Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.668
Filtrar
1.
Sci Rep ; 14(1): 21257, 2024 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-39261531

RESUMEN

The bacterium Clostridium botulinum, well-known for producing botulinum neurotoxins, which cause the severe paralytic illness known as botulism, produces C2 toxin, a binary AB-toxin with ADP-ribosyltranferase activity. C2 toxin possesses two separate protein components, an enzymatically active A-component C2I and the binding and translocation B-component C2II. After proteolytic activation of C2II to C2IIa, the heptameric structure binds C2I and is taken up via receptor-mediated endocytosis into the target cells. Due to acidification of endosomes, the C2IIa/C2I complex undergoes conformational changes and consequently C2IIa forms a pore into the endosomal membrane and C2I can translocate into the cytoplasm, where it ADP-ribosylates G-actin, a key component of the cytoskeleton. This modification disrupts the actin cytoskeleton, resulting in the collapse of cytoskeleton and ultimately cell death. Here, we show that the serine-protease inhibitor α1-antitrypsin (α1AT) which we identified previously from a hemofiltrate library screen for PT from Bordetella pertussis is a multitoxin inhibitor. α1AT inhibits intoxication of cells with C2 toxin via inhibition of binding to cells and inhibition of enzyme activity of C2I. Moreover, diphtheria toxin and an anthrax fusion toxin are inhibited by α1AT. Since α1AT is commercially available as a drug for treatment of the α1AT deficiency, it could be repurposed for treatment of toxin-mediated diseases.


Asunto(s)
Toxinas Bacterianas , Toxinas Botulínicas , alfa 1-Antitripsina , Toxinas Botulínicas/metabolismo , Toxinas Botulínicas/antagonistas & inhibidores , Toxinas Botulínicas/química , Humanos , alfa 1-Antitripsina/metabolismo , alfa 1-Antitripsina/química , Toxinas Bacterianas/metabolismo , Toxina Diftérica/metabolismo , Corynebacterium diphtheriae/metabolismo , Corynebacterium diphtheriae/efectos de los fármacos , Antígenos Bacterianos/metabolismo , Animales , Clostridium botulinum/metabolismo , Bacillus anthracis/metabolismo , Bacillus anthracis/efectos de los fármacos
2.
Sci Rep ; 14(1): 21648, 2024 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-39289452

RESUMEN

Helicobacter pylori (H. pylori) is one of the most common bacterial infections in the world, and its key virulence component CagA is the leading cause of gastric cancer. Mitophagy is a form of selective autophagy that eliminates damaged mitochondria and is essential for some viruses and bacteria to evade the immune system. However, the mechanisms by which CagA mediates H. pylori-induced mitophagy and NLRP3 inflammasome activation remain elusive. In this study, we reported that H. pylori primarily uses its CagA to induce mitochondrial oxidative damage, mitochondrial dysfunction, dynamic imbalance, and to block autophagic flux. Inhibition of mitophagy led to an increase in NLRP3 inflammasome activation and apoptosis and a decrease in the viability of H. pylori-infected cells. Our findings suggested that H. pylori induces mitochondrial dysfunction and mitophagy primarily via CagA. It reduces NLRP3 inflammasome activation to evade host immune surveillance and increases the survival and viability of infected cells, potentially leading to gastric cancer initiation and development. Our findings provide new insights into the pathogenesis of H. pylori-induced gastric cancer, and inhibition of mitophagy may be one of the novel techniques for the prevention and treatment of this disease.


Asunto(s)
Antígenos Bacterianos , Proteínas Bacterianas , Helicobacter pylori , Inflamasomas , Mitocondrias , Mitofagia , Proteína con Dominio Pirina 3 de la Familia NLR , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/metabolismo , Inflamasomas/metabolismo , Helicobacter pylori/patogenicidad , Helicobacter pylori/fisiología , Humanos , Mitocondrias/metabolismo , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/metabolismo , Infecciones por Helicobacter/inmunología , Supervivencia Celular , Neoplasias Gástricas/microbiología , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Apoptosis
3.
Cancer Biomark ; 41(2): 93-101, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39331090

RESUMEN

BACKGROUND: The association between infection with cagA-positive H. pylori and an elevated susceptibility to gastric cancer has been firmly established. PIM2 is known to be overexpressed in various types of cancers; however, the specific mechanism by which cagA influences the regulation of PIM2 expression in gastric cancer remains unidentified at present. MATERIALS AND METHODS: A mutant NCTC11637ΔcagA strain of H. pylori and the eukaryotic expression vector pcDNA-cagA were constructed for evaluating PIM2 expression levels in gastric cancer cells (HGC27, SGC7901, and AG) co-cultured with the NCTC11637 and NCTC11637ΔcagA strain, as well as pcDNA-cagA and the empty vector pcDNA3.1 (+). RESULTS: Co-culturing gastric cancer cells with NCTC11637 significantly increased PIM2 expression levels (P< 0.001) compared to the negative control group. Additionally, the expression of PIM2 in cells co-cultured with NCTC11637 was higher than that co-cultured with NCTC11637ΔcagA (P< 0.001). Furthermore, successful construction of the eukaryotic expression vector pcDNA-cagA resulted in a significant increase in PIM2 mRNA expression levels after its transfection into gastric cancer cells compared to the control group after 48 hours. CONCLUSIONS: The findings indicate that H. pylori/cagA A could be one of the key factors in regulating PIM2 expression levels, potentially influencing the progression of H. pylori-related Gastric Cancer.


Asunto(s)
Antígenos Bacterianos , Proteínas Bacterianas , Helicobacter pylori , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas , Neoplasias Gástricas , Neoplasias Gástricas/genética , Neoplasias Gástricas/microbiología , Neoplasias Gástricas/patología , Neoplasias Gástricas/metabolismo , Humanos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Antígenos Bacterianos/genética , Antígenos Bacterianos/metabolismo , Línea Celular Tumoral , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Infecciones por Helicobacter/genética , Infecciones por Helicobacter/microbiología , Regulación Neoplásica de la Expresión Génica , Técnicas de Cocultivo
4.
Microb Pathog ; 195: 106898, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39208956

RESUMEN

Helicobacter pylori infection has been thought to be associated with liver diseases, although the exact mechanisms remain elusive. This study identified H. pylori-induced liver inflammation and tissue damage in infected mice and examined the exosome-mediated mechanism underlying H. pylori infection's impact on liver injury. Exosomes were isolated from H. pylori-infected gastric epithelial GES-1 cells (Hp-GES-EVs), and the crucial virulence factor CagA was identified within these exosomes. Fluorescent labeling demonstrated that Hp-GES-EVs can be absorbed by liver cells. Treatment with Hp-GES-EVs enhanced the proliferation, migration, and invasion of Hep G2 and Hep 3B cells. Additionally, exposure to Hp-GES-EVs activated NF-κB and PI3K/AKT signaling pathways, which provides a reasonable explanation for the liver inflammation and neoplastic traits. Using a mouse model established via tail vein injection of Hp-GES-EVs, exosome-driven liver injury was evidenced by slight hepatocellular erosion around the central hepatic vein and elevated serum levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), and IL-6. Administering the exosome inhibitor GW4869 via intraperitoneal injection in mice resulted in a reduction of liver damage caused by H. pylori infection. These findings illuminate the exosome-mediated pathogenesis of H. pylori-induced liver injury and offer valuable insights into the extra-gastrointestinal manifestations of H. pylori infection.


Asunto(s)
Antígenos Bacterianos , Proteínas Bacterianas , Modelos Animales de Enfermedad , Exosomas , Infecciones por Helicobacter , Helicobacter pylori , Hígado , Transducción de Señal , Exosomas/metabolismo , Animales , Infecciones por Helicobacter/complicaciones , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/metabolismo , Helicobacter pylori/patogenicidad , Ratones , Humanos , Proteínas Bacterianas/metabolismo , Hígado/patología , Hígado/metabolismo , Hígado/microbiología , Antígenos Bacterianos/metabolismo , Compuestos de Bencilideno/farmacología , Compuestos de Anilina/farmacología , FN-kappa B/metabolismo , Células Hep G2 , Aspartato Aminotransferasas/sangre , Interleucina-6/metabolismo , Alanina Transaminasa/sangre , Proliferación Celular , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Células Epiteliales/microbiología , Células Epiteliales/metabolismo , Movimiento Celular , Línea Celular , Masculino , Factores de Virulencia/metabolismo
5.
Int J Biol Sci ; 20(10): 4007-4028, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39113698

