1
|
Yabaji SM, Zhernovkov V, Araveti PB, Lata S, Rukhlenko OS, Abdullatif SA, Vanvalkenburg A, Alekseev Y, Ma Q, Dayama G, Lau NC, Johnson WE, Bishai WR, Crossland NA, Campbell JD, Kholodenko BN, Gimelbrant AA, Kobzik L, Kramnik I. Lipid Peroxidation and Type I Interferon Coupling Fuels Pathogenic Macrophage Activation Causing Tuberculosis Susceptibility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.03.05.583602. [PMID: 38496444 PMCID: PMC10942339 DOI: 10.1101/2024.03.05.583602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
A quarter of human population is infected with Mycobacterium tuberculosis, but less than 10% of those infected develop pulmonary TB. We developed a genetically defined sst1-susceptible mouse model that uniquely reproduces a defining feature of human TB: the development of necrotic lung granulomas and determined that the sst1-susceptible phenotype was driven by the aberrant macrophage activation. This study demonstrates that the aberrant response of the sst1-susceptible macrophages to prolonged stimulation with TNF is primarily driven by conflicting Myc and antioxidant response pathways leading to a coordinated failure 1) to properly sequester intracellular iron and 2) to activate ferroptosis inhibitor enzymes. Consequently, iron-mediated lipid peroxidation fueled Ifn-beta superinduction and sustained the Type I Interferon (IFN-I) pathway hyperactivity that locked the sst1-susceptible macrophages in a state of unresolving stress and compromised their resistance to Mtb. The accumulation of the aberrantly activated, stressed, macrophages within granuloma microenvironment led to the local failure of anti-tuberculosis immunity and tissue necrosis. The upregulation of Myc pathway in peripheral blood cells of human TB patients was significantly associated with poor outcomes of TB treatment. Thus, Myc dysregulation in activated macrophages results in an aberrant macrophage activation and represents a novel target for host-directed TB therapies.
Collapse
|
2
|
Tian N, Chu H, Li Q, Sun H, Zhang J, Chu N, Sun Z. Host-directed therapy for tuberculosis. Eur J Med Res 2025; 30:267. [PMID: 40211397 PMCID: PMC11987284 DOI: 10.1186/s40001-025-02443-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 03/09/2025] [Indexed: 04/13/2025] Open
Abstract
Current TB treatment regimens are hindered by drug resistance, numerous adverse effects, and long treatment durations, highlighting the need for 'me-better' treatment regimens. Host-directed therapy (HDT) has gained recognition as a promising approach in TB treatment. It allows the repurposing of existing drugs approved for other conditions and aims to enhance the effectiveness of existing anti-TB therapies, minimize drug resistance, decrease treatment duration, and adverse effects. By modulating the host immune response, HDT ameliorates immunopathological damage and improves overall outcomes by promoting autophagy, antimicrobial peptide production, and other mechanisms. It holds promise for addressing the challenges posed by multiple and extensively drug-resistant Mycobacterium tuberculosis strains, which are increasingly difficult to treat using conventional therapies. This article reviews various HDT candidates, including repurposed drugs, explores their underlying mechanisms such as autophagy promotion and inflammation reduction, while emphasizing their potential to improve TB treatment outcomes and outlining future research directions.
Collapse
Affiliation(s)
- Na Tian
- Department of Tuberculosis, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Hongqian Chu
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
| | - Qi Li
- Department of Tuberculosis, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Hong Sun
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
| | - Jingfang Zhang
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
| | - Naihui Chu
- Department of Tuberculosis, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China.
| | - Zhaogang Sun
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China.
| |
Collapse
|
3
|
Mhlanga MM, Fanucchi S, Ozturk M, Divangahi M. Cellular and Molecular Mechanisms of Innate Memory Responses. Annu Rev Immunol 2025; 43:615-640. [PMID: 40279311 DOI: 10.1146/annurev-immunol-101721-035114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2025]
Abstract
There has been an increasing effort to understand the memory responses of a complex interplay among innate, adaptive, and structural cells in peripheral organs and bone marrow. Trained immunity is coined as the de facto memory of innate immune cells and their progenitors. These cells acquire epigenetic modifications and shift their metabolism to equip an imprinted signature to a persistent fast-responsive functional state. Recent studies highlight the contribution of noncoding RNAs and modulation of chromatin structures in establishing this epigenetic readiness for potential immune perturbations. In this review, we discuss recent studies that highlight trained immunity-mediated memory responses emerging intrinsically in innate immune cells and as a complex interplay with other cells at the organ level. Lastly, we survey epigenetic contributors to trained immunity phenotypes-specifically, a recently discovered regulatory circuit coordinating the regulation of a key driver of trained immunity.
Collapse
Affiliation(s)
- Musa M Mhlanga
- Epigenomics & Single Cell Biophysics Group, Department of Cell Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University, Nijmegen, The Netherlands;
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Mumin Ozturk
- Epigenomics & Single Cell Biophysics Group, Department of Cell Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University, Nijmegen, The Netherlands;
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Maziar Divangahi
- Departments of Medicine, Pathology, and Microbiology & Immunology, McGill University, Montreal, Quebec, Canada
- McGill University Health Centre, McGill International TB Centre, and Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada;
| |
Collapse
|
4
|
Lee OV, Ji DX, Rosa BA, Jaye DL, Suliman S, Mitreva M, Gabay C, Vance RE, Kotov DI. Interleukin-1 receptor antagonist is a conserved early factor for exacerbating tuberculosis susceptibility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.10.27.564420. [PMID: 37961447 PMCID: PMC10634924 DOI: 10.1101/2023.10.27.564420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Mycobacterium tuberculosis (Mtb) causes 1.25 million deaths a year. However, tuberculosis (TB) pathogenesis remains poorly understood and is not fully recapitulated in standard mouse models. Here we find that gene signatures from three different Mtb-susceptible mouse models predict active TB disease in humans significantly better than a signature from resistant C57BL/6 (B6) mice. Conserved among susceptible mice, non-human primates, and humans, but largely absent from B6 mice, was Mtb-induced differentiation of macrophages into an Spp1 + differentiation state. Spp1 + macrophages expressed high levels of immunosuppressive molecules including IL-1 receptor antagonist (IL-1Ra). IL-1Ra was previously reported to cause Mtb susceptibility in one mouse model, but whether IL-1Ra is broadly important remains uncertain. Here we report that enhancement of IL-1 signaling via deletion of IL-Ra promoted bacterial control across three susceptible mouse models. We found IL-1 signaling amplified production of multiple cytokines by lymphoid and stromal cells, providing a multifactorial mechanism for how IL-1 promotes Mtb control. Our results indicate that myeloid cell expression of immunosuppressive molecules, in particular IL-1 receptor antagonist, is a conserved early mechanism limiting Mtb control in mice, non-human primates, and humans.
Collapse
Affiliation(s)
- Ophelia V. Lee
- Divison of Immunology and Molecular Medicine, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Daisy X. Ji
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Bruce A. Rosa
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - David L. Jaye
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Sara Suliman
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, 94115, USA
| | - Makedonka Mitreva
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Cem Gabay
- Division of Rheumatology, Department of Medicine, University Hospital of Geneva, Geneva, Switzerland
| | - Russell E. Vance
- Divison of Immunology and Molecular Medicine, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Dmitri I. Kotov
- Division of Infectious Diseases, Department of Medicine, Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, 63110, USA
| |
Collapse
|
5
|
Rajamanickam A, Ann Daniel E, Dasan B, Thiruvengadam K, Chandrasekaran P, Gaikwad S, Pattabiraman S, Bhanu B, Sivaprakasam A, Kulkarni V, Karyakarte R, Paradkar M, Shivakumar SVBY, Mave V, Gupta A, Hanna LE, Babu S. Plasma Immune Biomarkers Predictive of Progression to Active Tuberculosis in Household Contacts of Patients With Tuberculosis. J Infect Dis 2025; 231:696-705. [PMID: 39028003 PMCID: PMC11911786 DOI: 10.1093/infdis/jiae365] [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: 01/09/2024] [Revised: 07/08/2024] [Accepted: 07/18/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND The progression from Mycobacterium tuberculosis infection to active tuberculosis disease varies among individuals, and identifying biomarkers to predict progression is crucial for guiding interventions. In this study, we aimed to determine plasma immune biomarker profiles in healthy household contacts of index patients with pulmonary tuberculosis, who either progressed to tuberculosis or remained as nonprogressors. METHODS A cohort of household contacts of adults with pulmonary tuberculosis was enrolled, consisting of 15 contacts who progressed to tuberculosis disease and 15 nonprogressors. Plasma samples were collected at baseline, 4 months, and 12 months to identify predictive tuberculosis progression markers. RESULTS Our findings revealed that individuals in the progressor group exhibited significantly decreased levels of interferon (IFN) γ, tumor necrosis factor α, interleukin 2, IL-1α, IL-1β, and 17A, and interleukin 1 receptor antagonist (IL-1Ra) at baseline, month 4, and month 12. In contrast, the progressor group displayed significantly elevated levels of IFN-α, IFN-β, interleukin 6 and 12, granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin 10 (IL-10) and 33 (IL-33), CCL2, CCL11, CXCL8, CXCL10, CX3CL1, vascular endothelial growth factor, granzyme B, and programmed death ligand -1 compared to the nonprogressor group at baseline, months 4 and 12. Receiver operating characteristic analysis (ROC) identified IFN-γ, GM-CSF, IL-1Ra, CCL2, and CXCL10 as the most promising predictive markers, with an area under the receiver operating characteristic curve of ≥90. Furthermore, combinatorial analysis demonstrated that GM-CSF, CXCL10, and IL-1Ra, when used in combination, exhibited high accuracy in predicting progression to active tuberculosis disease. CONCLUSIONS Our study suggests that a specific set of plasma biomarkers, GM-CSF, CXCL10, and IL-1Ra, can effectively identify household contacts at significant risk of developing tuberculosis disease. These findings have important implications for early intervention and preventive strategies in tuberculosis-endemic regions.
Collapse
Affiliation(s)
- Anuradha Rajamanickam
- National Institute of Health-National Institute of Allergy and Infectious Diseases–International Center for Excellence in Research, Chennai, India
| | - Evangeline Ann Daniel
- ICMR-National Institute for Research in Tuberculosis, Indian Council of Medical Research, Chennai, India
- University of Madras, Chennai, India
| | - Bindu Dasan
- National Institute of Health-National Institute of Allergy and Infectious Diseases–International Center for Excellence in Research, Chennai, India
| | - Kannan Thiruvengadam
- ICMR-National Institute for Research in Tuberculosis, Indian Council of Medical Research, Chennai, India
| | | | - Sanjay Gaikwad
- Department of Pulmonary Medicine, Byramjee Jeejeebhoy Government Medical College and Sassoon General Hospitals, Pune, India
| | - Sathyamurthi Pattabiraman
- ICMR-National Institute for Research in Tuberculosis, Indian Council of Medical Research, Chennai, India
| | - Brindha Bhanu
- ICMR-National Institute for Research in Tuberculosis, Indian Council of Medical Research, Chennai, India
| | - Amsaveni Sivaprakasam
- ICMR-National Institute for Research in Tuberculosis, Indian Council of Medical Research, Chennai, India
| | - Vandana Kulkarni
- Byramjee Jeejeebhoy Government Medical College–Johns Hopkins Clinical Research Site, Pune, India
- Johns Hopkins Center for Infectious Diseases in India, Pune, India
| | - Rajesh Karyakarte
- Department of Microbiology, Byramjee Jeejeebhoy Government Medical College and Sassoon General Hospitals, Pune, India
| | - Mandar Paradkar
- Byramjee Jeejeebhoy Government Medical College–Johns Hopkins Clinical Research Site, Pune, India
- Johns Hopkins Center for Infectious Diseases in India, Pune, India
| | | | - Vidya Mave
- Byramjee Jeejeebhoy Government Medical College–Johns Hopkins Clinical Research Site, Pune, India
- Johns Hopkins Center for Infectious Diseases in India, Pune, India
- Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Amita Gupta
- Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Luke Elizabeth Hanna
- ICMR-National Institute for Research in Tuberculosis, Indian Council of Medical Research, Chennai, India
| | - Subash Babu
- National Institute of Health-National Institute of Allergy and Infectious Diseases–International Center for Excellence in Research, Chennai, India
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
6
|
Luna MJ, Oluoch PO, Miao J, Culviner P, Papavinasasundaram K, Jaecklein E, Shell SS, Ioerger TR, Fortune SM, Farhat MR, Sassetti CM. Frequently arising ESX-1-associated phase variants influence Mycobacterium tuberculosis fitness in the presence of host and antibiotic pressures. mBio 2025; 16:e0376224. [PMID: 39873486 PMCID: PMC11898584 DOI: 10.1128/mbio.03762-24] [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: 12/04/2024] [Accepted: 12/18/2024] [Indexed: 01/30/2025] Open
Abstract
Mycobacterium tuberculosis (Mtb) exhibits an impressive ability to adapt to rapidly changing environments, despite its genome's apparent stability. Recently, phase variation through indel formation in homopolymeric tracts (HT) has emerged as a potentially important mechanism promoting adaptation in Mtb. This study examines the impact of common phase variants associated with the ESX-1 type VII secretion system, focusing on a highly variable HT upstream of the ESX-1 regulatory factor, espR. By engineering this frequently observed indel into an isogenic background, we demonstrate that a single nucleotide insertion in the espR 5'UTR causes post-transcriptional upregulation of EspR protein abundance and corresponding alterations in the EspR regulon. Consequently, this mutation increases the expression of ESX-1 components in the espACD operon and enhances ESX-1 substrate secretion. We find that this indel specifically increases isoniazid resistance without impacting the effectiveness of other drugs tested. Furthermore, we show that two distinct observed HT indels that regulate either espR translation or espACD transcription increase bacterial fitness in a mouse infection model. The presence of multiple ESX-1-associated HTs provides a mechanism to combinatorially tune protein secretion, drug sensitivity, and host-pathogen interactions. More broadly, these findings support emerging data that Mtb utilizes HT-mediated phase variation to direct genetic variation to certain sites across the genome in order to adapt to changing pressures. IMPORTANCE Mycobacterium tuberculosis (Mtb) is responsible for more deaths worldwide than any other single infectious agent. Understanding how this pathogen adapts to the varied environmental pressures imposed by host immunity and antibiotics has important implications for the design of more effective therapies. In this work, we show that the genome of Mtb contains multiple contingency loci that control the activity of the ESX-1 secretion system, which is critical for interactions with the host. These loci consist of homopolymeric DNA tracts in gene regulatory regions that are subject to high-frequency reversible variation and act to tune the activity of ESX-1. We find that variation at these sites increases the fitness of Mtb in the presence of antibiotic and/or during infection. These findings indicate that Mtb has the ability to diversify its genome in specific sites to create subpopulations of cells that are preadapted to new conditions.
Collapse
Affiliation(s)
- Michael J. Luna
- Department of Microbiology, UMass Chan Medical School, Worcester, Massachusetts, USA
| | - Peter O. Oluoch
- Department of Microbiology, UMass Chan Medical School, Worcester, Massachusetts, USA
| | - Jiazheng Miao
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts, USA
| | - Peter Culviner
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public Health, Boston, Massachusetts, USA
| | | | - Eleni Jaecklein
- Department of Microbiology, UMass Chan Medical School, Worcester, Massachusetts, USA
| | - Scarlet S. Shell
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| | - Thomas R. Ioerger
- Department of Computer Science and Engineering, Texas A&M University, College Station, Texas, USA
| | - Sarah M. Fortune
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public Health, Boston, Massachusetts, USA
| | - Maha R. Farhat
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts, USA
- Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | |
Collapse
|
7
|
Pullen KM, Finethy R, Ko SHB, Reames CJ, Sassetti CM, Lauffenburger DA. Cross-species transcriptomics translation reveals a role for the unfolded protein response in Mycobacterium tuberculosis infection. NPJ Syst Biol Appl 2025; 11:19. [PMID: 39955299 PMCID: PMC11830044 DOI: 10.1038/s41540-024-00487-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 12/25/2024] [Indexed: 02/17/2025] Open
Abstract
Numerous studies have identified similarities in blood transcriptomic signatures of tuberculosis (TB) phenotypes between mice and humans, including type 1 interferon production and innate immune cell activation. However, murine infection pathophysiology is distinct from human disease. We hypothesized that this is partly due to differences in the relative importance of biological pathways across species. To address this animal-to-human gap, we applied a systems modeling framework, Translatable Components Regression, to identify the axes of variation in the preclinical data most relevant to human TB disease state. Among the pathways our cross-species model pinpointed as highly predictive of human TB phenotype was the infection-induced unfolded protein response. To validate this mechanism, we confirmed that this cellular stress pathway modulates immune functions in Mycobacterium tuberculosis-infected mouse macrophages. Our work demonstrates how systems-level computational models enhance the value of animal studies for elucidating complex human pathophysiology.
Collapse
Affiliation(s)
- Krista M Pullen
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ryan Finethy
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, USA
| | - Seung-Hyun B Ko
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Charlotte J Reames
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, USA
| | - Christopher M Sassetti
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, USA.
| | - Douglas A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
8
|
Vance RE. Tuberculosis as an unconventional interferonopathy. Curr Opin Immunol 2025; 92:102508. [PMID: 39637776 DOI: 10.1016/j.coi.2024.102508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 10/31/2024] [Accepted: 11/12/2024] [Indexed: 12/07/2024]
Abstract
Tuberculosis is caused by Mycobacterium tuberculosis, a bacterium that accounts for more human mortality than any other. Evidence is accumulating for the view that tuberculosis is an interferonopathy - a disease driven by type I interferons. However, how type I interferons exacerbate tuberculosis remains poorly understood. As an infection, tuberculosis is distinct from conventional interferonopathies, which are autoinflammatory diseases. Here I consider the hypothesis that type I interferons promote bacterial replication by impairing key antibacterial immune responses, including those orchestrated by interleukin-1 and interferon γ. Paradoxically, during tuberculosis, the underlying state of impaired antibacterial immunity co-exists with overt (but ineffective) inflammation. Conceiving of tuberculosis as an unconventional interferonopathy may suggest fruitful avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Russell E Vance
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, CA USA.
| |
Collapse
|
9
|
Malik AA, Shariq M, Sheikh JA, Zarin S, Ahuja Y, Fayaz H, Alam A, Ehtesham NZ, Hasnain SE. Activation of the lysosomal damage response and selective autophagy: the coordinated actions of galectins, TRIM proteins, and CGAS-STING1 in providing immunity against Mycobacterium tuberculosis. Crit Rev Microbiol 2025; 51:108-127. [PMID: 38470107 DOI: 10.1080/1040841x.2024.2321494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 01/16/2024] [Accepted: 02/14/2024] [Indexed: 03/13/2024]
Abstract
Autophagy is a crucial immune defense mechanism that controls the survival and pathogenesis of M. tb by maintaining cell physiology during stress and pathogen attack. The E3-Ub ligases (PRKN, SMURF1, and NEDD4) and autophagy receptors (SQSTM1, TAX1BP1, CALCOCO2, OPTN, and NBR1) play key roles in this process. Galectins (LGALSs), which bind to sugars and are involved in identifying damaged cell membranes caused by intracellular pathogens such as M. tb, are essential. These include LGALS3, LGALS8, and LGALS9, which respond to endomembrane damage and regulate endomembrane damage caused by toxic chemicals, protein aggregates, and intracellular pathogens, including M. tb. They also activate selective autophagy and de novo endolysosome biogenesis. LGALS3, LGALS9, and LGALS8 interact with various components to activate autophagy and repair damage, while CGAS-STING1 plays a critical role in providing immunity against M. tb by activating selective autophagy and producing type I IFNs with antimycobacterial functions. STING1 activates cGAMP-dependent autophagy which provides immunity against various pathogens. Additionally, cytoplasmic surveillance pathways activated by ds-DNA, such as inflammasomes mediated by NLRP3 and AIM2 complexes, control M. tb. Modulation of E3-Ub ligases with small regulatory molecules of LGALSs and TRIM proteins could be a novel host-based therapeutic approach for controlling TB.
Collapse
Affiliation(s)
- Asrar Ahmad Malik
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Mohd Shariq
- ICMR-National Institute of Pathology, New Delhi, India
| | - Javaid Ahmad Sheikh
- Department of Biotechnology, School of Chemical and Life Sciences, New Delhi, India
| | - Sheeba Zarin
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
- Department of Molecular Medicine, School of Interdisciplinary Sciences and Technology, New Delhi, India
| | - Yashika Ahuja
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Haleema Fayaz
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Anwar Alam
- Department of Biotechnology, School of Science and Engineering Technology, Sharda University, Greater Noida, India
| | - Nasreen Z Ehtesham
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Seyed E Hasnain
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, New Delhi, India
| |
Collapse
|
10
|
Jani C, Jain N, Marsh AK, Uchil P, Doan T, Hudspith M, Glover OT, Baskir ZR, Boucau J, Root DE, van der Wel NN, Doench JG, Barczak AK. VPS18 contributes to phagosome membrane integrity in Mycobacterium tuberculosis-infected macrophages. SCIENCE ADVANCES 2025; 11:eadr6166. [PMID: 39888996 PMCID: PMC11784855 DOI: 10.1126/sciadv.adr6166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 12/31/2024] [Indexed: 02/02/2025]
Abstract
Mycobacterium tuberculosis (Mtb) has evolved to be exquisitely adapted to survive within host macrophages. The capacity to damage the phagosomal membrane has emerged as central to Mtb virulence. While Mtb factors driving membrane damage have been described, host factors that maintain phagosomal integrity or repair Mtb-induced damage to contain the pathogen remain largely unknown. We used a genome-wide CRISPR screen to identify host factors required to repair Mtb-damaged phagosomal membranes. Vacuolar protein sorting-associated protein 18 (VPS18), a member of the HOPS and CORVET trafficking complexes, was among the top hits. VPS18 colocalized with Mtb in macrophages beginning shortly after infection, and VPS18-knockout macrophages demonstrated increased damage of Mtb-containing phagosomes without impaired autophagy. Mtb grew more robustly in VPS18-knockout cells, and the first-line antituberculosis antibiotic pyrazinamide was less effective. Our results identify VPS18 as required for phagosomal membrane integrity in Mtb-infected cells and suggest that modulating phagosome integrity may hold promise for improving the efficacy of antibiotic treatment for TB.
Collapse
Affiliation(s)
- Charul Jani
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Neha Jain
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Amanda K. Marsh
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Pooja Uchil
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
- Institute of Clinical and Molecular Virology, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Triet Doan
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Meggie Hudspith
- Electron Microscopy Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Owen T. Glover
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Zach R. Baskir
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Julie Boucau
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | | | - Nicole N. van der Wel
- Electron Microscopy Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | | | - Amy K. Barczak
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
- The Broad Institute, Cambridge, MA 02139, USA
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
11
|
Saqib M, Das S, Nafiz TN, McDonough E, Sankar P, Mishra LK, Zhang X, Cai Y, Subbian S, Mishra BB. Pathogenic role for CD101-negative neutrophils in the type I interferon-mediated immunopathogenesis of tuberculosis. Cell Rep 2025; 44:115072. [PMID: 39693225 PMCID: PMC11829800 DOI: 10.1016/j.celrep.2024.115072] [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: 05/01/2024] [Revised: 09/13/2024] [Accepted: 11/25/2024] [Indexed: 12/20/2024] Open
Abstract
Neutrophils are vital for immunity against Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), yet their heterogeneous nature suggests a complex role in TB pathogenesis. Here, we identify two distinct neutrophil populations based on CD101 expression, highlighting their divergent roles in TB. CD101-negative (CD101-ve) neutrophils, which resemble immature, pro-inflammatory granulocytes, exhibit reduced Mtb phagocytosis compared to their mature, CD101-positive (CD101+ve) counterparts. Our findings reveal that type I interferons (IFN-Is) suppress neutrophil Mtb uptake and drive the recruitment of CD101-ve neutrophils to the lungs. Infiltration of these cells promotes Mtb extracellular persistence, exacerbates epithelial damage, and impairs surfactant production. Furthermore, we demonstrate that granulocyte colony-stimulating factor (G-CSF) and chemokine receptor CXCR2 are essential for the pulmonary accumulation of CD101-ve neutrophils. Our study uncovers a pathogenic role for CD101-ve neutrophils in TB and highlights the IFN-I-dependent recruitment of this functionally compromised immature neutrophil as a driver of TB immunopathogenesis.
Collapse
Affiliation(s)
- Mohd Saqib
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA
| | - Shreya Das
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA
| | - Tanvir N Nafiz
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA
| | - Elizabeth McDonough
- GE Healthcare Technology and Innovation Center, GE Research, Niskayuna, NY, USA
| | - Poornima Sankar
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA
| | - Lokesh K Mishra
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA
| | - Ximeng Zhang
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
| | - Yi Cai
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
| | - Selvakumar Subbian
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Bibhuti B Mishra
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA.
| |
Collapse
|
12
|
Sánchez-Carvajal JM, Godel A, Husson N, Summerfield A, García-Nicolás O. Plasmacytoid dendritic cell sensing of African swine fever virus-infected macrophages results in STING-dependent robust interferon-α production. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:130-140. [PMID: 40073264 DOI: 10.1093/jimmun/vkae008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 10/16/2024] [Indexed: 03/14/2025]
Abstract
While several African swine fever virus (ASFV)-encoded proteins potently interfere with the cGAS-STING (cyclic GMP-AMP synthetase-stimulator of interferon genes) pathway at different levels to suppress interferon (IFN) type I production in infected macrophages, systemic IFN-α is induced during the early stages of AFSV infection in pigs. The present study elucidates a mechanism by which such responses can be triggered, at least in vitro. We demonstrate that infection of monocyte-derived macrophages (MDMs) by ASFV genotype 2 strains is highly efficient but immunologically silent with respect to IFN type I, IFN-stimulated gene induction, and tumor necrosis factor production. Additionally, ASFV does not directly activate plasmacytoid dendritic cells (pDCs). However, coculturing pDCs with ASFV-infected MDMs results in a strong pDC response characterized by high levels of IFN-α and tumor necrosis factor. IFN type I, in turn, promoted interleukin-1 receptor antagonist production by macrophages. Similar to the sensing of infected cells by other viruses, pDC activation required integrin-mediated cognate interactions with ASFV-infected MDMs to form an interferogenic synapse. Inhibitor studies indicated that the activation of pDCs requires the STING pathway and the formation of gap junctions. While IL-4-polarized macrophages showed increased susceptibility, IFN-γ-polarized ASFV-infected macrophages induced higher pDC activation. Pretreatment of pDCs with IFN-β and IFN-γ also enhanced IFN-α production in response to ASFV-infected macrophages, highlighting the influence of the immunological microenvironment. These findings suggest that the IFN-α detected during ASFV infection in pigs may be a result of pDC sensing ASFV-infected macrophages.
Collapse
Affiliation(s)
- José María Sánchez-Carvajal
- Department of Anatomy and Comparative Pathology and Toxicology, Pathology and Immunology Group, Unidad de Investigación Competitiva Zoonosis y Enfermedades Emergentes, University of Córdoba, Córdoba, Spain
- Institute of Virology and Immunology, Mittelhäusern, Switzerland
| | - Aurélie Godel
- Institute of Virology and Immunology, Mittelhäusern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Nolwen Husson
- Institute of Virology and Immunology, Mittelhäusern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Artur Summerfield
- Institute of Virology and Immunology, Mittelhäusern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Multidisciplinary Center for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Obdulio García-Nicolás
- Institute of Virology and Immunology, Mittelhäusern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Multidisciplinary Center for Infectious Diseases, University of Bern, Bern, Switzerland
| |
Collapse
|
13
|
Meade RK, Smith CM. Immunological roads diverged: mapping tuberculosis outcomes in mice. Trends Microbiol 2025; 33:15-33. [PMID: 39034171 DOI: 10.1016/j.tim.2024.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/23/2024]
Abstract
The journey from phenotypic observation to causal genetic mechanism is a long and challenging road. For pathogens like Mycobacterium tuberculosis (Mtb), which causes tuberculosis (TB), host-pathogen coevolution has spanned millennia, costing millions of human lives. Mammalian models can systematically recapitulate host genetic variation, producing a spectrum of disease outcomes. Leveraging genome sequences and deep phenotyping data from infected mouse genetic reference populations (GRPs), quantitative trait locus (QTL) mapping approaches have successfully identified host genomic regions associated with TB phenotypes. Here, we review the ongoing optimization of QTL mapping study design alongside advances in mouse GRPs. These next-generation resources and approaches have enabled identification of novel host-pathogen interactions governing one of the most prevalent infectious diseases in the world today.
Collapse
Affiliation(s)
- Rachel K Meade
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA; University Program in Genetics and Genomics, Duke University, Durham, NC, USA
| | - Clare M Smith
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA; University Program in Genetics and Genomics, Duke University, Durham, NC, USA.
| |
Collapse
|
14
|
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
|
15
|
Luo Y, Huang CC, Howard NC, Wang X, Liu Q, Li X, Zhu J, Amariuta T, Asgari S, Ishigaki K, Calderon R, Raman S, Ramnarine AK, Mayfield JA, Moody DB, Lecca L, Fortune SM, Murray MB, Raychaudhuri S. Paired analysis of host and pathogen genomes identifies determinants of human tuberculosis. Nat Commun 2024; 15:10393. [PMID: 39613754 PMCID: PMC11607449 DOI: 10.1038/s41467-024-54741-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 11/19/2024] [Indexed: 12/01/2024] Open
Abstract
Infectious disease is the result of interactions between host and pathogen and can depend on genetic variations in both. We conduct a genome-to-genome study of paired human and Mycobacterium tuberculosis genomes from a cohort of 1556 tuberculosis patients in Lima, Peru. We identify an association between a human intronic variant (rs3130660, OR = 10.06, 95%CI: 4.87 - 20.77, P = 7.92 × 10-8) in the FLOT1 gene and a subclavaluee of Mtb Lineage 2. In a human macrophage infection model, we observe hosts with the rs3130660-A allele exhibited stronger interferon gene signatures. The interacting strains have altered redox states due to a thioredoxin reductase mutation. We investigate this association in a 2020 cohort of 699 patients recruited during the COVID-19 pandemic. While the prevalence of the interacting strain almost doubled between 2010 and 2020, its infection is not associated with rs3130660 in this recent cohort. These findings suggest a complex interplay among host, pathogen, and environmental factors in tuberculosis dynamics.
Collapse
Affiliation(s)
- Yang Luo
- Center for Data Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Chuan-Chin Huang
- Division of Global Health Equity, Brigham and Women's Hospital, Boston, MA, USA
- Department of Global Health and Social Medicine, Harvard Medical School, Boston, MA, USA
| | - Nicole C Howard
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Xin Wang
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Qingyun Liu
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Xinyi Li
- Committee on Genetics, Genomics, and Systems Biology, University of Chicago, Chicago, IL, USA
| | - Junhao Zhu
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Tiffany Amariuta
- Center for Data Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Halıcıoğlu Data Science Institute, University of California San Diego, La Jolla, CA, USA
- Division of Biomedical Informatics, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Samira Asgari
- Center for Data Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Genomic Health, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Kazuyoshi Ishigaki
- Center for Data Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Laboratory for Human Immunogenetics, RIKEN Center for Integrative Medical Sciences, Kobe, Japan
| | | | - Sahadevan Raman
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Alexandrea K Ramnarine
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jacob A Mayfield
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - D Branch Moody
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Leonid Lecca
- Department of Global Health and Social Medicine, Harvard Medical School, Boston, MA, USA
- Socios En Salud Sucursal Peru, Lima, Peru
| | - Sarah M Fortune
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA.
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.
| | - Megan B Murray
- Division of Global Health Equity, Brigham and Women's Hospital, Boston, MA, USA.
- Department of Global Health and Social Medicine, Harvard Medical School, Boston, MA, USA.
- Department of Epidemiology, Harvard T.H Chan School of Public Health, Boston, MA, USA.
| | - Soumya Raychaudhuri
- Center for Data Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA.
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
16
|
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
|
17
|
Nakamura H, Hikichi H, Seto S, Hijikata M, Keicho N. Transcriptional regulators SP110 and SP140 modulate inflammatory response genes in Mycobacterium tuberculosis-infected human macrophages. Microbiol Spectr 2024; 12:e0010124. [PMID: 39162523 PMCID: PMC11448263 DOI: 10.1128/spectrum.00101-24] [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: 01/15/2024] [Accepted: 07/16/2024] [Indexed: 08/21/2024] Open
Abstract
Understanding the functions of human transcriptional regulatory genes SP110 and SP140 during Mycobacterium tuberculosis infection is crucial; in a mouse model, homologous genes Sp110 and Sp140 have been shown to negatively regulate inflammatory response genes, including the type I interferon (IFN) response. The reduction of these genes in mice is associated with susceptibility to M. tuberculosis infection and the development of necrotizing granulomatous lesions. To investigate the involvement of SP110 and SP140 in human inflammatory response, we analyzed their regulatory manner in THP-1 macrophages infected with M. tuberculosis. Genome-wide transcriptional profiling revealed that the depletion of SP110 and/or SP140 impaired the induction of gene expression associated with inflammatory responses, including IFN response genes, although it had little effect on the intracellular proliferation of M. tuberculosis. By contrast, genes related to phosphorylation were upregulated in infected macrophages with SP110 and/or SP140 knockdown, but downregulated in infected control macrophages without their knockdown. Reverse transcription-quantitative PCR and ELISA further confirmed the impairment of the induction of IFN response genes by the depletion of SP110 and/or SP140 in M. tuberculosis-infected macrophages. These findings suggest that human SP110 and SP140 act as positive regulators for genes associated with inflammatory responses in M. tuberculosis-infected macrophages. IMPORTANCE Tuberculosis (TB) is one of the most serious infectious diseases, with high morbidity and mortality worldwide. C3HeB/FeJ mice are widely utilized for evaluating anti-TB drugs because their drug sensitivity and pathology during M. tuberculosis infection resemble those of human TB, including the development of necrotizing granulomas. Downregulation of the transcriptional regulatory genes Sp110 and Sp140 in C3HeB/FeJ mice has been demonstrated to activate gene expression associated with inflammatory responses during M. tuberculosis infection, resulting in susceptibility to the infection. Here, we examined the regulatory manner of SP110 and SP140 using transcriptomic analysis in M. tuberculosis-infected human macrophages. Depletion of SP110 and/or SP140 in M. tuberculosis-infected THP-1 macrophages impaired the induction of gene expression associated with inflammatory responses, including interferon response genes, compared with that in control macrophages. These results suggest that human SP110 and SP140 act as positive regulators for genes associated with inflammatory responses upon M. tuberculosis infection.
Collapse
Affiliation(s)
- Hajime Nakamura
- Department of Pathophysiology and Host Defense, The Research Institute of Tuberculosis, Japan Anti-Tuberculosis Association, Tokyo, Japan
- Department of Basic Mycobacteriosis, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Haruka Hikichi
- Department of Pathophysiology and Host Defense, The Research Institute of Tuberculosis, Japan Anti-Tuberculosis Association, Tokyo, Japan
- Department of Basic Mycobacteriosis, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Shintaro Seto
- Department of Pathophysiology and Host Defense, The Research Institute of Tuberculosis, Japan Anti-Tuberculosis Association, Tokyo, Japan
| | - Minako Hijikata
- Department of Pathophysiology and Host Defense, The Research Institute of Tuberculosis, Japan Anti-Tuberculosis Association, Tokyo, Japan
| | - Naoto Keicho
- Department of Basic Mycobacteriosis, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- The Research Institute of Tuberculosis, Japan Anti-Tuberculosis Association, Tokyo, Japan
| |
Collapse
|
18
|
Malik AA, Shariq M, Sheikh JA, Jaiswal U, Fayaz H, Shrivastava G, Ehtesham NZ, Hasnain SE. Mechanisms of immune evasion by Mycobacterium tuberculosis: the impact of T7SS and cell wall lipids on host defenses. Crit Rev Biochem Mol Biol 2024; 59:310-336. [PMID: 39378051 DOI: 10.1080/10409238.2024.2411264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/21/2024] [Accepted: 09/27/2024] [Indexed: 11/14/2024]
Abstract
Mycobacterium tuberculosis (M. tb) is one of the most successful human pathogens, causing a severe and widespread infectious disease. The frequent emergence of multidrug-resistant (MDR) strains has exacerbated this public health crisis, particularly in underdeveloped regions. M. tb employs a sophisticated array of virulence factors to subvert host immune responses, both innate and adaptive. It utilizes the early secretory antigenic target (ESAT6) secretion system 1 (ESX-1) type VII secretion system (T7SS) and cell wall lipids to disrupt phagosomal integrity, inhibiting phagosome maturation, and fusion with lysosomes. Although host cells activate mechanisms such as ubiquitin (Ub), Ub-ligase, and cyclic GMP-AMP synthase-stimulator of interferon genes 1 (CGAS-STING1)-mediated autophagy to inhibit M. tb survival within macrophages, the pathogen counteracts these defenses with its own virulence factors, thereby inhibiting autophagy and dampening host-directed responses. T7SSs are critical for transporting proteins across the complex mycobacterial cell envelope, performing essential functions, including metabolite uptake, immune evasion, and conjugation. T7SS substrates fall into two main families: ESAT-6 system proteins, which are found in both Firmicutes and Actinobacteria, and proline-glutamic acid (PE) and proline-proline-glutamic acid (PPE) proteins, which are unique to mycobacteria. Recent studies have highlighted the significance of T7SSs in mycobacterial growth, virulence, and pathogenesis. Understanding the mechanisms governing T7SSs could pave the way for novel therapeutic strategies to combat mycobacterial diseases, including tuberculosis (TB).
Collapse
Affiliation(s)
- Asrar Ahmad Malik
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Mohd Shariq
- GITAM School of Science, GITAM University, Rudraram, Telangana, India
| | - Javaid Ahmad Sheikh
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, Hamdard Nagar, New Delhi, India
| | - Udyeshita Jaiswal
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, Hamdard Nagar, New Delhi, India
| | - Haleema Fayaz
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Gauri Shrivastava
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Nasreen Z Ehtesham
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Seyed E Hasnain
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, Delhi (IIT-D), Hauz Khas, New Delhi, India
| |
Collapse
|
19
|
Naik SK, McNehlan ME, Mreyoud Y, Kinsella RL, Smirnov A, Sur Chowdhury C, McKee SR, Dubey N, Woodson R, Kreamalmeyer D, Stallings CL. Type I IFN signaling in the absence of IRGM1 promotes M. tuberculosis replication in immune cells by suppressing T cell responses. Mucosal Immunol 2024; 17:1114-1127. [PMID: 39038752 DOI: 10.1016/j.mucimm.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 07/09/2024] [Accepted: 07/15/2024] [Indexed: 07/24/2024]
Abstract
Polymorphisms in the IRGM gene are associated with susceptibility to tuberculosis in humans. A murine ortholog of Irgm, Irgm1, is also essential for controlling Mycobacterium tuberculosis (Mtb) infection in mice. Multiple processes have been associated with IRGM1 activity that could impact the host response to Mtb infection, including roles in autophagy-mediated pathogen clearance and expansion of activated T cells. However, what IRGM1-mediated pathway is necessary to control Mtb infection in vivo and the mechanistic basis for this control remains unknown. We dissected the contribution of IRGM1 to immune control of Mtb pathogenesis in vivo and found that Irgm1 deletion leads to higher levels of IRGM3-dependent type I interferon signaling. The increased type I interferon signaling precludes T cell expansion during Mtb infection. The absence of Mtb-specific T cell expansion in Irgm1-/- mice results in uncontrolled Mtb infection in neutrophils and alveolar macrophages, which directly contributes to susceptibility to infection. Together, our studies reveal that IRGM1 is required to promote T cell-mediated control of Mtb infection in neutrophils, which is essential for the survival of Mtb-infected mice. These studies also uncover new ways type I interferon signaling can impact TH1 immune responses.
Collapse
Affiliation(s)
- Sumanta K Naik
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Michael E McNehlan
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yassin Mreyoud
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rachel L Kinsella
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Asya Smirnov
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Chanchal Sur Chowdhury
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Samuel R McKee
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Neha Dubey
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Reilly Woodson
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Darren Kreamalmeyer
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Christina L Stallings
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
20
|
Schrijver B, Göpfert J, La Distia Nora R, Putera I, Nagtzaam NM, Smits te Nijenhuis MA, van Rijswijk AL, ten Berge JC, van Laar JA, van Hagen PM, Dik WA. Increased serum interferon activity in sarcoidosis compared to that in tuberculosis: Implication for diagnosis? Heliyon 2024; 10:e37103. [PMID: 39309852 PMCID: PMC11416298 DOI: 10.1016/j.heliyon.2024.e37103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/25/2024] Open
Abstract
Objectives In this study, we measured serum interferon (IFN) levels and activity in patients with sarcoidosis and tuberculosis (TB) with and without uveitis. We aimed to understand the role of IFN in the pathophysiology of both conditions and explore its potential as a discriminating marker for these clinically similar diseases. Methods Sera from an Indonesian TB and a Dutch sarcoidosis cohort were used in the analysis. IFNα2 and IFNγ concentrations were measured using Simoa® and Luminex assays, respectively. Serum IFN activity was assessed by incubating THP-1 cells with patient serum and measuring IFN-stimulated gene transcription using qPCR. Anti-IFNα2 and IFNγ autoantibodies were detected via Luminex assay and tested for neutralizing capacity using a flow cytometry-based signal transducer and activator of transcription (STAT) 1 phosphorylation inhibition assay. Results IFNα2 was detected in 74 % and 64 % of patients with sarcoidosis and pulmonary TB, respectively, while IFNγ was found in 78 % and 23 % of patients with sarcoidosis and TB, respectively. For uveitis cases specifically, IFNα2 was detected in 85 % of sarcoid uveitis (SU) and 33 % of tubercular uveitis (TBU) cases. Similarly, IFNγ was detected in 69 % of SU and 17 % of TBU cases. IFNγ serum concentrations were higher in sarcoidosis than that in TB patients (p < 0.0001). Focusing on patients with uveitis, SU showed increased IFNα2 (p = 0.004) and IFNγ (p < 0.002) serum concentrations compared to that in TBU. Notably, TBU displayed significantly reduced IFNα2 concentrations compared to that in healthy controls (p = 0.006). These results align with the increased interferon stimulated gene (ISG) transcriptional upregulation observed in THP-1 cells stimulated with serum from patients with sarcoidosis. Elevated levels of non-neutralizing anti-IFN autoantibodies were observed in patients with TB; however, these levels were similar to those observed in geographically matched healthy Indonesian controls. Conclusion Our results suggest decreased serum levels and activity of type I and II IFN in TB compared to those in sarcoidosis. This is indicative of distinct pathophysiological processes in these highly clinically similar diseases. We propose that the assessment of serum IFN levels and IFN activity has the potential to distinguish between sarcoidosis/SU and TB/TBU.
Collapse
Affiliation(s)
- Benjamin Schrijver
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC University Medical Center Rotterdam, the Netherlands
| | - Jens Göpfert
- Department of Applied Biomarkers and Immunoassays, NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Rina La Distia Nora
- Department of Ophthalmology, Faculty of Medicine, Universitas Indonesia, Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Ikhwanuliman Putera
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC University Medical Center Rotterdam, the Netherlands
- Department of Ophthalmology, Faculty of Medicine, Universitas Indonesia, Cipto Mangunkusumo Hospital, Jakarta, Indonesia
- Department of Internal Medicine, section Allergy & Clinical Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Nicole M.A.N. Nagtzaam
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC University Medical Center Rotterdam, the Netherlands
| | - Marja A.W. Smits te Nijenhuis
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC University Medical Center Rotterdam, the Netherlands
| | - Angelique L.C.T. van Rijswijk
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC University Medical Center Rotterdam, the Netherlands
| | | | - Jan A.M. van Laar
- Department of Internal Medicine, section Allergy & Clinical Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - P. Martin van Hagen
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC University Medical Center Rotterdam, the Netherlands
- Department of Internal Medicine, section Allergy & Clinical Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Willem A. Dik
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC University Medical Center Rotterdam, the Netherlands
| |
Collapse
|
21
|
Ravesloot-Chávez MM, Van Dis E, Fox D, Anaya Sanchez A, Espich S, Nguyenla XH, Rawal SL, Samani H, Ballinger MA, Thomas H, Kotov D, Vance R, Nachman MW, Stanley SA. Tuberculosis susceptibility in genetically diverse mice reveals functional diversity of neutrophils. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.29.547125. [PMID: 39211107 PMCID: PMC11361191 DOI: 10.1101/2023.06.29.547125] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Tuberculosis (TB) is a heterogenous disease in humans with individuals exhibiting a wide range of susceptibility. This heterogeneity is not captured by standard laboratory mouse lines. We used a new collection of 19 wild-derived inbred mouse lines collected from diverse geographic sites to identify novel phenotypes during Mycobacterium tuberculosis ( Mtb ) infection. Wild derived mice have heterogenous immune responses to infection that result in differential ability to control disease at early timepoints. Correlation analysis with multiple parameters including sex, weight, and cellular immune responses in the lungs revealed that enhanced control of infection is associated with increased numbers of CD4 T cells, CD8 T cells and B cells. Surprisingly, we did not observe strong correlations between IFN-γ production and control of infection. Although in most lines high neutrophils were associated with susceptibility, we identified a mouse line that harbors high neutrophils numbers yet controls infection. Using single-cell RNA sequencing, we identified a novel neutrophil signature associated with failure to control infection.
Collapse
|
22
|
Maciag K, Plumlee CR, Cohen SB, Gern BH, Urdahl KB. Reappraising the Role of T Cell-Derived IFN-γ in Restriction of Mycobacterium tuberculosis in the Murine Lung. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:339-346. [PMID: 38912839 PMCID: PMC11249196 DOI: 10.4049/jimmunol.2400145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 05/29/2024] [Indexed: 06/25/2024]
Abstract
T cells producing IFN-γ have long been considered a stalwart for immune protection against Mycobacterium tuberculosis (Mtb), but their relative importance to pulmonary immunity has been challenged by murine studies that achieved protection by adoptively transferred Mtb-specific IFN-γ-/- T cells. Using IFN-γ-/- T cell chimeric mice and adoptive transfer of IFN-γ-/- T cells into TCRβ-/-δ-/- mice, we demonstrate that control of lung Mtb burden is in fact dependent on T cell-derived IFN-γ, and, furthermore, mice selectively deficient in T cell-derived IFN-γ develop exacerbated disease compared with T cell-deficient control animals, despite equivalent lung bacterial burdens. Deficiency in T cell-derived IFN-γ skews infected and bystander monocyte-derived macrophages to an alternative M2 phenotype and promotes neutrophil and eosinophil influx. Our studies support an important role for T cell-derived IFN-γ in pulmonary immunity against tuberculosis.
Collapse
Affiliation(s)
- Karolina Maciag
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA
- Seattle Children’s Research Institute, Seattle, WA
| | | | | | - Benjamin H. Gern
- Seattle Children’s Research Institute, Seattle, WA
- Department of Pediatrics, University of Washington, Seattle, WA
| | - Kevin B. Urdahl
- Seattle Children’s Research Institute, Seattle, WA
- Department of Pediatrics, University of Washington, Seattle, WA
- Department of Immunology, University of Washington, Seattle, WA
| |
Collapse
|
23
|
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
|
24
|
Vu A, Glassman I, Campbell G, Yeganyan S, Nguyen J, Shin A, Venketaraman V. Host Cell Death and Modulation of Immune Response against Mycobacterium tuberculosis Infection. Int J Mol Sci 2024; 25:6255. [PMID: 38892443 PMCID: PMC11172987 DOI: 10.3390/ijms25116255] [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: 05/01/2024] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 06/21/2024] Open
Abstract
Mycobacterium tuberculosis (Mtb) is the causative agent of tuberculosis (TB), a prevalent infectious disease affecting populations worldwide. A classic trait of TB pathology is the formation of granulomas, which wall off the pathogen, via the innate and adaptive immune systems. Some key players involved include tumor necrosis factor-alpha (TNF-α), foamy macrophages, type I interferons (IFNs), and reactive oxygen species, which may also show overlap with cell death pathways. Additionally, host cell death is a primary method for combating and controlling Mtb within the body, a process which is influenced by both host and bacterial factors. These cell death modalities have distinct molecular mechanisms and pathways. Programmed cell death (PCD), encompassing apoptosis and autophagy, typically confers a protective response against Mtb by containing the bacteria within dead macrophages, facilitating their phagocytosis by uninfected or neighboring cells, whereas necrotic cell death benefits the pathogen, leading to the release of bacteria extracellularly. Apoptosis is triggered via intrinsic and extrinsic caspase-dependent pathways as well as caspase-independent pathways. Necrosis is induced via various pathways, including necroptosis, pyroptosis, and ferroptosis. Given the pivotal role of host cell death pathways in host defense against Mtb, therapeutic agents targeting cell death signaling have been investigated for TB treatment. This review provides an overview of the diverse mechanisms underlying Mtb-induced host cell death, examining their implications for host immunity. Furthermore, it discusses the potential of targeting host cell death pathways as therapeutic and preventive strategies against Mtb infection.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA (G.C.); (A.S.)
| |
Collapse
|
25
|
Lyu S, Zhang T, Peng P, Cao D, Ma L, Yu Y, Dong Y, Qi X, Wei C. Involvement of cGAS/STING Signaling in the Pathogenesis of Candida albicans Keratitis: Insights From Genetic and Pharmacological Approaches. Invest Ophthalmol Vis Sci 2024; 65:13. [PMID: 38848078 PMCID: PMC11166223 DOI: 10.1167/iovs.65.6.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 05/14/2024] [Indexed: 06/13/2024] Open
Abstract
Purpose Fungal keratitis (FK) is an invasive corneal infection associated with significant risk to vision. Although the cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes (STING) signaling pathway has been recognized for its role in defending against viral infections, its involvement in FK still remains largely unclear. This study sought to elucidate the contribution of the cGAS/STING signaling pathway to the pathogenesis of FK. Methods The expression of cGAS/STING signaling components was assessed in a murine model of Candida albicans keratitis through RNA sequencing, western blot analysis, immunofluorescence staining, and real-time PCR. Both genetic (utilizing Sting1gt/gt mice) and pharmacological (using C176) interventions were employed to inhibit STING activity, allowing for the evaluation of resultant pathogenic alterations in FK using slit-lamp examination, clinical scoring, hematoxylin and eosin (H&E) staining, fungal culture, and RNA sequencing. Subconjunctival administration of the NOD-like receptor protein 3 (NLRP3) inflammasome inhibitor MCC950 was performed to evaluate FK manifestations following STING activity blockade. Furthermore, the impact of the STING agonist diABZI on FK progression was investigated. Results Compared to uninfected corneas, those infected with C. albicans exhibited increased expression of cGAS/STING signaling components, as well as its elevated activity. Inhibiting cGAS/STING signaling exacerbated the advancement of FK, as evidenced by elevated clinical scores, augmented fungal load, and heightened inflammatory response, including NLRP3 inflammasome activation and pyroptosis. Pharmacological inhibition of the NLRP3 inflammasome effectively mitigated the exacerbated FK by suppressing STING activity. Conversely, pre-activation of STING exacerbated FK progression compared to the PBS control, characterized by increased fungal burden and reinforced inflammatory infiltration. Conclusions This study demonstrates the essential role of the cGAS/STING signaling pathway in FK pathogenesis and highlights the necessity of its proper activation for the host against FK.
Collapse
Affiliation(s)
- Shanmei Lyu
- Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
| | - Ting Zhang
- Eye Hospital of Shandong First Medical University, Eye Institute of Shandong First Medical University, Jinan, Shandong, China
| | - Peng Peng
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
| | - Dingwen Cao
- Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
| | - Li Ma
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
| | - Yang Yu
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
| | - Yanling Dong
- Qingdao Eye Hospital of Shandong First Medical University, Eye Institute of Shandong First Medical University, Qingdao, China
| | - Xiaolin Qi
- Eye Hospital of Shandong First Medical University, Eye Institute of Shandong First Medical University, Jinan, Shandong, China
- School of Ophthalmology, Shandong First Medical University, Jinan, Shandong, China
| | - Chao Wei
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China
| |
Collapse
|
26
|
Li Z, Han J, Jing J, Fan A, Zhang Y, Gao Y. Bovine DDX3X Restrains Bovine SP110c-Mediated Activation of Inflammasome in Macrophages. Animals (Basel) 2024; 14:1650. [PMID: 38891697 PMCID: PMC11171048 DOI: 10.3390/ani14111650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/21/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
The inflammasome is a vital part of the host's innate immunity activated by cellular infection or stress. Our previous research identified the bovine SP110c isoform (bSP110c) as a novel activator of the inflammasome that promoted the secretion of proinflammatory cytokines IL-1β and IL-18 in macrophages infected with Listeria monocytogenes or stimulated with lipopolysaccharide (LPS). However, the exact molecular mechanism for inhibiting bSP110c-induced inflammasome activation requires further clarification. Here, the researchers identified bovine DDX3X (bDDX3X) as an NLRP3-associated protein and an inhibitor of the bSP110c-induced inflammasome in the human THP1 macrophage cell line. Immunoprecipitation showed that bDDX3X interacted with the bSP110c CARD domain via its helicase domain. The co-expression of bSP110c and bDDX3X in THP1 macrophages significantly prevented the bSP110c-induced activation of inflammasomes. In addition, both bDDX3X and bSP110c interacted with bovine NLRP3 (bNLRP3), and bDDX3X enhanced the interaction between bSP110c and bNLRP3. The expression of bDDX3X in nigericin-stimulated THP1 macrophages significantly suppressed NLRP3 inflammasome activation, ASC speck formation, and pyroptosis. These findings demonstrate that bDDX3X negatively regulates the bSP110c-mediated inflammatory response by restricting the activation of the NLRP3 inflammasome. This discovery unveils a novel regulatory mechanism involving bDDX3X and bSP110c in coordinating inflammasome activation and subsequent cell-fate decisions in LPS-treated macrophages and, in turn, constitutes a step forward toward the implementation of marker-assisted selection in breeding programs aimed at utilizing cattle's immune defenses.
Collapse
|
27
|
Truong T, Martin K, Salemi M, Ray A, Phinney BS, Penn BH. The balance between antiviral and antibacterial responses during M. tuberculosis infection is regulated by the ubiquitin ligase CBL. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.15.594178. [PMID: 38798543 PMCID: PMC11118416 DOI: 10.1101/2024.05.15.594178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
As a first line of host defense, macrophages must be able to effectively sense and respond to diverse types of pathogens, and while a particular type of immune response may be beneficial in some circumstances, it can be detrimental in others. Upon infecting a macrophage, M. tuberculosis (Mtb) induces proinflammatory cytokines that activate antibacterial responses. Surprisingly, Mtb also triggers antiviral responses that actually hinder the ability of macrophages to control Mtb infection. The ubiquitin ligase CBL suppresses these antiviral responses and shifts macrophages toward a more antibacterial state during Mtb infection, however, the mechanisms by which CBL regulates immune signaling are unknown. We found that CBL controls responses to multiple stimuli and broadly suppresses the expression of antiviral effector genes. We then used mass-spectrometry to investigate potential CBL substrates and identified over 46,000 ubiquitylated peptides in Mtb-infected macrophages, as well as roughly 400 peptides with CBL-dependent ubiquitylation. We then performed genetic interaction analysis of CBL and its putative substrates, and identified the Fas associated factor 2 (FAF2) adapter protein as a key signaling molecule protein downstream of CBL. Together, these analyses identify thousands of new ubiquitin-mediated signaling events during the innate immune response and reveal an important new regulatory hub in this response.
Collapse
Affiliation(s)
- Tina Truong
- Department of Internal Medicine, University of California, Davis, Davis, California, United States of America
- Graduate Group in Immunology, University of California, Davis, Davis, California, United States of America
| | - Kelsey Martin
- Department of Internal Medicine, University of California, Davis, Davis, California, United States of America
| | - Michelle Salemi
- Proteomics Core Facility, University of California, Davis, Davis, California, United States of America
| | - Abigail Ray
- Department of Internal Medicine, University of California, Davis, Davis, California, United States of America
- Microbiology Graduate Group, University of California, Davis, Davis, California, United States of America
| | - Brett S. Phinney
- Proteomics Core Facility, University of California, Davis, Davis, California, United States of America
| | - Bennett H. Penn
- Department of Internal Medicine, University of California, Davis, Davis, California, United States of America
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, California, United States of America
| |
Collapse
|
28
|
Casanova JL, MacMicking JD, Nathan CF. Interferon- γ and infectious diseases: Lessons and prospects. Science 2024; 384:eadl2016. [PMID: 38635718 DOI: 10.1126/science.adl2016] [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: 10/04/2023] [Accepted: 03/13/2024] [Indexed: 04/20/2024]
Abstract
Infectious diseases continue to claim many lives. Prevention of morbidity and mortality from these diseases would benefit not just from new medicines and vaccines but also from a better understanding of what constitutes protective immunity. Among the major immune signals that mobilize host defense against infection is interferon-γ (IFN-γ), a protein secreted by lymphocytes. Forty years ago, IFN-γ was identified as a macrophage-activating factor, and, in recent years, there has been a resurgent interest in IFN-γ biology and its role in human defense. Here we assess the current understanding of IFN-γ, revisit its designation as an "interferon," and weigh its prospects as a therapeutic against globally pervasive microbial pathogens.
Collapse
Affiliation(s)
- Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, Necker Hospital for Sick Children, 75015 Paris, France
- Imagine Institute, Paris Cité University, 75015 Paris, France
- Department of Pediatrics, Necker Hospital for Sick Children, 75015 Paris, France
| | - John D MacMicking
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Systems Biology Institute, Yale University, West Haven, CT 06477, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Carl F Nathan
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
29
|
Maciag K, Plumlee C, Cohen S, Gern B, Urdahl K. Re-appraising the role of T-cell derived interferon gamma in restriction of Mycobacterium tuberculosis in the murine lung: T-cell derived IFNγ is required to restrict pulmonary Mtb. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.04.588086. [PMID: 38617280 PMCID: PMC11014638 DOI: 10.1101/2024.04.04.588086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
T cells producing interferon gamma (IFNγ) have long been considered a stalwart for immune protection against Mycobacterium tuberculosis (Mtb), but their relative importance to pulmonary immunity has been challenged by murine studies which achieved protection by adoptively transferred Mtb-specific IFNγ-/- T cells. Using IFNγ-/- T cell chimeric mice and adoptive transfer of IFNγ-/- T cells into TCRβ-/-δ-/- mice, we demonstrate that control of lung Mtb burden is in fact dependent on T cell-derived IFNγ, and furthermore, mice selectively deficient in T cell-derived IFNγ develop exacerbated disease compared to T cell-deficient controls despite equivalent lung bacterial burdens. Deficiency in T cell-derived IFNγ skews infected and bystander monocyte-derived macrophages (MDMs) to an alternative M2 phenotype, and promotes neutrophil and eosinophil influx. Our studies support an important role for T cell-derived IFNγ in pulmonary immunity against TB.
Collapse
Affiliation(s)
- Karolina Maciag
- Seattle Children's Research Institute
- Division of Allergy and Infectious Diseases, University of Washington
| | | | | | | | - Kevin Urdahl
- Seattle Children's Research Institute
- Department of Immunology, University of Washington
| |
Collapse
|
30
|
Bobba S, Chauhan KS, Akter S, Das S, Mittal E, Mathema B, Philips JA, Khader SA. A protective role for type I interferon signaling following infection with Mycobacterium tuberculosis carrying the rifampicin drug resistance-conferring RpoB mutation H445Y. PLoS Pathog 2024; 20:e1012137. [PMID: 38603763 PMCID: PMC11037539 DOI: 10.1371/journal.ppat.1012137] [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: 08/09/2023] [Revised: 04/23/2024] [Accepted: 03/19/2024] [Indexed: 04/13/2024] Open
Abstract
Interleukin-1 (IL-1) signaling is essential for controlling virulent Mycobacterium tuberculosis (Mtb) infection since antagonism of this pathway leads to exacerbated pathology and increased susceptibility. In contrast, the triggering of type I interferon (IFN) signaling is associated with the progression of tuberculosis (TB) disease and linked with negative regulation of IL-1 signaling. However, mice lacking IL-1 signaling can control Mtb infection if infected with an Mtb strain carrying the rifampin-resistance conferring mutation H445Y in its RNA polymerase β subunit (rpoB-H445Y Mtb). The mechanisms that govern protection in the absence of IL-1 signaling during rpoB-H445Y Mtb infection are unknown. In this study, we show that in the absence of IL-1 signaling, type I IFN signaling controls rpoB-H445Y Mtb replication, lung pathology, and excessive myeloid cell infiltration. Additionally, type I IFN is produced predominantly by monocytes and recruited macrophages and acts on LysM-expressing cells to drive protection through nitric oxide (NO) production to restrict intracellular rpoB-H445Y Mtb. These findings reveal an unexpected protective role for type I IFN signaling in compensating for deficiencies in IL-1 pathways during rpoB-H445Y Mtb infection.
Collapse
Affiliation(s)
- Suhas Bobba
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Kuldeep S. Chauhan
- Department of Microbiology, University of Chicago, Chicago, Illinois, United States of America
| | - Sadia Akter
- Department of Microbiology, University of Chicago, Chicago, Illinois, United States of America
| | - Shibali Das
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Ekansh Mittal
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Barun Mathema
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, New York, United States of America
| | - Jennifer A. Philips
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Shabaana A. Khader
- Department of Microbiology, University of Chicago, Chicago, Illinois, United States of America
| |
Collapse
|
31
|
Widmer G, Creasey HN. Fecal microbiota impacts development of Cryptosporidium parvum in the mouse. Sci Rep 2024; 14:5498. [PMID: 38448682 PMCID: PMC10917813 DOI: 10.1038/s41598-024-56184-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 03/03/2024] [Indexed: 03/08/2024] Open
Abstract
The dependence of Cryptosporidium parasites on host cell metabolites suggests that the development of nutritional interventions to limit parasite proliferation should be feasible. Based on this concept, we are testing dietary interventions to affect the enterocytes' metabolism in a manner that limits intracellular multiplication of the parasite. We hypothesize that changes in the metabolic pathways encoded by the gastro-intestinal tract microbiota may restrict parasite proliferation. To identify taxonomic and metabolic features of the microbiota associated with severity of cryptosporidiosis, as determined by estimating oocyst output, we characterized the fecal microbiota from mice experimentally infected with Cryptosporidium parvum. To eliminate the confounding effect of the interaction between co-housed mice, as well as facilitate the identification of microbiota markers associated with severity of cryptosporidiosis, fecal microbiota from individually caged mice were analyzed. Variation partitioning analysis applied to 16S sequence data from 25 mice belonging to four experiments shows that experiment was by far the biggest source of microbiota variation. Severity of cryptosporidiosis explained a smaller, though significant, fraction of microbiota variation. Notably, this effect was significant in the pre-patent phase of the infection, before mice excreted oocysts. These results are consistent with the pre-patent intestinal microbiota having a modest, but measurable, effect on cryptosporidiosis.
Collapse
Affiliation(s)
- Giovanni Widmer
- Cummings School of Veterinary Medicine at Tufts University, 200 Westboro Road, North Grafton, MA, 01536, USA.
| | - Hannah N Creasey
- Cummings School of Veterinary Medicine at Tufts University, 200 Westboro Road, North Grafton, MA, 01536, USA
- Department of Immunology, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA
| |
Collapse
|
32
|
Kubara K, Yamazaki K, Miyazaki T, Kondo K, Kurotaki D, Tamura T, Suzuki Y. Lymph node macrophages drive innate immune responses to enhance the anti-tumor efficacy of mRNA vaccines. Mol Ther 2024; 32:704-721. [PMID: 38243602 PMCID: PMC10928146 DOI: 10.1016/j.ymthe.2024.01.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/11/2023] [Accepted: 01/12/2024] [Indexed: 01/21/2024] Open
Abstract
mRNA vaccines are promising for cancer treatment. Efficient delivery of mRNAs encoding tumor antigens to antigen-presenting cells (APCs) is critical to elicit anti-tumor immunity. Herein, we identified a novel lipid nanoparticle (LNP) formulation, L17-F05, for mRNA vaccines by screening 34 ionizable lipids and 28 LNP formulations using human primary APCs. Subcutaneous delivery of L17-F05 mRNA vaccine encoding Gp100 and Trp2 inhibited tumor growth and prolonged the survival of mice bearing B16F10 melanoma. L17-F05 efficiently delivered mRNAs to conventional dendritic cells (cDCs) and macrophages in draining lymph nodes (dLNs). cDCs functioned as the main APCs by presenting antigens along with enhanced expression of co-stimulatory molecules. Macrophages triggered innate immune responses centered on type-I interferon (IFN-I) in dLNs. Lymph node (LN) macrophage depletion attenuated APC maturation and anti-tumor activity of L17-F05 mRNA vaccines. Loss-of-function studies revealed that L17-F05 works as a self-adjuvant by activating the stimulator of interferon genes (STING) pathway in macrophages. Collectively, the self-adjuvanticity of L17-F05 triggered innate immune responses in LN macrophages via the STING-IFN-I pathway, contributing to APC maturation and potent anti-tumor activity of L17-F05 mRNA vaccines. Our findings provide strategies for further optimization of mRNA vaccines based on the innate immune response driven by LN macrophages.
Collapse
Affiliation(s)
- Kenji Kubara
- Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3, Tokodai, Tsukuba, Ibaraki 300-2635, Japan; Department of Immunology, Yokohama City University Graduate School of Medicine, 3-9, Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan.
| | - Kazuto Yamazaki
- Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3, Tokodai, Tsukuba, Ibaraki 300-2635, Japan
| | - Takayuki Miyazaki
- Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3, Tokodai, Tsukuba, Ibaraki 300-2635, Japan
| | - Keita Kondo
- Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3, Tokodai, Tsukuba, Ibaraki 300-2635, Japan
| | - Daisuke Kurotaki
- Department of Immunology, Yokohama City University Graduate School of Medicine, 3-9, Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; Laboratory of Chromatin Organization in Immune Cell Development, International Research Center for Medical Sciences (IRCMS), Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan
| | - Tomohiko Tamura
- Department of Immunology, Yokohama City University Graduate School of Medicine, 3-9, Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Yuta Suzuki
- Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3, Tokodai, Tsukuba, Ibaraki 300-2635, Japan
| |
Collapse
|
33
|
Matsuda R, Sorobetea D, Zhang J, Peterson ST, Grayczyk JP, Yost W, Apenes N, Kovalik ME, Herrmann B, O’Neill RJ, Bohrer AC, Lanza M, Assenmacher CA, Mayer-Barber KD, Shin S, Brodsky IE. A TNF-IL-1 circuit controls Yersinia within intestinal pyogranulomas. J Exp Med 2024; 221:e20230679. [PMID: 38363547 PMCID: PMC10873131 DOI: 10.1084/jem.20230679] [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: 04/20/2023] [Revised: 11/22/2023] [Accepted: 01/19/2024] [Indexed: 02/17/2024] Open
Abstract
Tumor necrosis factor (TNF) is a pleiotropic inflammatory cytokine that mediates antimicrobial defense and granuloma formation in response to infection by numerous pathogens. We previously reported that Yersinia pseudotuberculosis colonizes the intestinal mucosa and induces the recruitment of neutrophils and inflammatory monocytes into organized immune structures termed pyogranulomas (PG) that control Yersinia infection. Inflammatory monocytes are essential for the control and clearance of Yersinia within intestinal PG, but how monocytes mediate Yersinia restriction is poorly understood. Here, we demonstrate that TNF signaling in monocytes is required for bacterial containment following enteric Yersinia infection. We further show that monocyte-intrinsic TNFR1 signaling drives the production of monocyte-derived interleukin-1 (IL-1), which signals through IL-1 receptors on non-hematopoietic cells to enable PG-mediated control of intestinal Yersinia infection. Altogether, our work reveals a monocyte-intrinsic TNF-IL-1 collaborative inflammatory circuit that restricts intestinal Yersinia infection.
Collapse
Affiliation(s)
- Rina Matsuda
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel Sorobetea
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jenna Zhang
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stefan T. Peterson
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - James P. Grayczyk
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Winslow Yost
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nicolai Apenes
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Maria E. Kovalik
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Beatrice Herrmann
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rosemary J. O’Neill
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrea C. Bohrer
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Matthew Lanza
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Charles-Antoine Assenmacher
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Katrin D. Mayer-Barber
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sunny Shin
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Igor E. Brodsky
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
34
|
Al-Qahtani AA, Alhamlan FS, Al-Qahtani AA. Pro-Inflammatory and Anti-Inflammatory Interleukins in Infectious Diseases: A Comprehensive Review. Trop Med Infect Dis 2024; 9:13. [PMID: 38251210 PMCID: PMC10818686 DOI: 10.3390/tropicalmed9010013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/29/2023] [Accepted: 12/10/2023] [Indexed: 01/23/2024] Open
Abstract
Interleukins (ILs) are signaling molecules that are crucial in regulating immune responses during infectious diseases. Pro-inflammatory ILs contribute to the activation and recruitment of immune cells, whereas anti-inflammatory ILs help to suppress excessive inflammation and promote tissue repair. Here, we provide a comprehensive overview of the role of pro-inflammatory and anti-inflammatory ILs in infectious diseases, with a focus on the mechanisms underlying their effects, their diagnostic and therapeutic potential, and emerging trends in IL-based therapies.
Collapse
Affiliation(s)
- Arwa A. Al-Qahtani
- Department of Family Medicine, College of Medicine, Al-Imam Mohammad Ibn Saud Islamic University, Riyadh 11432, Saudi Arabia;
| | - Fatimah S. Alhamlan
- Department of Infection and Immunity, King Faisal Specialist Hospital & Research Center, Riyadh 11211, Saudi Arabia;
- Department of Microbiology and Immunology, College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia
| | - Ahmed Ali Al-Qahtani
- Department of Infection and Immunity, King Faisal Specialist Hospital & Research Center, Riyadh 11211, Saudi Arabia;
- Department of Microbiology and Immunology, College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia
| |
Collapse
|
35
|
Zhang L, Yang Z, Wu F, Ge Q, Zhang Y, Li D, Gao M, Liu X. Multiple cytokine analysis based on QuantiFERON-TB gold plus in different tuberculosis infection status: an exploratory study. BMC Infect Dis 2024; 24:28. [PMID: 38166667 PMCID: PMC10762904 DOI: 10.1186/s12879-023-08943-0] [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: 06/05/2023] [Accepted: 12/21/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND More efficient and convenient diagnostic method is a desperate need to reduce the burden of tuberculosis (TB). This study explores the multiple cytokines secretion based on QuantiFERON-TB Gold Plus (QFT-Plus), and screens for optimal cytokines with diagnostic potential to differentiate TB infection status. METHODS Twenty active tuberculosis (ATB) patients, fifteen patients with latent TB infection (LTBI), ten patients with previous TB and ten healthy controls (HC) were enrolled. Whole blood samples were collected and stimulated by QFT-Plus TB1 and TB2 antigens. The levels of IFN-γ, TNF-α, IL-2, IL-6, IL-5, IL-10, IP-10, IL-1Ra, CXCL-1 and MCP-1 in supernatant were measured by Luminex bead-based multiplex assays. The receiver operating characteristic curve was used to evaluate the diagnostic accuracy of cytokine for distinguishing different TB infection status. RESULTS After stimulation with QFT-Plus TB1 and TB2 antigens, the levels of all cytokines, except IL-5 in TB2 tube, in ATB group were significantly higher than that in HC group. The levels of IL-1Ra concurrently showed the equally highest AUC for distinguishing TB infection from HC, followed by the levels of IP-10 in both TB1 tube and TB2 tube. Moreover, IP-10 levels displayed the largest AUC for distinguishing ATB patients from non-ATB patients. Meanwhile, the levels of IP-10 also demonstrated the largest AUC in both TB1 tube and TB2 tube for distinguishing ATB patients from LTBI. CONCLUSIONS In addition to conventional detection of IFN-γ, measuring IP-10 and IL-1Ra based on QFT-Plus may have the more tremendous potential to discriminate different TB infection status.
Collapse
Affiliation(s)
- Lifan Zhang
- Division of Infectious Diseases, Department of Internal medicine, State Key Laboratory of Complex Severe and Rare Disease, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Clinical Epidemiology Unit, Peking Union Medical College, International Clinical Epidemiology Network, Beijing, China
- Center for Tuberculosis Research, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhengrong Yang
- Division of Infectious Diseases, Department of Internal medicine, State Key Laboratory of Complex Severe and Rare Disease, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fengying Wu
- Division of Infectious Diseases, Department of Internal medicine, State Key Laboratory of Complex Severe and Rare Disease, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qiping Ge
- Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Yueqiu Zhang
- Division of Infectious Diseases, Department of Internal medicine, State Key Laboratory of Complex Severe and Rare Disease, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dongyu Li
- Division of Infectious Diseases, Department of Internal medicine, State Key Laboratory of Complex Severe and Rare Disease, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mengqiu Gao
- Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Xiaoqing Liu
- Division of Infectious Diseases, Department of Internal medicine, State Key Laboratory of Complex Severe and Rare Disease, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Clinical Epidemiology Unit, Peking Union Medical College, International Clinical Epidemiology Network, Beijing, China.
- Center for Tuberculosis Research, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
36
|
Amaral EP, Namasivayam S, Queiroz ATL, Fukutani E, Hilligan KL, Aberman K, Fisher L, Bomfim CCB, Kauffman K, Buchanan J, Santuo L, Gazzinelli-Guimaraes PH, Costa DL, Teixeira MA, Barreto-Duarte B, Rocha CG, Santana MF, Cordeiro-Santos M, Barber DL, Wilkinson RJ, Kramnik I, Igarashi K, Scriba T, Mayer-Barber KD, Andrade BB, Sher A. BACH1 promotes tissue necrosis and Mycobacterium tuberculosis susceptibility. Nat Microbiol 2024; 9:120-135. [PMID: 38066332 PMCID: PMC10769877 DOI: 10.1038/s41564-023-01523-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 10/11/2023] [Indexed: 01/07/2024]
Abstract
Oxidative stress triggers ferroptosis, a form of cellular necrosis characterized by iron-dependent lipid peroxidation, and has been implicated in Mycobacterium tuberculosis (Mtb) pathogenesis. We investigated whether Bach1, a transcription factor that represses multiple antioxidant genes, regulates host resistance to Mtb. We found that BACH1 expression is associated clinically with active pulmonary tuberculosis. Bach1 deletion in Mtb-infected mice increased glutathione levels and Gpx4 expression that inhibit lipid peroxidation. Bach1-/- macrophages exhibited increased resistance to Mtb-induced cell death, while Mtb-infected Bach1-deficient mice displayed reduced bacterial loads, pulmonary necrosis and lipid peroxidation concurrent with increased survival. Single-cell RNA-seq analysis of lungs from Mtb-infected Bach1-/- mice revealed an enrichment of genes associated with ferroptosis suppression. Bach1 depletion in Mtb-infected B6.Sst1S mice that display human-like necrotic lung pathology also markedly reduced necrosis and increased host resistance. These findings identify Bach1 as a key regulator of cellular and tissue necrosis and host resistance in Mtb infection.
Collapse
Affiliation(s)
- Eduardo P Amaral
- Immunobiology Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD, USA.
| | | | - Artur T L Queiroz
- Laboratório de Inflamação e Biomarcadores, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Bahia, Brazil
| | - Eduardo Fukutani
- Laboratório de Inflamação e Biomarcadores, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Bahia, Brazil
| | - Kerry L Hilligan
- Immunobiology Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD, USA
| | - Kate Aberman
- Immunobiology Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD, USA
| | - Logan Fisher
- Immunobiology Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine Graduate School of Medical Sciences, New York, NY, USA
| | - Caio Cesar B Bomfim
- Immunobiology Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD, USA
| | - Keith Kauffman
- T lymphocyte Biology Section, Laboratory of Parasitic Diseases, National Institutes of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
| | - Jay Buchanan
- T lymphocyte Biology Section, Laboratory of Parasitic Diseases, National Institutes of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
| | - Leslie Santuo
- T lymphocyte Biology Section, Laboratory of Parasitic Diseases, National Institutes of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
| | - Pedro Henrique Gazzinelli-Guimaraes
- Helminth Immunology Section, Laboratory of Parasitic Diseases, National Institutes of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
| | - Diego L Costa
- Immunobiology Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD, USA
- Departmento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Mariane Araujo Teixeira
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil
| | - Beatriz Barreto-Duarte
- Laboratório de Inflamação e Biomarcadores, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Bahia, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil
- Curso de Medicina, Universidade Salvador (UNIFACS), Laureate Universities, Salvador, Bahia, Brazil
| | - Clarissa Gurgel Rocha
- Department of Pathology, School of Medicine of the Federal University of Bahia, Salvador, Bahia, Brazil
- Center for Biotechnology and Cell Therapy, D'Or Institute for Research and Education (IDOR), Sao Rafael Hospital, Salvador, Bahia, Brazil
| | - Monique Freire Santana
- Departmento de Ensino e Pesquisa, Fundação Centro de Controle de Oncologia do Estado do Amazonas-FCECON, Manaus, Amazonas, Brazil
- Fundação Medicina Tropical Doutor Heitor Vieira Dourado, Manaus, Amazonas, Brazil
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus, Amazonas, Brazil
| | - Marcelo Cordeiro-Santos
- Fundação Medicina Tropical Doutor Heitor Vieira Dourado, Manaus, Amazonas, Brazil
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus, Amazonas, Brazil
- Faculdade de Medicina, Universidade Nilton Lins, Manaus, Amazonas, Brazil
| | - Daniel L Barber
- T lymphocyte Biology Section, Laboratory of Parasitic Diseases, National Institutes of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
| | - Robert J Wilkinson
- Wellcome Centre for Infectious Disease Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- The Francis Crick Institute, London, UK
- Department of Infectious Disease, Imperial College London, London, UK
| | - Igor Kramnik
- Boston University School of Medicine, Boston, MA, USA
| | - Kazuhiko Igarashi
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Thomas Scriba
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Observatory, South Africa
| | - Katrin D Mayer-Barber
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, MD, USA
| | - Bruno B Andrade
- Laboratório de Inflamação e Biomarcadores, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Bahia, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil
- Curso de Medicina, Universidade Salvador (UNIFACS), Laureate Universities, Salvador, Bahia, Brazil
- Department of Pathology, School of Medicine of the Federal University of Bahia, Salvador, Bahia, Brazil
- Curso de Medicina, Escola Bahiana de Medicina e Saúde Pública, Salvador, Bahia, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia, Salvador, Bahia, Brazil
- Curso de Medicina, Universidade Faculdade de Tecnologia e Ciências (UniFTC), Salvador, Bahia, Brazil
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Alan Sher
- Immunobiology Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD, USA.
| |
Collapse
|
37
|
Kotov DI, Lee OV, Fattinger SA, Langner CA, Guillen JV, Peters JM, Moon A, Burd EM, Witt KC, Stetson DB, Jaye DL, Bryson BD, Vance RE. Early cellular mechanisms of type I interferon-driven susceptibility to tuberculosis. Cell 2023; 186:5536-5553.e22. [PMID: 38029747 PMCID: PMC10757650 DOI: 10.1016/j.cell.2023.11.002] [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: 10/18/2022] [Revised: 06/16/2023] [Accepted: 11/01/2023] [Indexed: 12/01/2023]
Abstract
Mycobacterium tuberculosis (Mtb) causes 1.6 million deaths annually. Active tuberculosis correlates with a neutrophil-driven type I interferon (IFN) signature, but the cellular mechanisms underlying tuberculosis pathogenesis remain poorly understood. We found that interstitial macrophages (IMs) and plasmacytoid dendritic cells (pDCs) are dominant producers of type I IFN during Mtb infection in mice and non-human primates, and pDCs localize near human Mtb granulomas. Depletion of pDCs reduces Mtb burdens, implicating pDCs in tuberculosis pathogenesis. During IFN-driven disease, we observe abundant DNA-containing neutrophil extracellular traps (NETs) described to activate pDCs. Cell-type-specific disruption of the type I IFN receptor suggests that IFNs act on IMs to inhibit Mtb control. Single-cell RNA sequencing (scRNA-seq) indicates that type I IFN-responsive cells are defective in their response to IFNγ, a cytokine critical for Mtb control. We propose that pDC-derived type I IFNs act on IMs to permit bacterial replication, driving further neutrophil recruitment and active tuberculosis disease.
Collapse
Affiliation(s)
- Dmitri I Kotov
- Division of Immunology and Molecular Medicine, University of California, Berkeley, Berkeley, CA 94720, USA; Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA.
| | - Ophelia V Lee
- Division of Immunology and Molecular Medicine, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Stefan A Fattinger
- Division of Immunology and Molecular Medicine, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Charlotte A Langner
- Division of Immunology and Molecular Medicine, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Jaresley V Guillen
- Division of Immunology and Molecular Medicine, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Joshua M Peters
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Andres Moon
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30322, USA
| | - Eileen M Burd
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30322, USA
| | - Kristen C Witt
- Division of Immunology and Molecular Medicine, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Daniel B Stetson
- Department of Immunology, University of Washington, Seattle, WA 98195, USA
| | - David L Jaye
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30322, USA
| | - Bryan D Bryson
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Russell E Vance
- Division of Immunology and Molecular Medicine, University of California, Berkeley, Berkeley, CA 94720, USA; Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
38
|
Li LS, Yang L, Zhuang L, Ye ZY, Zhao WG, Gong WP. From immunology to artificial intelligence: revolutionizing latent tuberculosis infection diagnosis with machine learning. Mil Med Res 2023; 10:58. [PMID: 38017571 PMCID: PMC10685516 DOI: 10.1186/s40779-023-00490-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 11/06/2023] [Indexed: 11/30/2023] Open
Abstract
Latent tuberculosis infection (LTBI) has become a major source of active tuberculosis (ATB). Although the tuberculin skin test and interferon-gamma release assay can be used to diagnose LTBI, these methods can only differentiate infected individuals from healthy ones but cannot discriminate between LTBI and ATB. Thus, the diagnosis of LTBI faces many challenges, such as the lack of effective biomarkers from Mycobacterium tuberculosis (MTB) for distinguishing LTBI, the low diagnostic efficacy of biomarkers derived from the human host, and the absence of a gold standard to differentiate between LTBI and ATB. Sputum culture, as the gold standard for diagnosing tuberculosis, is time-consuming and cannot distinguish between ATB and LTBI. In this article, we review the pathogenesis of MTB and the immune mechanisms of the host in LTBI, including the innate and adaptive immune responses, multiple immune evasion mechanisms of MTB, and epigenetic regulation. Based on this knowledge, we summarize the current status and challenges in diagnosing LTBI and present the application of machine learning (ML) in LTBI diagnosis, as well as the advantages and limitations of ML in this context. Finally, we discuss the future development directions of ML applied to LTBI diagnosis.
Collapse
Affiliation(s)
- Lin-Sheng Li
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, the Eighth Medical Center of PLA General Hospital, Beijing, 100091, China
- Hebei North University, Zhangjiakou, 075000, Hebei, China
- Senior Department of Respiratory and Critical Care Medicine, the Eighth Medical Center of PLA General Hospital, Beijing, 100091, China
| | - Ling Yang
- Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Li Zhuang
- Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Zhao-Yang Ye
- Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Wei-Guo Zhao
- Senior Department of Respiratory and Critical Care Medicine, the Eighth Medical Center of PLA General Hospital, Beijing, 100091, China.
| | - Wen-Ping Gong
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, the Eighth Medical Center of PLA General Hospital, Beijing, 100091, China.
| |
Collapse
|
39
|
Mishra R, Hannebelle M, Patil VP, Dubois A, Garcia-Mouton C, Kirsch GM, Jan M, Sharma K, Guex N, Sordet-Dessimoz J, Perez-Gil J, Prakash M, Knott GW, Dhar N, McKinney JD, Thacker VV. Mechanopathology of biofilm-like Mycobacterium tuberculosis cords. Cell 2023; 186:5135-5150.e28. [PMID: 37865090 PMCID: PMC10642369 DOI: 10.1016/j.cell.2023.09.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 07/26/2023] [Accepted: 09/14/2023] [Indexed: 10/23/2023]
Abstract
Mycobacterium tuberculosis (Mtb) cultured axenically without detergent forms biofilm-like cords, a clinical identifier of virulence. In lung-on-chip (LoC) and mouse models, cords in alveolar cells contribute to suppression of innate immune signaling via nuclear compression. Thereafter, extracellular cords cause contact-dependent phagocyte death but grow intercellularly between epithelial cells. The absence of these mechanopathological mechanisms explains the greater proportion of alveolar lesions with increased immune infiltration and dissemination defects in cording-deficient Mtb infections. Compression of Mtb lipid monolayers induces a phase transition that enables mechanical energy storage. Agent-based simulations demonstrate that the increased energy storage capacity is sufficient for the formation of cords that maintain structural integrity despite mechanical perturbation. Bacteria in cords remain translationally active despite antibiotic exposure and regrow rapidly upon cessation of treatment. This study provides a conceptual framework for the biophysics and function in tuberculosis infection and therapy of cord architectures independent of mechanisms ascribed to single bacteria.
Collapse
Affiliation(s)
- Richa Mishra
- Global Health Institute, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Melanie Hannebelle
- Global Health Institute, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland; Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Vishal P Patil
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Anaëlle Dubois
- BioElectron Microscopy Facility, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | | | - Gabriela M Kirsch
- Global Health Institute, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Maxime Jan
- Bioinformatics Competence Centre, University of Lausanne, 1015 Lausanne, Switzerland; Bioinformatics Competence Centre, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Kunal Sharma
- Global Health Institute, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Nicolas Guex
- Bioinformatics Competence Centre, University of Lausanne, 1015 Lausanne, Switzerland; Bioinformatics Competence Centre, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Jessica Sordet-Dessimoz
- Histology Core Facility, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Jesus Perez-Gil
- Department of Biochemistry, University Complutense Madrid, 28040 Madrid, Spain
| | - Manu Prakash
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Graham W Knott
- BioElectron Microscopy Facility, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Neeraj Dhar
- Global Health Institute, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - John D McKinney
- Global Health Institute, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Vivek V Thacker
- Global Health Institute, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland.
| |
Collapse
|
40
|
Bobba S, Howard NC, Das S, Ahmed M, Tang L, Thirunavukkarasu S, Larsen MH, Mathema B, Divangahi M, Khader SA. Mycobacterium tuberculosis carrying the rifampicin drug-resistance-conferring rpoB mutation H445Y is associated with suppressed immunity through type I interferons. mBio 2023; 14:e0094623. [PMID: 37682004 PMCID: PMC10653897 DOI: 10.1128/mbio.00946-23] [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: 05/08/2023] [Accepted: 06/22/2023] [Indexed: 09/09/2023] Open
Abstract
IMPORTANCE This study highlights the impact of specific rifampicin-resistance-conferring mutations on the host immune response to Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB). Clinical reports have previously suggested that multi-drug-resistant) TB patients exhibit altered peripheral immune responses as compared with their drug-sensitive TB counterparts. The murine model of infection with Mtb strains carrying drug-resistance-conferring mutations recapitulated these findings and allowed us to mechanistically interrogate the pathways responsible for driving the divergent immune responses. Our findings underscore the need for greater investigation into bacterial heterogeneity to better appreciate the diversity in host-pathogen interactions during TB disease.
Collapse
Affiliation(s)
- Suhas Bobba
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nicole C. Howard
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Shibali Das
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Mushtaq Ahmed
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Microbiology, University of Chicago, Chicago, Illinois, USA
| | - Linrui Tang
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, New York, USA
| | - Shyamala Thirunavukkarasu
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Michelle H. Larsen
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Barun Mathema
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, New York, USA
| | - Maziar Divangahi
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montreal, Quebec, Canada
- Department of Microbiology and Immunology, McGill International TB Centre, Montreal, Quebec, Canada
- Department of Pathology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Shabaana A. Khader
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Microbiology, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
41
|
Sankar P, Mishra BB. Early innate cell interactions with Mycobacterium tuberculosis in protection and pathology of tuberculosis. Front Immunol 2023; 14:1260859. [PMID: 37965344 PMCID: PMC10641450 DOI: 10.3389/fimmu.2023.1260859] [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: 07/18/2023] [Accepted: 09/26/2023] [Indexed: 11/16/2023] Open
Abstract
Tuberculosis (TB) remains a significant global health challenge, claiming the lives of up to 1.5 million individuals annually. TB is caused by the human pathogen Mycobacterium tuberculosis (Mtb), which primarily infects innate immune cells in the lungs. These immune cells play a critical role in the host defense against Mtb infection, influencing the inflammatory environment in the lungs, and facilitating the development of adaptive immunity. However, Mtb exploits and manipulates innate immune cells, using them as favorable niche for replication. Unfortunately, our understanding of the early interactions between Mtb and innate effector cells remains limited. This review underscores the interactions between Mtb and various innate immune cells, such as macrophages, dendritic cells, granulocytes, NK cells, innate lymphocytes-iNKT and ILCs. In addition, the contribution of alveolar epithelial cell and endothelial cells that constitutes the mucosal barrier in TB immunity will be discussed. Gaining insights into the early cellular basis of immune reactions to Mtb infection is crucial for our understanding of Mtb resistance and disease tolerance mechanisms. We argue that a better understanding of the early host-pathogen interactions could inform on future vaccination approaches and devise intervention strategies.
Collapse
Affiliation(s)
| | - Bibhuti Bhusan Mishra
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, United States
| |
Collapse
|
42
|
Jani C, Marsh A, Uchil P, Jain N, Baskir ZR, Glover OT, Root DE, Doench JG, Barczak AK. Vps18 contributes to phagosome membrane integrity in Mycobacterium tuberculosis-infected macrophages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.01.560397. [PMID: 37873319 PMCID: PMC10592876 DOI: 10.1101/2023.10.01.560397] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Mycobacterium tuberculosis (Mtb) has evolved to be exquisitely adapted to survive within host macrophages. The capacity to damage the phagosomal membrane has emerged as central to Mtb virulence. While Mtb factors driving membrane damage have been described, host factors that repair that damage to contain the pathogen remain largely unknown. We used a genome-wide CRISPR screen to identify novel host factors required to repair Mtb-damaged phagosomal membranes. Vacuolar protein sorting-associated protein 18 (Vps18), a member of the HOPS and CORVET trafficking complexes, was among the top hits. Vps18 colocalized with Mtb in macrophages beginning shortly after infection, and Vps18-knockout macrophages demonstrated increased damage of Mtb-containing phagosomes without impaired autophagy. Mtb grew more robustly in Vps18-knockout cells, and the first-line anti-tuberculosis antibiotic pyrazinamide was less effective. Our results identify Vps18 as required for phagosomal membrane integrity in Mtb-infected cells and suggest that modulating phagosome integrity may hold promise for improving the efficacy of antibiotic treatment for TB.
Collapse
Affiliation(s)
| | | | - Pooja Uchil
- The Ragon Institute of MGH, MIT and Harvard
- Institute of Clinical and Molecular Virology, Friedrich-Alexander Universität Erlangen-Nürnberg
| | - Neha Jain
- The Ragon Institute of MGH, MIT and Harvard
| | | | | | | | | | - Amy K Barczak
- The Ragon Institute of MGH, MIT and Harvard
- The Broad Institute
- Division of Infectious Diseases, Massachusetts General Hospital
- Department of Medicine, Harvard Medical School
| |
Collapse
|
43
|
Mayer-Barber KD. Granulocytes subsets and their divergent functions in host resistance to Mycobacterium tuberculosis - a 'tipping-point' model of disease exacerbation. Curr Opin Immunol 2023; 84:102365. [PMID: 37437471 PMCID: PMC10543468 DOI: 10.1016/j.coi.2023.102365] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/29/2023] [Accepted: 06/14/2023] [Indexed: 07/14/2023]
Abstract
Granulocytes are innate immune effector cells with essential functions in host resistance to bacterial infections. I will discuss emerging evidence that during Mycobacterium tuberculosis infection, counter-intuitively, eosinophils are host-protective while neutrophils are host detrimental. Additionally, I will propose a 'tipping-point' model in which neutrophils are an integral part of a feedforward loop driving tuberculosis disease exacerbation.
Collapse
Affiliation(s)
- Katrin D Mayer-Barber
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, 20892, USA.
| |
Collapse
|
44
|
Yabaji SM, Rukhlenko OS, Chatterjee S, Bhattacharya B, Wood E, Kasaikina M, Kholodenko BN, Gimelbrant AA, Kramnik I. Cell state transition analysis identifies interventions that improve control of Mycobacterium tuberculosis infection by susceptible macrophages. SCIENCE ADVANCES 2023; 9:eadh4119. [PMID: 37756395 PMCID: PMC10530096 DOI: 10.1126/sciadv.adh4119] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023]
Abstract
Understanding cell state transitions and purposefully controlling them to improve therapies is a longstanding challenge in biological research and medicine. Here, we identify a transcriptional signature that distinguishes activated macrophages from the tuberculosis (TB) susceptible and resistant mice. We then apply the cSTAR (cell state transition assessment and regulation) approach to data from screening-by-RNA sequencing to identify chemical perturbations that shift the transcriptional state of tumor necrosis factor (TNF)-activated TB-susceptible macrophages toward that of TB-resistant cells, i.e., prevents their aberrant activation without suppressing beneficial TNF responses. Last, we demonstrate that the compounds identified with this approach enhance the resistance of the TB-susceptible mouse macrophages to virulent Mycobacterium tuberculosis.
Collapse
Affiliation(s)
- Shivraj M. Yabaji
- The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, USA
| | - Oleksii S. Rukhlenko
- Systems Biology Ireland, School of Medicine and Medical Science, University College Dublin, Belfield Dublin 4, Ireland
| | - Sujoy Chatterjee
- The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, USA
| | - Bidisha Bhattacharya
- The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, USA
| | - Emily Wood
- Systems Biology Ireland, School of Medicine and Medical Science, University College Dublin, Belfield Dublin 4, Ireland
| | - Marina Kasaikina
- The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, USA
| | - Boris N. Kholodenko
- Systems Biology Ireland, School of Medicine and Medical Science, University College Dublin, Belfield Dublin 4, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield Dublin 4, Ireland
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA
| | | | - Igor Kramnik
- The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, USA
- Pulmonary Center, The Department of Medicine, Boston University School of Medicine, Boston, MA, USA
- Department of Microbiology, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
45
|
Bloom BR. A half-century of research on tuberculosis: Successes and challenges. J Exp Med 2023; 220:e20230859. [PMID: 37552470 PMCID: PMC10407785 DOI: 10.1084/jem.20230859] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/20/2023] [Accepted: 07/21/2023] [Indexed: 08/09/2023] Open
Abstract
Great progress has been made over the past half-century, but TB remains a formidable global health problem, particularly in low- and middle-income countries. Understanding the mechanisms of pathogenesis and necessary and sufficient conditions for protection are critical. The need for inexpensive and sensitive point-of-care diagnostic tests for earlier detection of infection and disease, shorter and less-toxic drug regimens for drug-sensitive and -resistant TB, and a more effective vaccine than BCG is immense. New and better tools, greater support for international research, collaborations, and training will be required to dramatically reduce the burden of this devastating disease which still kills 1.6 million people annually.
Collapse
Affiliation(s)
- Barry R. Bloom
- Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
46
|
Baker PJ, Amaral EP, Castro E, Bohrer AC, Torres-Juárez F, Jordan CM, Nelson CE, Barber DL, Johnson RF, Hilligan KL, Mayer-Barber KD. Co-infection of mice with SARS-CoV-2 and Mycobacterium tuberculosis limits early viral replication but does not affect mycobacterial loads. Front Immunol 2023; 14:1240419. [PMID: 37720210 PMCID: PMC10502726 DOI: 10.3389/fimmu.2023.1240419] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 08/15/2023] [Indexed: 09/19/2023] Open
Abstract
Viral co-infections have been implicated in worsening tuberculosis (TB) and during the COVID-19 pandemic, the global rate of TB-related deaths has increased for the first time in over a decade. We and others have previously shown that a resolved prior or concurrent influenza A virus infection in Mycobacterium tuberculosis (Mtb)-infected mice resulted in increased pulmonary bacterial burden, partly through type I interferon (IFN-I)-dependent mechanisms. Here we investigated whether SARS-CoV-2 (SCV2) co-infection could also negatively affect bacterial control of Mtb. Importantly, we found that K18-hACE2 transgenic mice infected with SCV2 one month before, or months after aerosol Mtb exposure did not display exacerbated Mtb infection-associated pathology, weight loss, nor did they have increased pulmonary bacterial loads. However, pre-existing Mtb infection at the time of exposure to the ancestral SCV2 strain in infected K18-hACE2 transgenic mice or the beta variant (B.1.351) in WT C57Bl/6 mice significantly limited early SCV2 replication in the lung. Mtb-driven protection against SCV2 increased with higher bacterial doses and did not require IFN-I, TLR2 or TLR9 signaling. These data suggest that SCV2 co-infection does not exacerbate Mtb infection in mice, but rather the inflammatory response generated by Mtb infection in the lungs at the time of SCV2 exposure restricts viral replication.
Collapse
Affiliation(s)
- Paul J. Baker
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Eduardo P. Amaral
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Ehydel Castro
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Andrea C. Bohrer
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Flor Torres-Juárez
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Cassandra M. Jordan
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Christine E. Nelson
- T Lymphocyte Biology Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD, United States
| | - Daniel L. Barber
- T Lymphocyte Biology Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD, United States
| | - Reed F. Johnson
- SARS-CoV-2 Virology Core, Laboratory of Viral Diseases, NIAID, NIH, Bethesda, MD, United States
| | - Kerry L. Hilligan
- Immunobiology Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD, United States
| | - Katrin D. Mayer-Barber
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
47
|
Jani C, Solomon SL, Peters JM, Pringle SC, Hinman AE, Boucau J, Bryson BD, Barczak AK. TLR2 is non-redundant in the population and subpopulation responses to Mycobacterium tuberculosis in macrophages and in vivo. mSystems 2023; 8:e0005223. [PMID: 37439558 PMCID: PMC10506474 DOI: 10.1128/msystems.00052-23] [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: 01/16/2023] [Accepted: 06/02/2023] [Indexed: 07/14/2023] Open
Abstract
Tuberculosis (TB), caused by the pathogenic bacterium Mycobacterium tuberculosis (Mtb), is a global health threat. Targeting host pathways that modulate protective or harmful components of inflammation has been proposed as a therapeutic strategy that could aid sterilization or mitigate TB-associated permanent tissue damage. In purified form, many Mtb components can activate innate immune pathways. However, knowledge of the pathways that contribute most to the observed response to live Mtb is incomplete, limiting the possibility of precise intervention. We took a systematic, unbiased approach to define the pathways that drive the earliest immune response to Mtb. Using a macrophage model of infection, we compared the bulk transcriptional response to infection with the response to a panel of Mtb-derived putative innate immune ligands. We identified two axes of response: an NF-kB-dependent response similarly elicited by all Mtb pathogen-associated molecular patterns (PAMPs) and a type I interferon axis unique to cells infected with live Mtb. Consistent with growing literature data pointing to TLR2 as a dominant Mtb-associated PAMP, the TLR2 ligand PIM6 most closely approximated the NF-kB-dependent response to the intact bacterium. Quantitatively, the macrophage response to Mtb was slower and weaker than the response to purified PIM6. On a subpopulation level, the TLR2-dependent response was heterogeneously induced, with only a subset of infected cells expressing key inflammatory genes known to contribute to the control of infection. Despite potential redundancies in Mtb ligand/innate immune receptor interactions during in vivo infection, loss of the TLR2/PIM6 interaction impacted the cellular composition of both the innate and adaptive compartments. IMPORTANCE Tuberculosis (TB) is a leading cause of death globally. Drug resistance is outpacing new antibiotic discovery, and even after successful treatment, individuals are often left with permanent lung damage from the negative consequences of inflammation. Targeting host inflammatory pathways has been proposed as an approach that could either improve sterilization or improve post-treatment lung health. However, our understanding of the inflammatory pathways triggered by Mycobacterium tuberculosis (Mtb) in infected cells and lungs is incomplete, in part because of the complex array of potential molecular interactions between bacterium and host. Here, we take an unbiased approach to identify the pathways most central to the host response to Mtb. We examine how individual pathways are triggered differently by purified Mtb products or infection with the live bacterium and consider how these pathways inform the emergence of subpopulation responses in cell culture and in infected mice. Understanding how individual interactions and immune pathways contribute to inflammation in TB opens the door to the possibility of developing precise therapeutic interventions.
Collapse
Affiliation(s)
- Charul Jani
- The Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
| | - Sydney L. Solomon
- The Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Joshua M. Peters
- The Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | | | - Amelia E. Hinman
- The Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
| | - Julie Boucau
- The Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
| | - Bryan D. Bryson
- The Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Amy K. Barczak
- The Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
- The Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
48
|
Gander-Bui HTT, Schläfli J, Baumgartner J, Walthert S, Genitsch V, van Geest G, Galván JA, Cardozo C, Graham Martinez C, Grans M, Muth S, Bruggmann R, Probst HC, Gabay C, Freigang S. Targeted removal of macrophage-secreted interleukin-1 receptor antagonist protects against lethal Candida albicans sepsis. Immunity 2023; 56:1743-1760.e9. [PMID: 37478856 DOI: 10.1016/j.immuni.2023.06.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 03/02/2023] [Accepted: 06/27/2023] [Indexed: 07/23/2023]
Abstract
Invasive fungal infections are associated with high mortality rates, and the lack of efficient treatment options emphasizes an urgency to identify underlying disease mechanisms. We report that disseminated Candida albicans infection is facilitated by interleukin-1 receptor antagonist (IL-1Ra) secreted from macrophages in two temporally and spatially distinct waves. Splenic CD169+ macrophages release IL-1Ra into the bloodstream, impeding early neutrophil recruitment. IL-1Ra secreted by monocyte-derived tissue macrophages further impairs pathogen containment. Therapeutic IL-1Ra neutralization restored the functional competence of neutrophils, corrected maladapted hyper-inflammation, and eradicated the otherwise lethal infection. Conversely, augmentation of macrophage-secreted IL-1Ra by type I interferon severely aggravated disease mortality. Our study uncovers how a fundamental immunoregulatory mechanism mediates the high disease susceptibility to invasive candidiasis. Furthermore, interferon-stimulated IL-1Ra secretion may exacerbate fungal dissemination in human patients with secondary candidemia. Macrophage-secreted IL-1Ra should be considered as an additional biomarker and potential therapeutic target in severe systemic candidiasis.
Collapse
Affiliation(s)
- Hang Thi Thuy Gander-Bui
- Division of Experimental Pathology, Institute of Tissue Medicine and Pathology, University of Bern, 3008 Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Joëlle Schläfli
- Division of Experimental Pathology, Institute of Tissue Medicine and Pathology, University of Bern, 3008 Bern, Switzerland
| | - Johanna Baumgartner
- Division of Experimental Pathology, Institute of Tissue Medicine and Pathology, University of Bern, 3008 Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Sabrina Walthert
- Division of Experimental Pathology, Institute of Tissue Medicine and Pathology, University of Bern, 3008 Bern, Switzerland
| | - Vera Genitsch
- Institute of Tissue Medicine and Pathology, University of Bern, 3008 Bern, Switzerland
| | - Geert van Geest
- Interfaculty Bioinformatics Unit and Swiss Institute of Bioinformatics, University of Bern, 3012 Bern, Switzerland
| | - José A Galván
- Institute of Tissue Medicine and Pathology, University of Bern, 3008 Bern, Switzerland
| | - Carmen Cardozo
- Institute of Tissue Medicine and Pathology, University of Bern, 3008 Bern, Switzerland
| | | | - Mona Grans
- Institute for Immunology, University Medical Center Mainz, 55131 Mainz, Germany
| | - Sabine Muth
- Institute for Immunology, University Medical Center Mainz, 55131 Mainz, Germany
| | - Rémy Bruggmann
- Interfaculty Bioinformatics Unit and Swiss Institute of Bioinformatics, University of Bern, 3012 Bern, Switzerland
| | | | - Cem Gabay
- Division of Rheumatology, Department of Medicine, University Hospital of Geneva, 1211 Geneva, Switzerland
| | - Stefan Freigang
- Division of Experimental Pathology, Institute of Tissue Medicine and Pathology, University of Bern, 3008 Bern, Switzerland.
| |
Collapse
|
49
|
Zhu C, Xu L, Chen L, Zhang Z, Zhang Y, Wu W, Li C, Liu S, Xiang S, Dai S, Zhang J, Guo H, Zhou Y, Wang F. Epitope-Directed Antibody Elicitation by Genetically Encoded Chemical Cross-Linking Reactivity in the Antigen. ACS CENTRAL SCIENCE 2023; 9:1229-1240. [PMID: 37396855 PMCID: PMC10311653 DOI: 10.1021/acscentsci.3c00265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Indexed: 07/04/2023]
Abstract
No current methods can selectively elicit an antibody response to a specific conformational epitope in a whole antigen in vivo. Here, we incorporated Nε-acryloyl-l-lysine (AcrK) or Nε-crotonyl-l-lysine (Kcr) with cross-linking activities into the specific epitopes of antigens and immunized mice to generate antibodies that can covalently cross-link with the antigens. By taking advantage of antibody clonal selection and evolution in vivo, an orthogonal antibody-antigen cross-linking reaction can be generated. With this mechanism, we developed a new approach for facile elicitation of antibodies binding to specific epitopes of the antigen in vivo. Antibody responses were directed and enriched to the target epitopes on protein antigens or peptide-KLH conjugates after mouse immunization with the AcrK or Kcr-incorporated immunogens. The effect is so prominent that the majority of selected hits bind to the target epitope. Furthermore, the epitope-specific antibodies effectively block IL-1β from activating its receptor, indicating its potential for the development of protein subunit vaccines.
Collapse
Affiliation(s)
- Chaoyang Zhu
- Key
Laboratory of Protein and Peptide Pharmaceutical, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- College
of Life Sciences, University of Chinese
Academy of Sciences, Beijing 100101, China
| | - Liang Xu
- Key
Laboratory of Protein and Peptide Pharmaceutical, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- College
of Life Sciences, University of Chinese
Academy of Sciences, Beijing 100101, China
| | - Longxin Chen
- Key
Laboratory of Protein and Peptide Pharmaceutical, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- Molecular
Biology Laboratory, Zhengzhou Normal University, Zhengzhou 450044, China
| | - Zihan Zhang
- Key
Laboratory of Protein and Peptide Pharmaceutical, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yuhan Zhang
- Key
Laboratory of Protein and Peptide Pharmaceutical, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Weiping Wu
- Suzhou
Institute for Biomedical Research, Suzhou, Jiangsu 215028, China
| | - Chengxiang Li
- Key
Laboratory of Protein and Peptide Pharmaceutical, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- College
of Life Sciences, University of Chinese
Academy of Sciences, Beijing 100101, China
| | - Shuang Liu
- Key
Laboratory of Protein and Peptide Pharmaceutical, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- College
of Life Sciences, University of Chinese
Academy of Sciences, Beijing 100101, China
| | - Shuqin Xiang
- Key
Laboratory of Protein and Peptide Pharmaceutical, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- College
of Life Sciences, University of Chinese
Academy of Sciences, Beijing 100101, China
| | - Shengwang Dai
- Key
Laboratory of Protein and Peptide Pharmaceutical, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- College
of Life Sciences, University of Chinese
Academy of Sciences, Beijing 100101, China
| | - Jay Zhang
- Key
Laboratory of Protein and Peptide Pharmaceutical, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- Suzhou
Institute for Biomedical Research, Suzhou, Jiangsu 215028, China
| | - Hui Guo
- Key
Laboratory of Protein and Peptide Pharmaceutical, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- Suzhou
Institute for Biomedical Research, Suzhou, Jiangsu 215028, China
- Beijing
Translational Center for Biopharmaceuticals, Beijing 100101, China
| | - Yinjian Zhou
- Key
Laboratory of Protein and Peptide Pharmaceutical, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- Beijing
Translational Center for Biopharmaceuticals, Beijing 100101, China
| | - Feng Wang
- Key
Laboratory of Protein and Peptide Pharmaceutical, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- Suzhou
Institute for Biomedical Research, Suzhou, Jiangsu 215028, China
- Beijing
Translational Center for Biopharmaceuticals, Beijing 100101, China
| |
Collapse
|
50
|
Abstract
Mycobacteria are responsible for several human and animal diseases. NOD2 is a pattern recognition receptor that has an important role in mycobacterial recognition. However, the mechanisms by which mutations in NOD2 alter the course of mycobacterial infection remain unclear. Herein, we aimed to review the totality of studies directly addressing the relationship between NOD2 and mycobacteria as a foundation for moving the field forward. NOD2 was linked to mycobacterial infection at 3 levels: (1) genetic, through association with mycobacterial diseases of humans; (2) chemical, through the distinct NOD2 ligand in the mycobacterial cell wall; and (3) immunologic, through heightened NOD2 signaling caused by the unique modification of the NOD2 ligand. The immune response to mycobacteria is shaped by NOD2 signaling, responsible for NF-κB and MAPK activation, and the production of various immune effectors like cytokines and nitric oxide, with some evidence linking this to bacteriologic control. Absence of NOD2 during mycobacterial infection of mice can be detrimental, but the mechanism remains unknown. Conversely, the success of immunization with mycobacteria has been linked to NOD2 signaling and NOD2 has been targeted as an avenue of immunotherapy for diseases even beyond mycobacteria. The mycobacteria-NOD2 interaction remains an important area of study, which may shed light on immune mechanisms in disease.
Collapse
Affiliation(s)
- Jean-Yves Dubé
- Department of Microbiology and Immunology, McGill University, Montréal, Canada
| | - Marcel A. Behr
- Department of Medicine, McGill University Health Centre, Montréal, Canada
| |
Collapse
|