1
|
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
|
2
|
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
|
3
|
Dutt TS, Choreño-Parra JA. Editorial: Tuberculosis and humoral immunity. Front Immunol 2025; 16:1562567. [PMID: 39995662 PMCID: PMC11847867 DOI: 10.3389/fimmu.2025.1562567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 01/27/2025] [Indexed: 02/26/2025] Open
Affiliation(s)
- Taru S. Dutt
- Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - José Alberto Choreño-Parra
- Departamento de Enseñanza, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| |
Collapse
|
4
|
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
|
5
|
Ssekamatte P, Nabatanzi R, Sitenda D, Nakibuule M, Bagaya BS, Kibirige D, Kyazze AP, Kateete DP, Sande OJ, van Crevel R, Cose S, Biraro IA. Impaired Mycobacterium tuberculosis-specific T-cell memory phenotypes and functional profiles among adults with type 2 diabetes mellitus in Uganda. Front Immunol 2024; 15:1480739. [PMID: 39430752 PMCID: PMC11486641 DOI: 10.3389/fimmu.2024.1480739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 09/18/2024] [Indexed: 10/22/2024] Open
Abstract
Background Efforts to eradicate tuberculosis (TB) are threatened by diabetes mellitus (DM), which confers a 3-fold increase in the risk of TB disease. The changes in the memory phenotypes and functional profiles of Mycobacterium tuberculosis (Mtb)-specific T cells in latent TB infection (LTBI)-DM participants remain poorly characterised. We, therefore, assessed the effect of DM on T-cell phenotype and function in LTBI and DM clinical groups. Methods We compared the memory phenotypes and function profiles of Mtb-specific CD4+ and CD8+ T cells among participants with LTBI-DM (n=21), LTBI-only (n=17) and DM-only (n=16). Peripheral blood mononuclear cells (PBMCs) were stimulated with early secretory antigenic 6 kDa (ESAT-6) and culture filtrate protein 10 (CFP-10) peptide pools or phytohemagglutinin (PHA). The memory phenotypes (CCR7/CD45RA), and functional profiles (HLA-DR, PD-1, CD107a, IFN-γ, IL-2, TNF, IL-13, IL-17A) of Mtb-specific CD4+ and CD8+ T cells were characterised by flow cytometry. Results Naïve CD4+ T cells were significantly decreased in the LTBI-DM compared to the LTBI-only participants [0.47 (0.34-0.69) vs 0.91 (0.59-1.05); (p<0.001)]. Similarly, CD8+ HLA-DR expression was significantly decreased in LTBI-DM compared to LTBI-only participants [0.26 (0.19-0.33) vs 0.52 (0.40-0.64); (p<0.0001)], whereas CD4+ and CD8+ PD-1 expression was significantly upregulated in the LTBI-DM compared to the LTBI-only participants [0.61 (0.53-0.77) vs 0.19 (0.10-0.28); (p<0.0001) and 0.41 (0.37-0.56) vs 0.29 (0.17-0.42); (p=0.007)] respectively. CD4+ and CD8+ IFN-γ production was significantly decreased in the LTBI-DM compared to the LTBI-only participants [0.28 (0.19-0.38) vs 0.39 (0.25-0.53); (p=0.030) and 0.36 (0.27-0.49) vs 0.55 (0.41-0.88); (p=0.016)] respectively. CD4+ TNF and CD8+ IL-17A production were significantly decreased in participants with LTBI-DM compared to those with LTBI-only [0.38 (0.33-0.50) vs 0.62 (0.46-0.87); (p=0.004) and 0.29 (0.16-0.42) vs 0.47 (0.29-0.52); (0.017)] respectively. LTBI-DM participants had significantly lower dual-functional (IFN-γ+IL-2+ and IL-2+TNF+) and mono-functional (IFN-γ+ and TNF+) CD4+ responses than LTBI-only participants. LTBI-DM participants had significantly decreased dual-functional (IFN-γ+IL-2+, IFN-γ+ TNF+ and IL-2+TNF+) and mono-functional (IFN-γ+, IL-2+ and TNF+) central and effector memory CD4+ responses compared to LTBI-only participants. Conclusion Type 2 DM impairs the memory phenotypes and functional profiles of Mtb-specific CD4+ and CD8+ T cells, potentially indicating underlying immunopathology towards increased active TB disease risk.
Collapse
Affiliation(s)
- Phillip Ssekamatte
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine, Entebbe, Uganda
| | - Rose Nabatanzi
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Diana Sitenda
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Marjorie Nakibuule
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine, Entebbe, Uganda
| | - Bernard Ssentalo Bagaya
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Davis Kibirige
- Department of Medicine, Uganda Martyrs Lubaga Hospital, Kampala, Uganda
| | - Andrew Peter Kyazze
- Department of Internal Medicine, School of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| | - David Patrick Kateete
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Obondo James Sande
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Reinout van Crevel
- Department of Internal Medicine and Radboud Centre for Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Stephen Cose
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine, Entebbe, Uganda
| | - Irene Andia Biraro
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine, Entebbe, Uganda
- Department of Internal Medicine, School of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| |
Collapse
|
6
|
Li X, Luo X, Wang B, Fu L, Chen X, Lu Y. Clofazimine inhibits innate immunity against Mycobacterium tuberculosis by NF-κB. mSphere 2024; 9:e0025424. [PMID: 39046230 PMCID: PMC11351037 DOI: 10.1128/msphere.00254-24] [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: 03/26/2024] [Accepted: 06/20/2024] [Indexed: 07/25/2024] Open
Abstract
Tuberculosis (TB) remains one of the infectious diseases with high incidence and high mortality. About a quarter of the population has been latently infected with Mycobacterium tuberculosis. At present, the available TB treatment strategies have the disadvantages of too long treatment duration and serious adverse reactions. The sustained inflammatory response leads to permanent tissue damage. Unfortunately, the current selection of treatment regimens does not consider the immunomodulatory effects of various drugs. In this study, we preliminarily evaluated the effects of commonly used anti-tuberculosis drugs on innate immunity at the cellular level. The results showed that clofazimine (CFZ) has a significant innate immunosuppressive effect. CFZ significantly inhibited cytokines and type I interferons (IFNα and IFNβ) expression under both lipopolysaccharide stimulation and CFZ-resistant strain infection. In further mechanistic studies, CFZ strongly inhibited the phosphorylation of nuclear factor kappa B (NF-κB) p65 and had no significant effect on the phosphorylation of p38. In conclusion, our study found that CFZ suppresses innate immunity against Mycobacterium tuberculosis by NF-κB, which should be considered in future regimen development. IMPORTANCE The complete elimination of Mycobacterium tuberculosis (Mtb), the etiologic agent of TB, from TB patients is a complicated process that takes a long time. The excessive immune inflammatory response of the host for a long time causes irreversible organic damage to the lungs and liver. Current antibiotic-based treatment options involve multiple complex drug combinations, often targeting different physiological processes of Mtb. Given the high incidence of post-tuberculosis lung disease, we should also consider the immunomodulatory properties of other drugs when selecting drug combinations.
Collapse
Affiliation(s)
- Xinda Li
- Department of Pharmacology, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Drug Resistance Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Xiaoyi Luo
- Department of Pharmacology, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Drug Resistance Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Bin Wang
- Department of Pharmacology, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Drug Resistance Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Lei Fu
- Department of Pharmacology, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Drug Resistance Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Xi Chen
- Department of Pharmacology, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Drug Resistance Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Yu Lu
- Department of Pharmacology, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Drug Resistance Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| |
Collapse
|
7
|
Ishida E, Corrigan DT, Chen T, Liu Y, Kim RS, Song L, Rutledge TM, Magee DM, LaBaer J, Lowary TL, Lin PL, Achkar JM. Mucosal and systemic antigen-specific antibody responses correlate with protection against active tuberculosis in nonhuman primates. EBioMedicine 2024; 99:104897. [PMID: 38096687 PMCID: PMC10758715 DOI: 10.1016/j.ebiom.2023.104897] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 11/15/2023] [Accepted: 11/15/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Increasing evidence supports that antibodies can protect against active tuberculosis (TB) but knowledge of potentially protective antigens, especially in the airways, is limited. The main objective of this study was to identify antigen-specific airway and systemic immunoglobulin isotype responses associated with the outcome of controlled latent Mycobacterium tuberculosis (Mtb) infection (LTBI) versus uncontrolled infection (TB) in nonhuman primates. METHODS In a case-control design, using non-parametric group comparisons with false discovery rate adjustments, we assessed antibodies in 57 cynomolgus macaques which, following low-dose airway Mtb infection, developed either LTBI or TB. We investigated airway and systemic IgG, IgA, and IgM responses in paired bronchoalveolar lavage and plasma samples prior to, two-, and 5-6-months post Mtb infection using an antigen-unbiased approach with Mtb glycan and proteome-wide microarrays. FINDINGS Macaques that developed LTBI (n = 36) had significantly increased airway and plasma IgA reactivities to specific arabinomannan (AM) motifs prior to Mtb infection compared to those that developed TB (n = 21; p < 0.01, q < 0.05). Furthermore, LTBI macaques had higher plasma IgG reactivity to protein MTB32A (Rv0125) early post Mtb infection (p < 0.05) and increasing airway IgG responses to some proteins over time. INTERPRETATION Our results support a protective role of pre-existing mucosal (lung) and systemic IgA to specific Mtb glycan motifs, suggesting that prior exposure to nontuberculous mycobacteria could be protective against TB. They further suggest that IgG to Mtb proteins early post infection could provide an additional protective mechanism. These findings could inform TB vaccine development strategies. FUNDING NIH/NIAID AI117927, AI146329, and AI127173 to JMA.
Collapse
Affiliation(s)
- Elise Ishida
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Devin T Corrigan
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Tingting Chen
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yanyan Liu
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ryung S Kim
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Lusheng Song
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Tara M Rutledge
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - D Mitchell Magee
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Joshua LaBaer
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Todd L Lowary
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada; Institute of Biological Chemistry, Academia Sinica, Nangang Taipei, Taiwan; Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Philana Ling Lin
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jacqueline M Achkar
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
8
|
Mott D, Yang J, Baer C, Papavinasasundaram K, Sassetti CM, Behar SM. High Bacillary Burden and the ESX-1 Type VII Secretion System Promote MHC Class I Presentation by Mycobacterium tuberculosis-Infected Macrophages to CD8 T Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1531-1542. [PMID: 37000471 PMCID: PMC10159937 DOI: 10.4049/jimmunol.2300001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 02/21/2023] [Indexed: 04/01/2023]
Abstract
We used a mouse model to study how Mycobacterium tuberculosis subverts host defenses to persist in macrophages despite immune pressure. CD4 T cells can recognize macrophages infected with a single bacillus in vitro. Under identical conditions, CD8 T cells inefficiently recognize infected macrophages and fail to restrict M. tuberculosis growth, although they can inhibit M. tuberculosis growth during high-burden intracellular infection. We show that high intracellular M. tuberculosis numbers cause macrophage death, leading other macrophages to scavenge cellular debris and cross-present the TB10.4 Ag to CD8 T cells. Presentation by infected macrophages requires M. tuberculosis to have a functional ESX-1 type VII secretion system. These data indicate that phagosomal membrane damage and cell death promote MHC class I presentation of the immunodominant Ag TB10.4 by macrophages. Although this mode of Ag presentation stimulates cytokine production that we presume would be host beneficial, killing of uninfected cells could worsen immunopathology. We suggest that shifting the focus of CD8 T cell recognition to uninfected macrophages would limit the interaction of CD8 T cells with infected macrophages and impair CD8 T cell-mediated resolution of tuberculosis.
