
Abstract
The emergence of the genotype (G) 2 and re-emergence of the G1 porcine epidemic diarrhea virus (PEDV) has caused severe economic impacts in the past decade. Developments of efficient vaccines against new variants of PEDV have been challenging, not least because of the difficulties in eliciting mucosal and lactogenic immunity. A single-chain fragment variable (scFv) capable of efficient antigen recognition is an alternative to vaccination and treatment of a viral infection. In the present study, the variable regions of the light chain and the heavy chain of a G2b PEDV spike domain A (S1A)-specific neutralizing monoclonal antibody (mAb) were sequenced, constructed with a (G4S) x3 linker, and produced by a mammalian protein expression system. Our results demonstrated that the PEDV S1A domain scFv was able to bind to S proteins of both G1 and G2b PEDVs. Nevertheless, the scFv was only capable of neutralizing the homologous G2b PEDV but not the G1 PEDV. The binding ability of the G2b-specific neutralizing scFv was not able to predict the neutralizing ability toward heterologous PEDV. The anti-PEDV S1A scFv presented herein serves as a potential therapeutic candidate against the virulent G2b PEDV.
Collapse
Affiliation(s)
- Chia-Yu Chang
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan; (C.-Y.C.); (Y.-S.W.); (T.-J.Y.)
| | - Yong-Sheng Wang
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan; (C.-Y.C.); (Y.-S.W.); (T.-J.Y.)
- Institute of Biochemical Sciences, National Taiwan University, Taipei 10617, Taiwan
| | - Jou-Fei Wu
- Graduate Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan; (J.-F.W.); (Y.-C.C.); (H.-W.C.)
| | - Tzu-Jing Yang
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan; (C.-Y.C.); (Y.-S.W.); (T.-J.Y.)
- Institute of Biochemical Sciences, National Taiwan University, Taipei 10617, Taiwan
| | - Yen-Chen Chang
- Graduate Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan; (J.-F.W.); (Y.-C.C.); (H.-W.C.)
| | - Chanhee Chae
- Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, Seoul 151-742, Korea;
| | - Hui-Wen Chang
- Graduate Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan; (J.-F.W.); (Y.-C.C.); (H.-W.C.)
| | - Shang-Te Danny Hsu
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan; (C.-Y.C.); (Y.-S.W.); (T.-J.Y.)
- Institute of Biochemical Sciences, National Taiwan University, Taipei 10617, Taiwan
- Correspondence: ; Tel.: +886-2-2785-5696 (ext. 5120)
| |
Collapse
|
15
|
Stable trimer formation of spike protein from porcine epidemic diarrhea virus improves the efficiency of secretory production in silkworms and induces neutralizing antibodies in mice. Vet Res 2021; 52:102. [PMID: 34233749 PMCID: PMC8261802 DOI: 10.1186/s13567-021-00971-5] [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: 04/23/2021] [Accepted: 06/21/2021] [Indexed: 12/14/2022] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) is a highly infectious pathogen of watery diarrhea that causes serious economic loss to the swine industry worldwide. Especially because of the high mortality rate in neonatal piglets, a vaccine with less production cost and high protective effect against PEDV is desired. The intrinsically assembled homotrimer of spike (S) protein on the PEDV viral membrane contributing to the host cell entry is a target of vaccine development. In this study, we designed trimerized PEDV S protein for efficient production in the silkworm-baculovirus expression vector system (silkworm-BEVS) and evaluated its immunogenicity in the mouse. The genetic fusion of the trimeric motif improved the expression of S protein in silkworm-BEVS. A small-scale screening of silkworm strains to further improve the S protein productivity finally achieved the yield of about 2 mg from the 10 mL larval serum. Mouse immunization study demonstrated that the trimerized S protein could elicit strong humoral immunity, including the S protein-specific IgG in the serum. These sera contained neutralizing antibodies that can protect Vero cells from PEDV infection. These results demonstrated that silkworm-BEVS provides a platform for the production of trimeric S proteins, which are promising subunit vaccines against coronaviruses such as PEDV.
