1
|
Nie X, Ma B, Yuan X, Li M, Liu Y, Hou Y, Yang Y, Xu J, Wang Y. Endoplasmic Reticulum Stress Mediated NLRP3 Inflammasome Activation and Pyroptosis in THP-1 Macrophages Infected with Bacillus Calmette-Guérin. Int J Mol Sci 2023; 24:11692. [PMID: 37511451 PMCID: PMC10380357 DOI: 10.3390/ijms241411692] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/11/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
Tuberculosis (TB) is a zoonotic infectious disease caused by Mycobacterium tuberculosis (Mtb). Mtb is a typical intracellular parasite, and macrophages are its main host cells. NLRP3 inflammasome-mediated pyroptosis is a form of programmed cell death implicated in the clearance of pathogenic infections. The bidirectional regulatory effect of endoplasmic reticulum stress (ERS) plays a crucial role in determining cell survival and death. Whether ERS is involved in macrophage pyroptosis with Mtb infection remains unclear. This article aims to explore the regulation of the NLRP3 inflammasome and pyroptosis by ERS in THP-1 macrophages infected with Mycobacterium bovis Bacillus Calmette-Guérin (BCG). The results showed that BCG infection induced THP-1 macrophage ERS, NLRP3 inflammasome activation and pyroptosis, which was inhibited by ERS inhibitor TUDCA. NLRP3 inhibitor MCC950 inhibited THP-1 macrophage NLRP3 inflammasome activation and pyroptosis caused by BCG infection. Compared with specific Caspase-1 inhibitor VX-765, pan-Caspase inhibitor Z-VAD-FMK showed a more significant inhibitory effect on BCG infection-induced pyroptosis of THP-1 macrophages. Taken together, this study demonstrates that ERS mediated NLRP3 inflammasome activation and pyroptosis after BCG infection of THP-1 macrophages, and that BCG infection of THP-1 macrophages induces pyroptosis through canonical and noncanonical pathways.
Collapse
Affiliation(s)
- Xueyi Nie
- School of Life Sciences, Ningxia University, Yinchuan 750021, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan 750021, China
| | - Boli Ma
- School of Life Sciences, Ningxia University, Yinchuan 750021, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan 750021, China
| | - Xiaotan Yuan
- School of Life Sciences, Ningxia University, Yinchuan 750021, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan 750021, China
| | - Mengyuan Li
- School of Life Sciences, Ningxia University, Yinchuan 750021, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan 750021, China
| | - Yueyang Liu
- School of Life Sciences, Ningxia University, Yinchuan 750021, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan 750021, China
| | - Yuxin Hou
- School of Life Sciences, Ningxia University, Yinchuan 750021, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan 750021, China
| | - Yi Yang
- School of Life Sciences, Ningxia University, Yinchuan 750021, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan 750021, China
| | - Jinrui Xu
- School of Life Sciences, Ningxia University, Yinchuan 750021, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan 750021, China
| | - Yujiong Wang
- School of Life Sciences, Ningxia University, Yinchuan 750021, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan 750021, China
| |
Collapse
|
2
|
Evaluation of early innate and adaptive immune responses to the TB vaccine Mycobacterium bovis BCG and vaccine candidate BCGΔBCG1419c. Sci Rep 2022; 12:12377. [PMID: 35858977 PMCID: PMC9300728 DOI: 10.1038/s41598-022-14935-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 05/03/2022] [Indexed: 12/30/2022] Open
Abstract
The vaccine Mycobacterium bovis Bacillus Calmette-Guérin (BCG) elicits an immune response that is protective against certain forms of tuberculosis (TB); however, because BCG efficacy is limited it is important to identify alternative TB vaccine candidates. Recently, the BCG deletion mutant and vaccine candidate BCGΔBCG1419c was demonstrated to survive longer in intravenously infected BALB/c mice due to enhanced biofilm formation, and better protected both BALB/c and C57BL/6 mice against TB-induced lung pathology during chronic stages of infection, relative to BCG controls. BCGΔBCG1419c-elicited protection also associated with lower levels of proinflammatory cytokines (i.e. IL6, TNFα) at the site of infection in C57BL/6 mice. Given the distinct immune profiles of BCG- and BCGΔBCG1419c-immunized mice during chronic TB, we set out to determine if there are early immunological events which distinguish these two groups, using multi-dimensional flow cytometric analysis of the lungs and other tissues soon after immunization. Our results demonstrate a number of innate and adaptive response differences between BCG- and BCGΔBCG1419c-immunized mice which are consistent with the latter being longer lasting and potentially less inflammatory, including lower frequencies of exhausted CD4+ T helper (TH) cells and higher frequencies of IL10-producing T cells, respectively. These studies suggest the use of BCGΔBCG1419c may be advantageous as an alternative TB vaccine candidate.
Collapse
|
3
|
Gairola A, Benjamin A, Weatherston JD, Cirillo JD, Wu HJ. Recent Developments in Drug Delivery for Treatment of Tuberculosis by Targeting Macrophages. ADVANCED THERAPEUTICS 2022; 5:2100193. [PMID: 36203881 PMCID: PMC9531895 DOI: 10.1002/adtp.202100193] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Indexed: 11/10/2022]
Abstract
Tuberculosis (TB) is among the greatest public health and safety concerns in the 21st century, Mycobacterium tuberculosis, which causes TB, infects alveolar macrophages and uses these cells as one of its primary sites of replication. The current TB treatment regimen, which consist of chemotherapy involving a combination of 3-4 antimicrobials for a duration of 6-12 months, is marked with significant side effects, toxicity, and poor compliance. Targeted drug delivery offers a strategy that could overcome many of the problems of current TB treatment by specifically targeting infected macrophages. Recent advances in nanotechnology and material science have opened an avenue to explore drug carriers that actively and passively target macrophages. This approach can increase the drug penetration into macrophages by using ligands on the nanocarrier that interact with specific receptors for macrophages. This review encompasses the recent development of drug carriers specifically targeting macrophages actively and passively. Future directions and challenges associated with development of effective TB treatment is also discussed.
