1 s2.0 S0944501322000507 Main

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 19

Microbiological Research 259 (2022) 127010

Contents lists available at ScienceDirect

Microbiological Research
journal homepage: www.elsevier.com/locate/micres

The urinary microbiome and biological therapeutics: Novel therapies for


urinary tract infections
Ciara Kenneally, Craig P. Murphy, Roy D. Sleator, Eamonn P. Culligan *
Department of Biological Sciences, Munster Technological University, Bishopstown, Cork T12 P928, Ireland

A R T I C L E I N F O A B S T R A C T

Keywords: The discovery of microbial communities in the urinary tract (the urobiome) has fundamentally altered the
Antibiotic Resistance previous doctrine regarding urine sterility and associated urinary disorders. Recent advances in culturing and
Asymptomatic Bacteriuria culture-independent DNA sequencing technologies have characterised the resident microbial community in the
Biotherapeutics
urobiome, and has, in turn, demonstrated how community imbalances potentially contribute to infection and
Urobiome
Uropathogenic E. coli
disease. As we enter a post-antibiotic era, the effectiveness of standard antimicrobial treatments against multi-
UTI drug resistant (MDR) uropathogens is vastly diminished. Preliminary research is accumulating surrounding
microbiome-based therapies, and their potential as non-antibiotic therapeutics. In this context, the urobiome is
significantly underexplored, and knowledge regarding the fundamental role of its constituents is lacking. Herein,
we review the current state of the art concerning the urobiome; specifically, how it impacts health and disease
states, in the context of urinary tract infections (UTIs). Furthermore, we discuss the development of novel bio­
logical therapeutics that may have the potential to provide significant advancements in UTI therapy, with a
particular focus on bacterial interference, probiotics, antimicrobial peptides, bacteriocins, and bacteriophage.

1. Introduction the existence of a ‘core’ urobiome has been hypothesized, with periodic
shifts in the microbial community leading to infection (Lewis et al.,
The human body is host to a diverse community of microorganisms, 2013; Ammitzbøll et al., 2021; Zandbergen et al., 2021). Indeed, one of
establishing symbiotic relationships with one another and their host the most common bacterial infections in humans affects the urinary tract
(Sleator, 2010). Extensive studies examining the human microbiome (Guglietta, 2017). Globally, urinary tract infections (UTIs) account for
have generated an unprecedented understanding of the microorganisms ~150 million infections annually, and ~40% of all hospital-acquired
that colonise both the human body and its external environment (Cull­ infections (Nicolle, 2008; Totsika et al., 2011). Women are dispropor­
igan et al., 2014). In turn, much has been learned about the metabolic tionally affected by UTIs with > 50% women developing at least one UTI
and immune function of the microbiome, and the interrelated dynamics over the course of their lifetime (Foxman, 2014). Risk factors among
between a microorganism’s relative abundance (Rizzetto et al., 2018; different age groups tend to vary, and overall women develop more UTIs
Visconti et al., 2019). However, not all parts of the body were deemed than males due to anatomical and physiological differences, such as a
suitable for initial exploration. shorter urethra and hormone fluctuations (Abelson et al., 2018). A
The urinary tract was not included in early microbiome research, as higher frequency of sexual intercourse is a risk factor for younger co­
it was originally thought to be a sterile environment (Magistro and Stief, horts and lower oestrogen levels predisposes postmenopausal females to
2019). Urine samples containing bacteria were believed to be contam­ developing UTIs (Alperin et al., 2019; Jung and Brubaker, 2020; Price
inated by the external environment, due to difficulties associated with et al., 2020b). Furthermore, one-in-three women in the United States
sampling and microbiological analysis (Ainsworth, 2017). However, will have received antibiotic therapy for their first UTI before the age of
over the last decade the existence of a urinary microbiome (urobiome) twenty-four (Foxman et al., 2000). Uropathogenic reservoirs often pre­
has been acknowledged, and shown to consist of a complex microbial vail within the urinary tract, and the surrounding vaginal and gut
community (Wolfe et al., 2012; Whiteside et al., 2015; Antunes-Lopes microbiomes, causing recurrent UTIs in up to 30% of individuals within
et al., 2020; Jones et al., 2021). Although studies are relatively recent, 6-months of the initial infection (Vagios et al., 2020). This high

* Correspondence to: Department of Biological Sciences, Munster Technological University, Cork, Ireland.
E-mail address: eamonn.culligan@mtu.ie (E.P. Culligan).

https://doi.org/10.1016/j.micres.2022.127010
Received 6 January 2022; Received in revised form 15 March 2022; Accepted 16 March 2022
Available online 20 March 2022
0944-5013/© 2022 The Author(s). Published by Elsevier GmbH. This is an open access article under the CC BY license
(http://creativecommons.org/licenses/by/4.0/).
C. Kenneally et al. Microbiological Research 259 (2022) 127010

prevalence of recurrent infections results in significant healthcare costs. MacConkey agar, aerobically incubated at 35 ◦C (Hilt et al., 2014).
In the United States alone, the annual economic burden of UTIs is esti­ When diagnosing UTIs, such culture techniques are limited in their
mated to exceed 3.5 billion dollars (Flores-Mireles et al., 2015). detection abilities, favouring the growth of fastidious, aerobic,
Financial costs are not the only burden associated with UTIs, as Gram-negative uropathogens (Wolfe et al., 2012). The diagnostic
antimicrobial resistance (AMR) among uropathogens continues to “gold-standard” relies on a quantitative threshold of ≥ 105 CFU/ml for
evolve at an alarming rate (Hrbacek et al., 2020). Conventional anti­ adequate diagnosis of bacteriuria causing UTIs (Nicolle et al., 2005).
biotic therapy causes long-term alterations in the urobiome, chronic However, the validity of such detection methods is questionable as an
recurring cystitis, and the development of MDR pathogens; often estimated 90% false-negative rate has been reported for standard urine
relating to prolonged episodes of infection (Raz, 2001; Kostakioti et al., culturing (Hilt et al., 2014).
2012). Despite this, broad-spectrum antibiotics remain the first-line Enhanced quantitative urine culture (EQUC) could improve detec­
treatment for UTIs (Bonkat et al., 2020). Consequently, approximately tion of clinically relevant urinary microbes, while additionally
80% of uropathogens now display resistance to at least two antibiotics advancing our understanding of commensal and pathogenic microor­
(Ahmed et al., 2019). Thus, MDR UTIs now represent a real and sub­ ganisms among the urobiome (Price et al., 2016). Whilst the
stantial health threat (Sihra et al., 2018). expanded-spectrum EQUC protocol involves plating increased urine
At present, functional knowledge regarding the mechanistic rela­ volumes and additional growth media with differing oxygen or carbon
tionship between the urobiome and the host remains obscure (Jones-­ dioxide enriched atmospheres, a streamlined adaptation could provide
Freeman et al., 2021). However, resident microbial communities in sufficient information to clinicians when assessing the treatment re­
other body sites are known to play critical roles in maintaining host quirements for uropathogens (Hilt et al., 2014; Pearce et al., 2014; Price
health (Krishnan et al., 2017; Dimitri-Pinheiro et al., 2020; Manor et al., et al., 2016). Blood agar, colistin-nalidixic acid agar, and MacConkey
2020). Further application of systems biology principles and ‘omics’ agar are utilised in the streamlined EQUC protocol and incubated in 5%
technologies could unravel the functional capabilities of the urobiome, CO2, diversifying the detection of urotypes, the dominant taxa in urine
thus, benefitting diagnostic and therapeutic approaches in urogynae­ samples (Price et al., 2016). Urotypes describe community profiles
cology (Bullman et al., 2012). This will likely be of value in clinical within the urobiome, typically dominated in relative abundance by one
practice, potentially identifying diagnostic biomarkers for targeting organism (Gottschick et al., 2017). Additional advancements in DNA
during clinical investigations (Claesson et al., 2017). Moreover, micro­ sequencing have increased the identification of urotypes based on the
organisms derived from such communal ecosystems could play a central relative abundance of operational taxonomic units (OTUs) in cluster
role as natural biological tools, and, when engineered, show promising analysis (Pearce et al., 2015; Ammitzbøll et al., 2021). Only 33% of
potential as novel therapeutic interventions (Sleator and Hill, 2008a, known uropathogens were detected with standard urine culturing, while
2008b). Herein, we review the current knowledge of live bio­ the streamlined EQUC protocol identified 84% (Price et al., 2016).
therapeutics within the urinary tract, and their prospective beneficial Contrasting the ≥ 105 CFU/ml threshold, the EQUC method can
contributions to urinary tract health. We also discuss emerging methods generally detect between 102-105 CFU/ml of urine, useful in the context
and technologies for sequencing the urobiome and the insight such tools of a low biomass community, such as the urinary tract microbiota
have given in enabling us to interpret alterations between healthy and (Mueller et al., 2017).
infected microbial communities of the urinary tract.
3.2. Culture-independent sequencing of the urinary microbiome
2. Description of the methodology
On average, it is estimated that the EQUC protocol captures
A literature search was performed using SCOPUS, PubMed, and approximately 72% of the total microbial genera present in the uro­
Google Scholar from inception to September 2021. Keywords included biome (Thomas-White et al., 2018). The dawn of the ‘omics’ era has
the following terms: ‘Microbiome’ OR ‘Asymptomatic Bacteriuria’ OR provided increased detection capabilities, as DNA sequencing technol­
‘Urobiome’ AND (‘UTI’ OR ‘Interstitial Cystitis’ OR ‘Pyelonephritis’ OR ogies can assess the collective microbial composition inhabiting a spe­
‘Antimicrobial Resistance’ OR ‘Uropathogens’) OR ’UTI’ AND cific environment (Franzosa et al., 2015). Large-scale metagenomic
(‘Asymptomatic Bacteriuria’ OR ‘Probiotics’ OR ‘Bacteriophage’ OR projects have not only led to the detection of novel genes and species,
‘Antimicrobial Peptide’ OR ‘Bacteriocin’). Types of articles retrieved but have also expanded the functional characterisation of the human
included original research articles, animal model studies, randomised microbiome in terms of metabolic pathways and physiological adapta­
controlled trials, clinical trials, and reviews. The initial search identified tions required for survival (Sberro et al., 2019; Dekaboruah et al., 2020).
766 papers, all of which were reviewed by title and abstract, and Two sequencing strategies are commonly employed when assessing
screened for eligibility. Articles excluded were those without an ab­ the microbiome: 16 S ribosomal RNA (rRNA) gene amplicon sequencing
stract, not written in English, and clinical studies lacking information of and whole-genome sequencing (WGS) (Human Microbiome Project
urine sampling techniques. Consortium, 2012; Thomas-White et al., 2016). Combined with urine
culturing, 16 S rRNA gene amplicon sequencing has been more
3. The urinary microbiome commonly employed in studies characterising the urobiome (Karstens
et al., 2021). Nine hypervariable regions (V1-V9) exist in the 16 S rRNA
3.1. Urine culturing gene (Van de Peer et al., 1996). When assessing the phylogenetic
relatedness, the hypervariable regions are utilised as they contain spe­
The establishment of the Human Microbiome Project (HMP) has led cific polymorphisms (Brubaker and Wolfe, 2015; Thomas-White et al.,
to an increased understanding of the role of microbes in human health 2016). Deciding the chosen hypervariable region targeted during 16 S
and disease (Human Microbiome Project Consortium, 2012). Due to the, rRNA gene sequencing is important, as different hypervariable regions
now dated, belief that the urinary tract was a sterile environment, it was might limit the taxonomic identification, causing microbial taxa to be
not included in the HMP as a body site to characterise (Hilt et al., 2014). either under- or over-represented (Mizrahi-man et al., 2013). Both the
Significant limitations are associated with standard urine culture V1 and V6 regions, for example, have reportedly been inadequate when
methods in terms of ability to isolate bacteria, however, optimised compared to other hypervariable regions, demonstrating suboptimal
culturing and culture-independent DNA sequencing technologies have performance in phylogenetic analysis due to limited region length and
revealed the low-biomass microbial community within the urinary tract lower average sequence identities (Yu and Morrison, 2004; Grasp­
(Wolfe and Brubaker, 2015). euntner et al., 2018). A direct comparison evaluating how different
Traditional urine culturing employs 5% sheep blood agar and hypervariable regions perform when capturing the composition of the

2
C. Kenneally et al. Microbiological Research 259 (2022) 127010

urobiome has not yet been conducted (Karstens et al., 2018). WGS can Firmicutes, while Actinobacteria and Bacteroidetes are lacking in the
comprehensively characterise complete genomes by sequencing the male urobiome (Nelson et al., 2010; Lewis et al., 2013; Karstens et al.,
entire DNA sequence (Neugent et al., 2020). Although WGS is a more 2016). Overall, the male urobiome is depicted as a less complex envi­
expensive technique, it generates sequence reads to a higher resolution ronment, typically distinguished from the female urobiome based on the
and usually offers a more detailed taxonomic and functional profile relative abundance of different genera (Fouts et al., 2012; Ceprnja et al.,
(Franzosa et al., 2015). Mutations in the FimH adhesin, for example, 2021). Corynebacterium has been identified as the dominant genus of the
were significantly correlated to the B2 phylogroup of uropathogenic male urobiome, a genus commonly present within the skin microbiome
E. coli (UPEC) isolates, when performing a comparison on faecal and (Fouts et al., 2012). Other bacterial genera commonly identified in both
urinary isolates, using WGS (Nielsen et al., 2017). WGS has also enabled the male and female urobiome include Lactobacillus, Streptococcus, and
the identification of anaerobic members of the Actinobacteria, Firmi­ Staphylococcus (Gottschick et al., 2017). Generally, the urobiome of
cutes, and Bacteroidetes, which were not previously identified using males in the > 70-years cohort appears to be more diverse than that of
culture-only methods (Thomas-White et al., 2018). females, potentially correlated with the increased risk of prostate, kid­
A study by Moustafa et al. (2018) assessed the complementarity of ney, and bladder diseases (Wojciuk et al., 2019). For example, Propio­
the sequencing methods. 16 S rRNA gene sequencing exhibited greater nibacterium acnes strains similar to those present in the male skin
sensitivity when detecting low-abundance DNA in voided urine samples microbiome had a higher abundance in prostate cancer patients, while
(Moustafa et al., 2018). Nevertheless, taxonomic profiling and func­ Enterobacteriaceae and Actinobaculum urinale uropathogens were prev­
tional resolution is limited with 16 S rRNA gene sequencing, whilst WGS alent in benign tumours (Mak et al., 2013; Shrestha et al., 2018). A
provides an increasingly specific functional and taxonomic classification cluster of proinflammatory bacteria including Streptococcus anginosus,
(Jovel et al., 2016). However, WGS can be subject to amplifying con­ Anaerococcus spp., Varibaculum cambriense, and Propionimicrobium lym­
taminants attained from the human host, generally from the skin or phophilum have also been linked to urological infections and malignant
vaginal microbiomes, possibly as a result of urine sampling methods tissues in males (Domann et al., 2003; Williams, 2015; Shrestha et al.,
(Moustafa et al., 2018). A limitation associated with both sequencing 2018). Microorganisms present in the male urobiome seem to resemble
methods is that non-viable, ruptured bacteria can be detected (Karstens those present in the skin microbiome, whilst the female urobiome ex­
et al., 2018; Boers et al., 2019). It is hypothesised that the identification hibits a commonality with the vaginal microbiome (Fouts et al., 2012;
of non-viable bacteria is still beneficial as they once played a role within Lewis et al., 2013).
the microbiome, potentially providing increased functional knowledge Urotypes in the female urobiome are commonly dominated by
(Aragón et al., 2018). However, this could lead to misinterpretation Shigella, Lactobacillus, Enterococcus, Gardnerella, Prevotella, Sneathia,
when assessing the urobiome’s extant state. Therefore, a combined use Escherichia, and Streptococcus (Gottschick et al., 2017; Price et al.,
of EQUC and DNA sequencing technologies is recommended, where 2020a). Lactobacillus is identified as the dominant genus in a healthy
EQUC will identify live microorganisms (Karstens et al., 2018). female urobiome (Hilt et al., 2014; Pearce et al., 2015). Lactobacillus is
Both techniques have identified as-yet-uncultured organisms from also abundant in the vaginal microbiome, with certain species contrib­
urinary samples (Fouts et al., 2012; Garretto et al., 2018; Cummings uting to a healthy vaginal state, when present at adequate levels,
et al., 2020). Urine samples analysed with 16 S rRNA gene sequencing maintaining a lower pH and protecting against pathogens (Marrazzo,
contained the Atopobium genus, which was not identified using culturing 2011). A similar function has been proposed for Lactobacillus in the
methods (Aragón et al., 2018). Non-prokaryotic organisms are also more urobiome, correlating with a decrease of the genus in postmenopausal
frequently detected using WGS compared to culture-only protocols, women and the subsequent increase of recurrent UTIs (rUTIs) (Staple­
namely the eukaryotic yeast species Candida (Moustafa et al., 2018). ton, 2016). Inhibition of uropathogen growth has also been shown in the
Atopobium and Candida have previously been identified within the presence of lactic acid and hydrogen peroxide; by-products of Lactoba­
vaginal microbiome, where increased abundance correlates with the cillus rhamnosus and Lactobacillus reuteri metabolism (Cadieux et al.,
onset of vaginal infection. Atopobium has been identified in samples 2009). Low-to-moderate levels of Lactobacillus in premenopausal women
derived from individuals suffering with bacterial vaginosis (Ferris et al., also correlates with urinary incontinence, indicating an imbalance from
2004) and Candida species have been associated with sexually trans­ the healthy microbiome (Komesu et al., 2018). Whilst taxonomic
mitted diseases and vaginal thrush (Moustafa et al., 2018). Most studies profiling of the urobiome has advanced significantly, it is still not known
surrounding the urobiome thus far have been limited in size, focused on how temporal factors, such as sex hormones, menstrual cycle, and sexual
taxonomic profiling with 16 S rRNA gene sequencing, and thus knowl­ intercourse contribute in altering the microbial composition (Gümüş
edge regarding the functionality of members of the urobiome remains et al., 2019; Price et al., 2020b). Early studies focused on the temporal
limited (Jones-Freeman et al., 2021). dynamics of the female urobiome have determined a likeness to the skin
microbiome during menstruation, and an increase in Staphylococcus and
3.3. Profiling the urinary microbiome Streptococcus linked to sexual intercourse (Price et al., 2020b).
Gardnerella is another genus frequently isolated from urine samples,
Urine is typically comprised of bacteria from four phyla: Firmicutes, more specifically Gardnerella vaginalis (Pearce et al., 2014, 2015; Kars­
Bacteroidetes, Proteobacteria, and Actinobacteria (Karstens et al., tens et al., 2016). Gardnerella is also reportedly increased in the vaginal
2016). A ‘core’ urobiome is being elucidated, however, most studies to microbiome post sexual intercourse (Gajer et al., 2012). G. vaginalis is
date, analysing the urinary tract have focused on the female urobiome, abundantly present in females suffering with bacterial vaginosis, and is
due to the wide range of urological disorders affecting women associated with an increased risk of women developing a UTI (Ravel
(Schneeweiss et al., 2016; Brubaker and Wolfe, 2017). Discrepancies et al., 2011; Morrill et al., 2020). Gilbert and colleagues (2017), using a
also persist among urine sampling techniques, whereby voided urine is mouse model, determined that G. vaginalis can function as a driving
often subject to post-urethral contamination (Wolfe et al., 2012; Bru­ factor causing the re-emergence of UPEC from latent intracellular res­
baker et al., 2021). Additionally, a recent study has also unveiled dif­ ervoirs. This suggests that G. vaginalis overgrowth is a plausible trigger
ferences between bacteria detected in urine samples and bladder for pyelonephritis in women, causing increasingly serious manifesta­
mucosal tissue samples (Mansour et al., 2020). Capturing tissue samples tions, resulting in severe kidney infections and epithelial exfoliation
along with urine would give further insight into various bladder dis­ (Gilbert et al., 2017; O’Brien et al., 2020). High abundance of Gard­
eases, however sampling techniques are more invasive (Mansour et al., nerella in the female urobiome is associated with a subsequent decrease
2020). Nevertheless, differences between the male and female urobiome in Lactobacillus species, however a dysbiotic relationship has not been
have been reported (Gottschick et al., 2017). verified due to the presence of Gardnerella in healthy individuals (Pearce
Both the male and female urobiome are dominated by the phylum et al., 2014). Large-scale, longitudinal studies will be required to further

3
C. Kenneally et al. Microbiological Research 259 (2022) 127010

evaluate the implications of alterations in genus-level abundances and to increasing the risk of rUTIs (De Nisco et al., 2019).
identify promising biomarkers. UPEC strains and filamentous uropathogens involved in the forma­
tion of IBCs are highly pathogenic, contributing to re-infection, resis­
tance to antimicrobials, and infection chronicity (Hannan et al., 2012;
3.4. Modulation of the urinary microbiome during UTI McLellan and Hunstad, 2016). Phylogenetic analysis typically divides
E. coli strains into four main groups (A, B1, B2, and D), however updated
3.4.1. Recurrent UTIs PCR-based methods recognise three additional groups (designated C, E,
UPEC dominates UTI pathogenesis, being detected in 80% of clinical and F) (Clermont et al., 2000). Phylogroup B2 and D are associated with
urine samples tested using standard culture techniques (Klein and virulent UPEC strains (Schreiber et al., 2017; Biggel et al., 2020).
Hultgren, 2020). However, the detection of emerging uropathogens is Notably, UPEC clones originating in the gut derived from phylogroup B2
reducing the heretofore E. coli-centric depiction of UTIs (Price et al., and D possess a higher level of multidrug resistance (Nielsen et al.,
2018; Hochstedler et al., 2021). Several species, including Pseudomonas 2014a). UPEC presence in faeces demonstrates that the urogenital and
aeruginosa, Enterobacter aerogenes, Klebsiella pneumoniae, Aerococcus gut microbiomes are not mutually exclusive. Forde et al. (2019) evalu­
urinae, Proteus mirabilis, and Streptococcus anginosus are increasingly ated the population dynamics of a virulent E. coli ST131 lineage from
identified by modern DNA sequencing methods as causative agents of voided urine and faecal samples, over a 5-year period, to understand
UTIs (Price et al., 2016, 2018). A comparison of the urobiome in a rUTI aetiology. An individual harboured an intestinal reservoir of
healthy state and during UTI is presented in Fig. 1. UTI-inducing E. coli ST131 variants causing reoccurrence over a 5-year
UPEC pathogenesis remains the primary concern for recurring in­ period, further linking a relationship between the gut and urobiome
fections. Reinfection occurs in approximately 25%− 30% of women (Forde et al., 2019). Interestingly, the ST131 clone conferred resistance
within 6 months of a preceding UTI (Vagios et al., 2020). The pathobi­ against more antibiotics initially, losing multiple resistance plasmids by
ology of rUTIs is still not fully understood, and longitudinal meta­ the end the fifth year (Forde et al., 2019). The authors suggested that the
genomic analysis is lacking in rUTI patients. One proposed mechanism is loss of resistance plasmids was a mechanism of adaption to fluctuating
that reinfection evolves from UPEC populating the gastrointestinal tract antibiotic regimens (Forde et al., 2019). This warrants a requirement for
or urogenital epithelial reservoirs (Nielsen et al., 2014a). UPEC isolates temporal and long-term analysis of the evolution of bacteriuria in IBC’s
colonise the periurethral area, subsequently moving to the urethra and and the impact on the urobiome. Persistent community alterations
ascending through to the bladder during UTI pathogenesis (Flores-Mir­ appear to correlate with an increase in latent reservoirs amongst the
eles et al., 2015; Terlizzi et al., 2017). After adherence to urogenital elderly, and despite antibiotic therapy, > 60% of rUTIs are attributed to
epithelial cells, UPEC can invade the cytosol causing aggregation and the original uropathogenic strain (Russo et al., 1995; Thänert et al.,
formation of intracellular bacterial communities (IBCs), typically within 2019; Nielsen et al., 2021).
a biofilm-like matrix (Hirakawa et al., 2019). The presence of IBCs have
been detected in individuals suffering from acute cystitis and rUTIs 3.4.2. Polymicrobial UTIs
(Rosen et al., 2007; Robino et al., 2013). Schreiber and team (2017) Standard urine culture relies on the belief that UTIs are generally
detected same-strain rUTIs in 12 out of 18 rUTI incidences, likely monomicrobial infections (Smelov et al., 2016). This has led to the
resulting from latent reservoirs. Internal uropathogen reservoirs appear concept of ‘single-species UTIs’ in clinical laboratories, deeming
to persist in the bladder and vaginal walls of postmenopausal females;

Fig. 1. Comparison of the urinary microbiome during a healthy state and UTI. (A) Most bacteria, such as Prevotella and Shigella, are commonly found in the
urobiomes of both healthy males and females. Lactobacillus and Corynebacterium species dominate the urobiomes of females and males, respectively. (B) Overview
of risk factors predisposing UTI pathogenesis. Immunocompromised, elderly, indwelling devices, and urogenital tract infections or previous antibiotic use greatly
increase the risk of infection. (C) UTIs are associated with increased abundance of common uropathogens such as E. coli, Enterococcus and Staphylococcus and a
subsequent decrease in Lactobacillus (arrow). Gardnerella species are also frequently associated with infection.

4
C. Kenneally et al. Microbiological Research 259 (2022) 127010

multiple colony morphologies as contamination (Kline and Lewis, urogenital tract are yet to be elucidated. Proposed explanations behind
2016). Despite a lack of studies surrounding polymicrobial UTIs, it is such microbial interplay include metabolic signalling interactions that
hypothesised that such infections are more common than previously are often accompanied by upregulation of virulence factors, reduced
assumed (Fourcade et al., 2015). Approximately 20% of women with wound healing, and altered antibiotic susceptibility (Pastar et al., 2013;
symptomatic UTIs result in culture-negative urine samples (Ferry et al., De Vos et al., 2017; Trizna et al., 2020).
2007). Culture-negative is a misleading term as it includes samples with Accumulating evidence suggests that polymicrobial interactions
polymicrobial growth, and pathogens present at < 105 CFU/ml (Kline heighten antimicrobial resistance, ultimately by hindering antibiotic
and Lewis, 2016). One study reported that 42% of women presented efficacy (Orazi and O’Toole, 2020). In turn, biofilm-mediated resistance
with polymicrobial infections, characterised by the presence of at least enables polymicrobial communities to withstand high concentrations of
one Gram-positive and one Gram-negative bacterium (Kline and Lewis, antimicrobial agents (Gaston et al., 2020). The extracellular matrix also
2016; Price et al., 2018). It is also estimated that 33% of UTIs among the offers additional resistance mechanisms, containing β-lactamases and
elderly are polymicrobial (Croxall et al., 2011). Polymicrobial UTIs are degrading antibiotics upon entry, before they can reach their target
frequently described among the immunocompromised and individuals bacterium (Høiby et al., 2010). For instance, E. coli isolates obtained
with indwelling catheters (Kline and Lewis, 2016). Werneburg and from polymicrobial UTIs appear more invasive and display increased
co-authors (2020) detected polymicrobial uropathogenic biofilms on resistance to antibiotics such as ciprofloxacin and trimethoprim (Croxall
catheters as early as 7-hours post installation, which further progressed et al., 2011). Depending on interspecies interactions, mixed-species
as indwelling time increased resulting in biofilms on the proximal and biofilms either enhance resistance or exhibit tolerance towards antimi­
distal portions by 26 days on average. As a result, these infections either crobial therapy; a phenomenon termed “recalcitrance” (Lebeaux et al.,
go untreated or are subject to inappropriate antibiotic interventions. The 2014). Enterococcus and Proteus species regularly co-colonise the urinary
damaging effects of this have also been noted by Werneburg et al. (2020) tract and indwelling catheters, and are notorious biofilm producers
whereby four out of ten sequence samples were noted to confer (Fourcade et al., 2015; Armbruster et al., 2017). Alarmingly, P. mirabilis
beta-lactam, quinolone, aminoglycoside, and tetracycline resistance and E. faecalis biofilms severely limit antimicrobial agents, conferring
genes. resistance to β-lactam antibiotics, aminoglycosides, fluoroquinolones,
Detrimental effects of antibiotics on the genitourinary microbiome and carbapenems (Ch’ng et al., 2019; Gaston et al., 2020). Additionally,
prompting the onset of polymicrobial UTIs and rUTIs have previously urease production is primarily associated with P. mirabilis, causing urine
been proposed. Discrepancies were demonstrated in an earlier study by obstruction and crystalline biofilm formation on catheters (Soto, 2014;
Rani et al. (2017) analysing the bladder microbiome of kidney trans­ Pelling et al., 2019). The crystalline layer also provides heighted pro­
plant patients treated with prophylactic trimethoprim-sulfamethoxazole tection to multi-species biofilms on catheters, providing resistance
(TMP-SMX) via metagenomic sequencing. Two opportunistic pathogens, against pre-coated antimicrobial agents (Goh et al., 2017). The conse­
Enterococcus faecalis and E. coli, both showed increased abundance when quences of such interspecies interactions can lead to metabolite
sequenced after treatment, whilst a subsequent decrease in general mi­ cross-feeding and altered susceptibility towards antibiotics (Short et al.,
crobial diversity was observed (Rani et al., 2017). Interestingly, 2014; Bottery et al., 2021), contributing to enhanced disease progres­
E. faecalis and UPEC have previously been reported to cause poly­ sion and adverse patient outcomes compared to monomicrobial
microbial infections within the urinary tract and surgical wounds infections.
(Keogh et al., 2016). A synergistic relationship has been described be­
tween the two pathogens, upon which L-ornithine is secreted by 4. Antibiotic resistance
E. faecalis and subsequently stimulates E. coli’s iron uptake system,
efeUOB complex, in iron-limiting environments; thus, E. coli growth and UTIs have been disproportionately affected by antibiotic misuse
biofilm formation are increased (Keogh et al., 2016). E. coli has also (O’Grady et al., 2019), increasing the prevalence of MDR uropathogens
previously exhibited synergism with the yeast Candida albicans, simul­ (Mazzariol et al., 2017). Currently, the clinical guidelines that define
taneously increasing one-another’s growth, possibly an aetiological UTIs lends itself to equivocal interpretations, often leading to unnec­
factor in fungal UTIs (De Brucker et al., 2015). However, not all inter­ essary administration of broad-spectrum antibiotics (Finucane, 2017).
species interactions are beneficial to pathogens. For instance, Surveillance data accumulated by The Global Prevalence of Infections in
P. aeruginosa displays dominance over E. coli, causing decreased growth, Urology (GPIU) suggests that approximately 56% of patients in urology
likely due to secretion of virulence factors such as N-acyl-L-homoserine units were prescribed antibiotics, of which 26% were prescribed for
lactones (Qin et al., 2009; Cerqueira et al., 2013). Generally, there is a proven UTIs, while 21% were for suspected UTIs (Johansen et al., 2006;
lack of comprehensive studies surrounding polymicrobial relationships Wagenlehner et al., 2016). Ultimately, the consequences for over­
within the urobiome, thus, interpretations of microbial interplay largely prescribing antibiotics for non-confirmed UTIs correlates to a significant
remain obscure (Azevedo et al., 2017). increase in MDR uropathogens (Cek et al., 2014). Consequentially,
guidelines for antibiotic prescriptions for UTIs have been updated to
3.4.3. Polymicrobial Biofilms reflect the rise in resistance. Empiric fluoroquinolone treatment is now
Biofilm formation is a common attribute associated with poly­ only recommended if the frequency of local resistance in E. coli is ≤ 10%
microbial communities, resulting from the assemblage of a collection of and if the patient has not received fluoroquinolone treatment within the
organisms enclosed within an extracellular polymeric substance matrix last 6 months (Gupta et al., 2011; Bonkat et al., 2020).
(Penesyan et al., 2020). Microbial culture and culture-independent ex­ Long-term modifications to microbiome constituents are also
aminations of catheter-associated UTIs (CAUTI) have determined that impacted by antibiotic regimens. Antibiotics commonly employed for
biofilms contaminating urinary catheters are usually polymicrobial in UTIs are said to impact the gut microbiome, decreasing diversity and
nature (Frank et al., 2009; Galván et al., 2016). Holá et al. (2010) re­ abundance of commensals (Elvers et al., 2020). Studies investigating the
ported similar observations when only 12.5% of CAUTI were mono­ influence of antibiotics on the urobiome are limited, yet still concerning.
microbial. However, their findings relied on culture techniques, and as Antibiotics have been noted to significantly contribute to communal
such, it is likely that metagenomic methods could lead to the reported variations in the urobiome (Gottschick et al., 2017; Mulder et al., 2019).
12.5% being further reduced. Organisms previously associated with Gottschick et al. (2017) evaluated a two-fold increase in Lactobacillus
polymicrobial infections identified on urinary catheters include iners post metronidazole treatment, and the relative abundance of
E. faecalis, P. mirabilis, and K. pneumoniae (Frank et al., 2009; Fourcade Gardnerella vaginalis, Atopobium vaginae and Sneathia amni was signifi­
et al., 2015; Armbruster et al., 2017; Werneburg et al., 2020). The un­ cantly decreased. Although Lactobacillus are typically acknowledged for
derlying mechanisms of most interspecies interactions within the their health-optimizing characteristics, L. iners is controversial relating

5
C. Kenneally et al. Microbiological Research 259 (2022) 127010

to significant abundance in diseases such as bacterial vaginosis (Petrova prone to rUTIs, where one group was subject to antibiotic therapy while
et al., 2017). When investigating the effect of several antibiotics on the participants in the control group were not. The frequency of reoccur­
urobiome, Mulder et al. (2019) noted a subsequent increase in E. coli and rence was significantly higher in the group taking antibiotics (46.8%)
Parabacteroides abundance in voided urine samples, while Finegoldia and compared to the control group (13.1%), and presence of ABU was sug­
Lactobacillus were decreased. Lack of oestrogen in postmenopausal gested to play a protective role (Cai et al., 2012). Cai et al. (2012),
women also decreases Lactobacillus abundance in the urobiome whilst (2015) also concluded that E. coli isolates from the antibiotic group
simultaneously enhancing the diversity of uropathogens, increasing the displayed significantly higher resistance against amoxicillin–clavulanic
prevalence of infection and frequency of antibiotic treatment (Alperin acid, ciprofloxacin and TMP-SMX. Hence, treating ABU with antibiotic
et al., 2019). Allowing such women access to oestrogen therapy can aid therapy is likely detrimental and might, in fact, damage the urobiome.
in postmenopausal issues, including increasing Lactobacillus prolifera­ Thus, the European Association of Urology (EAU) and American Uro­
tion within the urogenital microbiome (Gliniewicz et al., 2019). How­ logical Association (AUA) recommend avoid treating ABU unless
ever, it is yet to be associated with the complete prevention of UTIs (Raz, examining a particular subgroup; pregnant women and individuals un­
2001; Thomas-White et al., 2020). Thus, developing new therapeutics dergoing surgery in which the bladder mucosa will be breached (Anger
for UTIs is warranted as generally the archetypal resident urobiome et al., 2019; Bonkat et al., 2020).
microbial population cannot be restored after responding to antibiotic
therapy (Gottschick et al., 2017). 6. Potential BioTherapeutics

5. Asymptomatic bacteriuria versus symptomatic UTI Table 1 summarises the biotherapeutics that have potential to
modulate the urobiome, while an overview of the mechanisms of action
Asymptomatic bacteriuria (ABU) is defined as two consecutive pos­ is presented in Fig. 2.
itive urine specimens (≥105 CFU/ml) isolated from an individual lack­
ing symptoms attributable to a UTI (Nicolle et al., 2005). E. coli
6.1. Bacterial Interference
predominantly cause ABU (Roos et al., 2006a; Dobrindt et al., 2016).
ABU prevalence increases with age, occurring in 20% of women by the
E. coli 83972 is currently the most well characterised strain associ­
age of 80%, and 3.6%− 19% in community-dwelling elderly men after
ated with ABU (Darouiche et al., 2005; Köves et al., 2014; Stork et al.,
60 years (Nicolle, 2003; Roos et al., 2006c). The frequency increases to
2018). E. coli 83972 was first isolated from a young girl, who harboured
25–50% in women, and 15–40% in men in long-term care facilities;
the strain for ~3 years without displaying symptoms of infection
likely due to complications such as impaired functional status and uri­
(Lindberg et al., 1975; Andersson et al., 1991). Although E. coli 83972
nary incontinence, ultimately leading to long-term catheterisation
resembles commensal E. coli, studies indicate that the strain evolved
(Nicolle, 1999; Leihof et al., 2021). However, a misunderstanding sur­
from a uropathogenic progenitor (Klemm et al., 2006). Indeed, the strain
rounds ABU, and as to whether such strains should be considered as
has been linked to the E. coli B2 phylogenetic group, the predominant
commensal organisms or potential pathogens due to their phylogenetic
phylogroup associated with UPEC strains (Roos et al., 2006b). Virulence
relatedness to uropathogens causing symptomatic infection (Finucane,
factors associated with UPEC strains that are present in E. coli 83972
2017).
include the expression of colibactin and cytotoxic necrotizing factor 1
Underlying and confounding medical issues predispose institution­
(CNF-1) (Stork et al., 2018). However, mutations differentiating the
alised adults to developing a higher frequency of ABU strains (Ajayi and
83972 ABU strain from E. coli strains that cause symptomatic infection,
Radhakrishnan, 2016). Simultaneously, this increases antibiotic overuse
including lack of motility, and the absence functional P, type 1, and F1C
as UTIs are diagnosed based on the presence of ≥ 105 CFU/ml bacteri­
fimbriae due to fim deletions and papG point mutations, are factors in
uria in urine samples (Nicolle et al., 2005). Thus, long-term care facil­
attenuating its virulence (Klemm et al., 2006; Roos et al., 2006b;
ities have become hotspots for the spread of resistant uropathogens
Zdziarski et al., 2008, 2010).
(Rowe and Juthani-Mehta, 2014). ABU strains present in the urobiome
Bacterial interference can be established with E. coli 83972 as the
typically present in a state of commensalism; not causing symptoms in
strain outcompetes UPEC isolates, decreasing UTI risk. E. coli 83972
their host. Cai et al. (2012) evaluated the influence of ABU in women
colonisation, as opposed to UPEC, has been previously documented

Table 1
Biotherapeutics displaying antimicrobial activity against uropathogens.
BiotherapeuticName Type Mode of Action Clinical Evidence Reference

E. coli 83972 Asymptomatic Bacterial interference.Competition for nutrients. Phase 2 – significant reduction of rUTIs Sundén, Bonkat et al.,
Bacteriuria Biofilm interference. Bacteriocin production. (N = 20). No active phase 3. Approved for use (2010, 2020)
in LUTD by the EAU.
Pyophage Bacteriophage Virus infecting bacteria. Phase 3 – non-inferior to antibiotics (N = 113). Leitner et al. (2021a)
RCT commencing in spinal cord patients. Leitner et al. (2021b)
Lactin-V (Lactobacillus Probiotic Lactic acid and hydrogen peroxide production. Phase 2 – rUTI incidence rate reduced to 15% Stapleton et al. (2011)
crispatus) Blocks adherence to uroepithelial cells. (N = 50). No active phase 3.
L. rhamnosus GG Probiotic Lactic acid and hydrogen peroxide production. Phase 2 – 1 or 2 doses safe in NLUTD and SCI Groah et al. (2019)
Downregulates NF-κB, P, and Type 1 fimbriae. patients (N = 80). No active phase 3.
Biofilm interference.
Mutaflor (E. coli Nissle Probiotic Microcin production. Competition for iron using In Vitro – no significant UPEC reduction.Phase Pradhan and Weiss
1917) iron uptake systems. 4 in active development for UTIs children. (2020)Tapiainen
(2020)
Faecal Microbiota Donor Stool Bacterial interference. Preliminary clinical study – significant Ghani et al. (2021)
Transplantation Transplant reduction of rUTIs (N = 90).
Cecropin A Antimicrobial Membrane permeabilization.Inhibits efflux pump In Vivo – significant UPEC biofilm reduction in Fenner, (2020)Kalsy
Peptide and nucleic acids interactions. Biofilm interference. G. mellonella model. et al. (2020)
Colicin E2 Bacteriocin Endonucleolytic degradation of DNA. Preliminary catheter trials – complete Trautner et al. (2005)
inhibition of susceptible E. coli.

*rUTIs: recurrent urinary tract infection, LUTD: lower urinary tract dysfunction, EAU: European Association of Urology, RCT: randomised controlled trial, NLUTD:
neurogenic lower urinary tract dysfunction, SCI: spinal cord injury.

6
C. Kenneally et al. Microbiological Research 259 (2022) 127010

Fig. 2. Mechanisms of action of biotherapeutics against uropathogens including probiotics, bacteriocins and antimicrobial peptides. (A) Probiotic bacteria are
competitive microorganisms fighting for nutrients and urothelium adherence, and interfering with biofilms. Production of lactic acid regulates the pH of the
environment, providing an unsubstantial medium for uropathogen survival. (B) Bacteriocins act as toxins against competing bacteria, degrading their DNA, inhibiting
peptidoglycan synthesis, and/or forming pores in membranes. Small molecules, amino acids, and ions leak from the cell, inducing cell death. (C) Efflux pumps can be
inhibited by additional antimicrobial peptides, affecting transport of toxic compounds and bacterial metabolites.

7
C. Kenneally et al. Microbiological Research 259 (2022) 127010

through pre-inoculation of urinary catheters and deliberate establish­ previously proposed the concept of urinary microbiota transplantation
ment in biofilms (Trautner et al., 2003; Ferrières et al., 2007). The (UMT) to target rUTIs (Jones et al., 2021). Enhancing this trans­
clinical effectiveness of the strain has been investigated in small scale plantation therapy with health-associated strains such as ABU or
studies, where introduction of E. coli 83972 into neurogenic bladders Lactobacillus could perhaps identify a novel biological therapy for rUTIs.
reduced the reoccurrence of UTIs (Sundén et al., 2010). There is limited
knowledge regarding how the ABU outcompetes UPEC, with suggestions 6.3. Phage therapy
including outcompeting for nutrients and attachment sites, gene
acquisition, and inhibiting biofilm production (Darouiche and Hull, Whilst relatively less is known about the urobiome when compared
2012; Beatson et al., 2015; Stork et al., 2018). Competition for attach­ to other human microbomes, even fewer studies have focused on the
ment sites has been disproved, as E. coli 83972 lacks the fimbriae viral constituents of the urinary tract, otherwise known as the urinary
required for adherence (Hull et al., 2002; Roos et al., 2006b; Zdziarski virome (Santiago-Rodriguez, 2018). Although shifts within the uro­
et al., 2010). Despite confusion surrounding the exact mechanism of biome community are significantly affected by health status, viral con­
action, clinical use of E. coli 83972 is endorsed for deliberate bladder stituents appear to be unaltered (Santiago-Rodriguez et al., 2015).
colonisation by the European Urology Guidelines (Dobrindt et al., 2016; Presently, metagenomic analysis of the urinary virome has primarily
Bonkat et al., 2020). Aside from the prototype 83972 strain, additional focused on eukaryotic viruses, where Human Papilloma Virus (HPV) is
ABU strains are not as well characterised. A study by Stork et al. (2018) the most abundant (Shigehara et al., 2010; Rani et al., 2016). Pre­
showed that two ABU isolates (strain 9 and 106) displayed an increased liminary metagenomic analysis has revealed that novel phage sequences
competitive advantage against two model strains, E. coli 83972 and the in the urinary virome exceed eukaryotic viruses (Miller-Ensminger et al.,
UPEC 536 strain. In total, nine ABU strains were evaluated, however as 2018). For instance, Santiago-Rodriguez and colleagues (2015) have
they were only competing against two model strains, their performance determined that 27% of viral sequences in urine were homologous to
against other uropathogenic strains remains unelucidated (Stork et al., known viruses, out of which 99% represented bacteriophage genes.
2018). Both strain 9 and 106 appeared to contain additional fitness However, this only represents a fraction of phages present, meaning a
factors, such as bacteriocin expression and siderophore aerobactin sys­ large unexplored viral community exists in the urinary tract (Miller-­
tems, potentially contributing to their interference (Stork et al., 2018). Ensminger et al., 2018; Garretto et al., 2019).
Deliberate colonisation with E. coli 83972 in humans is regarded as safe, Currently, studies surrounding phage therapy for UTIs are typically
thus larger studies with additional ABU strains could lead to further comprised of lytic proteins, phage cocktails, or phages used synergisti­
development of novel biotherapeutics (Köves et al., 2014; cally with antibiotics (Malik et al., 2020). Lytic phages naturally pro­
Jones-Freeman et al., 2021). duce two types of lytic proteins targeting lysis of the cell wall; endolysins
and virion-associated peptidoglycan hydrolases (PGH), also referred to
6.2. Microbiota transplantation as virion associated lysins (VALs) (Gutiérrez et al., 2018). Additionally,
lytic phages are reported as potential therapies for MDR uropathogens
Faecal microbiota transplantation (FMT) involves transferring the (Galtier et al., 2016; Bagińska et al., 2021). Sybesma and co-authors
microbial community in a suspension of minimally manipulated stool (2016) investigated the in vitro susceptibility of nine urine derived
from a healthy donor into the intestinal lumen of a patient, restoring a K. pneumoniae isolates to lytic phages. Notably, significant activity was
healthy microbial composition (Culligan and Sleator, 2016). Recently, displayed by one phage, v_BR–KpS10, as 100% of K. pneumoniae isolates
FMT therapy has repopulated commensals in the urobiome, which has were lysed (Sybesma et al., 2016). More recently, the lytic phage
been associated with a reduction of rUTIs. In a small case study per­ VB_ecoS-Golestan has displayed activity against troubling UPEC strains,
formed by Tariq et al. (2017), a significant decrease of rUTI incidences inhibiting 56% of MDR UPEC isolates (Yazdi et al., 2020). Restricting
was detected after FMT treatment for recurrent Clostridium difficile factors, such as limited host range and phage-resistant mutants, do limit
infection (rCDI). Susceptibility to antibiotics subsequently increased, the efficacy of single phage therapies for UPEC treatment (Malik et al.,
and abundance of E. coli and Klebsiella uropathogens reduced (Tariq 2020).
et al., 2017). Biehl et al. (2018) detected similar reductions in Entero­ The high specificity phages display in their single-phage form not
bacteriaceae from an abundance of 8.3–0.5% 84 days after FMT in a only restricts their ability to treat polymicrobial infections, but also in­
50-year-old female, while Lactobacillaceae abundance increased from creases the risk of phage resistant bacteria evolving (Gurney et al., 2017;
~0.5% to > 50%. Similarly, in a 93-year-old female, FMT reduced Wright et al., 2019). Thus, phages in combination with another anti­
symptomatic rUTIs and eradicated rCDIs (Aira et al., 2021). The authors microbial, and phage cocktails are typically preferred. Polyphage and
reasoned the vast reduction in symptomatic infections resulted from the combination therapies are particularly advantageous when treating
reduction of Enterobacteriaceae in the gut from 74% pre-FMT to 0.07% polymicrobial infections and biofilms (Malik et al., 2020). For instance,
post-FMT (Aira et al., 2021). FMT has shown positive results in a slightly a phage cocktail targeting MDR Acinetobacter baumannii led to > 90%
larger cohort of 9 patients in rUTI reduction, with only one episode of reduction in biofilm biomass when combined with TMP-SMX and cip­
recurrence from ESBL-producing strains (Ghani et al., 2021). Pre­ rofloxacin (Grygorcewicz et al., 2021). The synergistic effect typically
liminary case studies to date have investigated FMT in few participants results from the antibiotics’ mode of action, inhibiting dihydrofolate
and large-scale studies would benefit the confirmation of this therapy. reductase (DHFR), DNA topoisomerase II, and DNA topoisomerase IV,
FMT’s renowned success in reducing rCDI has opened the question as and provoking faster phage expansion and cell lysis (Chegini et al.,
to whether microbial transplantation from other niche’s would be an 2021). Synergism has also been reported between antipseudomonal
effective therapy. Vaginal microbiota transplantation (VMT) has not phages and a genetically modified ABU strain, E. coli HU2117, a deriv­
been trialled in individuals suffering with rUTIs, but has been presented ative of E. coli 83972 with an 800-base pair deletion in chromosomal
as a potentially viable treatment for bacterial vaginosis (Lev-Sagie et al., papG (Liao et al., 2012). As a result of the chromosomal gene deletion, P
2019). This was the first reported VMT performed in humans, at the time fimbriae, virulence factors associated with bacteremia and pyelone­
of writing, and yielded in long-term remission of bacterial vaginosis in 4 phritis (Hull et al., 2002), are no longer produced by E. coli HU2117.
out of the 5 participants (Lev-Sagie et al., 2019). Additionally, a Lacto­ Liao and co-authors (2012) reported an almost 4-log reduction in
bacillus-dominated microbiome was restored after VMT, re-forming P. aeruginosa biofilm formation on catheters when E. coli HU2117 and
healthy vaginal microbiome configurations (Lev-Sagie et al., 2019). the phage (ΦE2005-A) were combined, whilst neither displayed signif­
Given the interrelatedness of the vaginal microbiome with the uro­ icant inhibition alone. However, the effectiveness of antibiotic-phage
biome, it could be explored whether this treatment strategy could be combinations are highly dependent on the chosen antibiotic and bac­
viable for rUTIs and other urological diseases. On a similar note, we have terial pathogen, as demonstrated by the antagonistic activity seen

8
C. Kenneally et al. Microbiological Research 259 (2022) 127010

between a MDR A. baumannii phage cocktail and colistin (Grygorcewicz (Stapleton et al., 2011). Additionally, L. rhamnosus is one of the most
et al., 2021). Phage binding and reproduction were limited when com­ frequently studied Lactobacillus strains (Petrova et al., 2018; Pino et al.,
bined with colistin due to the degradation of the cell membrane (Dan­ 2019). It promotes the downregulation of UPEC virulence factors
is-Wlodarczyk et al., 2016). including nuclear factor-κB (NF-κB) eukaryotic transcription factors, P
Pyophage, a commercial phage produced by the Eliava Institute and type 1 fimbriae, and also interferes with biofilms (Cadieux et al.,
(Tbilisi, Georgia), has been investigated in initial clinical studies for the 2009; McMillan et al., 2011; Karlsson et al., 2012; Petrova et al., 2016).
treatment of UTI-causing uropathogens when administered (Leitner Randomised controlled trials were conducted with 252 postmenopausal
et al., 2017). Pyophage is a phage cocktail that typically targets E. coli, females, evaluating the efficacy of an oral probiotic containing
Pseudomonas, Proteus, Staphylococcus and Streptococcus strains causing L. rhamnosus GR-1 and L. reuteri RC-14 against antibiotic therapy
wound or skin infections (Abedon et al., 2011). In a primary in vitro (Beerepoot et al., 2012). When compared against TMP-SMX the pro­
clinical study, the commercial Pyophage was adapted after a series of biotic did not meet the non-inferiority criteria for UTI inhibition, how­
liquid titrations to determine the lowest concentration affective against ever, presence of antibiotic resistant uropathogens were significantly
bacteria (Ujmajuridze et al., 2018). Subsequently, sensitivity of bacteria reduced within the probiotic cohort (Beerepoot et al., 2012). Vaginal
to a 4-fold adapted Pyophage increased from 41% to 75% (Ujmajuridze administration of a L. rhamnosus GR-1 and L. reuteri RC-14 probiotic has
et al., 2018). Furthermore, the adapted phage on a catheter caused a previously produced promising results; reducing UTI reoccurrence from
1–5 log reduction in bacterial titers in six out of nine participants, 47% to 21% when used alongside antibiotics (N = 41) (Reid et al.,
whereby E. coli was detected in four participants, and both Streptococcus 1992). Additionally, Lactobacillus acidophilus’ inhibitory effects against
spp. and Enterococcus spp. were only present in two participants post E. coli displayed the highest inhibitory potential (60%), warranting
treatment (Ujmajuridze et al., 2018). Recently, in a randomized further investigation into the strain (Velraeds et al., 1998; Vagios et al.,
controlled clinical trial consisting of 113 patients, Leitner and team 2020). A new era of probiotic administration could be on the horizon, as
(2021b) reported that the efficacy and safety of the adapted intravesical intravesical delivery of lactobacilli have shown promising potential in
Pyophage is non-inferior to antibiotic treatment for UTIs. Intravesical individuals with neurogenic lower urinary tract dysfunction (Groah
administration is a method of prophylaxis where a drug is administered et al., 2019). Currently, intravesical gentamicin irrigation is deemed
directly into a patient’s bladder using a catheter (GuhaSarkar and effective for individuals suffering with rUTIs when all other systemic
Banerjee, 2010). Additionally, bladder irrigation with the phage cocktail treatments are unsuccessful (Dray and Clemens, 2017). When adminis­
appeared superior in males undergoing transurethral resection of the tered in one or two doses, a L. rhamnosus GG intravesical probiotic was
prostate (TURP) (Leitner et al., 2021b). Leitner and colleagues (2021a) found to be safe and well tolerated in participants with cloudy or
have also hypothesized the potential use of phage treatment for in­ foul-smelling urine (Groah et al., 2019). It was hypothesized that a
dividuals with spinal cord injuries evidently suffering with UTIs and community shift in the urobiome could result from intravesical pro­
were to commence clinical trials in 2020. Therefore, whilst phage biotics, however the study is still in preliminary stages (Forster et al.,
research shows promising results when eradicating uropathogens, 2019). Despite the success of in vitro studies with oral/vaginal
further clinical studies are required to validate their effectiveness. L. rhamnosus, the strain has not yet shown significant reduction in UTI
occurrence, or UPEC reduction (Forster et al., 2019; Groah et al., 2019).
6.4. Probiotics Although this is the first human trial utilizing intravesical probiotics,
Lactobacillus casei demonstrated significant antimicrobial activity
6.4.1. Gram-positive against chronic UTIs in murine models (Asahara et al., 2001).
Probiotics are widely defined as “live microorganisms that, when
administered in adequate amounts, confer a health benefit on the host” 6.4.2. Gram-negative
(Guarner et al., 2012). Antimicrobial compounds, such as organic acids, E. coli Nissle was one of the first strains utilized as a probiotic, and
bacteriocins, biosurfactants, and hydrogen peroxide, enhance the has been commercialised as the main active component of the probiotic
antagonistic properties of probiotic bacteria aiding in the prevention of Mutaflor for the treatment of gastrointestinal disorders (Schultz, 2008;
various diseases/disorders (Darouiche and Hull, 2012). Lactic acid from Scaldaferri et al., 2016). E. coli Nissle 1917 has also shown promise as a
Lactobacillus SCS, in concentrations above 30 mM, exhibits potent prophylactic agent against urological infections (Storm et al., 2011).
antibacterial properties by eliminating UPEC (Cadieux et al., 2009). Genomic analysis between the avirulent E. coli strains Nissle 1917 and
However, Cadieux et al. (2009) indicated that Lactobacillus, which 83972, and a virulent uropathogen E. coli CFT073 revealed a close ge­
produce lactic acid, could also transform it into a carbon source bene­ netic relationship between the three strains, suggesting that the strains
ficial to UPEC, explaining the cause of infections when Lactobacillus likely originated from a common ancestor (Hancock et al., 2010b; Vej­
colonisation still persists. Hydrogen peroxide production can adversely borg et al., 2010). Increased genetic similarity between Nissle 1917 and
affect UPEC adhesion factors through the upregulation of stress proteins CFT073 was identified due to the presence of a pathogenic island
OmpA and OmpX, inducing membrane distress (Stancik et al., 2002; PAI-CFT073-serU, predominantly identified in strains causing pyelone­
Wang, 2002; Kim et al., 2010). Upregulation of such proteins could lead phritis (Vejborg et al., 2010). Additionally, expression of F1C fimbriae is
to the reduction of biofilm formation in enteric and genitourinary en­ associated with Nissle 1917, contributing to the strain’s adherence
vironments (Li et al., 2018). capability to different cells (Grozdanov et al., 2004; Kleta et al., 2014).
Typically, probiotics are consumed orally, however alternative de­ Expression of fitness factors contribute to E. coli Nissle’s 1917
livery routes are possible, including vaginally (Wolff et al., 2019). As antagonism, subsequently promoting the bacterium’s biofilm formation
latent UPEC reservoirs can colonise the genital tract (Brannon et al., (Hancock et al., 2010a; Scaldaferri et al., 2016). A ferric iron uptake
2020), vaginal probiotics could potentially decrease UPEC reservoirs system comprising of the siderophore yersiniabactin, has previously
and subsequently decrease rUTIs. Commensal lactobacilli regulate the been identified in the strain, contributing to biofilm formation in
vaginal microbiome, working to maintain homeostasis by reducing iron-limiting environments (Grozdanov et al., 2004). The fyuA gene,
pathogen abundance (Marrazzo, 2011). Lactobacillus crispatus is one of encoding the yersiniabactin receptor, is one of the most upregulated
the most effective strains in maintaining a healthy microbiome (Nardini genes in biofilm-producing strains, including those causing UTIs (Han­
et al., 2016). L. crispatus CTV-05 is the active probiotic strain in Lactin-V, cock et al., 2008). E. coli Nissle 1917 contains an array of additional iron
a live biotherapeutic product in clinical trials for treating bacterial uptake systems (enterobactin, aerobactin, ferric dicitrate transport sys­
vaginosis (Cohen et al., 2020). When assessing Lactin-V’s potential in tem, and the chu heme transport locus) facilitating survival through
treating uropathogens only 15% of the 50 treated individuals developed heme import (Garcia et al., 2011; Scaldaferri et al., 2016). Expression of
rUTIs, while the placebo cohort had a 27% rUTI incidence rate two known catechol microcins, microcin M and microcin H47,

9
C. Kenneally et al. Microbiological Research 259 (2022) 127010

represents yet another advantageous function of E. coli Nissle (Patzer to uropathogens is yet to be elucidated (Ali et al., 2009; Becknell et al.,
et al., 2003; Vassiliadis et al., 2010). Catechol microcins are post­ 2015). β-defensins are more widely expressed in humans, and both
translational modified peptides characterized by a catechol-siderophore human β-defensin-1 (HBD-1) and human β-defensin-2 (HBD-2) have
moiety (Patzer et al., 2003; Thomas et al., 2004). Under iron-limited been identified in the urinary tract (Becknell and Spencer, 2016).
conditions, such microcins utilize their catechol-siderophores contrib­ Limited research has been conducted on HBD-2’s function in the urinary
uting to their bacterial competitiveness by binding ferric iron (Thomas tract, although the AMP has displayed activity against Enterococcus
et al., 2004; Vassiliadis et al., 2010). This mechanism has aided in their faecalis (Kandaswamy et al., 2013). HBD-1 is predominantly expressed
antibacterial function against Enterobacteriaceae causing intestinal in­ by epithelial cells in the kidney, and its presence is increased three-fold
fections (Schultz, 2008; Baquero et al., 2019). Despite E. coli Nissle’s during acute pyelonephritis (Hiratsuka et al., 2000; Nielsen et al.,
probiotic properties, the strain has not yet displayed significant reduc­ 2014b). Deficiency of mouse BD-1 did not alter UPEC pathogenesis in
tion in UPEC, possibly due to antimicrobial defences in UPEC strains vivo¸ while mouse BD-3 and BD-4 displayed bactericidal activity against
(Pradhan and Weiss, 2020). However, E. coli Nissle 1917 has gained UPEC (Becknell et al., 2013). Interestingly, human β-defensins are
increased attention as a model organism for the development of novel expressed in different levels in the genital tract throughout the men­
therapeutics (Geldart et al., 2018; Praveschotinunt et al., 2019). strual cycle (King et al., 2003; Ali et al., 2009), which could indicate a
correlation to the possible interconnectivity of the urinary and vaginal
6.5. Antimicrobial peptides microbiomes. Oestrogen supplementation has also been shown to
correlate with an increase in AMPs, including β-defensins and LL-37, in
Antimicrobial peptides are small polypeptide antimicrobials the urinary tract (Lüthje and Brauner, 2016).
(<10 kDa), part of the innate immune system, and secreted by host AMPs derived from insects (Manniello et al., 2021) and plants
tissues (Suda et al., 2017). As part of the host defence system, AMPs are (Monteiro et al., 2019), and antifungal peptides (Raman et al., 2016)
produced by the urothelium of the bladder, ureter, kidneys, and the also represent potential treatments for human UTIs. Recently, cecropin
urethra in the urinary tract (Becknell et al., 2015). AMPs exert direct A (CecA) derived from the wax moth Galleria mellonella displayed ac­
antimicrobial activity, and can also function as immunomodulatory tivity as a novel therapeutic against biofilm-forming UPEC strains (Kalsy
compounds (O’Sullivan et al., 2020). Their net cationic charge, amphi­ et al., 2020). This is achieved through a multi-target mechanism, where
pathicity, and secondary structure contribute to their ability to bind to the outer membrane is permeabilised, resistance-nodulation-division
target cells and disrupt the lipid bilayers (Yeaman and Yount, 2003; (RND) efflux pumps are suppressed, and nucleic acid interactions are
Ebenhan et al., 2014; Becknell et al., 2015). Along with the bio­ inhibited, subsequently inhibiting biofilm formation (Kalsy et al., 2020).
therapeutic potential of AMPs, they can also be eminent biomarkers as A synergistic reaction between CecA and the antibiotic nalidixic acid
expression levels can often correlate with infection (Gupta et al., 2018). (NAL) was also observed, ultimately improving the entry of NAL into
Several known antimicrobial peptides detected in the urinary tract have cells, leading to a new approach to eradicate uropathogen biofilms
been thoroughly evaluated to determine their function in both mice and (Fenner, 2020).
humans, with cathelicidin, human β-defensins, and human α-defensin 5
(HD5) appearing to be the most prevalent (Hiratsuka et al., 2000; 6.6. Bacteriocins
Chromek et al., 2006; Zasloff, 2007; Spencer et al., 2014).
Human cathelicidin (LL-37) is an α-helical AMP, primarily expressed Bacteriocins, a subcategory of AMPs, are ribosomally synthesised
in circulating neutrophils, epithelial cells, and myeloid bone marrow peptides produced by bacteria to kill competing organisms, which are
cells (Agerberth et al., 2000). Although evidence surrounding LL-37’s usually closely related to the producer strain (Cotter et al., 2005). Bac­
protective role in the urinary tract is limited, some studies have reported teriocins produced by Gram-positive bacteria have been the focal point
promising results (Spencer et al., 2014; Babikir et al., 2018; Gupta et al., of bacteriocin research, with many extensive reviews on the topic
2018). For example, when challenged with UPEC invasion, increased (Mathur et al., 2017; Acedo et al., 2018; Soltani et al., 2020).
expression of mRNA in the CAMP LL-37 gene, and subsequent increased Gram-negative bacteria were not originally regarded as good candidates
LL-37 secretion, was detected within minutes (Chromek et al., 2006). for bacteriocin treatment, due to their outer lipopolysaccharide mem­
Deletion of the murine LL-37 homologue, CRAMP, resulted in higher brane and subsequent reduced antibiotic efficacy (Egan et al., 2017).
UPEC infection rates as bladder colonisation increased (Ali et al., 2009). However, the rise in MDR Gram-negative bacteria has led to a renewed
Additionally, LL-37-resistant E. coli are increasingly present in more interest in alternative therapeutics, including Gram-negative
invasive infections, such as pyelonephritis, and increased resistance is bacteriocins.
displayed in urine and faecal samples of patients suffering with rUTIs as Gram-negative bacteriocins can be classified into two main groups,
opposed to healthy individuals (Chromek et al., 2006; Nielsen et al., colicins and microcins (Engevik and Versalovic, 2017). Colicins are
2014b). LL-37 can suppress biofilm formation by inhibition of CsgA proteins with a high molecular weight (>20 kDa), while microcins are
polymerization, a subunit of curli fimbriae (Chromek, 2015). Presence lower molecular weight peptides of < 10 kDa (Rebuffat, 2011; Karpiński
of excess curli fimbriae gives rise to resistance in UPEC strains, inhib­ and Szkaradkiewicz, 2016). Colicins are more extensively characterised,
iting LL-37 from disrupting the bacterial membrane (Kai-Larsen et al., and are synthesised by > 50% of E. coli strains, encoded by small
2010; Luna-Pineda et al., 2019). multi-copy plasmids, or large low-copy plasmids (Braun et al., 1994;
Defensins are biologically active peptides classified into α- and Gillor et al., 2004). Three main mechanisms are utilised by colicins
β-defensins in humans, dependent on the sequence of cysteine residues when damaging target cells; membrane permeabilization through
and disulphide bridges (Becknell and Spencer, 2016). Significant in­ voltage-dependent channels, cellular nuclease degradation, and inhibi­
creases in the presence of the HD5 have been associated with patients tion of peptidoglycan synthesis (Cascales et al., 2007; de Zamaroczy and
suffering from pyelonephritis, and with patients undergoing urinary Chauleau, 2011). Historically, colicins have primarily been identified in
diversion surgery (Townes et al., 2011; Spencer et al., 2012). The AMP’s enteric strains (Garcia-Gutierrez et al., 2019). Studies regarding their
full potential is yet to be elucidated, however a recombinant HD5 has inhibitory potential against uropathogens are limited, but promising
been shown to exhibit bactericidal activity against a variety of uro­ (Budič et al., 2011; Roy and Riley, 2019).
pathogens (Wang et al., 2010). Tikhonov et al. (1997) also detected an Notably, Šmajs and co-authors (2010) reported a higher percentage
eight-fold increase in urinary α-defensin HNP-1 during chronic pyelo­ of colicin E1 plasmids (ColE1) in UPEC strains when compared to con­
nephritis, correlating with an increase in cytokine IL-8 and leukocyte trols, detected in 22% of strains versus 10%, respectively. Thus, ColE1
levels, suggesting neutrophil recruitment (Tikhonov et al., 1997). was suggested to be a potential virulence factor (Rijavec et al., 2007;
However, microbiological information regarding the HNPs relationship Šmajs et al., 2010). Conversely, certain colicins have displayed

10
C. Kenneally et al. Microbiological Research 259 (2022) 127010

inhibitory action, reducing biofilm formation on urinary catheters. contributed to the production of bacteriocin derivatives with improved
Coating of colicin E2-producing E. coli K-12 on urinary catheters functionality, solubility, stability, and activity (Kuipers et al., 1992; Rink
exhibited complete antibacterial activity against susceptible isolates, et al., 2007; Field et al., 2008; Cotter et al., 2013).
but was not effective against mutant E. coli displaying colicin resistance Typically, bacteriocins are more commonly employed against Gram-
(Trautner et al., 2005). Additionally, combining a lubricant with the positive bacteria, primarily due to the outer membrane of Gram-
novel colicin SR4, creating a prophylactic, achieved the same antimi­ negative bacteria decreasing their efficacy (Egan et al., 2017). Bioen­
crobial activity against uropathogens as gentamicin, whilst using 20–30 gineering of the N-terminal domain and the ‘hinge’ region in nisin
times less drug (Roy and Riley, 2019). Another mechanism of imple­ through both site-directed and site-saturation mutagenesis have been
menting bacteriocin activity against UPEC involves the introduction of particularly advantageous, expanding its antimicrobial spectrum against
recombinant bacteria. For example, genetically engineering the pro­ Gram-negative pathogens such as Shigella and Pseudomonas spp. (Yuan
biotic Lactobacillus brevis DT24 to express colicin E2 can increase the et al., 2004; Field et al., 2010; Healy et al., 2013). Additionally, bio­
strain’s antimicrobial capabilities against UPEC (Trivedi et al., 2014). engineered nisin variants S29A and S29G exhibited increased potency
Employing this form of biotherapy could inhibit uropathogens while against Gram-negative species, including E. coli (Field et al., 2012).
subsequently benefitting the microbiome, however it has not yet been Gram-negative bacteriocins also offer a degree of flexibility to en­
evaluated in vivo. gineer new derivatives (Behrens et al., 2017). Pheromonicin, for
Characteristically, colicins are expressed by more E. coli strains, example, is a fusion product formed by the combination of a
however microcin encoding determinants have been identified more channel-forming Colicin Ia, and its cognate immunity protein from
often in UTI causing strains (Šmajs et al., 2010). Microcins are heat E. coli, with a pheromone-encoding gene (agrD) from Staphylococcus
resistant antimicrobial peptides encoded by plasmids, or chromoso­ aureus (Ji et al., 1995; Mayville et al., 1999). Pheromonocin exhibits
mally, and display a broad spectrum of activity, mostly against Entero­ specific activity against S. aureus, and has shown increased performance
bacteriaceae species (Lagos, 2013). Microcins achieve antimicrobial compared to penicillin in mouse models (Gillor et al., 2004). Genetic
action through the inhibition of bacterial enzymes, membrane per­ fusion has also been exhibited in the construction of chimeras combining
meabilization, and utilisation of membrane components (Rodríguez and S-type pyocins with colicins, which exhibited antimicrobial activity
Laviña, 2003; Rebuffat, 2011). UTI-inducing UPEC are associated with against both E. coli and P. aeruginosa due to inhibition of lipopolysac­
an increase in chromosomally encoded microcin H47 and microcin M charide synthesis and DNA breakdown (Kageyama et al., 1996). More
(Šmajs et al., 2010; Massip et al., 2020). Interestingly, when both of recently, a similar strategy was adapted to enable the secretion of a
these microcins are expressed in E. coli Nissle 1917 they confer a sig­ chimeric bacteriocin from engineered E. coli. Replacing the N-terminal
nificant competitive advantage against UPEC as they induce a previ­ and central domains of colicin E3 with those of pyocin S3 produced the
ously mentioned iron uptake system (Patzer et al., 2003; Vassiliadis chimeric bacteriocin CoPy (Gupta et al., 2013). Subsequent tagging of
et al., 2010). Massip and co-authors (2020) also identified microcin H47 the flagellar secretion, FlgM, generated the antibacterial agent
and microcin M precursors more frequently in ABU strains than in UPEC. FlgM-CoPy, designed to kill a wild-type P. aeruginosa after detection of a
Whether or not such ABU strains displayed a competitive advantage quorum sensing molecule N-(3-oxododecanoyl) homoserine lactone
against UPEC strains was not established. At the time of writing, no (3OC12HSL) (Gupta et al., 2013). Incorporation of non-canonical amino
naturally produced microcins have displayed significant reduction in acids into microcin J25’s peptide structure resulted in several variants
UPEC isolates. However, antimicrobial activity against both exhibiting antimicrobial activity similar to, or higher than, wild-type
Gram-positive and Gram-negative organisms has been analysed using microcin (Piscotta et al., 2015). Furthermore, combining chitosan
the recombinant hybrid bacteriocin Ent35-MccV, combining enterocin nanoparticles with microcin J25 formed a stable nano-antimicrobial,
CRL35 and microcin V (Acuña et al., 2012). Production of the hybrid CN-MccJ25, exhibiting bactericidal activity against both
bacteriocin was achieved by the fusion of the structural portions of Gram-negative and Gram-positive strains (Yu et al., 2021). Further
enterocin CRL35 and microcin V, encoded by munA and cvaC respec­ implementation of bioengineering strategies on antimicrobial com­
tively, by asymmetric PCR (Acuña et al., 2012). Potent activity was pounds could eventually be the key to creating novel therapeutics,
exhibited against various strains of E. coli, including five UPEC strains reducing the reliance on antibiotics for UTIs.
(Acuña et al., 2012, 2015). UPEC230 was the only strain which dis­
played resistance against Ent35-MccV, likely due to the strain’s own 8. Future outlook
production of microcin V and immunity proteins. Bacteriocins are
notoriously unstable, however bioengineering is a common method of It is apparent that the relative abundance of bacterial communities
improving stability whilst simultaneously expanding their antimicrobial has a significant influence on urinary tract health, contributing to the
activity (Navarro et al., 2020; Soltani et al., 2020). Thus, implementing risk of diseases and infections (Burnett et al., 2021; Zandbergen et al.,
bioengineering techniques could potentially produce a therapeutic 2021). However, a significant knowledge gap still exists. For instance,
bacteriocin which could be administered intravesically on urinary although the synergistic activities between E. coli and E. faecalis have
catheters (Roy and Riley, 2019) or within engineered probiotics (Trivedi been previously described (Keogh et al., 2016), overall the mechanistic
et al., 2014), for the inhibition of uropathogens. and functional relationships between microbes in the urobiome remains
undefined. Integrating advanced ‘omics’ technologies such as metab­
7. Bioengineering olomics and metatranscriptomics could further our insight into meta­
bolic signalling activities within the urobiome, and its interactions with
Bacteriocins are typically gene-encoded and thus are amenable to other microbial communities (Franzosa et al., 2015). Gaining this in­
bioengineering to alter the peptides structure with the aim of creating a formation could evaluate how potential treatment options such as nat­
mutant peptide with enhanced function (Culligan and Sleator, 2017). ural UMT could be of benefit to the urobiome constituents or if
Modification of bacteriocins can be induced by mutating manipulation of urine needs to occur for this to be a viable future
bacteriocin-encoding genes or fusing genes together with subsequent therapy. Lactobacillus strains have received the most attention for
expression in an alternative host (Gillor et al., 2005; Cotter, 2012). reducing E. coli abundance. As similar benefits have been seen from ABU
Nisin, a model bacteriocin, is extensively exploited for engineering strains (Stork et al., 2018), it may be warranted to deepen our functional
purposes resulting from the substantial understanding of the lantibiotic knowledge of these strains, adding to the existing data surrounding
structure (Field et al., 2010). Bioengineering strategies including alter­ E. coli 83972. This could further our understanding of their metabolic
ations of single amino acid residues, amino acid arrangement, and interactions with UPEC and could determine their unexplored potential
site-saturation mutagenesis of individual hinge residues have against other uropathogens. Additionally, whether or not these strains

11
C. Kenneally et al. Microbiological Research 259 (2022) 127010

display similarity to E. coli Nissle 1917 as potential probiotic organisms Conflicts of interest
would be an avenue to investigate. If this is the case, ABU strains
eventually may be main active components in probiotics, possibly The authors declare that there are no conflicts of interest regarding
treating urological disorders. Thus, initiating large-scale screening the publication of this paper of which we are aware.
projects to identify more ABU strains, and characterising them at a
molecular and genetic level could be particularly valuable in designing References
novel therapies.
The efficacy of the biological therapeutics identified to date have not Abedon, S.T., Kuhl, S.J., Blasdel, B.G., Kutter, E.M., 2011. Phage treatment of human
infections. Bacteriophage 1 (2), 66–85 doi: 10.4161/bact.1.2.15845.
currently met the non-inferiority criteria when compared to antibiotics Abelson, B., Sun, D., Que, L., Nebel, R.A., Baker, D., Popiel, P., Amundsen, C.L., Chai, T.,
in clinical studies to treat UTIs. Nevertheless, there is a movement to Close, C., Disanto, M., Fraser, M.O., Kielb, S.J., Kuchel, G., Mueller, E.R., Palmer, M.
exploit such biological tools from inside the microbiome and improve H., Parker-Autry, C., Wolfe, A.J., Damaser, M.S., 2018. Sex differences in lower
urinary tract biology and physiology. Biol. Sex. Differ. 9 (1), 1–13. https://doi.org/
their antimicrobial efficacy, developing novel therapies. Lactobacillus 10.1186/s13293-018-0204-8.
abundance in the vaginal microbiome does increase post oral adminis­ Acedo, J.Z., Chiorean, S., Vederas, J.C., van Belkum, M.J., 2018. The expanding
tration of probiotics, yet a significant increase has not been detected in structural variety among bacteriocins from Gram-positive bacteria. FEMS Microbiol.
Rev. 42 (6), 805–828. https://doi.org/10.1093/femsre/fuy033.
the urobiome (Vagios et al., 2020). Direct instillation of Lactobacillus Acuña, L., Picariello, G., Sesma, F., Morero, R.D., Bellomio, A., 2012. A new hybrid
into the bladder of animal models have shown more positive results, bacteriocin, Ent35-MccV, displays antimicrobial activity against pathogenic Gram-
suggesting a more promising route of administration when treating UTIs positive and Gram-negative bacteria. FEBS Open Biol. 2, 12–19. https://doi.org/
10.1016/j.fob.2012.01.002.
(Asahara et al., 2001). However, a healthcare professional would be
Acuña, L., Corbalan, N.S., Fernandez-No, I.C., Morero, R.D., Barros-Velazquez, J.,
required for bladder installation, which could be both costly and labo­ Bellomio, A., 2015. Inhibitory effect of the hybrid bacteriocin ent35-mccv on the
rious if repeat visits are necessary. Data regarding the treatment of UTIs growth of escherichia coli and listeria monocytogenes in model and food systems.
using Lactobacillus-containing probiotics is inconsistent, as studies vary Food Bioprocess Technol. 8 (5), 1063–1075. https://doi.org/10.1007/s11947-015-
1469-0.
heavily in terms of model systems, delivery routes, and probiotic agents Agerberth, B., Charo, J., Werr, J., Olsson, B., Idali, F., Lindbom, L., Kiessling, R.,
(Vagios et al., 2020). In turn, their true clinical potential remains un­ Jornvall, H., Wigzell, H., Gudmundsson, G.H., 2000. The human antimicrobial and
determined. Additionally, while bacteriocins also exhibit promising chemotactic peptides LL-37 and α-defensins are expressed by specific lymphocyte
and monocyte populations. Blood 96 (9), 3086–3093. https://doi.org/10.1182/
potential, a significant gap remains in relation to studies focusing on blood.v96.9.3086.
bacteriocins from Gram-negative bacteria. Bioinformatic tools have also Ahmed, S.S., Shariq, A., Alsalloom, A.A., Babikir, I.H., Alhomoud, B.N., 2019.
increased our ability to detect these antimicrobials. Approximately 2000 Uropathogens and their antimicrobial resistance patterns: relationship with urinary
tract infections. Int. J. Health Sci. 13 (2), 48–55 (Available at). 〈http://www.ncbi.
bacteriocin coding genes had been identified in Gram-negative bacteria nlm.nih.gov/pubmed/30983946%0Ahttp://www.pubmedcentral.nih.gov/articleren
by 2017, including in opportunistic pathogens such as E. coli, K. pneu­ der.fcgi?artid=PMC6436442〉.
moniae and P. aeruginosa (Sharp et al., 2017). The transition from Ainsworth, C., 2017. Microbiome: a bag of surprises. Nature 551 (7679), S40–S41.
https://doi.org/10.1038/551S40a.
genome mining to in vitro and in vivo studies is also significantly un­ Aira, A., Rubio, E., Vergara Gómez, A., Fehér, C., Casals-Pascual, C., González, B.,
derrepresented with bacteriocins from Gram-negative origins (Behrens Morata, L., Rico, V., Soriano, A., 2021. rUTI resolution after FMT for clostridioides
et al., 2017). In their natural form the efficacy of biological therapies is difficile infection: a case report. Infect. Dis. Ther. 10 (2), 1065–1071. https://doi.
org/10.1007/s40121-020-00365-8.
often limited, relating to the low stability and solubility, and their strain
Ajayi, T., Radhakrishnan, R., 2016. Urinary tract infection in older adults in long-term
specificity (Riley et al., 2012; Meade et al., 2020). Nonetheless, their care facilities. Cmaj 188 (12), 899. https://doi.org/10.1503/cmaj.150708.
antimicrobial potential against uropathogens warrants further explora­ Ali, A.S.M., Townes, C.L., Hall, J., Pickard, R.S., 2009. Maintaining a sterile urinary tract:
tion, proving advantageous when targeting uropathogen growth and the role of antimicrobial peptides. J. Urol. 182 (1), 21–28. https://doi.org/10.1016/
j.juro.2009.02.124.
biofilm formation. Alperin, M., Burnett, L., Lukacz, E., Brubaker, L., 2019. The mysteries of menopause and
Over the last decade, bioengineering strategies have advanced the urogynecologic health: clinical and scientific gaps. Menopause 26 (1), 103–111.
exploitation of biological tools to enhance their antimicrobial function, https://doi.org/10.1097/GME.0000000000001209.
Ammitzbøll, N., Bau, B.P.J., Bundgaard-Nielsen, C., Villadsen, A.B., Jensen, A.M.,
stress tolerance, and solubility (Sheehan et al., 2007; Field et al., 2008; Leutscher, P.D.C., Glavind, K., Hagstrøm, S., Arenholt, L.T.S., Sørensen, S., 2021.
Hayes et al., 2019). Other microbiome’s have appeared to be rich Pre- and postmenopausal women have different core urinary microbiota. Sci. Rep. 11
sources of antimicrobial compounds (Mousa et al., 2017; O’Sullivan (1), 1–10. https://doi.org/10.1038/s41598-021-81790-8.
Andersson, P., Engberg, I., Lidin-Janson, G., Lincoln, K., Hull, R., Hull, S., Svanborg, C.,
et al., 2019; Lagenaur et al., 2021), suggesting that screening the uro­ 1991. Persistence of Escherichia coli bacteriuria is not determined by bacterial
biome might also unveil a potentially rich source of novel antimicro­ adherence. Infect. Immun. 59 (9), 2915–2921. https://doi.org/10.1128/
bials. Bacteriocins detected in other environments have displayed iai.59.9.2915-2921.1991.
Anger, J., Lee, U., Ackerman, A.L., Chughtai, B., Clemens, J.Q., Hickling, D., 2019.
enhanced activity when expressed in a recombinant engineered host American Urological Association ( AUA)/ Canadian Urological Association ( CUA)/
(O’Sullivan et al., 2021). Similar strategies could be applied to potential Society of Urodynamics, Female Pelvic Medicine & Urogenital Reconstruction (
bacteriocins in the urobiome in an attempt to enhance their activity. SUFU) recurrent uncomplicated urinary tract infections in women: AUA / CUA /
SUFU guideline R. Am. Urol. Assoc. April, 1–36.
Bioengineering Lactobacillus or ABU, for instance, as host expression
Antunes-Lopes, T., Vale, L., Coelho, A.M., Silva, C., Rieken, M., Geavlete, B., Rashid, T.,
systems for urobiome derived bacteriocins could provide many advan­ Rahnama’i, S.M., Cornu, J.N., Marcelissen, T., 2020. The role of urinary microbiota
tages such as enhanced antimicrobial activity, improved yield, and in lower urinary tract dysfunction: a systematic review. Eur. Urol. Focus 6 (2),
potentially making bacteriocins more respondent to purification. 361–369. https://doi.org/10.1016/j.euf.2018.09.011.
Aragón, I.M., Herrera-Imbroda, B., Queipo-Ortuño, M.I., Castillo, E., Del Moral, J.S.G.,
Therefore, a mechanistic understanding of microbe-microbe and Gómez-Millán, J., Yucel, G., Lara, M.F., 2018. The urinary tract microbiome in
microbe-host interactions within the urobiome is still required to gain a health and disease. Eur. Urol. Focus 4 (1), 128–138. https://doi.org/10.1016/j.
deeper insight into how such potential biological therapeutics truly euf.2016.11.001.
Armbruster, C.E., Forsyth-DeOrnellas, V., Johnson, A.O., Smith, S.N., Zhao, L., Wu, W.,
interact with the host’s immune system. Ultimately, this would provide Mobley, H.L.T., 2017. Genome-wide transposon mutagenesis of Proteus mirabilis:
a means of identifying potential biomarkers for non-antibiotic therapies essential genes, fitness factors for catheter-associated urinary tract infection, and the
and combined with bioengineering strategies, lead to the development impact of polymicrobial infection on fitness requirements. PLoS Pathog. 13 (6),
1–43. https://doi.org/10.1371/journal.ppat.1006434.
of more effective treatments, paving the way of a new era of diagnosis Asahara, T., Nomoto, K., Watanuki, M., Yokokura, T., 2001. Antimicrobial activity of
and therapy in urology. intraurethrally administered probiotic Lactobacillus casei in a murine model of
Escherichia coli urinary tract infection. Antimicrob. Agents Chemother. 45 (6),
1751–1760. https://doi.org/10.1128/AAC.45.6.1751-1760.2001.
Funding Azevedo, A.S., Almeida, C., Melo, L.F., Azevedo, N.F., 2017. Impact of polymicrobial
biofilms in catheter-associated urinary tract infections. Crit. Rev. Microbiol. 43 (4),
Ciara Kenneally is in receipt of a RISAM PhD Scholarship at Munster 423–439. https://doi.org/10.1080/1040841X.2016.1240656.
Technological University.

12
C. Kenneally et al. Microbiological Research 259 (2022) 127010

Babikir, I.H., Abugroun, E.A., Bilal, N.E., Alghasham, A.A., Abdalla, E.E., Adam, I., 2018. Cai, T., Mazzoli, S., Mondaini, N., Meacci, F., Nesi, G., D’Elia, C., Malossini, G., Boddi, V.,
The impact of cathelicidin, the human antimicrobial peptide LL-37 in urinary tract Bartoletti, R., 2012. The role of asymptomatic bacteriuria in young women with
infections. BMC Infect. Dis. 18 (1), 1–8. https://doi.org/10.1186/s12879-017-2901- recurrent urinary tract infections: To Treat or not to treat? Clin. Infect. Dis. 55 (6),
z. 771–777. https://doi.org/10.1093/cid/cis534.
Bagińska, N., Cieślik, M., Górski, A., Jończyk-matysiak, E., 2021. The role of antibiotic Cai, T., Nesi, G., Mazzoli, S., Meacci, F., Lanzafame, P., Caciagli, P., Mereu, L., Tateo, S.,
resistant a. Baumannii in the pathogenesis of urinary tract infection and the potential Malossini, G., Selli, C., Bartoletti, R., 2015. Asymptomatic bacteriuria treatment is
of its treatment with the use of bacteriophage therapy. Antibiotics 10 (3). https:// associated with a higher prevalence of antibiotic resistant strains in women with
doi.org/10.3390/antibiotics10030281. urinary tract infections. Clin. Infect. Dis. 61 (11), 1655–1661. https://doi.org/
Baquero, F., Lanza, V.F., Baquero, M.R., del Campo, R., Bravo-Vázquez, D.A., 2019. 10.1093/cid/civ696.
Microcins in enterobacteriaceae: peptide antimicrobials in the eco-active intestinal Cascales, E., Buchanan, S.K., Duché, D., Kleanthous, C., Lloubès, R., Postle, K., Riley, M.,
chemosphere. Front. Microbiol. 10 (2261), 1–25. https://doi.org/10.3389/ Slatin, S., Cavard, D., 2007. COlicin Biology. Microbiol. Mol. Biol. Rev. 71 (1),
fmicb.2019.02261. 158–229. https://doi.org/10.1128/mmbr.00036-06.
Beatson, S.A., Ben Zakour, N.L., Totsika, M., Forde, B.M., Watts, R.E., Mabbett, A.N., Cek, M., Tandoğdu, Z., Wagenlehner, F., Tenke, P., Naber, K., Bjerklund-Johansen, T.E.,
Szubert, J.M., Sarkar, S., Phan, M.D., Peters, K.M., Petty, N.K., Alikhan, N.F., 2014. Healthcare-associated urinary tract infections in hospitalized urological
Sullivan, M.J., Gawthorne, J.A., Stanton-Cook, M., Nhu, N.T.K., Chong, T.M., Yin, W. patients—a global perspective: results from the GPIU studies 2003–2010. World J.
F., Chan, K.G., Hancock, V., Ussery, D.W., Ulett, G.C., Schembri, M.A., 2015. Urol. 32 (6), 1587–1594 doi: 10.1007/s00345-013-1218-9.
Molecular analysis of asymptomatic bacteriuria Escherichia coli strain VR50 reveals Ceprnja, M., Oros, D., Melvan, E., Svetlicic, E., Skrlin, J., Barisic, K., Starcevic, L.,
adaptation to the urinary tract by gene acquisition. Infect. Immun. 83 (5), Zucko, J., Starcevic, A., 2021. Modeling of urinary microbiota associated with
1749–1764 doi: 10.1128/IAI.02810-14. cystitis. Front. Cell. Infect. Microbiol. 11 (643638), 1–10. https://doi.org/10.3389/
Becknell, B., Spencer, J.D., 2016. AntimicrobIal peptides: maintaining sterility of the fcimb.2021.643638.
urinary tract. Birkhauser Adv. Infect. Dis. 53–65 doi: 10.1007/978-3-319-24199-9_4. Cerqueira, L., Oliveira, J.A., Nicolau, A., Azevedo, N.F., Vieira, M.J., 2013. Biofilm
Becknell, B., Spencer, J.D., Carpenter, A.R., Chen, X., Singh, A., Ploeger, S., Kline, J., formation with mixed cultures of Pseudomonas aeruginosa/Escherichia coli on
Ellsworth, P., Li, B., Proksch, E., Schwaderer, A.L., Hains, D.S., Justice, S.S., silicone using artificial urine to mimic urinary catheters. Biofouling 29 (7), 829–840.
McHugh, K.M., 2013. Expression and antimicrobial function of beta-defensin 1 in the https://doi.org/10.1080/08927014.2013.807913.
lower urinary tract. PLoS One 8 (10), 1–10. https://doi.org/10.1371/journal. Ch’ng, J.H., Chong, K.K.L., Lam, L.N., Wong, J.J., Kline, K.A., 2019. Biofilm-associated
pone.0077714. infection by enterococci. Nat. Rev. Microbiol. 17 (2), 82–94. https://doi.org/
Becknell, B., Schwaderer, A., Hains, D.S., Spencer, J.D., 2015. Amplifying renal 10.1038/s41579-018-0107-z.
immunity: the role of antimicrobial peptides in pyelonephritis. Nat. Rev. Nephrol. 11 Chegini, Z., Khoshbayan, A., Vesal, S., Moradabadi, A., Hashemi, A., Shariati, A., 2021.
(11), 642–655. https://doi.org/10.1038/nrneph.2015.105. Bacteriophage therapy for inhibition of multi drug-resistant uropathogenic bacteria:
Beerepoot, M.A.J., ter Riet, G., Nys, S., van der Wal, W.M., de Borgie, C.A.J.M., de a narrative review. Ann. Clin. Microbiol. 20 (1), 1–13 doi: 10.1186/s12941-021-
Reijke, T.M., Prins, J.M., Koeijers, J., Verbon, A., Stobberingh, E., Geerlings, S.E., 00433-y.
2012. Lactobacilli vs antibiotics to prevent urinary tract infections. Arch. Intern. Chromek, M., 2015. The role of the antimicrobial peptide cathelicidin in renal diseases.
Med. 172 (9), 704. https://doi.org/10.1001/archinternmed.2012.777. Pediatr. Nephrol. 30 (8), 1225–1232. https://doi.org/10.1007/s00467-014-2895-3.
Behrens, H.M., Six, A., Walker, D., Kleanthous, C., 2017. The therapeutic potential of Chromek, M., Slamová, Z., Bergman, P., Kovács, L., Podracká, L., Ehrén, I., Hökfelt, T.,
bacteriocins as protein antibiotics. Emerg. Top. Life Sci. 1 (1), 65–74. https://doi. Gudmundsson, G.H., Gallo, R.L., Agerberth, B., Brauner, A., 2006. The antimicrobial
org/10.1042/ETLS20160016. peptide cathelicidin protects the urinary tract against invasive bacterial infection.
Biehl, L.M., Cruz Aguilar, R., Farowski, F., Hahn, W., Nowag, A., Wisplinghoff, H., Nat. Med. 12 (6), 636–641. https://doi.org/10.1038/nm1407.
Vehreschild, M.J.G.T., 2018. Fecal microbiota transplantation in a kidney transplant Claesson, M.J., Clooney, A.G., O’Toole, P.W., 2017. A clinician’s guide to microbiome
recipient with recurrent urinary tract infection. Infect. Springe Berl. Heidelb. 46 (6), analysis. Nat. Rev. Gastroenterol. Hepatol. 14 (10), 585–595. https://doi.org/
871–874. https://doi.org/10.1007/s15010-018-1190-9. 10.1038/nrgastro.2017.97.
Biggel, M., Xavier, B.B., Johnson, J.R., Nielsen, K.L., Frimodt-Møller, N., Clermont, O., Bonacorsi, S., Bingen, E., 2000. Rapid and simple determination of the
Matheeussen, V., Goossens, H., Moons, P., Van Puyvelde, S., 2020. Horizontally Escherichia coli phylogenetic group. Appl. Environ. Microbiol. 66 (10), 4555–4558.
acquired papGII-containing pathogenicity islands underlie the emergence of invasive https://doi.org/10.1128/AEM.66.10.4555-4558.2000.
uropathogenic Escherichia coli lineages. Nat. Commun. 11 (1) https://doi.org/ Cohen, C.R., Wierzbicki, M.R., French, A.L., Morris, S., Newmann, S., Reno, H., Green, L.,
10.1038/s41467-020-19714-9. Miller, S., Powell, J., Parks, T., Hemmerling, A., 2020. Randomized trial of Lactin-V
Boers, S.A., Jansen, R., Hays, J.P., 2019. Understanding and overcoming the pitfalls and to prevent recurrence of bacterial vaginosis. N. Engl. J. Med. 382 (20), 1906–1915
biases of next-generation sequencing (NGS) methods for use in the routine clinical doi: 10.1056/nejmoa1915254.
microbiological diagnostic laboratory. Eur. J. Clin. Microbiol. Infect. Dis. 38 (6), Cotter, P.D., 2012. Bioengineering. Bioengineered 3 (6), 313–319. https://doi.org/
1059–1070. https://doi.org/10.1007/s10096-019-03520-3. 10.4161/bioe.21601.
Bonkat, G., Bartoletti, R., Bruyère, F., Cai, T., Geerlings, S.E., Köves, B., Schubert, S., Cotter, P.D., Hill, C., Ross, R.P., 2005. Food mIcrobiology: Bacteriocins: Developing
Wagenlehner, F., Mezei, T., Pilatz, A., Pradere, B. and Veeratterapillay, R., 2020. Innate Immunity For food. Nat. Rev. Microbiol. 3 (10), 777–788. https://doi.org/
EAU 2020 Guidelines on Urological Infections, European Association of Urology. 10.1038/nrmicro1273.
Available at: 〈http://www.uroweb.org/gls/pdf/15_Urological_Infections.pdf〉. Cotter, P.D., Ross, R.P., Hill, C., 2013. Bacteriocins-a viable alternative to antibiotics?
Bottery, M.J., Pitchford, J.W., Friman, V.P., 2021. Ecology and evolution of Nat. Rev. Microbiol. 11 (2), 95–105. https://doi.org/10.1038/nrmicro2937.
antimicrobial resistance in bacterial communities. ISME J. 15 (4), 939–948. https:// Croxall, G., Weston, V., Joseph, S., Manning, G., Cheetham, P., McNally, A., 2011.
doi.org/10.1038/s41396-020-00832-7. Increased human pathogenic potential of Escherichia coli from polymicrobial
Brannon, J.R., Dunigan, T.L., Beebout, C.J., Ross, T., Wiebe, M.A., Reynolds, W.S., urinary tract infections in comparison to isolates from monomicrobial culture
Hadjifrangiskou, M., 2020. Invasion of vaginal epithelial cells by uropathogenic samples. J. Med. Microbiol. 60 (1), 102–109. https://doi.org/10.1099/
Escherichia coli. Nat. Commun. 11 (1), 1–11. https://doi.org/10.1038/s41467-020- jmm.0.020602-0.
16627-5. Culligan, E.P., Sleator, R.D., 2016. Advances in the microbiome: applications to
Braun, V., Pilsl, H., Grob, P., 1994. Colicins: structures, modes of action, transfer through clostridium difficile infection. J. Clin. Med. 5 (9) https://doi.org/10.3390/
membranes, and evolution. Arch. Microbiol. 161 (3), 199–206. https://doi.org/ jcm5090083.
10.1007/BF00248693. Culligan, E.P., Sleator, R.D., 2017. Antibiotics v2.0: computational and synthetic biology
Brubaker, L., Wolfe, A.J., 2015. The new world of the urinary microbiota in women. Am. approaches to combat antibiotic resistance. Future Microbiol 12 (4), 267–269.
J. Obstet. Gynecol. 213 (5), 644–649. https://doi.org/10.1016/j.ajog.2015.05.032. https://doi.org/10.2217/fmb-2017-0005.
Brubaker, L., Wolfe, A.J., 2017. The female urinary microbiota/microbiome: clinical and Culligan, E.P., Sleator, R.D., Marchesi, J.R., Hill, C., 2014. Metagenomics and novel gene
research implications. Rambam Maimonides Med. J. 8 (2), e0015 doi: 10.5041/ discoVery: promise And potential for novel therapeutics. Virulence 5 (3). https://
rmmj.10292. doi.org/10.4161/viru.27208.
Brubaker, L., Gourdine, J.-P.F., Siddiqui, N.Y., Holland, A., Halverson, T., Limeria, R., Cummings, L.A., Hoogestraat, D.R., Rassoulian-Barrett, S.L., Rosenthal, C.A.,
Pride, D., Ackerman, L., Forster, C.S., Jacobs, K.M., Thomas-White, K.J., Putonti, C., Salipante, S.J., Cookson, B.T., Hoffman, N.G., 2020. Comprehensive evaluation of
Dong, Q., Weinstein, M., Lukacz, E.S., Karstens, L., Wolfe, A.J., 2021. Forming complex polymicrobial specimens using next generation sequencing and standard
consensus to advance urobiome research. mSystems 6 (4), 1–10. https://doi.org/ microbiological culture. Sci. Rep. 10 (1), 1–12. https://doi.org/10.1038/s41598-
10.1128/msystems.01371-20. 020-62424-x.
Budič, M., Rijavec, M., Petkovšek, Ž., Žgur-Bertok, D., 2011. Escherichia coli Danis-Wlodarczyk, K., Vandenheuvel, D., Jang, H., Bin, Briers, Y., Olszak, T., Arabski, M.,
bacteriocins: antimicrobial efficacy and prevalence among isolates from patients Wasik, S., Drabik, M., Higgins, G., Tyrrell, J., Harvey, B.J., Noben, J.P., Lavigne, R.,
with bacteraemia. PLoS One 6 (12). https://doi.org/10.1371/journal.pone.0028769. Drulis-Kawa, Z., 2016. A proposed integrated approach for the preclinical evaluation
Bullman, S., Lucey, B., Sleator, R.D., 2012. Molecular diagnostics: the changing culture of of phage therapy in Pseudomonas infections. Sci. Rep. 6 (28115), 1–13. https://doi.
medical microbiology. Bioeng. Bugs 3 (1), 1–7. https://doi.org/10.4161/ org/10.1038/srep28115.
bbug.3.1.19011. Darouiche, R.O., Hull, R.A., 2012. Bacterial interference for prevention of urinary tract
Burnett, L.A., Hochstedler, B.R., Weldon, K., Wolfe, A.J., Brubaker, L., 2021. Recurrent infection. Clin. Infect. Dis. 55 (10), 1400–1407. https://doi.org/10.1093/cid/cis639.
urinary tract infection: association of clinical profiles with urobiome composition in Darouiche, R.O., Thornby, J.I., Stewart, C.C., Donovan, W.H., Hull, R.A., 2005. Bacterial
women. Neurourol. Urodyn. 40 (6), 1479–1489 doi: 10.1002/nau.24707. interference for prevention of urinary tract infection: a prospective, randomized,
Cadieux, P.A., Burton, J.P., Devillard, E., Reid, G., 2009. Lactobacillus by-products placebo-controlled, double-blind pilot trial. Clin. Infect. Dis. 41 (10), 1531–1534.
inhibit the growth and virulence of uropathogenic Escherichia coli. J. Physiol. https://doi.org/10.1086/497272.
Pharmacol. SUPPL.6 ((), 13–18. De Brucker, K., Tan, Y., Vints, K., De Cremer, K., Braem, A., Verstraeten, N., Michiels, J.,
Vleugels, J., Cammue, B.P.A., Thevissen, K., 2015. Fungal β-1,3-glucan increases

13
C. Kenneally et al. Microbiological Research 259 (2022) 127010

ofloxacin tolerance of escherichia coli in a polymicrobial E. coli/candida albicans Fouts, D.E., Pieper, R., Szpakowski, S., Pohl, H., Knoblach, S., Suh, M.J., Huang, S.T.,
biofilm. Antimicrob. Agents Chemother. 59 (6), 3052–3058 doi: 10.1128/ Ljungberg, I., Sprague, B.M., Lucas, S.K., Torralba, M., Nelson, K.E., Groah, S.L.,
AAC.04650-14. 2012. Integrated next-generation sequencing of 16S rDNA and metaproteomics
De Nisco, N.J., Neugent, M., Mull, J., Chen, L., Kuprasertkul, A., de Souza Santos, M., differentiate the healthy urine microbiome from asymptomatic bacteriuria in
Palmer, K.L., Zimmern, P., Orth, K., 2019. Direct detection of tissue-resident bacteria neuropathic bladder associated with spinal cord injury. J. Transl. Med. 10 (1), 1–17.
and chronic inflammation in the bladder wall of postmenopausal women with https://doi.org/10.1186/1479-5876-10-174.
recurrent urinary tract infection. J. Mol. Biol. 431 (21), 4368–4379, 10.1016/j. Foxman, B., 2014. Urinary tract infection syndromes. Occurrence, recurrence,
jmb.2019.04.008. bacteriology, risk factors, and disease burden. Infect. Dis. Clin. North Am. 28 (1),
De Vos, M.G.J., Zagorski, M., McNally, A., Bollenbach, T., 2017. Interaction networks, 1–13. https://doi.org/10.1016/j.idc.2013.09.003.
ecological stability, and collective antibiotic tolerance in polymicrobial infections. Foxman, B., Barlow, R., D’Arcy, H., Gillespie, B., Sobel, J.D., 2000. Urinary tract
PNAS USA 114 (40), 10666–10671 doi: 10.1073/pnas.1713372114. infection: Self-reported incidence and associated costs. Ann. Epidemiol. 10 (8),
Dekaboruah, E., Suryavanshi, M.V., Chettri, D., Verma, A.K., 2020. Human microbiome: 509–515. https://doi.org/10.1016/S1047-2797(00)00072-7.
an academic update on human body site specific surveillance and its possible role. Frank, D.N., Wilson St., S.S., Amand, A.L., Pace, N.R., 2009. Culture-independent
Arch. Microbiol. 202 (8), 2147–2167. https://doi.org/10.1007/s00203-020-01931- microbiological analysis of Foley urinary catheter biofilms. PLoS One 4 (11), 1–7.
x. https://doi.org/10.1371/journal.pone.0007811.
Dimitri-Pinheiro, S., Soares, R., Barata, P., 2020. The microbiome of the nose—friend or Franzosa, E.A., Hsu, T., Sirota-Madi, A., Shafquat, A., Abu-Ali, G., Morgan, X.C.,
foe? Allergy Rhinol. 11, 1–10. https://doi.org/10.1177/2152656720911605. Huttenhower, C., 2015. Sequencing and beyond: integrating molecular “omics” for
Dobrindt, U., Wullt, B., Svanborg, C., 2016. Asymtomatic bacteriuria as a model to study microbial community profiling. Nat. Rev. Microbiol. 13 (6), 360–372. https://doi.
the coevolution of hosts and Bacteria. Pathogens 5 (1), 1–8. https://doi.org/ org/10.1038/nrmicro3451.
10.3390/pathogens5010021. Gajer, P., Brotman, R.M., Bai, G., Sakamoto, J., Schütte, U.M.E., Zhong, X., Koenig, S.S.
Domann, E., Hong, G., Imirzalioglu, C., Turschner, S., Kühle, J., Watzel, C., Hain, T., K., Fu, L., Ma, Z., Zhou, X., Abdo, Z., Forney, L.J., Ravel, J., 2012. Temporal
Hossain, H., Chakraborty, T., 2003. Culture-Independent Identification of dynamics of the human vaginal microbiota. Sci. Transl. Med. 4 (132) https://doi.
Pathogenic Bacteria and Polymicrobial Infections in the Genitourinary Tract of Renal org/10.1126/scitranslmed.3003605.
Transplant Recipients. J. Clin. Microbiol. 41 (12), 5500–5510. https://doi.org/ Galtier, M., De Sordi, L., Maura, D., Arachchi, H., Volant, S., Dillies, M.A., Debarbieux, L.,
10.1128/JCM.41.12.5500-5510.2003. 2016. Bacteriophages to reduce gut carriage of antibiotic resistant uropathogens
Dray, E.V., Clemens, J.Q., 2017. ‘Recurrent urinary tract infections in patients with with low impact on microbiota composition. Environ. Microbiol. 18 (7), 2237–2245
incomplete bladder emptYing: Is There a role for intravesical therapy?’. Transl. doi: 10.1111/1462-2920.13284.
Androl. Urol. 6 (4), S163–S170. https://doi.org/10.21037/tau.2017.04.08. Galván, E.M., Mateyca, C., Ielpi, L., 2016. Role of interspecies interactions in dual-species
Ebenhan, T., Gheysens, O., Kruger, H.G., Zeevaart, J.R., Sathekge, M.M., 2014. biofilms developed in vitro by uropathogens isolated from polymicrobial urinary
Antimicrobial peptides: their role as infection-selective tracers for molecular catheter-associated bacteriuria. Biofouling 32 (9), 1067–1077. https://doi.org/
imaging. Biomed. Res. Int. 2014. https://doi.org/10.1155/2014/867381. 10.1080/08927014.2016.1231300.
Egan, K., Paul Ross, R., Hill, C., 2017. Bacteriocins: antibiotics in the age of the Garcia, E.C., Brumbaugh, A.R., Mobley, H.L.T., 2011. Redundancy and specificity of
microbiome. Emerg. Top. Life Sci. 1 (1), 55–63. https://doi.org/10.1042/ Escherichia coli iron acquisition systems during urinary tract infection. Infect.
ETLS20160015. Immun. 79 (3), 1225–1235. https://doi.org/10.1128/IAI.01222-10.
Elvers, K.T., Wilson, V.J., Hammond, A., Duncan, L., Huntley, A.L., Hay, A.D., van der Garcia-Gutierrez, E., Mayer, M.J., Cotter, P.D., Narbad, A., 2019. Gut microbiota as a
Werf, E.T., 2020. Antibiotic-induced changes in the human gut microbiota for the source of novel antimicrobials. Gut Microbes 10 (1), 1–21. https://doi.org/10.1080/
most commonly prescribed antibiotics in primary care in the UK: a systematic 19490976.2018.1455790.
review. BMJ Open 10 (9), e035677. https://doi.org/10.1136/bmjopen-2019- Garretto, A., Thomas-White, K., Wolfe, A.J., Putonti, C., 2018. Detecting viral genomes in
035677. the female urinary microbiome. J. Gen. Virol. 99 (8), 1141–1146. https://doi.org/
Engevik, M.A., Versalovic, J., 2017. Biochemical features of beneficial microbes: 10.1099/jgv.0.001097.
foundations for therapeutic microbiology. Microbiol. Spectr. 5 (5) https://doi.org/ Garretto, A., Miller-Ensminger, T., Wolfe, A.J., Putonti, C., 2019. Bacteriophages of the
10.1128/microbiolspec.bad-0012-2016. lower urinary tract. Nat. Rev. Urol. 16 (7), 422–432. https://doi.org/10.1038/
Fenner, A., 2020. Antimicrobial peptide derived from moths can eradicate UPEC biofilms s41585-019-0192-4.
and could offer a novel therapeutic option. Nat. Rev. Urol. 17 (4), 191. https://doi. Gaston, J.R., Andersen, M.J., Johnson, A.O., Bair, K.L., Sullivan, C.M., Guterman, L.B.,
org/10.1038/s41585-020-0309-9. White, A.N., Brauer, A.L., Learman, B.S., Flores-Mireles, A.L., Armbruster, C.E.,
Ferrières, L., Hancock, V., Klemm, P., 2007. Biofilm exclusion of uropathogenic bacteria 2020. Enterococcus faecalis polymicrobial interactions facilitate biofilm formation,
by selected asymptomatic bacteriuria Escherichia coli strains. Microbiology 153 (6), antibiotic recalcitrance, and persistent colonization of the catheterized urinary tract.
1711–1719. https://doi.org/10.1099/mic.0.2006/004721-0. Pathogens 9 (10), 1–20. https://doi.org/10.3390/pathogens9100835.
Ferris, M.J., Masztal, A., Aldridge, K.E., Fortenberry, D., Fidel, P.L., Martin, D.H., 2004. Geldart, K.G., Kommineni, S., Forbes, M., Hayward, M., Dunny, G.M., Salzman, N.H.,
Association of Atopobium vaginae, a recently described metronidazole resistant Kaznessis, Y.N., 2018. Engineered E. coli Nissle 1917 for the reduction of
anaerobe, with bacterial vaginosis. BMC Infect. Dis. 4, 1–8. https://doi.org/ vancomycin-resistant Enterococcus in the intestinal tract. Bioeng. Transl. Med. 3 (3),
10.1186/1471-2334-4-5. 197–208 doi: 10.1002/btm2.10107.
Ferry, S.A., Holm, S.E., Stenlund, H., Lundholm, R., Monsen, T.J., 2007. Clinical and Ghani, R., Mullish, B.H., McDonald, J.A.K., Ghazy, A., Williams, H.R.T., Brannigan, E.T.,
bacteriological outcome of different doses and duration of pivmecillinam compared Mookerjee, S., Satta, G., Gilchrist, M., Duncan, N., Corbett, R., Innes, A.J., Pavlů, J.,
with placebo therapy of uncomplicated lower urinary tract infection in women: The Thursz, M.R., Davies, F., Marchesi, J.R., 2021. Disease prevention not
LUTIW project. Scand. J. Prim. Health Care 25 (1), 49–57 doi: 10.1080/ decolonization: a model for fecal microbiota transplantation in patients colonized
02813430601183074. with multidrug-resistant organisms. Clin. Infect. Dis. 72 (8), 1444–1447. https://doi.
Field, D., Connor, P.M.O., Cotter, P.D., Hill, C., Ross, R.P., 2008. The generation of nisin org/10.1093/cid/ciaa948.
variants with enhanced activity against specific Gram-positive pathogens. Mol. Gilbert, N.M., O’Brien, V.P., Lewis, A.L., 2017. Transient microbiota exposures activate
Microbiol. 69 (1), 218–230 doi: 10.1111/j.1365-2958.2008.06279.x. dormant Escherichia coli infection in the bladder and drive severe outcomes of
Field, D., Hill, C., Cotter, P.D., Ross, R.P., 2010. The dawning of a “Golden era” in recurrent disease. PLoS Pathog. 13 (3), 1–19. https://doi.org/10.1371/journal.
lantibiotic bioengineering. Mol. Microbiol. 78 (5), 1077–1087. https://doi.org/ ppat.1006238.
10.1111/j.1365-2958.2010.07406.x. Gillor, O., Kirkup, B.C., Riley, M.A., 2004. Colicins and microcins: the next generation
Field, D., Begley, M., O’Connor, P.M., Daly, K.M., Hugenholtz, F., Cotter, P.D., Hill, C., antimicrobials. Adv. Appl. Microbiol. 54, 129–146. https://doi.org/10.1016/S0065-
Ross, R.P., 2012. Bioengineered nisin a derivatives with enhanced activity against 2164(04)54005-4.
both gram positive and gram negative pathogens. PLoS One 7 (10). https://doi.org/ Gillor, O., Nigro, L., Riley, M., 2005. Genetically engineered bacteriocins and their
10.1371/journal.pone.0046884. potential as the next generation of antimicrobials. Curr. Pharm. Des. 11 (8),
Finucane, T.E., 2017. “Urinary tract infection”—requiem for a heavyweight. J. Am. 1067–1075. https://doi.org/10.2174/1381612053381666.
Geriatr. Soc. 65 (8), 1650–1655 doi: 10.1111/jgs.14907. Gliniewicz, K., Schneider, G.M., Ridenhour, B.J., Williams, C.J., Song, Y., Farage, M.A.,
Flores-Mireles, A.L., Walker, J.N., Caparon, M., Hultgren, S.J., 2015. Urinary tract Miller, K., Forney, L.J., 2019. Comparison of the vaginal microbiomes of
infections: epidemiology, mechanisms of infection and treatment options. Nat. Rev. premenopausal and postmenopausal women. Front. Microbiol. 10 (193), 1–9.
Microbiol. 13 (5), 269–284. https://doi.org/10.1038/nrmicro3432. https://doi.org/10.3389/fmicb.2019.00193.
Forde, B.M., Roberts, L.W., Phan, M.D., Peters, K.M., Fleming, B.A., Russell, C.W., Goh, H.M.S., Yong, M.H.A., Chong, K.K.L., Kline, K.A., 2017. Model systems for the study
Lenherr, S.M., Myers, J.B., Barker, A.P., Fisher, M.A., Chong, T.M., Yin, W.F., of Enterococcal colonization and infection. Virulence 8 (8), 1525–1562. https://doi.
Chan, K.G., Schembri, M.A., Mulvey, M.A., Beatson, S.A., 2019. Population dynamics org/10.1080/21505594.2017.1279766.
of an Escherichia coli ST131 lineage during recurrent urinary tract infection. Nat. Gottschick, C., Deng, Z.L., Vital, M., Masur, C., Abels, C., Pieper, D.H., Wagner-Döbler, I.,
Commun. 10 (1), 1–10. https://doi.org/10.1038/s41467-019-11571-5. 2017. The urinary microbiota of men and women and its changes in women during
Forster, C.S., Hsieh, M.H., Pérez-Losada, M., Caldovic, L., Pohl, H., Ljungberg, I., bacterial vaginosis and antibiotic treatment. Microbiome 5 (1), 99. https://doi.org/
Sprague, B., Stroud, C., Groah, S., 2019. A single intravesical instillation of 10.1186/s40168-017-0305-3.
Lactobacillus rhamnosus GG is safe in children and adults with neuropathic bladder: Graspeuntner, S., Loeper, N., Künzel, S., Baines, J.F., Rupp, J., 2018. Selection of
a phase Ia clinical trial. J. Spinal Cord. Med 0 (0), 1–8. https://doi.org/10.1080/ validated hypervariable regions is crucial in 16S-based microbiota studies of the
10790268.2019.1616456. female genital tract. Sci. Rep. 8 (1), 4–10. https://doi.org/10.1038/s41598-018-
Fourcade, C., Canini, L., Lavigne, J.P., Sotto, A., 2015. A comparison of monomicrobial 27757-8.
versus polymicrobial Enterococcus faecalis bacteriuria in a French University Groah, S.L., Rounds, A.K., Ljungberg, I.H., Sprague, B.M., Frost, J.K., Tractenberg, R.E.,
Hospital. Eur. J. Clin. Microbiol. Infect. Dis. 34 (8), 1667–1673. https://doi.org/ 2019. Intravesical Lactobacillus rhamnosus GG is safe and well tolerated in adults
10.1007/s10096-015-2403-0.

14
C. Kenneally et al. Microbiological Research 259 (2022) 127010

and children with neurogenic lower urinary tract dysfunction: first-in-human trial. Høiby, N., Bjarnsholt, T., Givskov, M., Molin, S., Ciofu, O., 2010. Antibiotic resistance of
Ther. Adv. Urol. 11, 1–13. https://doi.org/10.1177/1756287219875594. bacterial biofilms. Int. J. Antimicrob. Agents 35 (4), 322–332, 10.1016/j.
Grozdanov, L., Raasch, C., Schulze, J., Sonnenborn, U., Gottschalk, G., Hacker, J., ijantimicag.2009.12.011.
Dobrindt, U., 2004. Analysis of the genome structure of the nonpathogenic probiotic Holá, V., Ruzicka, F., Horka, M., 2010. Microbial diversity in biofilm infections of the
Escherichia coli strain Nissle 1917. J. Bacteriol. 186 (16), 5432–5441. https://doi. urinary tract with the use of sonication techniques. FEMS Immunol. Med. Microbiol.
org/10.1128/JB.186.16.5432-5441.2004. 59 (3), 525–528. https://doi.org/10.1111/j.1574-695X.2010.00703.x.
Grygorcewicz, B., Wojciuk, B., Roszak, M., Łubowska, N., Blstrokaejczak, P., Jursa- Hrbacek, J., Cermak, P., Zachoval, R., 2020. Current antibiotic resistance trends of
Kulesza, J., Rakoczy, R., Masiuk, H., Dogowska, B., 2021. Environmental phage- uropathogens in central europe: Survey from a tertiary hospital urology department
based cocktail and antibiotic combination effects on acinetobacter baumannii 2011–2019. Antibiotics 9 (9), 1–11. https://doi.org/10.3390/antibiotics9090630.
biofilm in a human urine model. Microb. Drug Resist. 27 (1), 25–35. https://doi.org/ Hull, R.A., Donovan, W.H., Del Terzo, M., Stewart, C., Rogers, M., Darouiche, R.O., 2002.
10.1089/mdr.2020.0083. Role of type 1 fimbria- and P fimbria-specific adherence in colonization of the
Guarner, F., Khan, A.G., Garisch, J., Eliakim, R., Gangl, A., Thomson, A., Krabshuis, J., neurogenic human bladder by Escherichia coli. Infect. Immun. 70 (11), 6481–6484.
Lemair, T., Kaufmann, P., de Paula, J.A., Fedorak, R., Shanahan, F., Sanders, M.E., https://doi.org/10.1128/IAI.70.11.6481-6484.2002.
Szajewska, H., Ramakrishna, B.S., Karakan, T., Kim, N., 2012. World Human Microbiome Project Consortium, 2012. A framework for human microbiome
gastroenterology organisation global guidelines. J. Clin. Gastroenterol. 46 (6), research. Nature 486 (7402), 215–221. https://doi.org/10.1038/nature11209.
468–481. https://doi.org/10.1097/MCG.0b013e3182549092. Ji, G., Beavis, R.C., Novick, R.P., 1995. Cell density control of staphylococcal virulence
Guglietta, A., 2017. Recurrent urinary tract infections in women: Risk factors, etiology, mediated by an octapeptide pheromone. PNAS USA 92 (26), 12055–12059 doi:
pathogenesis and prophylaxis. Future Microbiol 12 (3), 239–246. https://doi.org/ 10.1073/pnas.92.26.12055.
10.2217/fmb-2016-0145. Johansen, T.E.B., Çek, M., Naber, K.G., Stratchounski, L., Svendsen, M.V., Tenke, P.,
GuhaSarkar, S., Banerjee, R., 2010. Intravesical drug delivery: Challenges, current status, 2006. Hospital acquired urinary tract infections in urology departments: pathogens,
opportunities and novel strategies. J. Control. Release 148 (2), 147–159. https://doi. susceptibility and use of antibiotics. Data from the PEP and PEAP-studies. Int. J.
org/10.1016/j.jconrel.2010.08.031. Antimicrob. Agents 28 (SUPPL. 1), 91–107 doi: 10.1016/j.ijantimicag.2006.05.005.
Gümüş, D., Kalaycı Yüksek, F., Sefer, Ö., Yörük, E., Uz, G., Anğ Küçüker, M., 2019. The Jones, J., Murphy, C.P., Sleator, R.D., Culligan, E.P., 2021. The urobiome, urinary tract
roles of hormones in the modulation of growth and virulence genes’ expressions in infections, and the need for alternative therapeutics. Microb. Pathog. 110, 105295
UPEC strains. Microb. Pathog. 132, 319–324. https://doi.org/10.1016/j. https://doi.org/10.1016/j.micpath.2021.105295.
micpath.2019.05.019. Jones-Freeman, B., Chonwerawong, M., Marcelino, V.R., Deshpande, A.V., Forster, S.C.,
Gupta, K., Hooton, T.M., Naber, K.G., Wullt, B., Colgan, R., Miller, L.G., Moran, G.J., Starkey, M.R., 2021. The microbiome and host mucosal interactions in urinary tract
Nicolle, L.E., Raz, R., Schaeffer, A.J., Soper, D.E., 2011. International clinical diseases. Mucosal Immunol. 14 (4), 1–14. https://doi.org/10.1038/s41385-020-
practice guidelines for the treatment of acute uncomplicated cystitis and 00372-5.
pyelonephritis in women: a 2010 update by the Infectious Diseases Society of Jovel, J., Patterson, J., Wang, W., Hotte, N., O’Keefe, S., Mitchel, T., Perry, T., Kao, D.,
America and the European Society for Microbiology and Infectious Diseases. Clin. Mason, A.L., Madsen, K.L., Wong, G.K.S., 2016. Characterization of the gut
Infect. Dis. 52 (5), 103–120. https://doi.org/10.1093/cid/ciq257. microbiome using 16S or shotgun metagenomics. Front. Microbiol. 7 (459), 1–17.
Gupta, S., Bram, E.E., Weiss, R., 2013. Genetically programmable pathogen sense and https://doi.org/10.3389/fmicb.2016.00459.
destroy. ACS Synth. Biol. 2 (12), 715–723. https://doi.org/10.1021/sb4000417. Jung, C., Brubaker, L., 2020. The etiology and management of recurrent urinary tract
Gupta, S., Jackson, A.R., DaJusta, D.G., McLeod, D.J., Alpert, S.A., Jayanthi, V.R., infections in postmenopausal women. Climacteric 22 (3), 242–249. https://doi.org/
McHugh, K., Schwaderer, A.R., Becknell, B., Ching, C.B., 2018. Urinary 10.1080/13697137.2018.1551871.
antimicrobial peptides: potential novel biomarkers of obstructive uropathy. Kageyama, M., Kobayashi, M., Sano, Y., Masaki, H., 1996. Construction and
J. Pediatr. Urol. 14 (3), 238.e1–238.e6. https://doi.org/10.1016/j. characterization of pyocin-colicin chimeric proteins. J. Bacteriol. 178 (1), 103–110.
jpurol.2018.03.006. https://doi.org/10.1128/jb.178.1.103-110.1996.
Gurney, J., Aldakak, L., Betts, A., Gougat-Barbera, C., Poisot, T., Kaltz, O., Hochberg, M. Kai-Larsen, Y., Lüthje, P., Chromek, M., Peters, V., Wang, X., Holm, Å., Kádas, L.,
E., 2017. Network structure and local adapta tion in co-evolving bacteria-phage Hedlund, K.O., Johansson, J., Chapman, M.R., Jacobson, S.H., Römling, U.,
interactions. Mol. Ecol. 26 (7), 1764–1777 doi: 10.1111/mec.14008. Agerberth, B., Brauner, A., 2010. Uropathogenic Escherichia coli modulates immune
Gutiérrez, D., Fernández, L., Rodríguez, A., García, P., 2018. ‘Are phage lytic proteins the responses and its curli fimbriae interact with the antimicrobial peptide LL-37. PLoS
secret weapon to kill Staphylococcus aureus?’. mBio 9 (1), 1–17. https://doi.org/ Pathog. 6 (7), 1–16. https://doi.org/10.1371/journal.ppat.1001010.
10.1128/mBio.01923-17. Kalsy, M., Tonk, M., Hardt, M., Dobrindt, U., Zdybicka-Barabas, A., Cytrynska, M.,
Hancock, V., Ferrières, L., Klemm, P., 2008. The ferric yersiniabactin uptake receptor Vilcinskas, A., Mukherjee, K., 2020. The insect antimicrobial peptide cecropin A
FyuA is required for efficient biofilm formation by urinary tract infectious disrupts uropathogenic Escherichia coli biofilms. npj Biofilms Micro 6 (1). https://
Escherichia coli in human urine. Microbiology 154 (1), 167–175. https://doi.org/ doi.org/10.1038/s41522-020-0116-3.
10.1099/mic.0.2007/011981-0. Kandaswamy, K., Liew, T.H., Wang, C.Y., Huston-Warren, E., Meyer-Hoffert, U.,
Hancock, V., Dahl, M., Klemm, P., 2010a. Probiotic Escherichia coli strain Nissle 1917 Hultenby, K., Schröder, J.M., Caparon, M.G., Normark, S., Henriques-Normark, B.,
outcompetes intestinal pathogens during biofilm formation. J. Med. Microbiol. 59 Hultgren, S.J., Kline, K.A., 2013. Focal targeting by human β-defensin 2 disrupts
(4), 392–399. https://doi.org/10.1099/jmm.0.008672-0. localized virulence factor assembly sites in Enterococcus faecalis. PNAS USA 110
Hancock, V., Vejborg, R.M., Klemm, P., 2010b. Functional genomics of probiotic (50), 20230–20235. https://doi.org/10.1073/pnas.1319066110.
Escherichia coli Nissle 1917 and 83972, and UPEC strain CFT073: comparison of Karlsson, M., Scherbak, N., Khalaf, H., Olsson, P.E., Jass, J., 2012. Substances released
transcriptomes, growth and biofilm formation. Mol. Genet. Genom. 284 (6), from probiotic Lactobacillus rhamnosus GR-1 potentiate NF-κB activity in
437–454 doi: 10.1007/s00438-010-0578-8. Escherichia coli-stimulated urinary bladder cells. FEMS Immunol. Med. Microbiol.
Hannan, T.J., Totsika, M., Mansfield, K.J., Moore, K.H., Schembri, M.A., Hultgren, S.J., 66 (2), 147–156. https://doi.org/10.1111/j.1574-695X.2012.00994.x.
2012. Host–pathogen checkpoints and population bottlenecks in persistent and Karpiński, T.M., Szkaradkiewicz, A.K., 2016. Bacteriocins. Encycl. Food Health 312–319.
intracellular uropathogenic Escherichia coli bladder infection. FEMS Microbiol. Rev. https://doi.org/10.1016/B978-0-12-384947-2.00053-2.
36 (3), 616–648. https://doi.org/10.1111/j.1574-6976.2012.00339.x. Karstens, L., Asquith, M., Davin, S., Stauffer, P., Fair, D., Gregory, W.T., Rosenbaum, J.T.,
Hayes, K., Field, D., Hill, C., O’Halloran, F., Cotter, L., 2019. A novel bioengineered Mcweeney, S.K., Nardos, R., 2016. Does the urinary microbiome play a role in
derivative of nisin displays enhanced antimicrobial activity against clinical urgency urinary incontinence and its severity ?’. Front. Cell. Infect. Microbiol. 6
Streptococcus agalactiae isolates. J. Glob. Antimicrob. Resist. 19, 14–21, 10.1016/j. (78), 1–13. https://doi.org/10.3389/fcimb.2016.00078.
jgar.2019.04.010. Karstens, L., Asquith, M., Caruso, V., Rosenbaum, J.T., Fair, D.A., Braun, J., Gregory, W.
Healy, B., Field, D., O’Connor, P.M., Hill, C., Cotter, P.D., Ross, R.P., 2013. Intensive T., Nardos, R., McWeeney, S.K., 2018. Community profiling of the urinary
mutagenesis of the nisin hinge leads to the rational design of enhanced derivatives. microbiota: considerations for low-biomass samples. Nat. Rev. Microbiol. 15 (12),
PLoS ONE 8 (11), 1–8. https://doi.org/10.1371/journal.pone.0079563. 735–749. https://doi.org/10.1038/s41585-018-0104-z.
Hilt, E.E., McKinley, K., Pearce, M.M., Rosenfeld, A.B., Zilliox, M.J., Mueller, E.R., Karstens, L., Siddiqui, N.Y., Zaza, T., Barstad, A., Amundsen, C.L., Sysoeva, T.A., 2021.
Brubaker, L., Gai, X., Wolfe, A.J., Schreckenberger, P.C., 2014. Urine is not sterile: Benchmarking DNA isolation kits used in analyses of the urinary microbiome. Sci.
Use of enhanced urine culture techniques to detect resident bacterial flora in the Rep. 11 (1), 1–9. https://doi.org/10.1038/s41598-021-85482-1.
adult female bladder. J. Clin. Microbiol. 52 (3), 871–876. https://doi.org/10.1128/ Keogh, D., Tay, W.H., Ho, Y.Y., Dale, J.L., Chen, S., Umashankar, S., Williams, R.B.H.,
JCM.02876-13. Chen, S.L., Dunny, G.M., Kline, K.A., 2016. Enterococcal metabolite cues facilitate
Hirakawa, H., Suzue, K., Kurabayashi, K., Tomita, H., 2019. The tol-pal system of interspecies niche modulation and polymicrobial infection. Cell Host Microbe 20 (4),
uropathogenic Escherichia coli Is Responsible for optimal internalization into and 493–503. https://doi.org/10.1016/j.chom.2016.09.004.
aggregation within bladder epithelial cells, colonization of the urinary tract of mice, Kim, K., Kim, K.P., Choi, J., Lim, J.A., Lee, J., Hwang, S., Ryu, S., 2010. Outer membrane
and bacterial motility. Front. Microbiol. 10 (1827), 1–14. https://doi.org/10.3389/ proteins A (OmpA) and X (OmpX) are essential for basolateral invasion of
fmicb.2019.01827. cronobacter sakazakii. Appl. Environ. Microbiol. 76 (15), 5188–5198. https://doi.
Hiratsuka, T., Nakazato, M., Ihi, T., Minematsu, T., Chino, N., Nakanishi, T., Shimizu, A., org/10.1128/AEM.02498-09.
Kangawa, K., Matsukura, S., 2000. Structural analysis of human β-defensin-1 and its King, A.E., Fleming, D.C., Critchley, H.O.D., Kelly, R.W., 2003. Differential expression of
significance in urinary tract infection. Nephron 85 (1), 34–40. https://doi.org/ the natural antimicrobials, beta-defensins 3 and 4, in human endometrium.
10.1159/000045627. J. Reprod. Immunol. 59 (1), 1–16. https://doi.org/10.1016/S0165-0378(02)00083-
Hochstedler, B.R., Burnett, L., Price, T.K., Jung, C., Wolfe, A.J., Brubaker, L., 2021. 9.
Urinary microbiota of women with recurrent urinary tract infection: collection and Klein, R.D., Hultgren, S.J., 2020. Urinary tract infections: microbial pathogenesis,
culture methods. Int. Urogynecol. J. https://doi.org/10.1007/s00192-021-04780-4. host–pathogen interactions and new treatment strategies. Nat. Rev. Microbiol. 18
(4), 211–226. https://doi.org/10.1038/s41579-020-0324-0.

15
C. Kenneally et al. Microbiological Research 259 (2022) 127010

Klemm, P., Roos, V., Ulett, G.C., Svanborg, C., Schembri, M.A., 2006. Molecular Lüthje, P., Brauner, A., 2016. Novel strategies in the prevention and treatment of urinary
characterization of the Escherichia coli asymptomatic bacteriuria strain 83972: The tract infections. Pathogens 5 (1). https://doi.org/10.3390/pathogens5010013.
taming of a pathogen. Infect. Immun. 74 (1), 781–785. https://doi.org/10.1128/ Magistro, G., Stief, C.G., 2019. The urinary tract microbiome: the answer to all our open
IAI.74.1.781-785.2006. questions? Eur. Urol. Focus 5 (1), 36–38. https://doi.org/10.1016/j.
Kleta, S., Nordhoff, M., Tedin, K., Wieler, L.H., Kolenda, R., Oswald, S., Oelschlaeger, T. euf.2018.06.011.
A., Bleiß, W., Schierack, P., 2014. Role of F1C Fimbriae, Flagella, and secreted Mak, T.N., Sfanos, K.S., Brüggemann, H., 2013. Draft genome sequences of two strains of
bacterial components in the inhibitory effect of probiotic Escherichia coli Nissle Propionibacterium acnes isolated from radical prostatectomy specimens. Genome
1917 on atypical enteropathogenic E. coli infection. Infect. Immun. 82 (5), Announc. 1 (6), 2013. https://doi.org/10.1128/genomeA.01071-13.
1801–1812. https://doi.org/10.1128/IAI.01431-13. Malik, S., Sidhu, P.K., Rana, J.S., Nehra, K., 2020. Managing urinary tract infections
Kline, K.A., Lewis, A.L., 2016. Gram-positive uropathogens, polymicrobial urinary tract through phage therapy: a novel approach. Folia Microbiol 65 (2), 217–231. https://
infection, and the emerging microbiota of the urinary tract. Microbiol. Spectr. 4 (2) doi.org/10.1007/s12223-019-00750-y.
https://doi.org/10.1128/microbiolspec.uti-0012-2012. Manniello, M.D., Moretta, A., Salvia, R., Scieuzo, C., Lucchetti, D., Vogel, H.,
Komesu, Y.M., Richter, H.E., Carper, B., Dinwiddie, D.L., Lukacz, E.S., Siddiqui, N.Y., Sgambato, A., Falabella, P., 2021. Insect antimicrobial peptides: potential weapons
Sung, V.W., Zyczynski, H.M., Ridgeway, B., Rogers, R.G., Arya, L.A., to counteract the antibiotic resistance. Cell. Mol. Life Sci. 78 (9), 4259–4282.
Mazloomdoost, D., Gantz, M.G., 2018. The urinary microbiome in women with https://doi.org/10.1007/s00018-021-03784-z.
mixed urinary incontinence compared to similarly aged controls. Int. Urogynecol. J. Manor, O., Dai, C.L., Kornilov, S.A., Smith, B., Price, N.D., Lovejoy, J.C., Gibbons, S.M.,
29 (12), 1785–1795. https://doi.org/10.1007/s00192-018-3683-6. Magis, A.T., 2020. Health and disease markers correlate with gut microbiome
Kostakioti, M., Hultgren, S.J., Hadjifrangiskou, M., 2012. Molecular blueprint of composition across thousands of people. Nat. Commun. 11 (1), 1–12. https://doi.
uropathogenic Escherichia coli virulence provides clues toward the development of org/10.1038/s41467-020-18871-1.
anti-virulence therapeutics. Virulence 3 (7), 592–594. https://doi.org/10.4161/ Mansour, B., Monyók, Á., Makra, N., Gajdács, M., Vadnay, I., Ligeti, B., Juhász, J.,
viru.22364. Szabó, D., Ostorházi, E., 2020. Bladder cancer-related microbiota: examining
Köves, B., Salvador, E., Grönberg-Hernández, J., Zdziarski, J., Wullt, B., Svanborg, C., differences in urine and tissue samples. Sci. Rep. Nat. Publ. Group UK 10 (1), 1–10.
Dobrindt, U., 2014. Rare emergence of symptoms during long-term asymptomatic https://doi.org/10.1038/s41598-020-67443-2.
Escherichia coli 83972 carriage without an altered virulence factor repertoire. Marrazzo, J.M., 2011. Interpreting the epidemiology and natural history of bacterial
J. Urol. 191 (2), 519–528. https://doi.org/10.1016/j.juro.2013.07.060. vaginosis: are we still confused? Anaerobe 17 (4), 186–190. https://doi.org/
Krishnan, K., Chen, T., Paster, B.J., 2017. A practical guide to the oral microbiome and its 10.1016/j.anaerobe.2011.03.016.
relation to health and disease. Oral. Dis. 23 (3), 276–286 doi: 10.1111/odi.12509. Massip, C., Chagneau, C.V., Boury, M., Oswald, E., 2020. The synergistic triad between
Kuipers, O.P., Rollema, H.S., Yap, W.M.G.J., Boot, H.J., Siezen, R.J., De Vos, W.M., 1992. microcin, colibactin, and salmochelin gene clusters in uropathogenic Escherichia
Engineering dehydrated amino acid residues in the antimicrobial peptide nisin. coli. Microb. Infect. 22 (3), 144–147. https://doi.org/10.1016/j.
J. Biol. Chem. 267 (34), 24340–24346. https://doi.org/10.1016/s0021-9258(18) micinf.2020.01.001.
35771-5. Mathur, H., Field, D., Rea, M.C., Cotter, P.D., Hill, C., Ross, R.P., 2017. Bacteriocin-
Lagenaur, L.A., Hemmerling, A., Chiu, C., Miller, S., Lee, P.P., Cohen, C.R., Parks, T.P., antimicrobial synergy: a medical and food perspective. Front. Microbiol. 8 (1205),
2021. Connecting the dots: Translating the vaginal microbiome into a drug. J. Infect. 1–18. https://doi.org/10.3389/fmicb.2017.01205.
Dis. 223 (Suppl 3), S296–S306. https://doi.org/10.1093/infdis/jiaa676. Mayville, P., Ji, G., Beavis, R., Yang, H., Goger, M., Novick, R.P., Muir, T.W., 1999.
Lagos, R., 2013. Bacteriocins. Brenners Encyclopedia of Genetics. Elsevier, pp. 277–279. Structure-activity analysis of synthetic autoinducing thiolactone peptides from
https://doi.org/10.1016/B978-0-12-374984-0.00291-6. Staphylococcus aureus responsible for virulence. PNAS USA 96 (4), 1218–1223.
Lebeaux, D., Ghigo, J.-M., Beloin, C., 2014. Biofilm-related infections: bridging the gap https://doi.org/10.1073/pnas.96.4.1218.
between clinical management and fundamental aspects of recalcitrance toward Mazzariol, A., Bazaj, A., Cornaglia, G., 2017. Multi-drug-resistant Gram-negative
antibiotics. Microbiol. Mol. Biol. Rev. 78 (3), 510–543. https://doi.org/10.1128/ bacteria causing urinary tract infections: a review. J. Chemother. 29, 2–9. https://
mmbr.00013-14. doi.org/10.1080/1120009X.2017.1380395.
Leihof, R.F., Nielsen, K.L., Frimodt-Møller, N., 2021. Asymptomatic Bacteriuria (ABU) in McLellan, L.K., Hunstad, D.A., 2016. Urinary tract infection: pathogenesis and outlook.
elderly: prevalence, virulence, phylogeny, antibiotic resistance and complement C3 Trends Mol. Med. 22 (11), 946–957. https://doi.org/10.1016/j.
in urine. Microorganisms 9 (2), 390. https://doi.org/10.3390/ molmed.2016.09.003.
microorganisms9020390. McMillan, A., Dell, M., Zellar, M.P., Cribby, S., Martz, S., Hong, E., Fu, J., Abbas, A.,
Leitner, L., Sybesma, W., Chanishvili, N., Goderdzishvili, M., Chkhotua, A., Dang, T., Miller, W., Reid, G., 2011. Disruption of urogenital biofilms by lactobacilli.
Ujmajuridze, A., Schneider, M.P., Sartori, A., Mehnert, U., Bachmann, L.M., Colloids Surf. B 86 (1), 58–64. https://doi.org/10.1016/j.colsurfb.2011.03.016.
Kessler, T.M., 2017. Bacteriophages for treating urinary tract infections in patients Meade, E., Slattery, M.A., Garvey, M., 2020. Bacteriocins, potent antimicrobial peptides
undergoing transurethral resection of the prostate: a randomized, placebo- and the fight against multi drug resistant species: resistance is futile? Antibiotics 9
controlled, double-blind clinical trial. BMC Urol. 17 (1), 1–6. https://doi.org/ (1). https://doi.org/10.3390/antibiotics9010032.
10.1186/s12894-017-0283-6. Miller-Ensminger, T., Garretto, A., Brenner, J., Thomas-White, K., Zambom, A., Wolfe, A.
Leitner, L., McCallin, S., Kessler, T.M., 2021a. Bacteriophages: what role may they play in J., Putonti, C., 2018. Bacteriophages of the urinary microbiome. J. Bacteriol. 200 (7)
life after spinal cord injury? Spinal Cord. 59 (9), 967–970. https://doi.org/10.1038/ https://doi.org/10.1128/JB.00738-17.
s41393-021-00636-2. Mizrahi-man, O., Davenport, E.R., Gilad, Y., 2013. Taxonomic classification of bacterial
Leitner, L., Ujmajuridze, A., Chanishvili, N., Goderdzishvili, M., Chkonia, I., Rigvava, S., 16S rRNA genes using short sequencing reads: evaluation of effective study designs.
Chkhotua, A., Changashvili, G., McCallin, S., Schneider, M.P., Liechti, M.D., PLoS One 8 (1), 18–23. https://doi.org/10.1371/journal.pone.0053608.
Mehnert, U., Bachmann, L.M., Sybesma, W., Kessler, T.M., 2021b. Intravesical Monteiro, C., Costa, F., Pirttilä, A.M., Tejesvi, M.V., Martins, M.C.L., 2019. Prevention of
bacteriophages for treating urinary tract infections in patients undergoing urinary catheter-associated infections by coating antimicrobial peptides from
transurethral resection of the prostate: a randomised, placebo-controlled, double- crowberry endophytes. Sci. Rep. 9 (1), 1–14. https://doi.org/10.1038/s41598-019-
blind clinical trial. Lancet Infect. Dis. 21 (3), 427–436. https://doi.org/10.1016/ 47108-5.
S1473-3099(20)30330-3. Morrill, S., Gilbert, N.M., Lewis, A.L., 2020. Gardnerella vaginalis as a cause of bacterial
Lev-Sagie, A., Goldman-Wohl, D., Cohen, Y., Dori-Bachash, M., Leshem, A., Mor, U., vaginosis: appraisal of the evidence from in vivo models. Front. Cell. Infect.
Strahilevitz, J., Moses, A.E., Shapiro, H., Yagel, S., Elinav, E., 2019. Vaginal Microbiol. 10 (168) https://doi.org/10.3389/fcimb.2020.00168.
microbiome transplantation in women with intractable bacterial vaginosis. Nat. Mousa, W.K., Athar, B., Merwin, N.J., Magarvey, N.A., 2017. Antibiotics and specialized
Med. 25 (10), 1500–1504. https://doi.org/10.1038/s41591-019-0600-6. metabolites from the human microbiota. Nat. Prod. Rep. R. Soc. Chem. 34 (11),
Lewis, D.A., Brown, R., Williams, J., White, P., Jacobson, S.K., Marchesi, J.R., Drake, M. 1302–1331. https://doi.org/10.1039/c7np00021a.
J., 2013. The human urinary microbiome; bacterial DNA in voided urine of Moustafa, A., Li, W., Singh, H., Moncera, K.J., Torralba, M.G., Yu, Y., Manuel, O.,
asymptomatic adults. Front. Cell. Infect. Microbiol. 3, 1–14. https://doi.org/ Biggs, W., Venter, J.C., Nelson, K.E., Pieper, R., Telenti, A., 2018. Microbial
10.3389/fcimb.2013.00041. metagenome of urinary tract infection. Sci. Rep. 8 (1), 1–12. https://doi.org/
Li, B., Huang, Q., Cui, A., Liu, X., Hou, B., Zhang, L., Liu, M., Meng, X., Li, S., 2018. 10.1038/s41598-018-22660-8.
Overexpression of outer membrane protein X (OmpX) compensates for the effect of Mueller, E.R., Wolfe, A.J., Brubaker, L., Opin, C., Author, U., 2017. The female urinary
TolC inactivation on biofilm formation and curli production in extraintestinal microbiota. Curr. Opin. Urol. 27 (3), 282–286. https://doi.org/10.1097/
pathogenic Escherichia coli (ExPEC). Front. Cell. Infect. Microbiol. 8 (208), 1–8. MOU.0000000000000396.The.
https://doi.org/10.3389/fcimb.2018.00208. Mulder, M., Radjabzadeh, D., Hassing, R.J., Heeringa, J., Uitterlinden, A.G., Kraaij, R.,
Liao, K.S., Lehman, S.M., Tweardy, D.J., Donlan, R.M., Trautner, B.W., 2012. Stricker, B.H., Verbon, A., 2019. The effect of antimicrobial drug use on the
Bacteriophages are synergistic with bacterial interference for the prevention of composition of the genitourinary microbiota in an elderly population. BMC
Pseudomonas aeruginosa biofilm formation on urinary catheters. J. Appl. Microbiol. Microbiol. https://doi.org/10.1186/s12866-018-1379-1.
113 (6), 1530–1539 doi: 10.1111/j.1365-2672.2012.05432.x. Nardini, P., Nãhui Palomino, R.A., Parolin, C., Laghi, L., Foschi, C., Cevenini, R.,
Lindberg, U., Hanson, L.A., Jodal, U., Lidin-Janson, G., Lincoln, K., Olling, S., 1975. Vitali, B., Marangoni, A., 2016. Lactobacillus crispatus inhibits the infectivity of
Asymptomatic bacteriuria in schoolgirls. Acta Paediatr. 64 (3), 432–436 doi: Chlamydia trachomatis elementary bodies, in vitro study. Sci. Rep. 6 (29024), 1–11.
10.1111/j.1651-2227.1975.tb03860.x. https://doi.org/10.1038/srep29024.
Luna-Pineda, V.M., Moreno-Fierros, L., Cázares-Domínguez, V., Ilhuicatzi-Alvarado, D., Navarro, S.A., Lanza, L., Acuña, L., Bellomio, A., Chalón, M.C., 2020. Features and
Ochoa, S.A., Cruz-Córdova, A., Valencia-Mayoral, P., Rodríguez-Leviz, A., applications of Ent35-MccV hybrid bacteriocin: current state and perspectives. Appl.
Xicohtencatl-Cortes, J., 2019. Curli of uropathogenic escherichia coli enhance Microbiol. Biotechnol. 104 (14), 6067–6077. https://doi.org/10.1007/s00253-020-
urinary tract colonization as a fitness factor. Front. Microbiol. 10 (September) 10650-8.
https://doi.org/10.3389/fmicb.2019.02063. Nelson, D.E., van der Pol, B., Dong, Q., Revanna, K.V., Fan, B., Easwaran, S.,
Sodergren, E., Weinstock, G.M., Diao, L., Fortenberry, D.J., 2010. Characteristic

16
C. Kenneally et al. Microbiological Research 259 (2022) 127010

male urine microbiomes associate with asymptomatic sexually transmitted infection. vaginal probiotic lactobacillus rhamnosus GR-1. Front. Microbiol. 9 (1278), 1–12.
PLoS One 5 (11), 1–7. https://doi.org/10.1371/journal.pone.0014116. https://doi.org/10.3389/fmicb.2018.01278.
Neugent, M.L., Hulyalkar, N.V., Nguyen, V.H., Zimmern, P.E., De Nisco, N.J., 2020. Pino, A., Bartolo, E., Caggia, C., Cianci, A., Randazzo, C.L., 2019. Detection of vaginal
Advances in understanding the human urinary microbiome and its potential role in lactobacilli as probiotic candidates. Sci. Rep. 9 (1), 1–10. https://doi.org/10.1038/
urinary tract infection. mBio 11 (2), 1–15. https://doi.org/10.1128/mBio.00218-20. s41598-019-40304-3.
Nicolle, L.E., 1999. Urinary infections in the elderly: symptomatic or asymptomatic? Int. Piscotta, F.J., Tharp, J.M., Liu, W.R., Link, A.J., 2015. Expanding the chemical diversity
J. Antimicrob. Agents 11 (3–4), 265–268 doi: 10.1016/S0924-8579(99)00028-X. of lasso peptide MccJ25 with genetically encoded noncanonical amino acids.
Nicolle, L.E., 2003. Asymptomatic bacteriuria: when to screen and when to treat. Infect. ChemComm 51 (2), 409–412 doi: 10.1039/c4cc07778d.
Dis. Clin. North Am. 17 (2), 367–394. https://doi.org/10.1016/S0891-5520(03) Pradhan, S., Weiss, A.A., 2020. Probiotic properties of Escherichia coli nissle in human
00008-4. intestinal organoids. mBio 11 (4), 1–16. https://doi.org/10.1128/mBio.01470-20.
Nicolle, L.E., 2008. The prevention of hospital-acquired urinary tract infection. Clin. Praveschotinunt, P., Duraj-Thatte, A.M., Gelfat, I., Bahl, F., Chou, D.B., Joshi, N.S., 2019.
Infect. Dis. 46 (2), 251–253. https://doi.org/10.1086/524663. Engineered E. coli Nissle 1917 for the delivery of matrix-tethered therapeutic
Nicolle, L.E., Bradley, S., Colgan, R., Rice, J.C., Schaeffer, A., Hooton, T.M., 2005. domains to the gut. Nat. Commun. 10 (1) https://doi.org/10.1038/s41467-019-
Infectious diseases society of America guidelines for the diagnosis and treatment of 13336-6.
asymptomatic bacteriuria in adults. Clin. Infect. Dis. 40 (10), 1556. https://doi.org/ Price, T.K., Dune, T., Hilt, E.E., Thomas-White, K.J., Kliethermes, S., Brincat, C.,
10.1086/430607. Brubaker, L., Wolfe, A.J., Mueller, E.R., Schreckenberger, P.C., 2016. The clinical
Nielsen, K.L., Dynesen, P., Larsen, P., Frimodt-Møller, N., 2014a. Faecal Escherichia coli urine culture: enhanced techniques improve detection of clinically relevant
from patients with E. coli urinary tract infection and healthy controls who have microorganisms. J. Clin. Microbiol. 54 (5), 1216–1222. https://doi.org/10.1128/
never had a urinary tract infection’. J. Med. Microbiol. 63, 582–589. https://doi. JCM.00044-16.
org/10.1099/jmm.0.068783-0. Price, T.K., Hilt, E.E., Dune, T.J., Mueller, E.R., Wolfe, A.J., Brubaker, L., 2018. Urine
Nielsen, K.L., Dynesen, P., Larsen, P., Jakobsen, L., Andersen, P.S., Frimodt-Møller, N., trouble: should we think differently about UTI? Int. Urogynecol. J. 29 (2), 205–210.
2014b. Role of urinary cathelicidin LL-37 and human β-defensin 1 in uncomplicated https://doi.org/10.1007/s00192-017-3528-8.
Escherichia coli urinary tract infections. Infect. Immun. 82 (4), 1572–1578. https:// Price, T.K., Hilt, E.E., Thomas-White, K., Mueller, E.R., Wolfe, A.J., Brubaker, L., 2020a.
doi.org/10.1128/IAI.01393-13. The urobiome of continent adult women: a cross-sectional study. BJOG 127 (2),
Nielsen, K.L., Stegger, M., Kiil, K., Godfrey, P.A., Feldgarden, M., Lilje, B., Andersen, P.S., 193–201 doi: 10.1111/1471-0528.15920.
Frimodt-Møller, N., 2017. Whole-genome comparison of urinary pathogenic Price, Travis K., Wolff, B., Halverson, T., Limeira, R., Brubaker, L., Dong, Q., Mueller, E.
Escherichia coli and faecal isolates of UTI patients and healthy controls. Int. J. Med. R., Wolfe, A.J., 2020b. Temporal dynamics of the adult female lower urinary tract
Microbiol. 307 (8), 497–507. https://doi.org/10.1016/j.ijmm.2017.09.007. microbiota. mBio 11 (2), 1–14 doi: 10.1128/mBio.00475-20.
Nielsen, K.L., Stegger, M., Kiil, K., Lilje, B., Ejrnæs, K., Leihof, R.F., Skjøt-Rasmussen, L., Qin, Z., Yang, L., Qu, D., Molin, S., Tolker-Nielsen, T., 2009. Pseudomonas aeruginosa
Godfrey, P., Monsen, T., Ferry, S., Hammerum, A.M., Frimodt-Møller, N., 2021. extracellular products inhibit staphylococcal growth, and disrut established biofilms
Escherichia coli causing recurrent urinary tract infections: Comparison to non- produced by Staphylococcus epidermidis. Microbiology 155 (7), 2148–2156.
recurrent isolates and genomic adaptation in recurrent infections. Microorganisms 9 https://doi.org/10.1099/mic.0.028001-0.
(7). https://doi.org/10.3390/microorganisms9071416. Raman, N., Lee, M.R., Rodríguez López, A., de, L., Palecek, S.P., Lynn, D.M., 2016.
O’Brien, V.P., Joens, M.S., Lewis, A.L., Gilbert, N.M., 2020. Recurrent Escherichia coli Antifungal activity of a β-peptide in synthetic urine media: Toward materials-based
urinary tract infection triggered by gardnerella vaginalis bladder exposure in mice. approaches to reducing catheter-associated urinary tract fungal infections. Acta
J. Vis. Exp. 166, 1–20. https://doi.org/10.3791/61967. Biomater. 43, 240–250. https://doi.org/10.1016/j.actbio.2016.07.016.
O’Grady, M.C., Barry, L., Corcoran, G.D., Hooton, C., Sleator, R.D., Lucey, B., 2019. Rani, A., Ranjan, R., McGee, H.S., Metwally, A., Hajjiri, Z., Brennan, D.C., Finn, P.W.,
Empirical treatment of urinary tract infections: How rational are our guidelines? Perkins, D.L., 2016. A diverse virome in kidney transplant patients contains multiple
J. Antimicrob. Chemother. 74 (1), 214–217. https://doi.org/10.1093/jac/dky405. viral subtypes with distinct polymorphisms. Sci. Rep. 6, 1–13. https://doi.org/
O’Sullivan, J.N., Rea, M.C., O’Connor, P.M., Hill, C., Ross, R.P., 2019. Human skin 10.1038/srep33327.
microbiota is a rich source of bacteriocin-producing staphylococci that kill human Rani, A., Ranjan, R., McGee, H.S., Andropolis, K.E., Panchal, D.V., Hajjiri, Z., Brennan, D.
pathogens. FEMS Microbiol. Ecol. 95 (2), 1–10. https://doi.org/10.1093/femsec/ C., Finn, P.W., Perkins, D.L., 2017. Urinary microbiome of kidney transplant patients
fiy241. reveals dysbiosis with potential for antibiotic resistance. Transl. Res. 181 (3), 59–70.
O’Sullivan, J.N., Rea, M.C., Hill, C., Ross, R.P., 2020. Protecting the outside: biological https://doi.org/10.1016/j.trsl.2016.08.008.
tools to manipulate the skin microbiota. FEMS Microbiol. Ecol. 96 (6), 1–14. https:// Ravel, J., Gajer, P., Abdo, Z., Schneider, G.M., Koenig, S.S.K., McCulle, S.L.,
doi.org/10.1093/femsec/fiaa085. Karlebach, S., Gorle, R., Russell, J., Tacket, C.O., Brotman, R.M., Davis, C.C., Ault, K.,
O’Sullivan, J.N., O’Connor, P.M., Rea, M.C., Field, D., Hill, C., Ross, R.P., 2021. Nisin Peralta, L., Forney, L.J., 2011. Vaginal microbiome of reproductive-age women.
variants from Streptococcus and Staphylococcus successfully express in NZ980’. PNAS USA 108 (SUPPL. 1), 4680–4687 doi: 10.1073/pnas.1002611107.
J. Appl. Microbiol. 131 (5), 2223–2234 doi: 10.1111/jam.15107. Raz, R., 2001. Postmenopausal women with recurrent UTI. Int. J. Antimicrob. Agents 17
Orazi, G., O’Toole, G.A., 2020. “It takes a village”: mechanisms underlying antimicrobial (4), 269–271 doi: 10.1016/S0924-8579(00)00355-1.
recalcitrance of polymicrobial biofilms. J. Bacteriol. 202 (1) https://doi.org/ Rebuffat, S., 2011. Bacteriocins from gram-negative bacteria: a classification?
10.1128/JB.00530-19. Prokaryotic Antimicrobial Peptides: From Genes to Applications. Springer, New
Pastar, I., Nusbaum, A.G., Gil, J., Patel, S.B., Chen, J., Valdes, J., Stojadinovic, O., York, pp. 55–72. https://doi.org/10.1007/978-1-4419-7692-5_4.
Plano, L.R., Tomic-Canic, M., Davis, S.C., 2013. Interactions of methicillin resistant Reid, G., Bruce, A.W., Taylor, M., 1992. Influence of three-day antimicrobial therapy and
staphylococcus aureus USA300 and pseudomonas aeruginosa in polymicrobial lactobacillus vaginal suppositories on recurrence of urinary tract infections. Clin.
wound infection. PLoS ONE 8 (2), 1–11. https://doi.org/10.1371/journal. Ther. 14 (1), 11–16.
pone.0056846. Rijavec, M., Budič, M., Mrak, P., Müller-Premru, M., Podlesek, Z., Žgur-Bertok, D., 2007.
Patzer, S.I., Baquero, M.R., Bravo, D., Moreno, F., Hantke, K., 2003. The colicin G, H and Prevalence of ColE1-like plasmids and colicin K production among uropathogenic
X determinants encode microcins M and H47, which might utilize the catecholate Escherichia coli strains and quantification of inhibitory activity of colicin K. Appl.
siderophore receptors FepA, Cir, Fiu and Ironn. Microbiology 149 (9), 2557–2570. Environ. Microbiol. 73 (3), 1029–1032. https://doi.org/10.1128/AEM.01780-06.
https://doi.org/10.1099/mic.0.26396-0. Riley, M.A., Robinson, S.M., Roy, C.M., Dennis, M., Liu, V., Dorit, R.L., 2012. Resistance
Pearce, M.M., Hilt, E.E., Rosenfeld, A.B., Zilliox, M.J., Thomas-White, K., Fok, C., is futile: The bacteriocin model for addressing the antibiotic resistance challenge.
Kliethermes, S., Schreckenberger, P.C., Brubaker, L., Gai, X., Wolfe, A.J., 2014. The Biochem. Soc. Trans. 40 (6), 1438–1442. https://doi.org/10.1042/BST20120179.
female urinary microbiome: a comparison of women with and without urgency Rink, R., Wierenga, J., Kuipers, A., Kluskens, L.D., Driessen, A.J.M., Kuipers, O.P.,
urinary incontinence. mBio 5 (4), 1–12. https://doi.org/10.1128/mBio.01283-14. Moll, G.N., 2007. Dissection and modulation of the four distinct activities of nisin by
Pearce, M.M., Zilliox, M.J., Rosenfeld, A.B., Thomas-White, K.J., Richter, H.E., Nager, C. mutagenesis of rings A and B and by C-terminal truncation. Appl. Environ.
W., Visco, A.G., Nygaard, I.E., Barber, M.D., Schaffer, J., Moalli, P., Sung, V.W., Microbiol. 73 (18), 5809–5816. https://doi.org/10.1128/AEM.01104-07.
Smith, A.L., Rogers, R., Nolen, T.L., Wallace, D., Meikle, S.F., Gai, X., Wolfe, A.J., Rizzetto, L., Fava, F., Tuohy, K.M., Selmi, C., 2018. Connecting the immune system,
Brubaker, L., 2015. The female urinary microbiome in urgency urinary incontinence. systemic chronic inflammation and the gut microbiome: the role of sex.
Am. J. Obstet. Gynecol. 213 (3), 347.e1–347.e11. https://doi.org/10.1016/j. J. Autoimmun. 92, 12–34. https://doi.org/10.1016/j.jaut.2018.05.008.
ajog.2015.07.009. Robino, L., Scavone, P., Araujo, L., Algorta, G., Zunino, P., Vignoli, R., 2013. Detection of
Pelling, H., Nzakizwanayo, J., Milo, S., Denham, E.L., MacFarlane, W.M., Bock, L.J., intracellular bacterial communities in a child with Escherichia coli recurrent urinary
Sutton, J.M., Jones, B.V., 2019. Bacterial biofilm formation on indwelling urethral tract infections. Pathog. Dis. 68 (3), 78–81. https://doi.org/10.1111/2049-
catheters. Lett. Appl. Microbiol. 68 (4), 277–293 doi: 10.1111/lam.13144. 632X.12047.
Penesyan, A., Paulsen, I.T., Gillings, M.R., Kjelleberg, S., Manefield, M.J., 2020. Rodríguez, E., Laviña, M., 2003. The proton channel is the minimal structure of ATP
Secondary effects of antibiotics on microbial biofilms. Front. Microbiol. 11 (2109), synthase necessary and sufficient for microcin H47 antibiotic action. Antimicrob.
1–8. https://doi.org/10.3389/fmicb.2020.02109. Agents Chemother. 47 (1), 181–187. https://doi.org/10.1128/AAC.47.1.181-
Petrova, M.I., Imholz, N.C.E., Verhoeven, T.L.A., Balzarini, J., Van Damme, E.J.M., 187.2003.
Schols, D., Vanderleyden, J., Lebeer, S., 2016. Lectin-like molecules of lactobacillus Roos, V., Nielsen, E.M., Klemm, P., 2006a. Asymptomatic bacteriuria Escherichia coli
rhamnosus GG inhibit pathogenic escherichia coli and salmonella biofilm formation. strains: adhesins, growth and competition. FEMS Microbiol. Lett. 262 (1), 22–30 doi:
PLoS One 11 (8). https://doi.org/10.1371/journal.pone.0161337. 10.1111/j.1574-6968.2006.00355.x.
Petrova, M.I., Reid, G., Vaneechoutte, M., Lebeer, S., 2017. Lactobacillus iners: Friend or Roos, V., Schembri, M.A., Ulett, G.C., Klemm, P., 2006b. Asymptomatic bacteriuria
Foe? Trends Microbiol 25 (3), 182–191. https://doi.org/10.1016/j.tim.2016.11.007. Escherichia coli strain 83972 carries mutations in the foc locus and is unable to
Petrova, M.I., Macklaim, J.M., Wuyts, S., Verhoeven, T., Vanderleyden, J., Gloor, G.B., express F1C fimbriae. Microbiology 152 (6), 1799–1806. https://doi.org/10.1099/
Lebeer, S., Reid, G., 2018. Comparative genomic and phenotypic analysis of the mic.0.28711-0.

17
C. Kenneally et al. Microbiological Research 259 (2022) 127010

Roos, V., Ulett, G.C., Schembri, M.A., Klemm, P., 2006c. The asymptomatic bacteriuria and urinary tract. PLoS One 7 (2), 1–8. https://doi.org/10.1371/journal.
Escherichia coli strain 83972 outcompetes uropathogenic E. coli strains in human pone.0031712.
urine. Infect. Immun. 74 (1), 615–624. https://doi.org/10.1128/IAI.74.1.615- Spencer, J.D., Schwaderer, A.L., Becknell, B., Watson, J., Hains, D.S., 2014. The innate
624.2006. immune response during urinary tract infection and pyelonephritis. Pediatr.
Rosen, D.A., Hooton, T.M., Stamm, W.E., Humphrey, P.A., Hultgren, S.J., 2007. Nephrol. 29 (7), 1139–1149. https://doi.org/10.1007/s00467-013-2513-9.
Detection of intracellular bacterial communities in human urinary tract infection. Stancik, L.M., Stancik, D.M., Schmidt, B., Barnhart, D.M., Yoncheva, Y.N.,
PLoS Med 4 (12), 1949–1958. https://doi.org/10.1371/journal.pmed.0040329. Slonczewski, J.L., 2002. pH-dependent expression of periplasmic proteins and amino
Rowe, T.A., Juthani-Mehta, M., 2014. Diagnosis and management of urinary tract acid catabolism in Escherichia coli. J. Bacteriol. 184 (15), 4246–4258. https://doi.
infection in older adults. Infect. Dis. Clin. North Am. 28 (1), 75–89. https://doi.org/ org/10.1128/JB.184.15.4246-4258.2002.
10.1016/j.idc.2013.10.004. Stapleton, A.E., 2016. The vaginal microbiota and urinary tract infection. Microbiol.
Roy, S.M., Riley, M.A., 2019. Evaluation of the potential of colicins to prevent Spectr. 4 (6), 139–148. https://doi.org/10.1128/microbiolspec.UTI-0025-2016.
extraluminal contamination of urinary catheters by Escherichia coli. Int. J. Stapleton, A.E., Au-yeung, M., Hooton, T.M., Fredricks, D.N., Roberts, P.L., Czaja, C.A.,
Antimicrob. Agents 54 (5), 619–625 doi: 10.1016/j.ijantimicag.2019.07.004. Yarova-yarovaya, Y., Fiedler, T., Cox, M., Stamm, W.E., 2011. Randomized, placebo-
Russo, T.A., Stapleton, A., Wenderoth, S., Hooton, T.M., Stamm, W.E., 1995. controlled phase 2 trial of a lactobacillus crispatus probiotic given intravaginally for
Chromosomal restriction fragment length polymorphism analysis of Escherichia coli prevention of recurrent urinary tract infection. Clin. Infect. Dis. 52 (10), 1212–1217.
strains causing recurrent urinary tract infections in young women. J. Infect. Dis. 172 https://doi.org/10.1093/cid/cir183.
(2), 440–445. https://doi.org/10.1093/infdis/172.2.440. Stork, C., Kovács, B., Rózsai, B., Putze, J., Kiel, M., Dorn, Á., Kovács, J., Melegh, S.,
Santiago-Rodriguez, T.M., 2018. Identification and quantification of DNA viral Leimbach, A., Kovács, T., Schneider, G., Kerényi, M., Emödy, L., Dobrindt, U., 2018.
populations in human urine using next-generation sequencing approaches. Methods Characterization of asymptomatic bacteriuria Escherichia coli isolates in search of
Mol. Bio 1838, 191–200. https://doi.org/10.1007/978-1-4939-8682-8_14. alternative strains for efficient bacterial interference against uropathogens. Front.
Santiago-Rodriguez, T.M., Ly, M., Bonilla, N., Pride, D.T., 2015. The human urine virome Microbiol. 9 (214), 1–18. https://doi.org/10.3389/fmicb.2018.00214.
in association with urinary tract infections. Front. Microbiol. 6 (14), 1–12. https:// Storm, D.W., Patel, A.S., Koff, S.A., Justice, S.S., 2011. Novel management of urinary
doi.org/10.3389/fmicb.2015.00014. tract infections. Curr. Opin. Urol. 21 (4), 328–333. https://doi.org/10.1097/
Sberro, H., Fremin, B.J., Zlitni, S., Edfors, F., Greenfield, N., Snyder, M.P., MOU.0b013e328346d4ee.
Pavlopoulos, G.A., Kyrpides, N.C., Bhatt, A.S., 2019. Large-scale analyses of human Suda, S., Field, D., Barron, N., 2017. Antimicrobial peptide production and purification.
microbiomes reveal thousands of small, novel genes. Cell 178 (5), 1245–1259. Protein Chromatography. Humana Press, New York, pp. 401–410. https://doi.org/
https://doi.org/10.1016/j.cell.2019.07.016. 10.1007/978-1-4939-6412-3_22.
Scaldaferri, F., Gerardi, V., Mangiola, F., Lopetuso, L.R., Pizzoferrato, M., Petito, V., Sundén, F., Håkansson, L., Ljunggren, E., Wullt, B., 2010. Escherichia coli 83972
Papa, A., Stojanovic, J., Poscia, A., Cammarota, G., Gasbarrini, A., 2016. Role and bacteriuria protects against recurrent lower urinary tract infections in patients with
mechanisms of action of Escherichia coli nissle 1917 in the maintenance of remission incomplete bladder emptying. J. Urol. 184 (1), 179–185. https://doi.org/10.1016/j.
in ulcerative colitis patients: an update. World J. Gastroenterol. 22 (24), 5505–5511. juro.2010.03.024.
https://doi.org/10.3748/wjg.v22.i24.5505. Sybesma, W., Zbinden, R., Chanishvili, N., Kutateladze, M., Chkhotua, A.,
Schneeweiss, J., Koch, M., Umek, W., 2016. The human urinary microbiome and how it Ujmajuridze, A., Mehnert, U., Kessler, T.M., 2016. Bacteriophages as potential
relates to urogynecology. Int. Urogynecol. J. 27 (9), 1307–1312. https://doi.org/ treatment for urinary tract infections. Front. Microbiol. 7 (465), 1–9. https://doi.
10.1007/s00192-016-2944-5. org/10.3389/fmicb.2016.00465.
Schreiber, H.L., Conover, M.S., Chou, W.C., Hibbing, M.E., Manson, A.L., Dodson, K.W., Tapiainen, T. , 2020. Preventing Urinary Tract Infections With E. Coli Nissle: (FinNissle),
Hannan, T.J., Roberts, P.L., Stapleton, A.E., Hooton, T.M., Livny, J., Earl, A.M., [online] Clinicaltrials.gov. Available at: Identifier: NCT04608851 (Accessed 25
Hultgren, S.J., 2017. Bacterial virulence phenotypes of Escherichia coli and host February 2022).
susceptibility determine risk for urinary tract infections. Sci. Transl. Med. 9 (382), Tariq, R., Pardi, D.S., Tosh, P.K., Walker, R.C., Razonable, R.R., Khanna, S., 2017. Fecal
1–14. https://doi.org/10.1126/scitranslmed.aaf1283. microbiota transplantation for recurrent clostridium difficile infection reduces
Schultz, M., 2008. Clinical use of E. coli Nissle 1917 in inflammatory bowel disease. recurrent urinary tract infection frequency. Clin. Infect. Dis. 65 (10), 1745–1747.
Inflamm. Bowel Dis. 14 (7), 1012–1018. https://doi.org/10.1002/ibd.20377. https://doi.org/10.1093/cid/cix618.
Sharp, C., Bray, J., Housden, N.G., Maiden, M.C. J. and Kleanthous, C. (2017) ‘Diversity Terlizzi, M.E., Gribaudo, G., Maffei, M.E., 2017. UroPathogenic Escherichia coli (UPEC)
and distribution of nuclease bacteriocins in bacterial genomes revealed using Hidden infections: virulence factors, bladder responses, antibiotic, and non-antibiotic
Markov Models’, PLOS Comput. Bio. Edited by C. A. Orengo, 13(7), p. e1005652. antimicrobial strategies. Front. Microbiol. 8 (1566) https://doi.org/10.3389/
doi: 10.1371/journal.pcbi.1005652. fmicb.2017.01566.
Sheehan, V.M., Sleator, R.D., Hill, C., Fitzgerald, G.F., 2007. Improving gastric transit, Thänert, R., Reske, K.A., Hink, T., Wallace, M.A., Wang, B., Schwartz, D.J., Seiler, S.,
gastrointestinal persistence and therapeutic efficacy of the probiotic strain Cass, C., Burnham, C.A.D., Dubberke, E.R., Kwon, J.H., Dantas, G., 2019.
Bifidobacterium breve UCC2003. Microbiology 153 (10), 3563–3571. https://doi. Comparative genomics of antibiotic-resistant uropathogens implicates three routes
org/10.1099/mic.0.2007/006510-0. for recurrence of urinary tract infections. mBio 10 (4), 1–16. https://doi.org/
Shigehara, K., Sasagawa, T., Kawaguchi, S., Kobori, Y., Nakashima, T., Shimamura, M., 10.1128/mBio.01977-19.
Taya, T., Furubayashi, K., Namiki, M., 2010. Prevalence of human papillomavirus Thomas, X., Destoumieux-Garzón, D., Peduzzi, J., Afonso, C., Blond, A., Birlirakis, N.,
infection in the urinary tract of men with urethritis. Int. J. Urol. 17 (6), 563–568. Goulard, C., Dubost, L., Thai, R., Tabet, J.C., Rebuffat, S., 2004. Siderophore peptide,
https://doi.org/10.1111/j.1442-2042.2010.02521.x. a new type of post-translationally modified antibacterial peptide with potent
Short, F.L., Murdoch, S.L., Ryan, R.P., 2014. Polybacterial human disease: the ills of activity. J. Biol. Chem. 279 (27), 28233–28242. https://doi.org/10.1074/jbc.
social networking. Trends Microbiol 22 (9), 508–516. https://doi.org/10.1016/j. M400228200.
tim.2014.05.007. Thomas-White, K., Brady, M., Wolfe, A.J., Mueller, E.R., 2016. The bladder is not sterile:
Shrestha, E., White, J.R., Yu, S., Kulac, I., Ertunc, O., De Marzo, A.M., history and current discoveries on the urinary microbiome. Curr. Bladder Dysfunct.
Yegnasubramanian, S., Mangold, L.A., Partin, A.W., Sfanos, K.S., 2018. Profiling the Rep. 11 (1), 18–24 doi: 10.1007/s11884-016-0345-8.
urinary microbiome in men with positive versus negative biopsies for prostate Thomas-White, K., Forster, S.C., Kumar, N., Van Kuiken, M., Putonti, C., Stares, M.D.,
cancer. J. Urol. 199 (1), 161–171. https://doi.org/10.1016/j.juro.2017.08.001. Hilt, E.E., Price, T.K., Wolfe, A.J., Lawley, T.D., 2018. Culturing of female bladder
Sihra, N., Goodman, A., Zakri, R., Sahai, A., Malde, S., 2018. Nonantibiotic prevention bacteria reveals an interconnected urogenital microbiota. Nat. Commun. 9 (1), 1557.
and management of recurrent urinary tract infection. Nat. Rev. Microbiol. 15 (12), https://doi.org/10.1038/s41467-018-03968-5.
750–776. https://doi.org/10.1038/s41585-018-0106-x. Thomas-White, K., Taege, S., Limeira, R., Brincat, C., Joyce, C., Hilt, E.E., Mac-Daniel, L.,
Sleator, R.D., 2010. The human superorganism - of microbes and men. Med. Hypotheses Radek, K.A., Brubaker, L., Mueller, E.R., Wolfe, A.J., 2020. Vaginal estrogen therapy
74 (2), 214–215. https://doi.org/10.1016/j.mehy.2009.08.047. is associated with increased Lactobacillus in the urine of postmenopausal women
Sleator, R.D., Hill, C., 2008a. “Bioengineered Bugs” - a patho-biotechnology approach to with overactive bladder symptoms. Am. J. Obstet. Gynecol. 223 (5), 727.e1–727.
probiotic research and applications. Med. Hypotheses 70 (1), 167–169. https://doi. e11. https://doi.org/10.1016/j.ajog.2020.08.006.
org/10.1016/j.mehy.2007.03.008. Tikhonov, I., Rebenok, A., Chyzh, A., 1997. A study of interleukin-8 and defensins in
Sleator, R.D., Hill, C., 2008b. Engineered pharmabiotics with improved therapeutic urine and plasma of patients with pyelonephritis and glomerulonephritis. Nephrol.
potential. Hum. Vaccin. 4 (4), 271–274. https://doi.org/10.4161/hv.4.4.6315. Dial. Transplant. 12 (12), 2557–2561. https://doi.org/10.1093/ndt/12.12.2557.
Šmajs, D., Micenková, L., Šmarda, J., Vrba, M., Ševčíková, A., Vališová, Z., Totsika, M., Beatson, S.A., Sarkar, S., Phan, M.D., Petty, N.K., Bachmann, N., Szubert, M.,
Woznicová, V., 2010. Bacteriocin synthesis in uropathogenic and commensal Sidjabat, H.E., Paterson, D.L., Upton, M., Schembri, M.A., 2011. Insights into a
Escherichia coli: colicin E1 is a potential virulence factor. BMC Microbiol 10. multidrug resistant Escherichia coli pathogen of the globally disseminated ST131
https://doi.org/10.1186/1471-2180-10-288. lineage: genome analysis and virulence mechanisms. PLoS ONE 6 (10). https://doi.
Smelov, V., Naber, K., Bjerklund Johansen, T.E., 2016. Improved classification of urinary org/10.1371/journal.pone.0026578.
tract infection: future considerations. Eur. Urol. Suppl. 15 (4), 71–80. https://doi. Townes, C.L., Ali, A., Robson, W., Pickard, R., Hall, J., 2011. Tolerance of bacteriuria
org/10.1016/j.eursup.2016.04.002. after urinary diversion is linked to antimicrobial peptide activity. Urology 77 (2),
Soltani, S., Hammami, R., Cotter, P.D., Rebuffat, S., Said, L., Ben, Gaudreau, H., 509.e1–509.e8. https://doi.org/10.1016/j.urology.2010.08.019.
Bédard, F., Biron, E., Drider, D., Fliss, I., 2020. Bacteriocins as a new generation of Trautner, B.W., Hull, R.A., Darouiche, R.O., 2003. Escherichia coli 83972 inhibits
antimicrobials: toxicity aspects and regulations. FEMS Microbiol. Rev. 45 (1), 1–24. catheter adherence by a broad spectrum of uropathogens. Urology 61 (5),
https://doi.org/10.1093/femsre/fuaa039. 1059–1062. https://doi.org/10.1016/S0090-4295(02)02555-4.
Soto, S.M., 2014. Importance of biofilms in urinary tract infections: new therapeutic Trautner, B.W., Hull, R.A., Darouiche, R.O., 2005. Colicins prevent colonization of
approaches. Adv. Biol. 2014, 1–13 doi: 10.1155/2014/543974. urinary catheters. J. Antimicrob. Chemother. 56 (2), 413–415. https://doi.org/
Spencer, J.D., Hains, D.S., Porter, E., Bevins, C.L., DiRosario, J., Becknell, B., Wang, H., 10.1093/jac/dki228.
Schwaderer, A.L., 2012. Human alpha defensin 5 expression in the human kidney

18
C. Kenneally et al. Microbiological Research 259 (2022) 127010

Trivedi, D., Jena, P.K., Seshadri, S., 2014. Colicin E2 expression in lactobacillus brevis Williams, G.D., 2015. Two cases of urinary tract infection caused by Propionimicrobium
DT24, a vaginal probiotic isolate, against uropathogenic Escherichia coli. ISRN Urol. lymphophilum. J. Clin. Microbiol. 53 (9), 3077–3080. https://doi.org/10.1128/
2014, 1–7. https://doi.org/10.1155/2014/869610. JCM.00438-15.
Trizna, E.Y., Yarullina, M.N., Baidamshina, D.R., Mironova, A.V., Akhatova, F.S., Wojciuk, B., Salabura, A., Grygorcewicz, B., Kędzierska, K., Ciechanowski, K.,
Rozhina, E.V., Fakhrullin, R.F., Khabibrakhmanova, A.M., Kurbangalieva, A.R., Dołęgowska, B., 2019. Urobiome: in sickness and in health. Microorganisms 7 (11),
Bogachev, M.I., Kayumov, A.R., 2020. Bidirectional alterations in antibiotics 548. https://doi.org/10.3390/microorganisms7110548.
susceptibility in Staphylococcus aureus—pseudomonas aeruginosa dual-species Wolfe, A.J., Brubaker, L., 2015. Sterile urine and the presence of bacteria. Eur. Urol. 68
biofilm. Sci. Rep. 10 (1), 1–18. https://doi.org/10.1038/s41598-020-71834-w. (2), 173–174. https://doi.org/10.1016/j.eururo.2015.02.041.
Ujmajuridze, A., Chanishvili, N., Goderdzishvili, M., Leitner, L., Mehnert, U., Wolfe, A.J., Toh, E., Shibata, N., Rong, R., Kenton, K., FitzGerald, M.P., Mueller, E.R.,
Chkhotua, A., Kessler, T.M., Sybesma, W., 2018. Adapted bacteriophages for treating Schreckenberger, P., Dong, Q., Nelson, D.E., Brubaker, L., 2012. Evidence of
urinary tract infections. Front. Microbiol. 9 (1832), 1–7. https://doi.org/10.3389/ uncultivated bacteria in the adult female bladder. J. Clin. Microbiol. 50 (4),
fmicb.2018.01832. 1376–1383. https://doi.org/10.1128/JCM.05852-11.
Vagios, S., Hesham, H., Mitchell, C., 2020. Understanding the potential of lactobacilli in Wolff, B.J., Price, T.K., Joyce, C.J., Wolfe, A.J., Mueller, E.R., 2019. Oral probiotics and
recurrent UTI prevention. Microb. Pathog. 148, 104544 https://doi.org/10.1016/j. the female urinary microbiome: a double-blinded randomized placebo-controlled
micpath.2020.104544. trial. Int. Urol. Nephrol. 51 (12), 2149–2159. https://doi.org/10.1007/s11255-019-
Van de Peer, Y., Chapelle, S., De Wachter, R., 1996. A quantitative map of nucleotide 02282-3.
substitution rates in bacterial rRNA. Nucleic Acids Res. 24 (17), 3381–3391. https:// Wright, R.C.T., Friman, V.P., Smith, M.C.M., Brockhurst, M.A., 2019. Resistance
doi.org/10.1093/nar/24.17.3381. evolution against phage combinations depends on the timing and order of exposure.
Vassiliadis, G., Destoumieux-Garzón, D., Lombard, C., Rebuffat, S., Peduzzi, J., 2010. mBio 10 (5). https://doi.org/10.1128/mBio.01652-19.
Isolation and characterization of two members of the siderophore-microcin family, Yazdi, M., Bouzari, M., Ghaemi, E.A., Shahin, K., 2020. Isolation, characterization and
microcins M and H47. Antimicrob. Agents Chemother. 54 (1), 288–297. https://doi. genomic analysis of a novel bacteriophage VB_EcoS-golestan infecting multidrug-
org/10.1128/AAC.00744-09. resistant escherichia coli isolated from urinary tract infection. Sci. Rep. 10 (1), 1–13.
Vejborg, R.M., Friis, C., Hancock, V., Schembri, M.A., Klemm, P., 2010. A virulent parent https://doi.org/10.1038/s41598-020-63048-x.
with probiotic progeny: comparative genomics of Escherichia coli strains CFT073, Yeaman, M.R., Yount, N.Y., 2003. Mechanisms of antimicrobial peptide action and
Nissle 1917 and ABU 83972. Mol. Genet. Genom. 283 (5), 469–484 doi: 10.1007/ resistance. Pharmacol. Rev. 55 (1), 27–55. https://doi.org/10.1124/pr.55.1.2.
s00438-010-0532-9. Yu, H., Ma, Z., Meng, S., Qiao, S., Zeng, X., Tong, Z., Jeong, K.C., 2021. A novel
Velraeds, M.M.C., Van De Belt-Gritter, B., Van Der Mei, H.C., Reid, G., Busscher, H.J., nanohybrid antimicrobial based on chitosan nanoparticles and antimicrobial peptide
1998. Interference in initial adhesion of uropathogenic bacteria and yeasts to microcin J25 with low toxicity. Carbohydr. Polym. 253, 117309 https://doi.org/
silicone rubber by a Lactobacillus acidophilus biosurfactant. J. Med. Microbiol. 47 10.1016/j.carbpol.2020.117309.
(12), 1081–1085. https://doi.org/10.1099/00222615-47-12-1081. Yu, Z., Morrison, M., 2004. Comparisons of different hypervariable regions of rrs genes
Visconti, A., Le Roy, C.I., Rosa, F., Rossi, N., Martin, T.C., Mohney, R.P., Li, W., de for use in fingerprinting of microbial communities by PCR-denaturing gradient gel
Rinaldis, E., Bell, J.T., Venter, J.C., Nelson, K.E., Spector, T.D., Falchi, M., 2019. electrophoresis. Appl. Environ. Microbiol. 70 (8), 4800–4806 doi: 10.1128/
Interplay between the human gut microbiome and host metabolism. Nat. Commun. AEM.70.8.4800.
10 (1) https://doi.org/10.1038/s41467-019-12476-z. Yuan, J., Zhang, Z.Z., Chen, X.Z., Yang, W., Huan, L.D., 2004. Site-directed mutagenesis
Wagenlehner, F., Tandogdu, Z., Bartoletti, R., Cai, T., Cek, M., Kulchavenya, E., of the hinge region of nisinZ and properties of nisinZ mutants. Appl. Microbiol.
Köves, B., Naber, K., Perepanova, T., Tenke, P., Wullt, B., Bogenhard, F., Biotechnol. 64 (6), 806–815. https://doi.org/10.1007/s00253-004-1599-1.
Johansen, T.E.B., 2016. The global prevalence of infections in urology study: a long- de Zamaroczy, M., Chauleau, M., 2011. Colicin killing: foiled cell defense and hijacked
term,worldwide surveillance study on urological infections. Pathogens 5 (1), 1–8. cell functions. Prokaryotic Antimicrobial Peptides. Springer, New York,
https://doi.org/10.3390/pathogens5010010. pp. 255–287. https://doi.org/10.1007/978-1-4419-7692-5_14.
Wang, A.P., Su, Y.P., Wang, S., Shen, M.Q., Chen, F., Chen, M., Ran, X.Z., Chen, T.M., Zandbergen, L.E., Halverson, T., Brons, J.K., Wolfe, A.J., De Vos, M.G.J., 2021. ‘The good
Wang, J.P., 2010. Antibacterial activity and mechanism of recombinant human - and the bad: Ecological interaction measurements between the urinary microbiota
defensin 5 against clinical antibiotic-resistant strains. Afr. J. Microbiol. Res. 4 (8), and uropathogens’. Front. Microbiol. 12 (May), 1026. https://doi.org/10.3389/
626–633. fmicb.2021.659450.
Wang, Y., 2002. The function of OmpA in Escherichia coli. Biochem. Biophys. Res. Zasloff, M., 2007. Antimicrobial peptides, innate immunity, and the normally sterile
Commun. 292 (2), 396–401. https://doi.org/10.1006/bbrc.2002.6657. urinary tract. J. Am. Soc. Nephrol. 18 (11), 2810–2816. https://doi.org/10.1681/
Werneburg, G.T., Nguyen, A., Henderson, N.S., Rackley, R.R., Shoskes, D.A., Le Sueur, A. ASN.2007050611.
L., Corcoran, A.T., Katz, A.E., Kim, J., Rohan, A.J., Thanassi, D.G., 2020. The natural Zdziarski, J., Svanborg, C., Wullt, B., Hacker, J., Dobrindt, U., 2008. Molecular basis of
history and composition of urinary catheter biofilms: early uropathogen colonization commensalism in the urinary tract: low virulence or virulence attenuation? Infect.
with intraluminal and distal predominance. J. Urol. 203 (2), 357–364. https://doi. Immun. 76 (2), 695–703. https://doi.org/10.1128/IAI.01215-07.
org/10.1097/JU.0000000000000492. Zdziarski, J., Brzuszkiewicz, E., Wullt, B., Liesegang, H., Biran, D., Voigt, B., Grönberg-
Whiteside, S.A., Razvi, H., Dave, S., Reid, G., Burton, J.P., 2015. The microbiome of the Hernandez, J., Ragnarsdottir, B., Hecker, M., Ron, E.Z., Daniel, R., Gottschalk, G.,
urinary tract - a role beyond infection. Nat. Rev. Urol. 12 (2), 81–90. https://doi. Hacker, J., Svanborg, C., Dobrindt, U., 2010. Host imprints on bacterial genomes-
org/10.1038/nrurol.2014.361. rapid, divergent evolution in individual patients. PLoS Pathog. 6 (8), 95–96. https://
doi.org/10.1371/journal.ppat.1001078.

19

You might also like