Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Sci Rep ; 14(1): 19250, 2024 08 20.
Article in English | MEDLINE | ID: mdl-39164329

ABSTRACT

Radiolabelled puromycin analogues will allow the quantification of protein synthesis through nuclear medicine-based imaging. A particularly useful application could be the non-invasive longitudinal visualisation of mycobacterial activity through direct quantification of puromycin binding. This study assesses the value of [68Ga]Ga-DOTA-puromycin in the visualisation of mycobacteria through positron emission tomography combined with magnetic resonance imaging (µPET/MRI). The radiopharmaceutical was produced by previously published and validated methods. [68Ga]Ga-DOTA-Puromycin imaging was performed on severe immunodeficient mice infected with Bacille Calmette-Guérin-derived M. Bovis (BCG). Acute and chronic infection stages were examined by µPET/MRI. A follow-up group of animals acted as controls (animals bearing S. aureus-derived infection and sterile inflammation) to assess tracer selectivity. [68Ga]Ga-DOTA-puromycin-µPET/MRI images revealed the acute, widespread infection within the right upper shoulder and armpit. Also, [68Ga]Ga-DOTA-puromycin signal sensitivity measured after a 12-week period was lower than that of [18F]FDG-PET in the same animals. A suitable correlation between normalised uptake values (NUV) and gold standard histopathological analysis confirms accurate tracer accumulation in viable bacteria. The radiopharmaceutical showed infection selectivity over inflammation but accumulated in both M. Bovis and S. Aureus, lacking pathogen specificity. Overall, [68Ga]Ga-DOTA-puromycin exhibits potential as a tool for non-invasive protein synthesis visualization, albeit without pathogen selectivity.


Subject(s)
Gallium Radioisotopes , Magnetic Resonance Imaging , Mycobacterium bovis , Positron-Emission Tomography , Radiopharmaceuticals , Animals , Magnetic Resonance Imaging/methods , Positron-Emission Tomography/methods , Mice , Radiopharmaceuticals/chemistry , Organometallic Compounds , Heterocyclic Compounds, 1-Ring/chemistry , Mice, SCID , Female , Tuberculosis/diagnostic imaging , Tuberculosis/microbiology , Tuberculosis/metabolism , Mycobacterium Infections/diagnostic imaging , Mycobacterium Infections/microbiology
2.
Int J Mol Sci ; 25(15)2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39125711

ABSTRACT

Cystatin F (CstF) is a protease inhibitor of cysteine cathepsins, including those involved in activating the perforin/granzyme cytotoxic pathways. It is targeted at the endolysosomal pathway but can also be secreted to the extracellular milieu or endocytosed by bystander cells. CstF was shown to be significantly increased in tuberculous pleurisy, and during HIV coinfection, pleural fluids display high viral loads. In human macrophages, our previous results revealed a strong upregulation of CstF in phagocytes activated by interferon γ or after infection with Mycobacterium tuberculosis (Mtb). CstF manipulation using RNA silencing led to increased proteolytic activity of lysosomal cathepsins, improving Mtb intracellular killing. In the present work, we investigate the impact of CstF depletion in macrophages during the coinfection of Mtb-infected phagocytes with lymphocytes infected with HIV. The results indicate that decreasing the CstF released by phagocytes increases the major pro-granzyme convertase cathepsin C of cytotoxic immune cells from peripheral blood-derived lymphocytes. Consequently, an observed augmentation of the granzyme B cytolytic activity leads to a significant reduction in viral replication in HIV-infected CD4+ T-lymphocytes. Ultimately, this knowledge can be crucial for developing new therapeutic approaches to control both pathogens based on manipulating CstF.


