Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 4.193
Filtrer
2.
J Biosci ; 492024.
Article de Anglais | MEDLINE | ID: mdl-39046034

RÉSUMÉ

Nitric oxide (NO) and iNOS are crucial host factors in innate immunity against intracellular pathogens. However, the role of NO in Mycobacterium tuberculosis (M. tb) infection in humans remains controversial, unlike in the murine model of TB. To investigate this, levels of NO, iNOS, and L-arginine, as well as the NOS2A gene polymorphism rs57234985 at the promoter region of NOS2A, were evaluated in pulmonary TB (PTB) patients and their household contacts (HHCs). Increased levels of NO and iNOS expression in HHCs indicated exposure to M. tb infection which was confirmed by higher levels of iNOS and NO in Mantouxpositive individuals. Furthermore, higher levels of arginine were detected in HHCs, suggesting its potential role in promoting optimal NO synthesis. PTB patients had higher levels of these analytes due to ongoing active infection. Interestingly, iNOS and NO levels were inversely related to bacterial burden, suggesting their antimicrobial role. NOS2A gene polymorphism was found to be associated with disease susceptibility, with the TT genotype linked to increased iNOS expression. To conclude, iNOS plays a crucial role in controlling early M. tb infection in HHCs by inducing optimal NO production with help of L-arginine. Further longitudinal studies are needed to better understand the role of these host factors upon disease activation.


Sujet(s)
Arginine , Immunité innée , Mycobacterium tuberculosis , Nitric oxide synthase type II , Monoxyde d'azote , Tuberculose pulmonaire , Humains , Monoxyde d'azote/métabolisme , Tuberculose pulmonaire/immunologie , Tuberculose pulmonaire/génétique , Tuberculose pulmonaire/microbiologie , Nitric oxide synthase type II/génétique , Nitric oxide synthase type II/métabolisme , Mycobacterium tuberculosis/immunologie , Mycobacterium tuberculosis/pathogénicité , Femelle , Mâle , Immunité innée/génétique , Adulte , Arginine/métabolisme , Adulte d'âge moyen , Caractéristiques familiales , Prédisposition génétique à une maladie , Régions promotrices (génétique)/génétique , Polymorphisme de nucléotide simple
3.
PLoS Genet ; 20(6): e1011313, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38870230

RÉSUMÉ

A quarter of humanity is estimated to have been exposed to Mycobacterium tuberculosis (Mtb) with a 5-10% risk of developing tuberculosis (TB) disease. Variability in responses to Mtb infection could be due to host or pathogen heterogeneity. Here, we focused on host genetic variation in a Peruvian population and its associations with gene regulation in monocyte-derived macrophages and dendritic cells (DCs). We recruited former household contacts of TB patients who previously progressed to TB (cases, n = 63) or did not progress to TB (controls, n = 63). Transcriptomic profiling of monocyte-derived DCs and macrophages measured the impact of genetic variants on gene expression by identifying expression quantitative trait loci (eQTL). We identified 330 and 257 eQTL genes in DCs and macrophages (False Discovery Rate (FDR) < 0.05), respectively. Four genes in DCs showed interaction between eQTL variants and TB progression status. The top eQTL interaction for a protein-coding gene was with FAH, the gene encoding fumarylacetoacetate hydrolase, which mediates the last step in mammalian tyrosine catabolism. FAH expression was associated with genetic regulatory variation in cases but not controls. Using public transcriptomic and epigenomic data of Mtb-infected monocyte-derived dendritic cells, we found that Mtb infection results in FAH downregulation and DNA methylation changes in the locus. Overall, this study demonstrates effects of genetic variation on gene expression levels that are dependent on history of infectious disease and highlights a candidate pathogenic mechanism through pathogen-response genes. Furthermore, our results point to tyrosine metabolism and related candidate TB progression pathways for further investigation.


Sujet(s)
Cellules dendritiques , Macrophages , Mycobacterium tuberculosis , Locus de caractère quantitatif , Tuberculose , Humains , Pérou , Tuberculose/génétique , Tuberculose/microbiologie , Macrophages/métabolisme , Macrophages/microbiologie , Mycobacterium tuberculosis/pathogénicité , Mycobacterium tuberculosis/génétique , Femelle , Cellules dendritiques/métabolisme , Mâle , Adulte , Prédisposition génétique à une maladie , Variation génétique , Régulation de l'expression des gènes , Adulte d'âge moyen , Polymorphisme de nucléotide simple , Analyse de profil d'expression de gènes
4.
EMBO Rep ; 25(7): 3064-3089, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38866980

RÉSUMÉ

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.


Sujet(s)
Mort cellulaire , Interféron de type I , Macrophages , Mycobacterium tuberculosis , Humains , Mycobacterium tuberculosis/immunologie , Mycobacterium tuberculosis/pathogénicité , Macrophages/microbiologie , Macrophages/métabolisme , Macrophages/immunologie , Interféron de type I/métabolisme , Transduction du signal , Tuberculose/microbiologie , Tuberculose/immunologie , Tuberculose/métabolisme , Animaux , Souris , Cellules cultivées
5.
PLoS One ; 19(6): e0304876, 2024.
Article de Anglais | MEDLINE | ID: mdl-38848336

RÉSUMÉ

We have identified an acyl-carrier protein, Rv0100, that is up-regulated in a dormancy model. This protein plays a critical role in the fatty acid biosynthesis pathway, which is important for energy storage and cell wall synthesis in Mycobacterium tuberculosis (MTB). Knocking out the Rv0100 gene resulted in a significant reduction of growth compared to wild-type MTB in the Wayne model of non-replicating persistence. We have also shown that Rv0100 is essential for the growth and survival of this pathogen during infection in mice and a macrophage model. Furthermore, knocking out Rv0100 disrupted the synthesis of phthiocerol dimycocerosates, the virulence-enhancing lipids produced by MTB and Mycobacterium bovis. We hypothesize that this essential gene contributes to MTB virulence in the state of latent infection. Therefore, inhibitors targeting this gene could prove to be potent antibacterial agents against this pathogen.


Sujet(s)
Protéine ACP , Protéines bactériennes , Mycobacterium tuberculosis , Animaux , Mycobacterium tuberculosis/métabolisme , Mycobacterium tuberculosis/génétique , Mycobacterium tuberculosis/pathogénicité , Souris , Protéines bactériennes/métabolisme , Protéines bactériennes/génétique , Protéine ACP/métabolisme , Protéine ACP/génétique , Macrophages/microbiologie , Macrophages/métabolisme , Virulence , Régulation de l'expression des gènes bactériens , Tuberculose/microbiologie , Lipides/composition chimique
6.
Pathog Dis ; 822024 Feb 07.
Article de Anglais | MEDLINE | ID: mdl-38845379

RÉSUMÉ

Tuberculosis (TB) continues to pose a significant global health challenge, emphasizing the critical need for effective preventive measures. Although many studies have tried to develop new attenuated vaccines, there is no effective TB vaccine. In this study, we report a novel attenuated Mycobacterium tuberculosis (M. tb) strain, CHVAC-25, cultured continuously for 25 years in the laboratory. CHVAC-25 exhibited significantly reduced virulence compared to both the virulent H37Rv strain in C57BL/6J and severe combined immunodeficiency disease mice. The comparative genomic analysis identified 93 potential absent genomic segments and 65 single nucleotide polymorphic sites across 47 coding genes. Notably, the deletion mutation of ppsC (Rv2933) involved in phthiocerol dimycocerosate synthesis likely contributes to CHVAC-25 virulence attenuation. Furthermore, the comparative analysis of immune responses between H37Rv- and CHVAC-25-infected macrophages showed that CHVAC-25 triggered a robust upregulation of 173 genes, particularly cytokines crucial for combating M. tb infection. Additionally, the survival of CHVAC-25 was significantly reduced compared to H37Rv in macrophages. These findings reiterate the possibility of obtaining attenuated M. tb strains through prolonged laboratory cultivation, echoing the initial conception of H37Ra nearly a century ago. Additionally, the similarity of CHVAC-25 to genotypes associated with attenuated M. tb vaccine positions it as a promising candidate for TB vaccine development.


Sujet(s)
Macrophages , Mycobacterium tuberculosis , Vaccins antituberculeux , Vaccins atténués , Mycobacterium tuberculosis/génétique , Mycobacterium tuberculosis/immunologie , Mycobacterium tuberculosis/pathogénicité , Animaux , Vaccins antituberculeux/immunologie , Vaccins antituberculeux/génétique , Souris , Macrophages/immunologie , Macrophages/microbiologie , Virulence/génétique , Vaccins atténués/immunologie , Vaccins atténués/génétique , Génome bactérien , Génomique/méthodes , Souris de lignée C57BL , Cytokines/métabolisme , Tuberculose/microbiologie , Tuberculose/immunologie , Tuberculose/prévention et contrôle , Polymorphisme de nucléotide simple , Modèles animaux de maladie humaine
7.
Int J Mol Sci ; 25(11)2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38892443

RÉSUMÉ

Mycobacterium tuberculosis (Mtb) is the causative agent of tuberculosis (TB), a prevalent infectious disease affecting populations worldwide. A classic trait of TB pathology is the formation of granulomas, which wall off the pathogen, via the innate and adaptive immune systems. Some key players involved include tumor necrosis factor-alpha (TNF-α), foamy macrophages, type I interferons (IFNs), and reactive oxygen species, which may also show overlap with cell death pathways. Additionally, host cell death is a primary method for combating and controlling Mtb within the body, a process which is influenced by both host and bacterial factors. These cell death modalities have distinct molecular mechanisms and pathways. Programmed cell death (PCD), encompassing apoptosis and autophagy, typically confers a protective response against Mtb by containing the bacteria within dead macrophages, facilitating their phagocytosis by uninfected or neighboring cells, whereas necrotic cell death benefits the pathogen, leading to the release of bacteria extracellularly. Apoptosis is triggered via intrinsic and extrinsic caspase-dependent pathways as well as caspase-independent pathways. Necrosis is induced via various pathways, including necroptosis, pyroptosis, and ferroptosis. Given the pivotal role of host cell death pathways in host defense against Mtb, therapeutic agents targeting cell death signaling have been investigated for TB treatment. This review provides an overview of the diverse mechanisms underlying Mtb-induced host cell death, examining their implications for host immunity. Furthermore, it discusses the potential of targeting host cell death pathways as therapeutic and preventive strategies against Mtb infection.


Sujet(s)
Mycobacterium tuberculosis , Tuberculose , Humains , Mycobacterium tuberculosis/immunologie , Mycobacterium tuberculosis/pathogénicité , Tuberculose/immunologie , Tuberculose/microbiologie , Tuberculose/anatomopathologie , Animaux , Mort cellulaire/immunologie , Interactions hôte-pathogène/immunologie , Apoptose , Immunité innée , Autophagie/immunologie , Transduction du signal , Macrophages/immunologie , Macrophages/microbiologie
8.
Methods Mol Biol ; 2813: 1-17, 2024.
Article de Anglais | MEDLINE | ID: mdl-38888767

RÉSUMÉ

Intracellular pathogens comprise a diverse group of pathogens that all share a required location in a host cell to infect, survive, and replicate. Intracellular location allows pathogens to hide from host immune responses, avoid competition with other pathogens, mediate host cellular functions, replicate safely, and cause infection that is difficult to target with therapeutics. All intracellular pathogens have varying routes of infiltration into host cells and different host cell preferences. For example, bacteria Mycobacterium tuberculosis chooses to invade antigen-presenting cells, which allows them to moderate host antigen presentation to memory cells, whereas rabies virus prefers to invade neurons because they have pre-existing innate immunity protection systems. Regardless of the pathway that each intracellular pathogen follows, all share the capacity to cause disease if they succeed in entering host cells. Here, we give an overview of selected intracellular pathogens and infections they cause, immune responses they induce, and intervention strategies used to treat and control them.


Sujet(s)
Interactions hôte-pathogène , Humains , Animaux , Interactions hôte-pathogène/immunologie , Mycobacterium tuberculosis/immunologie , Mycobacterium tuberculosis/pathogénicité , Immunité innée , Virus de la rage/immunologie , Virus de la rage/pathogénicité
9.
PLoS Pathog ; 20(6): e1011915, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38861581

RÉSUMÉ

Mycobacterium tuberculosis infects two billion people across the globe, and results in 8-9 million new tuberculosis (TB) cases and 1-1.5 million deaths each year. Most patients have no known genetic basis that predisposes them to disease. Here, we investigate the complex genetic basis of pulmonary TB by modelling human genetic diversity with the Diversity Outbred mouse population. When infected with M. tuberculosis, one-third develop early onset, rapidly progressive, necrotizing granulomas and succumb within 60 days. The remaining develop non-necrotizing granulomas and survive longer than 60 days. Genetic mapping using immune and inflammatory mediators; and clinical, microbiological, and granuloma correlates of disease identified five new loci on mouse chromosomes 1, 2, 4, 16; and three known loci on chromosomes 3 and 17. Further, multiple positively correlated traits shared loci on chromosomes 1, 16, and 17 and had similar patterns of allele effects, suggesting these loci contain critical genetic regulators of inflammatory responses to M. tuberculosis. To narrow the list of candidate genes, we used a machine learning strategy that integrated gene expression signatures from lungs of M. tuberculosis-infected Diversity Outbred mice with gene interaction networks to generate scores representing functional relationships. The scores were used to rank candidates for each mapped trait, resulting in 11 candidate genes: Ncf2, Fam20b, S100a8, S100a9, Itgb5, Fstl1, Zbtb20, Ddr1, Ier3, Vegfa, and Zfp318. Although all candidates have roles in infection, inflammation, cell migration, extracellular matrix remodeling, or intracellular signaling, and all contain single nucleotide polymorphisms (SNPs), SNPs in only four genes (S100a8, Itgb5, Fstl1, Zfp318) are predicted to have deleterious effects on protein functions. We performed methodological and candidate validations to (i) assess biological relevance of predicted allele effects by showing that Diversity Outbred mice carrying PWK/PhJ alleles at the H-2 locus on chromosome 17 QTL have shorter survival; (ii) confirm accuracy of predicted allele effects by quantifying S100A8 protein in inbred founder strains; and (iii) infection of C57BL/6 mice deficient for the S100a8 gene. Overall, this body of work demonstrates that systems genetics using Diversity Outbred mice can identify new (and known) QTLs and functionally relevant gene candidates that may be major regulators of complex host-pathogens interactions contributing to granuloma necrosis and acute inflammation in pulmonary TB.


Sujet(s)
Mycobacterium tuberculosis , Animaux , Mycobacterium tuberculosis/génétique , Mycobacterium tuberculosis/pathogénicité , Souris , Locus de caractère quantitatif , Tuberculose pulmonaire/génétique , Tuberculose pulmonaire/microbiologie , Tuberculose pulmonaire/anatomopathologie , Modèles animaux de maladie humaine , Lignées animales non consanguines , Humains , Cartographie chromosomique , Biologie des systèmes
10.
Microb Pathog ; 193: 106757, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38908454

RÉSUMÉ

The PE and PPE family proteins of Mycobacterium tuberculosis (Mtb) is exclusively found in pathogenic Mycobacterium species, comprising approximately 8-10 % of the Mtb genome. These emerging virulent factors have been observed to play pivotal roles in Mtb pathogenesis and immune evasion through various strategies. These immunogenic proteins are known to modulate the host immune response and cell-death pathways by targeting the powerhouse of the cell, the mitochondria to support Mtb survival. In this article, we are focused on how PE/PPE family proteins target host mitochondria to induce mitochondrial perturbations, modulate the levels of cellular ROS (Reactive oxygen species) and control cell death pathways. We observed that the time of expression of these proteins at different stages of infection is crucial for elucidating their impact on the cell death pathways and eventually on the outcome of infection. This article focuses on understanding the contributions of the PE/PPE proteins by unravelling the triad of host mitochondria, oxidative stress and cell death pathways that facilitate the Mtb persistence. Understanding the role of these proteins in host cellular pathways and the intricate mechanisms paves the way for the development of novel therapeutic strategies to combat TB infections.


Sujet(s)
Protéines bactériennes , Mort cellulaire , Interactions hôte-pathogène , Mitochondries , Mycobacterium tuberculosis , Stress oxydatif , Espèces réactives de l'oxygène , Mycobacterium tuberculosis/pathogénicité , Mycobacterium tuberculosis/métabolisme , Mycobacterium tuberculosis/génétique , Mitochondries/métabolisme , Protéines bactériennes/métabolisme , Protéines bactériennes/génétique , Humains , Espèces réactives de l'oxygène/métabolisme , Tuberculose/microbiologie , Tuberculose/métabolisme , Facteurs de virulence/métabolisme , Antigènes bactériens/métabolisme , Antigènes bactériens/génétique
11.
Nat Microbiol ; 9(6): 1607-1618, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38740932

RÉSUMÉ

Phthiocerol dimycocerosate (PDIM) is an essential virulence lipid of Mycobacterium tuberculosis. In vitro culturing rapidly selects for spontaneous PDIM-negative mutants that have attenuated virulence and increased cell wall permeability, thus impacting the relevance of experimental findings. PDIM loss can also reduce the efficacy of the BCG Pasteur vaccine. Here we show that vancomycin susceptibility can rapidly screen for M. tuberculosis PDIM production. We find that metabolic deficiency of methylmalonyl-CoA impedes the growth of PDIM-producing bacilli, selecting for PDIM-negative variants. Supplementation with odd-chain fatty acids, cholesterol or vitamin B12 restores PDIM-positive bacterial growth. Specifically, we show that propionate supplementation enhances PDIM-producing bacterial growth and selects against PDIM-negative mutants, analogous to in vivo conditions. Our study provides a simple approach to screen for and maintain PDIM production, and reveals how discrepancies between the host and in vitro nutrient environments can attenuate bacterial pathogenicity.


Sujet(s)
Mycobacterium tuberculosis , Propionates , Mycobacterium tuberculosis/effets des médicaments et des substances chimiques , Mycobacterium tuberculosis/métabolisme , Mycobacterium tuberculosis/pathogénicité , Mycobacterium tuberculosis/génétique , Mycobacterium tuberculosis/croissance et développement , Propionates/pharmacologie , Propionates/métabolisme , Virulence , Lipides/composition chimique , Cholestérol ester/métabolisme , Tuberculose/microbiologie , Tuberculose/prévention et contrôle , Acides gras/métabolisme , Vitamine B12/pharmacologie , Vitamine B12/métabolisme , Humains , Mutation , Facteurs de virulence/métabolisme , Facteurs de virulence/génétique , Cholestérol/métabolisme , Acyl coenzyme A
12.
Cell Mol Life Sci ; 81(1): 203, 2024 May 02.
Article de Anglais | MEDLINE | ID: mdl-38698289

RÉSUMÉ

Nitrogen metabolism of M. tuberculosis is critical for its survival in infected host cells. M. tuberculosis has evolved sophisticated strategies to switch between de novo synthesis and uptake of various amino acids from host cells for metabolic demands. Pyridoxal phosphate-dependent histidinol phosphate aminotransferase-HspAT enzyme is critically required for histidine biosynthesis. HspAT is involved in metabolic synthesis of histidine, phenylalanine, tyrosine, tryptophan, and novobiocin. We showed that M. tuberculosis Rv2231c is a conserved enzyme with HspAT activity. Rv2231c is a monomeric globular protein that contains α-helices and ß-sheets. It is a secretory and cell wall-localized protein that regulates critical pathogenic attributes. Rv2231c enhances the survival and virulence of recombinant M. smegmatis in infected RAW264.7 macrophage cells. Rv2231c is recognized by the TLR4 innate immune receptor and modulates the host immune response by suppressing the secretion of the antibacterial pro-inflammatory cytokines TNF, IL-12, and IL-6. It also inhibits the expression of co-stimulatory molecules CD80 and CD86 along with antigen presenting molecule MHC-I on macrophage and suppresses reactive nitrogen species formation, thereby promoting M2 macrophage polarization. Recombinant M. smegmatis expressing Rv2231c inhibited apoptosis in macrophages, promoting efficient bacterial survival and proliferation, thereby increasing virulence. Our results indicate that Rv2231c is a moonlighting protein that regulates multiple functions of M. tuberculosis pathophysiology to increase its virulence. These mechanistic insights can be used to better understand the pathogenesis of M. tuberculosis and to design strategies for tuberculosis mitigation.


Sujet(s)
Macrophages , Mycobacterium tuberculosis , Transaminases , Souris , Mycobacterium tuberculosis/pathogénicité , Mycobacterium tuberculosis/immunologie , Mycobacterium tuberculosis/enzymologie , Mycobacterium tuberculosis/métabolisme , Animaux , Cellules RAW 264.7 , Virulence , Macrophages/microbiologie , Macrophages/immunologie , Macrophages/métabolisme , Transaminases/métabolisme , Transaminases/génétique , Protéines bactériennes/métabolisme , Protéines bactériennes/génétique , Mycobacterium smegmatis/pathogénicité , Mycobacterium smegmatis/métabolisme , Mycobacterium smegmatis/génétique , Mycobacterium smegmatis/enzymologie , Cytokines/métabolisme , Récepteur de type Toll-4/métabolisme , Humains , Immunité innée , Interactions hôte-pathogène/immunologie , Tuberculose/immunologie , Tuberculose/microbiologie
13.
Nat Commun ; 15(1): 4216, 2024 May 17.
Article de Anglais | MEDLINE | ID: mdl-38760394

RÉSUMÉ

Antimicrobial peptides (AMPs), ancient scavengers of bacteria, are very poorly induced in macrophages infected by Mycobacterium tuberculosis (M. tuberculosis), but the underlying mechanism remains unknown. Here, we report that L-alanine interacts with PRSS1 and unfreezes the inhibitory effect of PRSS1 on the activation of NF-κB pathway to induce the expression of AMPs, but mycobacterial alanine dehydrogenase (Ald) Rv2780 hydrolyzes L-alanine and reduces the level of L-alanine in macrophages, thereby suppressing the expression of AMPs to facilitate survival of mycobacteria. Mechanistically, PRSS1 associates with TAK1 and disruptes the formation of TAK1/TAB1 complex to inhibit TAK1-mediated activation of NF-κB pathway, but interaction of L-alanine with PRSS1, disables PRSS1-mediated impairment on TAK1/TAB1 complex formation, thereby triggering the activation of NF-κB pathway to induce expression of AMPs. Moreover, deletion of antimicrobial peptide gene ß-defensin 4 (Defb4) impairs the virulence by Rv2780 during infection in mice. Both L-alanine and the Rv2780 inhibitor, GWP-042, exhibits excellent inhibitory activity against M. tuberculosis infection in vivo. Our findings identify a previously unrecognized mechanism that M. tuberculosis uses its own alanine dehydrogenase to suppress host immunity, and provide insights relevant to the development of effective immunomodulators that target M. tuberculosis.


Sujet(s)
Alanine , Peptides antimicrobiens , Macrophages , Mycobacterium tuberculosis , Facteur de transcription NF-kappa B , Tuberculose , Mycobacterium tuberculosis/pathogénicité , Mycobacterium tuberculosis/métabolisme , Animaux , Souris , Facteur de transcription NF-kappa B/métabolisme , Humains , Macrophages/microbiologie , Macrophages/métabolisme , Macrophages/immunologie , Alanine/métabolisme , Peptides antimicrobiens/métabolisme , Peptides antimicrobiens/génétique , Tuberculose/microbiologie , Tuberculose/immunologie , Alanine dehydrogenase/métabolisme , Alanine dehydrogenase/génétique , MAP Kinase Kinase Kinases/métabolisme , MAP Kinase Kinase Kinases/génétique , Protéines bactériennes/métabolisme , Protéines bactériennes/génétique , Transduction du signal , Souris de lignée C57BL , Cellules RAW 264.7 , Femelle
14.
Sci Rep ; 14(1): 12211, 2024 05 28.
Article de Anglais | MEDLINE | ID: mdl-38806568

RÉSUMÉ

This paper deals with the mathematical analysis of Tuberculosis by using fractal fractional operator. Mycobacterium TB is the bacteria that causes tuberculosis. This airborne illness mostly impacts the lungs but may extend to other body organs. When the infected individual coughs, sneezes or speaks, the bacterium gets released into the air and travels from one person to another. Five classes have been formulated to study the dynamics of this disease: susceptible class, infected of DS, infected of MDR, isolated class, and recovered class. To study the suggested fractal fractional model's wellposedness associated with existence results, and boundedness of solutions. Further, the invariant region of the considered model, positive solutions, equilibrium point, and reproduction number. One would typically employ a fractional calculus approach to obtain numerical solutions for the fractional order Tuberculosis model using the Adams-Bashforth-Moulton method. The fractional order derivatives in the model can be approximated using appropriate numerical schemes designed for fractional order differential equations.


Sujet(s)
Fractales , Mycobacterium tuberculosis , Tuberculose , Humains , Tuberculose/microbiologie , Mycobacterium tuberculosis/pathogénicité , Modèles théoriques , Modèles biologiques , Algorithmes
15.
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
16.
FEBS Lett ; 598(9): 1034-1044, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38639734

RÉSUMÉ

MTS1338, a distinctive small RNA in pathogenic mycobacteria, plays a crucial role in host-pathogen interactions during infection. Mycobacterial cells encounter heterogeneous stresses in macrophages, which highly upregulate MTS1338. A dormancy regulatory factor DosR regulates the intracellular abundance of MTS1338. Herein, we investigated the interplay of DosR and a low pH-inducible gene regulator PhoP binding to the MTS1338 promoter. We identified that DosR strongly binds to two regions upstream of the MTS1338 gene. The proximal region possesses a threefold higher affinity than the distal site, but the presence of both regions increased the affinity for DosR by > 10-fold. PhoP did not bind to the MTS1338 gene but binds to the DosR-bound MTS1338 gene, suggesting a concerted mechanism for MTS1338 expression.


Sujet(s)
Protéines bactériennes , Régulation de l'expression des gènes bactériens , Mycobacterium tuberculosis , Régions promotrices (génétique) , Activation de la transcription , Protéines bactériennes/métabolisme , Protéines bactériennes/génétique , Mycobacterium tuberculosis/génétique , Mycobacterium tuberculosis/pathogénicité , Mycobacterium tuberculosis/métabolisme , ARN bactérien/génétique , ARN bactérien/métabolisme , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Virulence/génétique , Liaison aux protéines , Petit ARN non traduit/génétique , Petit ARN non traduit/métabolisme
17.
Front Immunol ; 15: 1302163, 2024.
Article de Anglais | MEDLINE | ID: mdl-38515752

RÉSUMÉ

Mechanistic understanding of antibiotic persistence is a prerequisite in controlling the emergence of MDR cases in Tuberculosis (TB). We have reported that the cholesterol-induced activation of VapC12 ribonuclease is critical for disease persistence in TB. In this study, we observed that relative to the wild type, mice infected with ΔvapC12 induced a pro-inflammatory response, had a higher pathogen load, and responded better to the anti-TB treatment. In a high-dose infection model, all the mice infected with ΔvapC12 succumbed early to the disease. Finally, we reported that the above phenotype of ΔvapC12 was dependent on the presence of the TLR4 receptor. Overall, the data suggests that failure of a timely resolution of the early inflammation by the ΔvapC12 infected mice led to hyperinflammation, altered T-cell response and high bacterial load. In conclusion, our findings suggest the role of the VapC12 toxin in modulating the innate immune response of the host in ways that favor the long-term survival of the pathogen inside the host.


Sujet(s)
Mycobacterium tuberculosis , Ribonucléases , Tuberculose , Animaux , Souris , Immunité innée , Mycobacterium tuberculosis/génétique , Mycobacterium tuberculosis/pathogénicité , Phénotype , Toxines biologiques , Tuberculose/immunologie , Tuberculose/métabolisme , Ribonucléases/génétique , Ribonucléases/métabolisme
18.
Tuberculosis (Edinb) ; 143: 102421, 2023 12.
Article de Anglais | MEDLINE | ID: mdl-37879126

RÉSUMÉ

Mycobacterium tuberculosis secrets various effector proteins to evade host immune responses for facilitating its intracellular survival. The bacterial genome encodes several unique PE/PPE family proteins, which have been implicated to play important role in mycobacterial pathogenesis. A member of this family, PPE2 have been shown to contain a monopartite nuclear localization signal (NLS) and a DNA binding domain. In this study, we demonstrate that PPE2 protein is present in the sera of mice infected with either M. smegmatis expressing PPE2 or a clinical strain of M. tuberculosis (CDC1551). It was found that exogenously added PPE2 can permeate through the macrophage cell membrane and eventually translocate into the nucleus which requires the presence of NLS which showed considerable homology to HIV-tat like cell permeable peptides. Exogenously added PPE2 could inhibit NO production and decreased mycobacterial survival in macrophages. PPE2-null mutant of M. tuberculosis failed to inhibit NO production and had poor survival in macrophages which could be rescued by complementation with full-length PPE2. PPE2-null mutants also had poor survival in the lungs of infected mice indicating that PPE2 even when present in the bloodstream can confer a survival advantage to mycobacteria.


Sujet(s)
Mycobacterium tuberculosis , Tuberculose , Animaux , Souris , Antigènes bactériens/métabolisme , Protéines bactériennes/métabolisme , Interactions hôte-pathogène , Mycobacterium smegmatis/génétique , Mycobacterium smegmatis/métabolisme , Mycobacterium tuberculosis/génétique , Mycobacterium tuberculosis/pathogénicité , Tuberculose/métabolisme , Tuberculose/microbiologie
19.
Microbes Infect ; 25(6): 105126, 2023.
Article de Anglais | MEDLINE | ID: mdl-36931492

RÉSUMÉ

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.


Sujet(s)
Protéines du choc thermique HSP27 , Protéines du choc thermique HSP70 , Macrophages , Infections à Mycobacterium , Mycobacterium tuberculosis , Humains , Protéines du choc thermique/métabolisme , Macrophages/microbiologie , Infections à Mycobacterium/métabolisme , Mycobacterium tuberculosis/pathogénicité , Lymphocytes T , Protéines du choc thermique HSP70/immunologie , Protéines du choc thermique HSP70/métabolisme , Protéines du choc thermique HSP27/immunologie , Protéines du choc thermique HSP27/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE