1
|
Shin E, Yun JS, Lee YR, Cha HR, Kim SM, Shin SJ, Lee SW, Chung GT, Kim D, Yoo JS, Kim JS, Jeong HS. Efficacy and immunogenicity of rKVAC85B in a BCG prime-boost regimen against H37Rv and HN878 Mycobacterium tuberculosis strains. PLoS One 2025; 20:e0322147. [PMID: 40367100 PMCID: PMC12077692 DOI: 10.1371/journal.pone.0322147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 03/17/2025] [Indexed: 05/16/2025] Open
Abstract
Mycobacterium tuberculosis infection accounted for 1.3 million deaths worldwide in 2022. Bacillus Calmette-Guérin (BCG) is the only licensed vaccine against tuberculosis (TB); however, it has limited protective efficacy in adults. In this study, we constructed a recombinant vaccinia virus expressing Ag85B from M. tuberculosis using a novel attenuated vaccinia virus (KVAC103). We then analyzed the immunogenicity of prime-boost inoculation strategies using recombinant KVAC103 expressing Ag85B (rKVAC85B) compared to BCG. In both rKVAC85B prime-boost and BCG prime-rKVAC85B boost inoculation regimens, rKVAC85B induced the generation of specific immunoglobulin G (IgG) and secretion of interferon-γ by immune cells. In vitro analysis of Mycobacterium growth inhibition revealed a comparable immune-mediated pattern of outcomes. Furthermore, bacterial loads in the lungs were significantly lower in mice inoculated with the BCG prime-rKVAC85B boost than in the BCG-only group following a rechallenge infection with both H37Rv and HN878 strains of M. tuberculosis. These findings collectively suggest that KVAC103, incorporated into a viral vector, is a promising candidate for the development of a novel TB vaccine platform that is effective against multiple M. tuberculosis strains, including H37Rv and HN878, and that rKVAC85B effectively stimulates immune responses against M. tuberculosis infection.
Collapse
Affiliation(s)
- Eunkyung Shin
- National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, CheongJu, Chungbuk, Republic of Korea
| | - Jin-Seung Yun
- National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, CheongJu, Chungbuk, Republic of Korea
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Young-Ran Lee
- Bio-Convergence R&D Division, Korea Institute of Ceramic Engineering and Technology, CheongJu, Chungbuk, Republic of Korea
| | - Hye-Ran Cha
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Graduate School of Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Soo-Min Kim
- National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, CheongJu, Chungbuk, Republic of Korea
| | - Sung-Jae Shin
- Department of Microbiology, College of Medicine, Yonsei University, Seoul, Republic of Korea
| | - Sang-Won Lee
- Department of Data Science, Korea Disease Control and Prevention Agency, CheongJu, Chungbuk, Republic of Korea
| | - Gyung Tae Chung
- National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, CheongJu, Chungbuk, Republic of Korea
| | - Dokeun Kim
- National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, CheongJu, Chungbuk, Republic of Korea
| | - Jung Sik Yoo
- National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, CheongJu, Chungbuk, Republic of Korea
| | - Jong-Seok Kim
- Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon, Republic of Korea
| | - Hye-Sook Jeong
- National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, CheongJu, Chungbuk, Republic of Korea
| |
Collapse
|
2
|
Kanipe C, Putz EJ, Palmer MV. Differential expression of vascular endothelial growth factor A (VEGFA) and M1 macrophage marker nitric oxide synthase 2 (NOS2) in lymph node granulomas of BCG-vaccinated and non-vaccinated cattle infected with Mycobacterium bovis. Tuberculosis (Edinb) 2025; 151:102609. [PMID: 39862443 DOI: 10.1016/j.tube.2025.102609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 01/13/2025] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
Bovine tuberculosis is mainly caused by Mycobacterium bovis. Bacillus Calmette-Guérin (BCG) is an attenuated strain of M. bovis which provides variable disease protection. Lesions have been characterized in infected cattle, but little comparison has been done with lesions which form in BCG-vaccinates. Here, in situ hybridization examined differences in expression of M. bovis RNA, inducible nitric oxide synthase 2, and vascular endothelial growth factor A in relation to vaccination status and granuloma grade, using two different groups of cattle. Data found no differences between vaccination groups or granuloma grade in average copies of M. bovis mRNA per μm2 of total granuloma area or per μm2 of necrotic areas. Within a vaccination group high-grade granulomas had more NOS2 per cell, per μm2 and a higher percentage of cells expressing NOS2 than low-grade granulomas. Non-vaccinates had a higher percentage of cells producing NOS2 than vaccinates. Differences in NOS2 expression varied by group. Vaccination status and granuloma grade did not affect the average copies of VEGFA per cell or the percent of cells expressing RNA, however VEGFA copies per μm2 varied between groups. These findings suggest NOS2 and VEGFA are likely not mechanisms of BCG vaccination protection but may impact disease severity.
Collapse
Affiliation(s)
- C Kanipe
- Infectious Bacterial Diseases Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, Iowa, USA; Immunobiology Program, Iowa State University, Ames, IA, 50010, USA.
| | - E J Putz
- Infectious Bacterial Diseases Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, Iowa, USA
| | - M V Palmer
- Infectious Bacterial Diseases Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, Iowa, USA
| |
Collapse
|
3
|
Díaz-Otero F, Jaramillo-Meza L, Manzo-Sandoval A, Olguín-Alor R, Diosdado-Vargas F. Comparative longitudinal analysis of T lymphocyte subpopulations in calves vaccinated with different doses of BCG-Phipps or with culture filtrate protein extract of Mycobacterium bovis in a natural transmission setting. BMC Vet Res 2025; 21:78. [PMID: 39972321 PMCID: PMC11837346 DOI: 10.1186/s12917-025-04572-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 02/05/2025] [Indexed: 02/21/2025] Open
Abstract
Currently, control programmes for bovine tuberculosis (bTB) contemplate the use of vaccines to reduce disease incidence rates. The BCG vaccine and the culture filtrate protein extract (CFPE) of Mycobacterium bovis are strong candidates for vaccination against bTB. We conducted an analysis of the immune response and evaluated activation and memory markers in CD4+ and CD8+ T-lymphocyte subpopulations in Holstein-Friesian calves immunised with different doses of M. bovis BCG-Phipps vaccine (1×10⁴ and 1×10⁶ CFU) or with CFPE (300 µg and 600 µg) in a natural transmission setting. The study was carried out in a dairy herd, selecting calves aged 1-4 months that tested negative in various bTB diagnostic tests. In the groups immunised with the BCG-Phipps vaccine, gamma interferon (IFN-γ) secretion levels increased significantly, with the highest increase observed in the group that received a dose of 1×10⁶ CFU (P ≤ 0.05). The CD4+/CD8+ ratio increased significantly over time in both vaccinated and unvaccinated groups, with no significant differences between them. However, notable differences were observed in activated (CD25+) and memory (CD45RO+) CD4 and CD8 T-cell populations across different times and treatments. Remarkably, the groups immunised with the BCG vaccine remained free of M. bovis infection, as evidenced by negative IFN-γ results using ESAT-6/CFP-10 antigens and negative PCR test results for bacterial detection. The comparative analysis of the immune response induced by the different doses of the BCG-Phipps and CFPE vaccines revealed that the group of animals vaccinated with the 1×10⁶ CFU dose exhibited greater production of gamma interferon and a higher percentage of CD4+ T cells, as well as activated and memory CD8+ T cells compared to the other vaccinated and control groups in the natural transmission environment.
Collapse
Affiliation(s)
- Fernando Díaz-Otero
- Laboratorio de Inmunología, Centro Nacional de Investigación Disciplinaria en Salud Animal e Inocuidad, Instituto Nacional de Investigaciones Forestales, Agrícolas y Pecuarias, Mexico City, Mexico.
| | - Laura Jaramillo-Meza
- Laboratorio de Bacteriología Experimental, Centro Nacional de Investigación Disciplinaria en Salud Animal e Inocuidad, Instituto Nacional de Investigaciones Forestales, Agrícolas y Pecuarias, Mexico City, Mexico
| | - Anabelle Manzo-Sandoval
- Laboratorio de Inmunología, Centro Nacional de Investigación Disciplinaria en Salud Animal e Inocuidad, Instituto Nacional de Investigaciones Forestales, Agrícolas y Pecuarias, Mexico City, Mexico
| | - Roxana Olguín-Alor
- Laboratorio Nacional de Citometría de Flujo, Instituto de Investigaciones Biomédicas. UNAM, Mexico City, Mexico
| | - Fernando Diosdado-Vargas
- Laboratorio de Virología, Centro Nacional de Investigación Disciplinaria en Salud Animal e Inocuidad, Instituto Nacional de Investigaciones Forestales, Agrícolas y Pecuarias, Mexico City, Mexico
| |
Collapse
|
4
|
Zeng L, Zuo Y, Tang M, Lei C, Li H, Ma X, Ji J, Li H. A subunit vaccine Ag85A-LpqH focusing on humoral immunity provides substantial protection against tuberculosis in mice. iScience 2025; 28:111568. [PMID: 39868033 PMCID: PMC11760819 DOI: 10.1016/j.isci.2024.111568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/04/2024] [Accepted: 12/06/2024] [Indexed: 01/28/2025] Open
Abstract
The importance of humoral immunity in combating TB has gained extensive recognition. In this study, a subunit vaccine named Ag85A-LpqH (AL) was prepared by fusing the antigen Ag85A proved to induce robust T cell immune responses, and LpqH was shown to produce protective antibodies. The prevention and BCG prime-boost mouse models were established to test the vaccine efficacy. The results indicate that Ag85A-LpqH can induce substantial protection by reducing bacterial loads and pathological lesions. This vaccine can induce robust antibody responses, as well as T cell immune responses especially strong CD8+ T cell responses. Moreover, the serum from AL-immunized mice can reduce the bacterial load and lung pathology in mice. B cell receptor (BCR) sequencing revealed a notable rise in BCR diversity among mice immunized with AL. These results indicate that Ag85A-LpqH can be a promising vaccine candidate for tuberculosis prevention and control.
Collapse
Affiliation(s)
- Lingyuan Zeng
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - You Zuo
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Minghui Tang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Chengrui Lei
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Huoming Li
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xiuling Ma
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jiahong Ji
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Hao Li
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
5
|
Franzoni G, Signorelli F, Mazzone P, Donniacuo A, De Matteis G, Grandoni F, Schiavo L, Zinellu S, Dei Giudici S, Bezos J, De Carlo E, Galiero G, Napolitano F, Martucciello A. Cytokines as potential biomarkers for the diagnosis of Mycobacterium bovis infection in Mediterranean buffaloes ( Bubalus bubalis). Front Vet Sci 2024; 11:1512571. [PMID: 39776597 PMCID: PMC11703857 DOI: 10.3389/fvets.2024.1512571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025] Open
Abstract
Mycobacterium bovis (M. bovis) is the primary agent of bovine tuberculosis (TB) in Mediterranean buffalo, which has a negative economic impact on buffalo herds. Improving TB diagnostic performance in this species represents a key step to eradicate efficiently this disease. We have recently shown the utility of the IFN-γ assay in the diagnosis of M. bovis infection in Mediterranean buffaloes (Bubalus bubalis), but other cytokines might be useful immunological biomarkers of this infection. We therefore investigated the utility of key immune cytokines as diagnostic biomarkers of M. bovis infection in this species. Thirty-six Italian Mediterranean buffaloes were used in this study: healthy animals (N = 11), infected (IFN-γ test positive, no post-mortem lesions, no M. bovis detection; N = 14), and affected (IFN-γ test positive, visible post-mortem lesions; N = 11). Heparin blood samples were stimulated with bovine purified protein derivative (PPD-B), alongside controls, and 18-24 h later plasma were collected. Levels of 14 key cytokines were measured: IFN-γ, IL-17, IL-4, IL-10, TNF, IL-1α, IL-1β, IL-6, IP-10, MIP-1α, MIP-1β, MCP-1, IL-36Ra, and VEGF-A. We observed that both infected and affected animals released higher levels of IFN-γ, IL-17, IL-10, TNF, IL-1α, IL-6, MIP-1β, in response to PPD-B compared to healthy subjects. Mycobacterium bovis infected animals released also higher levels of IL-1β and IP-10 in response to PPD-B compared to healthy subjects, whereas only tendencies were detected in affected animals. Affected animals only released MIP-1α in response to PPD-B compared to healthy subjects and in this group higher values of PPD-B specific TNF was also observed. Finally, canonical discriminant analysis (CDA) was used to generate predictive cytokine profiles by groups. Our data suggest that IL-10, TNF, IL-1α, IL-6, MIP-1β could be useful biomarkers of TB in Mediterranean Buffalo and can improve the TB diagnostic performance in this specie.
Collapse
Affiliation(s)
- Giulia Franzoni
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, Italy
| | - Federica Signorelli
- CREA-Consiglio per la Ricerca in Agricoltura e l’Analisi dell’Economia Agraria, Centro di ricerca Zootecnia e Acquacoltura (Research Centre for Animal Production and Aquaculture), Monterotondo (RM), Italy
| | - Piera Mazzone
- Istituto Zooprofilattico Sperimentale dell’Umbria e delle Marche “Togo Rosati”, Perugia, Italy
| | - Anna Donniacuo
- National Reference Centre for Hygiene and Technologies of Mediterranean Buffalo Farming and Productions, Istituto Zooprofilattico Sperimentale del Mezzogiorno, Salerno, Italy
| | - Giovanna De Matteis
- CREA-Consiglio per la Ricerca in Agricoltura e l’Analisi dell’Economia Agraria, Centro di ricerca Zootecnia e Acquacoltura (Research Centre for Animal Production and Aquaculture), Monterotondo (RM), Italy
| | - Francesco Grandoni
- CREA-Consiglio per la Ricerca in Agricoltura e l’Analisi dell’Economia Agraria, Centro di ricerca Zootecnia e Acquacoltura (Research Centre for Animal Production and Aquaculture), Monterotondo (RM), Italy
| | - Lorena Schiavo
- National Reference Centre for Hygiene and Technologies of Mediterranean Buffalo Farming and Productions, Istituto Zooprofilattico Sperimentale del Mezzogiorno, Salerno, Italy
| | - Susanna Zinellu
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, Italy
| | - Silvia Dei Giudici
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, Italy
| | - Javier Bezos
- VISAVET Health Surveillance Centre, Complutense University of Madrid, Madrid, Spain
- Facultad de Veterinaria, Departamento de Sanidad Animal, Universidad Complutense de Madrid, Madrid, Spain
| | - Esterina De Carlo
- National Reference Centre for Hygiene and Technologies of Mediterranean Buffalo Farming and Productions, Istituto Zooprofilattico Sperimentale del Mezzogiorno, Salerno, Italy
| | - Giorgio Galiero
- National Reference Centre for Hygiene and Technologies of Mediterranean Buffalo Farming and Productions, Istituto Zooprofilattico Sperimentale del Mezzogiorno, Salerno, Italy
| | - Francesco Napolitano
- CREA-Consiglio per la Ricerca in Agricoltura e l’Analisi dell’Economia Agraria, Centro di ricerca Zootecnia e Acquacoltura (Research Centre for Animal Production and Aquaculture), Monterotondo (RM), Italy
| | - Alessandra Martucciello
- National Reference Centre for Hygiene and Technologies of Mediterranean Buffalo Farming and Productions, Istituto Zooprofilattico Sperimentale del Mezzogiorno, Salerno, Italy
| |
Collapse
|
6
|
Hendrix EK, Sha J, Kilgore PB, Neil BH, Verma AK, Chopra AK. The Protective Effect of IL-17A in Pneumonic Plague Can Be Compensated by Effective Vaccines and Immunization Strategies in Mice. Vaccines (Basel) 2024; 12:1361. [PMID: 39772023 PMCID: PMC11680114 DOI: 10.3390/vaccines12121361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 11/28/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025] Open
Abstract
Plague, caused by Yersinia pestis, poses a public health threat not only due to sporadic outbreaks across the globe but also due to its potential as a biothreat agent. Ironically, among the seven deadliest pandemics in global history, three were caused by Y. pestis. Pneumonic plague, the more contagious and severe form of the disease, is difficult to contain, requiring either prophylactic antibiotic treatment or vaccination. However, no vaccine (live attenuated or subunit) is currently approved by the Food and Drug Administration, requiring rigorous preclinical studies in different animal models, thus forming the basis of this study. Objectives: The aim of this study was to evaluate the efficacy and immune responses of two live attenuated vaccines (LAVs), LMA and LMP, either alone or in combination with a trivalent adenoviral vector-based vaccine (Ad5-YFV), in IL-17A-depleted and IgG control mice by using an anti-IL-17A monoclonal antibody (mAb) or its matched isotype IgG, respectively. Methods: IL-17A mAb or IgG isotype control was administered to mice twice per week to their respective groups during the course of immunization. Serum, spleens, and broncho-alveolar lavage fluid (BALF) were collected for assessing immunological responses, and another cohort of mice was intranasally challenged with a lethal dose of parental Y. pestis CO92. Results: Robust humoral and cellular immune responses followed by complete protection were observed in all vaccinated animals against highly lethal intranasal challenge doses of parental Y. pestis CO92. Serum IgG titers to YscF and overall mucosal IgA titers to all three antigens of the Ad5-YFV vaccine were significantly lower, with slightly reduced serum LcrV-neutralizing antibodies when IL-17A was depleted compared to IgG control animals during the course of immunization. A remarkable reduction in Th1 (IFNγ or IL-2) and Th17 cell populations was observed in IL-17A-depleted mice compared to IgG controls in response to vaccination. On the other hand, B cell activities in germinal centers, overall activated antigen-specific T cells, and memory B and T cells remained at comparable levels in both vaccinated IL-17A-depleted and IgG control mice. Conclusions: These data demonstrated the effectiveness of our vaccines even under the reduced levels of both Th1 and Th17 responses and thus should be suitable for those individuals associated with certain immune deficiencies.
Collapse
Affiliation(s)
- Emily K. Hendrix
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (E.K.H.)
| | - Jian Sha
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (E.K.H.)
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Paul B. Kilgore
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (E.K.H.)
| | - Blake H. Neil
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (E.K.H.)
| | - Atul K. Verma
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (E.K.H.)
| | - Ashok K. Chopra
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (E.K.H.)
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX 77555, USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX 77555, USA
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555, USA
- Center for Biodefense and Emerging Infectious Diseases, Galveston, TX 77555, USA
| |
Collapse
|
7
|
Colombatti Olivieri MA, Cuerda MX, Moyano RD, Gravisaco MJ, Pinedo MFA, Delgado FO, Calamante G, Mundo S, de la Paz Santangelo M, Romano MI, Alonso MN, Del Medico Zajac MP. Superior protection against paratuberculosis by a heterologous prime-boost immunization in a murine model. Vaccine 2024; 42:126055. [PMID: 38880691 DOI: 10.1016/j.vaccine.2024.06.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 06/03/2024] [Accepted: 06/07/2024] [Indexed: 06/18/2024]
Abstract
Vaccination is the best strategy to control Paratuberculosis (PTB), which is a significant disease in cattle and sheep. Previously we showed the humoral and cellular immune response induced by a novel vaccine candidate against PTB based on the Argentinian Mycobacterium avium subspecies paratuberculosis (Map) 6611 strain. To improve 6611 immunogenicity and efficacy, we evaluated this vaccine candidate in mice with two different adjuvants and a heterologous boost with a recombinant modified vaccinia Ankara virus (MVA) expressing the antigen 85A (MVA85A). We observed that boosting with MVA85A did not improve total IgG or specific isotypes in serum induced by one or two doses of 6611 formulated with incomplete Freund's adjuvant (IFA). However, when 6611 was formulated with ISA201 adjuvant, MVA85A boost enhanced the production of IFNγ, Th1/Th17 cytokines (IL-2, TNF, IL-17A) and IL-6, IL-4 and IL-10. Also, this group showed the highest levels of IgG2b and IgG3 isotypes, both important for better protection against Map infection in the murine model. Finally, the heterologous scheme elicited the highest levels of protection after Map challenge (lowest CFU count and liver lesion score). In conclusion, our results encourage further evaluation of 6611 strain + ISA201 prime and MVA85A boost in bovines.
Collapse
MESH Headings
- Animals
- Mycobacterium avium subsp. paratuberculosis/immunology
- Immunization, Secondary/methods
- Mice
- Paratuberculosis/prevention & control
- Paratuberculosis/immunology
- Immunoglobulin G/blood
- Cytokines/metabolism
- Female
- Antibodies, Bacterial/blood
- Antibodies, Bacterial/immunology
- Adjuvants, Immunologic/administration & dosage
- Disease Models, Animal
- Bacterial Vaccines/immunology
- Bacterial Vaccines/administration & dosage
- Mice, Inbred BALB C
- Vaccinia virus/immunology
- Vaccinia virus/genetics
- Antigens, Bacterial/immunology
- Antigens, Bacterial/genetics
- Immunity, Cellular/immunology
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Freund's Adjuvant/administration & dosage
- Freund's Adjuvant/immunology
Collapse
Affiliation(s)
| | - María Ximena Cuerda
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), INTA-CONICET, Hurlingham, Buenos Aires 1686, Argentina
| | - Roberto Damián Moyano
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), INTA-CONICET, Hurlingham, Buenos Aires 1686, Argentina
| | - María José Gravisaco
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), INTA-CONICET, Hurlingham, Buenos Aires 1686, Argentina
| | - María Fiorella Alvarado Pinedo
- Centro de Diagnóstico e Investigaciones Veterinarias (CEDIVE) de la Facultad de Ciencias Veterinarias - Universidad de La Plata, Chascomús, Buenos Aires 7130, Argentina
| | - Fernando Oscar Delgado
- Instituto de Patobiologia Veterinaria (IPV), INTA-CONICET, Hurlingham, Buenos Aires 1686, Argentina
| | - Gabriela Calamante
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), INTA-CONICET, Hurlingham, Buenos Aires 1686, Argentina
| | - Silvia Mundo
- Cátedra de Inmunología de la Facultad de Ciencias Veterinarias - Universidad de Buenos Aires, Ciudad de Buenos Aires 1427, Argentina
| | - María de la Paz Santangelo
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), INTA-CONICET, Hurlingham, Buenos Aires 1686, Argentina
| | - María Isabel Romano
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), INTA-CONICET, Hurlingham, Buenos Aires 1686, Argentina
| | - María Natalia Alonso
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), INTA-CONICET, Hurlingham, Buenos Aires 1686, Argentina.
| | - María Paula Del Medico Zajac
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), INTA-CONICET, Hurlingham, Buenos Aires 1686, Argentina
| |
Collapse
|
8
|
Holder T, Coad M, Allan G, Hogarth PJ, Vordermeier HM, Jones GJ. Vaccination of calves with Bacillus Calmette-Guerin Danish strain 1331 results in a duration of immunity of at least 52 weeks. Vaccine 2023; 41:7290-7296. [PMID: 37925317 DOI: 10.1016/j.vaccine.2023.10.060] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/20/2023] [Accepted: 10/22/2023] [Indexed: 11/06/2023]
Abstract
Bacillus Calmette-Guérin (BCG) Danish strain 1331 (CattleBCG) is currently the lead vaccine candidate for the control of bovine tuberculosis (TB) in cattle in GB, where prior vaccination has shown to result in a significant reduction in bovine TB pathology induced by infection with Mycobacterium bovis (M. bovis). A critical knowledge gap in our understanding of CattleBCG is the duration of immunity post vaccination at the minimum intended vaccine dose. To this end, we performed an experiment where calves were vaccinated with a targeted dose of 106 CFU and, after a period of 52 weeks, experimentally infected with M. bovis. Post mortem examination performed 13 weeks after infection revealed a statistically significant reduction in the severity of TB pathology in the CattleBCG vaccinated group compared with the unvaccinated control group. Additionally, this study allowed us to further assess the diagnostic performance of a defined antigen DIVA reagent (DST-F) developed to detect infected amongst vaccinated animals. Our results demonstrate that when used in a skin test format, DST-F showed high specificity (100 %) in BCG-vaccinated animals when tested prior to infection, whilst detecting all infected animals when re-tested after infection. Furthermore, we also present results supporting the use of the DST-F reagent in an interferon-gamma release assay. In conclusion, the results of this study demonstrate a 52-week duration of immunity following administration of a minimum dose of CattleBCG. This evidence will be a fundamental component in our efforts to apply for UK marketing authorisation to enable vaccination of cattle as a significant additional control measure in the ongoing fight against bovine TB in GB.
Collapse
Affiliation(s)
- Thomas Holder
- Animal and Plant Health Agency, Bacteriology, Addlestone, United Kingdom
| | - Michael Coad
- Animal and Plant Health Agency, Bacteriology, Addlestone, United Kingdom
| | - Grace Allan
- Animal and Plant Health Agency, Bacteriology, Addlestone, United Kingdom
| | - Phillip J Hogarth
- Animal and Plant Health Agency, Bacteriology, Addlestone, United Kingdom
| | | | - Gareth J Jones
- Animal and Plant Health Agency, Bacteriology, Addlestone, United Kingdom.
| |
Collapse
|
9
|
Hope JC, Khalid H, Thom ML, Howard CJ, Shaw DJ. Protective Efficacy of BCG Vaccination in Calves Vaccinated at Different Ages. Pathogens 2023; 12:789. [PMID: 37375479 DOI: 10.3390/pathogens12060789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 05/30/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Mycobacterium bovis, the causative agent of bovine tuberculosis (bTB), is a globally prevalent pathogen with significant animal welfare, economic and public health impacts. In the UK, the control of bTB relies on detection via tuberculin skin tests with ancillary interferon gamma (IFN-γ) release assays, followed by culling infected animals. Vaccination with Bacille Calmette-Guérin (BCG) could be an important element of bTB control, and a number of studies have demonstrated its protective efficacy, particularly when young calves are vaccinated. Here, we compared immune responses and the protective efficacy of BCG in calves vaccinated within the first day of life and at three weeks of age. Significant protection from M. bovis infection was observed in BCG-vaccinated calves compared to non-vaccinated, age-matched controls. No significant differences were shown between calves vaccinated at one day and at three weeks of age when assessing the protective efficacy of BCG (measured as a reduction in lesions and bacterial burden). Antigen-specific IFN-γ levels were similar between the BCG-vaccinated groups, but significantly different from the non-vaccinated control animals. Antigen-specific IFN-γ expression post-BCG vaccination was correlated significantly with protection from M. bovis infection, whereas IFN-γ levels post-challenge correlated with pathology and bacterial burden. These results indicate that early-life vaccination with BCG could have a significant impact on M. bovis infection and, therefore, bTB incidence, and they demonstrate that age, at least within the first month of life, does not significantly impact the protective effect of vaccination.
Collapse
Affiliation(s)
- Jayne C Hope
- Division of Infection and Immunity, The Roslin Institute, and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
| | - Hamza Khalid
- Division of Infection and Immunity, The Roslin Institute, and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
| | - Michelle L Thom
- Institute for Animal Health, Compton RG20 7NN, UK
- The Pirbright Institute, Pirbright GU24 0NF, UK
| | | | - Darren J Shaw
- Division of Infection and Immunity, The Roslin Institute, and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
| |
Collapse
|
10
|
Safety and Immunogenicity of Recombinant Bacille Calmette-Guérin Strain VPM1002 and Its Derivatives in a Goat Model. Int J Mol Sci 2023; 24:ijms24065509. [PMID: 36982586 PMCID: PMC10058566 DOI: 10.3390/ijms24065509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 03/15/2023] Open
Abstract
A more effective vaccine against tuberculosis than Bacille Calmette-Guérin (BCG) is urgently needed. BCG derived recombinant VPM1002 has been found to be more efficacious and safer than the parental strain in mice models. Newer candidates, such as VPM1002 Δpdx1 (PDX) and VPM1002 ΔnuoG (NUOG), were generated to further improve the safety profile or efficacy of the vaccine. Herein, we assessed the safety and immunogenicity of VPM1002 and its derivatives, PDX and NUOG, in juvenile goats. Vaccination did not affect the goats’ health in regards to clinical/hematological features. However, all three tested vaccine candidates and BCG induced granulomas at the site of injection, with some of the nodules developing ulcerations approximately one month post-vaccination. Viable vaccine strains were cultured from the injection site wounds in a few NUOG- and PDX- vaccinated animals. At necropsy (127 days post-vaccination), BCG, VPM1002, and NUOG, but not PDX, still persisted at the injection granulomas. All strains, apart from NUOG, induced granuloma formation only in the lymph nodes draining the injection site. In one animal, the administered BCG strain was recovered from the mediastinal lymph nodes. Interferon gamma (IFN-γ) release assay showed that VPM1002 and NUOG induced a strong antigen-specific response comparable to that elicited by BCG, while the response to PDX was delayed. Flow cytometry analysis of IFN-γ production by CD4+, CD8+, and γδ T cells showed that CD4+ T cells of VPM1002- and NUOG-vaccinated goats produced more IFN-γ compared to BCG-vaccinated and mock-treated animals. In summary, the subcutaneous application of VPM1002 and NUOG induced anti-tuberculous immunity, while exhibiting a comparable safety profile to BCG in goats.
Collapse
|
11
|
Melgarejo C, Planas C, Cobos A, Arrieta-Villegas C, Sevilla IA, Bezos J, Moll X, Espada Y, Garrido JM, Domingo M, Vidal E, Pérez de Val B. A proof-of-concept study to investigate the efficacy of heat-inactivated autovaccines in Mycobacterium caprae experimentally challenged goats. Sci Rep 2022; 12:22132. [PMID: 36550177 PMCID: PMC9780325 DOI: 10.1038/s41598-022-26683-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
This study aimed to assess the efficacy of a heat-inactivated Mycobacterium caprae (HIMC) vaccine in goats experimentally challenged with the same strain of M. caprae. Twenty-one goats were divided into three groups of seven: vaccinated with heat-inactivated Mycobacterium bovis (HIMB), with HIMC and unvaccinated. At 7 weeks post-vaccination all animals were endobronchially challenged with M. caprae. Blood samples were collected for immunological assays and clinical signs were recorded throughout the experiment. All goats were euthanized at 9 weeks post-challenge. Gross pathological examination, analysis of lung pathology using computed tomography, and bacterial load quantification in pulmonary lymph nodes (LN) by qPCR were carried out. Only HIMC vaccinated goats showed a significant reduction of lung lesions volume and mycobacterial DNA load in LN compared to unvaccinated controls. Both vaccinated groups showed also a significant reduction of the other pathological parameters, an improved clinical outcome and a higher proportion of IFN-γ-producing central memory T cells after vaccination. The results indicated that homologous vaccination of goats with HIMC induced enhanced protection against M. caprae challenge by reducing lung pathology and bacterial load compared to the heterologous vaccine (HIMB). Further large-scale trials are necessary to assess the efficacy of autovaccines under field conditions.
Collapse
Affiliation(s)
- Cristian Melgarejo
- grid.424716.2Unitat Mixta d’investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Catalonia Spain ,grid.424716.2IRTA. Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la UAB, Bellaterra, Catalonia Spain
| | - Carles Planas
- grid.7080.f0000 0001 2296 0625Departament de Medicina i Cirurgía Animals, Universitat Autònoma de Barcelona, Bellaterra, Catalonia Spain
| | - Alex Cobos
- grid.424716.2Unitat Mixta d’investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Catalonia Spain ,grid.424716.2IRTA. Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la UAB, Bellaterra, Catalonia Spain ,grid.7080.f0000 0001 2296 0625Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona, Bellaterra, Catalonia Spain
| | - Claudia Arrieta-Villegas
- grid.424716.2Unitat Mixta d’investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Catalonia Spain ,grid.424716.2IRTA. Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la UAB, Bellaterra, Catalonia Spain
| | - Iker A. Sevilla
- grid.509696.50000 0000 9853 6743Animal Health Department, NEIKER-Instituto Vasco de Investigación y Desarrollo Agrario, Basque Research and Technology Alliance (BRTA). Derio, Bizkaia, Basque Country Spain
| | - Javier Bezos
- grid.4795.f0000 0001 2157 7667Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain ,grid.4795.f0000 0001 2157 7667VISAVET Health Surveillance Centre, Universidad Complutense de Madrid, Madrid, Spain
| | - Xavier Moll
- grid.7080.f0000 0001 2296 0625Departament de Medicina i Cirurgía Animals, Universitat Autònoma de Barcelona, Bellaterra, Catalonia Spain ,grid.7080.f0000 0001 2296 0625Fundació Hospital Clínic Veterinari, Universitat Autònoma de Barcelona, Bellaterra, Catalonia Spain
| | - Yvonne Espada
- grid.7080.f0000 0001 2296 0625Departament de Medicina i Cirurgía Animals, Universitat Autònoma de Barcelona, Bellaterra, Catalonia Spain ,grid.7080.f0000 0001 2296 0625Fundació Hospital Clínic Veterinari, Universitat Autònoma de Barcelona, Bellaterra, Catalonia Spain
| | - Joseba M. Garrido
- grid.509696.50000 0000 9853 6743Animal Health Department, NEIKER-Instituto Vasco de Investigación y Desarrollo Agrario, Basque Research and Technology Alliance (BRTA). Derio, Bizkaia, Basque Country Spain
| | - Mariano Domingo
- grid.424716.2Unitat Mixta d’investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Catalonia Spain ,grid.424716.2IRTA. Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la UAB, Bellaterra, Catalonia Spain ,grid.7080.f0000 0001 2296 0625Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona, Bellaterra, Catalonia Spain
| | - Enric Vidal
- grid.424716.2Unitat Mixta d’investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Catalonia Spain ,grid.424716.2IRTA. Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la UAB, Bellaterra, Catalonia Spain
| | - Bernat Pérez de Val
- grid.424716.2Unitat Mixta d’investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra, Catalonia Spain ,grid.424716.2IRTA. Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la UAB, Bellaterra, Catalonia Spain
| |
Collapse
|
12
|
Rawat BS, Kumar D, Soni V, Rosenn EH. Therapeutic Potentials of Immunometabolomic Modulations Induced by Tuberculosis Vaccination. Vaccines (Basel) 2022; 10:vaccines10122127. [PMID: 36560537 PMCID: PMC9781011 DOI: 10.3390/vaccines10122127] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/03/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022] Open
Abstract
Metabolomics is emerging as a promising tool to understand the effect of immunometabolism for the development of novel host-directed alternative therapies. Immunometabolism can modulate both innate and adaptive immunity in response to pathogens and vaccinations. For instance, infections can affect lipid and amino acid metabolism while vaccines can trigger bile acid and carbohydrate pathways. Metabolomics as a vaccinomics tool, can provide a broader picture of vaccine-induced biochemical changes and pave a path to potentiate the vaccine efficacy. Its integration with other systems biology tools or treatment modes can enhance the cure, response rate, and control over the emergence of drug-resistant strains. Mycobacterium tuberculosis (Mtb) infection can remodel the host metabolism for its survival, while there are many biochemical pathways that the host adjusts to combat the infection. Similarly, the anti-TB vaccine, Bacillus Calmette-Guerin (BCG), was also found to affect the host metabolic pathways thus modulating immune responses. In this review, we highlight the metabolomic schema of the anti-TB vaccine and its therapeutic applications. Rewiring of immune metabolism upon BCG vaccination induces different signaling pathways which lead to epigenetic modifications underlying trained immunity. Metabolic pathways such as glycolysis, central carbon metabolism, and cholesterol synthesis play an important role in these aspects of immunity. Trained immunity and its applications are increasing day by day and it can be used to develop the next generation of vaccines to treat various other infections and orphan diseases. Our goal is to provide fresh insight into this direction and connect various dots to develop a conceptual framework.
Collapse
Affiliation(s)
- Bhupendra Singh Rawat
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Deepak Kumar
- Department of Zoology, University of Rajasthan, Jaipur 302004, Rajasthan, India
| | - Vijay Soni
- Division of Infectious Diseases, Weill Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
- Correspondence:
| | - Eric H. Rosenn
- School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
13
|
Kanipe C, Boggiatto PM, Putz EJ, Palmer MV. Histopathologic differences in granulomas of Mycobacterium bovis bacille Calmette Guérin (BCG) vaccinated and non-vaccinated cattle with bovine tuberculosis. Front Microbiol 2022; 13:1048648. [PMID: 36425039 PMCID: PMC9678917 DOI: 10.3389/fmicb.2022.1048648] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 10/24/2022] [Indexed: 10/24/2023] Open
Abstract
Mycobacterium bovis (M. bovis) is the zoonotic bacterium responsible for bovine tuberculosis. An attenuated form of M. bovis, Bacillus Calmette-Guerin (BCG), is a modified live vaccine known to provide variable protection in cattle and other species. Protection for this vaccine is defined as a reduction in disease severity rather than prevention of infection and is determined by evaluation of the characteristic lesion of tuberculosis: the granuloma. Despite its recognized ability to decrease disease severity, the mechanism by which BCG imparts protection remains poorly understood. Understanding the histopathologic differences between granulomas which form in BCG vaccinates compared to non-vaccinates may help identify how BCG imparts protection and lead to an improved vaccine. Utilizing special stains and image analysis software, we examined 88 lymph nodes obtained from BGC-vaccinated and non-vaccinated animals experimentally infected with M. bovis. We evaluated the number of granulomas, their size, severity (grade), density of multinucleated giant cells (MNGC), and the amounts of necrosis, mineralization, and fibrosis. BCG vaccinates had fewer granulomas overall and smaller high-grade granulomas with less necrosis than non-vaccinates. The relative numbers of high- and low- grade lesions were similar as were the amounts of mineralization and the density of MNGC. The amount of fibrosis was higher in low-grade granulomas from vaccinates compared to non-vaccinates. Collectively, these findings suggest that BCG vaccination reduces bacterial establishment, resulting in the formation of fewer granulomas. In granulomas that form, BCG has a protective effect by containing their size, reducing the relative amount of necrosis, and increasing fibrosis in low-grade lesions. Vaccination did not affect the amount of mineralization or density of MNGC.
Collapse
Affiliation(s)
- C. Kanipe
- Infectious Bacterial Diseases Research Unit, National Animal Disease Center, Agricultural Research Service (USDA), Ames, IA, United States
- Immunobiology Graduate Program, Iowa State University, Ames, IA, United States
| | - P. M. Boggiatto
- Infectious Bacterial Diseases Research Unit, National Animal Disease Center, Agricultural Research Service (USDA), Ames, IA, United States
| | - E. J. Putz
- Infectious Bacterial Diseases Research Unit, National Animal Disease Center, Agricultural Research Service (USDA), Ames, IA, United States
| | - M. V. Palmer
- Infectious Bacterial Diseases Research Unit, National Animal Disease Center, Agricultural Research Service (USDA), Ames, IA, United States
| |
Collapse
|
14
|
Khalid H, van Hooij A, Connelley TK, Geluk A, Hope JC. Protein Levels of Pro-Inflammatory Cytokines and Chemokines as Biomarkers of Mycobacterium bovis Infection and BCG Vaccination in Cattle. Pathogens 2022; 11:pathogens11070738. [PMID: 35889984 PMCID: PMC9320177 DOI: 10.3390/pathogens11070738] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/23/2022] [Accepted: 06/25/2022] [Indexed: 12/10/2022] Open
Abstract
Bovine tuberculosis (bTB), caused by Mycobacterium bovis, is a globally prevalent infectious disease with significant animal welfare and economic impact. Difficulties in implementing test-and-slaughter measures in low- and middle-income countries (LMICs) and the underperformance of the current diagnostics establish a clear need to develop improved diagnostics. Adaptive immunity biomarkers other than IFNγ could be useful as suggested by various gene expression studies; however, a comprehensive assessment at the protein level is lacking. Here, we screened a range of chemokines and cytokines for their potential as biomarkers in samples from M. bovis experimentally challenged or naive animals. Although serum concentrations for most proteins were low, the pro-inflammatory markers, IL-2, CXCL-9, IP-10 and CCL4, in addition to IFNγ, were found to be significantly elevated in bovine tuberculin (PPDb)-stimulated whole blood supernatants. Further assessment of these molecules in BCG-vaccinated with or without subsequent M. bovis challenge or naive animals revealed that PPDb-specific IL-2 and IP-10, in addition to IFNγ, could discriminate naive and BCG-vaccinated from M. bovis challenged animals. Moreover, these proteins, along with CCL4, showed DIVA potential, i.e., enabling differentiation of M. bovis-infected animals from BCG-vaccinated animals. Combined analysis of cytokines and chemokines could also accurately identify M. bovis infection with strong correlations observed between PPDb-specific IFNγ, IL-2 and IP-10 levels. This provides proof of concept for utilizing multiple biomarker signatures for discrimination of animals with respect to M. bovis infection or BCG vaccination status.
Collapse
Affiliation(s)
- Hamza Khalid
- Division of Infection and Immunity, The Roslin Institute, University of Edinburgh, Easter Bush, Edinburgh EH25 9RG, UK;
- Center for Inflammation Research, The Queen’s Medical Research Institute, Edinburgh BioQuarter, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
- Correspondence: (H.K.); (J.C.H.)
| | - Anouk van Hooij
- Department of Infectious Diseases, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (A.v.H.); (A.G.)
| | - Timothy K. Connelley
- Division of Infection and Immunity, The Roslin Institute, University of Edinburgh, Easter Bush, Edinburgh EH25 9RG, UK;
| | - Annemieke Geluk
- Department of Infectious Diseases, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (A.v.H.); (A.G.)
| | - Jayne C. Hope
- Division of Infection and Immunity, The Roslin Institute, University of Edinburgh, Easter Bush, Edinburgh EH25 9RG, UK;
- Correspondence: (H.K.); (J.C.H.)
| |
Collapse
|
15
|
Hu Z, Lu SH, Lowrie DB, Fan XY. Research Advances for Virus-vectored Tuberculosis Vaccines and Latest Findings on Tuberculosis Vaccine Development. Front Immunol 2022; 13:895020. [PMID: 35812383 PMCID: PMC9259874 DOI: 10.3389/fimmu.2022.895020] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 05/27/2022] [Indexed: 11/13/2022] Open
Abstract
Tuberculosis (TB), caused by respiratory infection with Mycobacterium tuberculosis, remains a major global health threat. The only licensed TB vaccine, the one-hundred-year-old Bacille Calmette-Guérin has variable efficacy and often provides poor protection against adult pulmonary TB, the transmissible form of the disease. Thus, the lack of an optimal TB vaccine is one of the key barriers to TB control. Recently, the development of highly efficacious COVID-19 vaccines within one year accelerated the vaccine development process in human use, with the notable example of mRNA vaccines and adenovirus-vectored vaccines, and increased the public acceptance of the concept of the controlled human challenge model. In the TB vaccine field, recent progress also facilitated the deployment of an effective TB vaccine. In this review, we provide an update on the current virus-vectored TB vaccine pipeline and summarize the latest findings that might facilitate TB vaccine development. In detail, on the one hand, we provide a systematic literature review of the virus-vectored TB vaccines are in clinical trials, and other promising candidate vaccines at an earlier stage of development are being evaluated in preclinical animal models. These research sharply increase the likelihood of finding a more effective TB vaccine in the near future. On the other hand, we provide an update on the latest tools and concept that facilitating TB vaccine research development. We propose that a pre-requisite for successful development may be a better understanding of both the lung-resident memory T cell-mediated mucosal immunity and the trained immunity of phagocytic cells. Such knowledge could reveal novel targets and result in the innovative vaccine designs that may be needed for a quantum leap forward in vaccine efficacy. We also summarized the research on controlled human infection and ultra-low-dose aerosol infection murine models, which may provide more realistic assessments of vaccine utility at earlier stages. In addition, we believe that the success in the ongoing efforts to identify correlates of protection would be a game-changer for streamlining the triage of multiple next-generation TB vaccine candidates. Thus, with more advanced knowledge of TB vaccine research, we remain hopeful that a more effective TB vaccine will eventually be developed in the near future.
Collapse
Affiliation(s)
- Zhidong Hu
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of Ministry of Education (MOE)/Ministry of Health (MOH), Fudan University, Shanghai, China
- *Correspondence: Zhidong Hu, ; Xiao-Yong Fan,
| | - Shui-Hua Lu
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of Ministry of Education (MOE)/Ministry of Health (MOH), Fudan University, Shanghai, China
- National Medical Center for Infectious Diseases of China, Shenzhen Third People Hospital, South Science & Technology University, Shenzhen, China
| | - Douglas B. Lowrie
- National Medical Center for Infectious Diseases of China, Shenzhen Third People Hospital, South Science & Technology University, Shenzhen, China
| | - Xiao-Yong Fan
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of Ministry of Education (MOE)/Ministry of Health (MOH), Fudan University, Shanghai, China
- *Correspondence: Zhidong Hu, ; Xiao-Yong Fan,
| |
Collapse
|
16
|
Wedlich N, Figl J, Liebler-Tenorio EM, Köhler H, von Pückler K, Rissmann M, Petow S, Barth SA, Reinhold P, Ulrich R, Grode L, Kaufmann SHE, Menge C. Video Endoscopy-Guided Intrabronchial Spray Inoculation of Mycobacterium bovis in Goats and Comparative Assessment of Lung Lesions With Various Imaging Methods. Front Vet Sci 2022; 9:877322. [PMID: 35591868 PMCID: PMC9113525 DOI: 10.3389/fvets.2022.877322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 04/01/2022] [Indexed: 11/13/2022] Open
Abstract
Bovine tuberculosis (bTB) not only poses a zoonotic threat to humans but also has a significant economic impact on livestock production in many areas of the world. Effective vaccines for humans, livestock, and wildlife are highly desirable to control tuberculosis. Suitable large animal models are indispensable for meaningful assessment of vaccine candidates. Here, we describe the refinement of an animal model for bTB in goats. Intrabronchial inoculation procedure via video-guided endoscopy in anesthetized animals, collection of lungs after intratracheal fixation in situ, and imaging of lungs by computed tomography (CT) were established in three goats using barium sulfate as surrogate inoculum. For subsequent infection experiments, four goats were infected with 4.7 × 102 colony-forming units of M. bovis by intrabronchial inoculation using video-guided endoscopy with spray catheters. Defined amounts of inoculum were deposited at five sites per lung. Four age-matched goats were mock-inoculated. None of the goats developed clinical signs until they were euthanized 5 months post infection, but simultaneous skin testing confirmed bTB infection in all goats inoculated with M. bovis. In tissues collected at necropsy, M. bovis was consistently re-isolated from granulomas in lymph nodes, draining the lungs of all the goats infected with M. bovis. Further dissemination was observed in one goat only. Pulmonary lesions were quantified by CT and digital 2D radiography (DR). CT revealed mineralized lesions in all the infected goats ranging from <5 mm to >10 mm in diameter. Small lesions <5 mm predominated. The DR failed to detect small lesions and to determine the exact location of lesions because of overlapping of pulmonary lobes. Relative volume of pulmonary lesions was low in three but high in one goat that also had extensive cavitation. CT lesions could be correlated to gross pathologic findings and histologic granuloma types in representative pulmonary lobes. In conclusion, video-guided intrabronchial inoculation with spray catheters, mimicking the natural way of infection, resulted in pulmonary infection of goats with M. bovis. CT, but not DR, presented as a highly sensitive method to quantify the extent of pulmonary lesions. This goat model of TB may serve as a model for testing TB vaccine efficacy.
Collapse
Affiliation(s)
- Nadine Wedlich
- Institute of Molecular Pathogenesis, Friedrich-Loeffler-Institute (FLI), Jena, Germany
| | - Julia Figl
- Institute of Molecular Pathogenesis, Friedrich-Loeffler-Institute (FLI), Jena, Germany
| | - Elisabeth M. Liebler-Tenorio
- Institute of Molecular Pathogenesis, Friedrich-Loeffler-Institute (FLI), Jena, Germany
- *Correspondence: Elisabeth M. Liebler-Tenorio
| | - Heike Köhler
- Institute of Molecular Pathogenesis, Friedrich-Loeffler-Institute (FLI), Jena, Germany
| | - Kerstin von Pückler
- Clinic for Small Animals – Radiology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Melanie Rissmann
- Department of Experimental Animal Facilities and Biorisk Management, Friedrich-Loeffler-Institute, Greifswald-Insel Riems, Germany
| | - Stefanie Petow
- Institute for Animal Welfare and Animal Husbandry, Friedrich-Loeffler-Institute, Celle, Germany
| | - Stefanie A. Barth
- Institute of Molecular Pathogenesis, Friedrich-Loeffler-Institute (FLI), Jena, Germany
| | - Petra Reinhold
- Institute of Molecular Pathogenesis, Friedrich-Loeffler-Institute (FLI), Jena, Germany
| | - Reiner Ulrich
- Department of Experimental Animal Facilities and Biorisk Management, Friedrich-Loeffler-Institute, Greifswald-Insel Riems, Germany
| | | | - Stefan H. E. Kaufmann
- Director Emeritus, Max Planck Institute for Infection Biology, Berlin, Germany
- Emeritus Group for Systems Immunology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Hagler Institute for Advanced Study, Texas A&M University, College Station, TX, United States
| | - Christian Menge
- Institute of Molecular Pathogenesis, Friedrich-Loeffler-Institute (FLI), Jena, Germany
| |
Collapse
|
17
|
A century of attempts to develop an effective tuberculosis vaccine: Why they failed? Int Immunopharmacol 2022; 109:108791. [PMID: 35487086 DOI: 10.1016/j.intimp.2022.108791] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 04/18/2022] [Accepted: 04/18/2022] [Indexed: 11/23/2022]
Abstract
Tuberculosis (TB) remains a major global health problem despite widespread use of the Bacillus BCG vaccine. This situation is worsened by co-infection with HIV, and the development of multidrug-resistant Mycobacterium tuberculosis (Mtb) strains. Thus, novel vaccine candidates and improved vaccination strategies are urgently needed in order to reduce the incidence of TB and even to eradicate TB by 2050. Over the last few decades, 23 novel TB vaccines have entered into clinical trials, more than 13 new vaccines have reached various stages of preclinical development, and more than 50 potential candidates are in the discovery stage as next-generation vaccines. Nevertheless, why has a century of attempts to introduce an effective TB vaccine failed? Who should be blamed -scientists, human response, or Mtb strategies? Literature review reveals that the elimination of latent or active Mtb infections in a given population seems to be an epigenetic process. With a better understanding of the connections between bacterial infections and gene expression conditions in epigenetic events, opportunities arise in designing protective vaccines or therapeutic agents, particularly as epigenetic processes can be reversed. Therefore, this review provides a brief overview of different approaches towards novel vaccination strategies and the mechanisms underlying these approaches.
Collapse
|
18
|
Palmer MV, Kanipe C, Boggiatto PM. The Bovine Tuberculoid Granuloma. Pathogens 2022; 11:61. [PMID: 35056009 PMCID: PMC8780557 DOI: 10.3390/pathogens11010061] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 12/28/2021] [Accepted: 12/30/2021] [Indexed: 02/05/2023] Open
Abstract
The bovine tuberculoid granuloma is the hallmark lesion of bovine tuberculosis (bTB) due to Mycobacterium bovis infection. The pathogenesis of bTB, and thereby the process of bovine tuberculoid granuloma development, involves the recruitment, activation, and maintenance of cells under the influence of antigen, cytokines and chemokines in affected lungs and regional lymph nodes. The granuloma is key to successful control of bTB by preventing pathogen dissemination through containment by cellular and fibrotic layers. Paradoxically, however, it may also provide a niche for bacterial replication. The morphologic and cellular characteristics of granulomas have been used to gauge disease severity in bTB pathogenesis and vaccine efficacy studies. As such, it is critical to understand the complex mechanisms behind granuloma initiation, development, and maintenance.
Collapse
Affiliation(s)
- Mitchell V. Palmer
- Bacterial Diseases of Livestock Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA 50010, USA; (C.K.); (P.M.B.)
| | | | | |
Collapse
|
19
|
Characterization of BoHV-1 gG-/tk-/gE- Mutant in Differential Protein Expression, Virulence, and Immunity. Vet Sci 2021; 8:vetsci8110253. [PMID: 34822626 PMCID: PMC8621285 DOI: 10.3390/vetsci8110253] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 11/17/2022] Open
Abstract
Infectious bovine rhinotracheitis (IBR), caused by bovine alphaherpesvirus 1 (BoHV-1), is an important disease affecting cattle worldwide resulting in great economic losses. Marker vaccines are effective in controlling infectious diseases including IBR, because they allow the discrimination between the natural infection and the vaccination. Therefore, a triple gene deleted strain BoHV-1 gG-/tk-/gE- was developed and evaluated in vivo and in vitro as a marker vaccine. In cell culture, this triple mutant virus showed significantly slower growth kinetics and smaller plaques when compared to wild-type (wt) BoHV-1 and double mutant BoHV-1 gG-/tk- (p < 0.01). On proteomic level, it revealed downregulation of some virulence related proteins including thymidine kinase, glycoproteins G, E, I, and K when compared to the wt. In vitro, the triple mutant virus showed a significantly lower and shorter viral shedding period (p < 0.001) in calves compared to double mutant. Moreover, the immunized calves with triple mutant virus showed protection rates of 64.2% and 68.6% against wt BoHV-1 and wt BoHV-5 challenge, respectively, without reactivation of latency after dexamethasone injection. In conclusion, BoHV-1 gG-/tk-/gE- is a safer marker vaccine against IBR although its immunogenicity in calves was decreased when compared to double mutant virus.
Collapse
|
20
|
Cho T, Khatchadourian C, Nguyen H, Dara Y, Jung S, Venketaraman V. A review of the BCG vaccine and other approaches toward tuberculosis eradication. Hum Vaccin Immunother 2021; 17:2454-2470. [PMID: 33769193 PMCID: PMC8475575 DOI: 10.1080/21645515.2021.1885280] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/29/2021] [Indexed: 02/02/2023] Open
Abstract
Despite aggressive eradication efforts, Tuberculosis (TB) remains a global health burden, one that disproportionally affects poorer, less developed nations. The only vaccine approved for TB, the Bacillus of Calmette and Guérin (BCG) vaccine remains controversial because it's stated efficacy has been cited as anywhere from 0 to 80%. Nevertheless, there have been exciting discoveries about the mechanism of action of the BCG vaccine that suggests it has a role in immunization schedules today. We review recent data suggesting the vaccine imparts protection against both tuberculosis and non-tuberculosis pathogens via a newly discovered immune system called trained immunity. BCG's efficacy also appears to be tied to its affect on granulocytes at the epigenetic and hematopoietic stem cell levels, which we discuss in this article at length. We also write about how the different strains of the BCG vaccine elicit different immune responses, suggesting that certain BCG strains are more immunogenic than others. Finally, our review delves into how the current vaccine is being reformulated to be more efficacious, and track the development of the next generation vaccines against TB.
Collapse
Affiliation(s)
- Thomas Cho
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, USA
| | | | - Huy Nguyen
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, USA
| | - Yash Dara
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, USA
| | - Shuna Jung
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, USA
| | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, USA
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, USA
| |
Collapse
|
21
|
Bayissa B, Sirak A, Zewude A, Worku A, Gumi B, Berg S, Hewinson RG, Wood JLN, Jones GJ, Vordermeier HM, Ameni G. Field evaluation of specific mycobacterial protein-based skin test for the differentiation of Mycobacterium bovis-infected and Bacillus Calmette Guerin-vaccinated crossbred cattle in Ethiopia. Transbound Emerg Dis 2021; 69:e1-e9. [PMID: 34331511 PMCID: PMC8801543 DOI: 10.1111/tbed.14252] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Bovine tuberculosis (bTB) challenges intensive dairy production in Ethiopia and implementation of the test and slaughter control strategy is not economically acceptable in the country. Vaccination of cattle with Bacillus Calmette–Guerin (BCG) could be an important adjunct to control, which would require a diagnostic test to differentiate Mycobacterium bovis (M. bovis)‐infected and BCG‐vaccinated animals (DIVA role). This study describes an evaluation of a DIVA skin test (DST) that is based on a cocktail (DSTc) or fusion (DSTf) of specific (ESAT‐6, CFP‐10 and Rv3615c) M. bovis proteins in Zebu–Holstein–Friesians crossbred cattle in Ethiopia. The study animals used were 74 calves (35 BCG vaccinated and 39 unvaccinated) aged less than 3 weeks at the start of experiment and 68 naturally infected ‘TB reactor’ cows. Six weeks after vaccination, the 74 calves were tested with the DSTc and the single intradermal cervical comparative tuberculin (SICCT) test. The TB reactor cows were tested with the DSTc and the SICCT test. Reactions to the DSTc were not observed in BCG‐vaccinated and unvaccinated calves, while SICCT test reactions were detected in vaccinated calves. DSTc reactions were detected in 95.6% of the TB reactor cows and single intradermal tuberculin positive reactions were found in 98.2% (95% confidence interval, CI, 92.1–100%). The sensitivity of the DSTc was 95.6% (95% CI, 87.6–99.1%), and significantly (p < .001) higher than the sensitivity (75%, 95% CI, 63.0–84.7%) of the SICCT test at 4 mm cut‐off. DSTf and DSTc reactions were correlated (r = 0.75; 95% CI = 0.53–0.88). In conclusion, the DSTc could differentiate M. bovis‐infected from BCG‐vaccinated cattle in Ethiopia. DST had higher sensitivity than the SICCT test. Hence, the DSTc could be used as a diagnostic tool for bTB if BCG vaccination is implemented for the control of bTB in Ethiopia and other countries.
Collapse
Affiliation(s)
- Berecha Bayissa
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa, Ethiopia.,Vaccine Production and Drug Formulation Directorate, National Veterinary Institute, Bishoftu, Ethiopia
| | - Asegedech Sirak
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa, Ethiopia.,National Animal Health Diagnostic and Investigation Centre, Sebeta, Ethiopia
| | - Aboma Zewude
- Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | - Adane Worku
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa, Ethiopia
| | - Balako Gumi
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa, Ethiopia
| | - Stefan Berg
- Department of Bacteriology, Animal and Plant Health Agency, New Haw, Addlestone, Surrey, UK
| | | | - James L N Wood
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Gareth J Jones
- Department of Bacteriology, Animal and Plant Health Agency, New Haw, Addlestone, Surrey, UK
| | | | - H Martin Vordermeier
- Department of Bacteriology, Animal and Plant Health Agency, New Haw, Addlestone, Surrey, UK.,Aberystwyth University, Ceredigion, UK
| | - Gobena Ameni
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa, Ethiopia.,Department of Veterinary Medicine, College of Food and Agriculture, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
22
|
Boggiatto PM, Kanipe CR, Palmer MV. Enhanced Detection of Mycobacterium bovis-Specific T Cells in Experimentally-Infected Cattle. Front Vet Sci 2021; 8:676710. [PMID: 34336973 PMCID: PMC8317970 DOI: 10.3389/fvets.2021.676710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 06/08/2021] [Indexed: 11/13/2022] Open
Abstract
Bovine tuberculosis (bTB), caused by infection with Mycobacterium bovis, continues to be a major economic burden associated with production losses and a public health concern due to its zoonotic nature. As with other intracellular pathogens, cell-mediated immunity plays an important role in the control of infection. Characterization of such responses is important for understanding the immune status of the host, and to identify mechanisms of protective immunity or immunopathology. This type of information can be important in the development of vaccination strategies, diagnostic assays, and in predicting protection or disease progression. However, the frequency of circulating M. bovis-specific T cells are often low, making the analysis of such responses difficult. As previously demonstrated in a different cattle infection model, antigenic expansion allows us to increase the frequency of antigen-specific T cells. Moreover, the concurrent assessment of cytokine production and proliferation provides a deeper understanding of the functional nature of these cells. The work presented here, analyzes the T cell response following experimental M. bovis infection in cattle via in vitro antigenic expansion and re-stimulation to characterize antigen-specific CD4, CD8, and γδ T cells and their functional phenotype, shedding light on the variable functional ability of these cells. Data gathered from these studies can help us better understand the cellular response to M. bovis infection and develop improved vaccines and diagnostic tools.
Collapse
Affiliation(s)
- Paola M Boggiatto
- Infectious Bacterial Diseases Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, United States
| | - Carly R Kanipe
- Infectious Bacterial Diseases Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, United States.,Immunobiology Program, Iowa State University, Ames, IA, United States.,Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN, United States
| | - Mitchell V Palmer
- Infectious Bacterial Diseases Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, United States
| |
Collapse
|
23
|
Temporal dynamics of intradermal cytokine response to tuberculin in Mycobacterium bovis BCG-vaccinated cattle using sampling microneedles. Sci Rep 2021; 11:7074. [PMID: 33782422 PMCID: PMC8007627 DOI: 10.1038/s41598-021-86398-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 03/15/2021] [Indexed: 11/08/2022] Open
Abstract
Bovine tuberculosis (bTB) is a disease of livestock with severe and worldwide economic, animal welfare and zoonotic consequences. Application of test-and-slaughter-based control polices reliant on tuberculin skin testing has been the mainstay of bTB control in cattle. However, little is known about the temporal development of the bovine tuberculin skin test response at the dermal sites of antigen injection. To fill this knowledge gap, we applied minimally-invasive sampling microneedles (SMNs) for intradermal sampling of interstitial fluid at the tuberculin skin test sites in Mycobacterium bovis BCG-vaccinated calves and determined the temporal dynamics of a panel of 15 cytokines and chemokines in situ and in the peripheral blood. The results reveal an orchestrated and coordinated cytokine and local chemokine response, identified IL-1RA as a potential soluble biomarker of a positive tuberculin skin response, and confirmed the utility of IFN-γ and IP-10 for bTB detection in blood-based assays. Together, the results highlight the utility of SMNs to identify novel biomarkers and provide mechanistic insights on the intradermal cytokine and chemokine responses associated with the tuberculin skin test in BCG-sensitized cattle.
Collapse
|
24
|
Smith K, Kleynhans L, Warren RM, Goosen WJ, Miller MA. Cell-Mediated Immunological Biomarkers and Their Diagnostic Application in Livestock and Wildlife Infected With Mycobacterium bovis. Front Immunol 2021; 12:639605. [PMID: 33746980 PMCID: PMC7969648 DOI: 10.3389/fimmu.2021.639605] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 02/08/2021] [Indexed: 01/06/2023] Open
Abstract
Mycobacterium bovis has the largest host range of the Mycobacterium tuberculosis complex and infects domestic animal species, wildlife, and humans. The presence of global wildlife maintenance hosts complicates bovine tuberculosis (bTB) control efforts and further threatens livestock and wildlife-related industries. Thus, it is imperative that early and accurate detection of M. bovis in all affected animal species is achieved. Further, an improved understanding of the complex species-specific host immune responses to M. bovis could enable the development of diagnostic tests that not only identify infected animals but distinguish between infection and active disease. The primary bTB screening standard worldwide remains the tuberculin skin test (TST) that presents several test performance and logistical limitations. Hence additional tests are used, most commonly an interferon-gamma (IFN-γ) release assay (IGRA) that, similar to the TST, measures a cell-mediated immune (CMI) response to M. bovis. There are various cytokines and chemokines, in addition to IFN-γ, involved in the CMI component of host adaptive immunity. Due to the dominance of CMI-based responses to mycobacterial infection, cytokine and chemokine biomarkers have become a focus for diagnostic tests in livestock and wildlife. Therefore, this review describes the current understanding of host immune responses to M. bovis as it pertains to the development of diagnostic tools using CMI-based biomarkers in both gene expression and protein release assays, and their limitations. Although the study of CMI biomarkers has advanced fundamental understanding of the complex host-M. bovis interplay and bTB progression, resulting in development of several promising diagnostic assays, most of this research remains limited to cattle. Considering differences in host susceptibility, transmission and immune responses, and the wide variety of M. bovis-affected animal species, knowledge gaps continue to pose some of the biggest challenges to the improvement of M. bovis and bTB diagnosis.
Collapse
Affiliation(s)
- Katrin Smith
- Division of Molecular Biology and Human Genetics, Department of Science and Innovation-National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, Faculty of Medicine and Health Sciences, South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | - Léanie Kleynhans
- Division of Molecular Biology and Human Genetics, Department of Science and Innovation-National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, Faculty of Medicine and Health Sciences, South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | - Robin M Warren
- Division of Molecular Biology and Human Genetics, Department of Science and Innovation-National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, Faculty of Medicine and Health Sciences, South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | - Wynand J Goosen
- Division of Molecular Biology and Human Genetics, Department of Science and Innovation-National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, Faculty of Medicine and Health Sciences, South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | - Michele A Miller
- Division of Molecular Biology and Human Genetics, Department of Science and Innovation-National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, Faculty of Medicine and Health Sciences, South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
25
|
Sasso E, D'Alise AM, Zambrano N, Scarselli E, Folgori A, Nicosia A. New viral vectors for infectious diseases and cancer. Semin Immunol 2020; 50:101430. [PMID: 33262065 DOI: 10.1016/j.smim.2020.101430] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 10/23/2020] [Accepted: 11/16/2020] [Indexed: 12/12/2022]
Abstract
Since the discovery in 1796 by Edward Jenner of vaccinia virus as a way to prevent and finally eradicate smallpox, the concept of using a virus to fight another virus has evolved into the current approaches of viral vectored genetic vaccines. In recent years, key improvements to the vaccinia virus leading to a safer version (Modified Vaccinia Ankara, MVA) and the discovery that some viruses can be used as carriers of heterologous genes encoding for pathological antigens of other infectious agents (the concept of 'viral vectors') has spurred a new wave of clinical research potentially providing for a solution for the long sought after vaccines against major diseases such as HIV, TB, RSV and Malaria, or emerging infectious diseases including those caused by filoviruses and coronaviruses. The unique ability of some of these viral vectors to stimulate the cellular arm of the immune response and, most importantly, T lymphocytes with cell killing activity, has also reawakened the interest toward developing therapeutic vaccines against chronic infectious diseases and cancer. To this end, existing vectors such as those based on Adenoviruses have been improved in immunogenicity and efficacy. Along the same line, new vectors that exploit viruses such as Vesicular Stomatitis Virus (VSV), Measles Virus (MV), Lymphocytic choriomeningitis virus (LCMV), cytomegalovirus (CMV), and Herpes Simplex Virus (HSV), have emerged. Furthermore, technological progress toward modifying their genome to render some of these vectors incompetent for replication has increased confidence toward their use in infant and elderly populations. Lastly, their production process being the same for every product has made viral vectored vaccines the technology of choice for rapid development of vaccines against emerging diseases and for 'personalised' cancer vaccines where there is an absolute need to reduce time to the patient from months to weeks or days. Here we review the recent developments in viral vector technologies, focusing on novel vectors based on primate derived Adenoviruses and Poxviruses, Rhabdoviruses, Paramixoviruses, Arenaviruses and Herpesviruses. We describe the rationale for, immunologic mechanisms involved in, and design of viral vectored gene vaccines under development and discuss the potential utility of these novel genetic vaccine approaches in eliciting protection against infectious diseases and cancer.
Collapse
Affiliation(s)
- Emanuele Sasso
- Nouscom srl, Via di Castel Romano 100, 00128 Rome, Italy; Ceinge-Biotecnologie Avanzate S.C. A.R.L., via Gaetano Salvatore 486, 80145 Naples, Italy.
| | | | - Nicola Zambrano
- Ceinge-Biotecnologie Avanzate S.C. A.R.L., via Gaetano Salvatore 486, 80145 Naples, Italy; Department of Molecular Medicine and Medical Biotechnology, University Federico II, Via Pansini 5, 80131 Naples, Italy.
| | | | | | - Alfredo Nicosia
- Ceinge-Biotecnologie Avanzate S.C. A.R.L., via Gaetano Salvatore 486, 80145 Naples, Italy; Department of Molecular Medicine and Medical Biotechnology, University Federico II, Via Pansini 5, 80131 Naples, Italy.
| |
Collapse
|
26
|
Vrba SM, Kirk NM, Brisse ME, Liang Y, Ly H. Development and Applications of Viral Vectored Vaccines to Combat Zoonotic and Emerging Public Health Threats. Vaccines (Basel) 2020; 8:E680. [PMID: 33202961 PMCID: PMC7712223 DOI: 10.3390/vaccines8040680] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 02/07/2023] Open
Abstract
Vaccination is arguably the most cost-effective preventative measure against infectious diseases. While vaccines have been successfully developed against certain viruses (e.g., yellow fever virus, polio virus, and human papilloma virus HPV), those against a number of other important public health threats, such as HIV-1, hepatitis C, and respiratory syncytial virus (RSV), have so far had very limited success. The global pandemic of COVID-19, caused by the SARS-CoV-2 virus, highlights the urgency of vaccine development against this and other constant threats of zoonotic infection. While some traditional methods of producing vaccines have proven to be successful, new concepts have emerged in recent years to produce more cost-effective and less time-consuming vaccines that rely on viral vectors to deliver the desired immunogens. This review discusses the advantages and disadvantages of different viral vaccine vectors and their general strategies and applications in both human and veterinary medicines. A careful review of these issues is necessary as they can provide important insights into how some of these viral vaccine vectors can induce robust and long-lasting immune responses in order to provide protective efficacy against a variety of infectious disease threats to humans and animals, including those with zoonotic potential to cause global pandemics.
Collapse
Affiliation(s)
- Sophia M. Vrba
- Department of Veterinary & Biomedical Sciences, University of Minnesota, Twin Cities, St. Paul, MN 55108, USA; (S.M.V.); (Y.L.)
| | - Natalie M. Kirk
- Comparative Molecular Biosciences Graduate Program, Department of Veterinary & Biomedical Sciences, University of Minnesota, Twin Cities, St. Paul, MN 55108, USA;
| | - Morgan E. Brisse
- Biochemistry, Molecular Biology and Biophysics Graduate Program, Department of Veterinary & Biomedical Sciences, University of Minnesota, Twin Cities, St. Paul, MN 55108, USA;
| | - Yuying Liang
- Department of Veterinary & Biomedical Sciences, University of Minnesota, Twin Cities, St. Paul, MN 55108, USA; (S.M.V.); (Y.L.)
| | - Hinh Ly
- Department of Veterinary & Biomedical Sciences, University of Minnesota, Twin Cities, St. Paul, MN 55108, USA; (S.M.V.); (Y.L.)
| |
Collapse
|
27
|
Benedictus L, Steinbach S, Holder T, Bakker D, Vrettou C, Morrison WI, Vordermeier M, Connelley T. Hydrophobic Mycobacterial Antigens Elicit Polyfunctional T Cells in Mycobacterium bovis Immunized Cattle: Association With Protection Against Challenge? Front Immunol 2020; 11:588180. [PMID: 33281817 PMCID: PMC7688591 DOI: 10.3389/fimmu.2020.588180] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/20/2020] [Indexed: 01/09/2023] Open
Abstract
Bovine tuberculosis (bTB), caused by Mycobacterium bovis, is a chronic disease of cattle with a detrimental impact on food quality and production. Research on bTB vaccines has predominantly been focused on proteinaceous antigens. However, mycobacteria have a thick and intricate lipid outer layer and lipids as well as lipopeptides are important for immune-evasion and virulence. In humans, lipid extracts of M. tuberculosis have been shown to elicit immune responses effective against M. tuberculosis in vitro. Chloroform-methanol extraction (CME) was applied to M. bovis BCG to obtain a hydrophobic antigen extract (CMEbcg) containing lipids and lipopeptides. CMEbcg stimulated IFN-γ+IL-2+ and IL-17A+IL-22+ polyfunctional T cells and elicited T cell responses with a Th1 and Th17 cytokine release profile in both M. bovis BCG vaccinated and M. bovis challenged calves. Lipopeptides were shown to be the immunodominant antigens in CMEbcg, stimulating CD4 T cells via MHC class II. CMEbcg expanded T cells killed CMEbcg loaded monocytes and the CMEbcg-specific CD3 T cell proliferative response following M. bovis BCG vaccination was the best predictor for reduced pathology following challenge with M. bovis. Although the high predictive value of CMEbcg-specific immune responses does not confirm a causal relationship with protection against M. bovis challenge, when taking into account the in vitro antimycobacterial phenotype of CMEbcg-specific T cells (e.g. Th1/Th17 cytokine profile), it is indicative that CMEbcg-specific immune responses could play a functional role in immunity against M. bovis. Based on these findings we conclude that lipopeptides of M. bovis are potential novel subunit vaccine candidates and that further studies into the functional characterization of lipopeptide-specific immune responses together with their role in protection against bovine tuberculosis are warranted.
Collapse
Affiliation(s)
- Lindert Benedictus
- Division of Infection and Immunity, The Roslin Institute, The University of Edinburgh, Easter Bush, United Kingdom
| | - Sabine Steinbach
- Department of Bacteriology, Animal and Plant Health Agency, Weybridge, United Kingdom
| | - Thomas Holder
- Department of Bacteriology, Animal and Plant Health Agency, Weybridge, United Kingdom
| | - Douwe Bakker
- Independent Researcher and Technical Consultant, Lelystad, Netherlands
| | - Christina Vrettou
- Division of Infection and Immunity, The Roslin Institute, The University of Edinburgh, Easter Bush, United Kingdom
| | - W Ivan Morrison
- Division of Infection and Immunity, The Roslin Institute, The University of Edinburgh, Easter Bush, United Kingdom
| | - Martin Vordermeier
- Department of Bacteriology, Animal and Plant Health Agency, Weybridge, United Kingdom.,Centre for Bovine Tuberculosis, Institute for Biological, Environmental and Rural Sciences, University of Aberystwyth, Aberystwyth, United Kingdom
| | - Timothy Connelley
- Division of Infection and Immunity, The Roslin Institute, The University of Edinburgh, Easter Bush, United Kingdom
| |
Collapse
|
28
|
Lee DF, Stewart GR, Chambers MA. Modelling early events in Mycobacterium bovis infection using a co-culture model of the bovine alveolus. Sci Rep 2020; 10:18495. [PMID: 33116165 PMCID: PMC7595104 DOI: 10.1038/s41598-020-75113-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/21/2020] [Indexed: 01/01/2023] Open
Abstract
Bovine tuberculosis (bTB), a zoonosis mainly caused by Mycobacterium bovis has severe socio-economic consequences and impact on animal health. Host-pathogen interactions during M. bovis infection are poorly understood, especially early events which are difficult to follow in vivo. This study describes the utilisation of an in vitro co-culture model, comprising immortalised bovine alveolar type II (BATII) epithelial cells and bovine pulmonary arterial endothelial cells (BPAECs). When cultured at air-liquid interface, it was possible to follow the migration of live M. bovis Bacille Calmette-Guérin (BCG) and to observe interactions with each cell type, alongside cytokine release. Infection with BCG was shown to exert a detrimental effect primarily upon epithelial cells, with corresponding increases in IL8, TNFα, IL22 and IL17a cytokine release, quantified by ELISA. BCG infection increased expression of CD54, MHC Class I and II molecules in endothelial but not epithelial cells, which exhibited constitutive expression. The effect of peripheral blood mononuclear cell conditioned medium from vaccinated cattle upon apical-basolateral migration of BCG was examined by quantifying recovered BCG from the apical, membrane and basolateral fractions over time. The numbers of recovered BCG in each fraction were unaffected by the presence of PBMC conditioned medium, with no observable differences between vaccinated and naïve animals.
Collapse
Affiliation(s)
- Diane Frances Lee
- School of Veterinary Medicine, University of Surrey, Guildford, Surrey, UK.
| | | | - Mark Andrew Chambers
- School of Veterinary Medicine, University of Surrey, Guildford, Surrey, UK
- School of Biosciences and Medicine, University of Surrey, Guildford, Surrey, UK
| |
Collapse
|
29
|
Ullah I, Bibi S, Ul Haq I, Safia, Ullah K, Ge L, Shi X, Bin M, Niu H, Tian J, Zhu B. The Systematic Review and Meta-Analysis on the Immunogenicity and Safety of the Tuberculosis Subunit Vaccines M72/AS01 E and MVA85A. Front Immunol 2020; 11:1806. [PMID: 33133057 PMCID: PMC7578575 DOI: 10.3389/fimmu.2020.01806] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 07/06/2020] [Indexed: 01/30/2023] Open
Abstract
Background: Tuberculosis (TB) is a severe infectious disease with devastating effects on global public health. No TB vaccine has yet been approved for use on latent TB infections and healthy adults. In this study, we performed a systematic review and meta-analysis to evaluate the immunogenicity and safety of the M72/AS01E and MVA85A subunit vaccines. The M72/AS01E is a novel peptide-based vaccine currently in progress, which may increase the protection level against TB infection. The MVA85A was a viral vector-based TB subunit vaccine being used in the clinical trials. The vaccines mentioned above have been studied in various phase I/II clinical trials. Immunogenicity and safety is the first consideration for TB vaccine development. Methods: The PubMed, Embase, and Cochrane Library databases were searched for published studies (until October 2019) to find out information on the M72/AS01E and MVA85A candidate vaccines. The meta-analysis was conducted by applying the standard methods and processes established by the Cochrane Collaboration. Results: Five eligible randomized clinical trials (RCTs) were selected for the meta-analysis of M72/AS01E candidate vaccines. The analysis revealed that the M72/AS01E subunit vaccine had an abundance of polyfunctional M72-specific CD4+ T cells [standardized mean difference (SMD) = 2.37] in the vaccine group versus the control group, the highest seropositivity rate [relative risk (RR) = 5.09]. The M72/AS01E vaccinated group were found to be at high risk of local injection site redness (RR = 2.64), headache (RR = 1.59), malaise (RR = 3.55), myalgia (RR = 2.27), fatigue (RR = 2.16), pain (RR = 3.99), swelling (RR = 5.09), and fever (RR = 2.04) compared to the control groups. The incidences of common adverse events of M72/AS01E were local injection site redness, headache, malaise, myalgia, fatigue, pain, swelling, fever, etc. Six eligible RCTs were selected for the meta-analysis on MVA85A candidate vaccines. The analysis revealed that the subunit vaccine MVA85A had a higher abundance of overall pooled proportion polyfunctional MVA85A-specific CD4+ T cells SMD = 2.41 in the vaccine group vs. the control group, with the highest seropositivity rate [estimation rate (ER) = 0.55]. The MVA85A vaccinated group were found to be at high risk of local injection site redness (ER = 0.55), headache (ER = 0.40), malaise (ER = 0.29), pain (ER = 0.54), myalgia (ER = 0.31), and fever (ER = 0.20). The incidences of common adverse events of MVA85A were local injection site redness, headache, malaise, pain, myalgia, fever, etc. Conclusion: The M72/AS01E and MVA85A vaccines against TB are safe and had immunogenicity in diverse clinical trials. The M72/AS01E and MVA85A vaccines are associated with a mild adverse reaction. The meta-analysis on immunogenicity and safety of M72/AS01E and MVA85A vaccines provides useful information for the evaluation of available subunit vaccines in the clinic.
Collapse
Affiliation(s)
- Inayat Ullah
- Lanzhou Center for Tuberculosis Research and Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.,Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation, Lanzhou University, Lanzhou, China
| | - Shaheen Bibi
- Lanzhou Center for Tuberculosis Research and Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.,Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation, Lanzhou University, Lanzhou, China.,School of Life Science, Northwest Normal University, Lanzhou, China
| | - Ijaz Ul Haq
- College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou, China
| | - Safia
- Pakistan Institute of Community Ophthalmology (PICO), Hayatabad Medical Complex, KMU, Peshawar, Pakistan
| | - Kifayat Ullah
- Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Long Ge
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Xintong Shi
- Lanzhou Center for Tuberculosis Research and Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.,Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation, Lanzhou University, Lanzhou, China
| | - Ma Bin
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation, Lanzhou University, Lanzhou, China
| | - Hongxia Niu
- Lanzhou Center for Tuberculosis Research and Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.,Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation, Lanzhou University, Lanzhou, China
| | - Jinhui Tian
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation, Lanzhou University, Lanzhou, China
| | - Bingdong Zhu
- Lanzhou Center for Tuberculosis Research and Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.,Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation, Lanzhou University, Lanzhou, China
| |
Collapse
|
30
|
Saralahti AK, Uusi-Mäkelä MIE, Niskanen MT, Rämet M. Integrating fish models in tuberculosis vaccine development. Dis Model Mech 2020; 13:13/8/dmm045716. [PMID: 32859577 PMCID: PMC7473647 DOI: 10.1242/dmm.045716] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Tuberculosis is a chronic infection by Mycobacterium tuberculosis that results in over 1.5 million deaths worldwide each year. Currently, there is only one vaccine against tuberculosis, the Bacillus Calmette–Guérin (BCG) vaccine. Despite widespread vaccination programmes, over 10 million new M. tuberculosis infections are diagnosed yearly, with almost half a million cases caused by antibiotic-resistant strains. Novel vaccination strategies concentrate mainly on replacing BCG or boosting its efficacy and depend on animal models that accurately recapitulate the human disease. However, efforts to produce new vaccines against an M. tuberculosis infection have encountered several challenges, including the complexity of M. tuberculosis pathogenesis and limited knowledge of the protective immune responses. The preclinical evaluation of novel tuberculosis vaccine candidates is also hampered by the lack of an appropriate animal model that could accurately predict the protective effect of vaccines in humans. Here, we review the role of zebrafish (Danio rerio) and other fish models in the development of novel vaccines against tuberculosis and discuss how these models complement the more traditional mammalian models of tuberculosis. Summary: In this Review, we discuss how zebrafish (Danio rerio) and other fish models can complement the more traditional mammalian models in the development of novel vaccines against tuberculosis.
Collapse
Affiliation(s)
- Anni K Saralahti
- Laboratory of Experimental Immunology, BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere FI-33014, Finland
| | - Meri I E Uusi-Mäkelä
- Laboratory of Experimental Immunology, BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere FI-33014, Finland
| | - Mirja T Niskanen
- Laboratory of Experimental Immunology, BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere FI-33014, Finland
| | - Mika Rämet
- Laboratory of Experimental Immunology, BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere FI-33014, Finland .,Vaccine Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere FI-33014, Finland.,PEDEGO Research Unit, Medical Research Center, University of Oulu, Oulu FI-90014, Finland.,Department of Children and Adolescents, Oulu University Hospital, Oulu FI-90029, Finland
| |
Collapse
|
31
|
Smith AA, Villarreal-Ramos B, Mendum TA, Williams KJ, Jones GJ, Wu H, McFadden J, Vordermeier HM, Stewart GR. Genetic screening for the protective antigenic targets of BCG vaccination. Tuberculosis (Edinb) 2020; 124:101979. [PMID: 32814303 DOI: 10.1016/j.tube.2020.101979] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/10/2020] [Accepted: 07/13/2020] [Indexed: 11/18/2022]
Abstract
Bovine tuberculosis is an important animal health problem and the predominant cause of zoonotic tuberculosis worldwide. It results in serious economic burden due to losses in productivity and the cost of control programmes. Control could be greatly improved by the introduction of an efficacious cattle vaccine but the most likely candidate, BCG, has several limitations including variable efficacy. Augmentation of BCG with a subunit vaccine booster has been shown to increase protection but the selection of antigens has hitherto been left largely to serendipity. In the present study, we take a rational approach to identify the protective antigens of BCG, selecting a BCG transposon mutant library in naïve and BCG-vaccinated cattle. Ten mutants had increased relative survival in vaccinated compared to naïve cattle, consistent with loss of protective antigen targets making the mutants less visible to the BCG immune response. The immunogenicity of three putative protective antigens, BCG_0116, BCG_0205 (YrbE1B) and BCG_1448 (PPE20) was investigated using peptide pools and PBMCs from BCG vaccinated cattle. BCG vaccination induced PBMC to release elevated levels of IP10, IL-17a and IL-10 in response to all three antigens. Taken together, the data supports the further study of these antigens for use in subunit vaccines.
Collapse
MESH Headings
- Animals
- Antigens, Bacterial/administration & dosage
- Antigens, Bacterial/genetics
- Antigens, Bacterial/immunology
- BCG Vaccine/administration & dosage
- BCG Vaccine/immunology
- Cattle
- Cytokines/immunology
- Cytokines/metabolism
- DNA Transposable Elements
- Immunogenicity, Vaccine
- Leukocytes, Mononuclear/immunology
- Leukocytes, Mononuclear/metabolism
- Leukocytes, Mononuclear/microbiology
- Mutation
- Mycobacterium tuberculosis/genetics
- Mycobacterium tuberculosis/immunology
- Tuberculosis, Bovine/immunology
- Tuberculosis, Bovine/metabolism
- Tuberculosis, Bovine/microbiology
- Tuberculosis, Bovine/prevention & control
- Vaccination/veterinary
Collapse
Affiliation(s)
- Alex A Smith
- Department of Microbial Sciences, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - Bernardo Villarreal-Ramos
- Department of Bacteriology, Animal and Plant Health Agency, Weybridge, KT15 3NB, UK; Centre of Excellence for Bovine Tuberculosis, Institute for Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth, Wales, SY23 3DA, UK.
| | - Tom A Mendum
- Department of Microbial Sciences, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - Kerstin J Williams
- Department of Microbial Sciences, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - Gareth J Jones
- Department of Bacteriology, Animal and Plant Health Agency, Weybridge, KT15 3NB, UK
| | - Huihai Wu
- Department of Microbial Sciences, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - Johnjoe McFadden
- Department of Microbial Sciences, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - H Martin Vordermeier
- Department of Bacteriology, Animal and Plant Health Agency, Weybridge, KT15 3NB, UK; Centre of Excellence for Bovine Tuberculosis, Institute for Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth, Wales, SY23 3DA, UK.
| | - Graham R Stewart
- Department of Microbial Sciences, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK.
| |
Collapse
|
32
|
Li J, Zhao A, Tang J, Wang G, Shi Y, Zhan L, Qin C. Tuberculosis vaccine development: from classic to clinical candidates. Eur J Clin Microbiol Infect Dis 2020; 39:1405-1425. [PMID: 32060754 PMCID: PMC7223099 DOI: 10.1007/s10096-020-03843-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 02/05/2020] [Indexed: 12/12/2022]
Abstract
Bacillus Calmette-Guérin (BCG) has been in use for nearly 100 years and is the only licensed TB vaccine. While BCG provides protection against disseminated TB in infants, its protection against adult pulmonary tuberculosis (PTB) is variable. To achieve the ambitious goal of eradicating TB worldwide by 2050, there is an urgent need to develop novel TB vaccines. Currently, there are more than a dozen novel TB vaccines including prophylactic and therapeutic at different stages of clinical research. This literature review provides an overview of the clinical status of candidate TB vaccines and discusses the challenges and future development trends of novel TB vaccine research in combination with the efficacy of evaluation of TB vaccines, provides insight for the development of safer and more efficient vaccines, and may inspire new ideas for the prevention of TB.
Collapse
Affiliation(s)
- Junli Li
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, 100021, People's Republic of China
- Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious, Beijing, 100021, People's Republic of China
- Key Laboratory of Human Diseases Animal Model, State Administration of Traditional Chinese Medicine, Beijing, 100021, People's Republic of China
- Tuberculosis Center, Chinese Academy of Medical Sciences (CAMS), Beijing, 100021, People's Republic of China
| | - Aihua Zhao
- Division of Tuberculosis Vaccines, National Institutes for Food and Drug Control (NIFDC), Beijing, 102629, People's Republic of China
| | - Jun Tang
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, 100021, People's Republic of China
- Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious, Beijing, 100021, People's Republic of China
- Key Laboratory of Human Diseases Animal Model, State Administration of Traditional Chinese Medicine, Beijing, 100021, People's Republic of China
- Tuberculosis Center, Chinese Academy of Medical Sciences (CAMS), Beijing, 100021, People's Republic of China
| | - Guozhi Wang
- Division of Tuberculosis Vaccines, National Institutes for Food and Drug Control (NIFDC), Beijing, 102629, People's Republic of China
| | - Yanan Shi
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, 100021, People's Republic of China
- Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious, Beijing, 100021, People's Republic of China
- Key Laboratory of Human Diseases Animal Model, State Administration of Traditional Chinese Medicine, Beijing, 100021, People's Republic of China
- Tuberculosis Center, Chinese Academy of Medical Sciences (CAMS), Beijing, 100021, People's Republic of China
| | - Lingjun Zhan
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, 100021, People's Republic of China.
- Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious, Beijing, 100021, People's Republic of China.
- Key Laboratory of Human Diseases Animal Model, State Administration of Traditional Chinese Medicine, Beijing, 100021, People's Republic of China.
- Tuberculosis Center, Chinese Academy of Medical Sciences (CAMS), Beijing, 100021, People's Republic of China.
| | - Chuan Qin
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, 100021, People's Republic of China.
- Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious, Beijing, 100021, People's Republic of China.
- Key Laboratory of Human Diseases Animal Model, State Administration of Traditional Chinese Medicine, Beijing, 100021, People's Republic of China.
- Tuberculosis Center, Chinese Academy of Medical Sciences (CAMS), Beijing, 100021, People's Republic of China.
| |
Collapse
|
33
|
Balseiro A, Thomas J, Gortázar C, Risalde MA. Development and Challenges in Animal Tuberculosis Vaccination. Pathogens 2020; 9:pathogens9060472. [PMID: 32549360 PMCID: PMC7350370 DOI: 10.3390/pathogens9060472] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/11/2020] [Accepted: 06/12/2020] [Indexed: 02/06/2023] Open
Abstract
Vaccination with Bacillus Calmette-Guérin (BCG) constituted a major advance in the prevention of human tuberculosis (TB) in the beginning of the past century. BCG has also a clear potential for use in animals and, in particular, in the main domestic species subjected to TB control programs, cattle. Nowadays, the use of BCG vaccination against TB in cattle is not permitted by European Union legislation because BCG can induce a cellular immune response producing diagnostic interference in the eradication programs based on tuberculin single and comparative intradermal tests imposed worldwide. In this review we recall the history of TB vaccination as well as different vaccine trials and the response to vaccination in both domestic and wild animals. Promising potential inactivated vaccines are also reviewed. Research studies are mainly focused to improve vaccine efficacy, and at the same time to ensure its easy administration, safety and stability in the environment. Great challenges remain, particularly in terms of vaccine candidates and also in the acceptance of vaccination. Vaccination should be included in a strategic plan for integrated control of TB under a "one health" perspective, which also includes other measures such as improved biosafety on farms to avoid or decrease contact between domestic and wild animals or control of wildlife reservoirs to avoid overabundance that may favor infection maintenance.
Collapse
Affiliation(s)
- Ana Balseiro
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad de León, 24071 León, Spain
- Departamento de Sanidad Animal, Instituto de Ganadería de Montaña (CSIC-Universidad de León), Finca Marzanas, Grulleros, 24346 León, Spain
- Correspondence: ; Tel.: +34-98-729-1331
| | - Jobin Thomas
- SaBio-Instituto de Investigación en Recursos Cinegéticos IREC (UCLM-CSIC-JCCM), Universidad de Castilla-la Mancha (UCLM), 13071 Ciudad Real, Spain; (J.T.); (C.G.)
- Indian Council of Agricultural Research (ICAR), New Delhi 110001, India
| | - Christian Gortázar
- SaBio-Instituto de Investigación en Recursos Cinegéticos IREC (UCLM-CSIC-JCCM), Universidad de Castilla-la Mancha (UCLM), 13071 Ciudad Real, Spain; (J.T.); (C.G.)
| | - María A. Risalde
- Departamento de Anatomía y Anatomía Patológica Comparadas y Toxicología. Facultad de Veterinaria. Universidad de Córdoba (UCO), 14014 Córdoba, Spain;
- Unidad de Enfermedades Infecciosas, Grupo de Virología Clínica y Zoonosis, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Reina Sofía, Universidad de Córdoba (UCO), 14004 Córdoba, Spain
| |
Collapse
|
34
|
Lyashchenko KP, Vordermeier HM, Waters WR. Memory B cells and tuberculosis. Vet Immunol Immunopathol 2020; 221:110016. [PMID: 32050091 DOI: 10.1016/j.vetimm.2020.110016] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/30/2019] [Accepted: 01/29/2020] [Indexed: 02/09/2023]
Abstract
Immunological memory is a central feature of adaptive immunity. Memory B cells are generated upon stimulation with antigen presented by follicular dendritic cells in the peripheral lymphoid tissues. This process typically involves class-switch recombination and somatic hypermutation and it can be dependent or independent on germinal centers or T cell help. The mature B cell memory pool is generally characterized by remarkable heterogeneity of functionally and phenotypically distinct sub-populations supporting multi-layer immune plasticity. Memory B cells found in human patients infected with Mycobacterium tuberculosis include IgD+ CD27+ and IgM+ CD27+ subsets. In addition, expansion of atypical memory B cells characterized by the lack of CD27 expression and by inability to respond to antigen-induced re-activation is documented in human tuberculosis. These functionally impaired memory B cells are believed to have adverse effects on host immunity. Human and animal studies demonstrate recruitment of antigen-activated B cells to the infection sites and their presence in lung granulomas where proliferating B cells are organized into discrete clusters resembling germinal centers of secondary lymphoid organs. Cattle studies show development of IgM+, IgG+, and IgA+ memory B cells in M. bovis infection with the ability to rapidly differentiate into antibody-producing plasma cells upon antigen re-exposure. This review discusses recent advances in research on generation, re-activation, heterogeneity, and immunobiological functions of memory B cells in tuberculosis. The role of memory B cells in post-skin test recall antibody responses in bovine tuberculosis and implications for development of improved immunodiagnostics are also reviewed.
Collapse
Affiliation(s)
| | - H Martin Vordermeier
- Tuberculosis Research Group, Animal and Plant Health Agency, Addlestone, United Kingdom; Institute for Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth, United Kingdom
| | - W Ray Waters
- National Animal Disease Center, Agricultural Research Service, US Department of Agriculture, Ames, IA, USA
| |
Collapse
|
35
|
Palmer MV, Thacker TC, Rabideau MM, Jones GJ, Kanipe C, Vordermeier HM, Ray Waters W. Biomarkers of cell-mediated immunity to bovine tuberculosis. Vet Immunol Immunopathol 2019; 220:109988. [PMID: 31846797 DOI: 10.1016/j.vetimm.2019.109988] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 11/15/2019] [Accepted: 11/28/2019] [Indexed: 10/25/2022]
Abstract
Whole blood based assays, particularly interferon gamma (IFN-γ) release assays (IGRAs), are used for the diagnosis of both bovine and human tuberculosis (TB). The aim of the current study was to evaluate a panel of cytokines and chemokines for potential use as diagnostic readouts indicative of Mycobacterium bovis (M. bovis) infection in cattle. A gene expression assay was used to determine the kinetics of the response to M. bovis purified protein derivative and a fusion protein consisting of ESAT-6, CFP10, and Rv3615c upon aerosol infection with ∼104 cfu of M. bovis. The panel of biomarkers included: IFN-γ, CXCL9, CXCL10, CCL2, CCL3, TNF-α, IL-1α, IL-1β, IL-1Ra, IL-22, IL-21 and IL-13. Protein levels of IFN-γ, CXCL9, and CXCL10 were determined by ELISA. Findings suggest that CXCL9, CXCL10, IL-21, IL-13, and several acute phase cytokines may be worth pursuing as diagnostic biomarkers of M. bovis infection in cattle.
Collapse
Affiliation(s)
- Mitchell V Palmer
- National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture (USDA), Ames, Iowa, USA.
| | - Tyler C Thacker
- National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture (USDA), Ames, Iowa, USA
| | - Meaghan M Rabideau
- National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture (USDA), Ames, Iowa, USA
| | - Gareth J Jones
- TB Immunology and Vaccinology, Department of Bacteriology, Animal and Plant Health Agency, New Haw, Addlestone, Surry UK
| | - Carly Kanipe
- National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture (USDA), Ames, Iowa, USA; Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, Iowa, USA
| | - H Martin Vordermeier
- TB Immunology and Vaccinology, Department of Bacteriology, Animal and Plant Health Agency, New Haw, Addlestone, Surry UK
| | - W Ray Waters
- National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture (USDA), Ames, Iowa, USA
| |
Collapse
|
36
|
Biffar L, Blunt L, Atkins W, Anderson P, Holder T, Xing Z, Vordermeier M, McShane H, Villarreal-Ramos B. Evaluating the sensitivity of the bovine BCG challenge model using a prime boost Ad85A vaccine regimen. Vaccine 2019; 38:1241-1248. [PMID: 31759733 DOI: 10.1016/j.vaccine.2019.11.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 10/30/2019] [Accepted: 11/04/2019] [Indexed: 10/25/2022]
Abstract
In the absence of biomarkers of protective immunity, newly developed vaccines against bovine tuberculosis need to be evaluated in virulent Mycobacterium bovis challenge experiments, which require the use of expensive and highly in demand Biological Safety Level 3 (BSL3) animal facilities. The recently developed bovine BCG challenge model offers a cheaper and faster way to test new vaccine candidates and additionally reduces the severity of the challenge compared to virulent M. bovis challenge in line with the remits of the NC3Rs. In this work we sought to establish the sensitivity of the BCG challenge model by testing a prime boost vaccine regimen that previously increased protection over BCG alone against M. bovis challenge. All animals, except the control group, were vaccinated subcutaneously with BCG Danish, and half of those were then boosted with a recombinant adenoviral vector expressing Antigen 85A, Ad85A. All animals were challenged with BCG Tokyo into the prescapular lymph node and the bacterial load within the lymph nodes was established. All vaccinated animals, independent of the vaccination regimen, cleared BCG significantly faster from the lymph node than control animals, suggesting a protective effect. There was however, no difference between the BCG and the BCG-Ad85A regimens. Additionally, we analysed humoral and cellular immune responses taken prior to challenge for possible predictors of protection. Cultured ELISpot identified significantly higher IFN-ɣ responses in protected vaccinated animals, relative to controls, but not in unprotected vaccinated animals. Furthermore, a trend for protected animals to produce more IFN-ɣ by quantitative PCR and intracellular staining was observed. Thus, this model can also be an attractive alternative to M. bovis challenge models for the discovery of protective biomarkers.
Collapse
Affiliation(s)
- Lucia Biffar
- Jenner Institute Oxford, Old Road Campus Research Build, Roosevelt Dr, Oxford OX3 7DQ, UK; TB Immunology and Vaccinology Team, Department of Bacteriology, Animal and Plant Health Agency, Weybridge, Surrey KT15 3NB, UK
| | - Laura Blunt
- TB Immunology and Vaccinology Team, Department of Bacteriology, Animal and Plant Health Agency, Weybridge, Surrey KT15 3NB, UK
| | - William Atkins
- TB Immunology and Vaccinology Team, Department of Bacteriology, Animal and Plant Health Agency, Weybridge, Surrey KT15 3NB, UK
| | - Paul Anderson
- TB Immunology and Vaccinology Team, Department of Bacteriology, Animal and Plant Health Agency, Weybridge, Surrey KT15 3NB, UK
| | - Tom Holder
- TB Immunology and Vaccinology Team, Department of Bacteriology, Animal and Plant Health Agency, Weybridge, Surrey KT15 3NB, UK
| | - Zhou Xing
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Martin Vordermeier
- TB Immunology and Vaccinology Team, Department of Bacteriology, Animal and Plant Health Agency, Weybridge, Surrey KT15 3NB, UK; Institute of Biological, Environmental & Rural Sciences (IBERS), Aberystwyth University, Penglais, Aberystwyth, Ceredigion SY23 3DA, UK
| | - Helen McShane
- Jenner Institute Oxford, Old Road Campus Research Build, Roosevelt Dr, Oxford OX3 7DQ, UK
| | - Bernardo Villarreal-Ramos
- TB Immunology and Vaccinology Team, Department of Bacteriology, Animal and Plant Health Agency, Weybridge, Surrey KT15 3NB, UK; Institute of Biological, Environmental & Rural Sciences (IBERS), Aberystwyth University, Penglais, Aberystwyth, Ceredigion SY23 3DA, UK.
| |
Collapse
|
37
|
Characterization of local and circulating bovine γδ T cell responses to respiratory BCG vaccination. Sci Rep 2019; 9:15996. [PMID: 31690788 PMCID: PMC6831659 DOI: 10.1038/s41598-019-52565-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 10/21/2019] [Indexed: 12/14/2022] Open
Abstract
The Mycobacterium bovis Bacillus Calmette-Guerin (BCG) vaccine is administered parenterally to infants and young children to prevent tuberculosis (TB) infection. However, the protection induced by BCG is highly variable and the vaccine does not prevent pulmonary TB, the most common form of the illness. Until improved TB vaccines are available, it is crucial to use BCG in a manner which ensures optimal vaccine performance. Immunization directly to the respiratory mucosa has been shown to promote greater protection from TB in animal models. γδ T cells play a major role in host defense at mucosal sites and are known to respond robustly to mycobacterial infection. Their positioning in the respiratory mucosa ensures their engagement in the response to aerosolized TB vaccination. However, our understanding of the effect of respiratory BCG vaccination on γδ T cell responses in the lung is unknown. In this study, we used a calf model to investigate the immunogenicity of aerosol BCG vaccination, and the phenotypic profile of peripheral and mucosal γδ T cells responding to vaccination. We observed robust local and systemic M. bovis-specific IFN-γ and IL-17 production by both γδ and CD4 T cells. Importantly, BCG vaccination induced effector and memory cell differentiation of γδ T cells in both the lower airways and peripheral blood, with accumulation of a large proportion of effector memory γδ T cells in both compartments. Our results demonstrate the potential of the neonatal calf model to evaluate TB vaccine candidates that are to be administered via the respiratory tract, and suggest that aerosol immunization is a promising strategy for engaging γδ T cells in vaccine-induced immunity against TB.
Collapse
|
38
|
Risalde MA, Roy Á, Bezos J, Pineda C, Casal C, Díez-Guerrier A, Lopez-Villalba I, Fernández-Manzano Á, Moreno I, De Juan L, Domínguez L, Gortazar C. Hypervitaminosis D has no positive effects on goat tuberculosis and may cause chronic renal lesions. Vet Rec 2019; 185:759. [PMID: 31690642 DOI: 10.1136/vr.105411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 08/12/2019] [Accepted: 10/18/2019] [Indexed: 11/03/2022]
Abstract
BACKGROUND There is evidence for a link between vitamin D deficiency and active tuberculosis (TB). In human beings, several trials have evaluated the role of vitamin D supplementation in TB treatment with conflicting results. However, the role of vitamin D supplementation in animal TB control has received less attention. The authors evaluated the benefit of vitamin D supplementation for preventing mycobacterial infection or reducing TB lesions (TBL) in a controlled trial with goats naturally exposed to Mycobacterium caprae. METHODS Two groups of goats, a vitamin D-supplemented group and a non-supplemented control group, were housed for 10 months in direct contact with M caprae-infected adult goats. Upon contact with the infected adult goats, all animals were TB-tested every two months. RESULTS No experimental evidence of a protective effect of vitamin D supplementation based on M caprae culture prevalence, TBL prevalence, median TBL score or the proportion of single versus multiple organs presenting TBL was observed. CONCLUSION The results indicate that, in the conditions used in this study, vitamin D supplementation in goats does not reduce TB infection risk nor the diffusion and severity of TBL. In addition, vitamin D-supplemented goats presented hyperphosphataemia and renal injury with calcifications suggestive of vitamin D intoxication.
Collapse
Affiliation(s)
- Maria Angeles Risalde
- Department of Comparative Pathology, University of Córdoba-Agrifood Campus of International Excellence (ceiA3), Cordoba, Spain.,Infectious Diseases Unit. Instituto Maimonides de Investigación Biomédica de Córdoba (IMIBIC). Hospital Universitario Reina Sofía de Córdoba - Universidad de Córdoba, Córdoba, Spain
| | - Álvaro Roy
- Centro de Vigilancia Sanitaria Veterinaria (VISAVET), Universidad Complutense, Madrid, Spain.,CZ Vaccines, Porriño, Pontevedra, Spain
| | - Javier Bezos
- Centro de Vigilancia Sanitaria Veterinaria (VISAVET), Universidad Complutense, Madrid, Spain.,Dpto. de Sanidad Animal, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain
| | - Carmen Pineda
- Departamento de Medicina y Cirugía Animal, Universidad de Córdoba, Córdoba, Spain
| | - Carmen Casal
- Laboratorios SYVA S.A.U. Technological Park, León, Spain
| | - Alberto Díez-Guerrier
- Dpto. de Sanidad Animal, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain.,MAEVA SERVET S.L, Alameda del Valle, Madrid, Spain
| | | | | | - Inmaculada Moreno
- Servicio de Inmunología Microbiana, Centro Nacional de Microbiología, Instituto de Investigación Carlos III, Majadahonda, Madrid
| | - Lucía De Juan
- Centro de Vigilancia Sanitaria Veterinaria (VISAVET), Universidad Complutense, Madrid, Spain.,Dpto. de Sanidad Animal, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain
| | - Lucas Domínguez
- Centro de Vigilancia Sanitaria Veterinaria (VISAVET), Universidad Complutense, Madrid, Spain.,Dpto. de Sanidad Animal, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain
| | - Christian Gortazar
- SaBio (Health and Biotechnology), Instituto de Investigación en Recursos Cinegéticos IREC (CSIC-UCLM), Ciudad Real, Spain
| |
Collapse
|
39
|
IL-10 Dampens the Th1 and Tc Activation through Modulating DC Functions in BCG Vaccination. Mediators Inflamm 2019; 2019:8616154. [PMID: 31281230 PMCID: PMC6594250 DOI: 10.1155/2019/8616154] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 05/08/2019] [Indexed: 02/08/2023] Open
Abstract
BCG, the only registered vaccine against Mycobacterial Tuberculosis (TB) infection, has been questioned for its protective efficacy for decades. Although lots of efforts were made to improve the BCG antigenicity, few studies were devoted to understand the role of host factors in the variability of the BCG protection. Using the IL-10KO mice and pulmonary tuberculosis infection model, we have addressed the role of IL-10 in the BCG vaccination efficacy. The data showed that IL-10-deficient dendritic cells (DCs) could promote the immune responses through upregulation of the surface costimulatory molecule expression and play an orchestra role through activating CD4+T cell. IL-10-deficient mice had higher IFN γ, TNF α, and IL-6 production after BCG vaccination, which was consistent with the higher proportion of IFN γ+CD3+, IFN γ+CD4+, and IFN γ+CD8+ T cells in the spleen. Particularly, the BCG-vaccinated IL-10KO mice showed less inflammation after TB challenge compared to WT mice, which was supported by the promoted Th1 and Tc, as well as the downregulated Treg responses in IL-10 deficiency. In a conclusion, we demonstrated the negative relationship between Th1/Tc responses with IL-10 production. IL-10 deficiency restored the type 1 immune response through DC activation, which provided better protection against TB infection. Hence, our study offers the first experimental evidence that, contrary to the modulation of BCG, host immunity plays a critical role in the BCG protective efficacy against TB.
Collapse
|
40
|
Kilpeläinen A, Saubi N, Guitart N, Moyo N, Wee EG, Ravi K, Hanke T, Joseph J. Priming With Recombinant BCG Expressing Novel HIV-1 Conserved Mosaic Immunogens and Boosting With Recombinant ChAdOx1 Is Safe, Stable, and Elicits HIV-1-Specific T-Cell Responses in BALB/c Mice. Front Immunol 2019; 10:923. [PMID: 31156614 PMCID: PMC6530512 DOI: 10.3389/fimmu.2019.00923] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 04/10/2019] [Indexed: 11/13/2022] Open
Abstract
BCG is currently the only licensed vaccine against tuberculosis (TB) and confers protection against meningitis and miliary tuberculosis in infants, although pulmonary disease protection in adults is inconsistent. Recently, promising HIV-1 immunogens were developed, such as the T-cell immunogens "tHIVconsvX," designed using functionally conserved protein regions across group M strains, with mosaic immunogens to improve HIV-1 variant match and response breadth. In this study, we constructed an integrative E. coli-mycobacterial shuttle plasmid, p2auxo.HIVconsvXint, expressing the immunogens HIVconsv1&2. This expression vector used an antibiotic resistance-free mechanism for plasmid selection and maintenance. It was first transformed into a glycine auxotrophic E. coli strain and subsequently transformed into a lysine auxotrophic Mycobacterium bovis BCG strain to generate vaccines BCG.HIVconsv12auxo.int and BCG.HIVconsv22auxo.int. The DNA sequence coding for the HIVconsv1&2 immunogens and protein expression were confirmed and working vaccine stocks were genetically and phenotypically characterized. We demonstrated that BCG.HIVconsv1&22auxo.int in combination with ChAdOx1.tHIVconsv5&6 were well tolerated and induced HIV-1-specific T-cell responses in adult BALB/c mice. In addition, we showed that the BCG.HIVconsv1&22auxo.int vaccine strains were stable in vitro after 35 bacterial generations and in vivo 7 weeks after inoculation. The use of integrative expression vectors and novel HIV-1 immunogens are likely to have improved the mycobacterial vaccine stability and specific immunogenicity and may enable the development of a useful vaccine platform for priming protective responses against HIV-1/TB and other prevalent pediatric pathogens shortly following birth.
Collapse
Affiliation(s)
- Athina Kilpeläinen
- AIDS Research Unit, Infectious Diseases Department, Catalan Center for HIV Vaccine Research and Development, Hospital Clínic/IDIBAPS, Barcelona, Spain
| | - Narcís Saubi
- AIDS Research Unit, Infectious Diseases Department, Catalan Center for HIV Vaccine Research and Development, Hospital Clínic/IDIBAPS, Barcelona, Spain
| | - Núria Guitart
- AIDS Research Unit, Infectious Diseases Department, Catalan Center for HIV Vaccine Research and Development, Hospital Clínic/IDIBAPS, Barcelona, Spain
| | - Nathifa Moyo
- The Jenner Institute Laboratories, University of Oxford, Oxford, United Kingdom
| | - Edmund G. Wee
- The Jenner Institute Laboratories, University of Oxford, Oxford, United Kingdom
| | - Krupa Ravi
- The Jenner Institute Laboratories, University of Oxford, Oxford, United Kingdom
| | - Tomáš Hanke
- The Jenner Institute Laboratories, University of Oxford, Oxford, United Kingdom
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Joan Joseph
- AIDS Research Unit, Infectious Diseases Department, Catalan Center for HIV Vaccine Research and Development, Hospital Clínic/IDIBAPS, Barcelona, Spain
| |
Collapse
|
41
|
Lee D, Chambers M. A bilayer tissue culture model of the bovine alveolus. F1000Res 2019; 8:357. [PMID: 31448101 PMCID: PMC6685456 DOI: 10.12688/f1000research.18696.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/27/2019] [Indexed: 08/02/2024] Open
Abstract
The epithelial lining of the lung is often the first point of interaction between the host and inhaled pathogens, allergens and medications. Epithelial cells are therefore the main focus of studies which aim to shed light on host-pathogen interactions, to dissect the mechanisms of local host immunity and study toxicology. If these studies are not to be conducted exclusively in vivo, it is imperative that in vitro models are developed with a high in vitro- in vivo correlation. We describe here a co-culture bilayer model of the bovine alveolus, designed to overcome some of the limitations encountered with mono-culture and live animal models. Our system includes bovine pulmonary arterial endothelial cells (BPAECs) seeded onto a permeable membrane in 24 well Transwell format. The BPAECs are overlaid with immortalised bovine alveolar type II epithelial cells and the bilayer cultured at air-liquid interface for 14 days before use; in our case to study host-mycobacterial interactions. Characterisation of novel cell lines and the bilayer model have provided compelling evidence that immortalised bovine alveolar type II cells are an authentic substitute for primary alveolar type II cells and their culture as a bilayer in conjunction with BPAECs provides a physiologically relevant in vitro model of the bovine alveolus. The bilayer model may be used to study dynamic intracellular and extracellular host-pathogen interactions, using proteomics, genomics, live cell imaging, in-cell ELISA and confocal microscopy. The model presented in this article enables other researchers to establish an in vitro model of the bovine alveolus that is easy to set up, malleable and serves as a comparable alternative to in vivo models, whilst allowing study of early host-pathogen interactions, currently not feasible in vivo. The model therefore achieves one of the 3Rs objectives in that it replaces the use of animals in research of bovine respiratory diseases.
Collapse
Affiliation(s)
- Diane Lee
- School of Veterinary Medicine, University of Surrey, Guildford, Surrey, GU2 7AL, UK
| | - Mark Chambers
- School of Veterinary Medicine, University of Surrey, Guildford, Surrey, GU2 7AL, UK
| |
Collapse
|
42
|
Abstract
The epithelial lining of the lung is often the first point of interaction between the host and inhaled pathogens, allergens and medications. Epithelial cells are therefore the main focus of studies which aim to shed light on host-pathogen interactions, to dissect the mechanisms of local host immunity and study toxicology. If these studies are not to be conducted exclusively in vivo, it is imperative that in vitro models are developed with a high in vitro- in vivo correlation. We describe here a co-culture model of the bovine alveolus, designed to overcome some of the limitations encountered with mono-culture and live animal models. Our system includes bovine pulmonary arterial endothelial cells (BPAECs) seeded onto a permeable membrane in 24 well Transwell format. The BPAECs are overlaid with immortalised bovine alveolar type II epithelial cells and cultured at air-liquid interface for 14 days before use; in our case to study host-mycobacterial interactions. Characterisation of novel cell lines and the co-culture model have provided compelling evidence that immortalised bovine alveolar type II cells are an authentic substitute for primary alveolar type II cells and their co-culture with BPAECs provides a physiologically relevant in vitro model of the bovine alveolus. The co-culture model may be used to study dynamic intracellular and extracellular host-pathogen interactions, using proteomics, genomics, live cell imaging, in-cell ELISA and confocal microscopy. The model presented in this article enables other researchers to establish an in vitro model of the bovine alveolus that is easy to set up, malleable and serves as a comparable alternative to in vivo models, whilst allowing study of early host-pathogen interactions, currently not feasible in vivo. The model therefore achieves one of the 3Rs objectives in that it replaces the use of animals in research of bovine respiratory diseases.
Collapse
Affiliation(s)
- Diane Lee
- School of Veterinary Medicine, University of Surrey, Guildford, Surrey, GU2 7AL, UK
| | - Mark Chambers
- School of Veterinary Medicine, University of Surrey, Guildford, Surrey, GU2 7AL, UK
| |
Collapse
|
43
|
Reid MJA, Arinaminpathy N, Bloom A, Bloom BR, Boehme C, Chaisson R, Chin DP, Churchyard G, Cox H, Ditiu L, Dybul M, Farrar J, Fauci AS, Fekadu E, Fujiwara PI, Hallett TB, Hanson CL, Harrington M, Herbert N, Hopewell PC, Ikeda C, Jamison DT, Khan AJ, Koek I, Krishnan N, Motsoaledi A, Pai M, Raviglione MC, Sharman A, Small PM, Swaminathan S, Temesgen Z, Vassall A, Venkatesan N, van Weezenbeek K, Yamey G, Agins BD, Alexandru S, Andrews JR, Beyeler N, Bivol S, Brigden G, Cattamanchi A, Cazabon D, Crudu V, Daftary A, Dewan P, Doepel LK, Eisinger RW, Fan V, Fewer S, Furin J, Goldhaber-Fiebert JD, Gomez GB, Graham SM, Gupta D, Kamene M, Khaparde S, Mailu EW, Masini EO, McHugh L, Mitchell E, Moon S, Osberg M, Pande T, Prince L, Rade K, Rao R, Remme M, Seddon JA, Selwyn C, Shete P, Sachdeva KS, Stallworthy G, Vesga JF, Vilc V, Goosby EP. Building a tuberculosis-free world: The Lancet Commission on tuberculosis. Lancet 2019; 393:1331-1384. [PMID: 30904263 DOI: 10.1016/s0140-6736(19)30024-8] [Citation(s) in RCA: 240] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 12/20/2018] [Accepted: 12/25/2018] [Indexed: 11/22/2022]
Affiliation(s)
- Michael J A Reid
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA; Institute for Global Health Sciences, University of California San Francisco, San Francisco, CA, USA.
| | - Nimalan Arinaminpathy
- School of Public Health, Imperial College London, London, UK; Faculty of Medicine, Imperial College London, London, UK
| | - Amy Bloom
- Tuberculosis Division, United States Agency for International Development, Washington, DC, USA
| | - Barry R Bloom
- Department of Global Health and Population, Harvard University, Cambridge, MA, USA
| | | | - Richard Chaisson
- Departments of Medicine, Epidemiology, and International Health, Johns Hopkins School of Medicine, Baltimore, MA, USA
| | | | | | - Helen Cox
- Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | | | - Mark Dybul
- Department of Medicine, Centre for Global Health and Quality, Georgetown University, Washington, DC, USA
| | | | - Anthony S Fauci
- National Institute of Allergy and Infectious Diseases, US National Institutes of Health, Maryland, MA, USA
| | | | - Paula I Fujiwara
- Department of Tuberculosis and HIV, The International Union Against Tuberculosis and Lung Disease, Paris, France
| | - Timothy B Hallett
- School of Public Health, Imperial College London, London, UK; Faculty of Medicine, Imperial College London, London, UK
| | | | | | - Nick Herbert
- Global TB Caucus, Houses of Parliament, London, UK
| | - Philip C Hopewell
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Chieko Ikeda
- Department of GLobal Health, Ministry of Heath, Labor and Welfare, Tokyo, Japan
| | - Dean T Jamison
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA; Institute for Global Health Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Aamir J Khan
- Interactive Research & Development, Karachi, Pakistan
| | - Irene Koek
- Global Health Bureau, United States Agency for International Development, Washington, DC, USA
| | - Nalini Krishnan
- Resource Group for Education and Advocacy for Community Health, Chennai, India
| | - Aaron Motsoaledi
- South African National Department of Health, Pretoria, South Africa
| | - Madhukar Pai
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, QC, Canada; McGill International TB Center, McGill University, Montreal, QC, Canada
| | - Mario C Raviglione
- University of Milan, Milan, Italy; Global Studies Institute, University of Geneva, Geneva, Switzerland
| | - Almaz Sharman
- Academy of Preventive Medicine of Kazakhstan, Almaty, Kazakhstan
| | - Peter M Small
- Global Health Institute, School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | | | - Zelalem Temesgen
- Department of Infectious Diseases, Mayo Clinic, Rochester, MI, USA
| | - Anna Vassall
- Department of Global Health and Development, Faculty of Public Health and Policy, London School of Hygiene and Tropical Medicine, London, UK; Amsterdam Institute for Global Health and Development, University of Amsterdam, Amsterdam, Netherlands
| | | | | | - Gavin Yamey
- Center for Policy Impact in Global Health, Duke Global Health Institute, Duke University, Durham, NC, USA
| | - Bruce D Agins
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA; Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Sofia Alexandru
- Institutul de Ftiziopneumologie Chiril Draganiuc, Chisinau, Moldova
| | - Jason R Andrews
- Division of Infectious Diseases and Geographic Medicine, Stanford University, Stanford, CA, USA
| | - Naomi Beyeler
- Institute for Global Health Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Stela Bivol
- Center for Health Policies and Studies, Chisinau, Moldova
| | - Grania Brigden
- Department of Tuberculosis and HIV, The International Union Against Tuberculosis and Lung Disease, Paris, France
| | - Adithya Cattamanchi
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Danielle Cazabon
- McGill International TB Center, McGill University, Montreal, QC, Canada
| | - Valeriu Crudu
- Center for Health Policies and Studies, Chisinau, Moldova
| | - Amrita Daftary
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, QC, Canada; McGill International TB Center, McGill University, Montreal, QC, Canada
| | - Puneet Dewan
- Bill & Melinda Gates Foundation, New Delhi, India
| | - Laurie K Doepel
- National Institute of Allergy and Infectious Diseases, US National Institutes of Health, Maryland, MA, USA
| | - Robert W Eisinger
- National Institute of Allergy and Infectious Diseases, US National Institutes of Health, Maryland, MA, USA
| | - Victoria Fan
- T H Chan School of Public Health, Harvard University, Cambridge, MA, USA; Office of Public Health Studies, University of Hawaii, Mānoa, HI, USA
| | - Sara Fewer
- Institute for Global Health Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Jennifer Furin
- Division of Infectious Diseases & HIV Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Jeremy D Goldhaber-Fiebert
- Centers for Health Policy and Primary Care and Outcomes Research, Stanford University, Stanford, CA, USA
| | - Gabriela B Gomez
- Department of Global Health and Development, Faculty of Public Health and Policy, London School of Hygiene and Tropical Medicine, London, UK
| | - Stephen M Graham
- Department of Tuberculosis and HIV, The International Union Against Tuberculosis and Lung Disease, Paris, France; Department of Paediatrics, Center for International Child Health, University of Melbourne, Melbourne, VIC, Australia; Burnet Institute, Melbourne, VIC, Australia
| | - Devesh Gupta
- Revised National TB Control Program, New Delhi, India
| | - Maureen Kamene
- National Tuberculosis, Leprosy and Lung Disease Program, Ministry of Health, Nairobi, Kenya
| | | | - Eunice W Mailu
- National Tuberculosis, Leprosy and Lung Disease Program, Ministry of Health, Nairobi, Kenya
| | | | - Lorrie McHugh
- Office of the Secretary-General's Special Envoy on Tuberculosis, United Nations, Geneva, Switzerland
| | - Ellen Mitchell
- International Institute of Social Studies, Erasmus University Rotterdam, The Hague, Netherland
| | - Suerie Moon
- Department of Global Health and Population, Harvard University, Cambridge, MA, USA; Global Health Centre, The Graduate Institute Geneva, Geneva, Switzerland
| | | | - Tripti Pande
- McGill International TB Center, McGill University, Montreal, QC, Canada
| | - Lea Prince
- Centers for Health Policy and Primary Care and Outcomes Research, Stanford University, Stanford, CA, USA
| | | | - Raghuram Rao
- Ministry of Health and Family Welfare, New Delhi, India
| | - Michelle Remme
- International Institute for Global Health, United Nations University, Kuala Lumpur, Malaysia
| | - James A Seddon
- Department of Medicine, Imperial College London, London, UK; Faculty of Medicine, Imperial College London, London, UK; Department of Paediatrics and Child Health, Stellenbosch University, Stellenbosch, South Africa
| | - Casey Selwyn
- Bill & Melinda Gates Foundation, Seattle, WA, USA
| | - Priya Shete
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | | | | | - Juan F Vesga
- School of Public Health, Imperial College London, London, UK; Faculty of Medicine, Imperial College London, London, UK
| | | | - Eric P Goosby
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA; Institute for Global Health Sciences, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
44
|
Sarmiento ME, Alvarez N, Chin KL, Bigi F, Tirado Y, García MA, Anis FZ, Norazmi MN, Acosta A. Tuberculosis vaccine candidates based on mycobacterial cell envelope components. Tuberculosis (Edinb) 2019; 115:26-41. [PMID: 30948174 DOI: 10.1016/j.tube.2019.01.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 01/12/2019] [Accepted: 01/16/2019] [Indexed: 12/11/2022]
Abstract
Even after decades searching for a new and more effective vaccine against tuberculosis, the scientific community is still pursuing this goal due to the complexity of its causative agent, Mycobacterium tuberculosis (Mtb). Mtb is a microorganism with a robust variety of survival mechanisms that allow it to remain in the host for years. The structure and nature of the Mtb envelope play a leading role in its resistance and survival. Mtb has a perfect machinery that allows it to modulate the immune response in its favor and to adapt to the host's environmental conditions in order to remain alive until the moment to reactivate its normal growing state. Mtb cell envelope protein, carbohydrate and lipid components have been the subject of interest for developing new vaccines because most of them are responsible for the pathogenicity and virulence of the bacteria. Many indirect evidences, mainly derived from the use of monoclonal antibodies, support the potential protective role of Mtb envelope components. Subunit and DNA vaccines, lipid extracts, liposomes and membrane vesicle formulations are some examples of technologies used, with encouraging results, to evaluate the potential of these antigens in the protective response against Mtb.
Collapse
Affiliation(s)
- M E Sarmiento
- School of Health Sciences (PPSK), Universiti Sains Malaysia (USM), 16150 Kubang Kerian, Kelantan, Malaysia
| | - N Alvarez
- Rutgers New Jersey Medical School, Public Health Research Institute, Newark, NJ, USA
| | - K L Chin
- Department of Biomedical Sciences and Therapeutic, Faculty of Medicine and Health Sciences (FPSK), Universiti Malaysia Sabah (UMS), Sabah, Malaysia
| | - F Bigi
- Institute of Biotechnology, INTA, Buenos Aires, Argentina
| | - Y Tirado
- Finlay Institute of Vaccines, La Habana, Cuba
| | - M A García
- Finlay Institute of Vaccines, La Habana, Cuba
| | - F Z Anis
- School of Health Sciences (PPSK), Universiti Sains Malaysia (USM), 16150 Kubang Kerian, Kelantan, Malaysia
| | - M N Norazmi
- School of Health Sciences (PPSK), Universiti Sains Malaysia (USM), 16150 Kubang Kerian, Kelantan, Malaysia.
| | - A Acosta
- School of Health Sciences (PPSK), Universiti Sains Malaysia (USM), 16150 Kubang Kerian, Kelantan, Malaysia.
| |
Collapse
|
45
|
Satti I, McShane H. Current approaches toward identifying a correlate of immune protection from tuberculosis. Expert Rev Vaccines 2018; 18:43-59. [PMID: 30466332 DOI: 10.1080/14760584.2019.1552140] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Mycobacterium tuberculosis kills more people than any other pathogen. Vaccination is the most cost-effective control measure for any infectious disease. Development of an effective vaccine against tuberculosis is hindered by the uncertain predictive value of preclinical animal models, incomplete understanding of protective immunity and lack of validated immune correlates of protection (COP). AREAS COVERED Here we review what is known about protective immunity against M.tb, the preclinical and clinical cohorts that can be utilized to identify COP, and COP that have been identified to date. EXPERT COMMENTARY The identification of COP would allow the rational design and development of vaccine candidates which can then be optimized and prioritized based on the induction of these immune responses. Once validated in field efficacy trials, such COP could potentially facilitate the development and licensure of vaccines, in combination with human efficacy data. The identification and validation of COP would represent a very significant advance in TB vaccine development. Every opportunity to collect samples and cohorts on which to cross-validate pre-existing COP and identify novel COP should be exploited. Furthermore, global cooperation and collaboration on such samples will ensure that the utility of such precious samples is fully exploited.
Collapse
Affiliation(s)
- Iman Satti
- a Jenner Institute, Nuffield Department of Medicine , University of Oxford , Oxford , UK
| | - Helen McShane
- a Jenner Institute, Nuffield Department of Medicine , University of Oxford , Oxford , UK
| |
Collapse
|
46
|
AlMatar M, Makky EA, AlMandeal H, Eker E, Kayar B, Var I, Köksal F. Does the Development of Vaccines Advance Solutions for Tuberculosis? Curr Mol Pharmacol 2018; 12:83-104. [PMID: 30474542 DOI: 10.2174/1874467212666181126151948] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 10/06/2018] [Accepted: 10/17/2018] [Indexed: 11/22/2022]
Abstract
BACKGROUND Mycobacterium tuberculosis (Mtb) is considered as one of the most efficacious human pathogens. The global mortality rate of TB stands at approximately 2 million, while about 8 to 10 million active new cases are documented yearly. It is, therefore, a priority to develop vaccines that will prevent active TB. The vaccines currently used for the management of TB can only proffer a certain level of protection against meningitis, TB, and other forms of disseminated TB in children; however, their effectiveness against pulmonary TB varies and cannot provide life-long protective immunity. Based on these reasons, more efforts are channeled towards the development of new TB vaccines. During the development of TB vaccines, a major challenge has always been the lack of diversity in both the antigens contained in TB vaccines and the immune responses of the TB sufferers. Current efforts are channeled on widening both the range of antigens selection and the range of immune response elicited by the vaccines. The past two decades witnessed a significant progress in the development of TB vaccines; some of the discovered TB vaccines have recently even completed the third phase (phase III) of a clinical trial. OBJECTIVE The objectives of this article are to discuss the recent progress in the development of new vaccines against TB; to provide an insight on the mechanism of vaccine-mediated specific immune response stimulation, and to debate on the interaction between vaccines and global interventions to end TB.
Collapse
Affiliation(s)
- Manaf AlMatar
- Department of Biotechnology, Institute of Natural and Applied Sciences (Fen Bilimleri Enstitusu) Cukurova University, Adana, Turkey
| | - Essam A Makky
- Department of Biotechnology, Faculty of Industrial Sciences and Technology, Universiti Malaysia Pahang (UMP), Kuantan, Malaysia
| | - Husam AlMandeal
- Freiburg Universität, Moltkestraße 90, 76133 karlsruhe Augenklinik, Germany
| | - Emel Eker
- Department of Medical Microbiology, Faculty of Medicine, Cukurova University, Adana, Turkey
| | - Begüm Kayar
- Department of Medical Microbiology, Faculty of Medicine, Cukurova University, Adana, Turkey
| | - Işıl Var
- Department of Food Engineering, Agricultural Faculty, Cukurova University, Adana, Turkey
| | - Fatih Köksal
- Department of Medical Microbiology, Faculty of Medicine, Cukurova University, Adana, Turkey
| |
Collapse
|
47
|
Targeting the Mincle and TLR3 receptor using the dual agonist cationic adjuvant formulation 9 (CAF09) induces humoral and polyfunctional memory T cell responses in calves. PLoS One 2018; 13:e0201253. [PMID: 30063728 PMCID: PMC6067743 DOI: 10.1371/journal.pone.0201253] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 07/11/2018] [Indexed: 12/29/2022] Open
Abstract
There is a need for the rational design of safe and effective vaccines to protect against chronic bacterial pathogens such as Mycobacterium tuberculosis and Mycobacterium avium subsp. paratuberculosis in a number of species. One of the main challenges for vaccine development is the lack of safe adjuvants that induce protective immune responses. Cationic Adjuvant Formulation 01 (CAF01)—an adjuvant based on trehalose dibehenate (TDB) and targeting the Mincle receptor—has entered human trials based on promising pre-clinical results in a number of species. However, in cattle CAF01 only induces weak systemic immune responses. In this study, we tested the ability of three pattern recognition receptors, either alone or in combination, to activate bovine monocytes and macrophages. We found that addition of the TLR3 agonist, polyinosinic:polycytidylic acid (Poly(I:C)) to either one of the Mincle receptor agonists, TDB or monomycoloyl glycerol (MMG), enhanced monocyte activation, and calves vaccinated with CAF09 containing MMG and Poly(I:C) had increased cell-mediated and humoral immune response compared to CAF01 vaccinated animals. In contrast to the highly reactogenic Montanide ISA 61 VG, CAF09-primed T cells maintained a higher frequency of polyfunctional CD4+ T cells (IFN-γ+ TNF-α+ IL-2+). In conclusion, CAF09 supports the development of antibodies along with a high-quality cell-mediated immune response and is a promising alternative to oil-in-water adjuvant in cattle and other ruminants.
Collapse
|
48
|
Wernike K, Mundt A, Link EK, Aebischer A, Schlotthauer F, Sutter G, Fux R, Beer M. N-terminal domain of Schmallenberg virus envelope protein Gc delivered by recombinant equine herpesvirus type 1 and modified vaccinia virus Ankara: Immunogenicity and protective efficacy in cattle. Vaccine 2018; 36:5116-5123. [PMID: 30049630 DOI: 10.1016/j.vaccine.2018.07.047] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 07/11/2018] [Accepted: 07/18/2018] [Indexed: 01/08/2023]
Abstract
Schmallenberg virus (SBV), which emerged in 2011 in Central Europe and subsequently spread very rapidly throughout the continent, affects predominantly ruminants. SBV is transmitted by insect vectors, and therefore vaccination is one of the major tools of disease control. Only recently, a domain connected to virus neutralization has been identified at the amino-terminal part of the viral envelope protein Gc. Here, this Gc domain delivered by recombinant EHV-1 or MVA vector viruses was tested in a vaccination-challenge trial in cattle, one of the major target species of SBV. The EHV-1-based vaccine conferred protection in two of four animals, whereas immunization using the MVA vector vaccine efficiently induced an SBV-specific antibody response and full protection against SBV challenge infection in all the vaccinated animals. Moreover, due to the absence of antibodies against SBVs N-protein, both vector vaccines enable the differentiation between vaccinated and field-infected animals making them to a promising tool to control SBV spread as well as to prevent disease in domestic ruminants.
Collapse
Affiliation(s)
- Kerstin Wernike
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Südufer 10, 17493 Greifswald - Insel Riems, Germany.
| | - Alice Mundt
- Boehringer Ingelheim Veterinary Research Centre, Bemeroder Str. 31, 30559 Hannover, Germany
| | - Ellen Kathrin Link
- Institute for Infectious Diseases and Zoonoses, Department of Veterinary Sciences, Ludwig-Maximilians-Universität, Veterinärstraße 13, 80539 Munich, Germany
| | - Andrea Aebischer
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Südufer 10, 17493 Greifswald - Insel Riems, Germany
| | - Felicia Schlotthauer
- Boehringer Ingelheim Veterinary Research Centre, Bemeroder Str. 31, 30559 Hannover, Germany
| | - Gerd Sutter
- Institute for Infectious Diseases and Zoonoses, Department of Veterinary Sciences, Ludwig-Maximilians-Universität, Veterinärstraße 13, 80539 Munich, Germany
| | - Robert Fux
- Institute for Infectious Diseases and Zoonoses, Department of Veterinary Sciences, Ludwig-Maximilians-Universität, Veterinärstraße 13, 80539 Munich, Germany
| | - Martin Beer
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Südufer 10, 17493 Greifswald - Insel Riems, Germany
| |
Collapse
|
49
|
Metcalfe HJ, Biffar L, Steinbach S, Guzman E, Connelley T, Morrison I, Vordermeier HM, Villarreal-Ramos B. Ag85A-specific CD4 + T cell lines derived after boosting BCG-vaccinated cattle with Ad5-85A possess both mycobacterial growth inhibition and anti-inflammatory properties. Vaccine 2018; 36:2850-2854. [PMID: 29655632 PMCID: PMC5937909 DOI: 10.1016/j.vaccine.2018.03.068] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 03/20/2018] [Accepted: 03/25/2018] [Indexed: 12/28/2022]
Abstract
There is a need to improve the efficacy of the BCG vaccine against human and bovine tuberculosis. Previous data showed that boosting bacilli Calmette-Guerin (BCG)-vaccinated cattle with a recombinant attenuated human type 5 adenovirally vectored subunit vaccine (Ad5-85A) increased BCG protection and was associated with increased frequency of Ag85A-specific CD4+ T cells post-boosting. Here, the capacity of Ag85A-specific CD4+ T cell lines - derived before and after viral boosting - to interact with BCG-infected macrophages was evaluated. No difference before and after boosting was found in the capacity of these Ag85A-specific CD4+ T cell lines to restrict mycobacterial growth, but the secretion of IL-10 in vitro post-boost increased significantly. Furthermore, cell lines derived post-boost had no statistically significant difference in the secretion of pro-inflammatory cytokines (IL-1β, IL-12, IFNγ or TNFα) compared to pre-boost lines. In conclusion, the protection associated with the increased number of Ag85A-specific CD4+ T cells restricting mycobacterial growth may be associated with anti-inflammatory properties to limit immune-pathology.
Collapse
Affiliation(s)
- Hannah J Metcalfe
- TB Immunology and Vaccinology Team, Department of Bacteriology, Animal and Plant Health Agency, Weybridge, Surrey KT15 3NB, UK; Immunity Division, The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
| | - Lucia Biffar
- TB Immunology and Vaccinology Team, Department of Bacteriology, Animal and Plant Health Agency, Weybridge, Surrey KT15 3NB, UK
| | - Sabine Steinbach
- TB Immunology and Vaccinology Team, Department of Bacteriology, Animal and Plant Health Agency, Weybridge, Surrey KT15 3NB, UK
| | - Efrain Guzman
- The Pirbright Institute Ash Road, Pirbright, Surrey GU24 0NF, UK
| | - Tim Connelley
- Immunity Division, The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
| | - Ivan Morrison
- Immunity Division, The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
| | - H Martin Vordermeier
- TB Immunology and Vaccinology Team, Department of Bacteriology, Animal and Plant Health Agency, Weybridge, Surrey KT15 3NB, UK
| | - Bernardo Villarreal-Ramos
- TB Immunology and Vaccinology Team, Department of Bacteriology, Animal and Plant Health Agency, Weybridge, Surrey KT15 3NB, UK.
| |
Collapse
|
50
|
Xin T, Gao X, Yang H, Li P, Liang Q, Hou S, Sui X, Guo X, Yuan W, Zhu H, Ding J, Jia H. Limitations of Using IL-17A and IFN-γ-Induced Protein 10 to Detect Bovine Tuberculosis. Front Vet Sci 2018; 5:28. [PMID: 29560355 PMCID: PMC5845669 DOI: 10.3389/fvets.2018.00028] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 02/09/2018] [Indexed: 11/18/2022] Open
Abstract
Bovine tuberculosis (bTB) is primarily caused by infection with Mycobacterium bovis, which belongs to the Mycobacterium tuberculosis complex. The airborne route is considered the most common for transmission of M. bovis, and more than 15% of cattle with bTB shed the Mycobacterium, which can be detect by nested PCR to amplify mycobacterial mpb70 from a nasal swab from a cow. To screen for cytokines fostering early and accurate detection of bTB, peripheral blood mononuclear cells were isolated from naturally M. bovis-infected, experimentally M. bovis 68002-infected, and uninfected cattle, then these cells were stimulated by PPD-B, CFP-10-ESAT-6 (CE), or phosphate-buffered saline (PBS) for 6 h. The levels of interferon gamma (IFN-γ), IFN-γ-induced protein 10 (IP-10), IL-6, IL-12, IL-17A, and tumor necrosis factor alpha mRNA were measured using real-time PCR. To explore the cytokines associated with different periods of M. bovis infection, cattle were divided into three groups: PCR-positive, PCR-negative, and uninfected using the tuberculin skin test, CFP-10/ESAT-6/TB10.4 protein cocktail-based skin test, IFN-γ release assay (IGRA), CFP-10/ESAT-6 (CE)-based IGRA, and nested PCR. The expression of IP-10, IL-17A, and IFN-γ proteins induced by PPD-B, CE, or PBS was detected by ELISA. The results showed that levels of PPD-B-stimulated IL-17A and IP-10 (mRNA and protein), and CE-induced IP-10 (mRNA and protein) were significantly higher in cattle naturally or experimentally infected with M. bovis than in those that were uninfected. The levels of PPD-B- or CE-induced IL-17A and IP-10 (protein) could be used to differentiate M. bovis-infected calves from uninfected ones for 6 to 30 weeks post-infection, whereas PPD-B- and CE-induced IP-10 and IL-17A mRNA expression could be used to differentiate M. bovis-infected calves from uninfected ones between 6 and 58 weeks post-infection. However, CE-induced IL-17A (protein) was not a reliable indicator of M. bovis infection in cattle that were confirmed positive for infection by nested PCR. Furthermore, the levels of PPD-B- or CE-induced IP-10 and IL-17A protein were lower than IFN-γ in M. bovis-infected cattle. Therefore, IL-17A and IP-10 protein are not suitable biomarkers for bTB. Antigen-induced IP-10 mRNA should be analyzed further for their potential to be used in the diagnosis of bTB.
Collapse
Affiliation(s)
- Ting Xin
- Institute of Animal Sciences (IAS), Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Xintao Gao
- Institute of Animal Sciences (IAS), Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Hongjun Yang
- Dairy Cattle Research Center, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Pingjun Li
- Institute of Animal Sciences (IAS), Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Qianqian Liang
- Institute of Animal Sciences (IAS), Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Shaohua Hou
- Institute of Animal Sciences (IAS), Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Xiukun Sui
- Institute of Animal Sciences (IAS), Chinese Academy of Agricultural Sciences (CAAS), Beijing, China.,Molecular and Cellular Biology, Gembloux Agro-Bio Tech University of Liège (ULg), Gembloux, Belgium
| | - Xiaoyu Guo
- Institute of Animal Sciences (IAS), Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Weifeng Yuan
- Institute of Animal Sciences (IAS), Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Hongfei Zhu
- Institute of Animal Sciences (IAS), Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Jiabo Ding
- China Institute of Veterinary Drugs Control, Beijing, China
| | - Hong Jia
- Institute of Animal Sciences (IAS), Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| |
Collapse
|