1
|
Sharma N, Sharma N, Biswas A, Gupta S, Behura A, Rodriguez GM. Iron-restricted Mycobacterium tuberculosis exports pathogenicity factors packed in extracellular vesicles. PLoS One 2025; 20:e0324919. [PMID: 40445943 PMCID: PMC12124568 DOI: 10.1371/journal.pone.0324919] [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: 01/20/2025] [Accepted: 05/02/2025] [Indexed: 06/02/2025] Open
Abstract
Mycobacterium tuberculosis, the pathogen responsible for human tuberculosis, responds to iron limitation by increasing the production of extracellular vesicles. This study examined the protein composition of induced M. tuberculosis extracellular membrane vesicles using chromatography coupled with mass spectrometry. The results revealed that vesicles contain key pathogenicity factors, including proteins that enhance bacterial survival, immune evasion, and inflammation. These findings deepen our understanding of the potential role of extracellular vesicles in M. tuberculosis-host interactions. The data can also aid in identifying new biomarkers of infection and developing vesicle-based, culture-independent TB diagnostic platforms.
Collapse
Affiliation(s)
- Nishant Sharma
- The Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey, United States of America
| | - Nevadita Sharma
- The Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey, United States of America
| | - Ashis Biswas
- The Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey, United States of America
| | - Shamba Gupta
- The Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey, United States of America
| | - Assirbad Behura
- The Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey, United States of America
| | - Gloria Marcela Rodriguez
- The Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey, United States of America
| |
Collapse
|
2
|
Grace PS, Peters JM, Sixsmith J, Lu R, Irvine EB, Luedeman C, Fenderson BA, Vickers A, Slein MD, McKitrick T, Wei MH, Cummings RD, Wallace A, Cavacini LA, Choudhary A, Proulx MK, Sundling C, Källenius G, Reljic R, Ernst JD, Casadevall A, Locht C, Pinter A, Sassetti CM, Bryson BD, Fortune SM, Alter G. Antibody-Fab and -Fc features promote Mycobacterium tuberculosis restriction. Immunity 2025:S1074-7613(25)00225-0. [PMID: 40449485 DOI: 10.1016/j.immuni.2025.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 01/31/2025] [Accepted: 05/07/2025] [Indexed: 06/03/2025]
Abstract
Mycobacterium tuberculosis, the causative agent of tuberculosis (TB), a leading cause of death by an infectious disease globally, has no efficacious vaccine. Antibodies are implicated in M. tuberculosis control, but the mechanisms of action remain poorly understood. We assembled a library of monoclonal antibodies (mAb) and screened for M. tuberculosis-restrictive activity in mice, identifying protective antibodies targeting diverse antigens. To dissect the mechanism of mAb-mediated M. tuberculosis restriction, we optimized a protective lipoarabinomannan-specific mAb, generating Fc variants. In vivo analysis of these Fc variants revealed a role for Fc-effector function in M. tuberculosis restriction. Restrictive Fc variants altered distribution of M. tuberculosis across innate immune cells. Single-cell transcriptomics highlighted distinctly activated pathways within innate immune cell subpopulations, identifying early activation of neutrophils as a key signature of mAb-mediated M. tuberculosis restriction. Therefore, antibody-mediated restriction of M. tuberculosis is associated with reorganization of the tissue-level immune response to infection and depends on the collaboration of antibody Fab and Fc.
Collapse
Affiliation(s)
- Patricia S Grace
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Department of Immunology and Infectious Disease, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Joshua M Peters
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jaimie Sixsmith
- Department of Immunology and Infectious Disease, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Richard Lu
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Edward B Irvine
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Department of Immunology and Infectious Disease, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | | | | | - Andrew Vickers
- Department of Immunology and Infectious Disease, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Matthew D Slein
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Tanya McKitrick
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Mo-Hui Wei
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Richard D Cummings
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Aaron Wallace
- MassBiologics of the University of Massachusetts Medical School, Boston, MA, USA
| | - Lisa A Cavacini
- MassBiologics of the University of Massachusetts Medical School, Boston, MA, USA
| | - Alok Choudhary
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Megan K Proulx
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, USA
| | - Christopher Sundling
- Division of Infectious Diseases, Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden; Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Gunilla Källenius
- Division of Infectious Diseases, Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden; Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Rajko Reljic
- Institute for Infection and Immunity, St. George's University, London, UK
| | - Joel D Ernst
- Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Camille Locht
- University of Lille, CNRS, Inserm, CHU Lille Institut Pasteur de Lille, U1019-URM9017_Center for Infection and Immunity of Lille, 5900 Lille, France
| | - Abraham Pinter
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Christopher M Sassetti
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, USA
| | - Bryan D Bryson
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sarah M Fortune
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Department of Immunology and Infectious Disease, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA.
| |
Collapse
|
3
|
Navon M, Ben-Shalom N, Dadiani M, Mor M, Yefet R, Bakalenik-Gavry M, Chat D, Balint-Lahat N, Barshack I, Tsarfaty I, Nili Gal-Yam E, Freund NT. Unique characteristics of autoantibodies targeting MET in patients with breast and lung cancer. JCI Insight 2025; 10:e187392. [PMID: 40401526 DOI: 10.1172/jci.insight.187392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 04/09/2025] [Indexed: 05/23/2025] Open
Abstract
The presence of B cells in tumors is correlated with favorable prognosis and efficient response to immunotherapy. While tumor-reactive antibodies have been detected in several cancer types, identifying antibodies that specifically target tumor-associated antigens remains a challenge. Here, we investigated the antibodies spontaneously elicited during breast and lung cancer that bind the cancer-associated antigen MET. We screened patients with lung (n = 25) and breast (n = 75) cancer and found that 13% had antibodies binding to both the recombinant ectodomain of MET, and the ligand binding part of MET, SEMA. MET binding in the breast cancer cohort was significantly correlated with hormone receptor-positive status. We further conducted immunoglobulin sequencing of peripheral MET-enriched B cells from 6 MET-reactive patients. The MET-enriched B cell repertoire was found to be polyclonal and prone to non-IgG1 subclass. Nine monoclonal antibodies were cloned and analyzed, and these exhibited MET binding, low thermostability, and high polyreactivity. Among these, antibodies 87B156 and 69B287 effectively bound to tumor cells and inhibited MET-expressing breast cancer cell lines. Overall, our data demonstrate that some patients with breast and lung cancer develop polyreactive antibodies that cross-react with MET. These autoantibodies have a potential contribution to immune responses against tumors.
Collapse
Affiliation(s)
- Michal Navon
- Department of Microbiology and Clinical Immunology, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Noam Ben-Shalom
- Department of Microbiology and Clinical Immunology, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Maya Dadiani
- Cancer Research Center, Sheba Medical Center, Ramat Gan, Israel
| | - Michael Mor
- Department of Microbiology and Clinical Immunology, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Ron Yefet
- Department of Microbiology and Clinical Immunology, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | | | - Dana Chat
- Cancer Research Center, Sheba Medical Center, Ramat Gan, Israel
| | | | - Iris Barshack
- Department of Pathology, Sheba Medical Center, Ramat Gan, Israel
- Department of Pathology, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ilan Tsarfaty
- Department of Microbiology and Clinical Immunology, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Einav Nili Gal-Yam
- Institute of Breast Oncology, Jusidman Cancer Center, Sheba Medical Center, Ramat Gan, Israel
| | - Natalia T Freund
- Department of Microbiology and Clinical Immunology, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
4
|
Dzigba P, Dekhtyar GAT, Hartman MJ, Winstead-Leroy KJ, Greenlee-Wacker MC, Swarts BM. An Antibody-Recruiting Molecule Enhances Fcγ Receptor-Mediated Uptake and Killing of Mycobacterial Pathogens by Macrophages. ACS Infect Dis 2025. [PMID: 40312277 DOI: 10.1021/acsinfecdis.5c00097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Mycobacteria, which include the infectious agents for tuberculosis (TB) and nontuberculous mycobacteria (NTM) disease, pose a critical health challenge due to traits that allow them to evade host immune clearance and antibiotic action. Toward a novel immunotherapy approach for mycobacteria, we previously reported an antibody-recruiting molecule (ARM) strategy to specifically modify the surface glycans of mycobacteria with exogenous haptens, marking the bacteria for opsonization by endogenous antibodies and enhancing the antibody-mediated immune response. We showed that the ARM, a trehalose-dinitrophenyl conjugate (Tre-DNP), exploited a conserved metabolic pathway to metabolically label the surface of nonpathogenic Mycobacterium smegmatis with DNP, recruited anti-DNP antibodies to the bacterial surface, and enhanced phagocytosis of the bacteria by THP-1 cells. Here, we extend these findings by investigating the ability of the Tre-DNP ARM strategy to increase macrophage-mediated phagocytosis and killing of different pathogenic mycobacterial species and interrogating mechanisms associated with the outcome. We show that Tre-DNP successfully modified the surface of multiple pathogens, including Mycobacterium tuberculosis and the NTM species Mycobacterium abscessus and Mycobacterium avium, and that phagocytosis and killing of intracellular bacteria by differentiated THP-1 cells is significantly enhanced for all species. Furthermore, we find that enhanced uptake is dependent upon the Fcγ receptor (FcγR) and enhanced killing correlates with sustained production of reactive oxygen species (ROS) and increased phagosome-lysosome fusion. Overall, our data demonstrate that Tre-DNP efficiently promotes ingestion of mycobacteria by human macrophages via the FcγR and enhances host effector responses against the pathogen. Thus, ARMs are tools that can be exploited for the purposes of (i) conducting mechanistic studies on immune recognition and elimination of mycobacterial pathogens and (ii) developing immune-targeting strategies against mycobacterial pathogens.
Collapse
Affiliation(s)
- Priscilla Dzigba
- Biochemistry, Cell, and Molecular Biology Graduate Programs, Central Michigan University, Mount Pleasant, Michigan 48859, United States
| | | | | | | | | | - Benjamin M Swarts
- Biochemistry, Cell, and Molecular Biology Graduate Programs, Central Michigan University, Mount Pleasant, Michigan 48859, United States
| |
Collapse
|
5
|
Li H, Ji J, Qu M, Ma X, Zuo Y, Tang M, Zeng L, Li H. Isolation and characterization of a protective monoclonal antibody targeting outer membrane protein (OmpA) against tuberculosis. Microbiol Spectr 2025; 13:e0294224. [PMID: 39964152 PMCID: PMC11960079 DOI: 10.1128/spectrum.02942-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 01/10/2025] [Indexed: 04/02/2025] Open
Abstract
Tuberculosis (TB) caused by Mycobacterium tuberculosis (Mtb) complex is an important zoonotic infectious disease around the world, and the One Health approach is an essential strategy for TB prevention and control. The abundant surface antigens present on the cell wall of Mtb can induce protective antibodies; however, its full characterization remains incomplete. Previous research has shown that antibodies targeting surface proteins can enhance host defense mechanisms. Therefore, identifying antigens that stimulate the production of protective antibodies is essential. In this study, we focused on a select antigen, outer membrane protein (OmpA), located on the outer membrane of Mtb. By screening for antibodies with protective effects in phagocytosis and intracellular killing in vitro, we identified a protective antibody targeting OmpA. We prepared an antibody, designated as 1E1, belonging to the IgG2b isotype, which exhibited high titers of 1:2,048,000. Further research demonstrated that antibody-induced protection was achieved by promoting opsonophagocytosis in a dose-dependent manner, enhancing phagosome-lysosome fusion, and inhibiting mycobacterial intracellular growth. These findings were corroborated in vivo, with a reduction in bacterial loads of approximately 0.7 log observed in the preventive group and almost 1.0 log in the therapeutic group of mice treated with the antibodies, compared to the negative control group. Cytotoxicity assays, animal toxicity analyses, and pharmacokinetic evaluations confirmed the safety and sustained effectiveness of the antibody in vivo. These findings indicate that OmpA can elicit protective antibodies and may serve as a treatment strategy for drug-resistant TB and a promising antigen for TB vaccine development. IMPORTANCE In this study, we identified a protective antibody targeting the outer membrane protein (OmpA) of Mycobacterium tuberculosis. This monoclonal antibody (MAb) belongs to the IgG2b isotype and exhibits high titers of 1:2,048,000 to the antigen. The cell infection assays demonstrated that antibody protection was achieved by promoting opsonophagocytosis in a dose-dependent manner, enhancing phagosome-lysosome fusion, and inhibiting mycobacterial intracellular growth in vitro and ex vivo. Cytotoxicity assays, animal toxicity analyses, and pharmacokinetic evaluations confirmed the safety and sustained effectiveness of the antibody in vivo. Furthermore, the mAb 1E1 can reduce the organs' bacterial burdens and pathological damages in the prevention mouse model as well as the treatment models. Above all, in this study, we found a novel mAb named 1E1 with IgG2b isotype targeting OmpA can have protection against tuberculosis (TB) in mice, which may serve as a treatment strategy for drug-resistant TB and a promising antigen for TB vaccine development.
Collapse
Affiliation(s)
- Huoming Li
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jiahong Ji
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Mengjin Qu
- Key Laboratory of Veterinary Public Health of Ministry of Agriculture, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Xiuling Ma
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - You Zuo
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Minghui Tang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Lingyuan Zeng
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Hao Li
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
6
|
Dzigba P, Seth MA, Greenlee-Wacker MC, Swarts BM. Redirecting the host immune response to bacterial infection with antibody-recruiting molecules (ARMs). Curr Opin Chem Biol 2025; 86:102585. [PMID: 40117716 DOI: 10.1016/j.cbpa.2025.102585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 02/18/2025] [Accepted: 02/22/2025] [Indexed: 03/23/2025]
Abstract
The increasing prevalence of antibiotic resistance, the stagnation of antibiotic development, and the adaptive capacity of bacteria to subvert the host immune response combine to pose significant global health concerns. Consequently, there is an urgent need to develop alternative therapeutic approaches to combat bacterial infections. Antibody-recruiting molecules (ARMs), which are bispecific small molecules that recruit endogenous antibodies to pathogenic cells or viruses, offer a promising avenue to harness the host immune system to target various diseases. In this review, we cover ARM strategies that have been developed for bacterial pathogens, including Gram-positive bacteria, Gram-negative bacteria, and mycobacteria, and we discuss the prospects and challenges of utilizing ARMs as alternatives to traditional antibiotic therapies.
Collapse
Affiliation(s)
- Priscilla Dzigba
- Department of Chemistry and Biochemistry, Central Michigan University, Mount Pleasant, MI, USA; Biochemistry, Cell, and Molecular Biology Graduate Programs, Central Michigan University, Mount Pleasant, MI, 48859, USA
| | - Megan A Seth
- Biological Sciences Department, California Polytechnic State University, San Luis Obispo, CA, 93407, USA
| | - Mallary C Greenlee-Wacker
- Biological Sciences Department, California Polytechnic State University, San Luis Obispo, CA, 93407, USA.
| | - Benjamin M Swarts
- Department of Chemistry and Biochemistry, Central Michigan University, Mount Pleasant, MI, USA; Biochemistry, Cell, and Molecular Biology Graduate Programs, Central Michigan University, Mount Pleasant, MI, 48859, USA.
| |
Collapse
|
7
|
Lepe BA, Zheng CR, Leddy OK, Allsup BL, Solomon SL, Bryson BD. Protease shaving of Mycobacterium tuberculosis facilitates vaccine antigen discovery and delivery of novel cargoes to the Mtb surface. Microbiol Spectr 2025; 13:e0227724. [PMID: 39688428 PMCID: PMC11792546 DOI: 10.1128/spectrum.02277-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Tuberculosis (TB) is the leading cause of infectious disease death and lacks a vaccine capable of protecting adults from pulmonary TB. The bacterial surface is a critical interface that shapes host-pathogen interactions. Several knowledge gaps persist in our understanding of Mycobacterium tuberculosis (Mtb)-host interactions that may be addressed by an improved understanding of the Mtb surface proteome, including the identification of novel vaccine targets as well as developing new approaches to interrogate host-pathogen interactions. Here, we sought to expand our understanding of the Mtb surface proteome in service of these knowledge gaps by adapting protease shaving protocols for multiplexed quantitative mass spectrometry. Pairing quantitative mass spectrometry with the construction of validation strains, we revealed several novel Mtb proteins on the Mtb surface largely derived from the PE/PPE class of Mtb proteins, including PPE18, a component of a leading Mtb vaccine candidate. We next exploited the localization of PPE18 to decorate the Mtb surface with heterologous proteins. Together, these studies reveal potential novel targets for new Mtb vaccines as well as facilitate new approaches to study difficult-to-study cellular compartments during bacterial growth and infection.IMPORTANCEThe surface of a bacterial pathogen is a critical interface between the bacterium and the immune system. A better understanding of this interface would facilitate the discovery of new vaccine targets, new virulence proteins, and enable new technologies that modify the bacterial surface. In this study, we established a multiplexed and quantitative biochemical strategy to study the surface of Mycobacterium tuberculosis (Mtb) and identified new vaccine targets. We furthermore established design rules for new technologies aimed at modifying the composition of the bacterial surface. Specifically, we achieved a biological milestone that has not been rigorously reported previously, which is the successful modification of the Mtb surface with a non-native protein.
Collapse
Affiliation(s)
- Bianca A. Lepe
- Department of Biological Engineering, MIT, Cambridge, Massachusetts, USA
- Ragon Institute of Mass General, Harvard, and MIT, Cambridge, Massachusetts, USA
| | - Christine R. Zheng
- Department of Biological Engineering, MIT, Cambridge, Massachusetts, USA
- Ragon Institute of Mass General, Harvard, and MIT, Cambridge, Massachusetts, USA
| | - Owen K. Leddy
- Department of Biological Engineering, MIT, Cambridge, Massachusetts, USA
- Ragon Institute of Mass General, Harvard, and MIT, Cambridge, Massachusetts, USA
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, Massachusetts, USA
| | - Benjamin L. Allsup
- Department of Biological Engineering, MIT, Cambridge, Massachusetts, USA
- Ragon Institute of Mass General, Harvard, and MIT, Cambridge, Massachusetts, USA
| | - Sydney L. Solomon
- Department of Biological Engineering, MIT, Cambridge, Massachusetts, USA
- Ragon Institute of Mass General, Harvard, and MIT, Cambridge, Massachusetts, USA
| | - Bryan D. Bryson
- Department of Biological Engineering, MIT, Cambridge, Massachusetts, USA
- Ragon Institute of Mass General, Harvard, and MIT, Cambridge, Massachusetts, USA
| |
Collapse
|
8
|
An Y, Ni R, Zhuang L, Yang L, Ye Z, Li L, Parkkila S, Aspatwar A, Gong W. Tuberculosis vaccines and therapeutic drug: challenges and future directions. MOLECULAR BIOMEDICINE 2025; 6:4. [PMID: 39841361 PMCID: PMC11754781 DOI: 10.1186/s43556-024-00243-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 12/06/2024] [Accepted: 12/24/2024] [Indexed: 01/23/2025] Open
Abstract
Tuberculosis (TB) remains a prominent global health challenge, with the World Health Organization documenting over 1 million annual fatalities. Despite the deployment of the Bacille Calmette-Guérin (BCG) vaccine and available therapeutic agents, the escalation of drug-resistant Mycobacterium tuberculosis strains underscores the pressing need for more efficacious vaccines and treatments. This review meticulously maps out the contemporary landscape of TB vaccine development, with a focus on antigen identification, clinical trial progress, and the obstacles and future trajectories in vaccine research. We spotlight innovative approaches, such as multi-antigen vaccines and mRNA technology platforms. Furthermore, the review delves into current TB therapeutics, particularly for multidrug-resistant tuberculosis (MDR-TB), exploring promising agents like bedaquiline (BDQ) and delamanid (DLM), as well as the potential of host-directed therapies. The hurdles in TB vaccine and therapeutic development encompass overcoming antigen diversity, enhancing vaccine effectiveness across diverse populations, and advancing novel vaccine platforms. Future initiatives emphasize combinatorial strategies, the development of anti-TB compounds targeting novel pathways, and personalized medicine for TB treatment and prevention. Despite notable advances, persistent challenges such as diagnostic failures and protracted treatment regimens continue to impede progress. This work aims to steer future research endeavors toward groundbreaking TB vaccines and therapeutic agents, providing crucial insights for enhancing TB prevention and treatment strategies.
Collapse
Affiliation(s)
- Yajing An
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The Eighth Medical Center of PLA General Hospital, 17#Heishanhu Road, Haidian District, Beijing, 100091, China
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Ruizi Ni
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Li Zhuang
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Ling Yang
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Zhaoyang Ye
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Linsheng Li
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Seppo Parkkila
- Faculty of Medicine and Health Technology, Tampere University, 33014, Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories PLC, Tampere, Finland
| | - Ashok Aspatwar
- Faculty of Medicine and Health Technology, Tampere University, 33014, Tampere, Finland.
| | - Wenping Gong
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The Eighth Medical Center of PLA General Hospital, 17#Heishanhu Road, Haidian District, Beijing, 100091, China.
| |
Collapse
|
9
|
Zeng L, Zuo Y, Tang M, Lei C, Li H, Ma X, Ji J, Li H. A subunit vaccine Ag85A-LpqH focusing on humoral immunity provides substantial protection against tuberculosis in mice. iScience 2025; 28:111568. [PMID: 39868033 PMCID: PMC11760819 DOI: 10.1016/j.isci.2024.111568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/04/2024] [Accepted: 12/06/2024] [Indexed: 01/28/2025] Open
Abstract
The importance of humoral immunity in combating TB has gained extensive recognition. In this study, a subunit vaccine named Ag85A-LpqH (AL) was prepared by fusing the antigen Ag85A proved to induce robust T cell immune responses, and LpqH was shown to produce protective antibodies. The prevention and BCG prime-boost mouse models were established to test the vaccine efficacy. The results indicate that Ag85A-LpqH can induce substantial protection by reducing bacterial loads and pathological lesions. This vaccine can induce robust antibody responses, as well as T cell immune responses especially strong CD8+ T cell responses. Moreover, the serum from AL-immunized mice can reduce the bacterial load and lung pathology in mice. B cell receptor (BCR) sequencing revealed a notable rise in BCR diversity among mice immunized with AL. These results indicate that Ag85A-LpqH can be a promising vaccine candidate for tuberculosis prevention and control.
Collapse
Affiliation(s)
- Lingyuan Zeng
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - You Zuo
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Minghui Tang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Chengrui Lei
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Huoming Li
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xiuling Ma
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jiahong Ji
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Hao Li
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
10
|
Zeng L, Li H. Protocol for rapid evaluation of antibody-mediated protection in macrophages using immunofluorescence techniques. STAR Protoc 2024; 5:103324. [PMID: 39302838 PMCID: PMC11437938 DOI: 10.1016/j.xpro.2024.103324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 06/29/2024] [Accepted: 08/26/2024] [Indexed: 09/22/2024] Open
Abstract
Antibodies have been shown to provide protection against tuberculosis (TB). It is important to identify the qualitative and quantitative protective effects of antibodies before a clinical study. Here, we present a protocol to evaluate antibody-mediated protection in macrophages using confocal fluorescence microscopy and flow cytometry. We describe steps for bacteria and macrophage preparation, cell infection, phagocytosis, and phagosome maturation analysis. For complete details on the use and execution of this protocol, please refer to Zeng et al.1.
Collapse
Affiliation(s)
- Lingyuan Zeng
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Hao Li
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
11
|
Kelkar NS, Curtis NC, Lahey TP, Wieland-Alter W, Stout JE, Larson EC, Jauro S, Scanga CA, Darrah PA, Roederer M, Seder RA, von Reyn CF, Lee J, Ackerman ME. Humoral correlate of vaccine-mediated protection from tuberculosis identified in humans and non-human primates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.05.627012. [PMID: 39713388 PMCID: PMC11661070 DOI: 10.1101/2024.12.05.627012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Development of an effective tuberculosis (TB) vaccine has been challenged by incomplete understanding of specific factors that provide protection against Mycobacterium tuberculosis (Mtb) and the lack of a known correlate of protection (CoP). Using a combination of samples from a vaccine showing efficacy (DarDar [NCT00052195]) and Bacille Calmette-Guerin (BCG)-immunized humans and nonhuman primates (NHP), we identify a humoral CoP that translates across species and vaccine regimens. Antibodies specific to the DarDar vaccine strain (M. obuense) sonicate (MOS) correlate with protection from the efficacy endpoint of definite TB. In humans, antibodies to MOS also scale with vaccine dose, are elicited by BCG vaccination, are observed during TB disease, and demonstrate cross-reactivity with Mtb; in NHP, MOS-specific antibodies scale with dose and serve as a CoP mediated by BCG vaccination. Collectively, this study reports a novel humoral CoP and specific antigenic targets that may be relevant to achieving vaccine-mediated protection from TB.
Collapse
Affiliation(s)
- Natasha S. Kelkar
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH, USA
| | | | - Timothy P. Lahey
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, VT, USA
| | - Wendy Wieland-Alter
- Geisel School of Medicine at Dartmouth, 1 Medical Center Drive, Lebanon, NH, USA
| | - Jason E. Stout
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Erica C. Larson
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Solomon Jauro
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Charles A. Scanga
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Patricia A. Darrah
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institute of Health (NIH), Bethesda, MD, USA
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institute of Health (NIH), Bethesda, MD, USA
| | - Robert A. Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institute of Health (NIH), Bethesda, MD, USA
| | - C. Fordham von Reyn
- Dartmouth International Vaccine Initiative, Geisel School of Medicine, 1 Medical Center Drive, Lebanon, NH, USA
| | - Jiwon Lee
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Margaret E. Ackerman
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH, USA
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| |
Collapse
|
12
|
Du F, Deng Y, Deng L, Du B, Xing A, Tao H, Li H, Xie L, Zhang X, Sun T, Li H. T-cell receptor and B-cell receptor repertoires profiling in pleural tuberculosis. Front Immunol 2024; 15:1473486. [PMID: 39664375 PMCID: PMC11632106 DOI: 10.3389/fimmu.2024.1473486] [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: 07/31/2024] [Accepted: 10/31/2024] [Indexed: 12/13/2024] Open
Abstract
Background Tuberculosis (TB) is a leading cause of death worldwide from a single infectious agent. In China the most common extra-pulmonary TB (EPTB) is pleural tuberculosis (PLTB). An important clinical feature of PLTB is that the lymphocytes associated with TB will accumulate in the pleural fluid. The adaptive immune repertoires play important roles in Mycobacterium tuberculosis (Mtb) infection. Methods In this study, 10 PLTB patients were enrolled, and their Peripheral Blood Mononuclear Cells(PBMCs) and Pleural Effusion Mononuclear Cells(PEMCs) were collected. After T cells were purified from PBMCs and PEMCs, high-throughput immunosequencing of the TCRβ chain (TRB), TCRγ chain(TRG), and B cell receptor(BCR) immunoglobulin heavy chain (IGH) were conducted on these samples. Results The TRB, TRG, and BCR IGH repertoires were characterized between the pleural effusion and blood in PLTB patients, and the shared clones were analyzed and collected. The binding activity of antibodies in plasma and pleural effusion to Mtb antigens was tested which indicates that different antibodies responses to Mtb antigens in plasma and pleural effusion in PLTB patients. Moreover, GLIPH2 was used to identify the specificity groups of TRB clusters and Mtb-specific TRB sequences were analyzed and collected by VJ mapping. Conclusion We characterize the adaptive immune repertoires and identify the shared clones and Mtb-specific clones in pleural effusion and blood in PLTB patients which can give important clues for TB diagnosis, treatment, and vaccine development.
Collapse
MESH Headings
- Humans
- Receptors, Antigen, B-Cell/genetics
- Receptors, Antigen, B-Cell/immunology
- Tuberculosis, Pleural/immunology
- Tuberculosis, Pleural/diagnosis
- Male
- Female
- Middle Aged
- Mycobacterium tuberculosis/immunology
- Adult
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- Aged
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
Collapse
Affiliation(s)
- Fengjiao Du
- National Clinical Laboratory on Tuberculosis, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Yunyun Deng
- Hangzhou ImmuQuad Biotechnologies, Hangzhou, China
| | - Ling Deng
- Hangzhou ImmuQuad Biotechnologies, Hangzhou, China
| | - Boping Du
- National Clinical Laboratory on Tuberculosis, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Aiying Xing
- National Clinical Laboratory on Tuberculosis, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Hong Tao
- National Clinical Laboratory on Tuberculosis, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Hua Li
- National Clinical Laboratory on Tuberculosis, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Li Xie
- National Clinical Laboratory on Tuberculosis, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Xinyong Zhang
- National Clinical Laboratory on Tuberculosis, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Tao Sun
- Hangzhou ImmuQuad Biotechnologies, Hangzhou, China
- Institute of Wenzhou, Zhejiang University, Wenzhou, China
| | - Hao Li
- Center for Infectious Disease Research, School of Medicine, Tsinghua University, Beijing, China
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
13
|
Grace PS, Peters JM, Sixsmith J, Lu R, Luedeman C, Fenderson BA, Vickers A, Slein MD, Irvine EB, McKitrick T, Wei MH, Cummings RD, Wallace A, Cavacini LA, Choudhary A, Proulx MK, Sundling C, Källenius G, Reljic R, Ernst JD, Casadevall A, Locht C, Pinter A, Sasseti CM, Bryson BD, Fortune SM, Alter G. Antibody-Fab and -Fc features promote Mycobacterium tuberculosis restriction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.07.617070. [PMID: 39416184 PMCID: PMC11482752 DOI: 10.1101/2024.10.07.617070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), is a leading cause of death by an infectious disease globally, with no efficacious vaccine. Antibodies are implicated in Mtb control, but the mechanisms of antibody action remain poorly understood. We assembled a library of TB monoclonal antibodies (mAb) and screened for the ability to restrict Mtb in mice, identifying protective antibodies targeting known and novel antigens. To dissect the mechanism of mAb-mediated Mtb restriction, we optimized a protective lipoarabinomannan-specific mAb through Fc-swapping. In vivo analysis of these Fc-variants revealed a critical role for Fc-effector function in Mtb restriction. Restrictive Fc-variants altered distribution of Mtb across innate immune cells. Single-cell transcriptomics highlighted distinctly activated molecular circuitry within innate immune cell subpopulations, highlighting early activation of neutrophils as a key signature of mAb-mediated Mtb restriction. Therefore, improved antibody-mediated restriction of Mtb is associated with reorganization of the tissue-level immune response to infection and depends on the collaboration of antibody Fab and Fc.
Collapse
|
14
|
Wang Q, Nag D, Baldwin SL, Coler RN, McNamara RP. Antibodies as key mediators of protection against Mycobacterium tuberculosis. Front Immunol 2024; 15:1430955. [PMID: 39286260 PMCID: PMC11402706 DOI: 10.3389/fimmu.2024.1430955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 08/13/2024] [Indexed: 09/19/2024] Open
Abstract
Tuberculosis (TB) is caused by infection with the bacterial pathogen Mycobacterium tuberculosis (M.tb) in the respiratory tract. There was an estimated 10.6 million people newly diagnosed with TB, and there were approximately 1.3 million deaths caused by TB in 2022. Although the global prevalence of TB has remained high for decades and is an annual leading cause of death attributed to infectious diseases, only one vaccine, Bacillus Calmette-Guérin (BCG), has been approved so far to prevent/attenuate TB disease. Correlates of protection or immunological mechanisms that are needed to control M.tb remain unknown. The protective role of antibodies after BCG vaccination has also remained largely unclear; however, recent studies have provided evidence for their involvement in protection against disease, as biomarkers for the state of infection, and as potential predictors of outcomes. Interestingly, the antibodies generated post-vaccination with BCG are linked to the activation of innate immune cascades, providing further evidence that antibody effector functions are critical for protection against respiratory pathogens such as M.tb. In this review, we aim to provide current knowledge of antibody application in TB diagnosis, prevention, and treatment. Particularly, this review will focus on 1) The role of antibodies in preventing M.tb infections through preventing Mtb adherence to epithelium, antibody-mediated phagocytosis, and antibody-mediated cellular cytotoxicity; 2) The M.tb-directed antibody response generated after vaccination and how humoral profiles with different glycosylation patterns of these antibodies are linked with protection against the disease state; and 3) How antibody-mediated immunity against M.tb can be further explored as early diagnosis biomarkers and different detection methods to combat the global M.tb burden. Broadening the paradigm of differentiated antibody profiling and antibody-based detection during TB disease progression offers new directions for diagnosis, treatment, and preventative strategies. This approach involves linking the aforementioned humoral responses with the disease state, progression, and clearance.
Collapse
Affiliation(s)
- Qixin Wang
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, MA, United States
| | - Deepika Nag
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Susan L. Baldwin
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Rhea N. Coler
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, United States
- Department of Global Health, University of Washington, Seattle, WA, United States
| | - Ryan P. McNamara
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, MA, United States
| |
Collapse
|
15
|
Irvine EB, Nikolov A, Khan MZ, Peters JM, Lu R, Sixsmith J, Wallace A, van Woudenbergh E, Shin S, Karpinski W, Hsiao JC, Casadevall A, Bryson BD, Cavacini L, Grace PS, Alter G, Fortune SM. Fc-engineered antibodies promote neutrophil-dependent control of Mycobacterium tuberculosis. Nat Microbiol 2024; 9:2369-2382. [PMID: 39174703 PMCID: PMC11371646 DOI: 10.1038/s41564-024-01777-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 07/09/2024] [Indexed: 08/24/2024]
Abstract
Mounting evidence indicates that antibodies can contribute towards control of tuberculosis (TB). However, the underlying mechanisms of humoral immune protection and whether antibodies can be exploited in therapeutic strategies to combat TB are relatively understudied. Here we engineered the receptor-binding Fc (fragment crystallizable) region of an antibody recognizing the Mycobacterium tuberculosis (Mtb) capsule, to define antibody Fc-mediated mechanism(s) of Mtb restriction. We generated 52 Fc variants that either promote or inhibit specific antibody effector functions, rationally building antibodies with enhanced capacity to promote Mtb restriction in a human whole-blood model of infection. While there is likely no singular Fc profile that universally drives control of Mtb, here we found that several Fc-engineered antibodies drove Mtb restriction in a neutrophil-dependent manner. Single-cell RNA sequencing analysis showed that a restrictive Fc-engineered antibody promoted neutrophil survival and expression of cell-intrinsic antimicrobial programs. These data show the potential of Fc-engineered antibodies as therapeutics able to harness the protective functions of neutrophils to promote control of TB.
Collapse
Affiliation(s)
- Edward B Irvine
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Angel Nikolov
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Mehak Z Khan
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Joshua M Peters
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Richard Lu
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Jaimie Sixsmith
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Aaron Wallace
- MassBiologics of the University of Massachusetts Chan Medical School, Boston, MA, USA
| | | | - Sally Shin
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | | | - Jeff C Hsiao
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Bryan D Bryson
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Lisa Cavacini
- MassBiologics of the University of Massachusetts Chan Medical School, Boston, MA, USA
| | - Patricia S Grace
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.
- Division of Infectious Disease, Massachusetts General Hospital, Boston, MA, USA.
| | - Sarah M Fortune
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
16
|
Lepe BA, Zheng CR, Leddy OK, Allsup BL, Solomon SL, Bryson BD. Protease shaving of Mycobacterium tuberculosis facilitates vaccine antigen discovery and delivery of novel cargoes to the Mtb surface. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.02.601718. [PMID: 39005324 PMCID: PMC11245043 DOI: 10.1101/2024.07.02.601718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), is the leading cause of infectious disease death and lacks a vaccine capable of protecting adults from pulmonary TB. Studies have shown that Mtb uses a variety of mechanisms to evade host immunity. Secreted Mtb proteins such as Type VII secretion system substrates have been characterized for their ability to modulate anti-Mtb immunity; however, studies of other pathogens such as Salmonella Typhi and Staphylococcus aureus have revealed that outer membrane proteins can also interact with the innate and adaptive immune system. The Mtb outer membrane proteome has received relatively less attention due to limited techniques available to interrogate this compartment. We filled this gap by deploying protease shaving and quantitative mass spectrometry to identify Mtb outer membrane proteins which serve as nodes in the Mtb-host interaction network. These analyses revealed several novel Mtb proteins on the Mtb surface largely derived from the PE/PPE class of Mtb proteins, including PPE18, a component of a leading Mtb vaccine candidate. We next exploited the localization of PPE18 to decorate the Mtb surface with heterologous proteins and deliver these surface-engineered Mtb to the phagosome. Together, these studies reveal potential novel targets for new Mtb vaccines as well as facilitate new approaches to study difficult to study cellular compartments during infection.
Collapse
Affiliation(s)
- Bianca A. Lepe
- Department of Biological Engineering, MIT, Cambridge, USA
- Ragon Institute of Mass General, Harvard, and MIT, Cambridge, USA
| | - Christine R. Zheng
- Department of Biological Engineering, MIT, Cambridge, USA
- Ragon Institute of Mass General, Harvard, and MIT, Cambridge, USA
| | - Owen K. Leddy
- Department of Biological Engineering, MIT, Cambridge, USA
- Ragon Institute of Mass General, Harvard, and MIT, Cambridge, USA
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, USA
| | - Benjamin L. Allsup
- Department of Biological Engineering, MIT, Cambridge, USA
- Ragon Institute of Mass General, Harvard, and MIT, Cambridge, USA
| | - Sydney L. Solomon
- Department of Biological Engineering, MIT, Cambridge, USA
- Ragon Institute of Mass General, Harvard, and MIT, Cambridge, USA
| | - Bryan D. Bryson
- Department of Biological Engineering, MIT, Cambridge, USA
- Ragon Institute of Mass General, Harvard, and MIT, Cambridge, USA
| |
Collapse
|
17
|
Miles JR, Lu P, Bai S, Aguillón-Durán GP, Rodríguez-Herrera JE, Gunn BM, Restrepo BI, Lu LL. Antigen specificity shapes antibody functions in tuberculosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.03.597169. [PMID: 38895452 PMCID: PMC11185737 DOI: 10.1101/2024.06.03.597169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Tuberculosis (TB) is the number one infectious disease cause of death worldwide due to an incomplete understanding of immunity. Emerging data highlight antibody functions mediated by the Fc domain as immune correlates. However, the mechanisms by which antibody functions impact the causative agent Mycobacterium tuberculosis (Mtb) are unclear. Here, we examine how antigen specificity determined by the Fab domain shapes Fc effector functions against Mtb. Using the critical structural and secreted virulence proteins Mtb cell wall and ESAT-6 & CFP-10, we observe that antigen specificity alters subclass, antibody post-translational glycosylation, and Fc effector functions in TB patients. Moreover, Mtb cell wall IgG3 enhances disease through opsonophagocytosis of extracellular Mtb . In contrast, polyclonal and a human monoclonal IgG1 we generated targeting ESAT-6 & CFP-10 inhibit intracellular Mtb . These data show that antibodies have multiple roles in TB and antigen specificity is a critical determinant of the protective and pathogenic capacity.
Collapse
|
18
|
Ozeki Y, Yokoyama A, Nishiyama A, Yoshida Y, Ohara Y, Mashima T, Tomiyama C, Shaban AK, Takeishi A, Osada-Oka M, Yamaguchi T, Tateishi Y, Maeyama JI, Hakamata M, Moro H, Kikuchi T, Hayashi D, Suzuki F, Yamamoto T, Iho S, Katahira M, Yamamoto S, Matsumoto S. Recombinant mycobacterial DNA-binding protein 1 with post-translational modifications boosts IFN-gamma production from BCG-vaccinated individuals' blood cells in combination with CpG-DNA. Sci Rep 2024; 14:9141. [PMID: 38644371 PMCID: PMC11033290 DOI: 10.1038/s41598-024-58836-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 04/03/2024] [Indexed: 04/23/2024] Open
Abstract
Tuberculosis remains a large health threat, despite the availability of the tuberculosis vaccine, BCG. As BCG efficacy gradually decreases from adolescence, BCG-Prime and antigen-booster may be an efficient strategy to confer vaccine efficacy. Mycobacterial DNA-binding protein 1 (MDP1, namely Rv2986c, hupB or HU) is a major Mycobacterium tuberculosis protein that induces vaccine-efficacy by co-administration with CpG DNA. To produce MDP1 for booster-vaccine use, we have created recombinant MDP1 produced in both Escherichia coli (eMDP1) and Mycolicibacterium smegmatis (mMDP1), an avirulent rapid-growing mycobacteria. We tested their immunogenicity by checking interferon (IFN)-gamma production by stimulated peripheral blood cells derived from BCG-vaccinated individuals. Similar to native M. tuberculosis MDP1, we observed that most lysin resides in the C-terminal half of mMDP1 are highly methylated. In contrast, eMDP1 had less post-translational modifications and IFN-gamma stimulation. mMDP1 stimulated the highest amount of IFN-gamma production among the examined native M. tuberculosis proteins including immunodominant MPT32 and Antigen 85 complex. MDP1-mediated IFN-gamma production was more strongly enhanced when combined with a new type of CpG DNA G9.1 than any other tested CpG DNAs. Taken together, these results suggest that the combination of mMDP1 and G9.1 possess high potential use for human booster vaccine against tuberculosis.
Collapse
Affiliation(s)
- Yuriko Ozeki
- Department of Bacteriology, Niigata University School of Medicine, 1-757, Asahimachi-Dori, Chuo-Ku, Niigata, 951-8510, Japan.
| | - Akira Yokoyama
- Department of Bacteriology, Niigata University School of Medicine, 1-757, Asahimachi-Dori, Chuo-Ku, Niigata, 951-8510, Japan
- Department of Respiratory Medicine, Graduate School of Medicine, University of Tokyo, 7-3-1, Hongo, Bunkyo-Ku, Tokyo, 113-8654, Japan
| | - Akihito Nishiyama
- Department of Bacteriology, Niigata University School of Medicine, 1-757, Asahimachi-Dori, Chuo-Ku, Niigata, 951-8510, Japan
| | - Yutaka Yoshida
- Department of Bacteriology, Niigata University School of Medicine, 1-757, Asahimachi-Dori, Chuo-Ku, Niigata, 951-8510, Japan
| | - Yukiko Ohara
- Department of Bacteriology, Niigata University School of Medicine, 1-757, Asahimachi-Dori, Chuo-Ku, Niigata, 951-8510, Japan
| | - Tsukasa Mashima
- Institute of Advanced Energy, Kyoto University, Gokasho, Uji, Kyoto, 611-0011, Japan
| | - Chikako Tomiyama
- Graduate School of Health Sciences, Niigata University, 2-746, Asahimachi-Dori, Chuo-Ku, Niigata, 951-8518, Japan
| | - Amina K Shaban
- Department of Bacteriology, Niigata University School of Medicine, 1-757, Asahimachi-Dori, Chuo-Ku, Niigata, 951-8510, Japan
| | - Atsuki Takeishi
- Department of Bacteriology, Niigata University School of Medicine, 1-757, Asahimachi-Dori, Chuo-Ku, Niigata, 951-8510, Japan
| | - Mayuko Osada-Oka
- Food Hygiene and Environmental Health, Division of Applied Life Science, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, 1-5, Shimogamo-Nakaragi-Cho, Sakyo-Ku, Kyoto, 606-8522, Japan
| | - Takehiro Yamaguchi
- Department of Bacteriology 1, National Institute of Infectious Disease, 1-23-1, Sinjuku-Ku, Tokyo, 162-8640, Japan
| | - Yoshitaka Tateishi
- Department of Bacteriology, Niigata University School of Medicine, 1-757, Asahimachi-Dori, Chuo-Ku, Niigata, 951-8510, Japan
| | - Jun-Ichi Maeyama
- Department of Bacteriology, Niigata University School of Medicine, 1-757, Asahimachi-Dori, Chuo-Ku, Niigata, 951-8510, Japan
- Reseach Center for Biological Products in the Next Generation, National Institute of Infectious Diseases, 4-7-1, Musashimurayama, Tokyo, 208-0011, Japan
| | - Mariko Hakamata
- Department of Bacteriology, Niigata University School of Medicine, 1-757, Asahimachi-Dori, Chuo-Ku, Niigata, 951-8510, Japan
- Department of Respiratory Medicine and Infectious Disease, Niigata University Graduate School of Medical and Dental Sciences, 1-757, Asahimachi-Dori, Chuo-Ku, Niigata, 951-8510, Japan
| | - Hiroshi Moro
- Department of Respiratory Medicine and Infectious Disease, Niigata University Graduate School of Medical and Dental Sciences, 1-757, Asahimachi-Dori, Chuo-Ku, Niigata, 951-8510, Japan
| | - Toshiaki Kikuchi
- Department of Respiratory Medicine and Infectious Disease, Niigata University Graduate School of Medical and Dental Sciences, 1-757, Asahimachi-Dori, Chuo-Ku, Niigata, 951-8510, Japan
| | - Daisuke Hayashi
- Central Laboratory, Japan BCG Laboratory, 3-1-5 Matsuyama, Kiyose, Tokyo, 204-0022, Japan
| | - Fumiko Suzuki
- Faculty of Medical Sciences, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji-Cho, Yoshida-Gun, Fukui, 910-1193, Japan
| | - Toshiko Yamamoto
- Central Laboratory, Japan BCG Laboratory, 3-1-5 Matsuyama, Kiyose, Tokyo, 204-0022, Japan
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, 4-2-1 Aobacho, Higashi-Murayama, Tokyo, 189-0002, Japan
| | - Sumiko Iho
- Department of Bacteriology, Niigata University School of Medicine, 1-757, Asahimachi-Dori, Chuo-Ku, Niigata, 951-8510, Japan
- Faculty of Medical Sciences, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji-Cho, Yoshida-Gun, Fukui, 910-1193, Japan
- Louis Pasteur Center for Medical Research, 103-5 Tanaka Monzen-cho, Sakyo-ku, Kyoto, 606-8225, Japan
| | - Masato Katahira
- Institute of Advanced Energy, Kyoto University, Gokasho, Uji, Kyoto, 611-0011, Japan
| | - Saburo Yamamoto
- Central Laboratory, Japan BCG Laboratory, 3-1-5 Matsuyama, Kiyose, Tokyo, 204-0022, Japan
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, 4-2-1 Aobacho, Higashi-Murayama, Tokyo, 189-0002, Japan
| | - Sohkichi Matsumoto
- Department of Bacteriology, Niigata University School of Medicine, 1-757, Asahimachi-Dori, Chuo-Ku, Niigata, 951-8510, Japan.
- Laboratory of Tuberculosis, Institute of Tropical Disease, Universitas Airlangga, Kampus C JI. Mulyorejo, Surabaya, 60113, Indonesia.
- Division of Research Aids, Hokkaido University Institute for Vaccine Research and Development, Kita 20, Nishi 10, Kita-Ku, Sapporo, 001-0020, Japan.
| |
Collapse
|
19
|
Zeng L, Ma X, Qu M, Tang M, Li H, Lei C, Ji J, Li H. Immunogenicity and protective efficacy of Ag85A and truncation of PstS1 fusion protein vaccines against tuberculosis. Heliyon 2024; 10:e27034. [PMID: 38463854 PMCID: PMC10920368 DOI: 10.1016/j.heliyon.2024.e27034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 03/12/2024] Open
Abstract
Tuberculosis (TB) is an important public health problem, and the One Health approach is essential for controlling zoonotic tuberculosis. Therefore, a rationally designed and more effective TB vaccine is urgently needed. To enhance vaccine efficacy, it is important to design vaccine candidates that stimulate both cellular and humoral immunity against TB. In this study, we fused the secreted protein Ag85A as the T cell antigen with truncated forms of the mycobacterial cell wall protein PstS1 with B cell epitopes to generate vaccine candidates, Ag85A-tnPstS1 (AP1, AP2, and AP3), and tested their immunogenicity and protective efficacy in mice. The three vaccine candidates induced a significant increase in the levels of T cell-related cytokines such as IFN-γ and IL-17, and AP1 and AP2 can induce more balanced Th1/Th2 responses than AP3. Strong humoral immune responses were also observed in which the production of IgG antibodies including its subclasses IgG1, IgG2c, and IgG3 was tremendously stimulated. AP1 and AP2 induced early antibody responses and more IgG3 isotype antibodies than AP3. Importantly, the mice immunised with the subunit vaccine candidates, particularly AP1 and AP2, had lower bacterial burdens than the control mice. Moreover, the serum from immunised mice can enhance phagocytosis and phagosome-lysosome fusion in macrophages, which can help to eradicate intracellular bacteria. These results indicate that the subunit vaccines Ag85A-tnPstS1 can be promising vaccine candidates for tuberculosis prevention.
Collapse
Affiliation(s)
- Lingyuan Zeng
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Xiuling Ma
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Mengjin Qu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Minghui Tang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Huoming Li
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Chengrui Lei
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Jiahong Ji
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Hao Li
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| |
Collapse
|
20
|
Chugh S, Bahal RK, Dhiman R, Singh R. Antigen identification strategies and preclinical evaluation models for advancing tuberculosis vaccine development. NPJ Vaccines 2024; 9:57. [PMID: 38461350 PMCID: PMC10924964 DOI: 10.1038/s41541-024-00834-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 02/05/2024] [Indexed: 03/11/2024] Open
Abstract
In its myriad devastating forms, Tuberculosis (TB) has existed for centuries, and humanity is still affected by it. Mycobacterium tuberculosis (M. tuberculosis), the causative agent of TB, was the foremost killer among infectious agents until the COVID-19 pandemic. One of the key healthcare strategies available to reduce the risk of TB is immunization with bacilli Calmette-Guerin (BCG). Although BCG has been widely used to protect against TB, reports show that BCG confers highly variable efficacy (0-80%) against adult pulmonary TB. Unwavering efforts have been made over the past 20 years to develop and evaluate new TB vaccine candidates. The failure of conventional preclinical animal models to fully recapitulate human response to TB, as also seen for the failure of MVA85A in clinical trials, signifies the need to develop better preclinical models for TB vaccine evaluation. In the present review article, we outline various approaches used to identify protective mycobacterial antigens and recent advancements in preclinical models for assessing the efficacy of candidate TB vaccines.
Collapse
Affiliation(s)
- Saurabh Chugh
- Centre for Tuberculosis Research, Tuberculosis Research Laboratory, Translational Health Science and Technology Institute, Faridabad, 121001, Haryana, India
| | - Ritika Kar Bahal
- Marshall Centre, School of Biomedical Sciences, University of Western Australia, Perth, Australia
| | - Rohan Dhiman
- Laboratory of Mycobacterial Immunology, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Ramandeep Singh
- Centre for Tuberculosis Research, Tuberculosis Research Laboratory, Translational Health Science and Technology Institute, Faridabad, 121001, Haryana, India.
| |
Collapse
|
21
|
McIntyre S, Warner J, Rush C, Vanderven HA. Antibodies as clinical tools for tuberculosis. Front Immunol 2023; 14:1278947. [PMID: 38162666 PMCID: PMC10755875 DOI: 10.3389/fimmu.2023.1278947] [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: 08/17/2023] [Accepted: 11/27/2023] [Indexed: 01/03/2024] Open
Abstract
Tuberculosis (TB) is a leading cause of morbidity and mortality worldwide. Global research efforts to improve TB control are hindered by insufficient understanding of the role that antibodies play in protective immunity and pathogenesis. This impacts knowledge of rational and optimal vaccine design, appropriate diagnostic biomarkers, and development of therapeutics. Traditional approaches for the prevention and diagnosis of TB may be less efficacious in high prevalence, remote, and resource-poor settings. An improved understanding of the immune response to the causative agent of TB, Mycobacterium tuberculosis (Mtb), will be crucial for developing better vaccines, therapeutics, and diagnostics. While memory CD4+ T cells and cells and cytokine interferon gamma (IFN-g) have been the main identified correlates of protection in TB, mounting evidence suggests that other types of immunity may also have important roles. TB serology has identified antibodies and functional characteristics that may help diagnose Mtb infection and distinguish between different TB disease states. To date, no serological tests meet the World Health Organization (WHO) requirements for TB diagnosis, but multiplex assays show promise for improving the sensitivity and specificity of TB serodiagnosis. Monoclonal antibody (mAb) therapies and serum passive infusion studies in murine models of TB have also demonstrated some protective outcomes. However, animal models that better reflect the human immune response to Mtb are necessary to fully assess the clinical utility of antibody-based TB prophylactics and therapeutics. Candidate TB vaccines are not designed to elicit an Mtb-specific antibody response, but evidence suggests BCG and novel TB vaccines may induce protective Mtb antibodies. The potential of the humoral immune response in TB monitoring and control is being investigated and these studies provide important insight into the functional role of antibody-mediated immunity against TB. In this review, we describe the current state of development of antibody-based clinical tools for TB, with a focus on diagnostic, therapeutic, and vaccine-based applications.
Collapse
Affiliation(s)
- Sophie McIntyre
- Biomedical Sciences and Molecular Biology, College of Public Health, Medical and Veterinary Sciences, James Cook University, Douglas, QLD, Australia
- Australian Institute of Tropical Health and Medicine, James Cook University, Douglas, QLD, Australia
| | - Jeffrey Warner
- Biomedical Sciences and Molecular Biology, College of Public Health, Medical and Veterinary Sciences, James Cook University, Douglas, QLD, Australia
- Australian Institute of Tropical Health and Medicine, James Cook University, Douglas, QLD, Australia
| | - Catherine Rush
- Biomedical Sciences and Molecular Biology, College of Public Health, Medical and Veterinary Sciences, James Cook University, Douglas, QLD, Australia
- Australian Institute of Tropical Health and Medicine, James Cook University, Douglas, QLD, Australia
| | - Hillary A. Vanderven
- Biomedical Sciences and Molecular Biology, College of Public Health, Medical and Veterinary Sciences, James Cook University, Douglas, QLD, Australia
- Australian Institute of Tropical Health and Medicine, James Cook University, Douglas, QLD, Australia
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
22
|
Vergara EJ, Tran AC, Kim MY, Mussá T, Paul MJ, Harrison T, Reljic R. Mucosal and systemic immune responses after a single intranasal dose of nanoparticle and spore-based subunit vaccines in mice with pre-existing lung mycobacterial immunity. Front Immunol 2023; 14:1306449. [PMID: 38130713 PMCID: PMC10733481 DOI: 10.3389/fimmu.2023.1306449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 11/15/2023] [Indexed: 12/23/2023] Open
Abstract
Tuberculosis (TB) is a major global health threat that claims more than one million lives annually. With a quarter of the global population harbouring latent TB, post-exposure vaccination aimed at high-risk populations that could develop active TB disease would be of great public health benefit. Mucosal vaccination is an attractive approach for a predominantly lung disease like TB because it elicits both local and systemic immunity. However, the immunological consequence of mucosal immunisation in the presence of existing lung immunity remains largely unexplored. Using a mycobacterial pre-exposure mouse model, we assessed whether pre-existing mucosal and systemic immune responses can be boosted and/or qualitatively altered by intranasal administration of spore- and nanoparticle-based subunit vaccines. Analysis of lung T cell responses revealed an increasing trend in the frequency of important CD4 and CD8 T cell subsets, and T effector memory cells with a Th1 cytokine (IFNγ and TNFα) signature among immunised mice. Additionally, significantly greater antigen specific Th1, Th17 and IL-10 responses, and antigen-induced T cell proliferation were seen from the spleens of immunised mice. Measurement of antigen-specific IgG and IgA from blood and bronchoalveolar lavage fluid also revealed enhanced systemic and local humoral immune responses among immunised animals. Lastly, peripheral blood mononuclear cells (PBMCs) obtained from the TB-endemic country of Mozambique show that individuals with LTBI showed significantly greater CD4 T cell reactivity to the vaccine candidate as compared to healthy controls. These results support further testing of Spore-FP1 and Nano-FP1 as post-exposure TB vaccines.
Collapse
Affiliation(s)
- Emil Joseph Vergara
- Institute for Infection and Immunity, St. George’s University of London, London, United Kingdom
| | - Andy Cano Tran
- Institute for Infection and Immunity, St. George’s University of London, London, United Kingdom
| | - Mi-Young Kim
- Institute for Infection and Immunity, St. George’s University of London, London, United Kingdom
- Department of Molecular Biology, Jeonbuk National University, Jeonju, Republic of Korea
| | - Tufária Mussá
- Department of Microbiology, Faculty of Medicine, Eduardo Mondlane University, Maputo, Mozambique
| | - Matthew J. Paul
- Institute for Infection and Immunity, St. George’s University of London, London, United Kingdom
| | - Thomas Harrison
- Institute for Infection and Immunity, St. George’s University of London, London, United Kingdom
| | - Rajko Reljic
- Institute for Infection and Immunity, St. George’s University of London, London, United Kingdom
| |
Collapse
|
23
|
Kang H, Lee J, Jung J, Oh EJ. Humoral Response Kinetics and Cross-Immunity in Hospitalized Patients with SARS-CoV-2 WT, Delta, or Omicron Infections: A Comparison between Vaccinated and Unvaccinated Cohorts. Vaccines (Basel) 2023; 11:1803. [PMID: 38140207 PMCID: PMC10747008 DOI: 10.3390/vaccines11121803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 12/24/2023] Open
Abstract
With the ongoing evolution of severe acute respiratory virus-2 (SARS-CoV-2), the number of confirmed COVID-19 cases continues to rise. This study aims to investigate the impact of vaccination status, SARS-CoV-2 variants, and disease severity on the humoral immune response, including cross-neutralizing activity, in hospitalized COVID-19 patients. This retrospective cohort study involved 122 symptomatic COVID-19 patients hospitalized in a single center. Patients were categorized based on the causative specific SARS-CoV-2 variants (33 wild-type (WT), 54 Delta and 35 Omicron) and their vaccination history. Sequential samples were collected to assess binding antibody responses (anti-S/RBD and anti-N) and surrogate virus neutralization tests (sVNTs) against WT, Omicron BA.1, and BA.4/5. The vaccinated breakthrough infection group (V) exhibited higher levels of anti-S/RBD compared to the variant-matched unvaccinated groups (UVs). The Delta infection resulted in a more rapid production of anti-S/RBD levels compared to infections with WT or Omicron variants. Unvaccinated severe WT or Delta infections had higher anti-S/RBD levels compared to mild cases, but this was not the case with Omicron infection. In vaccinated patients, there was no difference in antibody levels between mild and severe infections. Both Delta (V) and Omicron (V) groups showed strong cross-neutralizing activity against WT and Omicron (BA.1 and BA.4/5), ranging from 79.3% to 97.0%. WT (UV) and Delta (UV) infections had reduced neutralizing activity against BA.1 (0.8% to 12.0%) and BA.4/5 (32.8% to 41.0%). Interestingly, patients who received vaccines based on the ancestral spike exhibited positive neutralizing activity against BA.4/5, even though none of the study participants had been exposed to BA.4/5 and it is antigenically more advanced. Our findings suggest that a previous vaccination enhanced the humoral immune response and broadened cross-neutralizing activity to SARS-CoV-2 variants in hospitalized COVID-19 patients.
Collapse
Affiliation(s)
- Hyunhye Kang
- Department of Laboratory Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (H.K.); (J.J.)
- Research and Development Institute for In Vitro Diagnostic Medical Devices, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jihyun Lee
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Republic of Korea;
| | - Jin Jung
- Department of Laboratory Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (H.K.); (J.J.)
- Research and Development Institute for In Vitro Diagnostic Medical Devices, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Eun-Jee Oh
- Department of Laboratory Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (H.K.); (J.J.)
- Research and Development Institute for In Vitro Diagnostic Medical Devices, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
24
|
Martinez-Rivera S, Guet-Revillet H, Herin F, Martin-Blondel G, Miedouge M, Billon L, Treiner E, Renaudineau Y. Low performance of interferon gamma release assay Quantiferon-TB gold coupled or not with Pst1/3/lipoglycan humoral detection to predict Mycobacterium tuberculosis complex disease in a low-burden area. Tuberculosis (Edinb) 2023; 143:102416. [PMID: 37844483 DOI: 10.1016/j.tube.2023.102416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/22/2023] [Accepted: 10/01/2023] [Indexed: 10/18/2023]
Abstract
Whole T cell interferon gamma release assays such as QuantiFERON-TB Gold Plus (QTF-TB) are used to evaluate Mycobacterium tuberculosis complex (MTC) exposure but fail to discriminate latent tuberculosis infection (LTBI) from active disease. In this study conducted in a low-burden area, 1215 patients presenting MTC risk and tested both for QTF-TB and mycobacterial infection (microscopy, culture, and/or PCR) were selected, as well as 1298 controls screened with QTF-TB before medical recruitment. The humoral response (LIODetect®TB-ST) was further evaluated in 199 selected patients. In patients with active disease, MTC positivity (culture and/or PCR with species identification) was associated with QTF-TB positivity (45/56, 80.4 %). Although QTF-TB1/TB2 peptides were not suitable for discriminating against active MTC disease from LTBI, the cut-off value of 4.4 IFN-γ IU/mL produced the best diagnostic performance for MTC detection. Lower levels of QTF-TB were reported among patients with isolated active pulmonary MTC as compared to a lymph-nodal location and a disseminated form. Next, antibodies were detected in 4/55 (7.3 %) active MTC disease cases, while negative in cases of LTBI and indeterminate/negative QTF-TB. In conclusion, the added value to combine cellular (QTF-TB) and humoral (LIODetect®TB-ST) assays to predict an active MTC disease is limited.
Collapse
Affiliation(s)
- Silvia Martinez-Rivera
- Immunology Department Laboratory, Institut Fédératif de Biologie, Toulouse University Hospital, France.
| | - Helene Guet-Revillet
- Bacteriology Department Laboratory, Institut Fédératif de Biologie, Toulouse University Hospital, France.
| | - Fabrice Herin
- INFINITy, Toulouse Institute for Infectious and Inflammatory Diseases, INSERM U1291, CNRS U5051, University Toulouse III, Toulouse, France; Occupational and Environmental Diseases Department, Toulouse University Hospital, France.
| | - Guillaume Martin-Blondel
- INFINITy, Toulouse Institute for Infectious and Inflammatory Diseases, INSERM U1291, CNRS U5051, University Toulouse III, Toulouse, France; Infectious and Tropical Disease Department, Toulouse University Hospital, France.
| | - Marcel Miedouge
- Virology Department Laboratory, Institut Fédératif de Biologie, Toulouse University Hospital, France.
| | | | - Emmanuel Treiner
- Immunology Department Laboratory, Institut Fédératif de Biologie, Toulouse University Hospital, France; INFINITy, Toulouse Institute for Infectious and Inflammatory Diseases, INSERM U1291, CNRS U5051, University Toulouse III, Toulouse, France.
| | - Yves Renaudineau
- Immunology Department Laboratory, Institut Fédératif de Biologie, Toulouse University Hospital, France; INFINITy, Toulouse Institute for Infectious and Inflammatory Diseases, INSERM U1291, CNRS U5051, University Toulouse III, Toulouse, France.
| |
Collapse
|
25
|
Polonsky K, Pupko T, Freund NT. Evaluation of the Ability of AlphaFold to Predict the Three-Dimensional Structures of Antibodies and Epitopes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1578-1588. [PMID: 37782047 DOI: 10.4049/jimmunol.2300150] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 09/06/2023] [Indexed: 10/03/2023]
Abstract
Being able to accurately predict the three-dimensional structure of an Ab can facilitate Ab characterization and epitope prediction, with important diagnostic and clinical implications. In this study, we evaluated the ability of AlphaFold to predict the structures of 222 recently published, high-resolution Fab H and L chain structures of Abs from different species directed against different Ags. We show that although the overall Ab prediction quality is in line with the results of CASP14, regions such as the complementarity-determining regions (CDRs) of the H chain, which are prone to higher variation, are predicted less accurately. Moreover, we discovered that AlphaFold mispredicts the bending angles between the variable and constant domains. To evaluate the ability of AlphaFold to model Ab-Ag interactions based only on sequence, we used AlphaFold-Multimer in combination with ZDOCK to predict the structures of 26 known Ab-Ag complexes. ZDOCK, which was applied on bound components of both the Ab and the Ag, succeeded in assembling 11 complexes, whereas AlphaFold succeeded in predicting only 2 of 26 models, with significant deviations in the docking contacts predicted in the rest of the molecules. Within the 11 complexes that were successfully predicted by ZDOCK, 9 involved short-peptide Ags (18-mer or less), whereas only 2 were complexes of Ab with a full-length protein. Docking of modeled unbound Ab and Ag was unsuccessful. In summary, our study provides important information about the abilities and limitations of using AlphaFold to predict Ab-Ag interactions and suggests areas for possible improvement.
Collapse
Affiliation(s)
- Ksenia Polonsky
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Tal Pupko
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Natalia T Freund
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
26
|
Simonis A, Kreer C, Albus A, Rox K, Yuan B, Holzmann D, Wilms JA, Zuber S, Kottege L, Winter S, Meyer M, Schmitt K, Gruell H, Theobald SJ, Hellmann AM, Meyer C, Ercanoglu MS, Cramer N, Munder A, Hallek M, Fätkenheuer G, Koch M, Seifert H, Rietschel E, Marlovits TC, van Koningsbruggen-Rietschel S, Klein F, Rybniker J. Discovery of highly neutralizing human antibodies targeting Pseudomonas aeruginosa. Cell 2023; 186:5098-5113.e19. [PMID: 37918395 DOI: 10.1016/j.cell.2023.10.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 07/17/2023] [Accepted: 10/02/2023] [Indexed: 11/04/2023]
Abstract
Drug-resistant Pseudomonas aeruginosa (PA) poses an emerging threat to human health with urgent need for alternative therapeutic approaches. Here, we deciphered the B cell and antibody response to the virulence-associated type III secretion system (T3SS) in a cohort of patients chronically infected with PA. Single-cell analytics revealed a diverse B cell receptor repertoire directed against the T3SS needle-tip protein PcrV, enabling the production of monoclonal antibodies (mAbs) abrogating T3SS-mediated cytotoxicity. Mechanistic studies involving cryoelectron microscopy identified a surface-exposed C-terminal PcrV epitope as the target of highly neutralizing mAbs with broad activity against drug-resistant PA isolates. These anti-PcrV mAbs were as effective as treatment with conventional antibiotics in vivo. Our study reveals that chronically infected patients represent a source of neutralizing antibodies, which can be exploited as therapeutics against PA.
Collapse
Affiliation(s)
- Alexander Simonis
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany; German Center for Infection Research (DZIF), partner site Bonn-Cologne, 50937 Cologne, Germany.
| | - Christoph Kreer
- Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Alexandra Albus
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Katharina Rox
- Department of Chemical Biology, Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany; German Center for Infection Research (DZIF), partner site Hannover-Braunschweig, 38124 Braunschweig, Germany
| | - Biao Yuan
- Institute of Structural and Systems Biology, University Medical Center Hamburg-Eppendorf (UKE), 22607 Hamburg, Germany; Centre for Structural Systems Biology (CSSB), 22607 Hamburg, Germany; Deutsches Elektronen-Synchrotron Zentrum (DESY), 22607 Hamburg, Germany
| | - Dmitriy Holzmann
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Joana A Wilms
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Sylvia Zuber
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Lisa Kottege
- Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Sandra Winter
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Meike Meyer
- CF Centre, Pediatric Pulmonology and Allergology, University Children's Hospital Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; Centre for Rare Diseases, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| | - Kristin Schmitt
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Henning Gruell
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany; Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Sebastian J Theobald
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Anna-Maria Hellmann
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany; Department of Experimental Pediatric Oncology, University Children's Hospital Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| | - Christina Meyer
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Meryem Seda Ercanoglu
- Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Nina Cramer
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany
| | - Antje Munder
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), German Center for Lung Research, 30625 Hannover, Germany
| | - Michael Hallek
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Gerd Fätkenheuer
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; German Center for Infection Research (DZIF), partner site Bonn-Cologne, 50937 Cologne, Germany
| | - Manuel Koch
- Institute for Dental Research and Oral Musculoskeletal Biology, Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Harald Seifert
- German Center for Infection Research (DZIF), partner site Bonn-Cologne, 50937 Cologne, Germany; Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine and University Hospital of Cologne, University of Cologne, 50935 Cologne, Germany
| | - Ernst Rietschel
- CF Centre, Pediatric Pulmonology and Allergology, University Children's Hospital Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; Centre for Rare Diseases, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| | - Thomas C Marlovits
- Institute of Structural and Systems Biology, University Medical Center Hamburg-Eppendorf (UKE), 22607 Hamburg, Germany; Centre for Structural Systems Biology (CSSB), 22607 Hamburg, Germany; Deutsches Elektronen-Synchrotron Zentrum (DESY), 22607 Hamburg, Germany
| | - Silke van Koningsbruggen-Rietschel
- CF Centre, Pediatric Pulmonology and Allergology, University Children's Hospital Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; Centre for Rare Diseases, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| | - Florian Klein
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany; German Center for Infection Research (DZIF), partner site Bonn-Cologne, 50937 Cologne, Germany; Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Jan Rybniker
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany; German Center for Infection Research (DZIF), partner site Bonn-Cologne, 50937 Cologne, Germany.
| |
Collapse
|
27
|
Liu Y, Chen T, Zhu Y, Furey A, Lowary TL, Chan J, Bournazos S, Ravetch JV, Achkar JM. Features and protective efficacy of human mAbs targeting Mycobacterium tuberculosis arabinomannan. JCI Insight 2023; 8:e167960. [PMID: 37733444 PMCID: PMC10619501 DOI: 10.1172/jci.insight.167960] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 09/06/2023] [Indexed: 09/23/2023] Open
Abstract
A better understanding of the epitopes most relevant for antibody-mediated protection against tuberculosis (TB) remains a major knowledge gap. We have shown that human polyclonal IgG against the Mycobacterium tuberculosis (M. tuberculosis) surface glycan arabinomannan (AM) and related lipoarabinomannan (LAM) is protective against TB. To investigate the impact of AM epitope recognition and Fcγ receptor (FcγR) binding on antibody functions against M. tuberculosis, we isolated a high-affinity human monoclonal antibody (mAb; P1AM25) against AM and showed its binding to oligosaccharide (OS) motifs we previously found to be associated with in vitro functions of human polyclonal anti-AM IgG. Human IgG1 P1AM25, but not 2 other high-affinity human IgG1 anti-AM mAbs reactive with different AM OS motifs, enhanced M. tuberculosis phagocytosis by macrophages and reduced intracellular growth in an FcγR-dependent manner. P1AM25 in murine IgG2a, but neither murine IgG1 nor a non-FcγR-binding IgG, given intraperitoneally prior to and after aerosolized M. tuberculosis infection, was protective in C57BL/6 mice. Moreover, we demonstrated the protective efficacy of human IgG1 P1AM25 in passive transfer with M. tuberculosis-infected FcγR-humanized mice. These data enhance our knowledge of the important interplay between both antibody epitope specificity and Fc effector functions in the defense against M. tuberculosis and could inform development of vaccines against TB.
Collapse
Affiliation(s)
- Yanyan Liu
- Department of Microbiology and Immunology and
| | - Tingting Chen
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Yongqi Zhu
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Aisha Furey
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Todd L. Lowary
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
- Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - John Chan
- Public Health Research Institute at the International Center for Public Health, New Jersey Medical School – Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| | | | | | - Jacqueline M. Achkar
- Department of Microbiology and Immunology and
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
28
|
Malatji K, Singh A, Thobakgale C, Alexandre K. Development of a Multiplex HIV/TB Diagnostic Assay Based on the Microarray Technology. BIOSENSORS 2023; 13:894. [PMID: 37754128 PMCID: PMC10526232 DOI: 10.3390/bios13090894] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/11/2023] [Accepted: 09/18/2023] [Indexed: 09/28/2023]
Abstract
Currently there are diagnostic tests available for human immunodeficiency virus (HIV) and tuberculosis (TB); however, they are still diagnosed separately, which can delay treatment in cases of co-infection. Here we report on a multiplex microarray technology for the detection of HIV and TB antibodies using p24 as well as TB CFP10, ESAT6 and pstS1 antigens on epoxy-silane slides. To test this technology for antigen-antibody interactions, immobilized antigens were exposed to human sera spiked with physiological concentrations of primary antibodies, followed by secondary antibodies conjugated to a fluorescent reporter. HIV and TB antibodies were captured with no cross-reactivity observed. The sensitivity of the slides was compared to that of high-binding plates. We found that the slides were more sensitive, with the detection limit being 0.000954 µg/mL compared to 4.637 µg/mL for the plates. Furthermore, stability studies revealed that the immobilized antigens could be stored dry for at least 90 days and remained stable across all pH and temperatures assessed, with pH 7.4 and 25 °C being optimal. The data collectively suggested that the HIV/TB multiplex detection technology we developed has the potential for use to diagnose HIV and TB co-infection, and thus can be developed further for the purpose.
Collapse
Affiliation(s)
- Kanyane Malatji
- Array Technology Laboratory, Synthetic Biology and Precision Medicine Centre: Next Generation Health Cluster, Council for Scientific and Industrial Research, Brummeria, Pretoria 0001, South Africa (K.A.)
- Department of Virology, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Braamfontein, Johannesburg 2000, South Africa;
| | - Advaita Singh
- Future Production: Chemicals Cluster, Council for Scientific and Industrial Research, Brummeria, Pretoria 0001, South Africa
| | - Christina Thobakgale
- Department of Virology, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Braamfontein, Johannesburg 2000, South Africa;
- Centre for HIV and STIs, National Institute for Communicable Diseases, Sandringham, Johannesburg 2192, South Africa
| | - Kabamba Alexandre
- Array Technology Laboratory, Synthetic Biology and Precision Medicine Centre: Next Generation Health Cluster, Council for Scientific and Industrial Research, Brummeria, Pretoria 0001, South Africa (K.A.)
| |
Collapse
|
29
|
Krishnananthasivam S, Li H, Bouzeyen R, Shunmuganathan B, Purushotorman K, Liao X, Du F, Friis CGK, Crawshay-Williams F, Boon LH, Xinlei Q, Chan CEZ, Sobota R, Kozma M, Barcelli V, Wang G, Huang H, Floto A, Bifani P, Javid B, MacAry PA. An anti-LpqH human monoclonal antibody from an asymptomatic individual mediates protection against Mycobacterium tuberculosis. NPJ Vaccines 2023; 8:127. [PMID: 37626082 PMCID: PMC10457302 DOI: 10.1038/s41541-023-00710-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 07/11/2023] [Indexed: 08/27/2023] Open
Abstract
Tuberculosis (TB) is an airborne disease caused by Mycobacterium tuberculosis (Mtb). Whilst a functional role for humoral immunity in Mtb protection remains poorly defined, previous studies have suggested that antibodies can contribute towards host defense. Thus, identifying the critical components in the antibody repertoires from immune, chronically exposed, healthy individuals represents an approach for identifying new determinants for natural protection. In this study, we performed a thorough analysis of the IgG/IgA memory B cell repertoire from occupationally exposed, immune volunteers. We detail the identification and selection of a human monoclonal antibody that exhibits protective activity in vivo and show that it targets a virulence factor LpqH. Intriguingly, protection in both human ex vivo and murine challenge experiments was isotype dependent, with most robust protection being mediated via IgG2 and IgA. These data have important implications for our understanding of natural mucosal immunity for Mtb and highlight a new target for future vaccine development.
Collapse
Affiliation(s)
- Shivankari Krishnananthasivam
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Hao Li
- College of Veterinary Medicine, China Agricultural University, Beijing, China
- Center for Infectious Disease Research, School of Medicine, Tsinghua University, Beijing, China
| | - Rania Bouzeyen
- Division of Experimental Medicine, University of California, San Francisco, USA
| | | | - Kiren Purushotorman
- Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Xinlei Liao
- National Clinical Laboratory on Tuberculosis, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing Chest Hospital, Capital Medical University, Beijing, P.R. China
| | - Fengjiao Du
- National Clinical Laboratory on Tuberculosis, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing Chest Hospital, Capital Medical University, Beijing, P.R. China
| | - Claudia Guldager Kring Friis
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Felicity Crawshay-Williams
- Molecular Immunity Unit, University of Cambridge, Department of Medicine, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Low Heng Boon
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Qian Xinlei
- Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Conrad En Zuo Chan
- National Centre for Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
- Defence Medical and Environmental Research Institute, DSO National Laboratories, Singapore, Singapore
| | - Radoslaw Sobota
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Mary Kozma
- Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Valeria Barcelli
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Guirong Wang
- National Clinical Laboratory on Tuberculosis, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing Chest Hospital, Capital Medical University, Beijing, P.R. China
| | - Hairong Huang
- National Clinical Laboratory on Tuberculosis, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing Chest Hospital, Capital Medical University, Beijing, P.R. China
| | - Andreas Floto
- Molecular Immunity Unit, University of Cambridge, Department of Medicine, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Pablo Bifani
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Babak Javid
- Center for Infectious Disease Research, School of Medicine, Tsinghua University, Beijing, China.
- Division of Experimental Medicine, University of California, San Francisco, USA.
| | - Paul A MacAry
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Life Sciences Institute, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
30
|
Aiello A, Najafi-Fard S, Goletti D. Initial immune response after exposure to Mycobacterium tuberculosis or to SARS-COV-2: similarities and differences. Front Immunol 2023; 14:1244556. [PMID: 37662901 PMCID: PMC10470049 DOI: 10.3389/fimmu.2023.1244556] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 07/31/2023] [Indexed: 09/05/2023] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb) and Coronavirus disease-2019 (COVID-19), whose etiologic agent is severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), are currently the two deadliest infectious diseases in humans, which together have caused about more than 11 million deaths worldwide in the past 3 years. TB and COVID-19 share several aspects including the droplet- and aerosol-borne transmissibility, the lungs as primary target, some symptoms, and diagnostic tools. However, these two infectious diseases differ in other aspects as their incubation period, immune cells involved, persistence and the immunopathological response. In this review, we highlight the similarities and differences between TB and COVID-19 focusing on the innate and adaptive immune response induced after the exposure to Mtb and SARS-CoV-2 and the pathological pathways linking the two infections. Moreover, we provide a brief overview of the immune response in case of TB-COVID-19 co-infection highlighting the similarities and differences of each individual infection. A comprehensive understanding of the immune response involved in TB and COVID-19 is of utmost importance for the design of effective therapeutic strategies and vaccines for both diseases.
Collapse
Affiliation(s)
| | | | - Delia Goletti
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| |
Collapse
|
31
|
Shahrear S, Islam ABMMK. Modeling of MT. P495, an mRNA-based vaccine against the phosphate-binding protein PstS1 of Mycobacterium tuberculosis. Mol Divers 2023; 27:1613-1632. [PMID: 36006502 PMCID: PMC9406248 DOI: 10.1007/s11030-022-10515-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/13/2022] [Indexed: 11/28/2022]
Abstract
Tuberculosis (TB) is a contagious disease that predominantly affects the lungs, but can also spread to other organs via the bloodstream. TB affects about one-fourth population of the world. With age, the effectiveness of Bacillus Calmette-Guérin (BCG), the only authorized TB vaccine, decreases. In the quest for a prophylactic and immunotherapeutic vaccine, in this study, a hypothetical mRNA vaccine is delineated, named MT. P495, implementing in silico and immunoinformatics approaches to evaluate key aspects and immunogenic epitopes across the PstS1, a highly conserved periplasmic protein of Mycobacterium tuberculosis (Mtb). PstS1 elicited the potential to generate 99.9% population coverage worldwide. The presence of T- and B-cell epitopes across the PstS1 protein were validated using several computational prediction tools. Molecular docking and dynamics simulation confirmed stable epitope-allele interaction. Immune cell response to the antigen clearance rate was verified by the in silico analysis of immune simulation. Codon optimization confirmed the efficient translation of the mRNA in the host cell. With Toll-like receptors, the vaccine exhibited stable and strong interactions. Findings suggest that the MT. P495 vaccine probably will elicit specific immune responses against Mtb. This mRNA vaccine model is a ready source for further wet-lab validation to confirm the efficacy of this proposed vaccine candidate.
Collapse
Affiliation(s)
- Sazzad Shahrear
- Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, 1000, Bangladesh
| | | |
Collapse
|
32
|
Dzigba P, Rylski AK, Angera IJ, Banahene N, Kavunja HW, Greenlee-Wacker MC, Swarts BM. Immune Targeting of Mycobacteria through Cell Surface Glycan Engineering. ACS Chem Biol 2023; 18:1548-1556. [PMID: 37306676 PMCID: PMC10782841 DOI: 10.1021/acschembio.3c00155] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Mycobacteria and other organisms in the order Mycobacteriales cause a range of significant human diseases, including tuberculosis, leprosy, diphtheria, Buruli ulcer, and non-tuberculous mycobacterial (NTM) disease. However, the intrinsic drug tolerance engendered by the mycobacterial cell envelope undermines conventional antibiotic treatment and contributes to acquired drug resistance. Motivated by the need to augment antibiotics with novel therapeutic approaches, we developed a strategy to specifically decorate mycobacterial cell surface glycans with antibody-recruiting molecules (ARMs), which flag bacteria for binding to human-endogenous antibodies that enhance macrophage effector functions. Mycobacterium-specific ARMs consisting of a trehalose targeting moiety and a dinitrophenyl hapten (Tre-DNPs) were synthesized and shown to specifically incorporate into outer-membrane glycolipids of Mycobacterium smegmatis via trehalose metabolism, enabling recruitment of anti-DNP antibodies to the mycobacterial cell surface. Phagocytosis of Tre-DNP-modified M. smegmatis by macrophages was significantly enhanced in the presence of anti-DNP antibodies, demonstrating proof-of-concept that our strategy can augment the host immune response. Because the metabolic pathways responsible for cell surface incorporation of Tre-DNPs are conserved in all Mycobacteriales organisms but absent from other bacteria and humans, the reported tools may be enlisted to interrogate host-pathogen interactions and develop immune-targeting strategies for diverse mycobacterial pathogens.
Collapse
Affiliation(s)
- Priscilla Dzigba
- Department of Chemistry and Biochemistry, Central Michigan University, Mount Pleasant, MI, 48859, USA
- Department of Biology, Central Michigan University, Mount Pleasant, MI, 48859, USA
- Biochemistry, Cell, and Molecular Biology Graduate Programs, Central Michigan University, Mount Pleasant, MI, 48859 United States
| | - Adrian K. Rylski
- Department of Chemistry and Biochemistry, Central Michigan University, Mount Pleasant, MI, 48859, USA
| | - Isaac J. Angera
- Department of Chemistry and Biochemistry, Central Michigan University, Mount Pleasant, MI, 48859, USA
| | - Nicholas Banahene
- Department of Chemistry and Biochemistry, Central Michigan University, Mount Pleasant, MI, 48859, USA
- Biochemistry, Cell, and Molecular Biology Graduate Programs, Central Michigan University, Mount Pleasant, MI, 48859 United States
| | - Herbert W. Kavunja
- Department of Chemistry and Biochemistry, Central Michigan University, Mount Pleasant, MI, 48859, USA
| | - Mallary C. Greenlee-Wacker
- Department of Biology, Central Michigan University, Mount Pleasant, MI, 48859, USA
- Biological Sciences Department, California Polytechnic State University, San Luis Obispo, CA, 93407, USA
| | - Benjamin M. Swarts
- Department of Chemistry and Biochemistry, Central Michigan University, Mount Pleasant, MI, 48859, USA
- Biochemistry, Cell, and Molecular Biology Graduate Programs, Central Michigan University, Mount Pleasant, MI, 48859 United States
| |
Collapse
|
33
|
Qerqez AN, Silva RP, Maynard JA. Outsmarting Pathogens with Antibody Engineering. Annu Rev Chem Biomol Eng 2023; 14:217-241. [PMID: 36917814 PMCID: PMC10330301 DOI: 10.1146/annurev-chembioeng-101121-084508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
There is growing interest in identifying antibodies that protect against infectious diseases, especially for high-risk individuals and pathogens for which no vaccine is yet available. However, pathogens that manifest as opportunistic or latent infections express complex arrays of virulence-associated proteins and are adept at avoiding immune responses. Some pathogens have developed strategies to selectively destroy antibodies, whereas others create decoy epitopes that trick the host immune system into generating antibodies that are at best nonprotective and at worst enhance pathogenesis. Antibody engineering strategies can thwart these efforts by accessing conserved neutralizing epitopes, generating Fc domains that resist capture or degradation and even accessing pathogens hidden inside cells. Design of pathogen-resistant antibodies can enhance protection and guide development of vaccine immunogens against these complex pathogens. Here, we discuss general strategies for design of antibodies resistant to specific pathogen defense mechanisms.
Collapse
Affiliation(s)
- Ahlam N Qerqez
- Department of Chemical Engineering, The University of Texas, Austin, Texas, USA;
| | - Rui P Silva
- Department of Molecular Biosciences, The University of Texas, Austin, Texas, USA
| | - Jennifer A Maynard
- Department of Chemical Engineering, The University of Texas, Austin, Texas, USA;
| |
Collapse
|
34
|
Qu M, Liang Z, Chen Y, Wang Y, Wang H, Liu Z, Liu Y, Dong Y, Ge X, Li H, Zhou X. Antibodies Targeting the Cell Wall Induce Protection against Virulent Mycobacterium bovis Infection. Microbiol Spectr 2023; 11:e0343122. [PMID: 36847491 PMCID: PMC10100962 DOI: 10.1128/spectrum.03431-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 12/11/2022] [Indexed: 03/01/2023] Open
Abstract
Accumulating evidence indicates that antibodies can protect against some intracellular pathogens. Mycobacterium bovis is an intracellular bacterium, and its cell wall (CW) is essential for its virulence and survival. However, the questions of whether antibodies play a protective role in immunity against M. bovis infection and what effects antibodies specific to the CW of M. bovis have still remain unclear. Here, we report that antibodies targeting the CW of an isolated pathogenic M. bovis strain and that of an attenuated bacillus Calmette-Guérin (BCG) strain could induce protection against virulent M. bovis infection in vitro and in vivo. Further research found that the antibody-induced protection was mainly achieved by promoting Fc gamma receptor (FcγR)-mediated phagocytosis, inhibiting bacterial intracellular growth, and enhancing the fusion of phagosomes and lysosomes, and it also depended on T cells for its efficacy. Additionally, we analyzed and characterized the B-cell receptor (BCR) repertoires of CW-immunized mice via next-generation sequencing. CW immunization stimulated BCR changes in the complementarity determining region 3 (CDR3) isotype distribution, gene usage, and somatic hypermutation. Overall, our study validates the idea that antibodies targeting the CW induce protection against virulent M. bovis infection. This study highlights the importance of antibodies targeting the CW in the defense against tuberculosis. IMPORTANCE M. bovis is the causative agent of animal tuberculosis (TB) and human TB. Research on M. bovis is of great public health significance. Currently, TB vaccines are mainly aimed at eliciting protection by enhancement of cell-mediated immunity, and there are few studies on protective antibodies. This is the first report of protective antibodies against M. bovis infection, and the antibodies had both preventive and even therapeutic effects in an M. bovis infection mouse model. Additionally, we reveal the relationship between CDR3 gene diversity and the immune characteristics of the antibodies. These results will provide valuable advice for the rational development of TB vaccines.
Collapse
Affiliation(s)
- Mengjin Qu
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Zhengmin Liang
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yulan Chen
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yuanzhi Wang
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Haoran Wang
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Ziyi Liu
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yiduo Liu
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yuhui Dong
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xin Ge
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Hao Li
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xiangmei Zhou
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
35
|
Gong W, Xie J, Li H, Aspatwar A. Editorial: Research advances of tuberculosis vaccine and its implication on COVID-19. Front Immunol 2023; 14:1147704. [PMID: 36845104 PMCID: PMC9948598 DOI: 10.3389/fimmu.2023.1147704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 01/26/2023] [Indexed: 02/11/2023] Open
Affiliation(s)
- Wenping Gong
- Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The 8Medical Center of PLA General Hospital, Beijing, China
| | - Jianping Xie
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Chongqing, China
| | - Hao Li
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Ashok Aspatwar
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| |
Collapse
|
36
|
Monkeypox infection elicits strong antibody and B cell response against A35R and H3L antigens. iScience 2023; 26:105957. [PMID: 36687315 PMCID: PMC9838220 DOI: 10.1016/j.isci.2023.105957] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/19/2022] [Accepted: 01/09/2023] [Indexed: 01/14/2023] Open
Abstract
Monkeypox virus (MPXV) resides in two forms; mature and enveloped, and depending on it, distinct proteins are displayed on the viral surface. Here, we expressed two MPXV antigens from the mature, and one from the enveloped form, and tested their reactivity to sera of 11 MPXV recoverees while comparing to sera from recently and past vaccinated individuals. 8 out of 11 recoverees exhibited detectable neutralization levels against Vaccinia Lister. Sera from all recoverees bound strongly to A35R and H3L antigens. Moreover, the responses to A35R were significantly higher within the recoverees compared to both recently and past vaccinated donors. Lastly, A35R- and H3L-specific IgG+ B cells ranging from 0.03-0.46% and 0.11-0.36%, respectively, were detected in all recoverees (A35R), and in 9 out of 11 recoverees (H3L). Therefore, A35R and H3L represent MPXV immune targets and could be used in a heat-inactivated serological ELISA for the identification of recent MPXV infection.
Collapse
|
37
|
Abstract
Computer-aided drug design (CADD) approaches are playing an increasingly important role in understanding the fundamentals of ligand-receptor interactions and helping medicinal chemists design therapeutics. About 5 years ago, we presented a chapter devoted to an overview of CADD methods and covered typical CADD protocols including structure-based drug design (SBDD) and ligand-based drug design (LBDD) approaches that were frequently used in the antibiotic drug design process. Advances in computational hardware and algorithms and emerging CADD methods are enhancing the accuracy and ability of CADD in drug design and development. In this chapter, an update to our previous chapter is provided with a focus on new CADD approaches from our laboratory and other peers that can be employed to facilitate the development of antibiotic therapeutics.
Collapse
Affiliation(s)
- Wenbo Yu
- Department of Pharmaceutical Sciences, Computer-Aided Drug Design Center, School of Pharmacy, University of Maryland, Baltimore, MD, USA.
- Institute for Bioscience and Biotechnology Research (IBBR), Rockville, MD, USA.
- Center for Biomolecular Therapeutics (CBT), School of Medicine, University of Maryland, Baltimore, MD, USA.
| | - David J Weber
- Institute for Bioscience and Biotechnology Research (IBBR), Rockville, MD, USA
- Center for Biomolecular Therapeutics (CBT), School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Alexander D MacKerell
- Department of Pharmaceutical Sciences, Computer-Aided Drug Design Center, School of Pharmacy, University of Maryland, Baltimore, MD, USA.
- Institute for Bioscience and Biotechnology Research (IBBR), Rockville, MD, USA.
- Center for Biomolecular Therapeutics (CBT), School of Medicine, University of Maryland, Baltimore, MD, USA.
| |
Collapse
|
38
|
Cheng P, Jiang F, Wang G, Wang J, Xue Y, Wang L, Gong W. Bioinformatics analysis and consistency verification of a novel tuberculosis vaccine candidate HP13138PB. Front Immunol 2023; 14:1102578. [PMID: 36825009 PMCID: PMC9942524 DOI: 10.3389/fimmu.2023.1102578] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 01/17/2023] [Indexed: 01/28/2023] Open
Abstract
Background With the increasing incidence of tuberculosis (TB) and the shortcomings of existing TB vaccines to prevent TB in adults, new TB vaccines need to be developed to address the complex TB epidemic. Method The dominant epitopes were screened from antigens to construct a novel epitope vaccine termed HP13138PB. The immune properties, structure, and function of HP13138PB were predicted and analyzed with bioinformatics and immunoinformatics. Then, the immune responses induced by the HP13138PB were confirmed by enzyme-linked immunospot assay (ELISPOT) and Th1/Th2/Th17 multi-cytokine detection kit. Result The HP13138PB vaccine consisted of 13 helper T lymphocytes (HTL) epitopes, 13 cytotoxic T lymphocytes (CTL) epitopes, and 8 B-cell epitopes. It was found that the antigenicity, immunogenicity, and solubility index of the HP13138PB vaccine were 0.87, 2.79, and 0.55, respectively. The secondary structure prediction indicated that the HP13138PB vaccine had 31% of α-helix, 11% of β-strand, and 56% of coil. The tertiary structure analysis suggested that the Z-score and the Favored region of the HP13138PB vaccine were -4.47 88.22%, respectively. Furthermore, the binding energies of the HP13138PB to toll-like receptor 2 (TLR2) was -1224.7 kcal/mol. The immunoinformatics and real-world experiments showed that the HP13138PB vaccine could induce an innate and adaptive immune response characterized by significantly higher levels of cytokines such as interferon-gamma (IFN-γ), tumor necrosis factor-α (TNF-α), interleukin-4 (IL-4), and IL-10. Conclusion The HP13138PB is a potential vaccine candidate to prevent TB, and this study preliminarily evaluated the ability of the HP13138PB to generate an immune response, providing a precursor target for developing TB vaccines.
Collapse
Affiliation(s)
- Peng Cheng
- Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The Eighth Medical Center of PLA General Hospital, Beijing, China.,Department of Geriatrics, The Eighth Medical Center of PLA General Hospital, Beijing, China
| | - Fan Jiang
- The Second Brigade of Cadet, Basic Medical School, Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Guiyuan Wang
- Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The Eighth Medical Center of PLA General Hospital, Beijing, China.,Hebei North University, Zhangjiakou, Hebei, China
| | - Jie Wang
- Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The Eighth Medical Center of PLA General Hospital, Beijing, China
| | - Yong Xue
- Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The Eighth Medical Center of PLA General Hospital, Beijing, China
| | - Liang Wang
- Department of Geriatrics, The Eighth Medical Center of PLA General Hospital, Beijing, China
| | - Wenping Gong
- Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The Eighth Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
39
|
Immune cell interactions in tuberculosis. Cell 2022; 185:4682-4702. [PMID: 36493751 DOI: 10.1016/j.cell.2022.10.025] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/15/2022] [Accepted: 10/26/2022] [Indexed: 12/13/2022]
Abstract
Despite having been identified as the organism that causes tuberculosis in 1882, Mycobacterium tuberculosis has managed to still evade our understanding of the protective immune response against it, defying the development of an effective vaccine. Technology and novel experimental models have revealed much new knowledge, particularly with respect to the heterogeneity of the bacillus and the host response. This review focuses on certain immunological elements that have recently yielded exciting data and highlights the importance of taking a holistic approach to understanding the interaction of M. tuberculosis with the many host cells that contribute to the development of protective immunity.
Collapse
|
40
|
Proteome Profile Changes Induced by Heterologous Overexpression of Mycobacterium tuberculosis-Derived Antigens PstS-1 (Rv0934) and Ag85B (Rv1886c) in Mycobacterium microti. Biomolecules 2022; 12:biom12121836. [PMID: 36551264 PMCID: PMC9775975 DOI: 10.3390/biom12121836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/01/2022] [Accepted: 11/26/2022] [Indexed: 12/13/2022] Open
Abstract
The development of new tuberculosis vaccines remains a global priority, and recombinant vaccines are a frequently investigated option. These vaccines follow a molecular strategy that may enhance protective efficacy. However, their functional differences, particularly with respect to glycosylation, remain unknown. Recent studies have shown that glycosylation plays a key role in the host-pathogen interactions during immune recognition. The aim of this study was to determine the differences in the glycosylation profiles of two recombinant strains of Mycobacterium microti, overexpressing Ag85B (Rv1886c) and PstS-1 (Rv0934) antigens of M. tuberculosis. For each strain, the glycosylation profile was determined by Western blotting with lectins. The results showed the presence of mannosylated proteins and evidence of linked sialic acid proteins. Interestingly, different proteome and glycoproteome profiles were observed between the two recombinant strains and the wild-type strain. We have shown here that the construction of the recombinant strains of M. microti has altered the proteome and glycosylation profiles of these strains, leading us to ask what impact these changes might have on the immune response.
Collapse
|
41
|
Chandra P, Grigsby SJ, Philips JA. Immune evasion and provocation by Mycobacterium tuberculosis. Nat Rev Microbiol 2022; 20:750-766. [PMID: 35879556 PMCID: PMC9310001 DOI: 10.1038/s41579-022-00763-4] [Citation(s) in RCA: 242] [Impact Index Per Article: 80.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/20/2022] [Indexed: 02/07/2023]
Abstract
Mycobacterium tuberculosis, the causative agent of tuberculosis, has infected humans for millennia. M. tuberculosis is well adapted to establish infection, persist in the face of the host immune response and be transmitted to uninfected individuals. Its ability to complete this infection cycle depends on it both evading and taking advantage of host immune responses. The outcome of M. tuberculosis infection is often a state of equilibrium characterized by immunological control and bacterial persistence. Recent data have highlighted the diverse cell populations that respond to M. tuberculosis infection and the dynamic changes in the cellular and intracellular niches of M. tuberculosis during the course of infection. M. tuberculosis possesses an arsenal of protein and lipid effectors that influence macrophage functions and inflammatory responses; however, our understanding of the role that specific bacterial virulence factors play in the context of diverse cellular reservoirs and distinct infection stages is limited. In this Review, we discuss immune evasion and provocation by M. tuberculosis during its infection cycle and describe how a more detailed molecular understanding is crucial to enable the development of novel host-directed therapies, disease biomarkers and effective vaccines.
Collapse
Affiliation(s)
- Pallavi Chandra
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, USA
| | - Steven J Grigsby
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, USA
| | - Jennifer A Philips
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA.
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, USA.
| |
Collapse
|
42
|
Singh K, Kumar R, Umam F, Kapoor P, Sinha S, Aggarwal A. Distinct and shared B cell responses of tuberculosis patients and their household contacts. PLoS One 2022; 17:e0276610. [PMID: 36282846 PMCID: PMC9595562 DOI: 10.1371/journal.pone.0276610] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/11/2022] [Indexed: 11/11/2022] Open
Abstract
This study was aimed at identifying the B cell responses which could distinguish between 'latent tuberculosis infection (LTBI)' and active TB disease. Study subjects were smear-positive TB patients (n = 54) and their disease-free household contacts (HHCs, n = 120). The sera were used for determination of antibody levels (ΔOD values) against Mycobacterium tuberculosis membrane (MtM) antigens by ELISA and for visualisation of seroreactive MtM antigens by immunoblotting. B cell subsets in whole blood samples were determined by flow cytometry. In TB sera, levels of IgG antibodies were significantly higher than IgM and IgA whereas IgM and IgA antibody levels were comparable. Conversely, HHC sera had significantly higher IgM antibody levels than IgG and IgA. The ratio of IgM to IgG antibodies in HHCs were also significantly higher than in patients. Immunoblotting revealed that some of the MtM antigens (<10, ~12 and ~25 kDa) reacted with TB as well as HHC sera whereas some other antigens (~16, ~36, ~45 and ~60 kDa) reacted with most of TB and a subset of HHC sera. Frequencies of classical memory B cells (cMBCs, CD19+CD27+) were significantly higher, and of IgG+ cMBCs were significantly lower in HHCs than in patients. Frequencies of IgA+ cMBCs in HHCs and patients were comparable but both were significantly higher than the corresponding frequencies of IgG+ cMBCs. Frequencies of IgA+ atypical MBCs (aMBCs, CD19+CD27-) in HHCs and patients were also comparable and significantly higher than the IgG+ aMBCs. The plasmablast (CD19+CD27++CD38++) frequencies in HHCs and patients were comparable. These results suggest that the IgM/IgG antibody ratio, antibody binding to selected MtM antigens and relative frequencies of MBC subsets could indicate protective or pathogenic immune responses following the primary infection with Mtb. Responses that orchestrate protection leading to a 'quiescent' LTBI may provide clues to an effective vaccination strategy against TB.
Collapse
Affiliation(s)
- Komal Singh
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Rajesh Kumar
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Fareha Umam
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Prerna Kapoor
- DOTS Centre, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Sudhir Sinha
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
- * E-mail: (AA); (SS)
| | - Amita Aggarwal
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
- * E-mail: (AA); (SS)
| |
Collapse
|
43
|
Paliwal D, Thom M, Hussein A, Ravishankar D, Wilkes A, Charleston B, Jones IM. Towards Reverse Vaccinology for Bovine TB: High Throughput Expression of Full Length Recombinant Mycobacterium bovis Proteins. Front Mol Biosci 2022; 9:889667. [PMID: 36032666 PMCID: PMC9402895 DOI: 10.3389/fmolb.2022.889667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 06/06/2022] [Indexed: 11/13/2022] Open
Abstract
Bovine tuberculosis caused by Mycobacterium bovis, is a significant global pathogen causing economic loss in livestock and zoonotic TB in man. Several vaccine approaches are in development including reverse vaccinology which uses an unbiased approach to select open reading frames (ORF) of potential vaccine candidates, produce them as recombinant proteins and assesses their immunogenicity by direct immunization. To provide feasibility data for this approach we have cloned and expressed 123 ORFs from the M. bovis genome, using a mixture of E. coli and insect cell expression. We used a concatenated open reading frames design to reduce the number of clones required and single chain fusion proteins for protein pairs known to interact, such as the members of the PPE-PE family. Over 60% of clones showed soluble expression in one or the other host and most allowed rapid purification of the tagged bTB protein from the host cell background. The catalogue of recombinant proteins represents a resource that may be suitable for test immunisations in the development of an effective bTB vaccine.
Collapse
Affiliation(s)
- Deepa Paliwal
- School of Biological Sciences, University of Reading, Reading, United Kingdom
| | | | - Areej Hussein
- School of Biological Sciences, University of Reading, Reading, United Kingdom
| | | | - Alex Wilkes
- School of Biological Sciences, University of Reading, Reading, United Kingdom
| | | | - Ian M. Jones
- School of Biological Sciences, University of Reading, Reading, United Kingdom
- *Correspondence: Ian M. Jones,
| |
Collapse
|
44
|
George E, Goswami A, Lodhiya T, Padwal P, Iyer S, Gauttam I, Sethi L, Jeyasankar S, Sharma PR, Dravid AA, Mukherjee R, Agarwal R. Immunomodulatory effect of mycobacterial outer membrane vesicles coated nanoparticles. BIOMATERIALS ADVANCES 2022; 139:213003. [PMID: 35882150 DOI: 10.1016/j.bioadv.2022.213003] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 06/21/2022] [Accepted: 06/22/2022] [Indexed: 06/15/2023]
Abstract
Tuberculosis (TB) is one of the most widely prevalent infectious diseases that cause significant mortality. Bacillus Calmette-Guérin (BCG), the current TB vaccine used in clinics, shows variable efficacy and has safety concerns for immunocompromised patients. There is a need to develop new and more effective TB vaccines. Outer membrane vesicles (OMVs) are vesicles released by Mycobacteria that contain several lipids and membrane proteins and act as a good source of antigens to prime immune response. However, the use of OMVs as vaccines has been hampered by their heterogeneous size and low stability. Here we report that mycobacterial OMVs can be stabilized by coating over uniform-sized 50 nm gold nanoparticles. The OMV-coated gold nanoparticles (OMV-AuNP) show enhanced uptake and activation of macrophages and dendritic cells. Proteinase K and TLR inhibitor studies demonstrated that the enhanced activation was attributed to proteins present on OMVs and was mediated primarily by TLR2 and TLR4. Mass spectrometry analysis revealed several potential membrane proteins that were common in both free OMVs and OMV-AuNP. Such strategies may open up new avenues and the utilization of novel antigens for developing TB vaccines.
Collapse
Affiliation(s)
- Edna George
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | - Avijit Goswami
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | - Tejan Lodhiya
- Indian Institute of Science Education and Research, Tirupati, India
| | - Priyanka Padwal
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | - Shalini Iyer
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | - Iti Gauttam
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | - Lakshay Sethi
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | - Sharumathi Jeyasankar
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | - Pallavi Raj Sharma
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | - Ameya Atul Dravid
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | - Raju Mukherjee
- Indian Institute of Science Education and Research, Tirupati, India
| | - Rachit Agarwal
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India.
| |
Collapse
|
45
|
Bouzeyen R, Javid B. Therapeutic Vaccines for Tuberculosis: An Overview. Front Immunol 2022; 13:878471. [PMID: 35812462 PMCID: PMC9263712 DOI: 10.3389/fimmu.2022.878471] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 05/26/2022] [Indexed: 11/13/2022] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis is the world’s deadliest bacterial infection, resulting in more than 1.4 million deaths annually. The emergence of drug-resistance to first-line antibiotic therapy poses a threat to successful treatment, and novel therapeutic options are required, particularly for drug-resistant tuberculosis. One modality emerging for TB treatment is therapeutic vaccination. As opposed to preventative vaccination – the aim of which is to prevent getting infected by M. tuberculosis or developing active tuberculosis, the purpose of therapeutic vaccination is as adjunctive treatment of TB or to prevent relapse following cure. Several candidate therapeutic vaccines, using killed whole-cell or live attenuated mycobacteria, mycobacterial fragments and viral vectored vaccines are in current clinical trials. Other modes of passive immunization, including monoclonal antibodies directed against M. tuberculosis antigens are in various pre-clinical stages of development. Here, we will discuss these various therapeutics and their proposed mechanisms of action. Although the full clinical utility of therapeutic vaccination for the treatment of tuberculosis is yet to be established, they hold potential as useful adjunct therapies.
Collapse
|
46
|
Identification of the Bartonella autotransporter CFA as a protective antigen and hypervariable target of neutralizing antibodies in mice. Proc Natl Acad Sci U S A 2022; 119:e2202059119. [PMID: 35714289 PMCID: PMC9231624 DOI: 10.1073/pnas.2202059119] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Bartonella infections represent a significant burden to human health and are difficult to cure. Protective Bartonella vaccines are not available. Acquired immunity to Bartonella infection could provide a blueprint for vaccine design but remains incompletely defined. Moreover, bacterial immune evasion mechanisms have the potential to thwart vaccination efforts. Our study in a model of a natural Bartonella–host relationship revealed that antibody-mediated prevention of bacterial attachment to erythrocytes is sufficient for protection. We identified the bacterial surface determinant CFA (CAMP-like factor autotransporter) as a target of protective antibodies. While immunization with CFA protected against challenge with the homologous Bartonella isolate, extensive variability of CFA already at the strain level revealed bacterial immune evasion mechanisms with implications for Bartonella vaccine design. The bacterial genus Bartonella comprises numerous emerging pathogens that cause a broad spectrum of disease manifestations in humans. The targets and mechanisms of the anti-Bartonella immune defense are ill-defined and bacterial immune evasion strategies remain elusive. We found that experimentally infected mice resolved Bartonella infection by mounting antibody responses that neutralized the bacteria, preventing their attachment to erythrocytes and suppressing bacteremia independent of complement or Fc receptors. Bartonella-neutralizing antibody responses were rapidly induced and depended on CD40 signaling but not on affinity maturation. We cloned neutralizing monoclonal antibodies (mAbs) and by mass spectrometry identified the bacterial autotransporter CFA (CAMP-like factor autotransporter) as a neutralizing antibody target. Vaccination against CFA suppressed Bartonella bacteremia, validating CFA as a protective antigen. We mapped Bartonella-neutralizing mAb binding to a domain in CFA that we found is hypervariable in both human and mouse pathogenic strains, indicating mutational antibody evasion at the Bartonella subspecies level. These insights into Bartonella immunity and immune evasion provide a conceptual framework for vaccine development, identifying important challenges in this endeavor.
Collapse
|
47
|
Nziza N, Cizmeci D, Davies L, Irvine EB, Jung W, Fenderson BA, de Kock M, Hanekom WA, Franken KLMC, Day CL, Ottenhoff THM, Alter G. Defining Discriminatory Antibody Fingerprints in Active and Latent Tuberculosis. Front Immunol 2022; 13:856906. [PMID: 35514994 PMCID: PMC9066635 DOI: 10.3389/fimmu.2022.856906] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 03/11/2022] [Indexed: 01/10/2023] Open
Abstract
Tuberculosis (TB) is among the leading causes of death worldwide from a single infectious agent, second only to COVID-19 in 2020. TB is caused by infection with Mycobacterium tuberculosis (Mtb), that results either in a latent or active form of disease, the latter associated with Mtb spread. In the absence of an effective vaccine, epidemiologic modeling suggests that aggressive treatment of individuals with active TB (ATB) may curb spread. Yet, clinical discrimination between latent (LTB) and ATB remains a challenge. While antibodies are widely used to diagnose many infections, the utility of antibody-based tests to diagnose ATB has only regained significant traction recently. Specifically, recent interest in the humoral immune response to TB has pointed to potential differences in both targeted antigens and antibody features that can discriminate latent and active TB. Here we aimed to integrate these observations and broadly profile the humoral immune response across individuals with LTB or ATB, with and without HIV co-infection, to define the most discriminatory humoral properties and diagnose TB disease more easily. Using 209 Mtb antigens, striking differences in antigen-recognition were observed across latently and actively infected individuals that was modulated by HIV serostatus. However, ATB and LTB could be discriminated, irrespective of HIV-status, based on a combination of both antibody levels and Fc receptor-binding characteristics targeting both well characterized (like lipoarabinomannan, 38 kDa or antigen 85) but also novel Mtb antigens (including Rv1792, Rv1528, Rv2435C or Rv1508). These data reveal new Mtb-specific immunologic markers that can improve the classification of ATB versus LTB.
Collapse
Affiliation(s)
- Nadege Nziza
- Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
| | - Deniz Cizmeci
- Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
| | - Leela Davies
- Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
- Division of Infectious Diseases, Brigham and Women’s Hospital, Boston, MA, United States
| | - Edward B. Irvine
- Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Wonyeong Jung
- Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
| | - Brooke A. Fenderson
- Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
| | - Marwou de Kock
- South African Tuberculosis Vaccine Initiative (SATVI) and School of Child and Adolescent Health, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Willem A. Hanekom
- Africa Health Research Institute, Durban, South Africa
- Division of Infection and Immunity, University College London, London, United Kingdom
| | | | - Cheryl L. Day
- Department of Microbiology and Immunology, Emory University School of Medicine, Emory University, Atlanta, GA, United States
| | | | - Galit Alter
- Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
| |
Collapse
|
48
|
Patangia DV, Anthony Ryan C, Dempsey E, Paul Ross R, Stanton C. Impact of antibiotics on the human microbiome and consequences for host health. Microbiologyopen 2022; 11:e1260. [PMID: 35212478 PMCID: PMC8756738 DOI: 10.1002/mbo3.1260] [Citation(s) in RCA: 327] [Impact Index Per Article: 109.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/10/2021] [Accepted: 12/10/2021] [Indexed: 12/12/2022] Open
Abstract
It is well established that the gut microbiota plays an important role in host health and is perturbed by several factors including antibiotics. Antibiotic-induced changes in microbial composition can have a negative impact on host health including reduced microbial diversity, changes in functional attributes of the microbiota, formation, and selection of antibiotic-resistant strains making hosts more susceptible to infection with pathogens such as Clostridioides difficile. Antibiotic resistance is a global crisis and the increased use of antibiotics over time warrants investigation into its effects on microbiota and health. In this review, we discuss the adverse effects of antibiotics on the gut microbiota and thus host health, and suggest alternative approaches to antibiotic use.
Collapse
Affiliation(s)
- Dhrati V. Patangia
- School of MicrobiologyUniversity College CorkCorkIreland
- Teagasc Food Research Centre, MooreparkFermoy Co.CorkIreland
- APC MicrobiomeCorkIreland
| | | | - Eugene Dempsey
- School of MicrobiologyUniversity College CorkCorkIreland
| | - Reynolds Paul Ross
- School of MicrobiologyUniversity College CorkCorkIreland
- APC MicrobiomeCorkIreland
| | - Catherine Stanton
- Teagasc Food Research Centre, MooreparkFermoy Co.CorkIreland
- APC MicrobiomeCorkIreland
| |
Collapse
|
49
|
Qu M, Zhou X, Li H. BCG vaccination strategies against tuberculosis: updates and perspectives. Hum Vaccin Immunother 2021; 17:5284-5295. [PMID: 34856853 DOI: 10.1080/21645515.2021.2007711] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Bacillus Calmette-Guérin (BCG) is the only licensed vaccine against tuberculosis (TB). However, BCG has variable efficacy and cannot completely prevent TB infection and transmission. Therefore, the worldwide prevalence of TB calls for urgent development of a more effective TB vaccine. In the absence of other approved vaccines, it is also necessary to improve the efficacy of BCG itself. Intravenous (IV) BCG administration and BCG revaccination strategies have recently shown promising results for clinical usage. Therefore, it is necessary for us to revisit the BCG vaccination strategies and summarize the current research updates related to BCG vaccination. This literature review provides an updated overview and perspectives of the immunization strategies against TB using BCG, which may inspire the following research on TB vaccine development.
Collapse
Affiliation(s)
- Mengjin Qu
- College of Veterinary Medicine, China Agricultural University, Beijing, China.,Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, China Agricultural University, Beijing, China
| | - Xiangmei Zhou
- College of Veterinary Medicine, China Agricultural University, Beijing, China.,Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, China Agricultural University, Beijing, China
| | - Hao Li
- College of Veterinary Medicine, China Agricultural University, Beijing, China.,Center for Infectious Disease Research, School of Medicine, Tsinghua University, Beijing, China
| |
Collapse
|
50
|
Irvine EB, O'Neil A, Darrah PA, Shin S, Choudhary A, Li W, Honnen W, Mehra S, Kaushal D, Gideon HP, Flynn JL, Roederer M, Seder RA, Pinter A, Fortune S, Alter G. Robust IgM responses following intravenous vaccination with Bacille Calmette-Guérin associate with prevention of Mycobacterium tuberculosis infection in macaques. Nat Immunol 2021; 22:1515-1523. [PMID: 34811542 PMCID: PMC8642241 DOI: 10.1038/s41590-021-01066-1] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 10/04/2021] [Indexed: 01/31/2023]
Abstract
Development of an effective tuberculosis (TB) vaccine has suffered from an incomplete understanding of the correlates of protection against Mycobacterium tuberculosis (Mtb). Intravenous (i.v.) vaccination with Bacille Calmette-Guérin (BCG) provides nearly complete protection against TB in rhesus macaques, but the antibody response it elicits remains incompletely defined. Here we show that i.v. BCG drives superior antibody responses in the plasma and the lungs of rhesus macaques compared to traditional intradermal BCG administration. While i.v. BCG broadly expands antibody titers and functions, IgM titers in the plasma and lungs of immunized macaques are among the strongest markers of reduced bacterial burden. IgM was also enriched in macaques that received protective vaccination with an attenuated strain of Mtb. Finally, an Mtb-specific IgM monoclonal antibody reduced Mtb survival in vitro. Collectively, these data highlight the potential importance of IgM responses as a marker and mediator of protection against TB.
Collapse
Affiliation(s)
- Edward B Irvine
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Anthony O'Neil
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Patricia A Darrah
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD, USA
| | - Sally Shin
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Alok Choudhary
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Wenjun Li
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - William Honnen
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Smriti Mehra
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA, USA
| | - Deepak Kaushal
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Hannah Priyadarshini Gideon
- Department of Microbiology and Molecular Genetics and Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - JoAnne L Flynn
- Department of Microbiology and Molecular Genetics and Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD, USA
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD, USA
| | - Abraham Pinter
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Sarah Fortune
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|