Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.422
Filtrar
1.
Cell Rep ; 43(4): 114051, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38564334

RESUMO

Uropathogenic Escherichia coli (UPEC) is the most common causative agent of urinary tract infection (UTI). UPEC invades bladder epithelial cells (BECs) via fusiform vesicles, escapes into the cytosol, and establishes biofilm-like intracellular bacterial communities (IBCs). Nucleoside-diphosphate kinase (NDK) is secreted by pathogenic bacteria to enhance virulence. However, whether NDK is involved in UPEC pathogenesis remains unclear. Here, we find that the lack of ndk impairs the colonization of UPEC CFT073 in mouse bladders and kidneys owing to the impaired ability of UPEC to form IBCs. Furthermore, we demonstrate that NDK inhibits caspase-1-dependent pyroptosis by consuming extracellular ATP, preventing superficial BEC exfoliation, and promoting IBC formation. UPEC utilizes the reactive oxygen species (ROS) sensor OxyR to indirectly activate the regulator integration host factor, which then directly activates ndk expression in response to intracellular ROS. Here, we reveal a signaling transduction pathway that UPEC employs to inhibit superficial BEC exfoliation, thus facilitating acute UTI.


Assuntos
Caspase 1 , Infecções por Escherichia coli , Núcleosídeo-Difosfato Quinase , Piroptose , Infecções Urinárias , Escherichia coli Uropatogênica , Escherichia coli Uropatogênica/patogenicidade , Animais , Infecções Urinárias/microbiologia , Infecções Urinárias/patologia , Camundongos , Caspase 1/metabolismo , Núcleosídeo-Difosfato Quinase/metabolismo , Núcleosídeo-Difosfato Quinase/genética , Infecções por Escherichia coli/microbiologia , Infecções por Escherichia coli/metabolismo , Infecções por Escherichia coli/patologia , Espécies Reativas de Oxigênio/metabolismo , Camundongos Endogâmicos C57BL , Humanos , Feminino , Bexiga Urinária/microbiologia , Bexiga Urinária/patologia , Células Epiteliais/microbiologia , Células Epiteliais/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Transdução de Sinais
2.
Front Cell Infect Microbiol ; 14: 1268243, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38606299

RESUMO

Introduction: Crohn's disease (CD) is a chronic inflammatory bowel disease, of which the etiology involves genetic, environmental and microbial factors. Adherent-invasive Escherichia coli (AIEC) and polymorphisms in autophagy-related genes have been implicated in CD etiology. Autophagy is a key process for the maintenance of cellular homeostasis, which allows the degradation of damaged cytoplasmic components and pathogens via lysosome. We have shown that a functional autophagy is necessary for AIEC clearance. Here, we aimed at identifying the autophagy receptor(s) responsible to target AIEC to autophagy for degradation. Methods: The levels of autophagy receptors p62, NDP52, NBR1, TAX1BP1 and Optineurin were knocked down in human intestinal epithelial cells T84 using siRNAs. The NDP52 knock-out (KO) and p62 KO HeLa cells, as well as NDP52 KO HeLa cells expressing the wild-type NDP52 or the mutated NDP52Val248Ala protein were used. Results and discussion: We showed that, among the tested autophagy receptors (p62, NDP52, NBR1, TAX1BP1 and Optineurin), diminished expression of p62 or NDP52 increased the number of the clinical AIEC LF82 strain inside epithelial cells. This was associated with increased pro-inflammatory cytokine production. Moreover, p62 or NDP52 directly colocalized with AIEC LF82 and LC3, an autophagy marker. As the NDP52Val248Ala polymorphism has been associated with increased CD susceptibility, we investigated its impact on AIEC control. However, in HeLa cell and under our experimental condition, no effect of this polymorphism neither on AIEC LF82 intracellular number nor on pro-inflammatory cytokine production was observed. Together, our results suggest that p62 and NDP52 act as autophagy receptors for AIEC recognition, controlling AIEC intracellular replication and inflammation.


Assuntos
Doença de Crohn , Infecções por Escherichia coli , Humanos , Células HeLa , Mucosa Intestinal/metabolismo , Infecções por Escherichia coli/metabolismo , Proteínas de Transporte/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Autofagia/fisiologia , Citocinas/metabolismo , Aderência Bacteriana
3.
Cell Rep ; 43(4): 114007, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38517889

RESUMO

Urinary tract infections (UTIs) commonly afflict people with diabetes. To better understand the mechanisms that predispose diabetics to UTIs, we employ diabetic mouse models and altered insulin signaling to show that insulin receptor (IR) shapes UTI defenses. Our findings are validated in human biosamples. We report that diabetic mice have suppressed IR expression and are more susceptible to UTIs caused by uropathogenic Escherichia coli (UPEC). Systemic IR inhibition increases UPEC susceptibility, while IR activation reduces UTIs. Localized IR deletion in bladder urothelium promotes UTI by increasing barrier permeability and suppressing antimicrobial peptides. Mechanistically, IR deletion reduces nuclear factor κB (NF-κB)-dependent programming that co-regulates urothelial tight junction integrity and antimicrobial peptides. Exfoliated urothelial cells or urine samples from diabetic youths show suppressed expression of IR, barrier genes, and antimicrobial peptides. These observations demonstrate that urothelial insulin signaling has a role in UTI prevention and link IR to urothelial barrier maintenance and antimicrobial peptide expression.


Assuntos
Receptor de Insulina , Transdução de Sinais , Bexiga Urinária , Infecções Urinárias , Urotélio , Receptor de Insulina/metabolismo , Infecções Urinárias/microbiologia , Infecções Urinárias/metabolismo , Infecções Urinárias/patologia , Animais , Urotélio/metabolismo , Urotélio/patologia , Urotélio/microbiologia , Humanos , Bexiga Urinária/microbiologia , Bexiga Urinária/patologia , Bexiga Urinária/metabolismo , Camundongos , Escherichia coli Uropatogênica/patogenicidade , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Feminino , Infecções por Escherichia coli/metabolismo , Infecções por Escherichia coli/microbiologia , Insulina/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Masculino
4.
Cell Rep ; 43(4): 114004, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38522070

RESUMO

During infections, host cells are exposed to pathogen-associated molecular patterns (PAMPs) and virulence factors that stimulate multiple signaling pathways that interact additively, synergistically, or antagonistically. The net effect of such higher-order interactions is a vital determinant of the outcome of host-pathogen interactions. Here, we demonstrate one such complex interplay between bacterial exotoxin- and PAMP-induced innate immune pathways. We show that two caspases activated during enterohemorrhagic Escherichia coli (EHEC) infection by lipopolysaccharide (LPS) and Shiga toxin (Stx) interact in a functionally antagonistic manner; cytosolic LPS-activated caspase-11 cleaves full-length gasdermin D (GSDMD), generating an active pore-forming N-terminal fragment (NT-GSDMD); subsequently, caspase-3 activated by EHEC Stx cleaves the caspase-11-generated NT-GSDMD to render it nonfunctional, thereby inhibiting pyroptosis and interleukin-1ß maturation. Bacteria typically subvert inflammasomes by targeting upstream components such as NLR sensors or full-length GSDMD but not active NT-GSDMD. Thus, our findings uncover a distinct immune evasion strategy where a bacterial toxin disables active NT-GSDMD by co-opting caspase-3.


Assuntos
Caspase 3 , Gasderminas , Peptídeos e Proteínas de Sinalização Intracelular , Macrófagos , Proteínas de Ligação a Fosfato , Piroptose , Piroptose/efeitos dos fármacos , Proteínas de Ligação a Fosfato/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Caspase 3/metabolismo , Humanos , Animais , Camundongos , Proteínas Reguladoras de Apoptose/metabolismo , Toxinas Bacterianas/metabolismo , Caspases/metabolismo , Lipopolissacarídeos/farmacologia , Escherichia coli Êntero-Hemorrágica/metabolismo , Escherichia coli Êntero-Hemorrágica/patogenicidade , Caspases Iniciadoras/metabolismo , Inflamassomos/metabolismo , Camundongos Endogâmicos C57BL , Infecções por Escherichia coli/metabolismo , Infecções por Escherichia coli/microbiologia , Infecções por Escherichia coli/imunologia , Interleucina-1beta/metabolismo
5.
Food Funct ; 15(7): 3583-3599, 2024 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-38469921

RESUMO

Lactobacillus probiotics exert their effects in a strain-specific and metabolite-specific manner. This study aims to identify lactobacilli that can effectively enhance the intestinal barrier function both in vitro and in vivo and to investigate the underlying metabolite and molecular mechanisms involved. Nine Lactobacillus isolates were evaluated for their ability to enhance the IPEC-J2 cellular barrier function and for their anti-inflammatory and anti-apoptotic effects in IPEC-J2 cells after an enterotoxigenic Escherichia coli challenge. Of the nine isolates, L. plantarum T10 demonstrated significant advantages in enhancing the cellular barrier function and displayed anti-inflammatory and anti-apoptotic activities in vitro. The bioactivities of L. plantarum T10 were primarily attributed to the production of exopolysaccharides, which exerted their effects through the TLR-mediated p38 MAPK pathway in ETEC-challenged IPEC-J2 cells. Furthermore, the production of EPS by L. plantarum T10 led to the alleviation of dextran sulfate sodium-induced colitis by reducing intestinal damage and enhancing the intestinal barrier function in mice. The EPS is classified as a heteropolysaccharide with an average molecular weight of 23.0 kDa. It is primarily composed of mannose, glucose, and ribose. These findings have practical implications for the targeted screening of lactobacilli used in the production of probiotics and postbiotics with strain-specific features of exopolysaccharides.


Assuntos
Infecções por Escherichia coli , Lactobacillus plantarum , Probióticos , Animais , Camundongos , Mucosa Intestinal/metabolismo , 60435 , Infecções por Escherichia coli/metabolismo , Lactobacillus , Anti-Inflamatórios/metabolismo
6.
Cell ; 187(8): 1874-1888.e14, 2024 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-38518773

RESUMO

Infections of the lung cause observable sickness thought to be secondary to inflammation. Signs of sickness are crucial to alert others via behavioral-immune responses to limit contact with contagious individuals. Gram-negative bacteria produce exopolysaccharide (EPS) that provides microbial protection; however, the impact of EPS on sickness remains uncertain. Using genome-engineered Pseudomonas aeruginosa (P. aeruginosa) strains, we compared EPS-producers versus non-producers and a virulent Escherichia coli (E. coli) lung infection model in male and female mice. EPS-negative P. aeruginosa and virulent E. coli infection caused severe sickness, behavioral alterations, inflammation, and hypothermia mediated by TLR4 detection of the exposed lipopolysaccharide (LPS) in lung TRPV1+ sensory neurons. However, inflammation did not account for sickness. Stimulation of lung nociceptors induced acute stress responses in the paraventricular hypothalamic nuclei by activating corticotropin-releasing hormone neurons responsible for sickness behavior and hypothermia. Thus, EPS-producing biofilm pathogens evade initiating a lung-brain sensory neuronal response that results in sickness.


Assuntos
Infecções por Escherichia coli , Escherichia coli , Pulmão , Polissacarídeos Bacterianos , Infecções por Pseudomonas , Pseudomonas aeruginosa , Animais , Feminino , Masculino , Camundongos , Biofilmes , Escherichia coli/fisiologia , Hipotermia/metabolismo , Hipotermia/patologia , Inflamação/metabolismo , Inflamação/patologia , Pulmão/microbiologia , Pulmão/patologia , Pneumonia/microbiologia , Pneumonia/patologia , Pseudomonas aeruginosa/fisiologia , Células Receptoras Sensoriais , Polissacarídeos Bacterianos/metabolismo , Infecções por Escherichia coli/metabolismo , Infecções por Escherichia coli/microbiologia , Infecções por Escherichia coli/patologia , Infecções por Pseudomonas/metabolismo , Infecções por Pseudomonas/microbiologia , Infecções por Pseudomonas/patologia , Nociceptores/metabolismo
7.
Pharmacol Res ; 201: 107096, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38320736

RESUMO

The uncontrolled bacterial infection-induced cytokine storm and sequential immunosuppression are commonly observed in septic patients, which indicates that the activation of phagocytic cells and the efficient and timely elimination of bacteria are crucial for combating bacterial infections. However, the role of dysregulated immune cells and their disrupted function in sepsis remains unclear. Here, we found that macrophages exhibited the impaired endocytosis capabilities in sepsis by Single-cell RNA sequencing and bulk RNA sequencing. Caveolae protein Caveolin-1 (Cav-1) of macrophages was inactivated by SHP2 rapidly during Escherichia coli (E.coli) infection. Allosteric inhibitor of SHP2 effectively maintains Cav-1 phosphorylation to enhance macrophage to endocytose and eliminate bacteria. Additionally, TLR4 endocytosis of macrophage was also enhanced upon E.coli infection by SHP099, inducing an increased and rapidly resolved inflammatory response. In vivo, pretreatment or posttreatment with inhibitor of SHP2 significantly reduced the bacterial burden in organs and mortality of mice subjected E.coli infection or CLP-induced sepsis. The cotreatment of inhibitor of SHP2 with an antibiotic conferred complete protection against mortality in mice. Our findings suggest that Cav-1-mediated endocytosis and bacterial elimination may play a critical role in the pathogenesis of sepsis, highlighting inhibitor of SHP2 as a potential therapeutic agent for sepsis.


Assuntos
Cavéolas , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Sepse , Animais , Humanos , Camundongos , Bactérias , Cavéolas/metabolismo , Endocitose , Infecções por Escherichia coli/tratamento farmacológico , Infecções por Escherichia coli/metabolismo , Macrófagos , Proteína Tirosina Fosfatase não Receptora Tipo 11/antagonistas & inibidores , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Sepse/tratamento farmacológico , Sepse/metabolismo
8.
Theriogenology ; 219: 75-85, 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38402700

RESUMO

Endometritis is a disease caused by a postpartum bacterial infection with a poor prognosis that primarily affects dairy cows. Three-dimensional organoids have been used as a model for endometritis, because they exhibit a structure comparable to that of the endometrium, demonstrating both expansibility and hormone responsiveness. These characteristics render them an ideal platform for in vitro investigations of endometrial diseases. Estradiol (E2) is an endogenous steroid hormone with demonstrated anti-inflammatory properties, and the objective of this study was to determine the mechanism by which E2 modulates the inflammatory response and the Wnt signal transduction pathway in bovine endometrial epithelial cells and organoids following E. coli infection. We present the techniques for isolating and culturing primary bovine endometrial epithelial cells (BEECs), and producing endometrial organoids. For the experiments, the endometrial epithelial cells and organoids were infected with E. coli for 1 h, followed by incubation with E2 for 12 h. The mRNA and protein expressions of the inflammation-related genes, IL-1ß, IL-6, TLR4, and NF-κB, as well as the Wnt pathway-related genes, Wnt4, ß-catenin, c-Myc, and CyclinD1, were assessed using real-time quantitative-PCR and western blotting, respectively. The CCK8 viable cell counting assay was utilized to determine the optimal concentration of the Wnt inhibitor, IWR-1. The mRNA and protein expression of Wnt pathway-related genes was assessed following IWR-1 treatment, while the expression levels of proliferation-associated genes (Ki67, PCNA) and barrier repair genes (occludin, claudin, and Zo-1) in BEECs and organoids were evaluated after E2 treatment. The results of this study show that mRNA expression of the inflammatory genes, IL-1ß, TLR4, and NF-κB (P < 0.05) decreased in BEECs following E2 treatment compared to the E. coli group. The protein expression of the IL-1ß, IL-6, TLR4 and NF-κB genes was also inhibited (P < 0.05). Similar results were observed in tests on the organoids. Our findings demonstrate that E2 significantly upregulates the expression of Wnt-related genes, including ß-catenin and c-Myc, while concurrently downregulating the expression of GSK3ß (P < 0.05). Next, we treated E. coli-infected BEECs and organoids with the Wnt inhibitor, IWR-1. Compared with E. coli and E. coli + E2, the expression of mRNA and protein from Wnt 4, ß-catenin, and CyclinD1 in E. coli + E2 and E. coli + IWR-1 was down-regulated (P < 0.05). The expression of the proliferation genes, Ki67, PCNA, and the tight junction genes, occludin, claudin1, and Zo-1, in organoids was significantly higher than that in BEECs (P < 0.05). In summary, we found strong potential for E2 mitigation of the E. coli-induced inflammatory response in BEECs and organoids, through activation of the Wnt pathway. In addition, the proliferation and repair capacity of organoids was much higher than that of BEECs.


Assuntos
Doenças dos Bovinos , Endometrite , Infecções por Escherichia coli , Feminino , Bovinos , Animais , Endometrite/veterinária , NF-kappa B/metabolismo , Via de Sinalização Wnt , Interleucina-6/metabolismo , Escherichia coli/metabolismo , Estradiol/farmacologia , Estradiol/metabolismo , Receptor 4 Toll-Like/metabolismo , beta Catenina , Antígeno Ki-67/metabolismo , Ocludina/metabolismo , Ocludina/farmacologia , Antígeno Nuclear de Célula em Proliferação/metabolismo , Endométrio/metabolismo , Células Epiteliais/metabolismo , Infecções por Escherichia coli/veterinária , Infecções por Escherichia coli/metabolismo , RNA Mensageiro/metabolismo , Doenças dos Bovinos/metabolismo
9.
Int J Biol Macromol ; 262(Pt 2): 130101, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38346619

RESUMO

Post-weaning diarrhea caused by enterotoxigenic E. coli F18 introduces enormous losses to the porcine industry. N6-methyladenosine (m6A) is a ubiquitous epitranscriptomic biomarker that modulates host cell resistance to pathogen infection, however, its significance in E. coli F18-treated IPEC-J2 cells remains unexplored. Herein, we revealed that m6A and associated modulators strongly controlled E. coli F18 susceptibility. The data indicated an enhancement of METTL3 contents in E. coli F18-treated IPEC-J2 cells. METTL3 is known to be a major modulator of E. coli F18 adhesion within IPEC-J2 cells. As expected, METTL3 deficiency was observed to reduce m6A content at the IKBKG 5'-UTR, leading to critical suppression of YTHDF1-dependent IKBKG translation. Therefore, the activation of the NF-κB axis was observed, which enhanced IPEC-J2 resistance to E. coli F18 infection. Taken together, these findings uncover a potential mechanism underlying the m6A-mediated control of E. coli F18 susceptibility. This information may contribute to the establishment of new approaches for combating bacteria-induced diarrhea in piglets.


Assuntos
Escherichia coli Enterotoxigênica , Infecções por Escherichia coli , Animais , Suínos , NF-kappa B/metabolismo , Infecções por Escherichia coli/metabolismo , Transdução de Sinais , Diarreia , Células Epiteliais/metabolismo
10.
Front Cell Infect Microbiol ; 14: 1337439, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38390621

RESUMO

Introduction: The etiology of Escherichia coli in yaks, along with its drug resistance, results in economic losses within the yak breeding industry. The utilization of lactic acid bacteria treatment has emerged as a viable alternative to antibiotics in managing colibacillosis. Methods: To elucidate the therapeutic mechanisms of Lactobacillus against Escherichia coli-induced intestinal barrier damage in yaks, we employed yak epithelial cells as the experimental model and established a monolayer epithelial barrier using Transwell. The study encompassed four groups: a control group, a model group (exposed to E. coli O78), a low-dose Lactobacillus group (E. coli O78 + 1 × 105CFU LAB), and a high-dose Lactobacillus group (E. coli O78 + 1 × 107CFU LAB). Various techniques, including transmembrane resistance measurement, CFU counting, RT-qPCR, and Western Blot, were employed to assess indicators related to cell barrier permeability and tight junction integrity. Results: In the Model group, Escherichia coli O78 significantly compromised the permeability and tight junction integrity of the yak epithelial barrier. It resulted in decreased transmembrane resistance, elevated FD4 flux, and bacterial translocation. Furthermore, it downregulated the mRNA and protein expression of MUC2, Occludin, and ZO-1, while upregulating the mRNA expression and protein expression of FABP2 and Zonulin, thereby impairing intestinal barrier function. Contrastingly, Lactobacillus exhibited a remarkable protective effect. It substantially increased transmembrane resistance, mitigated FD4 flux, and reduced bacterial translocation. Moreover, it significantly upregulated the mRNA and protein expression of MUC2, Occludin, and ZO-1, while downregulating the mRNA and protein expression of FABP2 and Zonulin. Notably, high-dose LAB demonstrated superior regulatory effects compared to the low-dose LAB group. Discussion: In conclusion, our findings suggest that Lactobacillus holds promise in treating yak colibacillosis by enhancing mucin and tight junction protein expression. Furthermore, we propose that Lactobacillus achieves these effects through the regulation of Zonulin.


Assuntos
Infecções por Escherichia coli , Lactobacillus , Animais , Bovinos , Lactobacillus/genética , Mucosa Intestinal/microbiologia , Escherichia coli/genética , Ocludina/metabolismo , Infecções por Escherichia coli/metabolismo , RNA Mensageiro/metabolismo , Junções Íntimas/metabolismo
11.
Protein Expr Purif ; 215: 106411, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38056514

RESUMO

Pathogenic strains of Escherichia coli F17+ are associated with various intestinal and extra-intestinal pathologies, including diarrhea, and result in significant animal mortality. These infections rely on the expression of virulence factors, such as F17 fimbriae, for adhesion. F17 fimbriae form a protective layer on the surface of E. coli bacteria, consisting of a major structural subunit, F17A, and a minor functional subunit, F17G. Because of the evolution of bacterial resistance, conventional antibiotic treatments have limited efficacy. Therefore, there is a pressing need to develop novel therapeutic tools. In this study, we cloned and produced the F17G protein. We then immunized a camel with the purified F17G protein and constructed a VHH library consisting of 2 × 109 clones. The library was then screened against F17G protein using phage display technology. Through this process, we identified an anti-F17G nanobody that was subsequently linked, via a linker, to an anti-F17A nanobody, resulting in the creation of an effective bispecific nanobody. Comprehensive characterization of this bispecific nanobody demonstrated excellent production, specific binding capacity to both recombinant forms of the two F17 antigens and the E. coli F17+ strain, remarkable stability in camel serum, and superior resistance to pepsin protease. The successful generation of this bispecific nanobody with excellent production, specific binding capacity and stability highlights its potential as a valuable tool for fighting infections caused by pathogenic E. coli F17+ strain.


Assuntos
Infecções por Escherichia coli , Escherichia coli , Animais , Escherichia coli/genética , Escherichia coli/química , Infecções por Escherichia coli/metabolismo , Infecções por Escherichia coli/microbiologia , Camelus , Fímbrias Bacterianas/química , Fímbrias Bacterianas/metabolismo , Diarreia/metabolismo , Diarreia/microbiologia
12.
PLoS Pathog ; 19(12): e1011877, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38127952

RESUMO

Shiga-toxin producing Escherichia coli (STEC) infections can cause from bloody diarrhea to Hemolytic Uremic Syndrome. The STEC intestinal infection triggers an inflammatory response that can facilitate the development of a systemic disease. We report here that neutrophils might contribute to this inflammatory response by secreting Interleukin 1 beta (IL-1ß). STEC stimulated neutrophils to release elevated levels of IL-1ß through a mechanism that involved the activation of caspase-1 driven by the NLRP3-inflammasome and neutrophil serine proteases (NSPs). Noteworthy, IL-1ß secretion was higher at lower multiplicities of infection. This secretory profile modulated by the bacteria:neutrophil ratio, was the consequence of a regulatory mechanism that reduced IL-1ß secretion the higher were the levels of activation of both caspase-1 and NSPs, and the production of NADPH oxidase-dependent reactive oxygen species. Finally, we also found that inhibition of NSPs significantly reduced STEC-triggered IL-1ß secretion without modulating the ability of neutrophils to kill the bacteria, suggesting NSPs might represent pharmacological targets to be evaluated to limit the STEC-induced intestinal inflammation.


Assuntos
Infecções por Escherichia coli , Escherichia coli O157 , Síndrome Hemolítico-Urêmica , Interleucina-1beta , Escherichia coli Shiga Toxigênica , Humanos , Caspases , Infecções por Escherichia coli/metabolismo , Infecções por Escherichia coli/microbiologia , Síndrome Hemolítico-Urêmica/metabolismo , Síndrome Hemolítico-Urêmica/microbiologia , Neutrófilos , Interleucina-1beta/metabolismo
13.
Microb Pathog ; 185: 106435, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37931825

RESUMO

Bacterial infections result in intestinal inflammation and injury, which affects gut health and nutrient absorption. Lipocalin 2 (Lcn2) is a protein that reacts to microbial invasion, inflammatory responses, and tissue damage. However, it remains unclear whether Lcn2 has a protective effect against bacterial induced intestinal inflammation. Therefore, this study endeavors to investigate the involvement of Lcn2 in the intestinal inflammation of mice infected with Enterohemorrhagic Escherichia coli O157:H7 (E. coli O157:H7). Lcn2 knockout (Lcn2-/-) mice were used to evaluate the changes of inflammatory responses. Lcn2 deficiency significantly exacerbated clinical symptoms of E. coli O157:H7 infection by reducing body weight and encouraging bacterial colonization of. Compared to infected wild type mice, infected Lcn2-/- mice had significantly elevated levels of pro-inflammatory cytokines in serum and ileum, including interleukin (IL)-6, IL-1ß, and tumor necrosis factor-α (TNF-α), as well as severe villi destruction in the jejunum. Furthermore, Lcn2 deficiency aggravated intestinal barrier degradation by significantly reducing the expression of tight junction proteins occludin and claudin 1, the content of myeloperoxidase (MPO) in the ileum, and the number of goblet cells in the colon. Our findings indicated that Lcn2 could alleviate inflammatory damage caused by E. coli O157:H7 infection in mice by enhancing intestinal barrier function.


Assuntos
Infecções por Escherichia coli , Escherichia coli O157 , Lipocalina-2 , Animais , Camundongos , Colo/metabolismo , Colo/microbiologia , Colo/patologia , Infecções por Escherichia coli/metabolismo , Infecções por Escherichia coli/patologia , Escherichia coli O157/genética , Escherichia coli O157/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Inflamação/metabolismo , Interleucina-6/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Lipocalina-2/genética , Lipocalina-2/metabolismo
14.
Gut Microbes ; 15(2): 2265578, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37800577

RESUMO

Polyunsaturated fatty acids (PUFAs) have been shown to exacerbate Crohn's disease (CD) by promoting lipid peroxidation (LPO) of intestinal epithelial cells (IECs). Dysbiosis of the gut microbiota may play a crucial role in this process. CD patients often exhibit an increased abundance of Escherichia coli (E. coli) in the gut, and the colonization of adherent-invasive E. coli (AIEC) is implicated in the initiation of intestinal inflammation in CD. However, the impact of AIEC on LPO remains unclear. In this study, we observed that AIEC colonization in the terminal ileum of CD patients was associated with decreased levels of glutathione peroxidase 4 (GPX4) and ferritin heavy chain (FTH) in the intestinal epithelium, along with elevated levels of 4-Hydroxynonenal (4-HNE). In vitro experiments demonstrated that AIEC infection reduced the levels of GPX4 and FTH, increased LPO, and induced ferroptosis in IECs. Furthermore, arachidonic acid (AA) and docosahexaenoic acid (DHA) supplementation in AIEC-infected IECs significantly aggravated LPO and ferroptosis. However, overexpression of GPX4 rescued AIEC-induced LPO and ferroptosis in IECs. Our results further confirmed that AIEC with AA supplementation, associated with excessive LPO and cell death in IECs, worsened colitis in the DSS mouse model and induced enteritis in the antibiotic cocktail pre-treatment mouse model in vivo. Moreover, treatment with ferrostatin-1, a ferroptosis inhibitor, alleviated AIEC with AA supplementation-induced enteritis in mice, accompanied by reduced LPO and cell death in IECs. Our findings suggest that AIEC, in combination with PUFA supplementation, can induce and exacerbate intestinal inflammation, primarily through increased LPO and ferroptosis in IECs.


Assuntos
Doença de Crohn , Enterite , Infecções por Escherichia coli , Microbioma Gastrointestinal , Humanos , Camundongos , Animais , Doença de Crohn/metabolismo , Escherichia coli , Peroxidação de Lipídeos , Infecções por Escherichia coli/metabolismo , Mucosa Intestinal/metabolismo , Ácidos Graxos Insaturados/metabolismo , Inflamação/metabolismo , Aderência Bacteriana
15.
Front Cell Infect Microbiol ; 13: 1255083, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37881369

RESUMO

Background: Adherent-invasive E. coli (AIEC) LF82 is capable of adhering to and invading intestinal epithelial cells, as well as replicating within macrophages without inducing host cell death. Methods: We compared the transcriptomics of LF82 at pH=7.5 and pH=5.8 by RNA-sequencing, and qRT-PCR verified differentially expressed genes (DEGs). The deletion mutants of DEGs in the treatment group (pH=5.8) compared to the control group (pH=7.5) were constructed by λ recombinant. The replication differences between the mutants and WT infected Raw 264.7 at 24 h.p.i were analyzed by combining LB solid plate count and confocal observation. NH4Cl and chloroquine diphosphate (CQ) were used for acid neutralization to study the effect of pH on the replication of LF82 in macrophages. Na2NO3 was added to RPMI 1640 to study the effect of nitrate on the replication of LF82 in macrophages. 0.3% solid LB was used for flagellar motility assay and Hela was used to study flagellar gene deletion mutants and WT adhesion and invasion ability. Results: In this study, we found that infection with LF82 results in acidification of macrophages. Subsequent experiments demonstrated that an intracellular acidic environment is necessary for LF82 replication. Transcriptome and phenotypic analysis showed that high expression of acid shock genes and acid fitness genes promotes LF82 replication in macrophages. Further, we found that the replication of LF82 in macrophages was increased under nitrate treatment, and nitrogen metabolism genes of LF82 were upregulated in acid treatment. The replication in macrophages of ΔnarK, ΔnarXL, ΔnarP, and Δhmp were decreased. In addition, we found that the expression of flagellar genes was downregulated in acidic pH and after LF82 invading macrophages. Motility assay shows that the movement of LF82 on an acidic semisolid agar plate was limited. Further results showed that ΔfliC and ΔfliD decreased in motility, adhesion ability, and invasion of host cells, but no significant effect on replication in macrophages was observed. Conclusion: In this study, we simulated the acidic environment in macrophages, combined with transcriptome technology, and explained from the genetic level that LF82 promotes replication by activating its acid shock and fitness system, enhancing nitrate utilization, and inhibiting flagellar function.


Assuntos
Doença de Crohn , Infecções por Escherichia coli , Humanos , Escherichia coli/genética , Infecções por Escherichia coli/metabolismo , Nitratos/metabolismo , Macrófagos/metabolismo , Aderência Bacteriana/genética , Mucosa Intestinal/metabolismo
16.
Med ; 4(11): 761-777.e8, 2023 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-37863058

RESUMO

BACKGROUND: Shiga toxin (Stx)-producing Escherichia coli hemolytic uremic syndrome (STEC-HUS) is the leading cause of acute kidney injury in children, with an associated mortality of up to 5%. The mechanisms underlying STEC-HUS and why the glomerular microvasculature is so susceptible to injury following systemic Stx infection are unclear. METHODS: Transgenic mice were engineered to express the Stx receptor (Gb3) exclusively in their kidney podocytes (Pod-Gb3) and challenged with systemic Stx. Human glomerular cell models and kidney biopsies from patients with STEC-HUS were also studied. FINDINGS: Stx-challenged Pod-Gb3 mice developed STEC-HUS. This was mediated by a reduction in podocyte vascular endothelial growth factor A (VEGF-A), which led to loss of glomerular endothelial cell (GEnC) glycocalyx, a reduction in GEnC inhibitory complement factor H binding, and local activation of the complement pathway. Early therapeutic inhibition of the terminal complement pathway with a C5 inhibitor rescued this podocyte-driven, Stx-induced HUS phenotype. CONCLUSIONS: This study potentially explains why systemic Stx exposure targets the glomerulus and supports the early use of terminal complement pathway inhibition in this devastating disease. FUNDING: This work was supported by the UK Medical Research Council (MRC) (grant nos. G0901987 and MR/K010492/1) and Kidney Research UK (grant nos. TF_007_20151127, RP42/2012, and SP/FSGS1/2013). The Mary Lyon Center is part of the MRC Harwell Institute and is funded by the MRC (A410).


Assuntos
Infecções por Escherichia coli , Síndrome Hemolítico-Urêmica , Nefropatias , Podócitos , Escherichia coli Shiga Toxigênica , Criança , Humanos , Camundongos , Animais , Podócitos/metabolismo , Podócitos/patologia , Toxina Shiga/genética , Toxina Shiga/metabolismo , Toxina Shiga/uso terapêutico , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/uso terapêutico , Infecções por Escherichia coli/complicações , Infecções por Escherichia coli/tratamento farmacológico , Infecções por Escherichia coli/metabolismo , Síndrome Hemolítico-Urêmica/tratamento farmacológico , Síndrome Hemolítico-Urêmica/metabolismo , Síndrome Hemolítico-Urêmica/patologia , Escherichia coli Shiga Toxigênica/metabolismo , Ativação do Complemento , Nefropatias/patologia
17.
Life Sci ; 331: 122036, 2023 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-37633417

RESUMO

AIMS: We focused on investigating the influence of Escherichia coli (E. coli) on the intestinal barrier. MATERIAL AND METHODS: We studied changes in the distribution and secretory activities of goblet cells and enteroendocrine cells (EECs), as well as changes in the population of mast cells (MCs) in the jejunal and colonic mucosa of germ-free (GF) piglets as a healthy control group and GF piglets whose intestines were colonised with E. coli bacteria on day 5. KEY FINDINGS: The results suggest that the colon of GF piglets is more resistant and less prone to coliform bacterial infection compared to the jejunum. This can be confirmed by a lower degree of histopathological injury index as well as an improvement of the morphometric parameters of the colonic mucosa, together with a significantly increased (p < 0.05) expression of MUC1/EMA, and ZO-3. We also observed a significant decrease in the population of activated MCs (p < 0.001) and EECs (p < 0.001). These findings may indicate a rapid response and better preparation of the intestinal barrier for possible pathological attacks and the subsequent development of mucosal lesions during the development and progression of the intestinal diseases. SIGNIFICANCE: To date, gut-targeted therapeutic approaches that can modulate bacterial translocation and chronic inflammation are still in their infancy but represent one of the most promising areas of research for the development of new effective treatments or clinical strategies in the future. Therefore, a better understanding of these processes can significantly contribute to the development of these targeted strategies for disease prevention and treatment.


Assuntos
Infecções por Escherichia coli , Escherichia coli , Animais , Suínos , Mucosa Intestinal/metabolismo , Infecções por Escherichia coli/veterinária , Infecções por Escherichia coli/metabolismo , Jejuno/patologia , Bactérias
18.
Int J Mol Sci ; 24(13)2023 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-37445610

RESUMO

Meningitis is a major clinical manifestation of Escherichia coli (E. coli) infection characterized by inflammation of the meninges and subarachnoid space. Many chemokines are secreted during meningitic E. coli infection, of which C-X-C motif chemokine 3 (CXCL3) is the most highly expressed. However, it is unclear how CXCL3 plays a role in meningitic E. coli infection. Therefore, this study used in vitro and in vivo assays to clarify these contributions and to identify novel therapeutic targets for central nervous system inflammation. We found a significantly upregulated expression of CXCL3 in human brain microvascular endothelial cells and U251 cells after meningitic E. coli infection, and the CXCL3 receptor, C-X-C motif chemokine receptor 2 (CXCR2), was expressed in microglia. Furthermore, CXCL3 induced M1 microglia by selectively activating mitogen-activated protein kinases signaling and significantly upregulating tumor necrosis factor-α (TNF-α), interleukin (IL)-1ß, IL-6, nitric oxide synthase 2 (NOS2), and cluster of differentiation 86 (CD86) expression levels, promoting an inflammatory response. Our findings clarify the role of CXCL3 in meningitic E. coli-induced neuroinflammation and demonstrate that CXCL3 may be a potential therapeutic target for future investigation and prevention of E. coli-induced neuroinflammation.


Assuntos
Infecções por Escherichia coli , Meningite , Humanos , Escherichia coli/metabolismo , Microglia/metabolismo , Doenças Neuroinflamatórias , Células Endoteliais , Quimiocinas/metabolismo , Inflamação/metabolismo , Infecções por Escherichia coli/metabolismo
19.
Int J Mol Sci ; 24(14)2023 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-37511208

RESUMO

The high-pathogenicity island (HPI) was initially identified in Yersinia and can be horizontally transferred to Escherichia coli to produce yersiniabactin (Ybt), which enhances the pathogenicity of E. coli by competing with the host for Fe3+. Pyroptosis is gasdermin-induced necrotic cell death. It involves the permeabilization of the cell membrane and is accompanied by an inflammatory response. It is still unclear whether Ybt HPI can cause intestinal epithelial cells to undergo pyroptosis and contribute to gut inflammation during E. coli infection. In this study, we infected intestinal epithelial cells of mice with E. coli ZB-1 and the Ybt-deficient strain ZB-1Δirp2. Our findings demonstrate that Ybt-producing E. coli is more toxic and exacerbates gut inflammation during systemic infection. Mechanistically, our results suggest the involvement of the NLRP3/caspase-1/GSDMD pathway in E. coli infection. Ybt promotes the assembly and activation of the NLRP3 inflammasome, leading to GSDMD cleavage into GSDMD-N and promoting the pyroptosis of intestinal epithelial cells, ultimately aggravating gut inflammation. Notably, NLRP3 knockdown alleviated these phenomena, and the binding of free Ybt to NLRP3 may be the trigger. Overall, our results show that Ybt HPI enhances the pathogenicity of E. coli and induces pyroptosis via the NLRP3 pathway, which is a new mechanism through which E. coli promotes gut inflammation. Furthermore, we screened drugs targeting NLRP3 from an existing drug library, providing a list of potential drug candidates for the treatment of gut injury caused by E. coli.


Assuntos
Células Epiteliais , Infecções por Escherichia coli , Escherichia coli , Mucosa Intestinal , Proteína 3 que Contém Domínio de Pirina da Família NLR , Piroptose , Animais , Camundongos , Enterócitos/metabolismo , Enterócitos/microbiologia , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Escherichia coli/metabolismo , Escherichia coli/patogenicidade , Infecções por Escherichia coli/metabolismo , Inflamassomos/metabolismo , Inflamação/metabolismo , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Piroptose/fisiologia
20.
Cells ; 12(10)2023 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-37408237

RESUMO

Macrophages are important effectors of inflammation resolution that contribute to the elimination of pathogens and apoptotic cells and restoration of homeostasis. Pre-clinical studies have evidenced the anti-inflammatory and pro-resolving actions of GILZ (glucocorticoid-induced leucine zipper). Here, we evaluated the role of GILZ on the migration of mononuclear cells under nonphlogistic conditions and Escherichia coli-evoked peritonitis. TAT-GILZ (a cell-permeable GILZ-fusion protein) injection into the pleural cavity of mice induced monocyte/macrophage influx alongside increased CCL2, IL-10 and TGF-ß levels. TAT-GILZ-recruited macrophages showed a regulatory phenotype, exhibiting increased expression of CD206 and YM1. During the resolving phase of E. coli-induced peritonitis, marked by an increased recruitment of mononuclear cells, lower numbers of these cells and CCL2 levels were found in the peritoneal cavity of GILZ-deficient mice (GILZ-/-) when compared to WT. In addition, GILZ-/- showed higher bacterial loads, lower apoptosis/efferocytosis counts and a lower number of macrophages with pro-resolving phenotypes. TAT-GILZ accelerated resolution of E. coli-evoked neutrophilic inflammation, which was associated with increased peritoneal numbers of monocytes/macrophages, enhanced apoptosis/efferocytosis counts and bacterial clearance through phagocytosis. Taken together, we provided evidence that GILZ modulates macrophage migration with a regulatory phenotype, inducing bacterial clearance and accelerating the resolution of peritonitis induced by E. coli.


Assuntos
Infecções por Escherichia coli , Peritonite , Fatores de Transcrição , Animais , Camundongos , Escherichia coli/metabolismo , Infecções por Escherichia coli/metabolismo , Inflamação/metabolismo , Macrófagos/metabolismo , Monócitos/metabolismo , Peritonite/metabolismo , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...