Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
J Clin Invest ; 133(6)2023 03 15.
Article in English | MEDLINE | ID: mdl-36757797

ABSTRACT

Induction of lipid-laden foamy macrophages is a cellular hallmark of tuberculosis (TB) disease, which involves the transformation of infected phagolysosomes from a site of killing into a nutrient-rich replicative niche. Here, we show that a terpenyl nucleoside shed from Mycobacterium tuberculosis, 1-tuberculosinyladenosine (1-TbAd), caused lysosomal maturation arrest and autophagy blockade, leading to lipid storage in M1 macrophages. Pure 1-TbAd, or infection with terpenyl nucleoside-producing M. tuberculosis, caused intralysosomal and peribacillary lipid storage patterns that matched both the molecules and subcellular locations known in foamy macrophages. Lipidomics showed that 1-TbAd induced storage of triacylglycerides and cholesterylesters and that 1-TbAd increased M. tuberculosis growth under conditions of restricted lipid access in macrophages. Furthermore, lipidomics identified 1-TbAd-induced lipid substrates that define Gaucher's disease, Wolman's disease, and other inborn lysosomal storage diseases. These data identify genetic and molecular causes of M. tuberculosis-induced lysosomal failure, leading to successful testing of an agonist of TRPML1 calcium channels that reverses lipid storage in cells. These data establish the host-directed cellular functions of an orphan effector molecule that promotes survival in macrophages, providing both an upstream cause and detailed picture of lysosome failure in foamy macrophages.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Humans , Terpenes , Nucleosides , Macrophages/microbiology , Lipids , Lysosomes
2.
Front Immunol ; 12: 663695, 2021.
Article in English | MEDLINE | ID: mdl-34691015

ABSTRACT

In order to mount an appropriate immune response to infection, the macrophage must alter its metabolism by increasing aerobic glycolysis and concomitantly decreasing oxidative phosphorylation; a process known as the Warburg effect. Consequently, lactate, the end-product of glycolysis, accumulates in the extracellular environment. The subsequent effect of lactate on surrounding macrophages is poorly understood. Mycobacterium tuberculosis (Mtb), the causative organism of Tuberculosis (TB), is phagocytosed by macrophages in the airways. Mtb infected macrophages upregulate aerobic glycolysis and effector functions to try to kill the bacteria. Our lab has previously shown that human macrophages produce lactate in response to infection with Mtb. Although lactate has largely been considered a waste product of aerobic glycolysis, we hypothesised that the presence of extracellular lactate would impact subsequent immunometabolic responses and modulate macrophage function. We demonstrate that the presence of exogenous lactate has an immediate effect on the cellular metabolism of resting human macrophages; causing a decrease in extracellular acidification rate (ECAR; analogous to the rate of glycolysis) and an increase in the oxygen consumption rate (OCR; analogous to oxidative phosphorylation). When lactate-treated macrophages were stimulated with Mtb or LPS, glycolysis proceeds to increase immediately upon stimulation but oxidative phosphorylation remains stable compared with untreated cells that display a decrease in OCR. This resulted in a significantly reduced ECAR/OCR ratio early in response to stimulation. Since altered metabolism is intrinsically linked to macrophage function, we examined the effect of lactate on macrophage cytokine production and ability to kill Mtb. Lactate significantly reduced the concentrations of TNF and IL-1ß produced by human macrophages in response to Mtb but did not alter IL-10 and IL-6 production. In addition, lactate significantly improved bacillary clearance in human macrophages infected with Mtb, through a mechanism that is, at least in part, mediated by promoting autophagy. These data indicate that lactate, the product of glycolysis, has a negative feedback effect on macrophages resulting in an attenuated glycolytic shift upon subsequent stimulation and reduced pro-inflammatory cytokine production. Interestingly, this pro-resolution effect of lactate is associated with increased capacity to kill Mtb.


Subject(s)
Glycolysis/drug effects , Lactic Acid/pharmacology , Macrophages/drug effects , Mycobacterium tuberculosis/pathogenicity , Cells, Cultured , Cytokines/metabolism , Host-Pathogen Interactions , Humans , Inflammation Mediators/metabolism , Lactic Acid/metabolism , Macrophage Activation/drug effects , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Microbial Viability , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/metabolism , Oxidative Phosphorylation/drug effects
3.
J Vis Exp ; (174)2021 08 16.
Article in English | MEDLINE | ID: mdl-34459817

ABSTRACT

Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), was the most significant infectious disease killer globally until the advent of COVID-19. Mtb has evolved to persist in its intracellular environment, evade host defenses, and has developed resistance to many anti-tubercular drugs. One approach to solving resistance is identifying existing approved drugs that will boost the host immune response to Mtb. These drugs could then be repurposed as adjunctive host-directed therapies (HDT) to shorten treatment time and help overcome antibiotic resistance. Quantification of intracellular Mtb growth in macrophages is a crucial aspect of assessing potential HDT. The gold standard for measuring Mtb growth is counting colony-forming units (CFU) on agar plates. This is a slow, labor-intensive assay that does not lend itself to rapid screening of drugs. In this protocol, an automated, broth-based culture system, which is more commonly used to detect Mtb in clinical specimens, has been adapted for preclinical screening of host-directed therapies. The capacity of the liquid culture assay system to investigate intracellular Mtb growth in macrophages treated with HDT was evaluated. The HDTs tested for their ability to inhibit Mtb growth were all-trans Retinoic acid (AtRA), both in solution and encapsulated in poly(lactic-co-glycolic acid) (PLGA) microparticles and the combination of interferon-gamma and linezolid. The advantages of this automated liquid culture-based technique over the CFU method include simplicity of setup, less labor-intensive preparation, and faster time to results (5-12 days compared to 21 days or more for agar plates).


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Automation , Humans , Macrophages , Tuberculosis/drug therapy
4.
J Clin Invest ; 131(15)2021 08 02.
Article in English | MEDLINE | ID: mdl-34338227

ABSTRACT

Immunometabolism is a burgeoning field of investigation in tuberculosis host defense, susceptibility, and pathophysiology. Unbiased approaches to studying tuberculosis have, as expected, confirmed that pathways of immunometabolism are crucial in these disease processes. In this issue of the JCI, Reichmann et al. studied carefully controlled human lymph node tuberculosis and uncovered Sphingosine kinase 1 as a druggable target of interest that could support the infected host. Future host-directed therapy research might seek to establish the different cellular consequences of sphingolipid pathway manipulation. Animal models will be especially useful to establish the role of this pathway, which might target diseased organs to improve mycobactericidal effect and limit pathology.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Animals , Granuloma/genetics , Host-Pathogen Interactions , Humans , Lymph Nodes , Mycobacterium tuberculosis/genetics , Transcriptome , Tuberculosis/drug therapy , Tuberculosis/genetics
5.
Front Immunol ; 11: 836, 2020.
Article in English | MEDLINE | ID: mdl-32477344

ABSTRACT

Tuberculosis is the single biggest infectious killer in the world and presents a major global health challenge. Antimicrobial therapy requires many months of multiple drugs and incidences of drug resistant tuberculosis continues to rise. Consequently, research is now focused on the development of therapies to support the function of infected immune cells. HIF1α-mediated induction of aerobic glycolysis is integral to the host macrophage response during infection with Mtb, as this promotes bacillary clearance. Some iron chelators have been shown to modulate cellular metabolism through the regulation of HIF1α. We examined if the iron chelator, desferrioxamine (DFX), could support the function of primary human macrophages infected with Mtb. Using RT-PCR, we found that DFX promoted the expression of key glycolytic enzymes in Mtb-infected primary human MDMs and human alveolar macrophages. Using Seahorse technology, we demonstrate that DFX enhances glycolytic metabolism in Mtb-stimulated human MDMs, while helping to enhance glycolysis during mitochondrial distress. Furthermore, the effect of DFX on glycolysis was not limited to Mtb infection as DFX also boosted glycolytic metabolism in uninfected and LPS-stimulated cells. DFX also supports innate immune function by inducing IL1ß production in human macrophages during early infection with Mtb and upon stimulation with LPS. Moreover, using hypoxia, Western blot and ChIP-qPCR analyses, we show that DFX modulates IL1ß levels in these cells in a HIF1α-mediated manner. Collectively, our data suggests that DFX exhibits potential to enhance immunometabolic responses and augment host immune function during early Mtb infection, in selected clinical settings.


Subject(s)
Deferoxamine/pharmacology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/microbiology , Mycobacterium tuberculosis/immunology , Siderophores/pharmacology , Tuberculosis/immunology , Blood Donors , Cell Count , Cell Survival/drug effects , Cells, Cultured , Deferoxamine/metabolism , Glycolysis/drug effects , Host-Pathogen Interactions/immunology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Interleukin-1beta/metabolism , Iron/metabolism , Macrophages, Alveolar/metabolism , Siderophores/metabolism , Signal Transduction/drug effects , Tuberculosis/microbiology
6.
Cell Rep ; 30(1): 124-136.e4, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31914380

ABSTRACT

Increased glycolytic metabolism recently emerged as an essential process driving host defense against Mycobacterium tuberculosis (Mtb), but little is known about how this process is regulated during infection. Here, we observe repression of host glycolysis in Mtb-infected macrophages, which is dependent on sustained upregulation of anti-inflammatory microRNA-21 (miR-21) by proliferating mycobacteria. The dampening of glycolysis by miR-21 is mediated through targeting of phosphofructokinase muscle (PFK-M) isoform at the committed step of glycolysis, which facilitates bacterial growth by limiting pro-inflammatory mediators, chiefly interleukin-1ß (IL-1ß). Unlike other glycolytic genes, PFK-M expression and activity is repressed during Mtb infection through miR-21-mediated regulation, while other less-active isoenzymes dominate. Notably, interferon-γ (IFN-γ), which drives Mtb host defense, inhibits miR-21 expression, forcing an isoenzyme switch in the PFK complex, augmenting PFK-M expression and macrophage glycolysis. These findings place the targeting of PFK-M by miR-21 as a key node controlling macrophage immunometabolic function.


Subject(s)
Glycolysis , Host-Pathogen Interactions , Interleukin-1beta/metabolism , MicroRNAs/metabolism , Mycobacterium tuberculosis/physiology , Phosphofructokinase-1/metabolism , Animals , Anti-Inflammatory Agents/metabolism , Base Sequence , Cell Proliferation , HEK293 Cells , Humans , Interferon-gamma/metabolism , Macrophage Activation , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Mice , MicroRNAs/genetics , Phosphofructokinase-1/genetics , RAW 264.7 Cells , Tuberculosis/microbiology
7.
Radiother Oncol ; 145: 128-136, 2020 04.
Article in English | MEDLINE | ID: mdl-31962255

ABSTRACT

BACKGROUND AND PURPOSE: Immunotherapies involving the adoptive transfer of ex vivo expanded autologous invariant natural killer (iNKT) cells are a potential option for cancer patients and are under investigation in clinical trials. Most cancer patients receive radiotherapy at some point during their treatment. We investigated the effects of therapeutic doses of radiation on the viability and function of human primary cultures of iNKT cells in vitro. MATERIALS AND METHODS: iNKT cell lines generated from 6 healthy donors were subjected to therapeutically-relevant doses of radiation. Cell cycle arrest and cell death were assessed by flow cytometry. Double strand DNA breaks were analysed by measuring phosphorylated histone H2AX expression by fluorescence microscopy. Cytolytic degranulation, cytokine production and cytotoxicity by antigen-stimulated iNKT cells were assessed by flow cytometry. RESULTS: Radiation inhibited viability of iNKT cells in a dose-dependent manner. Radiation caused double strand DNA breaks, which were rapidly repaired, and affected the cell cycle at high doses. Moderate doses of radiation did not inhibit degranulation or cytotoxicity by iNKT cells, but induced perforin expression and inhibited proliferation and interferon-γ production by surviving iNKT cells. DISCUSSION: Exposure of iNKT cell to radiation can negatively affect their viability and function.


Subject(s)
Natural Killer T-Cells , Neoplasms , Humans , Immunotherapy , Killer Cells, Natural
8.
Eur J Pharm Biopharm ; 134: 153-165, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30385419

ABSTRACT

Ending the tuberculosis (TB) epidemic by 2030 was recently listed in the United Nations (UN) Sustainable Development Goals alongside HIV/AIDS and malaria as it continues to be a major cause of death worldwide. With a significant proportion of TB cases caused by resistant strains of Mycobacterium tuberculosis (Mtb), there is an urgent need to develop new and innovative approaches to treatment. Since 1989, researchers have been assessing the anti-bacterial effects of the active metabolite of vitamin A, all trans-Retinoic acid (ATRA) solution, in Mtb models. More recently the antibacterial effect of ATRA has been shown to regulate the immune response to infection via critical gene expression, monocyte activation and the induction of autophagy leading to its application as a host-directed therapy (HDT). Inhalation is an attractive route for targeted treatment of TB, and therefore we have developed ATRA-loaded microparticles (ATRA-MP) within the inhalable size range (2.07 ±â€¯0.5 µm) offering targeted delivery of the encapsulated cargo (70.5 ±â€¯2.3%) to the site of action within the alveolar macrophage, which was confirmed by confocal microscopy. Efficient cellular delivery of ATRA was followed by a reduction in Mtb growth (H37Ra) in THP-1 derived macrophages evaluated by both the BACT/ALERT® system and enumeration of colony forming units (CFU). The antibacterial effect of ATRA-MP treatment was further assessed in BALB/c mice infected with the virulent strain of Mtb (H37Rv). ATRA-MP treatments significantly decreased the bacterial burden in the lungs alongside a reduction in pulmonary pathology following just three doses administered intratracheally. The immunomodulatory effects of targeted ATRA treatment in the lungs indicate a distinct yet effective mechanism of action amongst the formulations. This is the first study to-date of a controlled release ATRA treatment for TB suitable for inhalation that offers improved targeting of a HDT, retains antibacterial efficacy and improves pulmonary pathology compared to ATRA solution.


Subject(s)
Antitubercular Agents/administration & dosage , Drug Carriers/chemistry , Mycobacterium tuberculosis/drug effects , Tretinoin/administration & dosage , Tuberculosis, Pulmonary/drug therapy , Administration, Inhalation , Animals , Antitubercular Agents/pharmacokinetics , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/pharmacokinetics , Disease Models, Animal , Drug Compounding/methods , Drug Liberation , Female , Humans , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/metabolism , Mice , Mice, Inbred BALB C , Particle Size , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/microbiology , Pulmonary Alveoli/pathology , THP-1 Cells , Treatment Outcome , Tretinoin/pharmacokinetics , Tuberculosis, Pulmonary/microbiology , Tuberculosis, Pulmonary/pathology
9.
Am J Respir Cell Mol Biol ; 59(5): 572-579, 2018 11.
Article in English | MEDLINE | ID: mdl-29944387

ABSTRACT

Smoking is a major risk factor driving the tuberculosis epidemic, and smokers' alveolar macrophages (AM) demonstrate significant immune defects after infection. Recently, macrophage glycolytic reprogramming has emerged as crucial in the early host immune response to Mycobacterium tuberculosis (Mtb) infection. In the present study, we sought to compare baseline metabolic characteristics and the glycolytic response to infection of human AM from smokers and nonsmokers. AM were obtained at bronchoscopy, and extracellular flux analyses were performed to determine baseline metabolic characteristics compared with human monocyte-derived macrophages (MDM). Metabolic characterization of AM from smokers and nonsmokers was performed similarly. After infection with Mtb, differences in glycolytic response were measured by extracellular flux analyses and gene expression analyses and correlated with production of glycolysis-driven IL-1ß and prostaglandin E2. Similar experiments were performed in cigarette smoke extract-treated MDM as an alternative model. At baseline, human AM from nonsmokers have a significantly lower extracellular acidification rate/oxygen consumption rate ratio than MDM (P < 0.05), but they retain substantial glycolytic reserve. Compared with nonsmokers' AM, smokers' AM demonstrate reduced metabolic activity, reduced glycolytic reserve (P = 0.051), and reduced spare respiratory capacity (P < 0.01). After infection with Mtb, smokers' AM have significantly reduced glycolytic response, as measured by extracellular flux analyses (P < 0.05) and glycolytic gene expression analyses. Cigarette smoke extract-treated MDM similarly demonstrate reduced metabolic activity and reserves, as well as impaired glycolytic response to infection. Human AM demonstrate metabolic plasticity that allows glycolytic reprogramming to occur after Mtb infection. In smokers, this metabolic reserve is significantly attenuated, with consequent impairment of the glycolytic response to infection.


Subject(s)
Cigarette Smoking/adverse effects , Energy Metabolism/immunology , Macrophages, Alveolar/immunology , Metabolome , Mycobacterium tuberculosis/immunology , Pulmonary Alveoli/immunology , Tuberculosis/immunology , Cells, Cultured , Energy Metabolism/drug effects , Glycolysis , Humans , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/microbiology , Mycobacterium tuberculosis/drug effects , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/microbiology , Respiratory Function Tests , Tuberculosis/microbiology , Tuberculosis/pathology
10.
Oncoimmunology ; 7(6): e1428156, 2018.
Article in English | MEDLINE | ID: mdl-29872551

ABSTRACT

Immunotherapies that target CD1d-restricted invariant NKT (iNKT) cells can prevent tumor growth in murine models but trials in humans have shown limited clinical efficacy. Here, we show that iNKT cells are depleted from blood and bronchial lavage samples from patients with non-small cell lung cancer (NSCLC) suggesting a role for these cells in immunity against NSCLC. We interrogated the Lung Cancer Explorer and Kaplan-Meier Plotter databases of NSCLC patients and found that pulmonary CD1d expression is reduced in patients with NSCLC and that low expression of CD1d mRNA is significantly associated with poor patient survival. We hypothesized that CD1d expression in NSCLC is epigenetically regulated and can be modulated using epigenetic targeting therapies. Treatment of the CD1d-negative NSCLC cell lines, A549 and SK-MES-1, with DNA methyltransferase inhibitors and histone deacetylase inhibitors resulted in a dose-dependent induction of CD1d mRNA and protein expression. Chromatin immunoprecipitation analysis indicated that this induction of CD1d expression directly involved chromatin remodelling. Induction of CD1d expression by A549 and SK-MES-1 cells using therapeutic low doses of DNA methyltransferase inhibitors and histone deacetylase inhibitors made them targets for iNKT cell-mediated cytolytic degranulation. Thus, epigenetic manipulation of CD1d expression may augment the efficacy of iNKT cell-based immunotherapies for NSCLC.

11.
Immunity ; 47(3): 552-565.e4, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28844797

ABSTRACT

Mycobacterium tuberculosis (Mtb) enters the host in aerosol droplets deposited in lung alveoli, where the bacteria first encounter lung-resident alveolar macrophages. We studied the earliest mycobacterium-macrophage interactions in the optically transparent zebrafish. First-responding resident macrophages phagocytosed and eradicated infecting mycobacteria, suggesting that to establish a successful infection, mycobacteria must escape out of the initially infected resident macrophage into growth-permissive monocytes. We defined a critical role for mycobacterial membrane phenolic glycolipid (PGL) in engineering this transition. PGL activated the STING cytosolic sensing pathway in resident macrophages, inducing the production of the chemokine CCL2, which in turn recruited circulating CCR2+ monocytes toward infection. Transient fusion of infected macrophages with CCR2+ monocytes enabled bacterial transfer and subsequent dissemination, and interrupting this transfer so as to prolong mycobacterial sojourn in resident macrophages promoted clearing of infection. Human alveolar macrophages produced CCL2 in a PGL-dependent fashion following infection, arguing for the potential of PGL-blocking interventions or PGL-targeting vaccine strategies in the prevention of tuberculosis. VIDEO ABSTRACT.


Subject(s)
Glycolipids/immunology , Macrophages/microbiology , Macrophages/physiology , Mycobacterium tuberculosis/immunology , Animals , Chemokine CCL2/metabolism , Chemotaxis/immunology , Cytokines/metabolism , Disease Models, Animal , Gene Knockout Techniques , Humans , Inflammation Mediators/metabolism , Macrophages, Alveolar/microbiology , Macrophages, Alveolar/physiology , Membrane Proteins/metabolism , Monocytes/immunology , Monocytes/metabolism , Monocytes/microbiology , Mutation , Mycobacterium tuberculosis/genetics , Organ Specificity/immunology , Tuberculosis/immunology , Tuberculosis/metabolism , Tuberculosis/microbiology , Zebrafish
12.
Cell ; 165(1): 139-152, 2016 Mar 24.
Article in English | MEDLINE | ID: mdl-27015311

ABSTRACT

A zebrafish genetic screen for determinants of susceptibility to Mycobacterium marinum identified a hypersusceptible mutant deficient in lysosomal cysteine cathepsins that manifests hallmarks of human lysosomal storage diseases. Under homeostatic conditions, mutant macrophages accumulate undigested lysosomal material, which disrupts endocytic recycling and impairs their migration to, and thus engulfment of, dying cells. This causes a buildup of unengulfed cell debris. During mycobacterial infection, macrophages with lysosomal storage cannot migrate toward infected macrophages undergoing apoptosis in the tuberculous granuloma. The unengulfed apoptotic macrophages undergo secondary necrosis, causing granuloma breakdown and increased mycobacterial growth. Macrophage lysosomal storage similarly impairs migration to newly infecting mycobacteria. This phenotype is recapitulated in human smokers, who are at increased risk for tuberculosis. A majority of their alveolar macrophages exhibit lysosomal accumulations of tobacco smoke particulates and do not migrate to Mycobacterium tuberculosis. The incapacitation of highly microbicidal first-responding macrophages may contribute to smokers' susceptibility to tuberculosis.


Subject(s)
Disease Susceptibility , Lysosomes/metabolism , Macrophages/immunology , Macrophages/pathology , Mycobacterium Infections/immunology , Mycobacterium Infections/pathology , Animals , Granuloma/metabolism , Macrophages/cytology , Macrophages, Alveolar/immunology , Mycobacterium marinum , Pulmonary Alveoli/immunology , Smoking , Transcription Factors/genetics , Transcription Factors/metabolism , Transport Vesicles/metabolism , Tuberculosis/immunology , Tuberculosis/pathology , Zebrafish , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
13.
PLoS One ; 11(2): e0149167, 2016.
Article in English | MEDLINE | ID: mdl-26894562

ABSTRACT

The emergence of multiple-drug-resistant tuberculosis (MDR-TB) has pushed our available repertoire of anti-TB therapies to the limit of effectiveness. This has increased the urgency to develop novel treatment modalities, and inhalable microparticle (MP) formulations are a promising option to target the site of infection. We have engineered poly(lactic-co-glycolic acid) (PLGA) MPs which can carry a payload of anti-TB agents, and are successfully taken up by human alveolar macrophages. Even without a drug cargo, MPs can be potent immunogens; yet little is known about how they influence macrophage function in the setting of Mycobacterium tuberculosis (Mtb) infection. To address this issue we infected THP-1 macrophages with Mtb H37Ra or H37Rv and treated with MPs. In controlled experiments we saw a reproducible reduction in bacillary viability when THP-1 macrophages were treated with drug-free MPs. NFκB activity was increased in MP-treated macrophages, although cytokine secretion was unaltered. Confocal microscopy of immortalized murine bone marrow-derived macrophages expressing GFP-tagged LC3 demonstrated induction of autophagy. Inhibition of caspases did not influence the MP-induced restriction of bacillary growth, however, blockade of NFκB or autophagy with pharmacological inhibitors reversed this MP effect on macrophage function. These data support harnessing inhaled PLGA MP-drug delivery systems as an immunotherapeutic in addition to serving as a vehicle for targeted drug delivery. Such "added value" could be exploited in the generation of inhaled vaccines as well as inhaled MDR-TB therapeutics when used as an adjunct to existing treatments.


Subject(s)
Autophagy/drug effects , Cytotoxicity, Immunologic/drug effects , Lactic Acid/administration & dosage , Macrophages/drug effects , Macrophages/microbiology , Mycobacterium tuberculosis/immunology , NF-kappa B/metabolism , Polyglycolic Acid/administration & dosage , Animals , Caspases/metabolism , Cell Death/drug effects , Cell Death/immunology , Cell Line , Cytokines/biosynthesis , Humans , Macrophages/physiology , Mice , Phagocytosis , Polylactic Acid-Polyglycolic Acid Copolymer
14.
J Immunol ; 196(6): 2444-9, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26873991

ABSTRACT

Recent advances in immunometabolism link metabolic changes in stimulated macrophages to production of IL-1ß, a crucial cytokine in the innate immune response to Mycobacterium tuberculosis. To investigate this pathway in the host response to M. tuberculosis, we performed metabolic and functional studies on human alveolar macrophages, human monocyte-derived macrophages, and murine bone marrow-derived macrophages following infection with the bacillus in vitro. M. tuberculosis infection induced a shift from oxidative phosphorylation to aerobic glycolysis in macrophages. Inhibition of this shift resulted in decreased levels of proinflammatory IL-1ß and decreased transcription of PTGS2, increased levels of anti-inflammatory IL-10, and increased intracellular bacillary survival. Blockade or absence of IL-1R negated the impact of aerobic glycolysis on intracellular bacillary survival, demonstrating that infection-induced glycolysis limits M. tuberculosis survival in macrophages through induction of IL-1ß. Drugs that manipulate host metabolism may be exploited as adjuvants for future therapeutic and vaccination strategies.


Subject(s)
Immunity, Innate/immunology , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/microbiology , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/metabolism , Animals , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Glycolysis/immunology , Humans , Interleukin-1beta/biosynthesis , Interleukin-1beta/immunology , Macrophages, Alveolar/immunology , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis/immunology , Real-Time Polymerase Chain Reaction , Tuberculosis, Pulmonary/microbiology
15.
Am J Respir Crit Care Med ; 190(12): 1430-6, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25390734

ABSTRACT

RATIONALE: Cigarette smoking is linked to important aspects of tuberculosis, such as susceptibility to infection, disease reactivation, mortality, transmission, and persistent infectiousness. The mechanistic basis for this remains poorly understood. OBJECTIVES: To compare the functional impairment seen in human alveolar macrophages (AM) from nonsmokers, smokers, and ex-smokers after infection with Mycobacterium tuberculosis (Mtb). METHODS: AM were acquired at bronchoscopy, and number and viability from smoking donors were compared with nonsmoking donors. AM were challenged in vitro with Mtb and intracellular bacterial viability was measured. Cytokine secretion was measured 24 hours postinfection by ELISA. Previously we determined the frequency of CD4(+)FoxP3(+) T cells in the presence or absence of allogeneic AM, and data were reanalyzed to separate the patient subjects according to smoking status. MEASUREMENTS AND MAIN RESULTS: There were significantly more AM from smokers compared with nonsmokers or ex-smokers (P < 0.01). AM from smokers could not control intracellular Mtb growth. Nonsmokers' AM generated significantly more tumor necrosis factor (TNF)-α, IFN-γ, and IL-1ß after Mtb infection compared with uninfected AM (P < 0.05). However, Mtb-infected AM from smokers did not secrete significantly more TNF-α, IFN-γ, and IL-1ß compared with uninfected smokers' AM. AM taken from ex-smokers also failed to secrete significantly increased TNF-α, IFN-γ, and IL-1ß after Mtb infection. Both smokers' and nonsmokers' AM induced FoxP3(+) T regulatory cell phenotype responses in allogeneic admixed T cells (>4.8 fold; P < 0.05). Even after Mtb infection, AM continued to drive this regulatory phenotype. CONCLUSIONS: In smokers, the pulmonary compartment has a number of macrophage-specific immune impairments that provide some mechanistic explanations whereby cigarette smoking renders a patient susceptible to tuberculosis infection and disease.


Subject(s)
Lung/immunology , Smoking/adverse effects , Tuberculosis, Pulmonary/etiology , Aged , Bronchoscopy , Case-Control Studies , Cytokines/physiology , Disease Susceptibility/etiology , Disease Susceptibility/immunology , Flow Cytometry , Humans , Immunity, Cellular , Lung/microbiology , Macrophages, Alveolar/physiology , Middle Aged , Mycobacterium tuberculosis/immunology , Tuberculosis, Pulmonary/immunology
16.
Mol Pharm ; 8(4): 1100-12, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21591762

ABSTRACT

With an ever increasing number of particulate drug delivery systems being developed for the intracellular delivery of therapeutics a robust high-throughput method for studying particle-cell interactions is urgently required. Current methods used for analyzing particle-cell interaction include spectrofluorimetry, flow cytometry, and fluorescence/confocal microscopy, but these methods are not high throughput and provide only limited data on the specific number of particles delivered intracellularly to the target cell. The work herein presents an automated high-throughput method to analyze microparticulate drug delivery system (DDS) uptake byalveolar macrophages. Poly(lactic-co-glycolic acid) (PLGA) microparticles were prepared in a range of sizes using a solvent evaporation method. A human monocyte cell line (THP-1) was differentiated into macrophage like cells using phorbol 12-myristate 13-acetate (PMA), and cells were treated with microparticles for 1 h and studied using confocal laser scanning microscopy (CLSM), spectrofluorimetry and a high-content analysis (HCA). PLGA microparticles within the size range of 0.8-2.1 µm were found to be optimal for macrophage targeting (p < 0.05). Uptake studies carried out at 37 °C and 4 °C indicated that microparticles were internalized in an energy dependent manner. To improve particle uptake, a range of opsonic coatings were assessed. Coating PLGA particles with gelatin and ovalbumin was found to significantly increase particle uptake from 2.75 ± 0.98 particles per cell for particles coated with gelatin. Opsonic coating also significantly increased particle internalization into primary human alveolar macrophages (p < 0.01) with a 1.7-fold increase in uptake from 4.19 ± 0.48 for uncoated to 7.53 ± 0.88 particles per cell for coated particles. In comparison to techniques such as spectrofluorimetry and CLSM, HCA provides both qualitative and quantitative data on the influence of carrier design on cell targeting that can be gathered in a high-throughput format and therefore has great potential in the screening of intracellularly targeted DDS.


Subject(s)
Drug Delivery Systems/methods , Macrophages, Alveolar/metabolism , Cell Line , Humans , Lactic Acid/chemistry , Microscopy, Confocal , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Spectrometry, Fluorescence , Tetradecanoylphorbol Acetate/chemistry
17.
Am J Respir Cell Mol Biol ; 45(1): 172-80, 2011 Jul.
Article in English | MEDLINE | ID: mdl-20889800

ABSTRACT

Successful phagolysosomal maturation is an important innate immune response to intracellular infection. However, Mycobacterium tuberculosis (Mtb) can manipulate and inhibit this host response to ensure survival within its niche cell. We investigate the role of the anti-inflammatory cytokine IL-10 on Mtb-phagosome maturation. Blocking IL-10, which was secreted from Mtb-infected macrophages, allowed phagosome maturation to proceed. Macrophage cytokine gene expression profiles were not significantly altered by blocking IL-10 3 hours after infection with Mtb. We demonstrate that IL-10 can regulate this protective phenotype in phorbol myristate acetate (PMA)-treated THP-1 cells, monocyte-derived macrophages (MDMs), and human alveolar macrophages (AMs) infected with Mtb. The regulatory effect of endogenous IL-10 was evident in macrophages infected with virulent Mtb H37Rv, as well as in attenuated strains of mycobacteria. Unlike live Mtb, dead bacilli occupy a mature, acidic phagosome. However, the addition of IL-10 to cells infected with killed Mtb successfully inhibited the maturation of this compartment. Importantly, we demonstrate that the addition of IL-10 to MDMs results in enhanced mycobacterial survival and growth. Our results suggest that IL-10 exerts its effects on this early macrophage response in a partly signal transducer and activator of transcription 3 (STAT3)-dependent manner, and independent of mitogen activated protein kinase p38 (MAPKp38) and extracellular regulated kinase 1/2 (ERK1/2) activity. IL-10 is a feature of human tuberculous granuloma, and these new findings support the hypothesis that this cytokine can promote pathogen persistence by contributing to Mtb-phagosome maturation arrest in human macrophages.


Subject(s)
Interleukin-10/metabolism , Macrophages , Mycobacterium tuberculosis/growth & development , Phagosomes , Tuberculosis , Carcinogens/pharmacology , Cell Line , Granuloma/metabolism , Granuloma/microbiology , Granuloma/pathology , Humans , Macrophages/metabolism , Macrophages/microbiology , Macrophages/pathology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phagosomes/metabolism , Phagosomes/microbiology , Phagosomes/pathology , STAT3 Transcription Factor/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Tuberculosis/metabolism , Tuberculosis/microbiology , Tuberculosis/pathology , p38 Mitogen-Activated Protein Kinases/metabolism
18.
Infect Immun ; 76(1): 351-60, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17954721

ABSTRACT

Human macrophages infected with Mycobacterium tuberculosis may undergo apoptosis. Macrophage apoptosis contributes to the innate immune response against M. tuberculosis by containing and limiting the growth of mycobacteria and also by depriving the bacillus of its niche cell. Apoptosis of infected macrophages is well documented; however, bystander apoptosis of uninfected macrophages has not been described in the setting of M. tuberculosis. We observed that uninfected human macrophages underwent significant bystander apoptosis 48 and 96 h after they came into contact with macrophages infected with avirulent M. tuberculosis. The bystander apoptosis was significantly greater than the background apoptosis observed in uninfected control cells cultured for the same length of time. There was no evidence of the involvement of tumor necrosis factor alpha, Fas, tumor necrosis factor-related apoptosis-inducing ligand, transforming growth factor beta, Toll-like receptor 2, or MyD88 in contact-mediated bystander apoptosis. This newly described phenomenon may further limit the spread of M. tuberculosis by eliminating the niche cells on which the bacillus relies.


Subject(s)
Apoptosis , Macrophages/cytology , Macrophages/microbiology , Mycobacterium tuberculosis/physiology , Cell Communication , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Humans , Macrophages/drug effects , Myeloid Differentiation Factor 88/metabolism , Neutrophils/microbiology , Neutrophils/physiology , Staurosporine/pharmacology , TNF-Related Apoptosis-Inducing Ligand/metabolism , Toll-Like Receptor 2/metabolism , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/metabolism , fas Receptor/metabolism
19.
Infect Immun ; 75(4): 1984-93, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17283090

ABSTRACT

Macrophages can undergo apoptosis after infection with Mycobacterium tuberculosis. This macrophage response deprives the bacillus of its niche cell and supports the host response through better antigen presentation. The intracellular pathways of apoptosis that elaborate this macrophage response are not well understood. To address this issue, we investigated the contribution of various apoptosis pathways to M. tuberculosis-induced macrophage cell death. We found that macrophages die in a caspase-independent manner after infection with M. tuberculosis (at multiplicities of infection ranging from 1 to 20). There was evidence for the involvement of both the mitochondria (cleavage of Bid) and the lysosomes (cathepsin-mediated DNA fragmentation) in this cell death pathway. Dying macrophages displayed several features typical of apoptosis, including DNA fragmentation, nuclear condensation, and exposure of phosphatidylserine on the plasma membrane. However, nuclear fragmentation was not observed, which suggests that M. tuberculosis-induced cell death differs in some respects from classical apoptosis. This novel mechanism of cell death was blocked by serine protease inhibitors. A better understanding of this protective macrophage response may direct new vaccine and treatment options.


Subject(s)
Apoptosis , Macrophages/cytology , Macrophages/microbiology , Mycobacterium tuberculosis/immunology , BH3 Interacting Domain Death Agonist Protein/metabolism , Bacillus , Caspases/metabolism , Cathepsins/metabolism , Cell Line , Cell Survival , Cells, Cultured , DNA Fragmentation , Humans , Lysosomes/metabolism , Macrophages/metabolism , Mitochondria/metabolism , Phosphatidylserines/analysis , Serine Proteinase Inhibitors/pharmacology , Sulfones/pharmacology
20.
Res Vet Sci ; 81(2): 170-6, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16545848

ABSTRACT

Brucellosis is a highly infectious disease which is diagnosed using serological and microbiological methods. The objective of this study was to assess the viability of using conventional and real-time PCR assays as potential diagnostic tools for the detection of Brucella abortus in naturally infected cows. PCR assays that amplify various regions of the Brucella genome, IS711 genetic element, 31kDa outer membrane protein and 16S rRNA, were optimised using nine known Brucella strains. Real-time PCR was used to examine the detection efficiency of the IS711 assay which was estimated at 10 gene copies. Milk, blood and lymph tissue samples were collected from naturally infected animals. B. abortus was not detected in blood samples collected from naturally infected cows by conventional or real-time PCR, but was detected in a proportion of the culture-positive milk (44%) and lymph tissue (66% - retropharyngeal, 75% - supramammary) samples by the same methods. There was no difference between PCR and bacteriological detection methods. It is unlikely that conventional or real-time PCR will supersede current diagnostic methods for detection of B. abortus in clinical samples.


Subject(s)
Brucella abortus/isolation & purification , Brucellosis, Bovine/microbiology , Lymph Nodes/microbiology , Milk/microbiology , Animals , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/genetics , Brucella abortus/genetics , Brucellosis, Bovine/blood , Cattle , Colony Count, Microbial/veterinary , DNA Transposable Elements/genetics , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Female , Polymerase Chain Reaction/veterinary , RNA, Ribosomal, 16S/chemistry , RNA, Ribosomal, 16S/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...