Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 189
Filter
1.
Int J Mol Sci ; 25(6)2024 Mar 09.
Article in English | MEDLINE | ID: mdl-38542119

ABSTRACT

Mycobacterium tuberculosis (Mtb) employs various strategies to manipulate the host's cellular machinery, overriding critical molecular mechanisms such as phagosome-lysosome fusion, which are crucial for its destruction. The Protein Kinase C (PKC) signaling pathways play a key role in regulating phagocytosis. Recent research in Interferon-activated macrophages has unveiled that PKC phosphorylates Coronin-1, leading to a shift from phagocytosis to micropinocytosis, ultimately resulting in Mtb destruction. Therefore, this study aims to identify additional PKC targets that may facilitate Mycobacterium bovis (M. bovis) infection in macrophages. Protein extracts were obtained from THP-1 cells, both unstimulated and mycobacterial-stimulated, in the presence or absence of a general PKC inhibitor. We conducted an enrichment of phosphorylated peptides, followed by their identification through mass spectrometry (LC-MS/MS). Our analysis revealed 736 phosphorylated proteins, among which 153 exhibited alterations in their phosphorylation profiles in response to infection in a PKC-dependent manner. Among these 153 proteins, 55 are involved in various cellular processes, including endocytosis, vesicular traffic, autophagy, and programmed cell death. Importantly, our findings suggest that PKC may negatively regulate autophagy by phosphorylating proteins within the mTORC1 pathway (mTOR2/PKC/Raf-1/Tsc2/Raptor/Sequestosome-1) in response to M. bovis BCG infection, thereby promoting macrophage infection.


Subject(s)
Mycobacterium Infections , Mycobacterium bovis , Mycobacterium tuberculosis , Humans , Mycobacterium bovis/physiology , Chromatography, Liquid , Tandem Mass Spectrometry , Macrophages/metabolism , Autophagy , Mycobacterium Infections/metabolism , Protein Kinase C/metabolism
2.
Microbes Infect ; 26(3): 105271, 2024.
Article in English | MEDLINE | ID: mdl-38036036

ABSTRACT

Microorganisms present in the gut modulate host defence responses against infections in order to maintain immune homeostasis. This host-microbe crosstalk is regulated by gut metabolites. Butyrate is one such small chain fatty acid produced by gut microbes upon fermentation that has the potential to influence immune responses. Here we investigated the role of butyrate in macrophages during mycobacterial infection. Results demonstrate that butyrate significantly suppresses the growth kinetics of mycobacteria in culture medium as well as inhibits mycobacterial survival inside macrophages. Interestingly, butyrate alters the pentose phosphate pathway by inducing higher expression of Glucose-6-Phosphate Dehydrogenase (G6PDH) resulting in a higher oxidative burst via decreased Sod-2 and increased Nox-2 (NADPH oxidase-2) expression. Butyrate-induced G6PDH also mediated a decrease in mitochondrial membrane potential. This in turn lead to an induction of apoptosis as measured by lower expression of the anti-apoptotic protein Bcl-2 and a higher release of Cytochrome C as a result of induction of apoptosis. These results indicate that butyrate alters the metabolic status of macrophages and induces protective immune responses against mycobacterial infection.


Subject(s)
Butyrates , Mycobacterium Infections , Humans , Butyrates/pharmacology , Glucosephosphate Dehydrogenase/metabolism , Respiratory Burst , Macrophages/microbiology , Mycobacterium Infections/metabolism , Apoptosis
3.
Cell ; 186(23): 5114-5134.e27, 2023 11 09.
Article in English | MEDLINE | ID: mdl-37875108

ABSTRACT

Human inherited disorders of interferon-gamma (IFN-γ) immunity underlie severe mycobacterial diseases. We report X-linked recessive MCTS1 deficiency in men with mycobacterial disease from kindreds of different ancestries (from China, Finland, Iran, and Saudi Arabia). Complete deficiency of this translation re-initiation factor impairs the translation of a subset of proteins, including the kinase JAK2 in all cell types tested, including T lymphocytes and phagocytes. JAK2 expression is sufficiently low to impair cellular responses to interleukin-23 (IL-23) and partially IL-12, but not other JAK2-dependent cytokines. Defective responses to IL-23 preferentially impair the production of IFN-γ by innate-like adaptive mucosal-associated invariant T cells (MAIT) and γδ T lymphocytes upon mycobacterial challenge. Surprisingly, the lack of MCTS1-dependent translation re-initiation and ribosome recycling seems to be otherwise physiologically redundant in these patients. These findings suggest that X-linked recessive human MCTS1 deficiency underlies isolated mycobacterial disease by impairing JAK2 translation in innate-like adaptive T lymphocytes, thereby impairing the IL-23-dependent induction of IFN-γ.


Subject(s)
Interferon-gamma , Janus Kinase 2 , Mycobacterium Infections , Humans , Male , Cell Cycle Proteins/metabolism , Interferon-gamma/immunology , Interleukin-12 , Interleukin-23 , Janus Kinase 2/metabolism , Mycobacterium/physiology , Mycobacterium Infections/immunology , Mycobacterium Infections/metabolism , Oncogene Proteins/metabolism
4.
Microbes Infect ; 25(6): 105126, 2023.
Article in English | MEDLINE | ID: mdl-36931492

ABSTRACT

Mycobacterium tuberculosis attenuates many defence responses from alveolar macrophages to create a niche at sites of infection in the human lung. Levels of Heat Shock Proteins have been reported to increase many folds in the serum of active TB patients than in latently infected individuals. Here we investigated the regulation of key defence responses by HSPs during mycobacterial infection. We show that infection of macrophages with M. bovis BCG induces higher expression of HSP-27 and HSP-70. Inhibiting HSP-27 and HSP-70 prior to mycobacterial infection leads to a significant decrease in mycobacterial growth inside macrophages. Further, inhibiting HSPs resulted in a significant increase in intracellular oxidative burst levels. This was accompanied by an increase in the levels of T cell activation molecules CD40 and IL-12 receptor and a concomitant decrease in the levels of T cell inhibitory molecules PD-L1 and IL-10 receptor. Furthermore, inhibiting HSPs significantly increased the expression of key proteins in the autophagy pathway along with increased activation of pro-inflammatory promoting transcription factors NF-κB and p-CREB. Interestingly, we also show that both HSP-27 and HSP-70 are associated with anti-apoptotic proteins Bcl-2 and Beclin-1. These results point towards a suppressive role for host HSP-27 and HSP-70 during mycobacterial infection.


Subject(s)
HSP27 Heat-Shock Proteins , HSP70 Heat-Shock Proteins , Macrophages , Mycobacterium Infections , Mycobacterium tuberculosis , Humans , Heat-Shock Proteins/metabolism , Macrophages/microbiology , Mycobacterium Infections/metabolism , Mycobacterium tuberculosis/pathogenicity , T-Lymphocytes , HSP70 Heat-Shock Proteins/immunology , HSP70 Heat-Shock Proteins/metabolism , HSP27 Heat-Shock Proteins/immunology , HSP27 Heat-Shock Proteins/metabolism
5.
Front Immunol ; 13: 891475, 2022.
Article in English | MEDLINE | ID: mdl-35874747

ABSTRACT

Macrophages deploy a variety of antimicrobial programs to contain mycobacterial infection. Upon activation, they undergo extensive metabolic reprogramming to meet an increase in energy demand, but also to support immune effector functions such as secretion of cytokines and antimicrobial activities. Here, we report that mitochondrial import of pyruvate is linked to production of mitochondrial ROS and control of Mycobacterium avium (M. avium) infection in human primary macrophages. Using chemical inhibition, targeted mass spectrometry and single cell image analysis, we showed that macrophages infected with M. avium switch to aerobic glycolysis without any major imbalances in the tricarboxylic acid cycle volume or changes in the energy charge. Instead, we found that pyruvate import contributes to hyperpolarization of mitochondria in infected cells and increases production of mitochondrial reactive oxygen species by the complex I via reverse electron transport, which reduces the macrophage burden of M. avium. While mycobacterial infections are extremely difficult to treat and notoriously resistant to antibiotics, this work stresses out that compounds specifically inducing mitochondrial reactive oxygen species could present themself as valuable adjunct treatments.


Subject(s)
Mycobacterium Infections , Mycobacterium avium-intracellulare Infection , Humans , Macrophages , Mitochondria/metabolism , Mycobacterium Infections/metabolism , Mycobacterium avium/physiology , Proto-Oncogene Proteins c-ret/metabolism , Pyruvic Acid/metabolism , Reactive Oxygen Species/metabolism
6.
Sci Rep ; 12(1): 9681, 2022 06 11.
Article in English | MEDLINE | ID: mdl-35690602

ABSTRACT

Pathogenic mycobacteria including Mycobacterium avium subsp. paratuberculosis (MAP), the causative agent of Johne's disease, manipulate host macrophages to persist and cause disease. In mycobacterial infection, highly plastic macrophages, shift between inflammatory M1 and permissive M2 phenotypes which alter the disease outcome and allow bacteria to survive intracellularly. Here we examine the impact of MAP infection on polarised macrophages and how increased lipid availability alters macrophage phenotype and bacterial persistence. Further, we assess if host microRNA (miRNA) are sensitive to macrophage polarisation state and how MAP can drive their expression to overcome innate responses. Using in vitro MAP infection, we find that increasing lipid availability through supplementing culture media with exogenous lipid increases cellular nitric oxide production. Lipid-associated miRs -19a, -129, -24, and -24-3p are differentially expressed following macrophage polarisation and lipid supplementation and are further regulated during MAP infection. Collectively, our results highlight the importance of host lipid metabolism in MAP infection and demonstrate control of miRNA expression by MAP to favour intracellular persistence.


Subject(s)
MicroRNAs , Mycobacterium Infections , Mycobacterium avium subsp. paratuberculosis , Animals , Lipid Metabolism , Lipids , Macrophages/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Mycobacterium Infections/metabolism
7.
Int J Mol Sci ; 23(5)2022 Feb 24.
Article in English | MEDLINE | ID: mdl-35269631

ABSTRACT

Mycobacterium mucogenicum (Mmuc), a rapidly growing nontuberculous mycobacterium (NTM), can infect humans (posttraumatic wound infections and catheter-related sepsis). Similar to other NTM species, Mmuc exhibits colony morphologies of rough (Mmuc-R) and smooth (Mmuc-S) types. Although there are several case reports on Mmuc infection, no experimental evidence supports that the R-type is more virulent. In addition, the immune response and metabolic reprogramming of Mmuc have not been studied on the basis of morphological characteristics. Thus, a standard ATCC Mmuc strain and two clinical strains were analyzed, and macrophages were generated from mouse bone marrow. Cytokines and cell death were measured by ELISA and FACS, respectively. Mitochondrial respiration and glycolytic changes were measured by XF seahorse. Higher numbers of intracellular bacteria were found in Mmuc-R-infected macrophages than in Mmuc-S-infected macrophages. Additionally, Mmuc-R induced higher levels of the cytokines TNF-α, IL-6, IL-12p40, and IL-10 and induced more BMDM necrotic death. Furthermore, our metabolic data showed marked glycolytic and respiratory differences between the control and each type of Mmuc infection, and changes in these parameters significantly promoted glucose metabolism, extracellular acidification, and oxygen consumption in BMDMs. In conclusion, at least in the strains we tested, Mmuc-R is more virulent, induces a stronger immune response, and shifts bioenergetic metabolism more extensively than the S-type. This study is the first to report differential immune responses and metabolic reprogramming after Mmuc infection and might provide a fundamental basis for additional studies on Mmuc pathogenesis.


Subject(s)
Mycobacteriaceae , Mycobacterium Infections, Nontuberculous , Mycobacterium Infections , Animals , Cytokines/metabolism , Immunity , Macrophages/metabolism , Mice , Mycobacterium Infections/metabolism , Mycobacterium Infections, Nontuberculous/microbiology
8.
Front Immunol ; 13: 791136, 2022.
Article in English | MEDLINE | ID: mdl-35237260

ABSTRACT

The inflammasome complex is important for host defense against intracellular bacterial infections. Mycobacterium tuberculosis (Mtb) is a facultative intracellular bacterium which is able to survive in infected macrophages. Here we discuss how the host cell inflammasomes sense Mtb and other related mycobacterial species. Furthermore, we describe the molecular mechanisms of NLRP3 inflammasome sensing of Mtb which involve the type VII secretion system ESX-1, cell surface lipids (TDM/TDB), secreted effector proteins (LpqH, PPE13, EST12, EsxA) and double-stranded RNA acting on the priming and/or activation steps of inflammasome activation. In contrast, Mtb also mediates inhibition of the NLRP3 inflammasome by limiting exposure of cell surface ligands via its hydrolase, Hip1, by inhibiting the host cell cathepsin G protease via the secreted Mtb effector Rv3364c and finally, by limiting intracellular triggers (K+ and Cl- efflux and cytosolic reactive oxygen species production) via its serine/threonine kinase PknF. In addition, Mtb inhibits the AIM2 inflammasome activation via an unknown mechanism. Overall, there is good evidence for a tug-of-war between Mtb trying to limit inflammasome activation and the host cell trying to sense Mtb and activate the inflammasome. The detailed molecular mechanisms and the importance of inflammasome activation for virulence of Mtb or host susceptibility have not been fully investigated.


Subject(s)
Mycobacterium Infections , Mycobacterium tuberculosis , Humans , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Macrophages/metabolism , Mycobacterium Infections/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
9.
BMC Pulm Med ; 22(1): 57, 2022 Feb 09.
Article in English | MEDLINE | ID: mdl-35139852

ABSTRACT

BACKGROUND: Extracellular vesicles (EVs) play a key role in cell communication and the pathogenesis of some diseases. EVs may accelerate cell death during the course of mycobacterial infection and are also considered as a new vaccine design, drug delivery, and biomarker candidates. The current study evaluates the effects of EVs from serum samples of mycobacteria-infected patients on THP-1 monocytes and PBMC cells. METHOD: EVs were purified from the serum, then cultured separately with THP-1 monocytes and PBMCs. The cell death was determined through annexin V-FITC and PI staining. GW4869, an EVs inhibitor, was used to determine if EVs released from serum could increase THP-1 monocytes cell death. RESULTS: The cell death was significantly increased in the presence of 10 µg/ml and 5 µg/ml concentrations of the purified EVs (p < 0.05). Minimal cell death was determined in 2.5 µg/ml and 1.2 µg/ml (p < 0.05). Up to 85% of the cells were viable in the presence of the GW4869 inhibitor (p < 0.05). CONCLUSION: Direct infection of the cells with EVs released from mycobacteria-infected patients samples, the multiplicity of infection with the EVs, and virulent or avirulent mycobacteria may change the status of the cell death. The isolated EVs  from serum samples of patients with mycobacterial  infection accelerated cell death, which means that they might   not be considered as an optimal tool for developing drug delivery and vaccine against tuberculosis.


Subject(s)
Cell Death , Extracellular Vesicles/metabolism , Mycobacterium Infections/immunology , Mycobacterium/physiology , Adult , Aged, 80 and over , Female , Humans , Leukocytes, Mononuclear , Male , Middle Aged , Mycobacterium/immunology , Mycobacterium Infections/metabolism , Mycobacterium Infections/microbiology , THP-1 Cells , Virulence
10.
Front Immunol ; 12: 747387, 2021.
Article in English | MEDLINE | ID: mdl-34630426

ABSTRACT

Mycobacterial diseases are a major public health challenge. Their causative agents include, in order of impact, members of the Mycobacterium tuberculosis complex (causing tuberculosis), Mycobacterium leprae (causing leprosy), and non-tuberculous mycobacterial pathogens including Mycobacterium ulcerans. Macrophages are mycobacterial targets and they play an essential role in the host immune response to mycobacteria. This review aims to provide a comprehensive understanding of the immune-metabolic adaptations of the macrophage to mycobacterial infections. This metabolic rewiring involves changes in glycolysis and oxidative metabolism, as well as in the use of fatty acids and that of metals such as iron, zinc and copper. The macrophage metabolic adaptations result in changes in intracellular metabolites, which can post-translationally modify proteins including histones, with potential for shaping the epigenetic landscape. This review will also cover how critical tuberculosis co-morbidities such as smoking, diabetes and HIV infection shape host metabolic responses and impact disease outcome. Finally, we will explore how the immune-metabolic knowledge gained in the last decades can be harnessed towards the design of novel diagnostic and therapeutic tools, as well as vaccines.


Subject(s)
Adaptation, Physiological/immunology , Host-Pathogen Interactions/immunology , Macrophages/immunology , Mycobacterium Infections/immunology , Animals , Humans , Macrophages/metabolism , Mycobacterium/immunology , Mycobacterium Infections/metabolism
11.
Front Immunol ; 12: 696491, 2021.
Article in English | MEDLINE | ID: mdl-34322125

ABSTRACT

Mycobacterium tuberculosis (M. tb) is an intracellular pathogen that exploits moonlighting functions of its proteins to interfere with host cell functions. PE/PPE proteins utilize host inflammatory signaling and cell death pathways to promote pathogenesis. We report that M. tb PE6 protein (Rv0335c) is a secretory protein effector that interacts with innate immune toll-like receptor TLR4 on the macrophage cell surface and promotes activation of the canonical NFĸB signaling pathway to stimulate secretion of proinflammatory cytokines TNF-α, IL-12, and IL-6. Using mouse macrophage TLRs knockout cell lines, we demonstrate that PE6 induced secretion of proinflammatory cytokines dependent on TLR4 and adaptor Myd88. PE6 possesses nuclear and mitochondrial targeting sequences and displayed time-dependent differential localization into nucleus/nucleolus and mitochondria, and exhibited strong Nucleolin activation. PE6 strongly induces apoptosis via increased production of pro-apoptotic molecules Bax, Cytochrome C, and pcMyc. Mechanistic details revealed that PE6 activates Caspases 3 and 9 and induces endoplasmic reticulum-associated unfolded protein response pathways to induce apoptosis through increased production of ATF6, Chop, BIP, eIF2α, IRE1α, and Calnexin. Despite being a potent inducer of apoptosis, PE6 suppresses innate immune defense strategy autophagy by inducing inhibitory phosphorylation of autophagy initiating kinase ULK1. Inversely, PE6 induces activatory phosphorylation of autophagy master regulator MtorC1, which is reflected by lower conversion of autophagy markers LC3BI to LC3BII and increased accumulation of autophagy substrate p62 which is also dependent on innate immune receptor TLR4. The use of pharmacological agents, rapamycin and bafilomycin A1, confirms the inhibitory effect of PE6 on autophagy, evidenced by the reduced conversion of LC3BI to LC3BII and increased accumulation of p62 in the presence of rapamycin and bafilomycin A1. We also observed that PE6 binds DNA, which could have significant implications in virulence. Furthermore, our analyses reveal that PE6 efficiently binds iron to likely aid in intracellular survival. Recombinant Mycobacterium smegmatis (M. smegmatis) containing pe6 displayed robust growth in iron chelated media compared to vector alone transformed cells, which suggests a role of PE6 in iron acquisition. These findings unravel novel mechanisms exploited by PE6 protein to subdue host immunity, thereby providing insights relevant to a better understanding of host-pathogen interaction during M. tb infection.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Bacterial Proteins/pharmacology , Inflammation/metabolism , Macrophage Activation/drug effects , Macrophages/drug effects , Mycobacterium Infections/metabolism , Toll-Like Receptor 4/agonists , Animals , Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cytokines/metabolism , HEK293 Cells , Host-Pathogen Interactions , Humans , Inflammation/immunology , Inflammation/microbiology , Inflammation Mediators/metabolism , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Mice , Microbial Viability , Mycobacterium Infections/immunology , Mycobacterium Infections/microbiology , Mycobacterium smegmatis/genetics , Mycobacterium smegmatis/immunology , Mycobacterium smegmatis/metabolism , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , RAW 264.7 Cells , Signal Transduction , THP-1 Cells , Toll-Like Receptor 4/metabolism
12.
Int J Mol Sci ; 22(14)2021 Jul 15.
Article in English | MEDLINE | ID: mdl-34299217

ABSTRACT

The mycobacterial cell wall is composed of large amounts of lipids with varying moieties. Some mycobacteria species hijack host cells and promote lipid droplet accumulation to build the cellular environment essential for their intracellular survival. Thus, lipids are thought to be important for mycobacteria survival as well as for the invasion, parasitization, and proliferation within host cells. However, their physiological roles have not been fully elucidated. Recent studies have revealed that mycobacteria modulate the peroxisome proliferator-activated receptor (PPAR) signaling and utilize host-derived triacylglycerol (TAG) and cholesterol as both nutrient sources and evasion from the host immune system. In this review, we discuss recent findings that describe the activation of PPARs by mycobacterial infections and their role in determining the fate of bacilli by inducing lipid metabolism, anti-inflammatory function, and autophagy.


Subject(s)
Mycobacterium Infections/microbiology , Mycobacterium/metabolism , Peroxisome Proliferator-Activated Receptors/metabolism , Animals , Autophagy/physiology , Cholesterol/metabolism , Humans , Lipid Metabolism , Mycobacterium/growth & development , Mycobacterium/immunology , Mycobacterium Infections/immunology , Mycobacterium Infections/metabolism , Peroxisome Proliferator-Activated Receptors/genetics , Signal Transduction
13.
Biomol Concepts ; 12(1): 94-109, 2021 Jul 25.
Article in English | MEDLINE | ID: mdl-34304400

ABSTRACT

We previously reported that M. tb on its own as well as together with HIV inhibits macrophage apoptosis by upregulating the expression of Bcl2 and Inhibitor of Apoptosis (IAP). In addition, recent reports from our lab showed that stimulation of either macrophages or BMDCs results in the significant upregulation of Bcl2. In this report, we delineate the role of Bcl2 in mediating defense responses from dendritic cells (BMDCs) during mycobacterial infection. Inhibiting Bcl2 led to a significant decrease in intracellular bacterial burden in BMDCs. To further characterize the role of Bcl2 in modulating defense responses, we inhibited Bcl2 in BMDCs as well as human PBMCs to monitor their activation and functional status in response to mycobacterial infection and stimulation with M. tb antigen Rv3416. Inhibiting Bcl2 generated protective responses including increased expression of co-stimulatory molecules, oxidative burst, pro-inflammatory cytokine expression and autophagy. Finally, co-culturing human PBMCs and BMDCs with antigen-primed T cells increased their proliferation, activation and effector function. These results point towards a critical role for Bcl2 in regulating BMDCs defense responses to mycobacterial infection.


Subject(s)
Dendritic Cells/immunology , Immunity , Macrophages/immunology , Mycobacterium Infections/immunology , Mycobacterium bovis/immunology , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Apoptosis , Autophagy , Cytokines/metabolism , Dendritic Cells/metabolism , Dendritic Cells/microbiology , Dendritic Cells/pathology , Female , Humans , Macrophages/metabolism , Macrophages/microbiology , Macrophages/pathology , Mice , Mice, Inbred BALB C , Mycobacterium Infections/metabolism , Mycobacterium Infections/microbiology , Mycobacterium Infections/pathology , Proto-Oncogene Proteins c-bcl-2/genetics
14.
Int J Mol Sci ; 22(11)2021 May 22.
Article in English | MEDLINE | ID: mdl-34067256

ABSTRACT

Tumor necrosis factor (TNF) is one of the main cytokines regulating a pro-inflammatory environment. It has been related to several cell functions, for instance, phagocytosis, apoptosis, proliferation, mitochondrial dynamic. Moreover, during mycobacterial infections, TNF plays an essential role to maintain granuloma formation. Several effector mechanisms have been implicated according to the interactions of the two active forms, soluble TNF (solTNF) and transmembrane TNF (tmTNF), with their receptors TNFR1 and TNFR2. We review the impact of these interactions in the context of mycobacterial infections. TNF is tightly regulated by binding to receptors, however, during mycobacterial infections, upstream activation signalling pathways may be influenced by key regulatory factors either at the membrane or cytosol level. Detailing the structure and activation pathways used by TNF and its receptors, such as its interaction with solTNF/TNFRs versus tmTNF/TNFRs, may bring a better understanding of the molecular mechanisms involved in activation pathways which can be helpful for the development of new therapies aimed at being more efficient against mycobacterial infections.


Subject(s)
Membrane Proteins/metabolism , Mycobacterium Infections/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Membrane/metabolism , Humans , Signal Transduction/physiology
15.
PLoS Pathog ; 17(1): e1009124, 2021 01.
Article in English | MEDLINE | ID: mdl-33411813

ABSTRACT

Mycobacterial pathogens pose a sustained threat to human health. There is a critical need for new diagnostics, therapeutics, and vaccines targeting both tuberculous and nontuberculous mycobacterial species. Understanding the basic mechanisms used by diverse mycobacterial species to cause disease will facilitate efforts to design new approaches toward detection, treatment, and prevention of mycobacterial disease. Molecular, genetic, and biochemical approaches have been widely employed to define fundamental aspects of mycobacterial physiology and virulence. The recent expansion of genetic tools in mycobacteria has further increased the accessibility of forward genetic approaches. Proteomics has also emerged as a powerful approach to further our understanding of diverse mycobacterial species. Detection of large numbers of proteins and their modifications from complex mixtures of mycobacterial proteins is now routine, with efforts of quantification of these datasets becoming more robust. In this review, we discuss the "genetic proteome," how the power of genetics, molecular biology, and biochemistry informs and amplifies the quality of subsequent analytical approaches and maximizes the potential of hypothesis-driven mycobacterial research. Published proteomics datasets can be used for hypothesis generation and effective post hoc supplementation to experimental data. Overall, we highlight how the integration of proteomics, genetic, molecular, and biochemical approaches can be employed successfully to define fundamental aspects of mycobacterial pathobiology.


Subject(s)
Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Genomics , Mycobacterium Infections/metabolism , Mycobacterium/metabolism , Proteome/metabolism , Humans , Mycobacterium/genetics , Mycobacterium/pathogenicity , Mycobacterium Infections/genetics , Mycobacterium Infections/microbiology , Proteome/analysis
16.
Front Immunol ; 12: 782495, 2021.
Article in English | MEDLINE | ID: mdl-35003097

ABSTRACT

The mycobacterium genus contains a broad range of species, including the human pathogens M. tuberculosis and M. leprae. These bacteria are best known for their residence inside host cells. Neutrophils are frequently observed at sites of mycobacterial infection, but their role in clearance is not well understood. In this review, we discuss how neutrophils attempt to control mycobacterial infections, either through the ingestion of bacteria into intracellular phagosomes, or the release of neutrophil extracellular traps (NETs). Despite their powerful antimicrobial activity, including the production of reactive oxidants such as hypochlorous acid, neutrophils appear ineffective in killing pathogenic mycobacteria. We explore mycobacterial resistance mechanisms, and how thwarting neutrophil action exacerbates disease pathology. A better understanding of how mycobacteria protect themselves from neutrophils will aid the development of novel strategies that facilitate bacterial clearance and limit host tissue damage.


Subject(s)
Host-Pathogen Interactions/immunology , Mycobacterium Infections/immunology , Mycobacterium Infections/microbiology , Mycobacterium/immunology , Neutrophils/immunology , Neutrophils/metabolism , Animals , Biomarkers , Cytotoxicity, Immunologic , Disease Susceptibility/immunology , Extracellular Traps/genetics , Extracellular Traps/immunology , Host-Pathogen Interactions/genetics , Humans , Immunity, Innate , Mycobacterium Infections/diagnosis , Mycobacterium Infections/metabolism , Neutrophil Activation/genetics , Neutrophil Activation/immunology , Neutrophils/microbiology , Neutrophils/pathology , Oxidants/metabolism , Oxidative Stress , Phagocytosis/genetics , Phagocytosis/immunology , Phagosomes/metabolism
17.
Biochim Biophys Acta Gen Subj ; 1865(2): 129806, 2021 02.
Article in English | MEDLINE | ID: mdl-33253803

ABSTRACT

BACKGROUND: Host-directed therapy is considered a novel anti-tuberculosis strategy in tackling the tuberculosis burden through autophagy induction by various inducers to curtail the growth of intracellular Mycobacterium tuberculosis. METHODS: In this study, we investigated the anti-tubercular role of soybean lectin, a lectin isolated from Glycine max (Soybean). Effect of SBL on intracellular mycobacterial viability through autophagy and the mechanism involved in differentiated THP-1 cells was studied using different experimental approaches. RESULTS: We initially performed a time kinetic experiment with the non-cytotoxic dose of SBL (20 µg/ml) and observed autophagy induction after 24 h of treatment. Abrogation of autophagy in the presence of 3-MA and an increase in LC3 puncta formation upon Baf-A1 addition elucidated the specific effect on autophagy and autophagic flux. SBL treatment also led to autophagy induction in mycobacteria infected macrophages that restricted the intracellular mycobacterial growth, thus emphasizing the host defensive role of SBL induced autophagy. Mechanistic studies revealed an increase in P2RX7 expression, NF-κB activation and reactive oxygen species generation upon SBL treatment. Inhibition of P2RX7 expression suppressed NF-κB dependent ROS level in SBL treated cells. Moreover, SBL induced autophagy was abrogated in the presence of either different inhibitors or P2RX7 siRNA, leading to the reduced killing of intracellular mycobacteria. CONCLUSION: Taken together, these results conclude that SBL induced autophagy exerts an anti-mycobacterial effect in P2RX7-NF-κB dependent manner through the generation of ROS. GENERAL SIGNIFICANCE: This study has provided a novel anti-mycobacterial role of SBL, which may play an important role in devising new therapeutic interventions.


Subject(s)
Anti-Bacterial Agents/pharmacology , Mycobacterium/drug effects , NF-kappa B/metabolism , Plant Lectins/pharmacology , Signal Transduction/drug effects , Soybean Proteins/pharmacology , Anti-Bacterial Agents/isolation & purification , Antitubercular Agents/isolation & purification , Antitubercular Agents/pharmacology , Autophagy/drug effects , Cell Line , Humans , Macrophages/microbiology , Models, Molecular , Mycobacterium/physiology , Mycobacterium Infections/drug therapy , Mycobacterium Infections/metabolism , Mycobacterium Infections/microbiology , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/physiology , Plant Lectins/isolation & purification , Reactive Oxygen Species/metabolism , Soybean Proteins/isolation & purification , Glycine max/chemistry , Tuberculosis/drug therapy , Tuberculosis/metabolism , Tuberculosis/microbiology
18.
Cell Rep ; 31(11): 107772, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32553163

ABSTRACT

ISG15 is a ubiquitin-like modifier that also functions extracellularly, signaling through the LFA-1 integrin to promote interferon (IFN)-γ release from natural killer (NK) and T cells. The signals that lead to the production of extracellular ISG15 and the relationship between its two core functions remain unclear. We show that both epithelial cells and lymphocytes can secrete ISG15, which then signals in either an autocrine or paracrine manner to LFA-1-expressing cells. Microbial pathogens and Toll-like receptor (TLR) agonists result in both IFN-ß-dependent and -independent secretion of ISG15, and residues required for ISG15 secretion are mapped. Intracellular ISGylation inhibits secretion, and viral effector proteins, influenza B NS1, and viral de-ISGylases, including SARS-CoV-2 PLpro, have opposing effects on secretion of ISG15. These results establish extracellular ISG15 as a cytokine-like protein that bridges early innate and IFN-γ-dependent immune responses, and indicate that pathogens have evolved to differentially inhibit the intracellular and extracellular functions of ISG15.


Subject(s)
Cytokines/metabolism , Signal Transduction , Ubiquitins/metabolism , Animals , HEK293 Cells , Humans , Influenza, Human/immunology , Influenza, Human/metabolism , Interferon-gamma/immunology , Interferon-gamma/metabolism , Jurkat Cells , Mice , Mice, Inbred C57BL , Mycobacterium Infections/immunology , Mycobacterium Infections/metabolism , Pathogen-Associated Molecular Pattern Molecules , Typhoid Fever/immunology , Typhoid Fever/metabolism , Viral Nonstructural Proteins/metabolism
19.
Biochim Biophys Acta Proteins Proteom ; 1868(10): 140470, 2020 10.
Article in English | MEDLINE | ID: mdl-32535275

ABSTRACT

Mycobacterium is known for subverting the host defense machinery, and one such mechanism is the inhibition of autophagy. Here, we have demonstrated that Mycobacterium tuberculosis (MTB) secretes a virulence factor; an early secretory antigenic target protein (ESAT-6) into the phagosome, which induces the expression and activity of mitochondrial superoxide dismutase (SOD-2) of macrophages. Using a series of experiments, and Mycobacterium bovis BCG as a model strain (where ESAT-6 protein is not expressed), we have delineated that the protein regulates SOD-2 of macrophages. The expression and augmentation of SOD-2 activity were confirmed by either incubating the macrophages with ESAT-6 protein, transfection of macrophage by esat6 gene using a eukaryotic promoter vector, or by infection with different mycobacterial strains. The induction of acidification of phagosomal compartment containing bacteria was observed in cells that express low levels of SOD-2. This was further confirmed by observing a significant decrease in the M. bovis BCG intracellular load in the sod-2 knocked-down macrophages.


Subject(s)
Antigens, Bacterial/metabolism , Autophagy , Bacterial Proteins/metabolism , Host-Pathogen Interactions , Mycobacterium Infections/metabolism , Mycobacterium Infections/microbiology , Mycobacterium bovis/physiology , Superoxide Dismutase/metabolism , Animals , Autophagosomes , Cell Line , Enzyme Activation , Macrophages/immunology , Macrophages/metabolism , Mice , Microbial Viability , Recombination, Genetic , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
20.
Article in English | MEDLINE | ID: mdl-32229179

ABSTRACT

Lipid droplets (LDs) are organelles that have multiple roles in inflammatory and infectious diseases. LD act as essential platforms for immunometabolic regulation, including as sites for lipid storage and metabolism, inflammatory lipid mediator production, and signaling pathway compartmentalization. Accumulating evidence indicates that intracellular pathogens may exploit host LDs as source of nutrients and as part of their strategy to promote immune evasion. Notably, numerous studies have demonstrated the interaction between LDs and pathogen-containing phagosomes. However, the mechanism involved in this phenomenon remains elusive. Here, we observed LDs and PLIN2 surrounding M. bovis BCG-containing phagosomes, which included observations of a bacillus cell surrounded by lipid content inside a phagosome and LAM from mycobacteria co-localizing with LDs; these results were suggestive of exchange of contents between these compartments. By using beads coated with M.tb lipids, we demonstrated that LD-phagosome associations are regulated through the mycobacterial cell wall components LAM and PIM. In addition, we demonstrated that Rab7 and RILP, but not Rab5, localizes to LDs of infected macrophages and observed the presence of Rab7 at the site of interaction with an infected phagosome. Moreover, treatment of macrophages with the Rab7 inhibitor CID1067700 significantly inhibited the association between LDs and LAM-coated beads. Altogether, our data demonstrate that LD-phagosome interactions are controlled by mycobacterial cell wall components and Rab7, which enables the exchange of contents between LDs and phagosomes and may represent a fundamental aspect of bacterial pathogenesis and immune evasion.


Subject(s)
Lipid Droplets/metabolism , Mycobacterium Infections/metabolism , Mycobacterium tuberculosis/metabolism , Phagosomes/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis/cytology , rab7 GTP-Binding Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...