Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 192
Filtrar
1.
Sci Rep ; 14(1): 15387, 2024 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-38965339

RESUMEN

Probiotics offer a promising prophylactic approach against various pathogens and represent an alternative strategy to combat biofilm-related infections. In this study, we isolated vaginal commensal microbiota from 54 healthy Indian women to investigate their probiotic traits. We primarily explored the ability of cell-free supernatant (CFS) from Lactobacilli to prevent Uropathogenic Escherichia coli (UPEC) colonization and biofilm formation. Our findings revealed that CFS effectively reduced UPEC's swimming and swarming motility, decreased cell surface hydrophobicity, and hindered matrix production by downregulating specific genes (fimA, fimH, papG, and csgA). Subsequent GC-MS analysis identified Tryptamine, a monoamine compound, as the potent bioactive substance from Lactobacilli CFS, inhibiting UPEC biofilms with an MBIC of 4 µg/ml and an MBEC of 8 µg/ml. Tryptamine induced significant changes in E. coli colony biofilm morphology, transitioning from the Red, Dry, and Rough (RDAR) to the Smooth and White phenotype, indicating reduced extracellular matrix production. Biofilm time-kill assays demonstrated a four-log reduction in UPEC viability when treated with Tryptamine, highlighting its potent antibacterial properties, comparable to CFS treatment. Biofilm ROS assays indicated a significant elevation in ROS generation within UPEC biofilms, suggesting a potential antibacterial mechanism. Gene expression studies with Tryptamine-treated samples showed a reduction in expression of curli gene (csgA), consistent with CFS treatment. This study underscores the potential of Tryptamine from probiotic Lactobacilli CFS as a promising antibiofilm agent against UPEC biofilms.


Asunto(s)
Biopelículas , Lactobacillus , Probióticos , Triptaminas , Escherichia coli Uropatógena , Vagina , Biopelículas/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Humanos , Triptaminas/farmacología , Femenino , Escherichia coli Uropatógena/efectos de los fármacos , Escherichia coli Uropatógena/fisiología , Probióticos/farmacología , Vagina/microbiología , Lactobacillus/efectos de los fármacos , Lactobacillus/metabolismo , Lactobacillus/fisiología , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/tratamiento farmacológico , Infecciones por Escherichia coli/prevención & control , Adulto , Antibacterianos/farmacología
2.
Arch Microbiol ; 206(8): 344, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38967798

RESUMEN

Uropathogenic Escherichia coli, the most common cause for urinary tract infections, forms biofilm enhancing its antibiotic resistance. To assess the effects of compounds on biofilm formation of uropathogenic Escherichia coli UMN026 strain, a high-throughput combination assay using resazurin followed by crystal violet staining was optimized for 384-well microplate. Optimized assay parameters included, for example, resazurin and crystal violet concentrations, and incubation time for readouts. For the assay validation, quality parameters Z' factor, coefficient of variation, signal-to-noise, and signal-to-background were calculated. Microplate uniformity, signal variability, edge well effects, and fold shift were also assessed. Finally, a screening with known antibacterial compounds was conducted to evaluate the assay performance. The best conditions found were achieved by using 12 µg/mL resazurin for 150 min and 0.023% crystal violet. This assay was able to detect compounds displaying antibiofilm activity against UMN026 strain at sub-inhibitory concentrations, in terms of metabolic activity and/or biomass.


Asunto(s)
Antibacterianos , Biopelículas , Violeta de Genciana , Ensayos Analíticos de Alto Rendimiento , Oxazinas , Escherichia coli Uropatógena , Xantenos , Biopelículas/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Escherichia coli Uropatógena/efectos de los fármacos , Escherichia coli Uropatógena/fisiología , Ensayos Analíticos de Alto Rendimiento/métodos , Xantenos/química , Antibacterianos/farmacología , Violeta de Genciana/metabolismo , Oxazinas/farmacología , Oxazinas/metabolismo , Oxazinas/química , Pruebas de Sensibilidad Microbiana , Infecciones Urinarias/microbiología , Humanos
3.
Pathog Dis ; 822024 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-38794885

RESUMEN

Urinary tract infection (UTI), one of the most common bacterial infections worldwide, is a typical example of an infection that is often polymicrobial in nature. While the overall infection course is known on a macroscale, bacterial behavior is not fully understood at the cellular level and bacterial pathophysiology during multispecies infection is not well characterized. Here, using clinically relevant bacteria, human epithelial bladder cells and human urine, we establish co-infection models combined with high resolution imaging to compare single- and multi-species bladder cell invasion events in three common uropathogens: uropathogenic Escherichia coli (UPEC), Klebsiella pneumoniae and Enterococcus faecalis. While all three species invaded the bladder cells, under flow conditions the Gram-positive E. faecalis was significantly less invasive compared to the Gram-negative UPEC and K. pneumoniae. When introduced simultaneously during an infection experiment, all three bacterial species sometimes invaded the same bladder cell, at differing frequencies suggesting complex interactions between bacterial species and bladder cells. Inside host cells, we observed encasement of E. faecalis colonies specifically by UPEC. During subsequent dispersal from the host cells, only the Gram-negative bacteria underwent infection-related filamentation (IRF). Taken together, our data suggest that bacterial multispecies invasions of single bladder cells are frequent and support earlier studies showing intraspecies cooperation on a biochemical level during UTI.


Asunto(s)
Enterococcus faecalis , Células Epiteliales , Klebsiella pneumoniae , Infecciones Urinarias , Escherichia coli Uropatógena , Humanos , Infecciones Urinarias/microbiología , Enterococcus faecalis/fisiología , Células Epiteliales/microbiología , Escherichia coli Uropatógena/fisiología , Klebsiella pneumoniae/fisiología , Vejiga Urinaria/microbiología , Vejiga Urinaria/citología , Coinfección/microbiología , Línea Celular , Interacciones Huésped-Patógeno
4.
PLoS Pathog ; 19(5): e1011388, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37167325

RESUMEN

There is a growing consensus that a significant proportion of recurrent urinary tract infections are linked to the persistence of uropathogens within the urinary tract and their re-emergence upon the conclusion of antibiotic treatment. Studies in mice and human have revealed that uropathogenic Escherichia coli (UPEC) can persist in bladder epithelial cells (BECs) even after the apparent resolution of the infection. Here, we found that, following the entry of UPEC into RAB27b+ fusiform vesicles in BECs, some bacteria escaped into the cytoplasmic compartment via a mechanism involving hemolysin A (HlyA). However, these UPEC were immediately recaptured within LC3A/B+ autophagosomes that matured into LAMP1+ autolysosomes. Thereafter, HlyA+ UPEC-containing lysosomes failed to acidify, which is an essential step for bacterial elimination. This lack of acidification was related to the inability of bacteria-harboring compartments to recruit V-ATPase proton pumps, which was attributed to the defragmentation of cytosolic microtubules by HlyA. The persistence of UPEC within LAMP1+ compartments in BECs appears to be directly linked to HlyA. Thus, through intravesicular instillation of microtubule stabilizer, this host defense response can be co-opted to reduce intracellular bacterial burden following UTIs in the bladder potentially preventing recurrence.


Asunto(s)
Infecciones por Escherichia coli , Infecciones Urinarias , Escherichia coli Uropatógena , Animales , Ratones , Humanos , Vejiga Urinaria/microbiología , Escherichia coli Uropatógena/fisiología , Proteínas Hemolisinas , Infecciones por Escherichia coli/microbiología , Infecciones Urinarias/microbiología , Células Epiteliales/microbiología , Lisosomas/patología , Concentración de Iones de Hidrógeno
5.
Elife ; 112022 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-35881547

RESUMEN

A key attribute of persistent or recurring bacterial infections is the ability of the pathogen to evade the host's immune response. Many Enterobacteriaceae express type 1 pili, a pre-adapted virulence trait, to invade host epithelial cells and establish persistent infections. However, the molecular mechanisms and strategies by which bacteria actively circumvent the immune response of the host remain poorly understood. Here, we identified CD14, the major co-receptor for lipopolysaccharide detection, on mouse dendritic cells (DCs) as a binding partner of FimH, the protein located at the tip of the type 1 pilus of Escherichia coli. The FimH amino acids involved in CD14 binding are highly conserved across pathogenic and non-pathogenic strains. Binding of the pathogenic strain CFT073 to CD14 reduced DC migration by overactivation of integrins and blunted expression of co-stimulatory molecules by overactivating the NFAT (nuclear factor of activated T-cells) pathway, both rate-limiting factors of T cell activation. This response was binary at the single-cell level, but averaged in larger populations exposed to both piliated and non-piliated pathogens, presumably via the exchange of immunomodulatory cytokines. While defining an active molecular mechanism of immune evasion by pathogens, the interaction between FimH and CD14 represents a potential target to interfere with persistent and recurrent infections, such as urinary tract infections or Crohn's disease.


Asunto(s)
Infecciones por Escherichia coli , Escherichia coli Uropatógena , Adhesinas de Escherichia coli/química , Adhesinas de Escherichia coli/genética , Adhesinas de Escherichia coli/metabolismo , Animales , Infecciones por Escherichia coli/microbiología , Proteínas Fimbrias/metabolismo , Fimbrias Bacterianas/metabolismo , Inmunidad , Ratones , Escherichia coli Uropatógena/fisiología
6.
Cell Rep ; 39(4): 110758, 2022 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-35476986

RESUMEN

Urinary tract infections (UTIs) are a cause for alarm given the high rates of treatment failure. In a recent issue of Cell Reports, Pang et al. uncovered dueling molecular machinery at the host-pathogen interface in response to phosphate that points to new anti-infective strategies against UTIs.


Asunto(s)
Infecciones por Escherichia coli , Infecciones Urinarias , Escherichia coli Uropatógena , Femenino , Humanos , Masculino , Escherichia coli Uropatógena/fisiología , Urotelio
7.
Sci Rep ; 12(1): 486, 2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-35017565

RESUMEN

Uropathogenic Escherichia coli (UPEC) may undergo a cyclic cascade of morphological alterations that are believed to enhance the potential of UPEC to evade host responses and re-infect host cell. However, knowledge on the pathogenic potential and host activation properties of UPEC during the morphological switch is limited. Microarray analysis was performed on mRNA isolated from human bladder epithelial cells (HBEP) after exposure to three different morphological states of UPEC (normal coliform, filamentous form and reverted form). Cells stimulated with filamentous bacteria showed the lowest number of significant gene alterations, although the number of enriched gene ontology classes was high suggesting diverse effects on many different classes of host genes. The normal coliform was in general superior in stimulating transcriptional activity in HBEP cells compared to the filamentous and reverted form. Top-scored gene entities activated by all three morphological states included IL17C, TNFAIP6, TNF, IL20, CXCL2, CXCL3, IL6 and CXCL8. The number of significantly changed canonical pathways was lower in HBEP cells stimulated with the reverted form (32 pathways), than in cells stimulated with the coliform (83 pathways) or filamentous bacteria (138 pathways). A host cell invasion assay showed that filamentous bacteria were unable to invade bladder cells, and that the number of intracellular bacteria was markedly lower in cells infected with the reverted form compared to the coliform. In conclusion, the morphological state of UPEC has major impact on the host bladder response both when evaluating the number and the identity of altered host genes and pathways.


Asunto(s)
Células Epiteliales/metabolismo , Infecciones por Escherichia coli/genética , Transcripción Genética , Vejiga Urinaria/microbiología , Escherichia coli Uropatógena/fisiología , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Línea Celular , Quimiocina CXCL2/genética , Quimiocina CXCL2/metabolismo , Quimiocinas CXC/genética , Quimiocinas CXC/metabolismo , Células Epiteliales/microbiología , Infecciones por Escherichia coli/metabolismo , Infecciones por Escherichia coli/microbiología , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Vejiga Urinaria/metabolismo , Escherichia coli Uropatógena/crecimiento & desarrollo
8.
Appl Biochem Biotechnol ; 194(1): 504-516, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34618338

RESUMEN

Nanotechnology involves the synthesis of nanoparticles that have been used in the therapeutic application for treating diseases. In this present study, we have adopted the synthesis of myconanoparticles from the extracellular extract of endophytic fungi Penicillium sclerotiorum (PsNps) and validated its antibacterial potential against antibiotic-resistant uropathogenic E. coli and ATCC (25,922) strain of Escherichia coli. Endophytic fungi were isolated from the healthy leaves of Tamarindus indica. The genomic DNA from endophytic fungi was isolated and the ITS region was amplified by polymerase chain reaction (PCR) using universal fungal primers ITS1 and ITS4 and sequenced for the identification of endophytic fungal isolates. Penicillium sclerotiorum extract was used for the synthesis of silver nanoparticles (PsNps) and was characterized by UV-vis spectroscopy, Fourier transform infrared spectroscopy (FTIR), zeta potential, FE-SEM, and Energy dispersive X-ray analysis (EDAX). Antibacterial activity of PsNps was tested against the antibiotic-resistant uropathogenic E. coli and ATCC (25,922) strain of E. coli. Further experiments were carried out to explore the potential of PsNps in regulating the CTX-M-15 gene. The antimicrobial activity showed that the PsNps inhibited growth, biofilm formation in both the strains of E. coli. The expression of the gene encoding CTX-M-15 was downregulated in a resistant strain of uropathogenic E. coli. Our results suggest that the PsNps could be used as an alternative source for antibiotics. Thus, further studies can be conducted to prove the in vivo potential of PsNps and can be formulated for commercialization.


Asunto(s)
Biopelículas/efectos de los fármacos , Nanopartículas/química , Penicillium/química , Plata , Escherichia coli Uropatógena/fisiología , Biopelículas/crecimiento & desarrollo , Plata/química , Plata/farmacología
9.
Cell Rep ; 36(3): 109351, 2021 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-34289360

RESUMEN

Recurrence of uropathogenic Escherichia coli (UPEC) infections has been attributed to reactivation of quiescent intracellular reservoirs (QIRs) in deep layers of the bladder wall. QIRs are thought to arise late during infection following dispersal of bacteria from intracellular bacterial communities (IBCs) in superficial umbrella cells. Here, we track the formation of QIR-like bacteria in a bladder organoid model that recapitulates the stratified uroepithelium within a volume suitable for high-resolution live-cell imaging. Bacteria injected into the organoid lumen enter umbrella-like cells and proliferate to form IBC-like bodies. In parallel, single bacteria penetrate deeper layers of the organoid wall, where they localize within or between uroepithelial cells. These "solitary" bacteria evade killing by antibiotics and neutrophils and are morphologically distinct from bacteria in IBCs. We conclude that bacteria with QIR-like properties may arise at early stages of infection, independent of IBC formation and rupture.


Asunto(s)
Antibacterianos/farmacología , Modelos Biológicos , Neutrófilos/patología , Organoides/microbiología , Vejiga Urinaria/microbiología , Escherichia coli Uropatógena/fisiología , Animales , Diferenciación Celular/efectos de los fármacos , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/patología , Femenino , Humanos , Imagenología Tridimensional , Ratones Endogámicos C57BL , Viabilidad Microbiana/efectos de los fármacos , Movimiento , Neutrófilos/efectos de los fármacos , Organoides/efectos de los fármacos , Organoides/ultraestructura , Vejiga Urinaria/patología , Escherichia coli Uropatógena/efectos de los fármacos , Escherichia coli Uropatógena/crecimiento & desarrollo , Escherichia coli Uropatógena/ultraestructura
10.
PLoS Pathog ; 17(5): e1009617, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-34043736

RESUMEN

Urinary tract infections (UTIs) are a common bacterial infectious disease in humans, and strains of uropathogenic Escherichia coli (UPEC) are the most frequent cause of UTIs. During infection, UPEC must cope with a variety of stressful conditions in the urinary tract. Here, we demonstrate that the small RNA (sRNA) RyfA of UPEC strains is required for resistance to oxidative and osmotic stresses. Transcriptomic analysis of the ryfA mutant showed changes in expression of genes associated with general stress responses, metabolism, biofilm formation and genes coding for cell surface proteins. Inactivation of ryfA in UPEC strain CFT073 decreased urinary tract colonization in mice and the ryfA mutant also had reduced production of type 1 and P fimbriae (pili), adhesins which are known to be important for UTI. Furthermore, loss of ryfA also reduced UPEC survival in human macrophages. Thus, ryfA plays a key regulatory role in UPEC adaptation to stress, which contributes to UTI and survival in macrophages.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Infecciones por Escherichia coli/microbiología , ARN Pequeño no Traducido/genética , Infecciones Urinarias/microbiología , Escherichia coli Uropatógena/genética , Adaptación Fisiológica , Adhesinas Bacterianas/genética , Adhesinas Bacterianas/metabolismo , Animales , Fimbrias Bacterianas/metabolismo , Perfilación de la Expresión Génica , Humanos , Macrófagos/microbiología , Ratones , Osmorregulación , Estrés Oxidativo , ARN Bacteriano/genética , Eliminación de Secuencia , Escherichia coli Uropatógena/crecimiento & desarrollo , Escherichia coli Uropatógena/fisiología , Virulencia
11.
PLoS Pathog ; 17(5): e1009553, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-34015044

RESUMEN

Bacterial infection results in a veritable cascade of host responses, both local and systemic. To study the initial stages of host-pathogen interaction in living tissue we use spatially-temporally controlled in vivo models. Using this approach, we show here that within 4 h of a uropathogenic Escherichia coli (UPEC) infection in the kidney, an IFNγ response is triggered in the spleen. This rapid infection-mediated inter-organ communication was found to be transmitted via nerve signalling. Bacterial expression of the toxin α-hemolysin directly and indirectly activated sensory neurons, which were identified in the basement membrane of renal tubules. Nerve activation was transmitted via the splenic nerve, inducing upregulation of IFNγ in the marginal zones of the spleen that led to increasing concentrations of IFNγ in the circulation. We found that IFNγ modulated the inflammatory signalling generated by renal epithelia cells in response to UPEC infection. This demonstrates a new concept in the host response to kidney infection; the role of nerves in sensing infection and rapidly triggering a systemic response which can modulate inflammation at the site of infection. The interplay between the nervous and immune systems is an exciting, developing field with the appealing prospect of non-pharmaceutical interventions. Our study identifies an important role for systemic neuro-immune communication in modulating inflammation during the very first hours of a local bacterial infection in vivo.


Asunto(s)
Infecciones por Escherichia coli/complicaciones , Interacciones Huésped-Patógeno , Inflamación/patología , Interferón gamma/metabolismo , Riñón/microbiología , Neuroinmunomodulación , Bazo/metabolismo , Animales , Células Epiteliales/microbiología , Inflamación/etiología , Inflamación/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Escherichia coli Uropatógena/fisiología
12.
NPJ Biofilms Microbiomes ; 7(1): 35, 2021 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-33863914

RESUMEN

Nutrient gradients in biofilms cause bacteria to organize into metabolically versatile communities capable of withstanding threats from external agents including bacteriophages, phagocytes, and antibiotics. We previously determined that oxygen availability spatially organizes respiration in uropathogenic Escherichia coli biofilms, and that the high-affinity respiratory quinol oxidase cytochrome bd is necessary for extracellular matrix production and biofilm development. In this study we investigate the physiologic consequences of cytochrome bd deficiency in biofilms and determine that loss of cytochrome bd induces a biofilm-specific increase in expression of general diffusion porins, leading to elevated outer membrane permeability. In addition, loss of cytochrome bd impedes the proton mediated efflux of noxious chemicals by diminishing respiratory flux. As a result, loss of cytochrome bd enhances cellular accumulation of noxious chemicals and increases biofilm susceptibility to antibiotics. These results identify an undescribed link between E. coli biofilm respiration and stress tolerance, while suggesting the possibility of inhibiting cytochrome bd as an antibiofilm therapeutic approach.


Asunto(s)
Antibacterianos/farmacología , Biopelículas/efectos de los fármacos , Grupo Citocromo b/genética , Farmacorresistencia Bacteriana , Proteínas del Complejo de Cadena de Transporte de Electrón/genética , Proteínas de Escherichia coli/genética , Oxidorreductasas/genética , Escherichia coli Uropatógena/fisiología , Alelos , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo , Biopelículas/crecimiento & desarrollo , Grupo Citocromo b/metabolismo , Relación Dosis-Respuesta a Droga , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Proteínas de Escherichia coli/metabolismo , Técnicas de Silenciamiento del Gen , Pruebas de Sensibilidad Microbiana , Oxidorreductasas/metabolismo , Plancton/efectos de los fármacos , Plancton/genética , Escherichia coli Uropatógena/efectos de los fármacos
13.
Immunology ; 164(1): 3-14, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33763853

RESUMEN

Urinary tract infections (UTI) are among the most prevalent infectious diseases and the most common cause of nosocomial infections, worldwide. Uropathogenic E. coli (UPEC) are responsible for approximately 80% of all UTI, which most commonly affect the bladder. UPEC colonize the urinary tract by ascension of the urethra, followed by cell invasion, and proliferation inside and outside urothelial cells, thereby causing symptomatic infections and quiescent intracellular reservoirs that may lead to recurrence. Sugars, or glycans, are key molecules for host-pathogen interactions, and UTI are no exception. Surface glycans regulate many of the events associated with UPEC adhesion and infection, as well as induction of the host immune response. While the bacterial protein FimH binds mannose-containing host glycoproteins to initiate infection and UPEC-secreted polysaccharides block immune mechanisms to favour intracellular replication, host glycans on the urothelial surface and on secreted glycoproteins prevent or limit infection by inhibiting UPEC adhesion. Given the importance of glycans during UTI, here we review the glycobiology of UPEC infection to highlight fundamental sugar-mediated processes of immunological interest for their potential clinical applications. Interdisciplinary approaches incorporating glycomics and infection biology may help to develop novel non-antibiotic-based therapeutic strategies for bacterial infections as the spread of antimicrobial-resistant uropathogens is currently threatening modern healthcare systems.


Asunto(s)
Polisacáridos/metabolismo , Sistema Urinario/inmunología , Escherichia coli Uropatógena/fisiología , Animales , Infecciones por Escherichia coli , Glicómica , Interacciones Huésped-Patógeno , Humanos , Polisacáridos/inmunología , Infecciones Urinarias , Virulencia
14.
J Ethnopharmacol ; 274: 114053, 2021 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-33746003

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Phaseaoli pericarpium (bean pods) is a pharmacopeial plant material traditionally used as a diuretic and antidiabetic agents. Diuretic activity of pod extracts was reported first in 1608. Since then Phaseoli pericarpium tea figures in many textbooks as medicinal plant material used by patients. AIM OF THE STUDY: Despite the traditional use of extracts from Phaseolium vulgaris pericarp, limited information is available on bioactivity, chemical composition, and bioavailability of such preparations. The following study aimed to investigate the phytochemical composition, the in vitro permeability of selected extract's constituents over the Caco-2 permeation system, and potential antivirulence activity against uropathogenic Escherichia coli of a hydroalcoholic Phaseoli pericarpium extract (PPX) in vitro to support its traditional use as a remedy used in urinary tract infections. MATERIAL AND METHODS: The chemical composition of the extract PPX [ethanol:water 7:3 (v/v)] investigated by using UHPLC-DAD-MSn and subsequent dereplication. The permeability of compounds present in PPX was evaluated using the Caco-2 monolayer permeation system. The influence of PPX on uropathogenic E. coli (UPEC) strain NU14 proliferation and against the bacterial adhesion to T24 epithelial cells was determined by turbidimetric assay and flow cytometry, respectively. The influence of the extract on the mitochondrial activity of T24 host cells was monitored by MTT assay. RESULTS: LC-MSn investigation and dereplication, indicated PPX extract to be dominated by a variety of flavonoids, with rutin as a major compound, and soyasaponin derivatives. Rutin, selected soyasaponins and fatty acids were shown to permeate the Caco-2 monolayer system, indicating potential bioavailability following oral intake. The extract did not influence the viability of T24 cells after 1.5h incubation at 2 mg/mL and UPEC. PPX significantly reduced the bacterial adhesion of UPEC to human bladder cells in a concentration-dependent manner (0.5-2 mg/mL). Detailed investigations by different incubation protocols indicated that PPX seems to interact with T24 cells, which subsequently leads to reduced recognition and adhesion of UPEC to the host cell membrane. CONCLUSIONS: PPX is characterised by the presence of flavonoids (e.g. rutin) and saponins, from which selected compounds might be bioavailable after oral application, as indicated by the Caco-2 permeation experiments. Rutin and some saponins can be considered as potentially bioavailable after the oral intake. The concentration-dependent inhibition of bacterial adhesion of UPEC to T24 cells justifies the traditional use of Phaseoli pericarpium in the prevention and treatment of urinary tract infections.


Asunto(s)
Adhesión Bacteriana/efectos de los fármacos , Phaseolus , Extractos Vegetales/farmacología , Escherichia coli Uropatógena/efectos de los fármacos , Línea Celular , Células Epiteliales/metabolismo , Etanol/química , Flavonoides/análisis , Flavonoides/farmacología , Humanos , Permeabilidad/efectos de los fármacos , Fitoquímicos/análisis , Fitoquímicos/farmacología , Extractos Vegetales/química , Saponinas/análisis , Saponinas/farmacología , Semillas/química , Solventes/química , Escherichia coli Uropatógena/fisiología , Agua/química
15.
Front Immunol ; 12: 582858, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33679734

RESUMEN

The structural and functional destruction of the blood-testis barrier (BTB) following uropathogenic E. coli (UPEC) infection may be a critical component of the pathologic progress of orchitis. Recent findings indicate that the mammalian target of the rapamycin (mTOR)-signaling pathway is implicated in the regulation of BTB assembly and restructuring. To explore the mechanisms underlying BTB damage induced by UPEC infection, we analyzed BTB integrity and the involvement of the mTOR-signaling pathway using in vivo and in vitro UPEC-infection models. We initially confirmed that soluble virulent factors secreted from UPEC trigger a stress response in Sertoli cells and disturb adjacent cell junctions via down-regulation of junctional proteins, including occludin, zonula occludens-1 (ZO-1), F-actin, connexin-43 (CX-43), ß-catenin, and N-cadherin. The BTB was ultimately disrupted in UPEC-infected rat testes, and blood samples from UPEC-induced orchitis in these animals were positive for anti-sperm antibodies. Furthermore, we herein also demonstrated that mTOR complex 1 (mTORC1) over-activation and mTORC2 suppression contributed to the disturbance in the balance between BTB "opening" and "closing." More importantly, rapamycin (a specific mTORC1 inhibitor) significantly restored the expression of cell-junction proteins and exerted a protective effect on the BTB during UPEC infection. We further confirmed that short-term treatment with rapamycin did not aggravate spermatogenic degeneration in infected rats. Collectively, this study showed an association between abnormal activation of the mTOR-signaling pathway and BTB impairment during UPEC-induced orchitis, which may provide new insights into a potential treatment strategy for testicular infection.


Asunto(s)
Barrera Hematotesticular/inmunología , Infecciones por Escherichia coli/inmunología , Diana Mecanicista del Complejo 1 de la Rapamicina/inmunología , Diana Mecanicista del Complejo 2 de la Rapamicina/inmunología , Infecciones Urinarias/inmunología , Escherichia coli Uropatógena/inmunología , Animales , Barrera Hematotesticular/metabolismo , Células Cultivadas , Infecciones por Escherichia coli/metabolismo , Infecciones por Escherichia coli/microbiología , Humanos , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Orquitis/inmunología , Orquitis/metabolismo , Orquitis/microbiología , Ratas Sprague-Dawley , Células de Sertoli/inmunología , Células de Sertoli/metabolismo , Células de Sertoli/microbiología , Espermatogénesis/inmunología , Testículo/inmunología , Testículo/metabolismo , Proteínas de Uniones Estrechas/inmunología , Proteínas de Uniones Estrechas/metabolismo , Infecciones Urinarias/metabolismo , Infecciones Urinarias/microbiología , Escherichia coli Uropatógena/fisiología
16.
Microbiol Res ; 246: 126709, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33578264

RESUMEN

Uropathogenic Escherichia coli (UPEC) is a major cause of urinary tract infections (UTI). UPEC persister bacteria play crucial roles in clinical treatment failure and relapse. Although DNA methylation is known to regulate gene expression, its role in persister formation has not been investigated. Here, we show that Δdam (adenine methylase) mutant from UPEC strain UTI89 had significant defect in persister formation and complementation of the Δdam mutant restored this defect. Using PacBio sequencing of methylome and RNA sequencing of Δdam, we defined, for the first time, the role of Dam in persister formation. We found that Δdam mutation had an overwhelming effect on demethylation of the genome and the demethylation sites affected expression of genes involved in broad transcriptional and metabolic processes. Using comparative COG analysis of methylome and transcriptome, we demonstrate that Dam mediates persister formation through transcriptional control, cell motility, DNA repair and metabolite transport processes. These findings provide the first evidence and molecular basis for DNA methylation mediated persister formation and implicate Dam DNA methylation as a potential drug target for persister bacteria.


Asunto(s)
Adaptación Fisiológica , Adenina/metabolismo , Metilación de ADN , Metiltransferasas/genética , Escherichia coli Uropatógena/fisiología , Reparación del ADN , ADN Bacteriano , Epigenoma , Infecciones por Escherichia coli/microbiología , Proteínas de Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica , Humanos , Mutación , Análisis de Secuencia de ARN , Estrés Fisiológico , Transcriptoma , Infecciones Urinarias/microbiología , Virulencia
17.
Infect Immun ; 89(4)2021 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-33468577

RESUMEN

Uropathogenic Escherichia coli (UPEC), the primary etiologic agent of urinary tract infections (UTIs), encounters a restrictive population bottleneck within the female mammalian bladder. Its genetic diversity is restricted during establishment of cystitis because successful UPEC must invade superficial bladder epithelial cells prior to forming clonal intracellular bacterial communities (IBCs). In this study, we aimed to understand UPEC population dynamics during ascending pyelonephritis, namely, formation of kidney bacterial communities (KBCs) in the renal tubular lumen and nucleation of renal abscesses. We inoculated the bladders of both male and female C3H/HeN mice, a background which features vesicoureteral reflux; we have previously shown that in this model, males develop severe, high-titer pyelonephritis and renal abscesses much more frequently than females. Mice were infected with 40 isogenic, PCR-tagged ("barcoded") UPEC strains, and tags remaining in bladder and kidneys were ascertained at intervals following infection. In contrast to females, males maintained a majority of strains within both the bladder and kidneys throughout the course of infection, indicating only a modest host-imposed bottleneck on overall population diversity during successful renal infection. Moreover, the diverse population in the infected male kidneys obscured any restrictive bottleneck in the male bladder. Finally, using RNA in situ hybridization following mixed infections with isogenic UPEC bearing distinct markers, we found that despite their extracellular location (in the urinary space), KBCs are clonal in origin. This finding indicates that even with bulk reflux of infected bladder urine into the renal pelvis, successful ascension of UPEC to establish the tubular niche is an uncommon event.


Asunto(s)
Infecciones por Escherichia coli/microbiología , Nefritis/microbiología , Infecciones Urinarias/microbiología , Escherichia coli Uropatógena/fisiología , Animales , Modelos Animales de Enfermedad , Femenino , Regulación Bacteriana de la Expresión Génica , Humanos , Masculino , Ratones , Dinámica Poblacional , Factores Sexuales
18.
Immunobiology ; 226(1): 152020, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33246308

RESUMEN

The bladder epithelial cells elicit robust innate immune responses against urinary tract infections (UTIs) for preventing the bacterial colonization. Physiological fluctuations in circulating estrogen levels in women increase the susceptibility to UTI pathogenesis, often resulting in adverse health outcomes. Dr adhesin bearing Escherichia coli (Dr E. coli) cause recurrent UTIs in menopausal women and acute pyelonephritis in pregnant women. Dr E. coli bind to epithelial cells via host innate immune receptor CD55, under hormonal influence. The role of estrogens or estrogen receptors (ERs) in regulating the innate immune responses in the bladder are poorly understood. In the current study, we investigated the role of ERα, ERß and GPR30 in modulating the innate immune responses against Dr E. coli induced UTI using human bladder epithelial carcinoma 5637 cells (HBEC). Both ERα and ERß agonist treatment in bladder cells induced a protection against Dr E. coli invasion via upregulation of TNFα and downregulation of CD55 and IL10, and these effects were reversed by action of ERα and ERß antagoinsts. In contrast, the agonist-mediated activation of GPR30 led to an increased bacterial colonization due to suppression of innate immune factors in the bladder cells, and these effects were reversed by the antagonist-mediated suppression of GPR30. Further, siRNA-mediated ERα knockdown in the bladder cells reversed the protection against bacterial invasion observed in the ERα positive bladder cells, by modulating the gene expression of TNFα, CD55 and IL10, thus confirming the protective role of ERα. We demonstrate for the first time a protective role of nuclear ERs, ERα and ERß but not of membrane ER, GPR30 against Dr E. coli invasion in HBEC 5637 cells. These findings have many clinical implications and suggest that ERs may serve as potential drug targets towards developing novel therapeutics for regulating local innate immunity and treating UTIs.


Asunto(s)
Células Epiteliales/inmunología , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/metabolismo , Neoplasias de la Vejiga Urinaria/inmunología , Vejiga Urinaria/inmunología , Vejiga Urinaria/patología , Infecciones Urinarias/metabolismo , Escherichia coli Uropatógena/fisiología , Adhesinas de Escherichia coli/metabolismo , Animales , Proliferación Celular , Células Cultivadas , Susceptibilidad a Enfermedades , Receptor alfa de Estrógeno/genética , Receptor beta de Estrógeno/genética , Estrógenos/metabolismo , Femenino , Humanos , Inmunidad Innata , Menopausia , Ratones , Terapia Molecular Dirigida , Embarazo , ARN Interferente Pequeño/genética , Receptores de Estrógenos , Receptores Acoplados a Proteínas G
19.
Libyan J Med ; 16(1): 1845444, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33170767

RESUMEN

Biofilm is an important virulent marker attributed to the development of urinary tract infections (UTIs) by uropathogenic E. coli (UPEC). Drug-resistant and biofilm-producing UPEC are highly problematic causing catheter-associated or recurrent UTIs with significant morbidity and mortality. The aim of the current study was to investigate the prevalence of biofilm formation and phylogenetic groups in drug-resistant UPEC to predict their ability to cause disease. This prospective study was conducted at the Department of Microbiology, University of Karachi from January to June 2019. A total of 50 highly drug-resistant UPEC were selected for this study. UPEC isolates were screened to form biofilm by Congo-red agar (CRA) and microtiter plate (MTP) technique. The representative biofilm-producing isolates were analysed by scanning electron microscopy (SEM) monitoring. Phylogenetic analysis was done by PCR method based on two preserved genes; chuA, yjaA and TspE4-C2 DNA fragment. On CRA 34 (68%) UPEC were slime producers, while on MTP 20 (40%) were strong biofilm producers, 19 (38%) moderate and 11 (22%) were low to negligible biofilm producers. Molecular typing confirmed that phylogenetic group B2 was prevalent in drug resistant UPEC strains. Pathogenic strains belonged to phylogenetic group B2 and D were found to have greater biofilm forming ability as compare to non-pathogenic commensal strains that belonged to phylogenetic group A. Our results indicate that biofilm formation vary in drug resistant UPEC belonged to different phylogenetic groups. This study indicates possible link between in vitro biofilm formation and phylogenetic groups of UPEC, therefore this knowledge might be helpful to predict the pathogenic potential of UPEC and help design strategies for controlling UTIs.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Farmacorresistencia Bacteriana/genética , Infecciones por Escherichia coli/microbiología , Infecciones Urinarias/microbiología , Escherichia coli Uropatógena/fisiología , Humanos , Filogenia , Estudios Prospectivos , Escherichia coli Uropatógena/genética
20.
J Vis Exp ; (166)2020 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-33346201

RESUMEN

Recurrent urinary tract infections (rUTI) caused by uropathogenic Escherichia coli (UPEC) are common and costly. Previous articles describing models of UTI in male and female mice have illustrated the procedures for bacterial inoculation and enumeration in urine and tissues. During an initial bladder infection in C57BL/6 mice, UPEC establish latent reservoirs inside bladder epithelial cells that persist following clearance of UPEC bacteriuria. This model builds on these studies to examine rUTI caused by the emergence of UPEC from within latent bladder reservoirs. The urogenital bacterium Gardnerella vaginalis is used as the trigger of rUTI in this model because it is frequently present in the urogenital tracts of women, especially in the context of vaginal dysbiosis that has been associated with UTI. In addition, a method for in situ bladder fixation followed by scanning electron microscopy (SEM) analysis of bladder tissue is also described, with potential application to other studies involving the bladder.


Asunto(s)
Infecciones por Escherichia coli/microbiología , Gardnerella vaginalis/fisiología , Vejiga Urinaria/microbiología , Vejiga Urinaria/patología , Infecciones Urinarias/microbiología , Animales , Modelos Animales de Enfermedad , Reservorios de Enfermedades/microbiología , Infecciones por Escherichia coli/patología , Infecciones por Escherichia coli/orina , Femenino , Ratones Endogámicos C57BL , Recurrencia , Espectrofotometría , Vejiga Urinaria/ultraestructura , Infecciones Urinarias/patología , Infecciones Urinarias/orina , Orina/citología , Escherichia coli Uropatógena/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...