Collapse
Affiliation(s)
- Daniel Mott
- 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
| | - Jason Yang
- 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
| | - Christina Baer
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Kadamba Papavinasasundaram
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Christopher M. Sassetti
- 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
| | - 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
|
9
|
Caputo MB, Elias J, Cesar G, Alvarez MG, Laucella SA, Albareda MC. Role of the Complement System in the Modulation of T-Cell Responses in Chronic Chagas Disease. Front Cell Infect Microbiol 2022; 12:910854. [PMID: 35846776 PMCID: PMC9282465 DOI: 10.3389/fcimb.2022.910854] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 05/26/2022] [Indexed: 01/19/2023] Open
Abstract
Chagas disease, caused by the intracellular pathogen Trypanosoma cruzi, is the parasitic disease with the greatest impact in Latin America and the most common cause of infectious myocarditis in the world. The immune system plays a central role in the control of T. cruzi infection but at the same time needs to be controlled to prevent the development of pathology in the host. It has been shown that persistent infection with T. cruzi induces exhaustion of parasite-specific T cell responses in subjects with chronic Chagas disease. The continuous inflammatory reaction due to parasite persistence in the heart also leads to necrosis and fibrosis. The complement system is a key element of the innate immune system, but recent findings have also shown that the interaction between its components and immune cell receptors might modulate several functions of the adaptive immune system. Moreover, the findings that most of immune cells can produce complement proteins and express their receptors have led to the notion that the complement system also has non canonical functions in the T cell. During human infection by T. cruzi, complement activation might play a dual role in the acute and chronic phases of Chagas disease; it is initially crucial in controlling parasitemia and might later contributes to the development of symptomatic forms of Chagas disease due to its role in T-cell regulation. Herein, we will discuss the putative role of effector complement molecules on T-cell immune exhaustion during chronic human T. cruzi infection.
Collapse
Affiliation(s)
- María Belén Caputo
- Investigation Department, Instituto Nacional de Parasitología Dr. Fatala Chaben, Buenos Aires, Argentina
| | - Josefina Elias
- Investigation Department, Instituto Nacional de Parasitología Dr. Fatala Chaben, Buenos Aires, Argentina
| | - Gonzalo Cesar
- Investigation Department, Instituto Nacional de Parasitología Dr. Fatala Chaben, Buenos Aires, Argentina
| | - María Gabriela Alvarez
- Chagas Section, Hospital Interzonal General de Agudos Eva Perón, Buenos Aires, Argentina
| | - Susana Adriana Laucella
- Investigation Department, Instituto Nacional de Parasitología Dr. Fatala Chaben, Buenos Aires, Argentina
- Chagas Section, Hospital Interzonal General de Agudos Eva Perón, Buenos Aires, Argentina
| | - María Cecilia Albareda
- Investigation Department, Instituto Nacional de Parasitología Dr. Fatala Chaben, Buenos Aires, Argentina
| |
Collapse
|
10
|
Druszczyńska M, Godkowicz M, Kulesza J, Wawrocki S, Fol M. Cytokine Receptors-Regulators of Antimycobacterial Immune Response. Int J Mol Sci 2022; 23:1112. [PMID: 35163035 PMCID: PMC8835057 DOI: 10.3390/ijms23031112] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 01/16/2022] [Accepted: 01/17/2022] [Indexed: 12/18/2022] Open
Abstract
Cytokine receptors are critical regulators of the antimycobacterial immune response, playing a key role in initiating and coordinating the recruitment and activation of immune cells during infection. They recognize and bind specific cytokines and are involved in inducing intracellular signal transduction pathways that regulate a diverse range of biological functions, including proliferation, differentiation, metabolism and cell growth. Due to mutations in cytokine receptor genes, defective signaling may contribute to increased susceptibility to mycobacteria, allowing the pathogens to avoid killing and immune surveillance. This paper provides an overview of cytokine receptors important for the innate and adaptive immune responses against mycobacteria and discusses the implications of receptor gene defects for the course of mycobacterial infection.
Collapse
Affiliation(s)
- Magdalena Druszczyńska
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, Banacha 12/16, 90-237 Lodz, Poland; (M.G.); (S.W.); (M.F.)
| | - Magdalena Godkowicz
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, Banacha 12/16, 90-237 Lodz, Poland; (M.G.); (S.W.); (M.F.)
- Lodz Institutes of the Polish Academy of Sciences, The Bio-Med-Chem Doctoral School, University of Lodz, 90-237 Lodz, Poland
| | - Jakub Kulesza
- Department of Internal Diseases and Clinical Pharmacology, Medical University of Lodz, Kniaziewicza 1/5, 91-347 Lodz, Poland;
| | - Sebastian Wawrocki
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, Banacha 12/16, 90-237 Lodz, Poland; (M.G.); (S.W.); (M.F.)
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, 7265 Davos, Switzerland
| | - Marek Fol
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, Banacha 12/16, 90-237 Lodz, Poland; (M.G.); (S.W.); (M.F.)
| |
Collapse
|
11
|
Co DO, Hogan LH, Karman J, Herbath M, Fabry Z, Sandor M. T Cell Interactions in Mycobacterial Granulomas: Non-Specific T Cells Regulate Mycobacteria-Specific T Cells in Granulomatous Lesions. Cells 2021; 10:cells10123285. [PMID: 34943793 PMCID: PMC8699651 DOI: 10.3390/cells10123285] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 11/16/2021] [Accepted: 11/20/2021] [Indexed: 11/16/2022] Open
Abstract
Infections with pathogenic mycobacteria are controlled by the formation of a unique structure known as a granuloma. The granuloma represents a host–pathogen interface where bacteria are killed and confined by the host response, but also where bacteria persist. Previous work has demonstrated that the T cell repertoire is heterogenous even at the single granuloma level. However, further work using pigeon cytochrome C (PCC) epitope-tagged BCG (PCC-BCG) and PCC-specific 5CC7 RAG−/− TCR transgenic (Tg) mice has demonstrated that a monoclonal T cell population is able to control infection. At the chronic stage of infection, granuloma-infiltrating T cells remain highly activated in wild-type mice, while T cells in the monoclonal T cell mice are anergic. We hypothesized that addition of an acutely activated non-specific T cell to the monoclonal T cell system could recapitulate the wild-type phenotype. Here we report that activated non-specific T cells have access to the granuloma and deliver a set of cytokines and chemokines to the lesions. Strikingly, non-specific T cells rescue BCG-specific T cells from anergy and enhance the function of BCG-specific T cells in the granuloma in the chronic phase of infection when bacterial antigen load is low. In addition, we find that these same non-specific T cells have an inhibitory effect on systemic BCG-specific T cells. Taken together, these data suggest that T cells non-specific for granuloma-inducing agents can alter the function of granuloma-specific T cells and have important roles in mycobacterial immunity and other granulomatous disorders.
Collapse
Affiliation(s)
- Dominic O. Co
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA;
| | - Laura H. Hogan
- The Institute for Clinical and Translational Research, University of Wisconsin-Madison, Madison, WI 53705, USA;
| | - Jozsef Karman
- Cambridge Research Center, Abbvie, Inc., Cambridge, MA 02139, USA;
| | - Melinda Herbath
- Department of Pathology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA; (M.H.); (Z.F.)
| | - Zsuzsanna Fabry
- Department of Pathology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA; (M.H.); (Z.F.)
| | - Matyas Sandor
- Department of Pathology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA; (M.H.); (Z.F.)
- Correspondence: ; Tel.: +1-(608)-265-8715
| |
Collapse
|
12
|
Davids M, Pooran A, Smith L, Tomasicchio M, Dheda K. The Frequency and Effect of Granulocytic Myeloid-Derived Suppressor Cells on Mycobacterial Survival in Patients With Tuberculosis: A Preliminary Report. Front Immunol 2021; 12:676679. [PMID: 34149712 PMCID: PMC8207464 DOI: 10.3389/fimmu.2021.676679] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/05/2021] [Indexed: 12/31/2022] Open
Abstract
Introduction Protective host responses in those exposed to or infected with tuberculosis (TB) is thought to require a delicate balance between pro-inflammatory and regulatory immune responses. Myeloid-derived suppressor cells (MDSCs), regulatory cells that dampen T-cell function, have been described in cancer and other infectious diseases but there are limited data on their role in TB. Methods Peripheral blood was obtained from patients with active pulmonary TB and participants with presumed latent TB infection (LTBI) from Cape Town, South Africa. MDSC frequency was ascertained by flow cytometry. Purified MDSCs were used to assess (i) their suppressive effect on T-cell proliferation using a Ki67 flow cytometric assay and (ii) their effect on mycobacterial containment by co-culturing with H37Rv-infected monocyte-derived macrophages and autologous pre-primed effector T-cells with or without MDSCs. Mycobacterial containment was measured by plating colony forming units (CFU). Results MDSCs (CD15+HLA-DR-CD33+) had significantly higher median frequencies (IQR) in patients with active TB (n=10) versus LTBI (n= 10) [8.2% (6.8-10.7) versus 42.2% (27-56) respectively; p=0.001]. Compared to MDSC-depleted peripheral blood mononuclear and effector T cell populations, dilutions of purified MDSCs isolated from active TB patients suppressed T-cell proliferation by up to 72% (n=6; p=0.03) and significantly subverted effector T-cell-mediated containment of H37Rv in monocyte-derived macrophages (n=7; 0.6% versus 8.5%; p=0.02). Conclusion Collectively, these data suggest that circulating MDSCs are induced during active TB disease and can functionally suppress T-cell proliferation and subvert mycobacterial containment. These data may inform the design of vaccines and immunotherapeutic interventions against TB but further studies are required to understand the mechanisms underpinning the effects of MDSCs.
Collapse
Affiliation(s)
- Malika Davids
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine and UCT Lung Institute & South African MRC/UCT Centre for the Study of Antimicrobial Resistance, University of Cape Town, Cape Town, South Africa
| | - Anil Pooran
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine and UCT Lung Institute & South African MRC/UCT Centre for the Study of Antimicrobial Resistance, University of Cape Town, Cape Town, South Africa
| | - Liezel Smith
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine and UCT Lung Institute & South African MRC/UCT Centre for the Study of Antimicrobial Resistance, University of Cape Town, Cape Town, South Africa
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research; South African Medical Research Council Centre for Tuberculosis Research; Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Michele Tomasicchio
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine and UCT Lung Institute & South African MRC/UCT Centre for the Study of Antimicrobial Resistance, University of Cape Town, Cape Town, South Africa
| | - Keertan Dheda
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine and UCT Lung Institute & South African MRC/UCT Centre for the Study of Antimicrobial Resistance, University of Cape Town, Cape Town, South Africa
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
13
|
He X, Eddy JJ, Jacobson KR, Henderson AJ, Agosto LM. Enhanced Human Immunodeficiency Virus-1 Replication in CD4+ T Cells Derived From Individuals With Latent Mycobacterium tuberculosis Infection. J Infect Dis 2021; 222:1550-1560. [PMID: 32417884 DOI: 10.1093/infdis/jiaa257] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 05/09/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Mycobacterium tuberculosis (Mtb) and human immunodeficiency virus (HIV) coinfection increases mortality, accelerates progression to acquired immune deficiency syndrome, and exacerbates tuberculosis disease. However, the impact of pre-existing Mtb infection on subsequent HIV infection has not been fully explored. We hypothesized that Mtb infection creates an immunological environment that influences the course of HIV infection, and we investigated whether pre-existing Mtb infection impacts the susceptibility of CD4+ T cells to HIV-1 infection. METHODS Plasma and blood CD4+ T cells isolated from HIV-negative individuals across the Mtb infection spectrum and non-Mtb-infected control individuals were analyzed for inflammation markers and T-cell phenotypes. CD4+ T cells were infected with HIV-1 in vitro and were monitored for viral replication. RESULTS We observed differences in proinflammatory cytokines and the relative proportion of memory T-cell subsets depending on Mtb infection status. CD4+ T cells derived from individuals with latent Mtb infection supported more efficient HIV-1 transcription, release, and replication. Enhanced HIV-1 replication correlated with higher percentages of CD4+ TEM and TTD cells. CONCLUSIONS Pre-existing Mtb infection creates an immunological environment that reflects Mtb infection status and influences the susceptibility of CD4+ T cells to HIV-1 replication. These findings provide cellular and molecular insights into how pre-existing Mtb infection influences HIV-1 pathogenesis.
Collapse
Affiliation(s)
- Xianbao He
- Department of Medicine, Section of Infectious Diseases, Boston University School of Medicine and Boston Medical Center, Boston, Massachusetts, USA
| | - Jared J Eddy
- Department of Medicine, Section of Infectious Diseases, Boston University School of Medicine and Boston Medical Center, Boston, Massachusetts, USA
| | - Karen R Jacobson
- Department of Medicine, Section of Infectious Diseases, Boston University School of Medicine and Boston Medical Center, Boston, Massachusetts, USA
| | - Andrew J Henderson
- Department of Medicine, Section of Infectious Diseases, Boston University School of Medicine and Boston Medical Center, Boston, Massachusetts, USA.,Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Luis M Agosto
- Department of Medicine, Section of Infectious Diseases, Boston University School of Medicine and Boston Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
14
|
Abstract
Tuberculosis (TB) vaccine research has reached a unique point in time. Breakthrough findings in both the basic immunology of Mycobacterium tuberculosis infection and the clinical development of TB vaccines suggest, for the first time since the discovery of the Mycobacterium bovis bacillus Calmette-Guérin (BCG) vaccine more than a century ago, that a novel, efficacious TB vaccine is imminent. Here, we review recent data in the light of our current understanding of the immunology of TB infection and discuss the identification of biomarkers for vaccine efficacy and the next steps in the quest for an efficacious vaccine that can control the global TB epidemic.
Collapse
|
15
|
Locally-secreted interleukin-6 is related with radiological severity in smear-negative pulmonary tuberculosis. Cytokine 2020; 127:154950. [DOI: 10.1016/j.cyto.2019.154950] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 11/20/2019] [Accepted: 12/03/2019] [Indexed: 01/09/2023]
|
16
|
Kaufmann SHE. Vaccination Against Tuberculosis: Revamping BCG by Molecular Genetics Guided by Immunology. Front Immunol 2020; 11:316. [PMID: 32174919 PMCID: PMC7056705 DOI: 10.3389/fimmu.2020.00316] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 02/07/2020] [Indexed: 12/21/2022] Open
Abstract
Tuberculosis (TB) remains a major health threat. Although a vaccine has been available for almost 100 years termed Bacille Calmette-Guérin (BCG), it is insufficient and better vaccines are urgently needed. This treatise describes first the basic immunology and pathology of TB with an emphasis on the role of T lymphocytes. Better understanding of the immune response to Mycobacterium tuberculosis (Mtb) serves as blueprint for rational design of TB vaccines. Then, disease epidemiology and the benefits and failures of BCG vaccination will be presented. Next, types of novel vaccine candidates are being discussed. These include: (i) antigen/adjuvant subunit vaccines; (ii) viral vectored vaccines; and (III) whole cell mycobacterial vaccines which come as live recombinant vaccines or as dead whole cell or multi-component vaccines. Subsequently, the major endpoints of clinical trials as well as administration schemes are being described. Major endpoints for clinical trials are prevention of infection (PoI), prevention of disease (PoD), and prevention of recurrence (PoR). Vaccines can be administered either pre-exposure or post-exposure with Mtb. A central part of this treatise is the description of the viable BCG-based vaccine, VPM1002, currently undergoing phase III clinical trial assessment. Finally, new approaches which could facilitate design of refined next generation TB vaccines will be discussed.
Collapse
Affiliation(s)
- Stefan H. E. Kaufmann
- Max Planck Institute for Infection Biology, Berlin, Germany
- Hagler Institute for Advanced Study, Texas A&M University, College Station, TX, United States
| |
Collapse
|
17
|
Lyashchenko KP, Vordermeier HM, Waters WR. Memory B cells and tuberculosis. Vet Immunol Immunopathol 2020; 221:110016. [PMID: 32050091 DOI: 10.1016/j.vetimm.2020.110016] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/30/2019] [Accepted: 01/29/2020] [Indexed: 02/09/2023]
Abstract
Immunological memory is a central feature of adaptive immunity. Memory B cells are generated upon stimulation with antigen presented by follicular dendritic cells in the peripheral lymphoid tissues. This process typically involves class-switch recombination and somatic hypermutation and it can be dependent or independent on germinal centers or T cell help. The mature B cell memory pool is generally characterized by remarkable heterogeneity of functionally and phenotypically distinct sub-populations supporting multi-layer immune plasticity. Memory B cells found in human patients infected with Mycobacterium tuberculosis include IgD+ CD27+ and IgM+ CD27+ subsets. In addition, expansion of atypical memory B cells characterized by the lack of CD27 expression and by inability to respond to antigen-induced re-activation is documented in human tuberculosis. These functionally impaired memory B cells are believed to have adverse effects on host immunity. Human and animal studies demonstrate recruitment of antigen-activated B cells to the infection sites and their presence in lung granulomas where proliferating B cells are organized into discrete clusters resembling germinal centers of secondary lymphoid organs. Cattle studies show development of IgM+, IgG+, and IgA+ memory B cells in M. bovis infection with the ability to rapidly differentiate into antibody-producing plasma cells upon antigen re-exposure. This review discusses recent advances in research on generation, re-activation, heterogeneity, and immunobiological functions of memory B cells in tuberculosis. The role of memory B cells in post-skin test recall antibody responses in bovine tuberculosis and implications for development of improved immunodiagnostics are also reviewed.
Collapse
Affiliation(s)
| | - H Martin Vordermeier
- Tuberculosis Research Group, Animal and Plant Health Agency, Addlestone, United Kingdom; Institute for Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth, United Kingdom
| | - W Ray Waters
- National Animal Disease Center, Agricultural Research Service, US Department of Agriculture, Ames, IA, USA
| |
Collapse
|
18
|
Patankar YR, Sutiwisesak R, Boyce S, Lai R, Lindestam Arlehamn CS, Sette A, Behar SM. Limited recognition of Mycobacterium tuberculosis-infected macrophages by polyclonal CD4 and CD8 T cells from the lungs of infected mice. Mucosal Immunol 2020; 13:140-148. [PMID: 31636345 PMCID: PMC7161428 DOI: 10.1038/s41385-019-0217-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/23/2019] [Accepted: 09/28/2019] [Indexed: 02/04/2023]
Abstract
Immune responses following Mycobacterium tuberculosis (Mtb) infection or vaccination are frequently assessed by measuring T-cell recognition of crude Mtb antigens, recombinant proteins, or peptide epitopes. We previously showed that not all Mtb-specific T cells recognize Mtb-infected macrophages. Thus, an important question is what proportion of T cells elicited by Mtb infection recognize Mtb-infected macrophages. We address this question by developing a modified elispot assay using viable Mtb-infected macrophages, a low multiplicity of infection and purified T cells. In C57BL/6 mice, CD4 and CD8 T cells were classically MHC restricted. Comparable frequencies of T cells that recognize Mtb-infected macrophages were determined using interferon-γ elispot and intracellular cytokine staining, and lung CD4 T cells more sensitively recognized Mtb-infected macrophages than lung CD8 T cells. Compared to the relatively high frequencies of T cells specific for antigens such as ESAT-6 and TB10.4, low frequencies of total pulmonary T cells elicited by aerosolized Mtb infection recognize Mtb-infected macrophages. Finally, we demonstrate that BCG vaccination elicits T cells that recognize Mtb-infected macrophages. We propose that the frequency of T cells that recognize infected macrophages could correlate with protective immunity and may be an alternative approach to measuring T-cell responses to Mtb antigens.
Collapse
Affiliation(s)
- Yash R. Patankar
- 0000 0001 0742 0364grid.168645.8Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655 USA
| | - Rujapak Sutiwisesak
- 0000 0001 0742 0364grid.168645.8Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655 USA
| | - Shayla Boyce
- 0000 0001 0742 0364grid.168645.8Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655 USA
| | - Rocky Lai
- 0000 0001 0742 0364grid.168645.8Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655 USA
| | - Cecilia S. Lindestam Arlehamn
- 0000 0004 0461 3162grid.185006.aDepartment of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
| | - Alessandro Sette
- 0000 0004 0461 3162grid.185006.aDepartment of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA 92037 USA ,0000 0001 2107 4242grid.266100.3Department of Medicine, University of California San Diego, La Jolla, CA 92093 USA
| | - Samuel M. Behar
- 0000 0001 0742 0364grid.168645.8Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655 USA
| |
Collapse
|
19
|
Urbanowski ME, Ihms EA, Bigelow K, Kübler A, Elkington PT, Bishai WR. Repetitive Aerosol Exposure Promotes Cavitary Tuberculosis and Enables Screening for Targeted Inhibitors of Extensive Lung Destruction. J Infect Dis 2019; 218:53-63. [PMID: 29554286 DOI: 10.1093/infdis/jiy127] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 03/07/2018] [Indexed: 12/22/2022] Open
Abstract
Background Cavitation is a serious consequence of tuberculosis. We tested the hypothesis that repetitive exposure to the same total bacterial burden of Mycobacterium tuberculosis drives greater lung destruction than a single exposure. We also tested whether inhibition of endogenous matrix metalloproteinase-1 (MMP-1) may inhibit cavitation during tuberculosis. Methods Over a 3-week interval, we infected rabbits with either 5 aerosols of 500 colony-forming units (CFU) of M. tuberculosis or a single aerosol of 2500 CFU plus 4 sham aerosols. We administered the MMP-1 inhibitor cipemastat (100 mg/kg daily) during weeks 5-10 to a subset of the animals. Results Repetitive aerosol infection produced greater lung inflammation and more cavities than a single aerosol infection of the same bacterial burden (75% of animals vs 25%). Necropsies confirmed greater lung pathology in repetitively exposed animals. For cipemastat-treated animals, there was no significant difference in cavity counts, cavity volume, or disease severity compared to controls. Conclusions Our data show that repetitive aerosol exposure with M. tuberculosis drives greater lung damage and cavitation than a single exposure. This suggests that human lung destruction due to tuberculosis may be exacerbated in settings where individuals are repeatedly exposed. MMP-1 inhibition with cipemastat did not prevent the development of cavitation in our model.
Collapse
Affiliation(s)
- Michael E Urbanowski
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Elizabeth A Ihms
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kristina Bigelow
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - André Kübler
- Queen's Hospital, Barking, Havering and Redbridge University Hospital National Health Service Trust, Romford, Essex
| | - Paul T Elkington
- National Institute for Health Research Biomedical Research Centre, Clinical and Experimental Sciences Academic Unit, Faculty of Medicine, University of Southampton, United Kingdom.,Institute for Life Sciences, University of Southampton, United Kingdom
| | - William R Bishai
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
20
|
Abstract
Three decades of research in hematopoietic stem cell transplantation and HIV/AIDS fields have shaped a picture of immune restoration disorders. This manuscript overviews the molecular biology of interferon networks, the molecular pathogenesis of immune reconstitution inflammatory syndrome, and post-hematopoietic stem cell transplantation immune restoration disorders (IRD). It also summarizes the effects of thymic involution on T cell diversity, and the results of the assessment of diagnostic biomarkers of IRD, and tested targeted immunomodulatory treatments.
Collapse
Affiliation(s)
- Hesham Mohei
- Department of Medicine, University of Minnesota, Minneapolis, USA
| | - Usha Kellampalli
- Department of Medicine, University of Minnesota, Minneapolis, USA
| | | |
Collapse
|
21
|
Labonte AC, Kegerreis B, Geraci NS, Bachali P, Madamanchi S, Robl R, Catalina MD, Lipsky PE, Grammer AC. Identification of alterations in macrophage activation associated with disease activity in systemic lupus erythematosus. PLoS One 2018; 13:e0208132. [PMID: 30562343 PMCID: PMC6298676 DOI: 10.1371/journal.pone.0208132] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 11/12/2018] [Indexed: 12/17/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is characterized by abnormalities in B cell and T cell function, but the role of disturbances in the activation status of macrophages (Mϕ) has not been well described in human patients. To address this, gene expression profiles from isolated lymphoid and myeloid populations were analyzed to identify differentially expressed (DE) genes between healthy controls and patients with either inactive or active SLE. While hundreds of DE genes were identified in B and T cells of active SLE patients, there were no DE genes found in B or T cells from patients with inactive SLE compared to healthy controls. In contrast, large numbers of DE genes were found in myeloid cells (MC) from both active and inactive SLE patients. Among the DE genes were several known to play roles in Mϕ activation and polarization, including the M1 genes STAT1 and SOCS3 and the M2 genes STAT3, STAT6, and CD163. M1-associated genes were far more frequent in data sets from active versus inactive SLE patients. To characterize the relationship between Mϕ activation and disease activity in greater detail, weighted gene co-expression network analysis (WGCNA) was used to identify modules of genes associated with clinical activity in SLE patients. Among these were disease activity-correlated modules containing activation signatures of predominantly M1-associated genes. No disease activity-correlated modules were enriched in M2-associated genes. Pathway and upstream regulator analysis of DE genes from both active and inactive SLE MC were cross-referenced with high-scoring hits from the drug discovery Library of Integrated Network-based Cellular Signatures (LINCS) to identify new strategies to treat both stages of SLE. A machine learning approach employing MC gene modules and a generalized linear model was able to predict the disease activity status in unrelated gene expression data sets. In summary, altered MC gene expression is characteristic of both active and inactive SLE. However, disease activity is associated with an alteration in the activation of MC, with a bias toward the M1 proinflammatory phenotype. These data suggest that while hyperactivity of B cells and T cells is associated with active SLE, MC potentially direct flare-ups and remission by altering their activation status toward the M1 state.
Collapse
Affiliation(s)
- Adam C. Labonte
- AMPEL BioSolutions LLC, Charlottesville, Virginia, United States of America
- RILITE Research Institute, Charlottesville, Virginia, United States of America
| | - Brian Kegerreis
- AMPEL BioSolutions LLC, Charlottesville, Virginia, United States of America
- RILITE Research Institute, Charlottesville, Virginia, United States of America
| | - Nicholas S. Geraci
- AMPEL BioSolutions LLC, Charlottesville, Virginia, United States of America
- RILITE Research Institute, Charlottesville, Virginia, United States of America
| | - Prathyusha Bachali
- AMPEL BioSolutions LLC, Charlottesville, Virginia, United States of America
| | - Sushma Madamanchi
- RILITE Research Institute, Charlottesville, Virginia, United States of America
| | - Robert Robl
- RILITE Research Institute, Charlottesville, Virginia, United States of America
| | - Michelle D. Catalina
- AMPEL BioSolutions LLC, Charlottesville, Virginia, United States of America
- RILITE Research Institute, Charlottesville, Virginia, United States of America
| | - Peter E. Lipsky
- AMPEL BioSolutions LLC, Charlottesville, Virginia, United States of America
- RILITE Research Institute, Charlottesville, Virginia, United States of America
| | - Amrie C. Grammer
- AMPEL BioSolutions LLC, Charlottesville, Virginia, United States of America
- RILITE Research Institute, Charlottesville, Virginia, United States of America
- * E-mail:
| |
Collapse
|
22
|
Moguche AO, Musvosvi M, Penn-Nicholson A, Plumlee CR, Mearns H, Geldenhuys H, Smit E, Abrahams D, Rozot V, Dintwe O, Hoff ST, Kromann I, Ruhwald M, Bang P, Larson RP, Shafiani S, Ma S, Sherman DR, Sette A, Lindestam Arlehamn CS, McKinney DM, Maecker H, Hanekom WA, Hatherill M, Andersen P, Scriba TJ, Urdahl KB. Antigen Availability Shapes T Cell Differentiation and Function during Tuberculosis. Cell Host Microbe 2018; 21:695-706.e5. [PMID: 28618268 DOI: 10.1016/j.chom.2017.05.012] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 04/03/2017] [Accepted: 05/30/2017] [Indexed: 01/20/2023]
Abstract
CD4 T cells are critical for protective immunity against Mycobacterium tuberculosis (Mtb), the cause of tuberculosis (TB). Yet to date, TB vaccine candidates that boost antigen-specific CD4 T cells have conferred little or no protection. Here we examined CD4 T cell responses to two leading TB vaccine antigens, ESAT-6 and Ag85B, in Mtb-infected mice and in vaccinated humans with and without underlying Mtb infection. In both species, Mtb infection drove ESAT-6-specific T cells to be more differentiated than Ag85B-specific T cells. The ability of each T cell population to control Mtb in the lungs of mice was restricted for opposite reasons: Ag85B-specific T cells were limited by reduced antigen expression during persistent infection, whereas ESAT-6-specific T cells became functionally exhausted due to chronic antigenic stimulation. Our findings suggest that different vaccination strategies will be required to optimize protection mediated by T cells recognizing antigens expressed at distinct stages of Mtb infection.
Collapse
Affiliation(s)
- Albanus O Moguche
- Center for Infectious Disease Research (CIDR), Seattle, WA 98109, USA; Department of Immunology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Munyaradzi Musvosvi
- South African Tuberculosis Vaccine Initiative (SATVI), University of Cape Town, Cape Town 7925, South Africa; Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town 7925, South Africa; Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | - Adam Penn-Nicholson
- South African Tuberculosis Vaccine Initiative (SATVI), University of Cape Town, Cape Town 7925, South Africa; Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town 7925, South Africa; Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | | | - Helen Mearns
- South African Tuberculosis Vaccine Initiative (SATVI), University of Cape Town, Cape Town 7925, South Africa; Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town 7925, South Africa; Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | - Hennie Geldenhuys
- South African Tuberculosis Vaccine Initiative (SATVI), University of Cape Town, Cape Town 7925, South Africa; Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town 7925, South Africa; Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | - Erica Smit
- South African Tuberculosis Vaccine Initiative (SATVI), University of Cape Town, Cape Town 7925, South Africa; Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town 7925, South Africa; Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | - Deborah Abrahams
- South African Tuberculosis Vaccine Initiative (SATVI), University of Cape Town, Cape Town 7925, South Africa; Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town 7925, South Africa; Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | - Virginie Rozot
- South African Tuberculosis Vaccine Initiative (SATVI), University of Cape Town, Cape Town 7925, South Africa; Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town 7925, South Africa; Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | - One Dintwe
- South African Tuberculosis Vaccine Initiative (SATVI), University of Cape Town, Cape Town 7925, South Africa; Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town 7925, South Africa; Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | - Søren T Hoff
- Statens Serum Institut (SSI), 2300 Copenhagen, Denmark
| | | | | | - Peter Bang
- Statens Serum Institut (SSI), 2300 Copenhagen, Denmark
| | - Ryan P Larson
- Center for Infectious Disease Research (CIDR), Seattle, WA 98109, USA
| | - Shahin Shafiani
- Center for Infectious Disease Research (CIDR), Seattle, WA 98109, USA
| | - Shuyi Ma
- Center for Infectious Disease Research (CIDR), Seattle, WA 98109, USA
| | - David R Sherman
- Center for Infectious Disease Research (CIDR), Seattle, WA 98109, USA
| | - Alessandro Sette
- Department of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla 92037, USA
| | | | - Denise M McKinney
- Department of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla 92037, USA
| | - Holden Maecker
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Willem A Hanekom
- South African Tuberculosis Vaccine Initiative (SATVI), University of Cape Town, Cape Town 7925, South Africa; Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town 7925, South Africa; Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | - Mark Hatherill
- South African Tuberculosis Vaccine Initiative (SATVI), University of Cape Town, Cape Town 7925, South Africa; Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town 7925, South Africa; Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | | | - Thomas J Scriba
- South African Tuberculosis Vaccine Initiative (SATVI), University of Cape Town, Cape Town 7925, South Africa; Institute of Infectious Disease and Molecular Medicine (IDM), University of Cape Town, Cape Town 7925, South Africa; Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa.
| | - Kevin B Urdahl
- Center for Infectious Disease Research (CIDR), Seattle, WA 98109, USA; Department of Immunology, University of Washington School of Medicine, Seattle, WA 98195, USA.
| |
Collapse
|
23
|
Yang JD, Mott D, Sutiwisesak R, Lu YJ, Raso F, Stowell B, Babunovic GH, Lee J, Carpenter SM, Way SS, Fortune SM, Behar SM. Mycobacterium tuberculosis-specific CD4+ and CD8+ T cells differ in their capacity to recognize infected macrophages. PLoS Pathog 2018; 14:e1007060. [PMID: 29782535 PMCID: PMC6013218 DOI: 10.1371/journal.ppat.1007060] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 06/01/2018] [Accepted: 04/27/2018] [Indexed: 11/19/2022] Open
Abstract
Containment of Mycobacterium tuberculosis (Mtb) infection requires T cell recognition of infected macrophages. Mtb has evolved to tolerate, evade, and subvert host immunity. Despite a vigorous and sustained CD8+ T cell response during Mtb infection, CD8+ T cells make limited contribution to protection. Here, we ask whether the ability of Mtb-specific T cells to restrict Mtb growth is related to their capacity to recognize Mtb-infected macrophages. We derived CD8+ T cell lines that recognized the Mtb immunodominant epitope TB10.44-11 and compared them to CD4+ T cell lines that recognized Ag85b240-254 or ESAT63-17. While the CD4+ T cells recognized Mtb-infected macrophages and inhibited Mtb growth in vitro, the TB10.4-specific CD8+ T cells neither recognized Mtb-infected macrophages nor restricted Mtb growth. TB10.4-specific CD8+ T cells recognized macrophages infected with Listeria monocytogenes expressing TB10.4. However, over-expression of TB10.4 in Mtb did not confer recognition by TB10.4-specific CD8+ T cells. CD8+ T cells recognized macrophages pulsed with irradiated Mtb, indicating that macrophages can efficiently cross-present the TB10.4 protein and raising the possibility that viable bacilli might suppress cross-presentation. Importantly, polyclonal CD8+ T cells specific for Mtb antigens other than TB10.4 recognized Mtb-infected macrophages in a MHC-restricted manner. As TB10.4 elicits a dominant CD8+ T cell response that poorly recognizes Mtb-infected macrophages, we propose that TB10.4 acts as a decoy antigen. Moreover, it appears that this response overshadows subdominant CD8+ T cell response that can recognize Mtb-infected macrophages. The ability of Mtb to subvert the CD8+ T cell response may explain why CD8+ T cells make a disproportionately small contribution to host defense compared to CD4+ T cells. The selection of Mtb antigens for vaccines has focused on antigens that generate immunodominant responses. We propose that establishing whether vaccine-elicited, Mtb-specific T cells recognize Mtb-infected macrophages could be a useful criterion for preclinical vaccine development.
Collapse
Affiliation(s)
- Jason D. Yang
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Daniel Mott
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Rujapak Sutiwisesak
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Yu-Jung Lu
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Fiona Raso
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Britni Stowell
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Greg Hunter Babunovic
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Jinhee Lee
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Steve M. Carpenter
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Sing Sing Way
- Division of Infectious Disease, Cincinnati Children’s Hospital, Cincinnati, Ohio, United States of America
| | - Sarah M. Fortune
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Samuel M. Behar
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| |
Collapse
|
24
|
Lindenstrøm T, Moguche A, Damborg M, Agger EM, Urdahl K, Andersen P. T Cells Primed by Live Mycobacteria Versus a Tuberculosis Subunit Vaccine Exhibit Distinct Functional Properties. EBioMedicine 2017; 27:27-39. [PMID: 29249639 PMCID: PMC5828549 DOI: 10.1016/j.ebiom.2017.12.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 11/22/2017] [Accepted: 12/05/2017] [Indexed: 01/10/2023] Open
Abstract
Despite inducing strong T cell responses, Mycobacterium tuberculosis (Mtb) infection fails to elicit protective immune memory. As such latently infected or successfully treated Tuberculosis (TB) patients are not protected against recurrent disease. Here, using a mouse model of aerosol Mtb infection, we show that memory immunity to H56/CAF01 subunit vaccination conferred sustained protection in contrast to the transient natural immunity conferred by Mtb infection. Loss of protection to re-infection in natural Mtb memory was temporally linked to an accelerated differentiation of ESAT-6- and to a lesser extent, Ag85B-specific CD4 T cells in both the lung parenchyma and vasculature. This phenotype was characterized by high KLRG1 expression and low, dual production of IFN-γ and TNF. In contrast, H56/CAF01 vaccination elicited cells that expressed low levels of KLRG1 with copious expression of IL-2 and IL-17A. Co-adoptive transfer studies revealed that H56/CAF01 induced memory CD4 T cells efficiently homed into the lung parenchyma of mice chronically infected with Mtb. In comparison, natural Mtb infection- and BCG vaccine-induced memory CD4 T cells exhibited a poor ability to home into the lung parenchyma. These studies suggest that impaired lung migratory capacity is an inherent trait of the terminally differentiated memory responses primed by mycobacteria/mycobacterial vectors. Differentiation state of M. tuberculosis (Mtb)-specific CD4 memory T cells differ depending on their initial priming Live mycobacteria prime fully differentiated CD4 memory T cells with lower lung homing capacity than subunit vaccination Lung parenchymal Mtb memory CD4 T cells produce fewer & less cytokines, express more KLRG1 and cannot sustain protection
People latently infected with M. tuberculosis or successfully treated for Tuberculosis are not protected against recurrent disease, even in the presence of strong T cell responses. Here, using a well-established mouse model, we show that in contrast to subunit vaccination, live mycobacteria prime CD4 T cells that are highly differentiated, have an inferior lung homing capacity and show impaired function once in the parenchyma leading to lack of sustained protection against challenge. This indicates a central shortcoming of natural immunity that needs to be addressed in order to develop improved vaccines against TB.
Collapse
Affiliation(s)
- Thomas Lindenstrøm
- Department of Infectious Disease Immunology, Statens Serum Institut, Denmark.
| | | | - Mie Damborg
- Department of Infectious Disease Immunology, Statens Serum Institut, Denmark
| | - Else Marie Agger
- Department of Infectious Disease Immunology, Statens Serum Institut, Denmark
| | - Kevin Urdahl
- Center for Infectious Disease Research, Seattle, USA
| | - Peter Andersen
- Department of Infectious Disease Immunology, Statens Serum Institut, Denmark
| |
Collapse
|
25
|
Role of Granulocyte-Macrophage Colony-Stimulating Factor Production by T Cells during Mycobacterium tuberculosis Infection. mBio 2017; 8:mBio.01514-17. [PMID: 29066547 PMCID: PMC5654932 DOI: 10.1128/mbio.01514-17] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Mice deficient for granulocyte-macrophage colony-stimulating factor (GM-CSF−/−) are highly susceptible to infection with Mycobacterium tuberculosis, and clinical data have shown that anti-GM-CSF neutralizing antibodies can lead to increased susceptibility to tuberculosis in otherwise healthy people. GM-CSF activates human and murine macrophages to inhibit intracellular M. tuberculosis growth. We have previously shown that GM-CSF produced by iNKT cells inhibits growth of M. tuberculosis. However, the more general role of T cell-derived GM-CSF during infection has not been defined and how GM-CSF activates macrophages to inhibit bacterial growth is unknown. Here we demonstrate that, in addition to nonconventional T cells, conventional T cells also produce GM-CSF during M. tuberculosis infection. Early during infection, nonconventional iNKT cells and γδ T cells are the main source of GM-CSF, a role subsequently assumed by conventional CD4+ T cells as the infection progresses. M. tuberculosis-specific T cells producing GM-CSF are also detected in the peripheral blood of infected people. Under conditions where nonhematopoietic production of GM-CSF is deficient, T cell production of GM-CSF is protective and required for control of M. tuberculosis infection. However, GM-CSF is not required for T cell-mediated protection in settings where GM-CSF is produced by other cell types. Finally, using an in vitro macrophage infection model, we demonstrate that GM-CSF inhibition of M. tuberculosis growth requires the expression of peroxisome proliferator-activated receptor gamma (PPARγ). Thus, we identified GM-CSF production as a novel T cell effector function. These findings suggest that a strategy augmenting T cell production of GM-CSF could enhance host resistance against M. tuberculosis. Mycobacterium tuberculosis is the bacterium that causes tuberculosis, the leading cause of death by any infection worldwide. T cells are critical components of the immune response to Mycobacterium tuberculosis. While gamma interferon (IFN-γ) is a key effector function of T cells during infection, a failed phase IIb clinical trial and other studies have revealed that IFN-γ production alone is not sufficient to control M. tuberculosis. In this study, we demonstrate that CD4+, CD8+, and nonconventional T cells produce GM-CSF during Mycobacterium tuberculosis infection in mice and in the peripheral blood of infected humans. Under conditions where other sources of GM-CSF are absent, T cell production of GM-CSF is protective and is required for control of infection. GM-CSF activation of macrophages to limit bacterial growth requires host expression of the transcription factor PPARγ. The identification of GM-CSF production as a T cell effector function may inform future host-directed therapy or vaccine designs.
Collapse
|
26
|
Abstract
Infection with Mycobacterium tuberculosis, the causative agent of tuberculosis (TB), results in a range of clinical presentations in humans. Most infections manifest as a clinically asymptomatic, contained state that is termed latent TB infection (LTBI); a smaller subset of infected individuals present with symptomatic, active TB. Within these two seemingly binary states, there is a spectrum of host outcomes that have varying symptoms, microbiologies, immune responses and pathologies. Recently, it has become apparent that there is diversity of infection even within a single individual. A good understanding of the heterogeneity that is intrinsic to TB - at both the population level and the individual level - is crucial to inform the development of intervention strategies that account for and target the unique, complex and independent nature of the local host-pathogen interactions that occur in this infection. In this Review, we draw on model systems and human data to discuss multiple facets of TB biology and their relationship to the overall heterogeneity observed in the human disease.
Collapse
|
27
|
Abstract
This article describes the nature of the host response to Mycobacterium tuberculosis in the mouse and guinea pig models of infection. It describes the great wealth of information obtained from the mouse model, reflecting the general availability of immunological reagents, as well as genetic manipulations of the mouse strains themselves. This has led to a good understanding of the nature of the T-cell response to the infection, as well as an appreciation of the complexity of the response involving multiple cytokine- and chemokine-mediated systems. As described here and elsewhere, we have a growing understanding of how multiple CD4-positive T-cell subsets are involved, including regulatory T cells, TH17 cells, as well as the subsequent emergence of effector and central memory T-cell subsets. While, in contrast, our understanding of the host response in the guinea pig model is less advanced, considerable strides have been made in the past decade in terms of defining the basis of the immune response, as well as a better understanding of the immunopathologic process. This model has long been the gold standard for vaccine testing, and more recently is being revisited as a model for testing new drug regimens (bedaquiline being the latest example).
Collapse
|
28
|
Boggiano C, Eichelberg K, Ramachandra L, Shea J, Ramakrishnan L, Behar S, Ernst JD, Porcelli SA, Maeurer M, Kornfeld H. "The Impact of Mycobacterium tuberculosis Immune Evasion on Protective Immunity: Implications for TB Vaccine Design" - Meeting report. Vaccine 2017; 35:3433-3440. [PMID: 28476627 PMCID: PMC5718043 DOI: 10.1016/j.vaccine.2017.04.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 03/18/2017] [Accepted: 04/04/2017] [Indexed: 12/26/2022]
Abstract
Tuberculosis (TB) is the major cause of death from infectious diseases around the world, particularly in HIV infected individuals. TB vaccine design and development have been focused on improving Bacille Calmette-Guérin (BCG) and evaluating recombinant and viral vector expressed Mycobacterium tuberculosis (Mtb) proteins, for boosting BCG-primed immunity, but these approaches have not yet yielded significant improvements over the modest effects of BCG in protecting against infection or disease. On March 7-8, 2016, the National Institute of Allergy and Infectious Diseases (NIAID) convened a workshop on "The Impact of Mtb Immune Evasion on Protective Immunity: Implications for TB Vaccine Design" with the goal of defining immune mechanisms that could be targeted through novel research approaches, to inform vaccine design and immune therapeutic interventions for prevention of TB. The workshop addressed early infection events, the impact of Mtb evolution on the development and maintenance of an adaptive immune response, and the factors that influence protection against and progression to active disease. Scientific gaps and areas of study to revitalize and accelerate TB vaccine design were discussed and prioritized. These included a comprehensive evaluation of innate and Mtb-specific adaptive immune responses in the lung at different stages of disease; determining the role of B cells and antibodies (Abs) during Mtb infection; development of better assays to measure Mtb burden following exposure, infection, during latency and after treatment, and approaches to improving current animal models to study Mtb immunogenicity, TB disease and transmission.
Collapse
Affiliation(s)
- Cesar Boggiano
- Division of AIDS/NIAID/NIH, 5601 Fishers Lane, Rm: 9D10B, MSC: 9829, Rockville, MD 20852, USA.
| | - Katrin Eichelberg
- Division of Microbiology and Infectious Diseases/NIAID/NIH, Rockville, MD, USA
| | - Lakshmi Ramachandra
- Division of Allergy, Immunology and Transplantation/NIAID/NIH, Rockville, MD, USA
| | | | | | - Samuel Behar
- University of Massachusetts Medical School, Worcester, MA, USA
| | - Joel D Ernst
- New York University School of Medicine, New York, NY, USA
| | | | | | - Hardy Kornfeld
- University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|
29
|
Aberrant plasma IL-7 and soluble IL-7 receptor levels indicate impaired T-cell response to IL-7 in human tuberculosis. PLoS Pathog 2017; 13:e1006425. [PMID: 28582466 PMCID: PMC5472333 DOI: 10.1371/journal.ppat.1006425] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Revised: 06/15/2017] [Accepted: 05/22/2017] [Indexed: 12/22/2022] Open
Abstract
T-cell proliferation and generation of protective memory during chronic infections depend on Interleukin-7 (IL-7) availability and receptivity. Regulation of IL-7 receptor (IL-7R) expression and signalling are key for IL-7-modulated T-cell functions. Aberrant expression of soluble (s) and membrane-associated (m) IL-7R molecules is associated with development of autoimmunity and immune failure in acquired immune deficiency syndrome (AIDS) patients. Here we investigated the role of IL-7/IL-7R on T-cell immunity in human tuberculosis. We performed two independent case-control studies comparing tuberculosis patients and healthy contacts. This was combined with follow-up examinations for a subgroup of tuberculosis patients under therapy and recovery. Blood plasma and T cells were characterised for IL-7/sIL-7R and mIL-7R expression, respectively. IL-7-dependent T-cell functions were determined by analysing STAT5 phosphorylation, antigen-specific cytokine release and by analysing markers of T-cell exhaustion and inflammation. Tuberculosis patients had lower soluble IL-7R (p < 0.001) and higher IL-7 (p < 0.001) plasma concentrations as compared to healthy contacts. Both markers were largely independent and aberrant expression normalised during therapy and recovery. Furthermore, tuberculosis patients had lower levels of mIL-7R in T cells caused by post-transcriptional mechanisms. Functional in vitro tests indicated diminished IL-7-induced STAT5 phosphorylation and impaired IL-7-promoted cytokine release of Mycobacterium tuberculosis-specific CD4+ T cells from tuberculosis patients. Finally, we determined T-cell exhaustion markers PD-1 and SOCS3 and detected increased SOCS3 expression during therapy. Only moderate correlation of PD-1 and SOCS3 with IL-7 expression was observed. We conclude that diminished soluble IL-7R and increased IL-7 plasma concentrations, as well as decreased membrane-associated IL-7R expression in T cells, reflect impaired T-cell sensitivity to IL-7 in tuberculosis patients. These findings show similarities to pathognomonic features of impaired T-cell functions and immune failure described in AIDS patients. IL-7 is important for the development and homeostasis of T cells and promotes antigen-specific T-cell responses. Aberrant expression of plasma IL-7 and soluble IL-7R are found in autoimmune diseases and chronic viral infections. In AIDS patients—especially those who fail to reconstitute T-cell numbers during therapy—impaired IL-7-promoted T-cell functions indicated T-cell exhaustion/senescence. In order to evaluate the potential impact of IL-7 on tuberculosis, we characterised various parameters involved in the IL-7-response of tuberculosis patients and healthy contacts. Despite IL-7 being available at higher plasma levels among tuberculosis patients, the T-cell response to IL-7 was impaired when compared to healthy contacts. Soluble IL-7R levels were aberrantly low in plasma during acute tuberculosis but did not account for impaired IL-7 usage. Chronic inflammation in tuberculosis patients—reflected by increased IL-6 plasma levels—did not account for dysfunctional T-cell responses and analysed T-cell exhaustion markers were only moderately correlated. Our findings demonstrate that availability of IL-7 alone is not sufficient to promote protective T-cell immunity against tuberculosis. We describe aberrant IL-7/soluble IL-7R expression and impaired IL-7-mediated T-cell functions in tuberculosis patients with similarities and differences to described IL-7 dysregulation seen in patients with AIDS.
Collapse
|
30
|
Geadas C, Stoszek SK, Sherman D, Andrade BB, Srinivasan S, Hamilton CD, Ellner J. Advances in basic and translational tuberculosis research. Tuberculosis (Edinb) 2017; 102:55-67. [DOI: 10.1016/j.tube.2016.11.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 11/13/2016] [Accepted: 11/25/2016] [Indexed: 12/16/2022]
|
31
|
Abstract
ABSTRACT
Immunological memory is a central feature of the adaptive immune system and a prerequisite for generating effective vaccines. Understanding long-term memory responses to
Mycobacterium tuberculosis
will thus provide us with valuable insights that can guide us in the search for a novel vaccine against tuberculosis (TB). For many years, triggering CD4 T cells and, in particular, those secreting interferon-γ has been the goal of most TB vaccine research, and numerous data from animals and humans support the key role of this subset in protective immunity. More recently, we have learned that the memory response required for effective control of
M. tuberculosis
is much more complex, probably involving several phenotypically different CD4 T cell subsets as well as other cell types that are yet to be defined. Herein, we describe recent insights into memory immunity to TB in the context of both animal models and the human infection. With the increasing amount of data generated from clinical testing of novel TB vaccines, we also summarize recent knowledge of vaccine-induced memory immunity.
Collapse
|
32
|
Abstract
Peptide-specific conventional T cells have been major targets for designing most antimycobacterial vaccines. Immune responses mediated by conventional T cells exhibit a delayed onset upon primary infection and are highly variable in different human populations. In contrast, innate-like T cells quickly respond to pathogens and display effector functions without undergoing extensive clonal expansion. Specifically, the activation of innate-like T cells depends on the promiscuous interaction of highly conserved antigen-presenting molecules, non-peptidic antigens, and likely semi-invariant T cell receptors. In antimicrobial immune responses, mucosal-associated invariant T cells are activated by riboflavin precursor metabolites presented by major histocompatibility complex-related protein I, while lipid-specific T cells including natural killer T cells are activated by lipid metabolites presented by CD1 proteins. Multiple innate-like T cell subsets have been shown to be protective or responsive in mycobacterial infections. Through rapid cytokine secretion, innate-like T cells function in early defense and memory response, offering novel advantages over conventional T cells in the design of anti-tuberculosis strategies.
Collapse
Affiliation(s)
- Shouxiong Huang
- Department of Environmental Health, University of Cincinnati College of Medicine , Cincinnati, OH , USA
| |
Collapse
|
33
|
Coppola M, van Meijgaarden KE, Franken KLMC, Commandeur S, Dolganov G, Kramnik I, Schoolnik GK, Comas I, Lund O, Prins C, van den Eeden SJF, Korsvold GE, Oftung F, Geluk A, Ottenhoff THM. New Genome-Wide Algorithm Identifies Novel In-Vivo Expressed Mycobacterium Tuberculosis Antigens Inducing Human T-Cell Responses with Classical and Unconventional Cytokine Profiles. Sci Rep 2016; 6:37793. [PMID: 27892960 PMCID: PMC5125271 DOI: 10.1038/srep37793] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 11/03/2016] [Indexed: 12/16/2022] Open
Abstract
New strategies are needed to develop better tools to control TB, including identification of novel antigens for vaccination. Such Mtb antigens must be expressed during Mtb infection in the major target organ, the lung, and must be capable of eliciting human immune responses. Using genome-wide transcriptomics of Mtb infected lungs we developed data sets and methods to identify IVE-TB (in-vivo expressed Mtb) antigens expressed in the lung. Quantitative expression analysis of 2,068 Mtb genes from the predicted first operons identified the most upregulated IVE-TB genes during in-vivo pulmonary infection. By further analysing high-level conservation among whole-genome sequenced Mtb-complex strains (n = 219) and algorithms predicting HLA-class-Ia and II presented epitopes, we selected the most promising IVE-TB candidate antigens. Several of these were recognized by T-cells from in-vitro Mtb-PPD and ESAT6/CFP10-positive donors by proliferation and multi-cytokine production. This was validated in an independent cohort of latently Mtb-infected individuals. Significant T-cell responses were observed in the absence of IFN-γ-production. Collectively, the results underscore the power of our novel antigen discovery approach in identifying Mtb antigens, including those that induce unconventional T-cell responses, which may provide important novel tools for TB vaccination and biomarker profiling. Our generic approach is applicable to other infectious diseases.
Collapse
Affiliation(s)
- Mariateresa Coppola
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Kees L M C Franken
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Susanna Commandeur
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Gregory Dolganov
- Department Microbiology Immunology, Stanford Univ. School of Medicine, Stanford, USA
| | - Igor Kramnik
- Department Immunology Infectious Diseases, Harvard School of Public Health, Boston, USA
| | - Gary K Schoolnik
- Department Microbiology Immunology, Stanford Univ. School of Medicine, Stanford, USA
| | - Inaki Comas
- Institute of Biomedicine of Valencia (IBV-CSIC), Valencia, Spain.,CIBER in Epidemiology and Public Health, Madrid, Spain
| | - Ole Lund
- Dept. Systems Biology, Technical Univ., Denmark
| | - Corine Prins
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Susan J F van den Eeden
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Gro E Korsvold
- Department of Infectious Disease Immunology, Domain for Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Fredrik Oftung
- Department of Infectious Disease Immunology, Domain for Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Annemieke Geluk
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
34
|
Recombinant human lactoferrin modulates human PBMC derived macrophage responses to BCG and LPS. Tuberculosis (Edinb) 2016; 101S:S53-S62. [PMID: 27727130 DOI: 10.1016/j.tube.2016.09.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Lactoferrin, an iron-binding glycoprotein found in mammalian mucosal secretions and granules of neutrophils, possesses several immune modulatory properties. Published reports indicate that lactoferrin enhances the efficacy of the tuberculosis vaccine, BCG (Bacillus Calmette Guerin), both by increasing macrophage and dendritic cell ability to stimulate receptive T cells and by modulating the inflammatory response. This report is the first to demonstrate the effects of a recombinant human lactoferrin (10 μg/mL) on human PBMC derived CD14+ and CD16+ macrophages stimulated with a strong (LPS, 10 ng/mL) or weaker (BCG, MOI 1:1) stimulator of inflammation. After 3 days culture, LPS and human lactoferrin treated CD14+ cells significantly increased production of IL-10, IL-6, and MCP-1 compared to the LPS only group. In contrast, similarly treated CD16+ macrophages increased production of IL-12p40 and IL-10 and decreased TNF-α. Limited changes were observed in BCG stimulated CD14+ and CD16+ macrophages with and without lactoferrin. Analysis of surface expression of antigen presentation and co-stimulatory molecules demonstrated that CD14+ macrophages, when stimulated with BCG or LPS and cultured with lactoferrin, increased expression of CD86. CD16+ macrophages treated with lactoferrin showed a similar trend of increase in CD86 expression, but only when stimulated with BCG.
Collapse
|
35
|
Neonatal Fc Receptor Regulation of Lung Immunoglobulin and CD103+ Dendritic Cells Confers Transient Susceptibility to Tuberculosis. Infect Immun 2016; 84:2914-21. [PMID: 27481246 PMCID: PMC5038074 DOI: 10.1128/iai.00533-16] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 07/19/2016] [Indexed: 12/24/2022] Open
Abstract
The neonatal Fc receptor (FcRn) extends the systemic half-life of IgG antibodies by chaperoning bound Fc away from lysosomal degradation inside stromal and hematopoietic cells. FcRn also transports IgG across mucosal barriers into the lumen, and yet little is known about how FcRn modulates immunity in the lung during homeostasis or infection. We infected wild-type (WT) and FcRn-deficient (fcgrt−/−) mice with Pseudomonas aeruginosa or Mycobacterium tuberculosis to investigate whether recycling and transport of IgG via FcRn influences innate and adaptive immunity in the lung in response to bacterial infection. We found that FcRn expression maintains homeostatic IgG levels in lung and leads to preferential secretion of low-affinity IgG ligands into the lumen. Fcgrt−/− animals exhibited no evidence of developmental impairment of innate immunity in the lung and were able to efficiently recruit neutrophils in a model of acute bacterial pneumonia. Although local humoral immunity in lung increased independently of the presence of FcRn during tuberculosis, there was nonetheless a strong impact of FcRn deficiency on local adaptive immunity. We show that the quantity and quality of IgG in airways, as well as the abundance of dendritic cells in the lung, are maintained by FcRn. FcRn ablation transiently enhanced local T cell immunity and neutrophil recruitment during tuberculosis, leading to a lower bacterial burden in lung. This novel understanding of tissue-specific modulation of mucosal IgG isotypes in the lung by FcRn sheds light on the role of mucosal IgG in immune responses in the lung during homeostasis and bacterial disease.
Collapse
|
36
|
Jiang J, Chen X, An H, Yang B, Zhang F, Cheng X. Enhanced immune response of MAIT cells in tuberculous pleural effusions depends on cytokine signaling. Sci Rep 2016; 6:32320. [PMID: 27586092 PMCID: PMC5009363 DOI: 10.1038/srep32320] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 08/02/2016] [Indexed: 12/19/2022] Open
Abstract
The functions of MAIT cells at the site of Mycobacterium tuberculosis infection in humans are still largely unknown. In this study, the phenotypes and immune response of MAIT cells from tuberculous pleural effusions and peripheral blood were investigated. MAIT cells in tuberculous pleural effusions had greatly enhanced IFN-γ, IL-17F and granzyme B response compared with those in peripheral blood. The level of IFN-γ response in MAIT cells from tuberculous pleural effusions was inversely correlated with the extent of tuberculosis infection (p = 0.0006). To determine whether cytokines drive the immune responses of MAIT cells at the site of tuberculosis infection, the role of IL-1β, IL-2, IL-7, IL-12, IL-15 and IL-18 was investigated. Blockade of IL-2, IL-12 or IL-18 led to significantly reduced production of IFN-γ and/or granzyme B in MAIT cells from tuberculous pleural effusions. Majority of IL-2-producing cells (94.50%) in tuberculous pleural effusions had phenotype of CD3(+)CD4(+), and most IL-12p40-producing cells (91.39%) were CD14(+) cells. MAIT cells had significantly elevated expression of γc receptor which correlated with enhanced immune responses of MAIT cells. It is concluded that MAIT cells from tuberculous pleural effusions exhibited highly elevated immune response to Mtb antigens, which are controlled by cytokines produced by innate/adaptive immune cells.
