Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.662
Filtrer
1.
Virulence ; 15(1): 2375549, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38982595

RÉSUMÉ

CagA is a significant oncogenic factor injected into host cells by Helicobacter pylori, which is divided into two subtypes: East Asian type (CagAE), characterized by the EPIYA-D motif, and western type (CagAW), harboring the EPIYA-C motif. CagAE has been reported to have higher carcinogenicity than CagAW, although the underlying reason is not fully understood. SHIP2 is an intracellular phosphatase that can be recruited by CagA to perturb the homeostasis of intracellular signaling pathways. In this study, we found that SHIP2 contributes to the higher oncogenicity of CagAE. Co-Immunoprecipitation and Pull-down assays showed that CagAE bind more SHIP2 than CagAW. Immunofluorescence staining showed that a higher amount of SHIP2 recruited by CagAE to the plasma membrane catalyzes the conversion of PI(3,4,5)P3 into PI(3,4)P2. This alteration causes higher activation of Akt signaling, which results in enhanced IL-8 secretion, migration, and invasion of the infected cells. SPR analysis showed that this stronger interaction between CagAE and SHIP2 stems from the higher affinity between the EPIYA-D motif of CagAE and the SH2 domain of SHIP2. Structural analysis revealed the crucial role of the Phe residue at the Y + 5 position in EPIYA-D. After mutating Phe of CagAE into Asp (the corresponding residue in the EPIYA-C motif) or Ala, the activation of downstream Akt signaling was reduced and the malignant transformation of infected cells was alleviated. These findings revealed that CagAE hijacks SHIP2 through its EPIYA-D motif to enhance its carcinogenicity, which provides a better understanding of the higher oncogenic risk of H. pylori CagAE.


Sujet(s)
Motifs d'acides aminés , Antigènes bactériens , Protéines bactériennes , Infections à Helicobacter , Helicobacter pylori , Phosphatidylinositol-3,4,5-trisphosphate 5-phosphatases , Humains , Protéines bactériennes/métabolisme , Protéines bactériennes/génétique , Helicobacter pylori/génétique , Helicobacter pylori/pathogénicité , Phosphatidylinositol-3,4,5-trisphosphate 5-phosphatases/génétique , Phosphatidylinositol-3,4,5-trisphosphate 5-phosphatases/métabolisme , Antigènes bactériens/métabolisme , Antigènes bactériens/génétique , Infections à Helicobacter/microbiologie , Transduction du signal , Carcinogenèse , Liaison aux protéines , Peuples d'Asie de l'Est
2.
Front Cell Infect Microbiol ; 14: 1419568, 2024.
Article de Anglais | MEDLINE | ID: mdl-38983115

RÉSUMÉ

Background: Helicobacter pylori infection poses a significant health burden worldwide, and its virulence factor CagA plays a pivotal role in its pathogenesis. Methods: In this study, the interaction between H. pylori-infected AGS cells and silver nanoparticles (AgNPs) was investigated, with a focus on the modulation of CagA-mediated responses, investigated by western blotting. Both, the dose-dependent efficacy against H. pylori (growth curves, CFU assay) and the impact of the nanoparticles on AGS cells (MTT assay) were elucidated. Results: AGS cells infected with H. pylori displayed dramatic morphological changes, characterized by elongation and a migratory phenotype, attributed to CagA activity. Preincubation of H. pylori with AgNPs affected these morphological changes in a concentration-dependent manner, suggesting a correlation between AgNPs concentration and CagA function. Conclusion: Our study highlights the nuanced interplay between host-pathogen interactions and the therapeutic potential of AgNPs in combating H. pylori infection and offers valuable insights into the multifaceted dynamics of CagA mediated responses.


Sujet(s)
Antigènes bactériens , Protéines bactériennes , Infections à Helicobacter , Helicobacter pylori , Nanoparticules métalliques , Transduction du signal , Argent , Helicobacter pylori/effets des médicaments et des substances chimiques , Protéines bactériennes/métabolisme , Antigènes bactériens/métabolisme , Argent/pharmacologie , Argent/métabolisme , Humains , Infections à Helicobacter/microbiologie , Infections à Helicobacter/traitement médicamenteux , Transduction du signal/effets des médicaments et des substances chimiques , Interactions hôte-pathogène , Cellules épithéliales/microbiologie , Facteurs de virulence/métabolisme , Lignée cellulaire , Antibactériens/pharmacologie , Lignée cellulaire tumorale
3.
World J Microbiol Biotechnol ; 40(9): 273, 2024 Jul 20.
Article de Anglais | MEDLINE | ID: mdl-39030443

RÉSUMÉ

Helicobacter pylori is a common resident in the stomach of at least half of the world's population and recent evidence suggest its emergence in other organs such as the pancreas. In this organ, the presence of H. pylori DNA has been reported in cats, although the functional implications remain unknown. In this work, we determined distinct features related to the H. pylori manifestation in pancreas in a rodent model, in order to analyse its functional and structural effect. Gerbils inoculated with H. pylori exhibited the presence of this bacterium, as revealed by the expression of some virulence factors, as CagA and OMPs in stomach and pancreas, and confirmed by urease activity, bacterial culture, PCR and immunofluorescence assays. Non-apparent morphological changes were observed in pancreatic tissue of infected animals; however, delocalization of intercellular junction proteins (claudin-1, claudin-4, occludin, ZO-1, E-cadherin, ß-catenin, desmoglein-2 and desmoplakin I/II) and rearrangement of the actin-cytoskeleton were exhibited. This structural damage was consistent with alterations in the distribution of insulin and glucagon, and a systemic inflammation, event demonstrated by elevated IL-8 levels. Overall, these findings indicate that H. pylori can reach the pancreas, possibly affecting its function and contributing to the development of pancreatic diseases.


Sujet(s)
Gerbillinae , Infections à Helicobacter , Helicobacter pylori , Jonctions intercellulaires , Pancréas , Animaux , Helicobacter pylori/pathogénicité , Helicobacter pylori/génétique , Infections à Helicobacter/microbiologie , Pancréas/microbiologie , Pancréas/anatomopathologie , Jonctions intercellulaires/microbiologie , Protéines bactériennes/métabolisme , Protéines bactériennes/génétique , Antigènes bactériens/métabolisme , Antigènes bactériens/génétique , Facteurs de virulence/métabolisme , Facteurs de virulence/génétique , Estomac/microbiologie , Estomac/anatomopathologie , Modèles animaux de maladie humaine , Mâle , Protéines de la membrane externe bactérienne/métabolisme , Protéines de la membrane externe bactérienne/génétique
4.
Int J Mol Sci ; 25(13)2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-39000189

RÉSUMÉ

Impaired E-cadherin (Cdh1) functions are closely associated with cellular dedifferentiation, infiltrative tumor growth and metastasis, particularly in gastric cancer. The class-I carcinogen Helicobacter pylori (H. pylori) colonizes gastric epithelial cells and induces Cdh1 shedding, which is primarily mediated by the secreted bacterial protease high temperature requirement A (HtrA). In this study, we used human primary epithelial cell lines derived from gastroids and mucosoids from different healthy donors to investigate HtrA-mediated Cdh1 cleavage and the subsequent impact on bacterial pathogenesis in a non-neoplastic context. We found a severe impairment of Cdh1 functions by HtrA-induced ectodomain cleavage in 2D primary cells and mucosoids. Since mucosoids exhibit an intact apico-basal polarity, we investigated bacterial transmigration across the monolayer, which was partially depolarized by HtrA, as indicated by microscopy, the analyses of the transepithelial electrical resistance (TEER) and colony forming unit (cfu) assays. Finally, we investigated CagA injection and observed efficient CagA translocation and tyrosine phosphorylation in 2D primary cells and, to a lesser extent, similar effects in mucosoids. In summary, HtrA is a crucially important factor promoting the multistep pathogenesis of H. pylori in non-transformed primary gastric epithelial cells and organoid-based epithelial models.


Sujet(s)
Protéines bactériennes , Cadhérines , Cellules épithéliales , Muqueuse gastrique , Helicobacter pylori , Organoïdes , Humains , Cadhérines/métabolisme , Organoïdes/métabolisme , Cellules épithéliales/métabolisme , Cellules épithéliales/microbiologie , Muqueuse gastrique/métabolisme , Muqueuse gastrique/microbiologie , Muqueuse gastrique/anatomopathologie , Protéines bactériennes/métabolisme , Protéines bactériennes/génétique , Antigènes bactériens/métabolisme , Infections à Helicobacter/métabolisme , Infections à Helicobacter/microbiologie , Infections à Helicobacter/anatomopathologie , Antigènes CD/métabolisme , Estomac/microbiologie , Estomac/anatomopathologie , Lignée cellulaire , Tumeurs de l'estomac/métabolisme , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/microbiologie , Protéases à sérine
5.
Sci Rep ; 14(1): 14185, 2024 06 20.
Article de Anglais | MEDLINE | ID: mdl-38902391

RÉSUMÉ

Helicobacter pylori (H. pylori), together with its CagA, has been implicated in causing DNA damage, cell cycle arrest, apoptosis, and the development of gastric cancer. Although lncRNA H19 is abundantly expressed in gastric cancer and functions as a pro-oncogene, it remains unclear whether lncRNA H19 contributes to the oncogenic process of H. pylori CagA. This study investigates the role of H19 in the DNA damage response and malignancy induced by H. pylori. It was observed that cells infected with CagA+ H. pylori strain (GZ7/cagA) showed significantly higher H19 expression, resulting in increased γH2A.X and p-ATM expression and decreased p53 and Rad51 expression. Faster cell migration and invasion was also observed, which was reversed by H19 knockdown in H. pylori. YWHAZ was identified as an H19 target protein, and its expression was increased in H19 knockdown cells. GZ7/cagA infection responded to the increased YWHAZ expression induced by H19 knockdown. In addition, H19 knockdown stimulated cells to enter the G2-phase and attenuated the effect of GZ7/cagA infection on the cellular S-phase barrier. The results suggest that H. pylori CagA can upregulate H19 expression, participate in the DNA damage response and promote cell migration and invasion, and possibly affect cell cycle arrest via regulation of YWHAZ.


Sujet(s)
Antigènes bactériens , Protéines bactériennes , Mouvement cellulaire , Altération de l'ADN , Helicobacter pylori , ARN long non codant , Tumeurs de l'estomac , Humains , Antigènes bactériens/métabolisme , Antigènes bactériens/génétique , ARN long non codant/génétique , ARN long non codant/métabolisme , Protéines bactériennes/génétique , Protéines bactériennes/métabolisme , Helicobacter pylori/génétique , Tumeurs de l'estomac/microbiologie , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/métabolisme , Mouvement cellulaire/génétique , Lignée cellulaire tumorale , Infections à Helicobacter/microbiologie , Infections à Helicobacter/génétique , Infections à Helicobacter/métabolisme , Rad51 Recombinase/métabolisme , Rad51 Recombinase/génétique , Protéine p53 suppresseur de tumeur/métabolisme , Protéine p53 suppresseur de tumeur/génétique , Histone/métabolisme
6.
EBioMedicine ; 105: 105196, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38880068

RÉSUMÉ

BACKGROUND: The ability to detect evidence of Mycobacterium tuberculosis (Mtb) infection within human tissues is critical to the study of Mtb physiology, tropism, and spatial distribution within TB lesions. The capacity of the widely-used Ziehl-Neelsen (ZN) staining method for identifying Mtb acid-fast bacilli (AFB) in tissue is highly variable, which can limit detection of Mtb bacilli for research and diagnostic purposes. Here, we sought to circumvent these limitations via detection of Mtb mRNA and secreted antigens in human tuberculous tissue. METHODS: We adapted RNAscope, an RNA in situ hybridisation (RISH) technique, to detect Mtb mRNA in ante- and postmortem human TB tissues and developed a dual ZN/immunohistochemistry staining approach to identify AFB and bacilli producing antigen 85B (Ag85B). FINDINGS: We identified Mtb mRNA within intact and disintegrating bacilli as well as extrabacillary mRNA. Mtb mRNA was distributed zonally within necrotic and non-necrotic granulomas. We also found Mtb mRNA within, and adjacent to, necrotic granulomas in ZN-negative lung tissue and in Ag85B-positive bronchiolar epithelium. Intriguingly, we observed accumulation of Mtb mRNA and Ag85B in the cytoplasm of host cells. Notably, many AFB were negative for Ag85B staining. Mtb mRNA was observed in ZN-negative antemortem lymph node biopsies. INTERPRETATION: RNAscope and dual ZN/immunohistochemistry staining are well-suited for identifying subsets of intact Mtb and/or bacillary remnants in human tissue. RNAscope can identify Mtb mRNA in ZN-negative tissues from patients with TB and may have diagnostic potential in complex TB cases. FUNDING: Wellcome Leap Delta Tissue Program, Wellcome Strategic Core Award, the National Institutes of Health (NIH, USA), the Mary Heersink Institute for Global Health at UAB, the UAB Heersink School of Medicine.


Sujet(s)
Antigènes bactériens , Mycobacterium tuberculosis , ARN messager , Humains , Mycobacterium tuberculosis/génétique , Antigènes bactériens/génétique , Antigènes bactériens/métabolisme , ARN messager/génétique , ARN messager/métabolisme , Hybridation in situ , Tuberculose/microbiologie , ARN bactérien/génétique , Immunohistochimie , Granulome/microbiologie , Granulome/métabolisme , Poumon/microbiologie , Poumon/anatomopathologie , Poumon/métabolisme
7.
Infect Immun ; 92(7): e0015224, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38888310

RÉSUMÉ

The major gram-positive pathogen group A Streptococcus (GAS) is a model organism for studying microbial epidemics as it causes waves of infections. Since 1980, several GAS epidemics have been ascribed to the emergence of clones producing increased amounts of key virulence factors such as streptolysin O (SLO). Herein, we sought to identify mechanisms underlying our recently identified temporal clonal emergence among emm4 GAS, given that emergent strains did not produce augmented levels of virulence factors relative to historic isolates. By creating and analyzing isoallelic strains, we determined that a conserved mutation in a previously undescribed gene encoding a putative carbonic anhydrase was responsible for the defective in vitro growth observed in the emergent strains. We also identified that the emergent strains survived better inside macrophages and killed macrophages at lower rates than the historic strains. Via the creation of isogenic mutant strains, we linked the emergent strain "survival" phenotype to the downregulation of the SLO encoding gene and upregulation of the msrAB operon which encodes proteins involved in defense against extracellular oxidative stress. Our findings are in accord with recent surveillance studies which found a high ratio of mucosal (i.e., pharyngeal) relative to invasive infections among emm4 GAS. Since ever-increasing virulence is unlikely to be evolutionarily advantageous for a microbial pathogen, our data further understanding of the well-described oscillating patterns of virulent GAS infections by demonstrating mechanisms by which emergent strains adapt a "survival" strategy to outcompete previously circulating isolates.


Sujet(s)
Protéines bactériennes , Macrophages , Infections à streptocoques , Streptococcus pyogenes , Streptolysines , Facteurs de virulence , Streptococcus pyogenes/génétique , Streptococcus pyogenes/pathogénicité , Streptococcus pyogenes/immunologie , Infections à streptocoques/microbiologie , Infections à streptocoques/immunologie , Infections à streptocoques/mortalité , Humains , Macrophages/microbiologie , Macrophages/immunologie , Protéines bactériennes/génétique , Protéines bactériennes/métabolisme , Streptolysines/génétique , Streptolysines/métabolisme , Facteurs de virulence/génétique , Mutation , Interactions hôte-pathogène/immunologie , Virulence/génétique , Animaux , Antigènes bactériens/génétique , Antigènes bactériens/métabolisme , Antigènes bactériens/immunologie , Viabilité microbienne , Protéines de la membrane externe bactérienne/génétique , Protéines de la membrane externe bactérienne/métabolisme , Souris , Régulation de l'expression des gènes bactériens , Protéines de transport
8.
Helicobacter ; 29(3): e13100, 2024.
Article de Anglais | MEDLINE | ID: mdl-38873839

RÉSUMÉ

BACKGROUND: The formation of gallstones is often accompanied by chronic inflammation, and the mechanisms underlying inflammation and stone formation are not fully understood. Our aim is to utilize single-cell transcriptomics, bulk transcriptomics, and microbiome data to explore key pathogenic bacteria that may contribute to chronic inflammation and gallstone formation, as well as their associated mechanisms. METHODS: scRNA-seq data from a gallstone mouse model were extracted from the Gene Expression Omnibus (GEO) database and analyzed using the FindCluster() package for cell clustering analysis. Bulk transcriptomics data from patients with gallstone were also extracted from the GEO database, and intergroup functional differences were assessed using GO and KEGG enrichment analysis. Additionally, 16S rRNA sequencing was performed on gallbladder mucosal samples from asymptomatic patients with gallstone (n = 6) and liver transplant donor gallbladder mucosal samples (n = 6) to identify key bacteria associated with stone formation and chronic inflammation. Animal models were constructed to investigate the mechanisms by which these key pathogenic bacterial genera promote gallstone formation. RESULTS: Analysis of scRNA-seq data from the gallstone mouse model (GSE179524) revealed seven distinct cell clusters, with a significant increase in neutrophil numbers in the gallstone group. Analysis of bulk transcriptomics data from patients with gallstone (GSE202479) identified chronic inflammation in the gallbladder, potentially associated with dysbiosis of the gallbladder microbiota. 16S rRNA sequencing identified Helicobacter pylori as a key bacterium associated with gallbladder chronic inflammation and stone formation. CONCLUSIONS: Dysbiosis of the gallbladder mucosal microbiota is implicated in gallstone disease and leads to chronic inflammation. This study identified H. pylori as a potential key mucosal resident bacterium contributing to gallstone formation and discovered its key pathogenic factor CagA, which causes damage to the gallbladder mucosal barrier. These findings provide important clues for the prevention and treatment of gallstones.


Sujet(s)
Antigènes bactériens , Protéines bactériennes , Cellules épithéliales , Vésicule biliaire , Calculs biliaires , Helicobacter pylori , Animaux , Calculs biliaires/microbiologie , Calculs biliaires/anatomopathologie , Cellules épithéliales/microbiologie , Souris , Humains , Vésicule biliaire/microbiologie , Vésicule biliaire/anatomopathologie , Protéines bactériennes/génétique , Protéines bactériennes/métabolisme , Antigènes bactériens/génétique , Antigènes bactériens/métabolisme , Helicobacter pylori/génétique , Helicobacter pylori/pathogénicité , Helicobacter pylori/physiologie , ARN ribosomique 16S/génétique , Modèles animaux de maladie humaine , Perméabilité , Infections à Helicobacter/microbiologie , Infections à Helicobacter/anatomopathologie , Femelle , Mâle , Souris de lignée C57BL
9.
Life Sci Alliance ; 7(8)2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38834194

RÉSUMÉ

Vinculin is a cytoskeletal linker strengthening cell adhesion. The Shigella IpaA invasion effector binds to vinculin to promote vinculin supra-activation associated with head-domain-mediated oligomerization. Our study investigates the impact of mutations of vinculin D1D2 subdomains' residues predicted to interact with IpaA VBS3. These mutations affected the rate of D1D2 trimer formation with distinct effects on monomer disappearance, consistent with structural modeling of a closed and open D1D2 conformer induced by IpaA. Notably, mutations targeting the closed D1D2 conformer significantly reduced Shigella invasion of host cells as opposed to mutations targeting the open D1D2 conformer and later stages of vinculin head-domain oligomerization. In contrast, all mutations affected the formation of focal adhesions (FAs), supporting the involvement of vinculin supra-activation in this process. Our findings suggest that IpaA-induced vinculin supra-activation primarily reinforces matrix adhesion in infected cells, rather than promoting bacterial invasion. Consistently, shear stress studies pointed to a key role for IpaA-induced vinculin supra-activation in accelerating and strengthening cell-matrix adhesion.


Sujet(s)
Adhérence cellulaire , Contacts focaux , Vinculine , Vinculine/métabolisme , Vinculine/génétique , Humains , Contacts focaux/métabolisme , Protéines bactériennes/métabolisme , Protéines bactériennes/génétique , Mutation , Interactions hôte-pathogène , Cellules HeLa , Liaison aux protéines , Shigella/métabolisme , Shigella/génétique , Antigènes bactériens/métabolisme , Antigènes bactériens/génétique , Dysenterie bacillaire/microbiologie , Dysenterie bacillaire/métabolisme
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.
J Biol Chem ; 300(7): 107478, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38879009

RÉSUMÉ

Antigenically sequence variable M proteins of the major bacterial pathogen Streptococcus pyogenes (Strep A) are responsible for recruiting human C4b-binding protein (C4BP) to the bacterial surface, which enables Strep A to evade destruction by the immune system. The most sequence divergent portion of M proteins, the hypervariable region (HVR), is responsible for binding C4BP. Structural evidence points to the conservation of two C4BP-binding sequence patterns (M2 and M22) in the HVR of numerous M proteins, with this conservation applicable to vaccine immunogen design. These two patterns, however, only partially explain C4BP binding by Strep A. Here, we identified several M proteins that lack these patterns but still bind C4BP and determined the structures of two, M68 and M87 HVRs, in complex with a C4BP fragment. Mutagenesis of these M proteins led to the identification of amino acids that are crucial for C4BP binding, enabling formulation of new C4BP-binding patterns. Mutagenesis was also carried out on M2 and M22 proteins to refine or generate experimentally grounded C4BP-binding patterns. The M22 pattern was the most prevalent among M proteins, followed by the M87 and M2 patterns, while the M68 pattern was rare. These patterns, except for M68, were also evident in numerous M-like Enn proteins. Binding of C4BP via these patterns to Enn proteins was verified. We conclude that C4BP-binding patterns occur frequently in Strep A strains of differing M types, being present in their M or Enn proteins, or frequently both, providing further impetus for their use as vaccine immunogens.


Sujet(s)
Antigènes bactériens , Protéine de liaison à C4b , Streptococcus pyogenes , Streptococcus pyogenes/métabolisme , Streptococcus pyogenes/génétique , Streptococcus pyogenes/composition chimique , Protéine de liaison à C4b/métabolisme , Antigènes bactériens/métabolisme , Antigènes bactériens/composition chimique , Antigènes bactériens/génétique , Humains , Protéines de la membrane externe bactérienne/métabolisme , Protéines de la membrane externe bactérienne/composition chimique , Protéines de la membrane externe bactérienne/génétique , Protéines de transport/métabolisme , Protéines de transport/génétique , Protéines de transport/composition chimique , Liaison aux protéines , Séquence d'acides aminés , Protéines bactériennes/métabolisme , Protéines bactériennes/composition chimique , Protéines bactériennes/génétique
12.
Infect Immun ; 92(6): e0009024, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38700336

RÉSUMÉ

bb0616 of Borrelia burgdorferi, the Lyme disease pathogen, encodes a hypothetical protein of unknown function. In this study, we showed that BB0616 was not surface-exposed or associated with the membrane through localization analyses using proteinase K digestion and cell partitioning assays. The expression of bb0616 was influenced by a reduced pH but not by growth phases, elevated temperatures, or carbon sources during in vitro cultivation. A transcriptional start site for bb0616 was identified by using 5' rapid amplification of cDNA ends, which led to the identification of a functional promoter in the 5' regulatory region upstream of bb0616. By analyzing a bb0616-deficient mutant and its isogenic complemented counterparts, we found that the infectivity potential of the mutant was significantly attenuated. The inactivation of bb0616 displayed no effect on borrelial growth in the medium or resistance to oxidative stress, but the mutant was significantly more susceptible to osmotic stress. In addition, the production of global virulence regulators such as BosR and RpoS as well as virulence-associated outer surface lipoproteins OspC and DbpA was reduced in the mutant. These phenotypes were fully restored when gene mutation was complemented with a wild-type copy of bb0616. Based on these findings, we concluded that the hypothetical protein BB0616 is required for the optimal infectivity of B. burgdorferi, potentially by impacting B. burgdorferi virulence gene expression as well as survival of the spirochete under stressful conditions.


Sujet(s)
Protéines bactériennes , Borrelia burgdorferi , Régulation de l'expression des gènes bactériens , Maladie de Lyme , Borrelia burgdorferi/génétique , Borrelia burgdorferi/pathogénicité , Borrelia burgdorferi/métabolisme , Animaux , Souris , Maladie de Lyme/microbiologie , Protéines bactériennes/génétique , Protéines bactériennes/métabolisme , Régions promotrices (génétique) , Facteurs de virulence/génétique , Facteurs de virulence/métabolisme , Virulence , Souris de lignée C3H , Facteur sigma/génétique , Facteur sigma/métabolisme , Protéines de la membrane externe bactérienne/génétique , Protéines de la membrane externe bactérienne/métabolisme , Site d'initiation de la transcription , Antigènes bactériens/génétique , Antigènes bactériens/métabolisme , Test de complémentation , Concentration en ions d'hydrogène
13.
Cell Mol Biol Lett ; 29(1): 70, 2024 May 13.
Article de Anglais | MEDLINE | ID: mdl-38741147

RÉSUMÉ

BACKGROUND: Mycobacterium tuberculosis heat-resistant antigen (Mtb-HAg) is a peptide antigen released from the mycobacterial cytoplasm into the supernatant of Mycobacterium tuberculosis (Mtb) attenuated H37Ra strain after autoclaving at 121 °C for 20 min. Mtb-HAg can specifically induce γδ T-cell proliferation in vitro. However, the exact composition of Mtb-HAg and the protein antigens that are responsible for its function are currently unknown. METHODS: Mtb-HAg extracted from the Mtb H37Ra strain was subjected to LC‒MS mass spectrometry. Twelve of the identified protein fractions were recombinantly expressed in Escherichia coli by genetic engineering technology using pET-28a as a plasmid and purified by Ni-NTA agarose resin to stimulate peripheral blood mononuclear cells (PBMCs) from different healthy individuals. The proliferation of γδ T cells and major γδ T-cell subset types as well as the production of TNF-α and IFN-γ were determined by flow cytometry. Their proliferating γδ T cells were isolated and purified using MACS separation columns, and Mtb H37Ra-infected THP-1 was co-cultured with isolated and purified γδ T cells to quantify Mycobacterium viability by counting CFUs. RESULTS: In this study, Mtb-HAg from the attenuated Mtb H37Ra strain was analysed by LC‒MS mass spectrometry, and a total of 564 proteins were identified. Analysis of the identified protein fractions revealed that the major protein components included heat shock proteins and Mtb-specific antigenic proteins. Recombinant expression of 10 of these proteins in by Escherichia coli genetic engineering technology was used to successfully stimulate PBMCs from different healthy individuals, but 2 of the proteins, EsxJ and EsxA, were not expressed. Flow cytometry results showed that, compared with the IL-2 control, HspX, GroEL1, and GroES specifically induced γδ T-cell expansion, with Vγ2δ2 T cells as the main subset, and the secretion of the antimicrobial cytokines TNF-α and IFN-γ. In contrast, HtpG, DnaK, GroEL2, HbhA, Mpt63, EsxB, and EsxN were unable to promote γδ T-cell proliferation and the secretion of TNF-α and IFN-γ. None of the above recombinant proteins were able to induce the secretion of TNF-α and IFN-γ by αß T cells. In addition, TNF-α, IFN-γ-producing γδ T cells inhibit the growth of intracellular Mtb. CONCLUSION: Activated γδ T cells induced by Mtb-HAg components HspX, GroES, GroEL1 to produce TNF-α, IFN-γ modulate macrophages to inhibit intracellular Mtb growth. These data lay the foundation for subsequent studies on the mechanism by which Mtb-HAg induces γδ T-cell proliferation in vitro, as well as the development of preventive and therapeutic vaccines and rapid diagnostic reagents.


Sujet(s)
Antigènes bactériens , Prolifération cellulaire , Mycobacterium tuberculosis , Lymphocytes T , Humains , Antigènes bactériens/immunologie , Antigènes bactériens/métabolisme , Antigènes bactériens/génétique , Mycobacterium tuberculosis/immunologie , Mycobacterium tuberculosis/génétique , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Interféron gamma/métabolisme , Interféron gamma/immunologie , Récepteur lymphocytaire T antigène, gamma-delta/métabolisme , Récepteur lymphocytaire T antigène, gamma-delta/immunologie , Récepteur lymphocytaire T antigène, gamma-delta/génétique , Facteur de nécrose tumorale alpha/métabolisme , Agranulocytes/métabolisme , Agranulocytes/immunologie , Protéines bactériennes/métabolisme , Protéines bactériennes/génétique , Protéines bactériennes/immunologie
14.
Elife ; 122024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38805257

RÉSUMÉ

Mycobacterium tuberculosis (Mtb) is known to survive within macrophages by compromising the integrity of the phagosomal compartment in which it resides. This activity primarily relies on the ESX-1 secretion system, predominantly involving the protein duo ESAT-6 and CFP-10. CFP-10 likely acts as a chaperone, while ESAT-6 likely disrupts phagosomal membrane stability via a largely unknown mechanism. we employ a series of biochemical analyses, protein modeling techniques, and a novel ESAT-6-specific nanobody to gain insight into the ESAT-6's mode of action. First, we measure the binding kinetics of the tight 1:1 complex formed by ESAT-6 and CFP-10 at neutral pH. Subsequently, we demonstrate a rapid self-association of ESAT-6 into large complexes under acidic conditions, leading to the identification of a stable tetrameric ESAT-6 species. Using molecular dynamics simulations, we pinpoint the most probable interaction interface. Furthermore, we show that cytoplasmic expression of an anti-ESAT-6 nanobody blocks Mtb replication, thereby underlining the pivotal role of ESAT-6 in intracellular survival. Together, these data suggest that ESAT-6 acts by a pH-dependent mechanism to establish two-way communication between the cytoplasm and the Mtb-containing phagosome.


Sujet(s)
Antigènes bactériens , Protéines bactériennes , Macrophages , Mycobacterium tuberculosis , Phagosomes , Anticorps à domaine unique , Humains , Antigènes bactériens/métabolisme , Antigènes bactériens/immunologie , Protéines bactériennes/métabolisme , Concentration en ions d'hydrogène , Macrophages/immunologie , Macrophages/métabolisme , Macrophages/microbiologie , Simulation de dynamique moléculaire , Mycobacterium tuberculosis/immunologie , Mycobacterium tuberculosis/métabolisme , Phagosomes/métabolisme , Anticorps à domaine unique/métabolisme
15.
mSphere ; 9(5): e0000524, 2024 May 29.
Article de Anglais | MEDLINE | ID: mdl-38661343

RÉSUMÉ

The mycobacterial cell envelope is a major virulence determinant in pathogenic mycobacteria. Specific outer lipids play roles in pathogenesis, modulating the immune system and promoting the secretion of virulence factors. ESX-1 (ESAT-6 system-1) is a conserved protein secretion system required for mycobacterial pathogenesis. Previous studies revealed that mycobacterial strains lacking the outer lipid PDIM have impaired ESX-1 function during laboratory growth and infection. The mechanisms underlying changes in ESX-1 function are unknown. We used a proteo-genetic approach to measure phthiocerol dimycocerosate (PDIM)- and phenolic glycolipid (PGL)-dependent protein secretion in M. marinum, a non-tubercular mycobacterial pathogen that causes tuberculosis-like disease in ectothermic animals. Importantly, M. marinum is a well-established model for mycobacterial pathogenesis. Our findings showed that M. marinum strains without PDIM and PGL showed specific, significant reductions in protein secretion compared to the WT and complemented strains. We recently established a hierarchy for the secretion of ESX-1 substrates in four (I-IV) groups. Loss of PDIM differentially impacted secretion of Group III and IV ESX-1 substrates, which are likely the effectors of pathogenesis. Our data suggest that the altered secretion of specific ESX-1 substrates is responsible for the observed ESX-1-related effects in PDIM-deficient strains.IMPORTANCEMycobacterium tuberculosis, the cause of human tuberculosis, killed an estimated 1.3 million people in 2022. Non-tubercular mycobacterial species cause acute and chronic human infections. Understanding how these bacteria cause disease is critical. Lipids in the cell envelope are essential for mycobacteria to interact with the host and promote disease. Strains lacking outer lipids are attenuated for infection, but the reasons are unclear. Our research aims to identify a mechanism for attenuation of mycobacterial strains without the PDIM and PGL outer lipids in M. marinum. These findings will enhance our understanding of the importance of lipids in pathogenesis and how these lipids contribute to other established virulence mechanisms.


Sujet(s)
Protéines bactériennes , Glycolipides , Mycobacterium marinum , Facteurs de virulence , Mycobacterium marinum/pathogénicité , Mycobacterium marinum/génétique , Mycobacterium marinum/métabolisme , Protéines bactériennes/génétique , Protéines bactériennes/métabolisme , Facteurs de virulence/génétique , Facteurs de virulence/métabolisme , Glycolipides/métabolisme , Virulence , Lipides , Antigènes bactériens/métabolisme , Antigènes bactériens/génétique
16.
Microb Pathog ; 190: 106636, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38556103

RÉSUMÉ

Enterotoxigenic Escherichia coli (ETEC) is one of the main causes of diarrhea in children and travelers in low-income regions. The virulence of ETEC is attributed to its heat-labile and heat-stable enterotoxins, as well as its colonization factors (CFs). CFs are essential for ETEC adherence to the intestinal epithelium. However, its invasive capability remains unelucidated. In this study, we demonstrated that the CS6-positive ETEC strain 4266 can invade mammalian epithelial cells. The invasive capability was reduced in the 4266 ΔCS6 mutant but reintroduction of CS6 into this mutant restored the invasiveness. Additionally, the laboratory E. coli strain Top 10, which lacks the invasive capability, was able to invade Caco-2 cells after gaining the CS6-expressing plasmid pCS6. Cytochalasin D inhibited cell invasion in both 4266 and Top10 pCS6 cells, and F-actin accumulation was observed near the bacteria on the cell membrane, indicating that CS6-positive bacteria were internalized via actin polymerization. Other cell signal transduction inhibitors, such as genistein, wortmannin, LY294002, PP1, and Ro 32-0432, inhibited the CS6-mediated invasion of Caco-2 cells. The internalized bacteria of both 4266 and Top10 pCS6 strains were able to survive for up to 48 h, and 4266 cells were able to replicate within Caco-2 cells. Immunofluorescence microscopy revealed that the internalized 4266 cells were present in bacteria-containing vacuoles, which underwent a maturation process indicated by the recruitment of the early endosomal marker EEA-1 and late endosomal marker LAMP-1 throughout the infection process. The autophagy marker LC3 was also observed near these vacuoles, indicating the initiation of LC-3-associated phagocytosis (LAP). However, intracellular bacteria continued to replicate, even after the initiation of LAP. Moreover, intracellular filamentation was observed in 4266 cells at 24 h after infection. Overall, this study shows that CS6, in addition to being a major CF, mediates cell invasion. This demonstrates that once internalized, CS6-positive ETEC is capable of surviving and replicating within host cells. This capability may be a key factor in the extended and recurrent nature of ETEC infections in humans, thus highlighting the critical role of CS6.


Sujet(s)
Cytochalasine D , Escherichia coli entérotoxigène , Protéines Escherichia coli , Humains , Cellules Caco-2 , Escherichia coli entérotoxigène/pathogénicité , Escherichia coli entérotoxigène/génétique , Escherichia coli entérotoxigène/métabolisme , Protéines Escherichia coli/génétique , Protéines Escherichia coli/métabolisme , Cytochalasine D/pharmacologie , Actines/métabolisme , Cellules épithéliales/microbiologie , Adhérence bactérienne , Infections à Escherichia coli/microbiologie , Virulence , Facteurs de virulence/génétique , Facteurs de virulence/métabolisme , Antigènes bactériens/métabolisme , Antigènes bactériens/génétique , Morpholines/pharmacologie , Transduction du signal , Androstadiènes/pharmacologie , Wortmannine/pharmacologie , Endocytose , 4H-1-Benzopyran-4-ones/pharmacologie , Plasmides/génétique
17.
Front Immunol ; 15: 1363962, 2024.
Article de Anglais | MEDLINE | ID: mdl-38515758

RÉSUMÉ

Introduction: Pancreatic ductal adenocarcinoma (PDAC) is a lethal cancer associated with an immunosuppressive environment. Neutrophil extracellular traps (NETs) were initially described in the context of infection but have more recently been implicated in contributing to the tolerogenic immune response in PDAC. Thus, NETs are an attractive target for new therapeutic strategies. Group A Streptococcus (GAS) has developed defensive strategies to inhibit NETs. Methods: In the present work, we propose utilizing intra-tumoral GAS injection to stimulate anti-tumor activity by inhibiting cancer-promoting NETs. Mice harboring Panc02 or KPC subcutaneous tumors injected with three different M-type GAS strains. Tumors and spleens were harvested at the endpoint of the experiments to assess bacterial colonization and systemic spread, while sera were analyzed for humoral responses toward the streptococcal antigens, especially the M1 and Scl1 proteins. Role of the streptococcal collagen-like protein 1 (Scl1) in anti-PDAC activity was assessed in vivo after intratumoral injection with M1 GAS wild-type, an isogenic mutant strain devoid of Scl1, or a complemented mutant strain with restored scl1 expression. In addition, recombinant Scl1 proteins were tested for NET inhibition using in vitro and ex vivo assays assessing NET production and myeloperoxidase activity. Results: Injection of three different M-type GAS strains reduced subcutaneous pancreatic tumor volume compared to control in two different murine PDAC models. Limitation of tumor growth was dependent on Scl1, as isogenic mutant strain devoid of Scl1 did not reduce tumor size. We further show that Scl1 plays a role in localizing GAS to the tumor site, thereby limiting the systemic spread of bacteria and off-target effects. While mice did elicit a humoral immune response to GAS antigens, tested sera were weakly immunogenic toward Scl1 antigen following intra-tumoral treatment with Scl1-expressing GAS. M1 GAS inhibited NET formation when co-cultured with neutrophils while Scl1-devoid mutant strain did not. Recombinant Scl1 protein inhibited NETs ex vivo in a dose-dependent manner by suppressing myeloperoxidase activity. Discussion: Altogether, we demonstrate that intra-tumoral GAS injections reduce PDAC growth, which is facilitated by Scl1, in part through inhibition of cancer promoting NETs. This work offers a novel strategy by which NETs can be targeted through Scl1 protein and potentiates its use as a cancer therapeutic.


Sujet(s)
Adénocarcinome , Pièges extracellulaires , Tumeurs du pancréas , Animaux , Souris , Protéines bactériennes , Pièges extracellulaires/métabolisme , Collagène/métabolisme , Antigènes bactériens/métabolisme , Collagène de type I/métabolisme , Streptococcus pyogenes , Myeloperoxidase/métabolisme
18.
Mol Cell Proteomics ; 23(5): 100753, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38527648

RÉSUMÉ

Bacterial or viral antigens can contain subdominant protein regions that elicit weak antibody responses upon vaccination or infection although there is accumulating evidence that antibody responses against subdominant regions can enhance the protective immune response. One proposed mechanism for subdominant protein regions is the binding of host proteins that prevent antibody production against epitopes hidden within the protein binding interfaces. Here, we used affinity purification combined with quantitative mass spectrometry (AP-MS) to examine the level of competition between antigen-specific antibodies and host-pathogen protein interaction networks using the M1 protein from Streptococcus pyogenes as a model system. As most humans have circulating antibodies against the M1 protein, we first used AP-MS to show that the M1 protein interspecies protein network formed with human plasma proteins is largely conserved in naïve mice. Immunizing mice with the M1 protein generated a time-dependent increase of anti-M1 antibodies. AP-MS analysis comparing the composition of the M1-plasma protein network from naïve and immunized mice showed significant enrichment of 292 IgG peptides associated with 56 IgG chains in the immune mice. Despite the significant increase of bound IgGs, the levels of interacting plasma proteins were not significantly reduced in the immune mice. The results indicate that the antigen-specific polyclonal IgG against the M1 protein primarily targets epitopes outside the other plasma protein binding interfaces. In conclusion, this study demonstrates that AP-MS is a promising strategy to determine the relationship between antigen-specific antibodies and host-pathogen interaction networks that could be used to define subdominant protein regions of relevance for vaccine development.


Sujet(s)
Antigènes bactériens , Immunoglobuline G , Liaison aux protéines , Streptococcus pyogenes , Animaux , Streptococcus pyogenes/immunologie , Streptococcus pyogenes/métabolisme , Antigènes bactériens/immunologie , Antigènes bactériens/métabolisme , Souris , Humains , Immunoglobuline G/immunologie , Immunoglobuline G/métabolisme , Immunité acquise , Protéines de la membrane externe bactérienne/immunologie , Protéines de la membrane externe bactérienne/métabolisme , Anticorps antibactériens/immunologie , Cartes d'interactions protéiques , Spectrométrie de masse , Protéines de transport/métabolisme , Protéines de transport/immunologie , Femelle , Interactions hôte-pathogène/immunologie
19.
Helicobacter ; 29(2): e13066, 2024.
Article de Anglais | MEDLINE | ID: mdl-38468575

RÉSUMÉ

BACKGROUND: SHP1 has been documented as a tumor suppressor and it was thought to play an antagonistic role in the pathogenesis of Helicobacter pylori infection. In this study, the exact mechanism of this antagonistic action was studied. MATERIALS AND METHODS: AGS, MGC803, and GES-1 cells were infected with H. pylori, intracellular distribution changes of SHP1 were first detected by immunofluorescence. SHP1 overexpression and knockdown were then constructed in these cells to investigate its antagonistic roles in H. pylori infection. Migration and invasion of infected cells were detected by transwell assay, secretion of IL-8 was examined via ELISA, the cells with hummingbird-like alteration were determined by microexamination, and activation of JAK2/STAT3, PI3K/Akt, and ERK pathways were detected by immunoblotting. Mice infection model was established and gastric pathological changes were evaluated. Finally, the SHP1 activator sorafenib was used to analyze the attenuating effect of SHP1 activation on H. pylori pathogenesis in vitro and in vivo. RESULTS: The sub-localization of SHP1 changed after H. pylori infection, specifically that the majority of the cytoplasmic SHP1 was transferred to the cell membrane. SHP1 inhibited H. pylori-induced activation of JAK2/STAT3 pathway, PI3K/Akt pathway, nuclear translocation of NF-κB, and then reduced EMT, migration, invasion, and IL-8 secretion. In addition, SHP1 inhibited the formation of CagA-SHP2 complex by dephosphorylating phosphorylated CagA, reduced ERK phosphorylation and the formation of CagA-dependent hummingbird-like cells. In the mice infection model, gastric pathological changes were observed and increased IL-8 secretion, indicators of cell proliferation and EMT progression were also detected. By activating SHP1 with sorafenib, a significant curative effect against H. pylori infection was obtained in vitro and in vivo. CONCLUSIONS: SHP1 plays an antagonistic role in H. pylori pathogenesis by inhibiting JAK2/STAT3 and PI3K/Akt pathways, NF-κB nuclear translocation, and CagA phosphorylation, thereby reducing cell EMT, migration, invasion, IL-8 secretion, and hummingbird-like changes.


Sujet(s)
Infections à Helicobacter , Helicobacter pylori , Animaux , Souris , Protéines bactériennes/métabolisme , Antigènes bactériens/métabolisme , Helicobacter pylori/physiologie , Facteur de transcription NF-kappa B/métabolisme , Interleukine-8/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Infections à Helicobacter/anatomopathologie , Sorafénib/métabolisme , Cellules épithéliales/métabolisme
20.
Int J Mol Sci ; 25(6)2024 Mar 07.
Article de Anglais | MEDLINE | ID: mdl-38542074

RÉSUMÉ

Lethal toxin (LT) is the critical virulence factor of Bacillus anthracis, the causative agent of anthrax. One common symptom observed in patients with anthrax is thrombocytopenia, which has also been observed in mice injected with LT. Our previous study demonstrated that LT induces thrombocytopenia by suppressing megakaryopoiesis, but the precise molecular mechanisms behind this phenomenon remain unknown. In this study, we utilized 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced megakaryocytic differentiation in human erythroleukemia (HEL) cells to identify genes involved in LT-induced megakaryocytic suppression. Through cDNA microarray analysis, we identified Dachshund homolog 1 (DACH1) as a gene that was upregulated upon TPA treatment but downregulated in the presence of TPA and LT, purified from the culture supernatants of B. anthracis. To investigate the function of DACH1 in megakaryocytic differentiation, we employed short hairpin RNA technology to knock down DACH1 expression in HEL cells and assessed its effect on differentiation. Our data revealed that the knockdown of DACH1 expression suppressed megakaryocytic differentiation, particularly in polyploidization. We demonstrated that one mechanism by which B. anthracis LT induces suppression of polyploidization in HEL cells is through the cleavage of MEK1/2. This cleavage results in the downregulation of the ERK signaling pathway, thereby suppressing DACH1 gene expression and inhibiting polyploidization. Additionally, we found that known megakaryopoiesis-related genes, such as FOSB, ZFP36L1, RUNX1, FLI1, AHR, and GFI1B genes may be positively regulated by DACH1. Furthermore, we observed an upregulation of DACH1 during in vitro differentiation of CD34-megakaryocytes and downregulation of DACH1 in patients with thrombocytopenia. In summary, our findings shed light on one of the molecular mechanisms behind LT-induced thrombocytopenia and unveil a previously unknown role for DACH1 in megakaryopoiesis.


Sujet(s)
Maladie du charbon , Bacillus anthracis , Leucémie érythroblastique aigüe , Thrombopénie , Animaux , Humains , Souris , Antigènes bactériens/métabolisme , Bacillus anthracis/métabolisme , Facteur BRF-1/métabolisme , Différenciation cellulaire , Thrombopénie/induit chimiquement , Thrombopénie/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE