1
|
Li W, Li YA, Wang S, Shi H. A universal live vaccine platform against multiple serotypes Streptococcus suis based on polyvalent antigen protein. Vaccine 2025; 47:126700. [PMID: 39778475 DOI: 10.1016/j.vaccine.2024.126700] [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: 09/12/2024] [Revised: 12/09/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025]
Abstract
Streptococcus suis (S. suis) is a major pathogen that poses a long-term threat to swine populations. Due to its foodborne transmission, this pathogen has recently emerged as a leading cause of meningitis in humans, presenting a significant public health challenge. Currently, no vaccine is available to combat this disease, particularly a universal vaccine capable of addressing multiple subtypes of S. suis. In this study, we developed a universal live vaccine candidate against multiple serotypes S. suis based on the polyvalent antigen protein SE6. A live Salmonella Choleraesuis (S. Choleraesuis) vector was employed for the production and in vivo delivery of the polyvalent antigen. The SE6 protein was efficiently expressed within the S. Choleraesuis vector and delivered to the host's lymphatic system. The antiserum of mice immunized with SE6-delivering S. Choleraesuis vector produced a broader and potent opsonophagocytic response against multiple serotypes of S. suis. Finally, the SE6-delivering S. Choleraesuis vector demonstrated high efficacy in providing protection against S. suis serotypes 2, 7, and 9 in vivo.
Collapse
Affiliation(s)
- Wenjing Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yu-An Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.
| | - Shifeng Wang
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611-0880, USA.
| | - Huoying Shi
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University (JIRLAAPS), Yangzhou, China.
| |
Collapse
|
2
|
Jacobs-Lorena M, Cha SJ. Unbiased phage display screening identifies hidden malaria vaccine targets. Emerg Microbes Infect 2024; 13:2429617. [PMID: 39529575 PMCID: PMC11587725 DOI: 10.1080/22221751.2024.2429617] [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: 07/05/2024] [Revised: 10/18/2024] [Accepted: 11/10/2024] [Indexed: 11/16/2024]
Abstract
Malaria is among the deadliest infectious diseases. Over 200 million annual clinical malaria cases are reported and more than half a million people, mostly children, die every year. The most advanced RTS,S/AS01 vaccine based on the P. falciparum circumsporozoite protein (CSP), targets sporozoite liver infection but achieved modest efficacy. To reduce malaria death, novel malaria vaccine development is a high priority. Most malaria vaccine candidates target three infection steps: sporozoite liver infection, merozoite red blood cell (RBC) infection, and mosquito midgut infection. However, only few malaria vaccine candidates target specific parasite-host cell interactions. Our group has implemented the phage peptide-display approach to discover new parasite ligands and host cell receptors. Here we summarize our findings and discuss their potential for the development of novel vaccines.
Collapse
Affiliation(s)
- Marcelo Jacobs-Lorena
- Department of Molecular Microbiology and Immunology and Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Sung-Jae Cha
- Department of Medical Sciences, Mercer University School of Medicine, Macon, GA, USA
| |
Collapse
|
3
|
Liu J, Zhang Z, Pu W, Pan X, Li P, Bai Q, Liang S, Li C, Yu Y, Yao H, Ma J. A multi-epitope subunit vaccine providing broad cross-protection against diverse serotypes of Streptococcus suis. NPJ Vaccines 2024; 9:216. [PMID: 39543108 PMCID: PMC11564553 DOI: 10.1038/s41541-024-01015-7] [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: 08/08/2024] [Accepted: 10/28/2024] [Indexed: 11/17/2024] Open
Abstract
Streptococcus suis infection represents a major challenge in pig farming and public health due to its zoonotic potential and diverse serotypes, while existing vaccines lack effective cross-protection. This study employed reverse vaccinology and immunoinformatics to identify 8 conserved proteins across 11 prevalent serotypes of S. suis. 16 candidate epitopes were selected to design three multi-epitope antigens against S. suis (designated as MEASs), which fused with a dendritic cell-targeting peptide to improve antigen presentation in host. Purified MEASs displayed favorable cross-reactogenicity against 29 serotype-specific antiserums. Robust humoral and cellular immune responses can be induced by MEAS 1 and MEAS 3 in a mouse model, which provided substantial protection against virulent strains from two different serotypes. In particular, their immune serums exhibited positive opsonization effects within bloodstream and macrophage phagocytosis. Taken together, we identified two promising MEASs with excellent cross-protection, offering potential in preventing S. suis infections in a mouse model.
Collapse
Affiliation(s)
- Jianan Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China
- WOAH Reference Lab for Swine Streptococcosis, Nanjing, 210095, China
| | - Zhen Zhang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China
- WOAH Reference Lab for Swine Streptococcosis, Nanjing, 210095, China
| | - Wanxia Pu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China
- WOAH Reference Lab for Swine Streptococcosis, Nanjing, 210095, China
| | - Xinming Pan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China
- WOAH Reference Lab for Swine Streptococcosis, Nanjing, 210095, China
| | - Pei Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China
- WOAH Reference Lab for Swine Streptococcosis, Nanjing, 210095, China
| | - Qiankun Bai
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China
- WOAH Reference Lab for Swine Streptococcosis, Nanjing, 210095, China
| | - Song Liang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China
- WOAH Reference Lab for Swine Streptococcosis, Nanjing, 210095, China
| | - Caiying Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China
- WOAH Reference Lab for Swine Streptococcosis, Nanjing, 210095, China
| | - Yong Yu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China.
- WOAH Reference Lab for Swine Streptococcosis, Nanjing, 210095, China.
| | - Huochun Yao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China.
- WOAH Reference Lab for Swine Streptococcosis, Nanjing, 210095, China.
| | - Jiale Ma
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China.
- WOAH Reference Lab for Swine Streptococcosis, Nanjing, 210095, China.
| |
Collapse
|
4
|
Yan Z, Pan R, Zhang J, Sun J, Ma X, Dong N, Yao X, Wei J, Liu K, Qiu Y, Sealey K, Nichols H, Jarvis MA, Upton M, Li X, Ma Z, Liu J, Li B. Immunogenicity and Protective Capacity of Sugar ABC Transporter Substrate-Binding Protein against Streptococcus suis Serotype 2, 7 and 9 Infection in Mice. Vaccines (Basel) 2024; 12:544. [PMID: 38793795 PMCID: PMC11126002 DOI: 10.3390/vaccines12050544] [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: 03/15/2024] [Revised: 05/10/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
Background:Streptococcus suis (S. suis) is a Gram-positive bacterium that causes substantial disease in pigs. S. suis is also an emerging zoonoses in humans, primarily in Asia, through the consumption of undercooked pork and the handling of infected pig meat as well as carcasses. The complexity of S. suis epidemiology, characterized by the presence of multiple bacterial serotypes and strains with diverse sequence types, identifies a critical need for a universal vaccine with the ability to confer cross-protective immunity. Highly conserved immunogenic proteins are generally considered good candidate antigens for subunit universal vaccines. Methods: In this study, the cross-protection of the sugar ABC transporter substrate-binding protein (S-ABC), a surface-associated immunogenic protein of S. suis, was examined in mice for evaluation as a universal vaccine candidate. Results: S-ABC was shown to be highly conserved, with 97% amino acid sequence identity across 31 S. suis strains deposited in GenBank. Recombinantly expressed S-ABC (rS-ABC) was recognized via rabbit sera specific to S. suis serotype 2. The immunization of mice with rS-ABC induced antigen-specific antibody responses, as well as IFN-γ and IL-4, in multiple organs, including the lungs. rS-ABC immunization conferred high (87.5% and 100%) protection against challenges with S. suis serotypes 2 and 9, demonstrating high cross-protection against these serotypes. Protection, albeit lower (50%), was also observed in mice challenged with S. suis serotype 7. Conclusions: These data identify S-ABC as a promising antigenic target within a universal subunit vaccine against S. suis.
Collapse
Affiliation(s)
- Zujie Yan
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (Z.Y.); (R.P.); (J.Z.); (X.M.); (N.D.); (X.Y.); (J.W.); (K.L.); (Y.Q.); (Z.M.)
- College of Veterinary Medicine, Hebei Agricultural University, Baoding 071000, China
| | - Ruyi Pan
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (Z.Y.); (R.P.); (J.Z.); (X.M.); (N.D.); (X.Y.); (J.W.); (K.L.); (Y.Q.); (Z.M.)
| | - Junjie Zhang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (Z.Y.); (R.P.); (J.Z.); (X.M.); (N.D.); (X.Y.); (J.W.); (K.L.); (Y.Q.); (Z.M.)
| | - Jianhe Sun
- Shanghai Key Laboratory of Veterinary Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China;
| | - Xiaochun Ma
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (Z.Y.); (R.P.); (J.Z.); (X.M.); (N.D.); (X.Y.); (J.W.); (K.L.); (Y.Q.); (Z.M.)
| | - Nihua Dong
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (Z.Y.); (R.P.); (J.Z.); (X.M.); (N.D.); (X.Y.); (J.W.); (K.L.); (Y.Q.); (Z.M.)
| | - Xiaohui Yao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (Z.Y.); (R.P.); (J.Z.); (X.M.); (N.D.); (X.Y.); (J.W.); (K.L.); (Y.Q.); (Z.M.)
| | - Jianchao Wei
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (Z.Y.); (R.P.); (J.Z.); (X.M.); (N.D.); (X.Y.); (J.W.); (K.L.); (Y.Q.); (Z.M.)
| | - Ke Liu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (Z.Y.); (R.P.); (J.Z.); (X.M.); (N.D.); (X.Y.); (J.W.); (K.L.); (Y.Q.); (Z.M.)
| | - Yafeng Qiu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (Z.Y.); (R.P.); (J.Z.); (X.M.); (N.D.); (X.Y.); (J.W.); (K.L.); (Y.Q.); (Z.M.)
| | - Katie Sealey
- School of Cellular and Molecular Medicine, University of Bristol, University Walk, Bristol BS8 1TD, UK;
| | | | - Michael A. Jarvis
- The Vaccine Group Ltd., Plymouth PL6 8BU, UK; (H.N.)
- School of Biomedical Sciences, University of Plymouth, Plymouth PL4 8AA, UK; (M.U.); (X.L.)
| | - Mathew Upton
- School of Biomedical Sciences, University of Plymouth, Plymouth PL4 8AA, UK; (M.U.); (X.L.)
| | - Xiangdong Li
- School of Biomedical Sciences, University of Plymouth, Plymouth PL4 8AA, UK; (M.U.); (X.L.)
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Zhiyong Ma
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (Z.Y.); (R.P.); (J.Z.); (X.M.); (N.D.); (X.Y.); (J.W.); (K.L.); (Y.Q.); (Z.M.)
| | - Juxiang Liu
- College of Veterinary Medicine, Hebei Agricultural University, Baoding 071000, China
| | - Beibei Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (Z.Y.); (R.P.); (J.Z.); (X.M.); (N.D.); (X.Y.); (J.W.); (K.L.); (Y.Q.); (Z.M.)
| |
Collapse
|
5
|
Liang S, Zhang S, Bao Y, Zhang Y, Liu X, Yao H, Liu G. Combined Immunoinformatics to Design and Evaluate a Multi-Epitope Vaccine Candidate against Streptococcus suis Infection. Vaccines (Basel) 2024; 12:137. [PMID: 38400121 PMCID: PMC10892848 DOI: 10.3390/vaccines12020137] [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: 12/18/2023] [Revised: 01/22/2024] [Accepted: 01/24/2024] [Indexed: 02/25/2024] Open
Abstract
Streptococcus suis (S. suis) is a zoonotic pathogen with multiple serotypes, and thus, multivalent vaccines generating cross-protection against S. suis infections are urgently needed to improve animal welfare and reduce antibiotic abuse. In this study, we established a systematic and comprehensive epitope prediction pipeline based on immunoinformatics. Ten candidate epitopes were ultimately selected for building the multi-epitope vaccine (MVSS) against S. suis infections. The ten epitopes of MVSS were all derived from highly conserved, immunogenic, and virulence-associated surface proteins in S. suis. In silico analyses revealed that MVSS was structurally stable and affixed with immune receptors, indicating that it would likely trigger strong immunological reactions in the host. Furthermore, mice models demonstrated that MVSS elicited high titer antibodies and diminished damages in S. suis serotype 2 and Chz infection, significantly reduced sequelae, induced cytokine transcription, and decreased organ bacterial burdens after triple vaccination. Meanwhile, anti-rMVSS serum inhibited five important S. suis serotypes in vitro, exerted beneficial protective effects against S. suis infections and significantly reduced histopathological damage in mice. Given the above, it is possible to develop MVSS as a universal subunit vaccine against multiple serotypes of S. suis infections.
Collapse
Affiliation(s)
- Song Liang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- OIE Reference Lab for Swine Streptococcosis, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Key Laboratory of Animal Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Shidan Zhang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- OIE Reference Lab for Swine Streptococcosis, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Key Laboratory of Animal Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Yinli Bao
- Engineering Research Center for the Prevention and Control of Animal Original Zoonosis, Fujian Province University, College of Life Science, Longyan University, Longyan 364012, China
| | - Yumin Zhang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- OIE Reference Lab for Swine Streptococcosis, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Key Laboratory of Animal Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Xinyi Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- OIE Reference Lab for Swine Streptococcosis, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Key Laboratory of Animal Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Huochun Yao
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- OIE Reference Lab for Swine Streptococcosis, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Key Laboratory of Animal Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Guangjin Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- OIE Reference Lab for Swine Streptococcosis, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Key Laboratory of Animal Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Sanya Institute of Nanjing Agricultural University, Nanjing Agricultural University, Sanya 572000, China
| |
Collapse
|
6
|
Bian X, Liu Q, Chen Y, Zhang W, Li M, Zhang X, Yang L, Liao Y, Kong Q. Immunogenicity and cross-protective efficacy induced by delayed attenuated Salmonella with regulated length of lipopolysaccharide in mice. Gut Microbes 2024; 16:2424983. [PMID: 39529227 PMCID: PMC11559367 DOI: 10.1080/19490976.2024.2424983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Non-typhoidal Salmonella enterica (NTS) is a major global foodborne pathogen that poses a major public health concern worldwide, and no vaccines are available for protecting against infection of multiple Salmonella serotypes, therefore, the development of Salmonella vaccines to provide broad protection is valuable. In this work, we aimed to regulate lipopolysaccharide (LPS) synthesis of live Salmonella in vivo for exposing conserved protein antigens on the outer membrane while maintaining smooth LPS patterns in vitro to keep their original ability to invade host cells for inducing cross-protection against infection of multiple Salmonella serotypes. We generated a series of mutants defective in genes to affect the length of LPS. These mutants exhibit in vivo regulated-delayed attenuation and altered length of LPS, and all these mutants were derived from SW067 (ΔpagL7 ΔpagP81::Plpp lpxE ΔlpxR9 Δfur9) containing ∆pagP81::Plpp lpxE mutation to reduce their endotoxic activity. Animal experiments demonstrated that all regulated delayed attenuated mutants exhibited reduced ability to colonize the organs of the mice, and SW114 (waaI), SW116 (waaJ), SW118 (waaL), and SW120 (wbaP) induced a significant production of IgG and IgA against OMPs isolated from S. Typhimurium, S. Enteritidis, and S. Choleraesuis. SW114 (waaI), SW116 (waaJ), and SW118 (waaL) were capable of conferring significant protection against infection of wild-type S. Enteritidis and S. Choleraesuis, with SW118 (waaL) triggering significant CD4+ T-cell responses as well as the B220low IgG+ BM cell. In conclusion, regulated delayed attenuated Salmonella vaccines with the whole core oligosaccharides of LPS showed a goo.d ability to expose conserved outer antigens and to trigger strong cross-immune responses against both homologous and heterologous Salmonella infections. These results give new insight into the development of the Salmonella vaccine against multiple serotypes of Salmonella.
Collapse
Affiliation(s)
- Xiaoping Bian
- College of Veterinary Medicine, Southwest University, Beibei, Chongqing, China
- Yibin Academy of Southwest University, Yibin, Sichuan, China
| | - Qing Liu
- College of Veterinary Medicine, Southwest University, Beibei, Chongqing, China
- Yibin Academy of Southwest University, Yibin, Sichuan, China
| | - Yaolin Chen
- College of Veterinary Medicine, Southwest University, Beibei, Chongqing, China
| | - Wenjin Zhang
- College of Veterinary Medicine, Southwest University, Beibei, Chongqing, China
| | - Mengru Li
- College of Veterinary Medicine, Southwest University, Beibei, Chongqing, China
| | - Xiaofen Zhang
- College of Veterinary Medicine, Southwest University, Beibei, Chongqing, China
| | - Liu Yang
- National Center of Technology Innovation for Pigs, Rongchang, Chongqing, China
| | - Yonghong Liao
- College of Veterinary Medicine, Southwest University, Beibei, Chongqing, China
| | - Qingke Kong
- College of Veterinary Medicine, Southwest University, Beibei, Chongqing, China
- Yibin Academy of Southwest University, Yibin, Sichuan, China
| |
Collapse
|
7
|
Zhang G, Fu Y, Li Y, Li Q, Wang S, Shi H. Oral Immunization with Attenuated Salmonella Choleraesuis Expressing the FedF Antigens Protects Mice against the Shiga-Toxin-Producing Escherichia coli Challenge. Biomolecules 2023; 13:1726. [PMID: 38136597 PMCID: PMC10741478 DOI: 10.3390/biom13121726] [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: 10/19/2023] [Revised: 11/18/2023] [Accepted: 11/22/2023] [Indexed: 12/24/2023] Open
Abstract
Edema disease (ED) is a severe and lethal infectious ailment in swine, stemming from Shiga-toxin-producing Escherichia coli (STEC). An efficient, user-friendly, and safe vaccine against ED is urgently required to improve animal welfare and decrease antibiotic consumption. Recombinant attenuated Salmonella vaccines (RASV) administered orally induce both humoral and mucosal immune responses to the immunizing antigen. Their potential for inducing protective immunity against ED is significant through the delivery of STEC antigens. rSC0016 represents an enhanced recombinant attenuated vaccine vector designed for Salmonella enterica serotype Choleraesuis. It combines sopB mutations with a regulated delay system to strike a well-balanced equilibrium between host safety and immunogenicity. We generated recombinant vaccine strains, namely rSC0016 (pS-FedF) and rSC0016 (pS-rStx2eA), and assessed their safety and immunogenicity in vivo. The findings demonstrated that the mouse models immunized with rSC0016 (pS-FedF) and rSC0016 (pS-rStx2eA) generated substantial IgG antibody responses to FedF and rStx2eA, while also provoking robust mucosal and cellular immune responses against both FedF and rStx2eA. The protective impact of rSC0016 (pS-FedF) against Shiga-toxin-producing Escherichia coli surpassed that of rSC0016 (pS-rStx2eA), with percentages of 83.3%. These findings underscore that FedF has greater suitability for vaccine delivery via recombinant attenuated Salmonella vaccines (RASVs). Overall, this study provides a promising candidate vaccine for infection with STEC.
Collapse
Affiliation(s)
- Guihua Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (G.Z.); (Y.F.); (Q.L.)
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Yang Fu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (G.Z.); (Y.F.); (Q.L.)
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Yu’an Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (G.Z.); (Y.F.); (Q.L.)
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Quan Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (G.Z.); (Y.F.); (Q.L.)
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Shifeng Wang
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611-0880, USA;
| | - Huoying Shi
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (G.Z.); (Y.F.); (Q.L.)
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University (JIRLAAPS), Yangzhou 225009, China
| |
Collapse
|
8
|
Jun T, Zhimin L, Xi D, Hua W, Huilong S, Jiaofeng P, Kang Z, Xie Q. Immunisation with the glycolytic enzyme enolase inhibits dissemination of Treponema pallidum in C57BL/6 mice. Microb Pathog 2023; 184:106374. [PMID: 37802159 DOI: 10.1016/j.micpath.2023.106374] [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: 07/17/2023] [Revised: 09/21/2023] [Accepted: 09/22/2023] [Indexed: 10/08/2023]
Abstract
Treponema pallidum (T. pallidum), an obligate extracellular bacterium, is the causative agent of sexually transmitted bacterial diseases. In this study, the glycolytic enzyme enolase (Tp Eno) of T. pallidum were injected intramuscularly into C57BL/6 mice, resulting in higher levels of specific anti-Tp Eno antibodies and Tp Eno-specific splenocyte proliferation than those in the mice immunized with recombinant protein Tp Eno. Cytokine (IL-4, IL-6, IL-10, IFN-γ, and TNF-α) analysis of splenocytes showed that the Tp Eno could slightly trigger the Th1-biased immune response. Furthermore, immunization of mice with Tp Eno elicited a significant production of IFN-γ by CD4+ T-cells in the spleen. Subsequently, mice were inoculated intradermally (between the scapulae), intraperitoneally, intrarectally and via the corpora cavernosa with 2.5 × 106 organisms per site (1 × 107 total organisms). The bacterial organ burden detected in the blood, spleen, liver, testes or brain of immunized mice suggested that Tp Eno enhances protective immunity to inhibit T. pallidum colonization in distal tissues. Therefore, Tp Eno vaccination enhances Tp Eno-specific immunogenicity and provides protection against T. pallidum dissemination.
Collapse
Affiliation(s)
- Tang Jun
- Department of Laboratory Medicine, Hunan People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410000, HuNan, China
| | - Liu Zhimin
- Clinical Laboratory, Affiliated Hengyang Hospital, Southern Medical University (Hengyang Central Hospital), 12# Yancheng Road, Hengyang, 421001, Hunan, China
| | - Deng Xi
- Clinical Laboratory, Affiliated Hengyang Hospital, Southern Medical University (Hengyang Central Hospital), 12# Yancheng Road, Hengyang, 421001, Hunan, China
| | - Wu Hua
- Clinical Laboratory, Affiliated Hengyang Hospital, Southern Medical University (Hengyang Central Hospital), 12# Yancheng Road, Hengyang, 421001, Hunan, China
| | - Shen Huilong
- Clinical Laboratory, Affiliated Hengyang Hospital, Southern Medical University (Hengyang Central Hospital), 12# Yancheng Road, Hengyang, 421001, Hunan, China
| | - Peng Jiaofeng
- Clinical Laboratory, Affiliated Hengyang Hospital, Southern Medical University (Hengyang Central Hospital), 12# Yancheng Road, Hengyang, 421001, Hunan, China
| | - Zheng Kang
- Clinical Laboratory, Affiliated Hengyang Hospital, Southern Medical University (Hengyang Central Hospital), 12# Yancheng Road, Hengyang, 421001, Hunan, China.
| | - Qinghua Xie
- The Affiliated Changsha Central Hospital, Department of Laboratory Medicine, Hengyang Medical School, University of South China, Changsha, 410004, HuNan, China.
| |
Collapse
|
9
|
Zhou G, Tian J, Tian Y, Ma Q, Li Q, Wang S, Shi H. Recombinant-attenuated Salmonella enterica serovar Choleraesuis vector expressing the PlpE protein of Pasteurella multocida protects mice from lethal challenge. BMC Vet Res 2023; 19:128. [PMID: 37598169 PMCID: PMC10439597 DOI: 10.1186/s12917-023-03679-0] [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: 08/31/2022] [Accepted: 07/27/2023] [Indexed: 08/21/2023] Open
Abstract
BACKGROUND Bacterial surface proteins play key roles in pathogenicity and often contribute to microbial adhesion and invasion. Pasteurella lipoprotein E (PlpE), a Pasteurella multocida (P. multocida) surface protein, has recently been identified as a potential vaccine candidate. Live attenuated Salmonella strains have a number of potential advantages as vaccine vectors, including immunization with live vector can mimic natural infections by organisms, lead to the induction of mucosal, humoral, and cellular immune responses. In this study, a previously constructed recombinant attenuated Salmonella Choleraesuis (S. Choleraesuis) vector rSC0016 was used to synthesize and secrete the surface protein PlpE of P. multocida to form the vaccine candidate rSC0016(pS-PlpE). Subsequently, the immunogenicity of S. Choleraesuis rSC0016(pS-PlpE) as an oral vaccine to induce protective immunity against P. multocida in mice was evaluated. RESULTS After immunization, the recombinant attenuated S. Choleraesuis vector can efficiently delivered P. multocida PlpE protein in vivo and induced a specific immune response against this heterologous antigen in mice. In addition, compared with the inactivated vaccine, empty vector (rSC0016(pYA3493)) and PBS immunized groups, the rSC0016(pS-PlpE) vaccine candidate group induced higher antigen-specific mucosal, humoral and mixed Th1/Th2 cellular immune responses. After intraperitoneal challenge, the rSC0016(pS-PlpE) immunized group had a markedly enhanced survival rate (80%), a better protection efficiency than 60% of the inactivated vaccine group, and significantly reduced tissue damage. CONCLUSIONS In conclusion, our study found that the rSC0016(pS-PlpE) vaccine candidate provided good protection against challenge with wild-type P. multocida serotype A in a mouse infection model, and may potentially be considered for use as a universal vaccine against multiple serotypes of P. multocida in livestock, including pigs.
Collapse
Affiliation(s)
- Guodong Zhou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
| | - Jiashuo Tian
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
| | - Yichen Tian
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
| | - Qifeng Ma
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
| | - Quan Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
| | - Shifeng Wang
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, 32611-0880, USA
| | - Huoying Shi
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China.
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University (JIRLAAPS), Yangzhou, China.
| |
Collapse
|
10
|
Dong N, Nichols H, Sun Q, Chen X, Zheng J, Guan Z, Zhang H, Davison A, Wezel Y, Li Z, Li B, Liu K, Shao D, Qiu Y, Sun J, Li X, Upton M, Ma Z, Jarvis MA, Wei J. Bovine Herpesvirus-4 Based Vaccine Provides Protective Immunity against Streptococcus suis Disease in a Rabbit Model. Vaccines (Basel) 2023; 11:1004. [PMID: 37243109 PMCID: PMC10222682 DOI: 10.3390/vaccines11051004] [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: 04/13/2023] [Revised: 05/17/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
Streptococcus suis (S. suis) is a bacterial pathogen of pigs that has a major animal health and economic impact on the pig industry. Bovine herpesvirus-4 (BoHV-4) is a new virus-based vaccine vector that has been used for the immunogenic delivery of antigens from a variety of pathogens. In the present study, two recombinant BoHV-4-based vectors were evaluated for their ability to induce immunity and protection against S. suis in a rabbit model. The GMD protein is a fusion protein consisting of multiple dominant B-cell epitopes ((B-cell dominant epitopes of GAPDH, MRP, and DLDH antigens) (BoHV-4/GMD)) and the second suilysin (SLY) (BoHV-4/SLY) from S. suis serotype 2 (SS2). Both GMD and SLY delivered by the BoHV-4 vectors were recognized by sera from SS2-infected rabbits. The vaccination of rabbits with the BoHV-4 vectors induced antibodies against SS2, as well as against additional S. suis serotypes, SS7 and SS9. However, sera from BoHV-4/GMD-vaccinated animals promoted a significant level of phagocytic activity by pulmonary alveolar macrophages (PAMs) against SS2, SS7, and SS9. In contrast, sera from rabbits immunized with BoHV-4/SLY induced PAM phagocytic activity against only SS2. In addition, BoHV-4 vaccines differed in the associated level of protection against lethal SS2 challenge, which ranged from high (71.4%) to low (12.5%) for BoHV-4/GMD and BoHV-4/SLY, respectively. These data suggest BoHV-4/GMD as a promising vaccine candidate against S. suis disease.
Collapse
Affiliation(s)
- Nihua Dong
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (N.D.); (Q.S.); (X.C.); (J.Z.); (Z.G.); (H.Z.); (Z.L.); (B.L.); (K.L.); (D.S.); (Y.Q.); (Z.M.)
| | - Hester Nichols
- The Vaccine Group Ltd., Derriford Research Facility, Plymouth PL6 8BX, UK; (H.N.); (Y.W.)
| | - Qing Sun
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (N.D.); (Q.S.); (X.C.); (J.Z.); (Z.G.); (H.Z.); (Z.L.); (B.L.); (K.L.); (D.S.); (Y.Q.); (Z.M.)
| | - Xiaojun Chen
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (N.D.); (Q.S.); (X.C.); (J.Z.); (Z.G.); (H.Z.); (Z.L.); (B.L.); (K.L.); (D.S.); (Y.Q.); (Z.M.)
| | - Jiayang Zheng
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (N.D.); (Q.S.); (X.C.); (J.Z.); (Z.G.); (H.Z.); (Z.L.); (B.L.); (K.L.); (D.S.); (Y.Q.); (Z.M.)
| | - Zhixin Guan
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (N.D.); (Q.S.); (X.C.); (J.Z.); (Z.G.); (H.Z.); (Z.L.); (B.L.); (K.L.); (D.S.); (Y.Q.); (Z.M.)
| | - Hailong Zhang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (N.D.); (Q.S.); (X.C.); (J.Z.); (Z.G.); (H.Z.); (Z.L.); (B.L.); (K.L.); (D.S.); (Y.Q.); (Z.M.)
| | - Andrew Davison
- MRC-University of Glasgow Centre for Virus Research, Glasgow G61 1QH, UK;
| | - Yvonne Wezel
- The Vaccine Group Ltd., Derriford Research Facility, Plymouth PL6 8BX, UK; (H.N.); (Y.W.)
| | - Zongjie Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (N.D.); (Q.S.); (X.C.); (J.Z.); (Z.G.); (H.Z.); (Z.L.); (B.L.); (K.L.); (D.S.); (Y.Q.); (Z.M.)
| | - Beibei Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (N.D.); (Q.S.); (X.C.); (J.Z.); (Z.G.); (H.Z.); (Z.L.); (B.L.); (K.L.); (D.S.); (Y.Q.); (Z.M.)
| | - Ke Liu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (N.D.); (Q.S.); (X.C.); (J.Z.); (Z.G.); (H.Z.); (Z.L.); (B.L.); (K.L.); (D.S.); (Y.Q.); (Z.M.)
| | - Donghua Shao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (N.D.); (Q.S.); (X.C.); (J.Z.); (Z.G.); (H.Z.); (Z.L.); (B.L.); (K.L.); (D.S.); (Y.Q.); (Z.M.)
| | - Yafeng Qiu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (N.D.); (Q.S.); (X.C.); (J.Z.); (Z.G.); (H.Z.); (Z.L.); (B.L.); (K.L.); (D.S.); (Y.Q.); (Z.M.)
| | - Jianhe Sun
- Shanghai Key Laboratory of Veterinary Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China;
| | - Xiangdong Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China;
| | - Mathew Upton
- School of Biomedical Sciences, University of Plymouth, Plymouth PL4 8AA, UK;
| | - Zhiyong Ma
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (N.D.); (Q.S.); (X.C.); (J.Z.); (Z.G.); (H.Z.); (Z.L.); (B.L.); (K.L.); (D.S.); (Y.Q.); (Z.M.)
| | - Michael A. Jarvis
- The Vaccine Group Ltd., Derriford Research Facility, Plymouth PL6 8BX, UK; (H.N.); (Y.W.)
- School of Biomedical Sciences, University of Plymouth, Plymouth PL4 8AA, UK;
| | - Jianchao Wei
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (N.D.); (Q.S.); (X.C.); (J.Z.); (Z.G.); (H.Z.); (Z.L.); (B.L.); (K.L.); (D.S.); (Y.Q.); (Z.M.)
| |
Collapse
|
11
|
Zhou G, Zhao Y, Ma Q, Li Q, Wang S, Shi H. Manipulation of host immune defenses by effector proteins delivered from multiple secretion systems of Salmonella and its application in vaccine research. Front Immunol 2023; 14:1152017. [PMID: 37081875 PMCID: PMC10112668 DOI: 10.3389/fimmu.2023.1152017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 03/23/2023] [Indexed: 04/07/2023] Open
Abstract
Salmonella is an important zoonotic bacterial species and hazardous for the health of human beings and livestock globally. Depending on the host, Salmonella can cause diseases ranging from gastroenteritis to life-threatening systemic infection. In this review, we discuss the effector proteins used by Salmonella to evade or manipulate four different levels of host immune defenses: commensal flora, intestinal epithelial-mucosal barrier, innate and adaptive immunity. At present, Salmonella has evolved a variety of strategies against host defense mechanisms, among which various effector proteins delivered by the secretory systems play a key role. During its passage through the digestive system, Salmonella has to face the intact intestinal epithelial barrier as well as competition with commensal flora. After invasion of host cells, Salmonella manipulates inflammatory pathways, ubiquitination and autophagy processes with the help of effector proteins. Finally, Salmonella evades the adaptive immune system by interfering the migration of dendritic cells and interacting with T and B lymphocytes. In conclusion, Salmonella can manipulate multiple aspects of host defense to promote its replication in the host.
Collapse
Affiliation(s)
- Guodong Zhou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yuying Zhao
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Qifeng Ma
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Quan Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Shifeng Wang
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - Huoying Shi
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University (JIRLAAPS), Yangzhou, China
| |
Collapse
|
12
|
Yan Z, Yao X, Pan R, Zhang J, Ma X, Dong N, Wei J, Liu K, Qiu Y, Sealey K, Nichols H, Jarvis MA, Upton M, Li X, Ma Z, Liu J, Li B. Subunit Vaccine Targeting Phosphate ABC Transporter ATP-Binding Protein, PstB, Provides Cross-Protection against Streptococcus suis Serotype 2, 7, and 9 in Mice. Vet Sci 2023; 10:vetsci10010048. [PMID: 36669049 PMCID: PMC9953333 DOI: 10.3390/vetsci10010048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/03/2023] [Accepted: 01/05/2023] [Indexed: 01/12/2023] Open
Abstract
Streptococcus suis is a significant pathogen in pigs and a newly emerging zoonotic agent in humans. The presence of multiple serotypes and strains with diversified sequence types in pig herds highlights the need for the identification of broadly cross-reactive universal vaccine antigen targets, capable of providing cross-protection against S. suis infection. Subunit vaccines based on the conserved proteins shared between different S. suis serotypes are potential candidates for such a universally protective vaccine. In the present study, phosphate ABC transporter ATP-binding protein PstB (PstB), an immunogenic protein of the S. suis bacterium, was expressed and purified, and then subjected to cross-protection evaluation in mice. The PstB protein showed nearly 100% amino acid similarity across a panel of 31 S. suis isolates representing different serotypes, which were collected from different countries. A recombinant PstB (rPstB) protein (S. suis serotype 2) was recognized by rabbit sera specific to this serotype, and induced high levels of IFN-γ and IL-4 in mice immunized with the recombinant protein. These cytokines are considered important for protection against S. suis infection. Immunization of mice with rPstB resulted in an 87.5% protection against challenge with S. suis serotype 2 and 9 strains, suggesting a high level of cross-protection for S. suis serotypes 2 and 9. A lower protection rate (62.5%) was observed in mice challenged with the S. suis serotype 7 strain. These data demonstrate that PstB is a promising target antigen for development as a component of a universal subunit vaccine against multiple S. suis serotypes.
Collapse
Affiliation(s)
- Zujie Yan
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China
- College of Veterinary Medicine, Hebei Agricultural University, Baoding 071000, China
| | - Xiaohui Yao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China
| | - Ruyi Pan
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China
| | - Junjie Zhang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China
| | - Xiaochun Ma
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China
| | - Nihua Dong
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China
| | - Jianchao Wei
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China
| | - Ke Liu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China
| | - Yafeng Qiu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China
| | - Katie Sealey
- School of Biomedical Sciences, University of Plymouth, Plymouth, Devon PL4 8AA, UK
| | - Hester Nichols
- The Vaccine Group Ltd., Plymouth, Derriford Research Facility, Devon PL6 8BX, UK
| | - Michael A. Jarvis
- School of Biomedical Sciences, University of Plymouth, Plymouth, Devon PL4 8AA, UK
- The Vaccine Group Ltd., Plymouth, Derriford Research Facility, Devon PL6 8BX, UK
| | - Mathew Upton
- School of Biomedical Sciences, University of Plymouth, Plymouth, Devon PL4 8AA, UK
| | - Xiangdong Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Zhiyong Ma
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China
| | - Juxiang Liu
- College of Veterinary Medicine, Hebei Agricultural University, Baoding 071000, China
- Correspondence: (J.L.); (B.L.)
| | - Beibei Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China
- Correspondence: (J.L.); (B.L.)
| |
Collapse
|
13
|
Zhou G, Tian Y, Tian J, Ma Q, Huang S, Li Q, Wang S, Shi H. Oral Immunization with Attenuated Salmonella Choleraesuis Expressing the P42 and P97 Antigens Protects Mice against Mycoplasma hyopneumoniae Challenge. Microbiol Spectr 2022; 10:e0236122. [PMID: 36377878 PMCID: PMC9769600 DOI: 10.1128/spectrum.02361-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 10/28/2022] [Indexed: 11/16/2022] Open
Abstract
Mycoplasma hyopneumoniae (M. hyopneumoniae, Mhp) is the etiological agent of swine enzootic pneumonia (EP), which has been associated with considerable economic losses due to reduced daily weight gain and feed efficiency. Adhesion to the cilia is important for Mhp to colonize the respiratory epithelium. Therefore, a successful vaccine must induce broad Mhp-specific immune responses at the mucosal surface. Recombinant attenuated Salmonella strains are believed to act as powerful live vaccine vectors that are able to elicit mucosal immune responses against various pathogens. To develop efficacious and inexpensive vaccines against Mhp, the immune responses and protection induced by recombinant attenuated Salmonella vaccines based on the P42 and P97 antigens of Mhp were evaluated. In general, the oral inoculation of recombinant rSC0016(pS-P42) or rSC0016(pS-P97) resulted in strong mucosal immunity, cell-mediated immunity, and humoral immunity, which was a mixed Th1/Th2-type response. In addition, the levels of specific IL-4 and IFN-γ in the immunized mice were increased, and the proliferation of lymphocytes was also enhanced, confirming the production of a good cellular immune response. Finally, both vaccine candidate strains were able to improve the weight loss of mice after a challenge and reduce clinical symptoms, lung pathological damage, and the inflammatory cell infiltration. These results suggest that the delivery of protective antigens with recombinant attenuated Salmonella vectors may be an effective means by which to combat Mhp infection. IMPORTANCE Mhp is the main pathogen of porcine enzootic pneumonia, a highly infectious and economically significant respiratory disease that affects pigs of all ages. As the target tissue of Mhp infections are the mucosal sites of the respiratory tract, the induction of protective immunity at the mucosal tissues is the most efficient strategy by which to block disease transmission. Because the stimulation of mucosal immune responses is efficient, Salmonella-vector oral vaccines are expected to be especially useful against mucosal-invading pathogens. In this study, we expressed the immunogenic proteins of P42 and P97 with the attenuated Salmonella Choleraesuis vector rSC0016, thereby generating a low-cost and more effective vaccine candidate against Mhp by inducing significant mucosal, humoral and cellular immunity. Furthermore, rSC0016(pS-P42) effectively prevents Mhp-induced weight loss and the pulmonary inflammation of mice. Because of the effectiveness of rSC0016(pS-P42) against Mhp infection in mice, this novel vaccine candidate strain shows great potential for its use in the pig breeding industry.
Collapse
Affiliation(s)
- Guodong Zhou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yichen Tian
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Jiashuo Tian
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Qifeng Ma
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Shan Huang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Quan Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Shifeng Wang
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, USA
| | - Huoying Shi
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University (JIRLAAPS), Yangzhou, China
| |
Collapse
|
14
|
Li YA, Sun Y, Zhang Y, Wang S, Shi H. Live attenuated Salmonella enterica serovar Choleraesuis vector delivering a virus-like particles induces a protective immune response against porcine circovirus type 2 in mice. Vaccine 2022; 40:4732-4741. [PMID: 35773121 DOI: 10.1016/j.vaccine.2022.06.046] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/30/2022] [Accepted: 06/16/2022] [Indexed: 11/25/2022]
Abstract
The virus-like particles (VLPs) of porcine circovirus type 2 (PCV2) is an attractive vaccine candidate that retains the natural conformation of the virion but lacks the viral genome to replicate, thus balancing safety and immunogenicity. However, the assembly of VLPs requires cumbersome subsequent processes, hindering the development of related vaccines. In addition, as a subunit antigen, VLPs are defective in inducing cellular and mucosal immune responses. In this study, the capsid (Cap) protein of PCV2 was synthesized and self-assembled into VLPs in the recombinant attenuated S. Choleraesuis vector, rSC0016(pS-Cap). Furthermore, rSC0016(pS-Cap) induced a Cap-specific Th1-dominant immune response, mucosal immune responses, and neutralizing antibodies against PCV2. Finally, the virus genome copies in mice immunized with the rSC0016(pS-Cap) were significantly lower than those of the empty vector control group after challenge with PCV2. In conclusion, our study demonstrates the potential of using S. Choleraesuis vectors to delivery VLPs, providing new ideas for the development of PCV2 vaccines.
Collapse
Affiliation(s)
- Yu-An Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yanni Sun
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yuqin Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Shifeng Wang
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611-0880, USA
| | - Huoying Shi
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University (JIRLAAPS), Yangzhou, China.
| |
Collapse
|
15
|
Li YA, Sun Y, Fu Y, Zhang Y, Li Q, Wang S, Shi H. Salmonella enterica serovar Choleraesuis vector delivering a dual-antigen expression cassette provides mouse cross-protection against Streptococcus suis serotypes 2, 7, 9, and 1/2. Vet Res 2022; 53:46. [PMID: 35733156 PMCID: PMC9215036 DOI: 10.1186/s13567-022-01062-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 05/10/2022] [Indexed: 12/01/2022] Open
Abstract
A universal vaccine protecting against multiple serotypes of Streptococcus suis is urgently needed to improve animal welfare and reduce the consumption of antibiotics. In this study, a dual antigen expression cassette consisting of SS2-SaoA and SS9-Eno was delivered by a recombinant Salmonella Choleraesuis vector to form the vaccine candidate rSC0016(pS-SE). SaoA and Eno were simultaneously synthesized in rSC0016(pS-SE) without affecting the colonization of the recombinant vector in the lymphatic system. In addition, the antiserum of mice immunized with rSC0016(pS-SE) produced a broader and potent opsonophagocytic response against multiple serotypes of S. suis. Finally, rSC0016(pS-SE) provided mice with a 100% protection against a lethal dose of parent S. suis serotype 2 and serotype 9, and provided 90% and 80% protection against heterologous S. suis serotype 7 or 1/2. These values were significantly higher than those obtained with rSC0016(pS-SaoA) or rSC0016(pS-Eno). Together, this study serves as a foundation for developing a universal vaccine against multiple serotypes of S. suis.
Collapse
Affiliation(s)
- Yu-An Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China.,Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yanni Sun
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China.,Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yang Fu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China.,Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yuqin Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China.,Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Quan Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China.,Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Shifeng Wang
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, 32611-0880, USA
| | - Huoying Shi
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China. .,Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China. .,Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University (JIRLAAPS), Yangzhou, China.
| |
Collapse
|
16
|
Hussain M, Kohler C, Becker K. Enolase of Staphylococcus lugdunensis Is a Surface-Exposed Moonlighting Protein That Binds to Extracellular Matrix and the Plasminogen/Plasmin System. Front Microbiol 2022; 13:837297. [PMID: 35308335 PMCID: PMC8928124 DOI: 10.3389/fmicb.2022.837297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 01/20/2022] [Indexed: 11/27/2022] Open
Abstract
The coagulase-negative staphylococcal (CoNS) species Staphylococcus lugdunensis is unique in causing serious infections in humans that resemble those of Staphylococcus aureus rather than those of other CoNS species. The colonization and invasion of host tissue presupposes the presence of adherence factors, but only a few proteins mediating adhesion of S. lugdunensis to biotic surfaces are known yet. Here, we report on the functionality of the S. lugdunensis enolase (SlEno), which performs two distinct roles, first, as the metabolic enzyme of the glycolysis, and second, as an adherence factor to the extracellular matrix (ECM) of cells. Phylogenetic analyses of the SlEno confirmed their high conservation to enolases of other species and revealed a closer relationship to Staphylococcus epidermidis than to S. aureus. Using matrix-assisted laser desorption/ionization time of flight mass spectrometry and Western blot experiments, we identified SlEno to be located in the cytoplasm as well as on the cell surface of S. lugdunensis. Recombinantly generated and surface-associated SlEno showed the usual enolase activity by catalyzing the conversion of 2-phosphoglycerate to phosphoenolpyruvate but, in addition, also displayed strong binding to immobilized laminin, fibronectin, fibrinogen, and collagen type IV in a dose-dependent manner. We also showed a strong binding of SlEno to plasminogen (Plg) and observed a tissue plasminogen activator (tPA)-dependent conversion of Plg to plasmin (Pln) whereby the Plg activation significantly increased in the presence of SlEno. This interaction might be dependent on lysines of the SlEno protein as binding to Plg was inhibited by ε-aminocaproic acid. Furthermore, the enhanced activation of the Plg/Pln system by SlEno enabled S. lugdunensis to migrate through a fibrin matrix. This migration was about 10-fold higher than without exogenously added SlEno. Finally, we observed a significantly higher clearance of S. lugdunensis by freshly prepared granulocytes and in the presence of anti-SlEno antibodies. In conclusion, these data demonstrate for the first time a moonlighting function of the S. lugdunensis enolase, which is an underrated virulence factor for colonization and invasion of tissues. Hence, SlEno might be a potential vaccine candidate to prevent severe infections caused by this pathogen.
Collapse
Affiliation(s)
- Muzaffar Hussain
- Institute of Medical Microbiology, University Hospital Münster, Münster, Germany
| | - Christian Kohler
- Friedrich Loeffler Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Karsten Becker
- Institute of Medical Microbiology, University Hospital Münster, Münster, Germany
- Friedrich Loeffler Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
- Interdisciplinary Centre for Clinical Research (IZKF), University Hospital Münster, Münster, Germany
- *Correspondence: Karsten Becker,
| |
Collapse
|
17
|
Neutrophils in Streptococcus suis Infection: From Host Defense to Pathology. Microorganisms 2021; 9:microorganisms9112392. [PMID: 34835517 PMCID: PMC8624082 DOI: 10.3390/microorganisms9112392] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/11/2021] [Accepted: 11/17/2021] [Indexed: 01/02/2023] Open
Abstract
Streptococcus suis is a swine pathogen and zoonotic agent responsible for economic losses to the porcine industry. Infected animals may develop meningitis, arthritis, endocarditis, sepsis and/or sudden death. The pathogenesis of the infection implies that bacteria breach mucosal host barriers and reach the bloodstream, where they escape immune-surveillance mechanisms and spread throughout the organism. The clinical manifestations are mainly the consequence of an exacerbated inflammation, defined by an exaggerated production of cytokines and recruitment of immune cells. Among them, neutrophils arrive first in contact with the pathogens to combat the infection. Neutrophils initiate and maintain inflammation, by producing cytokines and deploying their arsenal of antimicrobial mechanisms. Furthermore, neutrophilic leukocytosis characterizes S. suis infection, and lesions of infected subjects contain a large number of neutrophils. Therefore, this cell type may play a role in host defense and/or in the exacerbated inflammation. Nevertheless, a limited number of studies addressed the role or functions of neutrophils in the context of S. suis infection. In this review, we will explore the literature about S. suis and neutrophils, from their interaction at a cellular level, to the roles and behaviors of neutrophils in the infected host in vivo.
Collapse
|
18
|
Weiße C, Dittmar D, Jakóbczak B, Florian V, Schütze N, Alber G, Klose K, Michalik S, Valentin-Weigand P, Völker U, Baums CG. Immunogenicity and protective efficacy of a Streptococcus suis vaccine composed of six conserved immunogens. Vet Res 2021; 52:112. [PMID: 34433500 PMCID: PMC8390293 DOI: 10.1186/s13567-021-00981-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 05/20/2021] [Indexed: 01/12/2023] Open
Abstract
A vaccine protecting against different Streptococcus suis serotypes is highly needed in porcine practice to improve animal welfare and reduce the use of antibiotics. We hypothesized that immunogens prominently recognized by convalescence sera but significantly less so by sera of susceptible piglets are putative protective antigens. Accordingly, we investigated immunogenicity and protective efficacy of a multicomponent vaccine including six main conserved immunogens, namely SSU0934, SSU1869, SSU0757, SSU1950, SSU1664 and SSU0187. Flow cytometry confirmed surface expression of all six immunogens in S. suis serotypes 2, 9 and 14. Although prime-booster vaccination after weaning resulted in significantly higher specific IgG levels against all six immunogens compared to the placebo-treated group, no significant differences between bacterial survival in blood from either vaccinated or control animals were recorded for serotype 2, 9 and 14 strains. Furthermore, vaccinated piglets were not protected against morbidity elicited through intranasal challenge with S. suis serotype 14. As ~50% of animals in both groups did not develop disease, we investigated putative other correlates of protection. Induction of reactive oxygen species (ROS) in blood granulocytes was not associated with vaccination but correlated with protection as all piglets with >5% ROS survived the challenge. Based on these findings we discuss that the main immunogens of S. suis might actually not be a priori good candidates for protective antigens. On the contrary, expression of immunogens that evoke antibodies that do not mediate killing of this pathogen might constitute an evolutionary advantage conserved in many different S. suis strains.
Collapse
Affiliation(s)
- Christine Weiße
- Institute of Bacteriology and Mycology, Centre for Infectious Diseases, Faculty of Veterinary Medicine, Leipzig University, Leipzig, Germany
| | - Denise Dittmar
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | | | | | - Nicole Schütze
- Institute of Immunology, Centre for Infectious Diseases, Faculty of Veterinary Medicine, Leipzig University, Leipzig, Germany
| | - Gottfried Alber
- Institute of Immunology, Centre for Infectious Diseases, Faculty of Veterinary Medicine, Leipzig University, Leipzig, Germany
| | - Kristin Klose
- Institute of Pathology, Faculty of Veterinary Medicine, Leipzig University, Leipzig, Germany
| | - Stephan Michalik
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Peter Valentin-Weigand
- Department of Infectious Diseases, Institute for Microbiology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Uwe Völker
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Christoph Georg Baums
- Institute of Bacteriology and Mycology, Centre for Infectious Diseases, Faculty of Veterinary Medicine, Leipzig University, Leipzig, Germany.
| |
Collapse
|
19
|
Li Q, Hu Y, Fei X, Du Y, Guo W, Chu D, Wang X, Wang S, Shi H. OmpC, a novel factor H-binding surface protein, is dispensable for the adherence and virulence of Salmonella enterica serovar Typhimurium. Vet Microbiol 2021; 259:109157. [PMID: 34197978 DOI: 10.1016/j.vetmic.2021.109157] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 06/15/2021] [Indexed: 11/24/2022]
Abstract
Salmonella enterica serovar Typhimurium utilizes a series of strategies to evade host innate immune defenses, including the serum complement system. Many microbial pathogens have evolved the ability to bind the complement regulatory protein factor H (FH) through their surface factor H-binding proteins (FHBPs) to circumvent the complement-mediated bactericidal effect. However, the roles of FHBPs in Salmonella pathogenesis are not well understood. In this study, we demonstrated that the survival of S. Typhimurium in human serum was decreased in a time and concentration dependent manner. Pre-incubation with FH attenuated the sensitivity of S. Typhimurium strain χ3761 to complement-mediated serum killing, suggesting FH binding enhance survival in serum. We aimed to identify novel S. Typhimurium FHBPs and characterize their biological functions. Here, six potential FHBPs were identified by two-dimensional (2D)-Far-western blot, and three of them were further confirmed to bind FH by Far-western blot and dot blot. We found that deletion of ompC (ΔompC) significantly inhibited the survival of S. Typhimurium strain χ3761 in human serum. Our results indicated that the ompC mutation does not affect χ3761 adhesion to HeLa cells. Furthermore, a mice infection model showed that deletion of ompC had no significant effect on the histopathological lesions or viability compared with the wild-type strain χ3761. In summary, these results suggested that OmpC is an important FHBP, but not a critical virulence factor of S. Typhimurium.
Collapse
Affiliation(s)
- Quan Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China; Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
| | - Yuhan Hu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China; Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
| | - Xia Fei
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China; Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
| | - Yuanzhao Du
- Yebio Bioengineering Co., Ltd of Qingdao, Qingdao, 266114, China.
| | - Weiwei Guo
- Yebio Bioengineering Co., Ltd of Qingdao, Qingdao, 266114, China.
| | - Dianfeng Chu
- Yebio Bioengineering Co., Ltd of Qingdao, Qingdao, 266114, China.
| | - Xiaobo Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China; Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
| | - Shifeng Wang
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, 32611-0880, USA.
| | - Huoying Shi
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China; Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China; Key Laboratory of Animal Infectious Diseases, Ministry of Agriculture, Yangzhou University, China; Jiangsu Key Laboratory of Preventive Veterinary Medicine, Yangzhou University, China.
| |
Collapse
|
20
|
Guo G, Kong X, Wang Z, Li M, Tan Z, Zhang W. Evaluation of the immunogenicity and protective ability of a pili subunit, SBP2', of Streptococcus suis serotype 2. Res Vet Sci 2021; 137:201-207. [PMID: 34020335 DOI: 10.1016/j.rvsc.2021.05.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/30/2021] [Accepted: 05/12/2021] [Indexed: 01/08/2023]
Abstract
Streptococcus suis is an important zoonotic pathogen that leads to huge economic losses in the swine industry. Because of the enormous genetic and phenotypic diversity within S. suis, it is necessary to develop effective vaccines to control this zoonotic pathogen. SBP2' is a major pili subunit in S. suis that belongs to an srtBCD pili cluster and has already been reported to be associated with the pathogenesis of this bacterium. In this study, we aimed to evaluate the immunogenicity and protective ability of SBP2'. The rSBP2' protein was expressed by an Escherichia coli expression system and emulsified with Montanide ISA 201 adjuvant to prepare the subunit vaccine. Through active immune assays, the results showed that rSBP2' exhibited good immunogenicity and could protect mice from a lethal dose challenge. Additionally, the qRT-PCR data showed that the transcription levels of cytokines associated with systemic symptoms caused by S. suis were decreased, indicating that immunization with rSBP2' could protect the host from cytokine storms caused by S. suis. Furthermore, the passive immune assay showed that the humoral immunity induced by rSBP2' played an important role against S. suis infection. Taken together, SBP2' could provide proper immune protection against S. suis challenge and could be a candidate for S. suis subunit vaccine. The results of this study could provide new ideas for the development of effective vaccines against S. suis.
Collapse
Affiliation(s)
- Genglin Guo
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China; OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing, China.
| | - Xuewei Kong
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China; OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing, China
| | - Zhuohao Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China; OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing, China
| | - Min Li
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China; OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing, China
| | - Zhongming Tan
- NHC Key laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210009, China.
| | - Wei Zhang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China; OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing, China.
| |
Collapse
|
21
|
Han Y, Luo P, Chen Y, Xu J, Sun J, Guan C, Wang P, Chen M, Zhang X, Zhu Y, Zhu T, Zhai R, Cheng C, Song H. Regulated delayed attenuation improves vaccine efficacy in preventing infection from avian pathogenic Escherichia coli O 78 and Salmonella typhimurium. Vet Microbiol 2021; 254:109012. [PMID: 33611126 DOI: 10.1016/j.vetmic.2021.109012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 02/03/2021] [Indexed: 10/22/2022]
Abstract
Avian pathogenic Escherichia coli (APEC) O78 and Salmonella typhimurium (S. Typhimurium) are two leading bacterial pathogens that cause significant economic loss in the poultry industry. O-antigen is an important immunogen of these two bacteria to induce host protective immune responses during infection. To develop a bivalent vaccine against APEC O78 and S. Typhimurium, the attenuated Salmonella ST01 (Δasd ΔrfbP Δcrp) was genetically constructed to deliver APEC O78 O-antigen polysaccharide (OPS), which stably expresses OPS with asd+ balanced-lethal system in vitro and in vivo. After oral immunization, the recombinant attenuated Salmonella vaccine (RASV) strain ST01 (pSS26-O78) provided insufficient protection against the APEC O78 challenge. Therefore, the regulated delayed attenuation strain ST02 (Δasd ΔrfbP ΔPcrp::TTaraC PBADcrp) was further constructed by regulating cyclic AMP receptor protein (crp) with araC PBAD cassette to better present the heterologous O-antigen to the host immune system. The innovative recombinant strain ST02 (pSS26-O78) stimulated robust antibody responses against APEC O78 and S. Typhimurium OPS, with serum titers over 1:800 for both IgG and IgA, thereby providing the complement-mediated bactericidal activity and stronger protection against APEC O78 and S. Typhimurium infection. Collectively, this study demonstrates a biologically-conjugated polysaccharide vaccine candidate that can enhance homologous protection against APEC O78 and S. Typhimurium.
Collapse
Affiliation(s)
- Yue Han
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology, College of Veterinary Medicine of Zhejiang A&F University, Lin'an, China
| | - Ping Luo
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology, College of Veterinary Medicine of Zhejiang A&F University, Lin'an, China
| | - Yuji Chen
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology, College of Veterinary Medicine of Zhejiang A&F University, Lin'an, China
| | - Jiali Xu
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology, College of Veterinary Medicine of Zhejiang A&F University, Lin'an, China
| | - Jing Sun
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology, College of Veterinary Medicine of Zhejiang A&F University, Lin'an, China
| | - Chiyu Guan
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology, College of Veterinary Medicine of Zhejiang A&F University, Lin'an, China
| | - Pu Wang
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology, College of Veterinary Medicine of Zhejiang A&F University, Lin'an, China
| | - Mianmian Chen
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology, College of Veterinary Medicine of Zhejiang A&F University, Lin'an, China
| | - Xian Zhang
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology, College of Veterinary Medicine of Zhejiang A&F University, Lin'an, China
| | - Yueyue Zhu
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology, College of Veterinary Medicine of Zhejiang A&F University, Lin'an, China
| | - Tingting Zhu
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology, College of Veterinary Medicine of Zhejiang A&F University, Lin'an, China
| | - Ruidong Zhai
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology, College of Veterinary Medicine of Zhejiang A&F University, Lin'an, China
| | - Changyong Cheng
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology, College of Veterinary Medicine of Zhejiang A&F University, Lin'an, China.
| | - Houhui Song
- China-Australian Joint Laboratory for Animal Health Big Data Analytics, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology, College of Veterinary Medicine of Zhejiang A&F University, Lin'an, China.
| |
Collapse
|
22
|
Loh FK, Nathan S, Chow SC, Fang CM. Immunogenicity and protection efficacy of enhanced fitness recombinant Salmonella Typhi monovalent and bivalent vaccine strains against acute toxoplasmosis. Pathog Glob Health 2021; 115:392-404. [PMID: 33525974 DOI: 10.1080/20477724.2021.1881369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
The proficiency of Salmonella Typhi to induce cell-mediated immunity has allowed its exploitation as a live vector against the obligate intracellular protozoan Toxoplasma gondii. T. gondii vaccine research is of great medical value due to the lack of a suitable toxoplasmosis vaccine. In the present work, we integrated T. gondii antigen into a growth-dependent chromosome locus guaBA of S. Typhi CVD910 strain to form recombinant S. Typhi monovalent CVD910-SAG1 expressed T. gondii SAG1 antigen and monovalent CVD910-GRA2 expressed T. gondii GRA2 antigen. Furthermore, a low-copy stabilized recombinant plasmid encoding SAG1 antigen was transformed into CVD910-GRA2 to form bivalent CVD910-GS strain. An osmolarity-regulated promoter was also incorporated to control the gene transcription, whereas clyA export protein was included to translocate the antigen out of the cytoplasm. Both CVD910-GRA2 and CVD910-GS displayed healthy growth fitness and readily expressed the encoded T. gondii antigens. When administered in vivo, CVD910-GS successfully induced both humoral and cellular immunity in the immunized BALB/c mice, and extended mice survival against virulent T. gondii. In particular, the mice immunized with bivalent CVD910-GS presented the highest titers of IgG, percentages of CD4+ T, CD8+ T, B cells and memory T cells, and total IgG+ memory B cells as compared to the CVD910-GRA2 and control strains. The CVD910-GS group also generated mixed Th1/Th2 cytokine profile with secretions of IFN-ɣ, IL-2 and IL-10. This study demonstrated the importance of enhancing live vector fitness to sustain heterologous antigen expression for eliciting robust immune responses and providing effective protection against pathogen.
Collapse
Affiliation(s)
- Fei-Kean Loh
- Division of Biomedical Sciences, School of Pharmacy, the University of Nottingham Malaysia Campus, Semenyih, Selangor, Malaysia
| | - Sheila Nathan
- Faculty of Science & Technology, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | - Sek-Chuen Chow
- School of Science, Monash University Malaysia, Selangor, Malaysia
| | - Chee-Mun Fang
- Division of Biomedical Sciences, School of Pharmacy, the University of Nottingham Malaysia Campus, Semenyih, Selangor, Malaysia
| |
Collapse
|
23
|
Salmonella Vaccine Vector System for Foot-and-Mouth Disease Virus and Evaluation of Its Efficacy with Virus-Like Particles. Vaccines (Basel) 2021; 9:vaccines9010022. [PMID: 33466461 PMCID: PMC7824887 DOI: 10.3390/vaccines9010022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 12/29/2020] [Accepted: 12/31/2020] [Indexed: 01/11/2023] Open
Abstract
Foot-and-mouth disease virus (FMDV) causes a highly contagious and devastating disease in livestock animals and has a great potential to cause severe economic loss worldwide. The major antigen of FMDV capsid protein, VP1, contains the major B-cell epitope responsible for effectively eliciting protective humoral immunity. In this study, irradiated Salmonella Typhimurium (KST0666) were used as transgenic vectors containing stress-inducible plasmid pRECN-VP1 to deliver the VP1 protein from FMDV-type A/WH/CHA/09. Mice were orally inoculated with ATOMASal-L3 harboring pRECN-VP1, and FMDV virus-like particles, where (VLPFMDV)-specific humoral, mucosal, and cellular immune responses were evaluated. Mice vaccinated with attenuated Salmonella (KST0666) expressing VP1 (named KST0669) showed high levels of VLP-specific IgA in feces and IgG in serum, with high FMDV neutralization titer. Moreover, KST0669-vaccinated mice showed increased population of IFN-γ (type 1 T helper cells; Th1 cells)-, IL-5 (Th2 cells)-, and IL-17A (Th17 cells)-expressing CD4+ as well as activated CD8+ T cells (IFN-γ+CD8+ cells), detected by stimulating VLPFMDV. All data indicate that our Salmonella vector system successfully delivered FMDV VP1 to immune cells and that the humoral and cellular efficacy of the vaccine can be easily evaluated using VLPFMDV in a Biosafety Level I (BSL1) laboratory.
Collapse
|