1
|
Wang H, Clapp B, Hoffman C, Yang X, Pascual DW. A Single Nasal Dose Vaccination with a Brucella abortus Mutant Potently Protects against Pulmonary Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1576-1588. [PMID: 37036290 PMCID: PMC10159994 DOI: 10.4049/jimmunol.2300071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 03/14/2023] [Indexed: 04/11/2023]
Abstract
The Brucella abortus double-mutant (ΔznuA ΔnorD Brucella abortus-lacZ [znBAZ]) was assessed for its protective efficacy after vaccination with a single nasal dose. Superior protection was achieved in znBAZ-vaccinated mice against pulmonary, wild-type B. abortus 2308 challenge when compared with conventional livestock Brucella abortus vaccines, the smooth S19 (smooth B. abortus strain 19 vaccine) and rough RB51 (rough mutant vaccine strain of B. abortus) strains. Nasal znBAZ vaccination reduced splenic and lung colonization by wild-type brucellae by >3-4 logs. In contrast, S19 reduced lung colonization by only 32-fold, and RB51 failed to reduce colonization. One profound attribute of znBAZ vaccination was the >3-fold increase in pulmonary CD8+ T cells when compared with other vaccinated groups. S19 vaccination increased only CD4+ T cells. All vaccines induced IFN-γ and TNF-α production by CD4+ T cells, but only znBAZ vaccination enhanced the recruitment of polyfunctional CD8+ T cells, by >100-fold. IL-17 by both CD4+ and CD8+ T cells was also induced by subsequent znBAZ vaccination. These results demonstrate that, in addition to achieving protective immunity by CD4+ T cells, CD8+ T cells, specifically resident memory T cells, also confer protection against brucellosis. The protection obtained by znBAZ vaccination was attributed to IFN-γ-producing CD8+ T cells, because depletion of CD8+ T cells throughout vaccination and challenge phases abrogated protection. The stimulation of only CD4+ T cells by RB51- and S19-vaccinated mice proved insufficient in protecting against pulmonary B. abortus 2308 challenge. Thus, nasal znBAZ vaccination offers an alternative means to elicit protection against brucellosis.
Collapse
Affiliation(s)
- Hongbin Wang
- Department of Infectious Diseases and Immunology, University of Florida, Gainesville, FL
| | - Beata Clapp
- Department of Infectious Diseases and Immunology, University of Florida, Gainesville, FL
| | - Carol Hoffman
- Department of Infectious Diseases and Immunology, University of Florida, Gainesville, FL
| | - Xinghong Yang
- Department of Infectious Diseases and Immunology, University of Florida, Gainesville, FL
| | - David W. Pascual
- Department of Infectious Diseases and Immunology, University of Florida, Gainesville, FL
| |
Collapse
|
2
|
Ali A, Waris A, Khan MA, Asim M, Khan AU, Khan S, Zeb J. Recent advancement, immune responses, and mechanism of action of various vaccines against intracellular bacterial infections. Life Sci 2023; 314:121332. [PMID: 36584914 DOI: 10.1016/j.lfs.2022.121332] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 12/20/2022] [Accepted: 12/23/2022] [Indexed: 12/29/2022]
Abstract
Emerging and re-emerging bacterial infections are a serious threat to human and animal health. Extracellular bacteria are free-living, while facultative intracellular bacteria replicate inside eukaryotic host cells. Many serious human illnesses are now known to be caused by intracellular bacteria such as Salmonella enterica, Escherichia coli, Staphylococcus aureus, Rickettsia massiliae, Chlamydia species, Brucella abortus, Mycobacterium tuberculosis and Listeria monocytogenes, which result in substantial morbidity and mortality. Pathogens like Mycobacterium, Brucella, MRSA, Shigella, Listeria, and Salmonella can infiltrate and persist in mammalian host cells, particularly macrophages, where they proliferate and establish a repository, resulting in chronic and recurrent infections. The current treatment for these bacteria involves the application of narrow-spectrum antibiotics. FDA-approved vaccines against obligate intracellular bacterial infections are lacking. The development of vaccines against intracellular pathogenic bacteria are more difficult because host defense against these bacteria requires the activation of the cell-mediated pathway of the immune system, such as CD8+ T and CD4+ T. However, different types of vaccines, including live, attenuated, subunit, killed whole cell, nano-based and DNA vaccines are currently in clinical trials. Substantial development has been made in various vaccine strategies against intracellular pathogenic bacteria. This review focuses on the mechanism of intracellular bacterial infection, host immune response, and recent advancements in vaccine development strategies against various obligate intracellular bacterial infections.
Collapse
Affiliation(s)
- Asmat Ali
- Department of Biotechnology and Genetic Engineering, Hazara University Mansehra, Pakistan
| | - Abdul Waris
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong.
| | - Muhammad Ajmal Khan
- Division of Life Sciences, Center for Cancer Research and State Key Laboratory of Molecular Neurosciences, The Hong Kong University of Science and Technology, Hong Kong
| | - Muhammad Asim
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong
| | - Atta Ullah Khan
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, China
| | - Sahrish Khan
- Department of Biotechnology, Quaid-i-Azam University, Islamabad, Pakistan
| | - Jehan Zeb
- Department of Infectious Diseases and Public Health, City University of Hong Kong, Hong Kong
| |
Collapse
|
3
|
Nasal vaccination stimulates CD8(+) T cells for potent protection against mucosal Brucella melitensis challenge. Immunol Cell Biol 2016; 94:496-508. [PMID: 26752510 PMCID: PMC4879022 DOI: 10.1038/icb.2016.5] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 12/10/2015] [Accepted: 01/05/2016] [Indexed: 12/31/2022]
Abstract
Brucellosis remains a significant zoonotic threat worldwide. Humans and animals acquire infection via their oropharynx and upper respiratory tract following oral or aerosol exposure. After mucosal infection, brucellosis develops into a systemic disease. Mucosal vaccination could offer a viable alternative to conventional injection practices to deter disease. Using a nasal vaccination approach, the ΔznuA B. melitensis was found to confer potent protection against pulmonary Brucella challenge, and reduce colonization of spleens and lungs by more than 2500-fold, with more than 50% of vaccinated mice showing no detectable brucellae. Furthermore, tenfold more brucellae-specific, IFN-γ-producing CD8+ T cells than CD4+ T cells were induced in the spleen and respiratory lymph nodes. Evaluation of pulmonary and splenic CD8+ T cells from mice vaccinated with ΔznuA B. melitensis revealed that these expressed an activated effector memory (CD44hiCD62LloCCR7lo) T cells producing elevated levels of IFN-γ, TNF-α, perforin, and granzyme B. To assess the relative importance of these increased numbers of CD8+ T cells, CD8−/− mice were challenged with virulent B. melitensis, and they showed markedly increased bacterial loads in organs in contrast to similarly challenged CD4−/− mice. Only ΔznuA B. melitensis- and Rev-1-vaccinated CD4−/− and wild-type mice, not CD8−/− mice, were completely protected against Brucella challenge. Determination of cytokines responsible for conferring protection showed the relative importance of IFN-γ, but not IL-17. Unlike wild-type mice, IL-17 was greatly induced in IFN-γ−/− mice, but IL-17 could not substitute for IFN-γ’s protection, although an increase in brucellae dissemination was observed upon in vivo IL-17 neutralization. These results show that nasal ΔznuA B. melitensis vaccination represents an attractive means to stimulate systemic and mucosal immune protection via CD8+ T cell engagement.
Collapse
|
4
|
Brucella abortusΔcydCΔcydD and ΔcydCΔpurD double-mutants are highly attenuated and confer long-term protective immunity against virulent Brucella abortus. Vaccine 2015; 34:237-244. [PMID: 26616550 DOI: 10.1016/j.vaccine.2015.11.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 11/04/2015] [Accepted: 11/13/2015] [Indexed: 11/24/2022]
Abstract
We constructed double deletion (ΔcydCΔcydD and ΔcydCΔpurD) mutants from virulent Brucella abortus biovar 1 field isolate (BA15) by deleting the genes encoding an ATP-binding cassette-type transporter (cydC and cydD genes) and a phosphoribosylamine-glycine ligase (purD). Both BA15ΔcydCΔcydD and BA15ΔcydCΔpurD double-mutants exhibited significant attenuation of virulence when assayed in murine macrophages or in BALB/c mice. Both double-mutants were readily cleared from spleens by 4 weeks post-inoculation even when inoculated at the dose of 10(8) CFU per mouse. Moreover, the inoculated mice showed no splenomegaly, which indicates that the mutants are highly attenuated. Importantly, the attenuation of in vitro and in vivo growth did not impair the ability of these mutants to confer long-term protective immunity in mice against challenge with B. abortus strain 2308. Vaccination of mice with either mutant induced humoral and cell-mediated immune responses, and provided significantly better protection than commercial B. abortus strain RB51 vaccine. These results suggest that highly attenuated BA15ΔcydCΔcydD and BA15ΔcydCΔpurD mutants can be used effectively as potential live vaccine candidates against bovine brucellosis.
Collapse
|
5
|
Afley P, Dohre SK, Prasad GBKS, Kumar S. Prediction of T cell epitopes of Brucella abortus and evaluation of their protective role in mice. Appl Microbiol Biotechnol 2015; 99:7625-37. [PMID: 26150246 DOI: 10.1007/s00253-015-6787-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Revised: 06/17/2015] [Accepted: 06/19/2015] [Indexed: 01/18/2023]
Abstract
Brucellae are Gram-negative intracellular bacteria that cause an important zoonotic disease called brucellosis. The animal vaccines are available but have disadvantage of causing abortions in a proportion of pregnant animals. The animal vaccines are also pathogenic to humans. Recent trend in vaccine design has shifted to epitope-based vaccines that are safe and specific. In this study, efforts were made to identify MHC-I- and MHC-II-restricted T cell epitopes of Brucella abortus and evaluate their vaccine potential in mice. The peptides were designed using online available immunoinformatics tools, and five MHC-I- and one MHC-II-restricted T cell peptides were selected on the basis of their ability to produce interferon gamma (IFN-γ) in in vivo studies. The selected peptides were co-administered with poly DL-lactide-co-glycolide (PLG) microparticles and evaluated for immunogenicity and protection in BALB/c mice. Mice immunized with peptides either entrapped in PLG microparticles (EPLG-Pep) or adsorbed on PLG particles (APLG-Pep) showed significantly higher splenocyte proliferation and IFN-γ generation to all selected peptides than the mice immunized with corresponding irrelevant peptides formulated PLG microparticles or phosphate-buffered saline (PBS). A significant protection compared to PBS control was also observed in EPLG-Pep and APLG-Pep groups. A plasmid DNA vaccine construct (pVaxPep) for peptides encoding DNA sequences was generated and injected to mice by in vivo electroporation. Significant protection was observed (1.66 protection units) when compared with PBS and empty vector control group animals. Overall, the MHC-I and MHC-II peptides identified in this study are immunogenic and protective in mouse model and support the feasibility of peptide-based vaccine for brucellosis.
Collapse
Affiliation(s)
- Prachiti Afley
- Defence Research & Development Establishment, Jhansi Road, Gwalior, Madhya Pradesh, 474002, India
| | | | | | | |
Collapse
|
6
|
Amani J, Ghasemi A, Ranjbar R, Shabani M, Zandemami M, Golmohammadi R. Immune reactivity of sera obtained from brucellosis patients and vaccinated-rabbits to a fusion protein from Brucella melitensis. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2015; 18:350-5. [PMID: 26019797 PMCID: PMC4439449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Accepted: 10/29/2014] [Indexed: 11/10/2022]
Abstract
OBJECTIVES Brucella spp. are facultative intracellular pathogens which can stay alive and multiply in professional and nonprofessional phagocytes. Immunity against Brucella melitensis involves antigen-specific CD4(+) and CD8(+) T-cells activation and humoral immune responses. Due to negative aspects of live attenuated vaccines, much attention has been focused on finding Brucella-protective antigens to introduce them as potential subunit vaccine candidates. MATERIALS AND METHODS A chimeric gene encoding trigger factor (TF), Omp31(48-74) and BP26(87-111) fragments (TOB) from B. melitensis was successfully cloned, expressed in Escherichia coli BL21-DE3 and purified by Ni-NTA agarose column. Antibodies to recombinant TOB (rTOB) have been investigated in Brucella-infected human sera and a pool serum prepared from B. melitensis-vaccinated rabbits. RESULTS Our results showed that the immunized rabbit pool serum strongly reacted with rTOB. In addition, antibodies against rTOB were detectable in 76.5% of sera obtained from infected patients. CONCLUSION These findings suggest that rTOB may provide a potential immunogenic candidate which could be considered in future vaccine studies.
Collapse
Affiliation(s)
- Jafar Amani
- Molecular Biology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Amir Ghasemi
- Applied Microbiology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran,*Corresponding author: Amir Ghasemi. Applied Microbiology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran. Vanak Sq. Molasadra St. Tehran- Iran. Tel: +98-21-82482568; Fax: +98-21-88068924;
| | - Reza Ranjbar
- Molecular Biology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mahdi Shabani
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Mahdi Zandemami
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Reza Golmohammadi
- Molecular Biology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Ghasemi A, Zarnani AH, Ghoodjani A, Rezania S, Salari MH, Jeddi-Tehrani M. Identification of a new immunogenic candidate conferring protection against Brucella melitensis infection in Mice. Mol Immunol 2014; 62:142-9. [DOI: 10.1016/j.molimm.2014.06.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 05/19/2014] [Accepted: 06/11/2014] [Indexed: 01/18/2023]
|
8
|
CD8 knockout mice are protected from challenge by vaccination with WR201, a live attenuated mutant of Brucella melitensis. Clin Dev Immunol 2013; 2013:686919. [PMID: 24288554 PMCID: PMC3830850 DOI: 10.1155/2013/686919] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 09/09/2013] [Indexed: 01/18/2023]
Abstract
CD8+ T cells have been reported to play an important role in defense against B. abortus infection in mouse models. In the present report, we use CD8 knockout mice to further elucidate the role of these cells in protection from B. melitensis infection. Mice were immunized orally by administration of B. melitensis WR201, a purine auxotrophic attenuated vaccine strain, then challenged intranasally with B. melitensis 16M. In some experiments, persistence of WR201 in the spleens of CD8 knockout mice was slightly longer than that in the spleens of normal mice. However, development of anti-LPS serum antibody, antigen-induced production of γ-interferon (IFN-γ) by immune splenic lymphocytes, protection against intranasal challenge, and recovery of nonimmunized animals from intranasal challenge were similar between normal and knockout animals. Further, primary Brucella infection was not exacerbated in perforin knockout and Fas-deficient mice and these animals' anti-Brucella immune responses were indistinguishable from those of normal mice. These results indicate that CD8+ T cells do not play an essential role as either cytotoxic cells or IFN-γ producers, yet they do participate in a specific immune response to immunization and challenge in this murine model of B. melitensis infection.
Collapse
|
9
|
Motaharinia Y, Rezaee MA, Rashidi A, Jalili A, Rezaie MJ, Shapouri R, Hossieni W, Rahmani MR. Induction of protective immunity against brucellosis in mice by vaccination with a combination of naloxone, alum, and heat-killed Brucella melitensis 16 M. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2012; 46:253-8. [PMID: 22727892 DOI: 10.1016/j.jmii.2012.03.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Revised: 02/29/2012] [Accepted: 03/16/2012] [Indexed: 02/07/2023]
Abstract
BACKGROUND/PURPOSE A T-helper cell type 1-specific response leads to the elimination of intracellular infection with Brucella. Studies have shown that naloxone (NLX) can promote a cellular immune response in this respect. The current study was carried out to evaluate the induction of protective immunity in mice against brucellosis by vaccination with a combination of NLX, alum, and heat-killed Brucella melitensis 16 M (HKB). METHODS Mice were categorized into five groups and received intraperitoneal vaccination on Days 0 and 7. Then serum levels of interferon (IFN)-γ and interleukin (IL)-4, the bacterial load, and the survival rate were measured 2 weeks after the last vaccination. RESULTS The serum levels of IFN-γ, IL-4, and immunoglobulin G in the NLX + alum + HKB group were shown to be significantly increased (p < 0.05). Furthermore, the lowest bacterial load was observed in this group. The survival rate in groups vaccinated with combinations containing adjuvants was 100%. CONCLUSION The combination of NLX and alum enhanced the immunogenicity of HKB, which can be used in the vaccination of animals and humans at risk of the disease.
Collapse
Affiliation(s)
- Yousef Motaharinia
- Department of Immunology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Protective live oral brucellosis vaccines stimulate Th1 and th17 cell responses. Infect Immun 2011; 79:4165-74. [PMID: 21768283 DOI: 10.1128/iai.05080-11] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Zoonotic transmission of brucellosis often results from exposure to Brucella-infected livestock, feral animals, or wildlife or frequently via consumption of unpasteurized milk products or raw meat. Since natural infection of humans often occurs by the oral route, mucosal vaccination may offer a means to confer protection for both mucosal and systemic tissues. Significant efforts have focused on developing a live brucellosis vaccine, and deletion of the znuA gene involved in zinc transport has been found to attenuate Brucella abortus. A similar mutation has been adapted for Brucella melitensis and tested to determine whether oral administration of ΔznuA B. melitensis can confer protection against nasal B. melitensis challenge. A single oral vaccination with ΔznuA B. melitensis rapidly cleared from mice within 2 weeks and effectively protected mice upon nasal challenge with wild-type B. melitensis 16M. In 83% of the vaccinated mice, no detectable brucellae were found in their spleens, unlike with phosphate-buffered saline (PBS)-dosed mice, and vaccination also enhanced the clearance of brucellae from the lungs. Moreover, vaccinated gamma interferon-deficient (IFN-γ(-/-)) mice also showed protection in both spleens and lungs, albeit protection that was not as effective as in immunocompetent mice. Although IFN-γ, interleukin 17 (IL-17), and IL-22 were stimulated by these live vaccines, only RB51-mediated protection was codependent upon IL-17 in BALB/c mice. These data suggest that oral immunization with the live, attenuated ΔznuA B. melitensis vaccine provides an attractive strategy to protect against inhalational infection with virulent B. melitensis.
Collapse
|
11
|
Nikolich MP, Warren RL, Lindler LE, Izadjoo MJ, Hoover DL. Attenuation of defined Brucella melitensis wboA mutants. Vaccine 2010; 28 Suppl 5:F12-6. [DOI: 10.1016/j.vaccine.2010.03.058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2010] [Accepted: 03/19/2010] [Indexed: 11/15/2022]
|
12
|
Fetherston JD, Kirillina O, Bobrov AG, Paulley JT, Perry RD. The yersiniabactin transport system is critical for the pathogenesis of bubonic and pneumonic plague. Infect Immun 2010; 78:2045-52. [PMID: 20160020 PMCID: PMC2863531 DOI: 10.1128/iai.01236-09] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2009] [Revised: 11/30/2009] [Accepted: 02/04/2010] [Indexed: 01/18/2023] Open
Abstract
Iron acquisition from the host is an important step in the pathogenic process. While Yersinia pestis has multiple iron transporters, the yersiniabactin (Ybt) siderophore-dependent system plays a major role in iron acquisition in vitro and in vivo. In this study, we determined that the Ybt system is required for the use of iron bound by transferrin and lactoferrin and examined the importance of the Ybt system for virulence in mouse models of bubonic and pneumonic plague. Y. pestis mutants unable to either transport Ybt or synthesize the siderophore were both essentially avirulent via subcutaneous injection (bubonic plague model). Surprisingly, via intranasal instillation (pneumonic plague model), we saw a difference in the virulence of Ybt biosynthetic and transport mutants. Ybt biosynthetic mutants displayed an approximately 24-fold-higher 50% lethal dose (LD(50)) than transport mutants. In contrast, under iron-restricted conditions in vitro, a Ybt transport mutant had a more severe growth defect than the Ybt biosynthetic mutant. Finally, a Delta pgm mutant had a greater loss of virulence than the Ybt biosynthetic mutant, indicating that the 102-kb pgm locus encodes a virulence factor, in addition to Ybt, that plays a role in the pathogenesis of pneumonic plague.
Collapse
Affiliation(s)
- Jacqueline D. Fetherston
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, Kentucky 40536-0298
| | - Olga Kirillina
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, Kentucky 40536-0298
| | - Alexander G. Bobrov
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, Kentucky 40536-0298
| | - James T. Paulley
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, Kentucky 40536-0298
| | - Robert D. Perry
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, Kentucky 40536-0298
| |
Collapse
|
13
|
Yang X, Thornburg T, Walters N, Pascual DW. DeltaznuADeltapurE Brucella abortus 2308 mutant as a live vaccine candidate. Vaccine 2010; 28:1069-74. [PMID: 19914192 PMCID: PMC9811401 DOI: 10.1016/j.vaccine.2009.10.113] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2009] [Revised: 10/15/2009] [Accepted: 10/19/2009] [Indexed: 01/18/2023]
Abstract
To create a new, safe brucellosis live vaccine, a double mutant strain was constructed from Brucella abortus 2308. Using the DeltaznuA B. abortus 2308 mutant, a second mutation was introduced by deleting purE gene. The DeltaznuA DeltapurE B. abortus 2308 strain was less capable of surviving in macrophages. When evaluated in vivo, it was cleared within 8 weeks (wks) from mice, causing significantly less inflammation than spleens obtained from wild-type B. abortus 2308-infected mice. Furthermore, two doses of DeltaznuA DeltapurE B. abortus 2308 conferred 0.79 log protection, similar to S19 as did a single dose of DeltaznuA B. abortus 2308. Thus, this study shows the DeltaznuA DeltapurE B. abortus 2308 strain to be a potential livestock vaccine candidate.
Collapse
Affiliation(s)
| | | | | | - David W. Pascual
- Corresponding author at: Veterinary Molecular Biology, Molecular Biosciences Building, 960 Technology Blvd., Bozeman, MT 59718-4000, United States. Tel.: +1 406 994 6244; fax: +1 406 994 4303. (D.W. Pascual)
| |
Collapse
|
14
|
Lu S, Wang S. Technical transformation of biodefense vaccines. Vaccine 2009; 27 Suppl 4:D8-D15. [PMID: 19837293 DOI: 10.1016/j.vaccine.2009.08.055] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2009] [Revised: 08/14/2009] [Accepted: 08/17/2009] [Indexed: 01/08/2023]
Abstract
Biodefense vaccines are developed against a diverse group of pathogens. Vaccines were developed for some of these pathogens a long time ago but they are facing new challenges to move beyond the old manufacturing technologies. New vaccines to be developed against other pathogens have to determine whether to follow traditional vaccination strategies or to seek new approaches. Advances in basic immunology and recombinant DNA technology have fundamentally transformed the process of formulating a vaccine concept, optimizing protective antigens, and selecting the most effective vaccine delivery approach for candidate biodefense vaccines.
Collapse
Affiliation(s)
- Shan Lu
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| | | |
Collapse
|
15
|
Izadjoo MJ, Mense MG, Bhattacharjee AK, Hadfield TL, Crawford RM, Hoover DL. A Study on the Use of Male Animal Models for Developing a Live Vaccine for Brucellosis. Transbound Emerg Dis 2008; 55:145-51. [DOI: 10.1111/j.1865-1682.2008.01019.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
16
|
ODBILEG R, PUREVTSEREN B, GANTSETSEG D, BOLDBAATAR B, BUYANNEMEKH T, GALMANDAKH Z, ERDENEBAATAR J, KONNAI S, ONUMA M, OHASHI K. Cytokine Responses in Camels (Camelus bactrianus) Vaccinated with Brucella abortus strain 19 Vaccine. J Vet Med Sci 2008; 70:197-201. [DOI: 10.1292/jvms.70.197] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Raadan ODBILEG
- Department of Disease Control, Graduate School of Veterinary Medicine, Hokkaido University
| | | | - Dorj GANTSETSEG
- Animal Disease Control Section, State Central Veterinary Sanitary and Diagnostic Laboratory
| | | | | | - Zagd GALMANDAKH
- Immunological Research Center, Institute of Veterinary Medicine
| | | | - Satoru KONNAI
- Department of Disease Control, Graduate School of Veterinary Medicine, Hokkaido University
| | - Misao ONUMA
- Department of Disease Control, Graduate School of Veterinary Medicine, Hokkaido University
| | - Kazuhiko OHASHI
- Department of Disease Control, Graduate School of Veterinary Medicine, Hokkaido University
| |
Collapse
|
17
|
Kahl-McDonagh MM, Arenas-Gamboa AM, Ficht TA. Aerosol infection of BALB/c mice with Brucella melitensis and Brucella abortus and protective efficacy against aerosol challenge. Infect Immun 2007; 75:4923-32. [PMID: 17664263 PMCID: PMC2044518 DOI: 10.1128/iai.00451-07] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Brucellosis is a zoonotic disease with a worldwide distribution that can be transmitted via intentional or accidental aerosol exposure. In order to engineer superior vaccine strains against Brucella species for use in animals as well as in humans, the possibility of challenge infection via aerosol needs to be considered to properly evaluate vaccine efficacy. In this study, we assessed the use of an aerosol chamber to infect deep lung tissue of mice to elicit systemic infections with either Brucella abortus or B. melitensis at various doses. The results reveal that B. abortus causes a chronic infection of lung tissue in BALB/c mice and peripheral organs at low doses. In contrast, B. melitensis infection diminishes more rapidly, and higher infectious doses are required to obtain infection rates in animals similar to those of B. abortus. Whether this difference translates to severity of human infection remains to be elucidated. Despite these differences, unmarked deletion mutants BADeltaasp24 and BMDeltaasp24 consistently confer superior protection to mice against homologous and heterologous aerosol challenge infection and should be considered viable candidates as vaccine strains against brucellosis.
Collapse
Affiliation(s)
- M M Kahl-McDonagh
- Texas A&M University, Department of Pathobiology, College Station, TX 77843-4467, USA
| | | | | |
Collapse
|
18
|
Bhattacharjee AK, Izadjoo MJ, Zollinger WD, Nikolich MP, Hoover DL. Comparison of protective efficacy of subcutaneous versus intranasal immunization of mice with a Brucella melitensis lipopolysaccharide subunit vaccine. Infect Immun 2006; 74:5820-5. [PMID: 16988260 PMCID: PMC1594895 DOI: 10.1128/iai.00331-06] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Groups of mice were immunized either subcutaneously or intranasally with purified Brucella melitensis lipopolysaccharide (LPS) or with LPS as a noncovalent complex with Neisseria meningitidis group B outer membrane protein (LPS-GBOMP). Control mice were inoculated with sterile saline. Two doses of vaccine were given 4 weeks apart. Mice were challenged intranasally with virulent B. melitensis strain 16M 4 weeks after the second dose of vaccine. Sera, spleens, lungs, and livers of mice were harvested 8 weeks after challenge. The bacterial loads in the organs were determined by culture on brucella agar plates. Protective efficacy was determined by comparing the clearance of bacteria from organs of immunized mice with the clearance of bacteria from organs of control mice. At 8 weeks postchallenge there was significant protection from disseminated infection of spleens and livers of mice intranasally immunized with either vaccine compared to infection of control mice (P < 0.01). There was no significant difference in clearance of bacteria from the lungs of immunized mice and control mice. However, mice immunized subcutaneously with either LPS or LPS-GBOMP vaccine showed significant protection against infection of the spleen (P < 0.001), liver (P < 0.001), and lungs (P < 0.05). These results show that intranasal immunization of mice with either vaccine provided significant protection against disseminated infection of the spleen and liver but subcutaneous immunization of mice with the vaccines conferred significant protection against infection of the spleen, liver, and lungs.
Collapse
Affiliation(s)
- Apurba K Bhattacharjee
- Department of Bacterial Diseases, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910-7500, USA.
| | | | | | | | | |
Collapse
|
19
|
Ding XZ, Paulsen IT, Bhattacharjee AK, Nikolich MP, Myers G, Hoover DL. A high efficiency cloning and expression system for proteomic analysis. Proteomics 2006; 6:4038-46. [PMID: 16800024 DOI: 10.1002/pmic.200600047] [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] [Indexed: 11/08/2022]
Abstract
The recent description of the complete genomes of the two most pathogenic species of Brucella opens the way for genome-based analysis of the antigenicity of their proteins. In the present report, we describe a bench-level high-efficiency cloning and expression system (HECES) that allow expression of large numbers of Brucella proteins based on genomic sequence information. Purified proteins are produced with high efficiency in a microarray format conducive to analysis of their sero-reactivity against serum from immunized animals. This method is applicable at either small or large scale of protein processing. While it does not require robotics, the format is amenable to robotic implementation for all aspects of the process and subsequent analysis of protein characteristics. This method will allow selection of new reagents for diagnosis of brucellosis and development of vaccine against Brucella, an important zoonotic disease and biothreat agent.
Collapse
Affiliation(s)
- Xuan Z Ding
- Department of Bacterial Diseases, Division of Communicable Diseases and Immunology, Walter Reed Army Institute of Reseach, Silver Spring, MD 20910-7500, USA.
| | | | | | | | | | | |
Collapse
|
20
|
Paranavitana CM, Zelazowska E, Das R, Izadjoo M, Jett M, Hoover D. Identification of novel genes in the memory response to Brucella infection by cDNA arrays. Mol Cell Probes 2005; 19:341-8. [PMID: 16146685 DOI: 10.1016/j.mcp.2005.06.009] [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: 04/13/2005] [Accepted: 06/15/2005] [Indexed: 10/25/2022]
Abstract
This study investigated memory responses in immune mice spleen cells to brucellosis by gene expression utilizing cDNA micro arrays. Out of a total of 1176 cDNA's 21 genes were differentially regulated in three independent experiments, and generally supported a Th1 type immune response. 10 genes were validated by real time PCR, and 3 genes (CD 86, CD 40 L and CD 132) were also analyzed by Flow Cytometry for surface protein expression. We extended these findings by studying the expression of five selected genes (IRF 1, SOCS 1, IL 2 R, IRF 7, and CXCR 4) in two independent groups of Brucella immunized mice. In this study we show the potential application of utilizing gene arrays to identify and establish new correlates of protection against a cell mediated immune response.
Collapse
Affiliation(s)
- Chrysanthi M Paranavitana
- Department of Bacterial Diseases, Walter Reed Army Institute of Research, Building 503, Forest Glen, Silver Spring, MD 20910, USA.
| | | | | | | | | | | |
Collapse
|
21
|
Ding XZ, Bhattacharjee A, Nikolich MP, Paulsen IT, Myers G, Seshadri R, L Hoover D. Cloning, expression, and purification of Brucella suis outer membrane proteins. Protein Expr Purif 2005; 40:134-41. [PMID: 15721781 DOI: 10.1016/j.pep.2004.12.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2004] [Revised: 12/01/2004] [Indexed: 10/26/2022]
Abstract
Brucella, an aerobic, nonsporeforming, nonmotile Gram-negative coccobacillus, is a NIH/CDC category B bioterror threat agent that causes incapacitating human illness. Medical defense against the bioterror threat posed by Brucella would be strengthened by development of a human vaccine and improved diagnostic tests. Central to advancement of these goals is discovery of bacterial constituents that are immunogenic or antigenic for humans. Outer membrane proteins (OMPs) are particularly attractive for this purpose. In this study, we cloned, expressed, and purified seven predicted OMPs of Brucella suis. The recombinant proteins were fused with 6-His and V5 epitope tags at their C termini to facilitate detection and purification. The B. suis surface genes were PCR synthesized based on their ORF sequences and directly cloned into an entry vector. The recombinant entry constructs were propagated in TOP 10 cells, recombined into a destination vector, pET-DEST42, then transformed into Escherichia coli BL21 cells for IPTG-induced protein expression. The expressed recombinant proteins were confirmed with Western blot analysis using anti-6-His antibody conjugated with alkaline phosphatase. These B. suis OMPs were captured and purified using a HisGrab plate. The purified recombinant proteins were examined for their binding activity with antiserum. Serum derived from a rabbit immunized intramuscularly with dialyzed cell lysate of Brucella rough mutant WRR51. The OMPs were screened using the rabbit antiserum and purified IgG. The results suggested that recombinant B. suis OMPs were successfully cloned, expressed and purified. Some of the expressed OMPs showed high binding activity with immunized rabbit antiserum.
Collapse
Affiliation(s)
- Xuan Z Ding
- Division of CD&I, Department of Bacterial Diseases, Walter Reed Army Institute of Research, Washington, DC, USA.
| | | | | | | | | | | | | |
Collapse
|
22
|
Paranavitana C, Zelazowska E, Izadjoo M, Hoover D. Interferon-gamma associated cytokines and chemokines produced by spleen cells from Brucella-immune mice. Cytokine 2005; 30:86-92. [PMID: 15804600 DOI: 10.1016/j.cyto.2004.12.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2004] [Revised: 11/30/2004] [Accepted: 12/17/2004] [Indexed: 11/22/2022]
Abstract
It is known that interferon (IFN)-gamma plays a critical role in protection against brucellosis. In this study we have investigated several cytokines and chemokines that are associated with IFN-gamma for potential in vitro correlates of protection. We cultured spleen cells in vitro from mice immunized orally with a live, attenuated Brucella melitensis vaccine candidate (WR201) and stimulated these cells with a lysate of B. melitensis. Differential gene expression of several cytokines and chemokines in stimulated spleen cells was analysed by real-time PCR, and secreted proteins were determined by ELISA. Immunized mice produced higher levels of both protein and gene transcripts for IFN-gamma, interleukin (IL)-2, IL-18 and MIP1-alpha. Immunized mice also had elevated gene expression levels for IL12-p40, IL23-p19, IP-10, MIG and MCP-1 when compared to normal mice. In this study we have identified new cytokines and chemokines as potential immune correlates in responses to protection in Brucella-vaccinated mice.
Collapse
Affiliation(s)
- Chrysanthi Paranavitana
- Department of Bacterial Diseases, Walter Reed Army Institute of Research, Bldg 503, Room 3E10, Robert Grant Avenue, Silver Spring, MD 20910, USA.
| | | | | | | |
Collapse
|
23
|
Vemulapalli R, Contreras A, Sanakkayala N, Sriranganathan N, Boyle SM, Schurig GG. Enhanced efficacy of recombinant Brucella abortus RB51 vaccines against B. melitensis infection in mice. Vet Microbiol 2005; 102:237-45. [PMID: 15327798 DOI: 10.1016/j.vetmic.2004.07.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2004] [Revised: 06/21/2004] [Accepted: 07/01/2004] [Indexed: 11/24/2022]
Abstract
Brucella abortus strain RB51 is an attenuated rough strain, currently being used as the official live vaccine for bovine brucellosis in the USA and several other countries. In strain RB51, the wboA gene, encoding a glycosyltransferase required for the O-side chain synthesis, is disrupted by an IS711 element. Recently, we have demonstrated that strain RB51WboA, RB51 complemented with a functional wboA gene, remains rough but expresses low quantities of O-side chain in the cytoplasm. Mice vaccinated with strain RB51WboA develop greatly enhanced resistance against challenge with B. abortus virulent strain 2308. We have also demonstrated that overexpression of Cu/Zn superoxide dismutase (SOD) in strain RB51 (RB51SOD) significantly increases its vaccine efficacy against strain 2308 challenge. In this study, we constructed a new recombinant strain, RB51SOD/WboA, that over expresses SOD with simultaneous expression of O-side chain in the cytoplasm. We tested the vaccine potential of strains RB51SOD, RB51WboA, RB51SOD/WboA against challenge with virulent Brucella melitensis 16M and B. abortus 2308 in mice. In comparison with strain RB51, strain RB51SOD induced better protection against strain 2308, but not strain 16M, challenge. Similar to strain RB51WboA, vaccination with strain RB51SOD/WboA resulted in complete protection of the mice from infection with strain 2308. When challenged with strain 16M, mice vaccinated with either strain RB51WboA or strain RB51SOD/WboA were significantly better protected than those vaccinated with strain RB51 or RB51SOD. These results suggest that strains RB51WboA and RB51SOD/WboA are good vaccine candidates for inducing enhanced protection against B. melitensis infection.
Collapse
Affiliation(s)
- Ramesh Vemulapalli
- Department of Veterinary Pathobiology, School of Veterinary Medicine, Purdue University, 725 Harrison Street, West Lafayette, IN 47907, USA.
| | | | | | | | | | | |
Collapse
|
24
|
Alcantara RB, Read RDA, Valderas MW, Brown TD, Roop RM. Intact purine biosynthesis pathways are required for wild-type virulence of Brucella abortus 2308 in the BALB/c mouse model. Infect Immun 2004; 72:4911-7. [PMID: 15271960 PMCID: PMC470684 DOI: 10.1128/iai.72.8.4911-4917.2004] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Brucella abortus 2308 derivatives with mini-Tn5 insertions in purE, purL, and purD display significant attenuation in the BALB/c mouse model, while isogenic mutants with mini-Tn5 insertions in pheA, trpB, and dagA display little or no attenuation in cultured murine macrophages or mice. These experimental findings confirm the importance of the purine biosynthesis pathways for the survival and replication of the brucellae in host macrophages. In contrast to previous reports, however, these results indicate that exogenous tryptophan and phenylalanine are available for use by the brucellae in the phagosomal compartment.
Collapse
Affiliation(s)
- Rosemarie B Alcantara
- Department of Microbiology and Immunology, East Carolina University School of Medicine, 600 Moye Blvd., Greenville, NC 27858-4354, USA
| | | | | | | | | |
Collapse
|
25
|
Izadjoo MJ, Bhattacharjee AK, Paranavitana CM, Hadfield TL, Hoover DL. Oral vaccination with Brucella melitensis WR201 protects mice against intranasal challenge with virulent Brucella melitensis 16M. Infect Immun 2004; 72:4031-9. [PMID: 15213148 PMCID: PMC427460 DOI: 10.1128/iai.72.7.4031-4039.2004] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human brucellosis can be acquired from infected animal tissues by ingestion, inhalation, or contamination of conjunctiva or traumatized skin by infected animal products. In addition, Brucella is recognized as a biowarfare threat agent. Although a vaccine to protect humans from natural or deliberate infection could be useful, vaccines presently used in animals are unsuitable for human use. We tested orally administered live, attenuated, purine auxotrophic B. melitensis WR201 bacteria for their ability to elicit cellular and humoral immune responses and to protect mice against intranasal challenge with B. melitensis 16M bacteria. Immunized mice made serum antibody to lipopolysaccharide and non-O-polysaccharide antigens. Splenocytes from immunized animals released interleukin-2 and gamma interferon when grown in cultures with Brucella antigens. Immunization led to protection from disseminated infection and enhanced clearance of the challenge inoculum from the lungs. Optimal protection required administration of live bacteria, was related to immunizing dose, and was enhanced by booster immunization. These results establish the usefulness of oral vaccination against respiratory challenge with virulent Brucella and suggest that WR201 should be further investigated as a vaccine to prevent human brucellosis.
Collapse
Affiliation(s)
- Mina J Izadjoo
- Department of Infectious and Parasitic Diseases, Armed Forces Institute of Pathology, Washington, DC 20306-6000, USA.
| | | | | | | | | |
Collapse
|
26
|
Mense MG, Borschel RH, Wilhelmsen CL, Pitt ML, Hoover DL. Pathologic changes associated with brucellosis experimentally induced by aerosol exposure in rhesus macaques ( Macaca mulatta ). Am J Vet Res 2004; 65:644-52. [PMID: 15141886 DOI: 10.2460/ajvr.2004.65.644] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To develop an aerosol exposure method for induction of brucellosis in rhesus macaques (Macaca mulatta). ANIMALS 10 adult rhesus macaques. PROCEDURE 8 rhesus macaques were challenge exposed with 10(2) to 10(5) colony-forming units of Brucella melitensis 16M by use of an aerosol-exposure technique, and 2 served as control animals. All macaques were euthanatized 63 days after challenge exposure. Gross and microscopic lesions, bacterial burden in target organs, and histologic changes in tissues were evaluated. RESULTS Grossly, spleen weights were increased in exposed macaques, compared with spleen weights in control macaques. Histologically, there was inflammation in the liver, kidneys, spleen, testes, and epididymides in exposed macaques. The spleen and lymph nodes had increased numbers of lymphohistiocytic cells. Morphometrically, the spleen also had an increased ratio of white pulp to red pulp. Areas of hepatitis and amount of splenic white pulp increased with increasing exposure dose. CONCLUSIONS AND CLINICAL RELEVANCE Pathologic findings in rhesus macaques after aerosol exposure to B melitensis are similar to those observed in humans with brucellosis. IMPACT FOR HUMAN MEDICINE These results may aid in the development of a vaccine against brucellosis that can be used in humans.
Collapse
Affiliation(s)
- Mark G Mense
- Department of Diagnostic Pathology, Walter Reed Army Institute of Research, Silver Spring, MD 20910-7500, USA
| | | | | | | | | |
Collapse
|
27
|
Bermúdez-Humarán LG, Langella P, Cortes-Perez NG, Gruss A, Tamez-Guerra RS, Oliveira SC, Saucedo-Cardenas O, Montes de Oca-Luna R, Le Loir Y. Intranasal immunization with recombinant Lactococcus lactis secreting murine interleukin-12 enhances antigen-specific Th1 cytokine production. Infect Immun 2003; 71:1887-96. [PMID: 12654805 PMCID: PMC152106 DOI: 10.1128/iai.71.4.1887-1896.2003] [Citation(s) in RCA: 102] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Interleukin-12 (IL-12), a heterodimeric cytokine, plays an important role in cellular immunity to several bacterial, viral, and parasitic infections and has adjuvant activity when it is codelivered with DNA vaccines. IL-12 has also been used with success in cancer immunotherapy treatments. However, systemic IL-12 therapy has been limited by high levels of toxicity. We describe here inducible expression and secretion of IL-12 in the food-grade lactic acid bacterium Lactococcus lactis. IL-12 was expressed as two separate polypeptides (p35-p40) or as a single recombinant polypeptide (scIL-12). The biological activity of IL-12 produced by the recombinant L. lactis strain was confirmed in vitro by its ability to induce gamma interferon (IFN-gamma) production by mouse splenocytes. Local administration of IL-12-producing strains at the intranasal mucosal surface resulted in IFN-gamma production in mice. The activity was greater with the single polypeptide scIL-12. An antigen-specific cellular response (i.e., secretion of Th1 cytokines, IL-2, and IFN-gamma) elicited by a recombinant L. lactis strain displaying a cell wall-anchored human papillomavirus type 16 E7 antigen was dramatically increased by coadministration with an L. lactis strain secreting IL-12 protein. Our data show that IL-12 is produced and secreted in an active form by L. lactis and that the strategy which we describe can be used to enhance an antigen-specific immune response and to stimulate local mucosal immunity.
Collapse
Affiliation(s)
- Luis G Bermúdez-Humarán
- Unité de Recherches Laitières et de Génétique Appliquée, INRA, Domaine de Vilvert, 78352 Jouy en Josas cedex, France
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Rouot B, Alvarez-Martinez MT, Marius C, Menanteau P, Guilloteau L, Boigegrain RA, Zumbihl R, O'Callaghan D, Domke N, Baron C. Production of the type IV secretion system differs among Brucella species as revealed with VirB5- and VirB8-specific antisera. Infect Immun 2003; 71:1075-82. [PMID: 12595417 PMCID: PMC148853 DOI: 10.1128/iai.71.3.1075-1082.2003] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Expression of the virB operon, encoding the type IV secretion system required for Brucella suis virulence, occurred in the acidic phagocytic vacuoles of macrophages and could be induced in minimal medium at acidic pH values. To analyze the production of VirB proteins, polyclonal antisera against B. suis VirB5 and VirB8 were generated. Western blot analysis revealed that VirB5 and VirB8 were detected after 3 h in acidic minimal medium and that the amounts increased after prolonged incubation. Unlike what occurs in the related organism Agrobacterium tumefaciens, the periplasmic sugar binding protein ChvE did not contribute to VirB protein production, and B. suis from which chvE was deleted was fully virulent in a mouse model. Comparative analyses of various Brucella species revealed that in all of them VirB protein production increased under acidic conditions. However, in rich medium at neutral pH, Brucella canis and B. suis, as well as the Brucella abortus- and Brucella melitensis-derived vaccine strains S19, RB51, and Rev.1, produced no VirB proteins or only small amounts of VirB proteins, whereas the parental B. abortus and B. melitensis strains constitutively produced VirB5 and VirB8. Thus, the vaccine strains were still able to induce virB expression under acidic conditions, but the VirB protein production was markedly different from that in the wild-type strains at pH 7. Taken together, the data indicate that VirB protein production and probably expression of the virB operon are not uniformly regulated in different Brucella species. Since VirB proteins were shown to modulate Brucella phagocytosis and intracellular trafficking, the differential regulation of the production of these proteins reported here may provide a clue to explain their role(s) during the infection process.
Collapse
Affiliation(s)
- Bruno Rouot
- INSERM U431, Université de Montpellier 2, 34095 Montpellier Cedex 05, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Ko J, Splitter GA. Molecular host-pathogen interaction in brucellosis: current understanding and future approaches to vaccine development for mice and humans. Clin Microbiol Rev 2003; 16:65-78. [PMID: 12525425 PMCID: PMC145300 DOI: 10.1128/cmr.16.1.65-78.2003] [Citation(s) in RCA: 210] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Brucellosis caused by Brucella spp. is a major zoonotic disease. Control of brucellosis in agricultural animals is a prerequisite for the prevention of this disease in human beings. Recently, Brucella melitensis was declared by the Centers for Disease Control and Prevention to be one of three major bioterrorist agents due to the expense required for the treatment of human brucellosis patients. Also, the economic agricultural loss due to bovine brucellosis emphasizes the financial impact of brucellosis in society. Thus, vaccination might efficiently solve this disease. Currently, B. abortus RB51 and B. melitensis REV.1 are used to immunize cattle and to immunize goats and sheep, respectively, in many countries. However, these genetically undefined strains still induce abortion and persistent infection, raising questions of safety and efficiency. In fact, the REV.1 vaccine is quite virulent and apparently unstable, creating the need for improved vaccines for B. melitensis. In addition, Brucella spp. may or may not provide cross-protection against infection by heterologous Brucella species, hampering the acceleration of vaccine development. This review provides our current understanding of Brucella pathogenesis and host immunity for the development of genetically defined efficient vaccine strains. Additionally, conditions required for an effective Brucella vaccine strain as well as the future research direction needed to investigate Brucella pathogenesis and host immunity are postulated.
Collapse
Affiliation(s)
- Jinkyung Ko
- Laboratory of Cellular and Molecular Immunology, Department of Animal Health and Biomedical Sciences, University of Wisconsin, Madison, Wisconsin 53706, USA
| | | |
Collapse
|
30
|
Abstract
The first effective Brucella vaccine was based on live Brucella abortus strain 19, a laboratory-derived strain attenuated by an unknown process during subculture. This induces reasonable protection against B. abortus, but at the expense of persistent serological responses. A similar problem occurs with the B. melitensis Rev.1 strain that is still the most effective vaccine against caprine and ovine brucellosis. Vaccines based on killed cells of virulent strains administered with adjuvant induced significant protection but also unacceptable levels of antibodies interfering with diagnostic tests. Attempts were made to circumvent this problem by using a live rough strain B. abortus 45/20, but this reverted to virulence in vivo. Use of killed cells of this strain in adjuvant met with moderate success but batch to batch variation in reactogenicity and agglutinogenicity limited application. This problem has been overcome by the development of the rifampicin-resistant mutant B. abortus RB51 strain. This strain has proved safe and effective in the field against bovine brucellosis and exhibits negligible interference with diagnostic serology. Attempts are being made to develop defined rough mutant vaccine strains that would be more effective against B. melitensis and B. suis. Various studies have examined cell-free native and recombinant proteins as candidate protective antigens, with or without adjuvants. Limited success has been obtained with these or with DNA vaccines encoding known protective antigens in experimental models and further work is indicated.
Collapse
Affiliation(s)
- Gerhardt G Schurig
- Center for Molecular Medicine and Infectious Diseases, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA
| | | | | |
Collapse
|
31
|
Bhattacharjee AK, Van de Verg L, Izadjoo MJ, Yuan L, Hadfield TL, Zollinger WD, Hoover DL. Protection of mice against brucellosis by intranasal immunization with Brucella melitensis lipopolysaccharide as a noncovalent complex with Neisseria meningitidis group B outer membrane protein. Infect Immun 2002; 70:3324-9. [PMID: 12065469 PMCID: PMC128042 DOI: 10.1128/iai.70.7.3324-3329.2002] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Intranasal immunization of mice with purified Brucella melitensis lipopolysaccharide (LPS) as a noncovalent complex with Neisseria meningitidis group B outer membrane protein (GBOMP) elicited a high-titer anti-LPS systemic antibody response and a significant mucosal antibody response. The anti-LPS immunoglobulin G (IgG) antibody was predominantly of the IgG1 subtype, although there was some response of the IgG2a, IgG2b, and IgG3 subtypes. The antibody titer remained high for 16 weeks postimmunization. Immunized mice and sham-immunized control mice were challenged intranasally with 10(4) CFU of virulent B. melitensis strain 16 M 4 weeks after the second dose of vaccine. The numbers of bacteria in lungs, livers, and spleens at 3 days, 9 days, and 8 weeks postchallenge were determined. Bacteria were found in lungs of all mice on day 3, but there was no disseminated infection of liver or spleen. By day 9, 40% of the mice had infected spleens and livers. At 8 weeks postchallenge, spleens of 25 of 62 immunized mice were infected, compared to 61 of 62 control mice (P < 0.0001). The livers of 12 of 43 immunized mice were infected, compared to 22 of 36 control mice (P = 0.005). In contrast, the lungs of 26 of 46 immunized mice were still infected, compared to 27 of 44 control mice. The numbers of bacterial CFU in lungs of immunized and control animals were identical. These studies show that intranasal immunization with B. melitensis LPS-GBOMP subunit vaccine significantly protects mice against intranasal challenge with virulent B. melitensis. Vaccination reduces bacterial dissemination to spleen and liver but has no effect on the course of lung infection.
Collapse
Affiliation(s)
- Apurba K Bhattacharjee
- Department of Bacterial Diseases, Walter Reed Army Institute of Research, Washington, D.C. 20307-5100, USA.
| | | | | | | | | | | | | |
Collapse
|
32
|
Campos E, Cravero SL, Delgui L, Mora I, Kahn N, Arese AI, Rossetti OL. Brucella abortus INTA2, a novel strain 19 (Delta)bp26::luc (Delta)bmp18 double mutant lacking drug resistance markers. Vet Microbiol 2002; 87:1-13. [PMID: 12079742 DOI: 10.1016/s0378-1135(02)00016-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Brucella abortus INTA2, a novel mutant strain, was constructed by inactivation of two B. abortus S19 genes: bp26 and bmp18, with the objective of obtaining a mutant strain that could be compatible with a diagnostic test and have less residual virulence than strain 19. The double mutant was constructed by replacing a large section of the bp26 coding region with the luciferase (luc) coding gene, resulting in mutant strain B. abortus M1luc, followed by partial deletion of bmp18 coding sequence. Both genes were inactivated by allelic replacement assisted by sacB counter-selection. Luciferase expression was evaluated and confirmed that it is a valid marker in the construction of mutant strains. When B. abortus INTA2 was inoculated in BALB/c mice, significantly fewer colony forming units (CFUs) were recovered from mice spleens during initial phase of infection. No splenomegaly was observed in strain INTA2-immunized mice at any time suggesting that strain INTA2 has lost some residual virulence of the parental strain. Nevertheless, similar protection levels against virulent challenge were observed in mice immunized with strains INTA2 or S19. Although strain INTA2 would still induce O-side antibodies, it does not express BP26. This would allow differentiation of INTA2-vaccinated animals from animals infected with field strains by measuring anti-BP26 antibodies, either by an agglutination test or ELISA using BP26 as antigen. Altogether these results indicate that B. abortus INTA2 might be a promising vaccine strain against brucellosis.
Collapse
Affiliation(s)
- Eleonora Campos
- Instituto de Biotecnología, Centro de Investigaciones en Ciencias Veterinarias y Agronómicas, P.O. Box 77, 1708 Morón, Buenos Aires, Argentina
| | | | | | | | | | | | | |
Collapse
|
33
|
Pasquali P, Adone R, Gasbarre LC, Pistoia C, Ciuchini F. Mouse cytokine profiles associated with Brucella abortus RB51 vaccination or B. abortus 2308 infection. Infect Immun 2001; 69:6541-4. [PMID: 11553603 PMCID: PMC98794 DOI: 10.1128/iai.69.10.6541-6544.2001] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2001] [Accepted: 07/25/2001] [Indexed: 11/20/2022] Open
Abstract
This study indicated that mice immunized with Brucella abortus RB51 bacteria and subsequently challenged with B. abortus 2308 were protected from reinfection. After vaccination, both Th1 and Th2 cytokine patterns were observed. Of those, the early production of gamma interferon seems to have the prominent role in inducing an immunologically based protection.
Collapse
Affiliation(s)
- P Pasquali
- Laboratory of Veterinary Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy.
| | | | | | | | | |
Collapse
|
34
|
Izadjoo MJ, Polotsky Y, Mense MG, Bhattacharjee AK, Paranavitana CM, Hadfield TL, Hoover DL. Impaired control of Brucella melitensis infection in Rag1-deficient mice. Infect Immun 2000; 68:5314-20. [PMID: 10948160 PMCID: PMC101794 DOI: 10.1128/iai.68.9.5314-5320.2000] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2000] [Accepted: 05/26/2000] [Indexed: 11/20/2022] Open
Abstract
After intranasal inoculation, Brucella melitensis chronically infects the mononuclear phagocyte system in BALB/c mice, but it causes no apparent illness. Adaptive immunity, which can be transferred by either T cells or antibody from immune to naive animals, confers resistance to challenge infection. The role of innate, non-B-, non-T-cell-mediated immunity in control of murine brucellosis, however, is unknown. In the present study, we documented that BALB/c and C57BL/6 mice had a similar course of infection after intranasal administration of 16M, validating the usefulness of the model in the latter mouse strain. We then compared the course of infection in Rag1 knockout mice (C57BL/6 background) (referred to here as RAG-1 mice) which have no B or T cells as a consequence of deletion of Rag1 (recombination-activating gene 1), with infection in normal C57BL/6 animals after intranasal administration of B. melitensis 16M. C57BL/6 mice cleared brucellae from their lungs by 8 to 12 weeks and controlled infection in the liver and spleen at a low level. In contrast, RAG-1 mice failed to reduce the number of bacteria in any of these organs. From 1 to 4 weeks after inoculation, the number of splenic bacteria increased from 2 to 4.5 logs and remained at that level. In contrast to the consistently high numbers of brucellae observed in the spleens, the number of bacteria rose in the livers sampled for up to 20 weeks. Immunohistologic examination at 8 weeks after infection disclosed foci of persistent pneumonia and large amounts of Brucella antigen in macrophages in lung, liver, and spleen in RAG-1, but not C57BL/6, mice. These studies indicate that T- and B-cell-independent immunity can control Brucella infection at a high level in the murine spleen, but not in the liver. Immunity mediated by T and/or B cells is required for clearance of bacteria from spleen and lung and for control of bacterial replication in the liver.
Collapse
Affiliation(s)
- M J Izadjoo
- American Registry of Pathology, Armed Forces Institute of Pathology, Washington, DC 20306-6000, USA.
| | | | | | | | | | | | | |
Collapse
|