1
|
Zhuang L, Ye Z, Li L, Yang L, Gong W. Next-Generation TB Vaccines: Progress, Challenges, and Prospects. Vaccines (Basel) 2023; 11:1304. [PMID: 37631874 PMCID: PMC10457792 DOI: 10.3390/vaccines11081304] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 07/28/2023] [Accepted: 07/28/2023] [Indexed: 08/27/2023] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (MTB), is a prevalent global infectious disease and a leading cause of mortality worldwide. Currently, the only available vaccine for TB prevention is Bacillus Calmette-Guérin (BCG). However, BCG demonstrates limited efficacy, particularly in adults. Efforts to develop effective TB vaccines have been ongoing for nearly a century. In this review, we have examined the current obstacles in TB vaccine research and emphasized the significance of understanding the interaction mechanism between MTB and hosts in order to provide new avenues for research and establish a solid foundation for the development of novel vaccines. We have also assessed various TB vaccine candidates, including inactivated vaccines, attenuated live vaccines, subunit vaccines, viral vector vaccines, DNA vaccines, and the emerging mRNA vaccines as well as virus-like particle (VLP)-based vaccines, which are currently in preclinical stages or clinical trials. Furthermore, we have discussed the challenges and opportunities associated with developing different types of TB vaccines and outlined future directions for TB vaccine research, aiming to expedite the development of effective vaccines. This comprehensive review offers a summary of the progress made in the field of novel TB vaccines.
Collapse
Affiliation(s)
- Li Zhuang
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, Eighth Medical Center of Chinese PLA General Hospital, Beijing 100091, China
- Hebei North University, Zhangjiakou 075000, China
| | - Zhaoyang Ye
- Hebei North University, Zhangjiakou 075000, China
| | - Linsheng Li
- Hebei North University, Zhangjiakou 075000, China
| | - Ling Yang
- Hebei North University, Zhangjiakou 075000, China
| | - Wenping Gong
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, Eighth Medical Center of Chinese PLA General Hospital, Beijing 100091, China
| |
Collapse
|
2
|
da Silva ASM, Albuquerque LHP, de Ponte CGG, de Almeida MR, de Faria SER, Ribeiro MDS, Pereira ENGDS, Antas PRZ. Time to face the proofs: the BCG Moreau vaccine promotes superior inflammatory cytokine profile in vitro when compared with Russia, Pasteur, and Danish strains. Hum Vaccin Immunother 2021; 18:1989913. [PMID: 34766868 PMCID: PMC8942427 DOI: 10.1080/21645515.2021.1989913] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Tuberculosis (TB) has been a major public health problem worldwide, and the Bacillus Calmette–Guérin (BCG) vaccine is the only available vaccine against this disease. The BCG vaccine is no longer a single organism; it consists of diverse strains. The early-shared strains of the BCG vaccine are stronger immunostimulators than the late-shared strains. In this study, we have employed a simple in vitro human model to broadly evaluate the differences among four widely used BCG vaccines during the characterization of strain-specific host immune responses. In general, the BCG Moreau vaccine generated a higher inflammatory cytokine profile and lower TGF-β levels compared with the Russia, Pasteur, and Danish strains in the context of early sensitization with TB; however, no changes were observed in the IL-23 levels between infected and noninfected cultures. Unsurprisingly, the BCG vaccines provided different features, and the variances among those strains may influence the activation of infected host cells, which ultimately leads to distinct protective efficacy to tackle TB.
Collapse
Affiliation(s)
- Andreon Santos Machado da Silva
- Laboratório de Imunologia Clínica, Instituto Oswaldo Cruz, Rio de Janeiro, andInstituto Nacional de Ciência e Tecnologia em Tuberculose (INCT-TB)
| | - Lawrence Henrique Paz Albuquerque
- Laboratório de Imunologia Clínica, Instituto Oswaldo Cruz, Rio de Janeiro, andInstituto Nacional de Ciência e Tecnologia em Tuberculose (INCT-TB)
| | - Carlos Germano Garrido de Ponte
- Laboratório de Imunologia Clínica, Instituto Oswaldo Cruz, Rio de Janeiro, andInstituto Nacional de Ciência e Tecnologia em Tuberculose (INCT-TB)
| | - Matheus Rogério de Almeida
- Laboratório de Imunologia Clínica, Instituto Oswaldo Cruz, Rio de Janeiro, andInstituto Nacional de Ciência e Tecnologia em Tuberculose (INCT-TB)
| | - Sandra Elizabete Ribeiro de Faria
- Laboratório de Imunologia Clínica, Instituto Oswaldo Cruz, Rio de Janeiro, andInstituto Nacional de Ciência e Tecnologia em Tuberculose (INCT-TB)
| | | | | | - Paulo Renato Zuquim Antas
- Laboratório de Imunologia Clínica, Instituto Oswaldo Cruz, Rio de Janeiro, andInstituto Nacional de Ciência e Tecnologia em Tuberculose (INCT-TB)
| |
Collapse
|
3
|
Dorneles J, Madruga AB, Seixas Neto ACP, Rizzi C, Bettin ÉB, Hecktheuer AS, Castro CCD, Fernandes CG, Oliveira TL, Dellagostin OA. Protection against leptospirosis conferred by Mycobacterium bovis BCG expressing antigens from Leptospira interrogans. Vaccine 2020; 38:8136-8144. [PMID: 33176938 DOI: 10.1016/j.vaccine.2020.10.086] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/22/2020] [Accepted: 10/27/2020] [Indexed: 12/29/2022]
Abstract
Leptospirosis is a zoonotic disease worldwide and caused by the pathogenic spirochetes of the genus Leptospira. Bacterins make up the vaccines used against leptospirosis, but they only succeed in providing short-term and serovar-specific protection. The use of Mycobacterium bovis BCG as a live vaccine vector expressing leptospiral antigens is a promising alternative, particularly due to its adjuvant properties. Four distinct portions P1 (lipL32), P2 (ligAni), P3 (lemA:ligAni) and P4 (lipL32:lemA) of a recombinant chimera composed of the lipL32, lemA and ligANI genes from Leptospira interrogans were cloned individually according to the BioBricks® strategy in the plasmid pUP500/PpAN. These constructs were individually transformed into a BCG Pasteur strain, and protein expression was detected by Western blot. For vaccination, 5 groups of 10 Golden Syrian hamsters were used, aged 4-6 weeks - group 1, rBCG (LipL32); group 2, rBCG (LigAni); group 3, rBCG (LemA:LigAni); group 4, (LipL32:LemA); group 5, wild-type BCG Pasteur (negative control). Two doses containing 106 CFU of rBCG were administered subcutaneously, the challenge was performed with 5 × LD50 of Leptospira interrogans serovar Copenhageni L1-130, and the animals were observed for a 30-day period until the endpoint was reached. Humoral immunity was assessed via indirect ELISA, while renal colonisation was assessed by culture and quantitative real-time PCR. All vaccinated groups were protected against lethal challenge and renal colonisation, in comparison with negative control group (P < 0.05). Recombinant vaccines were not effective at inducing significant humoral immunity, which suggests the induction of cellular immunity - a characteristic of M. bovis BCG. In conclusion, all formulations provide 100% significant protection against leptospirosis in hamsters with no renal colonisation. The use of rBCG as a vaccine vector represents a promising alternative for the control of animal leptospirosis, allowing for protection against clinical signs of leptospirosis and renal colonisation.
Collapse
Affiliation(s)
- Jessica Dorneles
- Programa de Pós-Graduação em Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Andriele Bonemann Madruga
- Programa de Pós-Graduação em Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Amilton Clair Pinto Seixas Neto
- Programa de Pós-Graduação em Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Caroline Rizzi
- Programa de Pós-Graduação em Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Éverton Burlamarque Bettin
- Programa de Pós-Graduação em Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Amanda Silva Hecktheuer
- Programa de Pós-Graduação em Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Clarissa Caetano de Castro
- Programa de Pós-Graduação em Veterinária, Departamento de Patologia Animal, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Cristina Gevehr Fernandes
- Programa de Pós-Graduação em Veterinária, Departamento de Patologia Animal, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Thaís Larré Oliveira
- Programa de Pós-Graduação em Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Odir Antonio Dellagostin
- Programa de Pós-Graduação em Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, RS, Brazil.
| |
Collapse
|
4
|
de Queiroz NMGP, Marinho FV, Chagas MA, Leite LCC, Homan EJ, de Magalhães MTQ, Oliveira SC. Vaccines for COVID-19: perspectives from nucleic acid vaccines to BCG as delivery vector system. Microbes Infect 2020; 22:515-524. [PMID: 32961274 PMCID: PMC7501874 DOI: 10.1016/j.micinf.2020.09.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 09/10/2020] [Indexed: 12/12/2022]
Abstract
This article discusses standard and new disruptive strategies in the race to develop an anti-COVID-19 vaccine. We also included new bioinformatic data from our group mapping immunodominant epitopes and structural analysis of the spike protein. Another innovative approach reviewed here is the use of BCG vaccine as priming strategy and/or delivery system expressing SARS-CoV-2 antigens.
Collapse
Affiliation(s)
- Nina Marí G P de Queiroz
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Fabio V Marinho
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Marcelo A Chagas
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luciana C C Leite
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, SP, Brazil
| | | | - Mariana T Q de Magalhães
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Sergio C Oliveira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), CNPq/MCT, BA, Brazil.
| |
Collapse
|
5
|
Rodriguez D, Goulart C, Pagliarone AC, Silva EP, Cunegundes PS, Nascimento IP, Borra RC, Dias WO, Tagliabue A, Boraschi D, Leite LCC. In vitro Evidence of Human Immune Responsiveness Shows the Improved Potential of a Recombinant BCG Strain for Bladder Cancer Treatment. Front Immunol 2019; 10:1460. [PMID: 31297119 PMCID: PMC6607967 DOI: 10.3389/fimmu.2019.01460] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 06/10/2019] [Indexed: 12/16/2022] Open
Abstract
The live attenuated mycobacterial strain BCG, in use as vaccine against tuberculosis, is considered the gold standard for primary therapy of carcinoma in situ of the bladder. Despite its limitations, to date it has not been surpassed by any other treatment. Our group has developed a recombinant BCG strain expressing the detoxified S1 pertussis toxin (rBCG-S1PT) that proved more effective than wild type BCG (WT-BCG) in increasing survival time in an experimental mouse model of bladder cancer, due to the well-known adjuvant properties of pertussis toxin. Here, we investigated the capacity of rBCG-S1PT to stimulate human immune responses, in comparison to WT-BCG, using an in vitro stimulation assay based on human whole blood cells that allows for a comprehensive evaluation of leukocyte activation. Blood leukocytes stimulated with rBCG-S1PT produced increased levels of IL-6, IL-8, and IL-10 as compared to WT-BCG, but comparable levels of IL-1β, IL-2, IFN-γ, and TNF-α. Stimulation of blood cells with the recombinant BCG strain also enhanced the expression of CD25 and CD69 on human CD4+ T cells. PBMC stimulated with rBCG-S1PT induced higher cytotoxicity to MB49 bladder cancer cells than WT-BCG-stimulated PBMC. These results suggest that the rBCG-S1PT strain is able to activate an immune response in human leukocytes that is higher than that induced by WT-BCG for parameters linked to better prognosis in bladder cancer (regulation of immune and early inflammatory responses), while fully comparable to WT-BCG for classical inflammatory parameters. This establishes rBCG-S1PT as a new highly effective candidate as immunotherapeutic agent against bladder cancer.
Collapse
Affiliation(s)
- Dunia Rodriguez
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, Brazil
| | - Cibelly Goulart
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, Brazil
| | - Ana C. Pagliarone
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, Brazil
| | - Eliane P. Silva
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, Brazil
- Programa de Pós-Graduação Interunidades em Biotecnologia USP-I.Butantan-IPT, São Paulo, Brazil
| | - Priscila S. Cunegundes
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, Brazil
- Programa de Pós-Graduação Interunidades em Biotecnologia USP-I.Butantan-IPT, São Paulo, Brazil
| | - Ivan P. Nascimento
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, Brazil
| | - Ricardo C. Borra
- Laboratório de Imunologia Aplicada, Departamento de Genética e Evolução, Universidade Federal de São Carlos, São Carlos, Brazil
| | - Waldely O. Dias
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, Brazil
| | - Aldo Tagliabue
- Institute of Genetic and Biomedical Research, National Research Council, Cagliari, Italy
| | - Diana Boraschi
- Institute of Protein Biochemistry, National Research Council, Naples, Italy
| | - Luciana C. C. Leite
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, Brazil
| |
Collapse
|
6
|
Gong W, Liang Y, Wu X. The current status, challenges, and future developments of new tuberculosis vaccines. Hum Vaccin Immunother 2018; 14:1697-1716. [PMID: 29601253 DOI: 10.1080/21645515.2018.1458806] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Mycobacterium tuberculosis complex causes tuberculosis (TB), one of the top 10 causes of death worldwide. TB results in more fatalities than multi-drug resistant (MDR) HIV strain related coinfection. Vaccines play a key role in the prevention and control of infectious diseases. Unfortunately, the only licensed preventive vaccine against TB, bacilli Calmette-Guérin (BCG), is ineffective for prevention of pulmonary TB in adults. Therefore, it is very important to develop novel vaccines for TB prevention and control. This literature review provides an overview of the innate and adaptive immune response during M. tuberculosis infection, and presents current developments and challenges to novel TB vaccines. A comprehensive understanding of vaccines in preclinical and clinical studies provides extensive insight for the development of safer and more efficient vaccines, and may inspire new ideas for TB prevention and treatment.
Collapse
Affiliation(s)
- Wenping Gong
- a Army Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Institute for Tuberculosis Research , Haidian District, Beijing , China
| | - Yan Liang
- a Army Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Institute for Tuberculosis Research , Haidian District, Beijing , China
| | - Xueqiong Wu
- a Army Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Institute for Tuberculosis Research , Haidian District, Beijing , China
| |
Collapse
|
7
|
New Recombinant Mycobacterium bovis BCG Expression Vectors: Improving Genetic Control over Mycobacterial Promoters. Appl Environ Microbiol 2016; 82:2240-2246. [PMID: 26850295 PMCID: PMC4959472 DOI: 10.1128/aem.03677-15] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 01/25/2016] [Indexed: 12/18/2022] Open
Abstract
The expression of many antigens, stimulatory molecules, or even metabolic pathways in mycobacteria such as Mycobacterium bovis BCG or M. smegmatis was made possible through the development of shuttle vectors, and several recombinant vaccines have been constructed. However, gene expression in any of these systems relied mostly on the selection of natural promoters expected to provide the required level of expression by trial and error. To establish a systematic selection of promoters with a range of strengths, we generated a library of mutagenized promoters through error-prone PCR of the strong PL5 promoter, originally from mycobacteriophage L5. These promoters were cloned upstream of the enhanced green fluorescent protein reporter gene, and recombinant M. smegmatis bacteria exhibiting a wide range of fluorescence levels were identified. A set of promoters was selected and identified as having high (pJK-F8), intermediate (pJK-B7, pJK-E6, pJK-D6), or low (pJK-C1) promoter strengths in both M. smegmatis and M. bovisBCG. The sequencing of the promoter region demonstrated that it was extensively modified (6 to 11%) in all of the plasmids selected. To test the functionality of the system, two different expression vectors were demonstrated to allow corresponding expression levels of the Schistosoma mansoni antigen Sm29 in BCG. The approach used here can be used to adjust expression levels for synthetic and/or systems biology studies or for vaccine development to maximize the immune response.
Collapse
|
8
|
Wang Y, Yang M, Yu Q, Yu L, Shao S, Wang X. Recombinant bacillus Calmette-Guérin in urothelial bladder cancer immunotherapy: current strategies. Expert Rev Anticancer Ther 2014; 15:85-93. [PMID: 25231670 DOI: 10.1586/14737140.2015.961430] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Bacillus Calmette-Guérin (BCG) has been used in the intravesical treatment of urothelial bladder cancer (UBC) for three decades. Despite its efficacy, intravesical BCG therapy is associated with some limitations such as side effects and BCG failure, which have inspired multiple ways to improve it. Recent advances have focused on recombinant BCG (rBCG) which provides a novel tactic for modification of BCG. To date, a number of rBCG strains have been developed and demonstrated to encourage efficacy and safety in preclinical and clinical studies. This review summarizes current rBCG strategies, concerns and future directions in UBC immunotherapy with an intention to encourage further research and eventually to inform clinical decisions.
Collapse
Affiliation(s)
- Yonghua Wang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, 266071, China
| | | | | | | | | | | |
Collapse
|
9
|
Liu D, Lu H, Shi K, Su F, Li J, Du R. Immunogenicity of recombinant BCGs expressing predicted antigenic epitopes of bovine viral diarrhea virus E2 gene. Res Vet Sci 2014; 97:430-8. [PMID: 25135492 DOI: 10.1016/j.rvsc.2014.07.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 06/09/2014] [Accepted: 07/03/2014] [Indexed: 12/18/2022]
Abstract
To develop a vaccine to prevent diseases caused by Mycobacterium tuberculosis and bovine viral diarrhea virus (BVDV) simultaneously, recombinant Bacillus Calmette-Guerin (rBCG) vaccines expressing different regions of the BVDV E2 gene were constructed. Using DNASTAR 6.0 software, potential antigenic epitopes were predicted, and six regions were chosen to generate recombinant plasmids with the pMV361 vector (pMV361-E2-1, pMV361-E2-2, pMV361-E2-3, pMV361-E2-4, pMV361-E2-5 and pMV361-E2-6, respectively). The recombinant plasmids were transformed into BCG, and protein expression was thermally induced at 45 °C. Mice were immunized with 5 × 10(6) CFU/200 µL of each rBCG strain. Compared with other groups, BVDV E2 specific antibody titers were higher in mice immunized with rBCG-E2-6. Ratios and numbers of CD4+, CD8+ and IL-12 expressing spleen lymphocytes of the rBCG-E2-6 group also were higher than those of other groups. Thus, the rBCG-E2-6 vaccine showed the highest immunogenicity of all groups based on the humoral and cellular responses to vaccination.
Collapse
Affiliation(s)
- Dongxu Liu
- College of Chinese Medicine Material, Jilin Agricultural University, Changchun 130118, China
| | - Huijun Lu
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun 130122, China
| | - Kun Shi
- College of Chinese Medicine Material, Jilin Agricultural University, Changchun 130118, China
| | - Fengyan Su
- College of Chinese Medicine Material, Jilin Agricultural University, Changchun 130118, China
| | - Jianming Li
- College of Chinese Medicine Material, Jilin Agricultural University, Changchun 130118, China
| | - Rui Du
- College of Chinese Medicine Material, Jilin Agricultural University, Changchun 130118, China.
| |
Collapse
|
10
|
Oral biosciences: The annual review 2012. J Oral Biosci 2013. [DOI: 10.1016/j.job.2013.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
11
|
Protection by a recombinant Mycobacterium bovis Bacillus Calmette-Guerin vaccine expressing Shiga toxin 2 B subunit against Shiga toxin-producing Escherichia coli in mice. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2012; 19:1932-7. [PMID: 23035176 DOI: 10.1128/cvi.00473-12] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We have developed a novel vaccine against Shiga toxin (Stx)-producing Escherichia coli (STEC) infection using a recombinant Mycobacterium bovis BCG (rBCG) system. Two intraperitoneal vaccinations with rBCG expressing the Stx2 B subunit (Stx2B) resulted in an increase of protective serum IgG and mucosal IgA responses to Stx2B in BALB/c mice. When orally challenged with 10(3) CFU of STEC strain B2F1 (O91: H21), the immunized mice survived statistically significantly longer than the nonvaccinated mice. We suggest that intraperitoneal immunization with rBCG expressing Stx2B would be a potential vaccine strategy for STEC.
Collapse
|
12
|
Generation of recombinant bacillus Calmette–Guérin and Mycobacterium smegmatis expressing BfpA and intimin as vaccine vectors against enteropathogenic Escherichia coli. Vaccine 2012; 30:5999-6005. [DOI: 10.1016/j.vaccine.2012.05.083] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2012] [Revised: 05/11/2012] [Accepted: 05/30/2012] [Indexed: 11/22/2022]
|
13
|
Enhanced priming of adaptive immunity by Mycobacterium smegmatis mutants with high-level protein secretion. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2012; 19:1416-25. [PMID: 22787192 DOI: 10.1128/cvi.00131-12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Mycobacteria have features that make them attractive as potential vaccine vectors. The nonpathogenic and rapidly growing Mycobacterium smegmatis can express both Mycobacterium tuberculosis antigens and heterologous antigens from other pathogens, and it has been used as a viable vector for the development of live vaccines. In order to further improve antigen-specific immunogenicity of M. smegmatis, we screened a random transposon mutant library for mutants displaying enhanced efficiency of protein secretion ("high secretors") and isolated 61 mutants showing enhanced endogenic and transgenic protein secretion. Sequence analysis identified a total of 54 genes involved in optimal secretion of insert proteins, as well as multiple independent transposon insertions localized within the same genomic loci and operons. The majority of transposon insertions occurred in genes that have no known protein secretion function. These transposon mutants were shown to prime antigen-specific CD8(+) T cell responses better than the parental strain. Specifically, upon introducing the simian immunodeficiency virus (SIV) gag gene into these transposon mutant strains, we observed that they primed SIV Gag-specific CD8(+) T cell responses significantly better than the control prime immunization in a heterologous prime/boost regimen. Our results reveal a dependence on bacterial secretion of mycobacterial and foreign antigens for the induction of antigen-specific CD8(+) T cells in vivo. The data also suggest that these M. smegmatis transposon mutants could be used as novel live attenuated vaccine strains to express foreign antigens, such as those of human immunodeficiency virus type 1 (HIV-1), and induce strong antigen-specific T cell responses.
Collapse
|
14
|
Progress in Oral Vaccination against Tuberculosis in Its Main Wildlife Reservoir in Iberia, the Eurasian Wild Boar. Vet Med Int 2012; 2012:978501. [PMID: 22848869 PMCID: PMC3400400 DOI: 10.1155/2012/978501] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Accepted: 05/08/2012] [Indexed: 11/17/2022] Open
Abstract
Eurasian wild boar (Sus scrofa) is the main wildlife reservoir for tuberculosis (TB) in Iberia. This review summarizes the current knowledge on wild boar vaccination including aspects of bait design, delivery and field deployment success; wild boar response to vaccination with Bacillus Calmette-Guérin (BCG) and inactivated Mycobacterium bovis; and wild boar vaccination biosafety issues as well as prospects on future research. Oral vaccination with BCG in captive wild boar has shown to be safe with significant levels of protection against challenge with virulent M. bovis. An oral vaccination with a new heat-killed M. bovis vaccine conferred a protection similar to BCG. The study of host-pathogen interactions identified biomarkers of resistance/susceptibility to tuberculosis in wild boar such as complement component 3 (C3) and methylmalonyl coenzyme A mutase (MUT) that were used for vaccine development. Finally, specific delivery systems were developed for bait-containing vaccines to target different age groups. Ongoing research includes laboratory experiments combining live and heat-killed vaccines and the first field trial for TB control in wild boar.
Collapse
|
15
|
Ohara N. Current status of tuberculosis and recombinant bacillus Calmette-Guérin vaccines. J Oral Biosci 2012. [DOI: 10.1016/j.job.2012.04.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
16
|
Kong CU, Ng LG, Nambiar JK, Spratt JM, Weninger W, Triccas JA. Targeted induction of antigen expression within dendritic cells modulates antigen-specific immunity afforded by recombinant BCG. Vaccine 2011; 29:1374-81. [DOI: 10.1016/j.vaccine.2010.12.070] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2010] [Revised: 11/24/2010] [Accepted: 12/16/2010] [Indexed: 11/26/2022]
|
17
|
Abstract
Elucidating the function of mycobacterial proteins, determining their contribution to virulence, and developing new vaccine candidates has been facilitated by systems permitting the heterologous expression of genes in mycobacteria. Mycobacterium bovis bacille Calmette Guérin (BCG) and Mycobacterium smegmatis have commonly been employed as host systems for the heterologous expression of mycobacterial genes as well as genes from other bacteria, viruses, and mammalian cells. Vectors that permit strong, constitutive expression of genes have been developed, and more recently systems that allow tightly regulated induction of gene expression have become available. In this chapter, we describe two complementary techniques relevant to the field of gene expression in mycobacteria. We first outline the methodology used for the expression and specific detection of recombinant products expressed in BCG. The expression vectors described use an epitope tag fused to the C-terminal end of the foreign protein, ablating the need for additional reagents to detect the recombinant product. Second, we describe the inducible expression of genes in recombinant M. smegmatis and the subsequent purification of gene products using affinity chromatography.
Collapse
|
18
|
Yuan S, Shi C, Liu L, Han W. MUC1-based recombinant Bacillus Calmette-Guerin vaccines as candidates for breast cancer immunotherapy. Expert Opin Biol Ther 2010; 10:1037-48. [PMID: 20420512 DOI: 10.1517/14712598.2010.485185] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
IMPORTANCE OF THE FIELD The challenge in breast cancer vaccine development is to find the best combination of antigen, adjuvant and delivery system to produce a strong and long-lasting immune response. Mucin 1 (MUC1) is a potential candidate target for breast cancer immunotherapy. Bacillus Calmette-Guerin (BCG) is used widely in human vaccines. Furthermore, it can potentially offer unique advantages for developing a safe and effective multi-vaccine vehicle. Due to these properties, the development of MUC1 based recombinant BCG (rBCG) vaccines for breast cancer immunotherapy has gained great momentum in recent years. AREAS COVERED IN THIS REVIEW Our aim is to discuss the recent progress in MUC1-based breast cancer immunotherapy and to highlight the advantages of MUC1-based rBCG vaccines as the new breast cancer vaccines. WHAT THE READER WILL GAIN Several promising MUC1-based rBCG vaccines have been shown to induce MUC1-specific antitumor immune responses in pre-clinical studies. This review updates and evaluates this very important and rapidly developing field, and provides a critical perspective and information source for its potential clinical applications. TAKE HOME MESSAGE MUC1-based rBCG vaccines have been shown to elicit an effective anti-tumor immune response in vivo demonstrating its potential utility in breast cancer treatment.
Collapse
Affiliation(s)
- Shifang Yuan
- Fourth Military Medical University, Xijing Hospital, Department of Vascular and Endocrine Surgery, Xi'an, 710032, People's Republic of China.
| | | | | | | |
Collapse
|
19
|
Molecular characterization of heterologous HIV-1gp120 gene expression disruption in mycobacterium bovis BCG host strain: a critical issue for engineering mycobacterial based-vaccine vectors. J Biomed Biotechnol 2010; 2010:357370. [PMID: 20617151 PMCID: PMC2896670 DOI: 10.1155/2010/357370] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2010] [Revised: 03/26/2010] [Accepted: 04/22/2010] [Indexed: 11/19/2022] Open
Abstract
Mycobacterium bovis Bacillus Calmette-Guérin (BCG) as a live vector of recombinant bacterial vaccine is a promising system to be used. In this study, we evaluate the disrupted expression of heterologous HIV-1gp120 gene in BCG Pasteur host strain using replicative vectors pMV261 and pJH222. pJH222 carries a lysine complementing gene in BCG lysine auxotrophs. The HIV-1 gp120 gene expression was regulated by BCG hsp60 promoter (in plasmid pMV261) and Mycobacteria spp. α-antigen promoter (in plasmid pJH222). Among 14 rBCG:HIV-1gp120 (pMV261) colonies screened, 12 showed a partial deletion and two showed a complete deletion. However, deletion was not observed in all 10 rBCG:HIV-1gp120 (pJH222) colonies screened. In this study, we demonstrated that E. coli/Mycobacterial expression vectors bearing a weak promoter and lysine complementing gene in a recombinant lysine auxotroph of BCG could prevent genetic rearrangements and disruption of HIV 1gp120 gene expression, a key issue for engineering Mycobacterial based vaccine vectors.
Collapse
|
20
|
Development of Th1 imprints to rBCG expressing a foreign protein: implications for vaccination against HIV-1 and diverse influenza strains. J Biomed Biotechnol 2010; 2010:591348. [PMID: 20625498 PMCID: PMC2896700 DOI: 10.1155/2010/591348] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2009] [Revised: 03/14/2010] [Accepted: 03/22/2010] [Indexed: 11/18/2022] Open
Abstract
We demonstrate here that immunizing naïve mice with low numbers of recombinant Bacille Calmette-Guérin (rBCG) expressing β-galactosidase (β-gal) generates predominant Th1 responses to both BCG and β-gal whereas infection with high numbers generates a mixed Th1/Th2 response to both BCG and β-gal. Furthermore, the Th1 response to both BCG and β-gal is stable when mice, pre-exposed to low numbers of rBCG, are challenged four months later with high numbers of rBCG. Thus the Th1/Th2 phenotypes of the immune responses to β-gal and to BCG are “coherently” regulated. Such rBCG vectors, encoding antigens of pathogens preferentially susceptible to cell-mediated attack, may be useful in vaccinating against such pathogens. We discuss vaccination strategies employing rBCG vectors that are designed to provide protection against diverse influenza strains or numerous variants of HIV-1 and consider what further experiments are essential to explore the possibility of realizing such strategies.
Collapse
|
21
|
Triccas JA. Recombinant BCG as a vaccine vehicle to protect against tuberculosis. Bioeng Bugs 2010; 1:110-5. [PMID: 21326936 PMCID: PMC3026451 DOI: 10.4161/bbug.1.2.10483] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2009] [Revised: 11/01/2009] [Accepted: 11/02/2009] [Indexed: 11/19/2022] Open
Abstract
Mycobacterium bovis Bacille Calmette Guérin (BCG) was first administered to humans in 1921 and has subsequently been delivered to an estimated 3 billion individuals, with a low incidence of serious complications. The vaccine is immunogenic and is stable and cheap to produce. Additionally, the vaccine can be engineered to express foreign molecules in a functional form, and this has driven the development of BCG as a recombinant vector to protect against infectious diseases and malignancies such as cancer. However, it is now clear that the existing BCG vaccine has proved insufficient to control the spread of tuberculosis, and a major focus of tuberculosis vaccine development programs is the construction and testing of modified forms of BCG. This review summarizes the strategies employed to develop recombinant forms of BCG and describes the potential of these vaccines to stimulate protective immunity and protect against Mycobacterium tuberculosis infection.
Collapse
Affiliation(s)
- James A Triccas
- Discipline of Infectious Diseases and Immunology, Blackburn Building, University of Sydney, NSW Australia.
| |
Collapse
|
22
|
Construction of an unmarked recombinant BCG expressing a pertussis antigen by auxotrophic complementation: protection against Bordetella pertussis challenge in neonates. Vaccine 2009; 27:7346-51. [PMID: 19782111 DOI: 10.1016/j.vaccine.2009.09.043] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2009] [Revised: 09/02/2009] [Accepted: 09/13/2009] [Indexed: 10/20/2022]
Abstract
Mycobacterium bovis BCG has long been investigated as a candidate for heterologous antigen presentation. We have previously described an rBCG-Pertussis that confers protection against challenge with Bordetella pertussis in neonate and adult mice. In order to obtain stable expression in vivo, we constructed an unmarked BCG lysine auxotrophic and a complementation vector containing the lysine and the genetically detoxified S1 pertussis toxin genes, both under control of the same promoter. Complemented BCG-Delta lysine growth and expression of the pertussis antigen were stable, without the use of an antibiotic marker. Our results show that the complemented rBCG-Delta lysA-S1PT-lysA(+)(kan(-)), which is now suitable to be evaluated in clinical trials, maintains similar characteristics of the original rBCG-pNL71S1PT strain, such as the antigen expression level, cellular immune response and protection against the same model challenge in neonatal-immunized mice.
Collapse
|
23
|
Chege GK, Thomas R, Shephard EG, Meyers A, Bourn W, Williamson C, Maclean J, Gray CM, Rybicki EP, Williamson AL. A prime-boost immunisation regimen using recombinant BCG and Pr55(gag) virus-like particle vaccines based on HIV type 1 subtype C successfully elicits Gag-specific responses in baboons. Vaccine 2009; 27:4857-66. [PMID: 19520196 DOI: 10.1016/j.vaccine.2009.05.064] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2008] [Revised: 05/13/2009] [Accepted: 05/21/2009] [Indexed: 12/11/2022]
Abstract
Mycobacterium bovis BCG is considered an attractive live bacterial vaccine vector. In this study, we investigated the immune response of baboons to a primary vaccination with recombinant BCG (rBCG) constructs expressing the gag gene from a South African HIV-1 subtype C isolate, and a boost with HIV-1 subtype C Pr55(gag) virus-like particles (Gag VLPs). Using an interferon enzyme-linked immunospot assay, we show that although these rBCG induced only a weak or an undetectable HIV-1 Gag-specific response on their own, they efficiently primed for a Gag VLP boost, which strengthened and broadened the immune responses. These responses were predominantly CD8+ T cell-mediated and recognised similar epitopes as those targeted by humans with early HIV-1 subtype C infection. In addition, a Gag-specific humoral response was elicited. These data support the development of HIV-1 vaccines based on rBCG and Pr55(gag) VLPs.
Collapse
Affiliation(s)
- Gerald K Chege
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town, South Africa
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Wang Q, Li J, Zhang X, Liu C, Cao L, Ren K, Gong P, Cai Y. Construction of EGFP-tagged rBCG of E.tenella and distribution in chickens. ACTA ACUST UNITED AC 2009; 52:278-83. [PMID: 19294353 DOI: 10.1007/s11427-009-0008-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2008] [Accepted: 09/09/2008] [Indexed: 11/26/2022]
Abstract
Chicken coccidiosis is a major parasitic disease with substantial economic burden to the poultry industry. Enhanced green fluorescent protein (EGFP) tagged recombinant Bacille Calmette-Guerin (rBCG), as a fusion protein with coccidian rhomboid antigen was constructed to track rBCG in vivo in chickens in this study. Immunization of chickens with one dose of rBCG pMV361-Rho/EGFP induced humoral immune response. The colonization of rBCG in liver, spleen, lung, kidney and caecum was observed by laser confocal microscopy. Real-time quantitative RT-PCR showed a rise expression level of rhomboid protein on the 7th day and a peak on the 14th day and disappearance on the 28th day after immunization. These results have significant implications for the development of rBCG vaccines against avian coccidiosis.
Collapse
Affiliation(s)
- QiuYue Wang
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun, 130062, China
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Wang Q, Li J, Zhang X, Liu Q, Liu C, Ma G, Cao L, Gong P, Cai Y, Zhang G. Protective immunity of recombinant Mycobacterium bovis BCG expressing rhomboid gene against Eimeria tenella challenge. Vet Parasitol 2009; 160:198-203. [DOI: 10.1016/j.vetpar.2008.11.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2008] [Revised: 10/22/2008] [Accepted: 11/05/2008] [Indexed: 10/21/2022]
|
26
|
da Silva Ramos Rocha A, Conceição FR, Grassmann AA, Lagranha VL, Dellagostin OA. B subunit ofEscherichia coliheat-labile enterotoxin as adjuvant of humoral immune response in recombinant BCG vaccination. Can J Microbiol 2008; 54:677-86. [DOI: 10.1139/w08-056] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The B subunit of Escherichia coli heat-labile enterotoxin (LTB), a nontoxic molecule with potent biological properties, is a powerful mucosal and parenteral adjuvant that induces a strong immune response against co-administered or coupled antigens. In this paper, the effect of LTB on the humoral immune response to recombinant BCG (rBCG) vaccination was evaluated. Isogenic mice were immunized with rBCG expressing the R1 repeat region of the P97 adhesin of Mycoplasma hyopneumoniae alone (rBCG/R1) or fused to LTB (rBCG/LTBR1). Anti-R1 systemic antibody levels (IgG1, IgG2a, IgG2b, IgG3, IgM, and IgA) were measured by ELISA using recombinant R1 as antigen. With the exception of IgM, LTB doubled the anti-R1 antibody levels in rBCG vaccination. The IgG1/IgG2a mean ratio showed that both rBCG/LTBR1 and rBCG/R1 induced a mixed Th1/Th2 immune response. Interestingly, anti-R1 serum IgA was induced only by rBCG/LTBR1. These results demonstrate that LTB has an adjuvant effect on the humoral immune response to recombinant antigens expressed in BCG.
Collapse
Affiliation(s)
- Andréa da Silva Ramos Rocha
- Centro de Biotecnologia, Universidade Federal de Pelotas, CP 354, Pelotas, RS 96010-900, Brazil
- Faculdade de Veterinária, Universidade Federal de Pelotas, Pelotas, RS 96010-900, Brazil
- Instituto de Biologia, Universidade Federal de Pelotas, Pelotas, RS 96010-900, Brazil
| | - Fabricio Rochedo Conceição
- Centro de Biotecnologia, Universidade Federal de Pelotas, CP 354, Pelotas, RS 96010-900, Brazil
- Faculdade de Veterinária, Universidade Federal de Pelotas, Pelotas, RS 96010-900, Brazil
- Instituto de Biologia, Universidade Federal de Pelotas, Pelotas, RS 96010-900, Brazil
| | - André Alex Grassmann
- Centro de Biotecnologia, Universidade Federal de Pelotas, CP 354, Pelotas, RS 96010-900, Brazil
- Faculdade de Veterinária, Universidade Federal de Pelotas, Pelotas, RS 96010-900, Brazil
- Instituto de Biologia, Universidade Federal de Pelotas, Pelotas, RS 96010-900, Brazil
| | - Valeska Lizzi Lagranha
- Centro de Biotecnologia, Universidade Federal de Pelotas, CP 354, Pelotas, RS 96010-900, Brazil
- Faculdade de Veterinária, Universidade Federal de Pelotas, Pelotas, RS 96010-900, Brazil
- Instituto de Biologia, Universidade Federal de Pelotas, Pelotas, RS 96010-900, Brazil
| | - Odir Antônio Dellagostin
- Centro de Biotecnologia, Universidade Federal de Pelotas, CP 354, Pelotas, RS 96010-900, Brazil
- Faculdade de Veterinária, Universidade Federal de Pelotas, Pelotas, RS 96010-900, Brazil
- Instituto de Biologia, Universidade Federal de Pelotas, Pelotas, RS 96010-900, Brazil
| |
Collapse
|
27
|
Martin C, Walker B. Attenuated strains of Mycobacterium tuberculosis complex for laboratory and clinical use. Tuberculosis (Edinb) 2008; 88:371-4. [PMID: 18550439 DOI: 10.1016/j.tube.2008.04.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Carlos Martin
- CIBER de Enfermedades Respiratorias, Department of Microbiology, Faculty of Medicine, University of Zaragoza, C/Domingo Miral sn, Zaragoza, Spain.
| | | |
Collapse
|
28
|
Wang JL, Qie YQ, Zhu BD, Zhang HM, Xu Y, Wang QZ, Chen JZ, Liu W, Wang HH. Evaluation of a recombinant BCG expressing antigen Ag85B and PPE protein Rv3425 from DNA segment RD11 of Mycobacterium tuberculosis in C57BL/6 mice. Med Microbiol Immunol 2008; 198:5-11. [PMID: 18491134 DOI: 10.1007/s00430-008-0098-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2007] [Indexed: 11/25/2022]
Abstract
Antigen 85B (Ag85B) is an important immunodominant antigen of Mycobacterium tuberculosis, and is a very promising vaccine candidate molecule. Rv3425 is a member of the subgroup 3 of the PPE family, which does not exist in all BCG strains. In this study we constructed a new rBCG which included this united gene (Ag85B-Rv3425). The level of antigen-stimulated T cells expressing IFN-gamma was significantly higher in the C57BL/6 mice vaccinated with rBCG::Ag85B-Rv3425 than with BCG. In addition, the sera from mice immunized with rBCG::Ag85B-Rv3425 revealed an increase in the specific immunoglobulin G titers than that from mice immunized with BCG. Antigen specific IgG subclass analysis showed that rBCG::Ag85B-Rv3425 tended to facilitate IgG2a production, suggesting enhancement of predominant Th1 response which in turn may facilitate increased production of protective IFN-gamma. These results suggested that this rBCG::Ag85B-Rv3425 could be a strong vaccine candidate for further study.
Collapse
Affiliation(s)
- Jiu ling Wang
- Institute of Genetics, Fudan University, 200433, Shanghai, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Asensio JAG, Arbués A, Pérez E, Gicquel B, Martin C. Live tuberculosis vaccines based onphoPmutants: a step towards clinical trials. Expert Opin Biol Ther 2008; 8:201-11. [DOI: 10.1517/14712598.8.2.201] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
30
|
Nascimento IP, Dias WO, Quintilio W, Christ AP, Moraes JF, Vancetto MDC, Ribeiro-Dos-Santos G, Raw I, Leite LCC. Neonatal immunization with a single dose of recombinant BCG expressing subunit S1 from pertussis toxin induces complete protection against Bordetella pertussis intracerebral challenge. Microbes Infect 2007; 10:198-202. [PMID: 18248757 DOI: 10.1016/j.micinf.2007.10.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2007] [Revised: 09/21/2007] [Accepted: 10/17/2007] [Indexed: 10/22/2022]
Abstract
The currently used pertussis vaccines are highly efficacious; however, neonates are susceptible to whooping cough up to the sixth month. In agreement, DTP-immunized neonate mice were not protected against intracerebral challenge with Bordetella pertussis. Neonate mice immunized with either DTP or a recombinant-BCG strain expressing the genetically detoxified S1 subunit of pertussis toxin do not show a humoral immune response against PT. On the other hand, rBCG-Pertussis induces higher PT-specific IFN-gamma production and an increase in both IFN-gamma(+) and TNF-alpha(+)-CD4(+)-T cells than the whole cell pertussis vaccine and confers protection against a lethal intracerebral challenge with B. pertussis.
Collapse
Affiliation(s)
- Ivan P Nascimento
- Centro de Biotecnologia, Instituto Butantan, Av. Vital Brasil 1500, 05503-900 São Paulo, SP, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Seixas FK, da Silva EF, Hartwig DD, Cerqueira GM, Amaral M, Fagundes MQ, Dossa RG, Dellagostin OA. Recombinant Mycobacterium bovis BCG expressing the LipL32 antigen of Leptospira interrogans protects hamsters from challenge. Vaccine 2007; 26:88-95. [PMID: 18063449 DOI: 10.1016/j.vaccine.2007.10.052] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2007] [Revised: 10/12/2007] [Accepted: 10/18/2007] [Indexed: 10/22/2022]
Abstract
The bacillus Calmette--Guerin (BCG), a live attenuated Mycobacterium bovis strain is considered a promising candidate as a vector system for delivery of foreign antigens to the immune system. The gene coding for the Leptospira interrogans external membrane protein LipL32, a highly immunogenic antigen found in all pathogenic leptospira, was cloned into several mycobacterial vectors for expression in BCG. Hamsters immunized with recombinant BCG (rBCG) expressing LipL32 were protected against mortality (P<or=0.05) upon challenge with a lethal inoculum of L. interrogans serovar Copenhageni. Autopsy examination did not reveal macroscopic or histological evidence of disease in rBCG immunized hamsters that survived lethal challenge. The data presented here further enhance the status of LipL32 as a promising candidate antigen for use in the control of leptospirosis, when presented to the immune system by an appropriate delivery system.
Collapse
Affiliation(s)
- Fabiana Kömmling Seixas
- Centro de Biotecnologia, Universidade Federal de Pelotas, Campus Universitário, 96010-900 Pelotas - RS, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Borsuk S, Mendum TA, Fagundes MQ, Michelon M, Cunha CW, McFadden J, Dellagostin OA. Auxotrophic complementation as a selectable marker for stable expression of foreign antigens in Mycobacterium bovis BCG. Tuberculosis (Edinb) 2007; 87:474-80. [PMID: 17888740 DOI: 10.1016/j.tube.2007.07.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2007] [Revised: 06/20/2007] [Accepted: 07/19/2007] [Indexed: 10/22/2022]
Abstract
Mycobacterium bovis BCG has the potential to be an effective live vector for multivalent vaccines. However, most mycobacterial cloning vectors rely on antibiotic resistance genes as selectable markers, which would be undesirable in any practical vaccine. Here we report the use of auxotrophic complementation as a selectable marker that would be suitable for use in a recombinant vaccine. A BCG auxotrophic for the amino acid leucine was constructed by knocking out the leuD gene by unmarked homologous recombination. Expression of leuD on a plasmid not only allowed complementation, but also acted as a selectable marker. Removal of the kanamycin resistance gene, which remained necessary for plasmid manipulations in Escherichia coli, was accomplished by two different methods: restriction enzyme digestion followed by re-ligation before BCG transformation, or by Cre-loxP in vitro recombination mediated by the bacteriophage P1 Cre Recombinase. Stability of the plasmid was evaluated during in vitro and in vivo growth of the recombinant BCG in comparison to selection by antibiotic resistance. The new system was highly stable even during in vivo growth, as the selective pressure is maintained, whereas the conventional vector was unstable in the absence of selective pressure. This new system will now allow the construction of potential recombinante vaccine strains using stable multicopy plasmid vectors without the inclusion of antibiotic resistance markers.
Collapse
Affiliation(s)
- Sibele Borsuk
- Centro de Biotecnologia, Universidade Federal de Pelotas, CP-354, 96010-900 Pelotas, RS, Brazil
| | | | | | | | | | | | | |
Collapse
|
33
|
Bourn WR, Jansen Y, Stutz H, Warren RM, Williamson AL, van Helden PD. Creation and characterisation of a high-copy-number version of the pAL5000 mycobacterial replicon. Tuberculosis (Edinb) 2007; 87:481-8. [PMID: 17888739 DOI: 10.1016/j.tube.2007.08.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2007] [Revised: 07/31/2007] [Accepted: 08/04/2007] [Indexed: 11/21/2022]
Abstract
The majority of mycobacterial plasmid vectors are derived from the pAL5000 replicon and maintained at approximately five copies per cell. We have devised a method that directly selects for high-copy-number plasmids. This involves enriching for high copy number plasmids by repeatedly isolating and retransforming plasmids from a mutant library. Using this method we have selected a copy-up version of the pAL5000 replicon. In Mycobacterium smegmatis the copy-number was shown to have increased 7-fold to between 32 and 64 copies/cell, and the plasmid remained relatively stable after 100 generations in the absence of antibiotic selection. The plasmid also has a high-copy-number phenotype in M. bovis BCG and can be used to increase expression of cloned genes, as we have demonstrated with the green fluorescent protein. The mutation was found to be the deletion of an alanine residue in the C-terminal end of the RepA replication protein. We argue that the mutation exerts its effect through altered RNA folding, thereby affecting the translationally coupled RepA-RepB expression.
Collapse
Affiliation(s)
- William R Bourn
- DST/NRF Centre of Excellence for Biomedical Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Health Sciences, Stellenbosch University, P.O. Box 19063, Tygerberg 7505, South Africa
| | | | | | | | | | | |
Collapse
|
34
|
Triccas JA, Shklovskaya E, Spratt J, Ryan AA, Palendira U, Fazekas de St Groth B, Britton WJ. Effects of DNA- and Mycobacterium bovis BCG-based delivery of the Flt3 ligand on protective immunity to Mycobacterium tuberculosis. Infect Immun 2007; 75:5368-75. [PMID: 17724075 PMCID: PMC2168302 DOI: 10.1128/iai.00322-07] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The control of intracellular pathogens such as Mycobacterium tuberculosis is dependent on the activation and maintenance of pathogen-reactive T cells. Dendritic cells (DCs) are the major antigen-presenting cells initiating antimycobacterial T-cell responses in vivo. To investigate if immunization strategies that aim to optimize DC function can improve protective immunity against virulent mycobacterial infection, we exploited the ability of the hematopoietic growth factor Fms-like tyrosine kinase 3 ligand (Flt3L) to expand the number of DCs in vivo. A DNA fusion of the genes encoding murine Flt3L and M. tuberculosis antigen 85B stimulated enhanced gamma interferon (IFN-gamma) release by T cells and provided better protection against virulent M. tuberculosis than DNA encoding the single components. Vaccination of mice with a recombinant Mycobacterium bovis BCG strain secreting Flt3L (BCG:Flt3L) led to early expansion of DCs compared to immunization with BCG alone, and this effect was associated with increased stimulation of BCG-reactive IFN-gamma-secreting T cells. BCG and BCG:Flt3L provided similar protective efficacies against low-dose aerosol M. tuberculosis; however, immunization of immunodeficient mice revealed that BCG:Flt3L was markedly less virulent than conventional BCG. These results demonstrate the potential of in vivo targeting of DCs to improve antimycobacterial vaccine efficacy.
Collapse
Affiliation(s)
- James A Triccas
- Microbial Pathogenesis and Immunity Group, Discipline of Infectious Diseases and Immunology, University of Sydney, Sydney, Australia.
| | | | | | | | | | | | | |
Collapse
|
35
|
Expression and immunogenicity of recombinant Mycobacterium bovis Bacillus Calmette-Guérin strains secreting the antigen ESAT-6 from Mycobacterium tuberculosis in mice. Chin Med J (Engl) 2007. [DOI: 10.1097/00029330-200707020-00003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
36
|
Seixas FK, Fernandes CH, Hartwig DD, Conceição FR, Aleixo JAG, Dellagostin OA. Evaluation of different ways of presenting LipL32 to the immune system with the aim of developing a recombinant vaccine against leptospirosis. Can J Microbiol 2007; 53:472-9. [PMID: 17612601 DOI: 10.1139/w06-138] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Leptospirosis, caused by bacteria of the genus Leptospira , is a direct zoonosis with wide geographical distribution. The implications in terms of public health and the economical losses caused by leptospirosis justify the use of a vaccine against Leptospira in human or animal populations at risk. In this study, we used the external membrane protein LipL32 as a model antigen, as it is highly immunogenic. The LipL32 coding sequence was cloned into several expression vectors: (i) pTarget, to create a DNA vaccine; (ii) pUS973, pUS974, and pUS977 for expression in BCG (rBCG); and (iii) pAE, to express the recombinant protein in Escherichia coli , for a subunit vaccine. Mice were immunized with the various constructs, and the immune response was evaluated. The highest humoral immune response was elicited by the subunit vaccine (rLipL32). However, with rBCG, the titer was still rising at the end of the experiment. The serum of vaccinated animals was able to recognize LipL32 on the membrane of the Leptospira, detected by indirect immunofluorescence. A monoclonal antibody anti-LipL32 was shown to inhibit the growth of Leptospira in vitro, indicating potential protection induced by the LipL32 antigen.
Collapse
|
37
|
Dennehy M, Bourn W, Steele D, Williamson AL. Evaluation of recombinant BCG expressing rotavirus VP6 as an anti-rotavirus vaccine. Vaccine 2007; 25:3646-57. [PMID: 17339069 DOI: 10.1016/j.vaccine.2007.01.087] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2006] [Revised: 12/19/2006] [Accepted: 01/11/2007] [Indexed: 11/16/2022]
Abstract
Recombinant BCG expressing rotavirus VP6 was explored as an anti-rotavirus vaccine in a mouse model. Three promoters and five ribosome-binding sites were used in episomal and integrative E. coli-mycobacterium shuttle vectors to express VP6 in BCG. The VP6 gene was configured for accumulation within the BCG cytoplasm, secretion from the BCG cell or targeting to the BCG cell membrane. Vectors were assessed in terms of stability, levels of antigen production, immunogenicity and protection in mice. Gross instability occurred in episomal vectors utilizing the hsp60 promoter. However, three integrative vectors using the same expression system and two episomal vectors using inducible promoters were successfully recovered from BCG. Growth rates of the former were not detectably reduced. Growth rates of the latter were considerably reduced, implying the existence of a significant metabolic load. In the absence of selection, loss rate of these plasmids was high. VP6 production levels (0.04-1.78% of total cytoplasmic protein) were on the lower end of the range reported for other rBCG. One episomal and one integrated vaccine reduced viral shedding in intraperitoneally vaccinated mice challenged with rotavirus. Compared to controls, infection-associated faecal shedding of virus was reduced by 66% and 62%, respectively. These protective vectors differ in promoter, ribosome-binding site and antigen production level, but both link the VP6 protein to the 19kDa lipoprotein signal sequence, suggesting that transport of VP6 to the BCG membrane is important for induction of a protective immune response. Protection occurred in the absence of detectable anti-rotavirus antibody in serum or faeces, implicating cellular immunity in protection.
Collapse
Affiliation(s)
- Maureen Dennehy
- Division of Medical Virology, Department of Clinical Laboratory Sciences, Faculty of Health Sciences, University of Cape Town, and National Health Laboratory Service, Groote Schuur Hospital, Cape Town, South Africa.
| | | | | | | |
Collapse
|
38
|
Santangelo MP, McIntosh D, Bigi F, Armôa GRG, Campos ASD, Ruybal P, Dellagostin OA, McFadden J, Mendum T, Gicquel B, Winter N, Farber M, Cataldi A. Mycobacterium bovis BCG as a delivery system for the RAP-1 antigen from Babesia bovis. Vaccine 2007; 25:1104-13. [PMID: 17049681 DOI: 10.1016/j.vaccine.2006.09.069] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2006] [Revised: 09/15/2006] [Accepted: 09/15/2006] [Indexed: 11/20/2022]
Abstract
Babesia bovis is the causative agent of babesiosis, a tick-borne disease that is a major cause of loss to livestock production in Latin America. Vaccination against Babesia species represents a major challenge against cattle morbidity and mortality in enzootic areas. The aim of this study was to evaluate the capacity of Bacille Calmette-Guerin (BCG) to deliver the rhoptry associated protein (RAP-1) antigen of B. bovis and to stimulate specific cellular and humoral immune responses in mice. Two of five mycobacterial expression vectors efficiently expressed the antigen. These constructs were subsequently studied in vivo following three immunization protocols. The construct with the greatest in vivo stability proved to be the one that induced the strongest immune responses. Our data support the hypothesis that specific T lymphocyte priming by rBCG can be employed as a component of a combined vaccine strategy to induce long-lasting humoral and cellular immune responsiveness towards B. bovis and encourage further work on the application of rBCG to the development of Babesia vaccines.
Collapse
Affiliation(s)
- M P Santangelo
- Institute of Biotechnology, CICVyA-INTA, Los Reseros y Las Cabañas, 1712 Castelar, Argentina
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Michelon A, Conceição FR, Binsfeld PC, da Cunha CW, Moreira AN, Argondizzo AP, McIntosh D, Armôa GRG, Campos AS, Farber M, McFadden J, Dellagostin OA. Immunogenicity of Mycobacterium bovis BCG expressing Anaplasma marginale MSP1a antigen. Vaccine 2006; 24:6332-9. [PMID: 16781025 DOI: 10.1016/j.vaccine.2006.05.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2006] [Revised: 05/11/2006] [Accepted: 05/16/2006] [Indexed: 11/16/2022]
Abstract
Humoral and cellular immune responses of mice inoculated with recombinant Mycobacterium bovis BCG expressing the MSP1a antigen of Anaplasma marginale were evaluated. The msp1a gene was amplified by PCR and cloned into the mycobacterial expression vectors pUS2000 and pMIP12. Immunization of isogenic BALB/c mice with the rBCG/pUS2000-msp1a construct induced significant seroconversion to MSP1a (p<0.001), which was 26 times above pre-immunization levels at day 63 post-initial immunization and which remained stable for the duration of the experiment (6 months). In contrast, rBCG/pMIP12-msp1a induced seroconversion at a level of 6 times above pre-immunization values, which peaked at day 63. Western blot analysis showed that sera derived from mice vaccinated with either rBCG construct recognized both native and recombinant forms of A. marginale MSP1a. In contrast to the humoral response data, immunization with rBCG/pMIP12-msp1a was found to induce a markedly stronger cellular response than that recorded for BCG/pUS2000-msp1a. These observations clearly demonstrated the immunogenicity of recombinant BCG expressing the MSP1a antigen and suggested that the immune responses were influenced by the level of antigen expression. The results of this research warrant studies of recombinant M. bovis BCG expressing MSP1a in cattle to test for protective antibody production for control of bovine anaplasmosis.
Collapse
Affiliation(s)
- André Michelon
- Centro de Biotecnologia, Universidade Federal de Pelotas, CP 354, 96010-900 Pelotas, RS, Brazil
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Medeiros MA, Armôa GRG, Dellagostin OA, McIntosh D. Induction of humoral immunity in response to immunization with recombinant Mycobacterium bovis BCG expressing the S1 subunit of Bordetella pertussis toxin. Can J Microbiol 2006; 51:1015-20. [PMID: 16462859 DOI: 10.1139/w05-095] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Two recombinant Mycobacterium bovis BCG (rBCG) vaccine strains were developed for the expression of cytoplasmically located S1 subunit of pertussis toxin, with expression driven by the hsp60 promoter of M. bovis (rBCG/pPB10) or the pAN promoter of Mycobacterium paratuberculosis (rBCG/pPB12). Both strains showed stable expression of equivalent levels of recombinant S1 in vitro and induced long-term (up to 8 months) humoral immune responses in BALB/c mice, although these responses differed quantitatively and qualitatively. Specifically, rBCG/pPB12 induced markedly higher levels of IgG1 than did rBCG/pPB10, and mice immunized with the former strain developed specific long-term memory to S1, as indicated by the production of high levels of S1-specific IgG in response to a sublethal challenge with pertussis toxin 15 months after initial immunization. When considered in combination with previous studies, our data encourage further evaluation of rBCG as a potential means of developing a low-cost whooping cough vaccine based on defined antigens.
Collapse
Affiliation(s)
- Marco A Medeiros
- Laboratory of Recombinant Technology, Bio-Manguinhos, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.
| | | | | | | |
Collapse
|
41
|
Cosgrove CA, Castello-Branco LRR, Hussell T, Sexton A, Giemza R, Phillips R, Williams A, Griffin GE, Dougan G, Lewis DJM. Boosting of cellular immunity against Mycobacterium tuberculosis and modulation of skin cytokine responses in healthy human volunteers by Mycobacterium bovis BCG substrain Moreau Rio de Janeiro oral vaccine. Infect Immun 2006; 74:2449-52. [PMID: 16552077 PMCID: PMC1418892 DOI: 10.1128/iai.74.4.2449-2452.2006] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Oral immunization of healthy adults with 10(7) CFU BCG Moreau Rio de Janeiro was well tolerated and significantly boosted gamma interferon responses to purified protein derivative, Ag85, and MPB70 from previous childhood intradermal BCG immunization. Oral BCG offers the possibility of a needle-free tuberculosis vaccine and of boosting the protective immunity from intradermal tuberculosis vaccines.
Collapse
Affiliation(s)
- Catherine A Cosgrove
- CMM (Infectious Diseases), St George's Hospital Medical School, Cranmer Terrace, London SW17 0RE, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Benévolo-de-Andrade TC, Monteiro-Maia R, Cosgrove C, Castello-Branco LRR. BCG Moreau Rio de Janeiro: an oral vaccine against tuberculosis - review. Mem Inst Oswaldo Cruz 2005; 100:459-65. [PMID: 16184220 DOI: 10.1590/s0074-02762005000500002] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The vaccine Bacillus of Calmette Guérin (BCG) was originally developed in France as an oral vaccine against tuberculosis. The oral use of this vaccine was replaced by the parenteral route in almost all countries after the Lubeck disaster. In contrast, Brazil retained the oral delivery of the vaccine until the mid-seventies when it was replaced by the intradermal route. This change in route of delivery was mainly secondary to pressure by medical practitioners based on the poor responses of oral immunized subjects to purified protein derivative (PPD) skin tests. Even after the change of route of delivery, Ataulpho de Paiva Foundation continued making the oral vaccine. Currently, BCG Moreau has been described as one of the most immunogenic and with fewer side effects than other BCGs. The genomics, proteomics and vaccine trials for oral BCG Moreau Rio de Janeiro are currently under investigation. In this review, we intend to describe the history of BCG Moreau Rio de Janeiro in Brazil.
Collapse
|
43
|
Nascimento IP, Leite LCC. The effect of passaging in liquid media and storage onMycobacterium bovisâ BCG growth capacity and infectivity. FEMS Microbiol Lett 2005; 243:81-6. [PMID: 15668004 DOI: 10.1016/j.femsle.2004.11.043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2004] [Revised: 11/08/2004] [Accepted: 11/24/2004] [Indexed: 11/19/2022] Open
Abstract
The effect of successive cultures--undergoing or not cycles of freezing, storage and thawing--on the growth curves of the Mycobacterium bovis Bacille Calmette-Guerin (BCG) Moreau strain and a recombinant-BCG (rBCG) vaccine preparation were evaluated. The results showed that both strains going through three rounds of freezing and thawing were not able to grow efficiently in the third stage of liquid culture. This effect and also long-term frozen storage appeared to be more preeminent in cultures that had been harvested at 0.8 optical density (OD at 600 nm) prior to freezing and storage, as in comparison to their 0.4 OD counterparts. Altogether, the data suggest that cultures inoculated with samples harvested at lower OD are less sensitive to the limiting effects of serial cultivation, regardless of being BCG or rBCG. Successive cultivations without freezing and thawing also affect growth of BCG culture inoculated with cells at later exponential phase (0.8 OD). Finally, macrophage infectivity with BCG cells from the third growth passage was significantly lower than from the first passage. These results draw attention to the importance of using fresh, low-passage and/or growth and infection capacity-controlled vaccine stocks for the evaluation of strains of BCG or rBCG.
Collapse
Affiliation(s)
- Ivan P Nascimento
- Centro de Biotecnologia, Instituto Butantan, Av. Vital Brasil 1500, São Paulo 05503-900, SP, Brazil
| | | |
Collapse
|
44
|
Spratt JM, Ryan AA, Britton WJ, Triccas JA. Epitope-tagging vectors for the expression and detection of recombinant proteins in mycobacteria. Plasmid 2005; 53:269-73. [PMID: 15848231 DOI: 10.1016/j.plasmid.2004.11.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2004] [Revised: 11/04/2004] [Accepted: 11/08/2004] [Indexed: 10/26/2022]
Abstract
New tools are required to study the growing number of uncharacterised genes derived from genome sequence projects that are specific to bacterial pathogens such as Mycobacterium tuberculosis. We have developed a series of vectors that permit the specific detection of recombinant proteins expressed in mycobacterial species. Gene expression in these vectors is driven by the strong hsp60 promoter of Mycobacterium bovis BCG and detection of expressed products is facilitated by C-terminal fusion of residues 409-419 of the human c-myc proto-oncogene. Using the M. tuberculosis Ag85B as a reporter of gene expression, we demonstrate that the vectors permit the specific detection of recombinant products expressed in the host species M. bovis BCG. BCG over-expressing Ag85B was a potent inducer of Ag85B-specific T cells in immunised mice, indicating that the C-terminal c-myc tag did not alter the characteristics of the recombinant protein. The versatility of the epitope-tagging vectors was demonstrated by the efficient secretion and detection of recombinant products in BCG. The vectors described in this study will facilitate the expression of foreign proteins in mycobacterial host systems.
Collapse
Affiliation(s)
- Joanne M Spratt
- Mycobacterial Research Group, Centenary Institute of Cancer Medicine and Cell Biology, Locked Bag No. 6, Newtown, NSW 2042, Australia
| | | | | | | |
Collapse
|
45
|
Dennehy M, Williamson AL. Factors influencing the immune response to foreign antigen expressed in recombinant BCG vaccines. Vaccine 2005; 23:1209-24. [PMID: 15652663 DOI: 10.1016/j.vaccine.2004.08.039] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2004] [Accepted: 08/26/2004] [Indexed: 11/30/2022]
Abstract
A wide range of recombinant BCG vaccine candidates containing foreign viral, bacterial, parasite or immunomodulatory genetic material have been developed and evaluated, primarily in animal models, for immune response to the foreign antigen. This review considers some of the factors that may influence the immunogenicity of these vaccines. The influence of levels and timing of expression of the foreign antigen and the use of targeting sequences are considered in the first section. Genetic and functional stability of rBCG is reviewed in the second section. In the last section, the influence of dose and route of immunization, strain of BCG and the animal model used are discussed.
Collapse
Affiliation(s)
- Maureen Dennehy
- The Biovac Institute, Private Bag X3, Pinelands, 7430 Cape Town, South Africa.
| | | |
Collapse
|
46
|
Antigen Delivery Systems II: Development of Live Recombinant Attenuated Bacterial Antigen and DNA Vaccine Delivery Vector Vaccines. Mucosal Immunol 2005. [DOI: 10.1016/b978-012491543-5/50060-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
47
|
Varaldo PB, Leite LCC, Dias WO, Miyaji EN, Torres FIG, Gebara VC, Armôa GRG, Campos AS, Matos DCS, Winter N, Gicquel B, Vilar MM, McFadden J, Almeida MS, Tendler M, McIntosh D. Recombinant Mycobacterium bovis BCG expressing the Sm14 antigen of Schistosoma mansoni protects mice from cercarial challenge. Infect Immun 2004; 72:3336-43. [PMID: 15155638 PMCID: PMC415698 DOI: 10.1128/iai.72.6.3336-3343.2004] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Sm14 antigen of Schistosoma mansoni was cloned and expressed in Mycobacterium bovis BCG as a fusion with the Mycobacterium fortuitum beta-lactamase protein under the control of its promoter, pBlaF*; the protein was localized in the bacterial cell wall. The rBCG-Sm14 strain was shown to be relatively stable in cultured murine and bovine monocytes in terms of infectivity, bacterial persistence, and plasmid stability. The immunization of mice with rBCG-Sm14 showed no induction of anti-Sm14 antibodies; however, splenocytes of immunized mice released increased levels of gamma interferon upon stimulation with recombinant Sm14 (rSm14), indicating an induction of a Th1-predominant cellular response against Sm14. Mice immunized with one or two doses of rBCG-Sm14 and challenged with live S. mansoni cercaria showed a 48% reduction in worm burden, which was comparable to that obtained by immunization with three doses of rSm14 purified from Escherichia coli. The data presented here further enhance the status of Sm14 as a promising candidate antigen for the control of schistosomiasis and indicate that a one-dose regimen of rBCG-Sm14 could be considered a convenient means to overcome many of the practical problems associated with the successful implementation of a multiple-dose vaccine schedule in developing countries.
Collapse
Affiliation(s)
- Paula B Varaldo
- Centro de Biotecnologia, Instituto Butantan, São Paulo SP, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Castañón-Arreola M, López-Vidal Y. A second-generation anti TB vaccine is long overdue. Ann Clin Microbiol Antimicrob 2004; 3:10. [PMID: 15176980 PMCID: PMC446207 DOI: 10.1186/1476-0711-3-10] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2004] [Accepted: 06/03/2004] [Indexed: 11/24/2022] Open
Abstract
Mycobacterium bovis BCG vaccine significantly reduces the risk of tuberculosis by 50% and continues to be used to prevent tuberculosis around the world. However, it has been shown to be ineffective in some geographical regions. The existence of different BCG strains was described more than 60 years ago, these vary in their antigenic content but the genetic mutations in BCG strains have yet been shown to affect their protection. After the declaration of tuberculosis as a global emergency in 1993, current research attempts to develop a novel more-effective vaccine. Using new technologies, recombinant, auxotroph, DNA, subunit and phylogenetically closely related mycobacteria, naturally or genetically attenuated, have been used as vaccines in animal models, but their protective efficacy, is less than that offered by the current BCG vaccine. Today it is mandatory that a major effort be made to understand how different BCG vaccine strains influence immune response and why in some cases vaccines have failed, so we can rationally develop the next generation of tuberculosis vaccines to reduce the prevalence from 10% to less than 2 % for developed countries.
Collapse
Affiliation(s)
- Mauricio Castañón-Arreola
- Programa de Inmunología Molecular Microbiana, Department of Microbiology and Parasitology, Faculty of Medicine, Universidad Nacional Autonoma de México (UNAM), Mexico City, Mexico
| | - Yolanda López-Vidal
- Programa de Inmunología Molecular Microbiana, Department of Microbiology and Parasitology, Faculty of Medicine, Universidad Nacional Autonoma de México (UNAM), Mexico City, Mexico
| |
Collapse
|
49
|
Bastos RG, Dellagostin OA, Barletta RG, Doster AR, Nelson E, Zuckermann F, Osorio FA. Immune response of pigs inoculated with Mycobacterium bovis BCG expressing a truncated form of GP5 and M protein of porcine reproductive and respiratory syndrome virus. Vaccine 2004; 22:467-74. [PMID: 14670329 DOI: 10.1016/s0264-410x(03)00572-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Pigs were immunised with recombinant BCG (rBCG) expressing a truncated form of GP5 (lacking the first 30 NH(2)-terminal residues) (rBCGGP5) and M protein (rBCGM) of porcine reproductive and respiratory syndrome virus (PRRSV). At 30 days post-inoculation (dpi), pigs inoculated with rBCGGP5 and rBCGM developed a specific humoral immune response against the viral proteins, as detected by commercial ELISA and Western blot tests, and at 60 dpi, three out of five animals developed neutralizing antibodies with titers ranging from 1:4 to 1:8. At 67 dpi, an IFN-gamma response against BCG antigens, but not against the viral proteins, was detected by ELISPOT in inoculated pigs. Following challenge with a pathogenic strain of PRRSV, pigs inoculated with rBCG showed lower (P<0.05) temperature, viremia and virus load in bronchial lymph nodes than control animals, suggesting the establishment of partial protection against PRRSV infection.
Collapse
Affiliation(s)
- Reginaldo G Bastos
- Department of Veterinary and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Dhar N, Rao V, Tyagi AK. Immunogenicity of recombinant BCG vaccine strains overexpressing components of the antigen 85 complex of Mycobacterium tuberculosis. Med Microbiol Immunol 2004; 193:19-25. [PMID: 12905016 DOI: 10.1007/s00430-002-0170-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2002] [Indexed: 11/29/2022]
Abstract
The components of antigen 85 complex of Mycobacterium tuberculosis (Ag 85A, Ag 85B and Ag 85C), due to their immunodominant and secretory nature, represent promising protective antigen candidates and have been used in numerous vaccine preparations. We have used recombinant Bacille Calmette Guerin (BCG) strains overexpressing Ag 85A and Ag 85C to immunize BALB/c mice to investigate the immunogenicity of these strains. Mice immunized with recombinant BCG strains exhibited an increased humoral immune response when compared to mice immunized with wild-type BCG. The recombinant BCG strain overexpressing Ag 85A also induced an increased Th1-like response, characterized by elevated levels of IFN-gamma in antigen stimulated splenocyte cultures and a strong IgG2a antibody response, when compared to wild-type BCG. Immunization with recombinant BCG strain overexpressing Ag 85C, on the other hand did not elicit increased IFN-gamma secretion on restimulation of splenocytes in vitro.
Collapse
Affiliation(s)
- Neeraj Dhar
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, New Delhi, 110021 India
| | | | | |
Collapse
|