1
|
Goig GA, Windels EM, Loiseau C, Stritt C, Biru L, Borrell S, Brites D, Gagneux S. Ecology, global diversity and evolutionary mechanisms in the Mycobacterium tuberculosis complex. Nat Rev Microbiol 2025:10.1038/s41579-025-01159-w. [PMID: 40133503 DOI: 10.1038/s41579-025-01159-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/03/2025] [Indexed: 03/27/2025]
Abstract
With the COVID-19 pandemic receding, tuberculosis (TB) is again the number one cause of human death to a single infectious agent. TB is caused by bacteria that belong to the Mycobacterium tuberculosis complex (MTBC). Recent advances in genome sequencing have provided new insights into the ecology and evolution of the MTBC. This includes the discovery of new phylogenetic lineages within the MTBC, a deeper understanding of the host tropism among the various animal-adapted lineages, enhanced knowledge on the evolutionary dynamics of antimicrobial resistance and transmission, as well as a better grasp of the within-host MTBC diversity. Moreover, advances in long-read sequencing are increasingly highlighting the relevance of structural genomic variation in the MTBC. These findings not only shed new light on the biology and epidemiology of TB, but also give rise to new questions and research avenues. The purpose of this Review is to summarize these new insights and discuss their implications for global TB control.
Collapse
Affiliation(s)
- Galo A Goig
- Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Etthel M Windels
- Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
- Swiss Institute for Bioinformatics, Lausanne, Switzerland
| | - Chloé Loiseau
- Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Christoph Stritt
- Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Loza Biru
- Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Sonia Borrell
- Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Daniela Brites
- Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Sebastien Gagneux
- Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Allschwil, Switzerland.
- University of Basel, Basel, Switzerland.
| |
Collapse
|
2
|
Hoerter A, Petrucciani A, Bonifacio J, Arnett E, Schlesinger LS, Pienaar E. Timing matters in macrophage/CD4+ T cell interactions: an agent-based model comparing Mycobacterium tuberculosis host-pathogen interactions between latently infected and naïve individuals. mSystems 2025; 10:e0129024. [PMID: 39918314 PMCID: PMC11915833 DOI: 10.1128/msystems.01290-24] [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: 10/04/2024] [Accepted: 12/17/2024] [Indexed: 03/19/2025] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), remains a significant health challenge. Clinical manifestations of TB exist across a spectrum with a majority of infected individuals remaining asymptomatic, commonly referred to as latent TB infection (LTBI). In vitro models have demonstrated that cells from individuals with LTBI can better control Mtb growth and form granuloma-like structures more quickly, compared to cells from uninfected (Mtb-naïve) individuals. These in vitro results agree with animal and clinical evidence that LTBI protects, to some degree, against reinfection. However, the mechanisms by which LTBI might offer protection against reinfection remain unclear, and quantifying the relative contributions of multiple control mechanisms is challenging using experimental methods alone. To complement in vitro models, we have developed an in silico agent-based model to help elucidate host responses that might contribute to protection against reinfection. Our simulations indicate that earlier contact between macrophages and CD4+ T cells leads to LTBI simulations having more activated CD4+ T cells and, in turn, more activated infected macrophages, all of which contribute to a decreased bacterial load early on. Our simulations also demonstrate that granuloma-like structures support this early macrophage activation in LTBI simulations. We find that differences between LTBI and Mtb-naïve simulations are driven by TNFα and IFNγ-associated mechanisms as well as macrophage phagocytosis and killing mechanisms. Together, our simulations show how important the timing of the first interactions between innate and adaptive immune cells is, how this impacts infection progression, and why cells from LTBI individuals might be faster to respond to reinfection.IMPORTANCETuberculosis (TB) remains a significant global health challenge, with millions of new infections and deaths annually. Despite extensive research, the mechanisms by which latent TB infection (LTBI) confers protection against reinfection remain unclear. In this study, we developed an in silico agent-based model to simulate early immune responses to Mycobacterium tuberculosis infection based on experimental in vitro infection of human donor cells. Our simulations reveal that early interactions between macrophages and CD4+ T cells, driven by TNFα and IFNγ, are critical for bacterial control and granuloma formation in LTBI. These findings offer new insights into the immune processes involved in TB, which could inform the development of targeted vaccines and host-directed therapies. By integrating experimental data with computational predictions, our research provides a robust framework for understanding TB immunity and guiding future interventions to mitigate the global TB burden.
Collapse
Affiliation(s)
- Alexis Hoerter
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
| | - Alexa Petrucciani
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
| | | | - Eusondia Arnett
- Texas Biomedical Research Institute, San Antonio, Texas, USA
| | | | - Elsje Pienaar
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
- Regenstrief Center for Healthcare Engineering, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
3
|
Yabaji SM, Lata S, Tseng AE, Araveti PB, Lo M, Gavrish I, O’Connell AK, Gertje HP, Belkina AC, Thurman CE, Kiyokawa H, Kotton D, Tan S, Endsley JJ, Bishai WR, Crossland N, Kobzik L, Kramnik I. Aberrant macrophage activation and maladaptive lung repair promote tuberculosis progression uniquely in the lung. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.10.17.562695. [PMID: 40093068 PMCID: PMC11908135 DOI: 10.1101/2023.10.17.562695] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Pulmonary tuberculosis (PTB) represents 85% of the disease burden caused by Mycobacterium tuberculosis (Mtb) and promotes aerosol transmission infecting about a quarter of people globally. Most Mtb infections are effectively limited within primary granulomatous lesions. Containment failures lead to hematogenous spread and the formation of post-primary destructive PTB lesions. Factors that favor Mtb survival and replication in the lungs after hematogenous spread despite systemic immunity represent appealing targets for host-directed TB therapies, but are currently unknown. We developed a novel mouse model that mimics progression of chronic post-primary PTB in humans: wherein PTB lesions form after hematogenous spread from a remote primary lesion in immunocompetent but TB-susceptible B6.Sst1S mice. The B6.Sst1S mice developed PTB lesions featuring granulomatous pneumonia, bronchogenic expansion and broncho-occlusion closely resembling post-primary PTB in humans. Using spatial transcriptomic and fluorescent multiplexed immunochemistry, we demonstrated the expansion of myeloid cell populations with the appearance of alternatively activated macrophages, dissolution of initial lymphoid follicles, and accumulation of de-differentiated lung epithelial cells in the advanced PTB lesions. To determine whether lung parenchymal cells or lung oxygenation were necessary for the pulmonary TB progression, we implanted lung and spleen fragments subcutaneously to serve as potential targets for hematogenous spread. The lung (but not spleen) implants displayed characteristic organized granulomas with necrosis and Mtb replication demonstrating that deleterious interactions of aberrantly activated macrophages with the inflammation-injured lung resident cells, and possibly hypoxia, not oxygenation, are critical determinants of PTB progression in immunocompetent hosts. Necrotic TB lesions also developed in subcutaneous implants of human lung tissue in mice with human immune system after respiratory infection. These animal models may serve to further dissect the lung-specific mechanisms of host susceptibility to virulent Mtb and for testing therapeutic interventions targeting these mechanisms.
Collapse
Affiliation(s)
- Shivraj M. Yabaji
- The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University
| | - Suruchi Lata
- The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University
| | - Anna E. Tseng
- The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University
| | | | - Ming Lo
- The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University
| | - Igor Gavrish
- The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University
| | - Aoife K O’Connell
- The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University
| | - Hans P Gertje
- The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University
| | - Anna C. Belkina
- The Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118
- Flow Cytometry Core Facility, Boston University School of Medicine, Boston, MA, USA
| | - Colleen E Thurman
- The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University
| | - Hirofumi Kiyokawa
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center and Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Darrell Kotton
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center and Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Shumin Tan
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA
| | - Janice J. Endsley
- Departments of Microbiology & Immunology and Pathology, The University of Texas Medical Branch, Galveston, TX 77555, USA
| | - William R Bishai
- Center for Tuberculosis Research School of Medicine, John Hopkins University Baltimore, Maryland
| | - Nicholas Crossland
- The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University
- The Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118
| | | | - Igor Kramnik
- The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University
- Pulmonary Center, The Department of Medicine, Boston University Chobanian & Aveedisian School of Medicine
| |
Collapse
|
4
|
Nieto Ramirez LM, Mehaffy C, Dobos KM. Systematic review of innate immune responses against Mycobacterium tuberculosis complex infection in animal models. Front Immunol 2025; 15:1467016. [PMID: 39949719 PMCID: PMC11821578 DOI: 10.3389/fimmu.2024.1467016] [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: 07/19/2024] [Accepted: 12/27/2024] [Indexed: 02/16/2025] Open
Abstract
Background Mycobacterium tuberculosis (Mtb) complex (MTBC) includes ten species that affect mammals and pose a significant global health concern. Upon infection, Mtb induces various stages in the host, including early bacterial elimination, which may or may not involve memory responses. Deciphering the role of innate immune responses during MTBC infection is crucial for understanding disease progression or protection. Over the past decade, there has been growing interest in the innate immune response to Mtb, with new preclinical models emerging. Methods We conducted a systematic review following PRISMA guidelines, focused on innate immune mediators linked to protection or disease progression in animal models of MTBC infection. We searched two databases: National Library of Medicine and Web of Science. Two researchers independently extracted data based on specific inclusion and exclusion criteria. Results Eighty-three articles were reviewed. Results were categorized in four groups: MTBC species, animal models, soluble factors and innate pathways, and other molecules (metabolites and drugs). Mtb and M. bovis were the only species studied. P2X7R receptor's role in disease progression and higher macrophage recruitment were observed differentially after infection with hypervirulent Mtb strains. Mice and non-human primates (NHPs) were the most used mammals, with emerging models like Galleria mellonella and planarians also studied. NHPs provided insights into age-dependent immunity and markers for active tuberculosis (ATB). Key innate immune factors/pathways identified included TNF-α, neutrophil recruitment, ROS/RNS responses, autophagy, inflammasomes, and antimicrobial peptides, with homologous proteins identified in insects. Metabolites like vitamin B5 and prostaglandin E2 were associated with protection. Immunomodulatory drugs targeting autophagy and other mechanisms were studied, exhibiting their potential as therapeutic alternatives. Conclusion Simpler, physiologically relevant, and ethically sound models, such as G. mellonella, are needed for studying innate responses in MTBC infection. While insects lack adaptive immunity, they could provide insights into "pure" innate immune responses. The dissection of "pure," "sustained" (later than 7 days post-infection), and trained innate immunity presents additional challenges that require high-resolution temporospatial analytical methods. Identifying early innate immune mediators and targetable pathways in the blood and affected tissues could identify biomarkers for immunization efficiency, disease progression, and potential synergistic therapies for ATB.
Collapse
Affiliation(s)
- Luisa Maria Nieto Ramirez
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, United States
| | | | - Karen Marie Dobos
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|
5
|
Miranda-Hernandez S, Kumar M, Henderson A, Graham E, Tan X, Taylor J, Meehan M, Ceja Z, Del Pozo-Ramos L, Pan Y, Tsui E, Donovan ML, Rentería ME, Flores-Valdez MA, Blumenthal A, Nguyen Q, Subbian S, Field MA, Kupz A. CD8 + T cells mediate vaccination-induced lymphatic containment of latent Mycobacterium tuberculosis infection following immunosuppression, while B cells are dispensable. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.23.634479. [PMID: 39896630 PMCID: PMC11785187 DOI: 10.1101/2025.01.23.634479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
It is estimated that two billion people are latently infected with Mycobacterium tuberculosis ( Mtb ), the causative agent of tuberculosis (TB). Latent Mtb infection (LTBI) can occur in multiple organs, including the lymphatics. The risk of LTBI reactivation increases in immunocompromised conditions, such as coinfection with human immunodeficiency virus (HIV), and during treatment of autoimmune diseases and organ transplantation. The immunological correlates of protection against TB, including against reactivation of LTBI, remain largely elusive. Here, we used a mouse model of latent lymphatic Mtb infection to dissect the immunological mechanisms underlying LTBI containment versus reactivation. We show that immunosuppression-mediated reactivation of lymphatic LTBI and the subsequent spread to non-lymphatic organs can be prevented by vaccination with multiple recombinant BCG (rBCG) strains despite the deficiency of CD4 + T cells. Using spatial transcriptomics, multi-parameter imaging, network analysis and bioinformatic integration of histopathological images, we reveal that immunosuppression is associated with a distinct repositioning of non-CD4 immune cells at the edge of TB lesions within the infection-draining cervical lymph nodes. While B cells increased in numbers, they are dispensable for the containment of LTBI. Lymphatic Mtb infection in different immune cell-deficient mouse strains, antibody-mediated cell depletion and adoptive transfer experiments into highly susceptible mice unequivocally show that vaccination-mediated prevention of LTBI reactivation is critically dependent on CD8 + T cells. These findings have profound implications for our understanding of immunity to TB and the management of LTBI.
Collapse
|
6
|
Russell DG, Simwela NV, Mattila JT, Flynn J, Mwandumba HC, Pisu D. How macrophage heterogeneity affects tuberculosis disease and therapy. Nat Rev Immunol 2025:10.1038/s41577-024-01124-3. [PMID: 39774813 DOI: 10.1038/s41577-024-01124-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2024] [Indexed: 01/11/2025]
Abstract
Macrophages are the primary host cell type for infection by Mycobacterium tuberculosis in vivo. Macrophages are also key immune effector cells that mediate the control of bacterial growth. However, the specific macrophage phenotypes that are required for optimal immune control of M. tuberculosis infection in vivo remain poorly defined. There are two distinct macrophage lineages in the lung, comprising embryonically derived, tissue-resident alveolar macrophages and recruited, blood monocyte-derived interstitial macrophages. Recent studies have shown that these lineages respond divergently to similar immune environments within the tuberculosis granuloma. Here, we discuss how the differing responses of macrophage lineages might affect the control or progression of tuberculosis disease. We suggest that the ability to reprogramme macrophage responses appropriately, through immunological or chemotherapeutic routes, could help to optimize vaccines and drug regimens for tuberculosis.
Collapse
Affiliation(s)
- David G Russell
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA.
| | - Nelson V Simwela
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Joshua T Mattila
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - JoAnne Flynn
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Henry C Mwandumba
- Malawi Liverpool Wellcome Research Programme, Kamuzu University of Health Sciences, Blantyre, Malawi
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Davide Pisu
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
- Department of Microbial Pathogenesis and Immunology, Texas A&M School of Medicine, Bryan, TX, USA
| |
Collapse
|
7
|
Peters JM, Irvine EB, Makatsa MS, Rosenberg JM, Wadsworth MH, Hughes TK, Sutton MS, Nyquist SK, Bromley JD, Mondal R, Roederer M, Seder RA, Darrah PA, Alter G, Seshadri C, Flynn JL, Shalek AK, Fortune SM, Bryson BD. High-dose intravenous BCG vaccination induces enhanced immune signaling in the airways. SCIENCE ADVANCES 2025; 11:eadq8229. [PMID: 39742484 PMCID: PMC11694782 DOI: 10.1126/sciadv.adq8229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 11/20/2024] [Indexed: 01/03/2025]
Abstract
Intradermal Bacillus Calmette-Guérin (BCG) is the most widely administered vaccine, but it does not sufficiently protect adults against pulmonary tuberculosis. Recent studies in nonhuman primates show that intravenous BCG administration offers superior protection against Mycobacterium tuberculosis (Mtb). We used single-cell analysis of bronchoalveolar lavage cells from rhesus macaques vaccinated via different routes and doses of BCG to identify alterations in the immune ecosystem in the airway following vaccination. Our findings reveal that high-dose intravenous BCG induces an influx of polyfunctional T cells and macrophages in the airways, with alveolar macrophages from high-dose intravenous BCG displaying a basal activation state in the absence of purified protein derivative stimulation, defined in part by interferon signaling. Enhanced intercellular immune signaling and stronger T helper 1-T helper 17 transcriptional responses were observed following purified protein derivative stimulation. These results suggest that high-dose intravenous BCG vaccination creates a specialized immune environment that primes airway cells for effective Mtb clearance.
Collapse
Affiliation(s)
- Joshua M. Peters
- Department of Biological Engineering, MIT, Cambridge, MA, USA
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Edward B. Irvine
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Mohau S. Makatsa
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Jacob M. Rosenberg
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Division of Infectious Diseases, MGH, Boston, MA, USA
| | - Marc H. Wadsworth
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering & Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
| | - Travis K. Hughes
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering & Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
| | | | - Sarah K. Nyquist
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering & Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
| | - Joshua D. Bromley
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering & Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
| | - Rajib Mondal
- Research Laboratory of Electronics, Harvard-MIT Program in Health Sciences and Technology, Cambridge, MA, USA
| | | | | | | | - Galit Alter
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA, USA
| | - Chetan Seshadri
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - JoAnne L. Flynn
- Department of Microbiology and Molecular Genetics and Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Alex K. Shalek
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering & Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
| | - Sarah M. Fortune
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Bryan D. Bryson
- Department of Biological Engineering, MIT, Cambridge, MA, USA
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA, USA
| |
Collapse
|
8
|
Joy N, Deshpande A, Lingamallu SM, Prabantu VM, Naveenkumar CN, Bharathkumar K, Bhat S, Alvarado-Martinez Z, Livraghi-Butrico A, Hagood JS, Boucher RC, Lafkas D, Byrd KM, Narayanan S, Shandil RK, Guha A. Notch signaling stabilizes lengths of motile cilia in multiciliated cells in the lung. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.12.628112. [PMID: 39713474 PMCID: PMC11661201 DOI: 10.1101/2024.12.12.628112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Airway multiciliated cells (MCs) maintain respiratory health by clearing mucus and trapped particles through the beating of motile cilia. While it is known that ciliary lengths decrease along the proximal-distal (P-D) axis of the tracheobronchial tree, how this is regulated is unclear. Here, we demonstrate that canonical Notch signaling in MCs plays a critical role in stabilizing ciliary length. Inhibition of Notch signaling in MCs results in ciliary shortening in the trachea, lengthening in the distal airway, and to region-specific alterations in gene expression. We probe how environmental challenges impact MC homeostasis using germ-free and Mycobacterium tuberculosis ( M. tb ) infection models. While germ-free conditions do not perturb ciliary lengths, M. tb infection leads to lengthening of distal airway cilia, correlating with a downregulation of Notch signaling. These findings reveal that ciliary length and the P-D gradient in the airways are actively regulated, with Notch signaling serving as a stabilizing mechanism.
Collapse
|
9
|
Lyu J, Narum DE, Baldwin SL, Larsen SE, Bai X, Griffith DE, Dartois V, Naidoo T, Steyn AJC, Coler RN, Chan ED. Understanding the development of tuberculous granulomas: insights into host protection and pathogenesis, a review in humans and animals. Front Immunol 2024; 15:1427559. [PMID: 39717773 PMCID: PMC11663721 DOI: 10.3389/fimmu.2024.1427559] [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: 05/04/2024] [Accepted: 11/18/2024] [Indexed: 12/25/2024] Open
Abstract
Granulomas, organized aggregates of immune cells which form in response to Mycobacterium tuberculosis (Mtb), are characteristic but not exclusive of tuberculosis (TB). Despite existing investigations on TB granulomas, the determinants that differentiate host-protective granulomas from granulomas that contribute to TB pathogenesis are often disputed. Thus, the goal of this narrative review is to help clarify the existing literature on such determinants. We adopt the a priori view that TB granulomas are host-protective organelles and discuss the molecular and cellular determinants that induce protective granulomas and those that promote their failure. While reports about protective TB granulomas and their failure may initially seem contradictory, it is increasingly recognized that either deficiencies or excesses of the molecular and cellular components in TB granuloma formation may be detrimental to the host. More specifically, insufficient or excessive expression/representation of the following components have been reported to skew granulomas toward the less protective phenotype: (i) epithelioid macrophages; (ii) type 1 adaptive immune response; (iii) type 2 adaptive immune response; (iv) tumor necrosis factor; (v) interleukin-12; (vi) interleukin-17; (vii) matrix metalloproteinases; (viii) hypoxia in the TB granulomas; (ix) hypoxia inducible factor-1 alpha; (x) aerobic glycolysis; (xi) indoleamine 2,3-dioxygenase activity; (xii) heme oxygenase-1 activity; (xiii) immune checkpoint; (xiv) leukotriene A4 hydrolase activity; (xv) nuclear-factor-kappa B; and (xvi) transforming growth factor-beta. Rather, more precise and timely coordinated immune responses appear essential for eradication or containment of Mtb infection. Since there are several animal models of infection with Mtb, other species within the Mtb complex, and the surrogate Mycobacterium marinum - whether natural (cattle, elephants) or experimental (zebrafish, mouse, guinea pig, rabbit, mini pig, goat, non-human primate) infections - we also compared the TB granulomatous response and other pathologic lung lesions in various animals infected with one of these mycobacteria with that of human pulmonary TB. Identifying components that dictate the formation of host-protective granulomas and the circumstances that result in their failure can enhance our understanding of the macrocosm of human TB and facilitate the development of novel remedies - whether they be direct therapeutics or indirect interventions - to efficiently eliminate Mtb infection and prevent its pathologic sequelae.
Collapse
Affiliation(s)
- Jiwon Lyu
- Division of Pulmonary and Critical Medicine, Soon Chun Hyang University Cheonan Hospital, Seoul, Republic of Korea
- Department of Academic Affairs, National Jewish Health, Denver, CO, United States
| | - Drew E. Narum
- Department of Academic Affairs, National Jewish Health, Denver, CO, United States
| | - Susan L. Baldwin
- Center for Global Infectious Diseases, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Sasha E. Larsen
- Center for Global Infectious Diseases, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Xiyuan Bai
- Department of Academic Affairs, National Jewish Health, Denver, CO, United States
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO, United States
| | - David E. Griffith
- Department of Medicine, National Jewish Health, Denver, CO, United States
| | - Véronique Dartois
- Center for Discovery and Innovation, Hackensack Meridian School of Medicine, Nutley, NJ, United States
| | - Threnesan Naidoo
- Departments of Forensic & Legal Medicine and Laboratory Medicine & Pathology, Faculty of Medicine & Health Sciences, Walter Sisulu University, Mthatha, South Africa
| | - Adrie J. C. Steyn
- Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
- Department of Microbiology and Centers for AIDS Research and Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Rhea N. Coler
- Center for Global Infectious Diseases, Seattle Children’s Research Institute, 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
| | - Edward D. Chan
- Department of Academic Affairs, National Jewish Health, Denver, CO, United States
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO, United States
- Department of Medicine, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, United States
| |
Collapse
|
10
|
Wang J, Fan XY, Hu Z. Immune correlates of protection as a game changer in tuberculosis vaccine development. NPJ Vaccines 2024; 9:208. [PMID: 39478007 PMCID: PMC11526030 DOI: 10.1038/s41541-024-01004-w] [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: 07/04/2024] [Accepted: 10/18/2024] [Indexed: 11/02/2024] Open
Abstract
The absence of validated correlates of protection (CoPs) hampers the rational design and clinical development of new tuberculosis vaccines. In this review, we provide an overview of the potential CoPs in tuberculosis vaccine research. Major hindrances and potential opportunities are then discussed. Based on recent progress, it is reasonable to anticipate that success in the ongoing efforts to identify CoPs would be a game-changer in tuberculosis vaccine development.
Collapse
Affiliation(s)
- Jing Wang
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai, 201508, China
| | - Xiao-Yong Fan
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai, 201508, China.
| | - Zhidong Hu
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai, 201508, China.
| |
Collapse
|
11
|
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
|
12
|
Bromley JD, Ganchua SKC, Nyquist SK, Maiello P, Chao M, Borish HJ, Rodgers M, Tomko J, Kracinovsky K, Mugahid D, Nguyen S, Wang QD, Rosenberg JM, Klein EC, Gideon HP, Floyd-O'Sullivan R, Berger B, Scanga CA, Lin PL, Fortune SM, Shalek AK, Flynn JL. CD4 + T cells re-wire granuloma cellularity and regulatory networks to promote immunomodulation following Mtb reinfection. Immunity 2024; 57:2380-2398.e6. [PMID: 39214090 PMCID: PMC11466276 DOI: 10.1016/j.immuni.2024.08.002] [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: 12/22/2023] [Revised: 06/03/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024]
Abstract
Immunological priming-in the context of either prior infection or vaccination-elicits protective responses against subsequent Mycobacterium tuberculosis (Mtb) infection. However, the changes that occur in the lung cellular milieu post-primary Mtb infection and their contributions to protection upon reinfection remain poorly understood. Using clinical and microbiological endpoints in a non-human primate reinfection model, we demonstrated that prior Mtb infection elicited a long-lasting protective response against subsequent Mtb exposure and was CD4+ T cell dependent. By analyzing data from primary infection, reinfection, and reinfection-CD4+ T cell-depleted granulomas, we found that the presence of CD4+ T cells during reinfection resulted in a less inflammatory lung milieu characterized by reprogrammed CD8+ T cells, reduced neutrophilia, and blunted type 1 immune signaling among myeloid cells. These results open avenues for developing vaccines and therapeutics that not only target lymphocytes but also modulate innate immune cells to limit tuberculosis (TB) disease.
Collapse
Affiliation(s)
- Joshua D Bromley
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Graduate Program in Microbiology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sharie Keanne C Ganchua
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sarah K Nyquist
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Pauline Maiello
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Michael Chao
- 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
| | - H Jacob Borish
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mark Rodgers
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jaime Tomko
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kara Kracinovsky
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Douaa Mugahid
- 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
| | - Son Nguyen
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Qianchang Dennis Wang
- Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jacob M Rosenberg
- 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
| | - Edwin C Klein
- Division of Laboratory Animal Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hannah P Gideon
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Roisin Floyd-O'Sullivan
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Bonnie Berger
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Charles A Scanga
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Philana Ling Lin
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, 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.
| | - Alex K Shalek
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - JoAnne L Flynn
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
13
|
Liu D, Mai D, Jahn AN, Murray TA, Aitchison JD, Gern BH, Urdahl KB, Aderem A, Diercks AH, Gold ES. APOE Protects Against Severe Infection with Mycobacterium tuberculosis by Restraining Production of Neutrophil Extracellular Traps. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.04.616580. [PMID: 39605723 PMCID: PMC11601580 DOI: 10.1101/2024.10.04.616580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
While neutrophils are the predominant cell type in the lungs of humans with active tuberculosis (TB), they are relatively scarce in the lungs of most strains of mice that are used to study the disease. However, similar to humans, neutrophils account for approximately 45% of CD45+ cells in the lungs of Apoe -/- mice on a high-cholesterol (HC) diet following infection with Mycobacterium tuberculosis (Mtb). We hypothesized that the susceptibility of Apoe -/- HC mice might arise from an unrestrained feed-forward loop in which production of neutrophil extracellular traps (NETs) stimulates production of type I interferons by pDCs which in turn leads to the recruitment and activation of more neutrophils, and demonstrated that depleting neutrophils, depleting plasmacytoid dendritic cells (pDCs), or blocking type I interferon signaling, improved the outcome of infection. In concordance with these results, we found that Mtb-infected in Apoe -/- HC mice produce high levels of LTB4 and 12-HETE, two eicosanoids known to act as neutrophil chemoattractants and showed that blocking leukotriene B4 (LTB4) receptor signaling also improved the outcome of tuberculosis. While production of NETs has been associated with severe tuberculosis in other mouse models and in humans, a causative role for NETs in the pathology has not been directly established. We demonstrate that blocking the activation of peptidylarginine deiminase 4 (PAD4), an enzyme critical to NET formation, leads to fewer NETs in the lungs and, strikingly, completely reverses the hypersusceptibility of Apoe -/- HC mice to tuberculosis.
Collapse
Affiliation(s)
- Dong Liu
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute; Seattle, WA 98109, USA
| | - Dat Mai
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute; Seattle, WA 98109, USA
| | - Ana N. Jahn
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute; Seattle, WA 98109, USA
| | - Tara A. Murray
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute; Seattle, WA 98109, USA
| | - John D. Aitchison
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute; Seattle, WA 98109, USA
| | - Benjamin H. Gern
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute; Seattle, WA 98109, USA
- University of Washington, Dept. of Pediatrics; Seattle, Washington, USA
| | - Kevin B. Urdahl
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute; Seattle, WA 98109, USA
- University of Washington, Dept. of Immunology; Seattle, Washington, USA
- University of Washington, Dept. of Pediatrics; Seattle, Washington, USA
| | - Alan Aderem
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute; Seattle, WA 98109, USA
| | - Alan H. Diercks
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute; Seattle, WA 98109, USA
| | - Elizabeth S. Gold
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute; Seattle, WA 98109, USA
- Virginia Mason Franciscan Health; Seattle, WA, 98101, USA
| |
Collapse
|
14
|
Church EC, Bishop E, Fiore-Gartland A, Yu KKQ, Chang M, Jones RM, Brache JK, Ballweber Fleming L, Phan JM, Makatsa MS, Heptinstall J, Chiong K, Dintwe O, Naidoo A, Voillet V, Mayer-Blackwell K, Nwanne G, Andersen-Nissen E, Vary JC, Tomaras GD, McElrath MJ, Sherman DR, Murphy SC, Kublin JG, Seshadri C. Probing Dermal Immunity to Mycobacteria through a Controlled Human Infection Model. Immunohorizons 2024; 8:695-711. [PMID: 39283647 PMCID: PMC11447685 DOI: 10.4049/immunohorizons.2400053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 08/13/2024] [Indexed: 09/22/2024] Open
Abstract
Cutaneous mycobacterial infections cause substantial morbidity and are challenging to diagnose and treat. An improved understanding of the dermal immune response to mycobacteria may inspire new therapeutic approaches. We conducted a controlled human infection study with 10 participants who received 2 × 106 CFUs of Mycobacterium bovis bacillus Calmette-Guérin (Tice strain) intradermally and were randomized to receive isoniazid or no treatment. Peripheral blood was collected at multiple time points for flow cytometry, bulk RNA sequencing (RNA-seq), and serum Ab assessments. Systemic immune responses were detected as early as 8 d postchallenge in this M. bovis bacillus Calmette-Guérin-naive population. Injection-site skin biopsies were performed at days 3 and 15 postchallenge and underwent immune profiling using mass cytometry and single-cell RNA-seq, as well as quantitative assessments of bacterial viability and burden. Molecular viability testing and standard culture results correlated well, although no differences were observed between treatment arms. Single-cell RNA-seq revealed various immune and nonimmune cell types in the skin, and communication between them was inferred by ligand-receptor gene expression. Day 3 communication was predominantly directed toward monocytes from keratinocyte, muscle, epithelial, and endothelial cells, largely via the migration inhibitory factor pathway and HLA-E-KLRK1 interaction. At day 15, communication was more balanced between cell types. These data reveal the potential role of nonimmune cells in the dermal immune response to mycobacteria and the utility of human challenge studies to augment our understanding of mycobacterial infections.
Collapse
Affiliation(s)
- E. Chandler Church
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
- Department of Medicine, University of Washington School of Medicine, Seattle, WA
- Seattle-King County Public Health, Seattle, WA
| | - Emma Bishop
- Department of Medicine, University of Washington School of Medicine, Seattle, WA
| | | | - Krystle K. Q. Yu
- Department of Medicine, University of Washington School of Medicine, Seattle, WA
| | - Ming Chang
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA
| | - Richard M. Jones
- Department of Microbiology, University of Washington School of Medicine, Seattle, WA
| | - Justin K. Brache
- Department of Microbiology, University of Washington School of Medicine, Seattle, WA
| | | | - Jolie M. Phan
- Department of Medicine, University of Washington School of Medicine, Seattle, WA
| | - Mohau S. Makatsa
- Department of Medicine, University of Washington School of Medicine, Seattle, WA
| | - Jack Heptinstall
- Duke Center for Human Systems Immunology, Duke University, Durham, NC
| | - Kelvin Chiong
- Duke Center for Human Systems Immunology, Duke University, Durham, NC
| | - One Dintwe
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
- Cape Town HVTN Immunology Laboratory, Hutchinson Centre Research Institute of South Africa, Cape Town, South Africa
| | - Anneta Naidoo
- Cape Town HVTN Immunology Laboratory, Hutchinson Centre Research Institute of South Africa, Cape Town, South Africa
| | - Valentin Voillet
- Cape Town HVTN Immunology Laboratory, Hutchinson Centre Research Institute of South Africa, Cape Town, South Africa
| | | | - Gift Nwanne
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Erica Andersen-Nissen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
- Cape Town HVTN Immunology Laboratory, Hutchinson Centre Research Institute of South Africa, Cape Town, South Africa
| | - Jay C. Vary
- Department of Dermatology, University of Washington School of Medicine, Seattle, WA
| | | | - M. Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
- Department of Medicine, University of Washington School of Medicine, Seattle, WA
| | - David R. Sherman
- Department of Microbiology, University of Washington School of Medicine, Seattle, WA
| | - Sean C. Murphy
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA
| | - James G. Kublin
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Chetan Seshadri
- Department of Medicine, University of Washington School of Medicine, Seattle, WA
| |
Collapse
|
15
|
Cohen SB, Plumlee CR, Engels L, Mai D, Murray TA, Jahn AN, Alexander B, Delahaye JL, Cross LM, Maciag K, Schrader S, Durga K, Gold ES, Aderem A, Gerner MY, Gern BH, Diercks AH, Urdahl KB. Host and pathogen genetic diversity shape vaccine-mediated protection to Mycobacterium tuberculosis. Front Immunol 2024; 15:1427846. [PMID: 39007152 PMCID: PMC11239334 DOI: 10.3389/fimmu.2024.1427846] [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: 05/04/2024] [Accepted: 06/13/2024] [Indexed: 07/16/2024] Open
Abstract
To investigate how host and pathogen diversity govern immunity against Mycobacterium tuberculosis (Mtb), we performed a large-scale screen of vaccine-mediated protection against aerosol Mtb infection using three inbred mouse strains [C57BL/6 (B6), C3HeB/FeJ (C3H), Balb/c x 129/SvJ (C129F1)] and three Mtb strains (H37Rv, CDC1551, SA161) representing two lineages and distinct virulence properties. We compared three protective modalities, all of which involve inoculation with live mycobacteria: Bacillus Calmette-Guérin (BCG), the only approved TB vaccine, delivered either subcutaneously or intravenously, and concomitant Mtb infection (CoMtb), a model of pre-existing immunity in which a low-level Mtb infection is established in the cervical lymph node following intradermal inoculation. We examined lung bacterial burdens at early (Day 28) and late (Day 98) time points after aerosol Mtb challenge and histopathology at Day 98. We observed substantial heterogeneity in the reduction of bacterial load afforded by these modalities at Day 28 across the combinations and noted a strong positive correlation between bacterial burden in unvaccinated mice and the degree of protection afforded by vaccination. Although we observed variation in the degree of reduction in bacterial burdens across the nine mouse/bacterium strain combinations, virtually all protective modalities performed similarly for a given strain-strain combination. We also noted dramatic variation in histopathology changes driven by both host and bacterial genetic backgrounds. Vaccination improved pathology scores for all infections except CDC1551. However, the most dramatic impact of vaccination on lesion development occurred for the C3H-SA161 combination, where vaccination entirely abrogated the development of the large necrotic lesions that arise in unvaccinated mice. In conclusion, we find that substantial TB heterogeneity can be recapitulated by introducing variability in both host and bacterial genetics, resulting in changes in vaccine-mediated protection as measured both by bacterial burden as well as histopathology. These differences can be harnessed in future studies to identify immune correlates of vaccine efficacy.
Collapse
Affiliation(s)
- Sara B. Cohen
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Courtney R. Plumlee
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Lindsay Engels
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Dat Mai
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Tara A. Murray
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Ana N. Jahn
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Bridget Alexander
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Jared L. Delahaye
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Lauren M. Cross
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Karolina Maciag
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
- Department of Medicine, Division of Infectious Diseases, University of Washington, Seattle, WA, United States
| | - Sam Schrader
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Kaitlin Durga
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Elizabeth S. Gold
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Alan Aderem
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
- Department of Pediatrics, University of Washington, Seattle, WA, United States
| | - Michael Y. Gerner
- Department of Immunology, University of Washington, Seattle, WA, United States
| | - Benjamin H. Gern
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
- Department of Pediatrics, University of Washington, Seattle, WA, United States
| | - Alan H. Diercks
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Kevin B. Urdahl
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
- Department of Pediatrics, University of Washington, Seattle, WA, United States
| |
Collapse
|
16
|
Gern BH, Klas JM, Foster KA, Cohen SB, Plumlee CR, Duffy FJ, Neal ML, Halima M, Gustin AT, Diercks AH, Aderem A, Gale M, Aitchison JD, Gerner MY, Urdahl KB. CD4-mediated immunity shapes neutrophil-driven tuberculous pathology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.12.589315. [PMID: 38659794 PMCID: PMC11042216 DOI: 10.1101/2024.04.12.589315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Pulmonary Mycobacterium tuberculosis (Mtb) infection results in highly heterogeneous lesions ranging from granulomas with central necrosis to those primarily comprised of alveolitis. While alveolitis has been associated with prior immunity in human post-mortem studies, the drivers of these distinct pathologic outcomes are poorly understood. Here, we show that these divergent lesion structures can be modeled in C3HeB/FeJ mice and are regulated by prior immunity. Using quantitative imaging, scRNAseq, and flow cytometry, we demonstrate that Mtb infection in the absence of prior immunity elicits dysregulated neutrophil recruitment and necrotic granulomas. In contrast, prior immunity induces rapid recruitment and activation of T cells, local macrophage activation, and diminished late neutrophil responses. Depletion studies at distinct infection stages demonstrated that neutrophils are required for early necrosis initiation and necrosis propagation at chronic stages, whereas early CD4 T cell responses prevent neutrophil feedforward circuits and necrosis. Together, these studies reveal fundamental determinants of tuberculosis lesion structure and pathogenesis, which have important implications for new strategies to prevent or treat tuberculosis.
Collapse
Affiliation(s)
- Benjamin H Gern
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, United States of America
- University of Washington, Dept. of Pediatrics, Seattle, Washington, United States of America
| | - Josepha M Klas
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, United States of America
| | - Kimberly A Foster
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, United States of America
- University of Washington, Dept. of Immunology, Seattle, Washington, United States of America
| | - Sara B Cohen
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, United States of America
| | - Courtney R Plumlee
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, United States of America
| | - Fergal J Duffy
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, United States of America
| | - Maxwell L Neal
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, United States of America
| | - Mehnaz Halima
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, United States of America
| | - Andrew T Gustin
- University of Washington, Dept. of Immunology, Seattle, Washington, United States of America
| | - Alan H Diercks
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, United States of America
| | - Alan Aderem
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, United States of America
| | - Michael Gale
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, United States of America
- University of Washington, Dept. of Immunology, Seattle, Washington, United States of America
| | - John D Aitchison
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, United States of America
| | - Michael Y Gerner
- University of Washington, Dept. of Immunology, Seattle, Washington, United States of America
| | - Kevin B Urdahl
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, United States of America
- University of Washington, Dept. of Pediatrics, Seattle, Washington, United States of America
- University of Washington, Dept. of Immunology, Seattle, Washington, United States of America
- Lead Contact
| |
Collapse
|
17
|
Ganchua SK, Maiello P, Chao M, Hopkins F, Mugahid D, Lin PL, Fortune SM, Flynn JL. Antibiotic treatment modestly reduces protection against Mycobacterium tuberculosis reinfection in macaques. Infect Immun 2024; 92:e0053523. [PMID: 38514467 PMCID: PMC11003231 DOI: 10.1128/iai.00535-23] [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: 12/21/2023] [Accepted: 03/03/2024] [Indexed: 03/23/2024] Open
Abstract
Concomitant immunity is generally defined as an ongoing infection providing protection against reinfection . Its role in prevention of tuberculosis (TB) caused by Mycobacterium tuberculosis (Mtb) is supported by epidemiological evidence in humans as well as experimental evidence in mice and non-human primates (NHPs). Whether the presence of live Mtb, rather than simply persistent antigen, is necessary for concomitant immunity in TB is still unclear. Here, we investigated whether live Mtb plays a measurable role in control of secondary Mtb infection. Using cynomolgus macaques, molecularly barcoded Mtb libraries, positron emission tomography-computed tomography (PET CT) imaging, flow cytometry, and cytokine profiling, we evaluated the effect of antibiotic treatment after primary infection on immunological response and bacterial establishment, dissemination, and burden post-secondary infection. Our data provide evidence that, in this experimental model, treatment with antibiotics after primary infection reduced inflammation in the lung but was not associated with a significant change in bacterial establishment, dissemination, or burden in the lung or lymph nodes. Nonetheless, treatment of the prior infection with antibiotics did result in a modest reduction in protection against reinfection: none of the seven antibiotic-treated animals demonstrated sterilizing immunity against reinfection, while four of the seven non-treated macaques were completely protected against reinfection. These findings support that antibiotic-treated animals were still able to restrict bacterial establishment and dissemination after rechallenge compared to naïve macaques, but not to the full extent of non-antibiotic-treated macaques.
Collapse
Affiliation(s)
- Sharie Keanne Ganchua
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Pauline Maiello
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Michael Chao
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, Massachusetts, USA
| | - Forrest Hopkins
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, Massachusetts, USA
| | - Douaa Mugahid
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, Massachusetts, USA
| | - Philana Ling Lin
- Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Sarah M. Fortune
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, Massachusetts, USA
| | - JoAnne L. Flynn
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
18
|
Tepekule Mueller B, Joerimann L, Schenkel CD, Opitz L, Tschumi J, Wolfensberger R, Neumann K, Kusejko K, Zeeb M, Boeck L, Kaelin M, Notter J, Furrer H, Hoffmann M, Hirsch HH, Calmy A, Cavassini M, Labhardt ND, Bernasconi E, Metzner KJ, Braun DL, Guenthard HF, Kouyos RD, Duffy F, Nemeth J. Mycobacterium tuberculosis infection associated immune perturbations correlate with antiretroviral immunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.14.548872. [PMID: 37649906 PMCID: PMC10465018 DOI: 10.1101/2023.07.14.548872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Infection with Mycobacterium tuberculosis (MTB) remains one of the most important opportunistic infections in people with HIV-1 (PWH). While active Tuberculosis (TB) leads to rapid progression of immunodeficiency in PWH, the interaction between MTB and HIV-1 during the asymptomatic phase of both infections remains poorly understood. In a cohort of individuals with HIV (PWH) with and without suppressed HIV-1 viral load, the transcriptomic profiles of peripheral blood mononuclear cells (PBMC) clustered in individuals infected with Mycobacterium tuberculosis (MTB) compared to carefully matched controls. Subsequent functional annotation analysis disclosed alterations in the IL-6, TNF, and KRAS pathways. Notably, MTB-associated genes demonstrated an inverse correlation with HIV-1 viremia, evident at both on individual gene level and when employed as a gene score. In sum, our data show that MTB infection in PWH is associated with a shift in the activation state of the immune system, displaying an inverse relationship with HIV-1 viral load. These results could provide an explanation for the observed increased antiretroviral control associated with MTB infection in PWH.
Collapse
|
19
|
Ganchua SK, Maiello P, Chao M, Hopkins F, Mugahid D, Lin PL, Fortune SM, Flynn JL. Antibiotic treatment modestly reduces protection against Mycobacterium tuberculosis reinfection in macaques. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.19.570845. [PMID: 38187678 PMCID: PMC10769216 DOI: 10.1101/2023.12.19.570845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Concomitant immunity is generally defined as an ongoing infection providing protection against reinfection1. Its role in prevention of tuberculosis (TB) caused by Mycobacterium tuberculosis (Mtb) is supported by epidemiological evidence in humans as well as experimental evidence in mice and non-human primates (NHPs). Whether the presence of live Mtb, rather than simply persistent antigen, is necessary for concomitant immunity in TB is still unclear. Here, we investigated whether live Mtb plays a measurable role in control of secondary Mtb infection. Using cynomolgus macaques, molecularly barcoded Mtb libraries, PET-CT imaging, flow cytometry and cytokine profiling we evaluated the effect of antibiotic treatment after primary infection on immunological response and bacterial establishment, dissemination, and burden post-secondary infection. Our data provide evidence that, in this experimental model, treatment with antibiotics after primary infection reduced inflammation in the lung but was not associated with a significant change in bacterial establishment, dissemination or burden in the lung or lymph nodes. Nonetheless, treatment of the prior infection with antibiotics did result in a modest reduction in protection against reinfection: none of the 7 antibiotic treated animals demonstrated sterilizing immunity against reinfection while 4 of the 7 non-treated macaques were completely protected against reinfection. These findings support that antibiotic-treated animals were still able to restrict bacterial establishment and dissemination after rechallenge compared to naïve macaques, but not to the full extent of non-antibiotic treated macaques.
Collapse
Affiliation(s)
- Sharie Keanne Ganchua
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, 523 Bridgeside Point 2, 450 Technology Drive, Pittsburgh, PA 15219
| | - Pauline Maiello
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, 523 Bridgeside Point 2, 450 Technology Drive, Pittsburgh, PA 15219
| | - Michael Chao
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, 677 Huntington Ave, Boston, MA 02115
| | - Forrest Hopkins
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, 677 Huntington Ave, Boston, MA 02115
| | - Douaa Mugahid
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, 677 Huntington Ave, Boston, MA 02115
| | - Philana Ling Lin
- Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh PA 15224
| | - Sarah M. Fortune
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, 677 Huntington Ave, Boston, MA 02115
| | - JoAnne L. Flynn
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, 523 Bridgeside Point 2, 450 Technology Drive, Pittsburgh, PA 15219
| |
Collapse
|
20
|
Grigsby SJ, Prasad GVRK, Wallach JB, Mittal E, Hsu FF, Schnappinger D, Philips JA. CpsA mediates infection of recruited lung myeloid cells by Mycobacterium tuberculosis. Cell Rep 2024; 43:113607. [PMID: 38127624 PMCID: PMC10900767 DOI: 10.1016/j.celrep.2023.113607] [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: 05/21/2023] [Revised: 10/27/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb) possesses an arsenal of virulence factors to evade host immunity. Previously, we showed that the Mtb protein CpsA, which protects Mtb against the host NADPH oxidase, is required in mice during the first 3 weeks of infection but is thereafter dispensable for full virulence. Using flow cytometry, we find that ΔcpsA Mtb is retained in alveolar macrophages, impaired in recruiting and disseminating into monocyte-derived cells, and more likely to be localized in airway cells than wild-type Mtb. The lungs of ΔcpsA-infected mice also have markedly fewer antigen-specific T cells, indicating a delay in adaptive immunity. Thus, we conclude that CpsA promotes dissemination of Mtb from alveolar macrophages and the airways and generation of an adaptive immune response. Our studies of ΔcpsA Mtb show that a more effective innate immune response against Mtb can be undermined by a corresponding delay in the adaptive immune response.
Collapse
Affiliation(s)
- 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
| | - G V R Krishna Prasad
- 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
| | - Joshua B Wallach
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York City, NY, USA
| | - Ekansh Mittal
- 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
| | - Fong-Fu Hsu
- Division of Endocrinology, Metabolism, & Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Dirk Schnappinger
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York City, NY, 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
|
21
|
Mai D, Jahn A, Murray T, Morikubo M, Lim PN, Cervantes MM, Pham LK, Nemeth J, Urdahl K, Diercks AH, Aderem A, Rothchild AC. Exposure to Mycobacterium remodels alveolar macrophages and the early innate response to Mycobacterium tuberculosis infection. PLoS Pathog 2024; 20:e1011871. [PMID: 38236787 PMCID: PMC10796046 DOI: 10.1371/journal.ppat.1011871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 11/27/2023] [Indexed: 01/22/2024] Open
Abstract
Alveolar macrophages (AMs) play a critical role during Mycobacterium tuberculosis (Mtb) infection as the first cells in the lung to encounter bacteria. We previously showed that AMs initially respond to Mtb in vivo by mounting a cell-protective, rather than pro-inflammatory response. However, the plasticity of the initial AM response was unknown. Here, we characterize how previous exposure to Mycobacterium, either through subcutaneous vaccination with Mycobacterium bovis (scBCG) or through a contained Mtb infection (coMtb) that mimics aspects of concomitant immunity, impacts the initial response by AMs. We find that both scBCG and coMtb accelerate early innate cell activation and recruitment and generate a stronger pro-inflammatory response to Mtb in vivo by AMs. Within the lung environment, AMs from scBCG vaccinated mice mount a robust interferon-associated response, while AMs from coMtb mice produce a broader inflammatory response that is not dominated by Interferon Stimulated Genes. Using scRNAseq, we identify changes to the frequency and phenotype of airway-resident macrophages following Mycobacterium exposure, with enrichment for both interferon-associated and pro-inflammatory populations of AMs. In contrast, minimal changes were found for airway-resident T cells and dendritic cells after exposures. Ex vivo stimulation of AMs with Pam3Cys, LPS and Mtb reveal that scBCG and coMtb exposures generate stronger interferon-associated responses to LPS and Mtb that are cell-intrinsic changes. However, AM profiles that were unique to each exposure modality following Mtb infection in vivo are dependent on the lung environment and do not emerge following ex vivo stimulation. Overall, our studies reveal significant and durable remodeling of AMs following exposure to Mycobacterium, with evidence for both AM-intrinsic changes and contributions from the altered lung microenvironments. Comparisons between the scBCG and coMtb models highlight the plasticity of AMs in the airway and opportunities to target their function through vaccination or host-directed therapies.
Collapse
Affiliation(s)
- Dat Mai
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Ana Jahn
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Tara Murray
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Michael Morikubo
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Pamelia N. Lim
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, United States of America
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, Amherst, Massachusetts, United States of America
| | - Maritza M. Cervantes
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, United States of America
| | - Linh K. Pham
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, United States of America
- Animal Biotechnology and Biomedical Sciences Graduate Program, University of Massachusetts Amherst, Amherst, Massachusetts, United States of America
| | - Johannes Nemeth
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Kevin Urdahl
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Alan H. Diercks
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Alan Aderem
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Alissa C. Rothchild
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, United States of America
| |
Collapse
|
22
|
Bromley JD, Ganchua SKC, Nyquist SK, Maiello P, Chao M, Borish HJ, Rodgers M, Tomko J, Kracinovsky K, Mugahid D, Nguyen S, Wang D, Rosenberg JM, Klein EC, Gideon HP, Floyd-O’Sullivan R, Berger B, Scanga CA, Lin PL, Fortune SM, Shalek AK, Flynn JL. CD4 + T cells are homeostatic regulators during Mtb reinfection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.20.572669. [PMID: 38187598 PMCID: PMC10769325 DOI: 10.1101/2023.12.20.572669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Immunological priming - either in the context of prior infection or vaccination - elicits protective responses against subsequent Mycobacterium tuberculosis (Mtb) infection. However, the changes that occur in the lung cellular milieu post-primary Mtb infection and their contributions to protection upon reinfection remain poorly understood. Here, using clinical and microbiological endpoints in a non-human primate reinfection model, we demonstrate that prior Mtb infection elicits a long-lasting protective response against subsequent Mtb exposure and that the depletion of CD4+ T cells prior to Mtb rechallenge significantly abrogates this protection. Leveraging microbiologic, PET-CT, flow cytometric, and single-cell RNA-seq data from primary infection, reinfection, and reinfection-CD4+ T cell depleted granulomas, we identify differential cellular and microbial features of control. The data collectively demonstrate that the presence of CD4+ T cells in the setting of reinfection results in a reduced inflammatory lung milieu characterized by reprogrammed CD8+ T cell activity, reduced neutrophilia, and blunted type-1 immune signaling among myeloid cells, mitigating Mtb disease severity. These results open avenues for developing vaccines and therapeutics that not only target CD4+ and CD8+ T cells, but also modulate innate immune cells to limit Mtb disease.
Collapse
Affiliation(s)
- Joshua D. Bromley
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Graduate Program in Microbiology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sharie Keanne C. Ganchua
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh PA USA
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh PA USA
| | - Sarah K. Nyquist
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Pauline Maiello
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh PA USA
| | - Michael Chao
- 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
| | - H. Jacob Borish
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh PA USA
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh PA USA
| | - Mark Rodgers
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh PA USA
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh PA USA
| | - Jaime Tomko
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh PA USA
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh PA USA
| | - Kara Kracinovsky
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh PA USA
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh PA USA
| | - Douaa Mugahid
- 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
| | - Son Nguyen
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Dennis Wang
- Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jacob M. Rosenberg
- 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
| | - Edwin C. Klein
- Division of Laboratory Animal Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hannah P. Gideon
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh PA USA
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh PA USA
| | - Roisin Floyd-O’Sullivan
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Bonnie Berger
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Charles A Scanga
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh PA USA
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh PA USA
| | - Philana Ling Lin
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh PA USA
- Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine
| | - 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
| | - Alex K. Shalek
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - JoAnne L. Flynn
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh PA USA
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh PA USA
- Lead contact
| |
Collapse
|
23
|
Silver RF, Xia M, Storer CE, Jarvela JR, Moyer MC, Blazevic A, Stoeckel DA, Rakey EK, Tennant JM, Goll JB, Head RD, Hoft DF. Distinct gene expression signatures comparing latent tuberculosis infection with different routes of Bacillus Calmette-Guérin vaccination. Nat Commun 2023; 14:8507. [PMID: 38129388 PMCID: PMC10739751 DOI: 10.1038/s41467-023-44136-8] [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: 12/17/2022] [Accepted: 12/01/2023] [Indexed: 12/23/2023] Open
Abstract
Tuberculosis remains an international health threat partly because of limited protection from pulmonary tuberculosis provided by standard intradermal vaccination with Bacillus of Calmette and Guérin (BCG); this may reflect the inability of intradermal vaccination to optimally induce pulmonary immunity. In contrast, respiratory Mycobacterium tuberculosis infection usually results in the immune-mediated bacillary containment of latent tuberculosis infection (LTBI). Here we present RNA-Seq-based assessments of systemic and pulmonary immune cells from LTBI participants and recipients of intradermal and oral BCG. LTBI individuals uniquely display ongoing immune activation and robust CD4 T cell recall responses in blood and lung. Intradermal BCG is associated with robust systemic immunity but only limited pulmonary immunity. Conversely, oral BCG induces limited systemic immunity but distinct pulmonary responses including enhanced inflammasome activation potentially associated with mucosal-associated invariant T cells. Further, IL-9 is identified as a component of systemic immunity in LTBI and intradermal BCG, and pulmonary immunity following oral BCG.
Collapse
Affiliation(s)
- Richard F Silver
- Division of Pulmonary, Critical Care and Sleep Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
- Pulmonary and Critical Care Medicine, The Louis Stokes Cleveland Department of Veterans' Affairs Medical Center, Cleveland, OH, USA.
| | - Mei Xia
- Division of Infectious Diseases, Allergy & Immunology, Saint Louis University School of Medicine, St. Louis, MO, USA
- Center for Vaccine Development, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Chad E Storer
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Jessica R Jarvela
- Division of Pulmonary, Critical Care and Sleep Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Pulmonary and Critical Care Medicine, The Louis Stokes Cleveland Department of Veterans' Affairs Medical Center, Cleveland, OH, USA
| | - Michelle C Moyer
- Division of Pulmonary, Critical Care and Sleep Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Pulmonary and Critical Care Medicine, The Louis Stokes Cleveland Department of Veterans' Affairs Medical Center, Cleveland, OH, USA
| | - Azra Blazevic
- Division of Infectious Diseases, Allergy & Immunology, Saint Louis University School of Medicine, St. Louis, MO, USA
- Center for Vaccine Development, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - David A Stoeckel
- Division of Pulmonary, Critical Care and Sleep Medicine, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Erin K Rakey
- Division of Pulmonary, Critical Care and Sleep Medicine, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Jan M Tennant
- Division of Infectious Diseases, Allergy & Immunology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | | | - Richard D Head
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Daniel F Hoft
- Division of Infectious Diseases, Allergy & Immunology, Saint Louis University School of Medicine, St. Louis, MO, USA.
- Center for Vaccine Development, Saint Louis University School of Medicine, St. Louis, MO, USA.
- Department of Molecular Microbiology & Immunology Saint Louis University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
24
|
Solomon SL, Bryson BD. Single-cell analysis reveals a weak macrophage subpopulation response to Mycobacterium tuberculosis infection. Cell Rep 2023; 42:113418. [PMID: 37963018 PMCID: PMC10842899 DOI: 10.1016/j.celrep.2023.113418] [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: 04/24/2023] [Revised: 08/28/2023] [Accepted: 10/25/2023] [Indexed: 11/16/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb) infection remains one of society's greatest human health challenges. Macrophages integrate multiple signals derived from ontogeny, infection, and the environment. This integration proceeds heterogeneously during infection. Some macrophages are infected, while others are not; therefore, bulk approaches mask the subpopulation dynamics. We establish a modular, targeted, single-cell protein analysis framework to study the immune response to Mtb. We demonstrate that during Mtb infection, only a small fraction of resting macrophages produce tumor necrosis factor (TNF) protein. We demonstrate that Mtb infection results in muted phosphorylation of p38 and JNK, regulators of inflammation, and leverage our single-cell methods to distinguish between pathogen-mediated interference in host signaling and weak activation of host pathways. We demonstrate that the inflammatory signal magnitude is decoupled from the ability to control Mtb growth. These data underscore the importance of developing pathogen-specific models of signaling and highlight barriers to activation of pathways that control inflammation.
Collapse
Affiliation(s)
- Sydney L Solomon
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; The Ragon Institute of MGH, Harvard & MIT, Cambridge, MA 02139, USA
| | - Bryan D Bryson
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; The Ragon Institute of MGH, Harvard & MIT, Cambridge, MA 02139, USA.
| |
Collapse
|
25
|
Horton KC, Richards AS, Emery JC, Esmail H, Houben RMGJ. Reevaluating progression and pathways following Mycobacterium tuberculosis infection within the spectrum of tuberculosis. Proc Natl Acad Sci U S A 2023; 120:e2221186120. [PMID: 37963250 PMCID: PMC10666121 DOI: 10.1073/pnas.2221186120] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 09/12/2023] [Indexed: 11/16/2023] Open
Abstract
Traditional understanding of the risk of progression from Mycobacterium tuberculosis (Mtb) infection to tuberculosis (TB) overlooks diverse presentations across a spectrum of disease. We developed a deterministic model of Mtb infection and minimal (pathological damage but not infectious), subclinical (infectious but no reported symptoms), and clinical (infectious and symptomatic) TB, informed by a rigorous evaluation of data from a systematic review of TB natural history. Using a Bayesian approach, we calibrated the model to data from historical cohorts that followed tuberculin-negative individuals to tuberculin conversion and TB, as well as data from cohorts that followed progression and regression between disease states, disease state prevalence ratios, disease duration, and mortality. We estimated incidence, pathways, and 10-y outcomes following Mtb infection for a simulated cohort. Then, 92.0% (95% uncertainty interval, UI, 91.4 to 92.5) of individuals self-cleared within 10 y of infection, while 7.9% (95% UI 7.4 to 8.5) progressed to TB. Of those, 68.6% (95% UI 65.4 to 72.0) developed infectious disease, and 33.2% (95% UI 29.9 to 36.4) progressed to clinical disease. While 98% of progression to minimal disease occurred within 2 y of infection, only 71% and 44% of subclinical and clinical disease, respectively, occurred within this period. Multiple progression pathways from infection were necessary to calibrate the model and 49.5% (95% UI 45.6 to 53.7) of those who developed infectious disease undulated between disease states. We identified heterogeneous pathways across disease states after Mtb infection, highlighting the need for clearly defined disease thresholds to inform more effective prevention and treatment efforts to end TB.
Collapse
Affiliation(s)
- Katherine C. Horton
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, LondonWC1E 7HT, United Kingdom
| | - Alexandra S. Richards
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, LondonWC1E 7HT, United Kingdom
| | - Jon C. Emery
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, LondonWC1E 7HT, United Kingdom
| | - Hanif Esmail
- Clinical Trials Unit, University College London, LondonWC1V 6LJ, United Kingdom
| | - Rein M. G. J. Houben
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, LondonWC1E 7HT, United Kingdom
| |
Collapse
|
26
|
Plumlee CR, Barrett HW, Shao DE, Lien KA, Cross LM, Cohen SB, Edlefsen PT, Urdahl KB. Assessing vaccine-mediated protection in an ultra-low dose Mycobacterium tuberculosis murine model. PLoS Pathog 2023; 19:e1011825. [PMID: 38011264 PMCID: PMC10703413 DOI: 10.1371/journal.ppat.1011825] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 12/07/2023] [Accepted: 11/14/2023] [Indexed: 11/29/2023] Open
Abstract
Despite widespread immunization with Bacille-Calmette-Guérin (BCG), the only currently licensed tuberculosis (TB) vaccine, TB remains a leading cause of mortality globally. There are many TB vaccine candidates in the developmental pipeline, but the lack of a robust animal model to assess vaccine efficacy has hindered our ability to prioritize candidates for human clinical trials. Here we use a murine ultra-low dose (ULD) Mycobacterium tuberculosis (Mtb) challenge model to assess protection conferred by BCG vaccination. We show that BCG confers a reduction in lung bacterial burdens that is more durable than that observed after conventional dose challenge, curbs Mtb dissemination to the contralateral lung, and, in a small percentage of mice, prevents detectable infection. These findings are consistent with the ability of human BCG vaccination to mediate protection, particularly against disseminated disease, in specific human populations and clinical settings. Overall, our findings demonstrate that the ultra-low dose Mtb infection model can measure distinct parameters of immune protection that cannot be assessed in conventional dose murine infection models and could provide an improved platform for TB vaccine testing.
Collapse
Affiliation(s)
- Courtney R. Plumlee
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Holly W. Barrett
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
- University of Washington, Dept. of Global Health, Seattle, Washington, United States of America
| | - Danica E. Shao
- Vaccine and Infectious Disease Division, Fred Hutch Cancer Center, Seattle, Washington, United States of America
| | - Katie A. Lien
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Lauren M. Cross
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Sara B. Cohen
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Paul T. Edlefsen
- Vaccine and Infectious Disease Division, Fred Hutch Cancer Center, Seattle, Washington, United States of America
| | - Kevin B. Urdahl
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
- University of Washington, Dept. of Immunology, Seattle, Washington, United States of America
- University of Washington, Dept. of Pediatrics, Seattle, Washington, United States of America
| |
Collapse
|
27
|
Larson EC, Ellis-Connell AL, Rodgers MA, Gubernat AK, Gleim JL, Moriarty RV, Balgeman AJ, Ameel CL, Jauro S, Tomko JA, Kracinovsky KB, Maiello P, Borish HJ, White AG, Klein E, Bucsan AN, Darrah PA, Seder RA, Roederer M, Lin PL, Flynn JL, O'Connor SL, Scanga CA. Intravenous Bacille Calmette-Guérin vaccination protects simian immunodeficiency virus-infected macaques from tuberculosis. Nat Microbiol 2023; 8:2080-2092. [PMID: 37814073 PMCID: PMC10627825 DOI: 10.1038/s41564-023-01503-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/13/2023] [Indexed: 10/11/2023]
Abstract
Tuberculosis, caused by Mycobacterium tuberculosis (Mtb), is the most common cause of death in people living with human immunodeficiency virus (HIV). Intra-dermal Bacille Calmette-Guérin (BCG) delivery is the only licensed vaccine against tuberculosis; however, it offers little protection from pulmonary tuberculosis in adults and is contraindicated in people living with HIV. Intravenous BCG confers protection against Mtb infection in rhesus macaques; we hypothesized that it might prevent tuberculosis in simian immunodeficiency virus (SIV)-infected macaques, a model for HIV infection. Here intravenous BCG-elicited robust airway T cell influx and elevated plasma and airway antibody titres in both SIV-infected and naive animals. Following Mtb challenge, all 7 vaccinated SIV-naive and 9 out of 12 vaccinated SIV-infected animals were protected, without any culturable bacteria detected from tissues. Peripheral blood mononuclear cell responses post-challenge indicated early clearance of Mtb in vaccinated animals, regardless of SIV infection. These data support that intravenous BCG is immunogenic and efficacious in SIV-infected animals.
Collapse
Affiliation(s)
- 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.
| | - Amy L Ellis-Connell
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI, USA
| | - Mark A Rodgers
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Abigail K Gubernat
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Janelle L Gleim
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ryan V Moriarty
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI, USA
| | - Alexis J Balgeman
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI, USA
| | - Cassaundra L Ameel
- Department of Microbiology and Molecular Genetics, 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
| | - Jaime A Tomko
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kara B Kracinovsky
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Pauline Maiello
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - H Jake Borish
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alexander G White
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Edwin Klein
- Division of Laboratory Animal Resources, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Allison N Bucsan
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Patricia A Darrah
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Philana Ling Lin
- Department of Pediatrics, Children's Hospital of Pittsburgh, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - JoAnne L Flynn
- 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
| | - Shelby L O'Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI, USA
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI, 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
| |
Collapse
|
28
|
Malainou C, Abdin SM, Lachmann N, Matt U, Herold S. Alveolar macrophages in tissue homeostasis, inflammation, and infection: evolving concepts of therapeutic targeting. J Clin Invest 2023; 133:e170501. [PMID: 37781922 PMCID: PMC10541196 DOI: 10.1172/jci170501] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023] Open
Abstract
Alveolar macrophages (AMs) are the sentinel cells of the alveolar space, maintaining homeostasis, fending off pathogens, and controlling lung inflammation. During acute lung injury, AMs orchestrate the initiation and resolution of inflammation in order to ultimately restore homeostasis. This central role in acute lung inflammation makes AMs attractive targets for therapeutic interventions. Single-cell RNA-Seq and spatial omics approaches, together with methodological advances such as the generation of human macrophages from pluripotent stem cells, have increased understanding of the ontogeny, function, and plasticity of AMs during infectious and sterile lung inflammation, which could move the field closer to clinical application. However, proresolution phenotypes might conflict with proinflammatory and antibacterial responses. Therefore, therapeutic targeting of AMs at vulnerable time points over the course of infectious lung injury might harbor the risk of serious side effects, such as loss of antibacterial host defense capacity. Thus, the identification of key signaling hubs that determine functional fate decisions in AMs is of the utmost importance to harness their therapeutic potential.
Collapse
Affiliation(s)
- Christina Malainou
- Department of Internal Medicine V, Universities of Giessen and Marburg Lung Center, Justus Liebig University Giessen, Member of the German Center for Lung Research (DZL), Giessen, Germany
- Institute for Lung Health, Justus Liebig University Giessen, Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute, Giessen, Germany
- German Center for Lung Research (DZL), Heidelberg, Germany
| | - Shifaa M. Abdin
- German Center for Lung Research (DZL), Heidelberg, Germany
- Department of Pediatric Pneumology, Allergology and Neonatology and
- REBIRTH Center for Translational and Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Nico Lachmann
- German Center for Lung Research (DZL), Heidelberg, Germany
- Department of Pediatric Pneumology, Allergology and Neonatology and
- REBIRTH Center for Translational and Regenerative Medicine, Hannover Medical School, Hannover, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
- RESIST (Resolving Infection Susceptibility), Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Ulrich Matt
- Department of Internal Medicine V, Universities of Giessen and Marburg Lung Center, Justus Liebig University Giessen, Member of the German Center for Lung Research (DZL), Giessen, Germany
- Institute for Lung Health, Justus Liebig University Giessen, Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute, Giessen, Germany
- German Center for Lung Research (DZL), Heidelberg, Germany
| | - Susanne Herold
- Department of Internal Medicine V, Universities of Giessen and Marburg Lung Center, Justus Liebig University Giessen, Member of the German Center for Lung Research (DZL), Giessen, Germany
- Institute for Lung Health, Justus Liebig University Giessen, Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute, Giessen, Germany
- German Center for Lung Research (DZL), Heidelberg, Germany
| |
Collapse
|
29
|
Corleis B, Bastian M, Hoffmann D, Beer M, Dorhoi A. Animal models for COVID-19 and tuberculosis. Front Immunol 2023; 14:1223260. [PMID: 37638020 PMCID: PMC10451089 DOI: 10.3389/fimmu.2023.1223260] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 07/21/2023] [Indexed: 08/29/2023] Open
Abstract
Respiratory infections cause tremendous morbidity and mortality worldwide. Amongst these diseases, tuberculosis (TB), a bacterial illness caused by Mycobacterium tuberculosis which often affects the lung, and coronavirus disease 2019 (COVID-19) caused by the Severe Acute Respiratory Syndrome Coronavirus type 2 (SARS-CoV-2), stand out as major drivers of epidemics of global concern. Despite their unrelated etiology and distinct pathology, these infections affect the same vital organ and share immunopathogenesis traits and an imperative demand to model the diseases at their various progression stages and localizations. Due to the clinical spectrum and heterogeneity of both diseases experimental infections were pursued in a variety of animal models. We summarize mammalian models employed in TB and COVID-19 experimental investigations, highlighting the diversity of rodent models and species peculiarities for each infection. We discuss the utility of non-human primates for translational research and emphasize on the benefits of non-conventional experimental models such as livestock. We epitomize advances facilitated by animal models with regard to understanding disease pathophysiology and immune responses. Finally, we highlight research areas necessitating optimized models and advocate that research of pulmonary infectious diseases could benefit from cross-fertilization between studies of apparently unrelated diseases, such as TB and COVID-19.
Collapse
Affiliation(s)
- Björn Corleis
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Max Bastian
- Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Donata Hoffmann
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Martin Beer
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Anca Dorhoi
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
- Faculty of Mathematics and Natural Sciences, University of Greifswald, Greifswald, Germany
| |
Collapse
|
30
|
Biswas VK, Sen K, Ahad A, Ghosh A, Verma S, Pati R, Prusty S, Nayak SP, Podder S, Kumar D, Gupta B, Raghav SK. NCoR1 controls Mycobacterium tuberculosis growth in myeloid cells by regulating the AMPK-mTOR-TFEB axis. PLoS Biol 2023; 21:e3002231. [PMID: 37590294 PMCID: PMC10465006 DOI: 10.1371/journal.pbio.3002231] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 08/29/2023] [Accepted: 07/04/2023] [Indexed: 08/19/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb) defends host-mediated killing by repressing the autophagolysosome machinery. For the first time, we report NCoR1 co-repressor as a crucial host factor, controlling Mtb growth in myeloid cells by regulating both autophagosome maturation and lysosome biogenesis. We found that the dynamic expression of NCoR1 is compromised in human peripheral blood mononuclear cells (PBMCs) during active Mtb infection, which is rescued upon prolonged anti-mycobacterial therapy. In addition, a loss of function in myeloid-specific NCoR1 considerably exacerbates the growth of M. tuberculosis in vitro in THP1 differentiated macrophages, ex vivo in bone marrow-derived macrophages (BMDMs), and in vivo in NCoR1MyeKO mice. We showed that NCoR1 depletion controls the AMPK-mTOR-TFEB signalling axis by fine-tuning cellular adenosine triphosphate (ATP) homeostasis, which in turn changes the expression of proteins involved in autophagy and lysosomal biogenesis. Moreover, we also showed that the treatment of NCoR1 depleted cells by Rapamycin, Antimycin-A, or Metformin rescued the TFEB activity and LC3 levels, resulting in enhanced Mtb clearance. Similarly, expressing NCoR1 exogenously rescued the AMPK-mTOR-TFEB signalling axis and Mtb killing. Overall, our data revealed a central role of NCoR1 in Mtb pathogenesis in myeloid cells.
Collapse
Affiliation(s)
- Viplov Kumar Biswas
- Immuno-genomics & Systems Biology Laboratory, Institute of Life Sciences (ILS), Bhubaneswar, India
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Bhubaneswar, India
| | - Kaushik Sen
- Immuno-genomics & Systems Biology Laboratory, Institute of Life Sciences (ILS), Bhubaneswar, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Abdul Ahad
- Immuno-genomics & Systems Biology Laboratory, Institute of Life Sciences (ILS), Bhubaneswar, India
| | - Arup Ghosh
- Immuno-genomics & Systems Biology Laboratory, Institute of Life Sciences (ILS), Bhubaneswar, India
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Bhubaneswar, India
| | - Surbhi Verma
- Molecular Medicine: Cellular Immunology, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Rashmirekha Pati
- Immuno-genomics & Systems Biology Laboratory, Institute of Life Sciences (ILS), Bhubaneswar, India
| | - Subhasish Prusty
- Immuno-genomics & Systems Biology Laboratory, Institute of Life Sciences (ILS), Bhubaneswar, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Sourya Prakash Nayak
- Immuno-genomics & Systems Biology Laboratory, Institute of Life Sciences (ILS), Bhubaneswar, India
| | - Sreeparna Podder
- Immuno-genomics & Systems Biology Laboratory, Institute of Life Sciences (ILS), Bhubaneswar, India
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Bhubaneswar, India
| | - Dhiraj Kumar
- Molecular Medicine: Cellular Immunology, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Bhawna Gupta
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Bhubaneswar, India
| | - Sunil Kumar Raghav
- Immuno-genomics & Systems Biology Laboratory, Institute of Life Sciences (ILS), Bhubaneswar, India
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Bhubaneswar, India
| |
Collapse
|
31
|
Lai R, Gong DN, Williams T, Ogunsola AF, Cavallo K, Lindestam Arlehamn CS, Acolatse S, Beamer GL, Ferris MT, Sassetti CM, Lauffenburger DA, Behar SM. Host genetic background is a barrier to broadly effective vaccine-mediated protection against tuberculosis. J Clin Invest 2023; 133:e167762. [PMID: 37200108 PMCID: PMC10313364 DOI: 10.1172/jci167762] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 05/11/2023] [Indexed: 05/20/2023] Open
Abstract
Heterogeneity in human immune responses is difficult to model in standard laboratory mice. To understand how host variation affects Bacillus Calmette Guerin-induced (BCG-induced) immunity against Mycobacterium tuberculosis, we studied 24 unique collaborative cross (CC) mouse strains, which differ primarily in the genes and alleles they inherit from founder strains. The CC strains were vaccinated with or without BCG and challenged with aerosolized M. tuberculosis. Since BCG protects only half of the CC strains tested, we concluded that host genetics has a major influence on BCG-induced immunity against M. tuberculosis infection, making it an important barrier to vaccine-mediated protection. Importantly, BCG efficacy is dissociable from inherent susceptibility to tuberculosis (TB). T cell immunity was extensively characterized to identify components associated with protection that were stimulated by BCG and recalled after M. tuberculosis infection. Although considerable diversity is observed, BCG has little impact on the composition of T cells in the lung after infection. Instead, variability is largely shaped by host genetics. BCG-elicited protection against TB correlated with changes in immune function. Thus, CC mice can be used to define correlates of protection and to identify vaccine strategies that protect a larger fraction of genetically diverse individuals instead of optimizing protection for a single genotype.
Collapse
Affiliation(s)
- Rocky Lai
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Diana N. Gong
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Travis Williams
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Abiola F. Ogunsola
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Kelly Cavallo
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | | | - Sarah Acolatse
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | | | - Martin T. Ferris
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Christopher M. Sassetti
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Douglas A. Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Samuel M. Behar
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
32
|
Plumlee C, Barrett H, Shao D, Lien K, Cross L, Cohen S, Edlefsen P, Urdahl K. Assessing vaccine-mediated protection in an ultra-low dose Mycobacterium tuberculosis murine model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.22.533820. [PMID: 36993415 PMCID: PMC10055404 DOI: 10.1101/2023.03.22.533820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Despite widespread immunization with Bacille-Calmette-Guerin (BCG), the only currently licensed tuberculosis (TB) vaccine, TB remains a leading cause of mortality globally. There are many TB vaccine candidates in the developmental pipeline, but the lack of a robust animal model to assess vaccine efficacy has hindered our ability to prioritize candidates for human clinical trials. Here we use a murine ultra-low dose (ULD) Mycobacterium tuberculosis (Mtb) challenge model to assess protection conferred by BCG vaccination. We show that BCGconfers a reduction in lung bacterial burdens that is more durable than that observed afterconventional dose challenge, curbs Mtb dissemination to the contralateral lung, and, in a smallpercentage of mice, prevents detectable infection. These findings are consistent with the ability of human BCG vaccination to mediate protection, particularly against disseminated disease, in specific human populations and clinical settings. Overall, our findings demonstrate that the ultra-low dose Mtb infection model can measure distinct parameters of immune protection that cannot be assessed in conventional dose murine infection models and could provide an improved platform for TB vaccine testing.
Collapse
Affiliation(s)
- C.R. Plumlee
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, 98109, USA
| | - H.W. Barrett
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, 98109, USA
- University of Washington, Dept. of Global Health, Seattle, WA, 98109, USA
| | - D.E. Shao
- Vaccine and Infectious Disease Division, Fred Hutch Cancer Center, Seattle, WA, 98109, USA
| | - K.A. Lien
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, 98109, USA
| | - L.M. Cross
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, 98109, USA
| | - S.B. Cohen
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, 98109, USA
| | - P.T Edlefsen
- Vaccine and Infectious Disease Division, Fred Hutch Cancer Center, Seattle, WA, 98109, USA
| | - K.B. Urdahl
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, 98109, USA
- University of Washington, Dept. of Immunology, Seattle, WA, 98109, USA
- University of Washington, Dept. of Pediatrics, Seattle, WA, 98109, USA
- Lead Contact
| |
Collapse
|
33
|
Zheng W, Chang IC, Limberis J, Budzik J, Zha BS, Howard Z, Chen L, Ernst J. Mycobacterium tuberculosis resides in lysosome-poor monocyte-derived lung cells during chronic infection. RESEARCH SQUARE 2023:rs.3.rs-3049913. [PMID: 37398178 PMCID: PMC10312915 DOI: 10.21203/rs.3.rs-3049913/v1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Mycobacterium tuberculosis (Mtb) persists in lung myeloid cells during chronic infection. However, the mechanisms allowing Mtb to evade elimination are not fully understood. Here, we determined that in chronic phase, CD11clo monocyte-derived lung cells termed MNC1 (mononuclear cell subset 1), harbor more live Mtb than alveolar macrophages (AM), neutrophils, and less permissive CD11chi MNC2. Transcriptomic and functional studies of sorted cells revealed that the lysosome biogenesis pathway is underexpressed in MNC1, which have less lysosome content, acidification, and proteolytic activity than AM, and less nuclear TFEB, a master regulator of lysosome biogenesis. Mtb infection does not drive lysosome deficiency in MNC1. Instead, Mtb recruits MNC1 and MNC2 to the lungs for its spread from AM to these cells via its ESX-1 secretion system. The c-Abl tyrosine kinase inhibitor nilotinib activates TFEB and enhances lysosome function of primary macrophages and MNC1 and MNC2 in vivo, improving control of Mtb infection. Our results indicate that Mtb exploits lysosome-poor monocyte-derived cells for in vivo persistence, suggesting a potential target for host-directed tuberculosis therapy.
Collapse
|
34
|
Sun J. A mathematic equation derived from host-pathogen interactions elucidates the significance of integrating modern medicine with traditional Chinese medicine to treat infectious diseases. JOURNAL OF INTEGRATIVE MEDICINE 2023:S2095-4964(23)00046-8. [PMID: 37349214 DOI: 10.1016/j.joim.2023.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 05/12/2023] [Indexed: 06/24/2023]
Abstract
The prognosis of infectious diseases is determined by host-pathogen interactions. Control of pathogens has been the central dogma of treating infectious diseases in modern medicine, but the pathogen-directed medicine is facing significant challenges, including a lack of effective antimicrobials for newly emerging pathogens, pathogen drug resistance, and drug side effects. Here, a mathematic equation (termed equation of host-pathogen interactions, HPI-Equation) is developed to dissect the key variables of host-pathogen interactions. It shows that control of pathogens does not necessarily lead to host recovery. Instead, a combination of promoting a host's power of self-healing and balancing immune responses provides the best benefit for host. Moreover, the HPI-Equation elucidates the scientific basis of traditional Chinese medicine (TCM), a host-based medicine that treats infectious diseases by promoting self-healing power and balancing immune responses. The importance of self-healing power elucidated in the HPI-Equation is confirmed by recent studies that the tolerance mechanism, which is discovered in plants and animals and conceptually similar to self-healing power, improves host survival without directly attacking pathogens. In summary, the HPI-Equation describes host-pathogen interactions with mathematical logic and precision; it translates the ancient wisdoms of TCM into apprehensible modern sciences and opens a new venue for integrating TCM and modern medicine for a future medicine.
Collapse
Affiliation(s)
- Jianjun Sun
- Department of Biological Sciences, Border Biomedical Research Center, University of Texas at El Paso, TX 79968, USA.
| |
Collapse
|
35
|
Zheng W, Chang IC, Limberis J, Budzik JM, Zha BS, Howard Z, Chen L, Ernst JD. Mycobacterium tuberculosis resides in lysosome-poor monocyte-derived lung cells during chronic infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.19.524758. [PMID: 36711606 PMCID: PMC9882350 DOI: 10.1101/2023.01.19.524758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Mycobacterium tuberculosis (Mtb) infects cells in multiple lung myeloid cell subsets and causes chronic infection despite innate and adaptive immune responses. However, the mechanisms allowing Mtb to evade elimination are not fully understood. Here, using new methods, we determined that after T cell responses have developed, CD11clo monocyte-derived lung cells termed MNC1 (mononuclear cell subset 1), harbor more live Mtb compared to alveolar macrophages (AM), neutrophils, and less permissive CD11chi MNC2. Bulk RNA sequencing of sorted cells revealed that the lysosome biogenesis pathway is underexpressed in MNC1. Functional assays confirmed that Mtb-permissive MNC1 have less lysosome content, acidification, and proteolytic activity than AM, and less nuclear TFEB, a master regulator of lysosome biogenesis. Mtb infection does not drive lysosome deficiency in MNC1 in vivo. Instead, Mtb recruits MNC1 and MNC2 to the lungs for its spread from AM to these cell subsets as a virulence mechanism that requires the Mtb ESX-1 secretion system. The c-Abl tyrosine kinase inhibitor nilotinib activates TFEB and enhances lysosome function of primary macrophages in vitro and MNC1 and MNC2 in vivo, improving control of Mtb infection. Our results indicate that Mtb exploits lysosome-poor monocyte-derived cells for in vivo persistence, suggesting a potential target for host-directed tuberculosis therapy.
Collapse
Affiliation(s)
- Weihao Zheng
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, California, USA
| | - I-Chang Chang
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, California, USA
| | - Jason Limberis
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, California, USA
| | - Jonathan M. Budzik
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, California, USA
| | - B. Shoshana Zha
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, California, USA
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, California, USA
| | - Zach Howard
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, California, USA
| | - Lucas Chen
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, California, USA
| | - Joel D. Ernst
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, California, USA
| |
Collapse
|
36
|
Larson EC, Ellis-Connell AL, Rodgers MA, Gubernat AK, Gleim JL, Moriarty RV, Balgeman AJ, Ameel CL, Jauro S, Tomko JA, Kracinovsky KB, Maiello P, Borish HJ, White AG, Klein E, Bucsan AN, Darrah PA, Seder RA, Roederer M, Lin PL, Flynn JL, O'Connor SL, Scanga CA. Vaccination with intravenous BCG protects macaques with pre-existing SIV infection from tuberculosis. RESEARCH SQUARE 2023:rs.3.rs-2802306. [PMID: 37090620 PMCID: PMC10120779 DOI: 10.21203/rs.3.rs-2802306/v1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Tuberculosis (TB) is the most common cause of death in people living with HIV. BCG delivered intradermally (ID) is the only licensed vaccine to prevent TB. However, it offers little protection from pulmonary TB in adults. Intravenous (IV) BCG, but not ID BCG, confers striking protection against Mycobacterium tuberculosis (Mtb) infection and disease in rhesus macaques. We investigated whether IV BCG could protect against TB in macaques with a pre-existing SIV infection. There was a robust influx of airway T cells following IV BCG in both SIV-infected and SIV-naïve animals, with elevated antibody titers in plasma and airways. Following Mtb challenge, all 7 SIV-naïve and 9 out of 12 SIV-infected vaccinated animals were completely protected, without any culturable bacilli in their tissues. PBMC responses post-challenge indicated early clearance of Mtb in vaccinated animals regardless of SIV infection. These data support that IV BCG is immunogenic and efficacious in SIV-infected animals.
Collapse
Affiliation(s)
- Erica C Larson
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
- Center for Vaccine Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Amy L Ellis-Connell
- Department of Pathology and Laboratory Medicine, University of Wisconsin - Madison, Madison, WI, USA
| | - Mark A Rodgers
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Abigail K Gubernat
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Janelle L Gleim
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Ryan V Moriarty
- Department of Pathology and Laboratory Medicine, University of Wisconsin - Madison, Madison, WI, USA
| | - Alexis J Balgeman
- Department of Pathology and Laboratory Medicine, University of Wisconsin - Madison, Madison, WI, USA
| | - Cassaundra L Ameel
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Solomon Jauro
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Jaime A Tomko
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Kara B Kracinovsky
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Pauline Maiello
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - H Jake Borish
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Alexander G White
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Edwin Klein
- Division of Laboratory Animal Resources, School of Medicine, University of Pittsburgh, PA, USA
| | - Allison N Bucsan
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Patricia A Darrah
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Philana Ling Lin
- Department of Pediatrics, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - JoAnne L Flynn
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
- Center for Vaccine Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Shelby L O'Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin - Madison, Madison, WI, USA
- Wisconsin National Primate Research Center, University of Wisconsin - Madison, Madison, WI, USA
| | - Charles A Scanga
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
- Center for Vaccine Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
37
|
Abstract
Tuberculosis (TB) remains the leading cause of bacterial disease-related death and is among the top 10 overall causes of death worldwide. The complex nature of this infectious lung disease has proven difficult to treat, and significant research efforts are now evaluating the feasibility of host-directed, adjunctive therapies. An attractive approach in host-directed therapy targets host epigenetics, or gene regulation, to redirect the immune response in a host-beneficial manner. Substantial evidence exists demonstrating that host epigenetics are dysregulated during TB and that epigenetic-based therapies may be highly effective to treat TB. However, the caveat is that much of the knowledge that exists on the modulation of the host epigenome during TB has been gained using in vitro, small-animal, or blood-derived cell models, which do not accurately reflect the pulmonary nature of the disease. In humans, the first and major target cells of Mycobacterium tuberculosis are alveolar macrophages (AM). As such, their response to infection and treatment is clinically relevant and ultimately drives the outcome of disease. In this review, we compare the fundamental differences between AM and circulating monocyte-derived macrophages in the context of TB and summarize the recent advances in elucidating the epigenomes of these cells, including changes to the transcriptome, DNA methylome, and chromatin architecture. We will also discuss trained immunity in AM as a new and emerging field in TB research and provide some perspectives for the translational potential of targeting host epigenetics as an alternative TB therapy.
Collapse
|
38
|
Larsen SE, Williams BD, Rais M, Coler RN, Baldwin SL. It Takes a Village: The Multifaceted Immune Response to Mycobacterium tuberculosis Infection and Vaccine-Induced Immunity. Front Immunol 2022; 13:840225. [PMID: 35359957 PMCID: PMC8960931 DOI: 10.3389/fimmu.2022.840225] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/08/2022] [Indexed: 11/18/2022] Open
Abstract
Despite co-evolving with humans for centuries and being intensely studied for decades, the immune correlates of protection against Mycobacterium tuberculosis (Mtb) have yet to be fully defined. This lapse in understanding is a major lag in the pipeline for evaluating and advancing efficacious vaccine candidates. While CD4+ T helper 1 (TH1) pro-inflammatory responses have a significant role in controlling Mtb infection, the historically narrow focus on this cell population may have eclipsed the characterization of other requisite arms of the immune system. Over the last decade, the tuberculosis (TB) research community has intentionally and intensely increased the breadth of investigation of other immune players. Here, we review mechanistic preclinical studies as well as clinical anecdotes that suggest the degree to which different cell types, such as NK cells, CD8+ T cells, γ δ T cells, and B cells, influence infection or disease prevention. Additionally, we categorically outline the observed role each major cell type plays in vaccine-induced immunity, including Mycobacterium bovis bacillus Calmette-Guérin (BCG). Novel vaccine candidates advancing through either the preclinical or clinical pipeline leverage different platforms (e.g., protein + adjuvant, vector-based, nucleic acid-based) to purposefully elicit complex immune responses, and we review those design rationales and results to date. The better we as a community understand the essential composition, magnitude, timing, and trafficking of immune responses against Mtb, the closer we are to reducing the severe disease burden and toll on human health inflicted by TB globally.
Collapse
Affiliation(s)
- Sasha E. Larsen
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States
| | - Brittany D. Williams
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States,Department of Global Health, University of Washington, Seattle, WA, United States
| | - Maham Rais
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States
| | - Rhea N. Coler
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States,Department of Global Health, University of Washington, Seattle, WA, United States,Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, United States
| | - Susan L. Baldwin
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States,*Correspondence: Susan L. Baldwin,
| |
Collapse
|
39
|
Tepekule B, Kusejko K, Zeeb M, Tarr PE, Calmy A, Battegay M, Furrer H, Cavassini M, Bernasconi E, Notter J, Günthard HF, Nemeth J, Kouyos RD. Impact of Latent Tuberculosis Infection on the Incidence of Type 2 Diabetes Mellitus in HIV-Infected Participants in the Swiss HIV Cohort Study. J Infect Dis 2022; 225:2229-2234. [PMID: 35172332 DOI: 10.1093/infdis/jiac054] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 02/14/2022] [Indexed: 11/13/2022] Open
Abstract
While an increased risk of active and latent tuberculosis infection (LTBI) in people with type-2 diabetes (DM) has been demonstrated, it is less well characterized whether LTBI is associated with an increased risk of developing DM. We investigated the link between LTBI and DM in people living with HIV (PHIV) in the Swiss HIV Cohort Study via time-dependent cox proportional hazards models. We found that LTBI significantly increased the risk of developing DM (HR=1.47), which was robust across different adjustment and censoring techniques. Our results thus suggest that LTBI may be associated with an increased risk of developing DM.
Collapse
Affiliation(s)
- Burcu Tepekule
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Switzerland.,Institute of Medical Virology, University of Zurich, Switzerland
| | - Katharina Kusejko
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Switzerland.,Institute of Medical Virology, University of Zurich, Switzerland
| | - Marius Zeeb
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Switzerland.,Institute of Medical Virology, University of Zurich, Switzerland
| | - Philip E Tarr
- Department of Medicine and Division of Infectious Diseases and Hospital Epidemiology, Kantonsspital Baselland, University of Basel, Bruderholz, Switzerland
| | - Alexandra Calmy
- Division of Infectious Diseases, University Hospital Geneva, University of Geneva, Geneva, Switzerland
| | - Manuel Battegay
- Division of Infectious Diseases & Hospital Hygiene, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Hansjakob Furrer
- Department of Infectious Diseases, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Matthias Cavassini
- Division of Infectious Diseases, University Hospital, Lausanne, Switzerland
| | - Enos Bernasconi
- Division of Infectious Diseases, Regional Hospital Lugano, Lugano, Switzerland
| | - Julia Notter
- Cantonal Hospital St Gallen, Division of Infectious Diseases and Hospital Epidemiology, St Gallen, Switzerland
| | - Huldrych F Günthard
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Switzerland
| | - Johannes Nemeth
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Switzerland
| | - Roger D Kouyos
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Switzerland.,Institute of Medical Virology, University of Zurich, Switzerland
| | | |
Collapse
|
40
|
Abstract
Pulmonary granulomas are widely considered the epicenters of the immune response to Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB). Recent animal studies have revealed factors that either promote or restrict TB immunity within granulomas. These models, however, typically ignore the impact of preexisting immunity on cellular organization and function, an important consideration because most TB probably occurs through reinfection of previously exposed individuals. Human postmortem research from the pre-antibiotic era showed that infections in Mtb-naïve individuals (primary TB) versus those with prior Mtb exposure (postprimary TB) have distinct pathologic features. We review recent animal findings in TB granuloma biology, which largely reflect primary TB. We also discuss our current understanding of postprimary TB lesions, about which much less is known. Many knowledge gaps remain, particularly regarding how preexisting immunity shapes granuloma structure and local immune responses at Mtb infection sites. Expected final online publication date for the Annual Review of Immunology, Volume 40 is April 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Sara B. Cohen
- Seattle Children's Research Institute, Seattle, Washington, USA
| | - Benjamin H. Gern
- Seattle Children's Research Institute, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Kevin B. Urdahl
- Seattle Children's Research Institute, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
- Department of Immunology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
41
|
Abstract
Alveolar macrophages (AMs) are lung-resident myeloid cells that sit at the interface of the airway and lung tissue. Under homeostatic conditions, their primary function is to clear debris, dead cells and excess surfactant from the airways. They also serve as innate pulmonary sentinels for respiratory pathogens and environmental airborne particles and as regulators of pulmonary inflammation. However, they have not typically been viewed as primary therapeutic targets for respiratory diseases. Here, we discuss the role of AMs in various lung diseases, explore the potential therapeutic strategies to target these innate cells and weigh the potential risks and challenges of such therapies. Additionally, in the context of the COVID-19 pandemic, we examine the role AMs play in severe disease and the therapeutic strategies that have been harnessed to modulate their function and protect against severe lung damage. There are many novel approaches in development to target AMs, such as inhaled antibiotics, liposomal and microparticle delivery systems, and host-directed therapies, which have the potential to provide critical treatment to patients suffering from severe respiratory diseases, yet there is still much work to be done to fully understand the possible benefits and risks of such approaches.
Collapse
|
42
|
Sher A, Flynn JL. Sterilizing immunity: New opportunities for rational TB vaccine design. J Exp Med 2021; 218:212188. [PMID: 34037668 PMCID: PMC8160586 DOI: 10.1084/jem.20210454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023] Open
Abstract
Recent studies have revealed situations of high-level naturally acquired and vaccine-induced immunity against Mycobacterium tuberculosis in animal models along with examples of significantly protective immunization in humans. These discoveries offer immunologists new opportunities to define effector mechanisms that when triggered by appropriately engineered vaccines could end TB's deadly reign.
Collapse
Affiliation(s)
- Alan Sher
- Laboratory of Parasitic Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Joanne L. Flynn
- Department of Microbiology and Molecular Genetics and Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
43
|
Wong EB. It Is Time to Focus on Asymptomatic Tuberculosis. Clin Infect Dis 2021; 72:e1044-e1046. [PMID: 33283223 PMCID: PMC8204778 DOI: 10.1093/cid/ciaa1827] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 12/03/2020] [Indexed: 01/26/2023] Open
Affiliation(s)
- Emily B Wong
- Africa Health Research Institute, Durban, KwaZulu-Natal, South Africa.,Division of Infectious Diseases, Department of Medicine, University of Alabama-Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
44
|
van Gisbergen KPJM, Zens KD, Münz C. T-cell memory in tissues. Eur J Immunol 2021; 51:1310-1324. [PMID: 33837521 DOI: 10.1002/eji.202049062] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/01/2021] [Accepted: 04/07/2021] [Indexed: 12/15/2022]
Abstract
Immunological memory equips our immune system to respond faster and more effectively against reinfections. This acquired immunity was originally attributed to long-lived, memory T and B cells with body wide access to peripheral and secondary lymphoid tissues. In recent years, it has been realized that both innate and adaptive immunity to a large degree depends on resident immune cells that act locally in barrier tissues including tissue-resident memory T cells (Trm). Here, we will discuss the phenotype of these Trm in mice and humans, the tissues and niches that support them, and their function, plasticity, and transcriptional control. Their unique properties enable Trm to achieve long-lived immunological memory that can be deposited in nearly every organ in response to acute and persistent infection, and in response to cancer. However, Trm may also induce substantial immunopathology in allergic and autoimmune disease if their actions remain unchecked. Therefore, inhibitory and activating stimuli appear to balance the actions of Trm to ensure rapid proinflammatory responses upon infection and to prevent damage to host tissues under steady state conditions.
Collapse
Affiliation(s)
- Klaas P J M van Gisbergen
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Kyra D Zens
- Viral Immunobiology, University of Zurich, Zurich, Switzerland.,Department of Public and Global Health, Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Zurich, Switzerland.,Department of Infectious Diseases and Hospital Epidemiology, University Hospital, Zurich, Switzerland
| | - Christian Münz
- Viral Immunobiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
45
|
Duffy FJ, Olson GS, Gold ES, Jahn A, Aderem A, Aitchison JD, Rothchild AC, Diercks AH, Nemeth J. Use of a Contained Mycobacterium tuberculosis Mouse Infection Model to Predict Active Disease and Containment in Humans. J Infect Dis 2021; 225:1832-1840. [PMID: 33693706 PMCID: PMC9113476 DOI: 10.1093/infdis/jiab130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 03/08/2021] [Indexed: 12/13/2022] Open
Abstract
Previous studies have identified whole-blood transcriptional risk and disease signatures for tuberculosis; however, several lines of evidence suggest that these signatures primarily reflect bacterial burden, which increases before symptomatic disease. We found that the peripheral blood transcriptome of mice with contained Mycobacterium tuberculosis infection (CMTI) has striking similarities to that of humans with active tuberculosis and that a signature derived from these mice predicts human disease with accuracy comparable to that of signatures derived directly from humans. A set of genes associated with immune defense are up-regulated in mice with CMTI but not in humans with active tuberculosis, suggesting that their up-regulation is associated with bacterial containment. A signature comprising these genes predicts both protection from tuberculosis disease and successful treatment at early time points where current signatures are not predictive. These results suggest that detailed study of the CMTI model may enable identification of biomarkers for human tuberculosis.
Collapse
Affiliation(s)
- Fergal J Duffy
- Seattle Children’s Research Institute, Seattle, Washington, USA,Correspondence: Fergal J. Duffy, Center for Global Infectious Disease Research, Seattle Children’s Research Institute, 307 Westlake Ave N #500, Seattle, WA 98109, USA ()
| | - Gregory S Olson
- Seattle Children’s Research Institute, Seattle, Washington, USA
| | | | - Ana Jahn
- Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Alan Aderem
- Seattle Children’s Research Institute, Seattle, Washington, USA
| | | | - Alissa C Rothchild
- Seattle Children’s Research Institute, Seattle, Washington, USA,Present affiliation: Department of Veterinary and Animal Sciences, University of Massachusetts Amherst
| | - Alan H Diercks
- Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Johannes Nemeth
- Seattle Children’s Research Institute, Seattle, Washington, USA,Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich,Switzerland
| |
Collapse
|
46
|
Huaman MA, Juchnowski SM, Zidar DA, Kityo CM, Nalukwago S, Nazzinda R, Fichtenbaum CJ, Longenecker CT. Monocyte activation in persons living with HIV and tuberculosis coinfection. AIDS 2021; 35:447-452. [PMID: 33252496 PMCID: PMC7855758 DOI: 10.1097/qad.0000000000002766] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES To characterize monocyte subsets and activation in persons living with HIV (PLWH) with tuberculosis coinfection. DESIGN Cross-sectional study within a cohort of PLWH and HIV-uninfected participants at the Joint Clinical Research Centre in Kampala, Uganda. METHODS Participants were at least 45 years old with at least one cardiovascular risk factor. PLWH had an HIV viral load 1000 copies/ml or less on stable antiretroviral therapy prior to cohort entry. QuantiFERON-TB testing was performed to define latent tuberculosis infection (LTBI). Prior active TB was defined by self-report and verified by medical records. Blood was stained with monocyte subset markers (CD14+, CD16), CD62p, CD69, CX3CR1, HLA-DR, and tissue factor, and examined with flow cytometry. RESULTS One hundred and twenty-five participants (83 PLWH and 42 without HIV) were included. Median CD4+ count was 582 cells/μl in PLWH. PLWH had a higher frequency of total monocytes (4.3% vs. 3.2%; P < 0.001) and inflammatory monocyte subset (15.5% vs. 11.7%; P = 0.016) compared with HIV-uninfected individuals. No differences in the frequency of monocyte subsets were observed by TB status. Among PLWH, prior active TB was associated with increased frequency of total monocytes compared with LTBI (5.1% vs. 3.7%; P = 0.013). HLA-DR density on monocytes was three-fold higher in PLWH with LTBI or prior TB compared with PLWH without LTBI (P = 0.002). In multivariate analysis, a higher monocyte HLA-DR density remained associated with LTBI or prior TB in PLWH (log-MFI; b = 1.17; P < 0.001). CONCLUSION Our findings indicate enhanced monocyte activation in PLWH with LTBI or prior active TB, which may contribute to the pathogenesis of noncommunicable diseases in HIV.
Collapse
Affiliation(s)
- Moises A Huaman
- Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati
| | | | - David A Zidar
- Case Western Reserve University School of Medicine
- Louis Stokes Cleveland Veterans Affairs Medical Center
- University Hospitals Harrington Heart & Vascular Institute, Cleveland, Ohio, USA
| | | | | | | | - Carl J Fichtenbaum
- Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati
| | - Chris T Longenecker
- Case Western Reserve University School of Medicine
- University Hospitals Harrington Heart & Vascular Institute, Cleveland, Ohio, USA
| |
Collapse
|
47
|
Huaman MA, De Cecco CN, Bittencourt MS, Ticona E, Kityo C, Ballena I, Nalukwago S, Nazzinda R, Ticona C, Azañero R, Zhang B, Farquhar C, Hawn TR, Sterling TR, Fichtenbaum CJ, Longenecker CT. Latent Tuberculosis Infection and Subclinical Coronary Atherosclerosis in Peru and Uganda. Clin Infect Dis 2021; 73:e3384-e3390. [PMID: 33388766 DOI: 10.1093/cid/ciaa1934] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Tuberculosis has been linked to an increased risk of atherosclerotic cardiovascular disease (ASCVD). We assessed whether latent tuberculosis infection (LTBI) is associated with subclinical coronary atherosclerosis in two TB-prevalent areas. METHODS We analyzed cross-sectional data from studies conducted in Lima, Peru, and Kampala, Uganda. Individuals ≥40 years old were included. We excluded persons with known history of ASCVD events or active TB. Participants underwent QuantiFERON®-TB (QFT) testing to define LTBI, and computed tomography angiography to examine coronary atherosclerosis. A Coronary Artery Disease-Reporting Data System (CAD-RADS) score ≥3 defined obstructive CAD (plaque causing ≥50% stenosis). RESULTS 113 persons with LTBI and 91 persons without LTBI were included. There were no significant differences between LTBI and non-LTBI participants in terms of age (median [interquartile range]; 56 [51-62] vs. 55 [49-64], p=0.829), male sex (38% vs. 42%; p=0.519), or 10-year ASCVD risk scores (7.1 [3.2-11.7] vs. 6.1 [2.8-10.8]; p=0.533). CAD prevalence (any plaque) was similar between groups (29% vs. 24%; p=0.421). Obstructive CAD was present in 9% of LTBI and 3% of non-LTBI individuals; p=0.095. LTBI was associated with obstructive CAD after adjusting for ASCVD risk score, HIV status, and study site (adjusted odds ratio, 4.96, 95% CI 1.05-23.44; p=0.043). Quantitative QFT TB antigen minus nil interferon-gamma responses were associated with obstructive CAD (adjusted odds ratio, 1.2, 95% CI 1.03-1.41; p=0.022). CONCLUSIONS LTBI was independently associated with an increased likelihood of subclinical obstructive CAD. Our data indicates that LTBI is a non-traditional correlate of ASCVD risk.
Collapse
Affiliation(s)
- Moises A Huaman
- Department of Internal Medicine, Division of Infectious Diseases, University of Cincinnati College of Medicine, Ohio, United States of America
| | - Carlo N De Cecco
- Division of Cardiothoracic Imaging, Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, United States of America
| | | | - Eduardo Ticona
- Hospital Nacional Dos de Mayo, Lima, Peru.,Department of Internal Medicine, Universidad Nacional Mayor de San Marcos, Lima, Peru
| | - Cissy Kityo
- Joint Clinical Research Centre, Kampala, Uganda
| | | | | | | | | | | | - Bin Zhang
- Division of Biostatistics and Epidemiology, Cincinnati Children's Medical Center, Ohio, United States of America
| | - Carey Farquhar
- Departments of Medicine and Global Health, University of Washington School of Medicine, Seattle, United States of America
| | - Thomas R Hawn
- Departments of Medicine and Global Health, University of Washington School of Medicine, Seattle, United States of America
| | - Timothy R Sterling
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University School of Medicine, United States of America
| | - Carl J Fichtenbaum
- Department of Internal Medicine, Division of Infectious Diseases, University of Cincinnati College of Medicine, Ohio, United States of America
| | - Chris T Longenecker
- Harrington Heart & Vascular Institute, Case Western Reserve University School of Medicine, Ohio, United States of America
| |
Collapse
|
48
|
Diagnosis of latent tuberculosis infection is associated with reduced HIV viral load and lower risk for opportunistic infections in people living with HIV. PLoS Biol 2020; 18:e3000963. [PMID: 33284802 PMCID: PMC7721132 DOI: 10.1371/journal.pbio.3000963] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 11/02/2020] [Indexed: 11/23/2022] Open
Abstract
Approximately 28% of the human population have been exposed to Mycobacterium tuberculosis (MTB), with the overwhelming majority of infected individuals not developing disease (latent TB infection (LTBI)). While it is known that uncontrolled HIV infection is a major risk factor for the development of TB, the effect of underlying LTBI on HIV disease progression is less well characterized, in part because longitudinal data are lacking. We sorted all participants of the Swiss HIV Cohort Study (SHCS) with at least 1 documented MTB test into one of the 3 groups: MTB uninfected, LTBI, or active TB. To detect differences in the HIV set point viral load (SPVL), linear regression was used; the frequency of the most common opportunistic infections (OIs) in the SHCS between MTB uninfected patients, patients with LTBI, and patients with active TB were compared using logistic regression and time-to-event analyses. In adjusted models, we corrected for baseline demographic characteristics, i.e., HIV transmission risk group and gender, geographic region, year of HIV diagnosis, and CD4 nadir. A total of 13,943 SHCS patients had at least 1 MTB test documented, of whom 840 (6.0%) had LTBI and 770 (5.5%) developed active TB. Compared to MTB uninfected patients, LTBI was associated with a 0.24 decreased log HIV SPVL in the adjusted model (p < 0.0001). Patients with LTBI had lower odds of having candida stomatitis (adjusted odds ratio (OR) = 0.68, p = 0.0035) and oral hairy leukoplakia (adjusted OR = 0.67, p = 0.033) when compared to MTB uninfected patients. The association of LTBI with a reduced HIV set point virus load and fewer unrelated infections in HIV/TB coinfected patients suggests a more complex interaction between LTBI and HIV than previously assumed. Surprisingly little is known about how latent tuberculosis infection alters human physiology and immune function. Extensive statistical analyses of the large Swiss HIV Cohort Study suggests that latent tuberculosis infection can be protective in individuals with HIV.
Collapse
|