Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.407
Filtrer
1.
Front Immunol ; 15: 1402024, 2024.
Article de Anglais | MEDLINE | ID: mdl-38873598

RÉSUMÉ

Mycobacterium tuberculosis (Mtb) is an intracellular pathogen capable of adapting and surviving within macrophages, utilizing host nutrients for its growth and replication. Cholesterol is the main carbon source during the infection process of Mtb. Cholesterol metabolism in macrophages is tightly associated with cell functions such as phagocytosis of pathogens, antigen presentation, inflammatory responses, and tissue repair. Research has shown that Mtb infection increases the uptake of low-density lipoprotein (LDL) and cholesterol by macrophages, and enhances de novo cholesterol synthesis in macrophages. Excessive cholesterol is converted into cholesterol esters, while the degradation of cholesterol esters in macrophages is inhibited by Mtb. Furthermore, Mtb infection suppresses the expression of ATP-binding cassette (ABC) transporters in macrophages, impeding cholesterol efflux. These alterations result in the massive accumulation of cholesterol in macrophages, promoting the formation of lipid droplets and foam cells, which ultimately facilitates the persistent survival of Mtb and the progression of tuberculosis (TB), including granuloma formation, tissue cavitation, and systemic dissemination. Mtb infection may also promote the conversion of cholesterol into oxidized cholesterol within macrophages, with the oxidized cholesterol exhibiting anti-Mtb activity. Recent drug development has discovered that reducing cholesterol levels in macrophages can inhibit the invasion of Mtb into macrophages and increase the permeability of anti-tuberculosis drugs. The development of drugs targeting cholesterol metabolic pathways in macrophages, as well as the modification of existing drugs, holds promise for the development of more efficient anti-tuberculosis medications.


Sujet(s)
Cholestérol , Macrophages , Mycobacterium tuberculosis , Tuberculose , Mycobacterium tuberculosis/immunologie , Cholestérol/métabolisme , Humains , Macrophages/immunologie , Macrophages/métabolisme , Macrophages/microbiologie , Tuberculose/immunologie , Tuberculose/métabolisme , Tuberculose/microbiologie , Animaux , Interactions hôte-pathogène/immunologie , Antituberculeux/pharmacologie , Antituberculeux/usage thérapeutique , Métabolisme lipidique
2.
Respir Res ; 25(1): 254, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38907347

RÉSUMÉ

Tuberculosis (TB) remains the second leading cause of death from a single infectious agent and long-term medication could lead to antituberculosis drug-induced liver injury (ATB-DILI). We established a prospective longitudinal cohort of ATB-DILI with multiple timepoint blood sampling and used untargeted metabolomics to analyze the metabolic profiles of 107 plasma samples from healthy controls and newly diagnosed TB patients who either developed ATB-DILI within 2 months of anti-TB treatment (ATB-DILI subjects) or completed their treatment without any adverse drug reaction (ATB-Ctrl subjects). The untargeted metabolome revealed that 77 metabolites (of 895 total) were significantly changed with ATB-DILI progression. Among them, levels of multiple fatty acids and bile acids significantly increased over time in ATB-DILI subjects. Meanwhile, metabolites of the same class were highly correlated with each other and pathway analysis indicated both fatty acids metabolism and bile acids metabolism were up-regulated with ATB-DILI progression. The targeted metabolome further validated that 5 fatty acids had prediction capability at the early stage of the disease and 6 bile acids had a better diagnostic performance when ATB-DILI occurred. These findings provide evidence indicating that fatty acids metabolism and bile acids metabolism play a vital role during ATB-DILI progression. Our report adds a dynamic perspective better to understand the pathological process of ATB-DILI in clinical settings.


Sujet(s)
Antituberculeux , Marqueurs biologiques , Lésions hépatiques dues aux substances , Métabolomique , Humains , Antituberculeux/effets indésirables , Mâle , Métabolomique/méthodes , Femelle , Lésions hépatiques dues aux substances/sang , Lésions hépatiques dues aux substances/diagnostic , Lésions hépatiques dues aux substances/métabolisme , Études longitudinales , Adulte , Adulte d'âge moyen , Marqueurs biologiques/sang , Études prospectives , Valeur prédictive des tests , Tuberculose/traitement médicamenteux , Tuberculose/sang , Tuberculose/métabolisme , Acides et sels biliaires/sang , Acides et sels biliaires/métabolisme
3.
Front Immunol ; 15: 1413947, 2024.
Article de Anglais | MEDLINE | ID: mdl-38881887

RÉSUMÉ

CD36 is a scavenger receptor that has been reported to function as a signaling receptor that responds to pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs) and could integrate metabolic pathways and cell signaling through its dual functions. Thereby influencing activation to regulate the immune response and immune cell differentiation. Recent studies have revealed that CD36 plays critical roles in the process of lipid metabolism, inflammatory response and immune process caused by Mycobacterium tuberculosis infection. This review will comprehensively investigate CD36's functions in lipid uptake and processing, inflammatory response, immune response and therapeutic targets and biomarkers in the infection process of M. tuberculosis. The study also raised outstanding issues in this field to designate future directions.


Sujet(s)
Antigènes CD36 , Mycobacterium tuberculosis , Tuberculose , Humains , Antigènes CD36/métabolisme , Mycobacterium tuberculosis/immunologie , Tuberculose/immunologie , Tuberculose/métabolisme , Tuberculose/microbiologie , Animaux , Métabolisme lipidique , Transduction du signal , Marqueurs biologiques , Interactions hôte-pathogène/immunologie
4.
Elife ; 122024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-38922679

RÉSUMÉ

During tuberculosis (TB), migration of dendritic cells (DCs) from the site of infection to the draining lymph nodes is known to be impaired, hindering the rapid development of protective T-cell-mediated immunity. However, the mechanisms involved in the delayed migration of DCs during TB are still poorly defined. Here, we found that infection of DCs with Mycobacterium tuberculosis (Mtb) triggers HIF1A-mediated aerobic glycolysis in a TLR2-dependent manner, and that this metabolic profile is essential for DC migration. In particular, the lactate dehydrogenase inhibitor oxamate and the HIF1A inhibitor PX-478 abrogated Mtb-induced DC migration in vitro to the lymphoid tissue-specific chemokine CCL21, and in vivo to lymph nodes in mice. Strikingly, we found that although monocytes from TB patients are inherently biased toward glycolysis metabolism, they differentiate into poorly glycolytic and poorly migratory DCs compared with healthy subjects. Taken together, these data suggest that because of their preexisting glycolytic state, circulating monocytes from TB patients are refractory to differentiation into migratory DCs, which may explain the delayed migration of these cells during the disease and opens avenues for host-directed therapies for TB.


Sujet(s)
Mouvement cellulaire , Cellules dendritiques , Glycolyse , Sous-unité alpha du facteur-1 induit par l'hypoxie , Monocytes , Mycobacterium tuberculosis , Tuberculose , Cellules dendritiques/métabolisme , Cellules dendritiques/immunologie , Monocytes/métabolisme , Monocytes/immunologie , Humains , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Mycobacterium tuberculosis/immunologie , Animaux , Tuberculose/immunologie , Tuberculose/métabolisme , Tuberculose/microbiologie , Souris , Récepteur de type Toll-2/métabolisme , Souris de lignée C57BL , Femelle
5.
Nat Commun ; 15(1): 5239, 2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-38937448

RÉSUMÉ

Tuberculosis remains a large global disease burden for which treatment regimens are protracted and monitoring of disease activity difficult. Existing detection methods rely almost exclusively on bacterial culture from sputum which limits sampling to organisms on the pulmonary surface. Advances in monitoring tuberculous lesions have utilized the common glucoside [18F]FDG, yet lack specificity to the causative pathogen Mycobacterium tuberculosis (Mtb) and so do not directly correlate with pathogen viability. Here we show that a close mimic that is also positron-emitting of the non-mammalian Mtb disaccharide trehalose - 2-[18F]fluoro-2-deoxytrehalose ([18F]FDT) - is a mechanism-based reporter of Mycobacteria-selective enzyme activity in vivo. Use of [18F]FDT in the imaging of Mtb in diverse models of disease, including non-human primates, successfully co-opts Mtb-mediated processing of trehalose to allow the specific imaging of TB-associated lesions and to monitor the effects of treatment. A pyrogen-free, direct enzyme-catalyzed process for its radiochemical synthesis allows the ready production of [18F]FDT from the most globally-abundant organic 18F-containing molecule, [18F]FDG. The full, pre-clinical validation of both production method and [18F]FDT now creates a new, bacterium-selective candidate for clinical evaluation. We anticipate that this distributable technology to generate clinical-grade [18F]FDT directly from the widely-available clinical reagent [18F]FDG, without need for either custom-made radioisotope generation or specialist chemical methods and/or facilities, could now usher in global, democratized access to a TB-specific PET tracer.


Sujet(s)
Mycobacterium tuberculosis , Tomographie par émission de positons , Tréhalose , Tuberculose , Animaux , Mycobacterium tuberculosis/métabolisme , Tomographie par émission de positons/méthodes , Tréhalose/métabolisme , Tuberculose/imagerie diagnostique , Tuberculose/microbiologie , Tuberculose/métabolisme , Humains , Souris , Radio-isotopes du fluor , Fluorodésoxyglucose F18/métabolisme , Fluorodésoxyglucose F18/composition chimique , Radiopharmaceutiques/métabolisme , Modèles animaux de maladie humaine , Femelle
6.
Front Cell Infect Microbiol ; 14: 1398077, 2024.
Article de Anglais | MEDLINE | ID: mdl-38836056

RÉSUMÉ

Mycobacterium tuberculosis (M.tb), the causative agent of Tuberculosis, is an intracellular bacterium well known for its ability to subvert host energy and metabolic pathways to maintain its intracellular survival. For this purpose, the bacteria utilize various mechanisms of which extracellular vehicles (EVs) related mechanisms attracted more attention. EVs are nanosized particles that are released by almost all cell types containing active biomolecules from the cell of origin and can target bioactive pathways in the recipient cells upon uptake. It is hypothesized that M.tb dictates the processes of host EV biogenesis pathways, selectively incorporating its molecules into the host EV to direct immune responses in its favor. During infection with Mtb, both mycobacteria and host cells release EVs. The composition of these EVs varies over time, influenced by the physiological and nutritional state of the host environment. Additionally, different EV populations contribute differently to the pathogenesis of disease at various stages of illness participating in a complex interplay between host cells and pathogens. These interactions ultimately influence immune responses and disease outcomes. However, the precise mechanisms and roles of EVs in pathogenicity and disease outcomes remain to be fully elucidated. In this review, we explored the properties and function of EVs in the context of M.tb infection within the host microenvironment and discussed their capacity as a novel therapeutic strategy to combat tuberculosis.


Sujet(s)
Vésicules extracellulaires , Interactions hôte-pathogène , Mycobacterium tuberculosis , Tuberculose , Vésicules extracellulaires/métabolisme , Vésicules extracellulaires/immunologie , Mycobacterium tuberculosis/immunologie , Humains , Tuberculose/immunologie , Tuberculose/microbiologie , Tuberculose/métabolisme , Interactions hôte-pathogène/immunologie , Animaux
7.
PLoS Pathog ; 20(5): e1012205, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38701094

RÉSUMÉ

Mycobacterium tuberculosis (Mtb) infects lung myeloid cells, but the specific Mtb-permissive cells and host mechanisms supporting Mtb persistence during chronic infection are incompletely characterized. We report that after the development of T cell responses, CD11clo monocyte-derived cells harbor more live Mtb than alveolar macrophages (AM), neutrophils, and CD11chi monocyte-derived cells. Transcriptomic and functional studies revealed that the lysosome pathway is underexpressed in this highly permissive subset, characterized by less lysosome content, acidification, and proteolytic activity than AM, along with less nuclear TFEB, a regulator of lysosome biogenesis. Mtb infection does not drive lysosome deficiency in CD11clo monocyte-derived cells but promotes recruitment of monocytes that develop into permissive lung cells, mediated by the Mtb ESX-1 secretion system. The c-Abl tyrosine kinase inhibitor nilotinib activates TFEB and enhances lysosome functions of macrophages in vitro and in vivo, improving control of Mtb infection. Our results suggest that Mtb exploits lysosome-poor lung cells for persistence and targeting lysosome biogenesis is a potential host-directed therapy for tuberculosis.


Sujet(s)
Lysosomes , Macrophages alvéolaires , Monocytes , Mycobacterium tuberculosis , Lysosomes/métabolisme , Lysosomes/microbiologie , Animaux , Monocytes/métabolisme , Monocytes/microbiologie , Souris , Macrophages alvéolaires/microbiologie , Macrophages alvéolaires/métabolisme , Poumon/microbiologie , Poumon/métabolisme , Souris de lignée C57BL , Maladie chronique , Tuberculose pulmonaire/microbiologie , Tuberculose pulmonaire/métabolisme , Tuberculose pulmonaire/immunologie , Tuberculose pulmonaire/anatomopathologie , Humains , Tuberculose/microbiologie , Tuberculose/immunologie , Tuberculose/métabolisme , Facteurs de transcription à motifs basiques hélice-boucle-hélice et à glissière à leucines/métabolisme
8.
PLoS Pathog ; 20(5): e1012148, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38728367

RÉSUMÉ

Previously, we found that Mycobacterium tuberculosis (Mtb) infection in type 2 diabetes mellitus (T2DM) mice enhances inflammatory cytokine production which drives pathological immune responses and mortality. In the current study, using a T2DM Mtb infection mice model, we determined the mechanisms that make T2DM mice alveolar macrophages (AMs) more inflammatory upon Mtb infection. Among various cell death pathways, necroptosis is a major pathway involved in inflammatory cytokine production by T2DM mice AMs. Anti-TNFR1 antibody treatment of Mtb-infected AMs from T2DM mice significantly reduced expression of receptor interacting protein kinase 3 (RIPK3) and mixed lineage kinase domain-like (MLKL) (necroptosis markers) and IL-6 production. Metabolic profile comparison of Mtb-infected AMs from T2DM mice and Mtb-infected AMs of nondiabetic control mice indicated that 2-ketohexanoic acid and deoxyadenosine monophosphate were significantly abundant, and acetylcholine and pyridoxine (Vitamin B6) were significantly less abundant in T2DM mice AMs infected with Mtb. 2-Ketohexanoic acid enhanced expression of TNFR1, RIPK3, MLKL and inflammatory cytokine production in the lungs of Mtb-infected nondiabetic mice. In contrast, pyridoxine inhibited RIPK3, MLKL and enhanced expression of Caspase 3 (apoptosis marker) in the lungs of Mtb-infected T2DM mice. Our findings demonstrate that metabolic changes in Mtb-infected T2DM mice enhance TNFR1-mediated necroptosis of AMs, which leads to excess inflammation and lung pathology.


Sujet(s)
Diabète de type 2 , Mycobacterium tuberculosis , Nécroptose , Animaux , Souris , Diabète de type 2/métabolisme , Diabète de type 2/microbiologie , Macrophages alvéolaires/métabolisme , Macrophages alvéolaires/immunologie , Macrophages alvéolaires/microbiologie , Souris de lignée C57BL , Tuberculose/immunologie , Tuberculose/métabolisme , Tuberculose/microbiologie , Diabète expérimental/métabolisme , Diabète expérimental/microbiologie , Receptor-Interacting Protein Serine-Threonine Kinases/métabolisme , Mâle , Cytokines/métabolisme
9.
PLoS Pathog ; 20(5): e1012214, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38722857

RÉSUMÉ

Epithelial cells function as the primary line of defense against invading pathogens. However, bacterial pathogens possess the ability to compromise this barrier and facilitate the transmigration of bacteria. Nonetheless, the specific molecular mechanism employed by Mycobacterium tuberculosis (M.tb) in this process is not fully understood. Here, we investigated the role of Rv2569c in M.tb translocation by assessing its ability to cleave E-cadherin, a crucial component of cell-cell adhesion junctions that are disrupted during bacterial invasion. By utilizing recombinant Rv2569c expressed in Escherichia coli and subsequently purified through affinity chromatography, we demonstrated that Rv2569c exhibited cell wall-associated serine protease activity. Furthermore, Rv2569c was capable of degrading a range of protein substrates, including casein, fibrinogen, fibronectin, and E-cadherin. We also determined that the optimal conditions for the protease activity of Rv2569c occurred at a temperature of 37°C and a pH of 9.0, in the presence of MgCl2. To investigate the function of Rv2569c in M.tb, a deletion mutant of Rv2569c and its complemented strains were generated and used to infect A549 cells and mice. The results of the A549-cell infection experiments revealed that Rv2569c had the ability to cleave E-cadherin and facilitate the transmigration of M.tb through polarized A549 epithelial cell layers. Furthermore, in vivo infection assays demonstrated that Rv2569c could disrupt E-cadherin, enhance the colonization of M.tb, and induce pathological damage in the lungs of C57BL/6 mice. Collectively, these results strongly suggest that M.tb employs the serine protease Rv2569c to disrupt epithelial defenses and facilitate its systemic dissemination by crossing the epithelial barrier.


Sujet(s)
Protéines bactériennes , Cadhérines , Cellules épithéliales , Mycobacterium tuberculosis , Protéases à sérine , Cadhérines/métabolisme , Mycobacterium tuberculosis/pathogénicité , Mycobacterium tuberculosis/métabolisme , Animaux , Humains , Souris , Protéases à sérine/métabolisme , Protéases à sérine/génétique , Cellules épithéliales/métabolisme , Cellules épithéliales/microbiologie , Protéines bactériennes/métabolisme , Protéines bactériennes/génétique , Cellules A549 , Tuberculose/microbiologie , Tuberculose/métabolisme , Femelle
10.
Life Sci Alliance ; 7(7)2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38803236

RÉSUMÉ

Neutrophils can be beneficial or deleterious during tuberculosis (TB). Based on the expression of MHC-II and programmed death ligand 1 (PD-L1), we distinguished two functionally and transcriptionally distinct neutrophil subsets in the lungs of mice infected with mycobacteria. Inflammatory [MHC-II-, PD-L1lo] neutrophils produced inflammasome-dependent IL-1ß in the lungs in response to virulent mycobacteria and "accelerated" deleterious inflammation, which was highly exacerbated in IFN-γR-/- mice. Regulatory [MHC-II+, PD-L1hi] neutrophils "brake" inflammation by suppressing T-cell proliferation and IFN-γ production. Such beneficial regulation, which depends on PD-L1, is controlled by IFN-γR signaling in neutrophils. The hypervirulent HN878 strain from the Beijing genotype curbed PD-L1 expression by regulatory neutrophils, abolishing the braking function and driving deleterious hyperinflammation in the lungs. These findings add a layer of complexity to the roles played by neutrophils in TB and may explain the reactivation of this disease observed in cancer patients treated with anti-PD-L1.


Sujet(s)
Antigène CD274 , Inflammation , Interleukine-1 bêta , Poumon , Granulocytes neutrophiles , Tuberculose , Animaux , Antigène CD274/métabolisme , Antigène CD274/génétique , Granulocytes neutrophiles/immunologie , Granulocytes neutrophiles/métabolisme , Souris , Interleukine-1 bêta/métabolisme , Inflammation/immunologie , Inflammation/métabolisme , Tuberculose/immunologie , Tuberculose/microbiologie , Tuberculose/métabolisme , Poumon/immunologie , Poumon/microbiologie , Poumon/métabolisme , Poumon/anatomopathologie , Souris de lignée C57BL , Souris knockout , Mycobacterium tuberculosis/immunologie , Modèles animaux de maladie humaine , Femelle , Humains
11.
Molecules ; 29(9)2024 Apr 29.
Article de Anglais | MEDLINE | ID: mdl-38731549

RÉSUMÉ

Targeting translation factor proteins holds promise for developing innovative anti-tuberculosis drugs. During protein translation, many factors cause ribosomes to stall at messenger RNA (mRNA). To maintain protein homeostasis, bacteria have evolved various ribosome rescue mechanisms, including the predominant trans-translation process, to release stalled ribosomes and remove aberrant mRNAs. The rescue systems require the participation of translation elongation factor proteins (EFs) and are essential for bacterial physiology and reproduction. However, they disappear during eukaryotic evolution, which makes the essential proteins and translation elongation factors promising antimicrobial drug targets. Here, we review the structural and molecular mechanisms of the translation elongation factors EF-Tu, EF-Ts, and EF-G, which play essential roles in the normal translation and ribosome rescue mechanisms of Mycobacterium tuberculosis (Mtb). We also briefly describe the structure-based, computer-assisted study of anti-tuberculosis drugs.


Sujet(s)
Protéines bactériennes , Mycobacterium tuberculosis , Mycobacterium tuberculosis/métabolisme , Mycobacterium tuberculosis/effets des médicaments et des substances chimiques , Mycobacterium tuberculosis/génétique , Protéines bactériennes/métabolisme , Protéines bactériennes/génétique , Protéines bactériennes/composition chimique , Biosynthèse des protéines , Facteurs élongation chaîne peptidique/métabolisme , Facteurs élongation chaîne peptidique/composition chimique , Facteurs élongation chaîne peptidique/génétique , Antituberculeux/pharmacologie , Antituberculeux/composition chimique , Ribosomes/métabolisme , Modèles moléculaires , Tuberculose/traitement médicamenteux , Tuberculose/microbiologie , Tuberculose/métabolisme , Conformation des protéines
12.
Biochem Biophys Res Commun ; 711: 149920, 2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38615574

RÉSUMÉ

Tuberculosis (TB), a deadly infectious disease induced by Mycobacterium tuberculosis (Mtb), continues to be a global public health issue that kill millions of patents every year. Despite significant efforts have been paid to identify effective TB treatments, the emergence of drug-resistant strains of the disease and the presence of comorbidities in TB patients urges us to explore the detailed mechanisms involved in TB immunity and develop more effective innovative anti-TB strategies. HIF-1α, a protein involved in regulating cellular immune responses during TB infection, has been highlighted as a promising target for the development of novel strategies for TB treatment due to its critical roles in anti-TB host immunity. This review provides a summary of current research progress on the roles of HIF-1α in TB infection, highlighting its importance in regulating the host immune response upon Mtb infection and summarizing the influences and mechanisms of HIF-1α on anti-TB immunological responses of host cells. This review also discusses the various challenges associated with developing HIF-1α as a target for anti-TB therapies, including ensuring specificity and avoiding off-target effects on normal cell function, determining the regulation and expression of HIF-1α in TB patients, and developing drugs that can inhibit HIF-1α. More deep understanding of the molecular mechanisms involved in HIF-1α signaling, its impact on TB host status, and systematic animal testing and clinical trials may benefit the optimization of HIF-1α as a novel therapeutic target for TB.


Sujet(s)
Antituberculeux , Sous-unité alpha du facteur-1 induit par l'hypoxie , Mycobacterium tuberculosis , Transduction du signal , Tuberculose , Humains , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Mycobacterium tuberculosis/effets des médicaments et des substances chimiques , Mycobacterium tuberculosis/métabolisme , Mycobacterium tuberculosis/immunologie , Transduction du signal/effets des médicaments et des substances chimiques , Tuberculose/traitement médicamenteux , Tuberculose/immunologie , Tuberculose/métabolisme , Tuberculose/microbiologie , Animaux , Antituberculeux/usage thérapeutique , Antituberculeux/pharmacologie , Thérapie moléculaire ciblée/méthodes
14.
Tuberculosis (Edinb) ; 147: 102493, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38547568

RÉSUMÉ

Tuberculosis, caused by Mycobacterium tuberculosis, remains one of the deadliest infections in humans. Because Mycobacterium bovis Bacillus Calmette-Guérin (BCG) share genetic similarities with Mycobacterium tuberculosis, it is often used as a model to elucidate the molecular mechanisms of more severe tuberculosis infection. Caveolin-1 has been implied in many physiological processes and diseases, but it's role in mycobacterial infections has barely been studied. We isolated macrophages from Wildtype or Caveolin-1 deficient mice and analyzed hallmarks of infection, such as internalization, induction of autophagy and apoptosis. For in vivo assays we intravenously injected mice with BCG and investigated tissues for bacterial load with colony-forming unit assays, bioactive lipids with mass spectrometry and changes of protein expressions by Western blotting. Our results revealed that Caveolin-1 was important for early killing of BCG infection in vivo and in vitro, controlled acid sphingomyelinase (Asm)-dependent ceramide formation, apoptosis and inflammatory cytokines upon infection with BCG. In accordance, Caveolin-1 deficient mice and macrophages showed higher bacterial burdens in the livers. The findings indicate that Caveolin-1 plays a role in infection of mice and murine macrophages with BCG, by controlling cellular apoptosis and inflammatory host response. These clues might be useful in the fight against tuberculosis.


Sujet(s)
Apoptose , Cavéoline-1 , Macrophages , Souris de lignée C57BL , Souris knockout , Mycobacterium bovis , Sphingomyeline phosphodiesterase , Tuberculose , Animaux , Cavéoline-1/métabolisme , Cavéoline-1/déficit , Cavéoline-1/génétique , Mycobacterium bovis/pathogénicité , Macrophages/microbiologie , Macrophages/métabolisme , Tuberculose/microbiologie , Tuberculose/immunologie , Tuberculose/métabolisme , Tuberculose/anatomopathologie , Sphingomyeline phosphodiesterase/métabolisme , Sphingomyeline phosphodiesterase/déficit , Autophagie , Interactions hôte-pathogène , Modèles animaux de maladie humaine , Charge bactérienne , Cytokines/métabolisme , Céramides/métabolisme , Foie/microbiologie , Foie/métabolisme , Foie/anatomopathologie , Cellules cultivées , Souris , Médiateurs de l'inflammation/métabolisme , Facteurs temps
15.
Org Lett ; 26(10): 2034-2038, 2024 Mar 15.
Article de Anglais | MEDLINE | ID: mdl-38486497

RÉSUMÉ

Tuberculosis (TB) is one of the most dreadful diseases, killing more than 3 million humans annually. M. tuberculosis (MTb) is the causative agent for TB and has a thick and waxy cell wall, making it an attractive target for immunological studies. In this study, a heptamannopyranoside containing 1 → 2 and 1 → 6 α-mannopyranosidic linkages has been explored for the immunological evaluations. The conjugation-ready heptamannopyranoside was synthesized by exploiting the salient features of recently discovered [Au]/[Ag]-glycosidation of ethynylcyclohexyl glycosyl carbonate donors. The glycan was conjugated to the ESAT6, an early secreted protein of MTb for further characterization as a potential subunit vaccine candidate.


Sujet(s)
Mycobacterium tuberculosis , Tuberculose , Humains , Tuberculose/métabolisme , Carbonates , Catalyse
16.
Autophagy ; 20(7): 1483-1504, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38390831

RÉSUMÉ

Infectious diseases, such as Mycobacterium tuberculosis (Mtb)-caused tuberculosis (TB), remain a global threat exacerbated by increasing drug resistance. Host-directed therapy (HDT) is a promising strategy for infection treatment through targeting host immunity. However, the limited understanding of the function and regulatory mechanism of host factors involved in immune defense against infections has impeded HDT development. Here, we identify the ubiquitin ligase (E3) TRIM27 (tripartite motif-containing 27) as a host protective factor against Mtb by enhancing host macroautophagy/autophagy flux in an E3 ligase activity-independent manner. Mechanistically, upon Mtb infection, nuclear-localized TRIM27 increases and functions as a transcription activator of TFEB (transcription factor EB). Specifically, TRIM27 binds to the TFEB promoter and the TFEB transcription factor CREB1 (cAMP responsive element binding protein 1), thus enhancing CREB1-TFEB promoter binding affinity and promoting CREB1 transcription activity toward TFEB, eventually inducing autophagy-related gene expression as well as autophagy flux activation to clear the pathogen. Furthermore, TFEB activator 1 can rescue TRIM27 deficiency-caused decreased autophagy-related gene transcription and attenuated autophagy flux, and accordingly suppressed the intracellular survival of Mtb in cell and mouse models. Taken together, our data reveal that TRIM27 is a host defense factor against Mtb, and the TRIM27-CREB1-TFEB axis is a potential HDT-based TB target that can enhance host autophagy flux.Abbreviations: ATG5: autophagy related 5; BMDMs: bone marrow-derived macrophages; CFU: colony-forming unit; ChIP-seq: chromatin immunoprecipitation followed by sequencing; CREB1: cAMP responsive element binding protein 1; CTSB: cathepsin B; E3: ubiquitin ligase; EMSA: electrophoretic mobility shift assay; HC: healthy control; HDT: host-directed therapy; LAMP: lysosomal associated membrane protein; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MCOLN1: mucolipin TPR cation channel 1; Mtb: Mycobacterium tuberculosis; NLS: nuclear localization signal; PBMCs: peripheral blood mononuclear cells; PRKA/PKA: protein kinase cAMP-activated; qRT-PCR: quantitative real-time PCR; RFP: RET finger protein; TB: tuberculosis; TBK1: TANK binding kinase 1; TFEB: transcription factor EB; TRIM: tripartite motif; TSS: transcription start site; ULK1: unc-51 like autophagy activating kinase 1.


Sujet(s)
Autophagie , Facteurs de transcription à motifs basiques hélice-boucle-hélice et à glissière à leucines , Mycobacterium tuberculosis , Tuberculose , Autophagie/physiologie , Facteurs de transcription à motifs basiques hélice-boucle-hélice et à glissière à leucines/métabolisme , Animaux , Tuberculose/immunologie , Tuberculose/microbiologie , Tuberculose/métabolisme , Humains , Souris , Protéine de liaison à l'élément de réponse à l'AMP cyclique/métabolisme , Ubiquitin-protein ligases/métabolisme , Souris de lignée C57BL , Protéines à motif tripartite/métabolisme , Protéines à motif tripartite/génétique , Macrophages/métabolisme , Macrophages/microbiologie , Cellules HEK293 , Régions promotrices (génétique)/génétique , Protéines de liaison à l'ADN , Protéines nucléaires
17.
Int J Mol Sci ; 25(4)2024 Feb 18.
Article de Anglais | MEDLINE | ID: mdl-38397085

RÉSUMÉ

Tuberculosis, caused by Mycobacterium tuberculosis (Mtb), remains a global health crisis with substantial morbidity and mortality rates. Type II alveolar epithelial cells (AEC-II) play a critical role in the pulmonary immune response against Mtb infection by secreting effector molecules such as antimicrobial peptides (AMPs). Here, human ß-defensin 1 (hBD1), an important AMP produced by AEC-II, has been demonstrated to exert potent anti-tuberculosis activity. HBD1 overexpression effectively inhibited Mtb proliferation in AEC-II, while mice lacking hBD1 exhibited susceptibility to Mtb and increased lung tissue inflammation. Mechanistically, in A549 cells infected with Mtb, STAT1 negatively regulated hBD1 transcription, while CEBPB was the primary transcription factor upregulating hBD1 expression. Furthermore, we revealed that the ERK1/2 signaling pathway activated by Mtb infection led to CEBPB phosphorylation and nuclear translocation, which subsequently promoted hBD1 expression. Our findings suggest that the ERK1/2-CEBPB-hBD1 regulatory axis can be a potential therapeutic target for anti-tuberculosis therapy aimed at enhancing the immune response of AEC-II cells.


Sujet(s)
Mycobacterium tuberculosis , Tuberculose , bêta-Défensines , Animaux , Humains , Souris , Pneumocytes , bêta-Défensines/génétique , bêta-Défensines/pharmacologie , Protéine bêta de liaison aux séquences stimulatrices de type CCAAT/génétique , Cellules épithéliales , Système de signalisation des MAP kinases , Tuberculose/métabolisme
18.
J Immunol ; 212(5): 765-770, 2024 Mar 01.
Article de Anglais | MEDLINE | ID: mdl-38251918

RÉSUMÉ

AIM2 (absent in melanoma 2), an inflammasome component, mediates IL-1ß release in murine macrophages and cell lines. AIM2 and IL-1ß contribute to murine control of Mycobacterium tuberculosis (M.tb) infection, but AIM2's impact in human macrophages, the primary niche for M.tb, remains unclear. We show that M.tb, Mycobacterium bovis bacillus Calmette-Guérin (BCG), and M. smegmatis induce AIM2 expression in primary human macrophages. M.tb-induced AIM2 expression is peroxisome proliferator-activated receptor γ (PPARγ)-dependent and M.tb ESX-1-independent, whereas BCG- and M. smegmatis-induced AIM2 expression is PPARγ-independent. PPARγ and NLRP3, but not AIM2, are important for IL-1ß release in response to M.tb, and NLRP3 colocalizes with M.tb. This is in contrast to the role for AIM2 in inflammasome activation in mice and peritoneal macrophages. Altogether, we show that mycobacteria induce AIM2 expression in primary human macrophages, but AIM2 does not contribute to IL-1ß release during M.tb infection, providing further evidence that AIM2 expression and function are regulated in a cell- and/or species-specific manner.


Sujet(s)
Mycobacterium bovis , Mycobacterium tuberculosis , Tuberculose , Animaux , Humains , Souris , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Inflammasomes/métabolisme , Interleukine-1 bêta/métabolisme , Macrophages/métabolisme , Mycobacterium tuberculosis/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Récepteur PPAR gamma/métabolisme , Tuberculose/métabolisme
19.
J Infect Dis ; 229(4): 1229-1238, 2024 Apr 12.
Article de Anglais | MEDLINE | ID: mdl-37788578

RÉSUMÉ

Positron emission tomography-computed tomography (PET-CT) has the potential to revolutionize research in infectious diseases, as it has done with cancer. There is growing interest in it as a biomarker in the setting of early-phase tuberculosis clinical trials, particularly given the limitations of current biomarkers as adequate predictors of sterilizing cure for tuberculosis. PET-CT is a real-time tool that provides a 3-dimensional view of the spatial distribution of tuberculosis within the lung parenchyma and the nature of lesions with uptake (ie, whether nodular, consolidative, or cavitary). Its ability to provide functional data on changes in metabolism, drug penetration, and immune control of tuberculous lesions has the potential to facilitate drug development and regimen selection for advancement to phase 3 trials in tuberculosis. In this narrative review, we discuss the role that PET-CT may have in evaluating responses to drug therapy in active tuberculosis treatment and the challenges in taking PET-CT forward as predictive biomarker of relapse-free cure in the setting of phase 2 clinical trials.


Sujet(s)
Tomographie par émission de positons couplée à la tomodensitométrie , Tuberculose , Humains , Tuberculose/imagerie diagnostique , Tuberculose/traitement médicamenteux , Tuberculose/métabolisme , Poumon/anatomopathologie , Récidive , Marqueurs biologiques , Fluorodésoxyglucose F18/usage thérapeutique , Tomographie par émission de positons , Essais cliniques de phase II comme sujet
20.
Front Immunol ; 14: 1254347, 2023.
Article de Anglais | MEDLINE | ID: mdl-37928531

RÉSUMÉ

Exosomes as double-membrane vesicles contain various contents of lipids, proteins, mRNAs and non-coding RNAs, and involve in multiple physiological processes, for instance intercellular communication and immunomodulation. Currently, numerous studies found that the components of exosomal proteins, nucleic acids or lipids released from host cells are altered following infection with Mycobacterium tuberculosis. Exosomal contents provide excellent biomarkers for the auxiliary diagnosis, efficacy evaluation, and prognosis of tuberculosis. This study aimed to review the current literatures detailing the functions of exosomes in the procedure of M. tuberculosis infection, and determine the potential values of exosomes as biomarkers to assist in the diagnosis and monitoring of tuberculosis.


Sujet(s)
Exosomes , Mycobacterium tuberculosis , Tuberculose , Humains , Exosomes/métabolisme , Marqueurs biologiques/métabolisme , Communication cellulaire , Tuberculose/diagnostic , Tuberculose/métabolisme , Lipides
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...