1
|
Shi Q, Wang Q, Shen Y, Chen S, Gan S, Lin T, Song F, Ma Y. Escherichia coli LTB26 mutant enhances immune responses to rotavirus antigen VP8 in a mouse model. Mol Immunol 2024; 173:10-19. [PMID: 39004021 DOI: 10.1016/j.molimm.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 06/03/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024]
Abstract
Adjuvant is a major supplementary component of vaccines to boost adaptive immune responses. To select an efficient adjuvant from the heat-labile toxin B subunit (LTB) of E. coli, four LTB mutants (numbered LTB26, LTB34, LTB57, and LTB85) were generated by multi-amino acid random replacement. Mice have been intranasally vaccinated with human rotavirus VP8 admixed. Among the four mutants, enzyme-linked immunosorbent assay (ELISA) revealed that LTB26 had enhanced mucosal immune adjuvanticity compared to LTB, showing significantly enhanced immune responses in both serum IgG and mucosal sIgA levels. The 3D modeling analysis suggested that the enhanced immune adjuvanticity of LTB26 might be due to the change of the first LTB α-helix to a β-sheet. The molecular mechanism was studied using transcriptomic and flow cytometric (FCM) analysis. The transcriptomic data demonstrated that LTB26 enhanced immune response by enhancing B cell receptor (BCR) and major histocompatibility complex (MHC) II+-related pathways. Furthermore, LTB26 promoted Th1 and Th2-type immune responses which were confirmed by detecting IFN-γ and IL-4 expression levels. Immunohistochemical analysis demonstrated that LTB26 enhanced both Th1 and Th2 type immunity. Therefore, LTB26 was a potent mucosal immune adjuvant meeting the requirement for use in human clinics in the future.
Collapse
Affiliation(s)
- Qinlin Shi
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Qiujuan Wang
- Department of Biochemistry and Molecular Biology, Basic Medical College, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No 1, Chongqing 400016, China
| | - Yanxi Shen
- Department of Biochemistry and Molecular Biology, Basic Medical College, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No 1, Chongqing 400016, China
| | - Sijing Chen
- Department of Biochemistry and Molecular Biology, Basic Medical College, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No 1, Chongqing 400016, China
| | - Sijie Gan
- Department of Biochemistry and Molecular Biology, Basic Medical College, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No 1, Chongqing 400016, China
| | - Tao Lin
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Fangzhou Song
- Department of Biochemistry and Molecular Biology, Basic Medical College, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No 1, Chongqing 400016, China
| | - Yongping Ma
- Department of Biochemistry and Molecular Biology, Basic Medical College, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No 1, Chongqing 400016, China.
| |
Collapse
|
2
|
Liu S, Yu Q, Li S, Li M, Yang L, Wang Q, Tu Z, Tao F, Yang P, Kong L, Xin X. Expression and immunogenicity of recombinant porcine epidemic diarrhea virus Nsp9. Virology 2023; 587:109861. [PMID: 37572518 DOI: 10.1016/j.virol.2023.109861] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 08/14/2023]
Abstract
Porcine epidemic diarrhea virus (PEDV) causes acute diarrhea, vomiting, dehydration, and high mortality in newborn piglets, which leads to significant economic losses. Coronavirus nonstructural protein 9 (Nsp9) is an essential RNA binding protein for coronavirus replication, which renders it a promising candidate for developing antiviral drugs and diagnosis targeting PEDV. In this study, PEDV Nsp9 protein fused with MBP protein and His-tag were expressed and purified in Escherichia coli. Furthermore, immunization of MBP-Nsp9 enhances both humoral and cellular immunity responses as compared with that of His-Nsp9 protein. Finally, the swine immunization showed that Nsp9 protein could stimulate the swine immunity system to carry out humoral immunity, and the generated antibody could inhibit the proliferation of PEDV in Vero cells. Altogether, our data provide direct evidence for the immunogenicity of PEDV Nsp9, which sheds light on the future developments of anti-PEDV drugs and vaccines for PED prevention.
Collapse
Affiliation(s)
- Shiguo Liu
- Institute of Pathogenic Microorganism and College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi, China; Nanchang Key Laboratory of Animal Virus and Genetic Engineering, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Qijia Yu
- Institute of Pathogenic Microorganism and College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi, China; Nanchang Key Laboratory of Animal Virus and Genetic Engineering, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Sha Li
- Institute of Pathogenic Microorganism and College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi, China; Nanchang Key Laboratory of Animal Virus and Genetic Engineering, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Mingzhi Li
- Institute of Pathogenic Microorganism and College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi, China; Nanchang Key Laboratory of Animal Virus and Genetic Engineering, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Li Yang
- Institute of Pathogenic Microorganism and College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi, China; Nanchang Key Laboratory of Animal Virus and Genetic Engineering, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Quansheng Wang
- Institute of Pathogenic Microorganism and College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi, China; Nanchang Key Laboratory of Animal Virus and Genetic Engineering, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Zewen Tu
- Institute of Pathogenic Microorganism and College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi, China; Nanchang Key Laboratory of Animal Virus and Genetic Engineering, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Feifei Tao
- Institute of Pathogenic Microorganism and College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi, China; Nanchang Key Laboratory of Animal Virus and Genetic Engineering, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Pingping Yang
- Institute of Pathogenic Microorganism and College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi, China; Nanchang Key Laboratory of Animal Virus and Genetic Engineering, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Lingbao Kong
- Institute of Pathogenic Microorganism and College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi, China; Nanchang Key Laboratory of Animal Virus and Genetic Engineering, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Xiu Xin
- Institute of Pathogenic Microorganism and College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi, China; Nanchang Key Laboratory of Animal Virus and Genetic Engineering, Jiangxi Agricultural University, Nanchang, 330045, China.
| |
Collapse
|
3
|
Zhang M, Chen X, Xue M, Jiang N, Li Y, Fan Y, Zhang P, Liu N, Xiao Z, Zhang Q, Zhou Y. Oral Vaccination of Largemouth Bass (Micropterus salmoides) against Largemouth Bass Ranavirus (LMBV) Using Yeast Surface Display Technology. Animals (Basel) 2023; 13:ani13071183. [PMID: 37048441 PMCID: PMC10093309 DOI: 10.3390/ani13071183] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/20/2023] [Accepted: 03/25/2023] [Indexed: 03/31/2023] Open
Abstract
Largemouth bass ranavirus (LMBV) infects largemouth bass, leading to significant mortality and economic losses. There are no safe and effective drugs against this disease. Oral vaccines that directly target the intestinal mucosal immune system play an important role in resisting pathogens. Herein, the B subunit of Escherichia coli heat-labile enterotoxin (LTB, a mucosal immune adjuvant) and the LMBV main capsid protein (MCP) were expressed using Saccharomyces cerevisiae surface display technology. The yeast-prepared oral vaccines were named EBY100-OMCP and EBY100-LTB-OMCP. The candidate vaccines could resist the acidic intestinal environment. After 7 days of continuous oral immunization, indicators of innate and adaptive immunity were measured on days 1, 7, 14, 21, 28, 35, and 42. High activities of immune enzymes (T-SOD, AKP, ACP, and LZM) in serum and intestinal mucus were detected. IgM in the head kidney was significantly upregulated (EBY100-OMCP group: 3.8-fold; BY100-LTB-OMCP group: 4.3-fold). IgT was upregulated in the intestines (EBY100-OMCP group: 5.6-fold; EBY100-LTB-OMCP group: 6.7-fold). Serum neutralizing antibody titers of the two groups reached 1:85. Oral vaccination protected against LMBV infection. The relative percent survival was 52.1% (EBY100-OMCP) and 66.7% (EBY100-LTB-OMCP). Thus, EBY100-OMCP and EBY100-LTB-OMCP are promising and effective candidate vaccines against LMBV infection.
Collapse
|
4
|
Immunogenicity analysis of the E. coli expressed structural protein VP1 of persistent infection foot-and-mouth disease virus. Virology 2023; 579:111-118. [PMID: 36634474 DOI: 10.1016/j.virol.2023.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 12/31/2022] [Accepted: 01/04/2023] [Indexed: 01/09/2023]
Abstract
The persistent infection of FMDV in cloven hoofed animals has made the epidemic prevention and control more difficult. VP1 is the main immunogenic protein and first candidate of vaccine development for FMDV prevention. However, the mutation of VP1 in host cell with persistent infection FMDV (PI-FMDV) caused the change of its immunogenicity. Hence, it is imperative to establish the expression system for VP1 of PI-FMDV (PI-VP1) and re-evaluate its immunogenicity. In this study, the PI-VP1 with His-tag was cloned into pET-28a vector. PI-VP1 protein was expressed and purified in E. coli, and further the antiserum of immunized mice was analyzed. Results showed that purified PI-VP1 protein produced a good humoral and cellular immune response after immunizing mice. Furthermore, our study showed that the antiserum could not only neutralize PI-FMDV, but also prevent the adsorption of WT-FMDV. In summarize, our work provides valuable implications for the FMDV vaccines and therapeutics development.
Collapse
|
5
|
Wang Y, Ling L, Zhang Z, Marin-Lopez A. Current Advances in Zika Vaccine Development. Vaccines (Basel) 2022; 10:vaccines10111816. [PMID: 36366325 PMCID: PMC9694033 DOI: 10.3390/vaccines10111816] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/19/2022] [Accepted: 10/25/2022] [Indexed: 11/16/2022] Open
Abstract
Zika virus (ZIKV), an emerging arthropod-borne flavivirus, was first isolated in Uganda in 1947 from monkeys and first detected in humans in Nigeria in 1952; it has been associated with a dramatic burden worldwide. Since then, interventions to reduce the burden of ZIKV infection have been mainly restricted to mosquito control, which in the end proved to be insufficient by itself. Hence, the situation prompted scientists to increase research on antivirals and vaccines against the virus. These efforts are still ongoing as the pathogenesis and immune evasion mechanisms of ZIKV have not yet been fully elucidated. Understanding the viral disease mechanism will provide a better landscape to develop prophylactic and therapeutic strategies against ZIKV. Currently, no specific vaccines or drugs have been approved for ZIKV. However, some are undergoing clinical trials. Notably, different platforms have been evaluated for the design of vaccines, including DNA, mRNA, viral vectors, virus-like particles (VLPs), inactivated virus, live attenuated virus, peptide and protein-based vaccines, passive immunizations by using monoclonal antibodies (MAbs), and vaccines that target vector-derived antigens. These vaccines have been shown to induce specific humoral and cellular immune responses and reduce viremia and viral RNA titers, both in vitro and in vivo. This review provides a comprehensive summary of current advancements in the development of vaccines against Zika virus.