Collapse
MESH Headings
- Adult
- Antibodies, Blocking/pharmacology
- Female
- Gene Expression Regulation
- Granzymes/genetics
- Granzymes/immunology
- Humans
- Immunity, Cellular
- Interferon-gamma/genetics
- Interferon-gamma/immunology
- Interleukin-12/antagonists & inhibitors
- Interleukin-12/genetics
- Interleukin-12/immunology
- Interleukin-12 Subunit p40/genetics
- Interleukin-12 Subunit p40/immunology
- Interleukin-17/genetics
- Interleukin-17/immunology
- Interleukin-18/antagonists & inhibitors
- Interleukin-18/genetics
- Interleukin-18/immunology
- Interleukin-2/antagonists & inhibitors
- Interleukin-2/genetics
- Interleukin-2/immunology
- Lipopolysaccharide Receptors/genetics
- Lipopolysaccharide Receptors/immunology
- Male
- Mucosal-Associated Invariant T Cells/immunology
- Mucosal-Associated Invariant T Cells/microbiology
- Mycobacterium tuberculosis/immunology
- Mycobacterium tuberculosis/pathogenicity
- Pleural Effusion/immunology
- Pleural Effusion/microbiology
- Pleural Effusion/pathology
- Primary Cell Culture
- Receptors, Antigen, T-Cell, gamma-delta/genetics
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Signal Transduction/immunology
- Tuberculosis, Pleural/immunology
- Tuberculosis, Pleural/microbiology
- Tuberculosis, Pleural/pathology
Collapse
Affiliation(s)
- Jing Jiang
- Shenzhen Key Laboratory of Infection and Immunity, Shenzhen Third People’s Hospital, Guangdong Medical College, Shenzhen, Guangdong, China
- Division of Research, Institute of Tuberculosis, 309th Hospital, Beijing, China
- Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Xinchun Chen
- Shenzhen Key Laboratory of Infection and Immunity, Shenzhen Third People’s Hospital, Guangdong Medical College, Shenzhen, Guangdong, China
- Department of Pathogen Biology, School of Medicine, Shenzhen University, Shenzhen, Guangdong, China
| | - Hongjuan An
- Division of Research, Institute of Tuberculosis, 309th Hospital, Beijing, China
| | - Bingfen Yang
- Division of Research, Institute of Tuberculosis, 309th Hospital, Beijing, China
| | - Fuping Zhang
- Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Xiaoxing Cheng
- Division of Research, Institute of Tuberculosis, 309th Hospital, Beijing, China
| |
Collapse
|
37
|
Do HLA class II genes protect against pulmonary tuberculosis? A systematic review and meta-analysis. Eur J Clin Microbiol Infect Dis 2016; 35:1567-80. [DOI: 10.1007/s10096-016-2713-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 06/17/2016] [Indexed: 01/25/2023]
|
38
|
Alvarez MG, Bertocchi GL, Cooley G, Albareda MC, Viotti R, Perez-Mazliah DE, Lococo B, Castro Eiro M, Laucella SA, Tarleton RL. Treatment Success in Trypanosoma cruzi Infection Is Predicted by Early Changes in Serially Monitored Parasite-Specific T and B Cell Responses. PLoS Negl Trop Dis 2016; 10:e0004657. [PMID: 27128444 PMCID: PMC4851297 DOI: 10.1371/journal.pntd.0004657] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 03/31/2016] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Chagas disease is the highest impact parasitic disease in Latin America. We have proposed that changes in Trypanosoma cruzi-specific immune responses might serve as surrogate indicators of treatment success. Herein, we addressed in a long-term follow-up study whether cure achieved after treatment can be predicted by changes in non-conventional indexes of anti-parasite serological and T cell activities. METHODOLOGY/PRINCIPAL FINDINGS T. cruzi-specific T cell responses, as measured by interferon-γ ELISPOT and T. cruzi-specific antibodies assessed by ELISA, hemagglutination and immunofluorescence tests as well as by a multiplex assay incorporating 14 recombinant T. cruzi proteins were measured in 33 patients at 48-150 months post-benznidazole treatment. Cure - as assessed by conventional serological tests - was associated with an early decline in T. cruzi-specific IFN-γ-producing T cells and in antibody titers measured by the multiplex serological assay. Changes in the functional status and potential of T. cruzi-specific T cells, indicative of reduced antigen stimulation, provided further evidence of parasitological cure following benznidazole treatment. Patients showing a significant reduction in T. cruzi-specific antibodies had higher pre-therapy levels of T. cruzi-specific IFN-γ- producing T cells compared to those with unaltered humoral responses post-treatment. CONCLUSIONS/SIGNIFICANCE Monitoring of appropriate immunological responses can provide earlier and robust measures of treatment success in T. cruzi infection.
Collapse
Affiliation(s)
- María G. Alvarez
- Hospital Interzonal General de Agudos Eva Perón, Buenos Aires, Argentina
| | | | - Gretchen Cooley
- Center for Tropical and Emerging Global Diseases, Athens, Georgia, United States of America
| | - María C. Albareda
- Instituto Nacional de Parasitología Dr. Mario Fatala Chaben, Buenos Aires, Argentina
| | - Rodolfo Viotti
- Hospital Interzonal General de Agudos Eva Perón, Buenos Aires, Argentina
| | | | - Bruno Lococo
- Hospital Interzonal General de Agudos Eva Perón, Buenos Aires, Argentina
| | - Melisa Castro Eiro
- Instituto Nacional de Parasitología Dr. Mario Fatala Chaben, Buenos Aires, Argentina
| | - Susana A. Laucella
- Hospital Interzonal General de Agudos Eva Perón, Buenos Aires, Argentina
- Instituto Nacional de Parasitología Dr. Mario Fatala Chaben, Buenos Aires, Argentina
| | - Rick L. Tarleton
- Center for Tropical and Emerging Global Diseases, Athens, Georgia, United States of America
- * E-mail:
| |
Collapse
|
39
|
Park AJ, Rendini T, Martiniuk F, Levis WR. Leprosy as a model to understand cancer immunosurveillance and T cell anergy. J Leukoc Biol 2016; 100:47-54. [PMID: 27106673 DOI: 10.1189/jlb.5ru1215-537rr] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 04/05/2016] [Indexed: 12/12/2022] Open
Abstract
Leprosy is a disease caused by Mycobacterium leprae that presents on a spectrum of both clinical manifestations and T cell response. On one end of this spectrum, tuberculoid leprosy is a well-controlled disease, characterized by a cell-mediated immunity and immunosurveillance. On the opposite end of the spectrum, lepromatous leprosy is characterized by M. leprae proliferation and T cell anergy. Similar to progressive tumor cells, M. leprae escapes immunosurveillance in more severe forms of leprosy. The mechanisms by which M. leprae is able to evade the host immune response involve many, including the alterations of lipid droplets, microRNA, and Schwann cells, and involve the regulation of immune regulators, such as the negative checkpoint regulators CTLA-4, programmed death 1, and V-domain Ig suppressor of T cell activation-important targets in today's cancer immunotherapies. The means by which tumor cells become able to escape immunosurveillance through negative checkpoint regulators are evidenced by the successes of treatments, such as nivolumab and ipilimumab. Many parallels can be drawn between the immune responses seen in leprosy and cancer. Therefore, the understanding of how M. leprae encourages immune escape during proliferative disease states has potential to add to our understanding of cancer immunotherapy.
Collapse
Affiliation(s)
- Andrew J Park
- Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - Tina Rendini
- Bellevue Hospital Center, National Hansen's Disease Program, New York, New York, USA; and
| | | | - William R Levis
- Bellevue Hospital Center, National Hansen's Disease Program, New York, New York, USA; and
| |
Collapse
|
40
|
Henao-Tamayo MI, Obregón-Henao A, Arnett K, Shanley CA, Podell B, Orme IM, Ordway DJ. Effect of bacillus Calmette-Guérin vaccination on CD4+Foxp3+ T cells during acquired immune response to Mycobacterium tuberculosis infection. J Leukoc Biol 2016; 99:605-17. [PMID: 26590147 PMCID: PMC4787291 DOI: 10.1189/jlb.4a0614-308rr] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 09/14/2015] [Accepted: 10/27/2015] [Indexed: 12/13/2022] Open
Abstract
Increasing information has shown that many newly emerging strains of Mycobacterium tuberculosis, including the highly prevalent and troublesome Beijing family of strains, can potently induce the emergence of Foxp3(+)CD4 Tregs Although the significance of this is still not fully understood, we have previously provided evidence that the emergence of this population can significantly ablate the protective effect of BCG vaccination, causing progressive fatal disease in the mouse model. However, whether the purpose of this response is to control inflammation or to directly dampen the acquired immune response is still unclear. In the present study, we have shown, using both cell depletion and adoptive transfer strategies, that Tregs can have either properties. Cell depletion resulted in a rapid, but transient, decrease in the lung bacterial load, suggesting release or temporary re-expansion of effector immunity. Transfer of Tregs into Rag2(-/-)or marked congenic mice worsened the disease course and depressed cellular influx of effector T cells into the lungs. Tregs from infected donors seemed to preferentially depress the inflammatory response and granulocytic influx. In contrast, those from BCG-vaccinated and then challenged donors seemed more focused on depression of acquired immunity. These qualitative differences might be related to increasing knowledge reflecting the plasticity of the Treg response.
Collapse
Affiliation(s)
- Marcela I Henao-Tamayo
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins CO, USA
| | - Andres Obregón-Henao
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins CO, USA
| | - Kimberly Arnett
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins CO, USA
| | - Crystal A Shanley
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins CO, USA
| | - Brendan Podell
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins CO, USA
| | - Ian M Orme
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins CO, USA
| | - Diane J Ordway
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins CO, USA
| |
Collapse
|
41
|
Influence of the polymorphism of the DUSP14 gene on the expression of immune-related genes and development of pulmonary tuberculosis. Genes Immun 2016; 17:207-12. [DOI: 10.1038/gene.2016.11] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 01/21/2016] [Accepted: 02/01/2016] [Indexed: 12/27/2022]
|
42
|
Jiang J, Yang B, An H, Wang X, Liu Y, Cao Z, Zhai F, Wang R, Cao Y, Cheng X. Mucosal-associated invariant T cells from patients with tuberculosis exhibit impaired immune response. J Infect 2016; 72:338-352. [PMID: 26724769 DOI: 10.1016/j.jinf.2015.11.010] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Revised: 11/02/2015] [Accepted: 11/19/2015] [Indexed: 12/13/2022]
Abstract
OBJECTIVES To identify factors which regulate MAIT cell response to Mycobacterium tuberculosis antigens, and to investigate the role of MAIT cells in patients with active tuberculosis. METHODS Immune response of MAIT cells to M. tuberculosis antigens were compared between patients with active TB and healthy controls by flow cytometry and RNA sequencing. RESULTS IFN-γ response of MAIT cells to M. tuberculosis lysates was dramatically improved by signal 3 cytokine IL-15 (p = 0.0002). Patients with active TB exhibited highly reduced IFN-γ production in MAIT cells stimulated with M. tuberculosis lysates/IL-15 compared with healthy controls (p < 0.0001) and individuals with latent TB infection (p = 0.0008). RNA sequencing of flow-sorted MAIT cells from patients with TB and healthy controls identified numerous differentially expressed genes, and the expression of genes that encode IFN-γ, TNF-α, IL-17F, granulysin and granzyme B were all down-regulated in patients with TB. MAIT cells from patients with TB has significantly lower expression of γc receptor than those from healthy controls under condition of Mtb lysates/IL-15 stimulation (p = 0.0028). Blockade of both γc and IL-2Rβ receptors resulted in highly reduced frequency of IFN-γ-producing MAIT cells (79.4%) (p = 0.0011). CONCLUSIONS MAIT cells from patients with active TB exhibited impaired cytokine and cytotoxic response to M. tuberculosis antigens.
Collapse
Affiliation(s)
- Jing Jiang
- Key Laboratory of Tuberculosis Prevention and Treatment, and Beijing Key Laboratory of New Techniques for Tuberculosis Diagnosis and Treatment, Division of Research, Institute of Tuberculosis, 309th Hospital, Beijing, China
| | - Bingfen Yang
- Key Laboratory of Tuberculosis Prevention and Treatment, and Beijing Key Laboratory of New Techniques for Tuberculosis Diagnosis and Treatment, Division of Research, Institute of Tuberculosis, 309th Hospital, Beijing, China
| | - Hongjuan An
- Key Laboratory of Tuberculosis Prevention and Treatment, and Beijing Key Laboratory of New Techniques for Tuberculosis Diagnosis and Treatment, Division of Research, Institute of Tuberculosis, 309th Hospital, Beijing, China
| | - Xinjing Wang
- Key Laboratory of Tuberculosis Prevention and Treatment, and Beijing Key Laboratory of New Techniques for Tuberculosis Diagnosis and Treatment, Division of Research, Institute of Tuberculosis, 309th Hospital, Beijing, China
| | - Yanhua Liu
- Key Laboratory of Tuberculosis Prevention and Treatment, and Beijing Key Laboratory of New Techniques for Tuberculosis Diagnosis and Treatment, Division of Research, Institute of Tuberculosis, 309th Hospital, Beijing, China
| | - Zhihong Cao
- Key Laboratory of Tuberculosis Prevention and Treatment, and Beijing Key Laboratory of New Techniques for Tuberculosis Diagnosis and Treatment, Division of Research, Institute of Tuberculosis, 309th Hospital, Beijing, China
| | - Fei Zhai
- Key Laboratory of Tuberculosis Prevention and Treatment, and Beijing Key Laboratory of New Techniques for Tuberculosis Diagnosis and Treatment, Division of Research, Institute of Tuberculosis, 309th Hospital, Beijing, China
| | - Ruo Wang
- Key Laboratory of Tuberculosis Prevention and Treatment, and Beijing Key Laboratory of New Techniques for Tuberculosis Diagnosis and Treatment, Division of Research, Institute of Tuberculosis, 309th Hospital, Beijing, China
| | - Yan Cao
- Key Laboratory of Tuberculosis Prevention and Treatment, and Beijing Key Laboratory of New Techniques for Tuberculosis Diagnosis and Treatment, Division of Research, Institute of Tuberculosis, 309th Hospital, Beijing, China
| | - Xiaoxing Cheng
- Key Laboratory of Tuberculosis Prevention and Treatment, and Beijing Key Laboratory of New Techniques for Tuberculosis Diagnosis and Treatment, Division of Research, Institute of Tuberculosis, 309th Hospital, Beijing, China.
| |
Collapse
|
43
|
Subbian S, Tsenova L, Holloway J, Peixoto B, O'Brien P, Dartois V, Khetani V, Zeldis JB, Kaplan G. Adjunctive Phosphodiesterase-4 Inhibitor Therapy Improves Antibiotic Response to Pulmonary Tuberculosis in a Rabbit Model. EBioMedicine 2016; 4:104-14. [PMID: 26981575 PMCID: PMC4776074 DOI: 10.1016/j.ebiom.2016.01.015] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 01/06/2016] [Accepted: 01/11/2016] [Indexed: 12/14/2022] Open
Abstract
Objectives Adjunctive host-directed therapy is emerging as a new potential approach to improve the outcome of conventional antimicrobial treatment for tuberculosis (TB). We tested the ability of a phosphodiesterase-4 inhibitor (PDE4i) CC-11050, co-administered with the first-line anti-TB drug isoniazid (INH), to accelerate bacillary killing and reduce chronic inflammation in the lungs of rabbits with experimental Mycobacterium tuberculosis (Mtb) infection. Methods A rabbit model of pulmonary TB that recapitulates the pathologic manifestations seen in humans was used. Rabbits were infected with virulent Mtb by aerosol exposure and treated for eight weeks with INH with or without CC-11050, starting at four weeks post infection. The effect of CC-11050 treatment on disease severity, pathology, bacillary load, T cell proliferation and global lung transcriptome profiles were analyzed. Results Significant improvement in bacillary clearance and reduced lung pathology and fibrosis were noted in the rabbits treated for eight weeks with INH + CC-11050, compared to those treated with INH or CC-11050 only. In addition, expression of host genes associated with tissue remodeling, tumor necrosis factor alpha (TNF-α) regulation, macrophage activation and lung inflammation networks was dampened in CC-11050-treated, compared to the untreated rabbits. Conclusions Adjunctive CC-11050 therapy significantly improves the response of rabbits with experimental pulmonary TB to INH treatment. We propose that CC-11050 may be a promising candidate for host directed therapy of patients with pulmonary TB, reducing the duration and improving clinical outcome of antibiotic treatment. CC-11050 is an anti-inflammatory molecule targeting host phosphodiesterase-4. CC-11050 plus isoniazid therapy significantly reduced bacillary load and pathology in a rabbit model pulmonary tuberculosis. CC-11050 can be a promising candidate for adjunctive host directed therapy of patients with active pulmonary tuberculosis.
In 2013, tuberculosis (TB) caused by Mycobacterium tuberculosis (Mtb) killed 1.5 million people worldwide. Current antibiotic therapy for tuberculosis is ineffective in eliminating the infecting bacilli and/or disease pathology such as lung fibrosis. Therefore, alternate approaches are urgently needed to control the TB epidemic. In this study, using a rabbit model of pulmonary TB, which closely mimics the human disease, we tested the hypothesis that reducing the host inflammatory response during Mtb infection would improve the outcome of antibiotic treatment; we show that adjunctive phosphodiesterase-4 inhibition therapy with isoniazid improves bacterial clearance and lung pathology.
Collapse
Affiliation(s)
- Selvakumar Subbian
- Public Health Research Institute (PHRI) of Rutgers Biomedical and Health Sciences (RBHS), Newark, NJ, USA
| | - Liana Tsenova
- Public Health Research Institute (PHRI) of Rutgers Biomedical and Health Sciences (RBHS), Newark, NJ, USA; Department of Biological Sciences, NYC College of Technology, Brooklyn, NY, USA
| | - Jennifer Holloway
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA
| | - Blas Peixoto
- Public Health Research Institute (PHRI) of Rutgers Biomedical and Health Sciences (RBHS), Newark, NJ, USA
| | - Paul O'Brien
- Public Health Research Institute (PHRI) of Rutgers Biomedical and Health Sciences (RBHS), Newark, NJ, USA
| | - Véronique Dartois
- Public Health Research Institute (PHRI) of Rutgers Biomedical and Health Sciences (RBHS), Newark, NJ, USA
| | | | | | - Gilla Kaplan
- Public Health Research Institute (PHRI) of Rutgers Biomedical and Health Sciences (RBHS), Newark, NJ, USA
| |
Collapse
|
44
|
Booty MG, Nunes-Alves C, Carpenter SM, Jayaraman P, Behar SM. Multiple Inflammatory Cytokines Converge To Regulate CD8+ T Cell Expansion and Function during Tuberculosis. THE JOURNAL OF IMMUNOLOGY 2016; 196:1822-31. [PMID: 26755819 DOI: 10.4049/jimmunol.1502206] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 12/10/2015] [Indexed: 11/19/2022]
Abstract
The differentiation of effector CD8(+) T cells is a dynamically regulated process that varies during different infections and is influenced by the inflammatory milieu of the host. In this study, we define three signals regulating CD8(+) T cell responses during tuberculosis by focusing on cytokines known to affect disease outcome: IL-12, type I IFN, and IL-27. Using mixed bone marrow chimeras, we compared wild-type and cytokine receptor knockout CD8(+) T cells within the same mouse following aerosol infection with Mycobacterium tuberculosis. Four weeks postinfection, IL-12, type 1 IFN, and IL-27 were all required for efficient CD8(+) T cell expansion in the lungs. We next determined if these cytokines directly promote CD8(+) T cell priming or are required only for expansion in the lungs. Using retrogenic CD8(+) T cells specific for the M. tuberculosis Ag TB10.4 (EsxH), we observed that IL-12 is the dominant cytokine driving both CD8(+) T cell priming in the lymph node and expansion in the lungs; however, type I IFN and IL-27 have nonredundant roles supporting pulmonary CD8(+) T cell expansion. Thus, IL-12 is a major signal promoting priming in the lymph node, but a multitude of inflammatory signals converge in the lung to promote continued expansion. Furthermore, these cytokines regulate the differentiation and function of CD8(+) T cells during tuberculosis. These data demonstrate distinct and overlapping roles for each of the cytokines examined and underscore the complexity of CD8(+) T cell regulation during tuberculosis.
Collapse
Affiliation(s)
- Matthew G Booty
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01655; and Program in Immunology, Division of Medical Sciences, Harvard Medical School, Boston, MA 02115
| | - Cláudio Nunes-Alves
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01655; and
| | - Stephen M Carpenter
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01655; and
| | - Pushpa Jayaraman
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01655; and
| | - Samuel M Behar
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01655; and
| |
Collapse
|
45
|
Achkar JM, Chan J, Casadevall A. B cells and antibodies in the defense against Mycobacterium tuberculosis infection. Immunol Rev 2015; 264:167-81. [PMID: 25703559 DOI: 10.1111/imr.12276] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Better understanding of the immunological components and their interactions necessary to prevent or control Mycobacterium tuberculosis (Mtb) infection in humans is critical for tuberculosis (TB) vaccine development strategies. Although the contributory role of humoral immunity in the protection against Mtb infection and disease is less defined than the role of T cells, it has been well-established for many other intracellular pathogens. Here we update and discuss the increasing evidence and the mechanisms of B cells and antibodies in the defense against Mtb infection. We posit that B cells and antibodies have a variety of potential protective roles at each stage of Mtb infection and postulate that such roles should be considered in the development strategies for TB vaccines and other immune-based interventions.
Collapse
|
46
|
Boule LA, Winans B, Lambert K, Vorderstrasse BA, Topham DJ, Pavelka MS, Lawrence BP. Activation of the aryl hydrocarbon receptor during development enhances the pulmonary CD4+ T-cell response to viral infection. Am J Physiol Lung Cell Mol Physiol 2015; 309:L305-13. [PMID: 26071552 DOI: 10.1152/ajplung.00135.2015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 06/03/2015] [Indexed: 12/13/2022] Open
Abstract
Respiratory infections are a threat to health and economies worldwide, yet the basis for striking variation in the severity of infection is not completely understood. Environmental exposures during development are associated with increased severity and incidence of respiratory infection later in life. Many of these exposures include ligands of the aryl hydrocarbon receptor (AHR), a transcription factor expressed by immune and nonimmune cells. In adult animals, AHR activation alters CD4(+) T cells and changes immunopathology. Developmental AHR activation impacts CD4(+) T-cell responses in lymphoid tissues, but whether skewed responses are also present in the infected lung is unknown. To determine whether pulmonary CD4(+) T-cell responses are modified by developmental AHR activation, mice were exposed to the prototypical AHR ligand 2,3,7,8-tetrachlorodibenzo-p-dioxin during development and infected with influenza virus as adults. Lungs of exposed offspring had greater bronchopulmonary inflammation compared with controls, and activated, virus-specific CD4(+) T cells contributed to the infiltrating leukocytes. These effects were CD4(+) T cell subset specific, with increases in T helper type 1 and regulatory T cells, but no change in the frequency of T helper type 17 cells in the infected lung. This is in direct contrast to prior reports of suppressed conventional CD4(+) T-cell responses in the lymph node. Using adoptive transfers and manipulating the pathogen properties, we determined that developmental exposure influenced factors intrinsic and extrinsic to CD4(+) T cells and may involve developmentally induced changes in signals from infected lung epithelial cells. Thus developmental exposures lead to context-dependent changes in pulmonary CD4(+) T-cell subsets, which may contribute to differential responses to respiratory infection.
Collapse
Affiliation(s)
- Lisbeth A Boule
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York
| | - Bethany Winans
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York; and
| | - Kris Lambert
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York
| | - Beth A Vorderstrasse
- Department of Public Health and Preventive Medicine, Oregon Health Sciences University, Portland, Oregon
| | - David J Topham
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York
| | - Martin S Pavelka
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York
| | - B Paige Lawrence
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York; Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York; and
| |
Collapse
|