1
|
Liu Y, Li H, Dai D, He J, Liang Z. Gene Regulatory Mechanism of Mycobacterium Tuberculosis during Dormancy. Curr Issues Mol Biol 2024; 46:5825-5844. [PMID: 38921019 PMCID: PMC11203133 DOI: 10.3390/cimb46060348] [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: 04/10/2024] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 06/27/2024] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb) complex, is a zoonotic disease that remains one of the leading causes of death worldwide. Latent tuberculosis infection reactivation is a challenging obstacle to eradicating TB globally. Understanding the gene regulatory network of Mtb during dormancy is important. This review discusses up-to-date information about TB gene regulatory networks during dormancy, focusing on the regulation of lipid and energy metabolism, dormancy survival regulator (DosR), White B-like (Wbl) family, Toxin-Antitoxin (TA) systems, sigma factors, and MprAB. We outline the progress in vaccine and drug development associated with Mtb dormancy.
Collapse
Affiliation(s)
- Yiduo Liu
- College of Animal Science and Technology, Guangxi University, No. 100 University West Road, Nanning 530004, China (D.D.)
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Han Li
- College of Animal Science and Technology, Guangxi University, No. 100 University West Road, Nanning 530004, China (D.D.)
| | - Dejia Dai
- College of Animal Science and Technology, Guangxi University, No. 100 University West Road, Nanning 530004, China (D.D.)
| | - Jiakang He
- College of Animal Science and Technology, Guangxi University, No. 100 University West Road, Nanning 530004, China (D.D.)
| | - Zhengmin Liang
- College of Animal Science and Technology, Guangxi University, No. 100 University West Road, Nanning 530004, China (D.D.)
| |
Collapse
|
2
|
Uddin MS, Kaldis A, Menassa R, Ortiz Guluarte J, Barreda DR, Guan LL, Alexander TW. Mucosal Immunization with Spore-Based Vaccines against Mannheimia haemolytica Enhances Antigen-Specific Immunity. Vaccines (Basel) 2024; 12:375. [PMID: 38675757 PMCID: PMC11054499 DOI: 10.3390/vaccines12040375] [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: 02/08/2024] [Revised: 03/05/2024] [Accepted: 03/06/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Mannheimia haemolytica is a bovine respiratory pathogen commonly associated with bacterial bronchopneumonia. Current vaccine strategies have shown variable efficacy in feedlot cattle, and therefore novel vaccines are needed. Bacillus subtilis spores have been investigated as a mucosal vaccine platform, due to their ability to bind and present antigens to the mucosa and act as an adjuvant. The aim of this study was to develop two spore-based mucosal vaccines targeting M. haemolytica and evaluate their immunogenicity in mice. METHODS Two antigen constructs composed of cholera toxin B subunit, M. haemolytica leukotoxin, and either the M. haemolytica outer membrane protein PlpE (MhCP1) or GS60 (MhCP2) were synthesized, purified and then bound to spores as vaccines. In two separate mice trials, the spore-bound vaccines (Spore-MhCP1 and Spore-MhCP2) were administered to mice through intranasal and intragastric routes, while free antigens were administered intranasally and intramuscularly. Unbound spores were also evaluated intranasally. Antigen-specific serum IgG and mucosal IgA from bronchoalveolar lavage, feces, and saliva were measured after vaccination. Mice sera from all treatment groups were assessed for their bactericidal activity against M. haemolytica. RESULTS In both mice experiments, intramuscular immunization induced the strongest serum IgG antibody response. However, the intranasal administration of Spore-MhCP1 and Spore-MhCP2 elicited the greatest secretory IgA-specific response against leukotoxin, PlpE, and GS60 in bronchoalveolar lavage, saliva, and feces (p < 0.05). Compared to the intranasal administration of free antigen, spore-bound antigen groups showed greater bactericidal activity against M. haemolytica (p < 0.05). CONCLUSIONS Since intranasally delivered Spore-MhCP1 and Spore-MhCP2 elicited both systemic and mucosal immune responses in mice, these vaccines may have potential to mitigate lung infection in cattle by restricting M. haemolytica colonization and proliferation in the respiratory tract. The efficacy of these mucosal spore-based vaccines merits further assessment against M. haemolytica in cattle.
Collapse
Affiliation(s)
- Muhammed Salah Uddin
- Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, Lethbridge, AB T1J 4B1, Canada; (M.S.U.); (J.O.G.)
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada; (D.R.B.); (L.L.G.)
| | - Angelo Kaldis
- London Research and Development Centre, Agriculture and Agri-Food Canada, London, ON N5V 4T3, Canada; (A.K.); (R.M.)
| | - Rima Menassa
- London Research and Development Centre, Agriculture and Agri-Food Canada, London, ON N5V 4T3, Canada; (A.K.); (R.M.)
| | - José Ortiz Guluarte
- Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, Lethbridge, AB T1J 4B1, Canada; (M.S.U.); (J.O.G.)
| | - Daniel R. Barreda
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada; (D.R.B.); (L.L.G.)
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Le Luo Guan
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada; (D.R.B.); (L.L.G.)
- Faculty of Land and Food Systems, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Trevor W. Alexander
- Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, Lethbridge, AB T1J 4B1, Canada; (M.S.U.); (J.O.G.)
| |
Collapse
|
3
|
Kwon KW, Choi HG, Kim KS, Park SA, Kim HJ, Shin SJ. BCG-booster vaccination with HSP90-ESAT-6-HspX-RipA multivalent subunit vaccine confers durable protection against hypervirulent Mtb in mice. NPJ Vaccines 2024; 9:55. [PMID: 38459038 PMCID: PMC10923817 DOI: 10.1038/s41541-024-00847-7] [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: 11/01/2023] [Accepted: 02/21/2024] [Indexed: 03/10/2024] Open
Abstract
The quest for effective and enhanced multiantigenic tuberculosis (TB) subunit vaccine necessitates the induction of a protective pathogen-specific immune response while circumventing detrimental inflammation within the lung milieu. In line with this goal, we engineered a modified iteration of the quadrivalent vaccine, namely HSP90-ESAT-6-HspX-RipA (HEHR), which was coupled with the TLR4 adjuvant, CIA09A. The ensuing formulation was subjected to comprehensive assessment to gauge its protective efficacy against the hypervirulent Mycobacterium tuberculosis (Mtb) Haarlem clinical strain M2, following a BCG-prime boost regimen. Regardless of vaccination route, both intramuscular and subcutaneous administration with the HEHR vaccine exhibited remarkable protective efficacy in significantly reducing the Mtb bacterial burden and pulmonary inflammation. This underscores its notably superior protective potential compared to the BCG vaccine alone or a former prototype, the HSP90-E6 subunit vaccine. In addition, this superior protective efficacy was confirmed when testing a tag-free version of the HEHR vaccine. Furthermore, the protective immune determinant, represented by durable antigen-specific CD4+IFN-γ+IL-17A+ T-cells expressing a CXCR3+KLRG1- cell surface phenotype in the lung, was robustly induced in HEHR-boosted mice at 12 weeks post-challenge. Collectively, our data suggest that the BCG-prime HEHR boost vaccine regimen conferred improved and long-term protection against hypervirulent Mtb strain with robust antigen-specific Th1/Th17 responses.
Collapse
Affiliation(s)
- Kee Woong Kwon
- Department of Microbiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, South Korea
- Department of Microbiology, College of Medicine, Gyeongsang National University, Jinju, 52727, South Korea
| | - Han-Gyu Choi
- Department of Microbiology, and Medical Science, College of Medicine, Chungnam National University, Daejeon, 35015, South Korea
| | | | - Shin Ae Park
- R&D Center, EyeGene Inc., Goyang, 10551, South Korea
| | - Hwa-Jung Kim
- Department of Microbiology, and Medical Science, College of Medicine, Chungnam National University, Daejeon, 35015, South Korea.
| | - Sung Jae Shin
- Department of Microbiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, South Korea.
- Institute for Immunology and Immunological Disease, Yonsei University College of Medicine, Seoul, 03722, South Korea.
| |
Collapse
|
4
|
Vergara EJ, Tran AC, Kim MY, Mussá T, Paul MJ, Harrison T, Reljic R. Mucosal and systemic immune responses after a single intranasal dose of nanoparticle and spore-based subunit vaccines in mice with pre-existing lung mycobacterial immunity. Front Immunol 2023; 14:1306449. [PMID: 38130713 PMCID: PMC10733481 DOI: 10.3389/fimmu.2023.1306449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 11/15/2023] [Indexed: 12/23/2023] Open
Abstract
Tuberculosis (TB) is a major global health threat that claims more than one million lives annually. With a quarter of the global population harbouring latent TB, post-exposure vaccination aimed at high-risk populations that could develop active TB disease would be of great public health benefit. Mucosal vaccination is an attractive approach for a predominantly lung disease like TB because it elicits both local and systemic immunity. However, the immunological consequence of mucosal immunisation in the presence of existing lung immunity remains largely unexplored. Using a mycobacterial pre-exposure mouse model, we assessed whether pre-existing mucosal and systemic immune responses can be boosted and/or qualitatively altered by intranasal administration of spore- and nanoparticle-based subunit vaccines. Analysis of lung T cell responses revealed an increasing trend in the frequency of important CD4 and CD8 T cell subsets, and T effector memory cells with a Th1 cytokine (IFNγ and TNFα) signature among immunised mice. Additionally, significantly greater antigen specific Th1, Th17 and IL-10 responses, and antigen-induced T cell proliferation were seen from the spleens of immunised mice. Measurement of antigen-specific IgG and IgA from blood and bronchoalveolar lavage fluid also revealed enhanced systemic and local humoral immune responses among immunised animals. Lastly, peripheral blood mononuclear cells (PBMCs) obtained from the TB-endemic country of Mozambique show that individuals with LTBI showed significantly greater CD4 T cell reactivity to the vaccine candidate as compared to healthy controls. These results support further testing of Spore-FP1 and Nano-FP1 as post-exposure TB vaccines.
Collapse
Affiliation(s)
- Emil Joseph Vergara
- Institute for Infection and Immunity, St. George’s University of London, London, United Kingdom
| | - Andy Cano Tran
- Institute for Infection and Immunity, St. George’s University of London, London, United Kingdom
| | - Mi-Young Kim
- Institute for Infection and Immunity, St. George’s University of London, London, United Kingdom
- Department of Molecular Biology, Jeonbuk National University, Jeonju, Republic of Korea
| | - Tufária Mussá
- Department of Microbiology, Faculty of Medicine, Eduardo Mondlane University, Maputo, Mozambique
| | - Matthew J. Paul
- Institute for Infection and Immunity, St. George’s University of London, London, United Kingdom
| | - Thomas Harrison
- Institute for Infection and Immunity, St. George’s University of London, London, United Kingdom
| | - Rajko Reljic
- Institute for Infection and Immunity, St. George’s University of London, London, United Kingdom
| |
Collapse
|
5
|
Zong R, Ruan H, Liu C, Fan S, Li J. Bacteria and Bacterial Components as Natural Bio-Nanocarriers for Drug and Gene Delivery Systems in Cancer Therapy. Pharmaceutics 2023; 15:2490. [PMID: 37896250 PMCID: PMC10610331 DOI: 10.3390/pharmaceutics15102490] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 10/06/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Bacteria and bacterial components possess multifunctional properties, making them attractive natural bio-nanocarriers for cancer diagnosis and targeted treatment. The inherent tropic and motile nature of bacteria allows them to grow and colonize in hypoxic tumor microenvironments more readily than conventional therapeutic agents and other nanomedicines. However, concerns over biosafety, limited antitumor efficiency, and unclear tumor-targeting mechanisms have restricted the clinical translation and application of natural bio-nanocarriers based on bacteria and bacterial components. Fortunately, bacterial therapies combined with engineering strategies and nanotechnology may be able to reverse a number of challenges for bacterial/bacterial component-based cancer biotherapies. Meanwhile, the combined strategies tend to enhance the versatility of bionanoplasmic nanoplatforms to improve biosafety and inhibit tumorigenesis and metastasis. This review summarizes the advantages and challenges of bacteria and bacterial components in cancer therapy, outlines combinatorial strategies for nanocarriers and bacterial/bacterial components, and discusses their clinical applications.
Collapse
Affiliation(s)
| | | | | | - Shaohua Fan
- School of Life Science, Jiangsu Normal University, Xuzhou 221116, China
| | - Jun Li
- School of Life Science, Jiangsu Normal University, Xuzhou 221116, China
| |
Collapse
|
6
|
White AD, Tran AC, Sibley L, Sarfas C, Morrison AL, Lawrence S, Dennis M, Clark S, Zadi S, Lanni F, Rayner E, Copland A, Hart P, Diogo GR, Paul MJ, Kim M, Gleeson F, Salguero FJ, Singh M, Stehr M, Cutting SM, Basile JI, Rottenberg ME, Williams A, Sharpe SA, Reljic R. Spore-FP1 tuberculosis mucosal vaccine candidate is highly protective in guinea pigs but fails to improve on BCG-conferred protection in non-human primates. Front Immunol 2023; 14:1246826. [PMID: 37881438 PMCID: PMC10594996 DOI: 10.3389/fimmu.2023.1246826] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 09/19/2023] [Indexed: 10/27/2023] Open
Abstract
Tuberculosis remains a major health threat globally and a more effective vaccine than the current Bacillus Calmette Guerin (BCG) is required, either to replace or boost it. The Spore-FP1 mucosal vaccine candidate is based on the fusion protein of Ag85B-Acr-HBHA/heparin-binding domain, adsorbed on the surface of inactivated Bacillus subtilis spores. The candidate conferred significant protection against Mycobacterium. tuberculosis challenge in naïve guinea pigs and markedly improved protection in the lungs and spleens of animals primed with BCG. We then immunized rhesus macaques with BCG intradermally, and subsequently boosted with one intradermal and one aerosol dose of Spore-FP1, prior to challenge with low dose aerosolized M. tuberculosis Erdman strain. Following vaccination, animals did not show any adverse reactions and displayed higher antigen specific cellular and antibody immune responses compared to BCG alone but this did not translate into significant improvement in disease pathology or bacterial burden in the organs.
Collapse
Affiliation(s)
- Andrew D. White
- United Kingdom Health Security Agency (UKHSA), Porton Down, Salisbury, United Kingdom
| | - Andy C. Tran
- Institute for Infection and Immunity, St George’s University of London, London, United Kingdom
| | - Laura Sibley
- United Kingdom Health Security Agency (UKHSA), Porton Down, Salisbury, United Kingdom
| | - Charlotte Sarfas
- United Kingdom Health Security Agency (UKHSA), Porton Down, Salisbury, United Kingdom
| | - Alexandra L. Morrison
- United Kingdom Health Security Agency (UKHSA), Porton Down, Salisbury, United Kingdom
| | - Steve Lawrence
- United Kingdom Health Security Agency (UKHSA), Porton Down, Salisbury, United Kingdom
| | - Mike Dennis
- United Kingdom Health Security Agency (UKHSA), Porton Down, Salisbury, United Kingdom
| | - Simon Clark
- United Kingdom Health Security Agency (UKHSA), Porton Down, Salisbury, United Kingdom
| | - Sirine Zadi
- United Kingdom Health Security Agency (UKHSA), Porton Down, Salisbury, United Kingdom
| | - Faye Lanni
- United Kingdom Health Security Agency (UKHSA), Porton Down, Salisbury, United Kingdom
| | - Emma Rayner
- United Kingdom Health Security Agency (UKHSA), Porton Down, Salisbury, United Kingdom
| | - Alastair Copland
- Institute for Infection and Immunity, St George’s University of London, London, United Kingdom
| | - Peter Hart
- Institute for Infection and Immunity, St George’s University of London, London, United Kingdom
| | - Gil Reynolds Diogo
- Institute for Infection and Immunity, St George’s University of London, London, United Kingdom
| | - Matthew J. Paul
- Institute for Infection and Immunity, St George’s University of London, London, United Kingdom
| | - Miyoung Kim
- Institute for Infection and Immunity, St George’s University of London, London, United Kingdom
| | - Fergus Gleeson
- Department of Oncology, The Churchill Hospital, Oxford, United Kingdom
| | - Francisco J. Salguero
- United Kingdom Health Security Agency (UKHSA), Porton Down, Salisbury, United Kingdom
| | | | | | - Simon M. Cutting
- School of Biological Sciences, Royal Holloway University of London, Surrey, United Kingdom
- Sporegen Ltd , London Bioscience Innovation Centre, London, United Kingdom
| | - Juan I. Basile
- Department of Microbiology, Tumour and Cell Biology and Centre for Tuberculosis Research, Karolinska Institute, Stockholm, Sweden
| | - Martin E. Rottenberg
- Department of Microbiology, Tumour and Cell Biology and Centre for Tuberculosis Research, Karolinska Institute, Stockholm, Sweden
| | - Ann Williams
- United Kingdom Health Security Agency (UKHSA), Porton Down, Salisbury, United Kingdom
| | - Sally A. Sharpe
- United Kingdom Health Security Agency (UKHSA), Porton Down, Salisbury, United Kingdom
| | - Rajko Reljic
- Institute for Infection and Immunity, St George’s University of London, London, United Kingdom
| |
Collapse
|
7
|
Uddin MS, Guluarte JO, Abbott DW, Inglis GD, Guan LL, Alexander TW. Development of a spore-based mucosal vaccine against the bovine respiratory pathogen Mannheimia haemolytica. Sci Rep 2023; 13:12981. [PMID: 37563163 PMCID: PMC10415371 DOI: 10.1038/s41598-023-29732-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 02/09/2023] [Indexed: 08/12/2023] Open
Abstract
Bovine respiratory disease (BRD) is a significant health issue in the North American feedlot industry, causing substantial financial losses due to morbidity and mortality. A lack of effective vaccines against BRD pathogens has resulted in antibiotics primarily being used for BRD prevention. The aim of this study was to develop a mucosal vaccine against the BRD pathogen, Mannheimia haemolytica, using Bacillus subtilis spores as an adjuvant. A chimeric protein (MhCP) containing a tandem repeat of neutralizing epitopes from M. haemolytica leukotoxin A (NLKT) and outer membrane protein PlpE was expressed to produce antigen for adsorption to B. subtilis spores. Adsorption was optimized by comparing varying amounts of antigen and spores, as well as different buffer pH and reaction temperatures. Using the optimal adsorption parameters, spore-bound antigen (Spore-MhCP) was prepared and administered to mice via two mucosal routes (intranasal and intragastric), while intramuscular administration of free MhCP and unvaccinated mice were used as positive and negative control treatments, respectively. Intramuscular administration of MhCP elicited the strongest serum IgG response. However, intranasal immunization of Spore-MhCP generated the best secretory IgA-specific response against both PlpE and NLKT in all samples evaluated (bronchoalveolar lavage, saliva, and feces). Since proliferation of M. haemolytica in the respiratory tract is a prerequisite to lung infection, this spore-based vaccine may offer protection in cattle by limiting colonization and subsequent infection, and Spore-MhCP warrants further evaluation in cattle as a mucosal vaccine against M. haemolytica.
Collapse
Affiliation(s)
- Muhammed Salah Uddin
- Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, 5403 1st Avenue South, Lethbridge, AB, T1J 4B1, Canada
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, T6G 2P5, Canada
| | - Jose Ortiz Guluarte
- Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, 5403 1st Avenue South, Lethbridge, AB, T1J 4B1, Canada
| | - D Wade Abbott
- Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, 5403 1st Avenue South, Lethbridge, AB, T1J 4B1, Canada
| | - G Douglas Inglis
- Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, 5403 1st Avenue South, Lethbridge, AB, T1J 4B1, Canada
| | - Le Luo Guan
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, T6G 2P5, Canada
| | - Trevor W Alexander
- Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, 5403 1st Avenue South, Lethbridge, AB, T1J 4B1, Canada.
| |
Collapse
|
8
|
MatRahim NA, Jones KM, Keegan BP, Strych U, Zhan B, Lee HY, AbuBakar S. TonB-Dependent Receptor Protein Displayed on Spores of Bacillus subtilis Stimulates Protective Immune Responses against Acinetobacter baumannii. Vaccines (Basel) 2023; 11:1106. [PMID: 37376495 DOI: 10.3390/vaccines11061106] [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: 04/19/2023] [Revised: 06/06/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
The emergence of antibiotic-resistant Acinetobacter baumannii strains with limited treatment options has become a significant global health concern. Efforts to develop vaccines against the bacteria have centred on several potential protein targets, including the TonB-dependent receptors (TBDRs). In the present study, TBDRs from A. baumannii were displayed on the surface of Bacillus subtilis spores. The immunogenicity of the recombinant spores was evaluated in orally vaccinated mice. None of the immunized mice demonstrated signs of illness and were observed to be healthy throughout the study. Sera and the intestinal secretions from the recombinant spores-treated mice demonstrated mucosal and humoral antibody responses to the vaccine antigen. In addition, bactericidal activities of the sera against A. baumannii clinical isolates were demonstrated. These observations suggest that the B. subtilis spore-displayed TBDRs should be further explored as much-needed potential oral vaccine candidates against A. baumannii.
Collapse
Affiliation(s)
- Nor-Aziyah MatRahim
- Tropical Infectious Diseases Research and Education Center (TIDREC), Universiti Malaya, Kuala Lumpur 50603, Malaysia
- Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX 77030, USA
- Virology Unit, Infectious Diseases Research Centre, Institute for Medical Research, National Institutes of Health, Shah Alam 40170, Malaysia
| | - Kathryn Marie Jones
- Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX 77030, USA
| | - Brian P Keegan
- Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ulrich Strych
- Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX 77030, USA
| | - Bin Zhan
- Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hai-Yen Lee
- Tropical Infectious Diseases Research and Education Center (TIDREC), Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Sazaly AbuBakar
- Tropical Infectious Diseases Research and Education Center (TIDREC), Universiti Malaya, Kuala Lumpur 50603, Malaysia
| |
Collapse
|
9
|
Liang X, Liang J, Cao J, Liu S, Wang Q, Ning Y, Liang Z, Zheng J, Zhang Z, Luo J, Chen Y, Huang X, Huang Y, Qin Q, Zhou S. Oral immunizations with Bacillus subtilis spores displaying VP19 protein provide protection against Singapore grouper iridovirus (SGIV) infection in grouper. FISH & SHELLFISH IMMUNOLOGY 2023:108860. [PMID: 37257567 DOI: 10.1016/j.fsi.2023.108860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 05/28/2023] [Accepted: 05/29/2023] [Indexed: 06/02/2023]
Abstract
Disease caused by Singapore grouper iridovirus (SGIV) results in major economic losses in the global grouper aquaculture industry. Vaccination is considered to be the most effective way to protect grouper from SGIV. In this study, the spores of Bacillus subtilis (B.subtilis) WB600 were utilized as the vehicle that the VP19 protein was displayed on the spores surface. To further investigate the effect of oral vaccination, the grouper were orally immunized with B.s-CotC-19 spores. After challenged, the survival rate of grouper orally vaccinated with B.s-CotC-19 spores was 34.5% and the relative percent survival (RPS) was 28.7% compared to the PBS group. Moreover, the viral load in the tissues of the B.s-CotC-19 group was significantly lower than that of the PBS group. The histopathological sections of head kidney and liver tissue from the B.s-CotC-19 group showed significantly less histopathology compared to the PBS group. In addition, the specific IgM levels in serum in the B.s-CotC-19 group was higher than those in the PBS group. In the hindgut tissue, the immune-related gene expression detected by quantitative real-time PCR (qRT-PCR) exhibited an increasing trend in different degrees in the B.s-CotC-19 group, suggesting that the innate and adaptive immune responses were activated. These results indicated that the oral administration of recombinant B.subtilis spores was effective for preventing SGIV infection. This study provided a feasible strategy for the controlling of fish virus diseases.
Collapse
Affiliation(s)
- Xia Liang
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Junjia Liang
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Jinqiao Cao
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Shijia Liu
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Quan Wang
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Yunshang Ning
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Zengjian Liang
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Jiaying Zheng
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Zemiao Zhang
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Jiayi Luo
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Yingjing Chen
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Xiaohong Huang
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China; College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China; Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266000, China
| | - Yan Huang
- ZhongShan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Qiwei Qin
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China; College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China; Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266000, China.
| | - Sheng Zhou
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China; College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China; Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266000, China.
| |
Collapse
|
10
|
Dijkman K, Lindenstrøm T, Rosenkrands I, Søe R, Woodworth JS, Lindestam Arlehamn CS, Mortensen R. A protective, single-visit TB vaccination regimen by co-administration of a subunit vaccine with BCG. NPJ Vaccines 2023; 8:66. [PMID: 37160970 PMCID: PMC10169149 DOI: 10.1038/s41541-023-00666-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 04/25/2023] [Indexed: 05/11/2023] Open
Abstract
The only licensed tuberculosis (TB) vaccine, Bacillus Calmette Guerin (BCG), fails to reliably protect adolescents and adults from pulmonary TB, resulting in ~1.6 million deaths annually. Protein subunit vaccines have shown promise against TB in clinical studies. Unfortunately, most subunit vaccines require multiple administrations, which increases the risk of loss to follow-up and necessitates more complex and costly logistics. Given the well-documented adjuvant effect of BCG, we hypothesized that BCG co-administration could compensate for a reduced number of subunit vaccinations. To explore this, we developed an expression-optimized version of our H107 vaccine candidate (H107e), which does not cross-react with BCG. In the CAF®01 adjuvant, a single dose of H107e induced inferior protection compared to three H107e/CAF®01 administrations. However, co-administering a single dose of H107e/CAF®01 with BCG significantly improved protection, which was equal to BCG co-administered with three H107e/CAF®01 doses. Importantly, combining BCG with a single H107e/CAF®01 dose also increased protection in previously BCG-primed animals. Overall, a single dose of H107e/CAF®01 with BCG induced long-lived immunity and triggered BCG-specific Th17 responses. These data support co-administration of BCG and subunit vaccines in both BCG naïve and BCG-primed individuals as an improved TB vaccine strategy with reduced number of vaccination visits.
Collapse
Affiliation(s)
- Karin Dijkman
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
- Janssen Vaccines & Prevention, Leiden, the Netherlands
| | - Thomas Lindenstrøm
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Ida Rosenkrands
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Rikke Søe
- Department of Vaccine Development, Statens Serum Institut, Copenhagen, Denmark
| | - Joshua S Woodworth
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | | | - Rasmus Mortensen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark.
| |
Collapse
|
11
|
Shurygina AP, Zabolotnykh N, Vinogradova T, Khairullin B, Kassenov M, Nurpeisova A, Sarsenbayeva G, Sansyzbay A, Vasilyev K, Buzitskaya J, Egorov A, Stukova M. Preclinical Evaluation of TB/FLU-04L-An Intranasal Influenza Vector-Based Boost Vaccine against Tuberculosis. Int J Mol Sci 2023; 24:ijms24087439. [PMID: 37108602 PMCID: PMC10138401 DOI: 10.3390/ijms24087439] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/14/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Tuberculosis is a major global threat to human health. Since the widely used BCG vaccine is poorly effective in adults, there is a demand for the development of a new type of boost tuberculosis vaccine. We designed a novel intranasal tuberculosis vaccine candidate, TB/FLU-04L, which is based on an attenuated influenza A virus vector encoding two mycobacterium antigens, Ag85A and ESAT-6. As tuberculosis is an airborne disease, the ability to induce mucosal immunity is one of the potential advantages of influenza vectors. Sequences of ESAT-6 and Ag85A antigens were inserted into the NS1 open reading frame of the influenza A virus to replace the deleted carboxyl part of the NS1 protein. The vector expressing chimeric NS1 protein appeared to be genetically stable and replication-deficient in mice and non-human primates. Intranasal immunization of C57BL/6 mice or cynomolgus macaques with the TB/FLU-04L vaccine candidate induced Mtb-specific Th1 immune response. Single TB/FLU-04L immunization in mice showed commensurate levels of protection in comparison to BCG and significantly increased the protective effect of BCG when applied in a "prime-boost" scheme. Our findings show that intranasal immunization with the TB/FLU-04L vaccine, which carries two mycobacterium antigens, is safe, and induces a protective immune response against virulent M. tuberculosis.
Collapse
Affiliation(s)
- Anna-Polina Shurygina
- Smorodintsev Research Institute of Influenza of the Ministry of Health of the Russian Federation, 197022 St. Petersburg, Russia
| | - Natalia Zabolotnykh
- Saint-Petersburg State Research Institute of Phthisiopulmonology of the Ministry of Health of the Russian Federation, 191036 St. Petersburg, Russia
| | - Tatiana Vinogradova
- Saint-Petersburg State Research Institute of Phthisiopulmonology of the Ministry of Health of the Russian Federation, 191036 St. Petersburg, Russia
| | - Berik Khairullin
- Research Institute for Biological Safety Problems, Gvardeiskiy 080409, Kazakhstan
| | - Markhabat Kassenov
- Research Institute for Biological Safety Problems, Gvardeiskiy 080409, Kazakhstan
| | - Ainur Nurpeisova
- Research Institute for Biological Safety Problems, Gvardeiskiy 080409, Kazakhstan
| | - Gulbanu Sarsenbayeva
- Research Institute for Biological Safety Problems, Gvardeiskiy 080409, Kazakhstan
| | - Abylai Sansyzbay
- Research Institute for Biological Safety Problems, Gvardeiskiy 080409, Kazakhstan
| | - Kirill Vasilyev
- Smorodintsev Research Institute of Influenza of the Ministry of Health of the Russian Federation, 197022 St. Petersburg, Russia
| | - Janna Buzitskaya
- Smorodintsev Research Institute of Influenza of the Ministry of Health of the Russian Federation, 197022 St. Petersburg, Russia
| | - Andrey Egorov
- Smorodintsev Research Institute of Influenza of the Ministry of Health of the Russian Federation, 197022 St. Petersburg, Russia
| | - Marina Stukova
- Smorodintsev Research Institute of Influenza of the Ministry of Health of the Russian Federation, 197022 St. Petersburg, Russia
| |
Collapse
|
12
|
Gonçalves G, Santos RA, Coutinho F, Pedrosa N, Curado M, Machado M, Costas B, Bonneville L, Serrano M, Carvalho AP, Díaz-Rosales P, Oliva-Teles A, Couto A, Serra CR. Oral vaccination of fish against vibriosis using spore-display technology. Front Immunol 2022; 13:1012301. [PMID: 36311700 PMCID: PMC9608137 DOI: 10.3389/fimmu.2022.1012301] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 09/29/2022] [Indexed: 10/06/2024] Open
Abstract
Oral vaccines are highly demanded by the aquaculture sector, to allow mass delivery of antigens without using the expensive and labor-intensive injectable vaccines. These later require individual handling of fish, provoking stress-related mortalities. One possible strategy to create injection-free vaccine delivery vehicles is the use of bacterial spores, extremely resistant structures with wide biotechnological applications, including as probiotics, display systems, or adjuvants. Bacterial spores, in particular those of Bacillus subtilis, have been shown to behave as mucosal vaccine adjuvants in mice models. However, such technology has not been extensively explored against fish bacterial disease. In this study, we used a laboratory strain of B. subtilis, for which a variety of genetic manipulation tools are available, to display at its spores surface either a Vibrio antigenic protein, OmpK, or the green fluorescence protein, GFP. When previously vaccinated by immersion with the OmpK- carrying spores, zebrafish survival upon a bacterial challenge with V. anguillarum and V. parahaemolyticus, increased up to 50 - 90% depending on the pathogen targeted. Further, we were able to detect anti-GFP-antibodies in the serum of European seabass juveniles fed diets containing the GFP-carrying spores and anti-V. anguillarum antibodies in the serum of European seabass juveniles fed the OmpK-carrying spores containing diet. More important, seabass survival was increased from 60 to 86% when previously orally vaccinated with in-feed OmpK- carrying spores. Our results indicate that B. subtilis spores can effectively be used as antigen-carriers for oral vaccine delivery in fish.
Collapse
Affiliation(s)
- Gabriela Gonçalves
- Departamento de Biologia, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
- Centro Interdisciplinar de Investigação Marinha e Ambiental, Universidade do Porto (CIMAR/CIIMAR), Matosinhos, Portugal
| | - Rafaela A. Santos
- Departamento de Biologia, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
- Centro Interdisciplinar de Investigação Marinha e Ambiental, Universidade do Porto (CIMAR/CIIMAR), Matosinhos, Portugal
| | - Filipe Coutinho
- Centro Interdisciplinar de Investigação Marinha e Ambiental, Universidade do Porto (CIMAR/CIIMAR), Matosinhos, Portugal
| | - Neide Pedrosa
- Centro Interdisciplinar de Investigação Marinha e Ambiental, Universidade do Porto (CIMAR/CIIMAR), Matosinhos, Portugal
| | - Maria Curado
- Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Marina Machado
- Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Benjamin Costas
- Centro Interdisciplinar de Investigação Marinha e Ambiental, Universidade do Porto (CIMAR/CIIMAR), Matosinhos, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Lourenço Bonneville
- Laboratory of Microbial Development, Instituto de Tecnologia Química e Biológica António Xavier (ITQB-NOVA), Oeiras, Portugal
| | - Mónica Serrano
- Laboratory of Microbial Development, Instituto de Tecnologia Química e Biológica António Xavier (ITQB-NOVA), Oeiras, Portugal
| | - António Paulo Carvalho
- Departamento de Biologia, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
- Centro Interdisciplinar de Investigação Marinha e Ambiental, Universidade do Porto (CIMAR/CIIMAR), Matosinhos, Portugal
| | - Patricia Díaz-Rosales
- Fish Immunology and Pathology Group, Animal Health Research Centre (CISA, INIA-CSIC), Madrid, Spain
| | - Aires Oliva-Teles
- Departamento de Biologia, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
- Centro Interdisciplinar de Investigação Marinha e Ambiental, Universidade do Porto (CIMAR/CIIMAR), Matosinhos, Portugal
| | - Ana Couto
- Departamento de Biologia, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
| | - Cláudia R. Serra
- Departamento de Biologia, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
- Centro Interdisciplinar de Investigação Marinha e Ambiental, Universidade do Porto (CIMAR/CIIMAR), Matosinhos, Portugal
| |
Collapse
|
13
|
Peng F, Xiang Y, Wang H, Hu Y, Zhou R, Hu Y. Biomimetic Assembly of Spore@ZIF-8 Microspheres for Vaccination. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2204011. [PMID: 35996807 DOI: 10.1002/smll.202204011] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Indexed: 06/15/2023]
Abstract
Vaccines have been one of the most powerful weapons to defend against infectious diseases for a long time now. Subunit vaccines are of increasing importance because of their safety and effectiveness. In this work, a Bacillus amyloliquefaciens spore@zeolitic imidazolate framework-8 (ZIF-8) vaccine platform is constructed. The ovalbumin (OVA) is encapsulated in the ZIF-8 shells as a model antigen to form a spore@OVA@ZIF-8 (SOZ) composite. The assembly of ZIF-8 improves the loading content of OVA on the spores and provides OVA with long-term protection. The SOZ composite enhances the immunization efficacy in multiple ways, such as facilitation of antigen uptake and lysosome escape, stimulation of dendritic cells to mature and secrete cytokines, boosting of antibody production and formation of an antigen depot. This platform shows several advantages including easy preparation, cost-effectiveness, long life, convenience of transportation and storage, and no need for the cold chain. These findings may have promising implications for the rational design of safe and effective spore-based composite vaccine platforms.
Collapse
Affiliation(s)
- Fei Peng
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
- Hongshan Laboratory, Wuhan, Hubei Province, 430070, China
| | - Yuqiang Xiang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
- Hongshan Laboratory, Wuhan, Hubei Province, 430070, China
| | - Hui Wang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
- Hongshan Laboratory, Wuhan, Hubei Province, 430070, China
| | - Yanjie Hu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
- Hongshan Laboratory, Wuhan, Hubei Province, 430070, China
| | - Rui Zhou
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Hongshan Laboratory, Wuhan, Hubei Province, 430070, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
- International Research Center for Animal Diseases (Ministry of Science & Technology of China), Wuhan, 430070, China
- Cooperative Innovation Center of Sustainable Pig Production, Wuhan, 430070, China
| | - Yonggang Hu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
- Hongshan Laboratory, Wuhan, Hubei Province, 430070, China
| |
Collapse
|
14
|
Chen C, Li YL, Lv FL, Xu LD, Huang YW. Surface Display of Peptides Corresponding to the Heptad Repeat 2 Domain of the Feline Enteric Coronavirus Spike Protein on Bacillus subtilis Spores Elicits Protective Immune Responses Against Homologous Infection in a Feline Aminopeptidase-N-Transduced Mouse Model. Front Immunol 2022; 13:925922. [PMID: 35837396 PMCID: PMC9273865 DOI: 10.3389/fimmu.2022.925922] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 05/23/2022] [Indexed: 11/22/2022] Open
Abstract
Although feline coronavirus (FCoV) infection is extremely common in cats, there are currently few effective treatments. A peptide derived from the heptad repeat 2 (HR2) domain of the coronavirus (CoV) spike protein has shown effective for inhibition of various human and animal CoVs in vitro, but further use of FCoV-HR2 in vivo has been limited by lack of practical delivery vectors and small animal infection model. To overcome these technical challenges, we first constructed a recombinant Bacillus subtilis (rBSCotB-HR2P) expressing spore coat protein B (CotB) fused to an HR2-derived peptide (HR2P) from a serotype II feline enteric CoV (FECV). Immunogenic capacity was evaluated in mice after intragastric or intranasal administration, showing that recombinant spores could trigger strong specific cellular and humoral immune responses. Furthermore, we developed a novel mouse model for FECV infection by transduction with its primary receptor (feline aminopeptidase N) using an E1/E3-deleted adenovirus type 5 vector. This model can be used to study the antiviral immune response and evaluate vaccines or drugs, and is an applicable choice to replace cats for the study of FECV. Oral administration of rBSCotB-HR2P in this mouse model effectively protected against FECV challenge and significantly reduced pathology in the digestive tract. Owing to its safety, low cost, and probiotic features, rBSCotB-HR2P is a promising oral vaccine candidate for use against FECV/FCoV infection in cats.
Collapse
Affiliation(s)
- Chu Chen
- Department of Veterinary Medicine, Zhejiang University, Hangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Ya-Li Li
- Department of Veterinary Medicine, Zhejiang University, Hangzhou, China
| | - Fang-Li Lv
- Department of Veterinary Medicine, Zhejiang University, Hangzhou, China
| | - Ling-Dong Xu
- Department of Veterinary Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Yao-Wei Huang, ; Ling-Dong Xu,
| | - Yao-Wei Huang
- Department of Veterinary Medicine, Zhejiang University, Hangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- *Correspondence: Yao-Wei Huang, ; Ling-Dong Xu,
| |
Collapse
|
15
|
La Manna MP, Shekarkar Azgomi M, Tamburini B, Badami GD, Mohammadnezhad L, Dieli F, Caccamo N. Phenotypic and Immunometabolic Aspects on Stem Cell Memory and Resident Memory CD8+ T Cells. Front Immunol 2022; 13:884148. [PMID: 35784300 PMCID: PMC9247337 DOI: 10.3389/fimmu.2022.884148] [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: 02/25/2022] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
The immune system, smartly and surprisingly, saves the exposure of a particular pathogen in its memory and reacts to the pathogen very rapidly, preventing serious diseases.Immunologists have long been fascinated by understanding the ability to recall and respond faster and more vigorously to a pathogen, known as “memory”.T-cell populations can be better described by using more sophisticated techniques to define phenotype, transcriptional and epigenetic signatures and metabolic pathways (single-cell resolution), which uncovered the heterogeneity of the memory T-compartment. Phenotype, effector functions, maintenance, and metabolic pathways help identify these different subsets. Here, we examine recent developments in the characterization of the heterogeneity of the memory T cell compartment. In particular, we focus on the emerging role of CD8+ TRM and TSCM cells, providing evidence on how their immunometabolism or modulation can play a vital role in their generation and maintenance in chronic conditions such as infections or autoimmune diseases.
Collapse
Affiliation(s)
- Marco Pio La Manna
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR) Azienda Ospedaliera Universitaria Policlinico (A.O.U.P.) Paolo Giaccone, University of Palermo, Palermo, Italy
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, Italy
| | - Mojtaba Shekarkar Azgomi
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR) Azienda Ospedaliera Universitaria Policlinico (A.O.U.P.) Paolo Giaccone, University of Palermo, Palermo, Italy
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, Italy
| | - Bartolo Tamburini
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR) Azienda Ospedaliera Universitaria Policlinico (A.O.U.P.) Paolo Giaccone, University of Palermo, Palermo, Italy
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, Italy
| | - Giusto Davide Badami
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR) Azienda Ospedaliera Universitaria Policlinico (A.O.U.P.) Paolo Giaccone, University of Palermo, Palermo, Italy
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, Italy
| | - Leila Mohammadnezhad
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR) Azienda Ospedaliera Universitaria Policlinico (A.O.U.P.) Paolo Giaccone, University of Palermo, Palermo, Italy
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, Italy
| | - Francesco Dieli
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR) Azienda Ospedaliera Universitaria Policlinico (A.O.U.P.) Paolo Giaccone, University of Palermo, Palermo, Italy
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, Italy
| | - Nadia Caccamo
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR) Azienda Ospedaliera Universitaria Policlinico (A.O.U.P.) Paolo Giaccone, University of Palermo, Palermo, Italy
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, Italy
- *Correspondence: Nadia Caccamo,
| |
Collapse
|
16
|
Therapeutic Effect of Subunit Vaccine AEC/BC02 on Mycobacterium tuberculosis Post-Chemotherapy Relapse Using a Latent Infection Murine Model. Vaccines (Basel) 2022; 10:vaccines10050825. [PMID: 35632581 PMCID: PMC9145927 DOI: 10.3390/vaccines10050825] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/14/2022] [Accepted: 05/21/2022] [Indexed: 11/17/2022] Open
Abstract
Tuberculosis (TB), caused by the human pathogen Mycobacterium tuberculosis (Mtb), is an infectious disease that presents a major threat to human health. Bacillus Calmette-Guérin (BCG), the only licensed TB vaccine, is ineffective against latent TB infection, necessitating the development of further TB drugs or therapeutic vaccines. Herein, we evaluated the therapeutic effect of a novel subunit vaccine AEC/BC02 after chemotherapy in a spontaneous Mtb relapse model. Immunotherapy followed 4 weeks of treatment with isoniazid and rifapentine, and bacterial loads in organs, pathological changes, and adaptive immune characteristics were investigated. The results showed slowly increased bacterial loads in the spleen and lungs of mice inoculated with AEC/BC02 with significantly lower loads than those of the control groups. Pathological scores for the liver, spleen, and lungs decreased accordingly. Moreover, AEC/BC02 induced antigen-specific IFN-γ-secreting or IL-2-secreting cellular immune responses, which decreased with the number of immunizations and times. Obvious Ag85b- and EC-specific IgG were observed in mice following the treatment with AEC/BC02, indicating a significant Th1-biased response. Taken together, these data suggest that AEC/BC02 immunotherapy post-chemotherapy may shorten future TB treatment.
Collapse
|
17
|
de Almeida MEM, Alves KCS, de Vasconcelos MGS, Pinto TS, Glória JC, Chaves YO, Neves WLL, Tarragô AM, de Souza Neto JN, Astolfi-Filho S, Pontes GS, da Silva Balieiro AA, Isticato R, Ricca E, Mariúba LAM. Bacillus subtilis spores as delivery system for nasal Plasmodium falciparum circumsporozoite surface protein immunization in a murine model. Sci Rep 2022; 12:1531. [PMID: 35087102 PMCID: PMC8795416 DOI: 10.1038/s41598-022-05344-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 11/29/2021] [Indexed: 11/15/2022] Open
Abstract
Malaria remains a widespread public health problem in tropical and subtropical regions around the world, and there is still no vaccine available for full protection. In recent years, it has been observed that spores of Bacillus subtillis can act as a vaccine carrier and adjuvant, promoting an elevated humoral response after co-administration with antigens either coupled or integrated to their surface. In our study, B. subtillis spores from the KO7 strain were used to couple the recombinant CSP protein of P. falciparum (rPfCSP), and the nasal humoral-induced immune response in Balb/C mice was evaluated. Our results demonstrate that the spores coupled to rPfCSP increase the immunogenicity of the antigen, which induces high levels of serum IgG, and with balanced Th1/Th2 immune response, being detected antibodies in serum samples for 250 days. Therefore, the use of B. subtilis spores appears to be promising for use as an adjuvant in a vaccine formulation.
Collapse
Affiliation(s)
- Maria Edilene M de Almeida
- Programa de Pós-Graduação Stricto Sensu em Biologia Celular e Molecular do Instituto Oswaldo Cruz (IOC/Fiocruz), Rio de Janeiro, RJ, Brazil.
- Instituto Leônidas e Maria Deane, Fiocruz Amazônia, Manaus, Brazil.
| | - Késsia Caroline Souza Alves
- Instituto Leônidas e Maria Deane, Fiocruz Amazônia, Manaus, Brazil
- Programa de Pós-Graduação em Biotecnologia, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, AM, Brazil
| | | | | | - Juliane Corrêa Glória
- Instituto Leônidas e Maria Deane, Fiocruz Amazônia, Manaus, Brazil
- Programa de Pós-Graduação em Biotecnologia, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, AM, Brazil
| | - Yury Oliveira Chaves
- Instituto Leônidas e Maria Deane, Fiocruz Amazônia, Manaus, Brazil
- Programa de Pós-Graduação Stricto Sensu em Biologia Parasitária do Instituto Oswaldo Cruz (IOC/Fiocruz), Rio de Janeiro, RJ, Brazil
| | - Walter Luiz Lima Neves
- Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas, HEMOAM, Manaus, AM, Brazil
| | - Andrea Monteiro Tarragô
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal Do Amazonas (UFAM), Manaus, AM, Brazil
- Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas, HEMOAM, Manaus, AM, Brazil
- Programa de Pós-Graduação Stricto Sensu em Ciências Aplicadas à Hematologia PPGH, Universidade do Estado do Amazonas (UEA), Manaus, AM, Brazil
| | - Júlio Nino de Souza Neto
- Centro de Apoio Multidisciplinar (CAM), Universidade Federal do Amazonas (UFAM), Manaus, AM, Brazil
| | - Spartaco Astolfi-Filho
- Instituto de Ciências Biológicas (ICB), Universidade Federal do Amazonas (UFAM), Manaus, AM, Brazil
| | | | - Antônio Alcirley da Silva Balieiro
- Instituto Leônidas e Maria Deane, Fiocruz Amazônia, Manaus, Brazil
- Programa de Pós-Graduação Stricto Sensu em Biologia Parasitária do Instituto Oswaldo Cruz (IOC/Fiocruz), Rio de Janeiro, RJ, Brazil
| | | | - Ezio Ricca
- Department of Biology, Federico II University, Naples, Italy
| | - Luis André M Mariúba
- Programa de Pós-Graduação Stricto Sensu em Biologia Celular e Molecular do Instituto Oswaldo Cruz (IOC/Fiocruz), Rio de Janeiro, RJ, Brazil.
- Instituto Leônidas e Maria Deane, Fiocruz Amazônia, Manaus, Brazil.
- Programa de Pós-Graduação em Biotecnologia, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, AM, Brazil.
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal Do Amazonas (UFAM), Manaus, AM, Brazil.
| |
Collapse
|
18
|
Lange J, Rivera-Ballesteros O, Buggert M. Human mucosal tissue-resident memory T cells in health and disease. Mucosal Immunol 2022; 15:389-397. [PMID: 34743182 PMCID: PMC8571012 DOI: 10.1038/s41385-021-00467-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 10/11/2021] [Accepted: 10/18/2021] [Indexed: 02/04/2023]
Abstract
Memory T cells are fundamental to maintain immune surveillance of the human body. During the past decade, it has become apparent that non-recirculating resident memory T cells (TRMs) form a first line memory response in tissues to tackle re-infections. The fact that TRMs are essential for local immunity highlights the therapeutic potential of targeting this population against tumors and infections. However, similar to other immune subsets, TRMs are heterogenous and may form distinct effector populations with unique functions at diverse tissue sites. Further insight into the mechanisms of how TRM function and respond to pathogens and malignancies at different mucosal sites will help to shape future vaccine and immunotherapeutic approaches. Here, we review the current understanding of TRM function and biology at four major mucosal sites: gastrointestinal tract, lung, head and neck, as well as female reproductive tract. We also summarize our current knowledge of how TRM targets invading pathogens and developing tumor cells at these mucosal sites and contemplate how TRMs may be exploited to protect from infections and cancer.
Collapse
Affiliation(s)
- Joshua Lange
- grid.4714.60000 0004 1937 0626Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Olga Rivera-Ballesteros
- grid.4714.60000 0004 1937 0626Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Marcus Buggert
- grid.4714.60000 0004 1937 0626Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
19
|
Enriquez AB, Izzo A, Miller SM, Stewart EL, Mahon RN, Frank DJ, Evans JT, Rengarajan J, Triccas JA. Advancing Adjuvants for Mycobacterium tuberculosis Therapeutics. Front Immunol 2021; 12:740117. [PMID: 34759923 PMCID: PMC8572789 DOI: 10.3389/fimmu.2021.740117] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 08/26/2021] [Indexed: 01/15/2023] Open
Abstract
Tuberculosis (TB) remains one of the leading causes of death worldwide due to a single infectious disease agent. BCG, the only licensed vaccine against TB, offers limited protection against pulmonary disease in children and adults. TB vaccine research has recently been reinvigorated by new data suggesting alternative administration of BCG induces protection and a subunit/adjuvant vaccine that provides close to 50% protection. These results demonstrate the need for generating adjuvants in order to develop the next generation of TB vaccines. However, development of TB-targeted adjuvants is lacking. To help meet this need, NIAID convened a workshop in 2020 titled “Advancing Vaccine Adjuvants for Mycobacterium tuberculosis Therapeutics”. In this review, we present the four areas identified in the workshop as necessary for advancing TB adjuvants: 1) correlates of protective immunity, 2) targeting specific immune cells, 3) immune evasion mechanisms, and 4) animal models. We will discuss each of these four areas in detail and summarize what is known and what we can advance on in order to help develop more efficacious TB vaccines.
Collapse
Affiliation(s)
- Ana B Enriquez
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States.,Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Angelo Izzo
- Tuberculosis Research Program, Centenary Institute, The University of Sydney, Sydney, NSW, Australia
| | - Shannon M Miller
- Center for Translational Medicine, University of Montana, Missoula, MT, United States.,Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT, United States
| | - Erica L Stewart
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.,Sydney Institute for Infectious Diseases and Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Robert N Mahon
- Division of AIDS, Columbus Technologies & Services Inc., Contractor to National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Daniel J Frank
- Division of AIDS, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, United States
| | - Jay T Evans
- Center for Translational Medicine, University of Montana, Missoula, MT, United States.,Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT, United States
| | - Jyothi Rengarajan
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States.,Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States.,Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, United States
| | - James A Triccas
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.,Sydney Institute for Infectious Diseases and Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
20
|
Bacillus subtilis Spore-Trained Dendritic Cells Enhance the Generation of Memory T Cells via ICAM1. Cells 2021; 10:cells10092267. [PMID: 34571913 PMCID: PMC8469252 DOI: 10.3390/cells10092267] [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: 06/07/2021] [Revised: 08/14/2021] [Accepted: 08/16/2021] [Indexed: 11/22/2022] Open
Abstract
Immunological memory is a cardinal feature of the immune system. The intestinal mucosa is the primary exposure and entry site of infectious organisms. For an effective and long-lasting safeguard, a robust immune memory system is required, especially by the mucosal immunity. It is well known that tissue-resident memory T cells (Trms) provide a first response against infections reencountered at mucosal tissues surfaces, where they accelerate pathogen clearance. However, their function in intestinal immunization remains to be investigated. Here, we report enhanced local mucosal and systemic immune responses through oral administration of H9N2 influenza whole inactivated virus (H9N2 WIV) plus Bacillus subtilis spores. Subsequently, H9N2 WIV plus spores led to the generation of CD103+ CD69+ Trms, which were independent of circulating T cells during the immune period. Meanwhile, we also found that Bacillus subtilis spores could stimulate Acrp30 expression in 3T3-L1 adipocytes. Moreover, spore-stimulated adipocyte supernatant also upregulated the expression of intercellular adhesion molecule-1 (ICAM1) in dendritic cells (DCs). Furthermore, the proportion of HA-tetramer+ cells was severely curtailed upon suppressed ICAM1 expression, which also depended on HA-loaded DCs. Taken together, our data demonstrated that spore-promoted H9N2 WIV induced an immune response by enhancing Trms populations, which were associated with the activation of ICAM1 in DCs.
Collapse
|
21
|
Lung Epithelial Signaling Mediates Early Vaccine-Induced CD4 + T Cell Activation and Mycobacterium tuberculosis Control. mBio 2021; 12:e0146821. [PMID: 34253059 PMCID: PMC8406195 DOI: 10.1128/mbio.01468-21] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Tuberculosis (TB) is one of the leading causes of death due to a single infectious agent. The development of a TB vaccine that induces durable and effective immunity to Mycobacterium tuberculosis (Mtb) infection is urgently needed. Early and superior Mtb control can be induced in M. bovis Bacillus Calmette-Guérin (BCG)-vaccinated hosts when the innate immune response is targeted to generate effective vaccine-induced immunity. In the present study, we show that innate activation of DCs is critical for mucosal localization of clonally activated vaccine-induced CD4+ T cells in the lung and superior early Mtb control. In addition, our study reveals that Th1/Th17 cytokine axis play an important role in superior vaccine-induced immunity. Our studies also show that activation of the nuclear factor kappa-light-chain enhancer of activated B cell (NF-κβ) pathway in lung epithelial cells is critical for the mucosal localization of activated vaccine-induced CD4+ T cells for rapid Mtb control. Thus, our study provides novel insights into the immune mechanisms that can overcome TB vaccine bottlenecks and provide early rapid Mtb control.
Collapse
|
22
|
Mat Rahim N, Lee H, Strych U, AbuBakar S. Facing the challenges of multidrug-resistant Acinetobacter baumannii: progress and prospects in the vaccine development. Hum Vaccin Immunother 2021; 17:3784-3794. [PMID: 34106809 PMCID: PMC8437540 DOI: 10.1080/21645515.2021.1927412] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
In 2017, the World Health Organization (WHO) named A. baumannii as one of the three antibiotic-resistant bacterial species on its list of global priority pathogens in dire need of novel and effective treatment. With only polymyxin and tigecycline antibiotics left as last-resort treatments, the need for novel alternative approaches to the control of this bacterium becomes imperative. Vaccines against numerous bacteria have had impressive records in reducing the burden of the respective diseases and addressing antimicrobial resistance; as in the case of Haemophilus influenzae type b . A similar approach could be appropriate for A. baumannii. Toward this end, several potentially protective antigens against A. baumannii were identified and evaluated as vaccine antigen candidates. A licensed vaccine for the bacteria, however, is still not in sight. Here we explore and discuss challenges in vaccine development against A. baumannii and the promising approaches for improving the vaccine development process.
Collapse
Affiliation(s)
- NorAziyah Mat Rahim
- Tropical Infectious Diseases Research and Education Center (TIDREC), Universiti Malaya, Kuala Lumpur, Malaysia.,Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA.,Virology Unit, Institute for Medical Research, National Institute of Health Complex, Setia Alam, Malaysia
| | - HaiYen Lee
- Tropical Infectious Diseases Research and Education Center (TIDREC), Universiti Malaya, Kuala Lumpur, Malaysia
| | - Ulrich Strych
- Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA
| | - Sazaly AbuBakar
- Tropical Infectious Diseases Research and Education Center (TIDREC), Universiti Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
23
|
Kim HS, Arellano K, Park H, Todorov SD, Kim B, Kang H, Park YJ, Suh DH, Jung ES, Ji Y, Holzapfel WH. Assessment of the safety and anti-inflammatory effects of three Bacillus strains in the respiratory tract. Environ Microbiol 2021; 23:3077-3098. [PMID: 33899316 DOI: 10.1111/1462-2920.15530] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 04/12/2021] [Indexed: 11/29/2022]
Abstract
Chronic respiratory diseases are part of accumulating health problems partly due to worldwide increase in air pollution. By their antimicrobial and immunomodulatory properties, some probiotics constitute promising alternatives for the prevention and treatment of chronic respiratory diseases. We have isolated Bacillus strains from Korean fermented foods and selected three potentially probiotic strains (two Bacillus subtilis and one Bacillus amyloliquefaciens) based on safety, antimicrobial efficacy, activity against airborne pathogens and their immunomodulatory properties in vivo. Safety evaluation included in silico analysis for confirming absence of virulence genes. Safety for the respiratory tract was confirmed by an in vivo pathogenicity test using a murine model. Antimicrobial activity was displayed against several airborne pathogens. Potential antimicrobial metabolites such as 2,3-butanediol and propylene glycol were identified as possible antagonistic agents. Immunomodulatory properties in vitro were confirmed by upregulation of IL-10 expression in a macrophage cell line. Intranasal instillation and inhalation in an ovalbumin (OVA)-induced lung inflammation murine model reduced T helper type 2 (Th2) cytokines at transcriptional and protein levels in the lungs. The safety and potentially beneficial role of these Bacillus strains could be demonstrated for the respiratory tract of a murine model.
Collapse
Affiliation(s)
- Hye-Shin Kim
- Department of Advanced Convergence, Handong Global University, Pohang, Gyungbuk, 37554, Republic of Korea
| | - Karina Arellano
- Department of Advanced Convergence, Handong Global University, Pohang, Gyungbuk, 37554, Republic of Korea
| | - Haryung Park
- HEM Inc., Pohang, Gyungbuk, 37554, Republic of Korea
| | - Svetoslav D Todorov
- Department of Advanced Convergence, Handong Global University, Pohang, Gyungbuk, 37554, Republic of Korea
| | - Bobae Kim
- HEM Inc., Pohang, Gyungbuk, 37554, Republic of Korea
| | - Hyeji Kang
- Department of Advanced Convergence, Handong Global University, Pohang, Gyungbuk, 37554, Republic of Korea.,HEM Inc., Pohang, Gyungbuk, 37554, Republic of Korea
| | - Yu Jin Park
- HEM Inc., 77, Changnyong-daero 256 Beon-gil, Suwon-si, Gyeonggi-do, 16229, Republic of Korea
| | - Dong Ho Suh
- HEM Inc., 77, Changnyong-daero 256 Beon-gil, Suwon-si, Gyeonggi-do, 16229, Republic of Korea
| | - Eun Sung Jung
- HEM Inc., 77, Changnyong-daero 256 Beon-gil, Suwon-si, Gyeonggi-do, 16229, Republic of Korea
| | - Yosep Ji
- Department of Advanced Convergence, Handong Global University, Pohang, Gyungbuk, 37554, Republic of Korea.,HEM Inc., 77, Changnyong-daero 256 Beon-gil, Suwon-si, Gyeonggi-do, 16229, Republic of Korea
| | - Wilhelm H Holzapfel
- Department of Advanced Convergence, Handong Global University, Pohang, Gyungbuk, 37554, Republic of Korea.,HEM Inc., Pohang, Gyungbuk, 37554, Republic of Korea
| |
Collapse
|
24
|
Ning H, Zhang W, Kang J, Ding T, Liang X, Lu Y, Guo C, Sun W, Wang H, Bai Y, Shen L. Subunit Vaccine ESAT-6:c-di-AMP Delivered by Intranasal Route Elicits Immune Responses and Protects Against Mycobacterium tuberculosis Infection. Front Cell Infect Microbiol 2021; 11:647220. [PMID: 33829000 PMCID: PMC8019782 DOI: 10.3389/fcimb.2021.647220] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 03/01/2021] [Indexed: 12/15/2022] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb) infection, remains the most common cause of death from a single infectious disease. More safe and effective vaccines are necessary for preventing the prevalence of TB. In this study, a subunit vaccine of ESAT-6 formulated with c-di-AMP (ESAT-6:c-di-AMP) promoted mucosal and systemic immune responses in spleen and lung. ESAT-6:c-di-AMP inhibited the differentiations of CD8+ T cells as well as macrophages, but promoted the differentiations of ILCs in lung. The co-stimulation also enhanced inflammatory cytokines production in MH-S cells. It was first revealed that ESAT-6 and c-di-AMP regulated autophagy of macrophages in different stages, which together resulted in the inhibition of Mtb growth in macrophages during early infection. After Mtb infection, the level of ESAT-6-specific immune responses induced by ESAT-6:c-di-AMP dropped sharply. Finally, inoculation of ESAT-6:c-di-AMP led to significant reduction of bacterial burdens in lungs and spleens of immunized mice. Our results demonstrated that subunit vaccine ESAT-6:c-di-AMP could elicit innate and adaptive immune responses which provided protection against Mtb challenge, and c-di-AMP as a mucosal adjuvant could enhance immunogenicity of antigen, especially for innate immunity, which might be used for new mucosal vaccine against TB.
Collapse
Affiliation(s)
- Huanhuan Ning
- Key Laboratory of Resources Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Xi'an, China.,Department of Microbiology and Pathogen Biology, Basic Medical School, Air Force Medical University, Xi'an, China
| | - Wei Zhang
- Department of Paediatrics, TangDu Hospital, Air Force Medical University, Xi'an, China
| | - Jian Kang
- Department of Microbiology and Pathogen Biology, Basic Medical School, Air Force Medical University, Xi'an, China
| | | | - Xuan Liang
- Key Laboratory of Resources Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Xi'an, China
| | - Yanzhi Lu
- Department of Microbiology and Pathogen Biology, Basic Medical School, Air Force Medical University, Xi'an, China
| | - Chengxuan Guo
- Student Brigade, Basic Medical School, Air Force Medical University, Xi'an, China
| | - Wenjie Sun
- Student Brigade, Basic Medical School, Air Force Medical University, Xi'an, China
| | - Huapeng Wang
- Student Brigade, Basic Medical School, Air Force Medical University, Xi'an, China
| | - Yinlan Bai
- Department of Microbiology and Pathogen Biology, Basic Medical School, Air Force Medical University, Xi'an, China
| | - Lixin Shen
- Key Laboratory of Resources Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Xi'an, China
| |
Collapse
|
25
|
Fan L, Wu Q, Kang S, Yang B, Wu C. The phenotypic and functional study of tissue B cells in respiratory system provided important information for diseases and development of vaccines. J Cell Mol Med 2021; 25:2621-2632. [PMID: 33481318 PMCID: PMC7933955 DOI: 10.1111/jcmm.16278] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/10/2020] [Accepted: 12/17/2020] [Indexed: 12/25/2022] Open
Abstract
The field of tissue-resident B cells has received increasing attention, yet the feature of tissue B cells in respiratory system is unclear. Here, we first show that non-circulating B cells obtained from nasal, trachea and lung tissues are numerically and phenotypically distinct from their circulating counterparts. Analysis of single cell transcriptome sequence identified multiple differentially expressed genes between non-circulating B cells and circulating B cells, which illustrated their heterogeneity. Furthermore, we found high expression of CXCR3 on non-circulating B cells, and the chemokine CXCL11 was also up-regulated in the respiratory tissues, suggesting that CXCR3-CXCL11 axis might accelerate the local resident of non-circulating B cells in respiratory tract. Interestingly, intranasal immunization with BCG in mice elicited a sustained humoral immune response via induction of IgA and IgG Abs, which revealed the role of B cells. Meanwhile, tissue-resident B cells, IgA+ and IgG+ memory B cells (MBCs) in respiratory tissues, as well as plasma cells in bone marrow, were expanded and maintained, and these subsets probably developed into antibody-producing cells to participate in the local humoral immunity. Our data illustrate the phenotype and function of tissue B cells in the upper and lower airways, provide references for the prospective development of vaccines.
Collapse
Affiliation(s)
- Li Fan
- Institute of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Medical College, Hubei University of Arts and Science, Xiangyang, China
| | - Qiongli Wu
- Institute of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Shuangpeng Kang
- Institute of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Binyan Yang
- Institute of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Changyou Wu
- Institute of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Clifford Hospital, Jinan University, Guangzhou, China
| |
Collapse
|
26
|
Dijkman K, Aguilo N, Boot C, Hofman SO, Sombroek CC, Vervenne RA, Kocken CH, Marinova D, Thole J, Rodríguez E, Vierboom MP, Haanstra KG, Puentes E, Martin C, Verreck FA. Pulmonary MTBVAC vaccination induces immune signatures previously correlated with prevention of tuberculosis infection. Cell Rep Med 2021; 2:100187. [PMID: 33521701 PMCID: PMC7817873 DOI: 10.1016/j.xcrm.2020.100187] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 10/23/2020] [Accepted: 12/17/2020] [Indexed: 11/29/2022]
Abstract
To fight tuberculosis, better vaccination strategies are needed. Live attenuated Mycobacterium tuberculosis-derived vaccine, MTBVAC, is a promising candidate in the pipeline, proven to be safe and immunogenic in humans so far. Independent studies have shown that pulmonary mucosal delivery of Bacillus Calmette-Guérin (BCG), the only tuberculosis (TB) vaccine available today, confers superior protection over standard intradermal immunization. Here we demonstrate that mucosal MTBVAC is well tolerated, eliciting polyfunctional T helper type 17 cells, interleukin-10, and immunoglobulins in the airway and yielding a broader antigenic profile than BCG in rhesus macaques. Beyond our previous work, we show that local immunoglobulins, induced by MTBVAC and BCG, bind to M. tuberculosis and enhance pathogen uptake. Furthermore, after pulmonary vaccination, but not M. tuberculosis infection, local T cells expressed high levels of mucosal homing and tissue residency markers. Our data show that pulmonary MTBVAC administration has the potential to enhance its efficacy and justifies further exploration of mucosal vaccination strategies in preclinical efficacy studies.
Collapse
Affiliation(s)
- Karin Dijkman
- Biomedical Primate Research Centre (BPRC), Rijswijk, the Netherlands
| | - Nacho Aguilo
- Department of Microbiology, Faculty of Medicine, IIS Aragon, University of Zaragoza, Zaragoza, Spain
- CIBERES, Instituto de Salud Carlos III, Madrid, Spain
| | - Charelle Boot
- Biomedical Primate Research Centre (BPRC), Rijswijk, the Netherlands
| | - Sam O. Hofman
- Biomedical Primate Research Centre (BPRC), Rijswijk, the Netherlands
| | | | | | | | - Dessislava Marinova
- Department of Microbiology, Faculty of Medicine, IIS Aragon, University of Zaragoza, Zaragoza, Spain
- CIBERES, Instituto de Salud Carlos III, Madrid, Spain
| | - Jelle Thole
- TuBerculosis Vaccine Initiative (TBVI), Lelystad, the Netherlands
| | | | | | | | | | - Carlos Martin
- Department of Microbiology, Faculty of Medicine, IIS Aragon, University of Zaragoza, Zaragoza, Spain
- CIBERES, Instituto de Salud Carlos III, Madrid, Spain
| | | |
Collapse
|
27
|
The Antimicrobial Peptide, Bactenecin 5, Supports Cell-Mediated but Not Humoral Immunity in the Context of a Mycobacterial Antigen Vaccine Model. Antibiotics (Basel) 2020; 9:antibiotics9120926. [PMID: 33352656 PMCID: PMC7766334 DOI: 10.3390/antibiotics9120926] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/16/2020] [Accepted: 12/17/2020] [Indexed: 01/28/2023] Open
Abstract
Bactenecin (Bac) 5 is a bovine antimicrobial peptide (AMP) capable of killing some species of bacteria through the inhibition of protein synthesis. Bac5 and other AMPs have also been shown to have chemotactic properties and can induce inflammatory cytokine expression by innate immune cells. Recently, AMPs have begun to be investigated for their potential use as novel vaccine adjuvants. In the current work, we characterise the functionality of Bac5 in vitro using murine macrophage-like cells, ex vivo using human tonsil tissue and in vivo using a murine model of vaccination. We report the effects of the peptide in isolation and in the context of co-presentation with mycobacterial antigen and whole, inert Bacillus subtilis spore antigens. We find that Bac5 can trigger the release of nitric oxide from murine macrophages and upregulate surface marker expression including CD86, MHC-I and MHC-II, in the absence of additional agonists. When coupled with mycobacterial Ag85 and B. subtilis spores, Bac5 also enhanced IFNγ secretion. We provide evidence that B. subtilis spores, but not the Bac5 peptide, act as strong adjuvants in promoting antigen-specific immunoglobulin production in Ag85B-vaccinated mice. Our findings suggest that Bac5 is an important regulator of the early cell-mediated host immune response.
Collapse
|
28
|
Counoupas C, Ferrell KC, Ashhurst A, Bhattacharyya ND, Nagalingam G, Stewart EL, Feng CG, Petrovsky N, Britton WJ, Triccas JA. Mucosal delivery of a multistage subunit vaccine promotes development of lung-resident memory T cells and affords interleukin-17-dependent protection against pulmonary tuberculosis. NPJ Vaccines 2020; 5:105. [PMID: 33298977 PMCID: PMC7665186 DOI: 10.1038/s41541-020-00255-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 10/02/2020] [Indexed: 11/29/2022] Open
Abstract
The development of effective vaccines against bacterial lung infections requires the induction of protective, pathogen-specific immune responses without deleterious inflammation within the pulmonary environment. Here, we made use of a polysaccharide-adjuvanted vaccine approach to elicit resident pulmonary T cells to protect against aerosol Mycobacterium tuberculosis infection. Intratracheal administration of the multistage fusion protein CysVac2 and the delta-inulin adjuvant Advax™ (formulated with a TLR9 agonist) provided superior protection against aerosol M. tuberculosis infection in mice, compared to parenteral delivery. Surprisingly, removal of the TLR9 agonist did not impact vaccine protection despite a reduction in cytokine-secreting T cell subsets, particularly CD4+IFN-γ+IL-2+TNF+ multifunctional T cells. CysVac2/Advax-mediated protection was associated with the induction of lung-resident, antigen-specific memory CD4+ T cells that expressed IL-17 and RORγT, the master transcriptional regulator of Th17 differentiation. IL-17 was identified as a key mediator of vaccine efficacy, with blocking of IL-17 during M. tuberculosis challenge reducing phagocyte influx, suppressing priming of pathogen-specific CD4+ T cells in local lymph nodes and ablating vaccine-induced protection. These findings suggest that tuberculosis vaccines such as CysVac2/Advax that are capable of eliciting Th17 lung-resident memory T cells are promising candidates for progression to human trials.
Collapse
Affiliation(s)
- Claudio Counoupas
- Discipline of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia.
- Tuberculosis Research Program Centenary Institute, The University of Sydney, Camperdown, NSW, Australia.
| | - Kia C Ferrell
- Discipline of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- Tuberculosis Research Program Centenary Institute, The University of Sydney, Camperdown, NSW, Australia
| | - Anneliese Ashhurst
- Discipline of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Nayan D Bhattacharyya
- Discipline of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- Tuberculosis Research Program Centenary Institute, The University of Sydney, Camperdown, NSW, Australia
| | - Gayathri Nagalingam
- Discipline of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Erica L Stewart
- Discipline of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- Tuberculosis Research Program Centenary Institute, The University of Sydney, Camperdown, NSW, Australia
- Vaxine Pty Ltd, 11 Walkley Avenue, Warradale and Flinders University, Adelaide, Australia
| | - Carl G Feng
- Discipline of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- Tuberculosis Research Program Centenary Institute, The University of Sydney, Camperdown, NSW, Australia
| | - Nikolai Petrovsky
- Vaxine Pty Ltd, 11 Walkley Avenue, Warradale and Flinders University, Adelaide, Australia
| | - Warwick J Britton
- Discipline of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- Tuberculosis Research Program Centenary Institute, The University of Sydney, Camperdown, NSW, Australia
- Department of Clinical Immunology, Royal Prince Alfred Hospital, Camperdown, NSW, 2050, Australia
| | - James A Triccas
- Discipline of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia.
- Tuberculosis Research Program Centenary Institute, The University of Sydney, Camperdown, NSW, Australia.
- Charles Perkins Centre and Marie Bashir Institute for Infectious Diseases and Biosecurity, The University of Sydney, Camperdown, NSW, Australia.
| |
Collapse
|
29
|
Applications of Bacillus subtilis Spores in Biotechnology and Advanced Materials. Appl Environ Microbiol 2020; 86:AEM.01096-20. [PMID: 32631858 DOI: 10.1128/aem.01096-20] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The bacterium Bacillus subtilis has long been an important subject for basic studies. However, this organism has also had industrial applications due to its easy genetic manipulation, favorable culturing characteristics for large-scale fermentation, superior capacity for protein secretion, and generally recognized as safe (GRAS) status. In addition, as the metabolically dormant form of B. subtilis, its spores have attracted great interest due to their extreme resistance to many environmental stresses, which makes spores a novel platform for a variety of applications. In this review, we summarize both conventional and emerging applications of B. subtilis spores, with a focus on how their unique characteristics have led to innovative applications in many areas of technology, including generation of stable and recyclable enzymes, synthetic biology, drug delivery, and material sciences. Ultimately, this review hopes to inspire the scientific community to leverage interdisciplinary approaches using spores to address global concerns about food shortages, environmental protection, and health care.
Collapse
|
30
|
Rasheed OK, Ettenger G, Buhl C, Child R, Miller SM, Evans JT, Ryter KT. 6,6'-Aryl trehalose analogs as potential Mincle ligands. Bioorg Med Chem 2020; 28:115564. [PMID: 32616186 PMCID: PMC7372699 DOI: 10.1016/j.bmc.2020.115564] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 04/27/2020] [Accepted: 05/20/2020] [Indexed: 12/17/2022]
Abstract
6,6'-Aryl trehalose derivatives have been synthesized with a view towards identifying novel Th-17-inducing vaccine adjuvants based on the high affinity Mincle ligand Brartemicin. The initial structure-activity relationships of these novel trehalose-based compounds were investigated. All compounds have been evaluated for their ability to engage the Mincle receptor and induce a potential pro-Th17 cytokine profile from human peripheral blood mononuclear cells based on IL-6 production in human peripheral blood mononuclear cells. The preliminary biological characterization of the designed analogs presented in this paper should aid in the future design and testing of more affine ligands that may foster the discovery of novel adjuvants with improved pharmacological properties.
Collapse
Affiliation(s)
- Omer K Rasheed
- Department of Chemistry and Biochemistry, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States
| | - George Ettenger
- Department of Chemistry and Biochemistry, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States
| | - Cassandra Buhl
- Division of Biological Sciences, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States
| | - Robert Child
- Division of Biological Sciences, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States
| | - Shannon M Miller
- Division of Biological Sciences, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States
| | - Jay T Evans
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States; Division of Biological Sciences, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States
| | - Kendal T Ryter
- Department of Chemistry and Biochemistry, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States; Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States.
| |
Collapse
|
31
|
Lee JE, Kye YC, Park SM, Shim BS, Yoo S, Hwang E, Kim H, Kim SJ, Han SH, Park TS, Park BC, Yun CH. Bacillus subtilis spores as adjuvants against avian influenza H9N2 induce antigen-specific antibody and T cell responses in White Leghorn chickens. Vet Res 2020; 51:68. [PMID: 32448402 PMCID: PMC7245620 DOI: 10.1186/s13567-020-00788-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 04/16/2020] [Indexed: 01/06/2023] Open
Abstract
Low-pathogenicity avian influenza H9N2 remains an endemic disease worldwide despite continuous vaccination, indicating the need for an improved vaccine strategy. Bacillus subtilis (B. subtilis), a gram-positive and endospore-forming bacterium, is a non-pathogenic species that has been used in probiotic formulations for both animals and humans. The objective of the present study was to elucidate the effect of B. subtilis spores as adjuvants in chickens administered inactivated avian influenza virus H9N2. Herein, the adjuvanticity of B. subtilis spores in chickens was demonstrated by enhancement of H9N2 virus-specific IgG responses. B. subtilis spores enhanced the proportion of B cells and the innate cell population in splenocytes from chickens administered both inactivated H9N2 and B. subtilis spores (Spore + H9N2). Furthermore, the H9N2 and spore administration induced significantly increased expression of the pro-inflammatory cytokines IL-1β and IL-6 compared to that in the H9N2 only group. Additionally, total splenocytes from chickens immunized with inactivated H9N2 in the presence or absence of B. subtilis spores were re-stimulated with inactivated H9N2. The subsequent results showed that the extent of antigen-specific CD4+ and CD8+ T cell proliferation was higher in the Spore + H9N2 group than in the group administered only H9N2. Taken together, these data demonstrate that B. subtilis spores, as adjuvants, enhance not only H9N2 virus-specific IgG but also CD4+ and CD8+ T cell responses, with an increase in pro-inflammatory cytokine production. This approach to vaccination with inactivated H9N2 together with a B. subtilis spore adjuvant in chickens produces a significant effect on antigen-specific antibody and T cell responses against avian influenza virus.
Collapse
Affiliation(s)
- Ji Eun Lee
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Yoon-Chul Kye
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Sung-Moo Park
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea.,Center for Food and Bioconvergence, Seoul National University, Seoul, Republic of Korea
| | | | - Sungsik Yoo
- Choong-Ang Vaccine Laboratory, Daejeon, Republic of Korea
| | - Eunmi Hwang
- Department of Biotechnology, Hoseo University, Asan, Chungcheongnam-do, Republic of Korea
| | - Hyungkuen Kim
- Department of Biotechnology, Hoseo University, Asan, Chungcheongnam-do, Republic of Korea
| | - Sung-Jo Kim
- Department of Biotechnology, Hoseo University, Asan, Chungcheongnam-do, Republic of Korea
| | - Seung Hyun Han
- Department of Oral Microbiology and Immunology, DRI and BK21 Program, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - Tae Sub Park
- Graduate School of International Agricultural Technology, Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang-gun, Gangwon-do, Republic of Korea
| | - Byung-Chul Park
- Graduate School of International Agricultural Technology, Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang-gun, Gangwon-do, Republic of Korea.
| | - Cheol-Heui Yun
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea. .,Center for Food and Bioconvergence, Seoul National University, Seoul, Republic of Korea. .,Graduate School of International Agricultural Technology, Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang-gun, Gangwon-do, Republic of Korea.
| |
Collapse
|
32
|
Recombinant HcGAPDH Protein Expressed on Probiotic Bacillus subtilis Spores Protects Sheep from Haemonchus contortus Infection by Inducing both Humoral and Cell-Mediated Responses. mSystems 2020; 5:5/3/e00239-20. [PMID: 32398277 PMCID: PMC7219552 DOI: 10.1128/msystems.00239-20] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Probiotic Bacillales are effective in controlling pathogens. Live probiotic bacteria improve the composition of the gastrointestinal microbiota, leading to a reduction in pathogen colonization. However, it remains largely unknown how probiotics regulate the host's immunologic responses and protect the host from parasitic infection. In this study, we addressed whether Bacillales were effective against Haemonchus contortus, a parasitic nematode that infects small ruminants worldwide. Using 16S rRNA sequencing, we found that Bacillales were largely depleted in the abomasal microbiota of sheep infected with H. contortus We constructed a recombinant Bacillus subtilis named rBS CotB-HcG that express the glyceraldehyde-3-phosphate dehydrogenase of H. contortus (HcGAPDH) on its spore surface using the Bacillus subtilis spore coat protein B (CotB) as a carrier. Mice receiving rBS CotB-HcG orally showed strong Th1-dominated immune responses. More importantly, sheep administered BS CotB-HcG per os showed increasing proliferation of the peripheral blood mononucleates, elevated anti-HcGAPDH IgG in sera, and higher anti-HcGAPDH sIgA in the intestinal mucus than the control sheep. The average weight gain of H. contortus-infected sheep treated with rBS CotB-HcG (Hc+rBS CotB-HcG ) was 48.73% greater than that of unvaccinated sheep. Furthermore, these Hc+rBS CotB-HcG sheep had fewer eggs per gram of feces by 84.1% and adult worms by 71.5%. They also demonstrated greatly lessened abomasal damage by H. contortus with an abundance of probiotic species in the abomasal microbiota. Collectively, our data unequivocally demonstrate the protective roles of CotB-HcGAPDH-expressing B. subtilis spores in against H. contortus infection and showed great potential of using probiotic-based strategy in controlling parasitic nematodes of socioeconomic importance in general.IMPORTANCE Initial analyses of the abomasal microbiota of sheep using 16S rRNA sequencing suggested that probiotic bacteria played a protective role in against H. contortus infection. A recombinant Bacillus subtilis expressing a fusion protein CotB-HcGAPDH on its spore's surface induced strong Th1 immune response in a murine model. The same probiotic recombinant, upon only one oral application, protected sheep against H. contortus infection by reducing egg shedding and decreasing adult worm loads of the parasite and increasing body weight gain of infected sheep. Both Th1 and Th2 immune responses were evident in these immunized sheep.
Collapse
|
33
|
Painter H, Prabowo SA, Cia F, Stockdale L, Tanner R, Willcocks S, Reljic R, Fletcher HA, Zelmer A. Adaption of the ex vivo mycobacterial growth inhibition assay for use with murine lung cells. Sci Rep 2020; 10:3311. [PMID: 32094451 PMCID: PMC7039920 DOI: 10.1038/s41598-020-60223-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 01/31/2020] [Indexed: 11/19/2022] Open
Abstract
In the absence of a correlate(s) of protection against human tuberculosis and a validated animal model of the disease, tools to facilitate vaccine development must be identified. We present an optimised ex vivo mycobacterial growth inhibition assay (MGIA) to assess the ability of host cells within the lung to inhibit mycobacterial growth, including Bacille Calmette-Guérin (BCG) and Mycobacterium tuberculosis (MTB) Erdman. Growth of BCG was reduced by 0.39, 0.96 and 0.73 log10 CFU following subcutaneous (s.c.) BCG, intranasal (i.n.) BCG, or BCG s.c. + mucosal boost, respectively, versus naïve mice. Comparatively, a 0.49 (s.c.), 0.60 (i.n.) and 0.81 (s.c. + mucosal boost) log10 reduction in MTB CFU was found. A BCG growth inhibitor, 2-thiophenecarboxylic acid hydrazide (TCH), was used to prevent quantification of residual BCG from i.n. immunisation and allow accurate MTB quantification. Using TCH, a further 0.58 log10 reduction in MTB CFU was revealed in the i.n. group. In combination with existing methods, the ex vivo lung MGIA may represent an important tool for analysis of vaccine efficacy and the immune mechanisms associated with vaccination in the organ primarily affected by MTB disease.
Collapse
Affiliation(s)
- Hannah Painter
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK.
| | - Satria A Prabowo
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - Felipe Cia
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - Lisa Stockdale
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
- Oxford Vaccine Group, Department of Paediatrics, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, OX4 2PG, UK
| | - Rachel Tanner
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Samuel Willcocks
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - Rajko Reljic
- Institute of Infection and Immunity, St George's University of London, Cranmer Terrrace, London, SW17 0RE, UK
| | - Helen A Fletcher
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - Andrea Zelmer
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| |
Collapse
|
34
|
Stylianou E, Paul MJ, Reljic R, McShane H. Mucosal delivery of tuberculosis vaccines: a review of current approaches and challenges. Expert Rev Vaccines 2019; 18:1271-1284. [PMID: 31876199 PMCID: PMC6961305 DOI: 10.1080/14760584.2019.1692657] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Introduction: Tuberculosis (TB) remains a major health threat and it is now clear that the current vaccine, BCG, is unable to arrest the global TB epidemic. A new vaccine is needed to either replace or boost BCG so that a better level of protection could be achieved. The route of entry of Mycobacterium tuberculosis, the causative organism, is via inhalation making TB primarily a respiratory disease. There is therefore good reason to hypothesize that a mucosally delivered vaccine against TB could be more effective than one delivered via the systemic route. Areas covered: This review summarizes the progress that has been made in the area of TB mucosal vaccines in the last few years. It highlights some of the strengths and shortcomings of the published evidence and aims to discuss immunological and practical considerations in the development of mucosal vaccines. Expert opinion: There is a growing body of evidence that the mucosal approach to vaccination against TB is feasible and should be pursued. However, further key studies are necessary to both improve our understanding of the protective immune mechanisms operating in the mucosa and the technical aspects of aerosolized delivery, before such a vaccine could become a feasible, deployable strategy.
Collapse
Affiliation(s)
- Elena Stylianou
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Matthew J Paul
- Institute for Infection and Immunity, St George's University of London, Tooting, London, UK
| | - Rajko Reljic
- Institute for Infection and Immunity, St George's University of London, Tooting, London, UK
| | - Helen McShane
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
35
|
Kuczkowska K, Øverland L, Rocha SDC, Eijsink VGH, Mathiesen G. Comparison of eight Lactobacillus species for delivery of surface-displayed mycobacterial antigen. Vaccine 2019; 37:6371-6379. [PMID: 31526620 DOI: 10.1016/j.vaccine.2019.09.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 09/04/2019] [Indexed: 12/26/2022]
Abstract
Lactobacillus spp. comprise a large group of Gram-positive lactic acid bacteria with varying physiological, ecological and immunomodulatory properties that are widely exploited by mankind, primarily in food production and as health-promoting probiotics. Recent years have shown increased interest in using lactobacilli for delivery of vaccines, mainly due to their ability to skew the immune system towards pro-inflammatory responses. We have compared the potential of eight Lactobacillus species, L. plantarum, L. brevis, L. curvatus, L. rhamnosus, L. sakei, L. gasseri, L. acidophilus and L. reuteri, as immunogenic carriers of the Ag85B-ESAT-6 antigen from Mycobacterium tuberculosis. Surface-display of the antigen was achieved in L. plantarum, L. brevis, L. gasseri and L. reuteri and these strains were further analyzed in terms of their in vitro and in vivo immunogenicity. All strains activated human dendritic cells in vitro. Immunization of mice using a homologous prime-boost regimen comprising a primary subcutaneous immunization followed by three intranasal boosters, led to slightly elevated IgG levels in serum in most strains, and, importantly, to significantly increased levels of antigen-specific mucosal IgA. Cellular immunity was assessed by studying antigen-specific T cell responses in splenocytes, which did not reveal proliferation as assessed by the expression of Ki67, but which showed clear antigen-specific IFN-γ and IL-17 responses for some of the groups. Taken together, the present results indicate that L. plantarum and L. brevis are the most promising carriers of TB vaccines.
Collapse
Affiliation(s)
- Katarzyna Kuczkowska
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences (NMBU), P.O. Box 5003, 1432 Aas, Norway.
| | - Lise Øverland
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences (NMBU), P.O. Box 5003, 1432 Aas, Norway
| | - Sergio D C Rocha
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences (NMBU), P.O. Box 5003, 1432 Aas, Norway
| | - Vincent G H Eijsink
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences (NMBU), P.O. Box 5003, 1432 Aas, Norway
| | - Geir Mathiesen
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences (NMBU), P.O. Box 5003, 1432 Aas, Norway
| |
Collapse
|
36
|
Tran AC, Kim MY, Reljic R. Emerging Themes for the Role of Antibodies in Tuberculosis. Immune Netw 2019; 19:e24. [PMID: 31501712 PMCID: PMC6722270 DOI: 10.4110/in.2019.19.e24] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/12/2019] [Accepted: 06/13/2019] [Indexed: 02/06/2023] Open
Abstract
The best way to debunk a scientific dogma is to throw irrefutable evidence at it. This is especially true if the dogma in question has been nurtured over many decades, as is the case with the apparent redundancy of antibodies (Abs) against intracellular pathogens. Although not fully compelling yet, that ‘hard core’ evidence is nevertheless now slowly beginning to emerge. This is true for several clinically relevant infections but none more so than Mycobacterium tuberculosis, the archetype intracellular pathogen that poses a great health challenge to the mankind. Here, prompted by a spate of recent high-profile reports on the effects of Abs in various experimental models of tuberculosis, we step back and take a critical look at the progress that has been made in the last 5 years and highlight some of the strengths and shortcomings of the presented evidence. We conclude that the tide of the opinion has begun to turn in favour of Abs but we also caution against overinterpreting the currently available limited evidence. For, until definitive evidence that can withstand even the most rigorous of experimental tests is produced, the dogma may yet survive. Or indeed, we may find that the truth is hidden somewhere in between the dogma and the unfulfilled scientific prophecy.
Collapse
Affiliation(s)
- Andy C Tran
- St George's, University of London, London SW17 0RE, UK
| | - Mi-Young Kim
- St George's, University of London, London SW17 0RE, UK.,Department of Molecular Biology and The Institute for Molecular Biology and Genetics, Chonbuk National University, Jeonju 54896, Korea
| | - Rajko Reljic
- St George's, University of London, London SW17 0RE, UK
| |
Collapse
|
37
|
De Maio F, Squeglia F, Goletti D, Delogu G. The Mycobacterial HBHA Protein: A Promising Biomarker for Tuberculosis. Curr Med Chem 2019; 26:2051-2060. [PMID: 30378481 DOI: 10.2174/0929867325666181029165805] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 10/01/2018] [Accepted: 10/18/2018] [Indexed: 02/06/2023]
Abstract
A major goal in tuberculosis (TB) research is the identification, among the subjects infected with Mycobacterium tuberculosis (Mtb), of those with active TB, or at higher risk of developing active disease, from the latently infected subjects. The classical heterogeneity of Mtb infection and TB disease is a major obstacle toward the identification of reliable biomarkers that can stratify Mtb infected subjects based on disease risk. The heparin-binding haemagglutinin (HBHA) is a mycobacterial surface antigen that is implicated in tuberculosis (TB) pathogenesis. The host immune response against HBHA varies depending on the TB status and several studies are supporting the role of HBHA as a useful biomarker of TB.
Collapse
Affiliation(s)
- Flavio De Maio
- Institute of Microbiology, Università Cattolica del Sacro Cuore, Milano, Italy.,Fondazione Policlinico Universitario A. Gemelli- IRCCS, Rome, Italy
| | - Flavia Squeglia
- Institute of Biostructures and Bioimaging, CNR, Via Mezzocannone 16. I-80134 Napoli, Italy
| | - Delia Goletti
- Translational Research Unit, Department of Epidemiology and Preclinical Research, "L. Spallanzani" National Institute for Infectious Diseases (INMI) IRCCCS, Rome, Italy
| | - Giovanni Delogu
- Institute of Microbiology, Università Cattolica del Sacro Cuore, Milano, Italy.,Fondazione Policlinico Universitario A. Gemelli- IRCCS, Rome, Italy
| |
Collapse
|
38
|
Kuczkowska K, Copland A, Øverland L, Mathiesen G, Tran AC, Paul MJ, Eijsink VGH, Reljic R. Inactivated Lactobacillus plantarum Carrying a Surface-Displayed Ag85B-ESAT-6 Fusion Antigen as a Booster Vaccine Against Mycobacterium tuberculosis Infection. Front Immunol 2019; 10:1588. [PMID: 31354727 PMCID: PMC6632704 DOI: 10.3389/fimmu.2019.01588] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 06/25/2019] [Indexed: 12/11/2022] Open
Abstract
Vaccination is considered the most effective strategy for controlling tuberculosis (TB). The existing vaccine, the Bacille Calmette-Guérin (BCG), although partially protective, has a number of limitations. Therefore, there is a need for developing new TB vaccines and several strategies are currently exploited including the use of viral and bacterial delivery vectors. We have previously shown that Lactobacillus plantarum (Lp) producing Ag85B and ESAT-6 antigens fused to a dendritic cell-targeting peptide (referred to as Lp_DC) induced specific immune responses in mice. Here, we analyzed the ability of two Lp-based vaccines, Lp_DC and Lp_HBD (in which the DC-binding peptide was replaced by an HBD-domain directing the antigen to non-phagocytic cells) to activate antigen-presenting cells, induce specific immunity and protect mice from Mycobacterium tuberculosis infection. We tested two strategies: (i) Lp as BCG boosting vaccine (a heterologous regimen comprising parenteral BCG immunization followed by intranasal Lp boost), and (ii) Lp as primary vaccine (a homologous regimen including subcutaneous priming followed by intranasal boost). The results showed that both Lp constructs applied as a BCG boost induced specific cellular immunity, manifested in T cell proliferation, antigen-specific IFN-γ responses and multifunctional T cells phenotypes. More importantly, intranasal boost with Lp_DC or Lp_HBD enhanced protection offered by BCG, as shown by reduced M. tuberculosis counts in lungs. These findings suggest that Lp constructs could be developed as a potential mucosal vaccine platform against mycobacterial infections.
Collapse
Affiliation(s)
- Katarzyna Kuczkowska
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | - Alastair Copland
- Institute for Infection and Immunity, St. George's University of London, London, United Kingdom.,College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Lise Øverland
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | - Geir Mathiesen
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | - Andy C Tran
- Institute for Infection and Immunity, St. George's University of London, London, United Kingdom
| | - Mathew J Paul
- Institute for Infection and Immunity, St. George's University of London, London, United Kingdom
| | - Vincent G H Eijsink
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | - Rajko Reljic
- Institute for Infection and Immunity, St. George's University of London, London, United Kingdom
| |
Collapse
|
39
|
Counoupas C, Triccas JA. The generation of T-cell memory to protect against tuberculosis. Immunol Cell Biol 2019; 97:656-663. [PMID: 31127962 DOI: 10.1111/imcb.12275] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 05/15/2019] [Accepted: 05/22/2019] [Indexed: 02/06/2023]
Abstract
Tuberculosis (TB) kills more individuals each year than any other single pathogen and a more effective vaccine is critical for the global control of the disease. Although there has been recent progress in the clinical testing of candidates, no new vaccine has been licensed for use and correlates of protective immunity in humans have not been defined. Prior Mycobacterium tuberculosis infection does not appear to confer long-term protective immunity in humans; thus mimicking the natural immune response to infection may not be a suitable approach to develop improved TB vaccines. Data from animal testing are used to progress vaccines through the "vaccine pipeline", but studies in animals have not been able to predict efficacy in humans. Furthermore, although the generation of conventional CD4+ T-cell responses are considered necessary to control infection with M. tuberculosis, these do not necessarily correlate with protection induced by candidate vaccines and other immune components may play a role, including donor unrestricted T cells, tissue-resident memory T cells and anti-M. tuberculosis antibodies. This review will summarize the current understanding of the protective immune responses following M. tuberculosis infection or vaccination, with a particular focus on vaccines that have recently entered clinical trials.
Collapse
Affiliation(s)
- Claudio Counoupas
- Discipline of Infectious Diseases and Immunology, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia.,Tuberculosis Research Program, Centenary Institute, Sydney, NSW, Australia
| | - James A Triccas
- Discipline of Infectious Diseases and Immunology, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia.,Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia.,Marie Bashir Institute for Infectious Diseases and Biosecurity, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
40
|
Ogongo P, Porterfield JZ, Leslie A. Lung Tissue Resident Memory T-Cells in the Immune Response to Mycobacterium tuberculosis. Front Immunol 2019; 10:992. [PMID: 31130965 PMCID: PMC6510113 DOI: 10.3389/fimmu.2019.00992] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 04/17/2019] [Indexed: 12/12/2022] Open
Abstract
Despite widespread BCG vaccination and effective anti-TB drugs, Tuberculosis (TB) remains the leading cause of death from an infectious agent worldwide. Several recent publications give reasons to be optimistic about the possibility of a more effective vaccine, but the only full-scale clinical trial conducted failed to show protection above BCG. The immunogenicity of vaccines in humans is primarily evaluated by the systemic immune responses they generate, despite the fact that a correlation between these responses and protection from TB disease has not been demonstrated. A novel approach to tackling this problem is to study the local immune responses that occur at the site of TB infection in the human lung, rather than those detectable in blood. There is a growing understanding that pathogen specific T-cell immunity can be highly localized at the site of infection, due to the existence of tissue resident memory T-cells (Trm). Notably, these cells do not recirculate in the blood and thus may not be represented in studies of the systemic immune response. Here, we review the potential role of Trms as a component of the TB immune response and discuss how a better understanding of this response could be harnessed to improve TB vaccine efficacy.
Collapse
Affiliation(s)
- Paul Ogongo
- Africa Health Research Institute, Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
- Institute of Primate Research, National Museums of Kenya, Nairobi, Kenya
| | - James Zachary Porterfield
- Africa Health Research Institute, Durban, South Africa
- College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
- Yale School of Public Health, Yale University, New Haven, CT, United States
| | - Alasdair Leslie
- Africa Health Research Institute, Durban, South Africa
- Department of Infection and Immunity, University College London, London, United Kingdom
| |
Collapse
|
41
|
Bacillus Calmette-Guérin Induces PD-L1 Expression on Antigen-Presenting Cells via Autocrine and Paracrine Interleukin-STAT3 Circuits. Sci Rep 2019; 9:3655. [PMID: 30842561 PMCID: PMC6403281 DOI: 10.1038/s41598-019-40145-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 02/01/2019] [Indexed: 12/18/2022] Open
Abstract
Bacillus Calmette-Guérin (BCG) is the only licensed vaccine for tuberculosis (TB), and is also used as an immunotherapy for bladder cancer and other malignancies due to its immunostimulatory properties. Mycobacteria spp., however, are well known for their numerous immune evasion mechanisms that limit the true potential of their therapeutic use. One such major mechanism is the induction of programmed death ligand-1 (PD-L1), which mitigates adaptive immune responses. Here, we sought to unravel the molecular pathways behind PD-L1 up-regulation on antigen-presenting cells (APCs) by BCG. We found that infection of APCs with BCG induced PD-L1 up-regulation, but that this did not depend on direct infection, suggesting a soluble mediator for this effect. BCG induced potent quantities of IL-6 and IL-10, and the downstream transcription factor STAT3 was hyper-phosphorylated. Intracellular analyses revealed that levels of PD-L1 molecules were associated with the STAT3 phosphorylation state, suggesting a causal link. Neutralisation of the IL-6 or IL-10 cytokine receptors dampened STAT3 phosphorylation and BCG-mediated up-regulation of PD-L1 on APCs. Pharmacological inhibition of STAT3 achieved the same effect, confirming an autocrine-paracrine cytokine loop as a mechanism for BCG-mediated up-regulation of PD-L1. Finally, an in vivo immunisation model showed that BCG vaccination under PD-L1 blockade could enhance antigen-specific memory CD4 T-cell responses. These novel findings could lead to refinement of BCG as both a vaccine for infectious disease and as a cancer immunotherapy.
Collapse
|
42
|
Vogt CM, Hilbe M, Ackermann M, Aguilar C, Eichwald C. Mouse intestinal microbiota reduction favors local intestinal immunity triggered by antigens displayed in Bacillus subtilis biofilm. Microb Cell Fact 2018; 17:187. [PMID: 30477481 PMCID: PMC6258259 DOI: 10.1186/s12934-018-1030-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 11/16/2018] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND We previously engineered Bacillus subtilis to express an antigen of interest fused to TasA in a biofilm. B. subtilis has several properties such as sporulation, biofilm formation and probiotic ability that were used for the oral application of recombinant spores harboring Echinococcus granulosus paramyosin and tropomyosin immunogenic peptides that resulted in the elicitation of a specific humoral immune response in a dog model. RESULTS In order to advance our understanding of the research in oral immunization practices using recombinant B. subtilis spores, we describe here an affordable animal model. In this study, we show clear evidence indicating that a niche is required for B. subtilis recombinant spores to colonize the densely populated mice intestinal microbiota. The reduction of intestinal microbiota with an antibiotic treatment resulted in a positive elicitation of local humoral immune response in BALB/c mice after oral application of recombinant B. subtilis spores harboring TasA fused to E. granulosus (102-207) EgTrp immunogenic peptide. Our results were supported by a lasting prevalence of spores in mice feces up to 50 days after immunization and by the presence of specific secretory IgA, isolated from feces, against E. granulosus tropomyosin. CONCLUSIONS The reduction of mouse intestinal microbiota allowed the elicitation of a local humoral immune response in mice after oral application with spores of B. subtilis harboring immunogenic peptides against E. granulosus.
Collapse
Affiliation(s)
- Cédric M Vogt
- Institute of Virology, University of Zurich, Winterthurerstrasse 266a, 8057, Zurich, Switzerland
| | - Monika Hilbe
- Laboratory for Animal Model Pathology, Institute of Pathology, Vetsuisse, University of Zurich, Zurich, Switzerland
| | - Mathias Ackermann
- Institute of Virology, University of Zurich, Winterthurerstrasse 266a, 8057, Zurich, Switzerland
| | | | - Catherine Eichwald
- Institute of Virology, University of Zurich, Winterthurerstrasse 266a, 8057, Zurich, Switzerland.
| |
Collapse
|
43
|
Flaxman A, Ewer KJ. Methods for Measuring T-Cell Memory to Vaccination: From Mouse to Man. Vaccines (Basel) 2018; 6:E43. [PMID: 30037078 PMCID: PMC6161152 DOI: 10.3390/vaccines6030043] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 07/16/2018] [Accepted: 07/20/2018] [Indexed: 12/27/2022] Open
Abstract
The development of effective vaccines continues to be a key goal for public health bodies, governments, funding bodies and pharmaceutical companies. With new vaccines such as Shingrix targeting Shingles and Bexsero for Meningitis B, licensed in recent years, today's population can be protected from more infectious diseases than ever before. Despite this, we are yet to license vaccines for some of the deadliest endemic diseases affecting children, such as malaria. In addition, the threat of epidemics caused by emerging pathogens is very real as exemplified by the 2014⁻2016 Ebola outbreak. Most licensed vaccines provide efficacy through humoral immunity and correlates of protection often quantify neutralising antibody titre. The role of T-cells in vaccine efficacy is less well understood and more complex to quantify. Defining T-cell responses which afford protection also remains a challenge, although more sophisticated assays for assessing cell-mediated immunity with the potential for higher throughput and scalability are now available and warrant review. Here we discuss the benefits of multiparameter cytokine analysis and omics approaches compared with flow cytometric and ELISpot assays. We also review technical challenges unique to clinical trial studies, including assay validation across laboratories and availability of sample type. Measuring T-cell immunogenicity alongside humoral responses provides information on the breadth of immune responses induced by vaccination. Accurately enumerating and phenotyping T-cell immunogenicity to vaccination is key for the determination of immune correlates of protection. However, identifying such T-cell parameters remains challenging without a clear understanding of the immunological mechanisms by which a T-cell-mediated response induces protection.
Collapse
Affiliation(s)
- Amy Flaxman
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK.
| | - Katie J Ewer
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK.
| |
Collapse
|