Collapse
Affiliation(s)
- Yuchen Wang
- Department of Inspection and Quarantine Technology Communication, Shanghai Customs College, Shanghai 201204, China
- Correspondence:
| | - Lin Ling
- Department of Inspection and Quarantine Technology Communication, Shanghai Customs College, Shanghai 201204, China
| | - Zilei Zhang
- Department of Inspection and Quarantine Technology Communication, Shanghai Customs College, Shanghai 201204, China
| | - Alejandro Marin-Lopez
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06420, USA
| |
Collapse
|
6
|
Feng T, Li M, Zhang L, Li S, Yang Z, Kang L, Guo Y, Kong L, Wang T. Immunity of two novel hepatitis C virus polyepitope vaccines. Vaccine 2022; 40:6277-6287. [PMID: 36150975 DOI: 10.1016/j.vaccine.2022.09.020] [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: 05/26/2022] [Revised: 08/01/2022] [Accepted: 09/05/2022] [Indexed: 11/17/2022]
Abstract
Hepatitis C virus (HCV) infection remains a serious public health burden around the world. So far there is no effective vaccine against this virus. Neutralizing antibody (NAb) responses to the epitopes within HCV E1 and E2 proteins are related to the resolution of hepatitis C infection. E. coli heat-labile enterotoxin B subunit (LTB) has been described as potent immunity adjuvants. In this study, we constructed recombinant pET vectors: pET-R9-Bp (B cell polyepitopes) expressing 7 epitopes from HCV E1 and E2 proteins including R9 (E2384-411aa)-Bp (E1313-327aa-E2396-424aa-E2436-447aa-E2523-540aa-E2610-627aa-E2631-648aa) and pET-LTB-R9-Bp expressing LTB adjuvant in combination with R9-Bp. Recombinant proteins R9-Bp and LTB-R9-Bp were expressed successfully in E. coli and purified by the Ni-NTA column. Both R9-Bp and LTB-R9-Bp in BALB/c mice induced robust humoral immune response in the context of intraperitoneal or intramuscular immunization but not oral immunization. Intraperitoneal administration of LTB-R9-Bp induced a higher antibody titer (peak titer: 1:341000) than that of R9-Bp (peak titer: 1:85000) after the second boost (P = 0.0036 or 0.0002). However, comparable antibody peak titers were elicited for both R9-Bp and LTB-R9-Bp in intramuscular immunization albeit with significant difference (P = 0.0032) a week after the second boost. In addition, both R9-Bp and LTB-R9-Bp induced the secretion of cytokines including IFN-γ and IL-4 at similar levels. anti-sera induced by both R9-Bp and LTB-R9-Bp recognized native HCV E1 and E2 proteins. Moreover, these HCV-specific antisera inhibited significantly the entry of HCV (P < 0.0001). Taken together, these findings showed that E. coli-based both R9-Bp and LTB-R9-Bp could become promising HCV vaccines.
Collapse
Affiliation(s)
- Tian Feng
- Nanchang City Key Laboratory of Animal Virus and Genetic Engineering, Nanchang, Jiangxi, China; Institute of Pathogenic Microorganism, Jiangxi Agricultural University, Nanchang, Jiangxi, China; College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Mingzhi Li
- Nanchang City Key Laboratory of Animal Virus and Genetic Engineering, Nanchang, Jiangxi, China; Institute of Pathogenic Microorganism, Jiangxi Agricultural University, Nanchang, Jiangxi, China; College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Lirong Zhang
- Nanchang City Key Laboratory of Animal Virus and Genetic Engineering, Nanchang, Jiangxi, China; Institute of Pathogenic Microorganism, Jiangxi Agricultural University, Nanchang, Jiangxi, China; College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Sha Li
- Nanchang City Key Laboratory of Animal Virus and Genetic Engineering, Nanchang, Jiangxi, China; Institute of Pathogenic Microorganism, Jiangxi Agricultural University, Nanchang, Jiangxi, China; College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Zibing Yang
- Nanchang City Key Laboratory of Animal Virus and Genetic Engineering, Nanchang, Jiangxi, China; Institute of Pathogenic Microorganism, Jiangxi Agricultural University, Nanchang, Jiangxi, China; College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Lumei Kang
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China; Center for Laboratory Animal Science, Nanchang University, Nanchang, Jiangxi, China
| | - Yunli Guo
- Nanchang City Key Laboratory of Animal Virus and Genetic Engineering, Nanchang, Jiangxi, China; Institute of Pathogenic Microorganism, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Lingbao Kong
- Nanchang City Key Laboratory of Animal Virus and Genetic Engineering, Nanchang, Jiangxi, China; Institute of Pathogenic Microorganism, Jiangxi Agricultural University, Nanchang, Jiangxi, China; College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi, China.
| | - Ting Wang
- Nanchang City Key Laboratory of Animal Virus and Genetic Engineering, Nanchang, Jiangxi, China; Institute of Pathogenic Microorganism, Jiangxi Agricultural University, Nanchang, Jiangxi, China; College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi, China.
| |
Collapse
|
7
|
Kumar S, Verma A, Yadav P, Dubey SK, Azhar EI, Maitra SS, Dwivedi VD. Molecular pathogenesis of Japanese encephalitis and possible therapeutic strategies. Arch Virol 2022; 167:1739-1762. [PMID: 35654913 PMCID: PMC9162114 DOI: 10.1007/s00705-022-05481-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 03/10/2022] [Indexed: 12/26/2022]
Abstract
Japanese encephalitis virus (JEV), a single-stranded, enveloped RNA virus, is a health concern across Asian countries, associated with severe neurological disorders, especially in children. Primarily, pigs, bats, and birds are the natural hosts for JEV, but humans are infected incidentally. JEV requires a few host proteins for its entry and replication inside the mammalian host cell. The endoplasmic reticulum (ER) plays a significant role in JEV genome replication and assembly. During this process, the ER undergoes stress due to its remodelling and accumulation of viral particles and unfolded proteins, leading to an unfolded protein response (UPR). Here, we review the overall strategy used by JEV to infect the host cell and various cytopathic effects caused by JEV infection. We also highlight the role of JEV structural proteins (SPs) and non-structural proteins (NSPs) at various stages of the JEV life cycle that are involved in up- and downregulation of different host proteins and are potentially relevant for developing efficient therapeutic drugs.
Collapse
Affiliation(s)
- Sanjay Kumar
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067 India
- Center for Bioinformatics, Computational and Systems Biology, Pathfinder Research and Training Foundation, Greater Noida, India
| | - Akanksha Verma
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067 India
| | - Pardeep Yadav
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh 201310 India
- Center for Bioinformatics, Computational and Systems Biology, Pathfinder Research and Training Foundation, Greater Noida, India
| | | | - Esam Ibraheem Azhar
- Special Infectious Agents Unit-BSL3, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, 21589 Saudi Arabia
| | - S. S. Maitra
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067 India
| | - Vivek Dhar Dwivedi
- Center for Bioinformatics, Computational and Systems Biology, Pathfinder Research and Training Foundation, Greater Noida, India
| |
Collapse
|
8
|
Li M, Weng S, Wang Q, Yang Z, Wang X, Yin Y, Zhou Q, Zhang L, Tao F, Li Y, Jia M, Yang L, Xin X, Li H, Kang L, Wang Y, Wang T, Li S, Kong L. Reduced binding activity of vaccine serum to omicron receptor-binding domain. Front Immunol 2022; 13:960195. [PMID: 35967350 PMCID: PMC9369000 DOI: 10.3389/fimmu.2022.960195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) vaccination regimens contribute to limiting the spread of severe acute respiratory syndrome Coronavirus-2 (SARS-CoV-2). However, the emergence and rapid transmission of the SARS-CoV-2 variant Omicron raise a concern about the efficacy of the current vaccination strategy. Here, we expressed monomeric and dimeric receptor-binding domains (RBDs) of the spike protein of prototype SARS-CoV-2 and Omicron variant in E. coli and investigated the reactivity of anti-sera from Chinese subjects immunized with SARS-CoV-2 vaccines to these recombinant RBDs. In 106 human blood samples collected from 91 participants from Jiangxi, China, 26 sera were identified to be positive for SARS-CoV-2 spike protein antibodies by lateral flow dipstick (LFD) assays, which were enriched in the ones collected from day 7 to 1 month post-boost (87.0%) compared to those harvested within 1 week post-boost (23.8%) (P < 0.0001). A higher positive ratio was observed in the child group (40.8%) than adults (13.6%) (P = 0.0073). ELISA results showed that the binding activity of anti-SARS-CoV-2 antibody-positive sera to Omicron RBDs dropped by 1.48- to 2.07-fold compared to its homogeneous recombinant RBDs. Thus, our data indicate that current SARS-CoV-2 vaccines provide restricted humoral protection against the Omicron variant.
Collapse
Affiliation(s)
- Mingzhi Li
- Nanchang City Key Laboratory of Animal Virus and Genetic Engineering, Institute of Pathogenic Microorganism, College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, China
| | - Shiqi Weng
- Nanchang City Key Laboratory of Animal Virus and Genetic Engineering, Institute of Pathogenic Microorganism, College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, China
| | - Quansheng Wang
- Nanchang City Key Laboratory of Animal Virus and Genetic Engineering, Institute of Pathogenic Microorganism, College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, China
| | - Zibing Yang
- Nanchang City Key Laboratory of Animal Virus and Genetic Engineering, Institute of Pathogenic Microorganism, College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, China
| | - Xiaoling Wang
- Nanchang City Key Laboratory of Animal Virus and Genetic Engineering, Institute of Pathogenic Microorganism, College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, China
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yanjun Yin
- Department of Clinical Laboratory, Jiangxi Provincial Children’s Hospital, Nanchang, China
| | - Qiuxiang Zhou
- Department of Clinical Laboratory, The Affiliated Hospital of Jiangxi Agricultural University, Nanchang, China
| | - Lirong Zhang
- Nanchang City Key Laboratory of Animal Virus and Genetic Engineering, Institute of Pathogenic Microorganism, College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, China
| | - Feifei Tao
- Nanchang City Key Laboratory of Animal Virus and Genetic Engineering, Institute of Pathogenic Microorganism, College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, China
| | - Yihan Li
- Nanchang City Key Laboratory of Animal Virus and Genetic Engineering, Institute of Pathogenic Microorganism, College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, China
| | - Mengle Jia
- Nanchang City Key Laboratory of Animal Virus and Genetic Engineering, Institute of Pathogenic Microorganism, College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, China
| | - Lingdi Yang
- Nanchang City Key Laboratory of Animal Virus and Genetic Engineering, Institute of Pathogenic Microorganism, College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, China
| | - Xiu Xin
- Nanchang City Key Laboratory of Animal Virus and Genetic Engineering, Institute of Pathogenic Microorganism, College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, China
| | - Hanguang Li
- Nanchang City Key Laboratory of Animal Virus and Genetic Engineering, Institute of Pathogenic Microorganism, College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, China
| | - Lumei Kang
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
- Center for Laboratory Animal Science, Nanchang University, Nanchang, China
| | - Yu Wang
- Nanchang City Key Laboratory of Animal Virus and Genetic Engineering, Institute of Pathogenic Microorganism, College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, China
- *Correspondence: Yu Wang, ; Ting Wang, ; Sha Li, ; Lingbao Kong,
| | - Ting Wang
- Nanchang City Key Laboratory of Animal Virus and Genetic Engineering, Institute of Pathogenic Microorganism, College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, China
- *Correspondence: Yu Wang, ; Ting Wang, ; Sha Li, ; Lingbao Kong,
| | - Sha Li
- Nanchang City Key Laboratory of Animal Virus and Genetic Engineering, Institute of Pathogenic Microorganism, College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, China
- *Correspondence: Yu Wang, ; Ting Wang, ; Sha Li, ; Lingbao Kong,
| | - Lingbao Kong
- Nanchang City Key Laboratory of Animal Virus and Genetic Engineering, Institute of Pathogenic Microorganism, College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, China
- *Correspondence: Yu Wang, ; Ting Wang, ; Sha Li, ; Lingbao Kong,
| |
Collapse
|
9
|
Han X, Cai Z, Dai Y, Huang H, Cao X, Wang Y, Fang Y, Liu G, Zhang M, Zhang Y, Yang B, Xue W, Zhao G, Tai W, Li M. Re-burying Artificially Exposed Surface of Viral Subunit Vaccines Through Oligomerization Enhances Vaccine Efficacy. Front Cell Infect Microbiol 2022; 12:927674. [PMID: 35846760 PMCID: PMC9278648 DOI: 10.3389/fcimb.2022.927674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 06/02/2022] [Indexed: 12/02/2022] Open
Abstract
Viral subunit vaccines often suffer low efficacy. We recently showed that when taken out of the context of whole virus particles, recombinant subunit vaccines contain artificially exposed surface regions that are non-neutralizing and reduce their efficacy, and thus these regions need to be re-buried in vaccine design. Here we used the envelope protein domain III (EDIII) of Japanese encephalitis virus (JEV), a subunit vaccine candidate, to further validate this important concept for subunit vaccine designs. We constructed monomeric EDIII, dimeric EDIII via a linear space, dimeric EDIII via an Fc tag, and trimeric EDIII via a foldon tag. Compared to monomeric EDIII or linearly linked dimeric EDIII, tightly packed EDIII oligomers via the Fc or foldon tag induce higher neutralizing antibody titers in mice and also protect mice more effectively from lethal JEV challenge. Structural analyses demonstrate that part of the artificially exposed surface areas on recombinant EDIII becomes re-buried in Fc or foldon-mediated oligomers. This study further establishes the artificially exposed surfaces as an intrinsic limitation of subunit vaccines, and suggests that re-burying these surfaces through tightly packed oligomerization is a convenient and effective approach to overcome this limitation.
Collapse
Affiliation(s)
- Xuelian Han
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Zhuming Cai
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, China
| | - Yulong Dai
- Institute of Hemu Biotechnology, Beijing Hemu Biotechnology Co., Ltd, Beijing, China
| | - He Huang
- Institute of Hemu Biotechnology, Beijing Hemu Biotechnology Co., Ltd, Beijing, China
| | - Xiangwen Cao
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China.,Public Health School, Mudanjiang Medical University, Mudanjiang, China
| | - Yuan Wang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Yingying Fang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China.,School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Gang Liu
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Min Zhang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Yuhang Zhang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Binhui Yang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Wei Xue
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Guangyu Zhao
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China.,Public Health School, Mudanjiang Medical University, Mudanjiang, China
| | - Wanbo Tai
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, China
| | - Min Li
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| |
Collapse
|
10
|
Chen T, Zhu S, Wei N, Zhao Z, Niu J, Si Y, Cao S, Ye J. Protective Immune Responses Induced by an mRNA-LNP Vaccine Encoding prM-E Proteins against Japanese Encephalitis Virus Infection. Viruses 2022; 14:1121. [PMID: 35746593 PMCID: PMC9227124 DOI: 10.3390/v14061121] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 05/17/2022] [Accepted: 05/19/2022] [Indexed: 02/01/2023] Open
Abstract
Japanese encephalitis virus (JEV) is an important zoonotic pathogen, which causes central nervous system symptoms in humans and reproductive disorders in swine. It has led to severe impacts on human health and the swine industry; however, there is no medicine available for treating yet. Therefore, vaccination is the best preventive measure for this disease. In the study, a modified mRNA vaccine expressing the prM and E proteins of the JEV P3 strain was manufactured, and a mouse model was used to assess its efficacy. The mRNA encoding prM and E proteins showed a high level of protein expression in vitro and were encapsulated into a lipid nanoparticle (LNP). Effective neutralizing antibodies and CD8+ T-lymphocytes-mediated immune responses were observed in vaccinated mice. Furthermore, the modified mRNA can protect mice from a lethal challenge with JEV and reduce neuroinflammation caused by JEV. This study provides a new option for the JE vaccine and lays a foundation for the subsequent development of a more efficient and safer JEV mRNA vaccine.
Collapse
Affiliation(s)
- Tao Chen
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (T.C.); (S.Z.); (N.W.); (Z.Z.); (J.N.); (Y.S.)
- Laboratory of Animal Virology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Shuo Zhu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (T.C.); (S.Z.); (N.W.); (Z.Z.); (J.N.); (Y.S.)
- Laboratory of Animal Virology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Ning Wei
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (T.C.); (S.Z.); (N.W.); (Z.Z.); (J.N.); (Y.S.)
- Laboratory of Animal Virology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Zikai Zhao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (T.C.); (S.Z.); (N.W.); (Z.Z.); (J.N.); (Y.S.)
- Laboratory of Animal Virology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Junjun Niu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (T.C.); (S.Z.); (N.W.); (Z.Z.); (J.N.); (Y.S.)
- Laboratory of Animal Virology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Youhui Si
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (T.C.); (S.Z.); (N.W.); (Z.Z.); (J.N.); (Y.S.)
- Laboratory of Animal Virology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Shengbo Cao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (T.C.); (S.Z.); (N.W.); (Z.Z.); (J.N.); (Y.S.)
- Laboratory of Animal Virology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Jing Ye
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (T.C.); (S.Z.); (N.W.); (Z.Z.); (J.N.); (Y.S.)
- Laboratory of Animal Virology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
11
|
Zhu L, Liu S, Zhuo Z, Lin Y, Zhang Y, Wang X, Kong L, Wang T. Expression and immunogenicity of nsp10 protein of porcine epidemic diarrhea virus. Res Vet Sci 2022; 144:34-43. [PMID: 35038674 PMCID: PMC8721950 DOI: 10.1016/j.rvsc.2021.12.024] [Citation(s) in RCA: 2] [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: 04/08/2021] [Revised: 12/01/2021] [Accepted: 12/28/2021] [Indexed: 12/16/2022]
Abstract
Porcine epidemic diarrhea virus (PEDV), a swine enteropathogenic coronavirus, causes lethal watery diarrhea to the piglets, which poses significant economic losses and public health concerns. The nsp10 protein of PEDV is essential regulatory subunits that are critical for virus replication. Since PEDV nsp10 is a crucial regulator of viral RNA synthesis, it is promising that nsp10 might become anti-virus drugs target or candidate for rapid diagnosis of PEDV infection. In this study, the PEDV nsp10 was inserted into pMAL-c2x-MBP / pET-28a vector, efficiently and stably expressed in E.coli system. Then the purified nsp10 protein was found to mediate potent antibody responses in immunized mice. The antibodies of immunized mice and PEDV infection swine strongly recognized purified nsp10 protein from cell lysates. Furthermore, cytokines test revealed that the expression of IL-2, IL-4, IL-10, TNF-α, IFN-γ were significantly higher than those in control group, indicated that purified nsp10 protein induce the cellular immune response mechanism in mice. Using modified seroneutralization test, we also demonstrated that sera from nsp10-immunized mice inhibited PEDV replication to some extent. These findings suggest that nsp10 has a high immunogenicity. This study may have implications for future development of PEDV detection or anti-virus drugs for swine.
Collapse
Affiliation(s)
- Liting Zhu
- Institute of Pathogenic Microorganism, College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Shiguo Liu
- Institute of Pathogenic Microorganism, College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Zewen Zhuo
- Institute of Pathogenic Microorganism, College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Yanxi Lin
- Institute of Pathogenic Microorganism, College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Yanni Zhang
- Jiangxi Province Center for Disease Control and Prevention, Nanchang, Jiangxi, China
| | - Xiaoling Wang
- Institute of Pathogenic Microorganism, College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Lingbao Kong
- Institute of Pathogenic Microorganism, College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Ting Wang
- Institute of Pathogenic Microorganism, College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi, China.
| |
Collapse
|