Subject(s)
Cathepsin C , Coinfection , Granzymes , HIV Infections , Macrophages , Mycobacterium tuberculosis , Humans , Granzymes/metabolism , Granzymes/genetics , HIV Infections/metabolism , HIV Infections/immunology , Macrophages/metabolism , Macrophages/immunology , Macrophages/microbiology , Macrophages/virology , Coinfection/microbiology , Cathepsin C/metabolism , Cathepsin C/genetics , Cystatins/metabolism , Cystatins/genetics , Tuberculosis/metabolism , Tuberculosis/immunology , Tuberculosis/microbiology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , HIV-1/physiology , Biomarkers, Tumor
3.
Metabolomics ; 20(5): 92, 2024 Aug 03.
Article in English | MEDLINE | ID: mdl-39096437

ABSTRACT

INTRODUCTION: The human immunodeficiency virus (HIV) and tuberculosis (TB) co-infection presents significant challenges due to the complex interplay between these diseases, leading to exacerbated metabolic disturbances. Understanding these metabolic profiles is crucial for improving diagnostic and therapeutic approaches. OBJECTIVE: This study aimed to characterise the urinary acylcarnitine and amino acid profiles, including 5-hydroxyindoleacetic acid (5-HIAA), in patients co-infected with HIV and TB using targeted liquid chromatography mass spectrometry (LC-MS) metabolomics. METHODS: Urine samples, categorised into HIV, TB, HIV/TB co-infected, and healthy controls, were analysed using HPLC-MS/MS. Statistical analyses included one-way ANOVA and a Kruskal-Wallis test to determine significant differences in the acylcarnitine and amino acid profiles between groups. RESULTS: The study revealed significant metabolic alterations, especially in TB and co-infected groups. Elevated levels of medium-chain acylcarnitines indicated increased fatty acid oxidation, commonly associated with cachexia in TB. Altered amino acid profiles suggested disruptions in protein and glucose metabolism, indicating a shift towards diabetes-like metabolic states. Notably, TB was identified as a primary driver of these changes, affecting protein turnover, and impacting energy metabolism in co-infected patients. CONCLUSION: The metabolic profiling of HIV/TB co-infection highlights the profound impact of TB on metabolic pathways, which may exacerbate the clinical complexities of co-infection. Understanding these metabolic disruptions can guide the development of targeted treatments and improve management strategies, ultimately enhancing the clinical outcomes for these patients. Further research is required to validate these findings and explore their implications in larger, diverse populations.


Subject(s)
Amino Acids , Carnitine , Coinfection , HIV Infections , Metabolomics , Tuberculosis , Adult , Female , Humans , Male , Middle Aged , Amino Acids/urine , Amino Acids/metabolism , Carnitine/analogs & derivatives , Carnitine/urine , Carnitine/metabolism , Chromatography, High Pressure Liquid/methods , Coinfection/urine , Coinfection/metabolism , HIV Infections/complications , HIV Infections/urine , HIV Infections/metabolism , Liquid Chromatography-Mass Spectrometry/methods , Metabolomics/methods , Tandem Mass Spectrometry/methods , Tuberculosis/urine , Tuberculosis/metabolism
4.
Front Immunol ; 15: 1369278, 2024.
Article in English | MEDLINE | ID: mdl-39021575

ABSTRACT

Matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) has recently gained prominence for its ability to provide molecular and spatial information in tissue sections. This technology has the potential to uncover novel insights into proteins and other molecules in biological and immunological pathways activated along diseases with a complex host-pathogen interaction, such as animal tuberculosis. Thus, the present study conducted a data analysis of protein signature in granulomas of cattle and pigs naturally infected with the Mycobacterium tuberculosis complex (MTC), identifying biological and immunological signaling pathways activated throughout the disease. Lymph nodes from four pigs and four cattle, positive for the MTC by bacteriological culture and/or real-time PCR, were processed for histopathological examination and MALDI-MSI. Protein identities were assigned using the MaTisse database, and protein-protein interaction networks were visualized using the STRING database. Gene Ontology (GO) analysis was carried out to determine biological and immunological signaling pathways in which these proteins could participate together with Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. Distinct proteomic profiles between cattle and pig granulomas were displayed. Noteworthy, the GO analysis revealed also common pathways among both species, such as "Complement activation, alternative pathway" and "Tricarboxylic acid cycle", which highlight pathways that are conserved among different species infected by the MTC. In addition, species-specific terms were identified in the current study, such as "Natural killer cell degranulation" in cattle or those related to platelet and neutrophil recruitment and activation in pigs. Overall, this study provides insights into the immunopathogenesis of tuberculosis in cattle and pigs, opening new areas of research and highlighting the importance, among others, of the complement activation pathway and the regulation of natural killer cell- and neutrophil-mediated immunity in this disease.


Subject(s)
Granuloma , Mycobacterium tuberculosis , Proteomics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tuberculosis , Animals , Swine , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/veterinary , Cattle , Proteomics/methods , Mycobacterium tuberculosis/immunology , Tuberculosis/immunology , Tuberculosis/veterinary , Tuberculosis/microbiology , Tuberculosis/metabolism , Granuloma/immunology , Granuloma/microbiology , Granuloma/metabolism , Granuloma/veterinary , Swine Diseases/immunology , Swine Diseases/microbiology , Protein Interaction Maps , Host-Pathogen Interactions/immunology , Proteome , Signal Transduction
5.
PLoS One ; 19(7): e0306752, 2024.
Article in English | MEDLINE | ID: mdl-38968285

ABSTRACT

PURPOSE: To analyze the causal relationship between 486 human serum metabolites and the active tuberculosis (ATB) in European population. METHODS: In this study, the causal relationship between human serum metabolites and the ATB was analyzed by integrating the genome-wide association study (GWAS). The 486 human serum metabolites were used as the exposure variable, three different ATB GWAS databases in the European population were set as outcome variables, and single nucleotide polymorphisms were used as instrumental variables for Mendelian Randomization. The inverse variance weighting was estimated causality, the MR-Egger intercept to estimate horizontal pleiotropy, and the combined effects of metabolites were also considered in the meta-analysis. Furthermore, the web-based MetaboAnalyst 6.0 was engaged for enrichment pathway analysis, while R (version 4.3.2) software and Review Manager 5.3 were employed for statistical analysis. RESULTS: A total of 21, 17, and 19 metabolites strongly associated with ATB were found in the three databases after preliminary screening (P < 0.05). The intersecting metabolites across these databases included tryptophan, betaine, 1-linoleoylglycerol (1-monolinolein) (1-LG), 1-eicosatrienoylglycerophosphocholine, and oleoylcarnitine. Among them, betaine (I2 = 24%, P = 0.27) and 1-LG (I2 = 0%, P = 0.62) showed the lowest heterogeneity among the different ATB databases. In addition, the metabolic pathways of phosphatidylethanolamine biosynthesis (P = 0.0068), methionine metabolism (P = 0.0089), betaine metabolism (P = 0.0205) and oxidation of branched-chain fatty acids (P = 0.0309) were also associated with ATB. CONCLUSION: Betaine and 1-LG may be biomarkers or auxiliary diagnostic tools for ATB. They may provide new guidance for medical practice in the early diagnosis and surveillance of ATB. In addition, by interfering with phosphatidylethanolamine biosynthesis, methionine metabolism, betaine metabolism, oxidation of branched-chain fatty acids, and other pathways, it is helpful to develop new anti-tuberculosis drugs and explore the virulence or pathogenesis of ATB at a deeper level, providing an effective reference for future studies.


Subject(s)
Betaine , Genome-Wide Association Study , Polymorphism, Single Nucleotide , Tuberculosis , Humans , Betaine/blood , Betaine/metabolism , Tuberculosis/genetics , Tuberculosis/blood , Tuberculosis/metabolism , Europe , White People/genetics , Metabolomics/methods
6.
Sci Rep ; 14(1): 15312, 2024 07 03.
Article in English | MEDLINE | ID: mdl-38961191

ABSTRACT

Nontuberculous mycobacteria (NTM) infection diagnosis remains a challenge due to its overlapping clinical symptoms with tuberculosis (TB), leading to inappropriate treatment. Herein, we employed noninvasive metabolic phenotyping coupled with comprehensive statistical modeling to discover potential biomarkers for the differential diagnosis of NTM infection versus TB. Urine samples from 19 NTM and 35 TB patients were collected, and untargeted metabolomics was performed using rapid liquid chromatography-mass spectrometry. The urine metabolome was analyzed using a combination of univariate and multivariate statistical approaches, incorporating machine learning. Univariate analysis revealed significant alterations in amino acids, especially tryptophan metabolism, in NTM infection compared to TB. Specifically, NTM infection was associated with upregulated levels of methionine but downregulated levels of glutarate, valine, 3-hydroxyanthranilate, and tryptophan. Five machine learning models were used to classify NTM and TB. Notably, the random forest model demonstrated excellent performance [area under the receiver operating characteristic (ROC) curve greater than 0.8] in distinguishing NTM from TB. Six potential biomarkers for NTM infection diagnosis, including methionine, valine, glutarate, 3-hydroxyanthranilate, corticosterone, and indole-3-carboxyaldehyde, were revealed from univariate ROC analysis and machine learning models. Altogether, our study suggested new noninvasive biomarkers and laid a foundation for applying machine learning to NTM differential diagnosis.


Subject(s)
Biomarkers , Machine Learning , Metabolomics , Mycobacterium Infections, Nontuberculous , Tuberculosis , Humans , Metabolomics/methods , Male , Biomarkers/urine , Female , Middle Aged , Tuberculosis/urine , Tuberculosis/diagnosis , Tuberculosis/microbiology , Tuberculosis/metabolism , Mycobacterium Infections, Nontuberculous/urine , Mycobacterium Infections, Nontuberculous/diagnosis , Mycobacterium Infections, Nontuberculous/microbiology , Nontuberculous Mycobacteria , Aged , Adult , Metabolome , ROC Curve , Diagnosis, Differential
7.
Biomolecules ; 14(7)2024 Jul 14.
Article in English | MEDLINE | ID: mdl-39062562

ABSTRACT

Tuberculosis and AIDS remain two of the most relevant human infectious diseases. The pathogens that cause them, Mycobacterium tuberculosis (Mtb) and HIV, individually elicit an immune response that treads the line between beneficial and detrimental to the host. Co-infection further complexifies this response since the different cytokines acting on one infection might facilitate the dissemination of the other. In these responses, the role of type I interferons is often associated with antiviral mechanisms, while for bacteria such as Mtb, their importance and clinical relevance as a suitable target for manipulation are more controversial. In this article, we review the recent knowledge on how these interferons play distinct roles and sometimes have opposite consequences depending on the stage of the pathogenesis. We highlight the dichotomy between the acute and chronic infections displayed by both infections and how type I interferons contribute to an initial control of each infection individually, while their chronic induction, particularly during HIV infection, might facilitate Mtb primo-infection and progression to disease. We expect that further findings and their systematization will allow the definition of windows of opportunity for interferon manipulation according to the stage of infection, contributing to pathogen clearance and control of immunopathology.


Subject(s)
HIV Infections , Interferon Type I , Mycobacterium tuberculosis , Tuberculosis , Humans , Interferon Type I/metabolism , Interferon Type I/immunology , Mycobacterium tuberculosis/immunology , HIV Infections/immunology , HIV Infections/microbiology , Tuberculosis/immunology , Tuberculosis/microbiology , Tuberculosis/metabolism , Coinfection/immunology , Coinfection/microbiology , Animals
8.
mSystems ; 9(8): e0050124, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39051782

ABSTRACT

Infection with Mycobacterium bovis precipitates a spectrum of pathologies in bovines, notably necrotic pneumonia, mastitis, and arthritis, impinging upon the health and nutritional assimilation of these animals. A pivotal factor, lipocalin 2 (Lcn2), is responsive to microbial invasion, inflammatory processes, and tissue damage, the extent of which Lcn2 modulates the gut environment, however, remains unclear in response to M. bovis-induced alterations. To explore the role of Lcn2 in shaping the gut milieu of mice during a 5-week period post-M. bovis infection, Lcn2 knockout Lcn2-/- mice were scrutinized for changes in the gut microbiota and metabolomic profiles. Results showed that Lcn2-/- mice infected with M. bovis exhibited notable shifts in the operational taxonomic units (OTUs) of gut microbiota, alongside significant disparities in α and ß diversity. Concomitantly, a marked increase was observed during the 5-week period in the abundance of Akkermansia, Oscillospira, and Bacteroides, coupled with a substantial decrease in Ruminococcus within the microbiome of Lcn2 knockout mice. Notably, Akkermansia muciniphila was significantly enriched in the gut flora of Lcn2-/- mice. Furthermore, the absence of Lcn2 significantly altered the gut metabolomic landscape, evidenced by elevated levels of metabolites such as taurodeoxycholic acid, 10-undecenoic acid, azelaic acid, and dodecanedioic acid in Lcn2-/- mice. Our findings demonstrated that the lack of Lcn2 in the context of M. bovis infection profoundly affected the regulation of gut microbiota and metabolomic components, culminating in a transformed gut environment. Our results revealed that Lcn2 may regulate gut microbiota and metabolome components, changing the intestinal environment, thereby affecting the infection status of M. bovis. IMPORTANCE: Our study addresses the critical knowledge gap regarding the specific influence of lipocalin 2 (LCN2) in the context of Mycobacterium bovis infection, particularly focusing on its role in the gut environment. Utilizing LCN2 knockout (Lcn2-/-) mice, we meticulously assessed changes in the gut microbiota and metabolic components following M. bovis infection. Our findings reveal alterations in the gut microbial community, emphasizing the potentially crucial role of LCN2 in maintaining stability. Furthermore, we observed significant shifts in specific microbial communities, including the enrichment of Akkermansia muciniphila, known for its positive impact on intestinal health and immune regulation. The implications of our study extend beyond understanding the dynamics of the gut microbiome, offering insights into the potential therapeutic strategies for gut-related health conditions and microbial dysbiosis.


Subject(s)
Gastrointestinal Microbiome , Lipocalin-2 , Metabolome , Mice, Knockout , Mycobacterium bovis , Animals , Lipocalin-2/genetics , Lipocalin-2/metabolism , Mice , Mice, Inbred C57BL , Tuberculosis/microbiology , Tuberculosis/genetics , Tuberculosis/metabolism , Tuberculosis/immunology , Female
9.
Metabolomics ; 20(4): 78, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39014031

ABSTRACT

INTRODUCTION: Amid the global health crisis, HIV/TB co-infection presents significant challenges, amplifying the burden on patients and healthcare systems alike. Metabolomics offers an innovative window into the metabolic disruptions caused by co-infection, potentially improving diagnosis and treatment monitoring. AIM: This study uses untargeted metabolomics to investigate the urinary metabolic signature of HIV/TB co-infection, enhancing understanding of the metabolic interplay between these infections. METHODS: Urine samples from South African adults, categorised into four groups - healthy controls, TB-positive, HIV-positive, and HIV/TB co-infected - were analysed using GCxGC-TOFMS. Metabolites showing significant differences among groups were identified through Kruskal-Wallis and Wilcoxon rank sum tests. RESULTS: Various metabolites (n = 23) were modulated across the spectrum of health and disease states represented in the cohorts. The metabolomic profiles reflect a pronounced disruption in biochemical pathways involved in energy production, amino acid metabolism, gut microbiome, and the immune response, suggesting a bidirectional exacerbation between HIV and TB. While both diseases independently perturb the host's metabolism, their co-infection leads to a unique metabolic phenotype, indicative of an intricate interplay rather than a simple additive effect. CONCLUSION: Metabolic profiling revealed a unique metabolic landscape shaped by HIV/TB co-infection. The findings highlight the potential of urinary differential metabolites for co-infection, offering a non-invasive tool for enhancing diagnostic precision and tailoring therapeutic interventions. Future research should focus on expanding sample sizes and integrating longitudinal analyses to build upon these foundational insights, paving the way for metabolomic applications in combating these concurrent pandemics.


Subject(s)
Coinfection , HIV Infections , Metabolomics , Tuberculosis , Humans , HIV Infections/complications , HIV Infections/metabolism , HIV Infections/urine , Metabolomics/methods , Coinfection/metabolism , Adult , Male , Tuberculosis/metabolism , Tuberculosis/urine , Female , Middle Aged , Metabolome , Biomarkers/urine
10.
Cytokine ; 181: 156693, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38986252

ABSTRACT

BACKGROUND: To delineate alterations in DNA methylation at high resolution within the genomic profile of monocyte-derived-dendritic cells (mo-DCs) in connection with Mycobacterium tuberculosis (MTB) infection, with particular emphasis on pro/ anti-inflammatory genes. METHODS: In the context of this investigation, mo-DCs were infected by various active strains of MTB (Rifampicin-resistant [RIFR], H37Rv, multidrug-resistant [MDR], and extensively drug-resistant [XDR]). Subsequently, the pro/anti-inflammatory hub gene expression levels within the IL-6, IL-12, IFN-γ, IL-1ß, TNF-α, and IL-10 pathways were evaluated employing real-time reverse transcription-polymerase chain reaction (RT-PCR). Additionally, the effects of MTB infection on mo-DC protein expression were examined through western blot analysis. The methylation status (%) of TNF-α and IL-10 was considered through Methylation Sensitive-High Resolution Melting (MS-HRM). RESULTS: The results revealed an up-regulation of all pro-inflammatory genes among all groups, with TNF-α exhibiting the highest expression level. Conversely, the anti-inflammatory gene (IL-10) showed a down-regulated expression level. Furthermore, the DNA methylation status (%) of TNF-α decreased significantly among all the groups (P < 0.001), although there were no notable distinctions in the DNA methylation status (%) of IL-10 when compared to the control group (P > 0.05). CONCLUSION: MTB infection induces DNA methylation changes in mo-DCs. The hypo-methylation of TNF-α may induce the up-regulation of this gene. This correlation revealed that the more resistant the MTB strain (XDR) is, the lower the methylation status (%) in the TNF-α gene.


Subject(s)
Cytokines , DNA Methylation , Dendritic Cells , Epigenesis, Genetic , Monocytes , Mycobacterium tuberculosis , Tuberculosis , Mycobacterium tuberculosis/immunology , Humans , Cytokines/metabolism , Dendritic Cells/metabolism , Monocytes/metabolism , Tuberculosis/microbiology , Tuberculosis/genetics , Tuberculosis/immunology , Tuberculosis/metabolism , Tumor Necrosis Factor-alpha/metabolism , Interleukin-10/metabolism , Interleukin-10/genetics
11.
Respir Res ; 25(1): 254, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38907347

ABSTRACT

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.


Subject(s)
Antitubercular Agents , Biomarkers , Chemical and Drug Induced Liver Injury , Metabolomics , Humans , Antitubercular Agents/adverse effects , Male , Metabolomics/methods , Female , Chemical and Drug Induced Liver Injury/blood , Chemical and Drug Induced Liver Injury/diagnosis , Chemical and Drug Induced Liver Injury/metabolism , Longitudinal Studies , Adult , Middle Aged , Biomarkers/blood , Prospective Studies , Predictive Value of Tests , Tuberculosis/drug therapy , Tuberculosis/blood , Tuberculosis/metabolism , Bile Acids and Salts/blood , Bile Acids and Salts/metabolism
12.
Front Immunol ; 15: 1413947, 2024.
Article in English | MEDLINE | ID: mdl-38881887

ABSTRACT

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.


Subject(s)
CD36 Antigens , Mycobacterium tuberculosis , Tuberculosis , Humans , CD36 Antigens/metabolism , Mycobacterium tuberculosis/immunology , Tuberculosis/immunology , Tuberculosis/metabolism , Tuberculosis/microbiology , Animals , Lipid Metabolism , Signal Transduction , Biomarkers , Host-Pathogen Interactions/immunology
13.
Elife ; 122024 Jun 26.
Article in English | MEDLINE | ID: mdl-38922679

ABSTRACT

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.


Subject(s)
Cell Movement , Dendritic Cells , Glycolysis , Hypoxia-Inducible Factor 1, alpha Subunit , Monocytes , Mycobacterium tuberculosis , Tuberculosis , Dendritic Cells/metabolism , Dendritic Cells/immunology , Monocytes/metabolism , Monocytes/immunology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mycobacterium tuberculosis/immunology , Animals , Tuberculosis/immunology , Tuberculosis/metabolism , Tuberculosis/microbiology , Mice , Toll-Like Receptor 2/metabolism , Mice, Inbred C57BL , Female
14.
Nat Commun ; 15(1): 5239, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38937448

ABSTRACT

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.


Subject(s)
Mycobacterium tuberculosis , Positron-Emission Tomography , Trehalose , Tuberculosis , Animals , Mycobacterium tuberculosis/metabolism , Positron-Emission Tomography/methods , Trehalose/metabolism , Tuberculosis/diagnostic imaging , Tuberculosis/microbiology , Tuberculosis/metabolism , Humans , Mice , Fluorine Radioisotopes , Fluorodeoxyglucose F18/metabolism , Fluorodeoxyglucose F18/chemistry , Radiopharmaceuticals/metabolism , Disease Models, Animal , Female
15.
Front Immunol ; 15: 1402024, 2024.
Article in English | MEDLINE | ID: mdl-38873598

ABSTRACT

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.


Subject(s)
Cholesterol , Macrophages , Mycobacterium tuberculosis , Tuberculosis , Mycobacterium tuberculosis/immunology , Cholesterol/metabolism , Humans , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Tuberculosis/immunology , Tuberculosis/metabolism , Tuberculosis/microbiology , Animals , Host-Pathogen Interactions/immunology , Antitubercular Agents/pharmacology , Antitubercular Agents/therapeutic use , Lipid Metabolism
16.
Biomed Pharmacother ; 177: 117030, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38917759

ABSTRACT

Ferroptosis is a lipid peroxidation-driven and iron-dependent form of programmed cell death, which is involved in a variety of physical processes and multiple diseases. Numerous reports have demonstrated that ferroptosis is closely related to the pathophysiological processes of Mycobacterium tuberculosis (M. tuberculosis) infection and is characterized by the accumulation of excess lipid peroxides on the cell membrane. In this study, the various functions of ferroptosis, and the therapeutic strategies and diagnostic biomarkers of tuberculosis, were summarized. Notably, this review provides insights into the molecular mechanisms and functions of M. tuberculosis-induced ferroptosis, suggesting potential future therapeutic and diagnostic markers for tuberculosis.


Subject(s)
Ferroptosis , Mycobacterium tuberculosis , Tuberculosis , Ferroptosis/physiology , Humans , Mycobacterium tuberculosis/pathogenicity , Tuberculosis/microbiology , Tuberculosis/pathology , Tuberculosis/metabolism , Animals , Lipid Peroxidation , Cell Death/physiology , Iron/metabolism , Biomarkers/metabolism
17.
Cell Signal ; 121: 111271, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38944259

ABSTRACT

Circular RNAs (circRNAs) play a critical role in pathological mechanisms of Mycobacterium tuberculosis (Mtb) and can be used as a new biomarker for active tuberculosis (ATB) diagnosis. Therefore, we identified significantly dysregulated circRNAs in ATB patients and healthy controls (HC) and explored their molecular mechanism. We found that hsa_circ_0002371 was significantly up-regulated in PBMCs of ATB patients and Mycobacterium tuberculosis H37Rv- or Mycobacterium bovis bacillus Calmette Guerin (BCG)-infected THP-1 cells. Functional experiments demonstrated that hsa_circ_0002371 inhibited autophagy in BCG-infected THP-1 cells and promoted intracellular BCG survival rate. In terms of mechanism, hsa_circ_0002371 facilitated the expression of hsa-miR-502-5p, as shown by bioinformatics and dual-luciferase reporter gene analysis, respectively. Notably, hsa-miR-502-5p inhibited autophagy via suppressing autophagy related 16 like 1 (ATG16L1) in BCG-infected macrophages and thus promoting intracellular BCG growth. In summation, hsa_circ_0002371 increased the suppression of hsa-miR-502-5p on ATG16L1 and inhibited autophagy to promote Mtb growth in macrophages. In Conclusion, our data suggested that hsa_circ_0002371 was significantly up-regulated in the PBMCs of ATB patients compared with HC. The hsa_circ_0002371/hsa-miR-502-5p/ATG16L1 axis promoted the survival of intracellular Mtb and inhibited autophagy in macrophages. Our findings suggested hsa_circ_0002371 could act as a potential diagnostic biomarker and therapeutic target.


Subject(s)
Autophagy-Related Proteins , Autophagy , Macrophages , MicroRNAs , Mycobacterium tuberculosis , RNA, Circular , Humans , MicroRNAs/metabolism , MicroRNAs/genetics , RNA, Circular/genetics , RNA, Circular/metabolism , Autophagy-Related Proteins/metabolism , Autophagy-Related Proteins/genetics , Macrophages/metabolism , Macrophages/microbiology , THP-1 Cells , Tuberculosis/metabolism , Tuberculosis/microbiology , Tuberculosis/genetics , Tuberculosis/pathology , Mycobacterium bovis , Male , Female
18.
Front Cell Infect Microbiol ; 14: 1398077, 2024.
Article in English | MEDLINE | ID: mdl-38836056

ABSTRACT

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.


Subject(s)
Extracellular Vesicles , Host-Pathogen Interactions , Mycobacterium tuberculosis , Tuberculosis , Extracellular Vesicles/metabolism , Extracellular Vesicles/immunology , Mycobacterium tuberculosis/immunology , Humans , Tuberculosis/immunology , Tuberculosis/microbiology , Tuberculosis/metabolism , Host-Pathogen Interactions/immunology , Animals
19.
EMBO Rep ; 25(7): 3064-3089, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38866980

ABSTRACT

Type I interferons (IFN-I) are implicated in exacerbation of tuberculosis (TB), but the mechanisms are unclear. Mouse macrophages infected with Mycobacterium tuberculosis (Mtb) produce IFN-I, which contributes to their death. Here we investigate whether the same is true for human monocyte-derived macrophages (MDM). MDM prepared by a conventional method markedly upregulate interferon-stimulated genes (ISGs) upon Mtb infection, while MDM prepared to better restrict Mtb do so much less. A mixture of antibodies inhibiting IFN-I signaling prevents ISG induction. Surprisingly, secreted IFN-I are undetectable until nearly two days after ISG induction. These same antibodies do not diminish Mtb-infected MDM death. MDM induce ISGs in response to picogram/mL levels of exogenous IFN-I while depleting similar quantities from the medium. Exogenous IFN-I increase the proportion of dead MDM. We speculate that Mtb-infected MDM produce and respond to minute levels of IFN-I, and that only some of the resultant signaling is susceptible to neutralizing antibodies. Many types of cells may secrete IFN-I in patients with TB, where IFN-I is likely to promote the death of infected macrophages.


Subject(s)
Cell Death , Interferon Type I , Macrophages , Mycobacterium tuberculosis , Humans , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/pathogenicity , Macrophages/microbiology , Macrophages/metabolism , Macrophages/immunology , Interferon Type I/metabolism , Signal Transduction , Tuberculosis/microbiology , Tuberculosis/immunology , Tuberculosis/metabolism , Animals , Mice , Cells, Cultured
20.
J Proteomics ; 305: 105245, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38942234

ABSTRACT

Tuberculosis (TB) is an infectious disease that remains one of the major global public health concerns. Early detection of Active Pulmonary TB is therefore of utmost importance for controlling lethality and disease spreading. Currently available TB diagnostics can be broadly categorized into microscopy, culture-based, and molecular approaches, all of which come with compromised sensitivity, limited efficacy, and high expenses. Hence, rapid, sensitive, and affordable diagnostic methods for TB is the current prerequisite for disease management. This review summarizes the proteomics investigations for host-specific biomarkers from serum, sputum, saliva, and urine samples of TB patients, along with patients having comorbidity. Thorough data mining from available literature led us to conclude that the host-specific proteins involved in immunity and defense, metabolic regulation, cellular adhesion, and motility, inflammatory responses, and tissue remodelling have shown significant deregulation upon Mycobacterium tuberculosis (Mtb) infection. Notably, the immunoregulatory protein orosomucoid (ORM) was up-regulated in active TB compared to non-TB individuals, as observed in multiple studies from diverse sample types. Mannose receptor C type 2 (MRC2) was identified as an upregulated, treatment response biomarker in two independent serum proteomics investigations. Thorough mechanistic investigation on these candidate proteins would be fascinating to dig into potential drug targets and customized therapeutics for TB patients, along with their diagnostic potentials.


Subject(s)
Biomarkers , Mycobacterium tuberculosis , Proteomics , Humans , Biomarkers/blood , Biomarkers/metabolism , Biomarkers/analysis , Proteomics/methods , Tuberculosis/diagnosis , Tuberculosis/blood , Tuberculosis/metabolism , Tuberculosis, Pulmonary/diagnosis , Tuberculosis, Pulmonary/blood , Tuberculosis, Pulmonary/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL