Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Mucosal Immunol ; 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38844208

ABSTRACT

Mycobacterium tuberculosis (Mtb)-infected neutrophils are often found in the airways of patients with active tuberculosis (TB), and excessive recruitment of neutrophils to the lung is linked to increased bacterial burden and aggravated pathology in TB. The basis for the permissiveness of neutrophils for Mtb and the ability to be pathogenic in TB has been elusive. Here, we identified metabolic and functional features of neutrophils that contribute to their permissiveness in Mtb infection. Using single-cell metabolic and transcriptional analyses, we found that neutrophils in the Mtb-infected lung displayed elevated mitochondrial metabolism, which was largely attributed to the induction of activated neutrophils with enhanced metabolic activities. The activated neutrophil subpopulation was also identified in the lung granulomas from Mtb-infected non-human primates. Functionally, activated neutrophils harbored more viable bacteria and displayed enhanced lipid uptake and accumulation. Surprisingly, we found that interferon-γ promoted the activation of lung neutrophils during Mtb infection. Lastly, perturbation of lipid uptake pathways selectively compromised Mtb survival in activated neutrophils. These findings suggest that neutrophil heterogeneity and metabolic diversity are key to their permissiveness for Mtb and that metabolic pathways in neutrophils represent potential host-directed therapeutics in TB.

3.
Immunometabolism (Cobham) ; 6(2): e00042, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38693938

ABSTRACT

Mycobacterium tuberculosis causes tuberculosis (TB), one of the world's most deadly infections. Lipids play an important role in M. tuberculosis pathogenesis. M. tuberculosis grows intracellularly within lipid-laden macrophages and extracellularly within the cholesterol-rich caseum of necrotic granulomas and pulmonary cavities. Evolved from soil saprophytes that are able to metabolize cholesterol from organic matter in the environment, M. tuberculosis inherited an extensive and highly conserved machinery to metabolize cholesterol. M. tuberculosis uses this machinery to degrade host cholesterol; the products of cholesterol degradation are incorporated into central carbon metabolism and used to generate cell envelope lipids, which play important roles in virulence. The host also modifies cholesterol by enzymatically oxidizing it to a variety of derivatives, collectively called oxysterols, which modulate cholesterol homeostasis and the immune response. Recently, we found that M. tuberculosis converts host cholesterol to an oxidized metabolite, cholestenone, that accumulates in the lungs of individuals with TB. M. tuberculosis encodes cholesterol-modifying enzymes, including a hydroxysteroid dehydrogenase, a putative cholesterol oxidase, and numerous cytochrome P450 monooxygenases. Here, we review what is known about cholesterol and its oxidation products in the pathogenesis of TB. We consider the possibility that the biological function of cholesterol metabolism by M. tuberculosis extends beyond a nutritional role.

4.
PLoS Pathog ; 20(4): e1012137, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38603763

ABSTRACT

Interleukin-1 (IL-1) signaling is essential for controlling virulent Mycobacterium tuberculosis (Mtb) infection since antagonism of this pathway leads to exacerbated pathology and increased susceptibility. In contrast, the triggering of type I interferon (IFN) signaling is associated with the progression of tuberculosis (TB) disease and linked with negative regulation of IL-1 signaling. However, mice lacking IL-1 signaling can control Mtb infection if infected with an Mtb strain carrying the rifampin-resistance conferring mutation H445Y in its RNA polymerase ß subunit (rpoB-H445Y Mtb). The mechanisms that govern protection in the absence of IL-1 signaling during rpoB-H445Y Mtb infection are unknown. In this study, we show that in the absence of IL-1 signaling, type I IFN signaling controls rpoB-H445Y Mtb replication, lung pathology, and excessive myeloid cell infiltration. Additionally, type I IFN is produced predominantly by monocytes and recruited macrophages and acts on LysM-expressing cells to drive protection through nitric oxide (NO) production to restrict intracellular rpoB-H445Y Mtb. These findings reveal an unexpected protective role for type I IFN signaling in compensating for deficiencies in IL-1 pathways during rpoB-H445Y Mtb infection.


Subject(s)
Bacterial Proteins , DNA-Directed RNA Polymerases , Interferon Type I , Mycobacterium tuberculosis , Rifampin , Signal Transduction , Interferon Type I/metabolism , Animals , Mice , Rifampin/pharmacology , DNA-Directed RNA Polymerases/metabolism , DNA-Directed RNA Polymerases/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Mutation , Mice, Inbred C57BL , Drug Resistance, Bacterial/genetics , Tuberculosis/microbiology , Tuberculosis/immunology , Tuberculosis/genetics , Mice, Knockout
5.
Nano Lett ; 24(1): 229-237, 2024 Jan 10.
Article in English | MEDLINE | ID: mdl-38146928

ABSTRACT

Rapid and accurate quantification of low-abundance protein biomarkers in biofluids can transform the diagnosis of a range of pathologies, including infectious diseases. Here, we harness ultrabright plasmonic fluors as "digital nanolabels" and demonstrate the detection and quantification of subfemtomolar concentrations of human IL-6 and SARS-CoV-2 alpha and variant proteins in clinical nasopharyngeal swab and saliva samples from COVID-19 patients. The resulting digital plasmonic fluor-linked immunosorbent assay (digital p-FLISA) enables detection of SARS-CoV-2 nucleocapsid protein, both in solution and in live virions. Digital p-FLISA outperforms the "gold standard" enzyme-linked immunosorbent assay (ELISA), having a nearly 7000-fold lower limit-of-detection, and outperforms a commercial antigen test, having over 5000-fold improvement in analytical sensitivity. Detection and quantification of very low concentrations of target proteins holds potential for early detection of pathological conditions, treatment monitoring, and personalized medicine.


Subject(s)
COVID-19 , Humans , Enzyme-Linked Immunosorbent Assay , COVID-19/diagnosis , Fluoroimmunoassay , SARS-CoV-2 , Biomarkers , Sensitivity and Specificity
6.
Proc Natl Acad Sci U S A ; 121(1): e2315865120, 2024 Jan 02.
Article in English | MEDLINE | ID: mdl-38147552

ABSTRACT

To define cellular immunity to the intracellular pathogen Toxoplasma gondii, we performed a genome-wide CRISPR loss-of-function screen to identify genes important for (interferon gamma) IFN-γ-dependent growth restriction. We revealed a role for the tumor suppressor NF2/Merlin for maximum induction of Interferon Stimulated Genes (ISG), which are positively regulated by the transcription factor IRF-1. We then performed an ISG-targeted CRISPR screen that identified the host E3 ubiquitin ligase RNF213 as necessary for IFN-γ-mediated control of T. gondii in multiple human cell types. RNF213 was also important for control of bacterial (Mycobacterium tuberculosis) and viral (Vesicular Stomatitis Virus) pathogens in human cells. RNF213-mediated ubiquitination of the parasitophorous vacuole membrane (PVM) led to growth restriction of T. gondii in response to IFN-γ. Moreover, overexpression of RNF213 in naive cells also impaired growth of T. gondii. Surprisingly, growth inhibition did not require the autophagy protein ATG5, indicating that RNF213 initiates restriction independent of a previously described noncanonical autophagy pathway. Mutational analysis revealed that the ATPase domain of RNF213 was required for its recruitment to the PVM, while loss of a critical histidine in the RZ finger domain resulted in partial reduction of recruitment to the PVM and complete loss of ubiquitination. Both RNF213 mutants lost the ability to restrict growth of T. gondii, indicating that both recruitment and ubiquitination are required. Collectively, our findings establish RNF213 as a critical component of cell-autonomous immunity that is both necessary and sufficient for control of intracellular pathogens in human cells.


Subject(s)
Toxoplasma , Toxoplasmosis , Humans , Interferon-gamma/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats , Toxoplasma/metabolism , Transcription Factors , Adenosine Triphosphatases/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
7.
Cell Rep ; 43(1): 113607, 2024 01 23.
Article in English | MEDLINE | ID: mdl-38127624

ABSTRACT

Mycobacterium tuberculosis (Mtb) possesses an arsenal of virulence factors to evade host immunity. Previously, we showed that the Mtb protein CpsA, which protects Mtb against the host NADPH oxidase, is required in mice during the first 3 weeks of infection but is thereafter dispensable for full virulence. Using flow cytometry, we find that ΔcpsA Mtb is retained in alveolar macrophages, impaired in recruiting and disseminating into monocyte-derived cells, and more likely to be localized in airway cells than wild-type Mtb. The lungs of ΔcpsA-infected mice also have markedly fewer antigen-specific T cells, indicating a delay in adaptive immunity. Thus, we conclude that CpsA promotes dissemination of Mtb from alveolar macrophages and the airways and generation of an adaptive immune response. Our studies of ΔcpsA Mtb show that a more effective innate immune response against Mtb can be undermined by a corresponding delay in the adaptive immune response.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Mice , Animals , Lung , Macrophages, Alveolar , Immunity, Innate
8.
Elife ; 122023 02 28.
Article in English | MEDLINE | ID: mdl-36852737

ABSTRACT

For decades, investigators have studied the interaction of Mycobacterium tuberculosis (Mtb) with macrophages, which serve as a major cellular niche for the bacilli. Because Mtb are prone to aggregation, investigators rely on varied methods to disaggregate the bacteria for these studies. Here, we examined the impact of routinely used preparation methods on bacterial cell envelope integrity, macrophage inflammatory responses, and intracellular Mtb survival. We found that both gentle sonication and filtering damaged the mycobacterial cell envelope and markedly impacted the outcome of infections in mouse bone marrow-derived macrophages. Unexpectedly, sonicated bacilli were hyperinflammatory, eliciting dramatically higher TLR2-dependent gene expression and elevated secretion of IL-1ß and TNF-α. Despite evoking enhanced inflammatory responses, sonicated bacilli replicated normally in macrophages. In contrast, Mtb that had been passed through a filter induced little inflammatory response, and they were attenuated in macrophages. Previous work suggests that the mycobacterial cell envelope lipid, phthiocerol dimycocerosate (PDIM), dampens macrophage inflammatory responses to Mtb. However, we found that the impact of PDIM depended on the method used to prepare Mtb. In conclusion, widely used methodologies to disaggregate Mtb may introduce experimental artifacts in Mtb-host interaction studies, including alteration of host inflammatory signaling, intracellular bacterial survival, and interpretation of bacterial mutants.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Animals , Mice , Mycobacterium tuberculosis/physiology , Tuberculosis/microbiology , Macrophages/microbiology , Tumor Necrosis Factor-alpha/metabolism , Phagosomes/metabolism , Host-Pathogen Interactions
9.
Cell Rep Methods ; 2(8): 100267, 2022 08 22.
Article in English | MEDLINE | ID: mdl-36046626

ABSTRACT

Secreted proteins mediate essential physiological processes. With conventional assays, it is challenging to map the spatial distribution of proteins secreted by single cells, to study cell-to-cell heterogeneity in secretion, or to detect proteins of low abundance or incipient secretion. Here, we introduce the "FluoroDOT assay," which uses an ultrabright nanoparticle plasmonic-fluor that enables high-resolution imaging of protein secretion. We find that plasmonic-fluors are 16,000-fold brighter, with nearly 30-fold higher signal-to-noise compared with conventional fluorescence labels. We demonstrate high-resolution imaging of different secreted cytokines in the single-plexed and spectrally multiplexed FluoroDOT assay that revealed cellular heterogeneity in secretion of multiple proteins simultaneously. Using diverse biochemical stimuli, including Mycobacterium tuberculosis infection, and a variety of immune cells such as macrophages, dendritic cells (DCs), and DC-T cell co-culture, we demonstrate that the assay is versatile, facile, and widely adaptable for enhancing biological understanding of spatial and temporal dynamics of single-cell secretome.


Subject(s)
Cytokines , Tuberculosis , Humans , Cytokines/metabolism , Tuberculosis/metabolism , Macrophages , T-Lymphocytes/metabolism
10.
Nat Rev Microbiol ; 20(12): 750-766, 2022 12.
Article in English | MEDLINE | ID: mdl-35879556

ABSTRACT

Mycobacterium tuberculosis, the causative agent of tuberculosis, has infected humans for millennia. M. tuberculosis is well adapted to establish infection, persist in the face of the host immune response and be transmitted to uninfected individuals. Its ability to complete this infection cycle depends on it both evading and taking advantage of host immune responses. The outcome of M. tuberculosis infection is often a state of equilibrium characterized by immunological control and bacterial persistence. Recent data have highlighted the diverse cell populations that respond to M. tuberculosis infection and the dynamic changes in the cellular and intracellular niches of M. tuberculosis during the course of infection. M. tuberculosis possesses an arsenal of protein and lipid effectors that influence macrophage functions and inflammatory responses; however, our understanding of the role that specific bacterial virulence factors play in the context of diverse cellular reservoirs and distinct infection stages is limited. In this Review, we discuss immune evasion and provocation by M. tuberculosis during its infection cycle and describe how a more detailed molecular understanding is crucial to enable the development of novel host-directed therapies, disease biomarkers and effective vaccines.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Humans , Immune Evasion , Tuberculosis/microbiology , Macrophages/microbiology , Virulence Factors/metabolism
11.
J Clin Invest ; 132(3)2022 02 01.
Article in English | MEDLINE | ID: mdl-35104812

ABSTRACT

Mycobacterium tuberculosis (M. tuberculosis) causes an enormous burden of disease worldwide. As a central aspect of its pathogenesis, M. tuberculosis grows in macrophages, and host and microbe influence each other's metabolism. To define the metabolic impact of M. tuberculosis infection, we performed global metabolic profiling of M. tuberculosis-infected macrophages. M. tuberculosis induced metabolic hallmarks of inflammatory macrophages and a prominent signature of cholesterol metabolism. We found that infected macrophages accumulate cholestenone, a mycobacterial-derived, oxidized derivative of cholesterol. We demonstrated that the accumulation of cholestenone in infected macrophages depended on the M. tuberculosis enzyme 3ß-hydroxysteroid dehydrogenase (3ß-Hsd) and correlated with pathogen burden. Because cholestenone is not a substantial human metabolite, we hypothesized it might be diagnostic of M. tuberculosis infection in clinical samples. Indeed, in 2 geographically distinct cohorts, sputum cholestenone levels distinguished subjects with tuberculosis (TB) from TB-negative controls who presented with TB-like symptoms. We also found country-specific detection of cholestenone in plasma samples from M. tuberculosis-infected subjects. While cholestenone was previously thought to be an intermediate required for cholesterol degradation by M. tuberculosis, we found that M. tuberculosis can utilize cholesterol for growth without making cholestenone. Thus, the accumulation of cholestenone in clinical samples suggests it has an alternative role in pathogenesis and could be a clinically useful biomarker of TB infection.


Subject(s)
Host-Pathogen Interactions , Macrophages/metabolism , Metabolomics , Mycobacterium tuberculosis/physiology , Signal Transduction , Tuberculosis/metabolism , Animals , Humans , Macrophages/microbiology , Mice
12.
Proc Natl Acad Sci U S A ; 117(52): 33561-33569, 2020 12 29.
Article in English | MEDLINE | ID: mdl-33376222

ABSTRACT

Streptococcus pneumoniae is a leading cause of pneumonia and invasive disease, particularly, in the elderly. S. pneumoniae lung infection of aged mice is associated with high bacterial burdens and detrimental inflammatory responses. Macrophages can clear microorganisms and modulate inflammation through two distinct lysosomal trafficking pathways that involve 1A/1B-light chain 3 (LC3)-marked organelles, canonical autophagy, and LC3-associated phagocytosis (LAP). The S. pneumoniae pore-forming toxin pneumolysin (PLY) triggers an autophagic response in nonphagocytic cells, but the role of LAP in macrophage defense against S. pneumoniae or in age-related susceptibility to infection is unexplored. We found that infection of murine bone-marrow-derived macrophages (BMDMs) by PLY-producing S. pneumoniae triggered Atg5- and Atg7-dependent recruitment of LC3 to S. pneumoniae-containing vesicles. The association of LC3 with S. pneumoniae-containing phagosomes required components specific for LAP, such as Rubicon and the NADPH oxidase, but not factors, such as Ulk1, FIP200, or Atg14, required specifically for canonical autophagy. In addition, S. pneumoniae was sequestered within single-membrane compartments indicative of LAP. Importantly, compared to BMDMs from young (2-mo-old) mice, BMDMs from aged (20- to 22-mo-old) mice infected with S. pneumoniae were not only deficient in LAP and bacterial killing, but also produced higher levels of proinflammatory cytokines. Inhibition of LAP enhanced S. pneumoniae survival and cytokine responses in BMDMs from young but not aged mice. Thus, LAP is an important innate immune defense employed by BMDMs to control S. pneumoniae infection and concomitant inflammation, one that diminishes with age and may contribute to age-related susceptibility to this important pathogen.


Subject(s)
Aging/immunology , Host-Pathogen Interactions/immunology , Macrophages/metabolism , Macrophages/microbiology , Microtubule-Associated Proteins/metabolism , Phagocytosis , Streptococcus pneumoniae/immunology , Animals , Autophagy , Bacterial Proteins/metabolism , Lipids/chemistry , Macrophages/ultrastructure , Male , Mice , Mice, Inbred C57BL , Microbial Viability , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Streptococcus pneumoniae/ultrastructure , Streptolysins/metabolism
13.
Cell Rep ; 33(5): 108339, 2020 11 03.
Article in English | MEDLINE | ID: mdl-33147451

ABSTRACT

Here, we report our studies of immune-mediated regulation of Zika virus (ZIKV), herpes simplex virus 1 (HSV-1), and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in the human cornea. We find that ZIKV can be transmitted via corneal transplantation in mice. However, in human corneal explants, we report that ZIKV does not replicate efficiently and that SARS-CoV-2 does not replicate at all. Additionally, we demonstrate that type III interferon (IFN-λ) and its receptor (IFNλR1) are expressed in the corneal epithelium. Treatment of human corneal explants with IFN-λ, and treatment of mice with IFN-λ eye drops, upregulates antiviral interferon-stimulated genes. In human corneal explants, blockade of IFNλR1 enhances replication of ZIKV and HSV-1 but not SARS-CoV-2. In addition to an antiviral role for IFNλR1 in the cornea, our results suggest that the human cornea does not support SARS-CoV-2 infection despite expression of ACE2, a SARS-CoV-2 receptor, in the human corneal epithelium.


Subject(s)
Betacoronavirus/physiology , Cornea/virology , Coronavirus Infections/transmission , Herpesvirus 1, Human/physiology , Interferons/immunology , Pneumonia, Viral/transmission , Zika Virus/physiology , Animals , Betacoronavirus/immunology , COVID-19 , Cornea/immunology , Coronavirus Infections/immunology , Coronavirus Infections/virology , Herpes Simplex/immunology , Herpes Simplex/transmission , Herpes Simplex/virology , Humans , Mice , Pandemics , Pneumonia, Viral/immunology , Pneumonia, Viral/virology , SARS-CoV-2 , Virus Replication/physiology , Zika Virus Infection/immunology , Zika Virus Infection/transmission , Zika Virus Infection/virology , Interferon Lambda
14.
mBio ; 11(4)2020 07 07.
Article in English | MEDLINE | ID: mdl-32636249

ABSTRACT

Macrophage activation involves metabolic reprogramming to support antimicrobial cellular functions. How these metabolic shifts influence the outcome of infection by intracellular pathogens remains incompletely understood. Mycobacterium tuberculosis (Mtb) modulates host metabolic pathways and utilizes host nutrients, including cholesterol and fatty acids, to survive within macrophages. We found that intracellular growth of Mtb depends on host fatty acid catabolism: when host fatty acid ß-oxidation (FAO) was blocked chemically with trimetazidine, a compound in clinical use, or genetically by deletion of the mitochondrial fatty acid transporter carnitine palmitoyltransferase 2 (CPT2), Mtb failed to grow in macrophages, and its growth was attenuated in mice. Mechanistic studies support a model in which inhibition of FAO generates mitochondrial reactive oxygen species, which enhance macrophage NADPH oxidase and xenophagy activity to better control Mtb infection. Thus, FAO inhibition promotes key antimicrobial functions of macrophages and overcomes immune evasion mechanisms of Mtb.IMPORTANCEMycobacterium tuberculosis (Mtb) is the leading infectious disease killer worldwide. We discovered that intracellular Mtb fails to grow in macrophages in which fatty acid ß-oxidation (FAO) is blocked. Macrophages treated with FAO inhibitors rapidly generate a burst of mitochondria-derived reactive oxygen species, which promotes NADPH oxidase recruitment and autophagy to limit the growth of Mtb. Furthermore, we demonstrate the ability of trimetazidine to reduce pathogen burden in mice infected with Mtb. These studies will add to the knowledge of how host metabolism modulates Mtb infection outcomes.


Subject(s)
Fatty Acids/metabolism , Host-Pathogen Interactions/drug effects , Macrophages/microbiology , Mycobacterium tuberculosis/immunology , Oxidation-Reduction/drug effects , Animals , Antitubercular Agents/pharmacology , Cells, Cultured , Cytokines/analysis , Female , Lipid Metabolism/drug effects , Macrophage Activation , Macrophages/drug effects , Metabolic Networks and Pathways , Mice , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Trimetazidine/pharmacology , Tuberculosis/microbiology
15.
Curr Opin Immunol ; 60: 81-90, 2019 10.
Article in English | MEDLINE | ID: mdl-31247378

ABSTRACT

The innate immune system has evolved to recognize diverse microbes and destroy them. At the same time, microbial pathogens undermine immunity to cause disease. Here, we highlight recent advances in understanding an antimicrobial pathway called LC3-associated phagocytosis (LAP), which combines features of autophagy with phagocytosis. Upon phagocytosis, many microbes, including bacteria, fungi, and parasites, are sequestered in an LC3-positive, single-membrane bound compartment, a hallmark of LAP. LAP depends upon NADPH oxidase activity at the incipient phagosome and culminates in lysosomal trafficking and microbial degradation. Most often LAP is an effective host defense, but some pathogens evade LAP or replicate successfully in this microenvironment. Here, we review how LAP targets microbial pathogens and strategies pathogens employ to circumvent LAP.


Subject(s)
Host-Pathogen Interactions , Microtubule-Associated Proteins/metabolism , Phagocytes/immunology , Phagocytes/metabolism , Phagocytosis/physiology , Animals , Biomarkers , Humans , Immunity, Innate , Macroautophagy , Oxidation-Reduction , Phagosomes/metabolism , Signal Transduction
16.
mBio ; 9(6)2018 11 27.
Article in English | MEDLINE | ID: mdl-30482832

ABSTRACT

Intracellular pathogens have varied strategies to breach the endolysosomal barrier so that they can deliver effectors to the host cytosol, access nutrients, replicate in the cytoplasm, and avoid degradation in the lysosome. In the case of Mycobacterium tuberculosis, the bacterium perforates the phagosomal membrane shortly after being taken up by macrophages. Phagosomal damage depends upon the mycobacterial ESX-1 type VII secretion system (T7SS). Sterile insults, such as silica crystals or membranolytic peptides, can also disrupt phagosomal and endolysosomal membranes. Recent work revealed that the host endosomal sorting complex required for transport (ESCRT) machinery rapidly responds to sterile endolysosomal damage and promotes membrane repair. We hypothesized that ESCRTs might also respond to pathogen-induced phagosomal damage and that M. tuberculosis could impair this host response. Indeed, we found that ESCRT-III proteins were recruited to M. tuberculosis phagosomes in an ESX-1-dependent manner. We previously demonstrated that the mycobacterial effectors EsxG/TB9.8 and EsxH/TB10.4, both secreted by the ESX-3 T7SS, can inhibit ESCRT-dependent trafficking of receptors to the lysosome. Here, we additionally show that ESCRT-III recruitment to sites of endolysosomal damage is antagonized by EsxG and EsxH, both within the context of M. tuberculosis infection and sterile injury. Moreover, EsxG and EsxH themselves respond within minutes to membrane damage in a manner that is independent of calcium and ESCRT-III recruitment. Thus, our study reveals that T7SS effectors and ESCRT participate in a series of measures and countermeasures for control of phagosome integrity.IMPORTANCEMycobacterium tuberculosis causes tuberculosis, which kills more people than any other infection. M. tuberculosis grows in macrophages, cells that specialize in engulfing and degrading microorganisms. Like many intracellular pathogens, in order to cause disease, M. tuberculosis damages the membrane-bound compartment (phagosome) in which it is enclosed after macrophage uptake. Recent work showed that when chemicals damage this type of intracellular compartment, cells rapidly detect and repair the damage, using machinery called the endosomal sorting complex required for transport (ESCRT). Therefore, we hypothesized that ESCRT might also respond to pathogen-induced damage. At the same time, our previous work showed that the EsxG-EsxH heterodimer of M. tuberculosis can inhibit ESCRT, raising the possibility that M. tuberculosis impairs this host response. Here, we show that ESCRT is recruited to damaged M. tuberculosis phagosomes and that EsxG-EsxH undermines ESCRT-mediated endomembrane repair. Thus, our studies demonstrate a battle between host and pathogen over endomembrane integrity.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Endosomal Sorting Complexes Required for Transport/antagonists & inhibitors , Host-Pathogen Interactions , Mycobacterium tuberculosis/pathogenicity , Type VII Secretion Systems/metabolism , Virulence Factors/metabolism , Animals , Cell Line , Endosomal Sorting Complexes Required for Transport/metabolism , Humans , Mice , Mycobacterium tuberculosis/metabolism , Phagosomes/metabolism , Phagosomes/microbiology , Protein Binding
17.
Autophagy ; 14(3): 552-554, 2018.
Article in English | MEDLINE | ID: mdl-29313424

ABSTRACT

M. tuberculosis causes an enormous worldwide burden of disease. Its success depends upon subverting the antimicrobial capacity of macrophages. We have known for decades that M. tuberculosis impairs phagosomal trafficking to avoid lysosomal degradation, but the mechanism is unclear. Recent work has described a phagolysosomal pathway called LC3-associated phagocytosis (LAP), in which LC3 associates with microbe-containing phagosomes. Macrophage pathogen recognition receptors (PRRs) initiate LAP, and NADPH oxidase and RUBCN/RUBICON are required for LAP. We discovered that CpsA, an exported M. tuberculosis virulence factor, blocks LAP by interfering with recruitment of CYBB/NOX2 (cytochrome b-245, beta polypeptide) to the mycobacterial phagosome. In macrophages and in mice, M. tuberculosis mutants lacking cpsA are successfully cleared by NADPH oxidase and the ensuing LC3-associated lysosomal trafficking pathway. CpsA belongs to the LytR-CpsA-Psr family, which is found widely in Gram-positive bacilli. This family is known for its enzymatic role in cell wall assembly. However, our data suggest that CpsA inhibits CYBB oxidase independently of a cell wall function. Thus, CpsA may have evolved from an enzyme involved in cell wall integrity to an indispensable virulence factor that M. tuberculosis uses to evade the innate immune response.


Subject(s)
Autophagy/physiology , Macrophages/metabolism , Mycobacterium tuberculosis/metabolism , Phagocytosis/physiology , Animals , Bacterial Proteins/metabolism , Host-Pathogen Interactions/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Phagosomes/metabolism
18.
Vaccine ; 36(7): 939-944, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29343411

ABSTRACT

Development of a new vaccine against tuberculosis is urgently needed. Recent work has demonstrated that two related LC3-associated trafficking pathways, autophagy and LC3-associated phagocytosis (LAP), enhance antigen presentation and might play a role in vaccine efficacy. Mycobacterium tuberculosis inhibits both LC3-trafficking pathways. Moreover, the vaccine strain, BCG, induces even less LC3-trafficking than M. tuberculosis, which may help explain its limited efficacy. To determine whether enhanced LC3-trafficking can improve efficacy of a live, attenuated M. tuberculosis vaccine, we took advantage of our recent finding that the bacterial virulence factor CpsA inhibits LAP. When we deleted cpsA in the mc26206 vaccine strain, it dramatically increased LC3-trafficking. We compared the protective efficacy of the strain lacking cpsA to the parent strain and to BCG in mice challenged with M. tuberculosis. We found that the strain lacking cpsA generated modestly enhanced protection in the spleen, but overall did not outperform BCG.


Subject(s)
Mycobacterium tuberculosis/immunology , Tuberculosis Vaccines/immunology , Tuberculosis/prevention & control , Vaccines, Attenuated/immunology , Animals , Antigen Presentation/immunology , BCG Vaccine/immunology , Humans , Immunization , Immunogenicity, Vaccine , Macrophages/immunology , Macrophages/microbiology , Mice , Mutation , Mycobacterium tuberculosis/genetics , Phagocytosis
19.
Bio Protoc ; 8(10): e2857, 2018 May 20.
Article in English | MEDLINE | ID: mdl-34285974

ABSTRACT

Macrophages are immune cells that contribute to host defense through various mechanisms including phagocytosis and antigen presentation. Their antimicrobial capacity is subverted by clinically important intracellular pathogens such as Mycobacterium tuberculosis. The study of host-pathogen interactions using these cells is therefore of considerable interest. Such studies often seek to express tagged proteins to characterize their activities, localizations, and protein-protein interactions. Here, we describe a robust method for transient protein expression in macrophages using mRNA lipoplex transfections.

20.
Proc Natl Acad Sci U S A ; 114(41): E8711-E8720, 2017 10 10.
Article in English | MEDLINE | ID: mdl-28973896

ABSTRACT

Mycobacterium tuberculosis' success as a pathogen comes from its ability to evade degradation by macrophages. Normally macrophages clear microorganisms that activate pathogen-recognition receptors (PRRs) through a lysosomal-trafficking pathway called "LC3-associated phagocytosis" (LAP). Although Mtuberculosis activates numerous PRRs, for reasons that are poorly understood LAP does not substantially contribute to Mtuberculosis control. LAP depends upon reactive oxygen species (ROS) generated by NADPH oxidase, but Mtuberculosis fails to generate a robust oxidative response. Here, we show that CpsA, a LytR-CpsA-Psr (LCP) domain-containing protein, is required for Mtuberculosis to evade killing by NADPH oxidase and LAP. Unlike phagosomes containing wild-type bacilli, phagosomes containing the ΔcpsA mutant recruited NADPH oxidase, produced ROS, associated with LC3, and matured into antibacterial lysosomes. Moreover, CpsA was sufficient to impair NADPH oxidase recruitment to fungal particles that are normally cleared by LAP. Intracellular survival of the ΔcpsA mutant was largely restored in macrophages missing LAP components (Nox2, Rubicon, Beclin, Atg5, Atg7, or Atg16L1) but not in macrophages defective in a related, canonical autophagy pathway (Atg14, Ulk1, or cGAS). The ΔcpsA mutant was highly impaired in vivo, and its growth was partially restored in mice deficient in NADPH oxidase, Atg5, or Atg7, demonstrating that CpsA makes a significant contribution to the resistance of Mtuberculosis to NADPH oxidase and LC3 trafficking in vivo. Overall, our findings reveal an essential role of CpsA in innate immune evasion and suggest that LCP proteins have functions beyond their previously known role in cell-wall metabolism.


Subject(s)
Bacterial Proteins/metabolism , Macrophages/immunology , Microtubule-Associated Proteins/physiology , NADPH Oxidase 2/physiology , Phagocytosis/physiology , Tuberculosis/prevention & control , Animals , Autophagy , Bacterial Proteins/genetics , Female , Host-Pathogen Interactions , Macrophages/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Mycobacterium tuberculosis/pathogenicity , Nitric Oxide Synthase Type II/physiology , Phagosomes , Reactive Oxygen Species/metabolism , Tuberculosis/immunology , Tuberculosis/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL
...