Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38463963

ABSTRACT

Low-abundance members of microbial communities are difficult to study in their native habitats. This includes Escherichia coli, a minor, but common inhabitant of the gastrointestinal tract and opportunistic pathogen, including of the urinary tract, where it is the primary pathogen. While multi-omic analyses have detailed critical interactions between uropathogenic Escherichia coli (UPEC) and the bladder that mediate UTI outcome, comparatively little is known about UPEC in its pre-infection reservoir, partly due to its low abundance there (<1% relative abundance). To accurately and sensitively explore the genomes and transcriptomes of diverse E. coli in gastrointestinal communities, we developed E. coli PanSelect which uses a set of probes designed to specifically recognize and capture E. coli's broad pangenome from sequencing libraries. We demonstrated the ability of E. coli PanSelect to enrich, by orders of magnitude, sequencing data from diverse E. coli using a mock community and a set of human stool samples collected as part of a cohort study investigating drivers of recurrent urinary tract infections (rUTI). Comparisons of genomes and transcriptomes between E. coli residing in the gastrointestinal tracts of women with and without a history of rUTI suggest that rUTI gut E. coli are responding to increased levels of oxygen and nitrate, suggestive of mucosal inflammation, which may have implications for recurrent disease. E. coli PanSelect is well suited for investigations of native in vivo biology of E. coli in other environments where it is at low relative abundance, and the framework described here has broad applicability to other highly diverse, low abundance organisms.

2.
Nat Microbiol ; 7(5): 630-639, 2022 05.
Article in English | MEDLINE | ID: mdl-35505248

ABSTRACT

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.


Subject(s)
Escherichia coli Infections , Gastrointestinal Microbiome , Urinary Tract Infections , Dysbiosis , Escherichia coli , Escherichia coli Infections/microbiology , Female , Humans , Leukocytes, Mononuclear , Male , Urinary Tract Infections/microbiology
3.
Genome Biol ; 23(1): 74, 2022 03 07.
Article in English | MEDLINE | ID: mdl-35255937

ABSTRACT

Human-associated microbial communities comprise not only complex mixtures of bacterial species, but also mixtures of conspecific strains, the implications of which are mostly unknown since strain level dynamics are underexplored due to the difficulties of studying them. We introduce the Strain Genome Explorer (StrainGE) toolkit, which deconvolves strain mixtures and characterizes component strains at the nucleotide level from short-read metagenomic sequencing with higher sensitivity and resolution than other tools. StrainGE is able to identify strains at 0.1x coverage and detect variants for multiple conspecific strains within a sample from coverages as low as 0.5x.


Subject(s)
Microbiota , Bacteria/genetics , Humans , Metagenome , Metagenomics , Microbiota/genetics
4.
BMC Microbiol ; 21(1): 53, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33596852

ABSTRACT

BACKGROUND: Urinary tract infections (UTIs) affect 15 million women each year in the United States, with > 20% experiencing frequent recurrent UTIs. A recent placebo-controlled clinical trial found a 39% reduction in UTI symptoms among recurrent UTI sufferers who consumed a daily cranberry beverage for 24 weeks. Using metagenomic sequencing of stool from a subset of these trial participants, we assessed the impact of cranberry consumption on the gut microbiota, a reservoir for UTI-causing pathogens such as Escherichia coli, which causes > 80% of UTIs. RESULTS: The overall taxonomic composition, community diversity, carriage of functional pathways and gene families, and relative abundances of the vast majority of observed bacterial taxa, including E. coli, were not changed significantly by cranberry consumption. However, one unnamed Flavonifractor species (OTU41), which represented ≤1% of the overall metagenome, was significantly less abundant in cranberry consumers compared to placebo at trial completion. Given Flavonifractor's association with negative human health effects, we sought to determine OTU41 characteristic genes that may explain its differential abundance and/or relationship to key host functions. Using comparative genomic and metagenomic techniques, we identified genes in OTU41 related to transport and metabolism of various compounds, including tryptophan and cobalamin, which have been shown to play roles in host-microbe interactions. CONCLUSION: While our results indicated that cranberry juice consumption had little impact on global measures of the microbiome, we found one unnamed Flavonifractor species differed significantly between study arms. This suggests further studies are needed to assess the role of cranberry consumption and Flavonifractor in health and wellbeing in the context of recurrent UTI. TRIAL REGISTRATION: Clinical trial registration number: ClinicalTrials.gov NCT01776021 .


Subject(s)
Bacteria/drug effects , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/genetics , Plant Extracts/administration & dosage , Vaccinium macrocarpon/chemistry , Adult , Bacteria/classification , Bacteria/genetics , Beverages , Double-Blind Method , Feces/microbiology , Female , Gastrointestinal Microbiome/physiology , Humans , Metagenome , Metagenomics/methods , Middle Aged , Reinfection/microbiology , Reinfection/prevention & control , Urinary Tract Infections/microbiology , Urinary Tract Infections/prevention & control
5.
Nat Rev Microbiol ; 19(4): 241-255, 2021 04.
Article in English | MEDLINE | ID: mdl-33093662

ABSTRACT

In a striking display of trans-kingdom symbiosis, gut bacteria cooperate with their animal hosts to regulate the development and function of the immune, metabolic and nervous systems through dynamic bidirectional communication along the 'gut-brain axis'. These processes may affect human health, as certain animal behaviours appear to correlate with the composition of gut bacteria, and disruptions in microbial communities have been implicated in several neurological disorders. Most insights about host-microbiota interactions come from animal models, which represent crucial tools for studying the various pathways linking the gut and the brain. However, there are complexities and manifest limitations inherent in translating complex human disease to reductionist animal models. In this Review, we discuss emerging and exciting evidence of intricate and crucial connections between the gut microbiota and the brain involving multiple biological systems, and possible contributions by the gut microbiota to neurological disorders. Continued advances from this frontier of biomedicine may lead to tangible impacts on human health.


Subject(s)
Brain Diseases/microbiology , Brain Diseases/pathology , Gastrointestinal Microbiome/physiology , Host Microbial Interactions/physiology , Animals , Bacteria/classification , Bacteria/isolation & purification , Brain/microbiology , Brain/pathology , Humans , Models, Animal , Symbiosis/physiology
6.
Article in English | MEDLINE | ID: mdl-29507749

ABSTRACT

The emergence of drug-resistant pathogens has led to a decline in the efficacy of traditional antimicrobial therapy. The rise in resistance has been driven by widespread use, and in some cases misuse, of antibacterial agents in treating a variety of infections. A growing body of research has begun to elucidate the harmful effects of broad-spectrum antibiotic therapy on the beneficial host microbiota. To combat these threats, increasing effort is being directed toward the development of precision antimicrobial therapeutics that target key virulence determinants of specific pathogens while leaving the remainder of the host microbiota undisturbed. This includes the recent development of small molecules termed "mannosides" that specifically target uropathogenic E. coli (UPEC). Mannosides are glycomimetics of the natural mannosylated host receptor for type 1 pili, extracellular appendages that promotes UPEC colonization in the intestine. Type 1 pili are also critical for colonization and infection in the bladder. In both cases, mannosides act as molecular decoys which potently prevent bacteria from binding to host tissues. In mice, oral treatment with mannosides simultaneously clears active bladder infection and removes intestinal UPEC while leaving the gut microbiota structure relatively unchanged. Similar treatment strategies successfully target other pathogens, like adherent-invasive E. coli (AIEC), an organism associated with Crohn's disease (CD), in mouse models. While not without its challenges, antibiotic-sparing therapeutic approaches hold great promise in a variety of disease systems, including UTI, CD, otitis media (OM), and others. In this perspective we highlight the benefits, progress, and roadblocks to the development of precision antimicrobial therapeutics.

7.
Genome Med ; 9(1): 110, 2017 Dec 14.
Article in English | MEDLINE | ID: mdl-29241446

ABSTRACT

Antibiotics have become the standard of care for bacterial infections. However, rising rates of antibiotic-resistant infections are outpacing the development of new antimicrobials. Broad-spectrum antibiotics also harm beneficial microbial communities inhabiting humans. To combat antibiotic resistance and protect these communities, new precision antimicrobials must be engineered to target specific pathogens.


Subject(s)
Anti-Infective Agents/therapeutic use , Gastrointestinal Microbiome/drug effects , Precision Medicine/methods , Animals , Anti-Infective Agents/administration & dosage , Anti-Infective Agents/adverse effects , Bacterial Infections/drug therapy , Humans
8.
Proc Natl Acad Sci U S A ; 114(41): E8721-E8730, 2017 10 10.
Article in English | MEDLINE | ID: mdl-28973850

ABSTRACT

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.


Subject(s)
Catheterization/adverse effects , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Staphylococcal Infections/microbiology , Urinary Bladder/microbiology , Urinary Tract Infections/microbiology , Urinary Tract/microbiology , Adhesins, Bacterial/metabolism , Animals , Female , Fibrinogen/metabolism , Humans , Mice , Mice, Inbred C57BL , Protein Binding , Staphylococcal Infections/metabolism , Staphylococcal Infections/pathology , Urinary Bladder/metabolism , Urinary Bladder/pathology , Urinary Tract/metabolism , Urinary Tract/pathology , Urinary Tract Infections/metabolism , Urinary Tract Infections/pathology
9.
Nature ; 546(7659): 528-532, 2017 06 22.
Article in English | MEDLINE | ID: mdl-28614296

ABSTRACT

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.


Subject(s)
Fimbriae Proteins/antagonists & inhibitors , Intestines/drug effects , Intestines/microbiology , Mannosides/pharmacology , Phthalic Acids/pharmacology , Urinary Tract Infections/prevention & control , Uropathogenic Escherichia coli/drug effects , Uropathogenic Escherichia coli/isolation & purification , Adhesins, Escherichia coli/metabolism , Amino Acid Sequence , Animals , Epithelial Cells/drug effects , Epithelial Cells/microbiology , Feces/microbiology , Female , Fimbriae Proteins/metabolism , Fimbriae, Bacterial/classification , Fimbriae, Bacterial/drug effects , Fimbriae, Bacterial/genetics , Fimbriae, Bacterial/metabolism , Humans , Intestines/cytology , Mannosides/therapeutic use , Mice , Models, Molecular , Phthalic Acids/therapeutic use , Urinary Bladder/drug effects , Urinary Bladder/microbiology , Urinary Tract Infections/drug therapy , Urinary Tract Infections/microbiology , Uropathogenic Escherichia coli/classification , Uropathogenic Escherichia coli/genetics
10.
Sci Transl Med ; 9(382)2017 03 22.
Article in English | MEDLINE | ID: mdl-28330863

ABSTRACT

Urinary tract infections (UTIs) are caused by uropathogenic Escherichia coli (UPEC) strains. In contrast to many enteric E. coli pathogroups, no genetic signature has been identified for UPEC strains. We conducted a high-resolution comparative genomic study using E. coli isolates collected from the urine of women suffering from frequent recurrent UTIs. These isolates were genetically diverse and varied in their urovirulence, that is, their ability to infect the bladder in a mouse model of cystitis. We found no set of genes, including previously defined putative urovirulence factors (PUFs), that were predictive of urovirulence. In addition, in some patients, the E. coli strain causing a recurrent UTI had fewer PUFs than the supplanted strain. In competitive experimental infections in mice, the supplanting strain was more efficient at colonizing the mouse bladder than the supplanted strain. Despite the lack of a clear genomic signature for urovirulence, comparative transcriptomic and phenotypic analyses revealed that the expression of key conserved functions during culture, such as motility and metabolism, could be used to predict subsequent colonization of the mouse bladder. Together, our findings suggest that UTI risk and outcome may be determined by complex interactions between host susceptibility and the urovirulence potential of diverse bacterial strains.


Subject(s)
Disease Susceptibility , Escherichia coli Infections/microbiology , Escherichia coli/pathogenicity , Host-Pathogen Interactions , Urinary Tract Infections/microbiology , Animals , Biomarkers/metabolism , Chronic Disease , Coinfection/microbiology , Colony Count, Microbial , Cystitis/microbiology , Cystitis/pathology , Escherichia coli/genetics , Escherichia coli/isolation & purification , Female , Gene Expression Regulation, Bacterial , Humans , Mice , Mice, Inbred Strains , Phenotype , Phylogeny , Recurrence , Risk Factors , Severity of Illness Index , Treatment Outcome , Urine/microbiology , Virulence/genetics , Virulence Factors/metabolism
12.
mBio ; 7(5)2016 10 25.
Article in English | MEDLINE | ID: mdl-27795399

ABSTRACT

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.


Subject(s)
Antibodies, Bacterial/administration & dosage , Catheter-Related Infections/therapy , Enterococcus/immunology , Gram-Positive Bacterial Infections/therapy , Immunotherapy/methods , Urinary Tract Infections/therapy , Adhesins, Bacterial/immunology , Animals , Catheter-Related Infections/prevention & control , Disease Models, Animal , Enterococcus/isolation & purification , Gram-Positive Bacterial Infections/prevention & control , Humans , Immunization, Passive/methods , Mice, Inbred C57BL , Treatment Outcome , United States , Urinary Tract Infections/prevention & control
13.
J Urol ; 196(2): 416-421, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26827873

ABSTRACT

PURPOSE: Catheter associated urinary tract infections account for approximately 40% of all hospital acquired infections worldwide with more than 1 million cases diagnosed annually. Recent data from a catheter associated urinary tract infection animal model has shown that inflammation induced by catheterization releases host fibrinogen, which accumulates on the catheter. Further, Enterococcus faecalis catheter colonization was found to depend on EbpA (endocarditis and biofilm-associated pilus), a fibrinogen binding adhesin. We evaluated this mechanism in a human model. MATERIALS AND METHODS: Urinary catheters were collected from patients hospitalized for surgical or nonsurgical urological procedures. Catheters were subjected to immunofluorescence analyses by incubation with antifibrinogen antibody and then staining for fluorescence. Fluorescence intensity was compared to that of standard catheters. Catheters were incubated with strains of Enterococcus faecalis, Staphylococcus aureus or Candida to assess binding of those strains to fibrinogen laden catheters. RESULTS: After various surgical and urological procedures, 50 catheters were collected. In vivo dwell time ranged from 1 hour to 59 days. All catheters had fibrinogen deposition. Accumulation depended on dwell time but not on surgical procedure or catheter material. Catheters were probed ex vivo with E. faecalis, S. aureus and Candida albicans, which bound to catheters only in regions where fibrinogen was deposited. CONCLUSIONS: Taken together, these data show that urinary catheters act as a binding surface for the accumulation of fibrinogen. Fibrinogen is released due to inflammation resulting from a urological procedure or catheter placement, creating a niche that can be exploited by uropathogens to cause catheter associated urinary tract infections.


Subject(s)
Bacterial Adhesion , Catheter-Related Infections/etiology , Cross Infection/etiology , Fibrinogen/analysis , Urinary Catheterization/adverse effects , Urinary Catheters/adverse effects , Urinary Tract Infections/etiology , Adult , Biomarkers/analysis , Biomarkers/metabolism , Candida albicans , Catheter-Related Infections/microbiology , Cross Infection/microbiology , Enterococcus faecalis , Female , Fibrinogen/metabolism , Humans , Male , Staphylococcus aureus , Urinary Catheters/microbiology , Urinary Tract Infections/microbiology , Urologic Surgical Procedures
14.
Curr Urol Rep ; 14(5): 448-56, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23832844

ABSTRACT

Urinary tract infections (UTIs) are common, and over half of women report having had at least one in their lifetime. Nearly a third of these women experience recurrent UTI episodes, but the mechanisms of these recurrences are not fully elucidated. Frequent use of antimicrobials for treatment and prevention of UTIs and other infections has contributed to the evolution of multidrug-resistant microorganisms globally. This is a looming worldwide crisis that has created an urgent need for novel strategies for the treatment and prevention of UTIs. Furthering our understanding of the mechanisms of recurrent UTIs, from both host and bacterial perspectives, will be paramount in developing targeted management strategies. In this review, we discuss recent findings regarding recurrent UTIs in women, including progress in our understanding of the mechanisms of recurrence as well as emerging treatments.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Urinary Tract Infections , Urodynamics/physiology , Humans , Secondary Prevention , Treatment Outcome , Urinary Tract Infections/drug therapy , Urinary Tract Infections/etiology , Urinary Tract Infections/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...