Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 100
Filter
Add more filters

Publication year range
1.
Proc Natl Acad Sci U S A ; 121(19): e2318003121, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38691588

ABSTRACT

Peptides presented by HLA-E, a molecule with very limited polymorphism, represent attractive targets for T cell receptor (TCR)-based immunotherapies to circumvent the limitations imposed by the high polymorphism of classical HLA genes in the human population. Here, we describe a TCR-based bispecific molecule that potently and selectively binds HLA-E in complex with a peptide encoded by the inhA gene of Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis in humans. We reveal the biophysical and structural bases underpinning the potency and specificity of this molecule and demonstrate its ability to redirect polyclonal T cells to target HLA-E-expressing cells transduced with mycobacterial inhA as well as primary cells infected with virulent Mtb. Additionally, we demonstrate elimination of Mtb-infected cells and reduction of intracellular Mtb growth. Our study suggests an approach to enhance host T cell immunity against Mtb and provides proof of principle for an innovative TCR-based therapeutic strategy overcoming HLA polymorphism and therefore applicable to a broader patient population.


Subject(s)
Histocompatibility Antigens Class I , Mycobacterium tuberculosis , Receptors, Antigen, T-Cell , T-Lymphocytes , Mycobacterium tuberculosis/immunology , Humans , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , T-Lymphocytes/immunology , HLA-E Antigens , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Tuberculosis/immunology
2.
Thorax ; 77(3): 304-311, 2022 03.
Article in English | MEDLINE | ID: mdl-34607905

ABSTRACT

Immune checkpoint inhibitors (ICIs) have revolutionised cancer treatment. However, immune-related adverse events (irAEs) are a common side effect which can mimic infection. Additionally, treatment of irAEs with corticosteroids and other immunosuppressant agents can lead to opportunistic infection, which we have classed as immunotherapy infections due to immunosuppression. However, emerging reports demonstrate that some infections can be precipitated by ICIs in the absence of immunosuppressive treatment, in contrast to the majority of reported cases. These infections are characterised by a dysregulated inflammatory immune response, and so we propose they are described as immunotherapy infections due to dysregulated immunity. This review summarises the rapidly emerging evidence of these phenomena and proposes a new framework for considering infection in the context of cancer immunotherapy.


Subject(s)
Neoplasms , Opportunistic Infections , Humans , Immune Checkpoint Inhibitors , Immunosuppressive Agents/adverse effects , Immunotherapy/adverse effects , Neoplasms/drug therapy , Opportunistic Infections/chemically induced
3.
Cancer Immunol Immunother ; 71(11): 2757-2764, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35429244

ABSTRACT

Recent advancements in cancer immunotherapy using immune checkpoint inhibitors (ICIs) have received considerable attention. Although advantageous, ICI therapies cause unique immune-related adverse events (irAEs) in some patients. Moreover, infectious diseases, such as tuberculosis, have been recognized as emerging concerns during immunotherapy. We aimed to evaluate the interferon-gamma release assay (IGRA) conversion rate and active tuberculosis incidence during immunotherapy to elucidate the incidence of tuberculosis reactivation after ICI therapy induction.We prospectively assessed IGRA results in lung cancer patients who received ICI monotherapy before ICI treatment and at 6 and 12 months after ICI treatment. We also assessed computed tomography findings to determine the presence of active tuberculosis when positive IGRA results were obtained. The ICIs used were nivolumab, pembrolizumab, atezolizumab, and durvalumab.In all, 178 patients were prospectively recruited between March 2017 and March 2020. Of these, 123 completed serial IGRAs, of whom 18, 101, and 4, respectively, had positive, negative, and indeterminate IGRAs at baseline. Three and four patients, respectively, showed IGRA reversion and conversion during immunotherapy. One patient with a sustained, stable positive IGRA and one with IGRA conversion developed active pulmonary tuberculosis during immunotherapy.We found that 3.3% and 1.6% of the patients developed IGRA conversion and active tuberculosis, respectively. Of the four patients who developed IGRA conversion, one developed active pulmonary tuberculosis during immunotherapy. Another patient with sustained, stable positive IGRA developed active tuberculosis. Physicians should be alert to tuberculosis development during ICI therapy, and IGRA testing is a useful tool to assess the risk of developing active tuberculosis.