Collapse
|
16
|
Jabif MF, Gumina E, Hall JW, Hernandez-Velasco X, Layton S. Evaluation of a Novel Mucosal Administered Subunit Vaccine on Colostrum IgA and Serum IgG in Sows and Control of Enterotoxigenic Escherichia coli in Neonatal and Weanling Piglets: Proof of Concept. Front Vet Sci 2021; 8:640228. [PMID: 33644156 PMCID: PMC7905019 DOI: 10.3389/fvets.2021.640228] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 01/20/2021] [Indexed: 11/21/2022] Open
Abstract
The purpose of the present study was to evaluate the ability of a novel experimental subunit vaccine (ESV), induce colostrum IgA and serum IgG in sows, and to control enterotoxigenic Escherichia coli (ETEC) disease in neonatal and weanling piglets. The vaccine was tested in three experiments. Experiment 1 consisted of two independent trials. In each trial, 20 pregnant sows/groups were vaccinated intramuscularly (IM) with a commercial E. coli vaccine or intranasally with ESV at weeks 11 and 13 of pregnancy. Blood and serum samples were obtained within 12 h post-partum. In Experiment 1, intranasal vaccination with ESV significantly increased the sample-to-positive (S/P) ratio of secretory IgA in the colostrum of sows (P < 0.01, trial 1; P < 0.05, trial 2) compared to the IM vaccine. In Experiment 2, twenty-five 3-day old piglets were randomly allocated into two groups, control (n = 13) or ESV (n = 12) and were oral gavaged with the respective treatments on days 3 and 14 of life. On days 17–19, all piglets were challenged using a mixed ETEC culture via oral gavage. Within 72 h, all control group animals developed disease consistent with colibacillosis. Conversely, the ESV treated group remained disease free over the 7-day observation period and had significant increases in body weight gain compared to the control group piglets. In Experiment 3, thirty 28-day old piglets were randomly allocated, control (n = 15) or ESV (n = 15), and on days 33 and 43 of life, piglets were either given by oral gavage 2.0 mL saline (control group) or 2.0 mL ESV. At days 46 and 47 of life, all pigs were challenged with a mixed culture of ETEC and observed for clinical signs of disease. Results of Experiment 3 were similar to those observed in Experiment 2. This study indicates the ESV can induce better levels of colostrum secretory IgA in pregnant sows than IM vaccination, which may be protective to neonatal piglets. Further, the vaccine can protect piglets as early as 3 days of age from an ETEC infection. Importantly, the data suggest a single vaccine could be used across the farrowing, suckling, and weaning program to protect against pathogenic E. coli.
Collapse
Affiliation(s)
| | | | | | - Xochitl Hernandez-Velasco
- Departamento de Medicina y Zootecnia de Aves, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autonoma de Mexico, Ciudad de Mexico, Mexico
| | - Sherry Layton
- Vetanco S.A., Buenos Aires, Argentina.,Vetanco USA, Inc., Saint Paul, MN, United States
| |
Collapse
|
17
|
Guest JD, Wang R, Elkholy KH, Chagas A, Chao KL, Cleveland TE, Kim YC, Keck ZY, Marin A, Yunus AS, Mariuzza RA, Andrianov AK, Toth EA, Foung SKH, Pierce BG, Fuerst TR. Design of a native-like secreted form of the hepatitis C virus E1E2 heterodimer. Proc Natl Acad Sci U S A 2021; 118:e2015149118. [PMID: 33431677 PMCID: PMC7826332 DOI: 10.1073/pnas.2015149118] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Hepatitis C virus (HCV) is a major worldwide health burden, and a preventive vaccine is needed for global control or eradication of this virus. A substantial hurdle to an effective HCV vaccine is the high variability of the virus, leading to immune escape. The E1E2 glycoprotein complex contains conserved epitopes and elicits neutralizing antibody responses, making it a primary target for HCV vaccine development. However, the E1E2 transmembrane domains that are critical for native assembly make it challenging to produce this complex in a homogenous soluble form that is reflective of its state on the viral envelope. To enable rational design of an E1E2 vaccine, as well as structural characterization efforts, we have designed a soluble, secreted form of E1E2 (sE1E2). As with soluble glycoprotein designs for other viruses, it incorporates a scaffold to enforce assembly in the absence of the transmembrane domains, along with a furin cleavage site to permit native-like heterodimerization. This sE1E2 was found to assemble into a form closer to its expected size than full-length E1E2. Preservation of native structural elements was confirmed by high-affinity binding to a panel of conformationally specific monoclonal antibodies, including two neutralizing antibodies specific to native E1E2 and to its primary receptor, CD81. Finally, sE1E2 was found to elicit robust neutralizing antibodies in vivo. This designed sE1E2 can both provide insights into the determinants of native E1E2 assembly and serve as a platform for production of E1E2 for future structural and vaccine studies, enabling rational optimization of an E1E2-based antigen.
Collapse
Affiliation(s)
- Johnathan D Guest
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742
| | - Ruixue Wang
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850
| | - Khadija H Elkholy
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850
- Molecular Biology Department, Genetic Engineering and Biotechnology Division, National Research Centre, Cairo 12622, Egypt
| | - Andrezza Chagas
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850
| | - Kinlin L Chao
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742
| | - Thomas E Cleveland
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850
- Biomolecular Measurement Division, National Institute of Standards and Technology, Gaithersburg, MD 20899
| | - Young Chang Kim
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305
| | - Zhen-Yong Keck
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305
| | - Alexander Marin
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850
| | - Abdul S Yunus
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850
| | - Roy A Mariuzza
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742
| | - Alexander K Andrianov
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850
| | - Eric A Toth
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850
| | - Steven K H Foung
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305
| | - Brian G Pierce
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850;
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742
| | - Thomas R Fuerst
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850;
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742
| |
Collapse
|
18
|
Won H, Lim J, Noh YH, Yoon I, Yoo HS. Efficacy of Porcine Epidemic Diarrhea Vaccines: A Systematic Review and Meta-Analysis. Vaccines (Basel) 2020; 8:vaccines8040642. [PMID: 33147824 PMCID: PMC7712170 DOI: 10.3390/vaccines8040642] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 12/16/2022] Open
Abstract
Porcine epidemic diarrhea (PED) is a devastating disease that causes considerable economic damage to the global pig industry. Although the causative agent, the porcine epidemic diarrhea virus (PEDV), was identified about a half century ago, there is still much debate on the preventive measures against the disease, especially regarding the PED vaccine. Recent reports on PEDV variants make the vaccination for PEDV more confusing. Therefore, we systematically reviewed published articles on PED and vaccines against the disease and performed a meta-analysis of vaccine efficacy based on the clinical signs, fecal score and survival rates. A total of 299 articles on the efficacy of PED vaccines were found online, and 21 articles were selected that fulfilled all the criteria. A meta-analysis was performed on the 21 articles based on the fecal scores and survival rates. This analysis showed the efficacy of PED vaccines, and no significant differences in the efficacy depending on vaccine type (killed vs. live) or administration route (intramuscular vs. oral) were found. The results from our study suggest that any vaccination against PED is a useful strategy to control the disease regardless of the type of vaccine and administration route.
Collapse
Affiliation(s)
- Hokeun Won
- Department of Infectious Diseases, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (H.W.); (J.L.)
- ChoongAng Vaccine Laboratories Co., Ltd., Daejeon 34055, Korea; (Y.H.N.); (I.Y.)
| | - Jeonggyo Lim
- Department of Infectious Diseases, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (H.W.); (J.L.)
- ChoongAng Vaccine Laboratories Co., Ltd., Daejeon 34055, Korea; (Y.H.N.); (I.Y.)
| | - Yun Hee Noh
- ChoongAng Vaccine Laboratories Co., Ltd., Daejeon 34055, Korea; (Y.H.N.); (I.Y.)
| | - Injoong Yoon
- ChoongAng Vaccine Laboratories Co., Ltd., Daejeon 34055, Korea; (Y.H.N.); (I.Y.)
| | - Han Sang Yoo
- Department of Infectious Diseases, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (H.W.); (J.L.)
- Bio-MAX/N-Bio Institute, Seoul National University, Seoul 08826, Korea
- Correspondence: ; Tel.: +82-2-880-1263
| |
Collapse
|
19
|
Hsu CW, Chang MH, Chang HW, Wu TY, Chang YC. Parenterally Administered Porcine Epidemic Diarrhea Virus-Like Particle-Based Vaccine Formulated with CCL25/28 Chemokines Induces Systemic and Mucosal Immune Protectivity in Pigs. Viruses 2020; 12:E1122. [PMID: 33023277 PMCID: PMC7600258 DOI: 10.3390/v12101122] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/30/2020] [Accepted: 09/30/2020] [Indexed: 01/12/2023] Open
Abstract
Generation of a safe, economical, and effective vaccine capable of inducing mucosal immunity is critical for the development of vaccines against enteric viral diseases. In the current study, virus-like particles (VLPs) containing the spike (S), membrane (M), and envelope (E) structural proteins of porcine epidemic diarrhea virus (PEDV) expressed by the novel polycistronic baculovirus expression vector were generated. The immunogenicity and protective efficacy of the PEDV VLPs formulated with or without mucosal adjuvants of CCL25 and CCL28 (CCL25/28) were evaluated in post-weaning pigs. While pigs intramuscularly immunized with VLPs alone were capable of eliciting systemic anti-PEDV S-specific IgG and cellular immunity, co-administration of PEDV VLPs with CCL25/28 could further modulate the immune responses by enhancing systemic anti-PEDV S-specific IgG, mucosal IgA, and cellular immunity. Upon challenge with PEDV, both VLP-immunized groups showed milder clinical signs with reduced fecal viral shedding as compared to the control group. Furthermore, pigs immunized with VLPs adjuvanted with CCL25/28 showed superior immune protection against PEDV. Our results suggest that VLPs formulated with CCL25/28 may serve as a potential PEDV vaccine candidate and the same strategy may serve as a platform for the development of other enteric viral vaccines.
Collapse
Affiliation(s)
- Chin-Wei Hsu
- Graduate Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, Taipei 106, Taiwan; (C.-W.H.); (H.-W.C.)
| | - Ming-Hao Chang
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan 320, Taiwan;
| | - Hui-Wen Chang
- Graduate Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, Taipei 106, Taiwan; (C.-W.H.); (H.-W.C.)
| | - Tzong-Yuan Wu
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan 320, Taiwan;
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 406, Taiwan
| | - Yen-Chen Chang
- Graduate Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, Taipei 106, Taiwan; (C.-W.H.); (H.-W.C.)
| |
Collapse
|
20
|
Li Z, Ma Z, Li Y, Gao S, Xiao S. Porcine epidemic diarrhea virus: Molecular mechanisms of attenuation and vaccines. Microb Pathog 2020; 149:104553. [PMID: 33011361 PMCID: PMC7527827 DOI: 10.1016/j.micpath.2020.104553] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/29/2020] [Accepted: 09/29/2020] [Indexed: 12/29/2022]
Abstract
Porcine epidemic diarrhea virus (PEDV) causes an emerging and re-emerging coronavirus disease characterized by vomiting, acute diarrhea, dehydration, and up to 100% mortality in neonatal suckling piglets, leading to huge economic losses in the global swine industry. Vaccination remains the most promising and effective way to prevent and control PEDV. However, effective vaccines for PEDV are still under development. Understanding the genomic structure and function of PEDV and the influence of the viral components on innate immunity is essential for developing effective vaccines. In the current review, we systematically describe the recent developments in vaccine against PEDV and the roles of structural proteins, non-structural proteins and accessory proteins of PEDV in affecting viral virulence and regulating innate immunity, which will provide insight into the rational design of effective and safe vaccines for PEDV or other coronaviruses. Advances in vaccines of PEDV, such as inactivated and attenuated live vaccines, subunit vaccines, and nucleic acid vaccines. The application of reverse genetics in the development of live attenuated PEDV vaccines. The roles of PEDV proteins in affecting viral virulence and regulating innate immunity.
Collapse
Affiliation(s)
- Zhiwei Li
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Zhiqian Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yang Li
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Sheng Gao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Shuqi Xiao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.
| |
Collapse
|
21
|
Adjuvants for swine vaccines: Mechanisms of actions and adjuvant effects. Vaccine 2020; 38:6659-6681. [DOI: 10.1016/j.vaccine.2020.08.054] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 02/07/2023]
|
22
|
Hsueh FC, Chang YC, Kao CF, Hsu CW, Chang HW. Intramuscular Immunization with Chemokine-Adjuvanted Inactive Porcine Epidemic Diarrhea Virus Induces Substantial Protection in Pigs. Vaccines (Basel) 2020; 8:vaccines8010102. [PMID: 32102459 PMCID: PMC7157555 DOI: 10.3390/vaccines8010102] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 02/17/2020] [Accepted: 02/21/2020] [Indexed: 12/18/2022] Open
Abstract
Intramuscular (IM) immunization is generally considered incapable of generating a protective mucosal immune response. In the swine industry, attempts to develop a safe and protective vaccine for controlling porcine epidemic diarrhea (PED) via an IM route of administration have been unsuccessful. In the present study, porcine chemokine ligand proteins CCL25, 27, and 28 were constructed and stably expressed in the mammalian expression system. IM co-administration of inactivated PEDV (iPEDV) particles with different CC chemokines and Freund’s adjuvants resulted in recruiting CCR9+ and/or CCR10+ inflammatory cells to the injection site, thereby inducing superior systemic PEDV specific IgG, fecal IgA, and viral neutralizing antibodies in pigs. Moreover, pigs immunized with iPEDV in combination with CCL25 and CCL28 elicited substantial protection against a virulent PEDV challenge. We show that the porcine CC chemokines could be novel adjuvants for developing IM vaccines for modulating mucosal immune responses against mucosal transmissible pathogens in pigs.
Collapse
Affiliation(s)
- Fu-Chun Hsueh
- Graduate Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan; (F.-C.H.); (Y.-C.C.); (C.-W.H.)
| | - Yen-Chen Chang
- Graduate Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan; (F.-C.H.); (Y.-C.C.); (C.-W.H.)
- School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan;
| | - Chi-Fei Kao
- School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan;
| | - Chin-Wei Hsu
- Graduate Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan; (F.-C.H.); (Y.-C.C.); (C.-W.H.)
| | - Hui-Wen Chang
- Graduate Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan; (F.-C.H.); (Y.-C.C.); (C.-W.H.)
- School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan;
- Correspondence: ; Tel.: +886-2-3366-3867
| |
Collapse
|
23
|
Chang CY, Hsu WT, Tsai PS, Chen CM, Cheng IC, Chao YC, Chang HW. Oral administration of porcine epidemic diarrhea virus spike protein expressing in silkworm pupae failed to elicit immune responses in pigs. AMB Express 2020; 10:20. [PMID: 31993764 PMCID: PMC6987277 DOI: 10.1186/s13568-020-0952-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 01/08/2020] [Indexed: 11/21/2022] Open
Abstract
The silkworm (Bombyx mori) and its pupae have been used for decades as nutritional additives and applied on the production of high-quality recombinant proteins via the baculovirus expression vector (BEV) system. The bio-capsule, the fat-rich body, and some body components of the silkworm pupae, which deliver antigens passing through the harsh environment of digestive tract and reaching the intestine, have been used as a vehicle for oral vaccines. In the present study, to develop a novel oral vaccine against porcine epidemic diarrhea virus (PEDV), the PEDV spike (S) protein was expressed in silkworm pupae and BmN cells using the BEV system. After three doses of oral administrations with 2-week intervals in pigs, neither PEDV S protein-specific humoral nor mucosal immune responses can be detected. The failure of eliciting the PEDV-specific immune response suggested that the BEV system using BmN cells or silkworm pupae as oral immunogen-expression vehicles was not able to overcome the immunological unresponsiveness, which was possibly due to gastrointestinal specific barriers and oral tolerance. Better strategies to enhance the delivery and immunogenicity of oral vaccines should be further investigated. Nevertheless, the PEDV S protein generated in the BmN cells and silkworm pupae herein provides an efficient tool to produce the recombinant antigen for future applications.
Collapse
|
24
|
Gestal MC, Johnson HM, Harvill ET. Immunomodulation as a Novel Strategy for Prevention and Treatment of Bordetella spp. Infections. Front Immunol 2019; 10:2869. [PMID: 31921136 PMCID: PMC6923730 DOI: 10.3389/fimmu.2019.02869] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 11/22/2019] [Indexed: 12/13/2022] Open
Abstract
Well-adapted pathogens have evolved to survive the many challenges of a robust immune response. Defending against all host antimicrobials simultaneously would be exceedingly difficult, if not impossible, so many co-evolved organisms utilize immunomodulatory tools to subvert, distract, and/or evade the host immune response. Bordetella spp. present many examples of the diversity of immunomodulators and an exceptional experimental system in which to study them. Recent advances in this experimental system suggest strategies for interventions that tweak immunity to disrupt bacterial immunomodulation, engaging more effective host immunity to better prevent and treat infections. Here we review advances in the understanding of respiratory pathogens, with special focus on Bordetella spp., and prospects for the use of immune-stimulatory interventions in the prevention and treatment of infection.
Collapse
Affiliation(s)
- Monica C Gestal
- Department of Infectious Diseases, College of Veterinary Sciences, University of Georgia, Athens, GA, United States
| | - Hannah M Johnson
- Department of Infectious Diseases, College of Veterinary Sciences, University of Georgia, Athens, GA, United States
| | - Eric T Harvill
- Department of Infectious Diseases, College of Veterinary Sciences, University of Georgia, Athens, GA, United States
| |
Collapse
|
25
|
Chang CY, Peng JY, Cheng YH, Chang YC, Wu YT, Tsai PS, Chiou HY, Jeng CR, Chang HW. Development and comparison of enzyme-linked immunosorbent assays based on recombinant trimeric full-length and truncated spike proteins for detecting antibodies against porcine epidemic diarrhea virus. BMC Vet Res 2019; 15:421. [PMID: 31775769 PMCID: PMC6880432 DOI: 10.1186/s12917-019-2171-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 11/08/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Since 2010, outbreaks of genotype 2 (G2) porcine epidemic diarrhea virus (PEDV) have caused high mortality in neonatal piglets and have had devastating impacts on the swine industry in many countries. A reliable serological assay for evaluating the PEDV-specific humoral and mucosal immune response is important for disease survey, monitoring the efficacy of immunization, and designing strategies for the prevention and control of PED. Two PEDV spike (S) glycoprotein-based indirect enzyme-linked immunosorbent assays (ELISAs) were developed using G2b PEDV-Pintung 52 (PEDV-PT) trimeric full-length S and truncated S1-501 proteins derived from the human embryonic kidney (HEK)-293 cell expression system. The truncated S1-501 protein was selected from a superior expressed stable cell line. The sensitivity and specificity of these two ELISAs were compared to immunostaining of G2b PEDV-PT infected cells and to a commercial nucleocapsid (N)-based indirect ELISA kit using a panel of PEDV negative and hyperimmune sera. RESULTS The commercial N-based ELISA exhibited a sensitivity of 37%, a specificity of 100%, and a fair agreement (kappa = 0.37) with the immunostaining result. In comparison, the full-length S-based ELISA showed a sensitivity of 97.8%, a specificity of 94%, and an almost perfect agreement (kappa = 0.90) with the immunostaining result. Interestingly, the S1-501-based ELISA had even higher sensitivity of 98.9% and specificity of 99.1%, and an almost perfect agreement (kappa = 0.97) with the immunostaining result. A fair agreement (kappa< 0.4) was seen between the commercial N-based ELISA and either of our S-based ELISAs. However, the results of the full-length S-based ELISA shared an almost perfect agreement (kappa = 0.92) with that of S1-501-based ELISA. CONCLUSIONS Both full-length S-based and S1-501-based ELISAs exhibit high sensitivity and high specificity for detecting antibodies against PEDVs. Considering the high protein yield and cost-effectiveness, the S1-501-based ELISA could be used as a reliable, sensitive, specific, and economic serological test for PEDV.
Collapse
Affiliation(s)
- Chia-Yu Chang
- Graduate Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, No. 1, Section 4, Roosevelt Rd., Taipei, 10617, Taiwan.,School of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan
| | - Ju-Yi Peng
- Graduate Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, No. 1, Section 4, Roosevelt Rd., Taipei, 10617, Taiwan
| | - Yun-Han Cheng
- School of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan
| | - Yen-Chen Chang
- Graduate Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, No. 1, Section 4, Roosevelt Rd., Taipei, 10617, Taiwan.,School of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan
| | - Yen-Tse Wu
- School of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan
| | - Pei-Shiue Tsai
- School of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan
| | - Hue-Ying Chiou
- Graduate Institute of Veterinary Pathobiology, College of Veterinary Medicine, National Chung Hsing University, Taichung, 402, Taiwan
| | - Chian-Ren Jeng
- Graduate Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, No. 1, Section 4, Roosevelt Rd., Taipei, 10617, Taiwan.,School of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan
| | - Hui-Wen Chang
- Graduate Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, No. 1, Section 4, Roosevelt Rd., Taipei, 10617, Taiwan. .,School of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan.
| |
Collapse
|
26
|
Peng X, Zhang R, Wang C, Yu F, Yu M, Chen S, Fan Q, Xi Y, Duan G. E. coli Enterotoxin LtB Enhances Vaccine-Induced Anti- H. pylori Protection by Promoting Leukocyte Migration into Gastric Mucus via Inflammatory Lesions. Cells 2019; 8:982. [PMID: 31461854 PMCID: PMC6770474 DOI: 10.3390/cells8090982] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 08/23/2019] [Accepted: 08/23/2019] [Indexed: 12/11/2022] Open
Abstract
Current studies indicate that the anti-H. pylori protective efficacy of oral vaccines to a large extent depends on using mucosal adjuvants like E. coli heat-lable enterotoxin B unit (LtB). However, the mechanism by which Th17/Th1-driven cellular immunity kills H. pylori and the role of LtB remains unclear. Here, two L.lactis strains, expressing H. pylori NapA and LtB, respectively, were orally administrated to mice. As observed, the administration of LtB significantly enhanced the fecal SIgA level and decreased gastric H. pylori colonization, but also markedly aggravated gastric inflammatory injury. Both NapA group and NapA+LtB group had elevated splenocyte production of IL-8, IL-10, IL-12, IL-17, IL-23 and INF-γ. Notably, gastric leukocytes' migration or leakage into the mucus was observed more frequently in NapA+LtB group than in NapA group. This report is the first that discusses how LtB enhances vaccine-induced anti-H. pylori efficacy by aggravating gastric injury and leukocytes' movement into the mucus layer. Significantly, it brings up a novel explanation for the mechanism underlying mucosal cellular immunity destroying the non-invasive pathogens. More importantly, the findings suggest the necessity to further evaluate LtB's potential hazards to humans before extending its applications. Thus, this report can provide considerable impact on the fields of mucosal immunology and vaccinology.
Collapse
Affiliation(s)
- Xiaoyan Peng
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
- Department of Basic Medicine, Chuxiong Medical College, Chuxiong 675005, China
| | - Rongguang Zhang
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China.
| | - Chen Wang
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Feiyan Yu
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Mingyang Yu
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Shuaiyin Chen
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Qingtang Fan
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Yuanlin Xi
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Guangcai Duan
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
27
|
Chang CY, Cheng IC, Chang YC, Tsai PS, Lai SY, Huang YL, Jeng CR, Pang VF, Chang HW. Identification of Neutralizing Monoclonal Antibodies Targeting Novel Conformational Epitopes of the Porcine Epidemic Diarrhoea Virus Spike Protein. Sci Rep 2019; 9:2529. [PMID: 30792462 PMCID: PMC6385244 DOI: 10.1038/s41598-019-39844-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 01/30/2019] [Indexed: 01/20/2023] Open
Abstract
Since 2010, newly identified variants of porcine epidemic diarrhoea virus (PEDV) have caused high mortality in neonatal piglets which has devastated the swine industry. The spike (S) glycoprotein of PEDV contains multiple neutralizing epitopes and is a major target for PEDV neutralization and vaccine development. To understand the antigenicity of the new PEDV variant, we characterized the neutralizing epitopes of a new genotype 2b PEDV isolate from Taiwan, PEDV Pintung 52 (PEDV-PT), by the generation of neutralizing monoclonal antibodies (NmAbs). Two NmAbs, P4B-1, and E10E-1–10 that recognized the ectodomain of the full-length recombinant PEDV S protein and exhibited neutralizing ability against the PEDV-PT virus were selected. Recombinant truncated S proteins were used to identify the target sequences for the NmAbs and P4B-1 was shown to recognize the C-terminus of CO-26K equivalent epitope (COE) at amino acids (a.a.) 575–639 of the PEDV S. Interestingly, E10E-1–10 could recognize a novel neutralizing epitope at a.a. 435–485 within the S1A domain of the PEDV S protein, whose importance and function are yet to be determined. Moreover, both NmAbs could not bind to linearized S proteins, indicating that only conformational epitopes are recognized. This data could improve our understanding of the antigenic structures of the PEDV S protein and facilitate future development of novel epitope-based vaccines.
Collapse
Affiliation(s)
- Chia-Yu Chang
- Graduate Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, Taipei, 106, Taiwan
| | - Ivan-Chen Cheng
- School of Veterinary Medicine, National Taiwan University, Taipei, 106, Taiwan
| | - Yen-Chen Chang
- Graduate Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, Taipei, 106, Taiwan
| | - Pei-Shiue Tsai
- School of Veterinary Medicine, National Taiwan University, Taipei, 106, Taiwan
| | - Seiu-Yu Lai
- School of Veterinary Medicine, National Taiwan University, Taipei, 106, Taiwan
| | - Yu-Liang Huang
- Animal Health Research Institute, Council of Agriculture, New Taipei City, 251, Taiwan
| | - Chian-Ren Jeng
- Graduate Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, Taipei, 106, Taiwan.,School of Veterinary Medicine, National Taiwan University, Taipei, 106, Taiwan
| | - Victor Fei Pang
- Graduate Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, Taipei, 106, Taiwan.,School of Veterinary Medicine, National Taiwan University, Taipei, 106, Taiwan
| | - Hui-Wen Chang
- Graduate Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, Taipei, 106, Taiwan. .,School of Veterinary Medicine, National Taiwan University, Taipei, 106, Taiwan.
| |
Collapse
|
28
|
Kao CF, Chiou HY, Chang YC, Hsueh CS, Jeng CR, Tsai PS, Cheng IC, Pang VF, Chang HW. The Characterization of Immunoprotection Induced by a cDNA Clone Derived from the Attenuated Taiwan Porcine Epidemic Diarrhea Virus Pintung 52 Strain. Viruses 2018; 10:E543. [PMID: 30287770 PMCID: PMC6213177 DOI: 10.3390/v10100543] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 09/27/2018] [Accepted: 10/02/2018] [Indexed: 12/11/2022] Open
Abstract
The porcine epidemic diarrhea virus (PEDV) poses a great threat to the global swine industries and the unreliable protection induced by the currently available vaccines remains a major challenge. We previously generated a genogroup 2b (G2b) PEDV Taiwan Pintung 52 (PEDVPT) strain, PEDVPT-P96, and determined its promising host immune response against the virulent PEDVPT-P5 strain. To study the attenuation determinants of PEDVPT-P96 and establish a PEDVPT-P96-based recombinant vector as a vaccine platform for further antigenicity modification, iPEDVPT-P96, a full-length cDNA clone of PEDVPT-P96, was established. Comparing to the parental PEDVPT-P96 virus, the iPEDVPT-P96 virus showed efficient replication kinetics with a delayed decline of viral load and similar but much more uniform plaque sizes in Vero cells. In the 5-week-old piglet model, fecal viral shedding was observed in the PEDVPT-P96-inoculated piglets, whereas those inoculated with iPEDVPT-P96 showed neither detectable fecal viral shedding nor PEDV-associated clinical signs. Moreover, inoculation with iPEDVPT-P96 elicited comparable levels of anti-PEDV specific plasma IgG and fecal/salivary IgA, neutralizing antibody titers, and similar but less effective immunoprotection against the virulent PEDVPT-P5 challenge compared to the parental PEDVPT-P96. In the present study, an infectious cDNA clone of an attenuated G2b PEDV strain was successfully generated for the first time, and the in vitro and in vivo data indicate that iPEDVPT-P96 is further attenuated but remains immunogenic compared to its parental PEDVPT-P96 viral stock. The successful development of the iPEDVPT-P96 cDNA clone could allow for the manipulation of the viral genome to study viral pathogenesis and facilitate the rapid development of effective vaccines.
Collapse
Affiliation(s)
- Chi-Fei Kao
- School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan.
| | - Hue-Ying Chiou
- Graduate Institute of Veterinary Pathobiology, College of Veterinary Medicine, National Chung Hsing University, 250 Kuo Kuang Rd, Taichung 402, Taiwan.
| | - Yen-Chen Chang
- School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan.
| | - Cheng-Shun Hsueh
- Graduate Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan.
| | - Chian-Ren Jeng
- School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan.
- Graduate Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan.
| | - Pei-Shiue Tsai
- School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan.
| | - Ivan-Chen Cheng
- School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan.
| | - Victor Fei Pang
- School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan.
- Graduate Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan.
| | - Hui-Wen Chang
- School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan.
- Graduate Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan.
| |
Collapse
|
29
|
Chang CY, Hsu WT, Chao YC, Chang HW. Display of Porcine Epidemic Diarrhea Virus Spike Protein on Baculovirus to Improve Immunogenicity and Protective Efficacy. Viruses 2018; 10:v10070346. [PMID: 29954081 PMCID: PMC6071207 DOI: 10.3390/v10070346] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 06/15/2018] [Accepted: 06/15/2018] [Indexed: 01/23/2023] Open
Abstract
A new variant of the porcine epidemic diarrhea virus (PEDV) is an emerging swine disease, killing considerable numbers of neonatal piglets in North America and Asia in recent years. To generate immunogens mimicking the complex spike (S) protein folding with proper posttranslational modification to mount a robust immune response against the highly virulent PEDV, two baculoviruses displaying the full-length S protein (S-Bac) and the S1 protein (S1-Bac) of the virulent Taiwan genotype 2b (G2b) PEDV Pintung 52 (PEDV-PT) strain were constructed. Intramuscular immunizations of mice and piglets with the S-Bac and S1-Bac demonstrated significantly higher levels of systemic anti-PEDV S-specific IgG, as compared with control group. Our results also showed that piglets in the S-Bac group elicited superior PEDV-specific neutralizing antibodies than those of the S1-Bac and control groups. The highly virulent PEDV-PT strain challenge experiment showed that piglets immunized with S-Bac and S1-Bac showed milder clinical symptoms with significantly less fecal viral shedding as compared with non-immunized control piglets. More importantly, piglets immunized with the S-Bac exhibited no to mild clinical signs, with a delayed, minimal viral shedding. Our results demonstrated that the S-Bac could serve as a safe, easy to manipulate, and effective vaccine candidate against the PEDV infection.
Collapse
Affiliation(s)
- Chia-Yu Chang
- School of Veterinary Medicine, National Taiwan University, Taipei 106, Taiwan.
| | - Wei-Ting Hsu
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei 115, Taiwan.
| | - Yu-Chan Chao
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei 115, Taiwan.
| | - Hui-Wen Chang
- School of Veterinary Medicine, National Taiwan University, Taipei 106, Taiwan.
- Graduate Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, Taipei 106, Taiwan.
| |
Collapse
|