RESUMEN

Cholesterol and Helicobacter pylori (H. pylori) are both risk factors for gastric cancer (GC). However, the relationship between cholesterol and H. pylori and their function in the progression of GC are controversial. In this study, we addressed that H. pylori could induce mitochondrial cholesterol accumulation and promote GC proliferation and protect GC cells against apoptosis via cholesterol. Metabolomic and transcriptomic sequencing were used to identify CYP11A1 responsible for H. pylori-induced cholesterol accumulation. In vitro and in vivo function experiments revealed that cholesterol could promote the proliferation of GC and inhibit apoptosis. Mechanically, the interaction of Cytotoxin-associated gene A (CagA) and CYP11A1 redistributed mitochondrial CYP11A1 outside the mitochondria and subsequently caused mitochondrial cholesterol accumulation. The CYP11A1-knockdown upregulated cholesterol accumulation and reproduced the effect of cholesterol on GC in a cholesterol-dependent manner. Moreover, CYP11A1-knockdown or H. pylori infection inhibited mitophagy and maintained the mitochondria homeostasis. H. pylori could contribute to the progression of GC through the CagA/CYP11A1-mitoCHO axis. This study demonstrates that H. pylori can contribute to the progression of GC via cholesterol, and eradicating H. pylori is still prognostically beneficial to GC patients.


Asunto(s)
Colesterol , Helicobacter pylori , Mitocondrias , Neoplasias Gástricas , Helicobacter pylori/metabolismo , Neoplasias Gástricas/microbiología , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Colesterol/metabolismo , Humanos , Mitocondrias/metabolismo , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/metabolismo , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/genética , Infecciones por Helicobacter/metabolismo , Infecciones por Helicobacter/microbiología , Animales , Antígenos Bacterianos/metabolismo , Antígenos Bacterianos/genética , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Línea Celular Tumoral , Ratones , Apoptosis , Masculino , Proliferación Celular
6.
Life Sci Alliance ; 7(11)2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39191487

RESUMEN

Helicobacter pylori infection predisposes carriers to a high risk of developing gastric cancer. The cell-of-origin of antral gastric cancer is the Lgr5+ stem cell. Here, we show that infection of antrum-derived gastric organoid cells with H. pylori increases the expression of the stem cell marker Lgr5 as determined by immunofluorescence microscopy, qRT-PCR, and Western blotting, both when cells are grown and infected as monolayers and when cells are exposed to H. pylori in 3D structures. H. pylori exposure increases stemness properties as determined by spheroid formation assay. Lgr5 expression and the acquisition of stemness depend on a functional type IV secretion system (T4SS) and at least partly on the T4SS effector CagA. The pharmacological inhibition or genetic ablation of NF-κB reverses the increase in Lgr5 and spheroid formation. Constitutively active Wnt/ß-catenin signaling because of Apc inactivation exacerbates H. pylori-induced Lgr5 expression and stemness, both of which persist even after eradication of the infection. The combined data indicate that H. pylori has stemness-inducing properties that depend on its ability to activate NF-κB signaling.


Asunto(s)
Infecciones por Helicobacter , Helicobacter pylori , FN-kappa B , Receptores Acoplados a Proteínas G , Neoplasias Gástricas , Vía de Señalización Wnt , Animales , Ratones , Antígenos Bacterianos/metabolismo , Antígenos Bacterianos/genética , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Mucosa Gástrica/metabolismo , Mucosa Gástrica/microbiología , Infecciones por Helicobacter/metabolismo , Infecciones por Helicobacter/microbiología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/microbiología , FN-kappa B/metabolismo , Organoides/metabolismo , Organoides/microbiología , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Células Madre/metabolismo , Estómago/microbiología , Estómago/patología , Neoplasias Gástricas/microbiología , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Sistemas de Secreción Tipo IV/metabolismo , Sistemas de Secreción Tipo IV/genética , Vía de Señalización Wnt/genética
7.
J Biotechnol ; 394: 73-84, 2024 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-39173715

RESUMEN

ApxII is a vaccine antigen used to protect against porcine contagious pleuropneumonia, which is a significant threat to the pig industry. Here, we aimed to improve the proteolytic degradation stability of ApxII during its secretion by establishing a complete screening process of stable variants through bioinformatics and site-directed mutagenesis. We employed a combination of semi-rational and rational design strategies to create 34 single-point variants of ApxII. Among them, R114E and T115D variants exhibited better stability without compromising antigen activity. Furthermore, we constructed a multi-site variant, R114E/T115D, which demonstrated the best stability, activity, and yield. Protein stability and molecular dynamic analysis indicated that the greater solubility and lower structural expansion coefficient might explain the increased stability of R114E/T115D. Additionally, site T115 was identified as a key point of truncated ApxII stability. The R114E/T115D variant, with its proven stability and intact antigenic activity, holds promising prospects for industrial-scale applications in the prevention of porcine contagious pleuropneumonia.


Asunto(s)
Antígenos Bacterianos , Corynebacterium glutamicum , Mutagénesis Sitio-Dirigida , Estabilidad Proteica , Corynebacterium glutamicum/genética , Corynebacterium glutamicum/metabolismo , Antígenos Bacterianos/genética , Antígenos Bacterianos/metabolismo , Antígenos Bacterianos/inmunología , Antígenos Bacterianos/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/química , Animales , Porcinos , Simulación de Dinámica Molecular
8.
PLoS Pathog ; 20(8): e1012144, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39172739

RESUMEN

Several reports suggest that intestinal tissue may be a natural niche for Chlamydia trachomatis infection and a reservoir for persistent infections in the human body. Due to the human specificity of the pathogen and the lack of suitable host models, there is limited knowledge on this topic. In our study, we modelled the course of the chlamydial infection in human primary gastrointestinal (GI) epithelial cells originating from patient-derived organoids. We show that GI cells are resistant to apical infection and C. trachomatis needs access to the basolateral membrane to establish an infection. Transmission electron microscopy analysis reveals the presence of both normal as well as aberrant chlamydial developmental forms in the infected cells, suggesting a possible cell-type specific nature of the infection. Furthermore, we show that the plasmid-encoded Pgp3 is an important virulence factor for the infection of human GI cells. This is the first report of C. trachomatis infection in human primary intestinal epithelial cells supporting a possible niche for chlamydial infection in the human intestinal tissue.


Asunto(s)
Infecciones por Chlamydia , Chlamydia trachomatis , Organoides , Humanos , Chlamydia trachomatis/fisiología , Organoides/microbiología , Organoides/patología , Infecciones por Chlamydia/microbiología , Mucosa Intestinal/microbiología , Células Epiteliales/microbiología , Antígenos Bacterianos/metabolismo , Proteínas Bacterianas
9.
PLoS One ; 19(8): e0307172, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39173001

RESUMEN

BACKGROUND: Helicobacter pylori (H. pylori) is frequently associated with non-cardia type gastric cancer, and it is designated as a group I carcinogen. This study aimed to systematically review and meta-analyze the evidence on the prevalence of CagA status in people with gastric disorders in the Indo-Pacific region, and to examine the association of CagA positive in the risk of gastric disorders. This study focused on the Indo-Pacific region owing to the high disability adjusted life-years related to these disorders, the accessibility of efficient treatments for this common bacterial infection, and the varying standard of care for these disorders, particularly among the elderly population in the region. METHODS: Relevant studies were identified in the health-related electronic databases including PubMed, Ovid, Medline, Ovid Embase, Index Medicus, and Google Scholar that were published in English between 1 January 2000, and 18 November 2023. For pooled prevalence, meta-analysis of proportional studies was done, after Freeman-Tukey double arcsine transformation of data. A random-effect model was used to compute the pooled odds ratio (OR) and 95% confidence interval (CI) to investigate the relationship between CagA positivity and gastric disorders. RESULTS: Twenty-four studies from eight Indo-Pacific countries (Bhutan, India, Indonesia, Malaysia, Myanmar, Singapore, Thailand, Vietnam) were included. Overall pooled prevalence of CagA positivity in H. pylori-infected gastric disorders was 83% (95%CI = 73-91%). Following stratification, the pooled prevalence of CagA positivity was 78% (95%CI = 67-90%) in H. pylori-infected gastritis, 86% (95%CI = 73-96%) in peptic ulcer disease, and 83% (95%CI = 51-100%) in gastric cancer. Geographic locations encountered variations in CagA prevalence. There was a greater risk of developing gastric cancer in those with CagA positivity compared with gastritis (OR = 2.53,95%CI = 1.15-5.55). CONCLUSION: Findings suggest that the distribution of CagA in H. pylori-infected gastric disorders varies among different type of gastric disorders in the study countries, and CagA may play a role in the development of gastric cancer. It is important to provide a high standard of care for the management of gastric diseases, particularly in a region where the prevalence of these disorders is high. Better strategies for effective treatment for high-risk groups are required for health programs to revisit this often-neglected infectious disease.


Asunto(s)
Antígenos Bacterianos , Proteínas Bacterianas , Infecciones por Helicobacter , Helicobacter pylori , Humanos , Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/epidemiología , Infecciones por Helicobacter/complicaciones , Estudios Observacionales como Asunto , Neoplasias Gástricas/microbiología , Neoplasias Gástricas/epidemiología , Prevalencia , Fenotipo
10.
Virulence ; 15(1): 2375549, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38982595

RESUMEN

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.


Asunto(s)
Secuencias de Aminoácidos , Antígenos Bacterianos , Proteínas Bacterianas , Infecciones por Helicobacter , Helicobacter pylori , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas , Humanos , Antígenos Bacterianos/metabolismo , Antígenos Bacterianos/genética , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Carcinogénesis , Pueblos del Este de Asia , Infecciones por Helicobacter/microbiología , Helicobacter pylori/genética , Helicobacter pylori/patogenicidad , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas/genética , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas/metabolismo , Unión Proteica , Transducción de Señal
11.
Mol Microbiol ; 122(2): 230-242, 2024 08.
Artículo en Inglés | MEDLINE | ID: mdl-38994873

RESUMEN

Enterococcus faecalis is an opportunistic pathogen frequently causing nosocomial infections. The virulence of this organism is underpinned by its capacity to evade phagocytosis, allowing dissemination in the host. Immune evasion requires a surface polysaccharide produced by all enterococci, known as the enterococcal polysaccharide antigen (EPA). EPA consists of a cell wall-anchored rhamnose backbone substituted by strain-specific polysaccharides called 'decorations', essential for the biological activity of this polymer. However, the structural determinants required for innate immune evasion remain unknown, partly due to a lack of suitable validated assays. Here, we describe a quantitative, in vitro assay to investigate how EPA decorations alter phagocytosis. Using the E. faecalis model strain OG1RF, we demonstrate that a mutant with a deletion of the locus encoding EPA decorations can be used as a platform strain to express heterologous decorations, thereby providing an experimental system to investigate the inhibition of phagocytosis by strain-specific decorations. We show that the aggregation of cells lacking decorations is increasing phagocytosis and that this process does not involve the recognition of lipoproteins by macrophages. Collectively, our work provides novel insights into innate immune evasion by enterococci and paves the way for further studies to explore the structure/function relationship of EPA decorations.


Asunto(s)
Enterococcus faecalis , Evasión Inmune , Lipoproteínas , Macrófagos , Fagocitosis , Enterococcus faecalis/inmunología , Enterococcus faecalis/metabolismo , Enterococcus faecalis/genética , Lipoproteínas/metabolismo , Lipoproteínas/genética , Macrófagos/microbiología , Macrófagos/inmunología , Macrófagos/metabolismo , Polisacáridos Bacterianos/metabolismo , Polisacáridos Bacterianos/inmunología , Humanos , Antígenos Bacterianos/metabolismo , Antígenos Bacterianos/inmunología , Antígenos Bacterianos/genética , Inmunidad Innata , Virulencia , Animales , Ratones
12.
Gut Microbes ; 16(1): 2382766, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39068523

RESUMEN

CagA, a virulence factor of Helicobacter pylori (H. pylori), is known to drive inflammation in gastric epithelial cells and is typically degraded through autophagy. However, the molecular mechanism by which CagA evades autophagy-mediated degradation remains elusive. This study found that H. pylori inhibits autophagic flux by upregulating the expression of AU-rich element RNA-binding factor 1 (AUF1). We confirmed that AUF1 does not affect autophagy initiation but instead hampers lysosomal clearance, as evidenced by treatments with 3-MA, CQ and BafA1. Upregulated AUF1 stabilizes CagA protein levels by inhibiting the autolysosomal degradation of intracellular CagA in H. pylori-infected gastric epithelial cells. Knocking down AUF1 promotes CagA degradation, an effect that can be reversed by the lysosome inhibitor BafA1 and CQ. Transcriptome analysis of AUF1-knockdown gastric epithelial cells infected with H. pylori indicated that AUF1 regulates the expression of lysosomal-associated hydrolase genes, specifically CTSD, to inhibit autolysosomal degradation. Moreover, we observed that knockdown of AUF1 enhanced the stability of CTSD mRNA and identified AUF1 binding to the 3'UTR region of CTSD mRNA. AUF1-mediated downregulation of CTSD expression contributes to CagA stability, and AUF1 overexpression leads to an increase in CagA levels in exosomes, thus promoting extracellular inflammation. In clinical gastric mucosa, the expression of AUF1 and its cytoplasmic translocation are associated with H. pylori-associated gastritis, with CagA being necessary for the translocation of AUF1 into the cytoplasm. Our findings suggest that AUF1 is a novel host-positive regulator of CagA, and dysregulation of AUF1 expression increases the risk of H. pylori-associated gastritis.


Asunto(s)
Antígenos Bacterianos , Autofagia , Proteínas Bacterianas , Células Epiteliales , Mucosa Gástrica , Infecciones por Helicobacter , Helicobacter pylori , Ribonucleoproteína Nuclear Heterogénea D0 , Ribonucleoproteína Heterogénea-Nuclear Grupo D , Lisosomas , Antígenos Bacterianos/metabolismo , Antígenos Bacterianos/genética , Ribonucleoproteína Nuclear Heterogénea D0/metabolismo , Helicobacter pylori/metabolismo , Helicobacter pylori/genética , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Humanos , Lisosomas/metabolismo , Lisosomas/microbiología , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/metabolismo , Infecciones por Helicobacter/patología , Ribonucleoproteína Heterogénea-Nuclear Grupo D/metabolismo , Ribonucleoproteína Heterogénea-Nuclear Grupo D/genética , Células Epiteliales/microbiología , Células Epiteliales/metabolismo , Mucosa Gástrica/microbiología , Mucosa Gástrica/metabolismo , Inflamación/metabolismo , Inflamación/microbiología , Factores de Virulencia/metabolismo , Factores de Virulencia/genética , Línea Celular
13.
J Biol Chem ; 300(9): 107613, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39079629

RESUMEN

Shigella spp. are highly pathogenic members of the Enterobacteriaceae family, causing ∼269 million cases of bacillary dysentery and >200,000 deaths each year. Like many Gram-negative pathogens, Shigella rely on their type three secretion system (T3SS) to inject effector proteins into eukaryotic host cells, driving both cellular invasion and evasion of host immune responses. Exposure to the bile salt deoxycholate (DOC) significantly enhances Shigella virulence and is proposed to serve as a critical environmental signal present in the small intestine that prepares Shigella's T3SS for efficient infection of the colonic epithelium. Here, we uncover critical mechanistic details of the Shigella-specific DOC signaling process by describing the role of a π-helix secondary structure element within the T3SS tip protein invasion plasmid antigen D (IpaD). Biophysical characterization and high-resolution structures of IpaD mutants lacking the π-helix show that it is not required for global protein structure, but that it defines the native DOC binding site and prevents off target interactions. Additionally, Shigella strains expressing the π-helix deletion mutants illustrate the pathogenic importance of its role in guiding DOC interaction as flow cytometry and gentamycin protection assays show that the IpaD π-helix is essential for DOC-mediated apparatus maturation and enhanced invasion of eukaryotic cells. Together, these findings add to our understanding of the complex Shigella pathogenesis pathway and its evolution to respond to environmental bile salts by identifying the π-helix in IpaD as a critical structural element required for translating DOC exposure to virulence enhancement.


Asunto(s)
Antígenos Bacterianos , Ácido Desoxicólico , Shigella flexneri , Virulencia , Ácido Desoxicólico/química , Ácido Desoxicólico/metabolismo , Antígenos Bacterianos/metabolismo , Antígenos Bacterianos/química , Antígenos Bacterianos/genética , Shigella flexneri/metabolismo , Shigella flexneri/genética , Shigella flexneri/patogenicidad , Sistemas de Secreción Tipo III/metabolismo , Sistemas de Secreción Tipo III/genética , Humanos , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Estructura Secundaria de Proteína
14.
Front Cell Infect Microbiol ; 14: 1419568, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38983115

RESUMEN

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.


Asunto(s)
Antígenos Bacterianos , Proteínas Bacterianas , Infecciones por Helicobacter , Helicobacter pylori , Nanopartículas del Metal , Transducción de Señal , Plata , Helicobacter pylori/efectos de los fármacos , Proteínas Bacterianas/metabolismo , Antígenos Bacterianos/metabolismo , Plata/farmacología , Plata/metabolismo , Humanos , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Interacciones Huésped-Patógeno , Células Epiteliales/microbiología , Factores de Virulencia/metabolismo , Línea Celular , Antibacterianos/farmacología , Línea Celular Tumoral
15.
Int J Mol Sci ; 25(13)2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-39000189

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas , Cadherinas , Células Epiteliales , Mucosa Gástrica , Helicobacter pylori , Organoides , Humanos , Cadherinas/metabolismo , Organoides/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Mucosa Gástrica/metabolismo , Mucosa Gástrica/microbiología , Mucosa Gástrica/patología , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Antígenos Bacterianos/metabolismo , Infecciones por Helicobacter/metabolismo , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/patología , Antígenos CD/metabolismo , Estómago/microbiología , Estómago/patología , Línea Celular , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Neoplasias Gástricas/microbiología , Serina Proteasas
16.
World J Microbiol Biotechnol ; 40(9): 273, 2024 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-39030443

RESUMEN

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.


Asunto(s)
Gerbillinae , Infecciones por Helicobacter , Helicobacter pylori , Uniones Intercelulares , Páncreas , Animales , Helicobacter pylori/patogenicidad , Helicobacter pylori/genética , Infecciones por Helicobacter/microbiología , Páncreas/microbiología , Páncreas/patología , Uniones Intercelulares/microbiología , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Antígenos Bacterianos/metabolismo , Antígenos Bacterianos/genética , Factores de Virulencia/metabolismo , Factores de Virulencia/genética , Estómago/microbiología , Estómago/patología , Modelos Animales de Enfermedad , Masculino , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas de la Membrana Bacteriana Externa/genética
17.
J Biol Chem ; 300(7): 107478, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38879009

RESUMEN

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.


Asunto(s)
Antígenos Bacterianos , Proteína de Unión al Complemento C4b , Streptococcus pyogenes , Streptococcus pyogenes/metabolismo , Streptococcus pyogenes/genética , Streptococcus pyogenes/química , Proteína de Unión al Complemento C4b/metabolismo , Antígenos Bacterianos/metabolismo , Antígenos Bacterianos/química , Antígenos Bacterianos/genética , Humanos , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas Portadoras/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/química , Unión Proteica , Secuencia de Aminoácidos , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética
18.
EBioMedicine ; 105: 105196, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38880068

RESUMEN

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.


Asunto(s)
Antígenos Bacterianos , Mycobacterium tuberculosis , ARN Mensajero , Humanos , Mycobacterium tuberculosis/genética , Antígenos Bacterianos/genética , Antígenos Bacterianos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Hibridación in Situ , Tuberculosis/microbiología , ARN Bacteriano/genética , Inmunohistoquímica , Granuloma/microbiología , Granuloma/metabolismo , Pulmón/microbiología , Pulmón/patología , Pulmón/metabolismo
19.
Infect Immun ; 92(7): e0015224, 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-38888310

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas , Macrófagos , Infecciones Estreptocócicas , Streptococcus pyogenes , Estreptolisinas , Factores de Virulencia , Streptococcus pyogenes/genética , Streptococcus pyogenes/patogenicidad , Streptococcus pyogenes/inmunología , Infecciones Estreptocócicas/microbiología , Infecciones Estreptocócicas/inmunología , Infecciones Estreptocócicas/mortalidad , Humanos , Macrófagos/microbiología , Macrófagos/inmunología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Estreptolisinas/genética , Estreptolisinas/metabolismo , Factores de Virulencia/genética , Mutación , Interacciones Huésped-Patógeno/inmunología , Virulencia/genética , Animales , Antígenos Bacterianos/genética , Antígenos Bacterianos/metabolismo , Antígenos Bacterianos/inmunología , Viabilidad Microbiana , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo , Ratones , Regulación Bacteriana de la Expresión Génica , Proteínas Portadoras
20.
Microb Pathog ; 193: 106757, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38908454

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas , Muerte Celular , Interacciones Huésped-Patógeno , Mitocondrias , Mycobacterium tuberculosis , Especies Reactivas de Oxígeno , Humanos , Antígenos Bacterianos/metabolismo , Antígenos Bacterianos/genética , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Mitocondrias/metabolismo , Mycobacterium tuberculosis/patogenicidad , Mycobacterium tuberculosis/metabolismo , Mycobacterium tuberculosis/genética , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Tuberculosis/microbiología , Tuberculosis/metabolismo , Factores de Virulencia/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA