Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
1.
J Med Chem ; 67(5): 3668-3678, 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38308631

RESUMEN

FmlH, a bacterial adhesin of uropathogenic Escherichia coli (UPEC), has been shown to provide a fitness advantage in colonizing the bladder during chronic urinary tract infections (UTIs). Previously reported ortho-biphenyl glycosides based on ßGal and ßGalNAc have excellent binding affinity to FmlH and potently block binding to its natural carbohydrate receptor, but they lack oral bioavailability. In this paper, we outline studies where we have optimized compounds for improved pharmacokinetics, leading to the discovery of novel analogues with good oral bioavailability. We synthesized galactosides with the anomeric O-linker replaced with more stable S- and C-linked linkers. We also investigated modifications to the GalNAc sugar and modifications to the biphenyl aglycone. We identified GalNAc 69 with an IC50 of 0.19 µM against FmlH and 53% oral bioavailability in mice. We also obtained a FimlH-bound X-ray structure of lead compound 69 (AM4085) which has potential as a new antivirulence therapeutic for UTIs.


Asunto(s)
Infecciones por Escherichia coli , Infecciones Urinarias , Escherichia coli Uropatógena , Ratones , Animales , Lectinas , Adhesinas de Escherichia coli/química , Infecciones Urinarias/tratamiento farmacológico , Compuestos de Bifenilo/química , Escherichia coli Uropatógena/metabolismo , Infecciones por Escherichia coli/tratamiento farmacológico
2.
bioRxiv ; 2024 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-38260261

RESUMEN

We have developed GmPcides from a peptidomimetic dihydrothiazolo ring-fused 2-pyridone scaffold that have antimicrobial activities against a broad-spectrum of Gram-positive pathogens. Here we examine the treatment efficacy of GmPcides using skin and soft tissue infection (SSTI) and biofilm formation models by Streptococcus pyogenes. Screening our compound library for minimal inhibitory (MIC) and minimal bactericidal (MBC) concentrations identified GmPcide PS757 as highly active against S. pyogenes. Treatment of S. pyogenes biofilm with PS757 revealed robust efficacy against all phases of biofilm formation by preventing initial biofilm development, ceasing biofilm maturation and eradicating mature biofilm. In a murine model of S. pyogenes SSTI, subcutaneous delivery of PS757 resulted in reduced levels of tissue damage, decreased bacterial burdens and accelerated rates of wound-healing, which were associated with down-regulation of key virulence factors, including M protein and the SpeB cysteine protease. These data demonstrate that GmPcides show considerable promise for treating S. pyogenes infections.

3.
Nat Commun ; 15(1): 61, 2024 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-38168042

RESUMEN

Catheter-associated urinary tract infections (CAUTIs), a common cause of healthcare-associated infections, are caused by a diverse array of pathogens that are increasingly becoming antibiotic resistant. We analyze the microbial occurrences in catheter and urine samples from 55 human long-term catheterized patients collected over one year. Although most of these patients were prescribed antibiotics over several collection periods, their catheter samples remain colonized by one or more bacterial species. Examination of a total of 366 catheter and urine samples identify 13 positive and 13 negative genus co-occurrences over 12 collection periods, representing associations that occur more or less frequently than expected by chance. We find that for many patients, the microbial species composition between collection periods is similar. In a subset of patients, we find that the most frequently sampled bacteria, Escherichia coli and Enterococcus faecalis, co-localize on catheter samples. Further, co-culture of paired isolates recovered from the same patients reveals that E. coli significantly augments E. faecalis growth in an artificial urine medium, where E. faecalis monoculture grows poorly. These findings suggest novel strategies to collapse polymicrobial CAUTI in long-term catheterized patients by targeting mechanisms that promote positive co-associations.


Asunto(s)
Infecciones Relacionadas con Catéteres , Infecciones Urinarias , Humanos , Escherichia coli , Infecciones Relacionadas con Catéteres/microbiología , Catéteres , Infecciones Urinarias/microbiología , Enterococcus faecalis , Bacterias
4.
Res Sq ; 2023 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-37609304

RESUMEN

Catheter-associated urinary tract infections (CAUTIs) contribute greatly to the burden of healthcare associated infections. Acinetobacter baumannii is a Gram-negative bacterium with high levels of antibiotic resistance that is of increasing concern as a CAUTI pathogen. A. baumannii expresses fibrinogen-binding adhesins (Abp1D and Abp2D) that mediate colonization and biofilm formation on catheters, which become coated with fibrinogen upon insertion. We developed a protein subunit vaccine against Abp1DRBD and Abp2DRBD and showed that vaccination significantly reduced bladder bacterial titers in a mouse model of CAUTI. We then determined that immunity to Abp2DRBD alone was sufficient for protection. Mechanistically, we defined the B cell response to Abp2DRBD vaccination and demonstrated that immunity was transferrable to naïve mice through passive immunization with Abp2DRBD-immune sera. This work represents a novel strategy in the prevention of A. baumannii CAUTI and has an important role to play in the global fight against antimicrobial resistance.

5.
Proc Natl Acad Sci U S A ; 120(4): e2212694120, 2023 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-36652481

RESUMEN

Multidrug-resistant Acinetobacter baumannii infections are an urgent clinical problem and can cause difficult-to-treat nosocomial infections. During such infections, like catheter-associated urinary tract infections (CAUTI), A. baumannii rely on adhesive, extracellular fibers, called chaperone-usher pathway (CUP) pili for critical binding interactions. The A. baumannii uropathogenic strain, UPAB1, and the pan-European subclone II isolate, ACICU, use the CUP pili Abp1 and Abp2 (previously termed Cup and Prp, respectively) in tandem to establish CAUTIs, specifically to facilitate bacterial adherence and biofilm formation on the implanted catheter. Abp1 and Abp2 pili are tipped with two domain tip adhesins, Abp1D and Abp2D, respectively. We discovered that both adhesins bind fibrinogen, a critical host wound response protein that is released into the bladder upon catheterization and is subsequently deposited on the catheter. The crystal structures of the Abp1D and Abp2D receptor-binding domains were determined and revealed that they both contain a large, distally oriented pocket, which mediates binding to fibrinogen and other glycoproteins. Genetic, biochemical, and biophysical studies revealed that interactions with host proteins are governed by several critical residues in and along the edge of the binding pocket, one of which regulates the structural stability of an anterior loop motif. K34, located outside of the pocket but interacting with the anterior loop, also regulates the binding affinity of the protein. This study illuminates the mechanistic basis of the critical fibrinogen-coated catheter colonization step in A. baumannii CAUTI pathogenesis.


Asunto(s)
Acinetobacter baumannii , Infecciones Urinarias , Humanos , Adhesinas Bacterianas/genética , Adhesinas Bacterianas/metabolismo , Infecciones Urinarias/microbiología , Catéteres , Acinetobacter baumannii/genética , Fibrinógeno/metabolismo
6.
Proc Natl Acad Sci U S A ; 119(43): e2210912119, 2022 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-36252016

RESUMEN

The alarming rise of multidrug-resistant Gram-positive bacteria has precipitated a healthcare crisis, necessitating the development of new antimicrobial therapies. Here we describe a new class of antibiotics based on a ring-fused 2-pyridone backbone, which are active against vancomycin-resistant enterococci (VRE), a serious threat as classified by the Centers for Disease Control and Prevention, and other multidrug-resistant Gram-positive bacteria. Ring-fused 2-pyridone antibiotics have bacteriostatic activity against actively dividing exponential phase enterococcal cells and bactericidal activity against nondividing stationary phase enterococcal cells. The molecular mechanism of drug-induced killing of stationary phase cells mimics aspects of fratricide observed in enterococcal biofilms, where both are mediated by the Atn autolysin and the GelE protease. In addition, combinations of sublethal concentrations of ring-fused 2-pyridones and standard-of-care antibiotics, such as vancomycin, were found to synergize to kill clinical strains of VRE. Furthermore, a broad range of antibiotic resistant Gram-positive pathogens, including those responsible for the increasing incidence of antibiotic resistant healthcare-associated infections, are susceptible to this new class of 2-pyridone antibiotics. Given the broad antibacterial activities of ring-fused 2-pyridone compounds against Gram-positive (GmP) bacteria we term these compounds GmPcides, which hold promise in combating the rising tide of antibiotic resistant Gram-positive pathogens.


Asunto(s)
Bacterias Grampositivas , Piridonas , Enterococos Resistentes a la Vancomicina , Antibacterianos/farmacología , Farmacorresistencia Bacteriana Múltiple , Bacterias Grampositivas/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , N-Acetil Muramoil-L-Alanina Amidasa/farmacología , Piridonas/farmacología , Vancomicina/farmacología , Enterococos Resistentes a la Vancomicina/efectos de los fármacos
7.
Nat Microbiol ; 7(5): 630-639, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35505248

RESUMEN

Recurrent urinary tract infections (rUTIs) are a major health burden worldwide, with history of infection being a significant risk factor. While the gut is a known reservoir for uropathogenic bacteria, the role of the microbiota in rUTI remains unclear. We conducted a year-long study of women with (n = 15) and without (n = 16) history of rUTI, from whom we collected urine, blood and monthly faecal samples for metagenomic and transcriptomic interrogation. During the study 24 UTIs were reported, with additional samples collected during and after infection. The gut microbiome of individuals with a history of rUTI was significantly depleted in microbial richness and butyrate-producing bacteria compared with controls, reminiscent of other inflammatory conditions. However, Escherichia coli gut and bladder populations were comparable between cohorts in both relative abundance and phylogroup. Transcriptional analysis of peripheral blood mononuclear cells revealed expression profiles indicative of differential systemic immunity between cohorts. Altogether, these results suggest that rUTI susceptibility is in part mediated through the gut-bladder axis, comprising gut dysbiosis and differential immune response to bacterial bladder colonization, manifesting in symptoms.


Asunto(s)
Infecciones por Escherichia coli , Microbioma Gastrointestinal , Infecciones Urinarias , Disbiosis , Escherichia coli , Infecciones por Escherichia coli/microbiología , Femenino , Humanos , Leucocitos Mononucleares , Masculino , Infecciones Urinarias/microbiología
8.
PLoS Pathog ; 17(1): e1009314, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33513212

RESUMEN

Type 1 pili have long been considered the major virulence factor enabling colonization of the urinary bladder by uropathogenic Escherichia coli (UPEC). The molecular pathogenesis of pyelonephritis is less well characterized, due to previous limitations in preclinical modeling of kidney infection. Here, we demonstrate in a recently developed mouse model that beyond bladder infection, type 1 pili also are critical for establishment of ascending pyelonephritis. Bacterial mutants lacking the type 1 pilus adhesin (FimH) were unable to establish kidney infection in male C3H/HeN mice. We developed an in vitro model of FimH-dependent UPEC binding to renal collecting duct cells, and performed a CRISPR screen in these cells, identifying desmoglein-2 as a primary renal epithelial receptor for FimH. The mannosylated extracellular domain of human DSG2 bound directly to the lectin domain of FimH in vitro, and introduction of a mutation in the FimH mannose-binding pocket abolished binding to DSG2. In infected C3H/HeN mice, type 1-piliated UPEC and Dsg2 were co-localized within collecting ducts, and administration of mannoside FIM1033, a potent small-molecule inhibitor of FimH, significantly attenuated bacterial loads in pyelonephritis. Our results broaden the biological importance of FimH, specify the first renal FimH receptor, and indicate that FimH-targeted therapeutics will also have application in pyelonephritis.


Asunto(s)
Adhesinas de Escherichia coli/metabolismo , Desmogleína 2/metabolismo , Infecciones por Escherichia coli/microbiología , Escherichia coli/patogenicidad , Proteínas Fimbrias/metabolismo , Pielonefritis/microbiología , Adhesinas de Escherichia coli/genética , Animales , Desmogleína 2/genética , Epitelio/microbiología , Escherichia coli/genética , Femenino , Proteínas Fimbrias/genética , Fimbrias Bacterianas/genética , Fimbrias Bacterianas/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C3H , Vejiga Urinaria/microbiología , Virulencia
9.
Aesthet Surg J ; 40(3): 281-295, 2020 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-30953053

RESUMEN

BACKGROUND: Staphylococcus epidermidis is a primary cause of breast implant-associated infection. S epidermidis possesses several virulence factors that enable it to bind both abiotic surfaces and host factors to form a biofilm. In addition S epidermidis colocalizes with matrix proteins coating explanted human breast implants. OBJECTIVES: The authors sought to identify matrix proteins that S epidermidis may exploit to infect various breast implant surfaces in vitro. METHODS: A combination of in vitro assays was used to characterize S epidermidis strains isolated from human breast implants to gain a better understanding of how these bacteria colonize breast implant surfaces. These included determining the (1) minimum inhibitory and bactericidal concentrations for irrigation solutions commonly used to prevent breast implant contamination; (2) expression and carriage of polysaccharide intercellular adhesin and serine-aspartate repeat proteins, which bind fibrinogen (SdrG) and collagen (SdrF), respectively; and (3) biofilm formation on varying implant surface characteristics, in different growth media, and supplemented with fibrinogen and Types I and III collagen. Scanning electron microscopy and immunofluorescence staining analyses were performed to corroborate findings from these assays. RESULTS: Textured breast implant surfaces support greater bacterial biofilm formation at baseline, and the addition of collagen significantly increases biomass on all surfaces tested. We found that S epidermidis isolated from breast implants all encoded SdrF. Consistent with this finding, these strains had a clear affinity for Type I collagen, forming dense, highly structured biofilms in its presence. CONCLUSIONS: The authors found that S epidermidis may utilize SdrF to interact with Type I collagen to form biofilm on breast implant surfaces.


Asunto(s)
Implantación de Mama , Implantes de Mama , Antibacterianos , Biopelículas , Implantación de Mama/efectos adversos , Implantes de Mama/efectos adversos , Humanos , Staphylococcus epidermidis
10.
Sci Rep ; 9(1): 10393, 2019 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-31316085

RESUMEN

Though rare, breast implant-associated anaplastic large cell lymphoma (BIA-ALCL), a CD30+ T-cell lymphoma associated with textured breast implants, has adversely impacted our perception of the safety of breast implants. Its etiology unknown, one hypothesis suggests an initiating inflammatory stimulus, possibly infectious, triggers BIA-ALCL. We analyzed microbiota of breast, skin, implant and capsule in BIA-ALCL patients (n = 7), and controls via culturing methods, 16S rRNA microbiome sequencing, and immunohistochemistry. Alpha and beta diversity metrics and relative abundance of Gram-negative bacteria were calculated, and phylogenetic trees constructed. Staphylococcus spp., the most commonly cultured microbes, were identified in both the BIA-ALCL and contralateral control breast. The diversity of bacterial microbiota did not differ significantly between BIA-ALCL and controls for any material analyzed. Further, there were no significant differences in the relative abundance of Gram-negative bacteria between BIA-ALCL and control specimens. Heat maps suggested substantial diversity in the composition of the bacterial microbiota of the skin, breast, implant and capsule between patients with no clear trend to distinguish BIA-ALCL from controls. While we identified no consistent differences between patients with BIA-ALCL-affected and contralateral control breasts, this study provides insights into the composition of the breast microbiota in this population.


Asunto(s)
Implantes de Mama/efectos adversos , Implantes de Mama/microbiología , Linfoma Anaplásico de Células Grandes/patología , Adulto , Bacterias , Implantación de Mama , Neoplasias de la Mama/patología , Femenino , Humanos , Linfoma Anaplásico de Células Grandes/microbiología , Microbiota , Persona de Mediana Edad , Filogenia , Complicaciones Posoperatorias/etiología , ARN Ribosómico 16S
11.
Plast Reconstr Surg Glob Open ; 7(2): e2037, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30881821

RESUMEN

BACKGROUND: Bacterial contamination of breast implants causes infection, can lead to capsular contracture, and is implicated in breast implant-associated anaplastic large cell lymphoma. Bacteria, however, also colonize clinically benign breast implants and little is known about the biologic signals that trigger the switch from a benign to pathologic state. METHODS: Explanted smooth as well as Biocell and Siltex textured breast implants associated with clinically normal and pathologic conditions were analyzed in this observational study. Immunofluorescence and bacterial culture techniques were performed. To avoid sampling bias, implant surfaces >25 sq cm were analyzed. RESULTS: Bacteria were detected on 9 of 22 clinically normal explanted devices or periprosthetic capsules, including 40% of Biocell tissue expanders and 75% of Biocell textured implants. Staphylococcus epidermidis was identified in 67% of the bacteria-positive capsular contractures. Fibrinogen was present on 17 of 18, and collagen on 13 of 18 analyzed breast implants. S. epidermidis co-localized with collagen, while group B streptococci and Klebsiella pneumoniae co-localized with fibrinogen. CONCLUSIONS: Bacteria are often detectable on clinically benign breast implants when a multimodal approach is applied to a substantial proportion of the device surface to avoid sampling bias. The impact of bacteria on breast implant pathology should be studied in the presence of an adequate negative control group to account for clinically benign bacteria. Disruption of the interaction of bacteria with matrix proteins coating the surface of breast implants may represent a nonantibiotic strategy for the prevention of breast implant bacterial contamination.

12.
J Med Chem ; 62(2): 467-479, 2019 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-30540910

RESUMEN

The F9/Yde/Fml pilus, tipped with the FmlH adhesin, has been shown to provide uropathogenic Escherichia coli (UPEC) a fitness advantage in urinary tract infections (UTIs). Here, we used X-ray structure guided design to optimize our previously described ortho-biphenyl Gal and GalNAc FmlH antagonists such as compound 1 by replacing the carboxylate with a sulfonamide as in 50. Other groups which can accept H-bonds were also tolerated. We pursued further modifications to the biphenyl aglycone resulting in significantly improved activity. Two of the most potent compounds, 86 (IC50 = 0.051 µM) and 90 (IC50 = 0.034 µM), exhibited excellent metabolic stability in mouse plasma and liver microsomes but showed only limited oral bioavailability (<1%) in rats. Compound 84 also showed a good pharmacokinetic (PK) profile in mice after IP dosing with compound exposure above the IC50 for 6 h. These new FmlH antagonists represent new antivirulence drugs for UTIs.


Asunto(s)
Adhesinas de Escherichia coli/química , Compuestos de Bifenilo/química , Diseño de Fármacos , Galactosamina/química , Galactosa/química , Adhesinas de Escherichia coli/metabolismo , Administración Oral , Animales , Galactosamina/farmacocinética , Galactosamina/uso terapéutico , Galactosa/farmacocinética , Semivida , Humanos , Ratones , Microsomas Hepáticos/metabolismo , Ratas , Relación Estructura-Actividad , Infecciones Urinarias/tratamiento farmacológico , Escherichia coli Uropatógena
13.
Nat Microbiol ; 3(12): 1362-1368, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30275511

RESUMEN

Chaperone-usher pathway pili are extracellular proteinaceous fibres ubiquitously found on Gram-negative bacteria, and mediate host-pathogen interactions and biofilm formation critical in pathogenesis in numerous human diseases1. During pilus assembly, an outer membrane macromolecular machine called the usher catalyses pilus biogenesis from the individual subunits that are delivered as chaperone-subunit complexes in the periplasm. The usher orchestrates pilus assembly using all five functional domains: a 24-stranded transmembrane ß-barrel translocation domain, a ß-sandwich plug domain, an amino-terminal periplasmic domain and two carboxy-terminal periplasmic domains (CTD1 and CTD2)2-6. Despite extensive structural and functional characterization, the mechanism by which the usher is activated to initiate pilus biogenesis is unknown. Here, we present the crystal structure of the full-length PapC usher from Escherichia coli in complex with its cognate PapDG chaperone-subunit complex in a pre-activation state, elucidating molecular details of how the usher is specifically engaged by allosteric interactions with its substrate preceding activation and how the usher facilitates the transfer of subunits from the amino-terminal periplasmic domain to the CTDs during pilus assembly. This work elucidates the intricate workings of a molecular machine that catalyses chaperone-usher pathway pilus assembly and opens the door for the development of potent inhibitors to block pilus biogenesis.


Asunto(s)
Proteínas de Escherichia coli/química , Escherichia coli/metabolismo , Fimbrias Bacterianas/química , Porinas/química , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Fimbrias Bacterianas/genética , Modelos Moleculares , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Proteínas Periplasmáticas/genética , Unión Proteica , Conformación Proteica , Dominios Proteicos
14.
mBio ; 9(4)2018 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-30018113

RESUMEN

Curli amyloid fibers are produced as part of the extracellular biofilm matrix and are composed primarily of the major structural subunit CsgA. The CsgE chaperone facilitates the secretion of CsgA through CsgG by forming a cap at the base of the nonameric CsgG outer membrane pore. We elucidated a series of finely tuned nonpolar and charge-charge interactions that facilitate the oligomerization of CsgE and its ability to transport unfolded CsgA to CsgG for translocation. CsgE oligomerization in vitro is temperature dependent and is disrupted by mutations in the W48 and F79 residues. Using nuclear magnetic resonance (NMR), we identified two regions of CsgE involved in the CsgE-CsgA interaction: a head comprising a positively charged patch centered around R47 and a stem comprising a negatively charged patch containing E31 and E85. Negatively charged residues in the intrinsically disordered N- and C-terminal "tails" were not implicated in this interaction. Head and stem residues were mutated and interrogated using in vivo measurements of curli production and in vitro amyloid polymerization assays. The R47 head residue of CsgE is required for stabilization of CsgA- and CsgE-mediated curli fiber formation. Mutation of the E31 and E85 stem residues to positively charged side chains decreased CsgE-mediated curli fiber formation but increased CsgE-mediated stabilization of CsgA. No single-amino-acid substitutions in the head, stem, or tail regions affected the ability of CsgE to cap the CsgG pore as determined by a bile salt sensitivity assay. These mechanistic insights into the directed assembly of functional amyloids in extracellular biofilms elucidate possible targets for biofilm-associated bacterial infections.IMPORTANCE Curli represent a class of functional amyloid fibers produced by Escherichia coli and other Gram-negative bacteria that serve as protein scaffolds in the extracellular biofilm matrix. Despite the lack of sequence conservation among different amyloidogenic proteins, the structural and biophysical properties of functional amyloids such as curli closely resemble those of amyloids associated with several common neurodegenerative diseases. These parallels are underscored by the observation that certain proteins and chemicals can prevent amyloid formation by the major curli subunit CsgA and by alpha-synuclein, the amyloid-forming protein found in Lewy bodies during Parkinson's disease. CsgA subunits are targeted to the CsgG outer membrane pore by CsgE prior to secretion and assembly into fibers. Here, we use biophysical, biochemical, and genetic approaches to elucidate a mechanistic understanding of CsgE function in curli biogenesis.


Asunto(s)
Amiloide/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Escherichia coli/fisiología , Proteínas de Transporte de Membrana/química , Proteínas de Transporte de Membrana/metabolismo , Biopelículas/crecimiento & desarrollo , Proteínas de Escherichia coli/genética , Matriz Extracelular de Sustancias Poliméricas/metabolismo , Lipoproteínas/metabolismo , Proteínas de Transporte de Membrana/genética , Modelos Biológicos , Modelos Moleculares , Mutación , Polimerizacion , Unión Proteica , Conformación Proteica , Estabilidad Proteica
15.
Proc Natl Acad Sci U S A ; 115(12): E2819-E2828, 2018 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-29507247

RESUMEN

Treatment of bacterial infections is becoming a serious clinical challenge due to the global dissemination of multidrug antibiotic resistance, necessitating the search for alternative treatments to disarm the virulence mechanisms underlying these infections. Uropathogenic Escherichia coli (UPEC) employs multiple chaperone-usher pathway pili tipped with adhesins with diverse receptor specificities to colonize various host tissues and habitats. For example, UPEC F9 pili specifically bind galactose or N-acetylgalactosamine epitopes on the kidney and inflamed bladder. Using X-ray structure-guided methods, virtual screening, and multiplex ELISA arrays, we rationally designed aryl galactosides and N-acetylgalactosaminosides that inhibit the F9 pilus adhesin FmlH. The lead compound, 29ß-NAc, is a biphenyl N-acetyl-ß-galactosaminoside with a Ki of ∼90 nM, representing a major advancement in potency relative to the characteristically weak nature of most carbohydrate-lectin interactions. 29ß-NAc binds tightly to FmlH by engaging the residues Y46 through edge-to-face π-stacking with its A-phenyl ring, R142 in a salt-bridge interaction with its carboxylate group, and K132 through water-mediated hydrogen bonding with its N-acetyl group. Administration of 29ß-NAc in a mouse urinary tract infection (UTI) model significantly reduced bladder and kidney bacterial burdens, and coadministration of 29ß-NAc and mannoside 4Z269, which targets the type 1 pilus adhesin FimH, resulted in greater elimination of bacteria from the urinary tract than either compound alone. Moreover, FmlH specifically binds healthy human kidney tissue in a 29ß-NAc-inhibitable manner, suggesting a key role for F9 pili in human kidney colonization. Thus, these glycoside antagonists of FmlH represent a rational antivirulence strategy for UPEC-mediated UTI treatment.


Asunto(s)
Adhesinas de Escherichia coli/química , Antibacterianos/química , Antibacterianos/farmacología , Adhesión Bacteriana/efectos de los fármacos , Infecciones Urinarias/microbiología , Adhesinas de Escherichia coli/metabolismo , Animales , Cristalografía por Rayos X , Evaluación Preclínica de Medicamentos/métodos , Infecciones por Escherichia coli/tratamiento farmacológico , Infecciones por Escherichia coli/microbiología , Femenino , Galactósidos/síntesis química , Galactósidos/química , Humanos , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/microbiología , Ligandos , Ratones Endogámicos C3H , Simulación del Acoplamiento Molecular , Imitación Molecular , Infecciones Urinarias/tratamiento farmacológico , Escherichia coli Uropatógena/efectos de los fármacos , Escherichia coli Uropatógena/patogenicidad
16.
Proc Natl Acad Sci U S A ; 114(41): E8721-E8730, 2017 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-28973850

RESUMEN

Methicillin-resistant Staphylococcus aureus (MRSA) is an emerging cause of catheter-associated urinary tract infection (CAUTI), which frequently progresses to more serious invasive infections. We adapted a mouse model of CAUTI to investigate how catheterization increases an individual's susceptibility to MRSA UTI. This analysis revealed that catheterization was required for MRSA to achieve high-level, persistent infection in the bladder. As shown previously, catheter placement induced an inflammatory response resulting in the release of the host protein fibrinogen (Fg), which coated the bladder and implant. Following infection, we showed that MRSA attached to the urothelium and implant in patterns that colocalized with deposited Fg. Furthermore, MRSA exacerbated the host inflammatory response to stimulate the additional release and accumulation of Fg in the urinary tract, which facilitated MRSA colonization. Consistent with this model, analysis of catheters from patients with S. aureus-positive cultures revealed colocalization of Fg, which was deposited on the catheter, with S. aureus Clumping Factors A and B (ClfA and ClfB) have been shown to contribute to MRSA-Fg interactions in other models of disease. We found that mutants in clfA had significantly greater Fg-binding defects than mutants in clfB in several in vitro assays. Paradoxically, only the ClfB- strain was significantly attenuated in the CAUTI model. Together, these data suggest that catheterization alters the urinary tract environment to promote MRSA CAUTI pathogenesis by inducing the release of Fg, which the pathogen enhances to persist in the urinary tract despite the host's robust immune response.


Asunto(s)
Cateterismo/efectos adversos , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Infecciones Estafilocócicas/microbiología , Vejiga Urinaria/microbiología , Infecciones Urinarias/microbiología , Sistema Urinario/microbiología , Adhesinas Bacterianas/metabolismo , Animales , Femenino , Fibrinógeno/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Infecciones Estafilocócicas/metabolismo , Infecciones Estafilocócicas/patología , Vejiga Urinaria/metabolismo , Vejiga Urinaria/patología , Sistema Urinario/metabolismo , Sistema Urinario/patología , Infecciones Urinarias/metabolismo , Infecciones Urinarias/patología
17.
Nature ; 546(7659): 528-532, 2017 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-28614296

RESUMEN

Urinary tract infections (UTIs) caused by uropathogenic Escherichia coli (UPEC) affect 150 million people annually. Despite effective antibiotic therapy, 30-50% of patients experience recurrent UTIs. In addition, the growing prevalence of UPEC that are resistant to last-line antibiotic treatments, and more recently to carbapenems and colistin, make UTI a prime example of the antibiotic-resistance crisis and emphasize the need for new approaches to treat and prevent bacterial infections. UPEC strains establish reservoirs in the gut from which they are shed in the faeces, and can colonize the periurethral area or vagina and subsequently ascend through the urethra to the urinary tract, where they cause UTIs. UPEC isolates encode up to 16 distinct chaperone-usher pathway pili, and each pilus type may enable colonization of a habitat in the host or environment. For example, the type 1 pilus adhesin FimH binds mannose on the bladder surface, and mediates colonization of the bladder. However, little is known about the mechanisms underlying UPEC persistence in the gut. Here, using a mouse model, we show that F17-like and type 1 pili promote intestinal colonization and show distinct binding to epithelial cells distributed along colonic crypts. Phylogenomic and structural analyses reveal that F17-like pili are closely related to pilus types carried by intestinal pathogens, but are restricted to extra-intestinal pathogenic E. coli. Moreover, we show that targeting FimH with M4284, a high-affinity inhibitory mannoside, reduces intestinal colonization of genetically diverse UPEC isolates, while simultaneously treating UTI, without notably disrupting the structural configuration of the gut microbiota. By selectively depleting intestinal UPEC reservoirs, mannosides could markedly reduce the rate of UTIs and recurrent UTIs.


Asunto(s)
Proteínas Fimbrias/antagonistas & inhibidores , Intestinos/efectos de los fármacos , Intestinos/microbiología , Manósidos/farmacología , Ácidos Ftálicos/farmacología , Infecciones Urinarias/prevención & control , Escherichia coli Uropatógena/efectos de los fármacos , Escherichia coli Uropatógena/aislamiento & purificación , Adhesinas de Escherichia coli/metabolismo , Secuencia de Aminoácidos , Animales , Células Epiteliales/efectos de los fármacos , Células Epiteliales/microbiología , Heces/microbiología , Femenino , Proteínas Fimbrias/metabolismo , Fimbrias Bacterianas/clasificación , Fimbrias Bacterianas/efectos de los fármacos , Fimbrias Bacterianas/genética , Fimbrias Bacterianas/metabolismo , Humanos , Intestinos/citología , Manósidos/uso terapéutico , Ratones , Modelos Moleculares , Ácidos Ftálicos/uso terapéutico , Vejiga Urinaria/efectos de los fármacos , Vejiga Urinaria/microbiología , Infecciones Urinarias/tratamiento farmacológico , Infecciones Urinarias/microbiología , Escherichia coli Uropatógena/clasificación , Escherichia coli Uropatógena/genética
18.
Sci Adv ; 3(2): e1601944, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28246638

RESUMEN

Positive selection in the two-domain type 1 pilus adhesin FimH enhances Escherichia coli fitness in urinary tract infection (UTI). We report a comprehensive atomic-level view of FimH in two-state conformational ensembles in solution, composed of one low-affinity tense (T) and multiple high-affinity relaxed (R) conformations. Positively selected residues allosterically modulate the equilibrium between these two conformational states, each of which engages mannose through distinct binding orientations. A FimH variant that only adopts the R state is severely attenuated early in a mouse model of uncomplicated UTI but is proficient at colonizing catheterized bladders in vivo or bladder transitional-like epithelial cells in vitro. Thus, the bladder habitat has barrier(s) to R state-mediated colonization possibly conferred by the terminally differentiated bladder epithelium and/or decoy receptors in urine. Together, our studies reveal the conformational landscape in solution, binding mechanisms, and adhesive strength of an allosteric two-domain adhesin that evolved "moderate" affinity to optimize persistence in the bladder during UTI.


Asunto(s)
Adhesinas de Escherichia coli , Infecciones por Escherichia coli , Escherichia coli , Proteínas Fimbrias , Interacciones Huésped-Parásitos/fisiología , Vejiga Urinaria , Infecciones Urinarias , Adhesinas de Escherichia coli/química , Adhesinas de Escherichia coli/genética , Adhesinas de Escherichia coli/metabolismo , Animales , Escherichia coli/química , Escherichia coli/patogenicidad , Escherichia coli/fisiología , Infecciones por Escherichia coli/genética , Infecciones por Escherichia coli/metabolismo , Infecciones por Escherichia coli/patología , Femenino , Proteínas Fimbrias/química , Proteínas Fimbrias/genética , Proteínas Fimbrias/metabolismo , Ratones , Dominios Proteicos , Vejiga Urinaria/metabolismo , Vejiga Urinaria/microbiología , Vejiga Urinaria/fisiología , Infecciones Urinarias/genética , Infecciones Urinarias/metabolismo , Infecciones Urinarias/microbiología , Infecciones Urinarias/patología
19.
mBio ; 7(5)2016 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-27795399

RESUMEN

Gram-positive bacteria in the genus Enterococcus are a frequent cause of catheter-associated urinary tract infection (CAUTI), a disease whose treatment is increasingly challenged by multiantibiotic-resistant strains. We have recently shown that E. faecalis uses the Ebp pilus, a heteropolymeric surface fiber, to bind the host protein fibrinogen as a critical step in CAUTI pathogenesis. Fibrinogen is deposited on catheters due to catheter-induced inflammation and is recognized by the N-terminal domain of EbpA (EbpANTD), the Ebp pilus's adhesin. In a murine model, vaccination with EbpANTD confers significant protection against CAUTI. Here, we explored the mechanism of protection using passive transfer of immune sera to show that antisera blocking EbpANTD-fibrinogen interactions not only is prophylactic but also can act therapeutically to reduce bacterial titers of an existing infection. Analysis of 55 clinical CAUTI, bloodstream, and gastrointestinal isolates, including E. faecalis, E. faecium, and vancomycin-resistant enterococci (VRE), revealed a diversity of levels of EbpA expression and fibrinogen-binding efficiency in vitro Strikingly, analysis of 10 strains representative of fibrinogen-binding diversity demonstrated that, irrespective of EbpA levels, EbpANTD antibodies were universally protective. The results indicate that, despite diversity in levels of fibrinogen binding, strategies that target the disruption of EbpANTD-fibrinogen interactions have considerable promise for treatment of CAUTI. IMPORTANCE: Urinary catheterization is a routine medical procedure, and it has been estimated that 30 million Foley catheters are used annually in the United States. Importantly, placement of a urinary catheter renders the patient susceptible to developing a catheter-associated urinary tract infection, accounting for 1 million cases per year. Additionally, these infections can lead to serious complications, including bloodstream infection and death. Enterococcus strains are a common cause of these infections, and management of enterococcal infections has been more difficult in recent years due to the development of antibiotic resistance and the ability of strains to disseminate, resulting in a major threat in hospital settings. In this study, we developed an antibiotic-sparing treatment that is effective against diverse enterococcal isolates, including vancomycin-resistant enterococci, during catheter-associated urinary tract infections.


Asunto(s)
Anticuerpos Antibacterianos/administración & dosificación , Infecciones Relacionadas con Catéteres/terapia , Enterococcus/inmunología , Infecciones por Bacterias Grampositivas/terapia , Inmunoterapia/métodos , Infecciones Urinarias/terapia , Adhesinas Bacterianas/inmunología , Animales , Infecciones Relacionadas con Catéteres/prevención & control , Modelos Animales de Enfermedad , Enterococcus/aislamiento & purificación , Infecciones por Bacterias Grampositivas/prevención & control , Humanos , Inmunización Pasiva/métodos , Ratones Endogámicos C57BL , Resultado del Tratamiento , Estados Unidos , Infecciones Urinarias/prevención & control
20.
J Med Chem ; 59(20): 9390-9408, 2016 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-27689912

RESUMEN

Gram-negative uropathogenic Escherichia coli (UPEC) bacteria are a causative pathogen of urinary tract infections (UTIs). Previously developed antivirulence inhibitors of the type 1 pilus adhesin, FimH, demonstrated oral activity in animal models of UTI but were found to have limited compound exposure due to the metabolic instability of the O-glycosidic bond (O-mannosides). Herein, we disclose that compounds having the O-glycosidic bond replaced with carbon linkages had improved stability and inhibitory activity against FimH. We report on the design, synthesis, and in vivo evaluation of this promising new class of carbon-linked C-mannosides that show improved pharmacokinetic (PK) properties relative to O-mannosides. Interestingly, we found that FimH binding is stereospecifically modulated by hydroxyl substitution on the methylene linker, where the R-hydroxy isomer has a 60-fold increase in potency. This new class of C-mannoside antagonists have significantly increased compound exposure and, as a result, enhanced efficacy in mouse models of acute and chronic UTI.


Asunto(s)
Antibacterianos/farmacología , Escherichia coli/efectos de los fármacos , Escherichia coli/patogenicidad , Manósidos/administración & dosificación , Manósidos/farmacología , Infecciones Urinarias/tratamiento farmacológico , Infecciones Urinarias/microbiología , Administración Oral , Animales , Antibacterianos/administración & dosificación , Antibacterianos/química , Relación Dosis-Respuesta a Droga , Femenino , Manósidos/química , Ratones , Ratones Endogámicos C3H , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Estructura Molecular , Relación Estructura-Actividad , Virulencia/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...