Collapse
Affiliation(s)
- Anirudh Gairola
- Department of Chemical Engineering, Texas A&M University, College Station, Texas, USA
| | - Aaron Benjamin
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Joshua D Weatherston
- Department of Chemical Engineering, Texas A&M University, College Station, Texas, USA
| | - Jeffrey D Cirillo
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Hung-Jen Wu
- Department of Chemical Engineering, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
4
|
Enriquez AB, Izzo A, Miller SM, Stewart EL, Mahon RN, Frank DJ, Evans JT, Rengarajan J, Triccas JA. Advancing Adjuvants for Mycobacterium tuberculosis Therapeutics. Front Immunol 2021; 12:740117. [PMID: 34759923 PMCID: PMC8572789 DOI: 10.3389/fimmu.2021.740117] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 08/26/2021] [Indexed: 01/15/2023] Open
Abstract
Tuberculosis (TB) remains one of the leading causes of death worldwide due to a single infectious disease agent. BCG, the only licensed vaccine against TB, offers limited protection against pulmonary disease in children and adults. TB vaccine research has recently been reinvigorated by new data suggesting alternative administration of BCG induces protection and a subunit/adjuvant vaccine that provides close to 50% protection. These results demonstrate the need for generating adjuvants in order to develop the next generation of TB vaccines. However, development of TB-targeted adjuvants is lacking. To help meet this need, NIAID convened a workshop in 2020 titled “Advancing Vaccine Adjuvants for Mycobacterium tuberculosis Therapeutics”. In this review, we present the four areas identified in the workshop as necessary for advancing TB adjuvants: 1) correlates of protective immunity, 2) targeting specific immune cells, 3) immune evasion mechanisms, and 4) animal models. We will discuss each of these four areas in detail and summarize what is known and what we can advance on in order to help develop more efficacious TB vaccines.
Collapse
Affiliation(s)
- Ana B Enriquez
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States.,Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Angelo Izzo
- Tuberculosis Research Program, Centenary Institute, The University of Sydney, Sydney, NSW, Australia
| | - Shannon M Miller
- Center for Translational Medicine, University of Montana, Missoula, MT, United States.,Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT, United States
| | - Erica L Stewart
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.,Sydney Institute for Infectious Diseases and Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Robert N Mahon
- Division of AIDS, Columbus Technologies & Services Inc., Contractor to National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Daniel J Frank
- Division of AIDS, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, United States
| | - Jay T Evans
- Center for Translational Medicine, University of Montana, Missoula, MT, United States.,Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT, United States
| | - Jyothi Rengarajan
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States.,Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States.,Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, United States
| | - James A Triccas
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.,Sydney Institute for Infectious Diseases and Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
5
|
Pu W, Zhao C, Wazir J, Su Z, Niu M, Song S, Wei L, Li L, Zhang X, Shi X, Wang H. Comparative transcriptomic analysis of THP-1-derived macrophages infected with Mycobacterium tuberculosis H37Rv, H37Ra and BCG. J Cell Mol Med 2021; 25:10504-10520. [PMID: 34632719 PMCID: PMC8581329 DOI: 10.1111/jcmm.16980] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 08/31/2021] [Accepted: 09/08/2021] [Indexed: 12/12/2022] Open
Abstract
Tuberculosis (TB) remains a worldwide healthcare concern, and the exploration of the host‐pathogen interaction is essential to develop therapeutic modalities and strategies to control Mycobacterium tuberculosis (M.tb). In this study, RNA sequencing (transcriptome sequencing) was employed to investigate the global transcriptome changes in the macrophages during the different strains of M.tb infection. THP‐1 cells derived from macrophages were exposed to the virulent M.tb strain H37Rv (Rv) or the avirulent M.tb strain H37Ra (Ra), and the M.tb BCG vaccine strain was used as a control. The cDNA libraries were prepared from M.tb‐infected macrophages and then sequenced. To assess the transcriptional differences between the expressed genes, the bioinformatics analysis was performed using a standard pipeline of quality control, reference mapping, differential expression analysis, protein‐protein interaction (PPI) networks, gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. Q‐PCR and Western blot assays were also performed to validate the data. Our findings indicated that, when compared to BCG or M.tb H37Ra infection, the transcriptome analysis identified 66 differentially expressed genes in the M.tb H37Rv‐infected macrophages, out of which 36 genes were up‐regulated, and 30 genes were down‐regulated. The up‐regulated genes were associated with immune response regulation, chemokine secretion, and leucocyte chemotaxis. In contrast, the down‐regulated genes were associated with amino acid biosynthetic and energy metabolism, connective tissue development and extracellular matrix organization. The Q‐PCR and Western blot assays confirmed increased expression of pro‐inflammatory factors, altered energy metabolic processes, enhanced activation of pro‐inflammatory signalling pathways and increased pyroptosis in H37Rv‐infected macrophage. Overall, our RNA sequencing‐based transcriptome study successfully identified a comprehensive, in‐depth gene expression/regulation profile in M.tb‐infected macrophages. The results demonstrated that virulent M.tb strain H37Rv infection triggers a more severe inflammatory immune response associated with increased tissue damage, which helps in understanding the host‐pathogen interaction dynamics and pathogenesis features in different strains of M.tb infection.
Collapse
Affiliation(s)
- Wenyuan Pu
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School of Nanjing University, Nanjing, China.,Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Chen Zhao
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School of Nanjing University, Nanjing, China.,Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Junaid Wazir
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School of Nanjing University, Nanjing, China.,Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Zhonglan Su
- Department of Dermatology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Mengyuan Niu
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School of Nanjing University, Nanjing, China.,Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Shiyu Song
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School of Nanjing University, Nanjing, China.,Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Lulu Wei
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School of Nanjing University, Nanjing, China.,Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Li Li
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School of Nanjing University, Nanjing, China.,Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Xia Zhang
- Nanjing Public Health Clinical Center, the Second hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xudong Shi
- Nanjing Public Health Clinical Center, the Second hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hongwei Wang
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School of Nanjing University, Nanjing, China.,Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
6
|
Foreman HCC, Frank A, Stedman TT. Determination of variable region sequences from hybridoma immunoglobulins that target Mycobacterium tuberculosis virulence factors. PLoS One 2021; 16:e0256079. [PMID: 34415957 PMCID: PMC8378720 DOI: 10.1371/journal.pone.0256079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 07/29/2021] [Indexed: 11/19/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) infects one-quarter of the world's population. Mtb and HIV coinfections enhance the comorbidity of tuberculosis (TB) and AIDS, accounting for one-third of all AIDS-associated mortalities. Humoral antibody to Mtb correlates with TB susceptibility, and engineering of Mtb antibodies may lead to new diagnostics and therapeutics. The characterization and validation of functional immunoglobulin (Ig) variable chain (IgV) sequences provide a necessary first step towards developing therapeutic antibodies against pathogens. The virulence-associated Mtb antigens SodA (Superoxide Dismutase), KatG (Catalase), PhoS1/PstS1 (regulatory factor), and GroES (heat shock protein) are potential therapeutic targets but lacked IgV sequence characterization. Putative IgV sequences were identified from the mRNA of hybridomas targeting these antigens and isotype-switched into a common immunoglobulin fragment crystallizable region (Fc region) backbone, subclass IgG2aκ. Antibodies were validated by demonstrating recombinant Ig assembly and secretion, followed by the determination of antigen-binding specificity using ELISA and immunoblot assay.
Collapse
Affiliation(s)
- Hui-Chen Chang Foreman
- BEI Resources, ATCC., Manassas, Virginia, United States of America
- * E-mail: (HCCF); (TTS)
| | - Andrew Frank
- BEI Resources, ATCC., Manassas, Virginia, United States of America
| | - Timothy T. Stedman
- BEI Resources, ATCC., Manassas, Virginia, United States of America
- * E-mail: (HCCF); (TTS)
| |
Collapse
|
7
|
Chen T, Blanc C, Liu Y, Ishida E, Singer S, Xu J, Joe M, Jenny-Avital ER, Chan J, Lowary TL, Achkar JM. Capsular glycan recognition provides antibody-mediated immunity against tuberculosis. J Clin Invest 2020; 130:1808-1822. [PMID: 31935198 DOI: 10.1172/jci128459] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 01/03/2020] [Indexed: 12/16/2022] Open
Abstract
A better understanding of all immune components involved in protecting against Mycobacterium tuberculosis infection is urgently needed to inform strategies for novel immunotherapy and tuberculosis (TB) vaccine development. Although cell-mediated immunity is critical, increasing evidence supports that antibodies also have a protective role against TB. Yet knowledge of protective antigens is limited. Analyzing sera from 97 US immigrants at various stages of M. tuberculosis infection, we showed protective in vitro and in vivo efficacy of polyclonal IgG against the M. tuberculosis capsular polysaccharide arabinomannan (AM). Using recently developed glycan arrays, we established that anti-AM IgG induced in natural infection is highly heterogeneous in its binding specificity and differs in both its reactivity to oligosaccharide motifs within AM and its functions in bacillus Calmette-Guérin vaccination and/or in controlled (latent) versus uncontrolled (TB) M. tuberculosis infection. We showed that anti-AM IgG from asymptomatic but not from diseased individuals was protective and provided data suggesting a potential role of IgG2 and specific AM oligosaccharides. Filling a gap in the current knowledge of protective antigens in humans, our data support the key role of the M. tuberculosis surface glycan AM and suggest the importance of targeting specific glycan epitopes within AM in antibody-mediated immunity against TB.
Collapse
Affiliation(s)
- Tingting Chen
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Caroline Blanc
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Yanyan Liu
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Elise Ishida
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Sarah Singer
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Jiayong Xu
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Maju Joe
- Alberta Glycomics Centre and Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | | | - John Chan
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA.,Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Todd L Lowary
- Alberta Glycomics Centre and Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Jacqueline M Achkar
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA.,Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
8
|
Ramos L, Lunney JK, Gonzalez-Juarrero M. Neonatal and infant immunity for tuberculosis vaccine development: importance of age-matched animal models. Dis Model Mech 2020; 13:dmm045740. [PMID: 32988990 PMCID: PMC7520460 DOI: 10.1242/dmm.045740] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Neonatal and infant immunity differs from that of adults in both the innate and adaptive arms, which are critical contributors to immune-mediated clearance of infection and memory responses elicited during vaccination. The tuberculosis (TB) research community has openly admitted to a vacuum of knowledge about neonatal and infant immune responses to Mycobacterium tuberculosis (Mtb) infection, especially in the functional and phenotypic attributes of memory T cell responses elicited by the only available vaccine for TB, the Bacillus Calmette-Guérin (BCG) vaccine. Although BCG vaccination has variable efficacy in preventing pulmonary TB during adolescence and adulthood, 80% of endemic TB countries still administer BCG at birth because it has a good safety profile and protects children from severe forms of TB. As such, new vaccines must work in conjunction with BCG at birth and, thus, it is essential to understand how BCG shapes the immune system during the first months of life. However, many aspects of the neonatal and infant immune response elicited by vaccination with BCG remain unknown, as only a handful of studies have followed BCG responses in infants. Furthermore, most animal models currently used to study TB vaccine candidates rely on adult-aged animals. This presents unique challenges when transitioning to human trials in neonates or infants. In this Review, we focus on vaccine development in the field of TB and compare the relative utility of animal models used thus far to study neonatal and infant immunity. We encourage the development of neonatal animal models for TB, especially the use of pigs.
Collapse
Affiliation(s)
- Laylaa Ramos
- Mycobacteria Research Laboratories, Microbiology Immunology and Pathology Department, Colorado State University, 1682 Campus Delivery, Fort Collins, CO 80523, USA
| | - Joan K Lunney
- Animal Parasitic Diseases Laboratory, BARC, NEA, ARS, USDA Building 1040, Room 103, Beltsville, MD 20705, USA
| | - Mercedes Gonzalez-Juarrero
- Mycobacteria Research Laboratories, Microbiology Immunology and Pathology Department, Colorado State University, 1682 Campus Delivery, Fort Collins, CO 80523, USA
| |
Collapse
|
9
|
Bickett TE, McLean J, Creissen E, Izzo L, Hagan C, Izzo AJ, Silva Angulo F, Izzo AA. Characterizing the BCG Induced Macrophage and Neutrophil Mechanisms for Defense Against Mycobacterium tuberculosis. Front Immunol 2020; 11:1202. [PMID: 32625209 PMCID: PMC7314953 DOI: 10.3389/fimmu.2020.01202] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Accepted: 05/14/2020] [Indexed: 12/16/2022] Open
Abstract
The live attenuated Mycobacterium bovis strain, Bacille Calmette Guérin (BCG) is a potent innate immune stimulator. In the C57BL/6 mouse model of tuberculosis, BCG vaccination leads to a significant reduction of Mycobacterium tuberculosis burden after aerogenic infection. Our studies indicated that BCG induced protection against pulmonary tuberculosis was independent of T cells and present as early as 7 days after vaccination. This protection showed longevity, as it did not wane when conventional T cell and TNF-α deficient mice were infected 30 days post-vaccination. As BCG induced mycobacterial killing after 7 days, this study investigated the contributions of the innate immune system after BCG vaccination to better understand mechanisms required for mycobacterial killing. Subcutaneous BCG inoculation resulted in significant CD11b+F4/80+ monocyte subset recruitment into the lungs within 7 days. Further studies revealed that killing of mycobacteria was dependent on the viability of BCG, because irradiated BCG did not have the same effect. Although others have identified BCG as a facilitator of trained innate immunity, we found that BCG reduced the mycobacterial burden in the absence of mechanisms required for trained innate immunity, highlighting a role for macrophages and neutrophils for vaccine induced killing of M. tuberculosis.
Collapse
Affiliation(s)
- Thomas E Bickett
- Department of Microbiology Immunology and Pathology, Cell and Molecular Biology, Colorado State University, Fort Collins, CO, United States
| | - Jennifer McLean
- Department of Microbiology Immunology and Pathology, Cell and Molecular Biology, Colorado State University, Fort Collins, CO, United States
| | - Elizabeth Creissen
- Department of Microbiology Immunology and Pathology, Cell and Molecular Biology, Colorado State University, Fort Collins, CO, United States
| | - Linda Izzo
- Department of Microbiology Immunology and Pathology, Cell and Molecular Biology, Colorado State University, Fort Collins, CO, United States
| | - Cassidy Hagan
- Department of Microbiology Immunology and Pathology, Cell and Molecular Biology, Colorado State University, Fort Collins, CO, United States
| | - Antonio J Izzo
- Department of Microbiology Immunology and Pathology, Cell and Molecular Biology, Colorado State University, Fort Collins, CO, United States
| | - Fabiola Silva Angulo
- Department of Microbiology Immunology and Pathology, Cell and Molecular Biology, Colorado State University, Fort Collins, CO, United States
| | - Angelo A Izzo
- Department of Microbiology Immunology and Pathology, Cell and Molecular Biology, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|
10
|
Liu S, Xie Y, Luo W, Dou Y, Xiong H, Xiao Z, Zhang XL. PE_PGRS31-S100A9 Interaction Promotes Mycobacterial Survival in Macrophages Through the Regulation of NF-κB-TNF-α Signaling and Arachidonic Acid Metabolism. Front Microbiol 2020; 11:845. [PMID: 32457723 PMCID: PMC7225313 DOI: 10.3389/fmicb.2020.00845] [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: 01/03/2020] [Accepted: 04/08/2020] [Indexed: 12/17/2022] Open
Abstract
Mycobacterium tuberculosis (M. tb) evades the surveillance of immune responses for survival in macrophages. However, the precise mechanism and toxins/proteins encoded by M. tb involved in the bacterial escape remain elusive. The function of Rv1768 protein (also referred to as PE_PGRS31, belonging to the PE_PGRS family) encoded by the region of deletion 14 (RD-14) in the virulent M. tb H37Rv strain has not, to the best of our knowledge, been reported previously. Here, we found that Rv1768 remarkably promotes bacterial survival in macrophages. Compared to wild type (WT) H37Rv, the Rv1768 deficient strain (H37RvΔ1768) showed significantly decreased colony-forming units in the lungs, spleen, and liver of the murine M. tb infection model. The bacterial burdens of WT H37Rv in WT macrophages and C57BL/6 mice were significantly higher than those in S100A9 deficiency cells and mice, but there were no significant differences for H37RvΔRv1768. Rv1768 binds S100A9 with the proline-glutamic acid domain (PE domain) and blocks the interaction between S100A9 and Toll-like receptor 4 (TLR4), and suppresses TLR4-myeloid differentiation factor 88-nuclear factor-kappa B (NF-κB)-tumor necrosis factor α (TNF-α) signaling in macrophages. Interestingly, Rv1768 binding to S100A9 also disturbs the metabolism of arachidonic acid by activating 5-lipoxygenase, increasing lipotoxin A4, and down-regulating cyclooxygenase-2 and prostaglandin E2 expression, thus, promoting mycobacterial survival. Our results revealed that M. tb Rv1768 promotes mycobacterial survival in macrophages by regulating NF-κB-TNF-α signaling and arachidonic acid metabolism via S100A9. Disturbing the interaction between Rv1768 and S100A9 may be a potential therapeutic target for tuberculosis.
Collapse
Affiliation(s)
- Sheng Liu
- Hubei Province Key Laboratory of Allergy and Immunology, Department of Immunology, Wuhan University School of Basic Medical Sciences and Department of Allergy, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Yan Xie
- Hubei Province Key Laboratory of Allergy and Immunology, Department of Immunology, Wuhan University School of Basic Medical Sciences and Department of Allergy, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Wei Luo
- Hubei Province Key Laboratory of Allergy and Immunology, Department of Immunology, Wuhan University School of Basic Medical Sciences and Department of Allergy, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Yafeng Dou
- Hubei Province Key Laboratory of Allergy and Immunology, Department of Immunology, Wuhan University School of Basic Medical Sciences and Department of Allergy, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Huan Xiong
- Hubei Province Key Laboratory of Allergy and Immunology, Department of Immunology, Wuhan University School of Basic Medical Sciences and Department of Allergy, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Zhen Xiao
- Hubei Province Key Laboratory of Allergy and Immunology, Department of Immunology, Wuhan University School of Basic Medical Sciences and Department of Allergy, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Xiao-Lian Zhang
- Hubei Province Key Laboratory of Allergy and Immunology, Department of Immunology, Wuhan University School of Basic Medical Sciences and Department of Allergy, Zhongnan Hospital, Wuhan University, Wuhan, China.,State Key Laboratory of Virology, Frontier Science Center for Immunology and Metabolism, Wuhan University School of Medicine, Wuhan, China
| |
Collapse
|
11
|
Angelidou A, Diray-Arce J, Conti MG, Smolen KK, van Haren SD, Dowling DJ, Husson RN, Levy O. BCG as a Case Study for Precision Vaccine Development: Lessons From Vaccine Heterogeneity, Trained Immunity, and Immune Ontogeny. Front Microbiol 2020; 11:332. [PMID: 32218774 PMCID: PMC7078104 DOI: 10.3389/fmicb.2020.00332] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 02/14/2020] [Indexed: 12/11/2022] Open
Abstract
Vaccines have been traditionally developed with the presumption that they exert identical immunogenicity regardless of target population and that they provide protection solely against their target pathogen. However, it is increasingly appreciated that vaccines can have off-target effects and that vaccine immunogenicity can vary substantially with demographic factors such as age and sex. Bacille Calmette-Guérin (BCG), the live attenuated Mycobacterium bovis vaccine against tuberculosis (TB), represents a key example of these concepts. BCG vaccines are manufactured under different conditions across the globe generating divergent formulations. Epidemiologic studies have linked early life immunization with certain BCG formulations to an unanticipated reduction (∼50%) in all-cause mortality, especially in low birthweight males, greatly exceeding that attributable to TB prevention. This mortality benefit has been related to prevention of sepsis and respiratory infections suggesting that BCG induces "heterologous" protection against unrelated pathogens. Proposed mechanisms for heterologous protection include vaccine-induced immunometabolic shifts, epigenetic reprogramming of innate cell populations, and modulation of hematopoietic stem cell progenitors resulting in altered responses to subsequent stimuli, a phenomenon termed "trained immunity." In addition to genetic differences, licensed BCG formulations differ markedly in content of viable mycobacteria key for innate immune activation, potentially contributing to differences in the ability of these diverse formulations to induce TB-specific and heterologous protection. BCG immunomodulatory properties have also sparked interest in its potential use to prevent or alleviate autoimmune and inflammatory diseases, including type 1 diabetes mellitus and multiple sclerosis. BCG can also serve as a model: nanoparticle vaccine formulations incorporating Toll-like receptor 8 agonists can mimic some of BCG's innate immune activation, suggesting that aspects of BCG's effects can be induced with non-replicating stimuli. Overall, BCG represents a paradigm for precision vaccinology, lessons from which will help inform next generation vaccines.
Collapse
Affiliation(s)
- Asimenia Angelidou
- Division of Newborn Medicine, Boston Children’s Hospital and Beth Israel Deaconess Medical Center, Boston, MA, United States
- Precision Vaccines Program, Boston Children’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Joann Diray-Arce
- Precision Vaccines Program, Boston Children’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
| | - Maria Giulia Conti
- Precision Vaccines Program, Boston Children’s Hospital, Boston, MA, United States
- Department of Maternal and Child Health, Sapienza University of Rome, Rome, Italy
| | - Kinga K. Smolen
- Precision Vaccines Program, Boston Children’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
| | - Simon Daniël van Haren
- Precision Vaccines Program, Boston Children’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
| | - David J. Dowling
- Precision Vaccines Program, Boston Children’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
| | - Robert N. Husson
- Harvard Medical School, Boston, MA, United States
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
| | - Ofer Levy
- Precision Vaccines Program, Boston Children’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
| |
Collapse
|
12
|
Ong E, He Y, Yang Z. Epitope promiscuity and population coverage of Mycobacterium tuberculosis protein antigens in current subunit vaccines under development. INFECTION GENETICS AND EVOLUTION 2020; 80:104186. [PMID: 31923726 DOI: 10.1016/j.meegid.2020.104186] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 01/05/2020] [Accepted: 01/07/2020] [Indexed: 02/06/2023]
Abstract
Tuberculosis (TB) is the leading infectious cause of death worldwide and claimed over 1.6 million lives in 2017. Furthermore, one-third of the world population is estimated to be latently infected with Mycobacterium tuberculosis (MTB). A safe and effective MTB vaccine that can prevent both the primary infection and the reactivation of latent tuberculosis infection (LTBI), and that can protect against all forms of TB in adults and adolescents is urgently needed. In this study, using computational approaches, we predicted the capacity of the epitopes to be presented by the HLA molecules for ten MTB protein antigens (Mtb39a, Mtb32a, Ag85B, ESAT-6, TB10.4, Rv2660, Rv2608, Rv3619, Rv3620, and Rv1813) constituting five MTB subunit vaccines (M72, H1, H4, H56, and ID93) that are currently in clinical trials. We also assessed the promiscuity of the predicted epitopes based on a reference set of alleles and supertype alleles, and estimated the population coverage of the ten antigens in three high TB burden countries (China, India, and South Africa). Among the ten antigens evaluated, Rv2608 was found to have the highest number of promiscuous epitopes predicted to bind the most MHC-I and MHC-II supertype alleles, highest predicted immunogenicity, and the broadest population coverage in three high burden countries. Between the two latency-related antigens (Rv1813 and Rv2660), Rv1813 was predicted to have a better epitope diversity and promiscuity, immunogenicity, and population coverage. As a result, the ID93 vaccine consisted of Rv2608, Rv1813, Rv3619, and Rv3620 was predicted to have the best potential for preventing both active and latent TB infection. Our results highlighted the importance and usefulness of a systematic and comprehensive assessment of protein antigens using computational approaches in MTB vaccine development.
Collapse
Affiliation(s)
- Edison Ong
- Department of Computational Medicine and Bioinformatics, University of Michigan, 500 S State St, Ann Arbor, MI 48109, United States.
| | - Yongqun He
- Unit for Laboratory Animal Medicine, University of Michigan, 500 S State St, Ann Arbor, MI 48109, United States; Department of Microbiology and Immunology Department, University of Michigan, 500 S State St, Ann Arbor, MI 48109, United States; Center of Computational Medicine and Bioinformatics, University of Michigan, 500 S State St, Ann Arbor, MI 48109, United States.
| | - Zhenhua Yang
- Department of Epidemiology, School of Public Health, University of Michigan, 500 S State St, Ann Arbor, MI 48109, United States.
| |
Collapse
|
13
|
Khader SA, Divangahi M, Hanekom W, Hill PC, Maeurer M, Makar KW, Mayer-Barber KD, Mhlanga MM, Nemes E, Schlesinger LS, van Crevel R, Vankayalapati R(K, Xavier RJ, Netea MG. Targeting innate immunity for tuberculosis vaccination. J Clin Invest 2019; 129:3482-3491. [PMID: 31478909 PMCID: PMC6715374 DOI: 10.1172/jci128877] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Vaccine development against tuberculosis (TB) is based on the induction of adaptive immune responses endowed with long-term memory against mycobacterial antigens. Memory B and T cells initiate a rapid and robust immune response upon encounter with Mycobacterium tuberculosis, thus achieving long-lasting protection against infection. Recent studies have shown, however, that innate immune cell populations such as myeloid cells and NK cells also undergo functional adaptation after infection or vaccination, a de facto innate immune memory that is also termed trained immunity. Experimental and epidemiological data have shown that induction of trained immunity contributes to the beneficial heterologous effects of vaccines such as bacille Calmette-Guérin (BCG), the licensed TB vaccine. Moreover, increasing evidence argues that trained immunity also contributes to the anti-TB effects of BCG vaccination. An interaction among immunological signals, metabolic rewiring, and epigenetic reprogramming underlies the molecular mechanisms mediating trained immunity in myeloid cells and their bone marrow progenitors. Future studies are warranted to explore the untapped potential of trained immunity to develop a future generation of TB vaccines that would combine innate and adaptive immune memory induction.
Collapse
Affiliation(s)
- Shabaana A. Khader
- Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Maziar Divangahi
- Meakins-Christie Laboratories, Department of Medicine, Department of Microbiology and Immunology, and Department of Pathology, McGill International TB Centre, McGill University Health Centre, Montreal, Quebec, Canada
| | - Willem Hanekom
- Bill & Melinda Gates Foundation, Seattle, Washington, USA
| | - Philip C. Hill
- Centre for International Health, Department of Preventive and Social Medicine, University of Otago Medical School, Dunedin, New Zealand
| | - Markus Maeurer
- Department of Oncology/Haematology, Krankenhaus Nordwest (KHNW), Frankfurt, Germany
- ImmunoSurgery Unit, Champalimaud Foundation, Lisbon, Portugal
| | - Karen W. Makar
- Bill & Melinda Gates Foundation, Seattle, Washington, USA
| | - Katrin D. Mayer-Barber
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Musa M. Mhlanga
- Division of Chemical Systems & Synthetic Biology, Institute for Infectious Disease & Molecular Medicine (IDM), Faculty of Health Sciences, Department of Integrative Biomedical Sciences, and
| | - Elisa Nemes
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | | | - Reinout van Crevel
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Raman (Krishna) Vankayalapati
- Department of Pulmonary Immunology, Center for Biomedical Research, University of Texas Health Science Center at Tyler, Tyler, Texas, USA
| | - Ramnik J. Xavier
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Center for Computational and Integrative Biology and
- Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Mihai G. Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
- Department for Genomics & Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | | |
Collapse
|
14
|
A multi-antigenic MVA vaccine increases efficacy of combination chemotherapy against Mycobacterium tuberculosis. PLoS One 2018; 13:e0196815. [PMID: 29718990 PMCID: PMC5931632 DOI: 10.1371/journal.pone.0196815] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 04/22/2018] [Indexed: 12/13/2022] Open
Abstract
Despite the existence of the prophylactic Bacille Calmette-Guérin (BCG) vaccine, infection by Mycobacterium tuberculosis (Mtb) remains a major public health issue causing up to 1.8 million annual deaths worldwide. Increasing prevalence of Mtb strains resistant to antibiotics represents an urgent threat for global health that has prompted a search for alternative treatment regimens not subject to development of resistance. Immunotherapy constitutes a promising approach to improving current antibiotic treatments through engagement of the host’s immune system. We designed a multi-antigenic and multiphasic vaccine, based on the Modified Vaccinia Ankara (MVA) virus, denoted MVATG18598, which expresses ten antigens classically described as representative of each of different phases of Mtb infection. In vitro analysis coupled with multiple-passage evaluation demonstrated that this vaccine is genetically stable, i.e. fit for manufacturing. Using different mouse strains, we show that MVATG18598 vaccination results in both Th1-associated T-cell responses and cytolytic activity, targeting all 10 vaccine-expressed Mtb antigens. In chronic post-exposure mouse models, MVATG18598 vaccination in combination with an antibiotic regimen decreases the bacterial burden in the lungs of infected mice, compared with chemotherapy alone, and is associated with long-lasting antigen-specific Th1-type T cell and antibody responses. In one model, co-treatment with MVATG18598 prevented relapse of the disease after treatment completion, an important clinical goal. Overall, results demonstrate the capacity of the therapeutic MVATG18598 vaccine to improve efficacy of chemotherapy against TB. These data support further development of this novel immunotherapeutic in the treatment of Mtb infections.
Collapse
|
15
|
Luo W, Qu Z, Zhang L, Xie Y, Luo F, Tan Y, Pan Q, Zhang XL. Recombinant BCG::Rv2645 elicits enhanced protective immunity compared to BCG in vivo with induced ISGylation-related genes and Th1 and Th17 responses. Vaccine 2018; 36:2998-3009. [PMID: 29681409 DOI: 10.1016/j.vaccine.2018.04.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 03/21/2018] [Accepted: 04/10/2018] [Indexed: 12/27/2022]
Abstract
There is a need to develop protective vaccines against tuberculosis (TB). Recently, we identified an immunodominant T-cell antigen, Rv2645, from the region of deletion 13 (RD13) of M. tuberculosis (M. tb) H37Rv, which is absent in Bacille Calmette-Guérin (BCG). Here, a recombinant BCG expressing Rv2645, namely, BCG::Rv2645, was constructed. Compared to BCG, we found that BCG::Rv2645 improved the antigen presentation capacity of dendritic cells (DCs) and elicited much stronger Th1 and Th17 responses, higher CD44highCD62low effector memory CD4+ T cells (TEM), and fewer T regulated cells (Treg) and regulatory B10 in mice. Importantly, BCG::Rv2645 exhibited enhanced protective efficacy against virulent M. tb H37Rv challenge in both mice and rhesus monkeys, showing less severe pathology and reduced pathogens. Further, transcriptomic analysis and reverse transcription-quantitative real time PCR revealed that the mRNA levels of ISGylation (Isg)-related genes such as interferon-stimulated gene 15 (Isg15), and Th1- and Th17-related genes such as interferon-γ (IFN-γ) and interleukin-17A (IL-17A) were significantly up-regulated in splenocytes and macrophages after stimulation with Rv2645. This study shows that BCG::Rv2645 is a promising TB vaccine candidate with enhanced protective immunity. The enhanced Th1/Th17 immune responses and up-regulation of ISGylation-related genes induced by Rv2645 may be major factors contributing to the protective immunity of BCG::Rv2645.
Collapse
Affiliation(s)
- Wei Luo
- State Key Laboratory of Virology and Department of Immunology, College of Basic Medical Sciences, Medical Research Institute and Key Laboratory of Allergy and Immunology, Wuhan University School of Medicine, Hubei Province, Wuhan 430071, China; Department of Clinical Laboratory, Tianjin Medical University General Hospital, Tianjin 30052, China
| | - Zilu Qu
- State Key Laboratory of Virology and Department of Immunology, College of Basic Medical Sciences, Medical Research Institute and Key Laboratory of Allergy and Immunology, Wuhan University School of Medicine, Hubei Province, Wuhan 430071, China
| | - Lingyun Zhang
- State Key Laboratory of Virology and Department of Immunology, College of Basic Medical Sciences, Medical Research Institute and Key Laboratory of Allergy and Immunology, Wuhan University School of Medicine, Hubei Province, Wuhan 430071, China
| | - Yan Xie
- State Key Laboratory of Virology and Department of Immunology, College of Basic Medical Sciences, Medical Research Institute and Key Laboratory of Allergy and Immunology, Wuhan University School of Medicine, Hubei Province, Wuhan 430071, China
| | - Fengling Luo
- State Key Laboratory of Virology and Department of Immunology, College of Basic Medical Sciences, Medical Research Institute and Key Laboratory of Allergy and Immunology, Wuhan University School of Medicine, Hubei Province, Wuhan 430071, China
| | - Yang Tan
- State Key Laboratory of Virology and Department of Immunology, College of Basic Medical Sciences, Medical Research Institute and Key Laboratory of Allergy and Immunology, Wuhan University School of Medicine, Hubei Province, Wuhan 430071, China
| | - Qin Pan
- State Key Laboratory of Virology and Department of Immunology, College of Basic Medical Sciences, Medical Research Institute and Key Laboratory of Allergy and Immunology, Wuhan University School of Medicine, Hubei Province, Wuhan 430071, China
| | - Xiao-Lian Zhang
- State Key Laboratory of Virology and Department of Immunology, College of Basic Medical Sciences, Medical Research Institute and Key Laboratory of Allergy and Immunology, Wuhan University School of Medicine, Hubei Province, Wuhan 430071, China.
| |
Collapse
|
16
|
Achkar JM, Prados-Rosales R. Updates on antibody functions in Mycobacterium tuberculosis infection and their relevance for developing a vaccine against tuberculosis. Curr Opin Immunol 2018; 53:30-37. [PMID: 29656063 DOI: 10.1016/j.coi.2018.04.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 03/31/2018] [Accepted: 04/02/2018] [Indexed: 11/16/2022]
Abstract
A more effective vaccine to control tuberculosis (TB), a major global public health problem, is urgently needed. Current vaccine candidates focus predominantly on eliciting cell-mediated immunity but other arms of the immune system also contribute to protection against TB. We review here recent studies that enhance our current knowledge of antibody-mediated functions against Mycobacterium tuberculosis. These findings, which contribute to the increasing evidence that antibodies have a protective role against TB, include demonstrations that firstly distinct human antibody Fc glycosylation patterns, found in latent M. tuberculosis infection but not in active TB, influence the efficacy of the host to control M. tuberculosis infection, secondly antibody isotype influences human antibody functions, and thirdly that antibodies targeting M. tuberculosis surface antigens are protective. We discuss these findings in the context of TB vaccine development and highlight the need for further research on antibody-mediated immunity in M. tuberculosis infection.
Collapse
Affiliation(s)
- Jacqueline M Achkar
- Department of Medicine, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461, United States; Department of Microbiology and Immunology, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461, United States.
| | - Rafael Prados-Rosales
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461, United States; Center for Cooperative Research bioGUNE (CICbioGUNE), Bizkaia Technology Park, 48160 Derio, Bizkaia, Spain
| |
Collapse
|
17
|
Abstract
Vaccinations have had tremendous success in the 20th century. However, in the 21st century, we are facing complex immunological issues in relation to controlling underlying infectious diseases. Therefore, new technologies are needed to develop vaccines against infectious diseases like respiratory syncytial virus, human immunodeficiency virus, and cytomegalovirus. In addition, recent emerging infections have taught us that we must prepare preventative measures in advance using our scientific abilities.
Collapse
Affiliation(s)
- Stanley A Plotkin
- Department of Pediatrics, University of Pennsylvania , Doylestown, Pennsylvania
| |
Collapse
|
18
|
Perez-Martinez AP, Ong E, Zhang L, Marrs CF, He Y, Yang Z. Conservation in gene encoding Mycobacterium tuberculosis antigen Rv2660 and a high predicted population coverage of H56 multistage vaccine in South Africa. INFECTION GENETICS AND EVOLUTION 2017; 55:244-250. [PMID: 28941991 DOI: 10.1016/j.meegid.2017.09.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 09/11/2017] [Accepted: 09/19/2017] [Indexed: 01/26/2023]
Abstract
H56/AERAS-456+IC31 (H56), composed of two early secretion proteins, Ag85B and ESAT-6, and a latency associated protein, Rv2660, and the IC31 Intercell adjuvant, is a new fusion subunit vaccine candidate designed to induce immunity against both new infection and reactivation of latent tuberculosis infection. Efficacy of subunit vaccines may be affected by the diversity of vaccine antigens among clinical strains and the extent of recognition by the diverse HLA molecules in the recipient population. Although a previous study showed the conservative nature of Ag85B- and ESAT-6-encoding genes, genetic diversity of Rv2660c that encodes RV2660 is largely unknown. The population coverage of H56 as a whole yet remains to be assessed. The present study was conducted to address these important knowledge gaps. DNA sequence analysis of Rv2660c found no variation among 83 of the 84 investigated clinical strains belonging to four genetic lineages. H56 was predicted to have as high as 99.6% population coverage in the South Africa population using the Immune Epitope Database (IEDB) Population Coverage Tool. Further comparison of H56 population coverage between South African Blacks and Caucasians based on the phenotypic frequencies of binding MHC Class I and Class II supertype alleles found that all of the nine MHC-I and six of eight MHC-II human leukocyte antigen (HLA) supertype alleles analyzed were significantly differentially expressed between the two subpopulations. This finding suggests the presence of race-specific functional binding motifs of MHC-I and MHC-II HLA alleles, which, in turn, highlights the importance of including diverse populations in vaccine clinical evaluation. In conclusion, H56 vaccine is predicted to have a promising population coverage in South Africa; this study demonstrates the utility of integrating comparative genomics and bioinformatics in bridging animal and clinical studies of novel TB vaccines.
Collapse
Affiliation(s)
- Angy P Perez-Martinez
- Department of Epidemiology, School of Public Health, University of Michigan, 1415 Washington Heights, Ann Arbor, MI 48109-2029, United States.
| | - Edison Ong
- Department of Computational Medicine and Bioinformatics, University of Michigan, 1415 Washington Heights, Ann Arbor, MI 48109-2029, United States.
| | - Lixin Zhang
- Department of Epidemiology, School of Public Health, University of Michigan, 1415 Washington Heights, Ann Arbor, MI 48109-2029, United States.
| | - Carl F Marrs
- Department of Epidemiology, School of Public Health, University of Michigan, 1415 Washington Heights, Ann Arbor, MI 48109-2029, United States.
| | - Yongqun He
- Unit for Laboratory Animal Medicine, University of Michigan, 1415 Washington Heights, Ann Arbor, MI 48109-2029, United States; Department of Microbiology and Immunology Department, University of Michigan, 1415 Washington Heights, Ann Arbor, MI 48109-2029, United States; Center of Computational Medicine and Bioinformatics, University of Michigan, 1415 Washington Heights, Ann Arbor, MI 48109-2029, United States.
| | - Zhenhua Yang
- Department of Epidemiology, School of Public Health, University of Michigan, 1415 Washington Heights, Ann Arbor, MI 48109-2029, United States.
| |
Collapse
|