Subject(s)
Lung Neoplasms , Tuberculosis, Pulmonary , Tuberculosis , Humans , Immune Checkpoint Inhibitors/adverse effects , Interferon-gamma Release Tests/methods , Lung Neoplasms/drug therapy , Nivolumab , Prospective Studies , Tuberculin Test/methods
4.
Eur Respir J ; 60(5)2022 11.
Article in English | MEDLINE | ID: mdl-35595321

ABSTRACT

By attenuating T-cell activation, immune checkpoints (ICs) limit optimal anti-tumour responses and IC inhibition (ICI) has emerged as a new therapy for a broad range of cancers. T-cell responses are indispensable to tuberculosis (TB) immunity in humans. However, boosting T-cell immunity in cancer patients by blocking the programmed cell death 1/programmed cell death ligand 1 (PD-1/PD-L1) axis can trigger re-activation of latent TB. This phenomenon appears to contradict the prevailing thought that enhancing T-cell immunity to Mycobacterium tuberculosis will improve immune control of this pathogen. In support of this anecdotal human data, several murine studies have shown that PD-1 deficiency leads to severe TB disease and rapid death. These observations warrant a serious reconsideration of what constitutes effective TB immunity and how ICs contribute to it. Through restraining T-cell responses, ICs are critical to preventing excessive tissue damage and maintaining a range of effector functions. Bolstering this notion, inhibitory receptors limit pathology in respiratory infections such as influenza, where loss of negative immune regulation resulted in progressive immunopathology. In this review, we analyse the mechanisms of ICs in general and their role in TB in particular. We conclude with a reflection on the emerging paradigm and avenues for future research.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Humans , Mice , Animals , Programmed Cell Death 1 Receptor/metabolism , Immune Checkpoint Inhibitors , Tuberculosis/drug therapy , Lymphocyte Activation
5.
Eur Respir J ; 60(6)2022 12.
Article in English | MEDLINE | ID: mdl-35777774

ABSTRACT

Respiratory diseases account for over 5 million deaths yearly and are a huge burden to healthcare systems worldwide. Murine models have been of paramount importance to decode human lung biology in vivo, but their genetic, anatomical, physiological and immunological differences with humans significantly hamper successful translation of research into clinical practice. Thus, to clearly understand human lung physiology, development, homeostasis and mechanistic dysregulation that may lead to disease, it is essential to develop models that accurately recreate the extraordinary complexity of the human pulmonary architecture and biology. Recent advances in micro-engineering technology and tissue engineering have allowed the development of more sophisticated models intending to bridge the gap between the native lung and its replicates in vitro Alongside advanced culture techniques, remarkable technological growth in downstream analyses has significantly increased the predictive power of human biology-based in vitro models by allowing capture and quantification of complex signals. Refined integrated multi-omics readouts could lead to an acceleration of the translational pipeline from in vitro experimental settings to drug development and clinical testing in the future. This review highlights the range and complexity of state-of-the-art lung models for different areas of the respiratory system, from nasal to large airways, small airways and alveoli, with consideration of various aspects of disease states and their potential applications, including pre-clinical drug testing. We explore how development of optimised physiologically relevant in vitro human lung models could accelerate the identification of novel therapeutics with increased potential to translate successfully from the bench to the patient's bedside.


Subject(s)
Lung , Respiratory Tract Diseases , Humans , Animals , Mice , Lung/physiology , Tissue Engineering/methods
6.
Clin Infect Dis ; 70(9): 1865-1874, 2020 04 15.
Article in English | MEDLINE | ID: mdl-31190065

ABSTRACT

BACKGROUND: Tuberculosis (TB) is the leading cause of mortality and morbidity in people living with human immunodeficiency virus (HIV) infection (PLWH). PLWH with TB disease are at risk of the paradoxical TB-associated immune reconstitution inflammatory syndrome (TB-IRIS) when they commence antiretroviral therapy. However, the pathophysiology is incompletely understood and specific therapy is lacking. We investigated the hypothesis that invariant natural killer T (iNKT) cells contribute to innate immune dysfunction associated with TB-IRIS. METHODS: In a cross-sectional study of 101 PLWH and HIV-uninfected South African patients with active TB and controls, iNKT cells were enumerated using α-galactosylceramide-loaded CD1d tetramers and subsequently functionally characterized by flow cytometry. In a second study of 49 people with HIV type 1 (HIV-1) and active TB commencing antiretroviral therapy, iNKT cells in TB-IRIS patients and non-IRIS controls were compared longitudinally. RESULTS: Circulating iNKT cells were reduced in HIV-1 infection, most significantly the CD4+ subset, which was inversely associated with HIV-1 viral load. iNKT cells in HIV-associated TB had increased surface CD107a expression, indicating cytotoxic degranulation. Relatively increased iNKT cell frequency in patients with HIV-1 infection and active TB was associated with development of TB-IRIS following antiretroviral therapy initiation. iNKT cells in TB-IRIS were CD4+CD8- subset depleted and degranulated around the time of TB-IRIS onset. CONCLUSIONS: Reduced iNKT cell CD4+ subsets as a result of HIV-1 infection may skew iNKT cell functionality toward cytotoxicity. Increased CD4- cytotoxic iNKT cells may contribute to immunopathology in TB-IRIS.


Subject(s)
HIV Infections , Immune Reconstitution Inflammatory Syndrome , Natural Killer T-Cells , Tuberculosis , Cross-Sectional Studies , HIV Infections/complications , HIV Infections/drug therapy , Humans , Tuberculosis/complications
7.
Thorax ; 75(7): 609-610, 2020 07.
Article in English | MEDLINE | ID: mdl-32409610

ABSTRACT

We report the first case of TB associated with triplet therapy (chemotherapy and immunotherapy concurrently) for lung cancer, developing just 44 days after treatment initiation. We feel that several important learning points arise from the discussion that are likely to be very relevant to the broad readership of Thorax, and have important clinical and scientific implications. In the three discussion paragraphs, we highlight that: 1) Triplet therapy is now standard first-line treatment for inoperable lung cancer. 2) TB reactivation is increasingly recognised as an adverse effect of immune checkpoint inhibition, but sending diagnostic samples is critical to avoid a missed diagnosis. 3) These insights from novel cancer immunotherapies are challenging the traditional views of the host-pathogen interaction in TB, with wide implications for future control strategies. We propose that the cases reported in the literature are likely to be the tip of the iceberg as most people with lung cancer managed with antiprogrammed death-1 agents who develop new lung lesions will be treated with standard antibiotics and then palliated when they do not respond.


Subject(s)
Antineoplastic Agents/adverse effects , Carcinoma, Non-Small-Cell Lung/therapy , Immunotherapy/adverse effects , Lung Neoplasms/therapy , Tuberculosis, Pulmonary/etiology , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/diagnosis , Humans , Image-Guided Biopsy , Lung Neoplasms/diagnosis , Male , Middle Aged , Radiography, Thoracic , Tomography, X-Ray Computed , Tuberculosis, Pulmonary/diagnosis
8.
Proc Natl Acad Sci U S A ; 114(51): E10956-E10964, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29158404

ABSTRACT

Tuberculosis (TB), caused by Mycobacterium tuberculosis, remains a major human pandemic. Germline-encoded mycolyl lipid-reactive (GEM) T cells are donor-unrestricted and recognize CD1b-presented mycobacterial mycolates. However, the molecular requirements governing mycolate antigenicity for the GEM T cell receptor (TCR) remain poorly understood. Here, we demonstrate CD1b expression in TB granulomas and reveal a central role for meromycolate chains in influencing GEM-TCR activity. Meromycolate fine structure influences T cell responses in TB-exposed individuals, and meromycolate alterations modulate functional responses by GEM-TCRs. Computational simulations suggest that meromycolate chain dynamics regulate mycolate head group movement, thereby modulating GEM-TCR activity. Our findings have significant implications for the design of future vaccines that target GEM T cells.


Subject(s)
Antigens, CD1/immunology , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/metabolism , Mycolic Acids/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tuberculosis/immunology , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , Antigens, CD1/chemistry , Antigens, CD1/genetics , Gene Expression , Granuloma/immunology , Granuloma/metabolism , Granuloma/microbiology , Granuloma/pathology , Humans , Immunohistochemistry , Lymphocyte Activation/immunology , Models, Molecular , Molecular Conformation , Mycolic Acids/chemistry , Mycolic Acids/metabolism , Protein Binding , Receptors, Antigen, T-Cell/metabolism , Tuberculosis/microbiology
9.
J Infect Dis ; 219(12): 1858-1866, 2019 05 24.
Article in English | MEDLINE | ID: mdl-30929010

ABSTRACT

Despite intensive research efforts, several fundamental disease processes for tuberculosis (TB) remain poorly understood. A central enigma is that host immunity is necessary to control disease yet promotes transmission by causing lung immunopathology. Our inability to distinguish these processes makes it challenging to design rational novel interventions. Elucidating basic immune mechanisms likely requires both in vivo and in vitro analyses, since Mycobacterium tuberculosis is a highly specialized human pathogen. The classic immune response is the TB granuloma organized in three dimensions within extracellular matrix. Several groups are developing cell culture granuloma models. In January 2018, NIAID convened a workshop, entitled "3-D Human in vitro TB Granuloma Model" to advance the field. Here, we summarize the arguments for developing advanced TB cell culture models and critically review those currently available. We discuss how integrating complementary approaches, specifically organoids and mathematical modeling, can maximize progress, and conclude by discussing future challenges and opportunities.


Subject(s)
Granuloma/immunology , Tuberculosis/immunology , Animals , Granuloma/microbiology , Humans , Models, Theoretical , Mycobacterium tuberculosis/immunology , Organoids/immunology , Organoids/microbiology , Tuberculosis/microbiology
10.
J Infect Dis ; 219(4): 633-636, 2019 01 29.
Article in English | MEDLINE | ID: mdl-29920600

ABSTRACT

Matrix metalloproteinases (MMPs) degrade extracellular matrix and are implicated in tuberculosis pathogenesis and cavitation. In particular, MMP-7 is induced by hypoxia and highly expressed around pulmonary cavities of Mycobacterium tuberculosis-infected C3HeB/FeJ mice. In this study, we evaluated whether administration of cipemastat, an orally available potent inhibitor of MMP-7, could reduce pulmonary cavitation in M. tuberculosis-infected C3HeB/FeJ mice. We demonstrate that, compared with untreated controls, cipemastat treatment paradoxically increases the frequency of cavitation (32% vs 7%; P = .029), immunopathology, and mortality. Further studies are needed to understand the role of MMP inhibitors as adjunctive treatments for pulmonary tuberculosis.


Subject(s)
Matrix Metalloproteinase 7/metabolism , Mycobacterium tuberculosis/growth & development , Tuberculosis, Pulmonary/pathology , Animals , Disease Models, Animal , Female , Matrix Metalloproteinase Inhibitors/administration & dosage , Mice, Inbred C3H , Survival Analysis , Tuberculosis, Pulmonary/mortality
11.
PLoS Pathog ; 13(6): e1006367, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28570642

ABSTRACT

Tuberculosis remains a global pandemic and drives lung matrix destruction to transmit. Whilst pathways driving inflammatory responses in macrophages have been relatively well described, negative regulatory pathways are less well defined. We hypothesised that Mycobacterium tuberculosis (Mtb) specifically targets negative regulatory pathways to augment immunopathology. Inhibition of signalling through the PI3K/AKT/mTORC1 pathway increased matrix metalloproteinase-1 (MMP-1) gene expression and secretion, a collagenase central to TB pathogenesis, and multiple pro-inflammatory cytokines. In patients with confirmed pulmonary TB, PI3Kδ expression was absent within granulomas. Furthermore, Mtb infection suppressed PI3Kδ gene expression in macrophages. Interestingly, inhibition of the MNK pathway, downstream of pro-inflammatory p38 and ERK MAPKs, also increased MMP-1 secretion, whilst suppressing secretion of TH1 cytokines. Cross-talk between the PI3K and MNK pathways was demonstrated at the level of eIF4E phosphorylation. Mtb globally suppressed the MMP-inhibitory pathways in macrophages, reducing levels of mRNAs encoding PI3Kδ, mTORC-1 and MNK-1 via upregulation of miRNAs. Therefore, Mtb disrupts negative regulatory pathways at multiple levels in macrophages to drive a tissue-destructive phenotype that facilitates transmission.


Subject(s)
Macrophages/immunology , Mycobacterium tuberculosis/immunology , Tuberculosis, Pulmonary/immunology , Animals , Humans , Macrophages/microbiology , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/immunology , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes/genetics , Multiprotein Complexes/immunology , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/physiology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/immunology , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/immunology , Tuberculosis, Pulmonary/genetics , Tuberculosis, Pulmonary/microbiology , Tuberculosis, Pulmonary/pathology
12.
Trends Immunol ; 37(12): 815-818, 2016 12.
Article in English | MEDLINE | ID: mdl-27773684

ABSTRACT

Tuberculosis (TB) is caused by Mycobacterium tuberculosis (Mtb) and provided original proof that an infectious agent can cause human disease. However, key steps in TB pathogenesis remain poorly understood. We propose that autoimmunity is a critical and overlooked process driving pathology in TB, and present clinical and experimental observations supporting this hypothesis.


Subject(s)
Autoimmune Diseases/immunology , Mycobacterium tuberculosis/immunology , Tuberculosis/immunology , Animals , Autoimmunity , Bacterial Load/immunology , Humans , Immunocompromised Host , Infection Control , Mice , Tuberculosis/transmission
13.
J Pathol ; 244(3): 311-322, 2018 03.
Article in English | MEDLINE | ID: mdl-29210073

ABSTRACT

Tuberculosis (TB) is characterized by extensive pulmonary matrix breakdown. Interleukin-17 (IL-17) is key in host defence in TB but its role in TB-driven tissue damage is unknown. We investigated the hypothesis that respiratory stromal cell matrix metalloproteinase (MMP) production in TB is regulated by T-helper 17 (TH -17) cytokines. Biopsies of patients with pulmonary TB were analysed by immunohistochemistry (IHC), and patient bronchoalveolar lavage fluid (BALF) MMP and cytokine concentrations were measured by Luminex assays. Primary human airway epithelial cells were stimulated with conditioned medium from human monocytes infected with Mycobacterium tuberculosis (Mtb) and TH -17 cytokines. MMP secretion, activity, and gene expression were determined by ELISA, Luminex assay, zymography, RT-qPCR, and dual luciferase reporter assays. Signalling pathways were examined using phospho-western analysis and siRNA. IL-17 is expressed in TB patient granulomas and MMP-3 is expressed in adjacent pulmonary epithelial cells. IL-17 had a divergent, concentration-dependent effect on MMP secretion, increasing epithelial secretion of MMP-3 (p < 0.001) over 72 h, whilst decreasing that of MMP-9 (p < 0.0001); mRNA levels were similarly affected. Both IL-17 and IL-22 increased fibroblast Mtb-dependent MMP-3 secretion but IL-22 did not modulate epithelial MMP-3 expression. Both IL-17 and IL-22, but not IL-23, were significantly up-regulated in BALF from TB patients. IL-17-driven MMP-3 was dependent on p38 MAP kinase and the PI3K p110α subunit. In summary, IL-17 drives airway stromal cell-derived MMP-3, a mediator of tissue destruction in TB, alone and with monocyte-dependent networks in TB. This is regulated by p38 MAP kinase and PI3K pathways. © 2017 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Interleukin-17/pharmacology , Lung/drug effects , Matrix Metalloproteinase 3/biosynthesis , Paracrine Communication/drug effects , Stromal Cells/drug effects , Th17 Cells/metabolism , Tuberculosis, Pulmonary/enzymology , Cells, Cultured , Class I Phosphatidylinositol 3-Kinases/metabolism , Enzyme Induction , Humans , Interleukin-17/immunology , Interleukin-17/metabolism , Interleukins/immunology , Interleukins/metabolism , Lung/enzymology , Lung/immunology , Lung/microbiology , Matrix Metalloproteinase 3/genetics , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/pathogenicity , Signal Transduction/drug effects , Stromal Cells/enzymology , Stromal Cells/immunology , Stromal Cells/microbiology , Th17 Cells/immunology , Th17 Cells/microbiology , Tuberculosis, Pulmonary/genetics , Tuberculosis, Pulmonary/immunology , p38 Mitogen-Activated Protein Kinases/metabolism , Interleukin-22
14.
Proc Natl Acad Sci U S A ; 113(9): E1266-75, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26884207

ABSTRACT

Cluster of differentiation 1c (CD1c)-dependent self-reactive T cells are abundant in human blood, but self-antigens presented by CD1c to the T-cell receptors of these cells are poorly understood. Here we present a crystal structure of CD1c determined at 2.4 Å revealing an extended ligand binding potential of the antigen groove and a substantially different conformation compared with known CD1c structures. Computational simulations exploring different occupancy states of the groove reenacted these different CD1c conformations and suggested cholesteryl esters (CE) and acylated steryl glycosides (ASG) as new ligand classes for CD1c. Confirming this, we show that binding of CE and ASG to CD1c enables the binding of human CD1c self-reactive T-cell receptors. Hence, human CD1c adopts different conformations dependent on ligand occupancy of its groove, with CE and ASG stabilizing CD1c conformations that provide a footprint for binding of CD1c self-reactive T-cell receptors.


Subject(s)
Antigens, CD1/immunology , Cholesterol Esters/metabolism , Glycoproteins/immunology , T-Lymphocytes/immunology , Antigens, CD1/chemistry , Antigens, CD1d , Glycoproteins/chemistry , Humans , Molecular Dynamics Simulation , Protein Conformation
15.
Int J Mol Sci ; 20(6)2019 Mar 18.
Article in English | MEDLINE | ID: mdl-30889803

ABSTRACT

Tuberculosis (TB) remains the single biggest infectious cause of death globally, claiming almost two million lives and causing disease in over 10 million individuals annually. Matrix metalloproteinases (MMPs) are a family of proteolytic enzymes with various physiological roles implicated as key factors contributing to the spread of TB. They are involved in the breakdown of lung extracellular matrix and the consequent release of Mycobacterium tuberculosis bacilli into the airways. Evidence demonstrates that MMPs also play a role in central nervous system (CNS) tuberculosis, as they contribute to the breakdown of the blood brain barrier and are associated with poor outcome in adults with tuberculous meningitis (TBM). However, in pediatric TBM, data indicate that MMPs may play a role in both pathology and recovery of the developing brain. MMPs also have a significant role in HIV-TB-associated immune reconstitution inflammatory syndrome in the lungs and the brain, and their modulation offers potential novel therapeutic avenues. This is a review of recent research on MMPs in pulmonary and CNS TB in adults and children and in the context of co-infection with HIV. We summarize different methods of MMP investigation and discuss the translational implications of MMP inhibition to reduce immunopathology.


Subject(s)
Matrix Metalloproteinases/metabolism , Tuberculosis, Central Nervous System/enzymology , Tuberculosis, Pulmonary/enzymology , Biomarkers/metabolism , Humans , Models, Biological , Tuberculosis, Central Nervous System/therapy , Tuberculosis, Meningeal/enzymology , Tuberculosis, Meningeal/therapy , Tuberculosis, Pulmonary/therapy
16.
J Infect Dis ; 218(1): 53-63, 2018 06 05.
Article in English | MEDLINE | ID: mdl-29554286

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.


Subject(s)
Aerosols/adverse effects , Environmental Exposure , Lung/pathology , Matrix Metalloproteinase 1/metabolism , Mycobacterium tuberculosis/growth & development , Tuberculosis, Pulmonary/epidemiology , Tuberculosis, Pulmonary/pathology , Animals , Disease Models, Animal , Female , Lung/microbiology , Protease Inhibitors/administration & dosage , Rabbits , Tuberculosis, Pulmonary/microbiology
17.
Am J Respir Cell Mol Biol ; 56(2): 223-232, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27654284

ABSTRACT

Tuberculosis (TB) causes disease worldwide, and multidrug resistance is an increasing problem. Matrix metalloproteinases (MMPs), particularly the collagenase MMP-1, cause lung extracellular matrix destruction, which drives disease transmission and morbidity. The role in such tissue damage of the stromelysin MMP-10, a key activator of the collagenase MMP-1, was investigated in direct Mycobacterium tuberculosis (Mtb)-infected macrophages and in conditioned medium from Mtb-infected monocyte-stimulated cells. Mtb infection increased MMP-10 secretion from primary human macrophages 29-fold, whereas Mtb-infected monocytes increased secretion by 4.5-fold from pulmonary epithelial cells and 10.5-fold from fibroblasts. Inhibition of MMP-10 activity decreased collagen breakdown. In two independent cohorts of patients with TB from different continents, MMP-10 was increased in both induced sputum and bronchoalveolar lavage fluid compared with control subjects and patients with other respiratory diseases (both P < 0.05). Mtb drove 3.5-fold greater MMP-10 secretion from human macrophages than the vaccine strain bacillus Calmette-Guerin (P < 0.001), whereas both mycobacteria up-regulated TNF-α secretion equally. Using overlapping, short, linear peptides covering the sequence of early secretory antigenic target-6, a virulence factor secreted by Mtb, but not bacillus Calmette-Guerin, we found that stimulation of human macrophages with a single specific 15-amino acid peptide sequence drove threefold greater MMP-10 secretion than any other peptide (P < 0.001). Mtb-driven MMP-10 secretion was inhibited in a dose-dependent manner by p38 and extracellular signal-related kinase mitogen-activated protein kinase blockade (P < 0.001 and P < 0.01 respectively), but it was not affected by inhibition of NF-κB. In summary, Mtb activates inflammatory and stromal cells to secrete MMP-10, and this is partly driven by the virulence factor early secretory antigenic target-6, implicating it in TB-associated tissue destruction.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Gene Expression Regulation, Enzymologic , Matrix Metalloproteinase 10/genetics , Matrix Metalloproteinase 10/metabolism , Tuberculosis/genetics , Tuberculosis/microbiology , Amino Acid Sequence , Antigens, Bacterial/chemistry , Bacterial Proteins/chemistry , Epithelial Cells/metabolism , Fibroblasts/metabolism , Humans , Lung/pathology , Macrophages/metabolism , Macrophages/microbiology , Matrix Metalloproteinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Mycobacterium tuberculosis/pathogenicity , NF-kappa B/metabolism , Tuberculosis Vaccines/immunology , Virulence
18.
Clin Infect Dis ; 65(1): 121-132, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28475709

ABSTRACT

Background: Extensive immunopathology occurs in human immunodeficiency virus (HIV)/tuberculosis (TB) coinfection, but the underlying molecular mechanisms are not well-defined. Excessive matrix metalloproteinase (MMP) activity is emerging as a key process but has not been systematically studied in HIV-associated TB. Methods: We performed a cross-sectional study of matrix turnover in HIV type 1 (HIV-1)-infected and -uninfected TB patients and controls, and a prospective cohort study of HIV-1-infected TB patients at risk of TB immune reconstitution inflammatory syndrome (TB-IRIS), in Cape Town, South Africa. Sputum and plasma MMP concentrations were quantified by Luminex, plasma procollagen III N-terminal propeptide (PIIINP) by enzyme-linked immunosorbent assay, and urinary lipoarabinomannan (LAM) by Alere Determine TB LAM assay. Peripheral blood mononuclear cells from healthy donors were cultured with Mycobacterium tuberculosis and extracellular matrix in a 3D model of TB granuloma formation. Results: MMP activity differed between HIV-1-infected and -uninfected TB patients and corresponded with specific TB clinical phenotypes. HIV-1-infected TB patients had reduced pulmonary MMP concentrations, associated with reduced cavitation, but increased plasma PIIINP, compared to HIV-1-uninfected TB patients. Elevated extrapulmonary extracellular matrix turnover was associated with TB-IRIS, both before and during TB-IRIS onset. The predominant collagenase was MMP-8, which was likely neutrophil derived and M. tuberculosis-antigen driven. Mycobacterium tuberculosis-induced matrix degradation was suppressed by the MMP inhibitor doxycycline in vitro. Conclusions: MMP activity in TB differs by HIV-1 status and compartment, and releases matrix degradation products. Matrix turnover in HIV-1-infected patients is increased before and during TB-IRIS, informing novel diagnostic strategies. MMP inhibition is a potential host-directed therapy strategy for prevention and treatment of TB-IRIS.


Subject(s)
Collagenases/metabolism , HIV Infections/complications , Immune Reconstitution Inflammatory Syndrome , Tuberculosis , Adult , Cross-Sectional Studies , Female , HIV Infections/epidemiology , HIV-1 , Humans , Immune Reconstitution Inflammatory Syndrome/complications , Immune Reconstitution Inflammatory Syndrome/epidemiology , Immune Reconstitution Inflammatory Syndrome/metabolism , Male , Matrix Metalloproteinase 8/metabolism , Peptide Fragments/metabolism , Procollagen/metabolism , Prospective Studies , South Africa , Tuberculosis/complications , Tuberculosis/epidemiology , Tuberculosis/metabolism , Young Adult
19.
Thorax ; 72(10): 946-949, 2017 10.
Article in English | MEDLINE | ID: mdl-28159773

ABSTRACT

The impact of immunosuppression on interferon-γ release assays and novel cytokine biomarkers of TB infection, mycobacteria-specific IL-2, IP-10 and TNF-α responses was investigated in an ex vivo model. Cytokine responses in standard QuantiFERON-TB Gold in-Tube (QFT-GIT) assays were compared with duplicate assays containing dexamethasone or infliximab. Dexamethasone converted QFT-GIT results from positive to negative in 30% of participants. Antigen-stimulated interferon-γ, IL-2 and TNF-α responses were markedly reduced, but IP-10 responses were preserved. Infliximab caused QFT-GIT result conversion in up to 30% of participants and substantial reductions in all cytokine responses. Therefore, corticosteroids and anti-TNF-α agents significantly impair interferon-γ release assay performance. IP-10 may be a more robust TB biomarker than interferon-γ in patients receiving corticosteroids.


Subject(s)
Adrenal Cortex Hormones/pharmacology , Antirheumatic Agents/pharmacology , Infliximab/pharmacology , Interferon-gamma Release Tests , Latent Tuberculosis/diagnosis , Adult , Aged , Dexamethasone/pharmacology , Female , Humans , Interferon-gamma/metabolism , Interleukin-2/metabolism , Male , Middle Aged , Tumor Necrosis Factor-alpha/metabolism
20.
J Neuroinflammation ; 14(1): 31, 2017 Feb 07.
Article in English | MEDLINE | ID: mdl-28173836

ABSTRACT

BACKGROUND: Central nervous system tuberculosis (CNS-TB) may be fatal even with treatment. Neutrophils are the key mediators of TB immunopathology, and raised CSF matrix metalloproteinase-9 (MMP-9) which correlates to neutrophil count in CNS-TB is associated with neurological deficit and death. The mechanisms by which neutrophils drive TB-associated CNS matrix destruction are not clearly defined. METHODS: Human brain biopsies with histologically proven CNS-TB were stained for neutrophils, neutrophil elastase, and MMP-9. Neutrophil MMP-9 secretion and gene expression were analyzed using Luminex and real-time PCR. Type IV collagen degradation was evaluated using confocal microscopy and quantitative fluorescent assays. Intracellular signaling pathways were investigated by immunoblotting and chemical inhibitors. RESULTS: MMP-9-expressing neutrophils were present in tuberculous granulomas in CNS-TB and neutrophil-derived MMP-9 secretion was upregulated by Mycobacterium tuberculosis (M.tb). Concurrent direct stimulation by M.tb and activation via monocyte-dependent networks had an additive effect on neutrophil MMP-9 secretion. Destruction of type IV collagen, a key component of the blood-brain barrier, was inhibited by neutralizing neutrophil MMP-9. Monocyte-neutrophil networks driving MMP-9 secretion in TB were regulated by MAP-kinase and Akt-PI3 kinase pathways and the transcription factor NF-kB. TNFα neutralization suppressed MMP-9 secretion to baseline while dexamethasone did not. CONCLUSIONS: Multiple signaling paths regulate neutrophil-derived MMP-9 secretion, which is increased in CNS-TB. These paths may be better targets for host-directed therapies than steroids currently used in CNS-TB.


Subject(s)
Gene Expression Regulation, Bacterial/physiology , Leukocytes/metabolism , Neutrophils/metabolism , Tuberculosis, Central Nervous System/pathology , Antibodies/pharmacology , Cells, Cultured , Chromones/pharmacology , Collagen Type IV/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Female , Humans , Male , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Morpholines/pharmacology , Mycobacterium tuberculosis/physiology , NF-kappa B/genetics , NF-kappa B/immunology , NF-kappa B/metabolism , Neutrophils/drug effects , Oncogene Protein v-akt/genetics , Oncogene Protein v-akt/metabolism , Peroxidase/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Up-Regulation/drug effects , Up-Regulation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL