1
|
Agulló-Ros I, Burucúa MM, Cheuquepán FA, Domínguez M, Sevilla IA, Martínez R, Plá N, Risalde MA, Marin MS. Heat-inactivated Mycobacterium bovis and P22PI protein immunocomplex: Two candidates for use as immunostimulants of innate immune response. Vet Microbiol 2025; 305:110527. [PMID: 40279721 DOI: 10.1016/j.vetmic.2025.110527] [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: 09/20/2024] [Revised: 03/22/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
Tuberculosis (TB), caused by members of the Mycobacterium tuberculosis complex, remains a critical global health challenge, affecting humans and a wide range of domestic and wild animals. Despite the availability of anti-TB drugs, cure rates remain suboptimal, exacerbated by the rise of multidrug-resistant TB strains. The Bacille Calmette-Guérin (BCG) vaccine, the only licensed vaccine against TB, has demonstrated efficacy in reducing lesion severity and bacterial burden in animals, as well as lowering TB-related and all-cause mortality in infants. However, BCG presents several safety concerns inherent to live vaccines. To overcome these limitations, exploring alternative vaccine candidates that do not incorporate live mycobacteria is crucial. This study aimed to evaluate and compare the immunostimulatory potential of two candidates based in mycobacteria inactivated or their derivatives, heat-inactivated Mycobacterium bovis (HIMB) and P22PI protein immunocomplex (P22PI), in bovine foetal lung cells. To assess the expression of innate immune components, including Toll-like receptors (TLRs), cathelicidins, and cytokines, bovine foetal lung were exposed to different concentrations of HIMB and P22PI immunostimulants, starting at 7.8 × 10⁶ CFU/ml and 10 µg/ml, respectively. These initial concentrations were subsequently diluted to 1/2 and 1/10 to evaluate dose-dependent effects. Our findings reveal that both HIMB and P22PI significantly stimulate innate immune mechanisms, as evidenced by the upregulation of TLR2 and TLR4, alongside the induction of BMAP28 cathelicidin, tumour necrosis factor alpha (TNFA) and interferons (IFNs). These results suggest their potential to orchestrate a robust innate immune response providing valuable insights into the immunological mechanisms underlying the protective effects of these immunostimulants. This underscores their potential role in in vivo studies as vaccine candidates. Furthermore, their ability to enhance antigen recognition via TLR and induce pro-inflammatory cytokines also indicates broader applications in immune modulation, potentially extending protection against heterologous pathogens through trained immunity.
Collapse
Affiliation(s)
- Irene Agulló-Ros
- Departamento de Anatomía y Anatomía Patológica Comparadas y Toxicología, Grupo de Investigación en Sanidad Animal y Zoonosis (GISAZ), UIC Zoonosis y Enfermedades Emergentes ENZOEM, Universidad de Córdoba, Córdoba, Spain
| | - Mercedes M Burucúa
- Departamento de Biología, Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar del Plata, Mar del Plata, Buenos Aires, Argentina
| | - Felipe A Cheuquepán
- Departamento de Biología, Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar del Plata, Mar del Plata, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Mercedes Domínguez
- Servicio de Inmunología Microbiana, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Iker A Sevilla
- Animal Health Department, NEIKER-Instituto Vasco de Investigación y Desarrollo Agrario, Derio, Bizkaia, Spain
| | - Remigio Martínez
- Departamento de Sanidad Animal, Grupo de Investigación en Sanidad Animal y Zoonosis (GISAZ), UIC Zoonosis y Enfermedades Emergentes ENZOEM, Universidad de Córdoba, Córdoba, Spain
| | - Natalia Plá
- Facultad de Ciencias Agrarias, Universidad Nacional de Mar del Plata, Mar del Plata, Buenos Aires, Argentina
| | - María A Risalde
- Departamento de Anatomía y Anatomía Patológica Comparadas y Toxicología, Grupo de Investigación en Sanidad Animal y Zoonosis (GISAZ), UIC Zoonosis y Enfermedades Emergentes ENZOEM, Universidad de Córdoba, Córdoba, Spain; CIBERINFEC, ISCIII - CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain.
| | - Maia S Marin
- Departamento de Biología, Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar del Plata, Mar del Plata, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
2
|
Prasanna P, Herrera B, Schlesinger LS, Paiardini M, Sharan R. Advances in host-directed therapy for tuberculosis and HIV coinfection: enhancing immune responses. Trends Microbiol 2025:S0966-842X(25)00117-9. [PMID: 40345900 DOI: 10.1016/j.tim.2025.04.006] [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: 01/24/2025] [Revised: 03/17/2025] [Accepted: 04/03/2025] [Indexed: 05/11/2025]
Abstract
Tuberculosis (TB) and HIV coinfection present a significant global health challenge worldwide. While most individuals infected with Mycobacterium tuberculosis (Mtb) have clinically asymptomatic latent TB infection (LTBI), those with immunosuppressive conditions, such as HIV, are at higher risk for reactivation and disseminated TB. HIV exacerbates TB progression by impairing immune responses, particularly through the depletion of Mtb-specific CD4+ T cells and chronic immune activation. Despite the success of combined antiretroviral therapy (cART) in managing HIV, it does not eliminate the risk of LTBI reactivation, highlighting the need for additional therapeutic strategies. Host-directed therapy (HDT) has emerged as a promising adjunct to current treatments. HDTs aim to restore immune function and counteract immune dysregulation caused by HIV, including T cell exhaustion and inflammation. This review explores key HDTs, including cytokine therapy, chimeric antigen receptor T cell (CAR-T cell) therapy and immune checkpoint inhibitors, which target Mtb-infected cells, enhance immune responses, and mitigate inflammation. By complementing cART and anti-TB therapy, HDTs could improve clinical outcomes by enhancing immune function, reducing inflammation, and preventing Mtb reactivation, offering hope for better management of TB/HIV coinfection.
Collapse
Affiliation(s)
- Preena Prasanna
- Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Bianca Herrera
- Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | | | - Mirko Paiardini
- Emory National Primate Research Center, Emory University, Atlanta, GA, USA; Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Riti Sharan
- Texas Biomedical Research Institute, San Antonio, TX 78227, USA.
| |
Collapse
|
3
|
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
|
4
|
Zapico D, Espinosa J, Criado M, Gutiérrez D, Ferreras MDC, Benavides J, Pérez V, Fernández M. Immunohistochemical expression of TLR1, TLR2, TLR4, and TLR9 in the different types of lesions associated with bovine paratuberculosis. Vet Pathol 2025; 62:305-318. [PMID: 39720873 DOI: 10.1177/03009858241302850] [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: 12/26/2024]
Abstract
The factors that determine the appearance of the different pathologic forms associated with bovine paratuberculosis are not fully understood, but new research suggests a critical role of innate immunity. Toll-like receptors (TLRs) trigger the recognition of invading pathogens by innate immune cells and the onset of specific immune responses. The aim of this work was to assess, immunohistochemically, the expression of TLR1, TLR2, TLR4, and TLR9 in intestinal samples of 20 cows showing different types of paratuberculous lesions: uninfected controls, focal lesions, paucibacillary, and multibacillary diffuse forms. The majority of labeled cells were morphologically consistent with macrophages. A differential cell count was performed in the intestinal lamina propria, gut-associated lymphoid tissue, and mesenteric lymph node. TLR9 immunolabeling between the different types of lesions was compared using a complete H-score. Focal and diffuse paucibacillary forms contained significantly increased TLR2-expressing macrophages outside of the lesions compared with the controls and diffuse multibacillary forms, and moderate TLR9 immunolabeling within granulomas. In the multibacillary granulomatous lesions, the expression of TLR1 and TLR4 was observed as well as increased TLR9 expression compared with the rest of the groups. Differences in the predominance of one type or another of TLR allows us to elucidate the importance of the innate immune response and its possible role in the development of the different types of paratuberculosis lesions.
Collapse
Affiliation(s)
- David Zapico
- Universidad de León, León, Spain
- Instituto de Ganadería de Montaña, León, Spain
| | - José Espinosa
- Universidad de León, León, Spain
- Instituto de Ganadería de Montaña, León, Spain
| | - Miguel Criado
- Universidad de León, León, Spain
- Instituto de Ganadería de Montaña, León, Spain
| | - Daniel Gutiérrez
- Universidad de León, León, Spain
- Instituto de Ganadería de Montaña, León, Spain
| | | | | | - Valentín Pérez
- Universidad de León, León, Spain
- Instituto de Ganadería de Montaña, León, Spain
| | | |
Collapse
|
5
|
Hilligan KL, Darrah PA, Seder RA, Sher A. Deconvoluting the interplay of innate and adaptive immunity in BCG-induced nonspecific and TB-specific host resistance. J Exp Med 2025; 222:e20240496. [PMID: 40100096 PMCID: PMC11917170 DOI: 10.1084/jem.20240496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/23/2025] [Accepted: 02/27/2025] [Indexed: 03/20/2025] Open
Abstract
BCG is the oldest vaccine in continuous use. While current intradermal vaccination regimens confer limited protection outside the context of pediatric extrapulmonary tuberculosis (TB), promising new data indicate that when administered mucosally or intravenously at a higher dose, BCG can induce sterilizing immunity against pulmonary TB in nonhuman primates. BCG is also known to promote nonspecific host resistance against a variety of unrelated infections and is a standard immunotherapy for bladder cancer, suggesting that this innate immune function may contribute to its protective role against TB. Here, we propose that both the mycobacterial-specific and off-target effects of BCG depend on the interplay of adaptive and innate cells and the cytokines they produce, and that the elucidation of this interaction should be a major strategy in the development of more effective BCG-based vaccines and immunotherapies.
Collapse
Affiliation(s)
| | - Patricia A. Darrah
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Robert A. Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Alan Sher
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
6
|
Bhaskar V, Kumar R, Praharaj MR, Gandham S, Maity HK, Sarkar U, Dey B. A bovine pulmosphere model and multiomics reveal early host response signature in tuberculosis. Commun Biol 2025; 8:559. [PMID: 40186000 PMCID: PMC11971429 DOI: 10.1038/s42003-025-07883-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 03/04/2025] [Indexed: 04/07/2025] Open
Abstract
Early interactions between tubercle bacilli and lung cells are critical in tuberculosis (TB) pathogenesis. Conventional two-dimensional cell cultures fail to replicate the multicellular complexity of lungs. We introduce a three-dimensional pulmosphere model for Mycobacterium tuberculosis infection in bovine systems, demonstrating through comprehensive transcriptome and proteome analyses that these multicellular spheroids closely mimic lung cell diversity, interactions, and extracellular matrix (ECM) composition. Cell viability, hypoxia, and reactive oxygen species assessments over three weeks confirm the model's suitability. To establish infection, we employed M. bovis BCG-an attenuated vaccine strain, and M. tuberculosis H37Rv-a laboratory adapted human clinical strain that is attenuated for cattle infection compared to M. bovis. Both infection upregulated key host pathways; however, M. tuberculosis induced distinct responses, including enhanced ECM receptors expression, neutrophil chemotaxis, interferon signaling, and RIG-1 signaling. A six genes/protein signature- IRF1, CCL5, CXCL8, CXCL10, SERPINE1, and CFB -emerges as an early host response marker to M. tuberculosis infection. Infection with virulent M. bovis and M. orygis revealed a shared upregulated gene signature across Mycobacterium tuberculosis complex species, but with pathogen-specific variations. This study presents a robust ex vivo bovine pulmosphere TB model with implications in biomarkers discovery, high-throughput drug screening, and TB control strategies.
Collapse
Affiliation(s)
- Vinay Bhaskar
- National Institute of Animal Biotechnology, Hyderabad, Telangana, India
| | - Rishi Kumar
- National Institute of Animal Biotechnology, Hyderabad, Telangana, India
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Manas Ranjan Praharaj
- National Institute of Animal Biotechnology, Hyderabad, Telangana, India
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Sripratyusha Gandham
- National Institute of Animal Biotechnology, Hyderabad, Telangana, India
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Hemanta Kumar Maity
- Department of Avian Sciences, West Bengal University of Animal and Fishery Sciences, Kolkata, West Bengal, India
| | - Uttam Sarkar
- Department of Animal Genetics and Breeding, West Bengal University of Animal and Fishery Sciences, Kolkata, West Bengal, India
| | - Bappaditya Dey
- National Institute of Animal Biotechnology, Hyderabad, Telangana, India.
- Regional Centre for Biotechnology, Faridabad, Haryana, India.
| |
Collapse
|
7
|
Han X, Wang X, Han F, Yan H, Sun J, Zhang X, Moog C, Zhang C, Su B. The cGAS-STING pathway in HIV-1 and Mycobacterium tuberculosis coinfection. Infection 2025; 53:495-511. [PMID: 39509013 DOI: 10.1007/s15010-024-02429-0] [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: 04/23/2024] [Accepted: 10/28/2024] [Indexed: 11/15/2024]
Abstract
Mycobacterium tuberculosis (M. tuberculosis) infection is the most common opportunistic infection in human immunodeficiency virus-1 (HIV-1)-infected individuals, and the mutual reinforcement of these two pathogens may accelerate disease progression and lead to rapid mortality. Therefore, HIV-1/M. tuberculosis coinfection is one of the major global public health concerns. HIV-1 infection is the greatest risk factor for M. tuberculosis infection and increases the likelihood of endogenous relapse and exogenous reinfection with M. tuberculosis. Moreover, M. tuberculosis further increases HIV-1 replication and the occurrence of chronic immune activation, accelerating the progression of HIV-1 disease. Exploring the pathogenesis of HIV-1/M. tuberculosis coinfections is essential for the development of novel treatments to reduce the global burden of tuberculosis. Innate immunity, which is the first line of host immune defense, plays a critical role in resisting HIV-1 and M. tuberculosis infections. The role of the cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway, which is a major DNA-sensing innate immune signaling pathway, in HIV-1 infection and M. tuberculosis infection has been intensively studied. This paper reviews the role of the cGAS-STING signaling pathway in HIV-1 infection and M. tuberculosis infection and discusses the possible role of this pathway in HIV-1/M. tuberculosis coinfection to provide new insight into the pathogenesis of HIV-1/M. tuberculosis coinfection and the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Xiaoxu Han
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
- Sino-French Joint Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Xiuwen Wang
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
- Sino-French Joint Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Fangping Han
- State Key Laboratory of Membrane Biology, School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084, China
| | - Hongxia Yan
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
- Sino-French Joint Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Jin Sun
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
- Sino-French Joint Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Xin Zhang
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
- Sino-French Joint Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Christiane Moog
- Sino-French Joint Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
- Laboratoire d'ImmunoRhumatologie Moléculaire, Faculté de Médecine, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR_S 1109, Institut Thématique Interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Fédération Hospitalo-Universitaire OMICARE, Université de Strasbourg, Transplantex, Strasbourg, NG, 67000, France
- Vaccine Research Institute (VRI), Créteil, 94000, France
| | - Conggang Zhang
- State Key Laboratory of Membrane Biology, School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China.
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084, China.
| | - Bin Su
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China.
- Sino-French Joint Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
8
|
O'Garra A. From Cytokines to Tuberculosis and Back: My Journey to Understanding the Immune Response to Infection. Annu Rev Immunol 2025; 43:1-28. [PMID: 40279305 DOI: 10.1146/annurev-immunol-010824-041601] [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
I felt honored by the invitation to write this autobiography, although it was an arduous task to describe my journey through science: first bacterial adhesion, then cytokine function, and then immune responses in tuberculosis. Since only seven women had been authors of autobiographies for the Annual Review of Immunology, I felt I couldn't refuse to contribute to Volume 43 of the journal. Moreover, this was a good occasion to record my appreciation to all the lab members and collaborators for their contributions over the last 40 years, to remember the exciting times, and to reflect on the obstacles we faced. I often reflect on this line that is commonly attributed to Winston Churchill: Success is not final; failure is not fatal: It is the courage to continue that counts. What kept me going was a burning desire to know how things work and find enjoyment in the discovery. This passion to understand immune responses to infection remains with me to this day. I thank all those I have interacted with for the support and friendship they provided.
Collapse
Affiliation(s)
- Anne O'Garra
- Laboratory of Immunoregulation and Infection, The Francis Crick Institute, London, United Kingdom;
| |
Collapse
|
9
|
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: 1] [Impact Index Per Article: 1.0] [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
|
10
|
Chang E, Cavallo K, Behar SM. CD4 T cell dysfunction is associated with bacterial recrudescence during chronic tuberculosis. Nat Commun 2025; 16:2636. [PMID: 40097414 PMCID: PMC11914476 DOI: 10.1038/s41467-025-57819-1] [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: 07/12/2024] [Accepted: 03/04/2025] [Indexed: 03/19/2025] Open
Abstract
While most people contain Mycobacterium tuberculosis infection, some individuals develop active disease, usually within two years of infection. Why immunity fails after initially controlling infection is unknown. C57BL/6 mice control Mycobacterium tuberculosis for up to a year but ultimately succumb to disease. We hypothesize that the development of CD4 T cell dysfunction permits bacterial recrudescence. We developed a reductionist model to assess antigen-specific T cells during chronic infection and found evidence of CD4 T cell senescence and exhaustion. In C57BL/6 mice, CD4 T cells upregulate coinhibitory receptors and lose effector cytokine production. Single cell RNAseq shows that only a small number of CD4 T cells in the lungs of chronically infected mice are polyfunctional. While the origin and causal relationship between T-cell dysfunction and recrudescence remains uncertain, we propose T cell dysfunction leads to a feed-forward loop that causes increased bacillary numbers, greater T cell dysfunction, and progressive disease.
Collapse
Affiliation(s)
- Evelyn Chang
- Immunology and Microbiology Program, Morningside Graduate School of Biomedical Sciences, Worcester, MA, USA
- Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Kelly Cavallo
- Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Samuel M Behar
- Immunology and Microbiology Program, Morningside Graduate School of Biomedical Sciences, Worcester, MA, USA.
- Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
11
|
Yabaji SM, Lata S, Tseng AE, Araveti PB, Lo M, Gavrish I, O’Connell AK, Gertje HP, Belkina AC, Thurman CE, Kiyokawa H, Kotton D, Tan S, Endsley JJ, Bishai WR, Crossland N, Kobzik L, Kramnik I. Aberrant macrophage activation and maladaptive lung repair promote tuberculosis progression uniquely in the lung. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.10.17.562695. [PMID: 40093068 PMCID: PMC11908135 DOI: 10.1101/2023.10.17.562695] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Pulmonary tuberculosis (PTB) represents 85% of the disease burden caused by Mycobacterium tuberculosis (Mtb) and promotes aerosol transmission infecting about a quarter of people globally. Most Mtb infections are effectively limited within primary granulomatous lesions. Containment failures lead to hematogenous spread and the formation of post-primary destructive PTB lesions. Factors that favor Mtb survival and replication in the lungs after hematogenous spread despite systemic immunity represent appealing targets for host-directed TB therapies, but are currently unknown. We developed a novel mouse model that mimics progression of chronic post-primary PTB in humans: wherein PTB lesions form after hematogenous spread from a remote primary lesion in immunocompetent but TB-susceptible B6.Sst1S mice. The B6.Sst1S mice developed PTB lesions featuring granulomatous pneumonia, bronchogenic expansion and broncho-occlusion closely resembling post-primary PTB in humans. Using spatial transcriptomic and fluorescent multiplexed immunochemistry, we demonstrated the expansion of myeloid cell populations with the appearance of alternatively activated macrophages, dissolution of initial lymphoid follicles, and accumulation of de-differentiated lung epithelial cells in the advanced PTB lesions. To determine whether lung parenchymal cells or lung oxygenation were necessary for the pulmonary TB progression, we implanted lung and spleen fragments subcutaneously to serve as potential targets for hematogenous spread. The lung (but not spleen) implants displayed characteristic organized granulomas with necrosis and Mtb replication demonstrating that deleterious interactions of aberrantly activated macrophages with the inflammation-injured lung resident cells, and possibly hypoxia, not oxygenation, are critical determinants of PTB progression in immunocompetent hosts. Necrotic TB lesions also developed in subcutaneous implants of human lung tissue in mice with human immune system after respiratory infection. These animal models may serve to further dissect the lung-specific mechanisms of host susceptibility to virulent Mtb and for testing therapeutic interventions targeting these mechanisms.
Collapse
Affiliation(s)
- Shivraj M. Yabaji
- The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University
| | - Suruchi Lata
- The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University
| | - Anna E. Tseng
- The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University
| | | | - Ming Lo
- The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University
| | - Igor Gavrish
- The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University
| | - Aoife K O’Connell
- The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University
| | - Hans P Gertje
- The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University
| | - Anna C. Belkina
- The Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118
- Flow Cytometry Core Facility, Boston University School of Medicine, Boston, MA, USA
| | - Colleen E Thurman
- The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University
| | - Hirofumi Kiyokawa
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center and Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Darrell Kotton
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center and Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Shumin Tan
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA
| | - Janice J. Endsley
- Departments of Microbiology & Immunology and Pathology, The University of Texas Medical Branch, Galveston, TX 77555, USA
| | - William R Bishai
- Center for Tuberculosis Research School of Medicine, John Hopkins University Baltimore, Maryland
| | - Nicholas Crossland
- The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University
- The Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118
| | | | - Igor Kramnik
- The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University
- Pulmonary Center, The Department of Medicine, Boston University Chobanian & Aveedisian School of Medicine
| |
Collapse
|
12
|
Seo H, Kim BJ, Oh J, Jung S, Lee JY, Kim BJ. Protection against tuberculosis by vaccination of secreted chorismate mutase (Rv1885c) combined with a hepatitis B virus (HBV)-derived peptide, Poly6, and alum adjuvants. Vaccine 2025; 47:126710. [PMID: 39787795 DOI: 10.1016/j.vaccine.2025.126710] [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: 08/09/2024] [Revised: 12/10/2024] [Accepted: 01/03/2025] [Indexed: 01/12/2025]
Abstract
Tuberculosis (TB) remains a significant global health issue due to the limited efficacy of the Bacillus Calmette-Guérin (BCG) vaccine, highlighting the need for the development of an improved TB vaccine. In this study, we created a novel TB subunit vaccine consisting of TB-secreted chorismate mutase (TBCM) (Rv1885c) and a hepatitis B virus (HBV)-derived peptide (Poly6), which elicits Type I interferon responses, both with and without an alum adjuvant. We evaluated the immunogenicity, protective efficacy, and therapeutic efficacy of this vaccine candidate in an in vivo mouse model. Our results revealed that subcutaneous vaccination with TBCM combined with Poly6 induced stronger antigen-specific humoral and cell-mediated immune responses than TBCM alone or TBCM combined with alum adjuvant. Furthermore, adding the alum adjuvant to TBCM combined with Poly6 significantly enhanced antigen-specific immune responses. Subcutaneous vaccination with TBCM combined with both Poly6 and alum adjuvants effectively protected mice against Mycobacterium tuberculosis (Mtb) K strain infection, demonstrating its potential as a TB vaccine. Additionally, this vaccine platform exhibited therapeutic potential by significantly reducing bacterial loads and lung inflammation in mice previously infected with the virulent Mtb K strain. In conclusion, our findings suggest that TBCM is highly immunogenic in mice and that TBCM combined with both Poly6 and alum adjuvants represents a promising vaccine platform for TB prevention and treatment.
Collapse
Affiliation(s)
- Hyejun Seo
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea; Cancer Research Institute, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea; Seoul National University Medical Research Center (SNUMRC), Seoul 03080, Republic of Korea
| | - Byung-Jun Kim
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
| | - Jaehun Oh
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea; BK21 FOUR Biomedical Science Project, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Sangkwon Jung
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea; Cancer Research Institute, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
| | - Ju-Young Lee
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea; BK21 FOUR Biomedical Science Project, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Bum-Joon Kim
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea; Cancer Research Institute, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea; Seoul National University Medical Research Center (SNUMRC), Seoul 03080, Republic of Korea; Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea; Liver Research Institute, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea; BK21 FOUR Biomedical Science Project, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.
| |
Collapse
|
13
|
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
|
14
|
Sarkar S, Carranza C, Gonzalez Y, Zhang J(J, Osornio Vargas ÁR, Ohman-Strickland P, Torres M, Schwander S. Exposure to Urban Air Pollution Particulate Matter Modifies Th1/Th2 Mtb Immunity in the Human Lung. Am J Respir Cell Mol Biol 2025; 72:222-225. [PMID: 39887972 PMCID: PMC11976655 DOI: 10.1165/rcmb.2024-0240le] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2025] Open
Affiliation(s)
- Srijata Sarkar
- Rutgers University School of Public HealthPiscataway, New Jersey
| | - Claudia Carranza
- Instituto Nacional de Enfermedades RespiratoriasMéxico City, México
| | - Yolanda Gonzalez
- Instituto Nacional de Enfermedades RespiratoriasMéxico City, México
| | | | | | | | - Martha Torres
- Instituto Nacional de Enfermedades RespiratoriasMéxico City, México
| | | |
Collapse
|
15
|
Chang E, Cavallo K, Behar SM. CD4 T cell dysfunction is associated with bacterial recrudescence during chronic tuberculosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.22.634376. [PMID: 39896548 PMCID: PMC11785196 DOI: 10.1101/2025.01.22.634376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
While most people contain Mycobacterium tuberculosis infection, some individuals develop active disease, usually within two years of infection. Why immunity fails after initially controlling infection is unknown. C57BL/6 mice control Mycobacterium tuberculosis for up to a year but ultimately succumb to disease. We hypothesize that the development of CD4 T cell dysfunction permits bacterial recrudescence. We developed a reductionist model to assess antigen-specific T cells during chronic infection and found evidence of CD4 T cell senescence and exhaustion. In C57BL/6 mice, CD4 T cells upregulate coinhibitory receptors and lose effector cytokine production. Single cell RNAseq shows that only a small number of CD4 T cells in the lungs of chronically infected mice are polyfunctional. While the origin and causal relationship between T-cell dysfunction and recrudescence remains uncertain, we propose T cell dysfunction leads to a feed-forward loop that causes increased bacillary numbers, greater T cell dysfunction, and progressive disease.
Collapse
Affiliation(s)
- Evelyn Chang
- Immunology and Microbiology Program, Graduate School of Biomedical Science, Worcester, Massachusetts, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Kelly Cavallo
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Samuel M. Behar
- Immunology and Microbiology Program, Graduate School of Biomedical Science, Worcester, Massachusetts, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
16
|
La Distia Nora R, Putera I, Schrijver B, Singh G, Bakker M, Riasanti M, Edwar L, Susiyanti M, Aziza Y, Ten Berge JCEM, Rombach SM, van Hagen PM, Sitompul R, Dik WA. Ocular Tuberculosis Diagnosis Through Biomarkers: Clinical Relevance of Serum C1q and Whole Blood Interferon Gene Signature Score. Ocul Immunol Inflamm 2025; 33:113-124. [PMID: 38913993 DOI: 10.1080/09273948.2024.2368670] [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: 04/05/2024] [Revised: 06/05/2024] [Accepted: 06/11/2024] [Indexed: 06/26/2024]
Abstract
PURPOSE To assess the clinical relevance of pathophysiology-based biomarkers, specifically serum C1q and whole blood interferon gene signature score (IGSS), in ocular tuberculosis (OTB) diagnosis by conducting an integrative analysis of clinical presentations and treatment response. METHODS This retrospective cohort study analysed data from 70 patients with suspected OTB at a tertiary care uveitis practice in Indonesia. Serum C1q levels and whole blood IGSS were quantified. Patients were categorized into four quadrants based on their biomarker profiles: quadrant 1 (high C1q & low IGSS), quadrant 2 (high C1q & high IGSS), quadrant 3 (low C1q & high IGSS), and quadrant 4 (low C1q & low IGSS). Characteristics of clinical presentations, work-up results, and treatment outcomes were explored according to the predefined quadrants. RESULTS We identified that the majority of OTB patients diagnosed with concurrent active pulmonary TB were in quadrant 1, 2, or 3 (20/23, 87.0%). Twenty-seven patients (27/47, 57.4%) with clinically undifferentiated uveitis were in quadrant 4 (p < 0.001). Among patients in quadrants 1, 2, and 3, completion of a full course of antitubercular treatment (ATT) was associated with a lower number of patients showing persistence or recurrence of ocular inflammation compared to those who were not fully treated with ATT (14.3% vs 85.7%, p = 0.001). CONCLUSIONS Based on the analysis of clinical features and treatment outcomes, patients with elevated levels of either or both serum C1q and whole blood IGSS may reflect active TB disease in the eye, necessitating full ATT management.
Collapse
Affiliation(s)
- Rina La Distia Nora
- Department of Ophthalmology, Faculty of Medicine, Universitas Indonesia, Cipto Mangunkusumo Hospital, Jakarta, Indonesia
- Laboratory Medical Immunology, Department of Immunology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Ikhwanuliman Putera
- Department of Ophthalmology, Faculty of Medicine, Universitas Indonesia, Cipto Mangunkusumo Hospital, Jakarta, Indonesia
- Laboratory Medical Immunology, Department of Immunology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Ophthalmology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine Section Allergy & Clinical Immunology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Benjamin Schrijver
- Laboratory Medical Immunology, Department of Immunology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Gurmeet Singh
- Department of Internal Medicine, Respirology and Critical Illness Division, Faculty of Medicine, Universitas Indonesia, Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Marleen Bakker
- Department of Pulmonary Diseases, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Mei Riasanti
- Department of Ophthalmology, Faculty of Medicine, Universitas Indonesia, Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Lukman Edwar
- Department of Ophthalmology, Faculty of Medicine, Universitas Indonesia, Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Made Susiyanti
- Department of Ophthalmology, Faculty of Medicine, Universitas Indonesia, Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Yulia Aziza
- Department of Ophthalmology, Faculty of Medicine, Universitas Indonesia, Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | | | - Saskia M Rombach
- 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 University Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine Section Allergy & Clinical Immunology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Ratna Sitompul
- Department of Ophthalmology, Faculty of Medicine, Universitas Indonesia, Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Willem A Dik
- Laboratory Medical Immunology, Department of Immunology, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
17
|
Scriba TJ, Maseeme M, Young C, Taylor L, Leslie AJ. Immunopathology in human tuberculosis. Sci Immunol 2024; 9:eado5951. [PMID: 39671470 DOI: 10.1126/sciimmunol.ado5951] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 11/15/2024] [Indexed: 12/15/2024]
Abstract
Mycobacterium tuberculosis (M.tb) is a bacterial pathogen that has evolved in humans, and its interactions with the host are complex and best studied in humans. Myriad immune pathways are involved in infection control, granuloma formation, and progression to tuberculosis (TB) disease. Inflammatory cells, such as macrophages, neutrophils, conventional and unconventional T cells, B cells, NK cells, and innate lymphoid cells, interact via cytokines, cell-cell communication, and eicosanoid signaling to contain or eliminate infection but can alternatively mediate pathological changes required for pathogen transmission. Clinical manifestations include pulmonary and extrapulmonary TB, as well as post-TB lung disease. Risk factors for TB progression, in turn, largely relate to immune status and, apart from traditional chemotherapy, interventions primarily target immune mechanisms, highlighting the critical role of immunopathology in TB. Maintaining a balance between effector mechanisms to achieve protective immunity and avoid detrimental inflammation is central to the immunopathogenesis of TB. Many research gaps remain and deserve prioritization to improve our understanding of human TB immunopathogenesis.
Collapse
Affiliation(s)
- Thomas J Scriba
- South African Tuberculosis Vaccine Initiative, Division of Immunology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Mahlatse Maseeme
- Africa Health Research Institute, Durban, South Africa
- College of Heath Sciences, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Carly Young
- South African Tuberculosis Vaccine Initiative, Division of Immunology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Laura Taylor
- Forensic Pathology Services, Western Cape Government/University of Cape Town, Cape Town, South Africa
| | - Alasdair J Leslie
- Africa Health Research Institute, Durban, South Africa
- University College London, London, UK
| |
Collapse
|
18
|
Chowdhury CS, Kinsella RL, McNehlan ME, Naik SK, Lane DS, Talukdar P, Smirnov A, Dubey N, Rankin AN, McKee SR, Woodson R, Hii A, Chavez SM, Kreamalmeyer D, Beatty W, Mattila JT, Stallings CL. Type I IFN-mediated NET release promotes Mycobacterium tuberculosis replication and is associated with granuloma caseation. Cell Host Microbe 2024; 32:2092-2111.e7. [PMID: 39637864 PMCID: PMC11637906 DOI: 10.1016/j.chom.2024.11.008] [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: 12/13/2022] [Revised: 10/16/2024] [Accepted: 11/11/2024] [Indexed: 12/07/2024]
Abstract
Neutrophils are the most abundant cell type in the airways of tuberculosis patients. Mycobacterium tuberculosis (Mtb) infection induces the release of neutrophil extracellular traps (NETs); however, the molecular regulation and impact of NET release on Mtb pathogenesis are unknown. We find that during Mtb infection in neutrophils, PAD4 citrullinates histones to decondense chromatin that gets released as NETs in a manner that can maintain neutrophil viability and promote Mtb replication. Type I interferon promotes the formation of chromatin-containing vesicles that allow NET release without compromising plasma membrane integrity. Analysis of nonhuman primate granulomas supports a model where neutrophils are exposed to type I interferon from macrophages as they migrate into the granuloma, thereby enabling the release of NETs associated with necrosis and caseation. Our data reveal NET release as a promising target to inhibit Mtb pathogenesis.
Collapse
Affiliation(s)
- Chanchal Sur Chowdhury
- 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
| | - Michael E McNehlan
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sumanta K Naik
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Daniel S Lane
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, PA 15261, USA
| | - Priyanka Talukdar
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, PA 15261, USA
| | - Asya Smirnov
- 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
| | - Ananda N Rankin
- 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
| | - Reilly Woodson
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Abigail Hii
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA; UCD School of Medicine, University College Dublin, Belfield, Dublin, Ireland
| | - Sthefany M Chavez
- 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
| | - Wandy Beatty
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Joshua T Mattila
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, PA 15261, 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
|
19
|
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
|
20
|
Shuster M, Lyu Z, Augenstreich J, Mathur S, Ganesh A, Ling J, Briken V. Salmonella Typhimurium infection inhibits macrophage IFNβ signaling in a TLR4-dependent manner. Infect Immun 2024; 92:e0009824. [PMID: 39269166 PMCID: PMC11475681 DOI: 10.1128/iai.00098-24] [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: 03/01/2024] [Accepted: 08/16/2024] [Indexed: 09/15/2024] Open
Abstract
Type I Interferons (IFNs) generally have a protective role during viral infections, but their function during bacterial infections is dependent on the bacterial species. Legionella pneumophila, Shigella sonnei and Mycobacterium tuberculosis can inhibit type I IFN signaling. Here we examined the role of type I IFN, specifically IFNβ, in the context of Salmonella enterica serovar Typhimurium (STm) macrophage infections and the capacity of STm to inhibit type I IFN signaling. We demonstrate that IFNβ has no effect on the intracellular growth of STm in infected bone marrow derived macrophages (BMDMs) derived from C57BL/6 mice. STm infection inhibits IFNβ signaling but not IFNγ signaling in a murine macrophage cell line. We show that this inhibition is independent of the type III and type VI secretion systems expressed by STm and is also independent of bacterial phagocytosis. The inhibition is Toll-like receptor 4 (TLR4)-dependent as the TLR4 ligand, lipopolysaccharide (LPS), alone is sufficient to inhibit IFNβ-mediated signaling. Cells downregulated their surface levels of IFNα/β receptor 1 (IFNAR1) in response to LPS, which may be mediating our observed inhibition. Lastly, we examined this inhibition in the context of TLR4-deficient BMDMs as well as TLR4 RNA interference and we observed a loss of inhibition with LPS stimulation as well as STm infection. In summary, we show that macrophages exposed to STm have reduced IFNβ signaling via crosstalk with TLR4 signaling, which may be mediated by reduced host cell surface IFNAR1, and that IFNβ signaling does not affect cell-autonomous host defense against STm.
Collapse
Affiliation(s)
- Michael Shuster
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA
| | - Zhihui Lyu
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA
| | - Jacques Augenstreich
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA
| | - Shrestha Mathur
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA
| | - Akshaya Ganesh
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA
| | - Jiqiang Ling
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA
| | - Volker Briken
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA
| |
Collapse
|
21
|
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
|
22
|
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
|
23
|
Theobald SJ, Müller TA, Lange D, Keck K, Rybniker J. The role of inflammasomes as central inflammatory hubs in Mycobacterium tuberculosis infection. Front Immunol 2024; 15:1436676. [PMID: 39324136 PMCID: PMC11422116 DOI: 10.3389/fimmu.2024.1436676] [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: 05/22/2024] [Accepted: 08/26/2024] [Indexed: 09/27/2024] Open
Abstract
Mycobacterium tuberculosis (Mtb) infection represents a global health problem and is characterized by formation of granuloma with a necrotic center and a systemic inflammatory response. Inflammasomes have a crucial role in the host immune response towards Mtb. These intracellular multi-protein complexes are assembled in response to pathogen-associated molecular patterns (PAMPs) or danger-associated molecular patterns (DAMPs). Inflammasome platforms activate caspases, leading to the maturation of the proinflammatory cytokines interleukin (IL)-1 and 18 and the cleavage of gasdermin D (GSDMD), a pore-forming protein responsible for cytokine release and pyroptotic cell death. Recent in vitro and in vivo findings have highlighted the importance of inflammasome signaling and subsequent necrotic cell death in Mtb-infected innate immune cells. However, we are just beginning to understand how inflammasomes contribute to disease or to a protective immune response in tuberculosis (TB). A detailed molecular understanding of inflammasome-associated pathomechanisms may foster the development of novel host-directed therapeutics or vaccines with improved activity. In this mini-review, we discuss the regulatory and molecular aspects of inflammasome activation and the associated immunological consequences for Mtb pathogenesis.
Collapse
Affiliation(s)
- Sebastian J. Theobald
- Department I of Internal Medicine, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| | - Tony A. Müller
- Department I of Internal Medicine, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Dinah Lange
- Department I of Internal Medicine, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| | - Katharina Keck
- Department I of Internal Medicine, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Jan Rybniker
- Department I of Internal Medicine, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| |
Collapse
|
24
|
Nobs E, Laschanzky K, Munke K, Movert E, Valfridsson C, Carlsson F. Cytosolic serpins act in a cytoprotective feedback loop that limits ESX-1-dependent death of Mycobacterium marinum-infected macrophages. mBio 2024; 15:e0038424. [PMID: 39087767 PMCID: PMC11389378 DOI: 10.1128/mbio.00384-24] [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: 02/06/2024] [Accepted: 05/28/2024] [Indexed: 08/02/2024] Open
Abstract
Serine protease inhibitors (serpins) constitute the largest family of protease inhibitors expressed in humans, but their role in infection remains largely unexplored. In infected macrophages, the mycobacterial ESX-1 type VII secretion system permeabilizes internal host membranes and causes leakage into the cytosol of host DNA, which induces type I interferon (IFN) production via the cyclic GMP-AMP synthase (cGAS) and stimulator of IFN genes (STING) surveillance pathway, and promotes infection in vivo. Using the Mycobacterium marinum infection model, we show that ESX-1-mediated type I IFN signaling in macrophages selectively induces the expression of serpina3f and serpina3g, two cytosolic serpins of the clade A3. The membranolytic activity of ESX-1 also caused leakage of cathepsin B into the cytosol where it promoted cell death, suggesting that the induction of type I IFN comes at the cost of lysosomal rupture and toxicity. However, the production of cytosolic serpins suppressed the protease activity of cathepsin B in this compartment and thus limited cell death, a function that was associated with increased bacterial growth in infected mice. These results suggest that cytosolic serpins act in a type I IFN-dependent cytoprotective feedback loop to counteract the inevitable toxic effect of ESX-1-mediated host membrane rupture. IMPORTANCE The ESX-1 type VII secretion system is a key virulence determinant of pathogenic mycobacteria. The ability to permeabilize host cell membranes is critical for several ESX-1-dependent virulence traits, including phagosomal escape and induction of the type I interferon (IFN) response. We find that it comes at the cost of lysosomal leakage and subsequent host cell death. However, our results suggest that ESX-1-mediated type I IFN signaling selectively upregulates serpina3f and serpina3g and that these cytosolic serpins limit cell death caused by cathepsin B that has leaked into the cytosol, a function that is associated with increased bacterial growth in vivo. The ability to rupture host membranes is widespread among bacterial pathogens, and it will be of interest to evaluate the role of cytosolic serpins and this type I IFN-dependent cytoprotective feedback loop in the context of human infection.
Collapse
Affiliation(s)
- Esther Nobs
- Department of Biology, Lund University, Lund, Sweden
| | | | - Kristina Munke
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Elin Movert
- Department of Biology, Lund University, Lund, Sweden
| | | | | |
Collapse
|
25
|
Krivošová M, Dohál M, Mäsiarová S, Pršo K, Gondáš E, Murín R, Fraňová S, Porvazník I, Solovič I, Mokrý J. Exploring cytokine dynamics in tuberculosis: A comparative analysis of patients and controls with insights from three-week antituberculosis intervention. PLoS One 2024; 19:e0305158. [PMID: 39208230 PMCID: PMC11361567 DOI: 10.1371/journal.pone.0305158] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/26/2024] [Indexed: 09/04/2024] Open
Abstract
Despite developing new diagnostics, drugs, and vaccines, treating tuberculosis (TB) remains challenging. Monitoring inflammatory markers can contribute to more precise diagnostics of TB, identifying its active and latent forms, or monitoring its treatment success. We assessed alterations in plasma levels of 48 cytokines in 20 patients (17 males) with active pulmonary TB compared to age-matched healthy controls (n = 18). Blood samples were collected from individuals hospitalised with TB prior to commencing antibiotic therapy, after the first week, and following the third week. The majority of patients received treatment with a combination of four first-line antituberculosis drugs: rifampicin, isoniazid, ethambutol, and pyrazinamide. Plasmatic cytokine levels from patients three times and controls were analyzed using a Bio-Plex Pro Human Cytokine Screening Panel. The results showed significantly higher levels of 31 cytokines (p<0.05) than healthy controls. Three-week therapy duration showed significantly decreased levels of nine cytokines: interferon alpha-2 (IFN-α2), interleukin (IL) 1 alpha (IL-1α), IL-1 receptor antagonist (IL-1ra), IL-6, IL-10, IL-12 p40, IL-17, leukemia inhibitory factor (LIF), and tumor necrosis factor alpha (TNF-α). Out of these, only levels of IL-1α and IL-6 remained significantly elevated compared to controls. Moreover, we have found a negative correlation of 18 cytokine levels with BMI of the patients but no correlation with age. Our results showed a clinical potential for monitoring the levels of specific inflammatory markers after a short treatment duration. The reduction in cytokine levels throughout the course of therapy could indicate treatment success but should be confirmed in studies with more individuals involved and a longer observation period.
Collapse
Affiliation(s)
- Michaela Krivošová
- Jessenius Faculty of Medicine in Martin, Biomedical Centre Martin, Comenius University Bratislava, Martin, Slovak Republic
| | - Matúš Dohál
- Jessenius Faculty of Medicine in Martin, Biomedical Centre Martin, Comenius University Bratislava, Martin, Slovak Republic
| | - Simona Mäsiarová
- Jessenius Faculty of Medicine in Martin, Department of Pharmacology, Comenius University Bratislava, Martin, Slovak Republic
| | - Kristián Pršo
- Jessenius Faculty of Medicine in Martin, Department of Pharmacology, Comenius University Bratislava, Martin, Slovak Republic
| | - Eduard Gondáš
- Jessenius Faculty of Medicine in Martin, Department of Pharmacology, Comenius University Bratislava, Martin, Slovak Republic
| | - Radovan Murín
- Jessenius Faculty of Medicine in Martin, Department of Medical Biochemistry, Comenius University Bratislava, Martin, Slovakia
| | - Soňa Fraňová
- Jessenius Faculty of Medicine in Martin, Department of Pharmacology, Comenius University Bratislava, Martin, Slovak Republic
| | - Igor Porvazník
- National Institute for Tuberculosis, Lung Diseases and Thoracic Surgery, Vyšné Hágy, Slovak Republic
- Faculty of Health, Catholic University, Ružomberok, Slovak Republic
| | - Ivan Solovič
- National Institute for Tuberculosis, Lung Diseases and Thoracic Surgery, Vyšné Hágy, Slovak Republic
- Faculty of Health, Catholic University, Ružomberok, Slovak Republic
| | - Juraj Mokrý
- Jessenius Faculty of Medicine in Martin, Department of Pharmacology, Comenius University Bratislava, Martin, Slovak Republic
| |
Collapse
|
26
|
Sharan R, Zou Y, Lai Z, Singh B, Shivanna V, Dick E, Hall-Ursone S, Khader S, Mehra S, Alvarez X, Rengarajan J, Kaushal D. Concurrent TB and HIV therapies effectively control clinical reactivation of TB during co-infection but fail to eliminate chronic immune activation. RESEARCH SQUARE 2024:rs.3.rs-4908400. [PMID: 39257997 PMCID: PMC11384027 DOI: 10.21203/rs.3.rs-4908400/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
The majority of Human Immunodeficiency Virus (HIV) negative individuals exposed to Mycobacterium tuberculosis (Mtb) control the bacillary infection as latent TB infection (LTBI). Co-infection with HIV, however, drastically increases the risk to progression to tuberculosis (TB) disease. TB is therefore the leading cause of death in people living with HIV (PLWH) globally. Combinatorial antiretroviral therapy (cART) is the cornerstone of HIV care in humans and reduces the risk of reactivation of LTBI. However, the immune control of Mtb infection is not fully restored by cART as indicated by higher incidence of TB in PLWH despite cART. In the macaque model of co-infection, skewed pulmonary CD4+ TEM responses persist, and new TB lesions form despite cART treatment. We hypothesized that regimens that concurrently administer anti-TB therapy and cART would significantly reduce TB in co-infected macaques than cART alone, resulting in superior bacterial control, mitigation of persistent inflammation and lasting protective immunity. We studied components of TB immunity that remain impaired after cART in the lung compartment, versus those that are restored by concurrent 3 months of once weekly isoniazid and rifapentine (3HP) and cART in the rhesus macaque (RM) model of LTBI and Simian Immunodeficiency Virus (SIV) co-infection. Concurrent administration of cART + 3HP did improve clinical and microbiological attributes of Mtb/SIV co-infection compared to cART-naïve or -untreated RMs. While RMs in the cART + 3HP group exhibited significantly lower granuloma volumes after treatment, they, however, continued to harbor caseous granulomas with increased FDG uptake. cART only partially restores the constitution of CD4 + T cells to the lung compartment in co-infected macaques. Concurrent therapy did not further enhance the frequency of reconstituted CD4+ T cells in BAL and lung of Mtb/SIV co-infected RMs compared to cART, and treated animals continued to display incomplete reconstitution to the lung. Furthermore, the reconstituted CD4+ T cells in BAL and lung of cART + 3HP treated RMs exhibited an increased frequencies of activated, exhausted and inflamed phenotype compared to LTBI RMs. cART + 3HP failed to restore the effector memory CD4+ T cell population that was significantly reduced in pulmonary compartment post SIV co-infection. Concurrent therapy was associated with the induction of Type I IFN transcriptional signatures and led to increased Mtb-specific TH1/TH17 responses correlated with protection, but decreased Mtb-specific TNFa responses, which could have a detrimental impact on long term protection. Our results suggest the mechanisms by which Mtb/HIV co-infected individuals remain at risk for progression due to subsequent infections or reactivation due of persisting defects in pulmonary T cell responses. By identifying lung-specific immune components in this model, it is possible to pinpoint the pathways that can be targeted for host-directed adjunctive therapies for TB/HIV co-infection.
Collapse
Affiliation(s)
| | | | - Zhao Lai
- The University of Texas Health San Antonio
| | | | | | | | | | | | | | | | | | - Deepak Kaushal
- Southwest National Primate Research Center, Texas Biomedical Research Institute
| |
Collapse
|
27
|
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
|
28
|
Meintjes G, Maartens G. HIV-Associated Tuberculosis. N Engl J Med 2024; 391:343-355. [PMID: 39047241 DOI: 10.1056/nejmra2308181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Affiliation(s)
- Graeme Meintjes
- From the Department of Medicine, University of Cape Town and Groote Schuur Hospital (G. Meintjes), and the Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine (G. Meintjes, G. Maartens), and the Division of Clinical Pharmacology, Department of Medicine (G. Maartens), University of Cape Town - all in Cape Town, South Africa; and Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom (G. Meintjes)
| | - Gary Maartens
- From the Department of Medicine, University of Cape Town and Groote Schuur Hospital (G. Meintjes), and the Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine (G. Meintjes, G. Maartens), and the Division of Clinical Pharmacology, Department of Medicine (G. Maartens), University of Cape Town - all in Cape Town, South Africa; and Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom (G. Meintjes)
| |
Collapse
|
29
|
Campo M, Dill-McFarland KA, Peterson GJ, Benson B, Skerrett SJ, Hawn TR. Human Alveolar and Monocyte-Derived Human Macrophage Responses to Mycobacterium tuberculosis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:161-169. [PMID: 38836816 PMCID: PMC11610518 DOI: 10.4049/jimmunol.2300885] [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: 12/20/2023] [Accepted: 04/29/2024] [Indexed: 06/06/2024]
Abstract
Alveolar macrophages (AMs) and recruited monocyte-derived macrophages (MDMs) mediate early lung immune responses to Mycobacterium tuberculosis. Differences in the response of these distinct cell types are poorly understood and may provide insight into mechanisms of tuberculosis pathogenesis. The objective of this study was to determine whether M. tuberculosis induces unique and essential antimicrobial pathways in human AMs compared with MDMs. Using paired human AMs and 5-d MCSF-derived MDMs from six healthy volunteers, we infected cells with M. tuberculosis H37Rv for 6 h, isolated RNA, and analyzed transcriptomic profiles with RNA sequencing. We found 681 genes that were M. tuberculosis dependent in AMs compared with MDMs and 4538 that were M. tuberculosis dependent in MDMs, but not AMs (false discovery rate [FDR] < 0.05). Using hypergeometric enrichment of DEGs in Broad Hallmark gene sets, we found that type I and II IFN Response were the only gene sets selectively induced in M. tuberculosis-infected AM (FDR < 0.05). In contrast, MYC targets, unfolded protein response and MTORC1 signaling, were selectively enriched in MDMs (FDR < 0.05). IFNA1, IFNA8, IFNE, and IFNL1 were specifically and highly upregulated in AMs compared with MDMs at baseline and/or after M. tuberculosis infection. IFNA8 modulated M. tuberculosis-induced proinflammatory cytokines and, compared with other IFNs, stimulated unique transcriptomes. Several DNA sensors and IFN regulatory factors had higher expression at baseline and/or after M. tuberculosis infection in AMs compared with MDMs. These findings demonstrate that M. tuberculosis infection induced unique transcriptional responses in human AMs compared with MDMs, including upregulation of the IFN response pathway and specific DNA sensors.
Collapse
Affiliation(s)
- Monica Campo
- Department of Medicine, University of Minnesota, Minneapolis, MN
| | | | | | - Basilin Benson
- Systems Immunology Program, Benaroya Research Institute, Seattle, WA
| | | | - Thomas R. Hawn
- Department of Medicine, University of Washington, Seattle, WA
| |
Collapse
|
30
|
Anes E, Azevedo-Pereira JM, Pires D. Role of Type I Interferons during Mycobacterium tuberculosis and HIV Infections. Biomolecules 2024; 14:848. [PMID: 39062562 PMCID: PMC11275242 DOI: 10.3390/biom14070848] [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: 06/18/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Tuberculosis and AIDS remain two of the most relevant human infectious diseases. The pathogens that cause them, Mycobacterium tuberculosis (Mtb) and HIV, individually elicit an immune response that treads the line between beneficial and detrimental to the host. Co-infection further complexifies this response since the different cytokines acting on one infection might facilitate the dissemination of the other. In these responses, the role of type I interferons is often associated with antiviral mechanisms, while for bacteria such as Mtb, their importance and clinical relevance as a suitable target for manipulation are more controversial. In this article, we review the recent knowledge on how these interferons play distinct roles and sometimes have opposite consequences depending on the stage of the pathogenesis. We highlight the dichotomy between the acute and chronic infections displayed by both infections and how type I interferons contribute to an initial control of each infection individually, while their chronic induction, particularly during HIV infection, might facilitate Mtb primo-infection and progression to disease. We expect that further findings and their systematization will allow the definition of windows of opportunity for interferon manipulation according to the stage of infection, contributing to pathogen clearance and control of immunopathology.
Collapse
Affiliation(s)
- Elsa Anes
- Host-Pathogen Interactions Unit, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (J.M.A.-P.); (D.P.)
| | - José Miguel Azevedo-Pereira
- Host-Pathogen Interactions Unit, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (J.M.A.-P.); (D.P.)
| | - David Pires
- Host-Pathogen Interactions Unit, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (J.M.A.-P.); (D.P.)
- Center for Interdisciplinary Research in Health, Católica Medical School, Universidade Católica Portuguesa, Estrada Octávio Pato, 2635-631 Rio de Mouro, Portugal
| |
Collapse
|
31
|
Crow YJ, Casanova JL. Human life within a narrow range: The lethal ups and downs of type I interferons. Sci Immunol 2024; 9:eadm8185. [PMID: 38968338 DOI: 10.1126/sciimmunol.adm8185] [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: 11/07/2023] [Accepted: 06/13/2024] [Indexed: 07/07/2024]
Abstract
The past 20 years have seen the definition of human monogenic disorders and their autoimmune phenocopies underlying either defective or enhanced type I interferon (IFN) activity. These disorders delineate the impact of type I IFNs in natural conditions and demonstrate that only a narrow window of type I IFN activity is beneficial. Insufficient type I IFN predisposes humans to life-threatening viral diseases (albeit unexpectedly few) with a central role in immunity to respiratory and cerebral viral infection. Excessive type I IFN, perhaps counterintuitively, appears to underlie a greater number of autoinflammatory and/or autoimmune conditions known as type I interferonopathies, whose study has revealed multiple molecular programs involved in the induction of type I IFN signaling. These observations suggest that the manipulation of type I IFN activity to within a physiological range may be clinically relevant for the prevention and treatment of viral and inflammatory diseases.
Collapse
Affiliation(s)
- Yanick J Crow
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- Laboratory of Neurogenetics and Neuroinflammation, Imagine Institute, INSERM UMR 1163, Paris, France
- University Paris Cité, Paris, France
| | - Jean-Laurent Casanova
- University Paris Cité, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Howard Hughes Medical Institute, New York, NY, USA
- Department of Pediatrics, Necker Hospital for Sick Children, Paris, France
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Imagine Institute, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France
| |
Collapse
|
32
|
Bernacchia A, Garcia AL, Raccio AG, Di Giovanni D. Disseminated BCG Disease in a Child with a Novel PSMG2 Deletion. J Clin Immunol 2024; 44:145. [PMID: 38847935 DOI: 10.1007/s10875-024-01738-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 05/16/2024] [Indexed: 06/29/2024]
Affiliation(s)
- Agustín Bernacchia
- Sección Inmunología, Hospital de Niños "Ricardo Gutiérrez", Ciudad de Buenos Aires, Argentina.
| | - Ana Luz Garcia
- Sección Inmunología, Hospital de Niños "Ricardo Gutiérrez", Ciudad de Buenos Aires, Argentina
| | - Andrea Gomez Raccio
- Sección Inmunología, Hospital de Niños "Ricardo Gutiérrez", Ciudad de Buenos Aires, Argentina
| | - Daniela Di Giovanni
- Sección Inmunología, Hospital de Niños "Ricardo Gutiérrez", Ciudad de Buenos Aires, Argentina
| |
Collapse
|
33
|
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: 12] [Impact Index Per Article: 12.0] [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
|
34
|
Lu Y, Li S, Su Z, Luo C, Gu M, Yuan D, Qin BE, Dai K, Xia H, Chen Y, Peng F, Jiang Y. Presence of Epstein-Barr virus in cerebrospinal fluid is associated with increased mortality in HIV-negative cryptococcal meningitis. Med Mycol 2024; 62:myae052. [PMID: 38710585 DOI: 10.1093/mmy/myae052] [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: 12/15/2023] [Revised: 04/15/2024] [Accepted: 05/05/2024] [Indexed: 05/08/2024] Open
Abstract
Cryptococcus neoformans is the most common cause of fungal meningitis and is associated with a high mortality. The clinical significance of concurrent Epstein-Barr virus (EBV) in the cerebrospinal fluid (CSF) of human immunodeficiency virus (HIV)-negative patients with cryptococcal meningitis (CM) remains unclear. A retrospective cohort study was performed by analyzing CSF samples from 79 HIV-negative Chinese Han patients with confirmed CM. We identified CSF viral DNA in these patients by metagenomic next-generation sequencing (mNGS) and compared 10-week survival rates among those with and without EBV DNA in CSF. Of the 79 CSF samples tested, 44.3% (35/79) had detectable viral DNA in CSF, while 55.7% (44/79) were virus-negative. The most frequent viral pathogen was EBV, which was detected in 22.8% (18/79) patients. The median number of CSF-EBV DNA reads was 4 reads with a range from 1 to 149 reads. The 10-week mortality rates were 22.2% (4/18) in those with positive CSF-EBV and 2.3% (1/44) in those with negative CSF-virus (hazard ratio 8.20, 95% confidence interval [CI] 1.52-81.80; P = 0.014), which remained significant after a multivariate adjustment for the known risk factors of mortality (adjusted hazard ratio 8.15, 95% CI 1.14-92.87; P = 0.037). mNGS can identify viruses that coexist in CSF of HIV-negative patients with CM. EBV DNA is most commonly found together with C. neoformans in CSF and its presence is associated with increased mortality in HIV-negative CM patients.
Collapse
Affiliation(s)
- Yi Lu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, PR China
- Department of Neurology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, PR China
| | - Shubo Li
- Department of Statistics, The Pennsylvania State University, State College, Pennsylvania, USA
| | - Zhihui Su
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, PR China
| | - Chongliang Luo
- Division of Public Health Sciences, Washington University School of Medicine in St. Louis, St Louis, Missouri, USA
| | - Meifeng Gu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, PR China
| | - Dasen Yuan
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, PR China
| | - Bang-E Qin
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, PR China
| | - Kai Dai
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, PR China
| | - Han Xia
- Department of Scientific Affairs, Hugobiotech Co, Beijing, PR China
| | - Yong Chen
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Fuhua Peng
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, PR China
| | - Ying Jiang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, PR China
| |
Collapse
|
35
|
Painter H, Willcocks S, Zelmer A, Reljic R, Tanner R, Fletcher H. Demonstrating the utility of the ex vivo murine mycobacterial growth inhibition assay (MGIA) for high-throughput screening of tuberculosis vaccine candidates against multiple Mycobacterium tuberculosis complex strains. Tuberculosis (Edinb) 2024; 146:102494. [PMID: 38367368 DOI: 10.1016/j.tube.2024.102494] [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: 12/06/2023] [Revised: 02/05/2024] [Accepted: 02/11/2024] [Indexed: 02/19/2024]
Abstract
Human tuberculosis (TB) is caused by various members of the Mycobacterium tuberculosis (Mtb) complex. Differences in host response to infection have been reported, illustrative of a need to evaluate efficacy of novel vaccine candidates against multiple strains in preclinical studies. We previously showed that the murine lung and spleen direct mycobacterial growth inhibition assay (MGIA) can be used to assess control of ex vivo mycobacterial growth by host cells. The number of mice required for the assay is significantly lower than in vivo studies, facilitating testing of multiple strains and/or the incorporation of other cellular analyses. Here, we provide proof-of-concept that the murine MGIA can be applied to evaluate vaccine-induced protection against multiple Mtb clinical isolates. Using an ancient and modern strain of the Mtb complex, we demonstrate that ex vivo bacillus Calmette-Guérin (BCG)-mediated mycobacterial growth inhibition recapitulates protection observed in the lung and spleen following in vivo infection of mice. Further, we provide the first report of cellular and transcriptional correlates of BCG-induced growth inhibition in the lung MGIA. The ex vivo MGIA represents a promising platform to gain early insight into vaccine performance against a collection of Mtb strains and improve preclinical evaluation of TB vaccine candidates.
Collapse
Affiliation(s)
- Hannah Painter
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK.
| | - Sam Willcocks
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - Andrea Zelmer
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - Rajko Reljic
- Institute of Infection and Immunity, St George's University of London, Cranmer Terrrace, London, SW17 0RE, UK
| | - Rachel Tanner
- Department of Biology, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Helen Fletcher
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| |
Collapse
|
36
|
Shaku MT, Um PK, Ocius KL, Apostolos AJ, Pires MM, Bishai WR, Kana BD. A modified BCG with depletion of enzymes associated with peptidoglycan amidation induces enhanced protection against tuberculosis in mice. eLife 2024; 13:e89157. [PMID: 38639995 PMCID: PMC11132681 DOI: 10.7554/elife.89157] [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: 04/17/2024] [Indexed: 04/20/2024] Open
Abstract
Mechanisms by which Mycobacterium tuberculosis (Mtb) evades pathogen recognition receptor activation during infection may offer insights for the development of improved tuberculosis (TB) vaccines. Whilst Mtb elicits NOD-2 activation through host recognition of its peptidoglycan-derived muramyl dipeptide (MDP), it masks the endogenous NOD-1 ligand through amidation of glutamate at the second position in peptidoglycan side-chains. As the current BCG vaccine is derived from pathogenic mycobacteria, a similar situation prevails. To alleviate this masking ability and to potentially improve efficacy of the BCG vaccine, we used CRISPRi to inhibit expression of the essential enzyme pair, MurT-GatD, implicated in amidation of peptidoglycan side-chains. We demonstrate that depletion of these enzymes results in reduced growth, cell wall defects, increased susceptibility to antibiotics, altered spatial localization of new peptidoglycan and increased NOD-1 expression in macrophages. In cell culture experiments, training of a human monocyte cell line with this recombinant BCG yielded improved control of Mtb growth. In the murine model of TB infection, we demonstrate that depletion of MurT-GatD in BCG, which is expected to unmask the D-glutamate diaminopimelate (iE-DAP) NOD-1 ligand, yields superior prevention of TB disease compared to the standard BCG vaccine. In vitro and in vivo experiments in this study demonstrate the feasibility of gene regulation platforms such as CRISPRi to alter antigen presentation in BCG in a bespoke manner that tunes immunity towards more effective protection against TB disease.
Collapse
Affiliation(s)
- Moagi Tube Shaku
- DST/NRF Centre of Excellence for Biomedical TB Research, Faculty of Health Sciences, University of the Witwatersrand, National Health Laboratory ServiceJohannesburgSouth Africa
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins School of MedicineBaltimoreUnited States
| | - Peter K Um
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins School of MedicineBaltimoreUnited States
| | - Karl L Ocius
- Department of Chemistry, University of VirginiaCharlottesvilleUnited States
| | - Alexis J Apostolos
- Department of Chemistry, University of VirginiaCharlottesvilleUnited States
| | - Marcos M Pires
- Department of Chemistry, University of VirginiaCharlottesvilleUnited States
| | - William R Bishai
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins School of MedicineBaltimoreUnited States
| | - Bavesh D Kana
- DST/NRF Centre of Excellence for Biomedical TB Research, Faculty of Health Sciences, University of the Witwatersrand, National Health Laboratory ServiceJohannesburgSouth Africa
| |
Collapse
|
37
|
Srikrishna G, Bullen CK, Bishai WR. Mycobacterial Nucleic Acids Modulate Host Innate Immune Responses. JOURNAL OF INFECTIOUS DISEASE AND THERAPY 2024; 12:1000585. [PMID: 38745994 PMCID: PMC11091829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Affiliation(s)
- Geetha Srikrishna
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, USA
| | - C Korin Bullen
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, USA
| | - William R Bishai
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, USA
| |
Collapse
|
38
|
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
|
39
|
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
|
40
|
Källenius G, Correia-Neves M, Sundling C. Diagnostic markers reflecting dysregulation of the host response in the transition to tuberculosis disease. Int J Infect Dis 2024; 141S:106984. [PMID: 38417614 DOI: 10.1016/j.ijid.2024.106984] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 03/01/2024] Open
Abstract
Sustained control of Mycobacterium tuberculosis infection without evidence of disease is based on a finely tuned balance between pro- and anti-inflammatory responses. Loss of this balance leads to tuberculosis (TB) disease, in which exacerbated myeloid and neutrophil activation is common. Proteomic and transcriptomic assessment of the host response can detect increasing immune activation associated with TB disease progression several months before clinical disease. Future diagnostic methods based on measuring host response biomarkers that are able to detect this dysregulation could therefore be valuable in the early detection of TB disease progression.
Collapse
Affiliation(s)
- Gunilla Källenius
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden; Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Margarida Correia-Neves
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden; Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's, PT Government Associate Laboratory, Braga, Portugal
| | - Christopher Sundling
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden; Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
41
|
Maure A, Lawarée E, Fiorentino F, Pawlik A, Gona S, Giraud-Gatineau A, Eldridge MJG, Danckaert A, Hardy D, Frigui W, Keck C, Gutierrez C, Neyrolles O, Aulner N, Mai A, Hamon M, Barreiro LB, Brodin P, Brosch R, Rotili D, Tailleux L. A host-directed oxadiazole compound potentiates antituberculosis treatment via zinc poisoning in human macrophages and in a mouse model of infection. PLoS Biol 2024; 22:e3002259. [PMID: 38683873 PMCID: PMC11081512 DOI: 10.1371/journal.pbio.3002259] [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: 07/05/2023] [Revised: 05/09/2024] [Accepted: 03/13/2024] [Indexed: 05/02/2024] Open
Abstract
Antituberculosis drugs, mostly developed over 60 years ago, combined with a poorly effective vaccine, have failed to eradicate tuberculosis. More worryingly, multiresistant strains of Mycobacterium tuberculosis (MTB) are constantly emerging. Innovative strategies are thus urgently needed to improve tuberculosis treatment. Recently, host-directed therapy has emerged as a promising strategy to be used in adjunct with existing or future antibiotics, by improving innate immunity or limiting immunopathology. Here, using high-content imaging, we identified novel 1,2,4-oxadiazole-based compounds, which allow human macrophages to control MTB replication. Genome-wide gene expression analysis revealed that these molecules induced zinc remobilization inside cells, resulting in bacterial zinc intoxication. More importantly, we also demonstrated that, upon treatment with these novel compounds, MTB became even more sensitive to antituberculosis drugs, in vitro and in vivo, in a mouse model of tuberculosis. Manipulation of heavy metal homeostasis holds thus great promise to be exploited to develop host-directed therapeutic interventions.
Collapse
Affiliation(s)
- Alexandra Maure
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Unit for Integrated Mycobacterial Pathogenomics, Paris, France
| | - Emeline Lawarée
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Unit for Integrated Mycobacterial Pathogenomics, Paris, France
| | - Francesco Fiorentino
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Rome, Italy
| | - Alexandre Pawlik
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Unit for Integrated Mycobacterial Pathogenomics, Paris, France
| | - Saideep Gona
- Department of Genetic Medicine, University of Chicago, Chicago, Illinois, United States of America
| | | | | | - Anne Danckaert
- Institut Pasteur, Université Paris Cité, UTechS BioImaging-C2RT, Paris, France
| | - David Hardy
- Institut Pasteur, Université Paris Cité, Histopathology Platform, Paris, France
| | - Wafa Frigui
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Unit for Integrated Mycobacterial Pathogenomics, Paris, France
| | - Camille Keck
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Unit for Integrated Mycobacterial Pathogenomics, Paris, France
| | - Claude Gutierrez
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Olivier Neyrolles
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Nathalie Aulner
- Institut Pasteur, Université Paris Cité, UTechS BioImaging-C2RT, Paris, France
| | - Antonello Mai
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Rome, Italy
- Pasteur Institute, Cenci-bolognetti Foundation, Sapienza University of Rome, Rome, Italy
| | - Mélanie Hamon
- Institut Pasteur, Université Paris Cité, Chromatine et Infection unit, Paris, France
| | - Luis B. Barreiro
- Department of Genetic Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Priscille Brodin
- Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Roland Brosch
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Unit for Integrated Mycobacterial Pathogenomics, Paris, France
| | - Dante Rotili
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Rome, Italy
| | - Ludovic Tailleux
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Unit for Integrated Mycobacterial Pathogenomics, Paris, France
| |
Collapse
|
42
|
Nore KG, Louet C, Bugge M, Gidon A, Jørgensen MJ, Jenum S, Dyrhol-Riise AM, Tonby K, Flo TH. The Cyclooxygenase 2 Inhibitor Etoricoxib as Adjunctive Therapy in Tuberculosis Impairs Macrophage Control of Mycobacterial Growth. J Infect Dis 2024; 229:888-897. [PMID: 37721470 PMCID: PMC10938220 DOI: 10.1093/infdis/jiad390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 08/26/2023] [Accepted: 09/14/2023] [Indexed: 09/19/2023] Open
Abstract
BACKGROUND Current tuberculosis treatment regimens could be improved by adjunct host-directed therapies (HDT) targeting host responses. We investigated the antimycobacterial capacity of macrophages from patients with tuberculosis in a phase 1/2 randomized clinical trial (TBCOX2) of the cyclooxygenase-2 inhibitor etoricoxib. METHODS Peripheral blood mononuclear cells from 15 patients with tuberculosis treated with adjunctive COX-2i and 18 controls (standard therapy) were collected on day 56 after treatment initiation. The ex vivo capacity of macrophages to control mycobacterial infection was assessed by challenge with Mycobacterium avium, using an in vitro culture model. Macrophage inflammatory responses were analyzed by gene expression signatures, and concentrations of cytokines were analyzed in supernatants by multiplex. RESULTS Macrophages from patients receiving adjunctive COX-2i treatment had higher M. avium loads than controls after 6 days, suggesting an impaired capacity to control mycobacterial infection compared to macrophages from the control group. Macrophages from the COX-2i group had lower gene expression of TNF, IL-1B, CCL4, CXCL9, and CXCL10 and lowered production of cytokines IFN-β and S100A8/A9 than controls. CONCLUSIONS Our data suggest potential unfavorable effects with impaired macrophage capacity to control mycobacterial growth in patients with tuberculosis receiving COX-2i treatment. Larger clinical trials are required to analyze the safety of COX-2i as HDT in patients with tuberculosis. CLINICAL TRIALS REGISTRATION NCT02503839.
Collapse
Affiliation(s)
- Kristin G Nore
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Claire Louet
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Marit Bugge
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Alexandre Gidon
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | | | - Synne Jenum
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Anne Ma Dyrhol-Riise
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Kristian Tonby
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Trude Helen Flo
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Infection, St Olav's Hospital, Trondheim, Norway
| |
Collapse
|
43
|
Shuster M, Lyu Z, Augenstreich J, Mathur S, Ganesh A, Ling J, Briken V. Salmonella Typhimurium infection inhibits macrophage IFNβ signaling in a TLR4-dependent manner. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.05.583530. [PMID: 38496427 PMCID: PMC10942315 DOI: 10.1101/2024.03.05.583530] [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
Type I Interferons (IFNs) generally have a protective role during viral infections, but their function during bacterial infections is dependent on the bacterial species. Legionella pneumophila, Shigella sonnei and Mycobacterium tuberculosis can inhibit type I IFN signaling. Here we examined the role of type I IFN, specifically IFNβ, in the context of Salmonella enterica serovar Typhimurium (STm) macrophage infections and the capacity of STm to inhibit type I IFN signaling. We demonstrate that IFNβ has no effect on the intracellular growth of STm in infected bone marrow derived macrophages (BMDMs) derived from C57BL/6 mice. STm infection inhibits IFNβ signaling but not IFNγ signaling in a murine macrophage cell line. We show that this inhibition is independent of the type III and type VI secretion systems expressed by STm and is also independent of bacterial phagocytosis. The inhibition is Toll-like receptor 4 (TLR4)-dependent as the TLR4 ligand, lipopolysaccharide (LPS), alone is sufficient to inhibit IFNβ-mediated signaling and STm-infected, TLR4-deficient BMDMs do not exhibit inhibited IFNβ signaling. In summary, we show that macrophages exposed to STm have reduced IFNβ signaling via crosstalk with TLR4 signaling, and that IFNβ signaling does not affect cell autonomous host defense against STm.
Collapse
Affiliation(s)
- Michael Shuster
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, USA
| | - Zhihui Lyu
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, USA
| | - Jacques Augenstreich
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, USA
| | - Shrestha Mathur
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, USA
| | - Akshaya Ganesh
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, USA
| | - Jiqiang Ling
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, USA
| | - Volker Briken
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, USA
| |
Collapse
|
44
|
Schorey JS, Vecchio J, McManus WR, Ongalo J, Webber K. Activation of host nucleic acid sensors by Mycobacterium: good for us or good for them? Crit Rev Microbiol 2024; 50:224-240. [PMID: 38153209 PMCID: PMC10985831 DOI: 10.1080/1040841x.2023.2294904] [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: 09/18/2023] [Revised: 12/06/2023] [Accepted: 12/10/2023] [Indexed: 12/29/2023]
Abstract
Although the importance of deoxyribonucleic acid (DNA) and ribonucleic acid (RNA) sensors in controlling viral infection is well established, their role in promoting an effective immune response to pathogens other than viruses is less clear. This is particularly true for infections with mycobacteria, as studies point to both protective and detrimental roles for activation of nucleic acid sensors in controlling a mycobacterial infection. Some of the contradiction likely stems from the use of different model systems and different mycobacterial species/strains as well as from which nucleic acid sensors were studied and what downstream effectors were evaluated. In this review, we will describe the different nucleic acid sensors that have been studied in the context of mycobacterial infections, and how the different studies compare. We conclude with a section on how nucleic acid sensor agonists have been used therapeutically and what further information is needed to enhance their potential as therapeutic agents.
Collapse
Affiliation(s)
- Jeffery S. Schorey
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556
| | - Joseph Vecchio
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556
| | - William R. McManus
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556
| | - Joshua Ongalo
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556
| | - Kylie Webber
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556
| |
Collapse
|
45
|
Lee AM, Laurent P, Nathan CF, Barrat FJ. Neutrophil-plasmacytoid dendritic cell interaction leads to production of type I IFN in response to Mycobacterium tuberculosis. Eur J Immunol 2024; 54:e2350666. [PMID: 38161237 DOI: 10.1002/eji.202350666] [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: 07/13/2023] [Revised: 12/13/2023] [Accepted: 12/14/2023] [Indexed: 01/03/2024]
Abstract
Mycobacterium tuberculosis (Mtb) can cause a latent infection that sometimes progresses to clinically active tuberculosis (TB). Type I interferons (IFN-I) have been implicated in initiating the progression from latency to active TB, in part because IFN-I stimulated genes are the earliest genes to be upregulated in patients as they advance to active TB. Plasmacytoid dendritic cells (pDCs) are major producers of IFN-I during viral infections and in response to autoimmune-induced neutrophil extracellular traps. pDCs have also been suggested to be the major producers of IFN-I during Mtb infection of mice and nonhuman primates, but direct evidence has been lacking. Here, we found that Mtb did not stimulate isolated human pDCs to produce IFN-I, but human neutrophils infected with Mtb-activated co-cultured pDCs to do so. Mtb-infected neutrophils produced neutrophil extracellular traps, whose exposed DNA is a well-known mechanism to activate pDCs to secrete IFN-I. We conclude that pDCs contribute to the IFN-I response during Mtb infection by interacting with infected neutrophils which may then promote Mtb pathogenesis.
Collapse
Affiliation(s)
- Angela M Lee
- Department of Microbiology & Immunology, Weill Cornell Medicine, New York, New York, USA
- Immunology & Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, New York, USA
| | - Paôline Laurent
- Department of Microbiology & Immunology, Weill Cornell Medicine, New York, New York, USA
- Hospital for Special Surgery, HSS Research Institute, New York, New York, USA
| | - Carl F Nathan
- Department of Microbiology & Immunology, Weill Cornell Medicine, New York, New York, USA
- Immunology & Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, New York, USA
| | - Franck J Barrat
- Department of Microbiology & Immunology, Weill Cornell Medicine, New York, New York, USA
- Immunology & Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, New York, USA
- Hospital for Special Surgery, HSS Research Institute, New York, New York, USA
| |
Collapse
|
46
|
Serene LG, Webber K, Champion PA, Schorey JS. Mycobacterium tuberculosis SecA2-dependent activation of host Rig-I/MAVs signaling is not conserved in Mycobacterium marinum. PLoS One 2024; 19:e0281564. [PMID: 38394154 PMCID: PMC10889897 DOI: 10.1371/journal.pone.0281564] [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: 02/06/2023] [Accepted: 11/02/2023] [Indexed: 02/25/2024] Open
Abstract
Retinoic acid inducible gene I (Rig-I) is a cytosolic pattern recognition receptor canonically described for its important role in sensing viral RNAs. Increasingly, bacterially-derived RNA from intracellular bacteria such as Mycobacterium tuberculosis, have been shown to activate the same host Rig-I/Mitochondrial antiviral sensing protein (MAVS) signaling pathway to drive a type-I interferon response that contributes to bacterial pathogenesis in vivo. In M. tuberculosis, this response is mediated by the protein secretion system SecA2, but little is known about whether this process is conserved in other pathogenic mycobacteria or the mechanism by which these nucleic acids gain access to the host cytoplasm. Because the M. tuberculosis and M. marinum SecA2 protein secretion systems share a high degree of genetic and functional conservation, we hypothesized that Rig-I/MAVS activation and subsequent induction of IFN-β secretion by host macrophages will also be conserved between these two mycobacterial species. To test this, we generated a ΔsecA2 M. marinum strain along with complementation strains expressing either the M. marinum or M. tuberculosis secA2 genes. Our results suggest that the ΔsecA2 strain has a growth defect in vitro but not in host macrophages. These intracellular growth curves also suggested that the calculation applied to estimate the number of bacteria added to macrophage monolayers in infection assays underestimates bacterial inputs for the ΔsecA2 strain. Therefore, to better examine secreted IFN-β levels when bacterial infection levels are equal across strains we plated bacterial CFUs at 2hpi alongside our ELISA based infections. This enabled us to normalize secreted levels of IFN-β to a standard number of bacteria. Applying this approach to both WT and MAVS-/- bone marrow derived macrophages we observed equal or higher levels of secreted IFN-β from macrophages infected with the ΔsecA2 M. marinum strain as compared to WT. Together our findings suggest that activation of host Rig-I/MAVS cytosolic sensors and subsequent induction of IFN-β response in a SecA2-dependent manner is not conserved in M. marinum under the conditions tested.
Collapse
Affiliation(s)
- Lindsay G. Serene
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, United States of America
- Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN, United States of America
| | - Kylie Webber
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, United States of America
- Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN, United States of America
| | - Patricia A. Champion
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, United States of America
- Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN, United States of America
| | - Jeffrey S. Schorey
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, United States of America
- Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN, United States of America
| |
Collapse
|
47
|
Minayoshi Y, Maeda H, Hamasaki K, Nagasaki T, Takano M, Fukuda R, Mizuta Y, Tanaka M, Sasaki Y, Otagiri M, Watanabe H, Maruyama T. Mouse Type-I Interferon-Mannosylated Albumin Fusion Protein for the Treatment of Chronic Hepatitis. Pharmaceuticals (Basel) 2024; 17:260. [PMID: 38399475 PMCID: PMC10893114 DOI: 10.3390/ph17020260] [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: 12/31/2023] [Revised: 02/03/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Although a lot of effort has been put into creating drugs and combination therapies against chronic hepatitis, no effective treatment has been established. Type-I interferon is a promising therapeutic for chronic hepatitis due to its excellent anti-inflammatory effects through interferon receptors on hepatic macrophages. To develop a type-I IFN equipped with the ability to target hepatic macrophages through the macrophage mannose receptor, the present study designed a mouse type-I interferon-mannosylated albumin fusion protein using site-specific mutagenesis and albumin fusion technology. This fusion protein exhibited the induction of anti-inflammatory molecules, such as IL-10, IL-1Ra, and PD-1, in RAW264.7 cells, or hepatoprotective effects on carbon tetrachloride-induced chronic hepatitis mice. As expected, such biological and hepatoprotective actions were significantly superior to those of human fusion proteins. Furthermore, the repeated administration of mouse fusion protein to carbon tetrachloride-induced chronic hepatitis mice clearly suppressed the area of liver fibrosis and hepatic hydroxyproline contents, not only with a reduction in the levels of inflammatory cytokine (TNF-α) and fibrosis-related genes (TGF-β, Fibronectin, Snail, and Collagen 1α2), but also with a shift in the hepatic macrophage phenotype from inflammatory to anti-inflammatory. Therefore, type-I interferon-mannosylated albumin fusion protein has the potential as a new therapeutic agent for chronic hepatitis.
Collapse
Affiliation(s)
- Yuki Minayoshi
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-Honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (Y.M.); (K.H.); (T.N.); (M.T.); (R.F.); (Y.M.); (H.W.)
| | - Hitoshi Maeda
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-Honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (Y.M.); (K.H.); (T.N.); (M.T.); (R.F.); (Y.M.); (H.W.)
| | - Keisuke Hamasaki
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-Honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (Y.M.); (K.H.); (T.N.); (M.T.); (R.F.); (Y.M.); (H.W.)
| | - Taisei Nagasaki
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-Honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (Y.M.); (K.H.); (T.N.); (M.T.); (R.F.); (Y.M.); (H.W.)
| | - Mei Takano
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-Honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (Y.M.); (K.H.); (T.N.); (M.T.); (R.F.); (Y.M.); (H.W.)
| | - Ryo Fukuda
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-Honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (Y.M.); (K.H.); (T.N.); (M.T.); (R.F.); (Y.M.); (H.W.)
| | - Yuki Mizuta
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-Honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (Y.M.); (K.H.); (T.N.); (M.T.); (R.F.); (Y.M.); (H.W.)
| | - Motohiko Tanaka
- Department of Gastroenterology and Hepatology, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan; (M.T.); (Y.S.)
- Public Health and Welfare Bureau, 5-1-1 Oe, Chuo-ku, Kumamoto 862-0971, Japan
| | - Yutaka Sasaki
- Department of Gastroenterology and Hepatology, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan; (M.T.); (Y.S.)
- Osaka Central Hospital, 3-3-30 Umeda, Kita-ku, Osaka 530-0001, Japan
| | - Masaki Otagiri
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 860-0082, Japan;
- DDS Research Institute, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 860-0082, Japan
| | - Hiroshi Watanabe
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-Honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (Y.M.); (K.H.); (T.N.); (M.T.); (R.F.); (Y.M.); (H.W.)
| | - Toru Maruyama
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-Honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (Y.M.); (K.H.); (T.N.); (M.T.); (R.F.); (Y.M.); (H.W.)
| |
Collapse
|
48
|
Hkimi C, Kamoun S, Khamessi O, Ghedira K. Mycobacterium tuberculosis-THP-1 like macrophages protein-protein interaction map revealed through dual RNA-seq analysis and a computational approach. J Med Microbiol 2024; 73. [PMID: 38314675 DOI: 10.1099/jmm.0.001803] [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: 02/06/2024] Open
Abstract
Introduction. Infection caused by Mycobacterium tuberculosis (M. tb) is still a leading cause of mortality worldwide with estimated 1.4 million deaths annually.Hypothesis/Gap statement. Despite macrophages' ability to kill bacterium, M. tb can grow inside these innate immune cells and the exploration of the infection has traditionally been characterized by a one-sided relationship, concentrating solely on the host or examining the pathogen in isolation.Aim. Because of only a handful of M. tb-host interactions have been experimentally characterized, our main goal is to predict protein-protein interactions during the early phases of the infection.Methodology. In this work, we performed an integrative computational approach that exploits differentially expressed genes obtained from Dual RNA-seq analysis combined with known domain-domain interactions.Results. A total of 2381 and 7214 genes were identified as differentially expressed in M. tb and in THP-1-like macrophages, respectively, revealing different transcriptional profiles in response to infection. Over 48 h of infection, the host-pathogen network revealed 25 016 PPIs. Analysis of the resulting predicted network based on cellular localization information of M. tb proteins, indicated the implication of interacting nodes including the bacterial PE/PPE/PE_PGRS family. In addition, M. tb proteins interacted with host proteins involved in NF-kB signalling pathway as well as interfering with the host apoptosis ability via the potential interaction of M. tb TB16.3 with human TAB1 and M. tb GroEL2 with host protein kinase C delta, respectively.Conclusion. The prediction of the full range of interactions between M. tb and host will contribute to better understanding of the pathogenesis of this bacterium and may provide advanced approaches to explore new therapeutic targets against tuberculosis.
Collapse
Affiliation(s)
- Chaima Hkimi
- Laboratory of Bioinformatics, Biomathematics and Biostatistics (LR20IPT09), Pasteur Institute of Tunis, Tunis 1002, Tunisia
- Higher Institute of Biotechnology of Sidi Thabet, University of Manouba, Ariana BP-66, Manouba 2010, Tunisia
| | - Selim Kamoun
- Laboratory of Bioinformatics, Biomathematics and Biostatistics (LR20IPT09), Pasteur Institute of Tunis, Tunis 1002, Tunisia
- Higher Institute of Biotechnology of Sidi Thabet, University of Manouba, Ariana BP-66, Manouba 2010, Tunisia
| | - Oussema Khamessi
- Laboratory of Bioinformatics, Biomathematics and Biostatistics (LR20IPT09), Pasteur Institute of Tunis, Tunis 1002, Tunisia
- Higher Institute of Biotechnology of Sidi Thabet, University of Manouba, Ariana BP-66, Manouba 2010, Tunisia
| | - Kais Ghedira
- Laboratory of Bioinformatics, Biomathematics and Biostatistics (LR20IPT09), Pasteur Institute of Tunis, Tunis 1002, Tunisia
| |
Collapse
|
49
|
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: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [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
|
50
|
Mousavian Z, Källenius G, Sundling C. From simple to complex: Protein-based biomarker discovery in tuberculosis. Eur J Immunol 2023; 53:e2350485. [PMID: 37740950 DOI: 10.1002/eji.202350485] [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: 06/27/2023] [Revised: 08/15/2023] [Accepted: 09/22/2023] [Indexed: 09/25/2023]
Abstract
Tuberculosis (TB) is a deadly infectious disease that affects millions of people globally. TB proteomics signature discovery has been a rapidly growing area of research that aims to identify protein biomarkers for the early detection, diagnosis, and treatment monitoring of TB. In this review, we have highlighted recent advances in this field and how it is moving from the study of single proteins to high-throughput profiling and from only using proteomics to include additional types of data in multi-omics studies. We have further covered the different sample types and experimental technologies used in TB proteomics signature discovery, focusing on studies of HIV-negative adults. The published signatures were defined as either coming from hypothesis-based protein targeting or from unbiased discovery approaches. The methodological approaches influenced the type of proteins identified and were associated with the circulating protein abundance. However, both approaches largely identified proteins involved in similar biological pathways, including acute-phase responses and T-helper type 1 and type 17 responses. By analysing the frequency of proteins in the different signatures, we could also highlight potential robust biomarker candidates. Finally, we discuss the potential value of integration of multi-omics data and the importance of control cohorts and signature validation.
Collapse
Affiliation(s)
- Zaynab Mousavian
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Gunilla Källenius
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Christopher Sundling
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|