1
|
Zhai K, Zhang Z, Liu X, Lv J, Zhang L, Li J, Ma Z, Wang Y, Guo H, Zhang Y, Pan L. Mucosal immune responses induced by oral administration of recombinant Lactococcus lactis expressing the S1 protein of PDCoV. Virology 2023; 578:180-189. [PMID: 36586181 DOI: 10.1016/j.virol.2022.12.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/20/2022] [Accepted: 12/20/2022] [Indexed: 12/27/2022]
Abstract
Porcine deltacoronavirus is an evolving coronavirus that primarily infects the intestine and may lead to intestinal disease in piglets. Up to now, no commercial vaccination is readily accessible to protect against the spread of PDCoV. Lactococcus lactis has been shown to have good immune efficacy and safety and can be used as a genetically engineered vaccine to deliver antigens. In this research, we utilized L. lactis NZ9000 to provide the S1 protein orally and improved the delivery efficiency by connecting the M cell targeting ligand Co1 with the S1 protein of PDCoV in tandem to obtain the recombinant protein S1-Co1. We successfully constructed two recombinant strains capable of expressing PDCoV-S1 and PDCoV-S1-Co1 proteins (i.e., L. lactis NZ9000-S1 and L. lactis NZ9000-S1-Co1), and their immunogenic capacity was evaluated in mice. Our study shows that Lactococcus is an advantageous bacterial live vector vaccine and is anticipated as a potential PDCoV vaccination option.
Collapse
Affiliation(s)
- Kaige Zhai
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China.
| | - Zhongwang Zhang
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China.
| | - Xinsheng Liu
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China.
| | - Jianliang Lv
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China.
| | - Liping Zhang
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China.
| | - Jiahao Li
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China.
| | - Zhongyuan Ma
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China.
| | - Yonglu Wang
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China.
| | - Huichen Guo
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China.
| | - Yongguang Zhang
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China.
| | - Li Pan
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China.
| |
Collapse
|
2
|
Li F, Mei Z, Ju N, Sui L, Fan X, Wang Z, Li J, Jiang Y, Cui W, Shan Z, Zhou H, Wang L, Qiao X, Tang L, Wang X, Li Y. Evaluation of the immunogenicity of auxotrophic Lactobacillus with CRISPR-Cas9D10A system-mediated chromosomal editing to express porcine rotavirus capsid protein VP4. Virulence 2022; 13:1315-1330. [PMID: 35920261 PMCID: PMC9351582 DOI: 10.1080/21505594.2022.2107646] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 07/13/2022] [Accepted: 07/26/2022] [Indexed: 11/06/2022] Open
Abstract
Porcine rotavirus (PoRV) is an important pathogen, leading to the occurrence of viral diarrhoea . As the infection displays obvious enterotropism, intestinal mucosal immunity is the significant line of defence against pathogen invasion. Moreover, as lactic acid bacteria (LAB) show acid resistance, bile salt resistance and immune regulation, it is of great significance to develop an oral vaccine. Most traditional plasmid delivery vectors use antibiotic genes as selective markers, easily leading to antibiotic accumulation. Therefore, to select a food-grade marker in genetically engineering food-grade microorganisms is vital. Based on the CRISPR-Cas9D10A system, we constructed a stable auxotrophic Lactobacillus paracasei HLJ-27 (Lactobacillus △Alr HLJ-27) strain. In addition, as many plasmids replicate in the host bacteria, resulting in internal gene deletions. In this study,we used a temperature-sensitive gene editing plasmidto insert the VP4 gene into the genome, yielding the insertion mutant strains VP4/△Alr HLJ-27, VP4/△Alr W56, and VP4/W56. This recombinant bacterium efficiently induced secretory immunoglobulin A (SIgA)-based mucosal and immunoglobulin G (IgG)-based humoral immune responses. These oral mucosal vaccines have the potential to act as an alternative to the application of antibiotics in the future and induce efficient immune responses against PEDV infection.
Collapse
Affiliation(s)
- Fengsai Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Zhuyuan Mei
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Ning Ju
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Ling Sui
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Xiaolong Fan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Zi Wang
- Tongliao Institute of agriculture and animal husbandry, Tongliao City, China
| | - Jiaxuan Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Yanping Jiang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Wen Cui
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Zhifu Shan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, Heilongjiang, China
| | - Han Zhou
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Li Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, Heilongjiang, China
| | - Xinyuan Qiao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, Heilongjiang, China
| | - Lijie Tang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, Heilongjiang, China
| | - Xiaona Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, Heilongjiang, China
| | - Yijing Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, Heilongjiang, China
| |
Collapse
|
3
|
Levit R, Cortes-Perez NG, de Moreno de Leblanc A, Loiseau J, Aucouturier A, Langella P, LeBlanc JG, Bermúdez-Humarán LG. Use of genetically modified lactic acid bacteria and bifidobacteria as live delivery vectors for human and animal health. Gut Microbes 2022; 14:2110821. [PMID: 35960855 PMCID: PMC9377234 DOI: 10.1080/19490976.2022.2110821] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
There is now strong evidence to support the interest in using lactic acid bacteria (LAB)in particular, strains of lactococci and lactobacilli, as well as bifidobacteria, for the development of new live vectors for human and animal health purposes. LAB are Gram-positive bacteria that have been used for millennia in the production of fermented foods. In addition, numerous studies have shown that genetically modified LAB and bifodobacteria can induce a systemic and mucosal immune response against certain antigens when administered mucosally. They are therefore good candidates for the development of new mucosal delivery strategies and are attractive alternatives to vaccines based on attenuated pathogenic bacteria whose use presents health risks. This article reviews the most recent research and advances in the use of LAB and bifidobacteria as live delivery vectors for human and animal health.
Collapse
Affiliation(s)
- Romina Levit
- Centro de Referencia para Lactobacilos (CERELA-CONICET), Chacabuco 145, (T4000ILC) San Miguel de Tucumán, Tucumán, Argentina
| | - Naima G. Cortes-Perez
- Université Paris-Saclay, INRAE, AgroParisTech, UMR 0496, 78350 Jouy-en-Josas, France
| | - Alejandra de Moreno de Leblanc
- Centro de Referencia para Lactobacilos (CERELA-CONICET), Chacabuco 145, (T4000ILC) San Miguel de Tucumán, Tucumán, Argentina
| | - Jade Loiseau
- Micalis Institute, Université Paris-Saclay, INRAE, AgroParisTech, 78350 Jouy-en-Josas, France
| | - Anne Aucouturier
- Micalis Institute, Université Paris-Saclay, INRAE, AgroParisTech, 78350 Jouy-en-Josas, France
| | - Philippe Langella
- Micalis Institute, Université Paris-Saclay, INRAE, AgroParisTech, 78350 Jouy-en-Josas, France
| | - Jean Guy LeBlanc
- Centro de Referencia para Lactobacilos (CERELA-CONICET), Chacabuco 145, (T4000ILC) San Miguel de Tucumán, Tucumán, Argentina
| | - Luis G. Bermúdez-Humarán
- Micalis Institute, Université Paris-Saclay, INRAE, AgroParisTech, 78350 Jouy-en-Josas, France,CONTACT Luis G. Bermúdez-Humarán Micalis Institute, Université Paris-Saclay, INRAE, AgroParisTech, 78350 Jouy-en-Josas, France
| |
Collapse
|
4
|
Qin D, Bai Y, Li Y, Huang Y, Li L, Wang G, Qu Y, Wang J, Yu LY, Hou X. Changes in Gut Microbiota by the Lactobacillus casei Anchoring the K88 Fimbrial Protein Prevented Newborn Piglets From Clinical Diarrhea. Front Cell Infect Microbiol 2022; 12:842007. [PMID: 35372106 PMCID: PMC8972131 DOI: 10.3389/fcimb.2022.842007] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/21/2022] [Indexed: 12/30/2022] Open
Abstract
In the last 20 years, accumulating evidence indicates that the gut microbiota contribute to the development, maturation, and regulation of the host immune system and mediate host anti-pathogen defenses. Lactobacillus casei (L.casei) is a normal flora of the gastrointestinal tract in mammals and, as a great mucosal delivery vehicle, has wide use in bioengineering. However, the diarrhea prevention role of commensal intestinal microbiota interfered by the recombinant L.casei (rL.casei) in newborn piglets is not well understood. In our study, newborn piglets orally fed with the rL.casei surface displayed the fimbrial protein K88 of enterotoxigenic Escherichia coli (ETEC) and their feces were collected for a period of time after feeding. The next-generation sequencing of these fecal samples showed that the relative abundance of L.casei was significantly increased. The oral administration of rL.casei altered the intestinal microbial community as evidenced by altered microbial diversity and microbial taxonomic composition. Remarkably, the functional enhancing of the intestinal bacterial community by rL.casei was positively correlated with membrane transport, replication, and repair (p < 0.05). The specific antibody detection indicates that high levels of anti-K88 secretory immunoglobulin A (sIgA) were induced in fecal samples and systemic immunoglobulin G was produced in serum. The diarrhea rate in piglets caused by ETEC K88 was decreased by about 24%. Thus, the oral administration of rL.casei not only activated the mucosal and humoral immune responses in vivo but also contributed to shape the intestinal probiotics in newborn piglets and to significantly reduce the diarrhea rates of newborn piglets.
Collapse
Affiliation(s)
- Da Qin
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Yongfei Bai
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Yan Li
- Colleges of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Yanmei Huang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Liyang Li
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Guihua Wang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Yi Qu
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Jiabin Wang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Li-Yun Yu
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
- Colleges of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
- *Correspondence: Li-Yun Yu, ; Xilin Hou,
| | - Xilin Hou
- Colleges of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
- *Correspondence: Li-Yun Yu, ; Xilin Hou,
| |
Collapse
|
5
|
Immunogenicity of Recombinant-Deficient Lactobacillus casei with Complementary Plasmid Expressing Alanine Racemase Gene and Core Neutralizing Epitope Antigen against Porcine Epidemic Diarrhea Virus. Vaccines (Basel) 2021; 9:vaccines9101084. [PMID: 34696192 PMCID: PMC8537014 DOI: 10.3390/vaccines9101084] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/22/2021] [Accepted: 09/24/2021] [Indexed: 01/16/2023] Open
Abstract
Porcine epidemic diarrhea (PED), which is caused by the porcine epidemic diarrhea virus (PEDV), has occurred worldwide and poses a serious threat to the pig industry. Intestine is the main function site of PEDV; therefore, it is important to develop an oral mucosal immunity vaccine against this virus infection. Most traditional plasmid delivery vectors use antibiotic genes as a selective marker, easily leading to antibiotic accumulation and gene contamination. In this study, to explore whether the alanine racemase gene (Alr) could be used as a screening marker and develop an efficient oral vaccine against PEDV infection, a recombinant strain was constructed using Lactobacillus casei with Alr deletion (L. casei ΔAlr W56) to deliver the Alr gene and a core-neutralizing epitope (COE) antigen. This recombinant bacterium efficiently induced secretory immunoglobulin A (SIgA)-based mucosal and immunoglobulin G (IgG)-based humoral immune responses via oral vaccination in mice. Compared to the other strains, the recombinant bacteria were able to grow without the addition of D-alanine, revealing that Alr in the plasmid could function normally in defective bacteria. This oral mucosal vaccine would provide a useful strategy to substitute the application of antibiotics in the future and induce efficient immune responses against PEDV infection.
Collapse
|
6
|
Oral Immunization with Lactobacillus casei Expressing the Porcine Circovirus Type 2 Cap and LTB Induces Mucosal and Systemic Antibody Responses in Mice. Viruses 2021; 13:v13071302. [PMID: 34372508 PMCID: PMC8310122 DOI: 10.3390/v13071302] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/28/2021] [Accepted: 06/28/2021] [Indexed: 02/03/2023] Open
Abstract
Porcine circovirus type 2 (PCV2) causes many diseases in weaned piglets, leading to serious economic losses to the pig industry. This study investigated the immune response following oral administration of Lactobacillus casei ATCC393 (L. casei 393) expressing PCV2 capsid protein (Cap) fusion with the Escherichia coli heat-labile toxin B subunit (LTB) in mice. Recombinant L. casei strains were constructed using plasmids pPG611.1 and pPG612.1. The expression and localization of proteins from recombinant pPG611.1-Cap-LTB (pPG-1-Cap-LTB)/L. casei 393 and pPG612.1-Cap-LTB (pPG-2-Cap-LTB)/L. casei 393 were detected. All recombinant strains were found to be immunogenic by oral administration in mice and developed mucosal and systemic immune responses against PCV2. The titers of specific antibodies in mice administered pPG-2-Cap-LTB/L. casei 393 were higher than those in mice administered pPG-1-Cap-LTB/L. casei 393 in serum and the mucosal samples. The mucosal immune response was not only limited to the gastrointestinal tract but was also generated in other mucosal parts. Thus, the application of recombinant L. casei could aid in vaccine development for PCV2.
Collapse
|
7
|
Li X, Zhang B, Zhang D, Liu S, Ren J. The construction of recombinant Lactobacillus casei vaccine of PEDV and its immune responses in mice. BMC Vet Res 2021; 17:184. [PMID: 33947419 PMCID: PMC8097780 DOI: 10.1186/s12917-021-02885-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 04/15/2021] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Porcine epidemic diarrhea (PED) is a contagious intestinal disease caused by porcine epidemic diarrhea virus (PEDV) characterized by vomiting, diarrhea, anorexia, and dehydration, which have caused huge economic losses around the world. At present, vaccine immunity is still the most effective method to control the spread of PED. In this study, we have constructed a novel recombinant L. casei-OMP16-PEDVS strain expressing PEDVS protein of PEDV and OMP16 protein of Brucella abortus strain. To know the immunogenicity of the recombinant L. casei-OMP16-PEDVS candidate vaccine, it was compared with BL21-OMP16-PEDVS-F, BL21-OMP16-PEDVS, and BL21-PEDVS recombinant protein. RESULTS The results showed that we could detect higher levels of IgG, neutralizing antibody, IL-4, IL-10, and INF-γ in serum and IgA in feces of L. casei-OMP16-PEDVS immunized mice, which indicated that L. casei-OMP16-PEDVS candidate vaccine could induce higher levels of humoral immunity, cellular immunity, and mucosal immunity. CONCLUSION Therefore, L. casei-OMP16-PEDVS is a promising candidate vaccine for prophylaxis of PEDV infection.
Collapse
Affiliation(s)
- Xiaowen Li
- Swine Research Institute of New Hope Group, Tai'an, China
- Shandong Swine Herd Health Big Data and Intelligent Monitoring Engineering Laboratory, Tai'an, China
- College of Animal Science and Technology, Shandong Agricultural University, Tai'an, China
| | - Bingzhou Zhang
- Swine Research Institute of New Hope Group, Tai'an, China
- Shandong Swine Herd Health Big Data and Intelligent Monitoring Engineering Laboratory, Tai'an, China
| | | | - Sidang Liu
- College of Animal Science and Technology, Shandong Agricultural University, Tai'an, China.
| | - Jing Ren
- Shandong Swine Herd Health Big Data and Intelligent Monitoring Engineering Laboratory, Tai'an, China.
- Shandong Provincial Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou, 253023, China.
| |
Collapse
|
8
|
Bai Y, Wang G, Qi H, Wang Y, Xu C, Yue L, Hou X, Yu L. Immunogenicity of 987P fimbriae of enterotoxigenic Escherichia coli surface-displayed on Lactobacillus casei. Res Vet Sci 2020; 128:308-314. [DOI: 10.1016/j.rvsc.2019.12.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 12/10/2019] [Accepted: 12/22/2019] [Indexed: 12/27/2022]
|
9
|
Kuczkowska K, Copland A, Øverland L, Mathiesen G, Tran AC, Paul MJ, Eijsink VGH, Reljic R. Inactivated Lactobacillus plantarum Carrying a Surface-Displayed Ag85B-ESAT-6 Fusion Antigen as a Booster Vaccine Against Mycobacterium tuberculosis Infection. Front Immunol 2019; 10:1588. [PMID: 31354727 PMCID: PMC6632704 DOI: 10.3389/fimmu.2019.01588] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 06/25/2019] [Indexed: 12/11/2022] Open
Abstract
Vaccination is considered the most effective strategy for controlling tuberculosis (TB). The existing vaccine, the Bacille Calmette-Guérin (BCG), although partially protective, has a number of limitations. Therefore, there is a need for developing new TB vaccines and several strategies are currently exploited including the use of viral and bacterial delivery vectors. We have previously shown that Lactobacillus plantarum (Lp) producing Ag85B and ESAT-6 antigens fused to a dendritic cell-targeting peptide (referred to as Lp_DC) induced specific immune responses in mice. Here, we analyzed the ability of two Lp-based vaccines, Lp_DC and Lp_HBD (in which the DC-binding peptide was replaced by an HBD-domain directing the antigen to non-phagocytic cells) to activate antigen-presenting cells, induce specific immunity and protect mice from Mycobacterium tuberculosis infection. We tested two strategies: (i) Lp as BCG boosting vaccine (a heterologous regimen comprising parenteral BCG immunization followed by intranasal Lp boost), and (ii) Lp as primary vaccine (a homologous regimen including subcutaneous priming followed by intranasal boost). The results showed that both Lp constructs applied as a BCG boost induced specific cellular immunity, manifested in T cell proliferation, antigen-specific IFN-γ responses and multifunctional T cells phenotypes. More importantly, intranasal boost with Lp_DC or Lp_HBD enhanced protection offered by BCG, as shown by reduced M. tuberculosis counts in lungs. These findings suggest that Lp constructs could be developed as a potential mucosal vaccine platform against mycobacterial infections.
Collapse
Affiliation(s)
- Katarzyna Kuczkowska
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | - Alastair Copland
- Institute for Infection and Immunity, St. George's University of London, London, United Kingdom.,College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Lise Øverland
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | - Geir Mathiesen
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | - Andy C Tran
- Institute for Infection and Immunity, St. George's University of London, London, United Kingdom
| | - Mathew J Paul
- Institute for Infection and Immunity, St. George's University of London, London, United Kingdom
| | - Vincent G H Eijsink
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | - Rajko Reljic
- Institute for Infection and Immunity, St. George's University of London, London, United Kingdom
| |
Collapse
|
10
|
Blocking HIV-1 Infection by Chromosomal Integrative Expression of Human CD4 on the Surface of Lactobacillus acidophilus ATCC 4356. J Virol 2019; 93:JVI.01830-18. [PMID: 30728264 DOI: 10.1128/jvi.01830-18] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 01/27/2019] [Indexed: 12/13/2022] Open
Abstract
Lactobacillus bacteria are potential delivery vehicles for biopharmaceutical molecules because they are well-recognized as safe microorganisms that naturally inhabit the human body. The goal of this study was to employ these lactobacilli to combat human immunodeficiency virus type 1 (HIV-1) infection and transmission. By using a chromosomal integration method, we engineered Lactobacillus acidophilus ATCC 4356 to display human CD4, the HIV-1 receptor, on the cell surface. Since human CD4 can bind to any infectious HIV-1 particles, the engineered lactobacilli can potentially capture HIV-1 of different subtypes and prevent infection. Our data demonstrate that the CD4-carrying bacteria are able to adsorb HIV-1 particles and reduce infection significantly in vitro and also block intrarectal HIV-1 infection in a humanized mouse model in preliminary tests in vivo Our results support the potential of this approach to decrease the efficiency of HIV-1 sexual transmission.IMPORTANCE In the absence of an effective vaccine, alternative approaches to block HIV-1 infection and transmission with commensal bacteria expressing antiviral proteins are being considered. This report provides a proof-of-concept by using Lactobacillus bacteria stably expressing the HIV-1 receptor CD4 to capture and neutralize HIV-1 in vitro and in a humanized mouse model. The stable expression of antiviral proteins, such as CD4, following genomic integration of the corresponding genes into this Lactobacillus strain may contribute to the prevention of HIV-1 sexual transmission.
Collapse
|
11
|
|
12
|
LeCureux JS, Dean GA. Lactobacillus Mucosal Vaccine Vectors: Immune Responses against Bacterial and Viral Antigens. mSphere 2018; 3:e00061-18. [PMID: 29769376 PMCID: PMC5956152 DOI: 10.1128/msphere.00061-18] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Lactic acid bacteria (LAB) have been utilized since the 1990s for therapeutic heterologous gene expression. The ability of LAB to elicit an immune response against expressed foreign antigens has led to their exploration as potential mucosal vaccine candidates. LAB vaccine vectors offer many attractive advantages: simple, noninvasive administration (usually oral or intranasal), the acceptance and stability of genetic modifications, relatively low cost, and the highest level of safety possible. Experimentation using LAB of the genus Lactobacillus has become popular in recent years due to their ability to elicit strong systemic and mucosal immune responses. This article reviews Lactobacillus vaccine constructs, including Lactobacillus species, antigen expression, model organisms, and in vivo immune responses, with a primary focus on viral and bacterial antigens.
Collapse
Affiliation(s)
- Jonathan S LeCureux
- Department of Natural and Applied Sciences, Evangel University, Springfield, Missouri, USA
| | - Gregg A Dean
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
13
|
Torres-Barredo GA, Atarashi H, Kajikawa A, Hirata A, Endo A, Nakagawa J. Intracellular localization of sirtuin and cell length analysis of Lactobacillus paracasei suggest possible role of sirtuin in cell division and cell shape regulation. Biosci Biotechnol Biochem 2018; 82:1-10. [PMID: 29499629 DOI: 10.1080/09168451.2018.1443790] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 02/13/2018] [Indexed: 10/17/2022]
Abstract
Sirtuin has been associated in prolonging lifespan of different model organisms. It has been shown to have an enzymatic activity of NAD+-dependent protein deacetylation targeting acetylated proteins. To determine targets and possible roles of sirtuin (LpSirA) in the Lactobacillus paracasei BL23, deletion (ΔsirA), sirtuin overexpressor (highsirA) and GFP fusion (highsirA-Venus) strains were generated, and microscopic localization and cell length analysis were done. Microscopic analysis revealed localization of LpSirA at cell division plates, at cell poles and all throughout the cell length in a spiral manner. Cell length analysis revealed that 46.9% of the ΔsirA cells were observed to be shorter (<2 μm), whereas 12.6% of the highsirA cells were observed to be longer (>4 μm) in comparison with the wild-type with only 17.1% short cells and 5.3% long cells. Our results suggest that sirtuin may have an essential role in cell division and cell shape regulation.
Collapse
Affiliation(s)
| | - Hotaka Atarashi
- a Graduate School of Bioindustry , Tokyo University of Agriculture , Abashiri City , Japan
| | - Akinobu Kajikawa
- b Department of Applied Biology and Chemistry , Tokyo University of Agriculture , Tokyo , Japan
| | - Aiko Hirata
- c Graduate School of Frontier Sciences , The University of Tokyo , Kashiwa , Japan
| | - Akihito Endo
- a Graduate School of Bioindustry , Tokyo University of Agriculture , Abashiri City , Japan
| | - Junichi Nakagawa
- a Graduate School of Bioindustry , Tokyo University of Agriculture , Abashiri City , Japan
| |
Collapse
|
14
|
Oral Immunization against PEDV with Recombinant Lactobacillus casei Expressing Dendritic Cell-Targeting Peptide Fusing COE Protein of PEDV in Piglets. Viruses 2018; 10:v10030106. [PMID: 29494530 PMCID: PMC5869499 DOI: 10.3390/v10030106] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 02/25/2018] [Accepted: 02/27/2018] [Indexed: 01/19/2023] Open
Abstract
Porcine epidemic diarrhea (PED) is a highly contagious disease in newborn piglets. In our previous study, a genetically engineered Lactobacillus casei oral vaccine (pPG-COE-DCpep/L393) expressing a dendritic cell (DC)-targeting peptide fused with porcine epidemic diarrhea virus (PEDV) COE antigen was developed. This vaccine induced significant levels of anti-PEDV specific IgG and IgA antibody responses in mice, indicating a potential strategy against PEDV infection. In this study, pPG-COE-DCpep/L393 was used for oral vaccination of newborn piglets against PEDV. We then assessed the immune responses and protection efficacy of pPG-COE-DCpep/L393. An indirect enzyme-linked immunosorbent assay (ELISA) showed that the recombinant Lactobacillus vaccine elicits a specific systemic and mucosal immune response. The T-helper cells mediated by pPG-COE-DCpep/L393 and PEDV infection display a Th1 phenotype. The histopathological results showed that pPG-COE-DCpep/L393 promotes lymphocyte proliferation and effectively protects piglets against PEDV infection. The transforming growth factor-β level indicated that the recombinant Lactobacillus vaccine plays a role in anti-inflammatory responses in mesenteric lymph nodes during PEDV infection. These results show that pPG-COE-DCpep/L393 is a potential vaccine against PEDV infection.
Collapse
|
15
|
Recombinant Lactobacillus casei expressing Clostridium perfringens toxoids α, β2, ε and β1 gives protection against Clostridium perfringens in rabbits. Vaccine 2017. [DOI: 10.1016/j.vaccine.2017.05.076] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
16
|
Trombert A. Recombinant lactic acid bacteria as delivery vectors of heterologous antigens: the future of vaccination? Benef Microbes 2016; 6:313-24. [PMID: 25245573 DOI: 10.3920/bm2014.0068] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
UNLABELLED Lactic acid bacteria (LABs) are good candidates for the development of new oral vaccines and are attractive alternatives to attenuated pathogens. This review focuses on the use of wild-type and recombinant lactococci and lactobacilli with emphasis on their molecular design, immunomodulation and treatment of bacterial infections. The majority of studies related to recombinant LABs have focused on Lactococcus lactis, however, molecular tools have been successfully used for Lactobacillus spp. RESEARCH Recombinant lactobacilli and lactococci have several health benefits, such as immunomodulation, restoration of the microbiota, synthesis of antimicrobial substances and inhibition of virulence factors. In addition, protective immune responses that are well tolerated are induced by the expression of heterologous antigens from recombinant probiotics.
Collapse
Affiliation(s)
- A Trombert
- Center for Genomics and Bioinformatics, Faculty of Science, Universidad Mayor, Camino La Piramide 5750, Huechuraba, Santiago, Chile
| |
Collapse
|
17
|
Al Hafid N, Christodoulou J. Phenylketonuria: a review of current and future treatments. Transl Pediatr 2015; 4:304-17. [PMID: 26835392 PMCID: PMC4728993 DOI: 10.3978/j.issn.2224-4336.2015.10.07] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 10/26/2015] [Indexed: 01/16/2023] Open
Abstract
Phenylketonuria (PKU) is an autosomal recessive inborn error of metabolism caused by a deficiency in the hepatic enzyme phenylalanine hydroxylase (PAH). If left untreated, the main clinical feature is intellectual disability. Treatment, which includes a low Phe diet supplemented with amino acid formulas, commences soon after diagnosis within the first weeks of life. Although dietary treatment has been successful in preventing intellectual disability in early treated PKU patients, there are major issues with dietary compliance due to palatability of the diet. Other potential issues associated with dietary therapy include nutritional deficiencies especially vitamin D and B12. Suboptimal outcomes in cognitive and executive functioning have been reported in patients who adhere poorly to dietary therapy. There have been continuous attempts at improving the quality of medical foods including their palatability. Advances in dietary therapy such as the use of large neutral amino acids (LNAA) and glycomacropeptides (GMP; found within the whey fraction of bovine milk) have been explored. Gene therapy and enzyme replacement or substitution therapy have yielded more promising data in the recent years. In this review the current and possible future treatments for PKU are discussed.
Collapse
|
18
|
Transformation Kinetics During Fermented Milk Production Using Lactobacillus Johnsonii (La1) and Streptococcus Thermophillus: A Comparison With Yogurt Inoculum. FOOD BIOPHYS 2015. [DOI: 10.1007/s11483-015-9406-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
19
|
da Silva AJ, Zangirolami TC, Novo-Mansur MTM, Giordano RDC, Martins EAL. Live bacterial vaccine vectors: an overview. Braz J Microbiol 2015; 45:1117-29. [PMID: 25763014 PMCID: PMC4323283 DOI: 10.1590/s1517-83822014000400001] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Accepted: 04/17/2014] [Indexed: 02/07/2023] Open
Abstract
Genetically attenuated microorganisms, pathogens, and some commensal bacteria can be engineered to deliver recombinant heterologous antigens to stimulate the host immune system, while still offering good levels of safety. A key feature of these live vectors is their capacity to stimulate mucosal as well as humoral and/or cellular systemic immunity. This enables the use of different forms of vaccination to prevent pathogen colonization of mucosal tissues, the front door for many infectious agents. Furthermore, delivery of DNA vaccines and immune system stimulatory molecules, such as cytokines, can be achieved using these special carriers, whose adjuvant properties and, sometimes, invasive capacities enhance the immune response. More recently, the unique features and versatility of these vectors have also been exploited to develop anti-cancer vaccines, where tumor-associated antigens, cytokines, and DNA or RNA molecules are delivered. Different strategies and genetic tools are constantly being developed, increasing the antigenic potential of agents delivered by these systems, opening fresh perspectives for the deployment of vehicles for new purposes. Here we summarize the main characteristics of the different types of live bacterial vectors and discuss new applications of these delivery systems in the field of vaccinology.
Collapse
Affiliation(s)
- Adilson José da Silva
- Departamento de Engenharia Química Universidade Federal de São Carlos São CarlosSP Brazil Departamento de Engenharia Química, Universidade Federal de São Carlos, São Carlos, SP, Brazil
| | - Teresa Cristina Zangirolami
- Departamento de Engenharia Química Universidade Federal de São Carlos São CarlosSP Brazil Departamento de Engenharia Química, Universidade Federal de São Carlos, São Carlos, SP, Brazil
| | - Maria Teresa Marques Novo-Mansur
- Departamento de Genética e Evolução Universidade Federal de São Carlos São CarlosSP Brazil Departamento de Genética e Evolução, Universidade Federal de São Carlos, São Carlos, SP, Brazil
| | - Roberto de Campos Giordano
- Departamento de Engenharia Química Universidade Federal de São Carlos São CarlosSP Brazil Departamento de Engenharia Química, Universidade Federal de São Carlos, São Carlos, SP, Brazil
| | - Elizabeth Angélica Leme Martins
- Centro de Biotecnologia Instituto Butantan São PauloSP Brazil Centro de Biotecnologia, Instituto Butantan, São Paulo, SP, Brazil
| |
Collapse
|
20
|
Su J, Li J, Zheng H, You Y, Luo X, Li Y, Li X, Ma X, Li J, Dou Y, Cai X. Adjuvant effects of L. acidophilus LW1 on immune responses to the foot-and-mouth disease virus DNA vaccine in mice. PLoS One 2014; 9:e104446. [PMID: 25119375 PMCID: PMC4131892 DOI: 10.1371/journal.pone.0104446] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Accepted: 07/09/2014] [Indexed: 01/18/2023] Open
Abstract
The adjuvant effects of Lactobacillus acidophilus on DNA vaccination are not fully understood. It has been hypothesized that swine-derived Lactobacillus acidophilus SW1 (LASW1) could function as an immune adjuvant to enhance antigen-specific immune responses after foot-and-mouth disease (FMD) DNA vaccination in mice. To evaluate the effect of oral LASW1 on the immune response to a DNA vaccine (pRC/CMV-vp1) harboring FMD VP1 gene, anti-FMDV antibody and its isotypes, T-cell proliferation, and cytokine detection were investigated. The results showed that LASW1 was able to enhance FMDV-specific antibody levels and FMDV-neutralizing antibodies. After a booster vaccine, the anti-FMDV antibody titers and FMDV-neutralizing antibodies levels induced by pRC/CMV-vp1 were higher in mice treated with LSAW1 than in the group immunized with pRC/CMV-vp1 alone (the control). Using T-cell proliferation, the stimulation index of the LASW1 group was significantly higher in response to ConA and 146S antigen (P<0.05) than in the control group. Importantly, higher concentrations of IFN-γ and IFN-γ-producing cells were also observed in splenocytes isolated from the experimental LASW1 mice, indicating that INF-γ secretion is important to the immune response to LASW1. The results indicate that LASW1 is a promising immune adjuvant in DNA vaccination against FMD when administrated orally.
Collapse
Affiliation(s)
- Junhong Su
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou, PR China
| | - Jidong Li
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou, PR China
- School of Agriculture, Ningxia University, Yinchuan, RP China
| | - Haixue Zheng
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou, PR China
| | - Yanan You
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou, PR China
| | - Xuenong Luo
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou, PR China
| | - Yungang Li
- Shandong Provincial Center for Animal Disease Prevention and Control, Jinan, PR China
| | - Xueqiang Li
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou, PR China
| | - Xusheng Ma
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou, PR China
| | - Junjun Li
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou, PR China
| | - Yongxi Dou
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou, PR China
- * E-mail: (XPC); (YXD)
| | - Xuepeng Cai
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou, PR China
- * E-mail: (XPC); (YXD)
| |
Collapse
|
21
|
Wen LJ, Hou XL, Wang GH, Yu LY, Wei XM, Liu JK, Liu Q, Wei CH. Immunization with recombinant Lactobacillus casei strains producing K99, K88 fimbrial protein protects mice against enterotoxigenic Escherichia coli. Vaccine 2012; 30:3339-49. [DOI: 10.1016/j.vaccine.2011.08.036] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Revised: 07/25/2011] [Accepted: 08/05/2011] [Indexed: 11/24/2022]
|
22
|
Liao N, Yin Y, Sun G, Xiang C, Liu D, Yu HD, Wang X. Colonization and distribution of segmented filamentous bacteria (SFB) in chicken gastrointestinal tract and their relationship with host immunity. FEMS Microbiol Ecol 2012; 81:395-406. [PMID: 22429007 DOI: 10.1111/j.1574-6941.2012.01362.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2011] [Revised: 03/03/2012] [Accepted: 03/06/2012] [Indexed: 11/30/2022] Open
Abstract
Uncultivable segmented filamentous bacteria (SFB) reside in the gastrointestinal (GI) tract of mammals and can boost the host immunity. Immunoglobulin A (IgA) from mother's milk has been previously shown to be a key factor in regulating SFB colonization. Because neonatal chicken cannot acquire IgA from maternal milk, they are a good model to examine the role of IgA in SFB colonization. Here, we used the fluorescent in situ hybridization (FISH) and quantitative PCR (qPCR) to monitor the colonization and distribution of SFB in chickens aged from 2-day-old to 6-week-old. Early SFB colonization, which primarily occurred in the ileal mucosa (< 13 days old), was IgA independent. From the age of 17-42 days, there was an increase in IgA in the gut mucosa, which was correlated with a decrease in SFB. To examine the effect of probiotics and immunosuppression on SFB colonization, we treated the chickens by feeding them Lactobacillus delbrueckii or giving them a subcutaneous injection of cyclophosphamide (CTX). Feeding lactobacilli at birth rendered SFB colonization occurring 4 days earlier, while CTX treatment increases the SFB colonization through reducing the other non-SFB bacteria. Altogether, our data suggest that early colonization of SFB in chicken occurs independently of IgA and the population of SFB in the GI tract of chicken may be manipulated from birth via probiotic or CTX treatment.
Collapse
Affiliation(s)
- Ningbo Liao
- State Key Laboratory Breeding Base for Zhejiang Sustainable Pest and Disease Control, Institute of Plant Protection and Microbiology, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | | | | | | | | | | | | |
Collapse
|
23
|
Zhao LL, Liu M, Ge JW, Qiao XY, Li YJ, Liu DQ. Expression of infectious pancreatic necrosis virus (IPNV) VP2-VP3 fusion protein in Lactobacillus casei and immunogenicity in rainbow trouts. Vaccine 2012; 30:1823-9. [PMID: 22234263 DOI: 10.1016/j.vaccine.2011.12.132] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Revised: 12/11/2011] [Accepted: 12/28/2011] [Indexed: 10/14/2022]
Abstract
Infectious pancreatic necrosis virus (IPNV) infects wild and cultured salmonids, causing high mortality in juvenile trouts and salmons. IPNV VP2-VP3 fusion gene was constructed by splicing overlap extension (SOE) PCR and inserted into Lactobacillus/Escherichia coli shuttle vectors (pPG1and pPG2) followed by transformation of Lactobacillus casei competent cell to yield two recombinant strains: Lc:PG1-VP2-VP3 (surface-displayed) and Lc:PG2-VP2-VP3 (secretory). Subsequently, juvenile rainbow trouts were inoculated with the recombinant strains via orogastric route. Our results demonstrated that Lactobacillus-derived VP2-VP3 fusion protein could induce production of serum IgM specific for IPNV with neutralizing activity in rainbow trouts. Statistical analyses of IgM levels showed that immunogenicity of Lc:PG1-VP2-VP3 was more powerful than that of Lc:PG2-VP2-VP3 (P<0.001) in rainbow trouts. This result has been confirmed by viral loads reduction analyzed by real-time RT-PCR in orogastrically immunized rainbow trouts after virus challenging. Comparing to trouts received Lactobacillus (control), rainbow trouts orogastrically dosed with Lc:PG1-VP2-VP3 resulted in ∼10-fold reduction in viral loads on day 10 post-virus challenging, and ∼4-fold did by Lc:PG2-VP2-VP3. Taken together, Lc:PG1-VP2-VP3 functions as novel mucosal vaccine against IPNV infection in rainbow trouts, which most likely come true.
Collapse
Affiliation(s)
- Li-Li Zhao
- Veterinary Microbiology Department, Veterinary Medicine College, Northeast Agricultural University, Harbin, China
| | | | | | | | | | | |
Collapse
|
24
|
Min L, Li-Li Z, Jun-Wei G, Xin-Yuan Q, Yi-Jing L, Di-Qiu L. Immunogenicity of Lactobacillus-expressing VP2 and VP3 of the infectious pancreatic necrosis virus (IPNV) in rainbow trout. FISH & SHELLFISH IMMUNOLOGY 2012; 32:196-203. [PMID: 22138084 DOI: 10.1016/j.fsi.2011.11.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Revised: 11/04/2011] [Accepted: 11/18/2011] [Indexed: 05/31/2023]
Abstract
Infectious pancreatic necrosis virus (IPNV) infects salmonid fish with high mortality and causes serious economic losses to salmonid aquaculture. Lactobacillus strains have a number of properties that make them attractive candidates as delivery vehicles for the presentation to the mucosa of compounds with pharmaceutical interest, in particular vaccines. Here, Lactobacilli/Escherichia coli shuttle vector pPG1 (surface-displayed) or pPG2 (secretory) with the capsid VP2 gene inserted was transformed into Lactobacillus casei to yield two recombinant strains: Lc:PG1-VP2 and Lc:PG2-VP2, respectively. Rainbow trout immunized respectively with Lc:PG1-VP2, Lc:PG2-VP2, Lc:PG1-VP3 and Lc:PG2-VP3 elicited anti-IPNV immune responses (serum IgM) via oral route. Statistical results of serum IgM titer with neutralizing activity showed that immunogenicity of Lc:PG2-VP2 was more powerful than that of Lc:PG1-VP2 (P < 0.001), Lc:PG1-VP3 (P < 0.001) and Lc:PG2-VP3 (P < 0.001), which was confirmed by viral loads reduction analyzed by real-time RT-PCR in orally immunized rainbow trout after virus challenge. Comparing with negative control, rainbow trout orally dosed with Lc:PG2-VP2 resulted in ∼46-fold reduction in virus load on days 10 post viral challenge as well as Lc:PG1-VP2(∼20-fold), Lc:PG2-VP3(∼6-fold) and Lc:PG1-VP3(∼3-fold). Taken together, Lc:PG2-VP2 exhibited a more appropriate candidate as live bacteria vaccine against IPNV infection in rainbow trout.
Collapse
Affiliation(s)
- Liu Min
- Veterinary Microbiology Department, Veterinary Medicine College, Northeast Agricultural University, Harbin 150030, PR China
| | | | | | | | | | | |
Collapse
|
25
|
Kim SK, Bhatnagar I, Kang KH. Development of marine probiotics: prospects and approach. ADVANCES IN FOOD AND NUTRITION RESEARCH 2012; 65:353-62. [PMID: 22361199 DOI: 10.1016/b978-0-12-416003-3.00023-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The term probiotics stands for bacterial strain acting for the upliftment of beneficial gut microflora as well as restricting the growth of decay or disease causing bacteria and other agents. They protect us from disease causing microorganisms and are vital for the digestion of nutrients and food. Researchers are now trying to explore the possibilities of therapeutic applications of probiotics in inflammatory bowel disease, treating diarrhea, eczema prevention in children, and reducing bladder cancer recurrence and urinary tract infections. The best known probiotic strains are Bifidobacteria, Lactobacilli, and Streptococcus thermophilus and can be found in food products such as yoghurts, fermented and unfermented milk, miso, tempeh, and some juices and soy beverages. Thus, as a substitute to the use of antibiotics, the dietary supplementation with probiotic bacteria is being widely employed in the aquaculture industry. This chapter is an attempt to shift the paradigm of research in probiotic area to the marine environment and to find ways of expanding the research in marine probiotics development.
Collapse
Affiliation(s)
- Se-Kwon Kim
- Department of Chemistry, Pukyong National University, Busan, Republic of Korea.
| | | | | |
Collapse
|
26
|
Prophylactic effect of Lactobacillus oral vaccine expressing a Japanese cedar pollen allergen. J Biosci Bioeng 2011; 113:536-41. [PMID: 22196937 DOI: 10.1016/j.jbiosc.2011.11.025] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2011] [Revised: 11/25/2011] [Accepted: 11/28/2011] [Indexed: 11/23/2022]
Abstract
Lactic acid bacteria (LAB) represent an attractive delivery vehicle for oral allergy vaccine because of their safety as a food microorganism as well as their potent adjuvant activity triggering anti-allergic immune response. Here, we report the generation of recombinant LAB expressing a major Japanese cedar pollen allergen Cry j 1 (Cry j 1-LAB), and their prophylactic effect in vivo. To facilitate heterologous expression, the codon usage in the Cry j 1 gene was optimized for the host LAB strain Lactobacillus plantarum by the recursive PCR-based exhaustive site-directed mutagenesis. Use of the codon-optimized Cry j 1 cDNA and a lactate dehydrogenase gene fusion system led to a successful production of recombinant Cry j 1 in L. plantarum NCL21. We also found that oral vaccination with the Cry j 1-LAB suppressed allergen-specific IgE response and nasal symptoms in a murine model of cedar pollinosis.
Collapse
|
27
|
Liu DQ, Ge JW, Qiao XY, Jiang YP, Liu SM, Li YJ. High-level mucosal and systemic immune responses induced by oral administration with Lactobacillus-expressed porcine epidemic diarrhea virus (PEDV) S1 region combined with Lactobacillus-expressed N protein. Appl Microbiol Biotechnol 2011; 93:2437-46. [PMID: 22134641 PMCID: PMC7080084 DOI: 10.1007/s00253-011-3734-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Revised: 10/31/2011] [Accepted: 11/15/2011] [Indexed: 12/14/2022]
Abstract
To develop effective mucosal vaccine formulation against porcine epidemic diarrhea virus (PEDV) infection, the DNA fragments encoding spike protein immunodominant region S1 and nucleocapsid N of PEDV were inserted into pPG1 (surface-displayed) or pPG2 (secretory) plasmids followed by electrotransformation into Lactobacillus casei (Lc) to yield four recombinant strains: PG1-S1, PG2-S1, PG1-N, and PG2-N. After intragastric administration, it was observed that live Lc-expressing S1 protein combined with Lc-expressing N protein could elicit much more potent mucosal and systemic immune responses than the former alone (P < 0.001), however slightly inferior to the latter alone (P > 0.05). Furthermore, the surface-displayed mixture (PG1-S1+ PG1-N) revealed stronger immunogenicity than the secretory mixture (PG2-S1+ PG2-N) as well as PEDV-neutralizing potency in vitro (P < 0.001). On 49th day after the last immunization, splenocytes were prepared from mice immunized with surface-displayed mixture, secretory mixture and negative control to be stimulated by purified N and S protein, respectively. The results of ELISA analysis showed that N protein was capable of inducing a higher level of IL-4 (P < 0.001) and IFN-γ (P < 0.001) than S1 protein in the immunized mice. Taken together, Lc-expressed N protein as molecular adjuvant or immunoenhancer was able to effectively facilitate the induction of mucosal and systemic immune responses by Lc-expressing S1 region.
Collapse
Affiliation(s)
- Di-qiu Liu
- Veterinary Microbiology Department, Veterinary Medicine College, Northeast Agricultural University, Harbin, People's Republic of China
| | | | | | | | | | | |
Collapse
|
28
|
Kajikawa A, Nordone SK, Zhang L, Stoeker LL, LaVoy AS, Klaenhammer TR, Dean GA. Dissimilar properties of two recombinant Lactobacillus acidophilus strains displaying Salmonella FliC with different anchoring motifs. Appl Environ Microbiol 2011; 77:6587-96. [PMID: 21784918 PMCID: PMC3187123 DOI: 10.1128/aem.05153-11] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2011] [Accepted: 07/12/2011] [Indexed: 11/20/2022] Open
Abstract
Display of heterologous antigens on the cell surface is considered a useful technique for vaccine delivery by recombinant lactobacilli. In this study, two recombinant Lactobacillus acidophilus derivatives displaying Salmonella flagellin (FliC) were constructed using different anchor motifs. In one instance, the FliC protein was fused to the C-terminal region of a cell envelope proteinase (PrtP) and was bound to the cell wall by electrostatic bonds. In the other case, the same antigen was conjugated to the anchor region of mucus binding protein (Mub) and was covalently associated with the cell wall by an LPXTG motif. These two recombinant L. acidophilus cell surface displays resulted in dissimilar maturation and cytokine production by human myeloid dendritic cells. The surface-associated antigen was highly sensitive to simulated gastric and small intestinal juices. By supplementation with bicarbonate buffer and soybean trypsin inhibitor, the cell surface antigen was protected from proteolytic enzymes during gastric challenge in vitro. The protective reagents also increased the viability of the L. acidophilus cells upon challenge with simulated digestive juices. These results demonstrate the importance of protecting cells and their surface-associated antigens during oral immunization.
Collapse
Affiliation(s)
- Akinobu Kajikawa
- Center for Comparative Medicine and Translational Research, Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Bermúdez-Humarán LG, Kharrat P, Chatel JM, Langella P. Lactococci and lactobacilli as mucosal delivery vectors for therapeutic proteins and DNA vaccines. Microb Cell Fact 2011; 10 Suppl 1:S4. [PMID: 21995317 PMCID: PMC3231930 DOI: 10.1186/1475-2859-10-s1-s4] [Citation(s) in RCA: 159] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Food-grade Lactic Acid Bacteria (LAB) have been safely consumed for centuries by humans in fermented foods. Thus, they are good candidates to develop novel oral vectors, constituting attractive alternatives to attenuated pathogens, for mucosal delivery strategies. Herein, this review summarizes our research, up until now, on the use of LAB as mucosal delivery vectors for therapeutic proteins and DNA vaccines. Most of our work has been based on the model LAB Lactococcus lactis, for which we have developed efficient genetic tools, including expression signals and host strains, for the heterologous expression of therapeutic proteins such as antigens, cytokines and enzymes. Resulting recombinant lactococci strains have been tested successfully for their prophylactic and therapeutic effects in different animal models: i) against human papillomavirus type 16 (HPV-16)-induced tumors in mice, ii) to partially prevent a bovine β-lactoglobulin (BLG)-allergic reaction in mice and iii) to regulate body weight and food consumption in obese mice. Strikingly, all of these tools have been successfully transposed to the Lactobacillus genus, in recent years, within our laboratory. Notably, anti-oxidative Lactobacillus casei strains were constructed and tested in two chemically-induced colitis models. In parallel, we also developed a strategy based on the use of L. lactis to deliver DNA at the mucosal level, and were able to show that L. lactis is able to modulate the host response through DNA delivery. Today, we consider that all of our consistent data, together with those obtained by other groups, demonstrate and reinforce the interest of using LAB, particularly lactococci and lactobacilli strains, to develop novel therapeutic protein mucosal delivery vectors which should be tested now in human clinical trials.
Collapse
Affiliation(s)
- Luis G Bermúdez-Humarán
- INRA, UMR1319 Micalis, Commensal and Probiotics-Host Interactions Laboratory, Domaine de Vilvert, 78352 Jouy-en-Josas Cedex, France
| | | | | | | |
Collapse
|
30
|
Tang L, Li Y. Oral immunization of mice with recombinant Lactococcus lactis expressing porcine transmissible gastroenteritis virus spike glycoprotein. Virus Genes 2011; 39:238-45. [PMID: 19629668 PMCID: PMC7089002 DOI: 10.1007/s11262-009-0390-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2009] [Accepted: 07/09/2009] [Indexed: 11/29/2022]
Abstract
Lactococcus lactis NZ9000 was selected as an antigen delivery vehicle for mucosal immunization against porcine transmissible gastroenteritis virus (TGEV) infection. An approximately 70 kDa fragment of the N-terminal globular domain of the spike (S) protein (SN protein) from the coronavirus TGEV was used as the transmissible gastroenteritis virus antigen model. Recombinant L. lactis, expressing the SN protein, was constructed with the pNZ8112 plasmid. Expression and localization of the transcribed SN protein from the recombinant LNZ9000-rTGEV-SN were detected via SDS-PAGE, Western blot, and immunofluorescence. BALB/c mice, orally immunized with LNZ9000-rTGEV-SN, produced local mucosal immune responses against TGEV. The induced antibodies demonstrated neutralizing effects on TGEV infection. These data indicated that the recombinant L. lactis could be a valuable tool in the development of future vaccines against TGEV.
Collapse
Affiliation(s)
- Lijie Tang
- Life Science Department, Northeast Agricultural University, Harbin 150030, People's Republic of China
| | | |
Collapse
|
31
|
Transformation of, and heterologous protein expression in, Lactobacillus agilis and Lactobacillus vaginalis isolates from the chicken gastrointestinal tract. Appl Environ Microbiol 2010; 77:220-8. [PMID: 21075881 DOI: 10.1128/aem.02006-10] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Lactobacilli are naturally found in the gastrointestinal tract of chickens, and there is interest in utilizing autochthonous strains for the delivery of therapeutic proteins. Previously we identified three chicken-derived Lactobacillus strains, Lactobacillus agilis La3, Lactobacillus vaginalis Lv5, and Lactobacillus crispatus Lc9, which persist in the gastrointestinal tract of chickens fed either a commercial or high-protein diet. In the current study, we investigated the ability to electrotransform these strains, determined plasmid vector stability, and compared reporter gene expression directed by several different promoters. The La3 and Lv5 strains were reproducibly transformed with efficiencies of 10(8) and 10(6) transformants per microgram of plasmid DNA, respectively. The third strain tested, L. crispatus Lc9, was recalcitrant to all transformation protocols examined. The plasmid vectors pTRK563 and pTRKH2 were maintained over 100 generations in La3 and Lv5, respectively. The ability of La3 and Lv5 to express the heterologous reporter gene gfp was analyzed using heterologous and homologous promoters. Transformants of both La3 and Lv5 containing the La3 ldhL promoter were the most fluorescent. To our knowledge, this is the first report of successful transformation and heterologous protein expression in L. agilis and L. vaginalis. The ability of these strains to express heterologous proteins in vitro indicates their potential utility as in vivo delivery vectors for therapeutic peptides to the chicken gastrointestinal tract.
Collapse
|
32
|
Kim SY, Shin KS, Lee H. Determination of primary factors with adhesive property of Lactobacillus brevis FSB-1 to rat colonic mucin. Food Sci Biotechnol 2010. [DOI: 10.1007/s10068-010-0188-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
33
|
del Rio B, Fuente JL, Neves V, Dattwyler R, Seegers JFML, Gomes-Solecki M. Platform technology to deliver prophylactic molecules orally: an example using the Class A select agent Yersinia pestis. Vaccine 2010; 28:6714-22. [PMID: 20699130 DOI: 10.1016/j.vaccine.2010.07.084] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2010] [Revised: 07/09/2010] [Accepted: 07/27/2010] [Indexed: 12/30/2022]
Abstract
Consumed for centuries, lactic acid bacteria are excellent candidates for the development of safe mucosal delivery vehicles for prophylactic and therapeutic molecules. We have recently reported that the immune response to an effective OspA-expressing L. plantarum vaccine for Lyme disease is modulated by the lipid modification of the antigen. In this study, we investigated if this technology can be applied to developing vaccines for other diseases by focusing on the Class A select agent, Yersinia pestis. We used a number of biochemistry and immunology techniques to determine the localization of the immunogen in our delivery vehicle and to evaluate the mucosal as well as the systemic immune response to the immunogen. We found that only LcrV cloned downstream of the signal sequence of B. burgdorferi OspA ((ss)LcrV), but not wildtype LcrV (LcrV), is localized to the desired peptidoglycan layer of the delivery vehicle. In addition, only mice that received L. plantarum expressing (ss)LcrV produced significant titers of IgG antibody as well as IgA in distant mucosal sites such as lungs and vagina. Furthermore, only L. plantarum expressing (ss)LcrV induced significant amounts of pro-inflammatory cytokines TNFα, IL-12, IFNγ and IL-6 as well as anti-inflammatory IL-10 in human peripheral blood mononuclear cells derived dendritic cells, suggesting that the mechanism by which LcrV-expressing L. plantarum stimulates the immune response involves polarization to Th1 mediated immunity with some involvement of Th2. The study reported here proves that this system is a platform technology to develop oral vaccines for multiple diseases.
Collapse
Affiliation(s)
- Beatriz del Rio
- Department of Molecular Sciences, UTHSC, Memphis, TN 38163, USA
| | | | | | | | | | | |
Collapse
|
34
|
Herich R, Bomba A, Nemcova R, Gancarcikova S. The Influence of Short-term and Continuous Administration of Lactobacillus casei on Basic Haematological and Immunological Parameters in Gnotobiotic Piglets. FOOD AGR IMMUNOL 2010. [DOI: 10.1080/09540109999672] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
|
35
|
del Rio B, Seegers JFML, Gomes-Solecki M. Immune response to Lactobacillus plantarum expressing Borrelia burgdorferi OspA is modulated by the lipid modification of the antigen. PLoS One 2010; 5:e11199. [PMID: 20585451 PMCID: PMC2887847 DOI: 10.1371/journal.pone.0011199] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2010] [Accepted: 05/25/2010] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Over the past decade there has been increasing interest in the use of lactic acid bacteria as mucosal delivery vehicles for vaccine antigens, microbicides and therapeutics. We investigated the mechanism by which a mucosal vaccine based in recombinant lactic acid bacteria breaks the immunological tolerance of the gut in order to elicit a protective immune response. METHODOLOGY/PRINCIPAL FINDINGS We analyzed how the lipid modification of OspA affects the localization of the antigen in our delivery vehicle using a number of biochemistry techniques. Furthermore, we examined how OspA-expressing L. plantarum breaks the oral tolerance of the gut by stimulating human intestinal epithelial cells, peripheral blood mononuclear cells and monocyte derived dendritic cells and measuring cytokine production. We show that the leader peptide of OspA targets the protein to the cell envelope of L. plantarum, and it is responsible for protein export across the membrane. Mutation of the lipidation site in OspA redirects protein localization within the cell envelope. Further, we show that lipidated-OspA-expressing L. plantarum does not induce secretion of the pro-inflammatory cytokine IL-8 by intestinal epithelial cells. In addition, it breaks oral tolerance of the gut via Th1/Th2 cell mediated immunity, as shown by the production of pro- and anti-inflammatory cytokines by human dendritic cells, and by the production of IgG2a and IgG1 antibodies, respectively. CONCLUSIONS/SIGNIFICANCE Lipid modification of OspA expressed in L. plantarum modulates the immune response to this antigen through a Th1/Th2 immune response.
Collapse
Affiliation(s)
- Beatriz del Rio
- Department of Molecular Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | | | - Maria Gomes-Solecki
- Department of Molecular Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Biopeptides Corp., Valhalla, New York, and Memphis, Tennessee, United States of America
| |
Collapse
|
36
|
Immunogenicity and protective efficacy of orally or intranasally administered recombinant Lactobacillus casei expressing ETEC K99. Vaccine 2010; 28:4113-8. [DOI: 10.1016/j.vaccine.2009.05.088] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2009] [Revised: 05/26/2009] [Accepted: 05/31/2009] [Indexed: 11/17/2022]
|
37
|
Probiotic Lactobacillus casei Expressing Human Lactoferrin Elevates Antibacterial Activity in the Gastrointestinal Tract. Biometals 2010; 23:543-54. [DOI: 10.1007/s10534-010-9298-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2009] [Accepted: 01/25/2010] [Indexed: 01/02/2023]
|
38
|
Qiao X, Li G, Wang X, Li X, Liu M, Li Y. Recombinant porcine rotavirus VP4 and VP4-LTB expressed in Lactobacillus casei induced mucosal and systemic antibody responses in mice. BMC Microbiol 2009; 9:249. [PMID: 19958557 PMCID: PMC2797526 DOI: 10.1186/1471-2180-9-249] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2008] [Accepted: 12/04/2009] [Indexed: 12/15/2022] Open
Abstract
Background Porcine rotavirus infection is a significant cause of morbidity and mortality in the swine industry necessitating the development of effective vaccines for the prevention of infection. Immune responses associated with protection are primarily mucosal in nature and induction of mucosal immunity is important for preventing porcine rotavirus infection. Results Lactobacillus casei expressing the major protective antigen VP4 of porcine rotavirus (pPG612.1-VP4) or VP4-LTB (heat-labile toxin B subunit from Echerichia coli) (pPG612.1-VP4-LTB) fusion protein was used to immunize mice orally. The expression of recombinant pPG612.1-VP4 and pPG612.1-VP4-LTB was confirmed by SDS-PAGE and Western blot analysis and surface-displayed expression on L. casei was verified by immunofluorescence. Mice orally immunized with recombinant protein-expressing L. casei produced high levels of serum immunoglobulin G (IgG) and mucosal IgA. The IgA titters from mice immunized with pPG612.1-VP4-LTB were higher than titters from pPG612.1-VP4-immunized mice. The induced antibodies demonstrated neutralizing effects on RV infection. Conclusion These results demonstrated that VP4 administered in the context of an L. casei expression system is an effective method for stimulating mucosal immunity and that LTB served to further stimulate mucosal immunity suggesting that this strategy can be adapted for use in pigs.
Collapse
Affiliation(s)
- Xinyuan Qiao
- Department of Preventive Veterinary, College of Veterinary, Northeast Agricultural University, 59 Mucai Street, Harbin, PR China.
| | | | | | | | | | | |
Collapse
|
39
|
Bermúdez-Humarán LG, Langella P. Utilisation des bactéries lactiques comme vecteurs vaccinaux. REVUE FRANCOPHONE DES LABORATOIRES 2009; 2009:79-89. [PMID: 32518601 PMCID: PMC7270964 DOI: 10.1016/s1773-035x(09)70312-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/17/2009] [Accepted: 10/12/2009] [Indexed: 11/26/2022]
Abstract
Aujourd’hui, nous disposons de données suffisantes qui confortent l’intérêt d’utiliser des bactéries lactiques (BL), notamment des souches des lactocoques et lactobacilles, pour le développement de nouvelles stratégies de vaccination mucosale. Les BL sont des bactéries à Gram positif utilisées depuis des millénaires dans la production d’aliments fermentés. Elles sont donc de bonnes candidates pour le développement de nouvelles stratégies de vectorisation orale et constituent des alternatives attractives aux stratégies vaccinales basées sur des bactéries pathogènes atténuées dont l’utilisation présente des risques sanitaires. Ce chapitre passe en revue la recherche et les progrès les plus récents dans l’utilisation des BL comme vecteurs de délivrance de protéines d’intérêt médical pour développer de nouveaux vaccins.
Collapse
|
40
|
Abstract
Vaccines consisting of transgenic plant-derived antigens offer a new strategy for development of safe, inexpensive vaccines. The vaccine antigens can be eaten with the edible part of the plant or purified from plant material. In phase 1 clinical studies of prototype potato- and corn-based vaccines, these vaccines have been safe and immunogenic without the need for a buffer or vehicle other than the plant cell. Transgenic plant technology is attractive for vaccine development because these vaccines are needle-less, stable, and easy to administer. This chapter examines some early human studies of oral transgenic plant-derived vaccines against enterotoxigenic Escherichia coli infection, norovirus, and hepatitis B.
Collapse
Affiliation(s)
- Alexander V. Karasev
- grid.266456.50000000122849900Department of Plant, Soil & Entomological Sciences, University of Idaho, Moscow, ID 83844-2339 USA
| |
Collapse
|
41
|
Charalampopoulos D, Rastall RA. Development of Mucosal Vaccines Based on Lactic Acid Bacteria. PREBIOTICS AND PROBIOTICS SCIENCE AND TECHNOLOGY 2009. [PMCID: PMC7121035 DOI: 10.1007/978-0-387-79058-9_29] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Today, sufficient data are available to support the use of lactic acid bacteria (LAB), notably lactococci and lactobacilli, as delivery vehicles for the development of new mucosal vaccines. These non-pathogenic Gram-positive bacteria have been safely consumed by humans for centuries in fermented foods. They thus constitute an attractive alternative to the attenuated pathogens (most popular live vectors actually studied) which could recover their pathogenic potential and are thus not totally safe for use in humans. This chapter reviews the current research and advances in the use of LAB as live delivery vectors of proteins of interest for the development of new safe mucosal vaccines. The use of LAB as DNA vaccine vehicles to deliver DNA directly to antigen-presenting cells of the immune system is also discussed.
Collapse
Affiliation(s)
| | - Robert A. Rastall
- Department of Food Biosciences, University of Reading Whiteknights, Reading, UK
| |
Collapse
|
42
|
Oral immunization with recombinant lactobacillus plantarum induces a protective immune response in mice with Lyme disease. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2008; 15:1429-35. [PMID: 18632920 PMCID: PMC2546682 DOI: 10.1128/cvi.00169-08] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Mucosal immunization is advantageous over other routes of antigen delivery because it can induce both mucosal and systemic immune responses. Our goal was to develop a mucosal delivery vehicle based on bacteria generally regarded as safe, such as Lactobacillus spp. In this study, we used the Lyme disease mouse model as a proof of concept. We demonstrate that an oral vaccine based on live recombinant Lactobacillus plantarum protects mice from tick-transmitted Borrelia burgdorferi infection. Our method of expressing vaccine antigens in L. plantarum induces both systemic and mucosal immunity after oral administration. This platform technology can be applied to design oral vaccine delivery vehicles against several microbial pathogens.
Collapse
|
43
|
Adjuvant effect of Lactobacillus casei in a mouse model of gluten sensitivity. Immunol Lett 2008; 119:78-83. [DOI: 10.1016/j.imlet.2008.04.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2008] [Revised: 04/08/2008] [Accepted: 04/28/2008] [Indexed: 01/12/2023]
|
44
|
Álvarez-García E, Alegre-Cebollada J, Batanero E, Monedero V, Pérez-Martínez G, García-Fernández R, Gavilanes JG, Martínez del Pozo Á. Lactococcus lactis as a vehicle for the heterologous expression of fungal ribotoxin variants with reduced IgE-binding affinity. J Biotechnol 2008; 134:1-8. [DOI: 10.1016/j.jbiotec.2007.12.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2007] [Revised: 09/29/2007] [Accepted: 12/13/2007] [Indexed: 02/03/2023]
|
45
|
Yigang XU, Yijing LI. Construction of recombinant Lactobacillus casei efficiently surface displayed and secreted porcine parvovirus VP2 protein and comparison of the immune responses induced by oral immunization. Immunology 2007; 124:68-75. [PMID: 18034821 PMCID: PMC2434381 DOI: 10.1111/j.1365-2567.2007.02738.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Lactobacillus casei ATCC 393 was selected as a bacterial carrier for the development of mucosal vaccine against porcine parvovirus (PPV) infection. The PPV major structural polypeptide VP2 was used as the model parvovirus antigen. Two inducible expression systems, namely pPG611.1 of the cell-surface expression system and pPG612.1 of the secretion expression system based on the xylose operon promoter were used to express the VP2 protein. The immunogenicity of recombinant strains producing VP2 protein in two cellular locations, cell-surface exposed and secreted, was compared to each other by immunizing mice through the intragastric administration. The two types of constructs were able to induce strong specific immune responses against VP2 via intragastric administration and maximum titres of IgA and IgG were attained on days 46 post oral immunization, while the highest antibody levels were obtained with the strain producing the VP2 protein in extracellular milieu. The induced antibodies demonstrated neutralizing effects on PPV infection.
Collapse
Affiliation(s)
- X U Yigang
- Veterinary Department, Northeast Agricultural University, Harbin, 150030, PR China
| | | |
Collapse
|
46
|
Silva AJ, Eko FO, Benitez JA. Exploiting cholera vaccines as a versatile antigen delivery platform. Biotechnol Lett 2007; 30:571-9. [PMID: 18008168 DOI: 10.1007/s10529-007-9594-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2007] [Accepted: 10/29/2007] [Indexed: 02/03/2023]
Abstract
The development of safe, immunogenic and protective cholera vaccine candidates makes possible their use as a versatile antigen delivery platform. Foreign antigens can be delivered to the immune system with cholera vaccines by expressing heterologous antigens in live attenuated vectors, as fusion proteins with cholera toxin subunits combined with inactivated Vibrio cholerae whole cells or by exposing them on the surface of V. cholerae ghosts. Progress in our understanding of the genes expressed by V. cholerae during infection creates unprecedented opportunities to develop an improved generation of vaccine vectors to induce immune protection against a broad range of pathogenic organisms.
Collapse
Affiliation(s)
- Anisia J Silva
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, 720 Westview Dr., SW Atlanta, GA 30310, USA.
| | | | | |
Collapse
|
47
|
Hultberg A, Tremblay DM, de Haard H, Verrips T, Moineau S, Hammarström L, Marcotte H. Lactobacillli expressing llama VHH fragments neutralise Lactococcus phages. BMC Biotechnol 2007; 7:58. [PMID: 17875214 PMCID: PMC2039727 DOI: 10.1186/1472-6750-7-58] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2007] [Accepted: 09/17/2007] [Indexed: 11/29/2022] Open
Abstract
Background Bacteriophages infecting lactic acid bacteria (LAB) are widely acknowledged as the main cause of milk fermentation failures. In this study, we describe the surface-expression as well as the secretion of two functional llama heavy-chain antibody fragments, one binding to the major capsid protein (MCP) and the other to the receptor-binding proteins (RBP) of the lactococcal bacteriophage p2, by lactobacilli in order to neutralise lactococcal phages. Results The antibody fragment VHH5 that is directed against the RBP, was fused to a c-myc tag and expressed in a secreted form by a Lactobacillus strain. The fragment VHH2 that is binding to the MCP, was fused to an E-tag and anchored on the surface of the lactobacilli. Surface expression of VHH2 was confirmed by flow cytometry using an anti-E-tag antibody. Efficient binding of both the VHH2 and the secreted VHH5 fragment to the phage antigens was shown in ELISA. Scanning electron microscopy showed that lactobacilli expressing VHH2 anchored at their surface were able to bind lactococcal phages. A neutralisation assay also confirmed that the secreted VHH5 and the anchored VHH2 fragments prevented the adsorption of lactococcal phages to their host cells. Conclusion Lactobacilli were able to express functional VHH fragments in both a secreted and a cell surface form and reduced phage infection of lactococcal cells. Lactobacilli expressing llama heavy-chain antibody fragments represent a novel way to limit phage infection.
Collapse
Affiliation(s)
- Anna Hultberg
- Division of Clinical Immunology at the Department of Laboratory Medicine, Karolinska Institutet at Karolinska University Hospital in Huddinge, Stockholm, Sweden
- Cellular Architecture and Dynamics (CAD), Utrecht University, The Netherlands
| | - Denise M Tremblay
- Groupe de recherche en écologie buccale, Faculté de médecine dentaire, Félix d'Hérelle Reference Center for Bacterial Viruses, Université Laval, Québec, G1K 7P4, Canada
| | - Hans de Haard
- Unilever Research and Development, Vlaardingen, The Netherlands
- Ablynx, Technologiepark 4, 9052 Ghent, Belgium
| | - Theo Verrips
- Unilever Research and Development, Vlaardingen, The Netherlands
- Cellular Architecture and Dynamics (CAD), Utrecht University, Utrecht, The Netherlands
| | - Sylvain Moineau
- Groupe de recherche en écologie buccale, Faculté de médecine dentaire, Félix d'Hérelle Reference Center for Bacterial Viruses, Université Laval, Québec, G1K 7P4, Canada
- Département de biochimie et de microbiologie, Faculté des sciences et de génie, Université Laval, Québec, G1K 7P4, Canada
| | - Lennart Hammarström
- Division of Clinical Immunology at the Department of Laboratory Medicine, Karolinska Institutet at Karolinska University Hospital in Huddinge, Stockholm, Sweden
| | - Harold Marcotte
- Division of Clinical Immunology at the Department of Laboratory Medicine, Karolinska Institutet at Karolinska University Hospital in Huddinge, Stockholm, Sweden
| |
Collapse
|
48
|
Xu Y, Li Y. Induction of immune responses in mice after intragastric administration of Lactobacillus casei producing porcine parvovirus VP2 protein. Appl Environ Microbiol 2007; 73:7041-7. [PMID: 17827311 PMCID: PMC2074969 DOI: 10.1128/aem.00436-07] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Lactobacillus casei ATCC 393 was selected as an antigen delivery vehicle for mucosal immunization against porcine parvovirus (PPV) infection. A 64-kDa fragment of PPV major protective antigen VP2 protein was used as the parvovirus antigen model. A recombinant Lactobacillus expressing VP2 protein was constructed with plasmid pPG611.1, where expression and localization of the VP2 protein from recombinant Lc393-rPPV-VP2 was detected via sodium dodecyl sulfate-polyacrylamide gel electrophoresis, Western blotting, and immunofluorescence. Both local mucosal and systemic immune responses against PPV were induced in BALB/c mice immunized orally with the recombinant Lactobacillus expressing VP2 protein. The induced antibodies demonstrated neutralizing effects on PPV infection. These data indicated that the use of recombinant lactobacilli could be a valuable strategy for future vaccine development of PPV.
Collapse
Affiliation(s)
- Yigang Xu
- Veterinary Department, Northeast Agricultural University, Harbin 150030, People's Republic of China
| | | |
Collapse
|
49
|
Yao XY, Yuan MM, Li DJ. Molecular adjuvant C3d3 improved the anti-hCGbeta humoral immune response in vaginal inoculation with live recombinant Lactobacillus expressing hCGbeta-C3d3 fusion protein. Vaccine 2007; 25:6129-39. [PMID: 17629363 DOI: 10.1016/j.vaccine.2007.04.090] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2006] [Revised: 04/02/2007] [Accepted: 04/28/2007] [Indexed: 11/25/2022]
Abstract
To enhance the contraceptive efficiency of human chorionic gonadotrophin (hCG)-beta contraceptive vaccine, we coupled hCG-beta gene with molecular adjuvant C3d3, and cloned into live Lactobacilli (Lb.) to express fusion protein hCGbeta-C3d3. The recombinant Lb. could survive in BALB/c murine vagina for at least 3 weeks. After inoculating BALB/c and C57BL/6 mice via vagina, we found that the antibody titer peaks induced by the Lb.hCGbeta-C3d3 inoculation were higher significantly than the Lb.hCGbeta. T and B cells in spleen and vagina were significantly increased, and anti-hCGbeta IgG and IgA antibody-secreting cells in uterus and vagina were significantly increased compared to the control in different strain mice. Our study shows that the C3d3 can display apparent adjuvant efficiency to induce more powerful humoral response to the hCGbeta antigen in vaginal mucosal immunization.
Collapse
Affiliation(s)
- Xiao-Ying Yao
- Laboratory for Reproductive Immunology, Hospital and Institute of Obstetrics & Gynecology, Fudan University Shanghai Medical College, Shanghai 200011, China
| | | | | |
Collapse
|
50
|
Kajikawa A, Satoh E, Leer RJ, Yamamoto S, Igimi S. Intragastric immunization with recombinant Lactobacillus casei expressing flagellar antigen confers antibody-independent protective immunity against Salmonella enterica serovar Enteritidis. Vaccine 2007; 25:3599-605. [PMID: 17287050 PMCID: PMC7115604 DOI: 10.1016/j.vaccine.2007.01.055] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2006] [Revised: 12/11/2006] [Accepted: 01/11/2007] [Indexed: 11/13/2022]
Abstract
A recombinant Lactobacillus casei expressing a flagellar antigen from Salmonella enterica serovar Enteritidis was constructed and evaluated as a mucosal vaccine. Intragastric immunization of the recombinant strain conferred protective immunity against Salmonella infection in mice. This immunization did not result in antigen-specific antibody in either feces or sera but induced the release of IFN-γ on restimulation of primed lymphocytes ex vivo. The results suggested that the protective efficacy provided by flagellin-expressing L. casei is mainly attributable to cell-mediated immune responses. In addition, an adjuvant-type effect of the antigen delivery system with L. casei was also observed.
Collapse
Affiliation(s)
- Akinobu Kajikawa
- Division of Biomedical Food Research, National Institute of Health Sciences, Tokyo, Japan.
| | | | | | | | | |
Collapse
|