1
|
Mollaei Alamuti M, Ravanshad M, Motamedi-Sedeh F, Nabizadeh A, Ahmadi E, Hossieni SM. Immune Response of Gamma-Irradiated Inactivated Bivalent Polio Vaccine Prepared plus Trehalose as a Protein Stabilizer in a Mouse Model. Intervirology 2021; 64:140-146. [PMID: 33853059 DOI: 10.1159/000515392] [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: 06/06/2020] [Accepted: 02/22/2021] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Poliovirus causes paralysis by infecting the nervous system. Currently, 2 types of polio vaccine are given in many countries in polio eradication program including inactivated polio vaccine (IPV) and oral polio vaccine (OPV). Because of OPV-related paralysis, OPV should be replaced by IPV. METHODS The aim of this study was to prepare the gamma-irradiated IPV and determine its effectiveness compared with the commercial vaccine (OPV) in the mouse model. The virus titration of OPV was determined and then inactivated by the appropriate dose of gamma radiation into an irradiated vaccine formula. The vaccine was inoculated in BALB/c mice in 2 different formulations of intramuscular injection with 2-week intervals. The level of anti-polio-neutralizing antibody and polio-specific splenocyte proliferation assay were evaluated by collecting the blood samples and spleens of the vaccinated groups with conventional vaccine and irradiated vaccine. RESULTS There was a significant increase in the neutralizing antibody titration between all of the vaccinated groups and negative control group (A) (p < 0.05). And it shows that the IPV by gamma irradiation has the highest antibody titration. Also, the increasing of stimulation index value in the B* group, F group, and G group was the most against other groups. Furthermore, the neutralizing anti-serum titer and splenic lymphocyte proliferation assay show humoral and cellular immunity were significantly increased in the irradiated vaccine group as compared with conventional group. CONCLUSION According to the results, gamma-irradiated IPV could induce humoral and cellular immunity in vaccinated mouse groups, so the irradiated poliovirus could be recommended as a good candidate vaccine to prevent the transport of poliovirus to the central nervous system and thus protect against paralysis.
Collapse
Affiliation(s)
- Maryam Mollaei Alamuti
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mehrdad Ravanshad
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Farahnaz Motamedi-Sedeh
- Nuclear Science and Technology Research Institute (NSTRI), Nuclear Agriculture Research School, Tehran, Iran
| | - Arezoo Nabizadeh
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Elham Ahmadi
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.,Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | | |
Collapse
|
2
|
Zang Y, Tian Y, Li Y, Xue R, Hu L, Zhang D, Sun S, Wang G, Chen J, Lan Z, Lin S, Jiang S. Recombinant Lactobacillus acidophilus expressing S 1 and S 2 domains of porcine epidemic diarrhea virus could improve the humoral and mucosal immune levels in mice and sows inoculated orally. Vet Microbiol 2020; 248:108827. [PMID: 32891955 PMCID: PMC7428733 DOI: 10.1016/j.vetmic.2020.108827] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 08/11/2020] [Indexed: 01/02/2023]
Abstract
The recombinant L. acidophilus expressing S1 and S2 domains of PEDV were generated. The oral vaccines for PED were based on a swine-origin L. acidophilus. The oral L. acidophilus vaccines induced humoral and mucosal immunity in mice. The L. acidophilus-S1 vaccine induced humoral and mucosal immunity in sows.
Porcine epidemic diarrhea (PED) is a highly contagious intestinal infectious disease caused by porcine epidemic diarrhea virus (PEDV), which is characterized by a high mortality rate in piglets. Since 2012, a remarkable growth in PED outbreaks occurred in many pig farms in China, landing a heavy blow on the pig industry. In order to develop a new effective vaccine for the current PEDV, oral vaccines were generated by transferring eukaryotic expression recombinant plasmids carrying the S1 and S2 (antigenic sites of the S protein) epitopes of PEDV into a swine-origin Lactobacillus acidophilus (L. acidophilus). After oral immunization of the BALB/c mice, higher levels of anti-PEDV specific IgG and SIgA antibodies and cellular immune responses were detected in mice orally administered with the recombinant L. acidophilus-S1 compared to the L. acidophilus-S2. Furthermore, L. acidophilus-S1 was used to inoculate the pregnant sows orally and the results showed that the recombinant L. acidophilus-S1 could elicit a specific systemic and mucosal immune response. In summary, our study demonstrated that oral immunization with L. acidophilus-S1 could improve the humoral and mucosal immune levels in sows and would be a promising candidate vaccine against PEDV infection in piglets.
Collapse
Affiliation(s)
- Yue Zang
- Shandong Provincial Center for Animal Disease Control and Prevention, Shandong, Jinan, 251000, China; College of Veterinary Medicine, Shandong Agricultural University, Shandong, Taian, 271018, China
| | - Ye Tian
- College of Veterinary Medicine, China Agricultural University, Beijing, 100094, China
| | - Yungang Li
- Shandong Provincial Center for Animal Disease Control and Prevention, Shandong, Jinan, 251000, China
| | - Ruixue Xue
- Shandong Provincial Center for Animal Disease Control and Prevention, Shandong, Jinan, 251000, China
| | - Liping Hu
- Shandong Provincial Center for Animal Disease Control and Prevention, Shandong, Jinan, 251000, China
| | - Dong Zhang
- Shandong Provincial Center for Animal Disease Control and Prevention, Shandong, Jinan, 251000, China
| | - Shengfu Sun
- Shandong Provincial Center for Animal Disease Control and Prevention, Shandong, Jinan, 251000, China
| | - Guisheng Wang
- Shandong Provincial Center for Animal Disease Control and Prevention, Shandong, Jinan, 251000, China
| | - Jing Chen
- Shandong Provincial Center for Animal Disease Control and Prevention, Shandong, Jinan, 251000, China
| | - Zouran Lan
- Shandong Provincial Center for Animal Disease Control and Prevention, Shandong, Jinan, 251000, China
| | - Shaoli Lin
- College of Veterinary Medicine, Shandong Agricultural University, Shandong, Taian, 271018, China
| | - Shijin Jiang
- College of Veterinary Medicine, Shandong Agricultural University, Shandong, Taian, 271018, China.
| |
Collapse
|
3
|
Sha Z, Shang H, Miao Y, Huang J, Niu X, Chen R, Hu L, Huang H, Wei K, Zhu R. Recombinant Lactococcus Lactis Expressing M1-HA2 Fusion Protein Provides Protective Mucosal Immunity Against H9N2 Avian Influenza Virus in Chickens. Front Vet Sci 2020; 7:153. [PMID: 32266297 PMCID: PMC7105734 DOI: 10.3389/fvets.2020.00153] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 03/03/2020] [Indexed: 01/16/2023] Open
Abstract
H9N2 subtype low pathogenicity avian influenza virus (LPAIV) is distributed worldwide and causes enormous economic losses in the poultry industry. Despite immunization of almost all chickens with inactivated vaccines, the disease still remains widespread. We speculated that improving mucosal or cellular immune responses could contribute to improved control of H9N2 viruses. In this study, we constructed a novel Lactococcus lactis (L. lactis) strain expressing a recombinant fusion protein consisting of the M1 and HA2 proteins derived from an antigenically conserved endemic H9N2 virus strain. The M1-HA2 fusion protein was cloned downstream of a gene encoding a secretory peptide, and we subsequently confirmed that the fusion protein was secreted from L. lactis by Western blotting. We assessed the immunogenicity and protective effects of this recombinant L. lactis strain. Eighty 1-day-old chickens were divided into four groups, and the experimental groups were orally vaccinated twice with the recombinant L. lactis strain. Fecal and intestinal samples, sera, and bronchoalveolar lavage fluid were collected at 7, 14, and 21 days post-vaccination (dpv). Chickens vaccinated with the recombinant L. lactis strain showed significantly increased levels of serum antibodies, T cell-mediated immune responses, and mucosal secretory IgA (SIgA). Following challenge with H9N2 virus at 21 dpv, chickens vaccinated with the recombinant L. lactis strain showed decreased weight loss, lower viral titers in the lung, and reduced lung pathological damage. In summary, our results demonstrated that a recombinant L. lactis strain expressing an H9N2 M1-HA2 fusion protein could induce protective mucosal and systemic immunity. This oral vaccine is H9N2 virus-specific and represents a significant design improvement compared with previous studies. Our study provides a theoretical basis for improving mucosal immune responses to prevent and control H9N2 virus infection.
Collapse
Affiliation(s)
- Zhou Sha
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an, China.,Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Tai'an, China
| | - Hongqi Shang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an, China.,Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Tai'an, China
| | - Yongqiang Miao
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an, China.,Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Tai'an, China
| | - Jin Huang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an, China.,Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Tai'an, China
| | - Xiangyun Niu
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an, China.,Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Tai'an, China
| | - Ruichang Chen
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an, China.,Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Tai'an, China
| | - Liping Hu
- Animal Disease Prevention and Control Center of Shandong Province, Animal Husbandry and Veterinary Bureau of Shandong Province, Jinan, China
| | - He Huang
- Shandong New Hope Liuhe Co., Ltd, New Hope Group, Qingdao, China
| | - Kai Wei
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an, China.,Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Tai'an, China
| | - Ruiliang Zhu
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an, China.,Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Tai'an, China
| |
Collapse
|
4
|
Foot-and-mouth disease vaccines: recent updates and future perspectives. Arch Virol 2019; 164:1501-1513. [PMID: 30888563 DOI: 10.1007/s00705-019-04216-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 03/16/2019] [Indexed: 02/07/2023]
Abstract
Foot-and-mouth disease (FMD) is a major worldwide viral disease in animals, affecting the national and international trade of livestock and animal products and leading to high economic losses and social consequences. Effective control measures of FMD involve prevention through vaccination with inactivated vaccines. These inactivated vaccines, unfortunately, require short-term protection and cold-chain and high-containment facilities. Major advances and pursuit of hot topics in vaccinology and vectorology are ongoing, involving peptide vaccines, DNA vaccines, live vector vaccines, and novel attenuated vaccines. DIVA capability and marker vaccines are very important in differentiating infected animals from vaccinated animals. This review focuses on updating the research progress of these novel vaccines, summarizing their merits and including ideas for improvement.
Collapse
|
5
|
Coelho-Rocha ND, de Castro CP, de Jesus LCL, Leclercq SY, de Cicco Sandes SH, Nunes AC, Azevedo V, Drumond MM, Mancha-Agresti P. Microencapsulation of Lactic Acid Bacteria Improves the Gastrointestinal Delivery and in situ Expression of Recombinant Fluorescent Protein. Front Microbiol 2018; 9:2398. [PMID: 30344518 PMCID: PMC6182071 DOI: 10.3389/fmicb.2018.02398] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 09/19/2018] [Indexed: 01/25/2023] Open
Abstract
The microencapsulation process of bacteria has been used for many years, mainly in the food industry and, among the different matrixes used, sodium alginate stands out. This matrix forms a protective wall around the encapsulated bacterial culture, increasing its viability and protecting against environmental adversities, such as low pH, for example. The aim of the present study was to evaluate both in vitro and in vivo, the capacity of the encapsulation process to maintain viable lactic acid bacteria (LAB) strains for a longer period of time and to verify if they are able to reach further regions of mouse intestine. For this purpose, a recombinant strain of LAB (L. lactis ssp. cremoris MG1363) carrying the pExu vector encoding the fluorescence protein mCherry [L. lactis MG1363 (pExu:mCherry)] was constructed. The pExu was designed by our group and acts as a vector for DNA vaccines, enabling the host cell to produce the protein of interest. The functionality of the pExu:mCherry vector, was demonstrated in vitro by fluorescence microscopy and flow cytometry after transfection of eukaryotic cells. After this confirmation, the recombinant strain was submitted to encapsulation protocol with sodium alginate (1%). Non-encapsulated, as well as encapsulated strains were orally administered to C57BL/6 mice and the expression of mCherry protein was evaluated at different times (0-168 h) in different bowel portions. Confocal microscopy showed that the expression of mCherry was higher in animals who received the encapsulated strain in all portions of intestine analyzed. These results were confirmed by qRT-PCR assay. Therefore, this is the first study comparing encapsulated and non-encapsulated L. lactis bacteria for mucosal DNA delivery applications. Our results showed that the microencapsulation process is an effective method to improve DNA delivery, ensuring a greater number of viable bacteria are able to reach different sections of the bowel.
Collapse
Affiliation(s)
- Nina D Coelho-Rocha
- Laboratório de Genética Celular e Molecular, Instituto de Ciências Biológicas, Departamento de Biologia Geral, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Camila P de Castro
- Laboratório de Genética Celular e Molecular, Instituto de Ciências Biológicas, Departamento de Biologia Geral, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Kroton Educacional, Faculdade Pitágoras, Contagem, Brazil
| | - Luis C L de Jesus
- Laboratório de Genética Celular e Molecular, Instituto de Ciências Biológicas, Departamento de Biologia Geral, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Sophie Y Leclercq
- Laboratório de Inovação Biotecnológica, Fundação Ezequiel Dias, Belo Horizonte, Brazil
| | - Savio H de Cicco Sandes
- Laboratório de Genética Molecular de Protozoários Parasitas, Instituto de Ciências Biológicas, Departamento de Biologia Geral, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Alvaro C Nunes
- Laboratório de Genética Molecular de Protozoários Parasitas, Instituto de Ciências Biológicas, Departamento de Biologia Geral, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Vasco Azevedo
- Laboratório de Genética Celular e Molecular, Instituto de Ciências Biológicas, Departamento de Biologia Geral, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mariana M Drumond
- Laboratório de Genética Celular e Molecular, Instituto de Ciências Biológicas, Departamento de Biologia Geral, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Centro Federal de Educação Tecnológica de Minas Gerais, Coordenação de Ciências, Belo Horizonte, Brazil
| | - Pamela Mancha-Agresti
- Laboratório de Genética Celular e Molecular, Instituto de Ciências Biológicas, Departamento de Biologia Geral, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
6
|
Zhang L, Yang W, Hu C, Wang Q, Wu Y. Properties and applications of nanoparticle/microparticle conveyors with adjuvant characteristics suitable for oral vaccination. Int J Nanomedicine 2018; 13:2973-2987. [PMID: 29861631 PMCID: PMC5968786 DOI: 10.2147/ijn.s154743] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Vaccination is one of the most effective approaches in the prevention and control of disease worldwide. Oral vaccination could have wide applications if effective protection cannot be achieved through traditional (eg, parenteral) routes of vaccination. However, oral administration is hampered by the difficulties in transferring vaccines in vivo. This has led to the development of materials such as carriers with potential adjuvant effects. Considering the requirements for selecting adjuvants for oral vaccines as well as the advantages of nanoparticle/microparticle materials as immune effectors and antigen conveyors, synthetic materials could improve the efficiency of oral vaccination. In this review, nanoparticles and microparticles with adjuvant characteristics are described with regard to their potential importance for oral immunization, and some promising and successful modification strategies are summarized.
Collapse
Affiliation(s)
- Lei Zhang
- College of Life Sciences, Fujian Normal University, Fuzhou, China.,State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, China
| | - Wendi Yang
- College of Life Sciences, Fujian Normal University, Fuzhou, China
| | - Chaohua Hu
- National Engineering Research Center for Sugarcane, College of Crop Science, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Qianchao Wang
- College of Life Sciences, Fujian Normal University, Fuzhou, China
| | - Yunkun Wu
- College of Life Sciences, Fujian Normal University, Fuzhou, China.,State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, China
| |
Collapse
|
7
|
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
|
8
|
Bhuyan AA, Memon AM, Bhuiyan AA, Zhonghua L, Zhang B, Ye S, Mengying L, He QG. The construction of recombinant Lactobacillus casei expressing BVDV E2 protein and its immune response in mice. J Biotechnol 2018; 270:51-60. [PMID: 29408654 DOI: 10.1016/j.jbiotec.2018.01.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 01/24/2018] [Accepted: 01/24/2018] [Indexed: 10/18/2022]
Abstract
Bovine viral diarrhea virus (BVDV) is the etiological agent of BVD causes substantial economic losses and endemic in world-wide cattle population. Mucosal immunity plays an important role in protection against BVDV infection and Lactobacillus casei is believed as an excellent live vaccine vector for expressing foreign genes. In this study, we have constructed a novel recombinant L. casei/pELX1-E2 strain expressing the most immunogenic E2 antigen of BVDV; using growth phage dependent surface expression system pELX1. The expression of E2 protein was verified by SDS-PAGE, Western blotting, and Immunofluorescence microscopic analysis. The immune responses triggered by the E2 producing recombinant L. casei were evaluated in BALB/c mice revealed that oral and intranasal (IN) administration of the recombinant strain was able to induce a significantly higher level of specific anti-E2 mucosal IgA and serum IgG as well as the greater level of cellular response by IFN-γ and IL-12 than those of intramuscular (IM) and control groups of mice. However, IN inoculation was found the most potent route of immunization. The ability of the recombinant strain to induce serum neutralizing antibody against BVDV and reduced viral load after viral challenge indicated better protection of BVDV infection. Therefore, this recombinant L. casei expressing E2 could be a safe and promising mucosal vaccine candidate against BVD.
Collapse
Affiliation(s)
- Anjuman Ara Bhuyan
- State Key Laboratory of Agricultural Microbiology, Ministry of Education, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; National Institute of Biotechnology, Savar, Dhaka, 1349, Bangladesh.
| | - Atta Muhammad Memon
- State Key Laboratory of Agricultural Microbiology, Ministry of Education, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Ali Akbar Bhuiyan
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Bangladesh Livestock Research Institute, Savar, Dhaka, 1341, Bangladesh.
| | - Li Zhonghua
- State Key Laboratory of Agricultural Microbiology, Ministry of Education, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Bingzhou Zhang
- State Key Laboratory of Agricultural Microbiology, Ministry of Education, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Shiyi Ye
- State Key Laboratory of Agricultural Microbiology, Ministry of Education, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Li Mengying
- State Key Laboratory of Agricultural Microbiology, Ministry of Education, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Qi-Gai He
- State Key Laboratory of Agricultural Microbiology, Ministry of Education, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
9
|
|
10
|
Lactic acid bacteria as mucosal delivery vehicles: a realistic therapeutic option. Appl Microbiol Biotechnol 2016; 100:5691-701. [DOI: 10.1007/s00253-016-7557-x] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 04/12/2016] [Accepted: 04/14/2016] [Indexed: 12/11/2022]
|
11
|
Motamedi-Sedeh F, Soleimanjahi H, Jalilian AR, Mahravani H, Shafaee K, Sotoodeh M, Taherkarami H, Jairani F. Development of Protective Immunity against Inactivated Iranian Isolate of Foot-and-Mouth Disease Virus Type O/IRN/2007 Using Gamma Ray-Irradiated Vaccine on BALB/c Mice and Guinea Pigs. Intervirology 2015. [PMID: 26202581 DOI: 10.1159/000433538] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVES Foot-and-mouth disease virus (FMDV) causes a highly contagious disease in cloven-hoofed animals and is the most damaging disease of livestock worldwide, leading to great economic losses. The aim of this research was the inactivation of FMDV type O/IRN/1/2007 to produce a gamma ray-irradiated (GRI) vaccine in order to immunize mice and guinea pigs. METHODS In this research, the Iranian isolated FMDV type O/IRN/1/2007 was irradiated by gamma ray to prepare an inactivated whole virus antigen and formulated as a GRI vaccine with unaltered antigenic characteristics. Immune responses against this vaccine were evaluated on mice and guinea pigs. RESULTS The comparison of the immune responses between the GRI vaccine and conventional vaccine did not show any significant difference in neutralizing antibody titer, memory spleen T lymphocytes or IFN-γ, IL-4, IL-2 and IL-10 concentrations (p > 0.05). In contrast, there were significant differences in all of the evaluated immune factors between the two vaccinated groups of mice and negative control mice (p < 0.05). The protective dose 50 for the conventional and GRI vaccines obtained were 6.28 and 7.07, respectively, which indicated the high potency of both vaccines. CONCLUSION GRI vaccine is suitable for both routine vaccination and control of FMDV in emergency outbreaks.
Collapse
|
12
|
Michon C, Christophe M, Kuczkowska K, Langella P, Eijsink VGH, Mathiesen G, Chatel JM. Surface display of an anti-DEC-205 single chain Fv fragment in Lactobacillus plantarum increases internalization and plasmid transfer to dendritic cells in vitro and in vivo. Microb Cell Fact 2015; 14:95. [PMID: 26141059 PMCID: PMC4491208 DOI: 10.1186/s12934-015-0290-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 06/24/2015] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Lactic acid bacteria (LAB) are promising vehicles for delivery of a variety of medicinal compounds, including antigens and cytokines. It has also been established that LAB are able to deliver cDNA to host cells. To increase the efficiency of LAB-driven DNA delivery we have constructed Lactobacillus plantarum strains targeting DEC-205, which is a receptor located at the surface of dendritic cells (DCs). The purpose was to increase uptake of bacterial cells, which could lead to improved cDNA delivery to immune cells. RESULTS Anti-DEC-205 antibody (aDec) was displayed at the surface of L. plantarum using three different anchoring strategies: (1) covalent anchoring of aDec to the cell membrane (Lipobox domain, Lip); (2) covalent anchoring to the cell wall (LPXTG domain, CWA); (3) non-covalent anchoring to the cell wall (LysM domain, LysM). aDec was successfully expressed in all three strains, but surface location of the antibody could only be demonstrated for the two strains with cell wall anchors (CWA and LysM). Co-incubation of the engineered strains and DCs showed increased uptake when anchoring aDec using the CWA or LysM anchors. In a competition assay, free anti-DEC abolished the increased uptake, showing that the internalization is due to specific interactions between the DEC-205 receptor and aDec. To test plasmid transfer, a plasmid for expression of GFP under control of an eukaryotic promoter was transformed into the aDec expressing strains and GFP expression in DCs was indeed increased when using the strains producing cell-wall anchored aDec. Plasmid transfer to DCs in the gastro intestinal tract was also detected using a mouse model. Surprisingly, in mice the highest expression of GFP was observed for the strain in which aDec was coupled to the cell membrane. CONCLUSION The results show that surface expression of aDec leads to increased internalization of L. plantarum and plasmid transfer in DCs and that efficiency depends on the type of anchor used. Interestingly, in vitro data indicates that cell wall anchoring is more effective, whereas in vivo data seem to indicate that anchoring to the cell membrane is preferable. It is likely that the more embedded localization of aDec in the latter case is favorable when cells are exposed to the harsh conditions of the gastro-intestinal tract.
Collapse
Affiliation(s)
| | - Michon Christophe
- INRA, UMR1319 MICALIS, Bat 440, R-2, 78352, Jouy-en-Josas, France. .,AgroParisTech, UMR MICALIS, 78352, Jouy-en-Josas, France.
| | - Katarzyna Kuczkowska
- Department of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Aas, Norway.
| | - Philippe Langella
- INRA, UMR1319 MICALIS, Bat 440, R-2, 78352, Jouy-en-Josas, France. .,AgroParisTech, UMR MICALIS, 78352, Jouy-en-Josas, France.
| | - Vincent G H Eijsink
- Department of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Aas, Norway.
| | - Geir Mathiesen
- Department of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Aas, Norway.
| | - Jean-Marc Chatel
- INRA, UMR1319 MICALIS, Bat 440, R-2, 78352, Jouy-en-Josas, France. .,AgroParisTech, UMR MICALIS, 78352, Jouy-en-Josas, France.
| |
Collapse
|
13
|
|
14
|
Abstract
This chapter reviews papers mostly written since 2005 that report results using live attenuated bacterial vectors to deliver after administration through mucosal surfaces, protective antigens, and DNA vaccines, encoding protective antigens to induce immune responses and/or protective immunity to pathogens that colonize on or invade through mucosal surfaces. Papers that report use of such vaccine vector systems for parenteral vaccination or to deal with nonmucosal pathogens or do not address induction of mucosal antibody and/or cellular immune responses are not reviewed.
Collapse
|
15
|
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
|
16
|
Bermúdez-Humarán LG, Aubry C, Motta JP, Deraison C, Steidler L, Vergnolle N, Chatel JM, Langella P. Engineering lactococci and lactobacilli for human health. Curr Opin Microbiol 2013; 16:278-83. [PMID: 23850097 DOI: 10.1016/j.mib.2013.06.002] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 06/11/2013] [Accepted: 06/11/2013] [Indexed: 02/06/2023]
Abstract
Food-grade lactic acid bacteria (LAB) are good candidates for the development of oral vectors, and are attractive alternatives to attenuated pathogens, for mucosal delivery strategies. In this review, we summarize recent results on the use of LAB as mucosal delivery vectors for therapeutic proteins and DNA vaccines. Most of this work has been based on the model LAB, Lactococcus lactis, which is suitable for the heterologous expression of therapeutic proteins. Recombinant lactococci and lactobacilli strains expressing antiproteases and antioxidant enzymes have been tested successfully for their prophylactic and therapeutic effects in murine models of colitis. Recombinant lactococci secreting autoantigens have been found to be effective for the treatment of type 1 diabetes. Also, recombinant lactococci delivering DNA were able to prevent a bovine β-lactoglobulin (BLG)-allergic reaction in mice. We believe that these various coherent findings demonstrate the potential value of using LAB, particularly lactococci and lactobacilli strains, to develop novel vectors for the therapeutic delivery of proteins to mucosal surfaces. Further tests and in particular human clinical trials are now important next steps to conclude on the benefit of these approaches for human health.
Collapse
Affiliation(s)
- Luis G Bermúdez-Humarán
- INRA, Commensal and Probiotics-Host Interactions Laboratory, UMR 1319 Micalis, F-78350 Jouy-en-Josas, France
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Fowler VL, Barnett PV. Progress in the development of DNA vaccines against foot-and-mouth disease. Expert Rev Vaccines 2012; 11:481-93. [PMID: 22551033 DOI: 10.1586/erv.11.198] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
DNA vaccines are, in principle, the simplest yet most versatile methods of inducing protective humoral and cellular immune responses. Research involving this type of vaccine against veterinary diseases began in the early 1990s and has since seen the evaluation of more than 30 important viral pathogens, including the economically important foot-and-mouth disease. With the demonstration that DNA vaccines protect against foot-and-mouth disease in sheep and pigs, and the advantages these DNA vaccines have over the conventional formulations, this approach may provide a better solution to the control of this disease. In this review, we provide a comprehensive overview of DNA vaccination strategies for foot-and-mouth disease reported in the literature, in which we highlight the studies that have reported protection in the key target species.
Collapse
Affiliation(s)
- Veronica L Fowler
- Institute for Animal Health, Pirbright Laboratory, Surrey GU24 0NF, UK.
| | | |
Collapse
|
18
|
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]
|
19
|
Wells J. Mucosal vaccination and therapy with genetically modified lactic acid bacteria. Annu Rev Food Sci Technol 2012; 2:423-45. [PMID: 22129390 DOI: 10.1146/annurev-food-022510-133640] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Lactic acid bacteria (LAB) have proved to be effective mucosal delivery vehicles that overcome the problem of delivering functional proteins to the mucosal tissues. By the intranasal route, both live and killed LAB vaccine strains have been shown to elicit mucosal and systemic immune responses that afford protection against infectious challenges. To be effective via oral administration, frequent dosing over several weeks is required but new targeting and adjuvant strategies have clearly demonstrated the potential to increase the immunogenicity and protective immunity of LAB vaccines. Oral administration of Lactococcus lactis has been shown to induce antigen-specific oral tolerance (OT) to secreted recombinant antigens. LAB delivery is more efficient at inducing OT than the purified antigen, thus avoiding the need for purification of large quantities of antigen. This approach holds promise for new therapeutic interventions in allergies and antigen-induced autoimmune diseases. Several clinical and research reports demonstrate considerable progress in the application of genetically modified L. lactis for the treatment of inflammatory bowel disease (IBD). New medical targets are on the horizon, and the approval by several health authorities and biosafety committees of a containment system for a genetically modified L. lactis that secretes Il-10 should pave the way for new LAB delivery applications in the future.
Collapse
Affiliation(s)
- Jerry Wells
- Host-Microbe-Interactomics, University of Wageningen, Animal Sciences Department, 6700 AH, Wageningen, The Netherlands.
| |
Collapse
|
20
|
Vesicular Stomatitis Virus glycoprotein G carrying a tandem dimer of Foot and Mouth Disease Virus antigenic site A can be used as DNA and peptide vaccine for cattle. Antiviral Res 2011; 92:219-27. [DOI: 10.1016/j.antiviral.2011.08.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Revised: 07/27/2011] [Accepted: 08/09/2011] [Indexed: 11/16/2022]
|
21
|
Rodriguez LL, Gay CG. Development of vaccines toward the global control and eradication of foot-and-mouth disease. Expert Rev Vaccines 2011; 10:377-87. [PMID: 21434805 DOI: 10.1586/erv.11.4] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Foot-and-mouth disease (FMD) is one of the most economically and socially devastating diseases affecting animal agriculture throughout the world. Although mortality is usually low in adult animals, millions of animals have been killed in efforts to rapidly control and eradicate FMD. The causing virus, FMD virus (FMDV), is a highly variable RNA virus occurring in seven serotypes (A, O, C, Asia 1, Sat 1, Sat 2 and Sat 3) and a large number of subtypes. FMDV is one of the most infectious agents known, affecting cloven-hoofed animals with significant variations in infectivity and virus transmission. Although inactivated FMD vaccines have been available for decades, there is little or no cross-protection across serotypes and subtypes, requiring vaccines that are matched to circulating field strains. Current inactivated vaccines require growth of virulent virus, posing a threat of escape from manufacturing sites, have limited shelf life and require re-vaccination every 4-12 months. These vaccines have aided in the eradication of FMD from Europe and the control of clinical disease in many parts of the world, albeit at a very high cost. However, FMDV persists in endemic regions impacting millions of people dependent on livestock for food and their livelihood. Usually associated with developing countries that lack the resources to control it, FMD is a global problem and the World Organization for Animal Health and the United Nations' Food Agriculture Organization have called for its global control and eradication. One of the main limitations to FMDV eradication is the lack of vaccines designed for this purpose, vaccines that not only protect against clinical signs but that can actually prevent infection and effectively interrupt the natural transmission cycle. These vaccines should be safely and inexpensively produced, be easy to deliver, and also be capable of inducing lifelong immunity against multiple serotypes and subtypes. Furthermore, there is a need for better integrated strategies that fit the specific needs of endemic regions. Availability of these critical components will greatly enhance the chances for the global control and eradication of FMDV.
Collapse
Affiliation(s)
- Luis L Rodriguez
- Agricultural Research Service, United States Department of Agriculture, Foreign Animal Disease Research Unit, Orient Point, New York, NY, USA.
| | | |
Collapse
|
22
|
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
|
23
|
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
|
24
|
Dory D, Rémond M, Béven V, Cariolet R, Backovic M, Zientara S, Jestin A. Pseudorabies virus glycoprotein B can be used to carry foot and mouth disease antigens in DNA vaccination of pigs. Antiviral Res 2009; 81:217-25. [DOI: 10.1016/j.antiviral.2008.11.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2008] [Revised: 11/17/2008] [Accepted: 11/24/2008] [Indexed: 02/03/2023]
|
25
|
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
|
26
|
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
|
27
|
Abstract
Important developments in the design of recombinant lactic acid bacteria (LAB) as mucosal carriers for a range of health-beneficial compounds, such as antigens, allergens, immune modulators, antimicrobial and trefoil peptides, single-chain antibodies and a few enzymes, have taken place in the past decade. The different approaches, strategies and proof-of-concept studies that have been conducted in animal models are reviewed in this article. The rationale for the use of lactic acid bacteria as mucosal delivery vehicles and key aspects of their interaction with the host mucosal surfaces are discussed. An overview of the progress in the field of LAB-based mucosal vaccines and a discussion of protection studies that have been conducted in rodents, mainly by intranasal and intragastric immunization, are provided. The latest developments in the use of LAB as vechicles for DNA vaccination are described. Studies that deal with successful delivery of cytokines or trefoil peptides to treat experimental colitis in rodents are reviewed. Notably, the first Phase I trial has been conducted with patients that suffer from inflammatory bowel disease using safe biologically contained recombinant lactococci that secrete human interleukin-10. Efforts to induce oral tolerance and develop preventive strategies against type I allergies using LAB are highlighted. Anti-infective strategies that are based on the delivery of microbicidal peptides are discussed, with a special emphasis on the prevention of HIV-1 infection. The concluding section captures the key learning points in the field, identifies major questions that remain to be answered and highlights challenges for the future.
The development of lactic acid bacteria as delivery vehicles for therapeutics, anti-infectives and vaccines at mucosa is discussed in this Review. Engineered LAB could be deployed to treat conditions such as allergy and inflammatory bowel disease, and might also be adopted in the fight against pathogens, including HIV-1 infection. Studies of lactic acid bacteria (LAB) as delivery vehicles have focused mainly on the development of mucosal vaccines, with much effort being devoted to the generation of genetic tools for antigen expression in different bacterial locations. Subsequently, interleukins have been co-expressed with antigens in LAB to enhance the immune response that is raised against the antigen. LAB have also been used as a delivery system for a range of molecules that have different applications, including anti-infectives, therapies for allergic diseases and therapies for gastrointestinal diseases. Now that the first human trial with a Lactococcus strain that expresses recombinant interleukin-10 has been completed, we discuss what we have learnt, what we do not yet understand and what the future holds for therapy and prophylaxis with LAB.
Collapse
|