Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 161
1.
Cell Rep ; 43(4): 114007, 2024 Apr 23.
Article En | MEDLINE | ID: mdl-38517889

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.


Receptor, Insulin , Signal Transduction , Urinary Bladder , Urinary Tract Infections , Urothelium , Receptor, Insulin/metabolism , Urinary Tract Infections/microbiology , Urinary Tract Infections/metabolism , Urinary Tract Infections/pathology , Animals , Urothelium/metabolism , Urothelium/pathology , Urothelium/microbiology , Humans , Urinary Bladder/microbiology , Urinary Bladder/pathology , Urinary Bladder/metabolism , Mice , Uropathogenic Escherichia coli/pathogenicity , Mice, Inbred C57BL , NF-kappa B/metabolism , Female , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Insulin/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Male
2.
Dev Cell ; 59(1): 33-47.e5, 2024 Jan 08.
Article En | MEDLINE | ID: mdl-38101412

Aging is a risk factor for disease via increased susceptibility to infection, decreased ability to maintain homeostasis, inefficiency in combating stress, and decreased regenerative capacity. Multiple diseases, including urinary tract infection (UTI), are more prevalent with age; however, the mechanisms underlying the impact of aging on the urinary tract mucosa and the correlation between aging and disease remain poorly understood. Here, we show that, relative to young (8-12 weeks) mice, the urothelium of aged (18-24 months) female mice accumulates large lysosomes with reduced acid phosphatase activity and decreased overall autophagic flux in the aged urothelium, indicative of compromised cellular homeostasis. Aged bladders also exhibit basal accumulation of reactive oxygen species (ROS) and a dampened redox response, implying heightened oxidative stress. Furthermore, we identify a canonical senescence-associated secretory phenotype (SASP) in the aged urothelium, along with continuous NLRP3-inflammasome- and Gasdermin-D-dependent pyroptotic cell death. Consequently, aged mice chronically exfoliate urothelial cells, further exacerbating age-related urothelial dysfunction. Upon infection with uropathogenic E. coli, aged mice harbor increased bacterial reservoirs and are more prone to spontaneous recurrent UTI. Finally, we discover that treatment with D-mannose, a natural bioactive monosaccharide, rescues autophagy flux, reverses the SASP, and mitigates ROS and NLRP3/Gasdermin/interleukin (IL)-1ß-driven pyroptotic epithelial cell shedding in aged mice. Collectively, our results demonstrate that normal aging affects bladder physiology, with aging alone increasing baseline cellular stress and susceptibility to infection, and suggest that mannose supplementation could serve as a senotherapeutic to counter age-associated urothelial dysfunction.


NLR Family, Pyrin Domain-Containing 3 Protein , Urinary Tract Infections , Mice , Female , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Urinary Bladder/metabolism , Urinary Bladder/microbiology , Urinary Bladder/pathology , Mannose/metabolism , Reactive Oxygen Species/metabolism , Escherichia coli/metabolism , Urothelium/metabolism , Urothelium/microbiology , Interleukin-1beta , Gasdermins , Urinary Tract Infections/metabolism , Urinary Tract Infections/microbiology , Urinary Tract Infections/pathology , Cellular Senescence
3.
Front Cell Infect Microbiol ; 13: 1128132, 2023.
Article En | MEDLINE | ID: mdl-37051302

Introduction: Murine models of urinary tract infection (UTI) have improved our understanding of host-pathogen interactions. However, given differences between rodent and human bladders which may modulate host and bacterial response, including certain biomarkers, urothelial thickness and the concentration of urine, the development of new human-based models is important to complement mouse studies and to provide a more complete picture of UTI in patients. Methods: We originally developed a human urothelial three-dimensional (3D) model which was urine tolerant and demonstrated several urothelial biomarkers, but it only achieved human thickness in heterogenous, multi-layered zones and did not demonstrate the comprehensive differentiation status needed to achieve barrier function. We optimised this model by altering a variety of conditions and validated it with microscopy, flow cytometry, transepithelial electrical resistance and FITC-dextran permeability assays to confirm tissue architecture, barrier integrity and response to bacterial infection. Results: We achieved an improved 3D urine-tolerant human urothelial model (3D-UHU), which after 18-20 days of growth, stratified uniformly to 7-8 layers comprised of the three expected, distinct human cell types. The apical surface differentiated into large, CD227+ umbrella-like cells expressing uroplakin-1A, II, III, and cytokeratin 20, all of which are important terminal differentiation markers, and a glycosaminoglycan layer. Below this layer, several layers of intermediate cells were present, with a single underlying layer of CD271+ basal cells. The apical surface also expressed E-cadherin, ZO-1, claudin-1 and -3, and the model possessed good barrier function. Infection with both Gram-negative and Gram-positive bacterial classes elicited elevated levels of pro-inflammatory cytokines and chemokines characteristic of urinary tract infection in humans and caused a decrease in barrier function. Discussion: Taken together, 3D-UHU holds promise for studying host-pathogen interactions and host urothelial immune response.


Urinary Tract Infections , Urothelium , Humans , Mice , Animals , Urothelium/microbiology , Urinary Bladder/microbiology , Cytokines/metabolism , Biomarkers/metabolism
4.
Molecules ; 27(1)2022 Jan 02.
Article En | MEDLINE | ID: mdl-35011504

Uropathogenic Escherichia coli (UPEC) is the most common pathogenic bacteria associated with urinary tract infection (UTI). UPEC can cause UTI by adhering to and invading uroepithelial cells. Fimbriae is the most important virulence factor of UPEC, and a potentially promising target in developing novel antibacterial treatments. In this study, the antibacterial properties and effects of the compound dictamnine, extracted from the traditional Chinese medicine Cortex Dictamni, on the bacterial morphology, cell adhesion, and invasion of UPEC were studied. Dictamnine exhibited no obvious antibacterial activity against UPEC, but significantly impeded the ability of UPEC to adhere to and invade uroepithelial cells. RT-qPCR analysis showed that treatment downregulated the expression of type 1 fimbriae, P fimbriae, and curli fimbriae adhesion genes, and also downregulated adhesion-related receptor genes of uroepithelial cells. Transmission electron microscopy showed that dictamnine destroyed the structure of the fimbriae and the surface of the bacteria became smooth. These results suggest that dictamnine may help to prevent UTI by simultaneously targeting UPEC fimbriae and urothelial adhesin receptors, and may have a potential use as a new anti-UPEC drug.


Bacterial Adhesion/drug effects , Escherichia coli Infections/drug therapy , Quinolines/pharmacology , Urinary Tract Infections/drug therapy , Uropathogenic Escherichia coli/metabolism , Urothelium/microbiology , Cell Line , Escherichia coli Infections/microbiology , Humans , Urinary Tract Infections/microbiology , Urothelium/metabolism
5.
Cell Rep ; 37(3): 109856, 2021 10 19.
Article En | MEDLINE | ID: mdl-34686330

Uropathogenic Escherichia coli (UPEC) cause urinary tract infections (UTIs) by invading urothelial cells. In response, the host mounts an inflammatory response to expel bacteria. Here, we show that the NF-E2-related factor 2 (NRF2) pathway is activated in response to UPEC-triggered reactive oxygen species (ROS) production. We demonstrate the molecular sequence of events wherein NRF2 activation in urothelial cells reduces ROS production, inflammation, and cell death, promotes UPEC expulsion, and reduces the bacterial load. In contrast, loss of NRF2 leads to increased ROS production, bacterial burden, and inflammation, both in vitro and in vivo. NRF2 promotes UPEC expulsion by regulating transcription of the RAB-GTPase RAB27B. Finally, dimethyl fumarate, a US Food and Administration-approved NRF2 inducer, reduces the inflammatory response, increases RAB27B expression, and lowers bacterial burden in urothelial cells and in a mouse UTI model. Our findings elucidate mechanisms underlying the host response to UPEC and provide a potential strategy to combat UTIs.


Escherichia coli Infections/metabolism , NF-E2-Related Factor 2/metabolism , Reactive Oxygen Species/metabolism , Urinary Tract Infections/metabolism , Uropathogenic Escherichia coli/pathogenicity , Urothelium/metabolism , rab27 GTP-Binding Proteins/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Bacterial Load , Cell Line, Tumor , Dimethyl Fumarate/pharmacology , Escherichia coli Infections/drug therapy , Escherichia coli Infections/genetics , Escherichia coli Infections/microbiology , Female , HEK293 Cells , Host-Pathogen Interactions , Humans , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/agonists , NF-E2-Related Factor 2/genetics , Oxidative Stress , Signal Transduction , Urinary Tract Infections/drug therapy , Urinary Tract Infections/genetics , Urinary Tract Infections/microbiology , Urothelium/drug effects , Urothelium/microbiology , rab GTP-Binding Proteins , rab27 GTP-Binding Proteins/genetics
6.
Sci Rep ; 11(1): 19263, 2021 09 28.
Article En | MEDLINE | ID: mdl-34584119

Urinary tract infection frequently caused by E. coli is one of the most common bacterial infections. Increasing antibiotic resistance jeopardizes successful treatment and alternative treatment strategies are therefore mandatory. Metformin, an oral antidiabetic drug, has been shown to activate macrophages in the protection against certain infecting microorganisms. Since epithelial cells often form the first line of defense, we here investigated the effect on uroepithelial cells during E. coli infection. Metformin upregulated the human antimicrobial peptides cathelicidin LL-37 and RNase7 via modulation of the TRPA1 channel and AMPK pathway. Interestingly, metformin stimulation enriched both LL-37 and TRPA1 in lysosomes. In addition, metformin specifically increased nitric oxide and mitochondrial, but not cytosolic ROS. Moreover, metformin also triggered mRNA expression of the proinflammatory cytokines IL1B, CXCL8 and growth factor GDF15 in human uroepithelial cells. The GDF15 peptide stimulated macrophages increased LL-37 expression, with increased bacterial killing. In conclusion, metformin stimulation strengthened the innate immunity of uroepithelial cells inducing enhanced extracellular and intracellular bacterial killing suggesting a favorable role of metformin in the host defense.


Escherichia coli Infections/drug therapy , Metformin/pharmacology , Urinary Tract Infections/drug therapy , Urothelium/drug effects , Antimicrobial Cationic Peptides/metabolism , Cell Line , Cytokines/metabolism , Drug Repositioning , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Humans , Immunity, Innate/drug effects , Metformin/therapeutic use , Ribonucleases/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , TRPA1 Cation Channel/metabolism , Up-Regulation/drug effects , Up-Regulation/immunology , Urinary Tract Infections/immunology , Urinary Tract Infections/microbiology , Uropathogenic Escherichia coli/immunology , Urothelium/immunology , Urothelium/microbiology , Cathelicidins
7.
Pathog Dis ; 79(6)2021 07 02.
Article En | MEDLINE | ID: mdl-34143186

Bacterial infection may have a pathophysiological role in refractory Detrusor Overactivity (DO). The aim of this study was to observe any impact of antibiotic therapy upon bacterial colonization of urothelial cells, and to determine whether a relationship existed between colonization and symptom severity. Mid-stream urine samples were collected as part of a clinical trial of antibiotics in women with refractory DO. Wright stained urothelial cells were categorized according to the degree of bacterial colonization as; 'clear' (free of bacteria), or as associated with bacteria that were 'adjacent' to the cell or 'intracellular' at low or high density. The average percentages were compared with routine microbiology cultures, over the 26 week trial, and with patient clinical outcome measures of DO severity. In patients receiving placebo, 'high-density intracellular bacteria' significantly increased during urinary tract infection (P = 0.0008). In antibiotic patients, 'clear' cells were more prevalent. Amoxicillin & Clavulanic Acid significantly decreased bacterial colonization within urothelial cells, suggesting that these antibiotics possess the greatest intracellular efficacy. 'High-density intracellular bacteria' positively correlated with symptom severity, measured by leakage on pad test (P = 0.014), leaks per day (P = 0.004), and voids per day (P = 0.005). Thus, by decreasing high density intracellular bacteria, antibiotic treatment may improve the refractory DO condition.


Amoxicillin/administration & dosage , Anti-Bacterial Agents/administration & dosage , Bacteria/drug effects , Bacterial Infections/drug therapy , Clavulanic Acid/administration & dosage , Cystitis/drug therapy , Urinary Bladder, Underactive/drug therapy , Urothelium/microbiology , Bacteriological Techniques , Cystitis/microbiology , Female , Humans , Microscopy, Confocal , Severity of Illness Index , Treatment Outcome , Urinary Tract Infections/drug therapy , Urine/microbiology , Urothelium/cytology
9.
J Microbiol Immunol Infect ; 54(2): 193-205, 2021 Apr.
Article En | MEDLINE | ID: mdl-31296484

BACKGROUND: Patients with diabetes mellitus (DM) have higher incidence and more severe urinary tract infections (UTIs) for longer duration than those of the patients without DM. It causes more complicated etiologies during uropathogenic Escherichia coli (UPEC) infection. However, studies regarding the molecular mechanism are scarce. METHODS: The present study (1) aimed to verify if sugar influences the process of UPEC-induced cystitis and invasion into the uroepithelial cells and (2) illustrated the mechanism of effects for sugar enhanced the UPEC infection into uroepithelial cells is related to TLR-4-mediated and JAK/STAT1-dependent pathway. RESULTS: The results of the present study indicated that sugar can enhance UPEC infection in uroepithelial cells by up-regulating the transduced circuit between TLR-4-mediated UPEC interaction and JAK/STAT-1 signal pathways. The results of the inhibitor-co-incubating experiments demonstrated that the mechanism involved in the synergistic amplification of TLR-4-mediated UPEC interaction and JAK/STAT1 signaling pathways is responsible for the increased UPEC infection in uroepithelial cells. CONCLUSION: The results also proved that STAT-1 plays a critical role in the regulation of UPEC invasion and infection in the uroepithelial cells, especially those pretreated with glucose. The present study suggests a possible therapeutic approach to preferentially suppress UPEC infection during UTIs in the patients with diabetes.


STAT1 Transcription Factor/metabolism , Signal Transduction , Sugars/metabolism , Toll-Like Receptor 4/metabolism , Uropathogenic Escherichia coli , Urothelium/metabolism , Cell Line , Epithelial Cells/metabolism , Escherichia coli Infections/pathology , Gene Expression Regulation , Humans , Toll-Like Receptor 4/genetics , Urinary Tract Infections/metabolism , Urinary Tract Infections/pathology , Urothelium/microbiology , Urothelium/pathology
10.
Clin Cancer Res ; 26(20): 5487-5493, 2020 10 15.
Article En | MEDLINE | ID: mdl-32933995

PURPOSE: Emerging evidence indicates that gut microbiota dysbiosis can reduce the effectiveness of immune checkpoint inhibitors (ICI). Proton pump inhibitors (PPI) are known to induce gut microbiota changes. However, little is known on the effects of PPIs on outcomes with ICI therapy, and it has not been explored in urothelial cancer treatment. EXPERIMENTAL DESIGN: Individual-participant data from the advanced urothelial cancer trials, IMvigor210 (single-arm atezolizumab trial, n = 429) and IMvigor211 (phase III randomized trial of atezolizumab vs. chemotherapy, n = 931) were pooled in a Cox proportional hazard analysis assessing the association between PPI use and overall survival (OS) and progression-free survival (PFS). PPI use was defined as any PPI administration between 30 days prior and 30 days after treatment initiation. RESULTS: Of the 1,360 participants, 471 (35%) received a PPI within the 60-day window. PPI use was associated with significantly worse OS [HR (95% confidence interval (CI)) = 1.52 (1.27-1.83), P < 0.001] and PFS [1.38 (1.18-1.62), P < 0.001] with atezolizumab, but not chemotherapy (P > 0.05). In the randomized cohort of IMvigor211, the OS treatment effect [HR (95% CI)] of atezolizumab versus chemotherapy was 1.04 (0.81-1.34) for PPI users, compared with 0.69 (0.56-0.84) for PPI nonusers (P interaction = 0.013). Similar associations were noted in the PD-L1 IC2/3 population. CONCLUSIONS: This study indicates PPI use is a negative prognostic marker in advanced urothelial carcinoma treated with ICI therapy, but not chemotherapy. Furthermore, the analysis suggests PPIs influence the magnitude of ICI efficacy, and this warrants further investigation.


Antibodies, Monoclonal, Humanized/administration & dosage , Dysbiosis/drug therapy , Gastrointestinal Microbiome/drug effects , Neoplasms/drug therapy , Adult , Aged , Antibodies, Monoclonal, Humanized/adverse effects , B7-H1 Antigen/genetics , Dysbiosis/complications , Dysbiosis/microbiology , Dysbiosis/pathology , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immune Checkpoint Inhibitors/administration & dosage , Immune Checkpoint Inhibitors/adverse effects , Male , Middle Aged , Neoplasms/microbiology , Neoplasms/pathology , Progression-Free Survival , Proportional Hazards Models , Proton Pump Inhibitors/administration & dosage , Proton Pump Inhibitors/adverse effects , Urothelium/drug effects , Urothelium/microbiology , Urothelium/pathology
11.
Infect Immun ; 88(9)2020 08 19.
Article En | MEDLINE | ID: mdl-32540870

Uropathogenic Escherichia coli (UPEC) is the leading cause of urinary tract infections. These bacteria undertake a multistage infection cycle involving invasion of and proliferation within urinary tract epithelial cells, leading to the rupture of the host cell and dispersal of the bacteria, some of which have a highly filamentous morphology. Here, we established a microfluidics-based model of UPEC infection of immortalized human bladder epithelial cells that recapitulates the main stages of bacterial morphological changes during the acute infection cycle in vivo and allows the development and fate of individual cells to be monitored in real time by fluorescence microscopy. The UPEC-infected bladder cells remained alive and mobile in nonconfluent monolayers during the development of intracellular bacterial communities (IBCs). Switching from a flow of growth medium to human urine resulted in immobilization of both uninfected and infected bladder cells. Some IBCs continued to develop and then released many highly filamentous bacteria via an extrusion-like process, whereas other IBCs showed strong UPEC proliferation, and yet no filamentation was detected. The filamentation response was dependent on the weak acidity of human urine and required component(s) in a low molecular-mass (<3,000 Da) fraction from a mildly dehydrated donor. The developmental fate for bacteria therefore appears to be controlled by multiple factors that act at the level of the whole IBC, suggesting that variable local environments or stochastic differentiation pathways influence IBC developmental fates during infection.


Epithelial Cells/microbiology , Microfluidic Analytical Techniques , Uropathogenic Escherichia coli/pathogenicity , Uropathogenic Escherichia coli/ultrastructure , Cell Line, Transformed , Cell Movement , Cell Proliferation , Epithelial Cells/pathology , Epithelial Cells/ultrastructure , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Hydrogen-Ion Concentration , Microscopy, Fluorescence , Models, Biological , Rheology , Urinary Bladder/microbiology , Urinary Bladder/pathology , Urinary Tract Infections/microbiology , Urinary Tract Infections/pathology , Uropathogenic Escherichia coli/physiology , Urothelium/microbiology , Urothelium/pathology
12.
J Infect Dev Ctries ; 14(4): 366-372, 2020 04 30.
Article En | MEDLINE | ID: mdl-32379713

INTRODUCTION: Approximately 5% of men and 40%-50% of women have experienced urinary tract infections (UTI), which are the most common infectious diseases and nosocomial infections in humans. Proteus mirabilis is susceptible to most antibiotics, but antibiotic treatment usually causes side effects. In this research, lactic acid bacteria (LAB) was assessed for its inhibitory activity against a urinary tract pathogen. METHODOLOGY: We studied the effect of pH adjustment, heat, and enzyme treatments on the inhibitory activity of LAB strains and their supernatants, using well-diffusion and co-culture assays. In the cell culture assay, anti-adhesion and anti-invasion activities against P. mirabilis were tested with SV-HUC-1 urothelial cells. RESULTS: LAB were able to adhere to the urothelial cells and inhibited P. mirabilis growth. LAB were also able to inhibit P. mirabilis adhesion to or invasion of SV-HUC-1 urothelial cells. Finally, in the competition assay, LAB showed inhibitory effects against P. mirabilis. LAB could also inhibit the invasion of P. mirabilis into urothelial cells. CONCLUSIONS: Two LAB strains (PM206 and 229) exhibited antagonistic activity against P. mirabilis adhesion or invasion of urothelial cells in culture. In the future, probiotics may be used in food or urinary tract cleansing and could replace antibiotic treatments.


Lactobacillales/physiology , Probiotics/pharmacology , Proteus Infections/prevention & control , Urinary Tract Infections/prevention & control , Urothelium/microbiology , Antibiosis , Bacterial Adhesion , Cell Line , Culture Media , Female , Humans , Proteus mirabilis/growth & development , Urinary Tract Infections/microbiology , Urothelium/cytology
13.
J Ethnopharmacol ; 257: 112889, 2020 Jul 15.
Article En | MEDLINE | ID: mdl-32311481

ETHNOPHARMACOLOGICAL RELEVANCE: Extracts from Cranberry fruits (Vaccinium macrocarpon) are traditionally used against urinary tract infections, mainly due to antiadhesive activity against uropathogenic E. coli (UPEC), but the exact mode of action and compounds, responsible for the activity, are unknown. AIM OF THE STUDY: i. To investigate if cranberry extract acts only by a single component or must be assessed as a multi-active-compound preparation; ii to screen isolated cranberry-related natural products under in vitro conditions to pinpoint natural products with antiadhesive effects against UPEC, followed by in silico calculations (QSAR) to predict potential antiadhesive compounds; iii. investigations by using urine samples from cranberry treated volunteers for evaluation on the bacterial transcriptome and the mannose-binding side of FimH, iv. to investigate if besides Tamm Horsfall Protein induction in the kidney, the extract acts also directly against UPEC. MATERIAL AND METHODS: Antiadhesive activity of 105 compounds was determined by flow cytometric adhesion assay (UPEC UTI89 on T24 bladder cells). Urine samples from 16 volunteers treated with cranberry extract (p.o., 7 days, 900 mg/day) were used for ex vivo testing concerning influence on the bacterial transcriptome (Illumina RNA-seq) and interaction with the mannose binding domain of type-1 fimbriae. RESULTS: i. The antiadhesive effect of cranberry extract cannot be attributed to a single compound or to a single fraction. ii. Unglycosylated flavones and flavonols with bulky substitution of the B ring contribute to the antiadhesive activity. 3'-8″-biflavones and flavolignans (not related to cranberry fruits) were identified as potent antiadhesive compounds against UPEC. iii. QSAR yielded a model with good statistical performance and sufficient internal and external predictive ability. iv. Urine samples from male cranberry-treated volunteers indicated significant interaction with the mannose binding domain of type-1 fimbriae, which correlated with the amount of Tamm-Horsfall Protein in the test samples. v Cranberry extract did not influence the UPEC transcriptome; gene expression of bacterial adhesins (P-, S-fimbrae, curli) was not influenced by the urine samples, while a slight, but non-significant upregulation of type 1 fimbriae was observed. CONCLUSIONS: B-ring substituted flavones and flavonols from cranberry contribute to the antiadhesive activity against UPEC by inhibition of the FimH-mediated interaction with the host cell bladder epithelium.


Anti-Bacterial Agents/pharmacology , Bacterial Adhesion/drug effects , Escherichia coli Infections/drug therapy , Plant Extracts/pharmacology , Urinary Tract Infections/drug therapy , Uropathogenic Escherichia coli/drug effects , Vaccinium macrocarpon , Adhesins, Escherichia coli/genetics , Adhesins, Escherichia coli/metabolism , Administration, Oral , Adult , Aged, 80 and over , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/isolation & purification , Anti-Bacterial Agents/urine , Cell Line , Escherichia coli Infections/microbiology , Escherichia coli Infections/urine , Female , Fimbriae Proteins/genetics , Fimbriae Proteins/metabolism , Fruit , Gene Expression Regulation, Bacterial , Host-Pathogen Interactions , Humans , Male , Middle Aged , Plant Extracts/administration & dosage , Plant Extracts/isolation & purification , Plant Extracts/urine , Urinary Bladder/drug effects , Urinary Bladder/microbiology , Urinary Tract Infections/microbiology , Urinary Tract Infections/urine , Urine/microbiology , Uromodulin/metabolism , Uropathogenic Escherichia coli/genetics , Uropathogenic Escherichia coli/pathogenicity , Urothelium/drug effects , Urothelium/microbiology , Vaccinium macrocarpon/chemistry , Young Adult
14.
Mucosal Immunol ; 13(4): 702-714, 2020 07.
Article En | MEDLINE | ID: mdl-32112048

The urothelium of the urinary bladder represents the first line of defense. However, uropathogenic E. coli (UPEC) damage the urothelium and cause acute bacterial infection. Here, we demonstrate the crosstalk between macrophages and the urothelium stimulating macrophage migration into the urothelium. Using spatial proteomics by MALDI-MSI and LC-MS/MS, a novel algorithm revealed the spatial activation and migration of macrophages. Analysis of the spatial proteome unravelled the coexpression of Myo9b and F4/80 in the infected urothelium, indicating that macrophages have entered the urothelium upon infection. Immunofluorescence microscopy additionally indicated that intraurothelial macrophages phagocytosed UPEC and eliminated neutrophils. Further analysis of the spatial proteome by MALDI-MSI showed strong expression of IL-6 in the urothelium and local inhibition of this molecule reduced macrophage migration into the urothelium and aggravated the infection. After IL-6 inhibition, the expression of matrix metalloproteinases and chemokines, such as CX3CL1 was reduced in the urothelium. Accordingly, macrophage migration into the urothelium was diminished in the absence of CX3CL1 signaling in Cx3cr1gfp/gfp mice. Conclusively, this study describes the crosstalk between the infected urothelium and macrophages through IL-6-induced CX3CL1 expression. Such crosstalk facilitates the relocation of macrophages into the urothelium and reduces bacterial burden in the urinary bladder.


Cell Communication , Chemokine CX3CL1/metabolism , Interleukin-6/metabolism , Macrophages/metabolism , Proteomics , Urothelium/immunology , Urothelium/metabolism , Animals , Disease Models, Animal , Disease Susceptibility , Fluorescent Antibody Technique , Immunohistochemistry , Macrophages/immunology , Mice , Proteomics/methods , Urinary Bladder/immunology , Urinary Bladder/metabolism , Urinary Bladder/microbiology , Urinary Tract Infections/etiology , Urinary Tract Infections/metabolism , Urinary Tract Infections/pathology , Urothelium/microbiology
15.
Bull Exp Biol Med ; 167(6): 795-800, 2019 Oct.
Article En | MEDLINE | ID: mdl-31656005

We performed an electron microscopic study of samples of urethral polyps obtained from 90 women (mean age 52.5±4.9 years). According to PCR and culture studies, the most common infectious agent in patients with urethral polyps is U. urealyticum (100% cases). In 70% cases, this infectious agent was present as monoinfection, of these, clinically significant concentration (>106 CFU/ml) were found in 53.3% cases. In 30% cases, associations with C. trachomatis, T. vaginalis, and M. genitalium were found. We observed significant ultrastructural heterogeneity of the epithelial cells in urethral polyps, which manifested in a combination of hyperplastic and metaplastic changes and signs of cytodestruction. Detection of mycoplasma-like bodies in connective tissue mononuclear cells and viral particles in epithelial cells during ultrastructural study, including cases with negative PCR results, indicates the pathogenetic role of latent infection in the formation of urethral polyps.


Polyps/complications , Polyps/pathology , Urethral Neoplasms/complications , Urethral Neoplasms/pathology , Urinary Tract Infections/complications , Urothelium/ultrastructure , Chlamydia Infections/epidemiology , Chlamydia Infections/pathology , Chlamydia trachomatis/genetics , Chlamydia trachomatis/isolation & purification , Female , Humans , Middle Aged , Mycoplasma Infections/epidemiology , Mycoplasma Infections/pathology , Mycoplasma genitalium/genetics , Mycoplasma genitalium/isolation & purification , Polymerase Chain Reaction , Polyps/epidemiology , Polyps/ultrastructure , Trichomonas Infections/epidemiology , Trichomonas Infections/pathology , Trichomonas vaginalis/genetics , Trichomonas vaginalis/isolation & purification , Ureaplasma Infections/epidemiology , Ureaplasma Infections/pathology , Ureaplasma urealyticum/genetics , Ureaplasma urealyticum/isolation & purification , Urethral Diseases/complications , Urethral Diseases/epidemiology , Urethral Diseases/microbiology , Urethral Diseases/pathology , Urethral Neoplasms/epidemiology , Urethral Neoplasms/ultrastructure , Urinary Tract Infections/epidemiology , Urinary Tract Infections/microbiology , Urothelium/microbiology , Urothelium/pathology
16.
Methods Mol Biol ; 2021: 129-137, 2019.
Article En | MEDLINE | ID: mdl-31309502

Bacterial adherence to eukaryotic cells is mediated by different adhesins that can act at different stages in bacteria-host interaction. Abundant evidence has suggested that adherence is critical for infection by bacterial pathogens. Proteus mirabilis is an opportunistic pathogen which frequently infects the human urinary tract, particularly in patients with indwelling urinary catheters. Sequencing of the genome of this pathogen has revealed the existence of a remarkable amount of complete fimbrial operons. In this chapter, we describe in vitro adherence assays of P. mirabilis to uroepithelial cells, which can provide relevant results to assess virulence of uropathogenic strains.


Adhesins, Bacterial/metabolism , Proteus mirabilis/physiology , Urothelium/microbiology , Animals , Bacterial Adhesion , Cell Line , Host Microbial Interactions , Humans , Proteus mirabilis/pathogenicity , Urothelium/cytology , Virulence
17.
Am J Physiol Cell Physiol ; 317(3): C544-C555, 2019 09 01.
Article En | MEDLINE | ID: mdl-31241987

The specific regulatory mechanism of bladder urothelial barrier dysfunction after infection with uropathogenic Escherichia coli (UPEC) is still unclear. The cross talk between bladder urothelial cells and mast cells may play an important role during UPEC infection. In this study, the pyroptosis of urothelial cells was investigated after UPEC infection both in vivo and in vitro. The levels of IL-1ß and IL-18 in exosomes derived from bladder urothelial cells after UPEC infection were detected. The role of these processes in the recruitment and activation of mast cells was measured. The mechanism of mast cell-induced disruption of bladder epithelial barrier function was also assessed. We found that UPEC infection induced pyroptosis of bladder urothelial cells and led to the release of IL-1ß and IL-18 in the form of exosomes, which promoted the migration of mast cells. Tryptase secreted by mast cells aggravated the damage to the barrier function of the bladder urothelium by acting on protease-activated receptor 2 (PAR2). Inhibition of pyroptosis or the tryptase-PAR2 axis reduced the disruption of bladder urothelial barrier function and decreased the bacterial burden. The present study supports a novel mechanism by which pyroptosis-dependent release of exosomes from bladder urothelial cells activates mast cells and regulates bladder urothelial barrier function during UPEC infection.


Escherichia coli Infections/metabolism , Exosomes/metabolism , Mast Cells/metabolism , Pyroptosis/physiology , Urinary Tract Infections/metabolism , Urothelium/metabolism , Animals , Cell Line , Escherichia coli Infections/immunology , Exosomes/immunology , Female , Humans , Mast Cells/immunology , Mice , Mice, Inbred C57BL , Urinary Bladder/immunology , Urinary Bladder/metabolism , Urinary Bladder/microbiology , Urinary Tract Infections/immunology , Uropathogenic Escherichia coli , Urothelium/immunology , Urothelium/microbiology
18.
Autophagy ; 15(3): 527-542, 2019 03.
Article En | MEDLINE | ID: mdl-30335568

50% of Caucasians carry a Thr300Ala variant (T300A) in the protein encoded by the macroautophagy/autophagy gene ATG16L1. Here, we show that the T300A variant confers protection against urinary tract infections (UTIs), the most common infectious disease in women. Using knockin mice carrying the human T300A variant, we show that the variant limits the UTI-causing bacteria, uropathogenic Escherichia coli (UPEC), from establishing persistent intracellular reservoirs, which can seed UTI recurrence. This phenotype is recapitulated in mice lacking Atg16l1 or Atg7 exclusively in the urothelium. We further show that mice with the T300A variant exhibit urothelial cellular abnormalities, including vesicular congestion and aberrant accumulation of UPK (uroplakin) proteins. Importantly, presence of the T300A variant in humans is associated with similar urothelial architectural abnormalities, indicating an evolutionarily conserved impact. Mechanistically, we show that the reduced bacterial persistence is independent of basal autophagic flux or proinflammatory cytokine responses and does not involve Atg14 or Epg5. However, the T300A variant is associated with increased expression of the small GTPase Rab33b; RAB33B interacts with ATG16L1, as well as other secretory RABs, RAB27B and RAB11A, important for UPEC exocytosis from the urothelium. Finally, inhibition of secretory RABs in bladder epithelial cells increases intracellular UPEC load. Together, our results reveal that UPEC selectively utilize genes important for autophagosome formation to persist in the urothelium, and that the presence of the T300A variant in ATG16L1 is associated with changes in urothelial vesicle trafficking, which disrupts the ability of UPEC to persist, thereby limiting the risk of recurrent UTIs. Abbreviations: 3-PEHPC: 3-pyridinyl ethylidene hydroxyl phosphonocarboxylate; ATG: autophagy; ATG16L1: autophagy related 16 like 1; BECs: bladder epithelial cells; dpi: days post infection; hpi: hours post infection; IF: immunofluorescence; IL1B: interleukin 1 beta; IL6: interleukin 6; MAP1LC3B/LC3B: microtubule-associated protein 1 light chain 3 beta; MVB: multivesicular bodies; T300A: Thr300Ala; TNF: tumor necrosis factor; QIR(s): quiescent intracellular reservoir(s); siRNA: short interfering RNA; UPEC: uropathogenic Escherichia coli; UTI(s): urinary tract infection(s); TEM: transmission electron microscopy; WT: wild type.


Autophagy/genetics , Escherichia coli Infections/metabolism , Urinary Tract Infections/metabolism , Uropathogenic Escherichia coli , Urothelium/microbiology , Animals , Autophagy-Related Protein 7/genetics , Autophagy-Related Protein 7/metabolism , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/metabolism , Cell Line , Epithelial Cells/metabolism , Escherichia coli Infections/genetics , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Female , Genetic Variation , Humans , Macrophages/metabolism , Mice , Mice, Knockout , Multivesicular Bodies/genetics , Multivesicular Bodies/microbiology , Multivesicular Bodies/pathology , Urinary Bladder/microbiology , Urinary Tract Infections/genetics , Urinary Tract Infections/microbiology , Urinary Tract Infections/pathology , Uroplakins/metabolism , Urothelium/cytology , Urothelium/metabolism , Urothelium/ultrastructure , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism , rab GTP-Binding Proteins/metabolism
19.
Med Mal Infect ; 49(5): 350-355, 2019 Aug.
Article En | MEDLINE | ID: mdl-30583869

PURPOSE: Intravesical BCG is the standard treatment of non-muscle invasive bladder cancer. No difference has yet been reported in the safety profiles of the various BCG strains. METHODS: A nationwide multidisciplinary retrospective survey was conducted between January 2013 and December 2016 to identify cases of BCG infection and differentiate them based on the type of BCG strain used. RESULTS: Forty patients were identified (BCG RIVM 28; other strains 8; unknown 4). Patients treated with BCG RIVM were less severely ill, with fewer occurrences of septic shock (3.6% vs. 50%, P=0.003) and ICU admission (7.1% vs. 62.5%, P=0.003). A higher frequency of pulmonary miliaries (71.4% vs. 12.5%, P=0.005) but lower transaminase levels (mean AST 65 vs. 264 U/L, P=0.001) were observed in these patients. No difference in terms of recovery was reported. CONCLUSION: The type of BCG strain could correlate with the frequency and severity of subsequent BCG infections.


BCG Vaccine/administration & dosage , BCG Vaccine/adverse effects , Bacillaceae Infections/etiology , Carcinoma, Transitional Cell/drug therapy , Urinary Bladder Neoplasms/drug therapy , Administration, Intravesical , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , BCG Vaccine/classification , Bacillaceae Infections/microbiology , Carcinoma, Transitional Cell/pathology , Female , Humans , Male , Middle Aged , Retrospective Studies , Urinary Bladder Neoplasms/pathology , Urothelium/microbiology , Urothelium/pathology
20.
Sci Rep ; 8(1): 17559, 2018 12 03.
Article En | MEDLINE | ID: mdl-30510202

Bacterial infections can be treated with bacteriophages that show great specificity towards their bacterial host and can be genetically modified for different applications. However, whether and how bacteriophages can kill intracellular bacteria in human cells remains elusive. Here, using CRISPR/Cas selection, we have engineered a fluorescent bacteriophage specific for E. coli K1, a nosocomial pathogen responsible for urinary tract infections, neonatal meningitis and sepsis. By confocal and live microscopy, we show that engineered bacteriophages K1F-GFP and E. coli EV36-RFP bacteria displaying the K1 capsule, enter human cells via phagocytosis. Importantly, we show that bacteriophage K1F-GFP efficiently kills intracellular E. coli EV36-RFP in T24 human urinary bladder epithelial cells. Finally, we provide evidence that bacteria and bacteriophages are degraded by LC3-associated phagocytosis and xenophagy.


Epithelial Cells/microbiology , Escherichia coli Infections/therapy , Escherichia coli/virology , Phage Therapy/methods , Podoviridae/growth & development , Urinary Tract Infections , Urothelium/microbiology , Cell Line , Epithelial Cells/pathology , Humans , Podoviridae/genetics , Urinary Tract Infections/microbiology , Urinary Tract Infections/therapy
...