1
|
Yang Y, Chen YZ, Xia T. Optimizing antigen selection for the development of tuberculosis vaccines. CELL INSIGHT 2024; 3:100163. [PMID: 38572176 PMCID: PMC10987857 DOI: 10.1016/j.cellin.2024.100163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 03/08/2024] [Accepted: 03/11/2024] [Indexed: 04/05/2024]
Abstract
Tuberculosis (TB) remains a prevalent global infectious disease caused by genetically closely related tubercle bacilli in Mycobacterium tuberculosis complex (MTBC). For a century, the Bacillus Calmette-Guérin (BCG) vaccine has been the primary preventive measure against TB. While it effectively protects against extrapulmonary forms of pediatric TB, it lacks consistent efficacy in providing protection against pulmonary TB in adults. Consequently, the exploration and development of novel TB vaccines, capable of providing broad protection to populations, have consistently constituted a prominent area of interest in medical research. This article presents a concise overview of the novel TB vaccines currently undergoing clinical trials, discussing their classification, protective efficacy, immunogenicity, advantages, and limitations. In vaccine development, the careful selection of antigens that can induce strong and diverse specific immune responses is essential. Therefore, we have summarized the molecular characteristics, biological function, immunogenicity, and relevant studies associated with the chosen antigens for TB vaccines. These insights gained from vaccines and immunogenic proteins will inform the development of novel mycobacterial vaccines, particularly mRNA vaccines, for effective TB control.
Collapse
Affiliation(s)
- Yang Yang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Yi-Zhen Chen
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Tian Xia
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China
| |
Collapse
|
2
|
Sun X, Li W, Zhao L, Fan K, Qin F, Shi L, Gao F, Zheng C. Current landscape of exosomes in tuberculosis development, diagnosis, and treatment applications. Front Immunol 2024; 15:1401867. [PMID: 38846947 PMCID: PMC11153741 DOI: 10.3389/fimmu.2024.1401867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 05/09/2024] [Indexed: 06/09/2024] Open
Abstract
Tuberculosis (TB), caused by the bacterial pathogen Mycobacterium tuberculosis (MTB), remains one of the most prevalent and deadly infectious diseases worldwide. Currently, there are complex interactions between host cells and pathogens in TB. The onset, progression, and regression of TB are correlated not only with the virulence of MTB but also with the immunity of TB patients. Exosomes are cell-secreted membrane-bound nanovesicles with lipid bilayers that contain a variety of biomolecules, such as metabolites, lipids, proteins, and nucleic acids. Exosome-mediated cell-cell communication and interactions with the microenvironment represent crucial mechanisms through which exosomes exert their functional effects. Exosomes harbor a wide range of regulatory roles in physiological and pathological conditions, including MTB infection. Exosomes can regulate the immune response, metabolism, and cellular death to remodel the progression of MTB infection. During MTB infection, exosomes display distinctive profiles and quantities that may act as diagnostic biomarkers, suggesting that exosomes provide a revealing glimpse into the evolving landscape of MTB infections. Furthermore, exosomes derived from MTB and mesenchymal stem cells can be harnessed as vaccine platforms and drug delivery vehicles for the precise targeting and treatment of TB. In this review, we highlight the functions and mechanisms through which exosomes influence the progression of TB. Additionally, we unravel the critical significance of exosomal constituents in the diagnosis and therapeutic applications of TB, aiming to offer novel perspectives and strategies for combating TB.
Collapse
Affiliation(s)
- Xuezhi Sun
- Department of Tuberculosis III, Wuhan Pulmonary Hospital, Wuhan, Hubei, China
| | - Wei Li
- Department of Tuberculosis III, Wuhan Pulmonary Hospital, Wuhan, Hubei, China
| | - Li Zhao
- Department of Tuberculosis III, Wuhan Pulmonary Hospital, Wuhan, Hubei, China
| | - Ke Fan
- Department of Tuberculosis III, Wuhan Pulmonary Hospital, Wuhan, Hubei, China
| | - Fenfen Qin
- Department of Tuberculosis III, Wuhan Pulmonary Hospital, Wuhan, Hubei, China
| | - Liwen Shi
- Department of Tuberculosis III, Wuhan Pulmonary Hospital, Wuhan, Hubei, China
| | - Feng Gao
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chunlan Zheng
- Department of Tuberculosis III, Wuhan Pulmonary Hospital, Wuhan, Hubei, China
| |
Collapse
|
3
|
Mo S, Shi C, Cai Y, Xu M, Xu H, Xu Y, Zhang K, Zhang Y, Liu J, Che S, Liu X, Xing C, Long X, Chen X, Liu E. Single-cell transcriptome reveals highly complement activated microglia cells in association with pediatric tuberculous meningitis. Front Immunol 2024; 15:1387808. [PMID: 38745656 PMCID: PMC11091396 DOI: 10.3389/fimmu.2024.1387808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 04/16/2024] [Indexed: 05/16/2024] Open
Abstract
Background Tuberculous meningitis (TBM) is a devastating form of tuberculosis (TB) causing high mortality and disability. TBM arises due to immune dysregulation, but the underlying immune mechanisms are unclear. Methods We performed single-cell RNA sequencing on peripheral blood mononuclear cells (PBMCs) and cerebrospinal fluid (CSF) cells isolated from children (n=6) with TBM using 10 xGenomics platform. We used unsupervised clustering of cells and cluster visualization based on the gene expression profiles, and validated the protein and cytokines by ELISA analysis. Results We revealed for the first time 33 monocyte populations across the CSF cells and PBMCs of children with TBM. Within these populations, we saw that CD4_C04 cells with Th17 and Th1 phenotypes and Macro_C01 cells with a microglia phenotype, were enriched in the CSF. Lineage tracking analysis of monocyte populations revealed myeloid cell populations, as well as subsets of CD4 and CD8 T-cell populations with distinct effector functions. Importantly, we discovered that complement-activated microglial Macro_C01 cells are associated with a neuroinflammatory response that leads to persistent meningitis. Consistently, we saw an increase in complement protein (C1Q), inflammatory markers (CRP) and inflammatory factor (TNF-α and IL-6) in CSF cells but not blood. Finally, we inferred that Macro_C01 cells recruit CD4_C04 cells through CXCL16/CXCR6. Discussion We proposed that the microglial Macro_C01 subset activates complement and interacts with the CD4_C04 cell subset to amplify inflammatory signals, which could potentially contribute to augment inflammatory signals, resulting in hyperinflammation and an immune response elicited by Mtb-infected tissues.
Collapse
Affiliation(s)
- Siwei Mo
- Department of Respiratory Medicine, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children’s Hospital of Chongqing Medical University, Chongqing Key Laboratory of Pediatrics, Chongqing, China
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, School of Medicine, Shenzhen University, Shenzhen, China
| | - Chenyan Shi
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, School of Medicine, Shenzhen University, Shenzhen, China
- School of Public Health, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China
| | - Yi Cai
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, School of Medicine, Shenzhen University, Shenzhen, China
| | - Maozhu Xu
- Maternal and Child Care Health Hospital of Zunyi City, Zunyi, Guizhou, China
| | - Hongmei Xu
- Department of Infectious Diseases, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Yuzhong Xu
- Department of Clinical Laboratory, Shenzhen Baoan Hospital, The Second Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Kehong Zhang
- Department of Clinical Laboratory, Shenzhen Baoan Hospital, The Second Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Yue Zhang
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, School of Medicine, Shenzhen University, Shenzhen, China
| | - Jiao Liu
- Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Siyi Che
- Department of Radiology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Xiangyu Liu
- Department of Respiratory Medicine, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children’s Hospital of Chongqing Medical University, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Chaonan Xing
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, School of Medicine, Shenzhen University, Shenzhen, China
| | - Xiaoru Long
- Department of Respiratory Medicine, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children’s Hospital of Chongqing Medical University, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Xinchun Chen
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, School of Medicine, Shenzhen University, Shenzhen, China
| | - Enmei Liu
- Department of Respiratory Medicine, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children’s Hospital of Chongqing Medical University, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| |
Collapse
|
4
|
Bellini C, Vergara E, Bencs F, Fodor K, Bősze S, Krivić D, Bacsa B, Surguta SE, Tóvári J, Reljic R, Horváti K. Design and Characterization of a Multistage Peptide-Based Vaccine Platform to Target Mycobacterium tuberculosis Infection. Bioconjug Chem 2023; 34:1738-1753. [PMID: 37606258 PMCID: PMC10587871 DOI: 10.1021/acs.bioconjchem.3c00273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/09/2023] [Indexed: 08/23/2023]
Abstract
The complex immunopathology ofMycobacterium tuberculosis(Mtb) is one of the main challenges in developing a novel vaccine against this pathogen, particularly regarding eliciting protection against both active and latent stages. Multistage vaccines, which contain antigens expressed in both phases, represent a promising strategy for addressing this issue, as testified by the tuberculosis vaccine clinical pipeline. Given this approach, we designed and characterized a multistage peptide-based vaccine platform containing CD4+ and CD8+ T cell epitopes previously validated for inducing a relevant T cell response against Mtb. After preliminary screening, CFP10 (32-39), GlfT2 (4-12), HBHA (185-194), and PPE15 (1-15) were selected as promising candidates, and we proved that the PM1 pool of these peptides triggered a T cell response in Mtb-sensitized human peripheral blood mononuclear cells (PBMCs). Taking advantage of the use of thiol-maleimide chemoselective ligation, we synthesized a multiepitope conjugate (Ac-CGHP). Our results showed a structure-activity relationship between the conjugation and a higher tendency to fold and assume an ordered secondary structure. Moreover, the palmitoylated conjugate (Pal-CGHP) comprising the same peptide antigens was associated with an enhanced cellular uptake in human and murine antigen-presenting cells and a better immunogenicity profile. Immunization study, conducted in BALB/c mice, showed that Pal-CGHP induced a significantly higher T cell proliferation and production of IFNγ and TNFα over PM1 formulated in the Sigma Adjuvant System.
Collapse
Affiliation(s)
- Chiara Bellini
- MTA-TTK
Lendület “Momentum” Peptide-Based Vaccines Research
Group, Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences, Budapest 1117, Hungary
- Hevesy
György PhD School of Chemistry, Eötvös
Loránd University, Budapest 1117, Hungary
| | - Emil Vergara
- Institute
for Infection and Immunity, St. George’s,
University of London, London SW17 0RE, U.K.
| | - Fruzsina Bencs
- Hevesy
György PhD School of Chemistry, Eötvös
Loránd University, Budapest 1117, Hungary
- Laboratory
of Structural Chemistry and Biology, Institute of Chemistry, Eötvös Loránd University, Budapest 1117, Hungary
| | - Kinga Fodor
- Department
of Laboratory Animal Science and Animal Protection, University of Veterinary Medicine, Budapest 1078, Hungary
| | - Szilvia Bősze
- ELKH-ELTE
Research Group of Peptide Chemistry, Eötvös Loránd
Research Network (ELKH), Eötvös
Loránd University, Budapest 1117, Hungary
| | - Denis Krivić
- Division
of Medical Physics and Biophysics, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria
| | - Bernadett Bacsa
- Division
of Medical Physics and Biophysics, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria
| | - Sára Eszter Surguta
- Department
of Experimental Pharmacology and National Tumor Biology Laboratory, National Institute of Oncology, Budapest 1122, Hungary
| | - József Tóvári
- Department
of Experimental Pharmacology and National Tumor Biology Laboratory, National Institute of Oncology, Budapest 1122, Hungary
| | - Rajko Reljic
- Institute
for Infection and Immunity, St. George’s,
University of London, London SW17 0RE, U.K.
| | - Kata Horváti
- MTA-TTK
Lendület “Momentum” Peptide-Based Vaccines Research
Group, Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences, Budapest 1117, Hungary
| |
Collapse
|
5
|
Miao B, Hu Z, Mezzadra R, Hoeijmakers L, Fauster A, Du S, Yang Z, Sator-Schmitt M, Engel H, Li X, Broderick C, Jin G, Gomez-Eerland R, Rozeman L, Lei X, Matsuo H, Yang C, Hofland I, Peters D, Broeks A, Laport E, Fitz A, Zhao X, Mahmoud MAA, Ma X, Sander S, Liu HK, Cui G, Gan Y, Wu W, Xiao Y, Heck AJR, Guan W, Lowe SW, Horlings HM, Wang C, Brummelkamp TR, Blank CU, Schumacher TNM, Sun C. CMTM6 shapes antitumor T cell response through modulating protein expression of CD58 and PD-L1. Cancer Cell 2023; 41:1817-1828.e9. [PMID: 37683639 PMCID: PMC11113010 DOI: 10.1016/j.ccell.2023.08.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 06/02/2023] [Accepted: 08/15/2023] [Indexed: 09/10/2023]
Abstract
The dysregulated expression of immune checkpoint molecules enables cancer cells to evade immune destruction. While blockade of inhibitory immune checkpoints like PD-L1 forms the basis of current cancer immunotherapies, a deficiency in costimulatory signals can render these therapies futile. CD58, a costimulatory ligand, plays a crucial role in antitumor immune responses, but the mechanisms controlling its expression remain unclear. Using two systematic approaches, we reveal that CMTM6 positively regulates CD58 expression. Notably, CMTM6 interacts with both CD58 and PD-L1, maintaining the expression of these two immune checkpoint ligands with opposing functions. Functionally, the presence of CMTM6 and CD58 on tumor cells significantly affects T cell-tumor interactions and response to PD-L1-PD-1 blockade. Collectively, these findings provide fundamental insights into CD58 regulation, uncover a shared regulator of stimulatory and inhibitory immune checkpoints, and highlight the importance of tumor-intrinsic CMTM6 and CD58 expression in antitumor immune responses.
Collapse
Affiliation(s)
- Beiping Miao
- German Cancer Research Center (DKFZ) Heidelberg, Division Immune Regulation in Cancer, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhaoqing Hu
- German Cancer Research Center (DKFZ) Heidelberg, Division Immune Regulation in Cancer, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Riccardo Mezzadra
- Division of Molecular Oncology & Immunology, Netherlands Cancer Institute, Oncode Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands; Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Lotte Hoeijmakers
- Division of Molecular Oncology & Immunology, Netherlands Cancer Institute, Oncode Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Astrid Fauster
- Division of Biochemistry, Netherlands Cancer Institute, Oncode Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Shangce Du
- German Cancer Research Center (DKFZ) Heidelberg, Division Immune Regulation in Cancer, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Faculty of Medicine, Heidelberg University, 69120 Heidelberg, Germany
| | - Zhi Yang
- Department of Gastrointestinal Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Melanie Sator-Schmitt
- German Cancer Research Center (DKFZ) Heidelberg, Division Immune Regulation in Cancer, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Helena Engel
- German Cancer Research Center (DKFZ) Heidelberg, Division Immune Regulation in Cancer, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Xueshen Li
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, USA
| | - Caroline Broderick
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Guangzhi Jin
- Department of Interventional Radiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, No. 1111, Xianxia Road, Shanghai 200336, China
| | - Raquel Gomez-Eerland
- Division of Molecular Oncology & Immunology, Netherlands Cancer Institute, Oncode Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Lisette Rozeman
- Division of Molecular Oncology & Immunology, Netherlands Cancer Institute, Oncode Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Xin Lei
- Department of Immunology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Hitoshi Matsuo
- German Cancer Research Center (DKFZ) Heidelberg, Division Immune Regulation in Cancer, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Chen Yang
- German Cancer Research Center (DKFZ) Heidelberg, Division Immune Regulation in Cancer, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ingrid Hofland
- Core Facility Molecular Pathology & Biobanking, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Dennis Peters
- Core Facility Molecular Pathology & Biobanking, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Annegien Broeks
- Core Facility Molecular Pathology & Biobanking, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Elke Laport
- German Cancer Research Center (DKFZ) Heidelberg, Division Immune Regulation in Cancer, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Annika Fitz
- German Cancer Research Center (DKFZ) Heidelberg, Division Immune Regulation in Cancer, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Xiyue Zhao
- German Cancer Research Center (DKFZ) Heidelberg, Division Immune Regulation in Cancer, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Mohamed A A Mahmoud
- German Cancer Research Center (DKFZ) Heidelberg, Division Immune Regulation in Cancer, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Xiujian Ma
- Faculty of Medicine, Heidelberg University, 69120 Heidelberg, Germany; German Cancer Research Center (DKFZ) Heidelberg, Division Molecular Neurogenetics, DKFZ-ZMBH Alliance, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Sandrine Sander
- German Cancer Research Center (DKFZ) Heidelberg, Division Adaptive Immunity and Lymphoma , Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Hai-Kun Liu
- German Cancer Research Center (DKFZ) Heidelberg, Division Molecular Neurogenetics, DKFZ-ZMBH Alliance, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Guoliang Cui
- German Cancer Research Center (DKFZ) Heidelberg, Division T Cell Metabolism, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Yu Gan
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, USA
| | - Wei Wu
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Padualaan 8, 3584 CH Utrecht, the Netherlands; Netherlands Proteomics Centre, Padualaan 8, 3584 CH Utrecht, the Netherlands; Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore; Department of Pharmacy, National University of Singapore, Singapore 117543, Singapore
| | - Yanling Xiao
- Department of Immunology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Albert J R Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Padualaan 8, 3584 CH Utrecht, the Netherlands; Netherlands Proteomics Centre, Padualaan 8, 3584 CH Utrecht, the Netherlands
| | - Wenxian Guan
- Department of Gastrointestinal Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Scott W Lowe
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Hugo M Horlings
- Department of Pathology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Cun Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Thijn R Brummelkamp
- Division of Biochemistry, Netherlands Cancer Institute, Oncode Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Christian U Blank
- Division of Molecular Oncology & Immunology, Netherlands Cancer Institute, Oncode Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands; Department of Medical Oncology, Netherlands Cancer Institute (NKI), Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands; Department of Medical Oncology, Leiden University Medical Centre (LUMC), Leiden, The Netherlands.
| | - Ton N M Schumacher
- Division of Molecular Oncology & Immunology, Netherlands Cancer Institute, Oncode Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands; Department of Hematology, Leiden University Medical Center (LUMC), Leiden, the Netherlands.
| | - Chong Sun
- German Cancer Research Center (DKFZ) Heidelberg, Division Immune Regulation in Cancer, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany.
| |
Collapse
|
6
|
Grant NL, Kelly K, Maiello P, Abbott H, O’Connor S, Lin PL, Scanga CA, Flynn JL. Mycobacterium tuberculosis-Specific CD4 T Cells Expressing Transcription Factors T-Bet or RORγT Associate with Bacterial Control in Granulomas. mBio 2023; 14:e0047723. [PMID: 37039646 PMCID: PMC10294621 DOI: 10.1128/mbio.00477-23] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 03/16/2023] [Indexed: 04/12/2023] Open
Abstract
Despite the extensive research on CD4 T cells within the context of Mycobacterium tuberculosis (Mtb) infections, few studies have focused on identifying and investigating the profile of Mtb-specific T cells within lung granulomas. To facilitate the identification of Mtb-specific CD4 T cells, we identified immunodominant epitopes for two Mtb proteins, namely, Rv1196 and Rv0125, using a Mauritian cynomolgus macaque model of Mtb infection, thereby providing data for the synthesis of MHC class II tetramers. Using tetramers, we identified Mtb-specific cells within different immune compartments, postinfection. We found that granulomas were enriched sites for Mtb-specific cells and that tetramer+ cells had increased frequencies of the activation marker CD69 as well as the transcription factors T-bet and RORγT, compared to tetramer negative cells within the same sample. Our data revealed that while the frequency of Rv1196 tetramer+ cells was positively correlated with the granuloma bacterial burden, the frequency of RORγT or T-bet within tetramer+ cells was inversely correlated with the granuloma bacterial burden, thereby highlighting the importance of having activated, polarized, Mtb-specific cells for the control of Mtb in lung granulomas. IMPORTANCE Tuberculosis, caused by the bacterial pathogen Mycobacterium tuberculosis, kills 1.5 million people each year, despite the existence of effective drugs and a vaccine that is given to infants in most countries. Clearly, we need better vaccines against this disease. However, our understanding of the immune responses that are necessary to prevent tuberculosis is incomplete. This study seeks to understand the functions of T cells that are specific for M. tuberculosis at the site of the disease in the lungs. For this, we developed specialized tools called MHC class II tetramers to identify those T cells that can recognize M. tuberculosis and applied the tools to the study of this infection in nonhuman primate models that mimic human tuberculosis. We demonstrate that M. tuberculosis-specific T cells in lung lesions are associated with control of the bacteria only when those T cells are expressing certain functions, thereby highlighting the importance of combining the identification of specific T cells with functional analyses. Thus, we surmise that these functions of specific T cells are critical to the control of infection and should be considered as a part of the development of vaccines against tuberculosis.
Collapse
Affiliation(s)
- Nicole L. Grant
- Department of Infectious Disease and Microbiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, USA
| | - Kristen Kelly
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Pauline Maiello
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Helena Abbott
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Shelby O’Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison Wisconsin, USA
| | - Philana Ling Lin
- Department of Pediatrics, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Charles A. Scanga
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - JoAnne L. Flynn
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
7
|
Leddy O, White FM, Bryson BD. Immunopeptidomics reveals determinants of Mycobacterium tuberculosis antigen presentation on MHC class I. eLife 2023; 12:e84070. [PMID: 37073954 PMCID: PMC10159623 DOI: 10.7554/elife.84070] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 04/17/2023] [Indexed: 04/20/2023] Open
Abstract
CD8+ T cell recognition of Mycobacterium tuberculosis (Mtb)-specific peptides presented on major histocompatibility complex class I (MHC-I) contributes to immunity to tuberculosis (TB), but the principles that govern presentation of Mtb antigens on MHC-I are incompletely understood. In this study, mass spectrometry (MS) analysis of the MHC-I repertoire of Mtb-infected primary human macrophages reveals that substrates of Mtb's type VII secretion systems (T7SS) are overrepresented among Mtb-derived peptides presented on MHC-I. Quantitative, targeted MS shows that ESX-1 activity is required for presentation of Mtb peptides derived from both ESX-1 substrates and ESX-5 substrates on MHC-I, consistent with a model in which proteins secreted by multiple T7SSs access a cytosolic antigen processing pathway via ESX-1-mediated phagosome permeabilization. Chemical inhibition of proteasome activity, lysosomal acidification, or cysteine cathepsin activity did not block presentation of Mtb antigens on MHC-I, suggesting involvement of other proteolytic pathways or redundancy among multiple pathways. Our study identifies Mtb antigens presented on MHC-I that could serve as targets for TB vaccines, and reveals how the activity of multiple T7SSs interacts to contribute to presentation of Mtb antigens on MHC-I.
Collapse
Affiliation(s)
- Owen Leddy
- Department of Biological Engineering, Massachusetts Institute of TechnologyCambridgeUnited States
- Ragon Institute of Massachusetts General Hospital, Harvard, and MITCambridgeUnited States
- Koch Institute for Integrative Cancer ResearchCambridgeUnited States
| | - Forest M White
- Department of Biological Engineering, Massachusetts Institute of TechnologyCambridgeUnited States
- Koch Institute for Integrative Cancer ResearchCambridgeUnited States
- Center for Precision Cancer MedicineCambridgeUnited States
| | - Bryan D Bryson
- Department of Biological Engineering, Massachusetts Institute of TechnologyCambridgeUnited States
- Ragon Institute of Massachusetts General Hospital, Harvard, and MITCambridgeUnited States
| |
Collapse
|
8
|
Larsen SE, Erasmus JH, Reese VA, Pecor T, Archer J, Kandahar A, Hsu FC, Nicholes K, Reed SG, Baldwin SL, Coler RN. An RNA-Based Vaccine Platform for Use against Mycobacterium tuberculosis. Vaccines (Basel) 2023; 11:vaccines11010130. [PMID: 36679975 PMCID: PMC9862644 DOI: 10.3390/vaccines11010130] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/21/2022] [Accepted: 12/28/2022] [Indexed: 01/07/2023] Open
Abstract
Mycobacterium tuberculosis (M.tb), a bacterial pathogen that causes tuberculosis disease (TB), exerts an extensive burden on global health. The complex nature of M.tb, coupled with different TB disease stages, has made identifying immune correlates of protection challenging and subsequently slowing vaccine candidate progress. In this work, we leveraged two delivery platforms as prophylactic vaccines to assess immunity and subsequent efficacy against low-dose and ultra-low-dose aerosol challenges with M.tb H37Rv in C57BL/6 mice. Our second-generation TB vaccine candidate ID91 was produced as a fusion protein formulated with a synthetic TLR4 agonist (glucopyranosyl lipid adjuvant in a stable emulsion) or as a novel replicating-RNA (repRNA) formulated in a nanostructured lipid carrier. Protein subunit- and RNA-based vaccines preferentially elicit cellular immune responses to different ID91 epitopes. In a single prophylactic immunization screen, both platforms reduced pulmonary bacterial burden compared to the controls. Excitingly, in prime-boost strategies, the groups that received heterologous RNA-prime, protein-boost or combination immunizations demonstrated the greatest reduction in bacterial burden and a unique humoral and cellular immune response profile. These data are the first to report that repRNA platforms are a viable system for TB vaccines and should be pursued with high-priority M.tb antigens containing CD4+ and CD8+ T-cell epitopes.
Collapse
Affiliation(s)
- Sasha E. Larsen
- Center for Global Infectious Disease Research, Seattle Childrens Research Institute, Seattle, WA 98109, USA
| | - Jesse H. Erasmus
- HDT BioCorp, Seattle, WA 98102, USA
- Department of Microbiology, University of Washington, Seattle, WA 98109, USA
| | - Valerie A. Reese
- Center for Global Infectious Disease Research, Seattle Childrens Research Institute, Seattle, WA 98109, USA
| | - Tiffany Pecor
- Center for Global Infectious Disease Research, Seattle Childrens Research Institute, Seattle, WA 98109, USA
| | | | | | | | | | | | - Susan L. Baldwin
- Center for Global Infectious Disease Research, Seattle Childrens Research Institute, Seattle, WA 98109, USA
| | - Rhea N. Coler
- Center for Global Infectious Disease Research, Seattle Childrens Research Institute, Seattle, WA 98109, USA
- Department of Pediatrics, University of Washington, School of Medicine, Seattle, WA 98105, USA
- Department of Global Health, University of Washington, Seattle, WA 98105, USA
- Correspondence:
| |
Collapse
|
9
|
Rais M, Abdelaal H, Reese VA, Ferede D, Larsen SE, Pecor T, Erasmus JH, Archer J, Khandhar AP, Cooper SK, Podell BK, Reed SG, Coler RN, Baldwin SL. Immunogenicity and protection against Mycobacterium avium with a heterologous RNA prime and protein boost vaccine regimen. Tuberculosis (Edinb) 2023; 138:102302. [PMID: 36586154 PMCID: PMC10361416 DOI: 10.1016/j.tube.2022.102302] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/16/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022]
Abstract
Prophylactic efficacy of two different delivery platforms for vaccination against Mycobacterium avium (M. avium) were tested in this study; a subunit and an RNA-based vaccine. The vaccine antigen, ID91, includes four mycobacterial antigens: Rv3619, Rv2389, Rv3478, and Rv1886. We have shown that ID91+GLA-SE is effective against a clinical NTM isolate, M. avium 2-151 smt. Here, we extend these results and show that a heterologous prime/boost strategy with a repRNA-ID91 (replicon RNA) followed by protein ID91+GLA-SE boost is superior to the subunit protein vaccine given as a homologous prime/boost regimen. The repRNA-ID91/ID91+GLA-SE heterologous regimen elicited a higher polyfunctional CD4+ TH1 immune response when compared to the homologous protein prime/boost regimen. More significantly, among all the vaccine regimens tested only repRNA-ID91/ID91+GLA-SE induced IFN-γ and TNF-secreting CD8+ T cells. Furthermore, the repRNA-ID91/ID91+GLA-SE vaccine strategy elicited high systemic proinflammatory cytokine responses and induced strong ID91 and an Ag85B-specific humoral antibody response a pre- and post-challenge with M. avium 2-151 smt. Finally, while all prophylactic prime/boost vaccine regimens elicited a degree of protection in beige mice, the heterologous repRNA-ID91/ID91+GLA-SE vaccine regimen provided greater pulmonary protection than the homologous protein prime/boost regimen. These data indicate that a prophylactic heterologous repRNA-ID91/ID91+GLA-SE vaccine regimen augments immunogenicity and confers protection against M. avium.
Collapse
Affiliation(s)
- Maham Rais
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98145, USA
| | - Hazem Abdelaal
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98145, USA
| | - Valerie A Reese
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98145, USA
| | - Debora Ferede
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98145, USA
| | - Sasha E Larsen
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98145, USA
| | - Tiffany Pecor
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98145, USA
| | | | | | | | - Sarah K Cooper
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98145, USA
| | - Brendan K Podell
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98145, USA
| | | | - Rhea N Coler
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98145, USA; Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, 98195, USA; Department of Global Health, University of Washington, Seattle, WA, 98195, USA
| | - Susan L Baldwin
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98145, USA.
| |
Collapse
|
10
|
Evaluating the Performance of PPE44, HSPX, ESAT-6 and CFP-10 Factors in Tuberculosis Subunit Vaccines. Curr Microbiol 2022; 79:260. [PMID: 35852636 PMCID: PMC9295111 DOI: 10.1007/s00284-022-02949-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 06/23/2022] [Indexed: 11/26/2022]
Abstract
Mycobacterium tuberculosis (M. tuberculosis) is an intracellular pathogen causing long-term infection in humans that mainly attacks macrophages and can escape from the immune system with the various mechanisms. The only FDA-approved vaccine against M. tuberculosis (MTB) is Mycobacterium bovis bacillus Calmette-Guérin (BCG). The protection of this vaccine typically lasts 10–15 years. Due to the increasing number of people becoming ill with MTB each year worldwide, the need to develop a new effective treatment against the disease has been increased. During the past two decades, the research budget for TB vaccine has quadrupled to over half a billion dollars. Most of these research projects were based on amplifying and stimulating the response of T-cells and developing the subunit vaccines. Additionally, these studies have demonstrated that secretory and immunogenic proteins of MTB play a key role in the pathogenesis of the bacteria. Therefore, these proteins were used to develop the new subunit vaccines. In this review, based on the use of these proteins in the successful new subunit vaccines, the PPE44, HSPX, CFP-10 and ESAT-6 antigens were selected and the role of these antigens in designing and developing new subunit vaccines against TB and for the prevention of TB were investigated.
Collapse
|
11
|
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: 7.5] [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
|
12
|
Larsen SE, Williams BD, Rais M, Coler RN, Baldwin SL. It Takes a Village: The Multifaceted Immune Response to Mycobacterium tuberculosis Infection and Vaccine-Induced Immunity. Front Immunol 2022; 13:840225. [PMID: 35359957 PMCID: PMC8960931 DOI: 10.3389/fimmu.2022.840225] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/08/2022] [Indexed: 11/18/2022] Open
Abstract
Despite co-evolving with humans for centuries and being intensely studied for decades, the immune correlates of protection against Mycobacterium tuberculosis (Mtb) have yet to be fully defined. This lapse in understanding is a major lag in the pipeline for evaluating and advancing efficacious vaccine candidates. While CD4+ T helper 1 (TH1) pro-inflammatory responses have a significant role in controlling Mtb infection, the historically narrow focus on this cell population may have eclipsed the characterization of other requisite arms of the immune system. Over the last decade, the tuberculosis (TB) research community has intentionally and intensely increased the breadth of investigation of other immune players. Here, we review mechanistic preclinical studies as well as clinical anecdotes that suggest the degree to which different cell types, such as NK cells, CD8+ T cells, γ δ T cells, and B cells, influence infection or disease prevention. Additionally, we categorically outline the observed role each major cell type plays in vaccine-induced immunity, including Mycobacterium bovis bacillus Calmette-Guérin (BCG). Novel vaccine candidates advancing through either the preclinical or clinical pipeline leverage different platforms (e.g., protein + adjuvant, vector-based, nucleic acid-based) to purposefully elicit complex immune responses, and we review those design rationales and results to date. The better we as a community understand the essential composition, magnitude, timing, and trafficking of immune responses against Mtb, the closer we are to reducing the severe disease burden and toll on human health inflicted by TB globally.
Collapse
Affiliation(s)
- Sasha E. Larsen
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States
| | - Brittany D. Williams
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States,Department of Global Health, University of Washington, Seattle, WA, United States
| | - Maham Rais
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States
| | - Rhea N. Coler
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States,Department of Global Health, University of Washington, Seattle, WA, United States,Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, United States
| | - Susan L. Baldwin
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States,*Correspondence: Susan L. Baldwin,
| |
Collapse
|
13
|
Masjedi M, Montahaei T, Sharafi Z, Jalali A. Pulmonary vaccine delivery: An emerging strategy for vaccination and immunotherapy. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
14
|
Cai Y, Wang Y, Shi C, Dai Y, Li F, Xu Y, Zhang P, Kong F, Deng G, Wen Z, Zhou Q, Kang BC, Singhal A, Yang Q, Feng CG, Chen X. Single-cell immune profiling reveals functional diversity of T cells in tuberculous pleural effusion. J Exp Med 2022; 219:212978. [PMID: 35061012 PMCID: PMC8789099 DOI: 10.1084/jem.20211777] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 11/04/2021] [Accepted: 12/23/2021] [Indexed: 12/31/2022] Open
Abstract
Orchestration of an effective T lymphocyte response at infection sites is critical for protection against Mycobacterium tuberculosis (Mtb) infection. However, the local T cell immunity landscape in human tuberculosis is poorly defined. Tuberculous pleural effusion (TPE), caused by Mtb, is characterized by an influx of leukocytes to the pleural space, providing a platform suitable for delineating complex tissue responses to Mtb infection. Using single-cell transcriptomics and T cell receptor sequencing, we analyzed mononuclear cell populations in paired pleural fluid and peripheral blood of TPE patients. While all major cell clusters were present in both tissues, their relative proportions varied significantly by anatomic location. Lineage tracking analysis revealed subsets of CD8 and CD4 T cell populations with distinct effector functions specifically expanded at pleural sites. Granzyme K–expressing CD8 T cells were preferentially enriched and clonally expanded in pleural fluid from TPE, suggesting that they are involved in the pathogenesis of the disease. The findings collectively reveal the landscape of local T cell immunity in tuberculosis.
Collapse
Affiliation(s)
- Yi Cai
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, China
| | - Yejun Wang
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, China
| | - Chenyan Shi
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, China
| | - Youchao Dai
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, China
| | - Fuxiang Li
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, China
| | - Yuzhong Xu
- Department of Clinical Laboratory, Shenzhen Baoan hospital, Shenzhen, China
| | - Peize Zhang
- Guangdong Key Lab for Diagnosis & Treatment of Emerging Infectious Diseases, Shenzhen Third People’s Hospital, Shenzhen, China
| | | | - Guofang Deng
- Guangdong Key Lab for Diagnosis & Treatment of Emerging Infectious Diseases, Shenzhen Third People’s Hospital, Shenzhen, China
| | - Zhihua Wen
- Shenzhen University and Yuebei Second People’s Hospital Joint Lab, Yuebei Second People's Hospital, Shaoguan, China
| | - Qi Zhou
- Analytical Biosciences Limited, Beijing, China
| | | | - Amit Singhal
- Infectious Diseases Labs, Agency for Science, Technology and Research, Singapore
| | - Qianting Yang
- Guangdong Key Lab for Diagnosis & Treatment of Emerging Infectious Diseases, Shenzhen Third People’s Hospital, Shenzhen, China
| | - Carl G. Feng
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, China
- Immunology and Host Defense Group, School of Medical Sciences, Faculty of Medicine and Health, the University of Sydney, Sydney, New South Wales, Australia
| | - Xinchun Chen
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University School of Medicine, Shenzhen, China
| |
Collapse
|
15
|
Sutiwisesak R, Hicks ND, Boyce S, Murphy KC, Papavinasasundaram K, Carpenter SM, Boucau J, Joshi N, Le Gall S, Fortune SM, Sassetti CM, Behar SM. A natural polymorphism of Mycobacterium tuberculosis in the esxH gene disrupts immunodomination by the TB10.4-specific CD8 T cell response. PLoS Pathog 2020; 16:e1009000. [PMID: 33075106 PMCID: PMC7597557 DOI: 10.1371/journal.ppat.1009000] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 10/29/2020] [Accepted: 09/23/2020] [Indexed: 12/20/2022] Open
Abstract
CD8 T cells provide limited protection against Mycobacterium
tuberculosis (Mtb) infection in the mouse model. As Mtb causes
chronic infection in mice and humans, we hypothesize that Mtb impairs T cell
responses as an immune evasion strategy. TB10.4 is an immunodominant antigen in
people, nonhuman primates, and mice, which is encoded by the
esxH gene. In C57BL/6 mice, 30–50% of pulmonary CD8 T cells
recognize the TB10.44−11 epitope. However, TB10.4-specific CD8 T
cells fail to recognize Mtb-infected macrophages. We speculate that Mtb elicits
immunodominant CD8 T cell responses to antigens that are inefficiently presented
by infected cells, thereby focusing CD8 T cells on nonprotective antigens. Here,
we leverage naturally occurring polymorphisms in esxH, which
frequently occur in lineage 1 strains, to test this “decoy hypothesis”. Using
the clinical isolate 667, which contains an EsxHA10T polymorphism, we
observe a drastic change in the hierarchy of CD8 T cells. Using isogenic
Erd.EsxHA10T and Erd.EsxHWT strains, we prove that
this polymorphism alters the hierarchy of immunodominant CD8 T cell responses.
Our data are best explained by immunodomination, a mechanism by which
competition for APC leads to dominant responses suppressing subdominant
responses. These results were surprising as the variant epitope can bind to
H2-Kb and is recognized by TB10.4-specific CD8 T cells. The
dramatic change in TB10.4-specific CD8 responses resulted from increased
proteolytic degradation of A10T variant, which destroyed the
TB10.44-11epitope. Importantly, this polymorphism affected T cell
priming and recognition of infected cells. These data support a model in which
nonprotective CD8 T cells become immunodominant and suppress subdominant
responses. Thus, polymorphisms between clinical Mtb strains, and BCG or H37Rv
sequence-based vaccines could lead to a mismatch between T cells that are primed
by vaccines and the epitopes presented by infected cells. Reprograming host
immune responses should be considered in the future design of vaccines. An important question for vaccine developers is the relative potency of CD4 vs.
CD8 T cells against Mtb, as strategies differ for eliciting these different T
cell subsets. Despite robust antigen-specific pulmonary CD8 T cell responses,
CD4 T cells mediate more protection than CD8 T cells in the murine model. Most
CD8 T cells recognize a single antigen, TB10.4, which is encoded by the
esxH gene. Based on finding that
TB10.44−11-specific CD8 T cells poorly recognize Mtb-infected
macrophages, we hypothesized that Mtb evades detection by CD8 T cells and
focuses the CD8 T cell response on non-protective antigen. We termed these
antigens “decoy antigens.” To test this hypothesis, we took advantage of a
natural variant of the esxH gene, which contains an A10T
polymorphism within the TB10.44−11 epitope. This polymorphism
drastically alters the hierarchy of CD8 T cell responses elicited by Mtb. These
data suggest that immunodomination by the TB10.4 epitope acts to suppress
subdominant CD8 T cell responses to other Mtb antigens, impairing the CD8 T cell
response to other Mtb antigens, some of which might be presented by Mtb-infected
macrophages and be targets of protective immunity. Importantly, this single
amino acid polymorphism, which does not significantly alter MHC-binding or T
cell recognition, alters the half-life of the epitope and consequently, has a
profound effect on CD8 T cell priming and recognition of infected cells. These
data also provide a mechanism that could be exploited to manipulate the
hierarchy of immunodominant responses.
Collapse
Affiliation(s)
- Rujapak Sutiwisesak
- Immunology and Microbiology Program, Graduate School of Biomedical
Science, University of Massachusetts Medical School, Worcester, Massachusetts,
United States of America
- Department of Microbiology and Physiological Systems, University of
Massachusetts Medical School, Worcester, Massachusetts, United States of
America
| | - Nathan D. Hicks
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan
School of Public Health, Boston, Massachusetts, United States of
America
| | - Shayla Boyce
- Department of Microbiology and Physiological Systems, University of
Massachusetts Medical School, Worcester, Massachusetts, United States of
America
| | - Kenan C. Murphy
- Immunology and Microbiology Program, Graduate School of Biomedical
Science, University of Massachusetts Medical School, Worcester, Massachusetts,
United States of America
- Department of Microbiology and Physiological Systems, University of
Massachusetts Medical School, Worcester, Massachusetts, United States of
America
| | - Kadamba Papavinasasundaram
- Department of Microbiology and Physiological Systems, University of
Massachusetts Medical School, Worcester, Massachusetts, United States of
America
| | - Stephen M. Carpenter
- Department of Microbiology and Physiological Systems, University of
Massachusetts Medical School, Worcester, Massachusetts, United States of
America
| | - Julie Boucau
- Ragon Institute of Massachusetts General Hospital, Massachusetts
Institute of Technology and Harvard University, Cambridge, MA, United States of
America
| | - Neelambari Joshi
- Ragon Institute of Massachusetts General Hospital, Massachusetts
Institute of Technology and Harvard University, Cambridge, MA, United States of
America
| | - Sylvie Le Gall
- Ragon Institute of Massachusetts General Hospital, Massachusetts
Institute of Technology and Harvard University, Cambridge, MA, United States of
America
| | - Sarah M. Fortune
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan
School of Public Health, Boston, Massachusetts, United States of
America
| | - Christopher M. Sassetti
- Immunology and Microbiology Program, Graduate School of Biomedical
Science, University of Massachusetts Medical School, Worcester, Massachusetts,
United States of America
- Department of Microbiology and Physiological Systems, University of
Massachusetts Medical School, Worcester, Massachusetts, United States of
America
| | - Samuel M. Behar
- Immunology and Microbiology Program, Graduate School of Biomedical
Science, University of Massachusetts Medical School, Worcester, Massachusetts,
United States of America
- Department of Microbiology and Physiological Systems, University of
Massachusetts Medical School, Worcester, Massachusetts, United States of
America
- * E-mail:
| |
Collapse
|
16
|
Kathamuthu GR, Moideen K, Sridhar R, Baskaran D, Babu S. Diminished Frequencies of Cytotoxic Marker Expressing T- and NK Cells at the Site of Mycobacterium tuberculosis Infection. Front Immunol 2020; 11:585293. [PMID: 33101317 PMCID: PMC7546427 DOI: 10.3389/fimmu.2020.585293] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/07/2020] [Indexed: 11/25/2022] Open
Abstract
Tuberculous lymphadenitis (TBL) individuals exhibit reduced frequencies of CD8+ T cells expressing cytotoxic markers in peripheral blood. However, the frequencies of cytotoxic marker expressing CD4+, CD8+ T cells, and NK cells at the site of infection is not known. Therefore, we measured the baseline and mycobacterial antigen specific frequencies of cytotoxic markers expressing CD4+, CD8+ T cells, and NK cells in the LN (n = 18) and whole blood (n = 10) of TBL individuals. TBL LN is associated with lower frequencies of CD4+ T cells expressing cytotoxic markers (Granzyme B, CD107a) compared to peripheral blood at baseline and in response to PPD, ESAT-6, and CFP-10 antigen stimulation. Similarly, lower frequencies of CD8+ T cells expressing cytotoxic markers (Perforin, Granzyme B, and CD107a) were also present in the TBL LN at baseline and following (except perforin) antigen stimulation. Finally, at baseline and after antigen (PPD, ESAT-6, and CFP-10) stimulation, frequencies of NK cells expressing cytotoxic markers were also significantly lower in TBL LN compared to whole blood. Hence, TBL is characterized by diminished frequencies of cytotoxic marker expressing CD4+, CD8+ T cells, and NK cells at the site of infection, which might reflect the lack of protective immune responses at the site of Mycobacterium tuberculosis infection.
Collapse
Affiliation(s)
- Gokul Raj Kathamuthu
- International Center for Excellence in Research, National Institutes of Health, National Institute for Research in Tuberculosis, Chennai, India.,National Institute for Research in Tuberculosis (NIRT), Chennai, India
| | - Kadar Moideen
- International Center for Excellence in Research, National Institutes of Health, National Institute for Research in Tuberculosis, Chennai, India
| | | | - Dhanaraj Baskaran
- National Institute for Research in Tuberculosis (NIRT), Chennai, India
| | - Subash Babu
- International Center for Excellence in Research, National Institutes of Health, National Institute for Research in Tuberculosis, Chennai, India.,Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
17
|
Gupta A, Saqib M, Singh B, Pal L, Nishikanta A, Bhaskar S. Mycobacterium indicus pranii Induced Memory T-Cells in Lung Airways Are Sentinels for Improved Protection Against M.tb Infection. Front Immunol 2019; 10:2359. [PMID: 31681272 PMCID: PMC6813244 DOI: 10.3389/fimmu.2019.02359] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 09/19/2019] [Indexed: 12/24/2022] Open
Abstract
The lungs are the most vulnerable site for air-borne infections. Immunologic compartmentalization of the lungs into airway lumen and interstitium has paved the way to determine the immune status of the site of pathogen entry, which is crucial for the outcome of any air-borne infections. Vaccination via the nasal route with Mycobacterium indicus pranii (MIP), a prospective candidate vaccine against tuberculosis (TB), has been reported to confer superior protection as compared to the subcutaneous (s.c.) route in small-animal models of TB. However, the immune mechanism remains only partly understood. Here, we showed that intranasal (i.n.) immunization of mice with MIP resulted in a significant recruitment of CD4+ and CD8+ T-cells expressing activation markers in the lung airway lumen. A strong memory T-cell response was observed in the lung airway lumen after i.n. MIP vaccination, compared with s.c. vaccination. The recruitment of these T-cells was regulated primarily by CXCR3–CXCL11 axis in “MIP i.n.” group. MIP-primed T-cells in the lung airway lumen effectively transferred protective immunity into naïve mice against Mycobacterium tuberculosis (M.tb) infection and helped reducing the pulmonary bacterial burden. These signatures of protective immune response were virtually absent or very low in unimmunized and subcutaneously immunized mice, respectively, before and after M.tb challenge. Our study provides mechanistic insights for MIP-elicited protective response against M.tb infection.
Collapse
Affiliation(s)
- Ananya Gupta
- National Institute of Immunology, Product Development Cell-I, New Delhi, India
| | - Mohd Saqib
- National Institute of Immunology, Product Development Cell-I, New Delhi, India.,Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, United States
| | - Bindu Singh
- National Institute of Immunology, Product Development Cell-I, New Delhi, India
| | - Lalit Pal
- National Institute of Immunology, Product Development Cell-I, New Delhi, India
| | - Akoijam Nishikanta
- National Institute of Immunology, Product Development Cell-I, New Delhi, India
| | - Sangeeta Bhaskar
- National Institute of Immunology, Product Development Cell-I, New Delhi, India
| |
Collapse
|
18
|
Multiplexed Quantitation of Intraphagocyte Mycobacterium tuberculosis Secreted Protein Effectors. Cell Rep 2019; 23:1072-1084. [PMID: 29694886 PMCID: PMC5946722 DOI: 10.1016/j.celrep.2018.03.125] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 02/16/2018] [Accepted: 03/26/2018] [Indexed: 01/21/2023] Open
Abstract
The pathogenic potential of Mycobacterium tuberculosis largely depends on ESX secretion systems exporting members of the multigenic Esx, Esp, and PE/PPE protein families. To study the secretion and regulation patterns of these proteins while circumventing immune cross-reactions due to their extensive sequence homologies, we developed an approach that relies on the recognition of their MHC class II epitopes by highly discriminative T cell receptors (TCRs) of a panel of T cell hybridomas. The latter were engineered so that each expresses a unique fluorescent reporter linked to specific antigen recognition. The resulting polychromatic and multiplexed imaging assay enabled us to measure the secretion of mycobacterial effectors inside infected host cells. We applied this novel technology to a large panel of mutants, clinical isolates, and host-cell types to explore the host-mycobacteria interplay and its impact on the intracellular bacterial secretome, which also revealed the unexpected capacity of phagocytes from lung granuloma to present mycobacterial antigens via MHC class II. T cell hybridomas detect individual mycobacterial proteins without cross-reactivity Detection of mycobacterial proteins by T cells allows visualization of their cellular topography Measurement of intraphagocyte mycobacterial proteins can be performed with T cells A multiplexed assay of mycobacterial protein quantitation has numerous applications
Collapse
|
19
|
Khan A, Bakhru P, Saikolappan S, Das K, Soudani E, Singh CR, Estrella JL, Zhang D, Pasare C, Ma Y, Sun J, Wang J, Hunter RL, Tony Eissa N, Dhandayuthapani S, Jagannath C. An autophagy-inducing and TLR-2 activating BCG vaccine induces a robust protection against tuberculosis in mice. NPJ Vaccines 2019; 4:34. [PMID: 31396406 PMCID: PMC6683161 DOI: 10.1038/s41541-019-0122-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 05/15/2019] [Indexed: 12/28/2022] Open
Abstract
Mycobacterium bovis BCG is widely used as a vaccine against tuberculosis due to M. tuberculosis (Mtb), which kills millions of people each year. BCG variably protects children, but not adults against tuberculosis. BCG evades phagosome maturation, autophagy, and reduces MHC-II expression of antigen-presenting cells (APCs) affecting T-cell activation. To bypass these defects, an autophagy-inducing, TLR-2 activating C5 peptide from Mtb-derived CFP-10 protein was overexpressed in BCG in combination with Ag85B. Recombinant BCG85C5 induced a robust MHC-II-dependent antigen presentation to CD4 T cells in vitro, and elicited stronger TH1 cytokines (IL-12, IL-1β, and TNFα) from APCs of C57Bl/6 mice increasing phosphorylation of p38MAPK and ERK. BCG85C5 also enhanced MHC-II surface expression of MΦs by inhibiting MARCH1 ubiquitin ligase that degrades MHC-II. BCG85C5 infected APCs from MyD88 or TLR-2 knockout mice showed decreased antigen presentation. Furthermore, BCG85C5 induced LC3-dependent autophagy in macrophages increasing antigen presentation. Consistent with in vitro effects, BCG85C5 markedly expanded both effector and central memory T cells in C57Bl/6 mice protecting them against both primary aerosol infection with Mtb and reinfection, but was less effective among TLR-2 knockout mice. Thus, BCG85C5 induces stronger and longer lasting immunity, and is better than BCG against tuberculosis of mice.
Collapse
Affiliation(s)
- Arshad Khan
- Department of Pathology and Laboratory Medicine, University of Texas Health Sciences Center, Houston, TX USA
| | - Pearl Bakhru
- Department of Pathology and Laboratory Medicine, University of Texas Health Sciences Center, Houston, TX USA
| | - Sankaralingam Saikolappan
- Molecular and Translational Medicine, Paul L. Foster School of Medicine Texas Tech University Health Sciences Center, El Paso, TX USA
| | - Kishore Das
- Molecular and Translational Medicine, Paul L. Foster School of Medicine Texas Tech University Health Sciences Center, El Paso, TX USA
| | - Emily Soudani
- Department of Pathology and Laboratory Medicine, University of Texas Health Sciences Center, Houston, TX USA
| | - Christopher R. Singh
- Department of Pathology and Laboratory Medicine, University of Texas Health Sciences Center, Houston, TX USA
| | - Jaymie L. Estrella
- Department of Pathology and Laboratory Medicine, University of Texas Health Sciences Center, Houston, TX USA
| | - Dekai Zhang
- Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX USA
| | - Chandrashekhar Pasare
- Division of Immunobiology, Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229 USA
| | - Yue Ma
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, Houston, TX USA
| | - Jianjun Sun
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, Houston, TX USA
| | - Jin Wang
- Methodist Hospital Research Institute, Houston, TX USA
| | - Robert L. Hunter
- Department of Pathology and Laboratory Medicine, University of Texas Health Sciences Center, Houston, TX USA
| | | | - Subramanian Dhandayuthapani
- Molecular and Translational Medicine, Paul L. Foster School of Medicine Texas Tech University Health Sciences Center, El Paso, TX USA
| | - Chinnaswamy Jagannath
- Department of Pathology and Laboratory Medicine, University of Texas Health Sciences Center, Houston, TX USA
- Methodist Hospital Research Institute, Houston, TX USA
| |
Collapse
|
20
|
Wang C, Lu J, Du W, Wang G, Li X, Shen X, Su C, Yang L, Chen B, Wang J, Xu M. Ag85b/ESAT6-CFP10 adjuvanted with aluminum/poly-IC effectively protects guinea pigs from latent mycobacterium tuberculosis infection. Vaccine 2019; 37:4477-4484. [PMID: 31266673 DOI: 10.1016/j.vaccine.2019.06.078] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 06/17/2019] [Accepted: 06/24/2019] [Indexed: 02/06/2023]
Abstract
The high global burden of tuberculosis (TB) underscores the urgent need for an effective TB vaccine since the only licensed Bacillus Calmette-Guérin (BCG) vaccine is ineffective in preventing adult pulmonary TB and affords no protection against latent TB infection (LTBI). Herein we investigated the potential of Mycobacterium tuberculosis (Mtb) antigen proteins AEC comprised of Ag85b and ESAT6-CFP10 proteins in conjunction with aluminum (Al) and polyriboinosinic-polyribocytidylic acid (poly-IC) as a novel subunit vaccine against TB. The immunogenicity and protection induced by the adjuvanted vaccine were evaluated in two animal models. Mice vaccinated with AEC/Al/poly-IC exhibited significant antigen-specific humoral immune responses and cell-mediated immunity as determined by immunoassay and multicolor flow cytometric assay, and the protective effect of the vaccine was demonstrated in a guinea pig model of latent Mtb infection. Compared to the control group, the mean pathological scores and bacterial loads in lungs and spleens of AEC/Al/poly-IC-immunized guinea pigs were significantly reduced. These data indicate that the AEC/Al/poly-IC is highly immunogenic in mice and can effectively protect guinea pigs against latent Mtb infection; it may represent a promising candidate vaccine for the control of latent TB.
Collapse
Affiliation(s)
- Chunhua Wang
- Division of Tuberculosis Vaccines, National Institutes for Food and Drug Control, Beijing 102629, China; Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Jinbiao Lu
- Division of Tuberculosis Vaccines, National Institutes for Food and Drug Control, Beijing 102629, China
| | - Weixin Du
- Division of Tuberculosis Vaccines, National Institutes for Food and Drug Control, Beijing 102629, China
| | - Guozhi Wang
- Division of Tuberculosis Vaccines, National Institutes for Food and Drug Control, Beijing 102629, China
| | - Xuguang Li
- Centre for Biologicals Evaluation, Biologics and Genetic Therapies Directorate, Health Canada, Ottawa, Ontario, Canada
| | - Xiaobin Shen
- Division of Tuberculosis Vaccines, National Institutes for Food and Drug Control, Beijing 102629, China
| | - Cheng Su
- Division of Tuberculosis Vaccines, National Institutes for Food and Drug Control, Beijing 102629, China
| | - Lei Yang
- Division of Tuberculosis Vaccines, National Institutes for Food and Drug Control, Beijing 102629, China
| | - Baowen Chen
- Division of Tuberculosis Vaccines, National Institutes for Food and Drug Control, Beijing 102629, China
| | - Junzhi Wang
- Division of Tuberculosis Vaccines, National Institutes for Food and Drug Control, Beijing 102629, China.
| | - Miao Xu
- Division of Tuberculosis Vaccines, National Institutes for Food and Drug Control, Beijing 102629, China.
| |
Collapse
|
21
|
Wu Y, Cai M, Ma J, Teng X, Tian M, Bassuoney EBMB, Fan X. Heterologous Boost Following Mycobacterium bovis BCG Reduces the Late Persistent, Rather Than the Early Stage of Intranasal Tuberculosis Challenge Infection. Front Immunol 2018; 9:2439. [PMID: 30425711 PMCID: PMC6218689 DOI: 10.3389/fimmu.2018.02439] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 10/02/2018] [Indexed: 01/06/2023] Open
Abstract
Adults are the leading population affected by tuberculosis (TB) epidemic and death. Developing an effective vaccine against adult TB is urgently needed. Mycobacterium bovis Bacillus Calmette-Guerin (BCG) prime-heterologous boost strategy has been explored extensively to protect adults against primary TB infection, but the majority of experimental regimens have not improved the protection primed by the BCG vaccine. The reason attributed to the failure remains unknown. In this study, CTT3H-based vaccines, namely DMT adjuvanted CTT3H subunit or DNA vaccine (pCTT3H-DMT), and recombinant adenovirus rAdCTT3H were constructed. Protective efficacy and immunogenicity of BCG prime-CTT3H based boosters were compared in C57BL/c mice models of primary or late persistent TB infection. Similar protective efficacy against early intranasal infection was provided by different CTT3H-based vaccines alone in vaccinated mice, and their protection was inferior to that of the BCG vaccine. In addition, CTT3H-based heterologous boosters did not enhance the protection conferred by the BCG vaccine against primary infection. However, all of these three boosters provided stronger protection against late persistent TB infection than BCG alone, regardless of vaccine types. Although BCG prime-boosters elicited Th1-biased responses to the antigen CTT3H, the number of CTT3H-sepcific IFN-γ-expressing TEM (CD62LloCD44hi) and IL-2-expressing TCM (CD62LhiCD44hi) cells in the spleen was not improved before exposure to Mycobacterium tuberculosis infection. In contrast, IFN-γ+ TEM and IL-2+ TCM cells in spleens, especially in lungs were significantly increased in BCG prime-boosters after exposure vaccination. Our results indicate that BCG prime-boost strategy might be a promising measure for the prevention against late persistent TB infection by induction of IFN-γ+ TEM and IL-2+ TCM cells in the lung, which can be used as alternative biomarkers for guiding the clinical practice and future development of TB vaccine for adults.
Collapse
Affiliation(s)
- Yaqi Wu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ming Cai
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jilei Ma
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xindong Teng
- Shandong International Travel Healthcare Center, Shandong Entry-Exit Inspection and Quarantine Bureau, Qingdao, China
| | - Maopeng Tian
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | - Xionglin Fan
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
22
|
Recombinant BCG Expressing ESX-1 of Mycobacterium marinum Combines Low Virulence with Cytosolic Immune Signaling and Improved TB Protection. Cell Rep 2017; 18:2752-2765. [PMID: 28297677 DOI: 10.1016/j.celrep.2017.02.057] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 01/18/2017] [Accepted: 02/16/2017] [Indexed: 12/30/2022] Open
Abstract
Recent insights into the mechanisms by which Mycobacterium tuberculosis, the etiologic agent of human tuberculosis, is recognized by cytosolic nucleotide sensors have opened new avenues for rational vaccine design. The only licensed anti-tuberculosis vaccine, Mycobacterium bovis BCG, provides limited protection. A feature of BCG is the partial deletion of the ESX-1 type VII secretion system, which governs phagosomal rupture and cytosolic pattern recognition, key intracellular phenotypes linked to increased immune signaling. Here, by heterologously expressing the esx-1 region of Mycobacterium marinum in BCG, we engineered a low-virulence, ESX-1-proficient, recombinant BCG (BCG::ESX-1Mmar) that induces the cGas/STING/TBK1/IRF-3/type I interferon axis and enhances AIM2 and NLRP3 inflammasome activity, resulting in both higher proportions of CD8+ T cell effectors against mycobacterial antigens shared with BCG and polyfunctional CD4+ Th1 cells specific to ESX-1 antigens. Importantly, independent mouse vaccination models show that BCG::ESX-1Mmar confers superior protection relative to parental BCG against challenges with highly virulent M. tuberculosis.
Collapse
|
23
|
Reactogenicity to major tuberculosis antigens absent in BCG is linked to improved protection against Mycobacterium tuberculosis. Nat Commun 2017; 8:16085. [PMID: 28706226 PMCID: PMC5519979 DOI: 10.1038/ncomms16085] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 05/25/2017] [Indexed: 12/14/2022] Open
Abstract
MTBVAC is a live-attenuated Mycobacterium tuberculosis vaccine, currently under clinical development, that contains the major antigens ESAT6 and CFP10. These antigens are absent from the current tuberculosis vaccine, BCG. Here we compare the protection induced by BCG and MTBVAC in several mouse strains that naturally express different MHC haplotypes differentially recognizing ESAT6 and CFP10. MTBVAC induces improved protection in C3H mice, the only of the three tested strains reactive to both ESAT6 and CFP10. Deletion of both antigens in MTBVAC reduces its efficacy to BCG levels, supporting a link between greater efficacy and CFP10- and ESAT6-specific reactogenicity. In addition, MTBVAC (but not BCG) triggers a specific response in human vaccinees against ESAT6 and CFP10. Our results warrant further exploration of this response as potential biomarker of protection in MTBVAC clinical trials. MTBVAC, a live attenuated Mycobacterium tuberculosis vaccine under clinical evaluation, contains the major tuberculosis antigens ESAT6 and CFP10, which are absent from the current vaccine, BCG. Here, the authors show that these antigens contribute to enhanced vaccine efficacy in mouse models.
Collapse
|
24
|
Kirschner D, Pienaar E, Marino S, Linderman JJ. A review of computational and mathematical modeling contributions to our understanding of Mycobacterium tuberculosis within-host infection and treatment. CURRENT OPINION IN SYSTEMS BIOLOGY 2017; 3:170-185. [PMID: 30714019 PMCID: PMC6354243 DOI: 10.1016/j.coisb.2017.05.014] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Tuberculosis (TB) is an ancient and deadly disease characterized by complex host-pathogen dynamics playing out over multiple time and length scales and physiological compartments. Computational modeling can be used to integrate various types of experimental data and suggest new hypotheses, mechanisms, and therapeutic approaches to TB. Here, we offer a first-time comprehensive review of work on within-host TB models that describe the immune response of the host to infection, including the formation of lung granulomas. The models include systems of ordinary and partial differential equations and agent-based models as well as hybrid and multi-scale models that are combinations of these. Many aspects of M. tuberculosis infection, including host dynamics in the lung (typical site of infection for TB), granuloma formation, roles of cytokine and chemokine dynamics, and bacterial nutrient availability have been explored. Finally, we survey applications of these within-host models to TB therapy and prevention and suggest future directions to impact this global disease.
Collapse
Affiliation(s)
- Denise Kirschner
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI
| | - Elsje Pienaar
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI
| | - Simeone Marino
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI
| | | |
Collapse
|
25
|
Robinson RT, Huppler AR. The Goldilocks model of immune symbiosis with Mycobacteria and Candida colonizers. Cytokine 2017; 97:49-65. [PMID: 28570933 DOI: 10.1016/j.cyto.2017.05.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 05/15/2017] [Accepted: 05/17/2017] [Indexed: 12/12/2022]
Abstract
Mycobacteria and Candida species include significant human pathogens that can cause localized or disseminated infections. Although these organisms may appear to have little in common, several shared pathways of immune recognition and response are important for both control and infection-related pathology. In this article, we compare and contrast the innate and adaptive components of the immune system that pertain to these infections in humans and animal models. We also explore a relatively new concept in the mycobacterial field: biological commensalism. Similar to the well-established model of Candida infection, Mycobacteria species colonize their human hosts in equilibrium with the immune response. Perturbations in the immune response permit the progression to pathologic disease at the expense of the host. Understanding the immune factors required to maintain commensalism may aid with the development of diagnostic and treatment strategies for both categories of pathogens.
Collapse
Affiliation(s)
- Richard T Robinson
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, USA.
| | - Anna R Huppler
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Pediatrics, Division of Infectious Disease, Medical College of Wisconsin, Children's Hospital and Health System, Children's Research Institute, Milwaukee, WI, USA.
| |
Collapse
|
26
|
Lau A, Singh V, Soualhine H, Hmama Z. Expression of Cathepsin S in BCG converts it into a pro-apoptotic and highly immunogenic strain. Vaccine 2017; 35:2060-2068. [PMID: 28318770 DOI: 10.1016/j.vaccine.2017.02.065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 02/16/2017] [Accepted: 02/28/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND BCG vaccine, introduced almost 100years ago, is the only option to prevent TB disease. It effectively protects newborns from meningeal TB but fails to prevent adult pulmonary TB. TB kills 1.3million people annually in areas where BCG vaccination is widely practiced. Thus, more effective TB vaccines are urgently needed. Others and we have shown that BCG mimics features of virulent M. tuberculosis, in particular attenuation of essential macrophage functions such as phagosome maturation and antigen presentation. One of these studies revealed that defect in antigen presentation is largely due to down-regulation of the cysteine protease Cathepsin S (CatS), which prevents MHC II molecule maturation and proper antigen peptide loading. Recent studies also suggested a potential role for cysteine proteases in the regulation of apoptosis, a key cellular process used by the macrophage to (i) contain and process ingested bacteria and (ii) facilitate cross-talk antigen presentation between the macrophage and dendritic cells. METHOD To reverse the phenotype of vaccine-mediated macrophage attenuation, we engineered a novel BCG strain that expresses and secretes active CatS (rBCG-CatS) to examine its pro-apoptotic properties in vitro, and subsequently, immunogenicity in mice. RESULTS Transcriptomic profiling of macrophages infected with rBCG-CatS, but not BCG, revealed upregulation of key pro-apoptotic genes and downregulation of anti-apoptotic genes, which were further confirmed by RT-qPCR analyses of expression of selected genes. Macrophages infected with rBCG-CatS undergo apoptosis as indicated by increased levels of annexin V staining and intracellular caspase-3 cleavage. Consistent with these findings, mice vaccinated with rBCG-CatS showed increased antigen-specific CD4+ T-cell responses, as well as enhanced cytokine production and proliferation in CD4+ upon ex vivo re-stimulation. CONCLUSION Collectively, this study shows that a pro-apoptotic BCG strain alleviates adverse traits of the wild-type strain, resulting in a highly immunogenic TB vaccine.
Collapse
Affiliation(s)
- Alice Lau
- Division of Infectious Diseases, Department of Medicine and Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Vijender Singh
- Division of Infectious Diseases, Department of Medicine and Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Hafid Soualhine
- Division of Infectious Diseases, Department of Medicine and Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Zakaria Hmama
- Division of Infectious Diseases, Department of Medicine and Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC V6H 3Z6, Canada.
| |
Collapse
|
27
|
Meng L, Tong J, Wang H, Tao C, Wang Q, Niu C, Zhang X, Gao Q. PPE38 Protein of Mycobacterium tuberculosis Inhibits Macrophage MHC Class I Expression and Dampens CD8 + T Cell Responses. Front Cell Infect Microbiol 2017; 7:68. [PMID: 28348981 PMCID: PMC5346565 DOI: 10.3389/fcimb.2017.00068] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 02/22/2017] [Indexed: 12/23/2022] Open
Abstract
Suppression of CD8+ T cell activation is a critical mechanism used by Mycobacterium tuberculosis (MTB) to escape protective host immune responses. PPE38 belongs to the unique PPE family of MTB and in our previous study, PPE38 protein was speculated to participate in manipulating macrophage MHC class I pathway. To test this hypothesis, the function of mycobacterial PPE38 protein was assessed here using macrophage and mouse infection models. Decreased amount of MHC class I was observed on the surface of macrophages infected with PPE38-expressing mycobacteria. The transcript of genes encoding MHC class I was also inhibited by PPE38. After infection of C57BL/6 mice with Mycobacterium smegmatis expressing PPE38 (Msmeg-PPE38), decreased number of CD8+ T cells was found in spleen, liver, and lungs through immunohistochemical analysis, comparing to the control strain harboring empty vector (Msmeg-V). Consistently, flow cytometry assay showed that fewer effector/memory CD8+ T cells (CD44highCD62Llow) were activated in spleen from Msmeg-PPE38 infected mice. Moreover, Msmeg-PPE38 confers a growth advantage over Msmeg-V in C57BL/6 mice, indicating an effect of PPE38 to favor mycobacterial persistence in vivo. Overall, this study shows a unique biological function of PPE38 protein to facilitate mycobacteria to escape host immunity, and provides hints for TB vaccine development.
Collapse
Affiliation(s)
- Lu Meng
- Key laboratory of Medical Molecular Virology, Institute of Biomedical Sciences and Institute of Medical Microbiology, Shanghai Medical College, Fudan University Shanghai, China
| | - Jingfeng Tong
- Key laboratory of Medical Molecular Virology, Institute of Biomedical Sciences and Institute of Medical Microbiology, Shanghai Medical College, Fudan University Shanghai, China
| | - Hui Wang
- Key laboratory of Medical Molecular Virology, Institute of Biomedical Sciences and Institute of Medical Microbiology, Shanghai Medical College, Fudan UniversityShanghai, China; The State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, School of Medicine, Shenzhen UniversityGuangdong, China
| | - Chengwu Tao
- Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences Shanghai, China
| | - Qinglan Wang
- Key laboratory of Medical Molecular Virology, Institute of Biomedical Sciences and Institute of Medical Microbiology, Shanghai Medical College, Fudan University Shanghai, China
| | - Chen Niu
- Key laboratory of Medical Molecular Virology, Institute of Biomedical Sciences and Institute of Medical Microbiology, Shanghai Medical College, Fudan University Shanghai, China
| | - Xiaoming Zhang
- Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences Shanghai, China
| | - Qian Gao
- Key laboratory of Medical Molecular Virology, Institute of Biomedical Sciences and Institute of Medical Microbiology, Shanghai Medical College, Fudan University Shanghai, China
| |
Collapse
|
28
|
Teng X, Tian M, Li J, Tan S, Yuan X, Yu Q, Jing Y, Zhang Z, Yue T, Zhou L, Fan X. Immunogenicity and protective efficacy of DMT liposome-adjuvanted tuberculosis subunit CTT3H vaccine. Hum Vaccin Immunother 2016; 11:1456-64. [PMID: 25905680 DOI: 10.1080/21645515.2015.1037057] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Different strategies have been proposed for the development of protein subunit vaccine candidates for tuberculosis (TB), which shows better safety than other types of candidates and the currently used Bacillus Calmette-Guérin (BCG) vaccine. In order to develop more effective protein subunits depending on the mechanism of cell-mediated immunity against TB, a polyprotein CTT3H, based on 5 immunodominant antigens (CFP10, TB10.4, TB8.4, Rv3615c, and HBHA) with CD8(+) epitopes of Mycobacterium tuberculosis, was constructed in this study. We vaccinated C57BL/6 mice with a TB subunit CTT3H protein in an adjuvant of dimethyldioctadecylammonium/monophosphoryl lipid A/trehalose 6,6'-dibehenate (DDA/MPL/TDB, DMT) liposome to investigate the immunogenicity and protective efficacy of this novel vaccine. Our results demonstrated that DMT liposome-adjuvanted CTT3H vaccine not only induced an antigen-specific CD4(+) Th1 response, but also raised the number of PPD- and CTT3H-specific IFN-γ(+) CD8(+) T cells and elicited strong CTL responses against TB10.4, which provided more effective protection against a 60 CFU M. tuberculosis aerosol challenge than PBS control and DMT adjuvant alone. Our findings indicate that DMT-liposome is an effective adjuvant to stimulate CD8(+) T cell responses and the DMT-adjuvanted subunit CTT3H vaccine is a promising candidate for the next generation of TB vaccine.
Collapse
Affiliation(s)
- Xindong Teng
- a Department of Pathogen Biology; School of Basic Medicine; Huazhong University of Science & Technology ; Wuhan , PR China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Booty MG, Nunes-Alves C, Carpenter SM, Jayaraman P, Behar SM. Multiple Inflammatory Cytokines Converge To Regulate CD8+ T Cell Expansion and Function during Tuberculosis. THE JOURNAL OF IMMUNOLOGY 2016; 196:1822-31. [PMID: 26755819 DOI: 10.4049/jimmunol.1502206] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 12/10/2015] [Indexed: 11/19/2022]
Abstract
The differentiation of effector CD8(+) T cells is a dynamically regulated process that varies during different infections and is influenced by the inflammatory milieu of the host. In this study, we define three signals regulating CD8(+) T cell responses during tuberculosis by focusing on cytokines known to affect disease outcome: IL-12, type I IFN, and IL-27. Using mixed bone marrow chimeras, we compared wild-type and cytokine receptor knockout CD8(+) T cells within the same mouse following aerosol infection with Mycobacterium tuberculosis. Four weeks postinfection, IL-12, type 1 IFN, and IL-27 were all required for efficient CD8(+) T cell expansion in the lungs. We next determined if these cytokines directly promote CD8(+) T cell priming or are required only for expansion in the lungs. Using retrogenic CD8(+) T cells specific for the M. tuberculosis Ag TB10.4 (EsxH), we observed that IL-12 is the dominant cytokine driving both CD8(+) T cell priming in the lymph node and expansion in the lungs; however, type I IFN and IL-27 have nonredundant roles supporting pulmonary CD8(+) T cell expansion. Thus, IL-12 is a major signal promoting priming in the lymph node, but a multitude of inflammatory signals converge in the lung to promote continued expansion. Furthermore, these cytokines regulate the differentiation and function of CD8(+) T cells during tuberculosis. These data demonstrate distinct and overlapping roles for each of the cytokines examined and underscore the complexity of CD8(+) T cell regulation during tuberculosis.
Collapse
Affiliation(s)
- Matthew G Booty
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01655; and Program in Immunology, Division of Medical Sciences, Harvard Medical School, Boston, MA 02115
| | - Cláudio Nunes-Alves
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01655; and
| | - Stephen M Carpenter
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01655; and
| | - Pushpa Jayaraman
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01655; and
| | - Samuel M Behar
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01655; and
| |
Collapse
|
30
|
Cliff JM, Cho JE, Lee JS, Ronacher K, King EC, van Helden P, Walzl G, Dockrell HM. Excessive Cytolytic Responses Predict Tuberculosis Relapse After Apparently Successful Treatment. J Infect Dis 2015; 213:485-95. [PMID: 26351358 PMCID: PMC4704670 DOI: 10.1093/infdis/jiv447] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 08/31/2015] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Currently, there are no tools to accurately predict tuberculosis relapse. This study aimed to determine whether patients who experience tuberculosis relapse have different immune responses to mycobacteria in vitro than patients who remain cured for 2 years. METHODS Patients with an initial episode of pulmonary tuberculosis were recruited in South Africa. Diluted blood, collected at diagnosis and after 2 and 4 weeks of treatment, was cultured with live Mycobacterium tuberculosis for 6 days, and cellular RNA was frozen. Gene expression in samples from 10 patients who subsequently experienced relapse, confirmed by strain genotyping, was compared to that in samples from patients who remained cured, using microarrays. RESULTS At diagnosis, expression of 668 genes was significantly different in samples from patients who experienced relapse, compared with expression in patients who remained successfully cured; these differences persisted for at least 4 weeks. Gene ontology and biological pathways analyses revealed significant upregulation of genes involved in cytotoxic cell-mediated killing. Results were confirmed by real-time quantitative reverse-transcription polymerase chain reaction analysis in a wider patient cohort. CONCLUSIONS These data show that patients who will subsequently experience relapse exhibit altered immune responses, including excessively robust cytolytic responses to M. tuberculosis in vitro, at the time of diagnosis, compared with patients who will achieve durable cure. Together with microbiological and clinical indices, these differences could be exploited in drug development.
Collapse
Affiliation(s)
- Jacqueline M Cliff
- TB Centre and Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, United Kingdom
| | - Jang-Eun Cho
- TB Centre and Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, United Kingdom
| | - Ji-Sook Lee
- TB Centre and Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, United Kingdom
| | - Katharina Ronacher
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences/MRC Centre for Molecular and Cellular Biology/DST NRF Centre of Excellence for Biomedical TB Research, Faculty of Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Elizabeth C King
- TB Centre and Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, United Kingdom
| | - Paul van Helden
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences/MRC Centre for Molecular and Cellular Biology/DST NRF Centre of Excellence for Biomedical TB Research, Faculty of Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Gerhard Walzl
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences/MRC Centre for Molecular and Cellular Biology/DST NRF Centre of Excellence for Biomedical TB Research, Faculty of Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Hazel M Dockrell
- TB Centre and Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, United Kingdom
| |
Collapse
|
31
|
Latent tuberculosis infection: myths, models, and molecular mechanisms. Microbiol Mol Biol Rev 2015; 78:343-71. [PMID: 25184558 DOI: 10.1128/mmbr.00010-14] [Citation(s) in RCA: 162] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The aim of this review is to present the current state of knowledge on human latent tuberculosis infection (LTBI) based on clinical studies and observations, as well as experimental in vitro and animal models. Several key terms are defined, including "latency," "persistence," "dormancy," and "antibiotic tolerance." Dogmas prevalent in the field are critically examined based on available clinical and experimental data, including the long-held beliefs that infection is either latent or active, that LTBI represents a small population of nonreplicating, "dormant" bacilli, and that caseous granulomas are the haven for LTBI. The role of host factors, such as CD4(+) and CD8(+) T cells, T regulatory cells, tumor necrosis factor alpha (TNF-α), and gamma interferon (IFN-γ), in controlling TB infection is discussed. We also highlight microbial regulatory and metabolic pathways implicated in bacillary growth restriction and antibiotic tolerance under various physiologically relevant conditions. Finally, we pose several clinically important questions, which remain unanswered and will serve to stimulate future research on LTBI.
Collapse
|
32
|
Orchestration of pulmonary T cell immunity during Mycobacterium tuberculosis infection: immunity interruptus. Semin Immunol 2014; 26:559-77. [PMID: 25311810 DOI: 10.1016/j.smim.2014.09.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 09/17/2014] [Accepted: 09/19/2014] [Indexed: 12/31/2022]
Abstract
Despite the introduction almost a century ago of Mycobacterium bovis BCG (BCG), an attenuated form of M. bovis that is used as a vaccine against Mycobacterium tuberculosis, tuberculosis remains a global health threat and kills more than 1.5 million people each year. This is mostly because BCG fails to prevent pulmonary disease--the contagious form of tuberculosis. Although there have been significant advances in understanding how the immune system responds to infection, the qualities that define protective immunity against M. tuberculosis remain poorly characterized. The ability to predict who will maintain control over the infection and who will succumb to clinical disease would revolutionize our approach to surveillance, control, and treatment. Here we review the current understanding of pulmonary T cell responses following M. tuberculosis infection. While infection elicits a strong immune response that contains infection, M. tuberculosis evades eradication. Traditionally, its intracellular lifestyle and alteration of macrophage function are viewed as the dominant mechanisms of evasion. Now we appreciate that chronic inflammation leads to T cell dysfunction. While this may arise as the host balances the goals of bacterial sterilization and avoidance of tissue damage, it is becoming clear that T cell dysfunction impairs host resistance. Defining the mechanisms that lead to T cell dysfunction is crucial as memory T cell responses are likely to be subject to the same subject to the same pressures. Thus, success of T cell based vaccines is predicated on memory T cells avoiding exhaustion while at the same time not promoting overt tissue damage.
Collapse
|
33
|
IL-10 and NOS2 modulate antigen-specific reactivity and nerve infiltration by T cells in experimental leprosy. PLoS Negl Trop Dis 2014; 8:e3149. [PMID: 25210773 PMCID: PMC4161319 DOI: 10.1371/journal.pntd.0003149] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 07/28/2014] [Indexed: 12/18/2022] Open
Abstract
Background Although immunopathology dictates clinical outcome in leprosy, the dynamics of early and chronic infection are poorly defined. In the tuberculoid region of the spectrum, Mycobacterium leprae growth is restricted yet a severe granulomatous lesion can occur. The evolution and maintenance of chronic inflammatory processes like those observed in the leprosy granuloma involve an ongoing network of communications via cytokines. IL-10 has immunosuppressive properties and IL-10 genetic variants have been associated with leprosy development and reactions. Methodology/Principal Findings The role of IL-10 in resistance and inflammation in leprosy was investigated using Mycobacterium leprae infection of mice deficient in IL-10 (IL-10−/−), as well as mice deficient in both inducible nitric oxide synthase (NOS2−/−) and IL-10 (10NOS2−/−). Although a lack of IL-10 did not affect M. leprae multiplication in the footpads (FP), inflammation increased from C57Bl/6 (B6)<IL-10−/−<NOS2−/−<10NOS2−/−. While IL-10−/− mice exhibited modest FP induration compared to B6, NOS2−/− and 10NOS2−/− mice developed markedly enlarged FP marking distinct phases: early (1 month), peak (3–4 months), and chronic (8 months). IFN-γ-producing CD4+CD44+ cells responding to M. leprae cell wall, membrane, and cytosol antigens and ML2028 (Ag85B) were significantly increased in the evolved granuloma in NOS2−/− FP compared to B6 and IL-10−/− during early and peak phases. In 10NOS2−/− FP, CD4+CD44+ and especially CD8+CD44+ responses were augmented even further to these antigens as well as to ML0380 (GroES), ML2038 (bacterioferritin), and ML1877 (EF-Tu). Moreover, fragmented nerves containing CD4+ cells were present in 10NOS2−/− FP. Conclusions/Significance The 10NOS2−/− strain offers insight on the regulation of granuloma formation and maintenance by immune modulators in the resistant forms of leprosy and presents a new model for investigating the pathogenesis of neurological involvement. Despite effective antimicrobial therapy, 30–50% of leprosy patients develop immunological complications called leprosy reactions before, during or even years after being cured. Leprosy reactions are a major risk for neuritis that leads to peripheral nerve damage, disfigurement and disability. Unfortunately, why and how leprosy reactions occur is not well understood. Based on the latest human genetic leprosy susceptibility research and mouse infection models, we generated a double knockout mouse strain (10NOS2−/−) which has deficiencies in two key immune factors, interleukin-10 (IL-10) and inducible nitric oxide synthase (NOS2). We investigated the dynamics of the immune response to Mycobacterium leprae infection and chronicled the types of immune cells recruited to the site of infection. 10NOS2−/− mice developed a substantial induration in response to infection, as well as an increased interferon-gamma response to components of the leprosy bacillus. Interestingly, these animals also exhibited CD4+ T cell infiltration into the nerves, a phenomenon which has not been previously reported in leprosy mouse models. This new model provides insight into potential mechanisms whereby immune modulators may regulate leprosy reactions and neuritis and could aid the development of tests for monitoring and treatment of leprosy patients.
Collapse
|
34
|
Kumar NP, Sridhar R, Hanna LE, Banurekha VV, Jawahar MS, Nutman TB, Babu S. Altered CD8(+) T cell frequency and function in tuberculous lymphadenitis. Tuberculosis (Edinb) 2014; 94:482-93. [PMID: 25027793 DOI: 10.1016/j.tube.2014.06.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 05/27/2014] [Accepted: 06/15/2014] [Indexed: 01/11/2023]
Abstract
CD8(+) T cells secreting Type1 and Type 17 cytokines and cytotoxic molecules play a major role in immunity and protection against pulmonary tuberculosis (PTB), although their role in tuberculous lymphadenitis (TBL) is not well known. To identify the distribution and function of CD8(+) T cells expressing Type1, Type 2 and Type 17 cytokines and cytotoxic molecules in TBL, we examined baseline and mycobacterial-antigen specific immune responses in the whole blood of individuals with PTB and compared them with TBL. TBL is characterized by elevated frequencies of baseline and mycobacterial-antigen stimulated CD8(+) T cells expressing Type 1 (IL-2 and TNFα) and Type 17 (IL-17A and IL-17F) cytokines in comparison to PTB individuals. In contrast, TBL individuals exhibited diminished frequency of CD8(+) T cells expressing perforin, granzyme B and CD107a. The blockade of IL-1R and IL-6R during antigenic stimulation resulted in significantly diminished frequencies of CD8(+) T cells expressing Type 1 and Type 17 cytokines in TBL. Therefore, our data suggest that TBL is characterized by an IL-1 and IL-6 dependent expansion of CD8(+) T cells expressing Type 1 and Type 17 cytokines as well as altered frequencies of cytotoxic molecules, reflecting an important association of these cells with the pathogenesis of TBL.
Collapse
Affiliation(s)
- Nathella Pavan Kumar
- National Institutes of Health-International Center for Excellence in Research, Chennai, India; National Institute for Research in Tuberculosis, Chennai, India.
| | | | | | | | | | - Thomas B Nutman
- Laboratory of Parasitic Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Subash Babu
- National Institutes of Health-International Center for Excellence in Research, Chennai, India; Laboratory of Parasitic Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
35
|
Hassan SS, Cho JE, Akram M, Fielding KL, Dockrell HM, Cliff JM. Modulation of NKG2D expression in human CD8(+) T cells corresponding with tuberculosis drug cure. PLoS One 2013; 8:e70063. [PMID: 23922903 PMCID: PMC3724721 DOI: 10.1371/journal.pone.0070063] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 06/18/2013] [Indexed: 11/18/2022] Open
Abstract
Background Biomarkers predicting tuberculosis treatment response and cure would facilitate drug development. This study investigated expression patterns of the co-stimulation molecule NKG2D in human tuberculosis and treatment to determine its potential usefulness as a host biomarker of tuberculosis drug efficacy. Methods Tuberculosis patients (n = 26) were recruited in Lahore, Pakistan, at diagnosis and followed up during treatment. Household contacts (n = 24) were also recruited. NKG2D expression was measured by qRT-PCR in RNA samples both ex vivo and following overnight mycobacterial stimulation in vitro. Protein expression of NKG2D and granzyme B was measured by flow cytometry. Results NKG2D expression in newly diagnosed tuberculosis patients was similar to household contacts in ex vivo RNA, but was higher following in vitro stimulation. The NKG2D expression was dramatically reduced by intensive phase chemotherapy, in both ex vivo blood RNA and CD8+ T cell protein expression, but then reverted to higher levels after the continuation phase in successfully treated patients. Conclusion The changes in NKG2D expression through successful treatment reflect modulation of the peripheral cytotoxic T cell response. This likely reflects firstly in vivo stimulation by live Mycobacterium tuberculosis, followed by the response to dead bacilli, antigen-release and finally immunopathology resolution. Such changes in host peripheral gene expression, alongside clinical and microbiological indices, could be developed into a biosignature of tuberculosis drug-induced cure to be used in future clinical trials.
Collapse
Affiliation(s)
- Syeda S. Hassan
- Department of Immunology and Infection, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Jang-Eun Cho
- Department of Immunology and Infection, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | | | - Katherine L. Fielding
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Hazel M. Dockrell
- Department of Immunology and Infection, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Jacqueline M. Cliff
- Department of Immunology and Infection, London School of Hygiene & Tropical Medicine, London, United Kingdom
- * E-mail:
| |
Collapse
|
36
|
Nagata T, Koide Y. Identification of T cell epitopes of Mycobacterium tuberculosis with biolistic DNA vaccination. Methods Mol Biol 2013; 940:285-303. [PMID: 23104350 DOI: 10.1007/978-1-62703-110-3_22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Tuberculosis (TB) has been listed as one of the most prevalent and serious infectious diseases worldwide. The etiological pathogen of TB is Mycobacterium tuberculosis (Mtb), a facultative intracellular bacterium. Mycobacterium bovis bacillus Calmette-Guérin (BCG) is the only approved vaccine against TB to date. BCG has been widely used, but the efficacy is questionable, especially in adult pulmonary TB. Therefore, more effective, safe and reliable TB vaccines have been urgently needed. T cell-mediated cellular immune response is a key immune response for effective protective immunity against TB. DNA vaccines using Mtb antigens have been studied as promising future TB vaccines. Most TB DNA vaccine studies so far reported used intramuscular or intradermal injection with needles, as these methods tend to induce a type 1 helper T lymphocyte (Th1)-type immune response that is critical for the protective immunity. We have been using DNA vaccines with gene gun bombardment for T cell epitope identification of various Mtb antigens. We show here our strategy to identify precise Mtb T cell epitopes using DNA vaccines with gene gun bombardment.
Collapse
Affiliation(s)
- Toshi Nagata
- Department of Health Science, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan.
| | | |
Collapse
|
37
|
Behar SM. Antigen-specific CD8(+) T cells and protective immunity to tuberculosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 783:141-63. [PMID: 23468108 DOI: 10.1007/978-1-4614-6111-1_8] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The continuing HIV/AIDS epidemic and the spread of multi-drug resistant Mycobacterium tuberculosis has led to the perpetuation of the worldwide tuberculosis epidemic. While M. bovis BCG is widely used as a vaccine, it lacks efficacy in preventing pulmonary tuberculosis in adults [1]. To combat this ongoing scourge, vaccine development for tuberculosis is a global priority. Most infected individuals develop long-lived protective immunity, which controls and contains M. tuberculosis in a T cell-dependent manner. An effective T cells response determines whether the infection resolves or develops into clinically evident disease. Consequently, there is great interest in determining which T cells subsets mediate anti-mycobacterial immunity, delineating their effector functions, and evaluating whether vaccination can elicit these T cells subsets and induce protective immunity. CD4(+) T cells are critical for resistance to M. tuberculosis in both humans and rodent models. CD4(+) T cells are required to control the initial infection as well as to prevent recrudescence in both humans and mice [2]. While it is generally accepted that class II MHC-restricted CD4(+) T cells are essential for immunity to tuberculosis, M. tuberculosis infection elicits CD8(+) T cells responses in both people and in experimental animals. CD8(+) T cells are also recruited to the lung during M. tuberculosis infection and are found in the granulomas of infected people. Thus, how CD8(+) T cells contribute to overall immunity to tuberculosis and whether antigens recognized by CD8(+) T cells would enhance the efficacy of vaccine strategies continue to be important questions.
Collapse
Affiliation(s)
- Samuel M Behar
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
38
|
Nikolova M, Markova R, Drenska R, Muhtarova M, Todorova Y, Dimitrov V, Taskov H, Saltini C, Amicosante M. Antigen-specific CD4- and CD8-positive signatures in different phases of Mycobacterium tuberculosis infection. Diagn Microbiol Infect Dis 2012; 75:277-81. [PMID: 23276770 DOI: 10.1016/j.diagmicrobio.2012.11.023] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Revised: 10/30/2012] [Accepted: 11/21/2012] [Indexed: 11/17/2022]
Abstract
Current diagnostic standards for Mycobacterium tuberculosis (MTB) infection do not distinguish between active and latent tuberculosis (TB). To identify specific biomarkers characterizing the different forms of TB infection, we investigated in parallel with the QuantiFERON -TB Gold In-Tube (QFT-IT) the use of flow cytometry measuring CD4 and CD8 MTB-specific immune response in 17 active-TB patients, 21 health care workers (HCW), 14 recent contacts of TB patients (RC-TB), and 10 bacille Calmette Guerin (BCG)-vaccinated healthy controls (BCG-HC). A correlation (r = 0.4526, P = 0.0002) was found only between the amount of IFN-γ measured by QFT-IT and the frequency of CD4+/CD69+/IFN-γ+ T cells. The frequency of CD4+/CD69+/IFNγ+ responding T cells was higher in active-TB patients (0.254 ± 0.336%, P < 0.01) compared to the other groups. The response of QFT-IT antigen-specific CD8+/CD69+/IFNγ+ T cells was significantly higher in RC-TB (0.245 ± 0.305%, P < 0.05) compared to the other study groups.
Collapse
Affiliation(s)
- Maria Nikolova
- Department of Immunology and Allergology, National Center of Infectious and Parasitic Diseases, 26 Yanko Sakazov, Sofia, Bulgaria
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Li W, Deng G, Li M, Liu X, Wang Y. Roles of Mucosal Immunity against Mycobacterium tuberculosis Infection. Tuberc Res Treat 2012; 2012:791728. [PMID: 23213508 PMCID: PMC3504404 DOI: 10.1155/2012/791728] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Revised: 09/12/2012] [Accepted: 09/27/2012] [Indexed: 12/21/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), is one of the world's leading infectious causes of morbidity and mortality. As a mucosal-transmitted pathogen, Mtb infects humans and animals mainly through the mucosal tissue of the respiratory tract. Apart from providing a physical barrier against the invasion of pathogen, the major function of the respiratory mucosa may be to serve as the inductive sites to initiate mucosal immune responses and sequentially provide the first line of defense for the host to defend against this pathogen. A large body of studies in the animals and humans have demonstrated that the mucosal immune system, rather than the systemic immune system, plays fundamental roles in the host's defense against Mtb infection. Therefore, the development of new vaccines and novel delivery routes capable of directly inducing respiratory mucosal immunity is emphasized for achieving enhanced protection from Mtb infection. In this paper, we outline the current state of knowledge regarding the mucosal immunity against Mtb infection, including the development of TB vaccines, and respiratory delivery routes to enhance mucosal immunity are discussed.
Collapse
Affiliation(s)
- Wu Li
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan, Ningxia 750021, China
- College of Life Science, Ningxia University, 539 W. Helanshan Road, Xixia District, Yinchuan, Ningxia 750021, China
| | - Guangcun Deng
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan, Ningxia 750021, China
- College of Life Science, Ningxia University, 539 W. Helanshan Road, Xixia District, Yinchuan, Ningxia 750021, China
| | - Min Li
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan, Ningxia 750021, China
- College of Life Science, Ningxia University, 539 W. Helanshan Road, Xixia District, Yinchuan, Ningxia 750021, China
| | - Xiaoming Liu
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan, Ningxia 750021, China
- College of Life Science, Ningxia University, 539 W. Helanshan Road, Xixia District, Yinchuan, Ningxia 750021, China
| | - Yujiong Wang
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan, Ningxia 750021, China
- College of Life Science, Ningxia University, 539 W. Helanshan Road, Xixia District, Yinchuan, Ningxia 750021, China
| |
Collapse
|
40
|
Rahman S, Magalhaes I, Rahman J, Ahmed RK, Sizemore DR, Scanga CA, Weichold F, Verreck F, Kondova I, Sadoff J, Thorstensson R, Spångberg M, Svensson M, Andersson J, Maeurer M, Brighenti S. Prime-boost vaccination with rBCG/rAd35 enhances CD8⁺ cytolytic T-cell responses in lesions from Mycobacterium tuberculosis-infected primates. Mol Med 2012; 18:647-58. [PMID: 22396020 DOI: 10.2119/molmed.2011.00222] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Accepted: 02/28/2012] [Indexed: 12/27/2022] Open
Abstract
To prevent the global spread of tuberculosis (TB) infection, a novel vaccine that triggers potent and long-lived immunity is urgently required. A plasmid-based vaccine has been developed to enhance activation of major histocompatibility complex (MHC) class I-restricted CD8⁺ cytolytic T cells using a recombinant Bacille Calmette-Guérin (rBCG) expressing a pore-forming toxin and the Mycobacterium tuberculosis (Mtb) antigens Ag85A, 85B and TB10.4 followed by a booster with a nonreplicating adenovirus 35 (rAd35) vaccine vector encoding the same Mtb antigens. Here, the capacity of the rBCG/rAd35 vaccine to induce protective and biologically relevant CD8⁺ T-cell responses in a nonhuman primate model of TB was investigated. After prime/boost immunizations and challenge with virulent Mtb in rhesus macaques, quantification of immune responses at the single-cell level in cryopreserved tissue specimen from infected organs was performed using in situ computerized image analysis as a technological platform. Significantly elevated levels of CD3⁺ and CD8⁺ T cells as well as cells expressing interleukin (IL)-7, perforin and granulysin were found in TB lung lesions and spleen from rBCG/rAd35-vaccinated animals compared with BCG/rAd35-vaccinated or unvaccinated animals. The local increase in CD8⁺ cytolytic T cells correlated with reduced expression of the Mtb antigen MPT64 and also with prolonged survival after the challenge. Our observations suggest that a protective immune response in rBCG/rAd35-vaccinated nonhuman primates was associated with enhanced MHC class I antigen presentation and activation of CD8⁺ effector T-cell responses at the local site of infection in Mtb-challenged animals.
Collapse
Affiliation(s)
- Sayma Rahman
- Center for Infectious Medicine, Department of Medicine, Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Singhal A, Mathys V, Kiass M, Creusy C, Delaire B, Aliouat EM, Dartois V, Kaplan G, Bifani P. BCG induces protection against Mycobacterium tuberculosis infection in the Wistar rat model. PLoS One 2011; 6:e28082. [PMID: 22162757 PMCID: PMC3230592 DOI: 10.1371/journal.pone.0028082] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Accepted: 10/31/2011] [Indexed: 11/25/2022] Open
Abstract
Our understanding of the correlation of Mycobacterium bovis Bacille Calmette-Guerin (BCG)-mediated immune responses and protection against Mycobacterium tuberculosis (Mtb) infection is still limited. We have recently characterized a Wistar rat model of experimental tuberculosis (TB). In the present study, we evaluated the efficacy of BCG vaccination in this model. Upon Mtb challenge, BCG vaccinated rats controlled growth of the bacilli earlier than unvaccinated rats. Histopathology analysis of infected lungs demonstrated a reduced number of granulomatous lesions and lower parenchymal inflammation in vaccinated animals. Vaccine-mediated protection correlated with the rapid accumulation of antigen specific CD4+ and CD8+ T cells in the infected lungs. Immunohistochemistry further revealed higher number of CD8+ cells in the pulmonary granulomas of vaccinated animals. Evaluation of pulmonary immune responses in vaccinated and Mtb infected rats by real time PCR at day 15 post-challenge showed reduced expression of genes responsible for negative regulation of Th1 immune responses. Thus, early protection observed in BCG vaccinated rats correlated with a similarly timed shift of immunity towards the Th1 type response. Our data support the importance of (i) the Th1-Th2 balance in the control of mycobacterial infection and (ii) the value of the Wistar rats in understanding the biology of TB.
Collapse
Affiliation(s)
- Amit Singhal
- Novartis Institute for Tropical Diseases, Singapore, Singapore
- Singapore Immunology Network, A*Star, Singapore, Singapore
- * E-mail: (AS); (PB)
| | - Vanessa Mathys
- Communicable and Infectious Diseases, Scientific Institute of Public Health, Brussels, Belgium
| | - Mehdi Kiass
- Communicable and Infectious Diseases, Scientific Institute of Public Health, Brussels, Belgium
| | - Colette Creusy
- Groupe Hospitalier de l'Institut Catholique Lillois, Hôpital Saint Vincent, Université Catholique de Lille, Lille, France
| | - Baptiste Delaire
- Groupe Hospitalier de l'Institut Catholique Lillois, Hôpital Saint Vincent, Université Catholique de Lille, Lille, France
| | - El Moukhtar Aliouat
- Department of Parasitology, Faculty of Biological and Pharmaceutical Sciences, University of Lille Nord de France, Lille, France
| | - Véronique Dartois
- Public Health Research Institute Center, University of Medicine and Dentistry of New Jersey, Newark, New Jersey, United States of America
| | - Gilla Kaplan
- Public Health Research Institute Center, University of Medicine and Dentistry of New Jersey, Newark, New Jersey, United States of America
| | - Pablo Bifani
- Novartis Institute for Tropical Diseases, Singapore, Singapore
- * E-mail: (AS); (PB)
| |
Collapse
|
42
|
Barnes PF, Samten B, Shams H, Vankayalapatib R. Progress in understanding the human immune responses to Mycobacterium tuberculosis. Tuberculosis (Edinb) 2011; 89 Suppl 1:S5-9. [PMID: 20006306 DOI: 10.1016/s1472-9792(09)70004-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Development of an effective vaccine against tuberculosis hinges on an improved understanding of the human immune response to Mycobacterium tuberculosis. Work in this area at the University of Texas Health Science Center at Tyler has led to advances in four areas: (1) natural killer cells contribute to innate immunity by lysing M. tuberculosis-infected mononuclear phagocytes, and to adaptive immunity by enhancing the CD8+ T-cell effector function and inhibiting expansion of T regulatory cells; (2) Interferon-gamma plays a central role in resistance to many intracellular pathogens, including M. tuberculosis, and we have identified three transcription factors that bind to the Interferon-gamma proximal promoter and increase Interferon-gamma transcription in live T-cells that are activated by M. tuberculosis antigens; (3) A DNA vaccine that encodes the M. tuberculosis 10fts;kDa culture filtrate protein and the lysosomal integral membrane protein-2 was produced to direct vaccine antigens to the MHC class II processing and presentation pathway. When this vaccine was coated with polyethylenimine and administered to mice, it yielded a remarkably potent pulmonary immune response that reduced the bacillary burden by 90% after M. tuberculosis challenge; (4) The early secreted antigenic target of 6fts;kDa (ESAT-6) is a putative vaccine antigen. We found that high concentrations of this antigen markedly inhibit Interferon-gamma production by T-cells and are working to understand the molecular mechanisms underlying this effect. Developing methods to enhance NK cell functions that favor protective immunity, increase interferon-gamma transcription, elicit protective pulmonary immune responses and prevent ESAT-6 from inhibiting T-cell function will contribute significantly to development of antituberculosis vaccines.
Collapse
Affiliation(s)
- Peter F Barnes
- Department of Medicine, The University of Texas Health Science Center at Tyler, Tyler, TX, USA.
| | | | | | | |
Collapse
|
43
|
Lin CW, Su IJ, Chang JR, Chen YY, Lu JJ, Dou HY. Recombinant BCG coexpressing Ag85B, CFP10, and interleukin-12 induces multifunctional Th1 and memory T cells in mice. APMIS 2011; 120:72-82. [PMID: 22151310 DOI: 10.1111/j.1600-0463.2011.02815.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Mycobacterium tuberculosis (MTB) continues to be a leading cause of human deaths due to an infectious agent. Current efforts are focused on making better TB vaccines. We describe the generation and immunological characterization of recombinant BCG (rBCG). This rBCG was generated by incorporating an expression plasmid encoding two mycobacterial antigens (Ag85B and CFP10) and human interleukin (IL)-12 into a BCG strain. Immunogenicity studies in mice showed that rBCG coexpressing Ag85B, CFP10, and IL-12 (rBCG::Ag85B-CFP10-IL-12) induces a robust immune response in mice. The rBCG vaccine promotes a T-cell response against MTB that is characterized by a high proportion of polyfunctional and memory T cells in spleen and lung. Our results showed strong immunogenicity and mycobacterial growth inhibition of rBCG::Ag85B-CFP10 plus IL-12 than that of BCG vaccine.
Collapse
Affiliation(s)
- Chih-Wei Lin
- Division of Infectious Diseases, National Health Research Institutes, Zhunan, Miaoli, Taiwan
| | | | | | | | | | | |
Collapse
|
44
|
T cell monitoring of chemotherapy in experimental rat tuberculosis. Antimicrob Agents Chemother 2011; 55:3677-83. [PMID: 21628535 DOI: 10.1128/aac.00136-11] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Mycobacterium tuberculosis is the causative agent of a pulmonary epidemic that is estimated to infect one-third of the world's population and that has an increased incidence of multidrug resistance. The evaluation of new chemical entities against M. tuberculosis is hampered by the lack of biological tools to help predict efficacy, from early drug development to clinical trials. As the rat is the animal species of choice in the pharmaceutical industry, we have developed a rat model of acute and chronic phases of M. tuberculosis infection for drug efficacy testing. In this model, we have evaluated the impact of tuberculosis drugs on T cell response using the enzyme-linked immunospot assay methodology. Infected rats treated with isoniazid (INH) or rifampin (RIF) responded to therapy, the potency of which was comparable to that seen in the mouse. Peripheral blood mononuclear cells from infected rats produced gamma interferon (IFN-γ) in response to RD-1 antigens, such as the 6-kDa early secretory antigen target (ESAT-6) and the 10-kDa culture filtrate protein (CFP-10). A decrease in IFN-γ spot-forming cells (SFCs) was consistently observed in response to drug treatment. In both the acute- and chronic-phase models, the T cell response was more sensitive to ESAT-6 than to CFP-10. The SFC count in response to ESAT-6 appears to be an indicator of bacterial killing in the rat. Collectively, our data suggest that the ESAT-6 response could be used as a potential surrogate of drug efficacy in the rat and that such a readout could help shorten drug testing during preclinical development.
Collapse
|
45
|
Kramp JC, McMurray DN, Formichella C, Jeevan A. The in vivo immunomodulatory effect of recombinant tumour necrosis factor-alpha in guinea pigs vaccinated with Mycobacterium bovis bacille Calmette-Guérin. Clin Exp Immunol 2011; 165:110-20. [PMID: 21545584 DOI: 10.1111/j.1365-2249.2011.04406.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Previous studies from our laboratory demonstrated that treatment in vitro with recombinant guinea pig tumour necrosis factor TNF (rgpTNF)-α-enhanced T cell and macrophage functions. Similarly, injection of Mycobacterium tuberculosis-infected guinea pigs with anti-TNF-α altered splenic granuloma organization and caused inflammatory changes and reduced the cell-associated mycobacteria in the tuberculous pluritis model. In this study, rgpTNF-α was injected into bacille Calmette-Guérin (BCG)-vaccinated guinea pigs to modulate immune functions in vivo. Guinea pigs were vaccinated intradermally with BCG, 2 × 10(3) colony-forming units (CFU) and injected intraperitoneally with either rgpTNF-α (25 µg/animal) or 1% bovine serum albumin (BSA) for a total of 12 injections given every other day. Treatment with rgpTNF-α significantly enhanced the skin test response to purified protein derivative (PPD), reduced the number of CFUs and increased the PPD-induced proliferation in the lymph nodes at 6 weeks after vaccination. The levels of interleukin (IL)-12 mRNA were increased in the lymph node and spleen cells stimulated with PPD. TNF-α treatment induced a decrease in TNF-α, IL-12p40 and IL-10 mRNA levels in peritoneal cells following PPD stimulation while live M. tuberculosis caused an increase in TNF-α mRNA and a decrease in the IL-10 mRNA expression. TNF-α injection also induced an increase in the infiltration of mononuclear cells and in the proportions of CD3(+) T cells in the lymph nodes. These results indicate that rgpTNF-α enhances some aspects of T cell immunity and promotes control of mycobacteria in the tissues. Future studies will address the role of TNF-α in BCG-vaccinated guinea pigs following low-dose pulmonary challenge with virulent M. tuberculosis.
Collapse
Affiliation(s)
- J C Kramp
- Department of Microbial and Molecular Pathogenesis, College of Medicine, Texas A&M Health Science Center, College of Medicine, College Station, TX, USA
| | | | | | | |
Collapse
|
46
|
Liu J, Zhang S, Tan S, Zheng B, Gao GF. Revival of the identification of cytotoxic T-lymphocyte epitopes for immunological diagnosis, therapy and vaccine development. Exp Biol Med (Maywood) 2011; 236:253-67. [PMID: 21330360 DOI: 10.1258/ebm.2010.010278] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Immunogenic T-cell epitopes have a central role in the cellular immunity against pathogens and tumors. However, in the early stage of cellular immunity studies, it was complicated and time-consuming to identify and characterize T-cell epitopes. Currently, the epitope screening is experiencing renewed enthusiasm due to advances in novel techniques and theories. Moreover, the application of T-cell epitope-based diagnoses for tuberculosis and new data on epitope-based vaccine development have also revived the field. There is a growing knowledge on the emphasis of epitope-stimulated T-cell immune responses in the elimination of pathogens and tumors. In this review, we outline the significance of the identification and characterization of T-cell epitopes. We also summarize the methods and strategies for epitope definition and, more importantly, address the relevance of cytotoxic T-lymphocyte epitopes to clinical diagnoses, therapy and vaccine development.
Collapse
Affiliation(s)
- Jun Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | | | | | | | | |
Collapse
|
47
|
Woodworth JS, Shin D, Volman M, Nunes-Alves C, Fortune SM, Behar SM. Mycobacterium tuberculosis directs immunofocusing of CD8+ T cell responses despite vaccination. THE JOURNAL OF IMMUNOLOGY 2010; 186:1627-37. [PMID: 21178003 DOI: 10.4049/jimmunol.1002911] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Vaccines that elicit T cell responses try to mimic protective memory T cell immunity after infection by increasing the frequency of Ag-specific T cells in the immune repertoire. However, the factors that determine immunodominance during infection and after vaccination and the relation between immunodominance and protection are incompletely understood. We previously identified TB10.4(20-28) as an immunodominant epitope recognized by H2-K(d)-restricted CD8(+) T cells after M. tuberculosis infection. Here we report a second epitope, EspA(150-158), that is recognized by a substantial number of pulmonary CD8(+) T cells. The relative abundance of these T cells in the naive repertoire only partially predicts their relative frequency after M. tuberculosis infection. Furthermore, although vaccination with recombinant vaccinia virus expressing these epitopes changes their relative immunodominance in the preinfection T cell repertoire, this change is transient after challenge with M. tuberculosis. We speculate that factors intrinsic to the chronic nature of M. tuberculosis infection establishes the hierarchy of immunodominance and may explain the failure of some vaccines to provide protection.
Collapse
Affiliation(s)
- Joshua S Woodworth
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | |
Collapse
|
48
|
Tang ST, van Meijgaarden KE, Caccamo N, Guggino G, Klein MR, van Weeren P, Kazi F, Stryhn A, Zaigler A, Sahin U, Buus S, Dieli F, Lund O, Ottenhoff THM. Genome-based in silico identification of new Mycobacterium tuberculosis antigens activating polyfunctional CD8+ T cells in human tuberculosis. THE JOURNAL OF IMMUNOLOGY 2010; 186:1068-80. [PMID: 21169544 DOI: 10.4049/jimmunol.1002212] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Although CD8(+) T cells help control Mycobacterium tuberculosis infection, their M. tuberculosis Ag repertoire, in vivo frequency, and functionality in human tuberculosis (TB) remains largely undefined. We have performed genome-based bioinformatics searches to identify new M. tuberculosis epitopes presented by major HLA class I supertypes A2, A3, and B7 (covering 80% of the human population). A total of 432 M. tuberculosis peptides predicted to bind to HLA-A*0201, HLA-A*0301, and HLA-B*0702 (representing the above supertypes) were synthesized and HLA-binding affinities determined. Peptide-specific CD8(+) T cell proliferation assays (CFSE dilution) in 41 M. tuberculosis-responsive donors identified 70 new M. tuberculosis epitopes. Using HLA/peptide tetramers for the 18 most prominently recognized HLA-A*0201-binding M. tuberculosis peptides, recognition by cured TB patients' CD8(+) T cells was validated for all 18 epitopes. Intracellular cytokine staining for IFN-γ, IL-2, and TNF-α revealed mono-, dual-, as well as triple-positive CD8(+) T cells, indicating these M. tuberculosis peptide-specific CD8(+) T cells were (poly)functional. Moreover, these T cells were primed during natural infection, because they were absent from M. tuberculosis-noninfected individuals. Control CMV peptide/HLA-A*0201 tetramers stained CD8(+) T cells in M. tuberculosis-infected and noninfected individuals equally, whereas Ebola peptide/HLA-A*0201 tetramers were negative. In conclusion, the M. tuberculosis-epitope/Ag repertoire for human CD8(+) T cells is much broader than hitherto suspected, and the newly identified M. tuberculosis Ags are recognized by (poly)functional CD8(+) T cells during control of infection. These results impact on TB-vaccine design and biomarker identification.
Collapse
Affiliation(s)
- Sheila T Tang
- Center for Biological Sequence Analysis, Technical University of Denmark, 2800 Lyngby, Denmark
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Schwander S, Dheda K. Human lung immunity against Mycobacterium tuberculosis: insights into pathogenesis and protection. Am J Respir Crit Care Med 2010; 183:696-707. [PMID: 21075901 DOI: 10.1164/rccm.201006-0963pp] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The study of human pulmonary immunity against Mycobacterium tuberculosis (M.tb) provides a unique window into the biological interactions between the human host and M.tb within the broncho-alveolar microenvironment, the site of natural infection. Studies of bronchoalveolar cells (BACs) and lung tissue evaluate innate, adaptive, and regulatory immune mechanisms that collectively contribute to immunological protection or its failure. In aerogenically M.tb-exposed healthy persons lung immune responses reflect early host pathogen interactions that may contribute to sterilization, the development of latent M.tb infection, or progression to active disease. Studies in these persons may allow the identification of biomarkers of protective immunity before the initiation of inflammatory and disease-associated immunopathological changes. In healthy close contacts of patients with tuberculosis (TB) and during active pulmonary TB, immune responses are compartmentalized to the lungs and characterized by an exuberant helper T-cell type 1 response, which as suggested by recent evidence is counteracted by local suppressive immune mechanisms. Here we discuss how exploring human lung immunity may provide insights into disease progression and mechanisms of failure of immunological protection at the site of the initial host-pathogen interaction. These findings may also aid in the identification of new biomarkers of protective immunity that are urgently needed for the development of new and the improvement of current TB vaccines, adjuvant immunotherapies, and diagnostic technologies. To facilitate further work in this area, methodological and procedural approaches for bronchoalveolar lavage studies and their limitations are also discussed.
Collapse
Affiliation(s)
- Stephan Schwander
- Department of Environmental and Occupational Health, UMDNJ-School of Public Health, 683 Hoes Lane West, Room 305, Piscataway, NJ 08854, USA.
| | | |
Collapse
|
50
|
Zhang H, Wen K, Shen J, Geng S, Huang J, Pan Z, Jiao X. Characterization of immune responses following intranasal immunization with the Mycobacterium bovis CFP-10 protein expressed by attenuated Salmonella typhimurium. Scand J Immunol 2010; 72:277-83. [PMID: 20883312 DOI: 10.1111/j.1365-3083.2010.02421.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Culture filtrate protein 10 (CFP-10) from Mycobacterium bovis or Mycobacterium tuberculosis (MTB) is an immunodominant T-cell antigen expressed during the early stages of infection. Because lungs are most commonly associated with primary M. bovis infections, specific immunity at this site is desirable for protection. Therefore, in this study, immune responses generated in mouse lung, spleen and Peyer's patches were examined following intranasal (i.n.) immunization with Salmonella typhimurium- expressing CFP-10. Cells harvested from the lungs and Peyer's patches of immunized mice and then stimulated with CFP-10 produced significant levels of IFN-γ and these mice developed elevated serum IgG and lung IgA anti-CFP-10 responses, suggesting that this approach induced potent anti-CFP-10 mucosal immunity. Our study demonstrates that i.n. administration of CFP-10 expressed by S. typhimurium represents an effective way to induce efficient immune response to M. bovis antigen.
Collapse
Affiliation(s)
- H Zhang
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China
| | | | | | | | | | | | | |
Collapse
|